+ All Categories
Home > Documents > EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

Date post: 08-Feb-2016
Category:
Upload: rajkaran-moorthy
View: 40 times
Download: 0 times
Share this document with a friend
Description:
EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34.pdf
Popular Tags:
283
University of Kentucky UKnowledge eses and Dissertations--Physiology Physiology 2013 EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THEPEUTIC APPROACH FOR THE TREATMENT OF TUMATIC BIN INJURY Shaun W. Carlson University of Kentucky, [email protected] is Doctoral Dissertation is brought to you for free and open access by the Physiology at UKnowledge. It has been accepted for inclusion in eses and Dissertations--Physiology by an authorized administrator of UKnowledge. For more information, please contact [email protected]. Recommended Citation Carlson, Shaun W., "EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THEPEUTIC APPROACH FOR THE TREATMENT OF TUMATIC BIN INJURY" (2013). eses and Dissertations--Physiology. Paper 9. hp://uknowledge.uky.edu/physiology_etds/9
Transcript
Page 1: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

University of KentuckyUKnowledge

Theses and Dissertations--Physiology Physiology

2013

EVALUATION OF INSULIN-LIKE GROWTHFACTOR-1 AS A THERAPEUTIC APPROACHFOR THE TREATMENT OF TRAUMATICBRAIN INJURYShaun W. CarlsonUniversity of Kentucky, [email protected]

This Doctoral Dissertation is brought to you for free and open access by the Physiology at UKnowledge. It has been accepted for inclusion in Thesesand Dissertations--Physiology by an authorized administrator of UKnowledge. For more information, please contact [email protected].

Recommended CitationCarlson, Shaun W., "EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPROACH FOR THETREATMENT OF TRAUMATIC BRAIN INJURY" (2013). Theses and Dissertations--Physiology. Paper 9.http://uknowledge.uky.edu/physiology_etds/9

Page 2: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

STUDENT AGREEMENT:

I represent that my thesis or dissertation and abstract are my original work. Proper attribution has beengiven to all outside sources. I understand that I am solely responsible for obtaining any needed copyrightpermissions. I have obtained and attached hereto needed written permission statements(s) from theowner(s) of each third‐party copyrighted matter to be included in my work, allowing electronicdistribution (if such use is not permitted by the fair use doctrine).

I hereby grant to The University of Kentucky and its agents the non-exclusive license to archive and makeaccessible my work in whole or in part in all forms of media, now or hereafter known. I agree that thedocument mentioned above may be made available immediately for worldwide access unless apreapproved embargo applies.

I retain all other ownership rights to the copyright of my work. I also retain the right to use in futureworks (such as articles or books) all or part of my work. I understand that I am free to register thecopyright to my work.

REVIEW, APPROVAL AND ACCEPTANCE

The document mentioned above has been reviewed and accepted by the student’s advisor, on behalf ofthe advisory committee, and by the Director of Graduate Studies (DGS), on behalf of the program; weverify that this is the final, approved version of the student’s dissertation including all changes requiredby the advisory committee. The undersigned agree to abide by the statements above.

Shaun W. Carlson, Student

Dr. Kathryn E. Saatman, Major Professor

Dr. Bret N. Smith, Director of Graduate Studies

Page 3: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPROACH FOR THE TREATMENT OF TRAUMATIC BRAIN INJURY

DISSERTATION

A dissertation submitted in partial fulfillment of the requirements for the degree of Doctor of Philosophy in the

College of Medicine, Department of Physiology at the University of Kentucky

By

Shaun William Carlson

Lexington, Kentucky

Director: Dr. Kathryn E. Saatman, Professor of Physiology

Lexington, Kentucky

2013

Copyright © Shaun William Carlson 2013

Page 4: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

ABSTRACT OF DISSERTATION

EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPROACH FOR THE TREATMENT OF TRAUMATIC BRAIN INJURY

Traumatic brain injury (TBI) is a prevalent CNS neurodegenerative condition that

results in lasting neurological dysfunction, including potentially debilitating cognitive impairments. Despite the advancements in understanding the complex damage that can culminate in cellular dysfunction and loss, no therapeutic treatment has been effective in clinical trials, highlighting that new approaches are desperately needed. A therapy that limits cell death while simultaneously promoting reparative mechanisms, including post-traumatic neurogenesis, in the injured brain may have maximum effectiveness in improving recovery of function after TBI.

Insulin-like growth factor-1 (IGF-1) is a potent growth factor that has previously been shown to promote recovery of function after TBI, but no studies have evaluated the efficacy of IGF-1 to promote cell survival and modulate neurogenesis following brain injury. Systemic infusion of IGF-1 resulted in undetectable levels of IGF-1 in the brain, but did promote increased cortical activation of Akt, a pro-survival downstream mediator of IGF-1 signaling, in mice subjected to controlled cortical impact (CCI), a well-established model of contusion TBI. However, systemic infusion of IGF-1 did not promote recovery of motor function in mice after CCI. A one week central infusion of IGF-1 elevated brain levels of IGF-1, increased Akt activation and improved motor and cognitive function after CCI. Central infusion of IGF-1 also significantly increased immature neuron density at 7 d post-injury for a range of doses and when administered with a clinically relevant delayed onset of 6 hr post-injury. To mitigate potential side effects of central infusion, an alternative conditional astrocyte-specific IGF-1 overexpressing mouse model was utilized to evaluate the efficacy of IGF-1 to promote post-traumatic neurogenesis. Overexpression of IGF-1 did not protect against acute immature neuron loss, but did increase immature neuron density above uninjured levels at 10 d post-injury. The increase in immature neuron density appeared to be driven by enhanced neuronal differentiation. In wildtype mice, immature neurons exhibited injury-induced reductions in dendritic arbor complexity following severe CCI, a previously unknown pathological phenomenon. Overexpression of IGF-1 in brain-injured mice promoted the restoration of dendritic arbor complexity to the dendritic morphology observed in uninjured mice. Together, these findings provide strong evidence that treatment with IGF-1 promotes the recovery of neurobehavioral function and enhances post-traumatic neurogenesis in a mouse model of contusion TBI.

Page 5: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

KEYWORDS: Traumatic Brain Injury, Insulin-like Growth Factor-1, Neurogenesis, Contusion, Neurobehavioral Function

Shaun W. Carlson__________________ Shaun W. Carlson

_July 5, 2013 _ ___ ________________

Date

Page 6: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPROACH FOR THE TREATMENT OF TRAUMATIC BRAIN INJURY

By

Shaun William Carlson

Kathryn E. Saatman ___ ____ Director of Dissertation

_Bret N. Smith ______________

Director of Graduate Studies

_July 5, 2013 __________ Date

Page 7: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

iii

ACKNOWLEDGMENTS

I wish to thank my mentor, Dr. Kathryn Saatman, for her continuous support and

guidance as she is an excellent mentor that fosters continual growth. It is with her

training and pursuit of excellence that I have become the scientist I am today. I would

also like to thank Drs. Edward Hall, Bret Smith, and Alexander Rabchevsky for their

valuable input and advice that helped improve the thesis work and guided me to think

critically and improve the quality of my work. I would also like to thank my outside

examiner Dr. Jimmi Hatton-Kolpek, who has provided valuable insight into enhancing the

clinical applicability of this project and future work.

I am especially appreciative of Dr. Sindhu Kizhakke-Madathil for her instruction

and support when I started in the laboratory and in the following years as I completed my

thesis work. I would also like to thank the many current and former members of Dr.

Saatman’s laboratory. Thank you to the many members of the Department of Physiology

and the Spinal Cord and Brain Injury research Center who have made this an enjoyable

time at the University of Kentucky.

This endeavor would not have been possible without the support from my close

friends and family. To Mike Fiandalo, I always looked forward to our science talks over a

cup of coffee and to your enthusiasm and support as we each completed our work. To

my parents, Dennis and Sandra Carlson and to my brother Jon Carlson, thank you for

your unending support and for always encouraging me to fully achieve my aspirations. I

also wish to thank my fiancé, Valerie Reeves. Valerie, thank you for being the pillar of

strength that helped make this journey possible and enjoyable. You have provided

endless encouragement and valuable input that has helped shape not only my thesis

work, but also the scientist, and more importantly, the person I am today. I look forward

to our future adventures in both our scientific careers and in life.

Page 8: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

iv

TABLE OF CONTENTS

Acknowledgments .......................................................................................................... iii

Table of Contents ........................................................................................................... iv

List of Tables ................................................................................................................. vii

List of Figures ............................................................................................................... viii

Chapter 1: Introduction ................................................................................................... 1 Traumatic Brain Injury Definition and Epidemiology ........................................................ 1 Considerations for Clinical Classification and Treatment of TBI ...................................... 1 Traumatic Brain Injury Pathology .................................................................................... 2 Primary Damage and Types of Injury.............................................................................. 5 Secondary Injury ............................................................................................................ 5 Neuronal Support System ............................................................................................... 6 TBI Models ..................................................................................................................... 8 Diffuse Models ................................................................................................................ 9 Focal Models: Controlled Cortical Impact ......................................................................10 CCI Pathology ...............................................................................................................13 Therapeutic Strategies for TBI .......................................................................................15 Recovery and plasticity in the brain after CCI ................................................................15 Insulin-like growth factor-1 .............................................................................................18 IGF-1 in the Brain ..........................................................................................................21 IGF-1 and Acute CNS Injury ..........................................................................................23 IGF-1 and TBI ................................................................................................................24 Endogenous changes in the IGF-1 axis after Experimental TBI .....................................24 Administration of IGF-1 after Experimental TBI ..............................................................25 IGF-1 Administration after Clinical TBI ...........................................................................25 Neurogenesis ................................................................................................................28 Neurogenesis and TBI ...................................................................................................31 IGF-1 and Neurogenesis ...............................................................................................32 Specific Aims and Hypothesis of the Dissertation ..........................................................33

Chapter 2: Rate of Neurodegeneration in the Mouse Controlled Cortical Impact Model is Influenced by Impactor Tip Shape: Implications for Mechanistic and Therapeutic Studies ......................................................................................................................................36 Introduction ...................................................................................................................36 Materials and Methods ..................................................................................................40 Finite Element Mouse Brain Model Development and CCI Simulations .........................40 Animals and Surgical Procedures ..................................................................................41 Tissue Preparation ........................................................................................................42 Cortical Contusion Volume ............................................................................................43 Regional Hippocampal Neurodegeneration ...................................................................43 Immunohistochemistry ...................................................................................................44 Immunoglobulin (IgG) ....................................................................................................44 Amyloid Precursor Protein .............................................................................................45 Behavioral Testing .........................................................................................................45 Neurological Severity Score ..........................................................................................45 Neuroscore ....................................................................................................................46 Morris Water Maze Cognitive Test .................................................................................47 Statistical Analysis .........................................................................................................47

Page 9: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

v

Results ..........................................................................................................................49 Finite element simulation of cortical impact injury ..........................................................49 Histological damage in Nissl-stained brain sections.......................................................53 Quantification of cortical tissue damage ........................................................................58 Regional hippocampal neurodegeneration ....................................................................61 Blood-brain barrier (BBB) damage .................................................................................65 Traumatic axonal injury .................................................................................................67 Motor and cognitive behavioral outcome .......................................................................69 Discussion .....................................................................................................................71

Chapter 3: Prolonged Continuous Systemic Infusion of IGF-1 after Severe Controlled Cortical Impact ..............................................................................................................79 Introduction ...................................................................................................................79 Materials and Methods ..................................................................................................83 Animals .........................................................................................................................83 Controlled Cortical Impact and Minipump Implantation ..................................................83 Motor Function Assessment ..........................................................................................84 Tissue Collection and Preparation .................................................................................85 Quantification of Human IGF-1 by ELISA ......................................................................86 Western Blot ..................................................................................................................86 Cresyl Violet and Fluorojade-B Staining ........................................................................87 Immunohistochemistry ...................................................................................................88 Image Acquisition and Quantification .............................................................................88 Statistical Analysis .........................................................................................................89 Results ..........................................................................................................................90 Discussion .....................................................................................................................98

Chapter 4: Central Infusion of IGF-1 Improves Neurobehavioral Function and Increases Immature Neuron Density following Controlled Cortical Impact ................................... 108 Introduction ................................................................................................................. 108 Materials and Methods ................................................................................................ 112 Animals ....................................................................................................................... 112 Controlled Cortical Impact and Central Infusion of Human IGF-1 ................................. 112 Central Infusion of IGF-1 ............................................................................................. 113 Study 1 (Effects of 10 µg/d hIGF-1 Infusion on Behavior and Hippocampal Neuron Survival) ...................................................................................................................... 114 Study 2 (Effects of 10 µg/d hIGF-1 on Regional Cerebral Edema) ............................... 115 Study 3 (Delayed Onset of hIGF-1 Infusion) ................................................................ 115 Study 4 (Dose Response for hIGF-1) .......................................................................... 116 Study 5 (Effect of Changing Infusate Flow Rate in Sham-injured Mice) ....................... 116 Study 6 (Efficacy of 3 µg/d hIGF-1 Infused at a Reduced Rate) ................................... 116 Motor Function Assessment Using a Modified Neurological Severity Score (NSS) ...... 117 Cognitive Performance Evaluation Using a Novel Object Recognition (NOR) Paradigm117 Tissue Collection ......................................................................................................... 118 Preparation of Tissue for ELISA and Western Blotting ................................................. 119 Quantification of IGF-1 by ELISA ................................................................................. 119 Western Blot ................................................................................................................ 120 Histological (Cresyl Violet) Staining ............................................................................. 120 Immunohistochemistry ................................................................................................. 121 Image Acquisition ........................................................................................................ 121 Cellular Quantification ................................................................................................. 122

Page 10: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

vi

Statistical Analysis ....................................................................................................... 123 Results ........................................................................................................................ 124 Discussion ................................................................................................................... 149

Chapter 5: Targeted Astrocyte-specific Conditional IGF-1 Overexpression Enhances Post-Traumatic Neurogenesis by Promoting Neuronal Differentiation ......................... 162 Introduction ................................................................................................................. 162 Materials and Methods ................................................................................................ 167 Animals ....................................................................................................................... 167 Controlled Cortical Impact ........................................................................................... 168 Tissue Collection and Processing ................................................................................ 169 Immunohistochemistry ................................................................................................. 169 Image Acquisition and Quantification ........................................................................... 170 Scholl Analysis ............................................................................................................ 171 Statistical Analysis ....................................................................................................... 172 Results ........................................................................................................................ 173 Discussion ................................................................................................................... 185

Chapter 6: Summary of Dissertation ............................................................................ 194 Controlled Cortical Impact and Impactor Tip Geometry ............................................... 195 Levels of Human IGF-1 in the Brain and Systemic Circulation after CCI ...................... 196 IGF-1 Signaling in the Brain after CCI ......................................................................... 199 Capacity of IGF-1 to Promote Survival of Hippocampal Immature Neurons ................. 199 Susceptibility of Immature Neurons after CCI .............................................................. 200 Calpain-mediated Proteolysis in the Hippocampal Neurogenic Niche .......................... 201 Recovery of Hippocampal Immature Neurons after CCI .............................................. 205 Proliferative Response of the Neurogenic Niche with Treatment of IGF-1 after CCI .... 206 IGF-1-mediated Enhancement of Neuronal Differentiation after CCI ........................... 207 Structural Plasticity with Treatment of IGF-1 after CCI ................................................. 210 Improvements in Motor Function with Treatment of IGF-1 after CCI ............................ 213 Improvements in Cognitive Function with Treatment of IGF-1 after CCI ...................... 214 Long-term Assessments of Cognitive Improvement after Treatment with IGF-1 .......... 215 Generation of Oligodendrocytes in the Injured Hippocampus and IGF-1 ..................... 216 Therapeutic Strategies for IGF-1 after TBI ................................................................... 217 Administration of IGF-1 Encapsulated PLGA Microspheres after TBI .......................... 220 Treatment with PEGylated IGF-1 after TBI .................................................................. 222 Intranasal Delivery of IGF-1 after TBI .......................................................................... 223 Final Conclusions ........................................................................................................ 223

Appendix 1: Long-term Cognitive Deficit following Severe Controlled Cortical Impact as Assessed by the Novel Object Recognition (NOR) Task ............................................. 225

Appendix 2: Formulation of Poly(lactic-co-glycolytic acid) (PLGA) Microspheres for the Development of a Subcutaneously-injected Sustained Release Delivery System for IGF-1 .................................................................................................................................. 227

Appendix 3: Permission for Article Reprint for Chapter 2. ............................................ 233

References .................................................................................................................. 234

Curriculum Vitae .......................................................................................................... 266

Page 11: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

vii

LIST OF TABLES

Table 4.1: Blood glucose levels were elevated in both vehicle (Veh) and insulin-like growth factor-1 (IGF-1) treated mice at an acute time point following sham injury (Sham) or controlled cortical impact (CCI) ................................................................................ 127

Page 12: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

viii

LIST OF FIGURES

Figure 1.1: Progression of primary and secondary injury mechanisms that can occur or become initiated within seconds to days following TBI. ................................................... 4

Figure 1.2: Controlled Cortical Impact (CCI) injury in the mouse positioned over the somatosensory cortex. ..................................................................................................12

Figure1.3: IGF-1 binding to its cognate receptor (IGFR) results in the autophosphorylation of the receptor and recruitment of the insulin response subunit (IRS) and growth factor receptor bound protein 2 (Grb2). ..............................................20

Figure 1.4: Hippocampal neurogenesis in the dentate gyrus granular layer. Proliferation of radial stem cells and neuronal differentiation of newly proliferated neural progenitor cells leads to the generation of immature neurons .........................................................30

Figure 2.1. Finite element model of mouse brain and impactor .....................................50

Figure 2.2. Effect of impactor shape on brain tissue strains ..........................................51

Figure 2.3. Strain profile as a function of cortical depth and tip geometry .....................52

Figure 2.4. Temporal progression of histological damage created by a flat tip versus a rounded tip CCI impactor ...............................................................................................55

Figure 2.5. Alterations in neocortical neuron morphology as a function of impactor tip shape ............................................................................................................................57

Figure 2.6. Quantification of cortical tissue damage ......................................................60

Figure 2.7. Hippocampal neurodegeneration after controlled cortical impact ...............62

Figure 2.8. Quantification of regional hippocampal neurodegeneration as a function of impactor tip shape .........................................................................................................64

Figure 2.9. Acute blood-brain barrier damage after severe controlled cortical impact using a flat tip impactor (A, C) or a rounded tip impactor (B, D). ....................................66

Figure 2.10. Traumatic axonal injury induced by injury with either a flat tip (A, C) or rounded tip (B, D) impactor. ...........................................................................................68

Figure 2.11. Effect of impactor tip shape on neurological motor and cognitive function of mice subjected to cortical impact injury ..........................................................................70

Figure 3.1: Systemic infusion of human insulin-like growth factor-1 (hIGF-1) for a period of 7 d promotes cortical activation of Akt, but does not reduce cortical neuron cytoskeletal protein loss after severe controlled cortical impact (CCI). ...........................92

Figure 3.2: Systemic infusion of human insulin-like growth factor-1 (hIGF-1) did not improve recovery of motor function following severe controlled cortical impact (CCI) ....94

Figure 3.3: Systemic infusion of human insulin-like growth factor-1 (hIGF-1) does not reduce tissue damage or regional cell loss at 3 d following severe controlled cortical impact (CCI) ..................................................................................................................97

Figure 4.1: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) over 7 d elevates brain levels of hIGF-1 and enhances Akt activation in the hippocampus after severe controlled cortical impact (CCI) ........................................................................ 125

Page 13: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

ix

Figure 4.2: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) over 7 d attenuates cognitive and motor impairment after severe controlled cortical impact (CCI) .................................................................................................................................... 130

Figure 4.3: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) protects immature neurons in the injured hippocampus after severe controlled cortical impact (CCI) ................................................................................................................ 132

Figure 4.4: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) promotes brain swelling and deformation in a subset of mice after severe controlled cortical impact (CCI) .................................................................................................... 135

Figure 4.6: Reductions in the concentration of centrally infused human insulin-like growth factor-1 (hIGF-1) appeared to reduce the severity and incidence of brain swelling following severe controlled cortical impact (CCI) ......................................................... 139

Figure 4.7: Central infusion of human insulin-like growth factor-1 (IGF-1) enhances hippocampal immature neuron density in an apparent dose-dependent manner after controlled cortical impact ............................................................................................. 141

Figure 4.8: Volume of infusate introduced into the influences brain swelling after craniotomy ................................................................................................................... 143

Figure 4.9 (cont.): Central infusion of 3 µg/d human insulin-like growth factor-1 (IGF-1) via a lower infusion rate (0.11 µL/hr) resulted in modest infusion-related brain swelling and enhanced post-traumatic neurogenesis at 7 d following severe controlled cortical impact (CCI) ................................................................................................................ 148

Figure 5.1: IGF-1 overexpression increases immature neuron density in the dentate gyrus granular layer following controlled cortical impact .............................................. 175

Figure 5.2: IGF-1 overexpression does not enhance brain injury-induced cellular proliferation in the hippocampal granular layer following controlled cortical impact. ..... 178

Figure 5.3: IGF-1 overexpression promotes neuronal differentiation of newly proliferated cells in the granular layer following controlled cortical impact ...................................... 181

Figure 5.4: IGF-1 overexpression restores immature neuron dendritic arbor complexity after controlled cortical impact (CCI). ........................................................................... 184

Fig. 6.1: Calpain-mediated spectrin cleavage was observed in nestin-positive, but not DCx-positive cells in the granular layer at 24 hr following controlled cortical impact (CCI) .................................................................................................................................... 204

Figure A1.1: Severe controlled cortical impact (CCI) results in cognitive deficits that persist for up to 6 weeks post-injury ............................................................................ 226

Figure A2.1: Protocol for the development of insulin-like growth factor-1 (IGF-1) encapsulated poly(lactic-co-glycolytic acid) (PLGA) microspheres by solvent extraction .................................................................................................................................... 230

Figure A2.2: Morphology and size of poly(lactic-co-glycolytic acid) (PLGA) microspheres generated by solvent extraction ................................................................................... 231

Figure A2.3: Morphology, size and release kinetics of insulin encapsulated poly(lactic-co-glycolytic acid) (PLGA) microspheres generated by spray drying ........................... 232

Page 14: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

1

Chapter 1: Introduction

Traumatic Brain Injury Definition and Epidemiology

Traumatic brain injury (TBI) is defined as an acquired insult following an initial

external, mechanical trauma that consequently results in damage to the brain (NINDS

2012). TBI is a leading cause of death and disability in the United States, with an

estimated 1.7 million cases annually (Summers et al. 2009; Faul M 2010). Stratification

of reported cases reveals that approximately 250,000 individuals are hospitalized and

50,000 patients do not survive their TBI each year (Langlois et al. 2006; Corrigan et al.

2010). Motor vehicle and transportation-related incidences, falls, and violence are the

leading causes of TBI (Thurman et al. 1999; Langlois et al. 2006). TBI afflicts individuals

independent of age and gender; however, the highest prevalence is reported in males

aged 20-30 (Langlois et al. 2006). A recent rise in the incidence of TBIs has also been

reported in individuals over the age of 65 (Thompson et al. 2006). In the United States,

the financial burden of TBI, including direct and indirect expenses, in 2000 alone was

over $76 billion (Finkelstein E 2006; Coronado et al. 2011).

Considerations for Clinical Classification and Treatment of TBI

The definition of TBI encompasses an array of pathoanatomical types of injury,

including contusion, hematoma, sub-arachnoid hemorrhage, and diffuse axonal injury.

The heterogeneity of TBI is a substantial hurdle for the development of effective

therapeutic strategies for treatment of TBI. The Glasgow Coma Scale (GCS) and

neuroimaging, including computed tomography scanning and magnetic resonance

imaging, of physical damage have been utilized to characterize injury severity, provide

improved prognosis and define clinical management of TBI (Teasdale and Jennett 1976;

Marshall et al. 1992; Maas et al. 2005). However, these diagnostic metrics to not provide

insight into the pathological mechanisms that produce neurological impairments that is

Page 15: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

2

necessary for implementation of therapeutic interventions tailored for specific

pathoanatomical types of TBI (Saatman et al. 2008).

Despite our increased understanding of TBI and the development of therapeutic

agents, clinical trials have continued to fail (Maas et al. 2007). Several factors have been

implicated in contributing to the clinical trial failures, including delayed treatment onset,

enrollment of patients in clinical trials based on injury severity determined by the GCS,

and a disconnect between injury heterogeneity and targeted therapeutic intervention

(Maas et al. 2007; Maas et al. 2008). The staggering number of clinical trial failures

highlights the need for improved diagnostic tools, such as biomarkers, to improve

classification parameters in the clinic based on pathoanatomical features and to pair

treatment strategies tailored to a specific pathoanatomical injury type (Saatman et al.

2008). In addition, the multitude of pathological processes after TBI may undermine the

efficacy of therapeutic agents aimed at targeting only a single process of injury.

However, therapeutic strategies designed to mitigate multiple pathological processes

may be efficacious for the treatment of TBI (Margulies and Hicks 2009).

Traumatic Brain Injury Pathology

Symptoms of a TBI can manifest as an innocuous headache, mild concussion,

prolonged coma, or death. Cognitive impairment is the most frequent and debilitating

functional consequence of TBI (Levin 1998; Lundin et al. 2006). Impairments in both

motor and cognitive function have been reported for weeks and years after a TBI, with

approximately 40% of patients developing long-term disabilities after being discharged

from the hospital (Capruso and Levin 1992; Levin 1998; Lundin et al. 2006; Selassie et

al. 2008; Jang 2009). Years after sustaining a TBI, patients often report that cognitive

Page 16: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

3

impairment contributes greatly to decreased satisfaction with their quality of life (Truelle

et al. 2010).

TBI is a complex injury that can be divided into two main sources of damage in

terms of cellular and histological pathology: 1) initial primary damage that occurs as an

external mechanical force is transferred to the head and 2) the activation of secondary

injury cascades (Fig. 1.1). Processes involved in recovery after TBI will be discussed in

subsequent sections of this chapter.

Page 17: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

4

Figure 1.1: Progression of primary and secondary injury mechanisms that can occur or become initiated within seconds to days following TBI. Several processes, including neurogenesis and hippocampal plasticity, can be targeted by therapeutic agents to enhance recovery following TBI.

Page 18: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

5

Primary Damage and Types of Injury

Primary damage occurs as a result of shearing, stretching or compression of

cells and vasculature with potential concomitant disruption to cellular membrane integrity

(Maas et al. 1999; Beauchamp et al. 2008). The primary source of injury cannot be

prevented nor targeted for therapeutic intervention, and subsequently leads to the onset

of the second source of injury, the activation of secondary injury cascades.

The types of initial force delivered to the brain can originate from contact with a

penetrating or nonpenetrating object, rapid acceleration/deceleration, waves of energy

emanating from a blast, or a combination of these forces (Maas et al. 2008). Focal injury

is characterized by damage located in a discrete area, while diffuse injury is reflective of

injury potentially distributed in non-discrete regions throughout the brain (Morales et al.

2005; Povlishock and Katz 2005; Andriessen et al. 2010). Focal injury is typically

characterized by the presence of contusions and hematomas while traumatic axonal

injury and edema are defining hallmarks of diffuse injury (Morales et al. 2005). However,

this distinction between focal and diffuse injury pathology can become obscured with

increasing injury severity.

Secondary Injury

Secondary injury is a term used to designate the activation of pathophysiological

mechanisms following the initial primary damage. The type of brain injury can greatly

influence the heterogeneity and duration of the secondary injury mechanisms that

manifest, which synergistically exacerbate injury pathology (McIntosh et al. 1998; Maas

et al. 1999). Each of the secondary injury mechanisms present and persist with varying

duration dependent upon injury type and severity. However, secondary injury cascades

Page 19: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

6

are the target of therapeutic and surgical intervention (Maas et al. 2005; Beauchamp et

al. 2008).

As a consequence of acutely disrupted cellular integrity and loss of ion

homeostasis, brain injury results in unregulated neurotransmitter release and the

activation of intracellular proteases, including calpains, within hours of the primary injury

(Baker et al. 1993; Saatman et al. 1996; Hall et al. 2005). Moreover, high concentrations

of intracellular calcium and the accumulation of free radicals contribute to mitochondria

dysfunction (Singh et al. 2006). Structural damage to endothelial tight junctions that

comprise the blood-brain barrier and the formation of microhemorrhages can both

contribute to brain edema (Sutton et al. 1993; Baldwin et al. 1996; Maas et al. 1999;

Greve and Zink 2009). Moreover, damage to the vasculature compromises cerebral

blood flow resulting in hypoxic-ischemia, impaired delivery of nutrients and, ultimately,

disrupted metabolic status of cells within the injured tissue (Martin et al. 1997).

Consequently, the numerous secondary injury mechanisms can overwhelm injured cells

yielding apoptosis or necrosis depending on the cell type and proximity to the primary

injury site (Rink et al. 1995; Conti et al. 1998; Fox et al. 1998). Cell loss is progressive as

dying cells may propagate dysfunction and death of adjacent cells (Conti et al. 1998;

Pleasant et al. 2011). Secondary axonal degeneration and demyelination can contribute

to neuronal dysfunction (Meaney et al. 2001; Mac Donald et al. 2007). The

consequences of the secondary injury mechanisms are not restricted exclusively to

neurons, but also extend to other cells of the brain.

Neuronal Support System

Collectively, the primary damage and various secondary injury cascades disrupt

neuronal cell function and the neuronal support system, comprised of astrocytes,

Page 20: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

7

endothelial cells, microglia and oligodendrocytes, contributing to neuronal cell loss within

the injured tissue. Glial cells play a critical, but complex role in the response to injury as

their actions can be both beneficial and detrimental to injured cells. Following TBI, glial

cells may mitigate or exacerbate the mechanisms of secondary injury depending on the

proximity to the site of injury and time after injury (Chen and Swanson 2003; Myer et al.

2006; Sofroniew and Vinters 2010; Graeber and Christie 2012). Each type of glial cell

including astrocytes, microglia and oligodendrocytes has unique properties that are

important for normal brain health and responses following injury.

Resident astrocytes are important in preserving homeostatic conditions in the

brain through maintenance of fluid and extracellular ion balance, scavenging of free

radicals, clearance of extracellular glutamate, production of growth factors and

inflammatory mediators, and supplying nutrients to neighboring cells. Reactive

astrocytosis is an endogenous response observed in human and experimental TBI,

characterized by cellular proliferation and hypertrophy, including cytoplasmic

enlargement, elongation of cellular processes, and increased expression of glial fibrillary

acidic protein (GFAP) (Baldwin et al. 1996; Ridet et al. 1997; Castejon 1998; Hausmann

and Betz 2000; Saatman et al. 2006). In the context of CNS trauma, astrocytes can

contain the products of injury, including cellular debris and cytotoxic cellular contents

released from dying cells, by formation of a physical barrier referred to as the glial scar

(Fitch et al. 1999; Myer et al. 2006; Fitch and Silver 2008). Conversely, astrocytes

produce cytokines that propagate inflammation in the brain and chondroitin sulfate

proteoglycan, a component of the glial scar, that inhibits regeneration and repair

mechanisms (Smith et al. 1986; Snow et al. 1990; McKeon et al. 1991; Dong and

Benveniste 2001). While astrocytes can be recruited to the site of injury, they are still

susceptible to mechanisms of injury, including hypoxia and reactive oxygen species that

Page 21: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

8

can promote cell death (Giffard et al. 1990; Swanson et al. 1997; Bondarenko and

Chesler 2001; Zhao et al. 2003).

Microglial cells, the resident immune cells in the CNS, play a critical role in the

surveillance of the CNS extracellular environment. Resident microglia or blood-borne

peripheral macrophages can be recruited to the site of injury after TBI (Holmin et al.

1995; Soares et al. 1995; Zhang et al. 2006). In the context of injury, microglia can

become activated to aid in the clearance of cellular debris and the recognition of foreign

antigens (Giulian et al. 1989; Kreutzberg 1996). Similar to astrocytes, microglia exhibit a

dual role after TBI by aiding in the elimination of injury debris, but, conversely, releasing

cytokines that potentiate the inflammatory response (Morganti-Kossman et al. 1997;

Morganti-Kossmann et al. 2002; Harting et al. 2008).

Oligodendrocytes myelinate neurons and facilitate rapid conduction of action

potentials in the CNS. Diffusion tensor imaging of brain-injured patients revealed

reduced fractional anisotropy, indicative of white matter track damage, which may be the

consequence of dysfunction or loss of oligodendrocytes (Levin et al. 2008; Rutgers et al.

2008). Damage to white matter tracks and loss of oligodendrocytes have been observed

within days and up to one year following experimental TBI (Pierce et al. 1998; Dixon et

al. 1999; Bramlett and Dietrich 2002; Lotocki et al. 2011; Flygt et al. 2013). Damage to

and subsequent loss of oligodendrocytes can impair axonal transmission and propagate

neuron loss following injury, in part, due to loss of trophic support for ensheathed

neurons (Povlishock 1992; Conti et al. 1998; Newcomb et al. 1999; Wilkins et al. 2001).

TBI Models

Several types of experimental brain injury models have been developed to

reproduce the continuum of human TBI in order to understand the mechanisms of

Page 22: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

9

secondary injury and to develop therapeutic strategies to mitigate the consequences of

secondary injury cascades. In order to reflect the condition of human TBI, models need

to collectively reproduce the spectrum of injury severity with fidelity to pathological

features observed in human TBI. Several of the most frequently utilized and well-

established experimental models of TBI will be discussed. The pathology observed in

these models can be broken into two general categories that aim to reproduce aspects

of diffuse or focal injury (Morales et al. 2005; Xiong et al. 2013).

Diffuse Models

The fluid percussion injury (FPI) produces an injury by delivering a bolus of fluid

rapidly onto the dural surface of the brain through a craniotomy in the skull (Dixon et al.

1987; McIntosh et al. 1989; Hicks et al. 1996; Xiong et al. 2013). Completion of a midline

craniotomy and fluid pulse over the sagittal suture of the brain (midline FPI) produces

features of diffuse injury, but increases in the magnitude of injury are associated with the

manifestation of features of focal injury (Dixon et al. 1987; Hayes et al. 1987; McIntosh

et al. 1989; Morales et al. 2005). Placement of the craniotomy and fluid pulse over the

lateral cortex of the brain (lateral FPI) results in a combination of focal contusion injury

with aspects of diffuse injury spread throughout the contused hemisphere (Cortez et al.

1989; Hicks et al. 1996). Increases in the injury magnitude for both lateral and midline

fluid percussion injury are achieved by increasing the fluid pressure pulse introduced

onto the dural surface of the brain (Dixon et al. 1987). The FPI model is widely used and

highly characterized for use in rats (Dixon et al. 1987; McIntosh et al. 1987; Saatman et

al. 1998), but the model has also been adapted for the utilization of mice (Carbonell et

al. 1998; Spain et al. 2010).

Page 23: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

10

Rodent weight drop models, including the Marmarou, Shohami, and Feeney

models, produce a brain injury by transferring force generated from a guided weight

falling at a prescribed height directly onto the intact skull or onto the dura through a

craniotomy (Feeney et al. 1981; Marmarou et al. 1994; Chen et al. 1996; Xiong et al.

2013). This model typically results in graded diffuse pathology and development of focal

injury with increasing heights and weights of the suspended weight (Feeney et al. 1981;

Tang et al. 1997).

Focal Models: Controlled Cortical Impact

The controlled cortical impact (CCI) model is a highly characterized and a widely

utilized model that produces a predominantly focal injury. Originally developed for use in

the ferret (Lighthall 1988) and later adapted for rodents (Dixon et al. 1991; Smith et al.

1995) to reduce the costs associated with therapeutic studies and to facilitate the usage

of transgenic and knockout animals (Schoch et al. 2012). The model utilizes a computer-

controlled pneumatic cylinder with an affixed impactor tip that transiently impacts the

brain at a specified velocity and depth (Fig. 1.2) (Lighthall 1988; Dixon et al. 1991). The

contact force is transferred to the intact dura of the brain by a metal impactor passing

through a craniotomy in the skull (Dixon et al. 1991). The advantages of the CCI model

include low mortality rates and control over biomechanical injury parameters including

the depth of injury, velocity and duration of impact. The CCI model is easily adapted for

mild, moderate, and severe injuries by changing the depth of impact facilitating graded

injury responses in altered physiology, histopathology, and behavioral function (Dixon et

al. 1991; Goodman et al. 1994; Saatman et al. 2006; Mao et al. 2010; Huh et al. 2011).

Additional parameters of injury, including impactor tip geometry, the velocity of impact,

and impact dwell time have a profound effect on the developing injury pathology

(Goodman et al. 1994; Hannay et al. 1999; Mao et al. 2010; Mao et al. 2011; Pleasant et

Page 24: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

11

al. 2011). Finite element modeling of CCI parameters suggested that the geometry of the

impactor tip ranked second to impact depth as an external mechanical parameter

capable of altering maximal principal strains during a contusion event (Mao et al. 2010).

Page 25: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

12

Figure 1.2: Controlled Cortical Impact (CCI) injury in the mouse positioned over the somatosensory cortex. The computer-controlled pneumatic cylinder rapidly impacts the intact dura of the brain with a metal impactor tip. The contusion site is designated by the red circle.

Page 26: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

13

CCI Pathology

The CCI model produces features of human contusion TBI with a focal injury

encompassing the spectrum of contusion pathology and regional hematomas (Dixon et

al. 1991; Xiong et al. 2013). Severe CCI in the rat and mouse produces pervasive

damage to the white and gray matter, hematoma and cortical cavitation as a result of

apoptotic and necrotic cell death (Smith et al. 1995; Hall et al. 2005; Saatman et al.

2006; Pleasant et al. 2011). Concurrent with cortical damage, loss of the subcortical

white matter at the site of injury occurs within hours and days following injury (Hall et al.

2005; Saatman et al. 2006; Mac Donald et al. 2007; Pleasant et al. 2011). Compromised

vascular integrity is evident within minutes and persists for days after injury (Smith et al.

1995; Baldwin et al. 1996; Baskaya et al. 1997; Saatman et al. 2006; Pleasant et al.

2011). Consequently, impaired cerebral blood flow can last for hours following contusion

injury (Bryan et al. 1995; Kochanek et al. 1995). Diffuse axonal injury and axonal

degeneration are observed within contused tissue and penumbral regions of the cortex

within hours and days after CCI (Dixon et al. 1991; Hall et al. 2005; Mac Donald et al.

2007; Pleasant et al. 2011). Regional hippocampal neuron loss occurs in the dentate

gyrus granular layer and hilus, CA3 and CA1 within hours after injury, the pattern and

extent of which is highly dependent on injury severity (Anderson et al. 2005; Hall et al.

2008; Dennis et al. 2009; Pleasant et al. 2011). Neuronal dysfunction and death

throughout the contused cortex, hippocampus, and penumbral tissue can culminate in

impaired motor and cognitive function.

Consistent with the manifestation of neurological impairments in clinical TBI, the

rodent CCI model of TBI produces impairments in motor and cognitive function (Fox et

al. 1998; Levin 1998; Saatman et al. 2006). Following CCI, rodents exhibit deficits in

motor function by 24 hr and can persist for up to two weeks, as evaluated by tasks that

Page 27: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

14

require a combination of balance and coordinated movement (Fox et al. 1998; Hannay et

al. 1999; Nakamura et al. 1999; Whalen et al. 1999; Pleasant et al. 2011). However, rats

and mice can exhibit spontaneous recovery of motor function over time and, as a result,

the rate of motor function recovery is utilized to evaluate improvements in motor

performance (Saatman et al. 2006; Pleasant et al. 2011).

In addition to motor dysfunction, CCI produces impairments in learning and

memory after injury. The Morris water maze (MWM) and the novel object recognition

task (NOR) are two routinely utilized assessments for evaluating cognitive impairment

after TBI (Smith et al. 1991; Tsenter et al. 2008; Scafidi et al. 2010; Pleasant et al. 2011;

Prins et al. 2013). MWM is sensitive to graded injury severity with rodents exhibiting

deficits in spatial memory as early as 2 days post-injury and spatial learning as early as

7 days following severe CCI (Hamm et al. 1992; Smith et al. 1995; Scheff et al. 1997;

Fox et al. 1998; Hannay et al. 1999; Saatman et al. 2006). The learning and memory

deficits observed in rats subjected to CCI persist for weeks to months post-injury (Hamm

et al. 1992; Dixon et al. 1999). The deficits reported in behavioral tasks evaluating

cognition after brain injury may reflect hippocampal neuron dysfunction and loss as

these tasks evaluate aspects of hippocampal-dependent learning and memory (Smith et

al. 1991; Hamm et al. 1992; Nakamura et al. 1999; Saatman et al. 2006; Pleasant et al.

2011). The fidelity of CCI to human contusion TBI and the neurobehavioral dysfunction

that manifests following injury highlights the appropriateness of the CCI model to

evaluate the efficacy of therapeutic strategies that aim to improve functional recovery

after TBI.

Page 28: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

15

Therapeutic Strategies for TBI

The research effort in experimental models of TBI has led to a better understanding

of secondary injury mechanisms and the identification of promising therapeutic targets.

The complex and diverse nature of the secondary injury mechanisms poses a

substantial challenge for the treatment of TBI. In the clinical setting, treatment with

mannitol to reduce brain edema, and surgical decompressive craniectomy to decrease

intracranial pressure, are two well-established treatment interventions following TBI

(Muizelaar et al. 1983; Munch et al. 2000). However, these interventions aim to

attenuate the consequences of secondary injury mechanisms and do not directly target

the mechanisms of injury.

Given the failure of clinical trials based on therapeutics targeting a single injury

mechanism, combinatorial approaches are gaining popularity as a means for targeting

multiple mechanisms of injury (Margulies and Hicks 2009). While combination therapies

using two or more drugs have promising potential for treating multiple aspects of brain

injury, the arduous task of identifying effective combinations and the necessary

comprehensive pharmacological characterization has tempered initial enthusiasm for

combination therapy after TBI. Alternatively, individual pleiotropic therapeutic agents that

target multiple injury mechanisms and promote regenerative processes are promising

strategies for attenuating the consequences of injury and improving recovery after TBI.

Recovery and plasticity in the brain after CCI

Brain injury initiates numerous pathological responses that perturb cellular

function and produce damage that can potentiate cell loss and impair brain function. The

brain possesses several endogenous repair mechanisms, including structural plasticity

and regeneration, which may help combat damage after injury. However, the magnitude

Page 29: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

16

of damage following TBI may overwhelm endogenous systems, and as a result limit the

ability of the brain to provide meaningful improvement. While CNS glial cells possess the

ability to proliferate and replenish lost cells, the brain has limited capacity to replenish

lost neurons. The subgranular zone of the hippocampus and subventricular zone of the

adult brain participate in ongoing neurogenesis (Zhao et al. 2008), but the extent of new

neuron generation is incapable of replenishing the number of neurons lost after brain

injury. Adult neurogenesis and the generation of new neurons in the context of brain

injury will be discussed in greater detail later in this chapter.

The developing brain possess several endogenous mechanisms of growth and

plasticity that aid in the formation of new neurons and new vessels, and structural

changes, for instance synaptogenesis and neurite outgrowth, that promote the growth

and development of the brain. Several of the mechanisms that mediate developmental

growth and plasticity become activated after CCI in the adult hippocampus, including

angiogenesis, neurogenesis, and synaptogenesis (Scheff et al. 2005; Rola et al. 2006;

Xiong et al. 2010). However, the re-establishment of these endogenous developmental

processes is insufficient to promote meaningful recovery, evident by the lasting cognitive

deficits associated with experimental TBI (Lyeth et al. 1990; Dixon et al. 1991; Hamm et

al. 1992; Adelson et al. 2000). Therapeutic agents that reduce neurodegeneration and

acute cell loss as well as promote and enhance endogenous plasticity may prove

efficacious in mitigating injury-induced damage and promoting recovery after TBI.

Growth factors are ideal therapeutic candidates to promote acute protective effects

with concurrent activation of regenerative mechanisms, including neurogenesis,

angiogenesis, and structural plasticity after TBI. Brain injury stimulates the production of

many neurotrophic factors including vascular endothelial growth factor (VEGF)

Page 30: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

17

(Chodobski et al. 2003), basic fibroblast growth factor (bFGF) (Logan et al. 1992), brain-

derived growth factor (BDNF) (Truettner et al. 1999), nerve growth factor (NGF)

(Truettner et al. 1999), (DeKosky et al. 2004) and insulin-like growth factor-1 (IGF-1)

(Madathil et al. 2010). Several of these studies demonstrate that TBI produces only

transient increases in growth factor expression and signaling (Truettner et al. 1999;

Chodobski et al. 2003; Madathil et al. 2010), suggesting that exogenous administration

of growth factors may be required to promote sustained increases in the levels of growth

factors after TBI.

Several studies have demonstrated that therapeutic strategies to boost

endogenous levels of growth factors, by genetic manipulation or exogenous

administration, reduce cell loss and attenuate neurobehavioral dysfunction following

experimental TBI. Reductions in cell loss after experimental TBI have been associated

with elevations in brain levels of bFGF (Dietrich et al. 1996), NGF (Philips et al. 2001)

and IGF-1 (Madathil 2013). Moreover, improvements in motor and cognitive function

after have been reported in studies evaluating the therapeutic efficacy of bFGF

(McDermott et al. 1997; Sun et al. 2009), NGF (Philips et al. 2001; Mahmood et al.

2004), VEGF (Thau-Zuchman et al. 2010), BDNF (Mahmood et al. 2004) and IGF-1

(Saatman et al. 1997; Rubovitch et al. 2010; Madathil 2013) after experimental TBI. The

improvements in neurobehavioral performance may be the related to improved cell

survival (Philips et al. 2001; Madathil 2013). Several studies also highlight that improved

recovery after TBI with supplementation of a growth factor, including bFGF, VEGF, and

BDNF, may be related to their capability to enhance post-traumatic neurogenesis. Taken

together, these studies illustrate that growth factors possess protective and regenerative

properties that may be effective at combating the pathological consequences of TBI.

Page 31: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

18

Several properties uniquely inherent to IGF-1 make this growth factor aptly suited for

treatment after TBI.

Insulin-like growth factor-1

Insulin-like growth factor-1 (IGF-1) is a promising therapeutic agent for the

treatment of TBI as it has been shown to attenuate motor and cognitive dysfunction after

experimental TBI (Saatman et al. 1997; Rubovitch et al. 2010; Madathil 2013). IGF-1 is a

potent growth factor that has many important roles in tissues throughout the body. The

70 amino acid, 7.6 kDa protein is highly conserved between species, including humans

and rodents, and is closely related in sequence and structure to insulin (Rinderknecht

and Humbel 1978). IGF-1 is post-translationally processed to produce the mature protein

comprised of two amino acid chains with 3 disulfide bonds (Narhi et al. 1993). IGF-1 is

primarily produced in the liver in response to growth hormone binding to its cognate

receptor. Once released into the blood from the liver, IGF-1 has a circulating half-life of

10-30 minutes (Guler et al. 1987; Baxter and Martin 1989). IGF-1 elicits effects in

several target tissues throughout the body.

Binding of IGF-1 to one of the six high affinity binding proteins (IGFBPs),

increases the half-life of IGF-1 in systemic circulation by impeding proteolysis and

decreasing excretion by the kidneys (Duan 2002). All binding proteins possess similar

structural features and sequence homology; however, decreased homology in cysteine-

free regions highlights the emergence of differential binding affinities and functional roles

of the individual binding proteins (Clemmons et al. 1993). IGFBP3 in complex with the

acid-labile subunit is the predominant carrier of IGF-1 in systemic circulation (Guler et al.

1987). The biological activity and bioavailability of IGF-1 is modulated by the actions of

the individual binding proteins (Duan 2002).

Page 32: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

19

Once present at the target tissue, IGF-1 binds to its cognate receptor, a receptor

tyrosine kinase (IGFR) (Fig. 1.3). Binding of IGF-1 to the IGFR promotes receptor

dimerization and autophosphorylation of the intracellular β subunit, and the subsequent

recruitment of scaffolding and secondary proteins, including, but not limited to, the

insulin response subunit (IRS) and the growth factor receptor bound protein 2 (Grb2)

(Laviola et al. 2007; Chitnis et al. 2008). The growth promoting effects of IGF-1 are

mediated by the activation of PI3K, Akt and ERK/MAPK. Activation of PI3K and Akt in

vitro leads to increased cell survival, enhanced growth and metabolism, increased

protein synthesis and the proliferation of new cells (Manning and Cantley 2007).

Phosphorylation of Akt is a potent anti-apoptotic signal that decreases caspase-9 activity

and increases Bcl-2 activity within the mitochondria thereby promoting cell survival

(Manning and Cantley 2007). Similarly, activation of the ERK/MAPK pathway is

associated with growth and metabolism, cell survival, cellular proliferation, and increased

gene transcription and translation (Seger and Krebs 1995). Activation of Akt and MAPK

via IGF-1 also leads to an upregulation in the production and release of IGF-1, which

can act in an autocrine and paracrine fashion to potentiate IGF-1 signaling in

neighboring cells (Underwood et al. 1986; Holly and Wass 1989).

Page 33: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

20

Figure 1.3: IGF-1 binding to its cognate receptor (IGFR) results in the autophosphorylation of the receptor and recruitment of the insulin response subunit (IRS) and growth factor receptor bound protein 2 (Grb2). Activation of mediators of IGF-1 signaling, including phosphatidylinositol 3-kinase (PI3K) and Ras GTPases (Ras) lead to the subsequent activation of Akt and MAPK. Yellow indicates the protein is activated by phosphorylation while blue indicates that protein activity is regulated by hydrolysis of GTP.

Page 34: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

21

IGF-1 signaling is critical for embryonic and postnatal development. Genetic

manipulation to reduce the biological activity of IGF-1, including IGF-1 binding protein-1

overexpression or mutations and homozygous knockout of either IGF-1 or the IGF-1

receptor during embryonic development, is associated with reduced postnatal viability

and retardation of brain growth (Baker et al. 1993; Ludwig et al. 1996). Conversely,

developmental overexpression of IGF-1 does not alter viability, but does increase body

weight and postnatal brain growth (Carson et al. 1993; Ye et al. 1995; Ye et al. 1995).

Together, these studies highlight that IGF-1 axis is important for systemic and brain

growth and development.

IGF-1 in the Brain

In addition to important systemic actions, IGF-1 is also critical for normal brain

development and function throughout life. IGF-1 is mainly expressed in neurons

throughout the brain (Bondy et al. 1992; Bondy and Lee 1993; D'Ercole et al. 1996), but

increased astrocytic expression of IGF-1 has been reported after injury to the brain

(Gluckman et al. 1992; Komoly et al. 1992; Lee et al. 1992; Gehrmann et al. 1994).

Although IGF-1 may be expressed within a single neuron or astrocyte, autocrine and

paracrine actions of IGF-1 can promote IGF-1 signaling in neighboring cells. In situ

hybridization demonstrates that IGF-1 mRNA is present in cells throughout the

developing brain with high levels of expression in the hippocampus and granular layer of

the dentate gyrus during postnatal development (Bondy et al. 1992; Bondy and Lee

1993). While the levels of IGF-1 expression are reduced after development, continued

postnatal expression has been reported in the cortex, striatum, olfactory bulb,

cerebellum and hippocampus of the adult brain (Bondy et al. 1992; Bondy and Lee

1993).

Page 35: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

22

IGF-1 receptor (IGF-1R) mRNA is abundantly expressed in the developing neural

tube with continued widespread postnatal expression in the mature brain, with

heightened expression in neuron dense regions, such as the granular layer of the

dentate gyrus, the olfactory bulb and the cerebellar granular layer (Bondy et al. 1992;

Bondy and Lee 1993). IGFBPs 2, 4, and 5 are the predominant carriers expressed in the

brain, but low concentrations of IGFBP3 have also been reported (D'Ercole et al. 1996).

IGF-1R mRNA is observed in the choroid plexus at the CSF-blood-brain barrier and in

the vasculature throughout the brain parenchyma, which may facilitate IGF-1

translocation into the brain (Bondy and Lee 1993; Reinhardt and Bondy 1994). While

IGF-1 is expressed by neurons throughout the brain, IGF-1 is a unique growth factor, in

that it can cross the blood-brain barrier by receptor-mediated endocytosis (Yu et al.

2006). The functional roles for brain-derived as compared to liver-derived IGF-1 are still

debated, but a handful of studies suggest that IGF-1 from the systemic circulation may

contribute to physiological processes important for cognition and hippocampal plasticity

(Trejo et al. 2001; Nishijima et al. 2010).

Consistent with growth-permissive effects during development, IGF-1 is an

important mediator of growth and survival in the adult brain. Overexpression of IGF-1 in

the brain, by glial or neuron-specific promoters, resulted in increased brain growth and

enhanced numbers and size of neurons, astrocytes and oligodendrocytes (Carson et al.

1993; Ye et al. 1995; Chrysis et al. 2001; Popken et al. 2004; Ye et al. 2004). The

observed increases in cell numbers are the result of an IGF-1-mediated enhancement of

cellular proliferation and reduced apoptosis (Ye et al. 1996; Chrysis et al. 2001). An

increase in IGF-1 signaling is associated with reduced apoptosis in cultured cells of

many cell types, including neurons (Russell et al. 1998; Delaney et al. 1999; Takadera et

al. 1999; Bendall et al. 2007). IGF-1 overexpression also reduces oligodendrocyte

Page 36: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

23

apoptosis and promotes remyelination during the weeks following cuprizone-induced

demyelination (Mason et al. 2000). The well-established growth permissive and anti-

apoptotic properties of IGF-1 are well suited to potentially attenuate the pathological

consequences of injury and promote functional recovery after TBI.

IGF-1 and Acute CNS Injury

IGF-1 has proven to be a promising therapeutic agent for the treatment of acute

CNS injury, as activation of the IGF-1 signaling cascade is associated with increased cell

survival and improved recovery. Transplantation of IGF-1-secreting marrow stromal cell

grafts into the spinal cords or subcortical lesions of rats subjected to either spinal cord

transection or subcortical axotomy resulted in improved survival of corticospinal neurons,

but promoted axonal regeneration of only raphespinal and rubrospinal neurons (Hollis et

al. 2009). Moreover, increased levels of IGF-1 by gene therapy initiated after spinal cord

transection enhanced Akt activation, reduced apoptosis and neuron loss, and promoted

recovery of motor function at 1 and 2 weeks following injury (Hung et al. 2007). Taken

together, these studies demonstrate that prolonged elevations in levels of IGF-1 promote

the survival of neurons and enhance aspects of regeneration after spinal cord injury.

In the context of hypoxic-ischemic injury in the brain, several studies have

demonstrated that IGF-1 is neuroprotective and promotes functional recovery. Central

administration of IGF-1 by intracerebroventricular injection acutely following hypoxic-

ischemic injury reduced neuron loss and infarct area (Guan et al. 1993; Guan et al.

2000; Guan et al. 2000; Brywe et al. 2005) and promoted the recovery of somatosensory

function (Guan et al. 2001). Increased Akt activation and reduced caspase activation

may underlie the improved neuron survival observed in these studies (Brywe et al.

2005). Alternative routes of administration, including intranasal (Lin et al. 2009) or topical

Page 37: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

24

application of IGF-1 also reduced neuron loss after hypoxic-injury (Wang et al. 2000).

Intracerebroventricular infusion of IGF-1 for a period of 3 days after ischemic-injury

resulted in reduced ischemic lesion area by 24 hr that was sustained for 1 week post-

injury (Schabitz et al. 2001). Prolonged elevations in brain IGF-1 by gene therapy,

initiated after hypoxic-injury, stimulated the formation of new vessels, increased cerebral

blood flow and enhanced neurogenesis at 8 weeks post-injury (Zhu et al. 2008). These

studies highlight that treatment of IGF-1 attenuates neuronal loss and enhances

regenerative processes that may contribute to improved functional recovery observed

after acute CNS injuries with similarities to TBI.

IGF-1 and TBI

Endogenous changes in the IGF-1 axis after Experimental TBI

Despite the promising results of IGF-1 promoting neuroprotection and recovery in

CNS injury models, little work has been done to evaluate the therapeutic potential of

IGF-1 after TBI. Several studies have, however, evaluated changes in the endogenous

IGF-1 axis after brain injury. In situ hybridization following a weight drop injury showed

an upregulation of cortical IGFBP2 and IGFBP4 mRNA within 24 hr and a maximal

increase in IGFBP2 mRNA at 3 days post injury, with no changes in IGFBP 3, 5, or 6

(Sandberg Nordqvist et al. 1996). IGF-1 mRNA abundance peaked at 3 days post-injury

at the site of injury, with no change in IGF-1R abundance (Sandberg Nordqvist et al.

1996). In a rat model of penetrating injury, the number of cortical and hippocampal cells

positively stained for IGF-1 was increased in the peritraumatic region at 1 week post-

injury (Kazanis et al. 2004). Kazanis et al. (2004) also demonstrated that administration

of IGF-1 2 hr post-injury reversed acute (4 and 12 hr) injury-induced reductions of BDNF

and neurotrophin-3 in the injured hippocampus. Mild TBI, in a model of weight drop,

Page 38: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

25

resulted in an injury-induced increase in the phosphorylation of the IGF-1R at 24 hr post-

injury, ERK1/2 at 1 hr post-injury, and Akt beginning at 1 hr and persisting for at least 72

hr post-injury in the hippocampus, which was abolished with treatment of a selective

IGF-1R inhibitor (AG1024) (Rubovitch et al. 2010). Following moderate CCI, IGF-1

protein levels were transiently increased in the injured cortex at 1 hr with a subsequent

increase in Akt phosphorylation at 6 hr post-injury but no alterations in levels of IGF-1R

after injury (Madathil et al. 2010), illustrating that exogenous administration of IGF-1 is

required to enhance the strength and duration to promote recovery after CCI.

Administration of IGF-1 after Experimental TBI

Only a handful of studies have evaluated the therapeutic efficacy of IGF-1

administered following TBI. Systemic injections of 4 µg/kg IGF-1 at 24 and 48 hr

following a mild TBI resulted in improved cognitive performance in a Y-maze task at 7

days post-injury (Rubovitch et al. 2010). Continuous systemic infusion of 4 mg/kg/d IGF-

1 for a period of 2 weeks in a rat model of moderate lateral fluid percussion injury

resulted in an attenuation of injury-induced deficits in both motor and cognitive

impairments (Saatman et al. 1997). While these studies highlight the effectiveness of

IGF-1 to reduce the neurobehavioral deficits associated with TBI, little is known about

the mechanisms underlying the neurobehavioral improvements observed after

experimental TBI.

IGF-1 Administration after Clinical TBI

In a phase II open-label, prospective randomized clinical trial of 33 patients,

treatment with 0.01 mg/kg/hr recombinant human IGF-1 by intravenous infusion was

initiated within 72 hr after injury for a period of 14 days in moderate-to-severe brain-

injured patients who presented with GCS scores ranging between 4 and 10 (Hatton et al.

Page 39: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

26

1997). Patients treated with IGF-1 exhibited improved metabolic activity and nitrogen

balance for 2 weeks post-injury, compared to non-treated control patients (Hatton et al.

1997). No serious or life-threatening events were observed with treatment of IGF-1 in

brain-injured patients. Moreover, in the same trial, stratification of IGF-1-treated patients

based on serum concentrations of IGF-1 revealed that patients who achieved a systemic

circulating IGF-1 concentration of 350 ng/ml or above exhibited improved Glasgow

Outcome Scale scores at 6 months after injury compared to IGF-1-treated patients that

did not achieve IGF-1 concentrations above 350 ng/ml (Hatton et al. 1997). A serum

concentration of 350 ng/mL of IGF-1 is in the upper range of physiological serum

concentrations of IGF-1, and may be indicative of a threshold concentration for the

therapeutic efficacy of IGF-1 after TBI. Despite the prolonged infusion, elevated levels of

IGF-1 were only sustained for an average of 8 d during the IGF-1 treatment regimen

(Hatton et al. 1997). The inability to maintain serum concentrations of IGF-1 was

attributed to decreased levels of growth hormone and IGFBP3 during the 14 d infusion of

IGF-1 (Hatton et al. 1997). These findings suggested that negative feedback of IGF-1 on

the growth hormone axis resulted in increased IGF-1 clearance as a result of decreased

levels of IGFBP3, an important protein that increases the half-life of IGF-1 in systemic

circulation (Guler et al. 1989). Future clinical trials would need to consider co-

administration of growth hormone to maintain binding protein levels and achieve

sustained increases in IGF-1 for the duration of the treatment regimen (Hatton et al.

1997).

In a randomized placebo-controlled trial of 23 patients, treatment with saline or

intravenous infusion of 0.01 mg/kg/hr recombinant human IGF-1 and daily subcutaneous

injections of 0.05 mg/kg of recombinant human growth hormone was initiated within 72

hr following injury for a period of 14 days in moderate-to-severe brain-injured patients

Page 40: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

27

that presented with a GCS score ranging between 4 and 10 (Rockich et al. 1999). Co-

administration of IGF-1 and growth hormone resulted in sustained supraphysiological

circulating concentrations of IGF-1 and increased IGFBP3 levels for the duration of

treatment (Rockich et al. 1999). These findings suggested that supplementation of

growth hormone with IGF-1 resulted in increased IGFBP3 expression and was needed

to achieve sustained concentrations above 350 ng/mL of IGF-1, which was previously

shown to improve outcome after TBI (Hatton et al. 1997).

In a randomized, double-blinded placebo-controlled clinical trial of 97 patients,

treatment of placebo or intravenous infusion of 0.01 mg/kg/hr IGF-1 and daily

subcutaneous injections of 0.05 mg/kg growth hormone beginning within 72 hr post-

injury for a period of 14 days in moderate-to-severe brain-injured patients presenting

with a GCS score ranging between 4 and 10 (Hatton et al. 2006). Treatment with IGF-1

resulted in an increased incidence of hypokalemia, hypocalcaemia, and bacteremia, but

no significant differences in the majority of safety parameters including infection rates or

fluid retention (Hatton et al. 2006). These results highlight that treatment with IGF-1 does

not pose safety concerns in brain-injured patients. Treatment with IGF-1 and growth

hormone resulted in significantly elevated circulating IGF-1 concentrations above 350

ng/mL, significantly increased daily serum glucose concentration and significantly higher

nitrogen balance during the duration of treatment compared to treatment with placebo

(Hatton et al. 2006). No differences in the Glasgow Outcome Scale scores were

observed between treatment groups at the cessation of treatment (Hatton et al. 2006).

This clinical trial for IGF-1 and growth hormone (Hatton et al. 2006) was halted in

response to reports from a European prospective, randomized clinical trial indicating that

treatment of growth hormone alone resulted in increased morbidity and mortality from

complications as a result of infection and septic shock (Takala et al. 1999). Brain-injured

Page 41: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

28

patients from placebo and IGF-1 treatment groups exhibited no difference in morbidity

from infectious complications or sepsis (Hatton et al. 2006). The findings reported by

Hatton et al. (2006) highlighted that co-administration of IGF-1 and growth hormone was

effective at achieving sustained IGF-1 concentrations during the dosing period, but

warranted future work in animal models of TBI to determine effective therapeutic

strategies to achieve sustained elevations in the levels of IGF-1 without growth hormone

supplementation. Moreover, the findings from these three clinical trials (Hatton et al.

1997; Rockich et al. 1999; Hatton et al. 2006) highlight the need for additional work in

experimental models of TBI to elucidate the IGF-1-mediated mechanisms that may

underlie the improved recovery observed in brain-injured patients after treatment with

IGF-1.

Neurogenesis

Neurogenesis is the process by which new neurons are generated from neural

stem cells and progenitor cells. The generation of newborn neurons in the hippocampus

of the adult human brain was originally demonstrated in postmortem tissue collected

from cancer patients (Eriksson et al. 1998). This fundamental breakthrough ignited a

field of study focused on trying to better understand the regenerative capacities and

plasticity of the adult brain. The subventricular zone and subgranular zone of the

hippocampus were identified as two regions of the brain that generate newborn neurons

throughout adulthood (Eriksson et al. 1998; Ming and Song 2011). While the process of

neurogenesis is similar for development of the immature brain, the remainder of this

section will discuss the properties of neurogenesis as they pertain to the generation of

newborn neurons in the hippocampus of the adult brain.

Page 42: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

29

Neurogenesis refers to a prescribed complex process, and not a single event

(Fig. 1.4). In the subgranular zone, asymmetrical division of a radial stem cell can give

rise to a neural progenitor cell that may begin to express immature neuron-specific

proteins (e.g. doublecortin, PSA-NCAM) (Ming and Song 2011). Over a period of 2 to 4

weeks after differentiation, the immature neuron will begin to migrate into the granular

layer, extend a primary axon into the hilus and ultimately CA-3, develop dendritic

projections concomitant with synaptogenesis, thereby developing into a mature neuron

that is functionally integrated into the hippocampal circuitry (Zhao et al. 2006; Deng et al.

2010; Kempermann et al. 2010; Ming and Song 2011). However, only a portion of the

immature neurons survive to fully mature and incorporate in the dentate gyrus granular

layer (Kee et al. 2007; Tashiro et al. 2007; Deng et al. 2009).

The trisynaptic circuit, critical for learning and memory, is defined by axonal

projections from the entorhinal cortex synapsing onto the dendrites of neurons in the

granular layer, including those of newly generated neurons, which in turn project axons

to CA3 pyramidal neurons, which project onto pyramidal neurons in CA1 (Wojtowicz

2012). Hippocampal neurogenesis is critical to cognition as several studies that

specifically deplete immature neurons using methods of targeted ablation demonstrate

detrimental effects on aspects of contextual and spatial learning and memory (Clelland

et al. 2009; Deng et al. 2009; Jessberger et al. 2009; Blaiss et al. 2011).

Page 43: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

30

Figure 1.4: Hippocampal neurogenesis in the dentate gyrus granular layer. Proliferation of radial stem cells and neuronal differentiation of newly proliferated neural progenitor cells leads to the generation of immature neurons. As a part of the developmental progression into mature granular neurons, immature neurons will project axonal and dendritic processes, migrate into the granular layer, and begin to functionally integrate into the granular layer.

Page 44: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

31

Neurogenesis and TBI

The subgranular zone of the dentate gyrus shows profound loss of newborn

neurons after CCI (Rola et al. 2006; Gao et al. 2008; Yu et al. 2008; Zhou et al. 2012). It

is possible that a reduction in immature neurons contributes to cognitive dysfunction

after injury, as immature neurons have been shown to be important for hippocampal-

dependent learning and memory (Clelland et al. 2009; Deng et al. 2009; Jessberger et

al. 2009). Following CCI, indicators of neurodegeneration and necrotic cell death are

found to colocalize with markers of immature neurons in the granular layer (Gao et al.

2008; Zhou et al. 2012). Nearly 50% of all immature neurons die within the first 24 hr

following moderate CCI (Gao et al. 2008), with a slow recovery to sham-injured levels

over the course of several weeks (Rola et al. 2006; Yu et al. 2008). Prevention of the

endogenous immature neuron recovery process, by targeted ablation of immature

neurons during the 4 week after brain injury results in an exacerbation of injury-induced

deficits in spatial learning and memory at 1 month following CCI (Blaiss et al. 2011).

Therapeutic strategies targeting enhanced survival and increased generation of

immature neurons may attenuate the cognitive impairment associated with experimental

TBI.

Brain injury elicits a robust increase in cellular proliferation within the subgranular

zone (Dash et al. 2001; Sun et al. 2005; Rola et al. 2006). Increased numbers of neural

progenitor cells may contribute to the injury-induced upregulation of neurogenesis and

the slow recovery of immature neurons within the granular layer of the hippocampus.

Proliferating cells can be identified by incorporation of bromo-deoxyuridine (BrdU), a

thymidine analog, into DNA during DNA synthesis (Taupin 2007). Time course

experiments of BrdU labeling within the granular layer highlight a maximal increase in

proliferation at 3 days post-injury with sustained proliferation for as long as two weeks

Page 45: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

32

after CCI (Dash et al. 2001; Rola et al. 2006). Many of the proliferated cells adopt an

astrocytic lineage, but a minority of the proliferated cells will differentiate into neurons

(Rola et al. 2006; Gao and Chen 2013). Treatment with erythropoietin or S100B, a

calcium binding protein, promoted cellular proliferation over 5 weeks and increased the

number of newborn neurons within 2 days after experimental TBI (Kleindienst et al.

2005; Lu et al. 2005). Enhanced post-traumatic neurogenesis and increased immature

neuron number have been reported in the dentate gyrus granular layer following

increased BDNF, VEGF, and bFGF in the brain following fluid percussion, weight drop

and CCI injury (Wu et al. 2008; Sun et al. 2009; Thau-Zuchman et al. 2010; Thau-

Zuchman et al. 2012). These studies highlight that the neurogenic niche of the

hippocampus is responsive to trophic support after injury, and corroborate the

hypothesis that treatment with a growth factor can enhance hippocampal neurogenesis

and immature neuron number following TBI.

IGF-1 and Neurogenesis

IGF-1 is an important mediator of brain developmental and adult neurogenesis.

The well-characterized anti-apoptotic effects of IGF-1 on neurons previously described in

this chapter extends to cultured pluripotent human embryonic stem cells. Blocking the

IGF-1 receptor reduced human embryonic stem cell survival within days following

inhibition of IGF-1 signaling (Bendall et al. 2007). IGF-1 is also a potent promoter of

cellular proliferation and neuronal differentiation of neural stem cells (Arsenijevic and

Weiss 1998; Aberg et al. 2000; Brooker et al. 2000; Arsenijevic et al. 2001; Yan et al.

2006). During maturation, immature neurons will extend dendritic processes and exhibit

the formation of functional synapses (Zhao et al. 2008). Targeted genetic depletion of

IGF-1 in the brain is associated with reduced dendritic length and arbor complexity

during postnatal development (Cheng et al. 2003). However, treatment with IGF-1 on

Page 46: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

33

primary organotypic cortical slices cultures resulted in increased branching and lengths

of dendrites pyramidal neurons (Niblock et al. 2000) and promoted neurite outgrowth in

dissociated neurons (Torres-Aleman et al. 1989). Collectively, these studies highlight

that IGF-1 is a potent neurotrophic factor that may promote several aspects of

neurogenesis, including proliferation and neuronal differentiation, in the hippocampus

following experimental TBI.

Specific Aims and Hypothesis of the Dissertation

TBI is an acquired injury that initiates detrimental secondary injury cascades that

culminate in cellular dysfunction and death. Clinical trials reveal that therapeutic agents

aimed at one aspect of injury pathology may not be sufficient in attenuating brain

damage following TBI. A therapeutic agent that reduces cell death while simultaneously

promoting regenerative mechanisms in the injured brain might have increased efficacy in

improving recovery of function after TBI. While previously published data demonstrate

that IGF-1 is effective at improving recovery after experimental TBI, it is unknown

whether IGF-1 is capable of protecting against cell death or promoting neurogenesis in

the traumatically injured brain. Utilizing targeted astrocyte-specific IGF-1 overexpression

in the brain or clinically relevant strategies to administer IGF-1 into the systemic

circulation or directly into the brain, this work investigates the recently discovered aspect

of injury-induced neurogenesis and how this reparative mechanism may be enhanced by

IGF-1, as enhanced neurogenesis could contribute to improved recovery after TBI.

The overall hypothesis of this dissertation is that treatment with IGF-1 attenuates

motor and cognitive impairment and enhances hippocampal neurogenesis in the mouse

brain following severe CCI. The long-term goal of this work is to provide additional data

that contributes to a preclinical evaluation of IGF-1 for the treatment of TBI.

Page 47: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

34

In chapter 2, we demonstrate that a simple modification to the well-established

CCI model slows the rate of cortical cell death and hippocampal neurodegeneration after

severe brain injury. We demonstrate that utilization of a rounded impactor tip reduces

maximal principal strain compared to a beveled impactor tip in a finite element

mathematical model of mouse CCI. We demonstrate in vivo that a rounded impactor tip,

compared to a beveled impactor tip, slows the rate of acute cortical cell loss and reduces

regional hippocampal neurodegeneration, without modulating other pathological

consequences of CCI, including blood-brain barrier damage, axonal injury, and

neurobehavioral dysfunction. In this chapter we provide evidence that CCI with a

rounded tip is an appropriate model to evaluate the therapeutic efficacy of IGF-1 to

attenuate hippocampal neurodegeneration and to enhance post-traumatic hippocampal

neurogenesis.

In chapter 3, we provide preliminary evidence that systemic infusion of IGF-1,

compared to infusion of vehicle, promotes the activation of Akt, a downstream mediator

of IGF-1 signaling, in the contused brain and modestly reduces neurodegeneration in the

dentate gyrus granular layer. However, systemic infusion of IGF-1 does not promote the

survival of hippocampal immature neurons at 3 days post-injury or improve motor

function over the first week after TBI.

In chapter 4, we demonstrate that central infusion of IGF-1 attenuates motor and

cognitive dysfunction associated with severe CCI. Treatment with IGF-1 increases

hippocampal immature neuron density at 7 d post-injury, even when treatment is

delayed up to 6 hr after injury. We provide evidence that suggests the observed

increases in immature neuron density with treatment IGF-1 after TBI is the result of

enhanced cellular proliferation and enhanced neuronal differentiation in the contused

Page 48: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

35

hippocampus. We also observed brain swelling in IGF-1-treated mice that could be

reduced with decreasing doses of IGF-1 or by alterations to the parameters of central

infusion.

In chapter 5, we utilize a conditional astrocyte-specific IGF-1 overexpressing

transgenic mouse model to evaluate the mechanisms by which IGF-1 enhances post-

traumatic neurogenesis after CCI. We demonstrate that overexpression of IGF-1 does

not promote the survival of immature neurons at 3 days post-injury. However,

overexpression of IGF-1 increases immature neuron density at 10 days after severe

CCI. We demonstrate that this increase in immature neuron number is the product of

enhanced neuronal differentiation of newly proliferated cells in the injured hippocampus.

Moreover, we demonstrate for the first time that hippocampal immature neurons exhibit

reduced dendritic arbor length and complexity at 10 d after severe CCI and provide

compelling evidence that overexpression of IGF-1 promotes the restoration of immature

neuron dendritic complexity to that observed in immature neurons of sham-injured mice.

Copyright © Shaun William Carlson 2013

Page 49: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

36

Chapter 2: Rate of Neurodegeneration in the Mouse Controlled Cortical Impact

Model is Influenced by Impactor Tip Shape: Implications for Mechanistic and

Therapeutic Studies

This chapter was published in Journal of Neurotrauma. Nov 2011; 28 (11); 2245-62.

Introduction

The annual incidence of traumatic brain injury (TBI) is estimated at over 1.5

million people in the United States alone, with many cases of mild TBI likely unreported.

Although numerous clinical trials have been conducted for moderate-to-severe TBI, no

therapeutic intervention has proven effective in improving functional outcome (Narayan

et al. 2002; Maas et al. 2010). The difficulty in establishing an effective pharmacological

treatment approach is thought to be related, in part, to the heterogeneity of TBI (Maas et

al. 1999; Saatman et al. 2008). At a cellular level, complexity arises from multiple,

interacting secondary injury cascades that, over time, amplify primary damage inflicted

during the traumatic insult. At a tissue level, TBI is heterogeneous in that it comprises

multiple pathoanatomical types of injury, such as contusion, diffuse axonal injury and

hematoma, which likely involve different combinations of primary damage and secondary

mediators. To overcome this challenge, treatment approaches tailored to a selected

pathoanatomical injury type may be utilized in future clinical trials for TBI. Contusion

injury is well suited to this targeted therapy approach for several reasons. Contusion

represents a common type of TBI which can be diagnosed using standard radiological

imaging (Marshall et al. 1992; Maas et al. 2005; Alahmadi et al. 2010). Furthermore,

animal models of contusion injury are widely used for identification of injury mechanisms

and the preclinical evaluation of therapeutic interventions.

Page 50: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

37

The controlled cortical impact (CCI) model is one of the most commonly used

models of contusion TBI. The CCI model utilizes a metal impactor rod to transiently and

rapidly deform the cortex exposed via a craniotomy. The model was originally

developed for use in ferrets (Lighthall 1988), but was subsequently adapted for rats

(Dixon et al. 1991) and mice (Smith et al. 1995). The use of CCI brain injury in mice has

dramatically increased over the past 15 years due to greater reliance on transgenic and

knockout mice for the study of cellular mechanisms and the cost effectiveness of mice

for therapeutic testing. In both mice (Smith et al. 1995; Fox et al. 1998; Hannay et al.

1999; Hall et al. 2005; Saatman et al. 2006) and rats (Dixon et al. 1991; Hamm et al.

1992; Sutton et al. 1993; Goodman et al. 1994; Hicks et al. 1997; Scheff et al. 1997),

experimental CCI reproduces important features of human contusion TBI such as

neocortical and hippocampal cell loss by necrosis and apoptosis, axonal injury, blood-

brain barrier (BBB) breakdown, and cognitive and motor deficits. In the process of

characterizing the CCI model, a number of laboratories have demonstrated that

changing the depth or velocity of impact or the number of craniotomies influences the

location and severity of injury (Dixon et al. 1991; Sutton et al. 1993; Goodman et al.

1994; Meaney et al. 1994; Fox et al. 1998; Hannay et al. 1999; Saatman et al. 2006).

In contrast, the effects of the shape of the impactor tip on the histopathological or

behavioral response to CCI brain injury has not been systematically investigated.

Interestingly, the shape of the impactor tip is frequently not described in the methodology

of published CCI studies. The model was originally developed with a rounded tip

impactor (Lighthall 1988). However, in mouse CCI studies that do describe the

impactor, nearly all describe the tip as ‘flat’ or ‘beveled flat’, that is, a flat surface where

the impactor contacts the exposed brain tissue (Fox et al. 1998; Dennis et al. 2009)

(Xiong et al. 2005; Thompson et al. 2006; Whalen et al. 2008; Sandhir and Berman

Page 51: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

38

2010). We recently demonstrated that, after impact depth, tip geometry (flat versus

rounded) was the most important parameter in estimating tissue strains and predicting

cortical contusion volume in a rat finite element simulation, exceeding the influence of

impact velocity, tip diameter or number of craniotomies (Mao et al. 2010). Simulations

demonstrated that impact with a flat tip resulted in high tissue strains localized at the

impactor rim, whereas strains produced by impact with a rounded (spherical) tip were

more radially distributed.

In our experience, the use of a flat tip impactor frequently resulted in very rapid

cortical neuron death, extensive cortical hemorrhage and distortion, and sometimes

even cortical tissue tearing in mice subjected to moderate or severe CCI. We reasoned

that by using a hemispheric (rounded) tip, we could decrease tensile/shear strains at the

edges of the impactor as it penetrated the cortical tissue, resulting in less ablative

primary cellular damage and a more gradual progression of neuronal death, thereby

enhancing the fidelity of the model to human closed head contusion injury.

To test this hypothesis, we utilized a finite element modeling approach to

visualize and compare the patterns of maximum principal (tensile) strains produced by

deformation of the mouse cortex with either a flat or rounded tip impactor. We then

compared the histological damage and the time course for neocortical contusion

formation from 1 h to 9 d after a 1.0 mm depth impact with these two tips. To provide a

comprehensive evaluation of the effects of modulating the tip shape, we also evaluated

other important features of CCI brain injury such as regional hippocampal

neurodegeneration, BBB breakdown, axonal injury, and neurobehavioral deficits. Our

data suggest that cortical impact with either a flat or rounded tip impactor results in

equivalent neocortical cell death at 9 d after injury, similar patterns of BBB breakdown

Page 52: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

39

and axonal injury, and equivalent neurobehavioral impairment and recovery.

Importantly, though, impact with a rounded tip impactor reduced cortical tissue strains at

the impactor edges, slowed the progression of cortical cell death and reduced the extent

of acute hippocampal neurodegeneration. These findings have important implications

for investigations of cellular mechanisms of damage and testing of therapeutic

interventions for contusion TBI.

Page 53: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

40

Materials and Methods

Finite Element Mouse Brain Model Development and CCI Simulations

Considering anatomical similarity between rat and mouse brains, a previously

developed rat brain finite element model (Mao et al. 2006) was used as the basis to

generate a finite element mouse brain model. The geometry used for developing the

mouse brain model was taken from a comprehensive three-dimensional C57BL/6J

mouse brain atlas reported by Ma et al. (2005). Ma and colleagues acquired microscopy

images from 10 adult male (12-14 weeks, weight range 25-30 g) C57BL/6J mice at a 47

µm isotropic resolution using a 17.6-T magnetic resonance imaging device (Ma et al.

2005). The ‘minimum deformation atlas’, a representation of the average structural

shape of the 10 mice, was adopted here for developing a mouse brain model. The

images were imported to an imaging processing software MIMICS 12 (Materialise,

Leuven, Belgium) to segment the outer surface of the brain, exterior surfaces of the

white matter, and ventricles. The Meshworks 5.0/Morpher (DEP, Troy, MI) was then

used to morph the exterior meshes of the rat brain model to the outer surface of the

mouse brain. After that, the white matter and ventricles were adjusted according to the

mouse brain anatomy. The morphed mouse brain model, consisting of the cerebrum,

cerebellum, corpus callosum, internal and external capsules, lateral ventricles, olfactory

bulb, brainstem, and part of spinal cord, has a total of 258,428 brick elements with a

typical spatial resolution of 150 µm (Figure 2.1). To the best of the authors’ knowledge,

material properties regarding mouse brain were not reported in the literature. Therefore,

the mouse brain material properties were defined based on combined in vitro and in situ

indentation tests of non-preconditioned adult rat brains reported by Gefen et al. (2003)

using a linear viscoelastic material law (LS-DYNA Material Type 61). For the gray

matter, cerebellum, and brainstem, a short-term shear modulus of 1.72 kPa and a long-

Page 54: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

41

term shear modulus of 0.51 kPa were assumed. For the white matter, a short-term

modulus of 1.2 kPa and a long-term modulus of 0.36 kPa were assumed. A decay

constant of 20 ms was assumed for both the white and gray matter. Additionally, the

ventricles were assumed to have a short-term modulus of 1 kPa and a long-term

modulus of 0.3 kPa while the corresponding parameters for the spinal cord were

assumed to be 3.1 and 0.92 kPa, respectively. For the membranes, linear elastic

material properties were assumed with a Young’s modulus of 12.5 MPa for the pia-

arachnoid complex (Jin et al., 2006) and 31.5 MPa for dura (Galford and McElhaney

1970).

Two CCI scenarios using a 2.5 mm diameter, flat tip and a 3.0 mm diameter,

hemispherical (rounded) tip impactor, identical to experimental conditions, were

simulated. The hemispherical tip was designed to be slightly larger in diameter to

partially offset the decrease in tip volume caused by rounding of the tip. A 4.5 mm

diameter craniotomy, centered at -2.5 mm Bregma and 2.75 mm lateral to the midline,

was removed from the mouse skull model. The impactor compressed the exposed dura

for up to 1.0 mm at a velocity of 3.51 m/s, the mean value calculated from experimental

records. The simulation duration was set as 2 ms and simulation results were plotted at

every 0.02 ms. The depth of impact was set at 1.0 mm for both impactors to mimic

commonly used experimental protocols and control for the variable most critical in

determining tissue strains in the CCI model (Mao et al. 2010).

Animals and Surgical Procedures

Adult male C57BL/6J mice (The Jackson Laboratory, Bar Harbor, ME), weighing

22-32 g, were housed in a room with a 14:10 light:dark photoperiod. Mice were given

food and water ad libitum. All animal experiments and procedures were in accordance

Page 55: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

42

with the National Institutes of Health Guide for the Care and Use of Laboratory Animals

and were approved by the University of Kentucky Institutional Animal Care and Use

Committee.

Mice were anesthetized using 2.5% isoflurane, their heads were shaved, and

they were placed in a stereotaxic frame (Kopf, Tujunga, CA) where they were

maintained on isoflurane delivered by a nose cone. A midline incision was made to

expose the skull, and a 4.5 mm craniotomy was made centered at -2.5mm Bregma and

2.75mm lateral to midline over the left hemisphere. Mice were subjected to CCI injury at

a 1.0 mm impact depth and a nominal velocity of 3.5 m/s. The CCI impactor device

(TBI-0310 Impactor, Precision Systems and Instrumentation, Fairfax Station, VA) uses a

computer controlled pneumatically driven piston to rapidly impact the brain. This system

has a sensitive mechanism for detecting the cortical surface after which the impactor is

automatically repositioned for a user determined impact depth. Mice were randomized

for injury with either a flat tip or rounded tip impactor (Figure 2.2A). After injury, a

cranioplast was placed over the exposed brain. The mice were sutured and placed on a

heating pad to maintain normal body temperature until they were fully awake, after which

they were returned to their home cages.

Tissue Preparation

At 1, 4, 12, or 24 h, or 9 d, (n=5 injured with a flat tip impactor, n=5 injured with a

rounded tip impactor per time point) mice were anesthetized with 65 mg/kg sodium

pentobarbital through an intraperitoneal injection. Mice were then intracardially perfused

with heparinized saline followed by 10% buffered formalin, and decapitated. After 24 h

of fixation in 10% buffered formalin, the brains were removed from the skull, post-fixed

for 24 h, cryoproteced in 30% sucrose, and quickly frozen in -30°C isopentane. Coronal

Page 56: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

43

40 μm sections were cut using a freezing sliding microtome (Dolby-Jamison, Pottstown,

PA).

Cortical Contusion Volume

Brain sections selected at 400 µm intervals between 0 and -3.5 mm Bregma

(Paxinos and Franklin 2001) were stained for Nissl substance using 2.5% Cresyl Violet

and mounted onto slides. Existing methods were modified slightly to quantify the

neocortical area containing surviving neurons (Royo et al. 2006; Saatman et al. 2006).

Sections were viewed using a light microscope (BH2, Olympus America Inc., Melville,

NY) equipped with an automated stage and camera. On a live image, the neocortices of

the contralateral and ipsilateral hemispheres were separately outlined using a tracing

tool (Bioquant Life Science version 8.40.20), alternating between 2x and 10x

magnification to evaluate the boundaries of the neocortex as well as cellular staining and

morphology. Only areas containing Nissl-stained cells with presumed neuronal

morphology were outlined, thereby excluding areas of necrotic tissue. Contusion area

was calculated as the difference between contralateral and spared ipsilateral neocortical

areas. The contusion volume was obtained using Cavalieri’s principle, integrating over

the inter-section distance, and is expressed as a percentage of the contralateral

neocortical volume to control for any potential variation in tissue shrinkage during fixation

and freezing.

Regional Hippocampal Neurodegeneration

Fluorojade-B staining for degenerating neurons (Schmued et al. 1997) has been

used to quantify trauma-induced hippocampal neuron death (Anderson et al. 2005;

Dennis et al. 2009). Brain sections containing hippocampus were selected every 800µm

between Bregma levels -1.06 mm and -3.5 mm (Paxinos and Franklin 2001) from mice

Page 57: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

44

that survived 1, 4, 12, and 24 h. After rinsing in Tris-buffered saline (TBS), sections

were immersed in diaminobenzidine (DAB) (Vector Laboratories, Inc., Burlingame, CA)

for 5 min to provide a substrate for endogenous peroxidases and eliminate

autofluorescence from parenchymal hemorrhage. Sections were then mounted onto

slides, warmed at 40-45ºC for 30 minutes, and kept at room temperature overnight. On

day 2, slides were immersed sequentially in 1% NaOH in 80% EtOH, 70% EtOH, and

ddH2O. Sections were placed in a 0.06% potassium permanganate solution for 10 min

and rinsed in ddH2O, before staining 10 min with 0.0001% Fluorojade-B (Millipore Co.,

Billerica, MA) in 0.1% acetic acid. Slides were then rinsed in ddH2O, dried on a slide

warmer at 50°C, immersed in Xylenes (Fisher Scientific, Fair Lawns, NJ), and

coverslipped in Cytoseal XYL (Richard-Allan Scientific, Kalamazoo, MI). For

quantification, Fluorojade-positive neurons were counted separately within the dentate

gyrus, CA3/CA3c, and CA1 of the ipsilateral hippocampus in three sections centered

within the impact site. Counting was performed at 20x under a fluorescence microscope

equipped with an FITC filter (AX80, Olympus America) by an individual blinded to the

injury conditions of each animal.

Immunohistochemistry

Immunoglobulin (IgG)

Free-floating sections, adjacent to both those used for Nissl and Fluorojade-B

staining, were used for immunohistochemical analysis of IgG extravasation, indicative of

BBB breakdown. Tissue was rinsed in TBS and treated with a 3% H2O2 solution to

quench endogenous peroxidases before blocking non-specific binding sites with 5%

normal horse serum (NHS). Biotinylated donkey anti-mouse IgG antibody (1:1000,

Jackson Immuno Research) was applied overnight at 4°C. Negative control sections

Page 58: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

45

were incubated in diluent without primary antibody. On day 2, the tissue was rinsed in

TBS and incubated in an avidin-biotin complex (AB Elite Kit, Vector Laboratories) and

reacted with DAB.

Amyloid Precursor Protein

Immunohistochemical detection of amyloid precursor protein (APP) accumulation

in axons is a commonly used marker for acute traumatic axonal injury (Sheriff et al.,

1994; Stone et al., 2000). Sections were rinsed in TBS and placed in 10M citric acid at

65ºC for 15 min for antigen retrieval. Tissue was then washed in TBS, blocked in 5%

NHS in 0.1% Triton X-100 and incubated overnight at 4°C in 1:500 anti-c-terminal APP

(Cat # 51-2700, Invitrogen, Carlsbad, CA). Following incubation in 1:1000 biotinylated

donkey anti-rabbit IgG secondary antibody, endogenous peroxidases were blocked

using 0.9% H2O2 in 50% methanol. Tissue was then rinsed and incubated in avidin-

biotin complex. Reaction product was visualized using DAB as a substrate.

Behavioral Testing

Following CCI injury, mice to be utilized for analysis of tissue damage at 9 d post-

injury (n=5/impactor tip) were subjected to behavior tests at several time points to

assess their level of motor and cognitive function. All behavior tests were performed by

a blinded evaluator.

Neurological Severity Score

A Neurological Severity Score (NSS), adapted from Tsenter et al. (2008), was

assessed at 1 h and 1, 2 and 7 d. NSS is a 14-point ordinal scale with 14 representing

normal motor function. Mice were scored as they traversed, in sequence, 3, 2, 1, and

0.5 cm wide Plexiglas beams (60 cm long) and a 0.5 cm diameter wooden rod (60 cm

long). The beams and rod were elevated 47 cm above the table top. Mice were

Page 59: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

46

acclimated to walking across the beams and rod 24 h before injury. Mice were allowed

up to 30 s to cross the beams and rod during both acclimation and testing. Each beam

had a maximum of 3 points; 3 points were given for successfully crossing the beam with

normal forelimb and hindlimb position, 2 points were given for successfully crossing the

beam despite either a forelimb or hindlimb hanging from the beam, and 1 point was

given for crossing the beam despite inverting underneath the beam one or more times.

If a mouse became inverted on the beam, it was righted and allowed to continue across.

The mouse scored a 0 if it did not cross the beam in the allotted time or if it fell off the

beam. For the wooden rod, there was a maximum of two points. A mouse received two

points for successfully crossing the rod and one point was given for crossing despite

inverting more than three times. A mouse received a 0 if it did not traverse or fell off of

the rod.

Neuroscore

Neurological motor function was also assessed at 1, 2, 5 and 7 d post-injury,

using a 12-point neuroscore evaluation. The neuroscore test was performed as

previously described (Scherbel et al. 1999) with slight modifications. The neuroscore

evaluation consisted of 3 tests: the grid walk (2 points), forelimb and hindlimb flexion

tests (3 points each), and lateral pulsion (4 points). For the grid walk, mice were placed

upon a wire grate with bars spaced 1.5 cm apart positioned 20 cm above a table. One

point was deducted if either a front or hind paw slipped through the grate without

immediate recovery during 60s of free exploration. Mice scored a 0 if they had both

forelimb and hindlimb slips. In the flexion tests, limb coordination and grip strength were

evaluated as mice were suspended by their tail over a metal rung cage top and lowered

to allow them to grasp the cage bars with both forelimbs. One point was deducted from

the forelimb score if the mouse had low grip strength, crossed its forelimbs, or had

Page 60: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

47

excessive hyperactivity. For the hindlimb score, abnormal limb extension or toe splaying

each resulted in a one point deduction, while 3 points were deducted if the mouse curled

its hindlimb up to its body. For the lateral pulsion test, mice were pushed 4 times from

left to right across a ribbed plastic mat at increasing speeds. Four points were deducted

for rolling over on the first trial, 3 points for rolling over on the second trial, 2 points for

rolling over on the third trial, and 1 point for rolling over on the fourth trial. If the mouse

did not roll over on any of the 4 trials, it received 4 points.

Morris Water Maze Cognitive Test

Learning ability was evaluated on days 7-9 post-injury using a paradigm similar

to those widely utilized in models of rodent TBI (Hamm et al. 1992; Saatman et al. 1997;

Prins and Hovda 2001). A 1 m diameter Morris water maze (MWM) was filled with 19-

21ºC water containing a non-toxic white paint (Rich Art Co., Northvale, NJ) to hide a 6.3

cm diameter platform that was submerged 0.5 cm below the surface of the water. Mice

were released into the water from one of four different starting points (North, East,

South, or West quadrant) and their latency to locate the platform (located in the West

quadrant) using large visual cues placed on the walls outside the tank was recorded.

Mice were tested in sets of four trials per day over three consecutive days. If the mouse

did not find the platform within the allotted 70 s trial time, they were placed onto the

platform for 5 s, then returned to a heated cage. Swimming trials were monitored using

an overhead video camera and tracking software (EZVideo version 5.51DV, Accuscan

Instruments Inc., Columbus, OH).

Statistical Analysis

All data are presented as means and standard deviation. Contusion volumes

were compared using a 2-way ANOVA. Fluorojade-positive cell counts were analyzed

Page 61: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

48

using a 2-way ANOVA for each hippocampal subfield. Morris water maze training

latencies, neuroscore and NSS motor function tests were analyzed using a repeated

measures 1-way ANOVA. In all cases, Neuman-Keuls post-hoc t-tests were performed

when appropriate. A value of p < 0.05 was considered statistically significant.

Page 62: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

49

Results

Finite element simulation of cortical impact injury

CCI brain injury at a 1.0 mm depth of impact was simulated for a flat tip impactor

and a rounded tip impactor (Figure 2.2A). Principal strain contours immediately after the

maximum dural compression was reached are illustrated in Figure 2.2B. The peak

maximum principal strains resulting from impact with a flat tip were > 49.5 - 55.0% and

above. In contrast, impact with a rounded tip generated lower peak maximum principal

strains, in the range of 38.5 - 44.0%. In addition, the largest strains induced by the flat

tip impactor were highly localized along the impactor rim, while there were no such

concentrations or ‘edge effects’ observed for the rounded tip impactor (Figure 2.2B).

The edge effect of the flat tip impactor is also clearly evident in strain profiles illustrating

the peak maximum principal strain as a function of element location along a lateral to

medial cortical arc (Figure 2.3), where element no.16 represents the position of the

center of either the flat tip or rounded tip impactor. At cortical layers 3 and 5, for

example, impact with the flat tip induced strains at the impactor rim which were almost

100% higher than the strains beneath impactor center. In contrast, the rounded tip

produced more uniform strain distributions beneath the impactor. The difference

between rounded tip and flat tip induced strains was less pronounced at the cortical

surface (layer 1), where the highest strains were induced due to tissue protrusion

between the impactor and the edge of the craniotomy. Strain elevations due to tissue

protrusion decreased as a function of cortical depth and were essentially eliminated by

layer 7. In the hippocampal region, the peak maximum principal strain was 24.8% for

the rounded tip group and 30.1% for flat tip group.

Page 63: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

50

Figure 2.1. Finite element model of mouse brain and impactor. (A) Isometric view of the three-dimensional finite element model of the mouse brain utilized for simulation of controlled cortical impact brain injury. (B) Cross-sectional view of the ipsilateral dorsal quadrant of the brain model illustrating interior structures, the mesh size and the position of the impactor.

Page 64: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

51

Figure 2.2. Effect of impactor shape on brain tissue strains. (A) Animals received cortical impact injury with either a flat tip (left) or rounded tip (right) metal impactor rod. (B) The impactor tip shapes were reproduced for the finite element simulations shown here, illustrating the maximum principal (tensile) strain contours in a coronal section through the impact center at an impact depth of 1.0 mm. Contours are shown with the tip in place (upper maps) and with strains mapped to the undeformed brain (lower maps). The fringe levels indicate the strain levels (0 to 0.5500) represented by colors in each contour map.

Page 65: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

52

Figure 2.3. Strain profile as a function of cortical depth and tip geometry. The peak maximum principal strain at any time during the impact is plotted for 31 elements along a lateral to medial cortical arc, or cortical layer, in the finite element model. The model consists of 8 cortical layers (see Figure 2.1B), with the cortical surface designated as Layer 1, and the deepest layer adjacent to the subcortical white matter designated as Layer 8. Element number 16 corresponds to the center of the impactor and craniotomy. Strain peaks labeled ‘A’ correspond to regions of tissue protrusion between the impactor and the craniotomy edge. Strain peaks labeled ‘B’ correspond to the edge effects created by the flat tip impactor.

Page 66: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

53

Histological damage in Nissl-stained brain sections

Histological analyses of cortical tissue damage using Nissl staining revealed that

injury with a flat tip impactor resulted in more pronounced initial tissue disruption and

more rapid cell death when compared to impact with a rounded tip impactor. Even

though cell loss progressed at different rates, neocortical tissue damage was ultimately

equivalent.

At 1 h post-injury, brains injured by the flat tip impactor exhibited marked cortical

swelling associated with extrusion at the craniotomy site (Figure 2.4A). Hemorrhage and

neuron loss was observed where cortical tissue strains were predicted to be maximal, at

the periphery of the impactor. Hemorrhage was also frequently found at the interface of

the cortex and the subcortical white matter. In most animals injured with the flat tip (3 of

5), hippocampal distortion and hemorrhage were prominent. In contrast, 1 h after injury

with a rounded tip impactor, brains exhibited no overt loss of Nissl staining, only mild to

moderate cortical swelling and minimal hemorrhage, typically along the cortical surface

(Figure 2.4B). The ipsilateral hippocampus was not distorted, and no hemorrhage was

present.

At 4 h after injury with a flat tip impactor, extensive neocortical cell death

compromised tissue integrity (Figure 2.4C), and began to result in tissue loss during the

cutting and mounting procedures. By 12 h, a cortical cavity or ‘lesion’ was well formed

(Figure 2.4E). At 4 and 12 h, hippocampal hemorrhage (6 of 10) and distortion (9 of 10)

were still prominent. After injury with a rounded tip impactor, the contused neocortex

exhibited mild to moderate swelling, mild intraparenchymal hemorrhage, and progressive

loss of Nissl staining intensity from 4 to 12 h (Figure 2.4D, F). The subcortical white

matter tract was intact but exhibited some disruption and hemorrhage. No hippocampal

Page 67: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

54

hemorrhage was observed at 4 or 12 h after injury with a rounded tip impactor, although

distortion of the hippocampal formation was evident by 12 h.

By 1 and 9 d postinjury, histological damage in Nissl-stained brain sections from

mice injured with either a flat and rounded tip impactor appeared much more similar than

at earlier time points. Both impactors resulted in the formation of a large cortical cavity

by 1 d, with evidence of intraparenchymal hemorrhage surrounding the cavity (Figures

2.4G, H). In both groups, the cortical cavity was lined by reactive glia by 9 d (Figures

2.4I, J). As with the flat tip impactor group, the rounded tip impactor produced

subcortical white matter disconnection and degeneration in nearly all mice (9 of 10) at 1

and 9 d postinjury. Delayed hippocampal hemorrhage was noted at 1 d after rounded tip

impact (3 of 5), while hemorrhage was not typical at 1 d after injury with a flat tip

impactor and was not observed at 9 d postinjury in either group.

Page 68: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

55

Figure 2.4. Temporal progression of histological damage created by a flat tip versus a rounded tip CCI impactor. Images are shown of the ipsilateral neocortex and hippocampus of Nissl-stained coronal sections taken at the epicenter of the impact at 1 h (A, B), 4 h (C, D), 12 h (E, F), 24 h (G, H) and 9 d (I, J) after 1.0 mm depth CCI. Injury with a flat tip impactor (A, C, E) created more profound tissue disruption, hemorrhage and cell loss within the first 12 h when compared to injury with a rounded tip impactor (B, D, F). By 24 h and 9 d post-injury, tissue damage resulting from the flat tip (G, I) and rounded tip (H, J) impactor was similar. Arrows mark the approximate location of the edges of the craniotomy. Arrowheads note the location of tissue hemorrhage and neuron loss, corresponding to the path of the impactor edges, at 1 h after impact.

Page 69: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

56

Differences in gross histological responses to injury using a flat tip versus a

rounded tip impactor (vide supra) were underscored by qualitative analysis of neocortical

neuron morphology. As early as 1 h after injury with a flat tip impactor, neocortical Nissl

staining was diminished in intensity, and scattered cell loss and shrinkage was evident

through all neocortical layers (Figure 2.5A) when compared to uninjured tissue (Figure

2.5E). Neurons in the neocortex injured by the rounded tip impactor were pyknotic, but

normal appearing in size, density, and Nissl intensity (Figure 2.5B). At 4 h after injury

with a flat tip impactor, neocortical neuronal damage had progressed substantially, with

few identifiable neurons visible in the contusion site (Figure 2.5C). In the neocortex of

mice injured with a rounded tip, Nissl-stained cells with neuronal morphology were still

clearly detectable at 4 h (Figure 2.5D), although pyknosis and loss of Nissl intensity was

greater than at 1h. The extent of neuronal damage was far less than that induced at 4 h

by a flat tip impactor (compare Figure 2.5D and 2.5C). Neuronal damage had increased

by 12 h after impact with a rounded tip (Figure 2.5F), with marked loss of Nissl staining

comparable to that observed 4 h after injury with the flat tip impactor (Figure 2.5C).

Page 70: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

57

Figure 2.5. Alterations in neocortical neuron morphology as a function of impactor tip shape. Compared to sham injury (E, with black border), cortical injury with a flat tip impactor (A) results in more severe neuron shrinkage and loss of neuronal Nissl staining at 1 h than injury with a rounded tip impactor (B). Neuron death is more rapid with the flat tip impactor, with nearly complete loss of cells with identifiable neuronal morphology by 4 h (C), while morphological damage increases progressively from 4 h (D) to 12 h (F) after injury with a rounded tip impactor. Scale bar represents 100 µm.

Page 71: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

58

Quantification of cortical tissue damage

To quantify cortical damage, the boundaries of the neocortex containing

morphologically identifiable neurons were outlined ipsilateral and contralateral to the

impact on Nissl-stained coronal sections. Gross patterns of early neocortical distortion

and cell death are shown in representative tracings in Figure 2.6A. Within the swollen

ipsilateral cortex, two regions of neocortical damage are clearly evident at 1 h after CCI

with a flat tip impactor, consistent with the regions of acute hemorrhage and neuron loss

illustrated in Figure 2.4A, and corresponding closely to the regions of high principle

strains predicted by finite element model simulation (Figure 2.2B, left panels). At 1 h

after CCI with a rounded tip impactor, neocortical tissue exhibited swelling at the

craniotomy site, but overt tissue damage was only noted at 4 h. The volume of

neocortical tissue damage increased significantly over time (main effect of time, F(4, 38)

= 52.15, p < 0.00001; Figure 2.6B). The temporal progression was strongly dependent

on the tip geometry used to create the injury (interaction of tip and time, F(4, 38) = 11.70,

p< 0.00001). At 1 h postinjury, both groups exhibited negative contusion volumes due to

neocortical swelling and herniation (see also Figures 2.4A, 2.4B and 2.6A). By 4 h

postinjury, the amount of cortical damage increased significantly for the group injured

with the flat tip (p<0.0005 compared to 1 h) but not the rounded tip impactor. No further

increase in contusion volume was measured for mice injured using the flat tip impactor,

suggesting that the majority of neocortical damage occurred by 4 h postinjury. In

contrast, mice injured with a rounded tip impactor showed a progressive increase in

contusion volume from 4 h to 12 h (p < 0.001) and from 12 h to 24 h (p < 0.001).

Cortical tissue damage was greater with the flat tip impactor than the rounded tip

impactor at both 4 h (p < 0.0005) and 12 h (p < 0.005), but was equivalent at 24 h and 9

d. These data demonstrate CCI injury with a rounded tip impactor resulted in a slower

Page 72: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

59

evolution of neocortical cell death over the first 12 h, but ultimately the same volume of

cortical tissue damage, as compared to injury with a flat tip impactor.

Page 73: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

60

Figure 2.6. Quantification of cortical tissue damage. (A) Representative tracings of the dorsal regions of the ipsilateral and contralateral neocortex containing morphologically identifiable neurons at the epicenter of injury 1h after impact with a flat tip impactor and 1h and 4h after injury with a rounded tip impact. Arrows mark the location of the craniotomy edges as in Figure 2.3. (B) The volume of cortical damage produced by injury with a flat tip impactor was maximal by 4 h and was unchanged out to 9 d. In contrast, cortical injury with a rounded tip impactor led to a more slowly developing cortical contusion that increased in size over 24 h, reaching a contusion volume equivalent to that of the flat tip impactor group. Square symbols denote flat tip impactor group means; diamond symbols denote round tip impactor group means. Error bars represent standard deviations. (+ p< 0.0005 compared to the previous time point for the flat tip impactor group; ** p< 0.001 compared to the previous time point for the rounded tip impactor group; # p < 0.005 comparing flat tip and rounded tip impactor groups at the same time point).

Page 74: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

61

Regional hippocampal neurodegeneration

Fluorojade staining was used to evaluate acute regional neurodegeneration in

the hippocampus. Only a few faintly labeled neurons were observed at 1 h postinjury

(data not shown). At 4, 12 and 24 h, Fluorojade-positive neurons were clearly visible in

both groups of mice after 1.0mm CCI brain injury. Injury with a flat tip impactor produced

widespread neurodegeneration in the granule layers and hilus of the dentate gyrus and

in the CA3 pyramidal layer, with less degeneration in the CA1 pyramidal layer (Figure

2.7A, C, E). Neurodegeneration was most prevalent in the dentate gyrus following injury

with a rounded tip impactor, with fewer Fluorojade-positive neurons in the CA3 pyramidal

layer and very few, if any, in the CA1 region (Figure 2.7B, D, F).

Page 75: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

62

Figure 2.7. Hippocampal neurodegeneration after controlled cortical impact (CCI) with a flat tip (A, C, E) or a rounded tip (B, D, F) impactor. Fluorojade-B staining of sections taken at the impact epicenter illustrate degenerating neurons in the dentate gyrus granule layers, dentate hilus, and CA3 and CA1 pyramidal layers at 4 h (A, B), 12 h (C, D) and 24 h (E, F) after 1.0 mm CCI brain injury. Scale bar represents 500 µm.

Page 76: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

63

Fluorojade-positive neurons were counted in the ipsilateral hippocampus at 4, 12,

and 24 h postinjury. In the dentate gyrus, there was no overall effect of tip geometry or

time, but the interaction between these variables was significant (F(2,24) = 6.15, p<

0.01; Figure 2.8A). The flat tip impactor resulted in a initial wave of neurodegeneration

in the dentate gyrus at 4 h that was reduced by 12 h, although this decrease did not

reach statistical significance (p = 0.06). The number of Fluorojade-positive dentate gyrus

neurons increased from 12 h to 24 h after injury with the flat tip impactor (p<0.05).

Numbers of degenerating dentate gyrus neurons did not change over time for the group

injured with a rounded tip impactor, and the numbers were not different from the flat tip

impactor group at any individual time point. In the CA3/CA3c pyramidal layer, CCI with

the flat tip impactor resulted in significantly greater neurodegeneration across the first

24h when compared to the rounded tip impactor (main effect of tip shape, F(1,24) =

7.30, p < 0.05; Figure 2.8B). Numbers of Fluorojade-positive cells decreased from 4 h to

12 h (p < 0.05) and then increased again at 24 h (p < 0.05 compared to 12 h), a

temporal trend that appeared more pronounced for the flat tip impactor. Injury with a flat

tip impactor also produced greater neuron death in the CA1 region than did the rounded

tip impactor (main effect of tip shape, F(1,24) = 22.90, p < 0.0001; Figure 2.8C), an

effect that was time dependent (interaction of tip and time, F(2, 24)=4.10, p < 0.05). At

4 h, numbers of Fluorojade-labeled CA1 neurons were more than ten-fold higher after

injury with a flat tip impactor than with a rounded tip impactor (p < 0.001), and were

significantly higher than those at 12 h or 24 h for either tip geometry (p < 0.05),

indicating a very acute wave of CA1 cell death induced only by the flat tip impactor.

Page 77: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

64

Figure 2.8. Quantification of regional hippocampal neurodegeneration as a function of impactor tip shape. Fluorojade-B positive (FJB+) neurons were counted in the (A) dentate gyrus, including the granule layers and hilar region, (B) CA3/CA3c pyramidal layer, and (C) CA1 pyramidal layer at 4 h, 12 h, and 24 h after brain injury with a flat tip impactor (dark grey bars) or rounded tip impactor (light gray bars). Data are shown as means + standard deviation. (+ p < 0.05 compared to the adjacent time point for the flat tip impactor group; # p < 0.001 comparing flat tip and rounded tip impactor groups at the same time point).

Page 78: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

65

Blood-brain barrier (BBB) damage

Regional patterns of BBB breakdown were comparable after CCI injury using a

flat tip or rounded tip impactor. At 1 h, IgG extravasation was localized to the impact site

spanning neocortical layers I through VI, as well as the subcortical white matter and CA1

stratum oriens (Figure 2.9A, B). By 4 h, BBB disruption had extended beyond the

impact site in the neocortex and encompassed the entire ipsilateral hippocampus at the

level of the impact epicenter (Figure 2.9C, D). At 12 and 24 h, in addition to the robust

IgG immunostaining in the ipsilateral cortex, subcortical white matter and hippocampus,

faint IgG labeling was observed in the dorsal thalamus and in the contralateral cortex

and hippocampus near midline. At 9 d, IgG extravasation was less pronounced than at

24h.

Page 79: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

66

Figure 2.9. Acute blood-brain barrier damage after severe controlled cortical impact using a flat tip impactor (A, C) or a rounded tip impactor (B, D). The location and progression of extravasation of immunoglobulin (IgG) was similar for injury with the flat tip and rounded tip impactors, as illustrated at 1 h (A, B) and 4 h (C, D) after injury.

Page 80: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

67

Traumatic axonal injury

Traumatic axonal injury was assessed qualitatively using immunohistochemistry

for APP, a well-established marker of axonal injury. Impact to the cortex with either a flat

tip or rounded tip impactor resulted in acute axonal injury throughout cortical and

subcortical structures in the ipsilateral hemisphere. There were no overt differences in

the distribution or time course of axonal injury between the two tip geometries. From 1 h

to 24 h post-injury, axonal injury was evident along the edges of the impact site in the

neocortex, and in the deep neocortex (layer VI) and the subcortical white matter tract

below the impacted cortex (Figure 2.10A, B). APP-positive injured axons were also

observed in the dorsal ipsilateral thalamus (Figure 2.10C, D) and occasionally within the

hippocampus, most often in the stratum oriens of the CA3 and CA1 regions and along

the hippocampal fissure. Qualitatively, the frequency and staining intensity of APP-

positive axons increased from 1 h to 12 h. No axonal injury was observed in the

contralateral hemisphere for any of the time points for either tip shape.

Page 81: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

68

Figure 2.10. Traumatic axonal injury induced by injury with either a flat tip (A, C) or rounded tip (B, D) impactor. The spatial patterns of axonal injury, detected using immunohistochemistry for amyloid precursor protein, were similar for mice injured with a flat tip or rounded tip impactor. In the neocortex, axonal injury was concentrated around the periphery of impact site and in the subjacent white matter and gray matter/white matter interface (12 h postinjury shown; A, B). Axonal injury was also clearly evident in subcortical structures such as the hippocampus (not shown) and dorsal thalamus (24 h shown; C, D; scale bar represents 200 µm).

Page 82: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

69

Motor and cognitive behavioral outcome

Deficits in coordinated motor function were evaluated using a modified NSS at 1

h and 1, 2, 5 and 7 d after CCI brain injury. Tip geometry was not a factor in determining

the level of initial motor dysfunction or the rate of recovery of function (Figure 2.11A).

Both groups of brain-injured mice exhibited profound motor deficits using the NSS at 1 h

post-injury and a significant spontaneous recovery over time (main effect of time, F(4,32)

= 37.71, p < 0.0001). Similarly, evaluation of basic motor functions including limb

flexion, extension and grip strength using a composite neuroscore revealed equivalent

deficits in mice injured using either a flat tip or rounded tip impactor (Figure 2.11B). Both

groups of mice showed notable motor dysfunction at 1 d after CCI, and significant

recovery of function over time (main effect of time, F(3,24) = 14.26, p < 0.0001).

As with posttraumatic motor deficits, the use of a flat tip or rounded tip impactor

did not significantly alter posttraumatic cognitive function assessed in a Morris water

maze at one week after severe CCI (Figure 2.11C). Learning latencies were equivalent

for the two groups, with both achieving significantly lower latencies to the platform on

days 8 and 9 compared to day 7 (p < 0.0005).

Page 83: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

70

Figure 2.11. Effect of impactor tip shape on neurological motor and cognitive function of mice subjected to cortical impact injury. Mice injured with a flat tip impactor or a rounded tip impactor exhibited equivalent motor deficits over the first week after injury, when measured using (A) a 14-point modified neurological severity score (NSS) or (B) a 12-point composite neuroscore. (C) Impactor tip shape also had no significant effect on the ability to learn the location of a hidden platform in a Morris water maze over a three day testing period. Dark gray bars or square symbols denote flat tip impactor group means; light grey bars or diamond symbols denote round tip impactor group means. Error bars represent standard deviations.

Page 84: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

71

Discussion

The CCI model of contusion brain injury is one of the most widely used models of

TBI in mice. Mouse models are being increasingly utilized to exploit transgenic and

knockout technologies for the study of specific proteins or for expression of tissue or cell

specific reporters. The low cost and small body weight of mice are also attractive for

testing therapeutic interventions, especially when large cohorts are needed to perform

comprehensive dose response studies or test behavioral outcomes. We have

demonstrated that in the mouse CCI model the rate of neocortical neurodegeneration is

highly dependent on the shape of the impactor tip. A 1.0 mm depth injury with a flat tip

impactor results in significantly faster cortical neuron death, more pronounced acute

hemorrhage, and greater initial hippocampal neurodegeneration than injury with a

rounded tip impactor. Injury severity, assessed by (a) impact depth and velocity, (b) the

size of the neocortical contusion at 9 d after injury, (c) the presence or pattern of BBB

breakdown and axonal injury, and (d) the magnitude of neurobehavioral deficits, was

equivalent for the two impactors. These data suggest that rounding the tip of the

impactor slows the temporal course of the neurovascular damage without reducing or

eliminating important pathophysiological features of CCI brain injury.

A three-dimensional finite element model was utilized to predict tissue strains in

response to a 1.0 mm 3.5 m/s CCI injury using flat tip or rounded tip impactor. Maximum

principal strains were predicted to be substantially higher for the flat tip impactor than for

the rounded tip impactor. For the flat tip impactor, tissue strains were predicted to be

highest around the edge of the impactor as it penetrated the cortical parenchyma.

Histological damage, including hemorrhage and loss of Nissl stain, was indeed observed

most frequently at the boundaries of the impactor’s path as early as 1 h after injury with

Page 85: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

72

a flat tip impactor (see Figures 2.4 and 2.6). Interestingly, the strain at which human

cerebral arteries fail under dynamic elongation ex vivo is estimated to be 0.50, or 50%

(Monson et al. 2003). Based on these data, arterial rupture or tearing would be

predicted along the edges of the path of the flat tip impactor, consistent with the

presence of hemorrhage in this region. For the rounded tip impactor, maximum strains

were predicted to be centered beneath the impactor, more diffusely distributed in the

deep cortex, consistent with strain contours generated for CCI injury in the rat (Mao et al.

2010). One limitation of the current mouse brain model is that the finite element meshes

were morphed from the previously validated rat brain model with some adjustments

made on the brain outer surface, ventricles, and white matter. Gray matter structures

were assigned the same material properties. Detailed meshes representing other

anatomical parts, such as hippocampus or thalamus, and incorporation of region-specific

material property heterogeneities, are required in the future to improve its biofidelity.

Testing mouse brains to acquire mouse-specific properties would increase the prediction

accuracy of the numerical mouse brain model. Tears within the brain parenchyma

during impact would necessarily alter the strain pattern in neighboring regions. However,

micro-tearing of brain tissues or intracranial vasculatures was not addressed in our finite

element model because a much higher resolution brain model would be needed for

predicting such injuries. Unfortunately, a very fine mesh model is computationally

challenging at present. Even if such a model existed and were computationally feasible,

very little is known regarding strain thresholds for individual cellular components within

the brain.

Dynamic strains of 0.10-0.20 (10-20%) or above are sufficient to elicit

morphological signs of injury or dysfunction in neurons or axons (Bain and Meaney

2000; Geddes et al. 2003; Cater et al. 2006; Elkin and Morrison 2007). During focal or

Page 86: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

73

diffuse TBI, tissue deformation leads to early compromise of the normal selective barrier

function of the plasmalemma (Farkas et al. 2006; Whalen et al. 2008), contributing to ion

dysregulation (Kilinc et al. 2009). Both enhanced neuronal membrane permeability and

increases in intracellular free calcium have been shown to be proportional to applied

strain (LaPlaca et al. 1997; Geddes et al. 2003), suggesting that in regions of highest

tissue strains, neurons will more likely experience greater and more sustained calcium

perturbations, which could lead to mitochondrial dysfunction, protease activation and cell

death. While trauma-induced membrane permeability increases may be transient under

some conditions, sustained leakiness results in cell death (LaPlaca et al. 1997; Geddes

et al. 2003; Geddes et al. 2003; Farkas et al. 2006). Collectively, these studies are

consistent with the premise that higher strains generated by a flat tip impactor would be

associated with more severe membrane permeability alterations and calcium

dysregulation with less likelihood of recovery, resulting in rapid cell death. Recent work

supports a direct correlation between maximum principal strains predicted by finite

element modeling and the degree of neuronal degeneration in a rat model of CCI brain

injury (Mao et al. 2010) (Mao et al., 2010a).

Quantification of the progression of cell death within the ipsilateral neocortex

following severe contusion injury with either a flat or rounded tip impactor revealed

marked differences in the evolution of neocortical damage. Injury with a flat tip impactor

produced rapid cell death, with maturation of the neocortical lesion by 4 h, whereas

impact with a rounded tip resulted in milder initial neocortical damage that progressively

increased out to 24 h post-injury to a level equivalent to that produced by the flat tip

impactor. Although relatively little information is available on the acute time course of

cortical damage after CCI, our findings are consistent with previous studies using CCI

with a flat tip impactor in mice. Using propidium iodide exclusion as an indicator of

Page 87: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

74

membrane integrity, Whalen et al. (2008) showed marked increases plasma membrane

compromise in the cortex at 1 h after 0.6mm, 6 m/s CCI, after which most membrane-

compromised neurons degenerated. Similarly, Hall and colleagues (Thompson et al.

2006; Hall et al. 2008) demonstrated that the size of the cortical cavitation (lesion

volume) is fully developed by 6 h after 1.0 mm, 3.5 m/s CCI. In the CCI model, the

amount of neocortical cell death can be altered by changing the depth or velocity of

impact in the mouse (Fox et al. 1998; Hannay et al. 1999; Saatman et al. 2006) and rat

(Sutton et al. 1993; Goodman et al. 1994). However, because these histological

evaluations were typically carried out days to weeks after the injury, it is unclear to what

extent velocity or depth of impact affects the rate of cortical neurodegeneration. Injury

severity and the rate of cell death may also vary for different injury devices. Fox et al.

(1998) use a custom-made CCI device with a flat tip impactor and report no cortical

cavitation at 7 d after a 1 mm depth, 4.5 m/s injury to C57BL/6 mice. Nevertheless, a 1

mm impact at a higher velocity (6 m/s) resulted in neocortical neurodegeneration

concentrated at the periphery of the flat tip impactor, a pattern consistent with our finite

element predictions, as described above.

In addition to a prominent neocortical contusion, CCI brain injury typically results

in neuron loss in the hippocampal dentate gyrus and CA3 pyramidal layers (Goodman et

al. 1994; Smith et al. 1995; Baldwin et al. 1997; Saatman et al. 2006; Hall et al. 2008).

Although the CA3 subregion is positioned further from the impact than the CA1 region,

CA3 neurons appear more vulnerable to CCI, perhaps due to greater tissue compliance

within the CA3 region compared to the CA1 (Elkin et al. 2010). However, involvement of

the CA1 pyramidal layer is also often reported as impact depth is increased (Goodman

et al. 1994; Scheff et al. 1997; Saatman et al. 2006; Whalen et al. 2008). Quantification

of degenerating neurons revealed that the rates and extent of neurodegeneration within

Page 88: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

75

the hippocampus are altered by the impactor tip geometry. Injury with a flat tip impactor

produced a wave of acute neurodegeneration at 4 h, with the largest number of

degenerating cells in the dentate gyrus, followed by the CA3 and then CA1 regions. A

similar acute phase (peak at 1-6 h) and regional distribution of degenerating neurons

with plasma membrane compromise was reported for mouse CCI with a flat tip impactor

(Whalen et al. 2008). In contrast, impact with a rounded tip appeared to produce a more

consistent progression of hippocampal neurodegeneration across the first 24 h after

injury, with minimal involvement of the CA1 region. Early hippocampal

neurodegeneration may be more extensive following injury with the flat tip impactor as a

result of larger tissue displacement during impact, with the bulk of the cortical tissue

being forced downward into the hippocampal structure. Despite the slightly larger

diameter of the rounded tip, a much smaller volume of tissue is displaced and some of

the cortical tissue may be compressed and deformed laterally, reducing the deformation

of the hippocampus. Results predicted by the finite element model corroborate this

assumption. A peak maximum principal strain of 30.1% was induced using the flat

impactor while the corresponding value was 24.8% for the rounded impactor. Despite

the lower strains that occur in the hippocampus compared to the neocortex with CCI,

hippocampal neurons in vitro exhibit larger intracellular calcium increases and more cell

death compared to cortical neurons in response to stretch injury, suggesting an

enhanced vulnerability to traumatic stimuli (Geddes et al. 2003; Elkin and Morrison

2007).

Blood-brain barrier breakdown is an important aspect of cerebrovascular damage

in TBI (DeWitt and Prough 2003) that has been reproduced in severe CCI(Smith et al.

1995; Hicks et al. 1997). In this study, damage to the BBB was visualized by IgG

immunoreactivity within the ipsilateral neocortex and hippocampus in patterns similar for

Page 89: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

76

both impactor tips. Extravasation of IgG was localized primarily to the neocortex at 1 h,

while at 4 h postinjury IgG labeling was also observed in the hippocampus, as previously

reported (Saatman et al. 2006). Regional patterns of disruption of the BBB in mice were

not substantially modulated in the present study by impactor tip geometry, or by severity

of injury (impact depth) for CCI injury with a rounded tip impactor (Saatman et al. 2006),

suggesting that the threshold for mechanically stimulated BBB damage may be low and

compromise of the BBB may be a common pathology across a range of injury severities.

Our analysis of BBB breakdown is limited in that it was not quantitative, leaving open the

possibility that the degree of extravasation caused by the two tips may be different. In

addition, IgG extravasation yields limited information about the time course of BBB

disruption. Further studies with Evan’s blue administered intravascularly at various

postinjury time intervals could provide more information regarding the time course of

BBB disruption or recovery for these two injury paradigms. Nonetheless, our data

confirm that impact injury with a rounded tip results in neocortical and hippocampal BBB

damage, reproducing an important aspect of CCI pathology associated with secondary

injury cascades such as inflammation and oxidative stress.

Axons are vulnerable to stretch injury induced by tensile/shear strains within the

brain tissue. Traumatic axonal injury results in rapidly altered membrane permeability,

intracellular calcium imbalances and mitochondrial damage, protease activation and

disruption in axonal transport (M.F 2009). Injured axons represent a vital therapeutic

target in diffuse brain injury, representing the defining pathology in diffuse axonal injury.

Sparing axonal integrity may also be important in focal brain trauma. Even in models

designed to produce focal contusions, such as the CCI model, axonal damage is present

in several regions outside the contused neocortex, such as the hippocampus and

thalamus (Lighthall et al. 1990; Dunn-Meynell and Levin 1997; Hall et al. 2005; Hall et al.

Page 90: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

77

2008). Here we show that impact with either a rounded tip or flat tip impactor produced

axonal injury within the contused neocortex and the underlying hippocampus and

thalamus. Although the number of injured axons was not quantified, the onset and

overall distribution of axonal injury was comparable for the two tip geometries.

In preclinical evaluation of therapeutic approaches for TBI, behavioral deficits are

an especially important outcome measure. Both motor and cognitive deficits have been

well described in both rat and mouse models of TBI, utilizing a number of different

behavioral tasks (Hamm 2001; Fujimoto et al. 2004). The severity of behavioral

dysfunction can be altered by changing certain biomechanical aspects of CCI injury,

such as the depth (Saatman et al. 2006; Yu et al. 2009) or velocity (Fox et al. 1998) of

impact. Despite the fact that CCI brain injury with a flat tip impactor resulted in greater

maximal tissue strains than impact with a rounded tip, motor and cognitive functions of

brain-injured mice were not grossly influenced by impactor tip geometry, although small

group sizes may have limited our ability to detect small differences. Behavior was

evaluated at several times after injury using a composite neuroscore, neurological

severity score, and a MWM learning task. This suggests that slowing the progression of

cortical cell death through the use of a rounded tip did not compromise the fidelity of the

behavioral response. Brain injury-induced neuromotor or cognitive impairments in CCI-

injured mice have been attenuated through various treatment approaches, illustrating the

influence of secondary injury cascades on posttraumatic behavioral responses (You et

al. 2008; Clausen et al. 2009; Longhi et al. 2009; Mbye et al. 2009). Therefore, the use

of a rounded tip impactor may provide a greater therapeutic window for targeting acute

cell dysfunction or death without diminishing the power of the CCI model for the

evaluation of behavioral impairment and recovery.

Page 91: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

78

In summary, we have quantitatively and qualitatively investigated the effects of

impactor tip geometry on predicted tissue strains, histological outcomes and behavioral

responses following severe lateral CCI brain injury in mice. Compared to CCI with the

commonly used flat tip impactor, injury with a rounded tip impactor resulted in more

uniform maximum principal strains with lower peak magnitudes, reduced acute

neurovascular disruption, and a slower progression of neocortical cell death. A more

gradual evolution of neocortical neuronal death was achieved with a rounded tip CCI

without altering the size of neocortical contusion at 9 days after injury or substantially

changing other clinically relevant aspects of this brain injury model such as BBB

breakdown, axonal injury or neurobehavioral dysfunction. By slowing the progression of

neuronal damage, the CCI model may better mimic the evolution of contusive damage in

human head injury. Furthermore, lengthening the time between the primary insult and

the eventual death of the cell is advantageous for mechanistic studies aimed at

distinguishing the onset, duration and interactions of upstream and downstream events,

or isolating specific aspects of injury cascades. Slowing the time course of neuronal

death also yields a longer therapeutic window, increasing the likelihood that therapeutic

compounds can be administered using a clinically relevant postinjury paradigm, reach

therapeutic levels in the CNS and affect the desired targets. These data suggest that

along with impact depth and velocity, impactor tip shape is an important determinant of

acute tissue response to rapid impact brain injury. Therefore, in studies of acute injury

mechanisms of contusive TBI or in evaluations of therapeutic interventions targeting

neuronal death, impactor tip geometry should be carefully considered.

Copyright © Shaun William Carlson 2013

Page 92: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

79

Chapter 3: Prolonged Continuous Systemic Infusion of IGF-1 after Severe

Controlled Cortical Impact

Introduction

There are an estimated 1.7 million annual cases of traumatic brain injury (TBI)

that result predominantly from motor vehicle incidents, falls, and violence (Langlois et al.

2006; Summers et al. 2009; Faul M 2010). TBI is an acquired condition that can afflict

individuals independent of age and gender, but the highest prevalence is found in males

aged 20-30 (Langlois et al. 2006). TBI is characterized by an initial mechanical

disturbance and the subsequent activation of secondary injury cascades that can

culminate in cellular dysfunction and death (Lynch and Dawson 1994; Marklund et al.

2006; Beauchamp et al. 2008). Neurological impairment, including motor and cognitive

dysfunction, is frequently reported in brain-injured patients (Capruso and Levin 1992;

Lundin et al. 2006; Selassie et al. 2008; Jang 2009).

The mouse model of lateral controlled cortical impact (CCI) reliably reproduces

cortical contusion, selective hippocampal neuron loss, and neurobehavioral deficits

including motor and cognitive dysfunction (Smith et al. 1995; Saatman et al. 2006;

Pleasant et al. 2011), recapitulating important features of the pathobiology associated

with human contusion TBI. CCI is a highly characterized model that is widely utilized for

preclinical assessment of therapeutic agents for the treatment of TBI.

The activation of multiple secondary cascades creates a substantial challenge for

therapeutic agents targeting a single mechanism of the secondary injury cascade after

TBI (Beauchamp et al. 2008; Margulies and Hicks 2009). Alternatively, a therapeutic

agent, for example a neurotrophic factor, that targets multiple mechanisms of secondary

injury and can promote regeneration, may prove efficacious for the treatment of TBI. The

Page 93: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

80

neurotrophic factor insulin-like growth factor-1 (IGF-1) is a highly conserved 7.6 kDa

protein, predominantly synthesized in the liver, which can cross the blood-brain barrier

by receptor-mediated endocytosis (Pardridge 1993; Reinhardt and Bondy 1994). IGF-1

is also synthesized in neurons and astrocytes of the adult rodent brain (Bondy et al.

1992; Bondy and Lee 1993; Bondy and Cheng 2004). Binding of IGF-1 to its cognate

receptor, which is highly expressed in the cortex, hippocampus and choroid plexus of the

adult rodent brain, results in the downstream activation of Akt (Bondy and Lee 1993).

Increased activation of Akt is linked with increased neuron survival after hypoxic-

ischemic injury and experimental TBI (Noshita et al. 2001; Noshita et al. 2002; Brywe et

al. 2005). In addition to enhancing neuron survival, IGF-1 also promotes neurogenesis

by enhancing proliferation and neuronal differentiation of newly proliferated cells in the

hippocampus (Arsenijevic and Weiss 1998; Aberg et al. 2000; Brooker et al. 2000;

Arsenijevic et al. 2001; Trejo et al. 2001; Yan et al. 2006). The neuroprotective and

neurogenic effects of IGF-1 make it a promising therapeutic agent for the treatment of

TBI.

Only a handful of studies have evaluated the therapeutic efficacy of IGF-1 in the

context of TBI. Systemic administration of IGF-1 attenuates deficits in spatial memory in

mice subjected to a mild brain injury (Rubovitch et al. 2010) and attenuates motor and

cognitive dysfunction in rats subjected to moderate TBI (Saatman et al. 1997). Previous

work from our lab also demonstrated that conditional astrocyte-specific overexpression

of IGF-1 attenuates motor and cognitive dysfunction and reduces hippocampal

neurodegeneration in CA-3 and the dentate gyrus granular layer at 3 d after severe CCI

(Madathil 2013). In a phase II clinical trial, brain-injured patients that received IGF-1 by

intravenous infusion demonstrated improved nitrogen balance and, in a subset of

patients in which serum IGF-1 concentrations were maintained above 350 ng/ml for 1

Page 94: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

81

week, improved outcome at 6 months (Hatton et al. 1997). Only one of these studies

(Madathil 2013) demonstrated elevated brain levels of IGF-1 and showed that increased

neuron survival may underlie the observed recovery of neurobehavioral function with

IGF-1 after TBI. Taken together, these studies suggest that IGF-1 is a viable and

promising treatment for TBI, and highlight the need for additional preclinical studies to

evaluate if systemically infused IGF-1 is neuroprotective and contributes to behavioral

improvement in mice subjected to contusion TBI.

A dose of 4 mg/kg/d IGF-1 was selected based on a previous study that

demonstrated this dose improved motor and cognitive function in moderately brain-

injured rats (Saatman et al. 1997). We sought to evaluate if treatment with IGF-1

improved recovery in motor function during the first week following CCI using the same

dose that provided behavioral benefit in a rodent contusion TBI model. We will expand

on previous work by quantifying levels of IGF-1 in the serum and brain, confirming IGF-1

signaling in the brain, and determining if treatment with IGF-1 protects against acute cell

loss.

In light of the previous observation that overexpression of IGF-1 reduced

hippocampal neurodegeneration at 3 d following severe CCI, we sought to evaluate if

systemic infusion of IGF-1 reduced cortical tissue damage and attenuated regional

hippocampal neurodegeneration at 3 d following severe CCI. Hippocampal

neurodegeneration is observed in the dentate gyrus granular layer, CA3 and CA1 at 2

and 3 d following CCI (Anderson et al. 2005; Hall et al. 2005; Saatman et al. 2006; Hall

et al. 2008; Cai et al. 2012; Zhou et al. 2012). Within the hippocampus, neuronal

differentiation of progenitor cells in the subgranular zone give rise to new neurons.

Newborn neurons are generated throughout adulthood in the dentate gyrus subgranular

Page 95: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

82

zone of the hippocampus (Eriksson et al. 1998; Zhao et al. 2008). A reduction in the

number of immature neurons in the hippocampus is associated with reductions in spatial

learning and memory, suggesting that newborn neurons are critical for cognition

(Clelland et al. 2009; Deng et al. 2009; Jessberger et al. 2009). Following CCI, the

number of hippocampal immature neurons is markedly reduced in the dentate gyrus

granular layer within 3 d of injury (Rola et al. 2006; Gao et al. 2008). We postulated that

reduced hippocampal neurodegeneration in the granular layer may be indicative of

enhanced immature neuron survival.

The data presented in this chapter provides preliminary evidence that prolonged

continuous systemic infusion of 4 mg/kg/d IGF-1 does not promote improved recovery of

motor function after severe CCI in mice. Although hIGF-1 could not be detected in the

brains of treated mice, systemic infusion of IGF-1 enhanced Akt activation in the

contused brain at 7 d post-injury. However, treatment with IGF-1 showed little efficacy in

reducing hippocampal neurodegeneration and did not attenuate loss of immature

neurons at 3 d following CCI.

Page 96: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

83

Materials and Methods

Animals

8-12 wk old C57BL/6 male mice utilized for this study were purchased from

Jackson Laboratories (Bar Harbor, ME). All protocols were approved by the University of

Kentucky’s Institutional Animal Care and Use Committee in accordance with established

guidelines from the Guide for the Care and Use of Laboratory Animals from the National

Institutes of Health. Animals were housed 5 mice per cage in the University of Kentucky

Medical Center animal vivarium with a 14:10 hr light:dark photoperiod and were provided

food and water ad libitum; however, food intake was not monitored in the mice utilized in

this study.

Controlled Cortical Impact and Minipump Implantation

The CCI injury was performed as previously described (Madathil et al. 2010;

Pleasant et al. 2011). Mice were anesthetized with 3% isoflurane to prepare the scalp for

surgery and for placement in the stereotaxic frame (David Kopf Instruments, CA).

Anesthesia was maintained with 3% isoflurane via a nose cone for the duration of the

surgical procedure. A midline incision was created and the skin reflected to expose the

skull. A 5 mm craniotomy was performed midway between bregma and lambda lateral to

the sagittal suture over the left hemisphere of the brain. Animals were randomly

assigned to receive either sham or CCI injury. Sham-injured mice received anesthesia

and only a craniotomy. In brain-injured mice, the CCI injury was induced using a

computer-controlled pneumatically driven piston that rapidly impacted the intact dura of

the brain at 1 mm depth with a velocity of 3.5 m/s using a 3 mm diameter rounded

impactor tip (TBI-0310 Impactor, Precision Systems and Instrumentation, VA). In the 7 d

Page 97: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

84

cohort, mice were subjected to sham injury (n=3/treatment group) or CCI (n=5/treatment

group) and euthanized at 7 d post-injury for analysis of brain tissue by western blot and

ELISA. In the 3 d cohort, mice were subjected to CCI (n=5/treatment group) and

euthanized at 3 d post-injury for histological assessment. Following injury, a small

circular disk of dental acrylic was adhered to the skull over the craniotomy site and the

scalp was sutured. At 15 minutes after injury, a small incision was made in the back to

facilitate subcutaneous implantation of a primed Alzet osmotic minipump (model 1003D

or 1007D) for the delivery of 4 mg/kg/d recombinant human IGF-1 (hIGF-1; National

Hormone and Peptide Program, CA) or vehicle (pH 7.4, 100mM acetic acid diluted in

USP grade PBS) over a period of 3 or 7 d, to provide infusion of hIGF-1 or vehicle until

the animal was euthanized. Following osmotic minipump implantation, mice were placed

on a heating pad to maintain normal body temperature. Once ambulatory, the mice were

returned to their home cage.

Motor Function Assessment

A previously described modified neurological severity score (NSS) (Pleasant et

al. 2011) task was used to evaluate motor dysfunction at 3 hr, 1, 3, 5, and 7d post-injury.

At 1 d prior to injury, mice were acclimated to each of four Plexiglas beams of varying

widths (3, 2, 1, 0.5 cm) and a 0.5 cm diameter wooden rod. The beams and rod were 60

cm in length and elevated 47 cm above the table top. Mice were allowed 30 seconds to

traverse the beams and rod during the periods of acclimation and testing. Each beam

was assigned a maximum of three points and the rod two points. Three points were

given for successful crossing of the beam with normal position and utilization of the

forelimb and hindlimb. Two points were given for successful crossing of the beam

despite either a forelimb or hindlimb hanging from the beam, and one point was given for

Page 98: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

85

crossing the beam despite inverting below the beam one or more times. The mouse was

righted and allowed to continue across the beam if it became inverted on the beam. A

score of zero was given if the mouse did not cross in the 30 second time or fell off the

beam. For the wooden rod, two points was given for successful crossing of the beam in

the allotted time. A score of one was given for crossing despite inverting greater than

three times. A score of zero was given if the mouse did not traverse in the allotted time

or fell off the rod.

Tissue Collection and Preparation

At 1 d post-injury, 25 µL of blood was collected from the saphenous vein of mice

from the 7 d cohort. Blood was collected in an eppendorf tube and allowed to coagulate

for 30 minutes at room temperature. Collected blood was centrifuged at 4500g for 10

minutes to separate the serum. Isolated serum was stored at -80°C.

In the 7 d cohort, mice received an overdose of Fatal-plus (65 mg/kg sodium

pentobarbital). Blood was collected transcardially with a 22G needle connected to a 1

mL syringe. To concentrate collection of cortical tissue on the injury site, 3 mm of the

rostral brain was blocked, and the remaining cortical regions of each hemisphere were

rapidly dissected and stored in separate tubes at -80°C. Cortical tissue from the injured

hemisphere was homogenized by wand sonicator in cold lysis buffer (1% triton X-100,

20 mM Tris-HCl, 150 mM NaCl, 5 mM EGTA, 10 mM EDTA, 10% glycerol, and a cocktail

of proteinase inhibitors (Roche, IN)) and centrifuged for 30 minutes at 4°C at a speed of

10,000xg. The supernatants were collected and utilized for analysis. Protein

concentrations were determined using a BCA assay kit (Pierce Biotechnology, IL). The

prepared cortical samples were utilized for western blot and ELISA analysis.

Page 99: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

86

In the 3 d cohort, mice received an overdose of Fatal-plus (65 mg/kg sodium

pentobarbital). Mice were transcardially perfused with heparinized saline to clear blood

from the vasculature. The mice were perfusion fixed using 10% neutral buffered

formalin. The mice were decapitated and the heads placed in 10% neutral buffered

formalin for 24 hr. The brains were carefully removed from the head and placed in 10%

neutral buffered formalin for an additional 24 hr. Brains were placed in 30% sucrose for

24 hr for cryoprotection prior to freezing in -30°C isopentanes cooled on dry ice. Brains

were cut into 40 µm coronal sections on a freezing sliding microtome (Microm, Dolbey-

Jamison, PA) at -20°C (Physitemp, NJ).

Quantification of Human IGF-1 by ELISA

Concentrations of hIGF-1 were quantified using a highly specific human IGF-1

ELISA with a sensitivity range of 16 ng/mL to 1137 ng/mL (Immunodiagnostic Systems,

kit #AC-27F1). The ELISA kit was used in accordance with the kit’s instructions. Serum

and brain samples were pretreated to dissociate IGF-1 from IGF-1 binding proteins.

Human IGF-1 standards (16-1137 ng/mL) and pretreated samples were pipetted in

duplicate into a 96 well plate coated with an antibody specific to hIGF-1. Absorbance of

each well was measured using a microplate reader (Tecan, NC) at the 450 nm and 540

nm wavelengths. The absorbance at 540 nm was subtracted from the 450 absorbance to

account for the background of each well.

Western Blot

Western blot analyses were performed as previously described (Madathil et al.

2010). Electrophoresis was completed using either 20 µg (neurofilament NF68) or 30 µg

(phosphorylated Akt) of protein supernatant from the injured cortex on a 3-8% Tris-HCl

Page 100: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

87

gel running at 150V. After transfer onto nitrocellulose membranes, the membranes were

blocked for 1 hr in 5% dry milk dissolved in 0.1% Tween 20 diluted in TBS, and

incubated overnight with either the primary antibody for p-Akt ser473 (rabbit monoclonal,

1:2000, Cell Signaling Technology, MA), or for NF68 (NR4, mouse monoclonal, 1:1000,

Sigma, MO). Secondary antibodies used were directly conjugated with an infrared dye

(1:10000 IRDye800CW, Rockland, PA). Membranes were imaged and optical densities

(OD) were quantified on the Li-Cor Odyssey Infrared Imaging System (Li-Cor

Biosciences, NE). After p-Akt or NF68 development, the membranes were reprobed for

actin using a primary antibody for anti-β-actin (mouse monoclonal 1:5000, Calbiochem

Inc, CA) and a secondary antibody for anti-mouse IgM conjugated to an infrared dye

(1:10000, Rockland). For quantification, the OD of each p-Akt or NF68 band was

normalized to its respective actin OD.

Cresyl Violet and Fluorojade-B Staining

A parallel set of 40 µm-thick coronal sections (n=10-12/mouse), spaced 400 µm

apart, were mounted and air-dried on gelatin-coated slides. The slides were hydrated in

graded ethanol solutions, rinsed in water, stained with 0.5% cresyl violet (Acros

Organics, NJ), dehydrated through graded ethanol solutions, and cleared in xylenes

(Fisher Scientific, NJ) prior to mounting with Permount (Fisher Scientific). Three 40 µm-

thick brain sections, 400 µm apart, centered on the injury epicenter, were stained with

Fluorojade-B (FJB; Millipore Co, MA) to visualize acute hippocampal neurodegeneration

after injury, as previously described (Pleasant et al. 2011).

Page 101: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

88

Immunohistochemistry

Three 40 µm-thick coronal brain sections, spaced 400 µm apart, centered on the

injury epicenter, were stained with doublecortin (DCx) to label immature neurons, using

standard free floating immunohistochemistry staining protocols, as previously described

(Cai et al 2012). A primary antibody for DCx (rabbit polyclonal, 1:500, Abcam, MA) and a

secondary antibody for anti-rabbit IgG directly conjugated to Alexa Fluor® 488 (donkey

polyclonal, 1:1000, Jackson ImmunoResearch, PA) were utilized to visualize immature

neurons in the granular layer. Sections were rinsed in TBS and treated with 5% normal

horse serum with 0.1% Triton-X-100 in TBS for 30 minutes to block nonspecific binding

sites. The DCx antibody was diluted in blocking solution and the tissue incubated

overnight at 4°C. The following day, tissue was rinsed with TBS and incubated with the

secondary antibody diluted in blocking solution for 1 hr. Tissue was rinsed with TBS,

incubated with Hoechst stain (1:10,000, Invitrogen, CA) for 1.5 minutes to label cell

nuclei and rinsed with TBS. Labeled sections were mounted on gelatin-coated slides,

coverslipped with Fluoromount (Southern Biotech, AL), and stored at 4°C.

Image Acquisition and Quantification

Images of cresyl violet, FJB, and DCx staining were acquired using an AX80

Olympus microscope (PA). Quantification for FJB and DCx was performed in the upper

and lower blades of the ipsilateral dentate gyrus granular layer of each section. The

dentate gyrus granular layer was utilized as an anatomical boundary and served as the

region of interest for quantification. FJB and DCx positive cells were quantified within

three sections centered around the injury core, as previously described for the

quantification of degenerating neurons in the hippocampus (Pleasant et al. 2011). All

positively labeled cells were manually counted through all focal planes within the

Page 102: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

89

granular layer (FJB and DCx), CA3 (FJB) and CA1 (FJB) at 40 x magnification on an

epifluorescent microscope (Olympus BX51) equipped with a FITC filter (41001, Chroma

Technology, VT). All quantification was performed by an investigator blinded to the

treatment and injury conditions of each animal. The DCx cell quantification is expressed

as a cellular density (cells/mm3) and reflects the summation of positively labeled cells

counted in the subgranular, inner, and outer granular layers. The Hoechst-stained

dentate gyrus granular layer was imaged (Olympus, AX80) and area measured using

ImagePro (MediaCybernetics, MD). The thickness of each section was measured, in

microns, using an epifluorescent scope (Olympus BX51) equipped with a stereology

stage. Measurements of granular layer area and thickness were used to calculate the

granular volume of each section in which quantification was completed.

Statistical Analysis

All data are presented as mean + standard error of the mean (SEM). Analyses of

hIGF-1 ELISA data and western blot data were performed by one-way analysis of

variance (ANOVA) and when applicable, the Newman-Keuls post-hoc t-test was used for

individual comparisons. Cellular counts (FJB and DCx) were analyzed by region using

Student’s t-tests. Neurological severity score data was analyzed by a repeated

measures one-way ANOVA. Statistical tests were completed using Statistica (Statsoft

Inc, OK). A p value less than 0.05 was considered significant for all tests.

Page 103: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

90

Results

To quantify the levels of hIGF-1 in the brain following systemic infusion of IGF-1,

we measured the human isoform of IGF-1 in serum collected at 1 and 7 d post-injury

using a highly-sensitive human-specific ELISA kit. Human IGF-1 was not detected in the

serum of mice infused with vehicle. Systemic infusion of 4 mg/kg/d hIGF-1 for a period of

7 d resulted in sustained elevated levels of hIGF-1 in sham-injured and brain-injured

mice during the infusion period (Fig. 3.1A). The serum concentrations of hIGF-1 in

sham-injured mice were not significantly different from those measured in brain-injured

mice. In order to verify that systemically delivered IGF-1 reached the brain, we quantified

the levels of hIGF-1 in the brain at 7 d post-injury using the same ELISA kit. Human IGF-

1 was not detected in cortical samples from IGF-1-infused sham-injured and brain-

injured mice suggesting that the levels of hIGF-1 in the brain may be lower than the

detection limits of the ELISA assay.

As an alternate means of verifying that IGF-1 administered subcutaneously

reached the brain, we evaluated Akt activation, a downstream mediator of IGF-1

signaling, as a surrogate measure of hIGF-1 activity in the brain. Western blot analysis

at 7 d post-injury revealed that brain-injured vehicle-treated mice exhibited a 21%

increase in cortical Akt activation compared to sham-injured vehicle-treated mice, but

this did not reach statistical significance (Fig. 3.1B). Brain-injured IGF-1-treated mice

exhibited a significant 57% increase in cortical Akt activation compared to brain-injured

vehicle-treated mice (p<0.05; Fig. 3.1B). Infusion of IGF-1 in sham-injured mice resulted

in only a 38% increase in cortical Akt activation compared to vehicle-treated sham-

injured mice, but this was not statistically significant. These findings highlight that

Page 104: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

91

systemic infusion of IGF-1 produced greater Akt activation in the injured brain compared

to the sham-injured brain.

To evaluate if systemic infusion of IGF-1 attenuated cortical cell loss after CCI,

we quantified cortical abundance for neurofilament 68 (NF68), a cytoskeletal protein

expressed exclusively in neurons, by western blot as an indicator of neuroprotection.

Brain injury resulted in a 36% decrease in NF68 abundance vehicle-treated mice

compared to vehicle-treated sham-injured mice, but this was not statistically significant

(ANOVA p=0.39; Fig. 3.1C). Brain-injured mice treated with hIGF-1 exhibited only a 12%

decrease in NF68 abundance compared to sham-injured mice treated with hIGF-1, but

the groups were not significantly different from each other. We were unable to detect a

brain-injury induced loss of NF68 following severe CCI. Taken together, these data

demonstrate that systemic infusion of 4 mg/kg hIGF-1 increased systemic circulating

levels of hIGF-1 and promoted activation of the pro-survival protein Akt at 7 d following

severe CCI.

Page 105: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

92

Figure 3.1: Systemic infusion of human insulin-like growth factor-1 (hIGF-1) for a period of 7 d promotes cortical activation of Akt, but does not reduce cortical neuron cytoskeletal protein loss after severe controlled cortical impact (CCI). (A) Levels of human IGF-1 in the systemic circulation were equivalent on days 1 and 7 of the subcutaneous infusion in sham and brain-injured mice, as quantified by human-specific ELISA. Human IGF-1 was not detected in mice treated with vehicle. (B) Systemic infusion of IGF-1 in brain-injured mice significantly enhanced phosphorylation of cortical Akt (*p<0.05 compared to all other groups). (C) Brain injury resulted in only a modest reduction in abundance of NF68, a neuron-specific cytoskeletal protein, in vehicle-treated mice. Treatment with IGF-1 did not significantly alter NF68 abundance compared to treatment with vehicle after sham injury or CCI. (n=3 sham-injured/treatment and n=5 brain-injured/treatment group).

Page 106: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

93

We evaluated if IGF-1 attenuated motor dysfunction in mice after severe CCI

using a dose of 4 mg/kg/d IGF-1, which was previously reported to improve motor

function in moderately brain-injured rats (Saatman et al. 1997). Sham-injured mice

exhibited no significant change in motor function during the 7 d of testing, independent of

treatment group (main ANOVA p<0.05, main time effect p<0.05, interaction p<0.05; Fig.

3.2A). Brain-injured mice, treated with either hIGF-1 or vehicle, showed significant

impairment in motor function at 3 hr, 1 and 3 d post-injury as compared to their

respective sham-injured mice (p<0.05, Fig. 3.2A). Systemic infusion of IGF-1 in brain-

injured mice did not result in improved motor function compared to brain-injured mice

infused with vehicle (p=0.16, Fig. 3.2A). A subsequent follow-up study that evaluated

acute motor function for 2 d following CCI showed that brain injured mice showed

significant improvement over time (main time effect p<0.01), but there was no significant

difference in motor function between mice treated with vehicle or IGF-1 (Fig. 3.2B). This

data suggest that IGF-1 infusion does improve motor function in mice following severe

CCI.

Page 107: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

94

Figure 3.2: Systemic infusion of human insulin-like growth factor-1 (hIGF-1) did not improve recovery of motor function following severe controlled cortical impact (CCI). (A) Brain injury produced significant motor dysfunction, as assessed by the neurological severity score, at 3 hr, 1 and 3 d post-injury, independent of treatment (p<0.001, compared to sham-injury). Treatment with IGF-1 over 7 days did not improve recovery of motor function during the week following severe CCI as compared to treatment with vehicle. (n=3 sham-injured/treatment group and n=5 brain-injured/treatment group). (B) Treatment with IGF-1 over 3 days after CCI, in a second cohort of mice, did not improve motor function compared to vehicle treatment. (n=5/treatment group).

Page 108: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

95

Histological assessment of cell loss by cresyl violet staining revealed that severe

CCI produced cortical cavitation and loss of subcortical white matter in vehicle-treated

mice at 3 d post-injury (Fig. 3.3A). Infusion of IGF-1 did not appear to reduce the extent

of cortical damage or subcortical white matter loss at 3 d following severe CCI.

Hippocampal neurodegeneration was evaluated at 3 d following CCI, as both

neurodegeneration and immature neuron loss can be observed at this time within the

contused hippocampus (Rola et al. 2006; Zhou et al. 2012). Fluorojade-B was utilized to

quantify the numbers of degenerating neurons in CA-1, CA-3 and in the dentate gyrus at

3 d after CCI. Representative images show Fluorojade-B positive cells within the

granular layer of the contused hemisphere (Fig. 3.3B). The abundance of degenerating

neurons did not appear to differ between mice treated with IGF-1 or vehicle after CCI

(Fig. 3.3B). Regional quantification of Fluorojade-B positive cells in the injured

hippocampus indicated that treatment with IGF-1 reduced the total number of

degenerating neurons by 21% compared to treatment with vehicle, but this did not reach

statistical significance (p=0.13; Fig. 3.3C, inset). Regional assessment of the numbers of

Fluorojade-B positive cells in the granular layer and CA3 and CA1 pyramidal layers

revealed that the greatest number of degenerating neurons were located in the granular

layer (Fig. 3.3C). The dentate gyrus granular layer was also the region with the greatest

reduction in the number of Fluorojade-B positive cells following treatment with IGF-1,

with a 25% decrease compared to vehicle treatment; however, this decrease was not

statistically significant (p=0.08; Fig. 3.3C).

Because immature neurons within the SGZ of the dentate gyrus have been

reported to be most vulnerable to TBI, immature neuron density was quantified in the

injured granular layer at 3 d post-injury to evaluate if IGF-1 promoted the survival of this

particular subpopulation of cells. Qualitative and quantitative analysis indicated that

Page 109: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

96

treatment of IGF-1 did not increase the density of immature neurons in the injured

granular layer at 3 d following severe CCI (Fig. 3.4D, E).

Page 110: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

97

Figure 3.3: Systemic infusion of human insulin-like growth factor-1 (hIGF-1) does not reduce tissue damage or regional cell loss at 3 d following severe controlled cortical impact (CCI). (A) Cortical cavitation and subcortical white matter damage were comparable in vehicle-treated and IGF-1-treated mice at 3d after severe CCI. Scale bar represents 500 µm. (B) Representative images of CCI-induced hippocampal neurodegeneration as detected by Fluorojade-B staining in the dentate gyrus granular layer (DG). Scale bar represents 50 µm. (C) Quantification of Fluorojade-B positive cells in the DG granular layer and CA3 and CA1 pyramidal layers. Inset shows the summation of numbers of degenerating neurons within all three regions. (D, E) Representative images of doublecortin (DCx) immunostaining (D) and quantification of immature neuron density in the injured granular layer of the dentate gyrus after injury (E). Scale bar represents 100 µm. (n=5 brain-injured/treatment group). Granular layer (GL) and Hilus (H).

Page 111: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

98

Discussion

In this chapter, we demonstrated that systemic infusion of 4 mg/kg/d hIGF-1 for a

period of 7 d resulted in sustained elevated levels of hIGF-1 in the serum. Levels of

hIGF-1 were not detectable in the brain, but we found that systemic infusion of hIGF-1

promoted the activation of Akt in the contused brain. We were unable to detect an injury-

induced loss in the abundance of cortical NF68 at 7 d following CCI. We also

demonstrated that continuous systemic infusion of hIGF-1 did not significantly reduce

acute hippocampal neurodegeneration or increase hippocampal immature neuron

density at 3 d post-injury.

The short 10-30 minute half-life of IGF-1 (Baxter and Martin 1989; Guler et al.

1989) poses a substantial obstacle for achieving sustained elevations of IGF-1 in

systemic circulation after a systemic bolus injection of IGF-1. We demonstrated that

prolonged continuous systemic infusion of IGF-1 established and sustained elevated

serum levels of IGF-1 for the duration of the infusion. Findings from a phase II clinical

trial showed that IGF-1-treated brain-injured patients who achieved systemic

concentrations of IGF-1 above 350 ng/mL exhibited improved metabolic health and

improved outcome (Hatton et al. 1997). Quantification of hIGF-1 in the current study

revealed that prolonged systemic infusion produced sustained concentrations of at least

150 ng/mL of hIGF-1 during the infusion period. Endogenous physiological levels of IGF-

1 in naïve C57/BL6 mice are reported to be approximately 250 ng/mL (Yuan R 2013),

suggesting that summation of exogenous hIGF-1 and endogenous IGF-1 may have

achieved the therapeutic concentration of 350 ng/mL IGF-1 in systemic circulation for the

duration of the infusion. However, we did not measure endogenous levels of IGF-1 in the

current study. Additional work will need to be completed to understand if exogenous

Page 112: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

99

administration of 4 mg/kg/d of IGF-1 for a period of 7 d modulated the production of

endogenous IGF-1 or resulted in a down regulation of IGF binding protein 3 as was

observed in the phase II clinical trial for TBI (Hatton et al. 1997). Similar to the findings

reported after intravenous infusion of IGF-1 in brain-injured patients (Hatton et al. 1997),

we have shown that prolonged subcutaneous infusion of hIGF-1 resulted in sustained

elevations in IGF-1 for 7 d following infusion.

Large proteins, including growth factors, do not normally gain access to the brain

parenchyma due to exclusion by the intact blood-brain barrier. Blood-brain barrier

breakdown, as observed after severe CCI (Baldwin et al. 1996), should greatly facilitate

growth factor delivery to the brain following severe contusive TBI. Breakdown of the

blood-brain barrier after severe CCI is biphasic as a first phase occurs between 5

minutes and 3 hours and a second phase is observed in the injured hippocampus

between 1 and 2 d following severe CCI (Baldwin et al. 1996). However, growth factor

based treatment strategies may require prolonged delivery to stimulate regenerative or

reparative effects in the injured brain, including include periods of time after the blood-

brain barrier is repaired, thereby limiting the movement of growth factors into the brain.

Moreover, damage to the blood-brain barrier may be less pronounced and shorter in

duration after mild and diffuse brain injuries, limiting the utility of many growth factors for

treatment of these types of injuries. However, IGF-1 is known to be transported across

the intact blood-brain barrier by receptor-mediated endocytosis (Pardridge 1993;

Reinhardt and Bondy 1994). The ability of IGF-1 to cross the intact blood-brain barrier

affords clinical applicability of IGF-1 across the spectrum of injury severity of TBI.

Despite prolonged elevation in the serum levels of hIGF-1, exogenous IGF-1 was

undetectable in the brains of uninjured or injured mice. The undetectable levels of

exogenous IGF-1 in the brain could suggest that IGF-1 did not gain enter the brain;

Page 113: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

100

however, we showed that treatment with IGF-1 increased activation of Akt, a

downstream mediator of IGF-1 signaling. This finding suggests that low levels of

exogenous IGF-1 entered the brain at 7 d post-injury, a time when the blood-brain

barrier has resealed (Baldwin et al. 1996). It is possible that we would have detected

exogenous IGF-1 in the injured brain during periods of compromised blood-brain barrier

integrity, including the first 3 hours and between 1 and 2 following CCI. Alternative

methods of detection and quantification, i.e. radiolabeled IGF-1 and high performance

liquid chromatography/mass spectroscopy, may afford increased sensitivity to quantify

lower levels of hIGF-1 in the brain. Additional work is also needed to define the temporal

prolife of changes in the levels of exogenous IGF-1 in the serum and brain after

prolonged infusion and in the context of brain injury.

Although exogenous IGF-1 was not detectable in the cortex of mice subjected to

sham injury or severe CCI, systemic infusion of IGF-1 enhanced cortical activation of Akt

following severe CCI, suggesting that exogenous IGF-1 entered the brain and promoted

the activation of IGF-1 signaling. Binding of IGF-1 to its cognate receptor promotes the

activation of Akt, a potent pro-survival and anti-apoptotic signal that blunts caspase-3

and 9 and NF-kB cell death-promoting actions (Fukunaga and Kawano 2003; Brywe et

al. 2005). Enhanced Akt activity is associated with improved neuron survival while,

conversely, blunted Akt phosphorylation increases neuron susceptibility to death in the

context of CNS injury (Noshita et al. 2001; Noshita et al. 2002). Experimental TBI results

in an acute increase in downstream mediators of endogenous IGF-1 signaling, including

Akt activation, but these elevations are not sustained after CCI (Zhang et al. 2006;

Madathil et al. 2010). We demonstrate that brain injury did not increase Akt activation in

vehicle-treated mice at 7 d post-injury. Treatment of IGF-1 resulted in a small increase in

Akt activation in sham-injured mice; however, treatment of IGF-1 resulted in a robust

Page 114: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

101

significant increase in Akt phosphorylation in brain-injured mice. Enhanced activation of

Akt following CCI corroborates our hypothesis that systemic infusion of IGF-1 increases

brain levels of IGF-1 and that breakdown of the blood-brain barrier may facilitate

increased exposure of circulating IGF-1 in the contused brain parenchyma. Additional

experiments need to determine if systemic infusion of IGF-1 after CCI results in

widespread activation of Akt throughout the contused hemisphere, including the

hippocampus. The levels of phosphorylated Akt need to be quantified in the injured

hippocampus to determine if lower levels of Akt activation accompany limited reductions

in neurodegeneration in the hippocampus at 3 d following severe CCI. While this study

demonstrates enhanced Akt activation at 7 d post-injury, additional studies also need to

evaluate the acute time course of Akt phosphorylation following the initiation of

systemically infused IGF-1. These studies will provide valuable insight into the kinetic

prolife of IGF-1-mediated Akt activation in the contused cortex acutely following CCI.

Systemic administration of IGF-1 accelerates recovery of motor function and

reduces cognitive impairments in rodent models of mild and moderate TBI (Saatman et

al. 1997; Rubovitch et al. 2010). Previous work from our lab demonstrated that repeated

systemic injections of 1 mg/kg hIGF-1 significantly improved motor function beginning 5

days after the initiation of IGF-1 treatment in rats subjected to moderate fluid percussion

injury (Saatman et al. 1997). Moreover, systemic infusion of 4 mg/kg/d hIGF-1, the same

dose utilized in our current study, was previous shown to significantly improve motor

function at 2 weeks post-injury in moderately brain injured rats (Saatman et al. 1997). In

our current study, systemic infusion of 4 mg/kg/d IGF-1 did not attenuate motor

impairment during the week following severe CCI. The findings reported by Saatman et

al. (1997), suggested that prolonged treatment with IGF-1 may promote delayed

recovery of motor function beginning nearly 1 week post-injury. In our current study,

Page 115: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

102

brain-injured mice exhibited spontaneous recovery of motor function, and did not exhibit

motor impairments after 3 d post-injury. Future studies may need to incorporate

additional behavioral tasks that are more sensitive at detecting motor dysfunction in mice

after brain injury. Alternatively, we may need to incorporate more sensitive assessments

of neurobehavioral function, including cognition, as a recent study by Rubovitch et al.

(2010) demonstrated that IGF-1 significantly improved performance in a Y maze

cognitive task in mice subjected to mild weight drop brain injury. Future studies will need

to incorporate assessments of motor and cognition function to evaluate the efficacy of

IGF-1 to attenuate neurobehavioral dysfunction after severe CCI.

Treatment with IGF-1 reduces apoptosis and enhances cell survival in a variety

of in vitro and in vivo models of CNS injury (Russell et al. 1998; Brywe et al. 2005; Zhu

et al. 2008; Hollis et al. 2009). Administration of IGF-1 following hypoxic-ischemic injury

produces improvements in neurobehavioral function that are concomitant with reduced

apoptosis and survival of neurons (Guan et al. 2001; Brywe et al. 2005; Lin et al. 2009).

The CCI model of experimental TBI is a reproducible and well-characterized model that

produces regional neurodegeneration and loss of cortical and hippocampal neurons

following contusion (Smith et al. 1995; Anderson et al. 2005; Saatman et al. 2006;

Pleasant et al. 2011). In the current study, cresyl violet staining revealed that severe CCI

produced cortical cavitation and subcortical white matter loss in vehicle-treated mice at 3

d post-injury. Continuous systemic infusion of IGF-1 did not appear to reduce cortical

tissue damage or loss of the subcortical white matter. Following CCI, acute cleavage

and sustained cortical breakdown of neurofilament 68, expressed exclusively in neurons,

is observed within 3 hr and sustained for at least 2 weeks post-injury and is indicative of

pathological proteolysis associated with neuronal death (Posmantur et al. 1994).

Quantification of NF68 in the cortex of vehicle-treated mice after CCI revealed a small,

Page 116: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

103

but non-significant reduction in the abundance of intact NF68 compared to vehicle-

treated mice after sham injury, highlighting that we were unable to detect injury-induced

loss using a well-established indicator of cortical cell loss. Moreover, at 3 d post-injury

we observed cortical cavitation consistent with severe CCI, suggesting that our inability

to detect a reduction in NF68 abundance after CCI may be reflective of the small group

sizes. Similar to our findings for the quantification of hippocampal neurodegeneration, a

power analysis revealed that 7 animals are required to achieve statistical significance.

We observed an apparent increase in NF68 abundance in brain-injured mice treated

with IGF-1 compared to brain-injured mice treated with vehicle, but our observations

from the cresyl violet staining suggests that IGF-1 may not significantly attenuate cortical

cell loss. Future studies, using appropriately sized groups, need to demonstrate that

severe CCI produces significant reductions in NF68 abundance in order to subsequently

evaluate the efficacy of systemically infused IGF-1 to attenuate reductions in cortical

NF68 abundance after brain injury.

Of the few studies that have evaluated the efficacy of IGF-1 to improve

neurobehavioral function in the context of brain injury (Saatman et al. 1997; Rubovitch et

al. 2010), only one study has evaluated if IGF-1-mediated neuroprotection contributes to

the improved neurobehavioral performance after TBI (Madathil 2013). Previous work

from our lab has demonstrated that conditional astrocyte-specific overexpression of IGF-

1 in the injured mouse brain reduces neurodegeneration in the dentate gyrus, but not

CA3 at 3 d following severe CCI (Madathil 2013). In the current study, quantification of

regional hippocampal neurodegeneration revealed that systemic infusion of IGF-1

resulted in a small, but non-significant reduction in the number of degenerating neurons

in the granular layer. We did not observe a notable reduction in the number of

degenerating neurons in CA3, similar to findings previously reported from our lab in IGF-

Page 117: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

104

1 overexpressing mice subjected to severe CCI (Madathil 2013). Our study utilized 5

brain-injured mice per treatment group. A power analysis using the data acquired from

the quantification of degenerating neurons in the granular layer at 3 d post-injury

indicated that 9 mice are required in each treatment group to achieve statistical

significance. The results of this power analysis are reasonable considering the IGF-1

overexpressing mouse study reported a significant reduction in the number of

degenerating neurons in the granular layer with 8 brain-injured mice per genotype group

(Madathil 2013). Quantification of hippocampal neurodegeneration by Fluorojade-B at a

single time point provides only an assessment of neurodegeneration at that moment

after brain injury and does not provide insight into a possible modulation of the temporal

progression of neurodegeneration. Future studies may need to evaluate hippocampal

neurodegeneration and neuron survival at multiple time points, including hours and days

after severe CCI to determine if IGF-1 modulates the time course of neurodegeneration

after injury. Our findings warrant future studies, using appropriately sized groups, to

evaluate the efficacy of systemically infused IGF-1 to attenuate hippocampal

neurodegeneration following severe CCI.

Indicators of hippocampal neurodegeneration, including Fluoro-jade-B, are

observed within the granular layer of the dentate gyrus in the days following CCI (Gao et

al. 2008; Hall et al. 2008; Cai et al. 2012; Zhou et al. 2012). Fluorojade-B has been

shown to colocalize with doublecortin, a marker of immature neurons, in the subgranular

zone at 24 hr following moderate CCI. Several studies highlight that immature neurons

are selectively vulnerable to CCI, and consequently, the density of immature neurons is

greatly reduced by 3 d post-injury (Rola et al. 2006; Gao et al. 2008; Yu et al. 2008). We

postulated that reductions in hippocampal neurodegeneration in the granular layer may

be indicative of improved immature neuron survival. In the current study, we

Page 118: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

105

demonstrate that systemic infusion of IGF-1 did not reduce hippocampal immature

neuron loss in the granular layer after CCI. This suggests that IGF-1 may not promote

the acute survival of immature neuron at 3 d post-injury. Additional work is needed to

identify if Fluorojade-B colocalizes predominantly with markers of immature or mature

neurons in the context of severe CCI. Subsequent to the acute loss of immature

neurons, there is a recovery of immature neurons in the weeks following CCI (Rola et al.

2006; Yu et al. 2008). Considering IGF-1 has been shown to promote neurogenesis by

increasing cellular proliferation and enhancing neuronal differentiation (Arsenijevic and

Weiss 1998; Aberg et al. 2000; Brooker et al. 2000; Arsenijevic et al. 2001), future

studies will evaluate the efficacy of IGF-1 to promote post-traumatic neurogenesis in the

hippocampus following severe CCI.

The modest neuroprotective effects of IGF-1 observed in quantification of

hippocampal neurodegeneration, immature neuron density, and cytoskeletal protein

levels in cortical neurons after prolonged systemic infusion may be related to delayed

increases in levels of IGF-1 in the contused brain. We have previously demonstrated

that vascular compromise occurs within 1 hr in the cortex and between 1 and 4 hr in the

hippocampus following severe CCI with a rounded tip (Pleasant et al. 2011). Guan et al.

(2000) demonstrated that delayed central administration of IGF-1 for 2 hr, but not 6 hr

protected against cortical cell loss in rats subjected to hypoxic-ischemic injury. It is

possible that delayed elevations in brain levels of IGF-1 reduce the acute

neuroprotective capacity of IGF-1. As mentioned previously, additional studies to

evaluate the acute prolife of IGF-1 signaling after systemic administration will provide

valuable information about the capacity of IGF-1 to promote neuron survival. The

efficacy of IGF-1-mediated mechanisms of plasticity, including enhancement of post-

traumatic neurogenesis, may not be dependent upon acute accumulations of IGF-1 in

Page 119: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

106

the injured brain. Future studies will investigate if IGF-1 enhances post-traumatic

neurogenesis to accelerate recovery of immature neurons lost after brain-injury.

The dose of 4 mg/kg/d IGF-1 was selected to evaluate the efficacy of IGF-1 to

attenuate neurobehavioral impairment and hippocampal neuron loss following severe

CCI in the mouse. Previously, this dose significantly enhanced recovery of motor

function and significantly improved learning and memory in the Morris water maze task

at 2 weeks post-injury in rats subjected to moderate TBI (Saatman et al. 1997).

Moreover, this dose promoted regeneration and recovery of function after sciatic nerve

crush injury (Contreras et al. 1995). While the largely negative effects observed with

infusion of 4 mg/kg/d hIGF-1 might argue for testing higher doses, systemic infusion of

doses higher than 4 mg/kg/d hIGF-1 should be avoided as continuous infusion of 8

mg/kg/d hIGF-1 produced undesirable side effects including prolonged hypoglycemia,

weight loss and increased mortality in brain-injured rats (Saatman et al. 1997).

Moreover, prolonged hypometabolism occurs within hours of experimental TBI, and

potentiation of metabolic depression in the brain by high levels of circulating IGF-1 could

exacerbate injury pathology (Yoshino et al. 1991; Dietrich et al. 1994). Because systemic

administration of exogenous IGF-1 at 4mg/kg/d appeared to result in very low brain

levels of IGF-1, in Chapter 4 central infusion of hIGF-1 will be utilized to accelerate and

maximize delivery of IGF-1 to the contused brain, in an effort to attenuate motor and

cognitive impairments, and increase immature neuron density after severe CCI.

In this chapter, we have demonstrated that prolonged systemic infusion of IGF-1

enhanced a pro-survival signaling cascade in the injured brain, but did not reduce

cytoskeletal protein loss in cortical neurons and hippocampal neurodegeneration. IGF-1

did not improve recovery of motor function in mice subjected to severe CCI. The work

presented in this chapter demonstrates that additional preclinical work is required to

Page 120: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

107

identify the appropriate administration paradigm and to more comprehensively evaluate

acute and long-term efficacy of IGF-1 to promote recovery after TBI.

Copyright © Shaun William Carlson 2013

Page 121: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

108

Chapter 4: Central Infusion of IGF-1 Improves Neurobehavioral Function and

Increases Immature Neuron Density following Controlled Cortical Impact

Introduction

In Chapter 3, we evaluated the efficacy of systemic administration of recombinant

human IGF-1 (hIGF-1), using a dose previously shown to improve functional outcome in

a rat model of TBI (Saatman et al. 1997), to promote recovery of motor dysfunction and

reduce hippocampal neuron loss after CCI in mice. We found that the dose of 4 mg/kg/d

resulted in undetectable levels of hIGF-1 in the brain, but increased activation of Akt in

the cortex suggesting that brain levels of IGF-1 were elevated following systemic

administration of IGF-1. Systemic administration of hIGF-1 over a period of 3 or 7 d post-

injury did not improve recovery of motor function. We also demonstrated that systemic

administration of hIGF-1 for 3 d was not sufficient to reduce the cortical contusion size,

hippocampal neurodegeneration or loss of hippocampal immature neurons after severe

CCI.

Therefore, we postulated that central infusion of hIGF-1 into the lateral ventricle

would elevate levels of IGF-1 in the brain acutely after injury, above the levels achieved

with systemic infusion, and minimize potential side effects associated with using doses

higher than 4 mg/kg/d for systemic administration of IGF-1. Central administration of

IGF-1 has been utilized as a treatment strategy in models of CNS neurodegeneration.

Central infusion of IGF-1 reduced the loss of Purkinje cells and inferior olive neurons and

reduced motor coordination impairments associated with cerebellar ataxia (Fernandez et

al. 1998; Tolbert and Clark 2003). IGF-1 also promoted improved spatial memory

following kainic acid-induced cognitive impairment in mice (Bluthe et al. 2005).

Moreover, intracerebroventricular infusion of IGF-1 reduced striatal lesion volume

following injections of quinolinate in a rat model of Huntington’s disease (Escartin et al.

Page 122: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

109

2004). Intracerebroventricular injections or prolonged infusion of IGF-1 has also been

shown to reduce infarct areas and promote the survival of neurons (Guan et al. 1993;

Guan et al. 2000; Brywe et al. 2005; Zhu et al. 2008; Selvamani and Sohrabji 2010) and

improve neurological function after hypoxic-ischemic injury (Guan et al. 2001; Schabitz

et al. 2001). Collectively, these studies highlight that administration of IGF-1 directly into

the brain has therapeutic efficacy in promoting neurobehavioral recovery in models of

neuronal degeneration and CNS acute injury. However, the therapeutic efficacy of

centrally infused IGF-1 to attenuate neurobehavioral impairment has not been evaluated

in the context of TBI.

Intracerebroventricular administration has been utilized to evaluate the

effectiveness of multiple neuroprotective compounds (Fink et al. 1999; Wang et al. 2012)

and anti-inflammatory agents (Toulmond and Rothwell 1995; Jones et al. 2005;

Marklund et al. 2005; Byrnes et al. 2012) after experimental TBI. Intracerebroventricular

administration of neurotrophic or neurotrophic-like factors, including S100B, vascular

endothelial growth factor (VEGF), epidermal growth factor (EGF), and basic fibroblast

growth factor (bFGF), has also been utilized to evaluate improvements in

neurobehavioral function or alterations in post-traumatic neurogenesis following

experimental TBI (Kleindienst et al. 2005; Sun et al. 2009; Sun et al. 2010; Thau-

Zuchman et al. 2010; Thau-Zuchman et al. 2012). Central infusion of therapeutic agents

represents a clinically relevant route of administration for severe brain-injured patients

requiring intracranial pressure shunts or decompressive craniotomies. Ideally therapeutic

agents demonstrating efficacy after central administration also need to be evaluated

using systemic administration in order to translate their application to TBIs of reduced

severity that do not necessitate an opening of the skull or dura. In this regard, IGF-1 is a

Page 123: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

110

unique therapeutic agent as systemic administration is a viable route of administration

after TBI, as described in Chapter 2.

Hippocampal immature neurons are selectively vulnerable to brain injury with

marked progressive immature neuron loss within hours and days following CCI (Rola et

al. 2006; Gao et al. 2008). Brain injury also initiates an endogenous self-renewal process

that promotes the slow recovery of immature neurons via increased proliferation of

progenitor cells in the subgranular zone of the dentate gyrus granular layer (Dash et al.

2001; Chirumamilla et al. 2002; Rola et al. 2006). The majority of newly proliferated cells

will become glial cells after CCI, but a minority of the proliferated cells will adopt a

neuronal phenotype after injury (Rola et al. 2006; Gao and Chen 2013). Immature

neurons generated after injury have been shown to integrate anatomically into the

granular layer and project to CA-3 (Emery et al. 2005; Sun et al. 2007) and express

synaptic markers at 10 weeks post injury in rats subjected to moderate fluid percussion

injury (Sun et al. 2007). However, only a portion of the newly generated immature

neurons will survive to develop into mature granular neurons and integrate into the

hippocampus (Sun et al. 2005; Sun et al. 2007). Central infusion of bFGF or VEGF has

been shown to promote post-traumatic proliferation and increase the generation of

newborn neurons without altering neuronal differentiation (Sun et al. 2009; Thau-

Zuchman et al. 2010; Thau-Zuchman et al. 2012). While the efficacy of IGF-1 to promote

neurogenesis after TBI has not been evaluated, several studies demonstrate that

administration of IGF-1 increases neurogenesis by promoting cellular proliferation and

neuronal differentiation (Arsenijevic and Weiss 1998; Aberg et al. 2000; Brooker et al.

2000; Arsenijevic et al. 2001; Trejo et al. 2001).

Page 124: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

111

In this chapter we evaluate the efficacy of intracerebroventricular infusion of IGF-

1 to improve motor and cognitive function during the first week following severe CCI in

mice. In Chapter 3, we showed that IGF-1 did not appear to promote the survival of

immature neurons at 3 d following CCI. In the current study, we assess immature neuron

density at 7 d post-injury, a time point at which the rate of new neurons being born is

increased in the injured hippocampus, to evaluate the efficacy of IGF-1 to increase

immature neuron density after CCI.

Page 125: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

112

Materials and Methods

Animals

All experimental protocols were approved by the University of Kentucky

Institutional Animal Care and Use Committee in accordance with the established

guidelines from the Guide for the Care and Use of Laboratory Animals from the National

Institutes of Health. Animals were housed 5 mice per cage in the University of Kentucky

Medical Center animal vivarium with a 14:10 hr light:dark photoperiod and provided food

and water ad libitum, but food consumption was not monitored in the mice utilized for

this study. Adult male C57/BL6 mice aged 8-10 weeks, weighing 25-30 g, were utilized

for all experiments.

Controlled Cortical Impact and Central Infusion of Human IGF-1

Mice were anesthetized using 3% isoflurane for preparation of the scalp and

placement in the stereotaxic frame (David Kopf Instruments, CA) and anesthesia was

maintained via nose cone for the duration of the surgery. An incision was made on the

midline of the scalp to expose the skull. A 5 mm craniotomy was completed midway

between bregma and lambda, lateral to the sagittal suture on the left hemisphere of the

skull with the dura left intact. Sham-injured animals received anesthesia and only a

craniotomy. CCI injury was performed with a computer-controlled pneumatically driven

piston to rapidly and transiently (500 msec) impact the exposed dura of the brain (TBI-

0310 Impactor, Precision Systems and Instrumentation, VA). The CCI injury was

produced by a 1 mm impact depth at a velocity of 3.5 m/s using a 3 mm diameter

rounded impactor tip. Animals were randomly assigned to either sham injury or brain

injury and treatment with either vehicle or recombinant human IGF-1 (hIGF-1) in each of

the studies detailed below.

Page 126: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

113

Central Infusion of IGF-1

While the mouse was anesthetized, a cannula was placed in the lateral ventricle

at 15 minutes following sham injury or CCI through a burr hole (AP -0.5 mm bregma, ML

-1.0 mm, DV -3.0 mm; Paxinos and Franklin 2001) in the hemisphere contralateral to the

site of injury. The cannula (brain infusion kit 3, Alzet, CA) was attached to an osmotic

minipump (model 1007D, 1002, or 1004, Alzet, CA) for continuous

intracerebroventricular (i.c.v.) infusion for a period of 7 d. Minipumps were primed in a

37°C water bath for at least 6 hr prior to surgical implantation, with the exception of the

delayed treatment study, in which an approximately 6 hr delay in treatment initiation was

achieved via implantation of a non-primed minipump. The base of the cannula was

secured to the skull using an instant adhesive (Loctite 454, Alzet, CA) and a set screw

(MX-0090-01F-C; AmazonSupply.com, WA) was placed adjacent to the base of the

cannula with a small amount of dental acrylic. The minipump was positioned in the

subcutaneous space over the scapula of the back, lateral to the spine. The mice

exhibited no observable impairment in mobility as a result of the location of the

minipump. The minipumps were filled with 0.026, 0.085, 0.26, or 0.85 mg/mL

recombinant hIGF-1 to deliver 0.3, 1, 3, or 10 µg/d; National Hormone and Peptide

Program, CA) or vehicle (pH 7.4, 24 mM acetic acid diluted in USP grade PBS). The

doses of IGF-1 utilized in this study were extrapolated from previous studies that

demonstrated efficacy of IGF-1 to attenuate somatosensory impairment and reduce cell

loss after cerebral hypoxic-ischemia (Guan et al. 1993; Zhu and Auer 1994; Guan et al.

2001; Selvamani and Sohrabji 2010), reduce cell loss in a model of Huntington’s disease

(Escartin et al. 2004), and promote hippocampal neurogenesis (Trejo et al. 2001).

Following cannula implantation, a small circular disk of dental acrylic was adhered to the

skull over the craniotomy site, the scalp was sutured, and the mouse was placed on a

Page 127: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

114

heating pad to maintain normal body temperature. All mice received a subcutaneous

injection of 1 mL of sterilized saline to prevent dehydration from the surgical procedure

and implantation of the minipump. Once ambulatory, the mice were returned to their

home cage.

Study 1 (Effects of 10 µg/d hIGF-1 Infusion on Behavior and Hippocampal Neuron

Survival)

Sham-injured (n=10 per treatment group) and brain-injured mice (n=19 per

treatment group) received central infusion of either 10 µg/d hIGF-1 or vehicle for 7 d

post-injury at a flow rate of 0.5 µL/hr (Alzet minipump model 1007D). Blood from non-

fasted mice was collected onto test strips via tail prick by a 25G needle and glucose

levels measured using a handheld glucometer (Onetouch Ultra, CA) prior to anesthesia,

and at 90 minutes, 1 d and 7 d post-injury. Motor and cognitive deficits were assessed

during the week following injury by a modified neurological severity score (NSS) and

novel object recognition (NOR) task, respectively, as described below. At 7 d post-injury,

mice received an overdose of Fatal-plus (65mg/kg sodium pentobarbital, Vortech MI),

and blood was collected by transcardial puncture for measurement of serum hIGF-1.

Mice were randomized into one of two groups to be utilized for quantification of hIGF-1

and Akt activation by ELISA and western blot, respectively (n=5 sham-injured per

treatment group and n=9 brain-injured per treatment group), or for cresyl violet

histological and immature neuron immunohistochemical assessments (n=5 sham-injured

per treatment group and n=10 brain-injured per treatment group), as described below.

Accurate placement of the cannula in the contralateral ventricle was determined in the

histology cohort by visual verification of the needle track during coronal sectioning of the

brain. As a result, two animals (n=1 Sham vehicle, n=1 Sham IGF-1) were excluded from

Page 128: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

115

the histological cohorts for inaccurate placement of the cannula. Cannula placement

could not be verified in brains utilized for western blot and ELISA. If an animal was

excluded for inaccurate cannula placement, it was excluded from all outcome measures

including blood glucose measurements, neurobehavioral assessments and cellular

quantification.

Study 2 (Effects of 10 µg/d hIGF-1 on Regional Cerebral Edema)

Mice were subjected to CCI and received no cannulation (n=6), or i.c.v infusion of

either 10 µg/d hIGF-1 (n=6) or vehicle (n=6) for 7 d via osmotic minipump (model

1007D). At 7 d post-injury, mice received an overdose of Fatal-plus and were euthanized

by decapitation. The ipsilateral and contralateral cortical and hippocampal regions were

rapidly dissected for quantification of water content using the wet weight/dry weight

method (Stewart-Wallace 1939, Sun 2003 J Neurosurg, Whalen 2000 J Leuko Biol).

Each brain sample was weighed on an aluminum foil packet to acquire the wet weight

immediately following dissection. Samples were dried in a desiccating oven at 70°C for

24 hr, and re-weighed to acquire the dry weight. The percent water content was

calculated as (wet weight – dry weight) / wet weight x 100.

Study 3 (Delayed Onset of hIGF-1 Infusion)

Mice were subjected to 1 mm CCI and infused with either 10 µg/d IGF-1 (n=5) or

vehicle (n=3) beginning 6 hr post-injury via implantation of a nonprimed minipump at 15

minutes post-injury. Histological assessment and quantification of immature neuron

density were performed at 7 d post-injury. No animals were excluded from this study.

Page 129: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

116

Study 4 (Dose Response for hIGF-1)

Mice subjected to CCI brain injury were randomized to receive infusion of 0.3, 1,

3, or 10 µg/d of hIGF-1 (n=5 per dose) or vehicle (n=4) beginning 15 minutes post-injury

for a period of 7 d (minipump model 1007D). Histological assessment and quantification

of immature neuron density was performed at 7 d post-injury. No animals were excluded

from this study.

Study 5 (Effect of Changing Infusate Flow Rate in Sham-injured Mice)

Mice were subjected to sham injury (n=5 per flow rate) and infused with vehicle,

beginning 15 minutes post-injury, for 7 d. Differential volumes of vehicle solution were

introduced into the brain by utilization of osmotic minipumps with different flow rates

(model 1002: 0.25 µL/hr; model 1004: 0.11 µL/hr). Histological assessment was

performed at 7 d post-injury and data were compared to that from sham-injured mice

(n=4) infused with vehicle at 0.5 µL/hr from Study 1. No animals were excluded from this

study.

Study 6 (Efficacy of 3 µg/d hIGF-1 Infused at a Reduced Rate)

Sham-injured (n=4 per treatment) and brain-injured mice (n=8 per treatment)

were infused with 3 µg/d hIGF-1 or vehicle beginning 15 minutes post-injury for 7 d at a

flow rate of 0.11 µL/hr (minipump model 1004:). At 4 d after injury, a peak time of post-

traumatic cellular proliferation, mice received three injections of 50 mg/kg (i.p.) 5-bromo-

2-deoxyuridine (BrdU, Fisher Scientific, NJ) given at 4 hr intervals to label proliferating

cells. Histological assessment and quantification of immature neuron and proliferating

cell densities were performed at 7 d post-injury. Two animals (n=1 vehicle CCI, n=1

hIGF-1 CCI) were excluded for inaccurate cannula placement.

Page 130: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

117

Motor Function Assessment Using a Modified Neurological Severity Score (NSS)

A previously described modification of the NSS task (Pleasant, Carlson 2011)

was utilized to assess motor dysfunction at 3 hr, 1, 3, 5, and 7 d post-injury. At 24 hr

prior to injury, mice were acclimated to each of four Plexiglas beams of varying widths

(3, 2, 1, 0.5 cm) and a 0.5 cm diameter wooden rod. The beams and rod were 60 cm in

length and elevated 47 cm above a table top. Mice were given 30 seconds to traverse

the beams and rod during both acclimation and testing. Each beam was assigned a

maximum of three points. Three points were given for successful crossing of the beam

with normal position and usage of the forelimb and hindlimb. Two points were given for

successful crossing of the beam despite either a forelimb or hindlimb hanging below the

beam, and one point was given for crossing the beam despite inverting below the beam

one or more times. The mouse was righted and allowed to continue across the beam if it

became inverted on the beam. A score of zero was given if the mouse did not cross in

the allotted time or fell off the beam. For the rod, two points were given for successful

crossing of the beam in the allotted time. A score of one was given for crossing despite

inverting more than three times. A score of zero was given if the mouse did not traverse

in the allotted time or fell off the rod.

Cognitive Performance Evaluation Using a Novel Object Recognition (NOR)

Paradigm

At 24 hr prior to injury, mice were acclimated to a clear plastic cage (32x20x14

cm) with an open top for a period of one hour. Mice were introduced to two identical

Lego®Duplo® figures placed in opposite corners of the cage for 5 minutes. At 7 d post-

injury, mice were re-introduced to the testing cage and allowed to explore for one hour.

The two original objects were re-introduced in the cage and the time spent exploring

Page 131: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

118

each object was recorded for a period of 5 minutes. After a delay of 4 hr, the mouse was

returned to the testing cage where one of the original objects had been replaced with a

novel object. The time spent exploring each object was recorded for 5 minutes. A

recognition index was calculated as the time spent exploring the novel object as a

percentage of total object exploration time.

Tissue Collection

Following transcardial collection, blood was transferred to a 1.65 mL eppendorf

tube and allowed to coagulate for 30 minutes. To extract serum, coagulated blood was

centrifuged at 4,500xg for 10 minutes at room temperature. Serum was transferred to a

new tube and stored at -80°C.

For ELISA and western blotting, mice received an overdose of Fatal-plus. The

brains were rapidly removed from the skull and placed onto an ice cold dissection plate.

To concentrate collection of tissue on the injury site, 3 mm of the anterior brain was

blocked in the coronal plane and the remaining ipsilateral and contralateral cortical and

hippocampal regions were dissected and separately placed in eppendorf tubes and

stored at -80°C.

For histology and immunohistochemistry, mice received an overdose of Fatal-

plus at 7 d post-injury. Mice were then transcardially perfused with heparinized saline to

clear blood from the vasculature followed by 10% neutral buffered formalin. The mice

were decapitated and the head placed in 10% neutral buffered formalin for 24 hr. The

brain was dissected from the head and placed in 10% neural buffered formalin for an

additional 24 hr. Brains were then placed in 30% sucrose for 24 hr for cryoprotection

prior to freezing in -30°C isopentanes cooled on dry ice. Brains were cut into 40 µm-thick

Page 132: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

119

coronal sections on a freezing sliding microtome (Microm, Dolbey-Jamison, PA) at -20°C

(Physitemp, NJ).

Preparation of Tissue for ELISA and Western Blotting

Cortical and hippocampal samples were homogenized by wand sonicator in cold

lysis buffer (1% triton X-100, 20 mM Tris-HCl, 150 mM NaCl, 5 mM EGTA, 10 mM

EDTA, 10% glycerol, and a cocktail of proteinase inhibitors (Roche, IN)) and centrifuged

for 30 minutes at 4°C at a speed of 10,000xg. The supernatants were collected and

utilized for analysis. Protein concentrations were determined using a BCA assay kit

(Pierce Biotechnology, IL).

Quantification of IGF-1 by ELISA

Human IGF-1 was quantified using the highly-specific Quantikine® human IGF-1

ELISA kit (DG100, R&D Systems Inc., MN) according to the manufacturer’s instructions.

Serum and brain samples were pretreated for dissociation of IGF-1 from the binding

proteins. Human IGF-1 standards (0.094-6.0 ng/mL) and pretreated samples were

pipetted in duplicate into a 96 well plate coated with monoclonal antibody specific to

human IGF-1. Following a 2 hr incubation, the wells were rinsed with the supplied wash

buffer to remove unbound antigens. A polyclonal antibody conjugated with HRP enzyme

was incubated in the wells for 1 hr. The wells were rinsed and incubated with a substrate

solution for 30 minutes. Development was halted by the additional of the 1N hydrochloric

acid stopping solution. Absorbance of each well was measured using a microplate

reader (Tecan, NC) at 450 nm and 540 nm wavelengths. The absorbance at 540 nm

was subtracted from the 450 nm absorbance to account for the background of each well.

Page 133: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

120

Western Blot

Western blot analyses were performed as previously described (Madathil et al.

2010). Electrophoresis was completed in triplicate using 30 µg of protein supernatant

from the ipsilateral hippocampal samples, on a 3-8% Tris-HCl gel at 150V. After transfer

onto nitrocellulose membranes, the membranes were blocked for 1 hr in 5% dry milk

dissolved in 0.1% Tween 20 in TBS and incubated overnight with a primary antibody for

p-Akt ser473 (rabbit monoclonal, 1:2000 concentration, Cell Signaling Technology, MA).

The membranes was rinsed the following day and incubated for 1 hr with a secondary

antibody for anti-rabbit IgG (1:5000 IRDye800CW, Rockland, PA). The membrane was

rinsed and then imaged and the optical density was quantified using the Li-Cor Odyssey

Infrared Imaging System (Li-Cor Biosciences, NE). After pAkt development, the

membranes were reprobed for actin using an anti-β-actin primary antibody (mouse

monoclonal 1:5000, Calbiochem Inc, CA) and an anti-mouse IgM conjugated to an

infrared dye (1:10000, Rockland). For quantification, OD of each pAkt band was divided

by its respective actin OD. A mean OD was calculated for each animal from the OD

values from the triplicate samples and normalized to the mean normalized OD for

vehicle-treated sham-injured mice to permit comparison across gels.

Histological (Cresyl Violet) Staining

For each brain, every tenth brain section spaced 400µm apart was mounted and

air-dried on gelatin-coated slides. The slides were hydrated in graded ethanol solutions,

stained with 0.5% cresyl violet (Acros Organics, NJ), rinsed in water, dehydrated through

graded ethanol solutions, cleared in xylenes (Fisher Scientific, NJ), and mounted with

Permount (Fisher Scientific, NJ).

Page 134: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

121

Immunohistochemistry

Coronal brain sections (n=6-7 per animal), spaced 200 µm apart and centered

with respect to the injury epicenter, were immunolabeled for doublecortin (DCx), a

microtubule protein expressed in immature neurons, or BrdU, a thymidine analog that

incorporates into DNA during DNA synthesis, using standard free floating

immunohistochemistry protocols, as previously described (Cai et al. 2012). Only sections

to be double labeled for BrdU (Study 6) were initially rinsed in TBS (pH 7.4), treated with

2N HCl for 1 hr at room temperature to expose the BrdU epitope, and subsequently

treated with 100 mM borate buffer (pH 8.0, Fisher Scientific, NJ) for 10 minutes.

Sections were then rinsed and incubated in TBS overnight at 4°C. Sections from all

studies were rinsed in TBS and blocked with 5% normal horse serum with 0.1% Triton-

X-100 in TBS for 30 minutes. Either DCx (rabbit polyclonal, 1:500, Abcam, MA) or a

cocktail of DCx and BrdU (rat polyclonal, 1:1000, Abcam, MA) primary antibodies, where

applicable, diluted in blocking solution, were placed on the tissues for overnight

incubation at 4°C. The sections were rinsed with TBS and incubated with secondary

antibodies (anti-rabbit IgG conjugated with Alexa Fluor® 488 or anti-rat IgG conjugated

with Cyanine Cy3, Jackson ImmunoResearch, PA) diluted in blocking solution for 1 hr

and rinsed with TBS. Sections were incubated with Hoechst (1:10,000, Invitrogen, CA)

for 1.5 minutes to label all nuclei and rinsed with TBS. Labeled sections were mounted

on gelatin-coated slides, coverslipped with Fluoromount (Southern Biotech, AL), and

stored at 4°C.

Image Acquisition

Representative images of cresyl violet stained sections and images of DCx and

BrdU immunoreactivity were acquired using an AX80 Olympus microscope (PA).

Page 135: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

122

Cellular Quantification

In comparison to Chapter 3, quantification of DCx and BrdU positive cells was

performed in a larger number of sections to expand the anatomical region of interest to

the rostrocaudal extent of the ipsilateral dentate gyrus granular layer, and also the

contralateral granular layer in Studies 3 and 4, of each section. Quantification of

additional sections provided a more comprehensive assessment of cell populations

throughout the dentate gyrus granular layer and not isolated to the injury core, as

previously described for quantification of degenerating neurons within the dentate gyrus

(Madathil 2013). All positively labeled cells were manually counted through all focal

planes within the dentate gyrus granular layer at 40x magnification on an epifluorescent

microscope (Olympus BX51) equipped with a FITC filter (41001, Chroma Technology,

VT) or TRITC filter (31002, Chroma Technology). Colocalization of DCx and BrdU was

determined using a wide band filter (51004v2, Chroma Technology) that permits

simultaneous viewing of the emissions of both fluorophores of the secondary antibodies.

Cells exhibiting colocalization of DCx and BrdU immunostaining were manually counted

through all focal planes within the granular layer at 40x magnification on the same

microscope. Colocalization was confirmed by scanning through the z-axis to ensure

colabeling within each cell. Cellular counts were normalized to the volume of the dentate

gyrus granular layer of each section to control for differences in dentate gyrus size

across sections. The area of the Hoechst-labeled dentate gyrus granular layer was

imaged (Olympus, AX80) and was measured using ImagePro (MediaCybernetics, MD).

The thickness of each section was measured, in microns, using an epifluorescent scope

(Olympus BX51) equipped with a stereology stage. Granular layer area and thickness

measurements were used to calculate granular layer volume. Volumetric density

measurements (1000 cells/mm3) represent the entire granular layer, inclusive of the

Page 136: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

123

subgranular, inner, and outer granular layers. A mean cell density was calculated for all

quantified sections for each animal. To reduce animal usage, sham-injured mice were

not included in Studies 3 and 4. Therefore, immature neuron densities in the ipsilateral

hemisphere were normalized to those in the non-impacted hemisphere (contralateral)

hemisphere and were expressed as a percentage of contralateral immature neuron

density. Due to the fact that brain injury results in immature neuron loss predominantly in

the impacted hemisphere (ipsilateral), the contralateral hippocampus can provide an

approximation of immature neuron density in the uninjured brain. Quantification of all

sections was performed by an investigator blinded to the treatment and injury conditions

of each animal.

Statistical Analysis

All data is presented as mean + standard error of the mean (SEM). Serum levels

of hIGF-1 (Fig. 4.1D) and cellular densities (Fig. 4.5C) were compared by Student’s t-

test. Brain levels of hIGF-1 (Fig. 4.1A) and the NSS motor function scores (Fig. 4.2B)

were compared using a repeated measures one-way analysis of variance (ANOVA).

NOR recognition indices (Fig. 4.2A), regional water content (Fig. 4.4G) and cellular

densities (Fig. 4.7B) were compared by one-way ANOVA. Baseline blood glucose levels

(Table 4.1), Akt activation (Fig. 4.1C), (Fig. 4.2A), and cellular densities (Fig. 4.3B, 4.9I,

J, K) were compared using a two-way ANOVA. Blood glucose levels (Table 4.1) were

compared using a repeated measures two-way ANOVA. In all cases, the Newman-Keuls

post-hoc t-tests were performed when appropriate. Statistical tests were completed

using Statistica (Statsoft Inc, OK). A p value less than 0.05 was considered statistically

significant.

Page 137: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

124

Results

By using recombinant human insulin-like growth factor-1 (hIGF-1), exogenously

infused IGF-1 could be distinguished from endogenous mouse IGF-1 using an ELISA kit

highly-specific for hIGF-1. Human IGF-1 was not detected in serum or brain samples of

vehicle-treated mice. A 7 d intracerebroventricular infusion of 10 µg/d hIGF-1 into the

contralateral ventricle in Study 1 resulted in measureable hIGF-1 in the hemisphere

ipsilateral to the impact, with equivalent levels in the cortex and hippocampus,

suggesting widespread delivery. The contralateral hippocampus (same hemisphere as

the cannula) had significantly higher concentrations of IGF-1 compared to the ipsilateral

cortex or hippocampus (#p<0.05, Fig. 4.1A).

Brain injury in vehicle-treated mice resulted in a small, but non-significant

decrease in hippocampal levels of phosphorylated Akt compared to sham-injury in

vehicle-treated mice. Consistent with appropriate signaling after binding to its cognate

receptor, infusion of IGF-1 in sham-injured and brain-injured mice resulted in significantly

enhanced phosphorylation of Akt, a potent pro-survival and growth permissive signal, in

the ipsilateral hippocampus, compared to infusion of vehicle (main treatment effect,

*p<0.05, Fig. 4.1B, C). These data demonstrated that central infusion of IGF-1 results in

a measureable and widespread increase of IGF-1 in the brain and promotes the

activation of a pro-survival pathway after severe CCI.

Page 138: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

125

Figure 4.1: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) over 7 d elevates brain levels of hIGF-1 and enhances Akt activation in the hippocampus after severe controlled cortical impact (CCI). (A) hIGF-1 was detected in the ipsilateral (ipsi) cortex (CTX) and ipsilateral and contralateral (contra) hippocampi (Hipp) at 7 d after sham injury (Sham) or severe (CCI) using a human-specific ELISA (#p<0.05, contra Hipp compared to ipsi CTX or Hipp). (B) Representative western blot images of phosphorylated Akt (pAkt) and actin at 7 d post-injury for vehicle (Veh) or hIGF-1 treated mice. (C) Hippocampal levels of pAkt as determined by semiquantitative Western blot. Infusion of hIGF-1 resulted in a significant increase in Akt activation within the hippocampi of sham-injured and CCI-injured mice at 7 d post-injury (*p<0.005, IGF-1 treated compared to vehicle treated). The optical density (OD) for each pAkt band was normalized to its respective actin band OD; the mean of the triplicate samples for each mouse was normalized to the mean OD value of the vehicle-treated sham group. The results are presented as mean + SEM. (n=5 sham-injured and n= 9 CCI-injured per treatment group).

Page 139: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

126

A previous study showed that IGF-1 is cleared from cerebrospinal fluid and

enters systemic circulation (Nagaraja et al. 2005). We used the same highly-specific

hIGF-1 kit to quantify the levels of hIGF-1 in systemic circulation. Human IGF-1 was not

detected in the serum of vehicle-treated mice. IGF-1-infused, brain-injured mice

exhibited significantly higher concentrations of hIGF-1 in the systemic circulation (79.4 +

4.7 ng/mL), as compared to sham-injured mice infused with IGF-1 (*p<0.05; 58.2 + 10.8

ng/mL). To determine whether elevated serum IGF-1 resulted in systemic hypoglycemia,

systemic blood glucose was measured at several time points following surgical

procedures. Blood glucose levels were not significant among treatment groups (Table

4.1). Anesthesia and surgical procedures induced a significant elevation in blood

glucose levels at 90 minutes post-injury, independent of injury and treatment condition

(*p<0.05 compared to all other time points), but blood glucose levels returned to

baseline at 1 d and 7 d post-injury (Table 4.1).

Page 140: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

127

Table 4.1: Blood glucose levels were elevated in both vehicle (Veh) and insulin-like growth factor-1 (IGF-1) treated mice at an acute time point following sham injury (Sham) or controlled cortical impact (CCI). Blood glucose was measured prior to injury, and at several time points after surgical procedures in non-fasted mice. Blood glucose levels were significantly higher at 90 minutes after surgical procedures compared to all time points, independent of treatment and injury status (*p<0.05). Data are presented as mean + SEM. (n= 9 sham-injured per treatment and n=19 brain-injured per treatment).

Blood glucose measurements following central infusion (mg/dL)

Baseline 90 minutes 1 d 7 d

Veh Sham 176+ 7 233+ 11 * 183+ 8 200+ 11

IGF-1 Sham 165+ 6 210+ 10 * 171+ 11 163+ 7

Veh CCI 187+ 9 229+ 9 * 203+ 17 192+ 16

IGF-1 CCI 179+ 9 231+ 12 * 170+ 9 189+ 11

Page 141: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

128

To evaluate if central infusion of hIGF-1 improves neurobehavioral performance

after severe CCI, we evaluated cognitive performance using the novel object recognition

task at 7 d after injury in mice from Study 1. Vehicle-infused mice exhibited significant

memory impairment after CCI as compared to sham-injured vehicle-treated mice, as the

recognition index scores indicated impaired recognition of the familiar object (*p<0.01,

Fig. 4.2A). Infusion of IGF-1 reduced the brain injury associated decline in cognitive

performance, improving posttraumatic memory function of IGF-1-treated brain-injured

mice to a level equivalent to IGF-1-treated sham-injured mice (Fig. 4.2A). Recognition

indices of sham-injured mice treated with IGF-1 were not significantly different from

sham-injured vehicle-treated mice (Fig. 4.2A).

A modified neurological severity score (NSS) was utilized to evaluate motor

coordination during the week following severe CCI. Motor performance was equivalent

for sham-injured mice treated with either vehicle or IGF-1 (Fig. 4.2B). Sham-injured

exhibited a small, but significant impairment in motor function at 1 d post-injury, but

exhibited no impairment for the remainder of the testing, independent of treatment status

(p<0.05, Fig. 4.2B). Brain-injured mice exhibited significantly impaired motor function

during the week post-injury, compared to sham-injured mice, independent of treatment

condition (p<0.01). Vehicle-treated brain-injured mice exhibited a significant reduction in

motor function at 1, 2, 3, and 5 d after injury, compared to sham-injured mice (*p<0.001,

Fig. 4.2B). Conversely, IGF-1-treated brain-injured mice exhibited a significant

impairment in motor function at only 1, 2, and 3 d post-injury (p<0.001 compared to IGF-

1-treated sham-injured mice, Fig. 4.2B). Mice infused with IGF-1 after brain injury

showed significantly improved motor function at 1, 2, 3, and 5 d after CCI, compared to

vehicle-infused brain-injured mice (#p<0.01, Fig. 4.2B). These data demonstrated that

Page 142: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

129

central infusion of hIGF-1 reduces cognitive impairment and attenuates motor

dysfunction associated with severe CCI.

Page 143: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

130

Figure 4.2: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) over 7 d attenuates cognitive and motor impairment after severe controlled cortical impact (CCI). (A) Brain injury resulted in a significant reduction in the novel object recognition index of vehicle-treated mice at 7 d post-injury (*p<0.05, compared to sham-injured (Sham) vehicle-treated (Veh) mice. Infusion of IGF-1 reduced the brain injury-induced decline in cognitive performance. (B) Vehicle-infused brain-injured mice exhibited significant impairment in motor function at 1, 2, 3, and 5 d post-injury as assessed by a modified neurological severity score (NSS) (*p<0.001, compared to vehicle-treated sham-injured mice). IGF-1-infused brain-injured mice exhibited a significant motor impairment at 1, 2, and 3 d post-injury compared to IGF-1 infused sham-injured mice (p<0.001). Treatment with IGF-1 improved motor function at 1, 2, 3, and 5 d post-injury compared to vehicle-infused brain-injured mice (#p<0.01). Data are presented as mean + SEM. (n=9 sham-injured per treatment and n=19 CCI-injured per treatment).

Page 144: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

131

In order to evaluate if a 7 d infusion of 10 μg/d hIGF-1 enhances hippocampal

immature neuron density after injury, we quantified DCx+ cells in the dentate gyrus

granular layer from mice in Study 1. CCI induced a significant reduction in DCx+ cell

density in vehicle-treated mice at 7 d post-injury compared to sham-injured mice

(*p<0.05, Fig. 4.3A, B). Brain-injured mice infused with IGF-1 had significantly higher

DCx+ cell densities compared to brain-injured mice infused with vehicle (#p<0.05, Fig.

4.3B). The density of immature neurons in brain-injured mice infused with IGF-1 was not

significantly different from sham-injured mice receiving IGF-1 (Fig. 4.3B). IGF-1

treatment did not affect immature neuron density in sham-injured mice (Fig. 4.3A, B).

Page 145: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

132

Figure 4.3: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) protects immature neurons in the injured hippocampus after severe controlled cortical impact (CCI). (A) Representative images of doublecortin (DCx, green) immunoreactivity in the ipsilateral hippocampus of vehicle (Veh) and IGF-1 infused sham-injured (Sham) and CCI-injured mice at 7 d post-injury. Scale bar represents 100 µm. Granular layer (GL) and Hilus (H). (B) DCx+ cell density is significantly decreased following brain injury in vehicle-treated mice (*p<0.05, compared to sham-injured vehicle-treated mice). Infusion of IGF-1 significantly attenuated the reduction in DCx+ cell density at 7 d after CCI injury (#p<0.05, compared to vehicle-treated brain-injured mice). (n=4 sham-injured per treatment, n=10 vehicle-treated brain-injured and n=9 IGF-1-treated brain-injured). Immature neuron counts obtained from the ipsilateral granular layer were normalized to the volume of the ipsilateral granular layer to calculate cellular density (1000/mm3). Data are expressed as mean + SEM.

Page 146: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

133

Histological evaluation by cresyl violet staining indicated that severe CCI

produced cortical cell loss and cavitation, subcortical white matter loss and thinning of

the hippocampal granular layer at 7 d post-injury (Fig. 4.4C). Qualitative assessment of

cresyl violet staining revealed that the majority of IGF-1-infused mice subjected to CCI

exhibited similar gross pathology (7 of 10, Fig. 4.4D, E), although the ipsilateral

hippocampus was distorted in some mice (Fig 4.4E). However, a subset of mice showed

profound swelling of subcortical regions and hippocampal distortion after CCI (3 of 10,

Fig. 4.4F) raising concerns that central infusion of IGF-1 may promote or exacerbate

posttraumatic cerebral edema Assessment of sham-injured vehicle-treated mice

revealed a small degree of brain swelling 7 d post-injury without any indications of

damage to the exposed dura at the time of craniotomy (Fig. 4.4A). Central infusion of

IGF-1 in sham-injured mice resulted in mild cortical tissue swelling at the craniotomy site

(Fig. 4.4B) that appeared more pronounced than for vehicle infusion.

To evaluate if cerebral edema contributed to the brain swelling observed in a

subset of IGF-1-infused, brain-injured mice, in Study 2 we generated a cohort of brain-

injured mice for evaluation of regional water content in the absence of infusion, with

infusion of vehicle, or with infusion of 10 µg/d hIGF-1. Brain injury-induced edema

develops within 24 after CCI brain injury and resolves towards non-injured levels by 7 d

post-injury (Kochanek et al. 1995). Analysis of the cortices and contralateral hippocampi

of all mice revealed no statistically significant changes in water content across

experimental groups (Fig. 4.4G). The water content percentages observed in these

regions (77-80%) were comparable to those reported after sham injury (Trabold et al.

2008; Kenne et al. 2012; Terpolilli et al. 2013). However, a modest increase in water

content in the ipsilateral hippocampus of brain–injured mice infused with 10 µg/d hIGF-1

appeared to be driven by a subset of mice (2 of 6) that exhibited a much higher than

Page 147: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

134

normal water content (81-82%; Fig 4.4G). This data suggested that edema in the injured

hippocampus of IGF-1-infused mice may contribute to the brain swelling and tissue

distortion observed in a subset of mice from Study 1. Differences in age or body weight

did not correlate with the incidence of brain swelling. We sought to modulate parameters

of the infusion paradigm that may attenuate brain swelling observed in IGF-1-infused

mice after severe CCI.

Page 148: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

135

Figure 4.4: Central infusion of 10 µg/d human insulin-like growth factor-1 (IGF-1) promotes brain swelling and deformation in a subset of mice after severe controlled cortical impact (CCI). (A-F) Representative images of cresyl violet staining of the cortex (CTX) and hippocampus (Hipp) ipsilateral (ipsi) to impact at 7 d post-injury from vehicle (Veh) and IGF-1-treated mice subjected to sham injury (Sham) or brain injury. Sham-injured mice infused with either (A) vehicle (Veh) or (B) IGF-1 exhibited brain swelling at the craniotomy site. (C) Brain-injured mice treated with vehicle exhibited cortical cavitation and hippocampal cell loss consistent with severe CCI. The majority of IGF-1-infused brain-injured mice (7 of 10) exhibited characteristic histopathology with either (D) minimal or (E) modest brain swelling. (F) However, a subset of IGF-1-infused brain-injured mice (3 of 10) showed severe distortion of the hippocampus at 7 d post-injury. (n=4 sham-injured per treatment and n=10 brain-injured per treatment). Scale bar represents 500 µm. (G) Water content was quantified in brain-injured mice receiving CCI without cannulation or infusion or CCI with central infusion of either vehicle or 10 µg/d IGF-1 (n=6 per group). Infusion of IGF-1 did not result in significant regional cerebral edema; however a small subset of mice (2 of 6) had markedly increased water content in the ipsilateral hippocampus. Contralateral (contra). Data are presented as mean + SEM.

Page 149: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

136

In an effort to minimize central infusion of infusate during the development of

edema within the initial hours following CCI (Kochanek et al. 1995), we sought to delay

infusion of IGF-1 until 6 hr following severe CCI. After induction of brain injury, a

nonprimed minipump was implanted to deliver 10 µg/d hIGF-1, achieving a delay in

infusion onset of approximately 6 hr (Study 3). Brain-injured mice infused with vehicle

beginning 6 hr post-injury exhibited cortical cavitation and dentate gyrus granular layer

thinning consistent with severe CCI (Fig. 4.5A). Delayed infusion of 10 µg/d of IGF-1

resulted in no observable brain swelling in the majority of mice (4 of 5), while a subset of

mice exhibited hippocampal distortion qualitatively similar to that observed with the acute

onset of infusion (1 of 5; Fig. 4.5A). Although delaying IGF-1 administration did not

appear to ameliorate infusion-related brain swelling, we evaluated whether efficacy for

protection of immature neurons was retained. Given that the initiation of treatment in

clinical trials typically occurs between 3 and 8 hr following the TBI (Maas et al. 2007),

this delayed IGF-1 infusion paradigm is highly clinically relevant. DCx immunoreactivity

appeared to be reduced in vehicle-treated mice, but was conserved in IGF-1-treated

mice (Fig. 4.5B). Quantification of DCx+ cell density demonstrated that delayed treatment

with IGF-1 significantly enhanced immature neuron density at 7 d post-injury, as

compared to vehicle treatment (#p<0.05, Fig. 4.5C). Thus, IGF-1-mediated

enhancement of immature neuron density was retained in this clinically relevant

treatment paradigm.

Page 150: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

137

Figure 4.5: Delayed infusion of 10 µg/d human insulin-like growth factor-1 (hIGF-1) resulted in brain swelling and increased hippocampal immature neuron density at 7 d after severe controlled cortical impact (CCI). (A) Images of cresyl violet staining of vehicle (Veh) and hIGF-1-treated brain-injured mice. Brain-injured vehicle-treated mice showed cortical and hippocampal cell loss consistent with severe CCI. The majority of IGF-1-treated brain-injured mice (4 of 5) exhibited minimal to modest brain swelling, but one mouse (shown) exhibited pronounced brain swelling at 7 d post-injury. Scale bar represents 500 µm. (B) Compared to vehicle-treated CCI-injured mice, doublecortin (DCx, green) immunoreactivity appeared to be enhanced in IGF-1-treated brain-injured mice. Scale bar represents 100 µm. (C) IGF-1-treated brain-injured mice showed significantly increased immature neuron density at 7 d post-injury, compared to vehicle-treated brain-injured mice (*p<0.05). Data are presented as mean + SEM. (n=3 vehicle-treated brain-injured and n=5 IGF-1-treated brain-injured mice).

Page 151: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

138

Because infusion-related brain distortion appeared worse with hIGF-1 than with

vehicle (see Fig. 4.4C, F), we next sought to evaluate if the incidence and severity of

brain swelling could be reduced by decreasing the dose of IGF-1. In Study 4, 10 µg/d

hIGF-1 was compared to doses of 3, 1, 0.3, and 0 (vehicle) µg/d. As assessed in cresyl

violet stained tissue sections, infusion of 10 µg/d hIGF-1 resulted in marked cortical and

hippocampal distortion in the ipsilateral hemisphere in a subset of mice (2 of 5; Fig

4.6A), consistent with observations in Study 1 (see Fig. 4.4F). However, the severity

and incidence of infusion-related brain swelling at 7 d following CCI appeared to be

dose-dependent. Infusion of 3 µg/d hIGF-1 reduced the incidence of exacerbated brain

swelling (1 of 5), and decreased the severity of brain swelling (Fig. 4.6B), as compared

to infusion of 10 µg/d hIGF-1. Animals infused with 1 or 0.3 µg/d hIGF-1 exhibited mild or

no hippocampal swelling (Fig. 4.6C, D) similar to mice infused with vehicle (Fig. 4.6E).

These observations suggest that the dose of IGF-1 may be a contributing component of

i.c.v infusion-related brain swelling after severe CCI.

Page 152: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

139

Figure 4.6: Reductions in the concentration of centrally infused human insulin-like growth factor-1 (hIGF-1) appeared to reduce the severity and incidence of brain swelling following severe controlled cortical impact (CCI). (A-E) For doses that resulted in brain swelling, images of cresyl violet staining were selected from the subgroup of hIGF-1-treated mice in each dose that exhibited notable brain swelling at 7 d post-injury. While the majority of mice infused with 10 µg/d hIGF-1 (3 of 5) exhibited characteristic histopathology after severe CCI with minimal hippocampal swelling, (A) a subset of mice (2 of 5) showed brain swelling and gross distortion of the hippocampus at 7 d after injury. (B) Infusion of 3 µg/d hIGF-1 resulted in reduced incidence (1 of 5) and severity of hippocampal swelling, when compared to infusion of 10 µg/d hIGF-1. Infusion of (C) 1 µg/d or (D) 0.3 µg/d hIGF-1 largely mitigated the infusion-induced hippocampal swelling after CCI, with pathology appearing comparable to vehicle-infused mice (E). Scale bar represents 500 µm. (n=4 vehicle-treated and n=5 per dose IGF-1-treated brain-injured mice).

Page 153: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

140

To evaluate if the effects of IGF-1 on immature neuron density were dose

dependent, DCx+ cell staining and immature neuron density was assessed in brain-

injured mice for each dose of IGF-1 examined in Study 4. DCx immunoreactivity was

greatly diminished in brain-injured vehicle-treated mice. Brain injury-induced loss of DCx

immunoreactivity was attenuated by IGF-1, and the effect appeared more robust with

increasing doses of infused IGF-1 (Fig. 4.7A). After CCI, DCx+ cell density was

decreased indicating a reduction in immature neurons in vehicle-treated mice. Analysis

of immature neuron densities as a function of dose by one-way ANOVA did not reach

statistical significance. However, IGF-1-treated mice exhibited an apparent dose-

dependent enhancement of immature neuron density at 7 d post-injury (Fig. 4.7B).

Although this relationship was not statistically significant, these data suggest a dose-

dependent increase in hippocampal immature neuron density with central infusion of

IGF-1.

Page 154: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

141

Figure 4.7: Central infusion of human insulin-like growth factor-1 (IGF-1) enhances hippocampal immature neuron density in an apparent dose-dependent manner after controlled cortical impact. (A) Brain injury in vehicle-treated (Veh) mice results in a robust loss of DCx immunoreactivity in the dentate gyrus granular layer. In IGF-1-treated mice, DCx immunoreactivity appeared to increase in a dose-dependent manner. Granular layer (GL) and hilus (H). Scale bar represents 50 µm. (B) Increases in immature neuron density were proportional to increasing concentrations of IGF-1 infusate. Data are presented as mean + SEM. (n=4 vehicle-treated brain injured and n=5 per dose IGF-1-treated brain-injured mice).

GL

H

Page 155: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

142

Because infusion of vehicle alone at a 0.5 µL/hr flow rate over 7 d resulted in a

small amount of brain swelling in sham-injured mice (Fig 4.4A, reproduced in Fig. 4.8A),

we next evaluated whether reducing the infusate volume would eliminate brain swelling

after prolonged central infusion. In Study 5, osmotic minipumps with different flow rates

were utilized to reduce the volume of vehicle infusate introduced into the contralateral

ventricle of sham-injured mice over a 7d infusion period. The brains of the sham-injured

mice exhibited no indications of dural damage or herniation at the completion of the

craniotomy. Compared to the flow rate of 0.5 µL/hr used previously (Alzet model 1007D;

Fig. 4.8A), reducing the rate to 0.25 µL/hr (model 1002; Fig. 4.8B) appeared to decrease

brain swelling at the craniotomy site. Swelling was negligible at a flow rate of 0.11 µL/hr

(model 1004; Fig 4.8C). Therefore, the volume of infusate introduced into the

contralateral ventricle appears to influence the development of brain swelling after

craniotomy.

Page 156: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

143

Figure 4.8: Volume of infusate introduced into the influences brain swelling after craniotomy. Representative images of cresyl violet staining of sham-injured vehicle-treated mice at 7 d post-surgery. (A) The 1007D osmotic minipump (0.5 µL/hr flow rate) produced mild brain swelling at 7 d post-injury (reproduced from Fig 4.4A). Utilization of osmotic minipumps with lower flow rates of (B) 0.25 µL/hr (model 1002) and (C) 0.11 µL/hr (1004) resulted in reduced cortical swelling at the craniotomy site. Scale bar represents 500 µm. (n=4 1007D, n=5 1002, and n=5 1004).

Page 157: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

144

In light of these data implicating IGF-1 concentration and infusate volume as

contributing factors to brain swelling after CCI, we sought to complete a follow up

experiment to determine if utilizing a lower dose of hIGF-1 (3 µg/d) in combination with a

slower infusion rate (0.11 µL/hr, model 1004) would reduce brain swelling while retaining

the beneficial effects of IGF-1 infusion on immature neuron density after injury. The dose

of 3 µg/d hIGF-1 was selected as this dose had reduced brain swelling and enhanced

immature neuron density in the previous studies. In Study 6, mice were subjected to

sham injury or CCI and centrally infused with vehicle or 3 µg/d hIGF-1 beginning 15

minutes after injury. At 7 d post-injury, cresyl violet stained tissue sections from vehicle-

infused sham-injured mice revealed reduced brain swelling (Fig. 4.9A) compared to

vehicle-treated sham-injured mice from Study 1 (Fig. 4.4A). Infusion of 3 µg/d hIGF-1 in

sham-injured mice appeared to reduce the severity of brain swelling compared to IGF-1-

treated sham-injured mice from Study 1; however, evidence of cortical swelling at the

craniotomy site was observed in a subset of sham-injured mice (2 of 4, Fig. 4.9B).

Similar to the previously described observations, severe CCI resulted in a characteristic

cortical cavitation and granular layer thinning with minimal hippocampal distortion

associated with infusion of vehicle (Fig. 4.9C). Infusion of 3 µg/d hIGF-1 at 0.11 µL/hr

after brain injury resulted in minimal brain swelling in the majority of mice (6 of 8) with a

subset of mice (2 of 8) exhibiting brain swelling at 7 d post-injury (Fig. 4.9D). However,

the severity of brain swelling observed following infusion of 3 µg/d hIGF-1 was notably

reduced compared to central infusion of 10 µg/d IGF-1 at 0.5 µL/hr (see Fig. 4.4F).

DCx immunoreactivity was assessed in these mice to evaluate if IGF-1-

stimulated enhancement of immature neuron density after severe CCI was retained

using this altered infusion paradigm. Sham-injured mice treated with either vehicle or

IGF-1 exhibited a similar extent of DCx immunoreactivity (Fig. 4.9E, F, I). Brain-injured

Page 158: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

145

mice exhibited a marked reduction in immature neuron density at 7 d post-injury,

independent of genotype (main effect of injury, p<0.005, Fig. 4.9I). Central infusion of

IGF-1 resulted in significantly elevated immature neuron density at 7 d post-injury (main

effect of treatment, p<0.05, Fig. 4.9I), but this effect was not dependent on injury

(interaction effect, p=0.1). Brain-injured mice treated with vehicle exhibited a 51%

reduction in immature neuron density at 7 d post-injury compared to sham-injured mice

treated with vehicle (Fig. 4.9I). In contrast, brain-injured mice treated with IGF-1

exhibited only a 17% reduction in immature neuron density compared to sham-injured

mice treated with IGF-1 (Fig. 4.9I). Infusion of 3 µg/d hIGF-1 at a flow rate of 0.11 µL/hr

resulted in increased immature neuron density at 7 d following severe CCI.

We next sought to evaluate if an increase in post-traumatic granular cell

proliferation contributed to the observed increase in immature neuron density in IGF-1-

treated mice. BrdU immunoreactivity was utilized to identify the number of newly

proliferated cells in the dentate gyrus granular layer after CCI. Brain injury resulted in a

significant increase in the density of BrdU+ cell density compared to sham injury,

independent of treatment (main effect of injury, p<0.05, Fig. 4.9J). Brain-injured mice

treated with vehicle exhibited a 53% increase in newly proliferated cells compared to

sham-injured mice treated with vehicle. In contrast, brain-injured IGF-1-treated mice

showed a 90% increase in the density of proliferated cells compared to sham-injured

IGF-1-treated mice at 7 d post-injury. Sham-injured mice treated with IGF-1 showed a

small increase in BrdU+ cell density compared to sham-injured mice treated with vehicle

(Fig. 4.9J). These data suggest that enhanced post-traumatic proliferation with treatment

with IGF-1 may contribute to the increased immature neuron density observed at 7 d

post-injury.

Page 159: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

146

To evaluate if IGF-1 enhanced the generation of newborn neurons after CCI,

DCx immunoreactivity was colocalized with BrdU to identify immature neurons born after

brain injury. Quantification revealed that treatment with IGF-1 resulted in a significant

increase in the density of newborn neurons at 7 d post-injury, independent of injury

(main effect of treatment, p<0.01, Fig. 4.9K). Brain-injured mice treated with vehicle

exhibited a 55% reduction in newborn neuron density compared to sham-injured mice

treated with vehicle. In contrast, brain-injured mice treated with IGF-1 showed no

reduction in the number of newborn neurons generated after injury compared to IGF-1

infused sham-injured mice (Fig. 4.9K) and an approximate 3-fold increase relative to

vehicle-treated, brain-injured mice. Sham-injured mice treated with IGF-1 showed a 42%

increase in newborn neuron density compared to sham-injured mice treated with vehicle

(Fig. 4.9K). These data demonstrated that although the decreased dose of 3 µg/d of

IGF-1 and the slower flow rate of the osmotic minipump did not fully eliminate infusion-

related brain swelling, the severity of brain swelling was reduced with these infusion

modifications. Moreover, these data highlight that increased immature neuron density

with treatment of 3 µg/d hIGF-1 may be due in part to the increased generation of

newborn neurons following severe CCI.

Page 160: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

147

Page 161: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

148

Figure 4.9 (cont.): Central infusion of 3 µg/d human insulin-like growth factor-1 (IGF-1) via a lower infusion rate (0.11 µL/hr) resulted in modest infusion-related brain swelling and enhanced post-traumatic neurogenesis at 7 d following severe controlled cortical impact (CCI). (A-D) Representative images of cresyl violet staining. Compared to (A) infusion of vehicle (Veh), (B) infusion of hIGF-1 in sham-injured mice (Sham) resulted in minor cortical swelling with no appearance of hippocampal distortion. (C) Brain-injured mice treated with vehicle exhibited characteristic CCI-induced cortical cavitation with minimal indication of brain swelling. (D) Infusion of 3 µg/d hIGF-1 at 0.11 µL/hr produced mild or no hippocampal swelling in the majority of mice (6 of 8, shown). Scale bar represents 500 µm. (E-H) Representative images of doublecortin (DCx, green) and 5-bromo-2-deoxyuridine (BrdU, red) immunohistochemistry. Scale bar represents 100 µm. (E-F) DCx and BrdU immunoreactivity and colocalization of DCx and BrdU appeared equivalent between treatment groups of sham-injured mice. (G) DCx immunoreactivity and the incidence of colocalization appeared to be reduced in vehicle-treated mice after controlled cortical impact compared to vehicle-treated sham-injured mice. (H) IGF-1-treated brain-injured mice exhibited increased DCx immunoreactivity and an increased incidence of DCx and BrdU colocalization compared to vehicle-treated brain-injured mice. (I) Brain-injured mice resulted in significantly reduced immature neuron density (main effect of injury, p<0.005). Treatment with IGF-1 significantly increased immature neuron density (main effect of treatment, p<0.05). (J) Brain injury produced a significant increase in BrdU+ cell density (main effect of injury, p<0.05). (K) Quantification of DCx and BrdU colocalization revealed a significant increase in the density of newborn neurons of mice infused with IGF-1 at 7 d compared to mice infused with vehicle (main effect of treatment, p<0.05). Data are presented as mean + SEM. (n=4 sham-injured per treatment and n=8 brain-injured per treatment group).

Page 162: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

149

Discussion

Systemic administration of IGF-1 has been shown to improve motor function

(Saatman et al. 1997) and/or cognitive performance (Saatman et al. 1997; Rubovitch et

al. 2010) in rodents subjected to experimental TBI; however, in Chapter 3 we showed

that systemic infusion of IGF-1 did not attenuate motor impairment in mice subjected to

severe CCI. We also demonstrated that we were unable to detect hIGF-1 in the brain

following systemic infusion of 4 mg/kg/d of hIGF-1. Considering that increasing doses of

systemically administered IGF-1 can produce hypoglycemia and increase mortality in

rats after experimental TBI (Saatman et al. 1997), we chose to pursue central infusion to

achieve greater levels of IGF-1 in the brain and reduce potential complications

associated with systemic administration of doses above 4 mg/kg/d. In the current study,

we demonstrate for the first time that central administration of IGF-1 improves motor

function, reduces cognitive impairment and enhances immature neuron density in mice

after experimental TBI.

Intracerebroventricular injection of radiolabeled IGF-1 into adult rats results in

increased radioactivity and IGF-1 immunoreactivity throughout the brain parenchyma,

including the cortex, white matter tracts and the hippocampus within 30 minutes post-

injection (Guan et al. 1996; Thorne et al. 2004). These studies highlight that

intracerebroventricular administration results in widespread distribution of IGF-1 in the

brain, and suggest that central administration is applicable for broad delivery of IGF-1

throughout the brain following experimental TBI. However, prolonged central infusion

may be necessary to achieve sustained elevations of IGF-1 in the injured brain.

Nagajara et al. (2005) demonstrated that following a single intracerebroventricular

injection, radiolabeled IGF-1 is rapidly cleared into systemic circulation with limited

penetrance of radiolabeled IGF-1 in the brain parenchyma by 3 hr after injection. By

Page 163: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

150

utilizing a continuous central infusion of hIGF-1 into the lateral ventricle contralateral to

the contusion injury we established that levels of hIGF-1 were elevated in the injured

cortex and bilaterally in the hippocampus, two of the most vulnerable brain regions

following severe CCI, even at 7 days after injury. While we observed a significantly

greater level of hIGF-1 in the contralateral hippocampus, this is likely due to its proximity

to the site of infusion.

Although it is well established that IGF-1 in the systemic circulation can pass into

the brain through receptor-mediated endocytosis (Reinhardt and Bondy 1994), a

previous study demonstrated the converse, that centrally infused IGF-1 can be cleared

from cerebrospinal fluid (CSF) and enter systemic circulation, likely via the lymphatic

system and cranial and/or spinal nerve roots (Nagaraja et al. 2005). We demonstrate

that hIGF-1 infused into the lateral ventricle was detected at concentrations of

approximately 60-80 ng/mL in the systemic circulation at the cessation of central

infusion. Surprisingly, the levels of hIGF-1 in the systemic circulation were comparable to

levels quantified in the ipsilateral cortex or hippocampus, suggesting that a moderate

proportion of centrally infused hIGF-1 was cleared from the brain and entered the

systemic circulation. Therefore it is possible that elevated levels of hIGF-1 in the brain

reflect, in part, secondary entry of hIGF-1 into the brain from systemic circulation via

receptor-mediated endocytosis. However, it is likely that this contribution is minimal, in

light of data in Chapter 3 demonstrating that serum levels of hIGF-1 nearly 2-fold higher

following systemic infusion of hIGF-1 resulted in undetectable levels in the brain at 7 d

following severe CCI. Our findings from Chapter 3 suggest that the elevation of brain

levels of hIGF-1 in the current study is likely reflective of central infusion and not

increased brain uptake of hIGF-1 from systemic circulation.

Page 164: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

151

Central infusion was selected to minimize systemic hypoglycemia in IGF-1-

infused brain-injured animals at 1 d following brain injury. Even though central infusion

resulted in elevated serum hIGF-1, central infusion of IGF-did not produce systemic

hypoglycemia at 24 hr after brain injury, as was previously reported following systemic

infusion of IGF-1 in rats after moderate TBI (Saatman et al. 1997). This finding highlights

that central infusion of IGF-1 eliminated a potential side effect of systemic administration

of IGF-1 after experimental TBI.

The IGF-1 receptor is expressed by neurons and astrocytes, and in the

vasculature, choroid plexus and the hippocampus throughout adulthood (Bondy and Lee

1993; Bondy and Cheng 2004). Binding of IGF-1 to its cognate receptor elicits activation

of Akt and the subsequent inhibition of pro-apoptotic factors including caspase-9, Bad

and NF-kB (Peruzzi et al. 1999; Fukunaga and Kawano 2003). Enhanced Akt activation

is associated with increased neuronal survival after CNS injury; conversely, low levels of

Akt activation have been linked to increased cellular susceptibility to damage and death

(Noshita et al. 2001; Noshita et al. 2002). Brain injury results in an acute and transitory

rise in endogenous levels of IGF-1 in the brain (Madathil et al. 2010) and a transient

increase in Akt activation for hours after experimental TBI (Zhang et al. 2006; Madathil et

al. 2010; Rubovitch et al. 2010). Subsequently, activity of Akt is reduced by 24 hr and

this depression persists for at least 72 hr in the hippocampus following CCI (Zhang et al.

2006), suggesting that continuous infusion of IGF-1 may be required to promote

sustained elevations in Akt activity after brain injury. IGF-1-treated mice in this study

exhibited a significant increase in Akt phosphorylation in the hippocampus after 7 d of

IGF-1 infusion. While increased activation of Akt suggested enhanced protective

signaling during the IGF-1 infusion, future studies will need to elucidate the acute time

course of Akt activation in response to IGF-1 treatment and assess other mediators of

Page 165: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

152

IGF-1 signaling including MAPK and mTOR. Additional work is also needed to

investigate the potency of IGF-1 to exert long-lasting protective and reparative effects

after the cessation of treatment. Together, these studies will afford a better

understanding of the time course of IGF-1 signaling and provide valuable insight into

designing effective therapeutic treatment paradigms for IGF-1 in the context of TBI.

Previous work from our lab demonstrated that astrocyte-specific overexpression

of IGF-1 in the contused mouse brain significantly attenuated motor dysfunction during

the week following severe CCI (Madathil 2013). In the current study, we corroborate

these findings using a more clinically relevant strategy for elevating brain IGF-1. Central

infusion of IGF-1 significantly attenuated motor impairments at 1, 2, 3, and 5 d post-

injury in mice subjected to severe CCI. An earlier study in which systemic injections of 1

mg/kg IGF-1 twice a day resulted in significantly improved motor function, beginning only

after 5 d of treatment in rats subjected to moderate TBI (Saatman et al. 1997),

suggested that IGF-1 promotes delayed plasticity or regeneration that may underlie the

observed recovery of motor function. Our findings of acute recovery of motor function

after central infusion of IGF-1 differ somewhat from these previously published findings.

It is possible that elevated levels of IGF-1 in the brain, as observed after targeted

astrocyte-specific overexpression or central infusion of IGF-1 is needed to promote

acute recovery of motor function, as this was not observed following systemic

administration of IGF-1. The observation that central infusion of IGF-1 attenuates motor

dysfunction during the week after severe CCI supports our hypothesis that exogenous

administration of IGF-1 promotes neurobehavioral improvement after experimental TBI.

We demonstrate that central infusion of IGF-1 reduces injury-induced cognitive

dysfunction associated with severe CCI at 7 d post-injury. Our findings corroborate

Page 166: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

153

previous work from our lab demonstrating that targeted astrocyte-specific

overexpression of IGF-1 attenuated cognitive deficits in the novel object recognition task

at 7 d following severe CCI (Madathil 2013). Exogenous administration of IGF-1 has also

been shown to promote recovery of cognitive function after experimental TBI. Following

mild weight drop injury, systemic injections of 4 µg/kg IGF-1 at 24 and 48 hr post-injury

improved performance in a Y-maze assessment of cognitive function at 7 d post-injury

(Rubovitch et al. 2010). Continuous systemic infusion of 4 mg/kg/d IGF-1 improved

spatial learning and memory at 2 weeks post-injury in rats subjected to moderate TBI

(Saatman et al. 1997). Collectively these previous reports and our current findings

highlight that administration of IGF-1 promotes recovery of cognitive function in rats and

mice in multiple models of TBI and across the spectrum of severity of brain injury. The

observed improvements in cognitive function may be related to neuroprotection or post-

traumatic neurogenesis.

Reductions in hippocampal immature neuron density result in impaired spatial

learning and memory (Clelland et al. 2009; Deng et al. 2009; Jessberger et al. 2009).

Immature neurons are particularly vulnerable to CCI, as demonstrated by a marked

reduction in the numbers of immature neurons within days after injury (Rola et al. 2006;

Gao et al. 2008; Yu et al. 2008). Similarly, we observed a marked reduction in immature

neuron density in the injured dentate gyrus granular layer at 7 d after severe CCI, a time

point shown to be near the peak of immature neuron loss (Rola et al. 2006). In this

report, mice centrally infused with either 10 g/d or 3 g/d IGF-1 after CCI exhibited

enhanced hippocampal immature neuron density at 7 d post-injury, as compared to

vehicle-infused brain-injured mice. This response appeared dose-dependent over the

range of 0.3 to 10 μg/d hIGF-1. In a rat model of cerebral hypoxic-ischemic injury, doses

of 50 g and 5g of IGF-1, but not 0.5µg of IGF-1, injected into the lateral ventricle

Page 167: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

154

promoted enhanced neuroprotection in the cortex and dentate gyrus of the hippocampus

(Guan et al. 1993), corroborating dose-dependent effects of IGF-1 in the brain after

acute CNS injury. In the clinical setting, the average administration window of

therapeutic agents in clinical trials is 3-8 hr after TBI for a variety of reasons including

transportation, stabilization and enrollment of the patient into the trial (Maas et al. 2007).

Using a clinically relevant delayed administration paradigm, we demonstrate that

infusion of IGF-1 initiated 6 hr following severe CCI was effective at increasing immature

neuron density.

Increases in immature neuron density at 7 d after TBI may reflect survival of

existing immature neurons or enhanced generation of new immature neurons. IGF-1 is a

potent neurotrophic factor that promotes cell survival of cultured neurons and reduces

cell loss following acute CNS injury (Guan et al. 1993; Russell et al. 1998; Guan et al.

2001; Brywe et al. 2005; Bendall et al. 2007), but can also increase cellular proliferation

and promote neuronal differentiation (Arsenijevic and Weiss 1998; Aberg et al. 2000;

Brooker et al. 2000; Arsenijevic et al. 2001; Yan et al. 2006). In our current study, we did

not evaluate survival of immature neurons acutely after injury; this would need to be

investigated in future studies. However, our findings in Chapter 3 (and in Chapter 5)

suggest that IGF-1 may not protect against acute immature neuron loss. Instead IGF-1

may enhance the generation of new neurons in the injured brain. Following the robust

loss of immature neurons after CCI, a slow recovery of immature neuron density to pre-

injury levels occurs in the days to weeks following CCI (Rola et al. 2006; Yu et al. 2008).

IGF-1 enhances the generation of newborn neurons in the hippocampus by enhancing

neuronal differentiation of newly proliferated cells (Aberg et al. 2000; Trejo et al. 2001).

Increased immature neuron density at 7 d following severe CCI with treatment of IGF-1

Page 168: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

155

may be indicative of post-traumatic neurogenesis, potentially driven by enhanced cellular

proliferation and neuronal differentiation.

Brain injury results in increased cellular proliferation in the hippocampus following

CCI (Dash et al. 2001; Rola et al. 2006; Zhou et al. 2012) and increased proliferation of

neural progenitor cells can contribute to the generation of newborn neurons following

brain injury (Rola et al. 2006; Sun et al. 2009). We confirmed that severe CCI produced

a significant increase in BrdU-positive cells in the dentate gyrus granular layer at 7 d

after CCI. Injury-induced cellular proliferation can be enhanced by

intracerebroventricular infusion of growth factors, including bFGF, VEGF and EGF

following experimental TBI (Sun et al. 2009; Sun et al. 2010; Thau-Zuchman et al. 2010;

Thau-Zuchman et al. 2012). Consistent with the cellular proliferative effects of IGF-1

(Aberg et al. 2000; Arsenijevic et al. 2001), we observed a small, but not statistically

significant increase in BrdU positive cells following central infusion of 3 g/d of hIGF-1,

suggesting that IGF-1-mediated increases in cellular proliferation may contribute to the

increased immature neurons density observed after severe CCI.

Colocalization of DCx and BrdU revealed that brain injury in vehicle-infused mice

resulted in a marked reduction in the number of neurons born between 4 and 5 days

after severe CCI. Central infusion of IGF-1 significantly increased the number of

newborn neurons, suggesting that IGF-1 enhanced the generation of newborn neurons

after brain injury. This is likely the result of enhanced neuronal differentiation of newly

proliferated cells after brain injury. A small proportion of newly proliferated cells in the

granular layer of the dentate gyrus will adopt a neuronal lineage after CCI (Rola et al.

2006; Gao et al. 2008), but the majority of the newly proliferated cells will express

markers characteristic of glial cells including astrocytes, microglia, and oligodendrocytes

Page 169: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

156

(Rola et al. 2006; Urrea et al. 2007). Our data suggests that treatment with IGF-1 may

shift the ratio of cellular differentiation of newly proliferated cells from a predominantly

glial lineage towards a neuronal lineage after CCI. Peripheral infusion of IGF-1 in

hypophysectomized rats results in increased generation of newborn neurons and not

astrocytes (Aberg et al. 2003), consistent with our observations of an IGF-1-mediated

enhancement of neuronal differentiation. IGF-1 is also a potent promoter of

oligodendrocyte differentiation (Aberg et al. 2007), and future studies will need to

elucidate if prolonged central infusion of IGF-1 also enhances the generation of new

oligodendrocytes in the dentate gyrus granular layer after CCI.

Immature neurons generated after fluid percussion injury project processes to

CA-3 and incorporate into the dentate gyrus during the weeks following injury (Emery et

al. 2005; Sun et al. 2007). Of the newborn neurons generated after brain injury, only a

small proportion will integrate into the dentate gyrus granular layer (Sun et al. 2007). In

the current study we showed that IGF-1 overexpression increases the number of

immature neurons at 7 d post-injury. Future studies will need to determine if treatment

with IGF-1 promotes long-term survival, maturation, and functional incorporation of the

newly generated immature neurons into the hippocampus after severe CCI. Newborn

neurons are speculated to contribute to improved spatial learning at 8 weeks post-injury

in rats subjected to moderate fluid percussion injury (Sun et al. 2007). Experimental

blockage of the immature neuron recovery phase after CCI exacerbates injury-induced

spatial learning deficits at 1 month after CCI (Blaiss et al. 2011), suggesting that

immature neurons are important for cognitive function after brain injury. The findings

reported by Blaiss et al. (2011) also support the hypothesis that therapeutic strategies

that attenuate immature neuron loss or enhance the generation of immature neurons

may improve cognitive outcome following TBI.

Page 170: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

157

Severe CCI produces cortical cavitation, loss of subcortical white matter, and

regional hippocampal neurodegeneration by one week post-injury (Hall et al. 2005;

Saatman et al. 2006; Hall et al. 2008; Pleasant et al. 2011). Treatment with hIGF-1

following severe CCI did not appear to reduce overt cortical or hippocampal cell loss

after brain injury. While previous work from our lab showed that targeted astrocyte-

specific overexpression of IGF-1 reduced hippocampal neurodegeneration at 3 d post-

injury (Madathil 2013), no previous study evaluating the efficacy of exogenous

administration of IGF-1 examined neuroprotection. In Chapter 3, we showed that

systemic administration of IGF-1 did not show overt neuroprotection, but brain levels of

IGF may have been below a therapeutic threshold. Quantification of the number of

neurons in the dentate gyrus granular layer, CA3 and CA1 may be needed to determine

if centrally infused IGF-1 protects against hippocampal neuron loss. Future studies may

also need to determine if the brain swelling observed in the subset of mice led to tissue

damage that may have counteracted potential neuroprotective effects in the injured

brain.

Infusion of hIGF-1 resulted in notable brain swelling in a subset of mice.

Parameters of body weight, age, and blood glucose did not predict the occurrence of

infusion related swelling. The observance of increased brain swelling may be related to

fluid effects such as raised intracranial pressure in the brain, exacerbation of brain injury-

induced edema, or to effects of IGF-1 itself.

Increases in fluid loading in the brain during central infusion may contribute to

brain swelling at 7 d following continuous infusion. Prolonged intracerebroventricular

infusion in mice resulted in modest indications of cortical brain swelling that were

particularly evident at the craniotomy site in sham-injured mice. Cerebrospinal fluid

Page 171: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

158

turnover occurs approximately four times a day with a total volume of approximately 40

L of fluid present in the adult mouse brain depending on the age and weight of the

mouse (Pardridge 1991). Utilization of osmotic minipumps with flow rates of 0.11 L/hr to

0.5 L/hr resulted in infusion of approximately 2.5 L to 12 L during a period of 24 hr,

suggesting that the volume of infusate represents a small volume (<10%) of total

cerebrospinal fluid volumes in the brain. However, the volume of infusate may have

contributed to brain swelling as we showed that a reduction in the volume of infusate in

sham-injured mice resulted in decreased cortical swelling at the craniotomy site. To the

best of our knowledge, brain swelling has not been reported as a consequence of

prolonged central infusion using osmotic minipumps similar to those utilized in the

current study. The completion of a decompressive craniotomy prevents increases in

intracranial pressure associated with severe CCI (Zweckberger et al. 2003). The

presence of a craniotomy, in sham-injured and brain-injured mice in the current study

may have facilitated brain swelling in response to increases in intracranial pressure

during prolonged infusion. The physical shape and proximity of the cannula base used in

the current study prevented complete sealing of the cranioplasty over the craniotomy

site, potentially allowing swelling of the brain tissue and equilibration of intracranial

pressure during the infusion period. There is a gap in our understanding of the effect of

increased fluid loading on intracranial pressure in the mouse brain after central infusion

in the context of TBI. Only a handful of studies have evaluated prolonged

intracerebroventricular infusion in mice following experimental TBI, including weight drop

injury (Thau-Zuchman et al. 2010; Thau-Zuchman et al. 2012). A single bolus injection of

2 µL saline into the lateral ventricle of CCI-injured mice resulted in an approximate 2-4

mmHg or 8-15% increase in intracranial pressure to 26 mmHg at 24 hr post-injury

(Trabold et al. 2008). With our highest flow rate, 2 L was infused over a period of

Page 172: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

159

approximately 4 hr. The cannula base utilized in the present study prevented the

implantation of an intracranial pressure-monitoring device into the lateral ventricle, but

our findings highlight that future studies should examine potential temporal changes in

intracranial pressures that may result from prolonged continuous central infusion after

severe CCI. It is unlikely that fluid loading alone resulted in brain swelling as sham-

injured mice exhibited less brain swelling than brain injured mice. While increases in fluid

loading does not appear to be the only cause of brain swelling, increasing volumes of

infusate may contribute to the incidence of brain swelling after prolonged central

infusion.

Brain edema develops in the contused cortex and hippocampus ipsilateral to

injury within hours, reaching maximal levels by 24 hr post-injury and resolving towards

non-injured values by 7 d after CCI (Kochanek et al. 1995; Trabold et al. 2008; Terpolilli

et al. 2013). The mean water content values reported in this study for all groups (77-

80%) are comparable to water content values reported in sham-injured mice

(approximately 79%) and consistent with the resolution of injury-induced edema by one

week post-injury (Trabold et al. 2008). However, central infusion of hIGF-1 after severe

CCI resulted in edema (81-82%) far above the normal range in the injured hippocampus

of a subset of mice. Delaying the onset of infusion for approximately 6 hr post-injury in

an effort to reduce fluid loading during acute development of post-traumatic edema did

not reduce brain swelling in the subset of mice at 7 d post-injury. Our findings suggested

that prolonged central infusion was not a main contributor to the exacerbation of brain

injury-induced edema in the subset of mice, and instead may be related to IGF-1 itself.

Because cortical brain swelling in sham-injured mice and hippocampal distortion

in brain-injured mice appear much more pronounced in IGF-1 infused mice, IGF-1 may

Page 173: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

160

be mediating direct or indirect effects that potentiate the development of brain swelling.

Reductions in the dose of infused IGF-1 resulted in decreases in the incidence and

severity of brain swelling after injury, with mice infused with the lowest doses of IGF-1

(0.3 and 1 µg/d) exhibiting no notable brain swelling after severe CCI. Our findings

suggest that IGF-1 may change fluid dynamics in the brain that could promote the

development of exacerbated brain-injury-induced brain swelling in after severe CCI.

Changes in CSF production or absorption could lead to alterations in intracranial

pressure after brain injury. Evidence is emerging that growth factors can disrupt normal

CSF formation and absorption. Intracerebroventricular infusion of bFGF promoted the

development of hydrocephalus, possibly as a result of reduced CSF absorption

(Johanson et al. 1999; Krishnamurthy et al. 2009). Increased levels of endogenous

transforming growth factor β (TGFβ) have been observed in the CSF of patients with

posthemorrhagic hydrocephalus, suggesting that TGFβ is associated with the

development of hydrocephaly after hemorrhage in the brain (Whitelaw et al. 1999; Flood

et al. 2001). Although little is known about the capacity of IGF-1 to disrupt CSF

dynamics, brain-specific overexpression of IGF-1 results in increased brain size and

weight (Ye et al. 1996; Popken et al. 2004; Ye et al. 2004), and one study observed that

overexpression of IGF-1 appeared to reduce the volumes of the lateral, third and fourth

ventricles in the postnatal brain (Popken et al. 2004), suggesting that elevated levels of

IGF-1 in the brain does not promote the development of hydrocephalus. Systemic co-

administration of IGF-1 and growth hormone for a period of 14 d to moderate-to-severely

brain-injured patients did not alter fluid retention or increase intracranial pressure (Hatton

et al. 2006), suggesting that systemic infusion of IGF-1 does not disrupt CSF balance in

the context of human contusion TBI. However, additional work is needed to ensure that

central infusion of IGF-1 does not modulate CSF balance in the context of TBI, as brain

Page 174: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

161

injury-induced increases in intracranial pressure could be exacerbated by changes in

CSF dynamics (Marmarou et al. 1978).

In summary, we have demonstrated that prolonged intracerebroventricular

infusion of IGF-1 attenuates motor and cognitive dysfunction after severe CCI in mice.

Treatment with IGF-1 showed an apparent dose-dependent increase in immature neuron

density after brain injury. Central infusion of either 10 µg/d or 3 µg/d resulted in

significantly increased immature neuron density, an effect retained even with a clinically

relevant 6 hr delayed infusion onset. This IGF-1-mediated increase in immature neuron

density at 7 d post-injury may be the result of increased cellular proliferation and

enhanced neuronal differentiation, important aspects of post-traumatic neurogenesis.

This study also highlights the need for careful consideration of the administration

paradigm utilized for central infusion of growth factor like therapeutic agents for the

treatment of TBI.

Copyright © Shaun William Carlson 2013

Page 175: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

162

Chapter 5: Targeted Astrocyte-specific Conditional IGF-1 Overexpression

Enhances Post-Traumatic Neurogenesis by Promoting Neuronal Differentiation

Introduction

In Chapter 4 we demonstrated that central infusion of IGF-1 attenuated motor

and cognitive impairments and increased immature neuron density in the dentate gyrus

granular layer after severe CCI. Moreover, we provided evidence that increased

immature neuron density may be the result of an IGF-1-mediated enhancement of post-

traumatic neurogenesis. However, we observed that direct infusion of IGF-1 can

exacerbate brain swelling after severe CCI. In order to minimize complications of

hippocampal distortion after central infusion of IGF-1, we chose to pursue our evaluation

of the efficacy of IGF-1 to promote post-traumatic neurogenesis in a conditional

astrocyte-specific IGF-1 overexpressing transgenic mouse model. In this chapter we

sought to evaluate the potential mechanisms, i.e. cell survival, cellular proliferation,

neuronal differentiation, morphological maturation of immature neurons, by which IGF-1

may promote hippocampal neurogenesis after severe CCI.

The controlled cortical impact (CCI) model of brain injury is well characterized

and widely utilized to study not only widespread cortical cell death after contusion, but

also selective regional hippocampal cell loss and the manifestations of functional

impairments including altered hippocampal long term potentiation and cognitive

dysfunction (Hamm et al. 1992; Smith et al. 1995; Fox et al. 1998; Albensi et al. 2000;

Saatman et al. 2006; Hall et al. 2008; Norris and Scheff 2009; Blaiss et al. 2011; Gao et

al. 2011; Pleasant et al. 2011). The severity of hippocampal cell loss after CCI can be

modulated by altering injury parameters, such as impact depth (Goodman et al. 1994;

Saatman et al. 2006; Huh et al. 2011), velocity (Goodman et al. 1994), or impactor tip

geometry (Nishibe et al. 2010; Pleasant et al. 2011). Mild CCI typically results in acute

Page 176: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

163

degeneration in the dentate gyrus and hilus, whereas increases in the injury severity

result in observable degeneration in CA3 and ultimately CA1 (Goodman et al. 1994;

Anderson et al. 2005; Saatman et al. 2006; Elkin et al. 2010; Pleasant et al. 2011). Due

to the fact that the dentate gyrus appears to be most sensitive to trauma after impact,

the CCI model is applicable for investigating the evolution and mechanisms of post-

traumatic neurogenesis (Rola et al. 2006; Gao et al. 2008; Yu et al. 2008; Zhou et al.

2012).

Newborn neurons are generated throughout adulthood in the subgranular (SGZ)

and subventricular zones of the brains of humans and rodents (Eriksson et al. 1998;

Zhao et al. 2008). In the dentate gyrus of the hippocampus, this population of neurons

migrates from the SGZ into the inner and outer layers of the granular layer, and

ultimately extends axonal and dendritic processes to integrate into the dentate gyrus

circuitry (van Praag et al. 2002; Jessberger and Kempermann 2003; Zhao et al. 2008).

This population of newborn neurons is thought to contribute to learning and the

formation of new memories as demonstrated by the fact that genetic depletion of the

immature neuron population impairs spatial learning at 1 month following CCI (Clelland

et al. 2009; Deng et al. 2009; Jessberger et al. 2009).

CCI results in a marked reduction in the numbers of immature neurons in the

hippocampal granular layer within days after injury (Rola et al. 2006; Gao et al. 2008; Yu

et al. 2008). Markers of degenerating neurons are observed in the inner granular layer

and found to predominantly colocalize with markers of immature neurons (Gao et al.

2008). Brain injury promotes cellular proliferation in the granular layer and the expansion

of the SGZ neurogenic niche (Dash et al. 2001; Kernie et al. 2001; Rola et al. 2006; Gao

et al. 2009) that contributes to the recovery of the newborn neuron population in the

Page 177: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

164

days to weeks following injury (Rola et al. 2006; Yu et al. 2008). A recent study

highlights that blocking post-traumatic neurogenesis for the 4 weeks following brain

injury, by genetic depletion of the immature neuron population, is sufficient to exacerbate

injury-induced deficits in spatial learning at 1 month following CCI (Blaiss et al. 2011).

Only a portion of the newborn neurons generated after brain injury will survive and

develop into mature granular neurons that incorporate into the granular layer (Sun et al.

2005; Rola et al. 2006; Sun et al. 2007). Due to the importance of immature neurons in

cognition, it is possible that enhancement of post-traumatic neurogenesis attenuates

learning and memory deficits associated with TBI.

Several studies have demonstrated that post-traumatic neurogenesis can be

enhanced after TBI with therapeutic agents and growth factors, including S100B,

erythropoietin, vascular endothelial growth factor and basic fibroblast growth factor

(Kleindienst et al. 2005; Lu et al. 2005; Sun et al. 2005; Thau-Zuchman et al. 2010).

Similar to the aforementioned growth factors, IGF-1 has been shown to promote

neurogenesis in the rodent hippocampus (Aberg et al. 2000; Trejo et al. 2001; Trejo et

al. 2008); however, the efficacy of IGF-1 to promote neurogenesis in the context of TBI

is not known. Insulin-like growth factor-1 (IGF-1) is a potent neurotropic factor, which

upon binding to its cognate receptor increases activation of Akt and MAPK (Seger and

Krebs 1995; Manning and Cantley 2007). Numerous studies have demonstrated that

enhanced IGF-1 signaling promotes stages of neurogenesis including cell survival,

proliferation, dendritic growth, and neuronal differentiation of newly proliferated cells

(Arsenijevic and Weiss 1998; Brooker et al. 2000; Niblock et al. 2000; Arsenijevic et al.

2001; Cheng et al. 2003; McCurdy et al. 2005). Overexpression of IGF-1 by glial and

neuronal-specific promoters results in increased numbers of neurons and glial cells in

the brain (Carson et al. 1993; Ye et al. 1995; Chrysis et al. 2001; Popken et al. 2004; Ye

Page 178: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

165

et al. 2004), due to increased cellular proliferation and reduced apoptosis (Ye et al.

1996; Chrysis et al. 2001). IGF-1 has been implicated as a mediator of exercise-induced

hippocampal neurogenesis evident by increased serum levels and increased brain

uptake of IGF-1 that accompany increases in hippocampal proliferation and newborn

neuron production (Trejo et al. 2001; Llorens-Martin et al. 2010). Furthermore, blocking

uptake of serum IGF-1 into the brain abolishes exercise-induced increases in

neurogenesis (Trejo et al. 2001; Llorens-Martin et al. 2010). In addition to promoting

neurogenesis, central administration of IGF-1 in aged rats reverses an age-induced

reduction in hippocampal neurogenesis (Lichtenwalner et al. 2001). Collectively, these

studies highlight that IGF-1 is a potent promoter of neurogenesis in the adult brain.

To evaluate the ability of IGF-1 to promote neurogenesis after TBI, we utilized an

astrocyte-specific conditional IGF-1 overexpressing transgenic mouse (Ye et al. 2004) to

increase brain levels of IGF-1 following severe CCI. The expression of IGF-1 is restricted

to glial fibrillary acidic protein (GFAP) expressing cells and is regulated by a Tet-off

expression system. Dietary supplementation of doxycycline suppresses expression of

IGF-1 during postnatal development, thereby minimizing potentially confounding effects

of IGF-1 overexpression during brain development. Brain injury is associated with

increased astrogliosis and increased GFAP expression in injured tissue after injury.

Reactive astrocytosis and enhanced GFAP expression in the hippocampus after CCI

(Saatman et al. 2006; Sandhir et al. 2008) provides targeted expression of IGF-1 in the

damaged hippocampus (Madathil 2013).

Using the conditional astrocyte-specific IGF-1 overexpression transgenic mouse

model, we will assess the densities of immature neurons, newly proliferated cells, and

immature neurons generated in the hippocampal granular layer after CCI in order to

Page 179: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

166

evaluate the efficacy of IGF-1 to enhance neurogenesis in the context of severe CCI. In

this chapter we demonstrate that elevated brain levels of IGF-1 promotes post-traumatic

neurogenesis by enhancing neuronal differentiation of newborn cells. We demonstrate

that severe CCI induced a significant reduction in immature neuron dendritic arbor

complexity, a previously unidentified pathological consequence in this population of

neurons. We also establish that overexpression of IGF-1 in brain-injured mice restores

immature neuron dendritic process complexity to that observed in mice subjected to

sham injury.

Page 180: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

167

Materials and Methods

Animals

All experimental procedures were approved by the University of Kentucky

Institutional Animal Care and Use Committee in accordance with the Guide for the Care

and Use of Laboratory Animals established guidelines from the National Institutes of

Health. Animals were housed 5 mice per cage in the University of Kentucky Medical

Center vivarium with a 14:10 hour light/dark photoperiod and were provided food and

water ad libitum, but food consumption was not monitored in animals utilized for this

study. Adult male transgenic IGF-1 overexpressing mice (IGF TG) and wild-type (WT)

littermates aged 8-12 weeks, weighing 22-30g, were utilized for all experiments.

The astrocyte-specific conditional overexpression of human IGF-1 in the

transgenic mouse has been previously described (Ye et al. 2004). The production of the

tetracycline transactivator (tTA) protein is linked to the glial fibrillary acidic protein

(GFAP) promoter in tTAGFAP mice. The transgene for human IGF-1 was inserted into a

pTRE plasmid and utilized to generate IGF-1pTRE mice. Crossing of tTAGFAP and IGF-

1pTRE mice yields double transgenic mice that carry both transgenes and conditionally

express IGF-1 in GFAP expressing cells. Expression of IGF-1 was suppressed for the

first 4 weeks of postnatal development by utilizing a Tet-off strategy and dietary

supplementation of doxycycline in mouse chow (200 mg/kg). The binding of tTA to the

tetracycline response element that drives IGF-1 expression is blocked by the binding of

doxycycline to the tTA protein. Mice received standard mouse chow for at least 4 weeks

to provide sufficient time for transgene expression prior to surgical procedures.

Page 181: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

168

Controlled Cortical Impact

Mice were anesthetized using 3% isoflurane in order to shave their heads and

place them in a stereotaxic frame (David Kopf Instruments, CA), where anesthesia was

maintained using 3% isoflurane via a nose cone for the duration of the surgical

procedure. A midline scalp incision was performed and the skin reflected to expose the

skull. A 5 mm craniotomy was performed centered between bregma and lambda lateral

to the sagittal suture of the left hemisphere of the brain with the dura left intact. Animals

were randomly assigned to receive either sham or CCI injury. Sham-injured mice

received anesthesia and only a craniotomy prior to the cranioplasty and closure of the

incision. The CCI injury was completed using a computer-controlled pneumatically driven

piston that rapidly impacts the brain (TBI-0310 Impactor, Precision Systems and

Instrumentation, VA). Mice subjected to brain injury received a 1 mm severe CCI injury

with a velocity of 3.5 m/s using a 3 mm diameter rounded impactor tip. Following injury,

a cranioplasty was completed using a small circular disk of dental acrylic adhered to the

skull over the craniotomy site, the scalp was sutured, and the mice were placed on a

heating pad to maintain normal body temperature. Once ambulatory, the mice were

returned to their home cage.

In one cohort, mice (n=4 sham-injured/genotype and n=8 CCI-injured/genotype)

were injected with 50 mg/kg (i.p.) 5-bromo-2-deoxyuridine (BrdU; Fisher Scientific, NJ) to

identify proliferating cells 4 hr prior to perfusion at 3 d post-injury. A second cohort of

mice (n=8 sham-injured/genotype and n=13 brain-injured/genotype) received daily BrdU

injections (50 mg/kg i.p.) beginning 1 hr after injury for 7 d to identify proliferating cells

during the first week following injury. After 3 additional days of survival to provide

sufficient time for newly proliferated cells to differentiate into immature neurons and

express DCx (Encinas 2003), mice were euthanized at 10 d post-injury.

Page 182: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

169

Tissue Collection and Processing

Animals received an overdose of Fatal-plus (65 mg/kg sodium pentobarbital, i.p.)

at either 3 or 10 d post-injury, and were transcardially perfused with heparinized saline

followed by 10% neutral buffered formalin. Brains were removed from the skull 24 hr

following post-fixation in 10% neutral buffered formalin, post-fixed for an additional 24 hr

in 10% neutral buffered formalin, cryoprotected in 30% sucrose for 24 hr and rapidly

frozen in -30°C isopentanes cooled on dry ice. Brains were cut into 40 µm-thick coronal

sections using a sliding microtome (Dolby-Jamison, Pottstown, PA) and stored in

cryoprotectant in parallel sets at -20°C.

Immunohistochemistry

Coronal brain sections, spaced 400 µm apart, between -1 mm and -3.5 mm

bregma (Paxinos and Franklin 2001) were stained using standard free-floating

immunohistochemistry staining protocols. Primary antibodies utilized were anti-

doublecortin (DCx; rabbit polyclonal, 1:500, Abcam, MA) to label the microtubule-

associated protein expressed exclusively in immature neurons and anti-BrdU (1:1000,

Abcam) to label proliferating cells. Secondary antibodies were conjugated with Alexa

Fluor® 488 (1:1000, Invitrogen, CA) or Cy3 (1:1000, Jackson ImmunoResearch, PA).

Sections were initially rinsed in TBS and treated with 2N HCl (Fisher Scientific) for 1 hr

at room temperature to expose the BrdU epitope, followed by treatment of 100mM

borate buffer (pH 8.0, Fisher Scientific) for 10 minutes. Sections were rinsed in TBS

overnight at 4°C. On the second day, sections were rinsed in TBS and blocked in 5%

normal horse serum and 0.1% Triton-X-100 in TBS for 30 minutes. Both DCx and BrdU

primary antibodies were diluted in blocking solution and sections incubated overnight at

4°C. The following day tissue was rinsed with TBS, incubated with secondary antibodies

for 1hr, rinsed with TBS, incubated with Hoechst (1:10,000, Invitrogen, CA) for 1.5

Page 183: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

170

minutes and rinsed with TBS. Labeled sections were mounted on gelatin-coated slides,

coverslipped with fluoromount (Southern Biotech, AL), and stored at 4°C.

Image Acquisition and Quantification

Representative images utilized in the figures are maximum intensity projections

of images collected as a z stack (1.5 µm step size) at 20X magnification using a C2+ TiE

Nikon confocal microscope. The densities of immature neurons and proliferated cells

were quantified in the ipsilateral dentate gyrus granular layer (DGGL) of each section,

with sections spanning the rostrocaudal extent of the hippocampus from -1.06 to -3.5

mm to bregma (Paxinos and Franklin 2001). We sampled one set of parallel sections (an

average of 6 sections), spaced 400 µm apart, for each animal. All DCx or BrdU positive

cells were manually counted in the upper and lower blades of the DGGL for each section

at 40X (Olympus, BX51) using a FITC filter (41001, Chroma Technology, VT), or a

TRITC filter (31002, Chroma Technology). The incidence of DCx and BrdU

colocalization was assessed using a wide band filter (51004v2, Chroma Technology)

that permits simultaneous viewing of the emissions of both fluorophores of the

secondary antibodies. Colocalization was verified by scanning through the z-axis to

ensure colabeling within each quantified cell. Cell counts were normalized to the volume

of the DGGL of each section in order to control for differences in granular DGGL size

across sections. The Hoechst-stained DGGL of each quantified section was imaged

(Olympus AX80, PA) and the area was measured using ImagePro (MediaCybernetics,

MD). The thickness of each section was measured, in microns, using a stereology stage

(Olympus BX51). Density measurements reflect the entire granular layer inclusive of

subgranular, inner, and outer granular layers. For each animal, a mean hippocampal cell

density was calculated for all quantified sections. In order to evaluate cell density as a

function of proximity to injury, we evaluated the location of each section and assigned

Page 184: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

171

the section to a specific bregma level according to the plates in the Paxinos and Franklin

mouse brain atlas (2001). Sections were sorted into one of five bregma intervals

spanning approximately 500 µm, with the center bregma level interval (-2.06 to -2.46

mm) representing the epicenter of injury. In the event that two sections for a given

animal fell within a single bregma interval, an average density measurement was

determined for the two sections and this single value was used for that interval. Newborn

neurons (DCx+/BrdU+) were also expressed as a percentage of total proliferated cells

(BrdU+) to illustrate differences in cellular differentiation between the genotypes following

injury. Quantification was performed by an investigator blinded to the genotype and

injury conditions of each animal.

Scholl Analysis

Images of DCx were acquired as a z-stack (1.5 µm step size) at 20X

magnification using a CKX31 A1 Nikon confocal microscope from a subset of mice (n=6 /

sham-injured / genotype and n=6 brain-injured / genotype) were randomly selected from

the 10 d cohort. Confocal images were obtained of eight randomly selected immature

neurons from the upper blade of the DGGL from a section at the epicenter of injury (-

2.06 to -2.46 mm). The neuron was not utilized if dendritic fibers could not be traced

through the confocal z-stack images. Dendritic processes of DCx expressing

hippocampal granular neurons were manually traced on maximum intensity projection

images, created from the z-stacks, and total dendritic length was analyzed using the

NeuronJ plug-in for NIH ImageJ, as previously described (Cai et al. 2012). The number

of dendritic intersections was quantified for each DCx+ neuron by a series of concentric

circles (10 µm intervals) drawn from the center of the cell body using a Scholl analysis

plug-in for NIH ImageJ. All image acquisition and subsequent analysis was completed by

an investigator blinded to the genotype and injury status of each animal.

Page 185: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

172

Statistical Analysis

All data is presented as mean + standard error of the mean (SEM). Cellular

densities and measurements of dendritic morphology were compared using a two-way

analysis of variance (ANOVA) followed by Newman-Keuls post-hoc t-tests when

appropriate. Statistical tests were completed using Statistica (Statsoft Inc, OK). A p

value less than 0.05 was considered statistically significant for all tests.

Page 186: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

173

Results

TBI results in acute cell loss throughout the hippocampus, with the dentate gyrus

being particularly susceptible to contusion injury (Anderson et al. 2005; Saatman et al.

2006; Hall et al. 2008). To examine if IGF-1 overexpression confers protection to

immature neurons in the hippocampus after severe CCI, we labeled immature neurons

using doublecortin (DCx). Immunohistochemical staining for DCx in WT mice revealed

loss of immunoreactivity consistent with cell loss scattered throughout the upper and

lower blades of the granular layer 3 d after CCI, and an apparent recovery of cell

number, similar to sham injury, at 10 d post-injury (Fig. 5.1A). Qualitative assessment of

DCx immunoreactivity demonstrated that IGF TG mice exhibited a reduction in

immunoreactivity consistent with cell loss throughout the upper and lower blades of the

granular layer at 3 d after CCI, similar to WT mice. At 10 d after CCI, brain-injured IGF

TG mice exhibited a robust increase in DCx immunoreactivity within each positive cell

and an apparent increase in cell number above IGF TG mice subjected to sham injury

(Fig. 5.1B).

CCI resulted in a 37% reduction in the numbers of DCx+ neurons in WT mice (25

+ 2 cells/mm3) at 3 d post-injury with a recovery to sham levels (40 + 3 x 1000/mm3) by

10 d (40 + 3 x 1000/mm3) following injury (Fig. 5.1C). IGF-1 overexpressing mice

exhibited a similar basal immature neuron density in sham-injured mice (43 + 7 x

1000/mm3), and a 23% decrease in DCx density at 3 d post injury (33 + 2 x 1000/mm3).

However, brain-injured IGF TG mice exhibited an 86% increase over sham–injured IGF

TG mice and an enhanced recovery of immature neuron density at 10 d post-injury (80 +

8 x 1000/mm3) compared to brain-injured WT mice (#p<0.05, Fig. 5.1C).

Page 187: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

174

To determine if immature neuron loss varied as a function of proximity to injury,

DCx+ density was evaluated along the rostrocaudal extent of the dentate gyrus granular

layer (-1.06mm to -3.4mm bregma). At 3 d following CCI, WT mice exhibited the greatest

reduction in immature neuron density (53%), compared to sham-injured mice, at the

epicenter of impact (-2.06 to -2.46mm), but reductions in immature neuron density were

notable in regions as far as approximately 500 µm rostral or caudal to the injury

epicenter (Fig. 5.1D). At 3 d following CCI, immature neuron loss in IGF TG mice was

evident in regions approximately 500 μm rostral and caudal to the epicenter of injury,

similar to CCI in WT mice (Fig. 5.1D). While DCx+ density returned to sham levels by 10

d post-injury at most bregma levels in WT mice, IGF-1 TG mice showed significant

increases, above sham levels, at the epicenter of injury and as far as approximately 500

µm rostral and caudal to the epicenter (# p<0.05, compared to all groups within that

bregma interval; Fig. 5.1D).

Page 188: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

175

Figure 5.1: IGF-1 overexpression increases immature neuron density in the

dentate gyrus granular layer following controlled cortical impact. (A, B)

Representative images of doublecortin (DCx, green) immunoreactivity in the upper blade

of the dentate gyrus granular layer of sham and brain-injured (A) wild-type (WT) and (B)

IGF-1 overexpressing transgenic (IGF TG) mice at 3d and 10d after injury. Scale bar

represents 50 µm. Granular layer (GL). (C) DCx+ cell density in WT mice recovered to

sham levels by 10 d post-injury. IGF TG mice exhibited a significant increase in DCx+ cell

Page 189: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

176

Figure 5.1 (cont.): density at 10 d post-injury (# p<0.05, compared to all groups). (D)

The increase in DCx+ cell density with IGF-1 overexpression is present over a large

rostrocaudal extent of the injured granular layer, presented along the x-axis as intervals

spanning Bregma coordinates (mm). Data are presented as mean cell density (x

1000/mm3) + SEM. (n=8 sham-injured/genotype, n=8 brain-injured/genotype at 3 d, and

n=13 brain-injured/genotype at 10 d post-injury). Two-way ANOVAs were utilized for

statistical analyses in (C) and at each bregma interval in (D), and followed by Newman-

Keuls post-hoc t-tests where appropriate.

Page 190: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

177

Severe CCI results in increased cellular proliferation within the granular layer of

the hippocampus within days following brain injury (Dash et al. 2001; Rola et al. 2006).

To evaluate if IGF-1 overexpression enhances immature neuron density at 10 d post-

injury by promoting cellular proliferation within the SGZ, we evaluated the extent of

proliferation using two BrdU injection paradigms: 1) a single injection at 3 d post-injury, a

peak time of proliferation (Dash et al. 2001) and 2) a cumulative injection paradigm to

capture proliferation over the first 7 d following CCI. Immunohistochemical labeling of

BrdU+ cells demonstrated no qualitative difference in the extent of proliferation in the

granular layer of WT and IGF-TG sham-injured mice in either injection paradigm (Fig.

5.2A, B). While CCI stimulated a significant 5-fold increase in cellular proliferation within

the ipsilateral granular layer mice at 3 d post-injury (main injury effect, p<0.01), IGF-1

overexpression did not affect early proliferation (Fig. 5.2C). The cumulative labeling

paradigm of repeated daily BrdU injections for 7 d revealed a similar extent of cellular

proliferation between WT and IGF TG sham-injured mice (Fig. 5.2B, D). Brain injury

produced a significant 3-fold increase in proliferation during the week following the injury

(main injury effect, p<0.01); however, proliferation was not enhanced by IGF-1

overexpression (Fig. 5.2D). These data suggest that the increased immature neuron

density observed in IGF TG mice at 10 d after CCI was not afforded by an IGF-1-

meditated enhancement of cellular proliferation.

Page 191: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

178

Figure 5.2: IGF-1 overexpression does not enhance brain injury-induced cellular proliferation in the hippocampal granular layer following controlled cortical impact. (A, B) Representative images of proliferated cells identified by bromo-deoxyuridine (BrdU, red) immunoreactivity counterstained with Hoechst stain (blue) in the upper blade of the granular layer of wildtype (WT) and IGF-1 overexpressing (IGF TG) mice 3 d (A) and 10 d (B) post-injury. Scale bar represents 50 µm. (C, D) BrdU+ cell density is significantly increased at 3 d (C) and 10 d (D) post-injury (main injury effect, p<0.01), independent of genotype. Data are presented as mean cell density (x 1000/mm3) + SEM. (n=8 sham-injured/genotype, n=8 brain-injured/genotype at 3 d, and n=13 brain-injured/genotype at 10 d post-injury). Two-way ANOVAs were utilized for statistical analyses.

Page 192: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

179

New neurons are constantly generated in the hippocampal SGZ throughout

adulthood by neuronal differentiation of newly proliferated progenitor cells (Zhao et al.

2008). To evaluate if IGF-1 enhances total immature neuron density at 10 d post-injury

by promoting neuronal differentiation, we assessed DCx+ and BrdU+ colocalization to

identify the population of immature neurons generated during the week after injury.

Colocalization of DCx and BrdU was observed in the SGZ, with DCx immunoreactivity in

the cell body encapsulating the nucleus containing the BrdU-labeled DNA (Fig. 5.3A).

IGF TG mice exhibited increased density of newborn neurons compared to WT mice

(main genotype effect, p<0.05), but this effect was more pronounced in brain-injured

mice (interaction effect, p<0.05). IGF-1 overexpression elicited a small, but non-

significant increase in the density of newborn neurons in sham-injured mice (10 + 2 x

1000/mm3) compared to WT sham-injured mice (6 + 1 x 1000/mm3; Fig 5.3B). In WT

mice, brain injury did not result in a change in the density of newborn neurons (7 + 1 x

1000/mm3), compared to sham injury (Fig. 5.3B). However, IGF-1 overexpression

resulted in a significant 2.5-fold increase in newborn neuron density 10 d following CCI

(25 + 5 x 1000/mm3), compared to the density of IGF-1 overexpressing sham-injured

mice and a significant 3.5 fold increase in density compared to WT brain-injured mice (#

p<0.005; Fig. 5.3B). When evaluated by proximity to injury, as a function of bregma

level, the selective increase in newborn neurons in IGF TG brain-injured mice was

observed at all levels except the most rostral (-1.06 to -1.46 mm bregma)(#p<0.05,

compared to all groups within each bregma interval; Fig. 5.3C).

In sham WT and IGF TG mice, newborn neurons represent approximately 40-

50% of all proliferating cells in the dentate gyrus granular layer. The percentage of

newborn neurons that comprised total proliferated cells was slightly increased in IGF TG

sham-injured mice (50%) compared to sham-injured WT mice (35%, Fig 5.3D). Although

Page 193: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

180

proliferation is markedly increased after brain injury in WT mice, newborn neurons

represented only approximately 10% of all proliferating cells (Fig. 5.3D). Conversely,

overexpression of IGF-1 in brain-injured mice resulted in an increased the proportion of

proliferated cells that became neurons to approximately 42% (Fig 5.3D).

Page 194: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

181

Figure 5.3: IGF-1 overexpression promotes neuronal differentiation of newly

proliferated cells in the granular layer following controlled cortical impact. (A)

Representative images of doublecortin (DCx, green) and bromo-deoxyuridine (BrdU,

red) immunoreactivity and Hoechst staining (blue) in the granular layer of sham-injured

and brain-injured wildtype (WT) and IGF-1 overexpressing transgenic (IGF TG) mice 10

d post-injury. Newborn neurons were identified by colocalization of DCx and BrdU. Scale

bar represents 50 µm. Granular layer (GL). (B) Newborn neuron (DCX+/BrdU+) density in

WT mice following brain injury was similar to WT sham-injured mice. However, IGF TG

mice exhibited a significant increase in newborn neuron density 10 d post-injury (#

p<0.05, compared to all other groups). (C) Overexpression of IGF-1 appeared to

increase newborn neuron density throughout most of the injured hippocampus (#

p<0.05, compared to all other groups classified by bregma interval). (D) Following brain

injury in WT mice, newborn neurons comprised a much smaller proportion of the total

proliferated cells than in sham injured controls. However, in IGF TG brain-injured mice a

Page 195: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

182

Figure 5.3 (cont.): greater proportion of proliferated cells adopted a neuronal

phenotype. Data are presented as mean cell density (1000/mm3) + SEM. (n=8 sham-

injured/genotype and n=13 brain-injured/genotype at 10 d post-injury). Two-way

ANOVAs were utilized for statistical analyses in (B) and (C), and were followed by

Newman-Keuls post-hoc t-tests where appropriate.

Page 196: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

183

In addition to increased immature neuron density in IGF TG mice after CCI, we

observed a qualitative increase in the complexity of the dendritic arbors IGF TG mice,

compared to WT mice at 10 d following CCI (Fig. 5.1A). Examination of the dendritic

arbor of DCx+ labeled cells located within the SGZ of WT mice at 10 d post-injury

demonstrated a brain injury-induced reduction in dendritic complexity (Fig. 5.4B). To

evaluate the efficacy of IGF-1 to attenuate or reverse damage to dendritic processes

after CCI, we quantified total dendritic process length and dendritic arbor complexity of

DCx+ neurons within the granular layer. WT sham-injured and IGF TG sham-injured

mice exhibited similar total dendritic lengths (Fig. 5.4C). Following CCI, WT mice

exhibited a significant reduction in the total length of dendritic processes (143 + 8 µm),

as compared to WT sham-injured mice (305 + 21 µm, *p<0.005; Fig. 5.4C). The total

dendritic length for IGF TG brain-injured mice (292 + 18 µm) was significantly increased

(#p<0.001) compared to WT brain-injured mice and was not significantly different from

WT sham-injured or IGF TG sham-injured mice (346 + 16 µm). Quantification of the

intersections per shell, a measure of dendritic bifurcations, revealed no difference in the

number of intersections between WT and IGF TG sham-injured mice (Fig. 5.4D). Brain

injury in WT mice induced a pronounced reduction in the number of intersections as well

as early termination of the dendritic arbor, compared to WT sham-injured mice (p<0.001,

Fig. 5.4D). Brain-injured IGF TG mice exhibited similar complexity of dendritic

arborization to IGF TG sham-injured mice. These data demonstrate that IGF-1

overexpression ameliorates the disruption of immature neuron dendritic morphology

associated with CCI.

Page 197: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

184

Figure 5.4: IGF-1 overexpression restores immature neuron dendritic arbor

complexity after controlled cortical impact (CCI). (A, B) Representative images (A)

and tracings (B) of doublecortin (DCx+, green) neurons in the granular layer of wildtype

(WT) sham-injured (top), WT brain-injured (middle) and IGF-1 overexpressing transgenic

(IGF-1 TG) brain-injured mice (bottom). (C) WT brain-injured mice exhibited a significant

reduction in total dendritic length that was attenuated in IGF TG brain-injured mice (*

p<0.005, compared to WT sham-injured mice; # p<0.001, compared to WT brain-injured

mice). (D) Scholl analysis revealed that IGF-1 overexpression protected against injury-

induced reduction in dendritic arbor complexity at 10 d after brain injury. Data are

presented as mean + SEM. (n=6 sham-injured/genotype and n=6 brain-

injured/genotype). Scale bars represent 50 µm.

Page 198: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

185

Discussion

In Chapter 4 we presented compelling evidence that central infusion of IGF-1

increases hippocampal immature neuron density at 7 d post-injury, and that this may be

the result of enhanced post-traumatic neurogenesis. In this chapter, we build upon our

findings in Chapter 4 using a complementary approach and demonstrate that brain-

specific overexpression of IGF-1 robustly increases immature neuron density at 10 d

following severe CCI. This increase in immature neuron density appears to be driven not

by protection against acute cell death but by enhanced generation of newborn neurons

during the week following brain injury. Following CCI, we also show that overexpression

of IGF-1 restores immature neuron dendritic arbor complexity to that observed following

sham injury.

In this chapter, we utilized a conditional astrocyte-specific IGF-1 overexpressing

transgenic mouse model to increase levels of IGF-1 in regions of enhanced GFAP

expression after CCI. Reactive astrocytosis has been reported in the dentate gyrus

within 24 hr after CCI injury (Saatman et al. 2006; Sandhir et al. 2008; Madathil 2013).

Previous work from our lab has shown that reactive astrocytosis in the injured

hippocampus increased expression of IGF-1 at 24 hr with further increases at 72 hr

following CCI in the IGF-1 overexpressing mouse model utilized in this chapter (Madathil

2013). Similar to infusion of exogenous IGF-1 in Chapters 3 and 4, targeted

overexpression of IGF-1 in astrocytes led to a biologically appropriate increase in Akt

activation following severe CCI (Madathil 2013). Targeted overexpression of IGF-1 also

protected against acute hippocampal neuron loss in the dentate gyrus, CA3, and CA1

and improved cognitive performance following severe CCI (Madathil 2013). While the

observation of improved neurobehavioral function may be related to hippocampal

Page 199: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

186

neuroprotection, an IGF-1-mediated enhancement of post-traumatic neurogenesis may

also contribute to improved cognitive function.

In Chapter 3, systemic administration of rhIGF-1 was shown to result in a modest

reduction in hippocampal neurodegeneration in the granular layer, visualized by

Fluorojade-B staining, suggesting that IGF-1 reduces neurodegeneration acutely after

brain injury. Moreover, previous work from our lab, utilizing the conditional astrocyte-

specific IGF-1 overexpressing mouse model demonstrated that overexpression of IGF-1

reduced the number of Fluorojade-C positive cells, an indication of degenerating

neurons, in the dentate gyrus granular layer at 3 d post-injury (Madathil 2013). We

postulated that reductions in hippocampal neurodegeneration in the granular layer may

be reflective of improved immature neuron survival. Several studies demonstrate that

immature neurons are selectively vulnerability to moderate and severe CCI and,

consequently, immature neuron density is markedly reduced in the days following CCI

(Rola et al. 2006; Gao et al. 2008; Yu et al. 2008). Consistent with these studies

reporting acute immature neuron loss, we show an approximate 35% reduction in

numbers of DCx-positive cells in the injured granular layer at 3 d after severe CCI.

However, overexpression of IGF-1 did not significantly increase immature neurons

density at 3 d following brain injury. These findings are also consistent with our findings

in Chapter 3 that systemic infusion of IGF-1 did not increase immature density at 3 d

following systemic infusion. Taken together, these studies highlight that IGF-1 reduces

hippocampal neurodegeneration, but may not promote the survival of immature neurons

acutely following CCI.

One possible explanation for the lack of IGF-1-mediated protection of immature

neurons could be related to immature neuron insensitivity to IGF-1. The IGF receptor is

Page 200: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

187

highly expressed in hippocampal neurons throughout the granular layer in the

developing and adult brain, suggesting that the niche should be capable of responding to

IGF-1 (Werther et al. 1990; Bondy and Lee 1993; Bracko et al. 2012). However, changes

in IGF-1 receptor cellular localization or expression levels could reduce the actions of

IGF-1 in the injured brain. In the context of brain injury, internalization of plasma

membrane receptors, including NMDA and AMPA, has been reported to occur within

hours after experimental TBI (Biegon et al. 2004). A reduction in the levels or

presentation of IGF-1 receptors on the plasma membrane could blunt the anti-apoptotic

effects of IGF-1 on hippocampal neurons, including immature neurons in the granular

layer. Our lab has previously demonstrated that levels of the IGF-1 receptor do not

change in the cortex for at least 72 hr following severe CCI (Madathil et al. 2010).

However, this work did not evaluate if expression patterns differ in the hippocampus or if

cellular localization and presentation of IGF-1 receptor on the plasma membrane is

altered as a consequence of brain injury. A more comprehensive evaluation of IGF-1

receptor expression in hippocampal immature neurons and receptor localization after

injury may provide insight to help explain why IGF-1 does not appear to protect against

acute hippocampal immature neuron loss.

In order to better understand immature neuron loss relative to proximity of injury,

we analyzed the density of immature neurons as a function of bregma level. The

epicenter of injury exhibited the greatest reduction in immature neuron density with

observable reductions at least 500 µm from the epicenter of injury. To the best of our

knowledge, this is the first study to evaluate hippocampal immature neuron loss as a

function of proximity to injury. These data demonstrate that future studies need to

incorporate methodology to quantify post-traumatic alterations in neurogenesis

throughout the entire dentate gyrus granular layer, and not restrict quantification to the

Page 201: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

188

site of maximal injury. We demonstrate that overexpression of IGF-1 enhanced newborn

neuron density throughout the granular layer and not only at the epicenter of injury.

Adoption of rigorous quantification methodology throughout the granular layer may be

especially critical for therapeutic studies evaluating immature neurons loss and

modulation of neurogenesis following experimental TBI.

CCI results in a marked reduction in immature neurons within the DGGL, but

brain injury also initiates an endogenous self-renewal process, including increased

cellular proliferation that promotes a gradual recovery of the number of immature

neurons to pre-injury levels (Rola et al. 2006; Yu et al. 2008). The data presented in this

chapter demonstrates that IGF-1 does not confer acute protection of the immature

neurons, but instead enhances the recovery of immature neurons at 10 d post-injury.

Brain injury induces proliferation of glial cells and stem cells within the dentate gyrus,

during the first week after brain injury (Dash et al. 2001; Chirumamilla et al. 2002; Emery

et al. 2005; Sun et al. 2005; Rola et al. 2006; Urrea et al. 2007). Several studies

demonstrate that cellular proliferation can be enhanced with the treatment of growth

factors, including VEGF, bFGF, and EGF, after experimental TBI (Yoshimura et al. 2003;

Sun et al. 2009; Sun et al. 2010). However, only a portion of the newly proliferated cells

will adopt a neuronal phenotype following severe CCI (Rola et al. 2006). IGF-1 has been

shown to enhance the generation of new neurons by promoting progenitor cell

proliferation and enhancing neuronal differentiation (Arsenijevic and Weiss 1998; Aberg

et al. 2000; Brooker et al. 2000; Arsenijevic et al. 2001).

We were surprised that overexpression of IGF-1 did not enhance post-traumatic

proliferation. Previous in vitro and in vivo studies have demonstrated that IGF-1

promotes proliferation of cultured neural stems cells (Brooker et al. 2000; Arsenijevic et

Page 202: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

189

al. 2001) and increases the proliferation of neural progenitor cells in the granular layer of

the hippocampus (Aberg et al. 2000; Trejo et al. 2001; Yan et al. 2006). IGF-1 is an

important mediator of exercise-induced neurogenesis by enhancing proliferation in the

DGGL (Trejo et al. 2001; Trejo et al. 2008). Together, these studies suggested that we

might expect an IGF-1-mediated enhancement of cellular proliferation in the DGGL.

However, in the current study we did not observe an IGF-1-mediated enhancement of

cellular proliferation at 3 or 10 d post-injury. Our findings from Chapter 4 showed that

central infusion of IGF-1 may have promoted increased cellular proliferation after CCI.

The BrdU injections paradigms utilized in the studies from Chapter 4 and this chapter

are different and may explain the disparity between our findings. Differences in the BrdU

injection paradigms including the time post-injury in which injections are given, the

frequency of injections and the duration between last injection and euthanasia may

begin to reflect a combination of both cellular proliferation and cell survival. Future

studies will need to incorporate evaluations of cellular proliferation using BrdU injection

paradigms that utilize single injections of BrdU with euthanasia within hours in order to

minimize contributions of cell survival in assessments of cellular proliferation.

Overexpression of IGF-1 appeared to promote the generation of newborn

immature neurons by promoting neuronal differentiation of newly proliferated cells after

injury. This effect was present throughout most bregma levels of the hippocampus, and

not restricted to regions exhibiting maximal injury or DCx+ cell loss. IGF-1

overexpression appears to promote neurogenesis in the neurogenic niche throughout

the injured hippocampus. Our observations of enhanced neuronal differentiation are

consistent with a previous report showing that systemic administration of IGF-1 in

hypophysectomized rats increased the generation of new neurons, but did not promote

the generation of astrocytes in the DGGL (Aberg et al. 2000). Although only a portion of

Page 203: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

190

newborn neurons generated after brain injury develop into mature granular neurons and

functionally incorporate, these newborn neurons may contribute to delayed

improvements in cognitive function (Sun et al. 2007), suggesting that therapies that

increase the number of immature neurons generated after brain injury may promote

cognitive recovery. Moreover, a recent study highlights that supraphysiological increases

in immature neuron numbers, similar to our observation at 10 d post-injury in IGF TG

mice, are sufficient to improve cognition as assessed by a contextual pattern separation

task (Sahay et al. 2011). Previous work from our lab demonstrated that IGF-1

overexpressing mice subjected to brain injury exhibited improved cognitive performance

compared to wildtype mice at 7 d following severe CCI (Madathil 2013). The findings in

this current study suggest that an IGF-1-mediated enhancement of post-traumatic

neurogenesis may contribute to the improved cognitive performance reported in this

transgenic mouse model following severe CCI.

It is also possible that the increased number of newborn neurons observed in

IGF-1 overexpressing mice at 10 d post-injury is reflective of increased survival of new

neurons generated after CCI and not exclusively due to increased likelihood of neuronal

differentiation. Additional work is needed to determine if IGF-1 reduces death of newborn

neurons, using markers of neurodegeneration, apoptosis, and necrosis, in the post-

traumatic immature neuron recovery phase between 7 and 10 d following severe CCI.

Future studies will also need to elucidate if overexpression of IGF-1 promotes long-term

survival and maturation of newborn neurons generated after brain injury.

Despite the recovery of immature neuron density in WT mice at 10 d post-injury,

we demonstrate that the immature neurons in the injured granular of WT mice exhibit

significant reductions in dendritic complexity following CCI. Recent studies highlight that

Page 204: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

191

brain injury results in damage to the dendritic processes of neocortical and hippocampal

mature granular neurons after CCI (Gao et al. 2011), but changes in immature neuron

dendritic morphology after brain injury have not been evaluated. Immature neurons from

WT mice exhibited reduced total dendrite length and arbor complexity after injury,

however, immature neurons from brain-injured IGF-1 overexpressing mice exhibited

significant restoration of dendritic morphology that was equivalent to the morphology

observed in sham-injured mice. There are two possible explanations for the IGF-1-

mediated restoration of dendritic arbor complexity after severe CCI. IGF-1 protects

against injury-induced damage to immature neuron dendritic processes or IGF-1

promotes the more complete formation of dendritic arbors of immature neurons during

the process of neuronal differentiation and maturation. At 3 d following CCI, both WT

and IGF-1 overexpressing mice exhibited a visible reduction in dendritic arbor

complexity, suggesting that IGF-1 did not protect against injury-induced damage.

However, future studies will need to quantify dendritic arbor complexity at 3 d following

CCI to definitively determine if IGF-1 protects against reductions in dendritic arbor

complexity in immature neurons after brain injury.

Alternatively, overexpression of IGF-1 may have enhanced the growth of

dendritic arbors in newly generated immature neurons after brain injury. Reductions in

levels of endogenous IGF-1 are associated with reduced dendritic growth and branching

(Cheng et al. 2003). Conversely, IGF-1 stimulates dendritic growth and enhances

branching of pyramidal neurons in organotypic primary cortical slice cultures and neurite

outgrowth in dissociated neurons (Torres-Aleman et al. 1989; Niblock et al. 2000).

Knockout of PTEN, the negative regulator of phosphatidylinositol-3-kinase signaling,

resulted in prolonged Akt activation and produced immature granular neurons with

elongated dendrites and increased arborization (Amiri et al. 2012). Taken together,

Page 205: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

192

these studies suggest that the re-establishment of sham injury-like dendritic morphology

in IGF-1 overexpressing brain-injured mice may be the result of enhanced dendritic

growth within newborn neurons. Reductions in mature neuron dendritic arbor complexity

and synaptic spine density likely contribute to reduced neuronal excitability of dentate

gyrus granular cells after CCI (Gao et al. 2011). Decreases in the length and complexity

of dendritic arbors of immature neurons may contribute to reduced synaptic connections

in the molecular layer and lead to functional impairments in long-term potentiation in the

hippocampus after CCI. Restoration of normal dendritic morphology in immature

neurons may have important functional implications in promoting recovery of function in

the hippocampus and contribute to improved cognitive function after TBI. While this

study did not evaluate the precise mechanism by which IGF-1 overexpression improves

dendritic morphology after brain injury, these data reveal a novel pathological

phenomenon and identify a new therapeutic target in experimental TBI in a population of

cells critical for learning and memory. A better understanding of the time course of

dendritic process recovery may provide better insight into the regenerative capacity of

dendritic arbors in the injured brain and help develop effective therapeutic strategies,

including IGF-1, that promote structural plasticity in the injured brain.

In summary, we demonstrate that targeted overexpression of IGF-1 does not

attenuate early loss of hippocampal immature neurons in the SGZ after severe CCI, but

promotes a robust recovery of the immature neuron population, most likely by enhancing

neuronal differentiation of hippocampal progenitors. We also establish that IGF-1

overexpression promotes a more complete recovery of the dendritic arbor of newborn

neurons after trauma. The data presented in this chapter provides additional evidence

for IGF-1 as a promising therapeutic agent that promotes post-traumatic neurogenesis

and structural plasticity in the hippocampus following experimental TBI.

Page 206: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

193

Copyright © Shaun William Carlson 2013

Page 207: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

194

Chapter 6: Summary of Dissertation

The major findings in this dissertation provide evidence for the therapeutic

efficacy of insulin-like growth factor-1 (IGF-1) in attenuating motor and cognitive

dysfunction and enhancing post-traumatic neurogenesis in a mouse model of contusion

traumatic brain injury (TBI). In Chapter 2, we demonstrated that a modification to the

geometry of the impactor tip utilized in the well-established controlled cortical impact

(CCI) injury model modulated the rate of acute cortical cell loss and extent of acute

regional hippocampal neurodegeneration, without altering other important pathological

features of contusion brain injury. In Chapter 3, we showed that prolonged continuous

systemic infusion of IGF-1 promoted the activation of Akt in the contused brain and

modestly reduced hippocampal neurodegeneration in the granular layer, but did not

attenuate motor dysfunction in mice subjected to severe CCI. In Chapter 4, we

demonstrated that prolonged intracerebroventricular infusion of IGF-1 significantly

improved motor function and reduced cognitive impairment associated with severe CCI.

We also highlighted that central infusion of IGF-1 increased immature neuron density in

a number of infusion paradigms, and this increase may be driven by enhanced post-

traumatic neurogenesis following brain injury. In Chapter 5, we established that IGF-1-

mediated increases in hippocampal immature neuron density occurred by enhanced

neuronal differentiation of newly proliferated cells in the dentate gyrus granular layer

after CCI. We also showed that IGF-1 attenuated a reduction in dendritic arbor

complexity of immature neurons following CCI, a previously unknown pathological

phenomenon in immature neurons. We hypothesized that treatment with IGF-1

attenuates motor and cognitive impairments and enhances hippocampal neurogenesis in

the mouse brain following brain injury. Taken together, the findings reported in this

dissertation supported our hypothesis and demonstrated that treatment with IGF-1

Page 208: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

195

improved neurobehavioral function and enhanced post-traumatic neurogenesis following

severe CCI in mice.

Controlled Cortical Impact and Impactor Tip Geometry

CCI is advantageous for studies evaluating mechanisms of injury or therapeutic

interventions of TBI, as the model is highly reproducible, well-characterized, and

produces pathology, including neurological and cognitive impairments (Dixon et al. 1991;

Hamm et al. 1992; Smith et al. 1995; Saatman et al. 2006), reflective of contusion injury

observed in brain-injured patients (Levin 1998; Serra-Grabulosa et al. 2005; Alahmadi et

al. 2010). Results from finite element mathematic models indicated that the geometrical

shape of the impactor tip can influence maximal tissue strain during the initial tissue

deformation and that an impactor tip with rounded shape reduced the predicted maximal

principal strains compared to an a beveled shape (Mao et al. 2011; Pleasant et al.

2011). Data from Chapter 2 demonstrated that use of a rounded impactor tip, as

compared to a beveled impactor tip, slowed the progression of cortical cell loss and

reduced acute hippocampal neurodegeneration in CA1 without altering other key

aspects of contusion injury, including axonal injury, breakdown of the blood-brain barrier,

and motor and cognitive impairment after severe CCI (Pleasant et al. 2011). The

accelerated rate of cortical cell loss within 4 hr of CCI with a beveled tip creates difficult

technical and experimental constraints for studies in which elucidation of acute injury

mechanisms is a primary outcome measure. The extent or rate of neurodegeneration in

the granular layer or CA3 regions of the hippocampus was not greatly modulated by

geometry of the impactor tip. This is an important finding and suggests that evaluation of

hippocampal neurodegeneration in these regions can be compared between studies

utilizing different impactor tip shapes.

Page 209: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

196

The slowed progression of tissue damage after impact with a rounded impactor

tip more closely recapitulates the progression of injury pathology observed by

radiological scans of patients following a contusion TBI (Alahmadi et al. 2010; Kurland et

al. 2012). Furthermore, the average treatment onset of therapeutic agents in TBI clinical

trials occurs between 3 and 8 hr post-injury (Maas et al. 2007). Considering the delay of

therapeutic intervention in the clinical setting, utilization of the rounded impactor tip may

provide a more clinically relevant and experimentally feasible evaluation of therapeutic

agents intended to reduce acute secondary injury and promote cell survival. Use of a

rounded impactor tip may also be critically important for therapeutic studies that aim to

initiate delivery of therapeutic agents in a clinically relevant treatment window. Our

findings in Chapter 2 highlight that the parameter of impactor tip geometry, in addition to

impact depth and velocity, should be considered as injury parameters that can modify

contusion impact pathology. The rounded impactor tip was therefore used in all studies

to evaluate the efficacy of IGF-1 to attenuate neurobehavioral dysfunction and enhance

hippocampal post-traumatic neurogenesis after TBI.

Levels of Human IGF-1 in the Brain and Systemic Circulation after CCI

The long-term goal of this dissertation work is to provide additional insight and

data that contributes to a preclinical evaluation of IGF-1 as a therapeutic strategy for the

treatment of TBI. In this dissertation research, we utilize multiple therapeutic paradigms

including prolonged systemic and central infusions and targeted astrocyte-specific

overexpression of IGF-1 to facilitate delivery of IGF-1 to the injured brain. In order to

compare effects of IGF-1 across these treatment strategies, we needed to establish the

levels of human IGF-1 (hIGF-1) in the injured brain.

Page 210: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

197

Findings from a Phase II open-label, prospective randomized clinical trial showed

that brain-injured patients treated with 0.01 mg/kg/hr IGF-1 by intravenous infusion who

achieved plasma levels of IGF-1 greater than 350 ng/mL showed improved metabolic

health and Glasgow Outcome Scale scores at 6 months post-injury (Hatton et al. 1997),

suggesting that systemic levels of IGF-1 need to exceed 350 ng/mL to improve outcome

after brain injury. In Chapter 3, we demonstrated that prolonged systemic infusion of 4

mg/kg/d hIGF-1 resulted in sustained levels of approximately 150 ng/mL of hIGF-1 in

systemic circulation over 1 week, but did not produce detectable levels of hIGF-1 in the

brain. The endogenous systemic circulating levels of IGF-1 in C57/BL6 naïve mice are

reported to be approximately 250 ng/mL (Yuan R 2013), suggesting that systemic

infusion of hIGF-1 resulted in total serum levels of IGF-1, including endogenous and

exogenous, above the therapeutic threshold of 350 ng/mL. We did not quantify

endogenous levels of IGF-1, warranting future studies to evaluate endogenous levels of

IGF-1 to ensure that continuous infusion of hIGF-1 did not reduce production of IGF-1.

Our inability to detect hIGF-1 in the brain following systemic infusion may be reflective of

several possible explanations, including limited penetrance of hIGF-1 into the brain as a

result of sealing of the blood-brain barrier at the time point in which hIGF-1 was

quantified, a short half-life of IGF-1 in the brain, or limited sensitivity of the ELISA kit that

was utilized. Additional studies may need to evaluate the penetrance and residence time

of systemically infused IGF-1 in the brain by additional techniques, including

radiolabeled or tagged IGF-1 that would afford increased sensitivity in detecting low

levels of IGF-1 in the brain parenchyma.

In Chapter 4 we show that supplementation of hIGF-1 resulted in quantifiable

increases in the levels of hIGF-1 in the cortex and hippocampus. Following central

infusion of 10 µg/d hIGF-1, we measured approximately 30 to 50 ng/mL hIGF-1 in both

Page 211: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

198

the injured cortex and hippocampus, two of the most vulnerable regions following severe

CCI. Previous work from our lab demonstrated that targeted astrocyte-specific

overexpression of IGF-1 resulted in progressively increasing levels of hIGF-1 that

reached 15 ng/mL hIGF-1 in the hippocampus at 72 hr post-injury and promoted

improved neurobehavioral function at 7 d following severe CCI (Madathil 2013),

suggesting that levels of 15 ng/mL hIGF-1 or greater in the brain can promote functional

recovery after brain injury. We measured levels of hIGF-1 2 to 3-fold higher in the brain

following central infusion, demonstrating that we achieved a therapeutic level of IGF-1 in

the injured brain. In Chapter 5, we utilized the aforementioned targeted astrocyte-

specific overexpression to elevate levels of hIGF-1 in areas exhibiting reactive

astrocytosis following brain injury. Currently, we have not fully examined the time course

of elevation of hIGF-1 following CCI for either central infusion or overexpression of hIGF-

1. Additional work is needed to elucidate the temporal profile of increasing levels of

hIGF-1 in the brain as this has important implications for the efficacy of IGF-1 to protect

against acute cell loss after TBI. In Chapter 4, we also demonstrated that prolonged

central infusion of 10 µg/d hIGF-1 into the lateral ventricle resulted in approximately 60-

80 ng/mL of hIGF-1 in systemic circulation at 7 d after the initiation of infusion. While it is

known that IGF-1 is rapidly cleared from the brain and enters systemic circulation

following intracerebroventricular injection, we were surprised to observe that central

infusion of IGF-1 resulted in relatively similar systemic circulation concentrations as

observed after systemic infusion. These findings argue that additional work may be

needed to better understand the interplay between the levels of IGF-1 in systemic

circulation and the brain following prolonged infusion. We did not measure the

concentrations of hIGF-1 in serum collected from IGF-1 overexpressing mice, but our

findings argue that future studies should incorporate this as an additional assessment of

Page 212: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

199

IGF-1 overexpression. The findings in this dissertation provide an important first step in

understanding of how plasma levels of IGF-1 may be used as a clinical indicator of

levels of IGF-1 in the brain parenchyma of TBI patients.

IGF-1 Signaling in the Brain after CCI

In light of our findings, we sought to determine if elevated levels of hIGF-1

resulted in increased activation of Akt, a potent pro-survival downstream mediator of

IGF-1 signaling. In Chapters 3 and 4 we demonstrated that both systemic and central

infusion of exogenous IGF-1 promoted activation of Akt in the brain. Previous work from

our lab demonstrated that targeted overexpression of IGF-1 significantly increased Akt

activation in the hippocampus at both 24 and 72 hr following severe CCI (Madathil

2013). Taken together, these studies show that supplementation of IGF-1, independent

of treatment strategy, significantly increased Akt activity in the brain after severe CCI.

These findings highlight the ability of IGF-1 to promote protective signaling in the brain,

and strongly support the development of IGF-1 as a therapeutic strategy across the

continuum of severity of TBI.

Capacity of IGF-1 to Promote Survival of Hippocampal Immature Neurons

Following moderate or severe CCI, a precipitous drop in hippocampal immature

neuron density occurs within 24 hr and continues for days following injury (Rola et al.

2006; Gao et al. 2008; Yu et al. 2008). Consistent with published reports of immature

neuron susceptibility after TBI, we demonstrated at 3 d (Chapter 3 and 5) and at 7 d

(Chapter 4) that brain-injured vehicle-treated mice exhibited significant reductions in

immature neuron density at 7 d post-injury.

Page 213: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

200

Increases in IGF-1 signaling are associated with improved neuron survival both

in vitro and in vivo (Guan et al. 1993; Russell et al. 1998; Chrysis et al. 2001; Zheng et

al. 2002; Bendall et al. 2007). Targeted astrocyte-specific overexpression of IGF-1

reduced the number of Fluorojade-C-positive cells, indicative of degenerating neurons,

in the dentate gyrus granular layer, but not CA3 at 3 d following severe CCI (Madathil

2013). In Chapter 3, we observed a modest decrease in the number of Fluorojade-B

positive cells in the dentate gyrus of mice systemically infused with IGF-1 after severe

CCI. Considering the neurodegenerative marker Fluorojade-B is found to colocalize with

doublecortin, a marker of immature neurons, in the granular layer at 24 hr after CCI (Gao

et al. 2008), we postulated that the reduced number of degenerating neurons observed

after systemic infusion or overexpression of hIGF-1 was reflective of improved immature

neuron survival. However, neither systemic infusion of hIGF-1 (Chapter 3) or astrocyte-

driven IGF-1 overexpression (Chapter 5) increased immature neuron density at 3 d

following severe CCI, suggesting that the reduced number of degenerating neurons

likely reflects improved survival of mature neurons and not immature neurons. Additional

studies are required to quantify the frequency that immature and mature neurons are

found to positively label with markers of neurodegeneration following treatment with IGF-

1 after brain injury. To the best of our knowledge, no studies have reported that

immature neuron loss can be attenuated by therapeutic interventions within the first 3 d

following CCI. Together, our findings suggest that IGF-1 provides limited protection

against acute loss of immature neurons following severe CCI.

Susceptibility of Immature Neurons after CCI

Immature neurons are particularly vulnerable to brain injury, but the mechanism,

including the mode of cell death, underlying this susceptibility is unknown. If immature

Page 214: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

201

neurons are found to die predominately from necrotic cell death, this may help explain

why IGF-1, a potent anti-apoptotic signal, did not protect against acute immature neuron

loss after severe CCI. Rola et al (2006) previously demonstrated that cells positively-

labeled with the cell death marker TUNEL (terminal deoxynucleotidly transferase dUTP

nick end labeling) were predominantly observed in the subgranular zone, within the 2 d

following severe CCI. TUNEL is a marker of DNA damage and may be reflective of

either apoptotic or necrotic cellular death (de Torres et al. 1997). Caspase-3, a marker of

apoptosis, was rarely observed in the granular layer at 4 hr, 1, 3, or 7 d following CCI,

but receptor interacting protein-1 (RIP-1), a marker of necrosis, was observed at 1 d

after CCI and was also found to colocalize with Fluorojade-B (Zhou et al. 2012). These

observations suggest that degenerating neurons in the granular layer may predominantly

die by necrosis and not apoptosis. Additional work is needed to colocalize indicators of

apoptosis and necrosis with markers of immature neurons to quantify and delineate the

cell death mechanisms in this subpopulation of neurons, and to elucidate if the

mechanisms of cell death change as a function of time following CCI.

Calpain-mediated Proteolysis in the Hippocampal Neurogenic Niche

Necrotic cell death of immature neurons within 24 hr following brain injury may be

related to calcium dysregulation and the acute activation of calcium mediated proteases,

including calpain. Cultured primary hippocampal neurons subjected to mechanical

stretch injury show increased membrane permeability and a rapid elevation in

concentrations of intracellular calcium within seconds that is sustained for minutes after

the initial stretch (Geddes et al. 2003). While regulated cellular microdomains of elevated

calcium are critical for normal cellular function, the increases in intracellular calcium

observed post-stretch can result in the pathological activation of proteases (Pike et al.

Page 215: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

202

2000). Increased calpain-mediated and caspase-meditated breakdown of α-spectrin, a

cytoskeletal protein, in hippocampal cell cultures occurs within hours following stretch

injury (Pike et al. 2000; DeRidder et al. 2006).

Considering the selective vulnerability of immature neurons after brain injury, we

sought to determine if calpains were activated in immature neurons. Calpain-mediated

cleavage of spectrin can be visualized by Ab37 immunoreactivity; as the antibody

recognizes the COOH-terminus of the NH2-terminal fragment generated during calpain

cleavage of spectrin (Saatman et al. 2000). In a preliminary study, in a small number of

mice (n=3) at 24 hr following severe CCI, Ab37 immunoreactivity in the subgranular zone

did not colocalize with DCx (Fig. 6.1A), but instead was found to colocalize with nestin, a

marker of radial stem cells and astrocytes within the subgranular zone (Fig. 6.1B). The

cellular morphology and location of nestin in these images is suggestive of neural

progenitor cells; however, based on these parameters alone we cannot eliminate the

possibility that Ab37 is also present in astrocytes within the subgranular zone. These

images suggest that calpain-mediated spectrin cleavage occurs in neural progenitor

cells, and not immature neurons. This observation argues that activation of calpains may

not be related to the loss of immature neurons, but may contribute to post-traumatic

changes in neural progenitor cells in the subgranular zone of the hippocampus.

Calpain has been identified as a mediator of cellular migration and structural

plasticity (Huttenlocher et al. 1997; Zadran et al. 2010), and may be important for neural

progenitor proliferation, differentiation, migration, and regulation of the neurogenic niche

(Santos et al. 2012). Alternatively, the presence of calpain-mediated spectrin breakdown

in nestin-positive cells could be a pathological response to injury in the niche. Following

moderate CCI, the number of quiescent progenitor cells increases robustly between 4

and 72 hr post-injury in the subgranular zone (Gao et al. 2009), which likely contributes

Page 216: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

203

to increased neurogenesis following CCI. The acute increase in proliferation at 4 hr post-

injury is consistent with the timing of injury-induced activation of calpain (Hall et al.

2005), suggesting that pathological activation of calpains could be involved in injury-

induced responses of the neurogenic niche. While these findings are preliminary

observations, they propose intriguing questions about the role of calpain-mediated

proteolysis in the neurogenic niche after brain injury.

Page 217: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

204

Fig. 6.1: Calpain-mediated spectrin cleavage was observed in nestin-positive, but not DCx-positive cells in the granular layer at 24 hr following controlled cortical impact (CCI). (A) A marker of immature neurons (DCx, green) did not colocalize with a marker of calpain-mediated spectrin cleavage (Ab37, red) in the dentate gyrus inner granular layer at 24 hr following severe CCI. (B) Nestin (red) a marker of neural progenitor cells and astrocytes in the dentate gyrus granular layer, colocalized with a marker of calpain-mediated spectrin cleavage (Ab37, green) at 24 hr after severe CCI. Granular layer (GL) and hilus (H). Scale bar represents 50 µm.

Page 218: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

205

Recovery of Hippocampal Immature Neurons after CCI

In Chapter 4, we observed a significant reduction in immature neuron density in

brain-injured mice treated with vehicle compared to sham-injured mice at 7 d post-injury.

This finding is consistent with the observation that mice subjected to severe CCI exhibit

maximal immature neuron loss at 7 d post-injury (Rola et al. 2006). Subsequent to this

initial reduction, immature neuron numbers recover towards baseline in the days to

weeks following CCI (Rola et al. 2006; Yu et al. 2008). Consistent with post-traumatic

recovery of immature neurons, in Chapter 5 we showed that wildtype mice subjected to

severe CCI exhibit immature neuron density similar to sham-injured mice at 10 d post-

injury. Considering we observed a marked reduction in immature neuron density in

wildtype mice at 3 d post-injury, it is likely that increased post-traumatic neurogenesis,

including increased cellular proliferation and neuronal differentiation, contributed to the

recovery of immature neuron numbers in wildtype mice after severe CCI.

IGF-1 is a potent promoter of neurogenesis by increasing cellular proliferation

and enhancing neuronal differentiation of newly proliferated cells (Ye et al. 1996;

Arsenijevic and Weiss 1998; Aberg et al. 2000; Brooker et al. 2000; Arsenijevic et al.

2001; Yan et al. 2006). We postulated that the neurogenic properties of IGF-1 would

enhance post-traumatic neurogenesis and promote the recovery of immature neuron

numbers in the dentate gyrus granular layer after severe CCI. In Chapter 4 we found that

central infusion of 10 μg/d and 3 μg/d of hIGF-1 resulted in a significant increase in

immature neuron density in brain-injured mice compared to treatment with vehicle, and

that the density of immature neurons was equivalent to sham-injured mice treated with

IGF-1. We showed that increasing concentrations of centrally infused IGF-1 resulted in

proportional elevations in immature neuron density following severe CCI. Importantly,

delayed central infusion of IGF-1 with a clinically relevant 6 hr post-injury onset retained

Page 219: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

206

the significantly increased immature neuron density that was previously observed using

an acute 15 minute post-injury treatment onset. We also found in Chapter 5 that targeted

overexpression of IGF-1 significantly enhanced the recovery of immature neuron density

in brain-injured mice, exceeding the immature neuron density of IGF-1 overexpressing

sham-injured mice. Findings from Chapter 4 and 5 highlight that elevated levels of IGF-1

in the brain are associated with increased immature neuron density following brain

injury. Additional work is needed to determine if systemically infused IGF-1 promotes the

recovery of immature neuron numbers after severe CCI or if our observations from

Chapters 4 and 5 are exclusive to central infusion of IGF-1. Taken together, our findings

strongly support that IGF-1 enhances post-traumatic neurogenesis which may be the

result of increased cellular proliferation or enhanced neuronal differentiation of newly

proliferated cells in the granular layer after severe CCI.

Proliferative Response of the Neurogenic Niche with Treatment of IGF-1 after CCI

Several reports demonstrate that brain injury results in cellular proliferation in the

injured dentate gyrus granular layer within 24 hr which is sustained for days following

experimental TBI (Dash et al. 2001; Kernie et al. 2001; Sun et al. 2005; Rola et al.

2006). In Chapters 4 and 5 we observed significant increases in hippocampal cellular

proliferation in mice subjected to severe CCI compared to mice subjected to sham injury.

Central infusion of IGF-1 appeared to augment brain injury-induced cellular proliferation,

but this effect was not statistically significant (Chapter 4). Overexpression of IGF-1 did

not enhance cellular proliferation at 3 d post-injury, a time of increased post-traumatic

proliferation (Dash et al. 2001; Rola et al. 2006; Gao et al. 2009; Gao and Chen 2013),

or cumulatively over the first 7 d following CCI (Chapter 5).

Page 220: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

207

While our data provide strong evidence that IGF-1 does not significantly

modulate cellular proliferation in the injured brain, we need to acknowledge that bromo-

deoxyuridine (BrdU) incorporation may not be reflective of only cellular proliferation. The

dose of BrdU, number of injections, duration between injections, and euthanasia time

after last dose can introduce aspects of cellular survival, and ongoing cellular

proliferation that can dilute BrdU staining in neural progenitor cells or proliferating

astrocytes (Taupin 2007), thereby complicating interpretation. Several studies evaluating

proliferation after experimental TBI euthanize the animal between 4 and 24 hr following a

single injection of BrdU to evaluate cellular proliferation (Sun et al. 2005; Gao and Chen

2013). A single BrdU injection with 4 hr euthanasia post-injection was utilized in Chapter

5 to evaluate cellular proliferation. Injection paradigms that utilize short durations

between the last injection and the time of euthanasia reduce, but do not fully eliminate,

aspects of cellular survival, particularly during times of widespread cell loss after severe

CCI. However, euthanasia within hours of the BrdU injection does not provide insight

into neuronal differentiation. Alternatively, repeated injections, as utilized in Chapters 4

and 5 provide insight into IGF-1-mediated changes in cellular differentiation.

IGF-1-mediated Enhancement of Neuronal Differentiation after CCI

Genetic ablation of immature neurons is associated with learning and memory

impairments (Clelland et al. 2009; Deng et al. 2009; Jessberger et al. 2009), suggesting

that the profound loss of immature neurons may contribute to cognitive impairments

observed after brain injury, and that enhanced recovery of immature neurons after CCI

could contribute to improvements in cognitive function. In addition to promoting cellular

proliferation, IGF-1 has also been shown to be a potent promoter of neuronal

differentiation of newly proliferated cells (Arsenijevic and Weiss 1998; Aberg et al. 2000;

Page 221: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

208

Brooker et al. 2000; Arsenijevic et al. 2001). In Chapter 4, we demonstrated that central

infusion of hIGF-1 promoted the generation of newborn neurons, which likely contributed

to the recovery of total immature neuron density to near control (sham-injured) levels.

Similarly, in Chapter 5 we demonstrated that hIGF-1 overexpression promoted the

generation of newborn neurons above sham-injured levels after CCI. Taken together, the

data from our two treatment paradigms illustrates that IGF-1-mediated neuronal

differentiation is likely a predominant factor in promoting the recovery and enhancement

of immature neuron density in the hippocampus after severe CCI. However, it is also

possible that IGF-1 promotes the survival of newly differentiation neurons after injury that

would increase newborn neuron density independent of enhanced differentiation.

Additional studies are needed to delineate these two possibilities to determine if the IGF-

1-mediated increase in newborn neurons is reflective of enhanced neuronal

differentiation, increased immature neuron survival, or potentially a combination of both

possibilities.

The findings in this dissertation show that IGF-1 resulted in increased post-

traumatic neurogenesis in the subgranular zone of the hippocampus. However, we did

not evaluate the efficacy of IGF-1 to enhance neurogenesis in the subventricular zone

(SVZ), a second site of ongoing neurogenesis in the adult brain. Several studies have

demonstrated that experimental TBI results in increased cellular proliferation and may

promote the generation of newborn neurons in the subventricular zone (Kernie et al.

2001; Rice et al. 2003; Ramaswamy et al. 2005; Sun et al. 2009). Central infusion of

either basic fibroblast growth factor or S100B after fluid percussion injury results in

increased cellular proliferation (Kleindienst et al. 2005; Sun et al. 2009), suggesting that

administration of therapeutic agents can enhance post-traumatic neurogenesis in the

SVZ. Preliminary findings from our lab suggest that IGF-1 may enhance the generation

Page 222: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

209

of newborn neurons in the SVZ (Kizhakke Madathil S 2010), but additional work is

needed to determine if IGF-1 promotes neurogenesis in the SVZ after experimental TBI.

A handful of studies highlight that newly proliferated cells can express markers of

neurons, astrocytes, oligodendrocytes, and microglia after experimental TBI (Sun et al.

2005; Rola et al. 2006; Sun et al. 2009). While the observations discussed in this

dissertation have been focused on the generation of newborn neurons after brain injury,

it is imperative to understand the potential implications of other cell types generated in

injured areas of the brain following treatment of IGF-1, considering glial cells likely

contribute to recovery after brain injury (Myer et al. 2006). The potential for the

generation of new oligodendrocytes with treatment of IGF-1 after TBI will be discussed

later in this chapter.

In only the last couple years, the methodology for assessing changes in

neurogenesis has been revolutionized to incorporate additional markers of neural

progenitor cells and neuroblasts in an effort to understand the complex hierarchy of

progenitor cells in the neurogenic niche of the adult brain. Recent work by Gao et al

(2013, 2009) illustrates that future studies evaluating post-traumatic neurogenesis may

need to incorporate confocal microscopy and detailed assessments of cellular

morphology to investigate aspects of neural progenitor cell proliferation and cellular

differentiation in the subgranular zone after experimental TBI. Considering the ongoing

debate of neural progenitor self-renewal and potential differences in multi-lineage

differentiation (Lugert et al. 2010; Ming and Song 2011), we need to strive for a more

comprehensive evaluation of the responses of neural progenitor cells to therapeutic

agents. This may provide valuable insight into understanding how therapeutic agents

can modulate the neurogenic niche and be used to target enhanced differentiation of

specific cells types, i.e. neurons or oligodendrocytes, in the neurogenic niche after TBI.

Page 223: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

210

Structural Plasticity with Treatment of IGF-1 after CCI

Formation and extension of dendritic processes is a hallmark morphological

feature of immature neuron maturation (Zhao et al. 2006; Deng et al. 2010). In Chapter

5, we demonstrate for the first time that immature neurons exhibit reduced dendritic

arbor complexity after brain injury. Moreover, we demonstrate that elevated levels of

IGF-1 in the brain restore dendritic arbor complexity to control (uninjured) levels

following severe CCI. Future studies need to evaluate the time course of reductions and

the potential endogenous recovery of dendritic arbor complexity in immature neurons

following severe CCI. Additional work is also needed to elucidate if IGF-1 protects

against acute reductions in dendritic arbor complexity or if IGF-1 promotes the formation

of dendrites in immature neurons generated after brain injury. Enhanced growth of new

dendrites is plausible as IGF-1 promotes dendritic growth and arborization during

development of neurons in the cerebellum and cerebral cortex (Niblock et al. 2000;

Ozdinler and Macklis 2006). Protection or enhanced formation of dendritic arbor

complexity following treatment with IGF-1 may have important functional implications in

promoting recovery in the injured hippocampus.

Reductions in dendritic arbor complexity may have functional consequences in

and potentiate the manifestation of neurobehavioral deficits after brain injury. Reductions

in dendritic length, as observed in brain-injured wildtype mice in Chapter 5, may reduce

the number of synaptic connections of immature neurons with axonal projections from

the entorhinal cortex. Following moderate CCI, mature granular neurons in the

hippocampal dentate gyrus exhibit a robust reduction in dendritic arbor complexity and

synaptic spine density, likely contributing to their decreased neuronal excitability at 3 d

post-injury (Gao et al. 2011). The juxtaposition of immature and mature neurons in the

dentate gyrus granular layer suggests that following CCI, immature neurons could also

Page 224: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

211

exhibit reductions in synaptic density of the remaining dendritic processes. CCI has also

been shown to result in a marked reduction in synaptic number at 2 d post-injury (Scheff

et al. 2005). Multiple studies demonstrate that functional deficits in hippocampal long-

term potentiation occur within hours and can lasting at least two weeks following brain

injury (Miyazaki et al. 1992; Reeves et al. 1995; Albensi et al. 2000; Schwarzbach et al.

2006; Atkins 2011). The hippocampus may possess capabilities of endogenous

recovery, as suggested by a slow recovery of synapse strength in CA1 within 14 d post-

injury after an initial deficit at 2 d post-injury (Norris and Scheff 2009). However, CCI is

associated with cognitive dysfunction that persists for at least weeks post injury (Scheff

et al. 1997; Dixon et al. 1999), suggesting that recovery of dendritic arborization, in

addition to recovery of synaptic number and strength, in the dentate gyrus may be

necessary to help promote recovery of cognitive function after TBI.

Synaptogenesis is another defining property of immature neuron maturation.

IGF-1 overexpression promotes synaptogenesis in dendritic processes of hippocampal

granular layer neurons in the brains of postnatal mice (O'Kusky et al. 2000). Increased

activation of Akt has been implicated as a crucial event in synapse plasticity and

maintenance of late-phase long-term potentiation (Horwood et al. 2006; Karpova et al.

2006). In light of our observations of reduced immature neuron dendritic arborization

complexity at 10 d following CCI, future studies need to evaluate if brain injury reduces

synaptic density in immature neurons following severe CCI. Moreover, in the event that

the reduction in synaptic density is observed, future studies will also need to evaluate

the efficacy of IGF-1 to protect against synapse loss or promote synaptogenesis in

immature neurons after experimental TBI.

Page 225: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

212

While IGF-1-mediated hippocampal plasticity may contribute to improved

recovery after brain injury, excessive plasticity could result in aberrant sprouting, altered

hippocampal function, or potentiate the development of maladaptive conditions, such as

post-traumatic epilepsy. Following a 1 mm CCI, aberrant mossy fiber sprouting was

observed in the inner molecular layer between 6 and 10 weeks after injury, a time in

which increased spontaneous seizures were also observed (Hunt et al. 2009).

Hippocampal granular neuron excitability is also increased as a result of reduced

inhibitory synaptic input from hilar GABAergic interneurons following brain injury (Hunt et

al. 2011). The formation of basal dendrites of mature and more recently immature

granular neurons has also been implicated as a precipitating feature of aberrant

sprouting and the development of seizure activity after pilocarpine-induced status

epilepticus (Parent et al. 1997; Spigelman et al. 1998; Scharfman et al. 2000; Dashtipour

et al. 2003; Walter et al. 2007). These studies suggest that enhanced outgrowth of

dendrites may facilitate the development of epilepsy after contusion injury. In our

studies, treatment with IGF-1 restored dendritic complexity to the morphology observed

with sham injury, suggesting that IGF-1 did not promote aberrant sprouting. Moreover,

we rarely observed basal dendrites on immature neurons, and the isolated observations

of basal dendrites may reflect a normal and transient structural property of immature

neurons (Jones et al. 2003; Ribak et al. 2004). While we did not observe indications of

aberrant sprouting at 10 d post-injury, future studies need to evaluate if immature

neurons begin to show indications of aberrant sprouting in the weeks following injury,

particularly during 6 to 10 weeks post-injury in which spontaneous seizures are observed

after severe CCI (Hunt et al. 2009). This additional work will be imperative to elucidate if

IGF-1 promotes appropriate and long-term plasticity in the injured hippocampus after

CCI.

Page 226: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

213

Improvements in Motor Function with Treatment of IGF-1 after CCI

Systemic administration of IGF-1 has been shown to promote delayed recovery

of motor function in rats following moderate fluid percussion injury (Saatman et al. 1997).

Using either repeated injections or continuous infusion, IGF-1 significantly improved

motor function as early as 5 days or as late as 2 weeks post-injury (Saatman et al.

1997). In Chapter 4 we showed that central infusion of IGF-1 significantly attenuated

motor dysfunction at 1, 2, 3, and 5 d following severe CCI. These data corroborate

previous findings from our lab demonstrating that targeted overexpression of IGF-1

significantly attenuated motor dysfunction at 1, 2, 3, and 4 d following severe CCI

(Madathil 2013). IGF-1 overexpressing mice and mice centrally infused with IGF-1

exhibited significantly improved motor function by 1 d following severe CCI. Previous

work from our lab showed that IGF-1 promoted delayed recovery of motor function in

rats systemically treated with IGF-1 (Saatman et al. 1997). In Chapter 3 we showed that

systemic infusion of 4 mg/kg/d hIGF-1, the same dose previously used by Saatman et al.

(1997), did not result in measureable levels of hIGF-1 in the brain and did not promote

acute recovery of motor function. Elevated levels of IGF-1 in the brain, as observed after

central infusion and overexpression of IGF-1, may be needed to promote acute recovery

of motor function in addition to the delayed recovery of motor function observed after

systemic infusion (Saatman et al. 1997). In Chapter 3 we were unable to assess delayed

recovery of motor function as CCI in the mouse results in more transient motor deficits

with nearly complete recovery of motor function by 7 d post-injury, which is not observed

after lateral fluid percussion in the rat (Saatman et al. 1997). Future studies will need to

incorporate more sensitive tests or other models of experimental TBI with more

persistent motor deficits to evaluate the efficacy of IGF-1 to promote delayed recovery of

motor function.

Page 227: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

214

Improvements in Cognitive Function with Treatment of IGF-1 after CCI

Lasting cognitive dysfunction is reported in brain-injured patients (Levin 1998;

Lundin et al. 2006; Jang 2009) and has been observed for up to one year following CCI

(Smith et al. 1997; Dixon et al. 1999). The Morris water maze is a frequently utilized tool

to evaluate learning and memory dysfunction after CCI (Hamm et al. 1992; Scheff et al.

1997; Dixon et al. 1999; Saatman et al. 2006; Pleasant et al. 2011). The Morris water

maze cognitive task has more recently been utilized to demonstrate that reductions in

the number of immature neurons results in significant impairments in learning and

memory (Clelland et al. 2009; Deng et al. 2009; Jessberger et al. 2009). Moreover,

prevention of the endogenous recovery of immature neurons after TBI results in

impaired learning of the platform location in the Morris water maze task (Blaiss et al.

2011), highlighting the importance of post-traumatic neurogenesis in cognitive function

following CCI. The novel object recognition task is also utilized to assess cognitive

function following experimental TBI (Pullela et al. 2006; Tsenter et al. 2008; Schoch et

al. 2012; Prins et al. 2013). In Chapter 4 we showed that mice subjected to severe CCI

exhibited significant cognitive impairment compared to mice subjected to sham injury, as

assessed by the novel object recognition task at 7 d post-injury.

Several studies have demonstrated that treatment with IGF-1 promotes cognitive

improvement after experimental TBI (Saatman et al. 1997; Rubovitch et al. 2010;

Madathil 2013). Supplementation of IGF-1 in mice significantly attenuated brain injury-

induced cognitive dysfunction in a Y maze at 7 d following mild TBI (Rubovitch et al.

2010) or in a novel object recognition task at 7 d following severe CCI (Madathil 2013).

Systemic infusion of IGF-1 significantly improved spatial learning and memory in the

Morris water maze cognitive task at 2 weeks post-injury in rats subjected to moderate

fluid percussion injury (Saatman et al. 1997). In Chapter 4 we demonstrated that central

Page 228: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

215

infusion of IGF-1 reduced brain injury-induced impairments in cognitive function at 7 d

following CCI. Taken together, these studies demonstrate that treatment with IGF-1

attenuates cognitive dysfunction across a spectrum of injury severities in rodent models

of TBI.

Future studies may need to incorporate additional cognitive tasks to evaluate the

specific contribution of post-traumatic neurogenesis in improvements of cognitive

function after TBI. A recent study highlights that an enhancement of neurogenesis

improves pattern separation in a contextual discrimination task, but does not improve

spatial memory in Morris water maze (Sahay et al. 2011), suggesting that enhancement

of post-traumatic neurogenesis may improve specific aspects of cognition. Considering

we observed an increase in immature neuron density after brain injury above sham

injury levels, future studies may need to incorporate additional assessments of cognitive

performance, including pattern separation and aspects of contextual learning and

memory, to evaluate potential functional implications of hippocampal plasticity after

experimental TBI.

Long-term Assessments of Cognitive Improvement after Treatment with IGF-1

The development of immature neurons into mature granular neurons is an

important stage of adult neurogenesis as surviving mature granular neurons can

functionally incorporate into the granular layer circuitry in the weeks following

differentiating into an immature neuron (Zhao et al. 2006). In a newly generated neuron,

expression of NeuN, a marker of mature neurons, and gradual cessation of DCx

expression occurs approximately 2 to 3 weeks following neuronal differentiation and is

indicative of the transition from an immature neuron into a mature neuron (van Praag et

al. 2002; Zhao et al. 2006). Future studies will need to evaluate if IGF-1 promotes the

survival, maturation and incorporation of newborn neurons generated after injury into the

Page 229: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

216

hippocampus by using markers of mature granular neurons, i.e. NeuN, and indicators of

neuronal activity, i.e. c-fos, in the weeks following CCI. In addition, future studies will

also need to evaluate if the neurobehavioral improvements afforded by IGF-1 are

sustained and correlate with increases in survival and maturation of newborn neurons. In

Appendix 1, we show that the novel object recognition task can be utilized to evaluate

persistent injury-induced cognitive impairments for at least 6 weeks following CCI

(Figure A1.1). The novel object recognition task will need to be incorporated into future

studies to evaluate if the IGF-1-mediated improvements in neurobehavioral function

described in Chapter 4 are sustained following severe CCI.

Generation of Oligodendrocytes in the Injured Hippocampus and IGF-1

In addition to enhanced neuronal differentiation, it is possible that treatment with

IGF-1 promotes oligodendrocyte survival and promotes the generation of new

oligodendrocytes in the injured brain. As demonstrated by Rola et al. (2006), cellular

proliferation in the dentate gyrus granular layer after CCI results in increased numbers of

oligodendrocytes as well as neurons, astrocytes, and microglia. The anti-apoptotic

capacity of IGF-1 may also promote oligodendrocyte survival. Administration of IGF-1

into the lateral ventricle of fetal sheep reduced demyelination and caspase-3 activation

in oligodendrocytes, and increased the number of phospholipid protein (PLP) positive

cells after ischemic injury (Guan et al. 2001). Overexpression of IGF-1 reduces

oligodendrocyte apoptosis and promotes remyelination by 5 weeks following cuprizone-

induced demyelination (Mason et al. 2000). Systemic administration of IGF-1 in

hypophysectomized rats resulted in increased cellular proliferation in the piriform and

parietal cortex after 20 d of daily BrdU injections and increased the number of the newly

proliferated cells that expressed markers of oligodendrocytes, including myelin basic

Page 230: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

217

protein and 2,3-cyclic nucleotide 3-phophodiesterase (Aberg et al. 2007). A previous

study suggests that IGF-1 signaling and increased inhibition of bone morphogenic

proteins (BMP) is instructive to drive oligodendrocyte and neuronal differentiation, but

not astrocytic differentiation, of the neural stem cells (Hsieh et al. 2004). Future studies

will need to evaluate if IGF-1 promotes the survival and generation of oligodendrocytes

following brain injury.

Therapeutic Strategies for IGF-1 after TBI

The incidence of TBI has been on the rise for many years, with an estimated 1.7

million cases in 2010, an increase of 200,000 cases annually since 2000 (Summers et

al. 2009; Faul M 2010). The majority of the estimated cases encompass mild or less

severe cases of TBI (Faul M 2010). Despite the increasing incidence of mild TBI, the

frequency of fatal TBIs has also steadily increased (Summers et al. 2009; Faul M 2010),

highlighting the need for therapeutic strategies that encompass the entire range of TBI

severities. Therapeutic interventions that aim to reduce injurious mechanisms while also

promoting regenerative mechanisms hold promise in counteracting a wide range of

pathological consequences of TBI. The pleiotropic effects of IGF-1 make it a promising

therapeutic agent for the treatment of TBI. While the findings in this dissertation and

additional work from our lab highlight the therapeutic potential of IGF-1 in the context of

severe contusion brain injury, we need to consider and pursue clinically relevant

therapeutic paradigms tailored for varying severities and pathoanatomical types of TBI.

While central administration of IGF-1 may be feasible for severely brain-injured patients,

additional considerations for therapeutic strategies need to be evaluated to afford

treatment of IGF-1 for less severe cases of TBI.

Page 231: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

218

In this dissertation we found that conditional astrocyte-specific overexpression of

IGF-1 enhanced post-traumatic hippocampal neurogenesis by promoting neuronal

differentiation of newly proliferated cells. While the transgenic overexpressing mouse

model provides valuable information about IGF-1-mediated effects in the brain, the

findings need to be verified in a clinically relevant systemic or central administration

paradigm. We corroborated our findings from the astrocyte-specific IGF-1

overexpressing transgenic mouse model using a more clinically relevant treatment

intervention in which IGF-1 was infused into the lateral ventricle following severe CCI.

We also demonstrated that IGF-1 consistently increased immature neuron density after

CCI in a number of infusion paradigms, including a clinically relevant delayed treatment

onset of 6 hr post-injury. Together these findings underscore the efficacy of central

delivery of IGF-1 in the brain to attenuate neurobehavioral dysfunction and enhance

post-traumatic neurogenesis. In order to expand the therapeutic applicability of IGF-1

across the spectrum of brain injury, we need to continue our evaluation of systemic

administration of IGF-1.

A Phase II safety and efficacy trial demonstrated that intravenous infusion of 0.01

mg/kg/hr IGF-1 for a period of 14 d in moderate-to-severely brain-injured patients

promoted metabolic stability and improved outcome at 6 months post-injury in patients

(Hatton et al. 1997). IGF-1-treated brain-injured patients achieved average peak levels

of approximately 460 ng/mL during the infusion period, with 11 out of 17 patients

achieving serum levels of IGF-1 above 350 ng/mL, while control patients achieved an

average of approximately 160 ng/mL, a value at the low end of the 150 to 400 ng/mL

physiological range of IGF-1 in systemic circulation (Hatton et al. 1997). In this same

study, intravenous infusion of IGF-1 resulted in elevated levels of IGF-1 above 350

ng/mL for an average of 8 d, but the heightened concentrations of plasma IGF-1 could

Page 232: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

219

not be sustained for the entire infusion period of 14 d (Hatton et al. 1997). The inability to

maintain sustained elevations of IGF-1 was attributed to a significant reduction in the

concentration of IGF binding protein 3 (IGFBP3). IGFBP3 is the primary carrier protein of

IGF-1 and formation of the IGF-1/IGFBP3 complex results in a prolonged circulating half-

life of IGF-1 (Guler et al. 1989). Prolonged infusion of IGF-1 likely resulted in negative

feedback of IGF-1 on the growth hormone axis and resulted in reduced IGFBP3

production.

In two subsequent placebo-controlled clinical trials, intravenous infusion of 0.01

mg/kg/hr IGF-1 was paired with daily subcutaneous injections of 0.05 mg/kg growth

hormone because growth hormone promotes the production of IGFBP3. Co-

administration of IGF-1 and growth hormone resulted in peak concentrations of IGF-1

above 1000 ng/mL in systemic circulation, and the elevated levels were sustained during

the infusion period (Rockich et al. 1999; Hatton et al. 2006). Co-administration of IGF-1

and growth hormone resulted in elevated levels of IGFBP3 in systemic circulation,

suggesting that increased IGFBP3 expression likely contributed to the sustain elevations

of IGF-1 above the therapeutic threshold of 350 ng/mL (Rockich et al. 1999; Hatton et al.

2006). The clinical trial (Hatton 2006) for co-administration of IGF-1 and growth hormone

was halted in response to findings from a European clinical trial in which administration

of growth hormone alone resulted in increased morbidity and mortality from increased

infection and septic shock (Takala et al. 1999). While no adverse side effects were

observed in brain-injured patients receiving IGF-1 and growth hormone, a moratorium on

growth hormone administration prevented additional investigation of this therapeutic

approach to deliver IGF-1 and growth hormone after TBI. The promising findings

reported by Hatton et al. (1997, 2006) warrant additional studies to investigate

alternative approaches to achieving sustained elevations of IGF-1 in systemic circulation

Page 233: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

220

without co-administration of growth hormone. In Chapters 3 and 4, we demonstrated that

systemic and central infusion of IGF-1 resulted in elevated and sustained levels of hIGF-

1 in systemic circulation. We also demonstrated that elevated levels of IGF-1 in the brain

resulted in improved neurobehavioral function and enhanced post-traumatic

neurogenesis in the injured hippocampus. Future studies will need to explore alternative

methods to achieve sustained elevations in the levels of IGF-1 using treatment

strategies that may prevent degradation and improve the circulating half-life of IGF-1.

Administration of IGF-1 Encapsulated PLGA Microspheres after TBI

Due to the short half-life of IGF-1, repeated injections or continuous infusion are

often required to sustain elevated therapeutic concentrations of IGF-1 in the systemic

circulation. A therapeutic strategy requiring less frequent administration may provide

greater compliance and convenience for hospital staff, caregivers, and brain-injured

patients with milder TBIs. Poly(lactic-co-glycolitic acid) is an FDA approved copolymer

frequently utilized to create microspheres to encapsulate and provide controlled

sustained release of a therapeutic agent (Crotts and Park 1998). Administration of

microsphere encapsulated IGF-1 promotes prolonged residence time of IGF-1 in

circulation and can provide continuous delivery of IGF-1 for weeks following a single

injection of microspheres (Meinel et al. 2001; Singh et al. 2001; Carrascosa et al. 2004).

Carrascosa et al. (2004) demonstrated that a single injection of 1.4mg/kg IGF-1

encapsulated microspheres increased serum concentrations of IGF-1 for 12 d and brain

concentrations for 5 d after injection. Repeated injections of IGF-1 encapsulated

microspheres also improved motor function, as assessed by rotarod, in a mouse model

of purkinje cell degeneration (Carrascosa et al. 2004). These finding suggest that IGF-1

Page 234: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

221

encapsulated microspheres could be utilized to promote sustained elevations in plasma

concentrations of IGF-1 following TBI.

In collaboration with Heidi Mansour, Ph.D. at the University of Kentucky, we

sought to produce IGF-1 encapsulated microspheres that could provide controlled and

sustained release of IGF-1 for days to weeks after brain injury. In Appendix 2, we

highlight our concerted efforts to produce PLGA microspheres using the solvent

extraction method (Yuan et al. 2009) and the spray dry method (Vehring 2008). We

aimed to produce spherical IGF-1 encapsulated microspheres 1-5 µm in diameter with a

drug loading efficiency of 75% to provide sustained systemic release of 4.5 mg/kg of

IGF-1 over a period of 2 weeks. Using the solvent extraction method (Figure A.2.1), we

produced microspheres of the target diameter and shape (Figure A.2.2A, B), but were

unable to encapsulate sufficient concentrations of IGF-1. We next pursued the spray

drying method to improve the IGF-1 loading efficiency.

Using the spray drying method, we produced spherical microspheres 1-5 µm in

diameter (Figure A2.3.A, B). We achieved a drug loading efficiency of approximately

75% using insulin as a structural analog to reduce cost during the development process.

In vitro release kinetics revealed that insulin encapsulated PLGA microspheres resulted

in an initial burst release of insulin within the first 12 hr and sustained release of insulin

for at least 7 d after reconstitution (Figure A2.3C), suggesting that microspheres

produced with IGF-1 could provide controlled sustained release of IGF-1 during the

week post-injury. However, additional work is required to determine if the release pattern

is altered in vivo and that encapsulation of IGF-1 does not produce different release

kinetics. Our preliminary studies demonstrate that PLGA microspheres of target shape

and size could be produced, and warrant future studies to consider IGF-1 encapsulated

Page 235: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

222

PLGA microspheres as a viable clinically relevant strategy to achieve sustained delivery

of IGF-1 after TBI.

Treatment with PEGylated IGF-1 after TBI

The addition of a polyethylene glycol moiety to proteins, referred to as protein

PEGylation, represents a successful strategy to prolong the half-life of subcutaneously

injected IGF-1. PEGylation is thought to enhance the pharmacokinetics of therapeutic

agents by reducing antigenicity, immunological degradation and renal clearance of the

PEGylated molecule (Milla et al. 2012). PEGylation of IGF-1 at Lys68 reduced the

association rates of IGF-1 for its receptor and the binding proteins; however, this

modification did not alter Akt or MAPK signaling or glucose uptake in cultured myoblasts

(Metzger et al. 2011). In vivo, subcutaneous injection of PEGylated IGF-1 increased the

circulating half-life of IGF-1, and increased systemic levels of IGF binding proteins 2 and

3 compared to recombinant human IGF-1 (Metzger et al. 2011). Two week

administration of PEGylated IGF-1, injected subcutaneously twice a week, reduced

soluble Aβ and increased synaptic markers (SNAP25 and PSD95) in a mouse model of

amyloidosis (Saenger et al. 2012). Moreover, treatment of PEGylated IGF-1 improves

muscle force generation, motor coordination, and improved rotarod performance in a

mouse model of amyotrophic lateral sclerosis (Saenger et al. 2012), suggesting that the

PEGylation process improves therapeutic applicability of IGF-1. PEGylation of IGF-1

prolongs the circulating half-life and reduces the potential detrimental side effects

associated with systemic administration of IGF-1. Considering the studies that

demonstrate the efficacy of systemically administered IGF-1 after TBI (Hatton et al.

1997; Saatman et al. 1997; Hatton et al. 2006; Rubovitch et al. 2010), PEGylated IGF-1

Page 236: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

223

should be evaluated as an alternative approach to achieving sustained plasma

concentrations of IGF-1 for the treatment of TBI.

Intranasal Delivery of IGF-1 after TBI

Intranasal delivery of IGF-1 represents an alternative non-invasive therapeutic

approach to paradigms designed to protect against degradation of IGF-1, and instead

would afford rapid delivery of IGF-1 to the brain. Intranasal administration of radiolabeled

IGF-1 results in a widespread distribution and rapid delivery of IGF-1 throughout several

regions of the brain, including the hippocampus, via movement along olfactory and

trigeminal pathways within 30 minutes following initial delivery (Thorne et al. 2004).

Previous studies illustrate that intranasal administration of IGF-1 protects against cell

loss and promotes improved neurological function after hypoxic-ischemic injury (Liu et al.

2001; Lin et al. 2009). IGF-1 encapsulated microspheres could also be adapted for

intranasal administration to achieve sustained delivery of IGF-1 throughout the injured

brain. Intranasal administration of IGF-1 is a promising therapeutic strategy for TBI due

in part to the non-invasive delivery and rapid uptake and distribution in the brain.

Final Conclusions

The findings reported in this dissertation provide strong evidence that

supplementation of IGF-1 attenuates neurobehavioral dysfunction and enhances

hippocampus neurogenesis in the controlled cortical impact mouse model of contusion

brain injury. Using clinically relevant treatment strategies of continuous systemic and

intracerebroventricular infusion, we showed that administration of IGF-1 promoted the

activation of Akt, indicative of increased IGF-1 signaling, in the injured brain.

Intracerebroventricular infusion of IGF-1 attenuated motor and cognitive dysfunction

Page 237: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

224

after brain injury. Elevated levels of IGF-1 in the brain resulted in significant increases in

immature neuron density and enhanced the generation of newborn neurons in the

injured hippocampus. We demonstrated that the significant increase in immature neuron

density was retained in a clinically relevant delayed treatment onset of 6 hr post-injury.

Targeted astrocyte-specific overexpression of IGF-1 promoted the generation of

newborn neurons in the injured hippocampus by promoting neuronal differentiation of

newly proliferated cells. We showed that immature neurons exhibited reduced dendritic

arbor complexity after brain injury, a previously unknown pathological consequence of

brain injury. Targeted overexpression of IGF-1 in the brain promoted the restoration of

dendritic arbor complexity to the morphology observed in uninjured mice. Taken

together, we provide compelling evidence and important groundwork for pursuing a

comprehensive preclinical evaluation of IGF-1 in an effort to create a solid foundation for

future clinical trials evaluating the administration of IGF-1 in brain-injured patients.

Copyright © Shaun William Carlson 2013

Page 238: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

225

Appendix 1: Long-term Cognitive Deficit following Severe Controlled Cortical

Impact as Assessed by the Novel Object Recognition (NOR) Task

Deficits in spatial learning and memory, as assessed by Morris Water Maze,

have been reported for up to one year following CCI in rats (Dixon et al. 1999). The

novel object recognition task was utilized to evaluate brain injury-induced memory

deficits at 7 d post-injury in Chapter 4 of this dissertation. We sought to determine if the

novel object recognition task is capable of detecting persistent cognitive dysfunction in

mice subjected to severe 1 mm CCI. A cohort of 8 week old male C57/BL6 mice were

subjected to sham injury (n= 7) or severe 1 mm CCI (n=12) and evaluated for cognitive

dysfunction at 8 d, and 2, 4, and 6 weeks following injury. We demonstrate that mice

subjected to severe CCI exhibit a sustained cognitive deficit for weeks at least 6 weeks

post-injury. Analysis by a repeated measures one-way ANOVA revealed that mice

subjected to 1 mm CCI exhibited significantly lower recognition indices, indicative of

reduced recognition of the novel object, at 8 d, and 2, 4, and 6 weeks post-injury

compared to sham-injured mice at each respective time point (*p<0.01, Newman-Keuls

post-hoc t-test; Fig. A1.1). Brain-injured mice show no change in recognition indices over

the 6 week period post-injury; however, sham-injured mice show significantly higher

recognition indices compared to sham-injured mice at 8 d and 4 week post-injury

(p<0.05, Newman-Keuls post-hoc t-test).

Page 239: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

226

Figure A1.1: Severe controlled cortical impact (CCI) results in cognitive deficits that persist for up to 6 weeks post-injury. Brain-injured mice exhibited significant reductions in recognition indices at 8 d, 2, 4, and 6 weeks after brain injury, as compared to sham-injured mice at each respective time point (*p<0.01, Newman-Keuls post-hoc t-test following a one-way repeated measures ANOVA). Brain-injured mice showed no change in recognition indices during the 6 weeks following CCI.

Copyright © Shaun William Carlson 2013

*

Page 240: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

227

Appendix 2: Formulation of Poly(lactic-co-glycolytic acid) (PLGA) Microspheres

for the Development of a Subcutaneously-injected Sustained Release Delivery

System for IGF-1

Poly(lactic-co-glycolytic acid) (PLGA) is an FDA-approved biodegradable

polymer often utilized to produce microspheres for sustained delivery of therapeutic

agents (Crotts and Park 1998). Due to the short plasma half of IGF-1, we postulated that

subcutaneous injection of IGF-1-encapsulated microspheres would provide sustained

delivery of IGF-1 into the systemic circulation after brain injury. The work described

below was completed in collaboration with Heidi Mansour, Ph.D. in the College of

Pharmacy at the University of Kentucky, and personnel in her lab, including Yun Seok

Rhee, Ph.D., Minji Sohn and Jackie Ma. Previous studies demonstrated that IGF-1 is

compatible with the microsphere encapsulation process (Meinel et al. 2001; Singh et al.

2001; Carrascosa et al. 2004). We developed a protocol to target production of spherical

microspheres (1-5 µm in diameter) with an IGF-1 encapsulation efficiency of 75% in

order to deliver 4.5 mg/kg of IGF-1 for a duration of 2 weeks post-injury. These

parameters were selected to mimic a previous study that demonstrated efficacy of IGF-1

encapsulated microspheres to achieve sustained delivery of IGF-1 for 2 weeks and

improve motor function in a mouse model of cerebellar ataxia (Carrascosa et al. 2004).

A 1-5 µm diameter was selected to reduce microsphere degradation by host immune

cells following subcutaneous injection.

The morphology and size of the microspheres were evaluated by scanning

electron microscopy (SEM) and particle size analyzer, respectively. Loading efficiencies

of IGF-1 and insulin, a structural surrogate of IGF-1 utilized to reduce the expense of

protein during developmental stages, were analyzed by high performance liquid

Page 241: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

228

chromatography. The methods of solvent extraction and spray drying were utilized to

produce IGF-1 or insulin encapsulated PLGA microspheres in this study.

The solvent extraction protocol that was utilized to produce IGF-1 encapsulated

PLGA microspheres is detailed in Fig. A2.1. A water/oil/water (W/O/W) emulsion was

created by the sequential addition of IGF-1 diluted in sodium succinate buffer to create

the first aqueous phase, and PLGA dissolved in methylene chloride to create the primary

water and oil emulsion (W/O). Addition of polyvinyl alcohol (PVA) and an initial

homogenization of the solution resulted in the generation of the W/O/W emulsion. This

was followed by sustained agitation to initiate microsphere formation. After a 16 hr

period of continued agitation, the solution was centrifuged for washing and collection of

the microspheres.

Several iterations were completed to generate microspheres of the target

spherical shape (Fig. A2.2A) and size (Fig. A2.2B), but a loading efficiency of less than

1% was achieved. Subsequent iterations to alter concentrations of PLGA and PVA did

not improve the efficiency of IGF-1 encapsulation. In light of the low IGF-1 encapsulation

efficiency, additional studies pursued the production of microspheres by the spray drying

method, as IGF-1 had been shown to be compatible with the spray drying procedure

(Lam et al. 2000). The previous morphology, size, and IGF-1 loading efficiency remained

the target parameters for subsequent experiments.

The spray drying method atomizes a primary PLGA and IGF-1 solution into small

droplets that are rapidly dried at elevated temperatures while being ejected from a small

nozzle of the apparatus (Vehring 2008). The dried microspheres were collected and

remained lyophilized until reconstituted for analysis. One disadvantage of the spray

drying method is the need for high drying temperatures, which despite the brief exposure

Page 242: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

229

to temperatures exceeding 60°C, could promote the denaturation of IGF-1. Conversely,

a great advantage of the spray drying method is increased yield as the lyophilization

process facilitates improved collection of the microspheres.

In an effort to reduce potential IGF-1 denaturation, we reduced the drying

temperatures below 60°C. Insulin was also utilized to evaluate drug loading efficiency.

Several iterations evaluating the concentrations of PLGA and insulin resulted in an

approximate 50% improvement in yield and produced insulin encapsulated microspheres

that exhibited the desired morphology (Fig. A2.3A), with a 75% loading efficiency of

insulin. Analysis of the insulin release kinetics in vitro revealed a rapid release of

approximately 40% of encapsulated insulin within 8 hr of reconstitution, followed by a

sustained release of insulin for up to 7 d following reconstitution (Fig. A2.3C). Additional

work is needed to determine if the in vitro release kinetics detailed in Fig. A2.3C is

reproducible in vivo after subcutaneous injection. Subsequent studies evaluating the

IGF-1 encapsulation efficiency and release profiles were not completed. It was

suggested that increased loading efficiency and sustained release patterns for delivery

of IGF-1 for 2 weeks would require higher drying temperatures that would likely lead to

protein denaturation. While this study did not achieve the desired sustained release of

IGF-1 for a period of 2 weeks, this work may be continued to evaluate if sustained

release of IGF-1 for 1 week post-injury is a viable subcutaneous continuous delivery

system for the administration of IGF-1 after TBI.

Page 243: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

230

Figure A2.1: Protocol for the development of insulin-like growth factor-1 (IGF-1) encapsulated poly(lactic-co-glycolytic acid) (PLGA) microspheres by solvent extraction. IGF-1 is initially dissolved in sodium succinate buffer and mixed with PLGA polymer to create the primary water/oil (W/O) emulsion. Addition of polyvinyl alcohol (PVA) to the solution produced the W/O/W emulsion after homogenization. Prolonged stirring results in the formation of IGF-1 encapsulated PLGA microspheres. Centrifugation is utilized to rinse and collect the microspheres. This experiment schematic was prepared by Dr. Yun Seok Rhee.

Page 244: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

231

Figure A2.2: Morphology and size of poly(lactic-co-glycolytic acid) (PLGA) microspheres generated by solvent extraction. (A) Spherical microspheres were observed by SEM that (B) ranged in size from 1 µm to greater than 10 µm, with a median diameter of 5 µm.

SHIMADZU SALD-7101 (SALD-7101-WEA1:V1.02)

(File Name) Blank MS06

(Sample ID) Blank MS6 (Sample #) 6

( Date ) 09/12/22 ( Time ) 10:56:56

R Index=1.60-0.10i Median D : 5.568

Modal D : 4.467

Mean V : 5.745

Std Dev : 0.263

10.0%D : 2.671

50.0%D : 5.568

90.0%D : 12.779

S Level : 0

D Func :None

D Shift : 0

Q 0 (%) q0(%)

Normalized Particle Amount

Particle Diameter ( m)

0.01 0.05 0.1 0.5 1 5 10 50 100 500 1000

0

10

20

30

40

50

60

70

80

90

100

Diam

x( m)

Cum

Q (%)0

Diff

q0(%)

Diam

x( m)

Cum

Q (%)0

Diff

q0(%)

Diam

x( m)

Cum

Q (%)0

Diff

q0(%)

1 300.000 100.000 0.000

2 244.106 100.000 0.000

3 198.626 100.000 0.000

4 161.620 100.000 0.000

5 131.508 100.000 0.000

6 107.006 100.000 0.000

7 87.070 100.000 0.000

8 70.847 100.000 0.000

9 57.648 100.000 0.007

10 46.907 99.993 0.091

11 38.168 99.902 0.353

12 31.057 99.549 0.898

13 25.270 98.651 1.707

14 20.562 96.944 2.488

15 16.731 94.457 3.166

16 13.614 91.291 4.705

17 11.078 86.586 8.108

18 9.014 78.478 11.479

19 7.334 67.000 12.723

20 5.968 54.276 12.735

21 4.856 41.542 12.651

22 3.951 28.891 10.814

23 3.215 18.076 9.246

24 2.616 8.830 5.357

25 2.129 3.473 2.818

26 1.732 0.654 0.654

27 1.409 0.000 0.000

28 1.147 0.000 0.000

29 0.933 0.000 0.000

30 0.759 0.000 0.000

31 0.618 0.000 0.000

32 0.503 0.000 0.000

33 0.409 0.000 0.000

34 0.333 0.000 0.000

35 0.271 0.000 0.000

36 0.220 0.000 0.000

37 0.179 0.000 0.000

38 0.146 0.000 0.000

39 0.119 0.000 0.000

40 0.097 0.000 0.000

41 0.079 0.000 0.000

42 0.064 0.000 0.000

43 0.052 0.000 0.000

44 0.042 0.000 0.000

45 0.034 0.000 0.000

46 0.028 0.000 0.000

47 0.023 0.000 0.000

48 0.019 0.000 0.000

49 0.015 0.000 0.000

50 0.012 0.000 0.000

51 0.010 0.000 0.000

Sampling Mode : Manual Refractive Index : 1.60-0.10i

Signal Accumulation Count : 1 Interval (sec) : ___ Signal Averaging Count : 128

Max of Absorbance Range : 0.200 Min of Absorbance Range : 0.010

Ultrasonic Dispersion Time (sec) : ___ Waiting Time After Ultrasonic Dispersion(sec) : ___

Range for Analysis : OFF Starting Point : 1 S/B Sensor : Enable

Blank microspheres

A

B

Page 245: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

232

Figure A2.3: Morphology, size and release kinetics of insulin encapsulated poly(lactic-co-glycolytic acid) (PLGA) microspheres generated by spray drying. (A) Spherical microspheres were observed by SEM that (B) ranged in size from the targeted 1 to 5 µm. Release kinetics demonstrate an initial burst release of insulin in the initial 12 hr followed by sustained delivery for up to 7 d after microsphere reconstitution.

Copyright © Shaun William Carlson 2013

SHIMADZU SALD-7101 (SALD-7101-WEA1:V1.02)

(File Name) PLGA LOW IV 5%65T42P50Q-

(Sample ID) LOWPLGA65T42P50QACE (Sample #) ACETONE-1

( Date ) 10/07/20 ( Time ) 19:17:03

R Index=2.40-0.20i Median D : 1.679

Modal D : 1.413

Mean V : 1.734

Std Dev : 0.143

10.0%D : 1.236

50.0%D : 1.679

90.0%D : 2.431

S Level : 0

D Func :None

D Shift : 0

Q 0 (%) q0(%)

Normalized Particle Amount

Particle Diameter ( m)

0.01 0.05 0.1 0.5 1 5 10 50 100 500 1000

0

10

20

30

40

50

60

70

80

90

100

Diam

x( m)

Cum

Q (%)0

Diff

q0(%)

Diam

x( m)

Cum

Q (%)0

Diff

q0(%)

Diam

x( m)

Cum

Q (%)0

Diff

q0(%)

1 300.000 100.000 0.000

2 244.106 100.000 0.000

3 198.626 100.000 0.000

4 161.620 100.000 0.000

5 131.508 100.000 0.000

6 107.006 100.000 0.000

7 87.070 100.000 0.003

8 70.847 99.997 0.008

9 57.648 99.988 0.020

10 46.907 99.969 0.038

11 38.168 99.930 0.061

12 31.057 99.869 0.082

13 25.270 99.787 0.087

14 20.562 99.701 0.071

15 16.731 99.630 0.046

16 13.614 99.584 0.022

17 11.078 99.562 0.015

18 9.014 99.547 0.070

19 7.334 99.477 0.199

20 5.968 99.278 0.327

21 4.856 98.951 0.374

22 3.951 98.577 0.482

23 3.215 98.095 3.484

24 2.616 94.611 15.245

25 2.129 79.366 24.735

26 1.732 54.631 29.478

27 1.409 25.153 20.641

28 1.147 4.512 4.512

29 0.933 0.000 0.000

30 0.759 0.000 0.000

31 0.618 0.000 0.000

32 0.503 0.000 0.000

33 0.409 0.000 0.000

34 0.333 0.000 0.000

35 0.271 0.000 0.000

36 0.220 0.000 0.000

37 0.179 0.000 0.000

38 0.146 0.000 0.000

39 0.119 0.000 0.000

40 0.097 0.000 0.000

41 0.079 0.000 0.000

42 0.064 0.000 0.000

43 0.052 0.000 0.000

44 0.042 0.000 0.000

45 0.034 0.000 0.000

46 0.028 0.000 0.000

47 0.023 0.000 0.000

48 0.019 0.000 0.000

49 0.015 0.000 0.000

50 0.012 0.000 0.000

51 0.010 0.000 0.000

Sampling Mode : Manual Refractive Index : 2.40-0.20i

Signal Accumulation Count : 1 Interval (sec) : ___ Signal Averaging Count : 128

Max of Absorbance Range : 0.250 Min of Absorbance Range : 0.010

Ultrasonic Dispersion Time (sec) : ___ Waiting Time After Ultrasonic Dispersion(sec) : ___

Range for Analysis : OFF Starting Point : 1 S/B Sensor : Enable

WATER AS DISPERSANT

A

B

C

Page 246: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

233

Appendix 3: Permission for Article Reprint for Chapter 2.

Copyright © Shaun William Carlson 2013

Page 247: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

234

References

Aberg, M. A., N. D. Aberg, H. Hedbacker, J. Oscarsson and P. S. Eriksson (2000). "Peripheral infusion of IGF-I selectively induces neurogenesis in the adult rat hippocampus." J Neurosci 20(8): 2896-2903.

Aberg, M. A., N. D. Aberg, T. D. Palmer, A. M. Alborn, C. Carlsson-Skwirut, P. Bang, L. E. Rosengren, T. Olsson, F. H. Gage and P. S. Eriksson (2003). "IGF-I has a direct proliferative effect in adult hippocampal progenitor cells." Mol Cell Neurosci 24(1): 23-40.

Aberg, N. D., U. E. Johansson, M. A. Aberg, N. A. Hellstrom, J. Lind, C. Bull, J. Isgaard, M. F. Anderson, J. Oscarsson and P. S. Eriksson (2007). "Peripheral infusion of insulin-like growth factor-I increases the number of newborn oligodendrocytes in the cerebral cortex of adult hypophysectomized rats." Endocrinology 148(8): 3765-3772.

Adelson, P. D., C. E. Dixon and P. M. Kochanek (2000). "Long-term dysfunction following diffuse traumatic brain injury in the immature rat." J Neurotrauma 17(4): 273-282.

Alahmadi, H., S. Vachhrajani and M. D. Cusimano (2010). "The natural history of brain contusion: an analysis of radiological and clinical progression." J Neurosurg 112(5): 1139-1145.

Albensi, B. C., P. G. Sullivan, M. B. Thompson, S. W. Scheff and M. P. Mattson (2000). "Cyclosporin ameliorates traumatic brain-injury-induced alterations of hippocampal synaptic plasticity." Exp Neurol 162(2): 385-389.

Amiri, A., W. Cho, J. Zhou, S. G. Birnbaum, C. M. Sinton, R. M. McKay and L. F. Parada (2012). "Pten deletion in adult hippocampal neural stem/progenitor cells causes cellular abnormalities and alters neurogenesis." J Neurosci 32(17): 5880-5890.

Anderson, K. J., K. M. Miller, I. Fugaccia and S. W. Scheff (2005). "Regional distribution of fluoro-jade B staining in the hippocampus following traumatic brain injury." Exp Neurol 193(1): 125-130.

Andriessen, T. M., B. Jacobs and P. E. Vos (2010). "Clinical characteristics and pathophysiological mechanisms of focal and diffuse traumatic brain injury." J Cell Mol Med 14(10): 2381-2392.

Arsenijevic, Y. and S. Weiss (1998). "Insulin-like growth factor-I is a differentiation factor for postmitotic CNS stem cell-derived neuronal precursors: distinct actions from those of brain-derived neurotrophic factor." J Neurosci 18(6): 2118-2128.

Arsenijevic, Y., S. Weiss, B. Schneider and P. Aebischer (2001). "Insulin-like growth factor-I is necessary for neural stem cell proliferation and demonstrates distinct actions of epidermal growth factor and fibroblast growth factor-2." J Neurosci 21(18): 7194-7202.

Page 248: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

235

Atkins, C. M. (2011). "Decoding Hippocampal Signaling Deficits after Traumatic Brain Injury." Transl Stroke Res 2(4): 546-555.

Bain, A. C. and D. F. Meaney (2000). "Tissue-level thresholds for axonal damage in an experimental model of central nervous system white matter injury." J Biomech Eng 122(6): 615-622.

Baker, A. J., R. J. Moulton, V. H. MacMillan and P. M. Shedden (1993). "Excitatory amino acids in cerebrospinal fluid following traumatic brain injury in humans." J Neurosurg 79(3): 369-372.

Baker, J., J. P. Liu, E. J. Robertson and A. Efstratiadis (1993). "Role of insulin-like growth factors in embryonic and postnatal growth." Cell 75(1): 73-82.

Baldwin, S. A., I. Fugaccia, D. R. Brown, L. V. Brown and S. W. Scheff (1996). "Blood-brain barrier breach following cortical contusion in the rat." J Neurosurg 85(3): 476-481.

Baldwin, S. A., T. Gibson, C. T. Callihan, P. G. Sullivan, E. Palmer and S. W. Scheff (1997). "Neuronal cell loss in the CA3 subfield of the hippocampus following cortical contusion utilizing the optical disector method for cell counting." J Neurotrauma 14(6): 385-398.

Baskaya, M. K., A. M. Rao, A. Dogan, D. Donaldson and R. J. Dempsey (1997). "The biphasic opening of the blood-brain barrier in the cortex and hippocampus after traumatic brain injury in rats." Neurosci Lett 226(1): 33-36.

Baxter, R. C. and J. L. Martin (1989). "Binding proteins for the insulin-like growth factors: structure, regulation and function." Prog Growth Factor Res 1(1): 49-68.

Beauchamp, K., H. Mutlak, W. R. Smith, E. Shohami and P. F. Stahel (2008). "Pharmacology of traumatic brain injury: where is the "golden bullet"?" Mol Med 14(11-12): 731-740.

Bendall, S. C., M. H. Stewart, P. Menendez, D. George, K. Vijayaragavan, T. Werbowetski-Ogilvie, V. Ramos-Mejia, A. Rouleau, J. Yang, M. Bosse, G. Lajoie and M. Bhatia (2007). "IGF and FGF cooperatively establish the regulatory stem cell niche of pluripotent human cells in vitro." Nature 448(7157): 1015-1021.

Biegon, A., P. A. Fry, C. M. Paden, A. Alexandrovich, J. Tsenter and E. Shohami (2004). "Dynamic changes in N-methyl-D-aspartate receptors after closed head injury in mice: Implications for treatment of neurological and cognitive deficits." Proc Natl Acad Sci U S A 101(14): 5117-5122.

Blaiss, C. A., T. S. Yu, G. Zhang, J. Chen, G. Dimchev, L. F. Parada, C. M. Powell and S. G. Kernie (2011). "Temporally specified genetic ablation of neurogenesis impairs cognitive recovery after traumatic brain injury." J Neurosci 31(13): 4906-4916.

Page 249: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

236

Bluthe, R. M., F. Frenois, K. W. Kelley and R. Dantzer (2005). "Pentoxifylline and insulin-like growth factor-I (IGF-I) abrogate kainic acid-induced cognitive impairment in mice." J Neuroimmunol 169(1-2): 50-58.

Bondarenko, A. and M. Chesler (2001). "Rapid astrocyte death induced by transient hypoxia, acidosis, and extracellular ion shifts." Glia 34(2): 134-142.

Bondy, C., H. Werner, C. T. Roberts, Jr. and D. LeRoith (1992). "Cellular pattern of type-I insulin-like growth factor receptor gene expression during maturation of the rat brain: comparison with insulin-like growth factors I and II." Neuroscience 46(4): 909-923.

Bondy, C. A. and C. M. Cheng (2004). "Signaling by insulin-like growth factor 1 in brain." Eur J Pharmacol 490(1-3): 25-31.

Bondy, C. A. and W. H. Lee (1993). "Patterns of insulin-like growth factor and IGF receptor gene expression in the brain. Functional implications." Ann N Y Acad Sci 692: 33-43.

Bracko, O., T. Singer, S. Aigner, M. Knobloch, B. Winner, J. Ray, G. D. Clemenson, Jr., H. Suh, S. Couillard-Despres, L. Aigner, F. H. Gage and S. Jessberger (2012). "Gene expression profiling of neural stem cells and their neuronal progeny reveals IGF2 as a regulator of adult hippocampal neurogenesis." J Neurosci 32(10): 3376-3387.

Bramlett, H. M. and W. D. Dietrich (2002). "Quantitative structural changes in white and gray matter 1 year following traumatic brain injury in rats." Acta Neuropathol 103(6): 607-614.

Brooker, G. J., M. Kalloniatis, V. C. Russo, M. Murphy, G. A. Werther and P. F. Bartlett (2000). "Endogenous IGF-1 regulates the neuronal differentiation of adult stem cells." J Neurosci Res 59(3): 332-341.

Bryan, R. M., Jr., L. Cherian and C. Robertson (1995). "Regional cerebral blood flow after controlled cortical impact injury in rats." Anesth Analg 80(4): 687-695.

Brywe, K. G., C. Mallard, M. Gustavsson, M. Hedtjarn, A. L. Leverin, X. Wang, K. Blomgren, J. Isgaard and H. Hagberg (2005). "IGF-I neuroprotection in the immature brain after hypoxia-ischemia, involvement of Akt and GSK3beta?" Eur J Neurosci 21(6): 1489-1502.

Byrnes, K. R., D. J. Loane, B. A. Stoica, J. Zhang and A. I. Faden (2012). "Delayed mGluR5 activation limits neuroinflammation and neurodegeneration after traumatic brain injury." J Neuroinflammation 9: 43.

Cai, W., S. W. Carlson, J. M. Brelsfoard, C. E. Mannon, C. L. Moncman, K. E. Saatman and D. A. Andres (2012). "Rit GTPase signaling promotes immature hippocampal neuronal survival." J Neurosci 32(29): 9887-9897.

Page 250: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

237

Capruso, D. X. and H. S. Levin (1992). "Cognitive impairment following closed head injury." Neurol Clin 10(4): 879-893.

Carbonell, W. S., D. O. Maris, T. McCall and M. S. Grady (1998). "Adaptation of the fluid percussion injury model to the mouse." J Neurotrauma 15(3): 217-229.

Carrascosa, C., I. Torres-Aleman, C. Lopez-Lopez, E. Carro, L. Espejo, S. Torrado and J. J. Torrado (2004). "Microspheres containing insulin-like growth factor I for treatment of chronic neurodegeneration." Biomaterials 25(4): 707-714.

Carson, M. J., R. R. Behringer, R. L. Brinster and F. A. McMorris (1993). "Insulin-like growth factor I increases brain growth and central nervous system myelination in transgenic mice." Neuron 10(4): 729-740.

Castejon, O. J. (1998). "Morphological astrocytic changes in complicated human brain trauma. A light and electron microscopic study." Brain Inj 12(5): 409-427; discussion 407.

Cater, H. L., L. E. Sundstrom and B. Morrison, 3rd (2006). "Temporal development of hippocampal cell death is dependent on tissue strain but not strain rate." J Biomech 39(15): 2810-2818.

Chen, Y., S. Constantini, V. Trembovler, M. Weinstock and E. Shohami (1996). "An experimental model of closed head injury in mice: pathophysiology, histopathology, and cognitive deficits." J Neurotrauma 13(10): 557-568.

Chen, Y. and R. A. Swanson (2003). "Astrocytes and brain injury." J Cereb Blood Flow Metab 23(2): 137-149.

Cheng, C. M., R. F. Mervis, S. L. Niu, N. Salem, Jr., L. A. Witters, V. Tseng, R. Reinhardt and C. A. Bondy (2003). "Insulin-like growth factor 1 is essential for normal dendritic growth." J Neurosci Res 73(1): 1-9.

Chirumamilla, S., D. Sun, M. R. Bullock and R. J. Colello (2002). "Traumatic brain injury induced cell proliferation in the adult mammalian central nervous system." J Neurotrauma 19(6): 693-703.

Chitnis, M. M., J. S. Yuen, A. S. Protheroe, M. Pollak and V. M. Macaulay (2008). "The type 1 insulin-like growth factor receptor pathway." Clin Cancer Res 14(20): 6364-6370.

Chodobski, A., I. Chung, E. Kozniewska, T. Ivanenko, W. Chang, J. F. Harrington, J. A. Duncan and J. Szmydynger-Chodobska (2003). "Early neutrophilic expression of vascular endothelial growth factor after traumatic brain injury." Neuroscience 122(4): 853-867.

Chrysis, D., A. S. Calikoglu, P. Ye and A. J. D'Ercole (2001). "Insulin-like growth factor-I overexpression attenuates cerebellar apoptosis by altering the expression of Bcl family proteins in a developmentally specific manner." J Neurosci 21(5): 1481-1489.

Page 251: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

238

Clausen, F., A. Hanell, M. Bjork, L. Hillered, A. K. Mir, H. Gram and N. Marklund (2009). "Neutralization of interleukin-1beta modifies the inflammatory response and improves histological and cognitive outcome following traumatic brain injury in mice." Eur J Neurosci 30(3): 385-396.

Clelland, C. D., M. Choi, C. Romberg, G. D. Clemenson, Jr., A. Fragniere, P. Tyers, S. Jessberger, L. M. Saksida, R. A. Barker, F. H. Gage and T. J. Bussey (2009). "A functional role for adult hippocampal neurogenesis in spatial pattern separation." Science 325(5937): 210-213.

Clemmons, D. R., J. I. Jones, W. H. Busby and G. Wright (1993). "Role of insulin-like growth factor binding proteins in modifying IGF actions." Ann N Y Acad Sci 692: 10-21.

Conti, A. C., R. Raghupathi, J. Q. Trojanowski and T. K. McIntosh (1998). "Experimental brain injury induces regionally distinct apoptosis during the acute and delayed post-traumatic period." J Neurosci 18(15): 5663-5672.

Contreras, P. C., C. Steffler, E. Yu, K. Callison, D. Stong and J. L. Vaught (1995). "Systemic administration of rhIGF-I enhanced regeneration after sciatic nerve crush in mice." J Pharmacol Exp Ther 274(3): 1443-1449.

Coronado, V. G., L. Xu, S. V. Basavaraju, L. C. McGuire, M. M. Wald, M. D. Faul, B. R. Guzman and J. D. Hemphill (2011). "Surveillance for traumatic brain injury-related deaths--United States, 1997-2007." MMWR Surveill Summ 60(5): 1-32.

Corrigan, J. D., A. W. Selassie and J. A. Orman (2010). "The epidemiology of traumatic brain injury." J Head Trauma Rehabil 25(2): 72-80.

Cortez, S. C., T. K. McIntosh and L. J. Noble (1989). "Experimental fluid percussion brain injury: vascular disruption and neuronal and glial alterations." Brain Res 482(2): 271-282.

Crotts, G. and T. G. Park (1998). "Protein delivery from poly(lactic-co-glycolic acid) biodegradable microspheres: release kinetics and stability issues." J Microencapsul 15(6): 699-713.

D'Ercole, A. J., P. Ye, A. S. Calikoglu and G. Gutierrez-Ospina (1996). "The role of the insulin-like growth factors in the central nervous system." Mol Neurobiol 13(3): 227-255.

Dash, P. K., S. A. Mach and A. N. Moore (2001). "Enhanced neurogenesis in the rodent hippocampus following traumatic brain injury." J Neurosci Res 63(4): 313-319.

Dashtipour, K., A. M. Wong, A. Obenaus, I. Spigelman and C. E. Ribak (2003). "Temporal profile of hilar basal dendrite formation on dentate granule cells after status epilepticus." Epilepsy Res 54(2-3): 141-151.

Page 252: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

239

de Torres, C., F. Munell, I. Ferrer, J. Reventos and A. Macaya (1997). "Identification of necrotic cell death by the TUNEL assay in the hypoxic-ischemic neonatal rat brain." Neurosci Lett 230(1): 1-4.

DeKosky, S. T., K. M. Taffe, E. E. Abrahamson, C. E. Dixon, P. M. Kochanek and M. D. Ikonomovic (2004). "Time course analysis of hippocampal nerve growth factor and antioxidant enzyme activity following lateral controlled cortical impact brain injury in the rat." J Neurotrauma 21(5): 491-500.

Delaney, C. L., H. L. Cheng and E. L. Feldman (1999). "Insulin-like growth factor-I prevents caspase-mediated apoptosis in Schwann cells." J Neurobiol 41(4): 540-548.

Deng, W., J. B. Aimone and F. H. Gage (2010). "New neurons and new memories: how does adult hippocampal neurogenesis affect learning and memory?" Nat Rev Neurosci 11(5): 339-350.

Deng, W., M. D. Saxe, I. S. Gallina and F. H. Gage (2009). "Adult-born hippocampal dentate granule cells undergoing maturation modulate learning and memory in the brain." J Neurosci 29(43): 13532-13542.

Dennis, A. M., M. L. Haselkorn, V. A. Vagni, R. H. Garman, K. Janesko-Feldman, H. Bayir, R. S. Clark, L. W. Jenkins, C. E. Dixon and P. M. Kochanek (2009). "Hemorrhagic shock after experimental traumatic brain injury in mice: effect on neuronal death." J Neurotrauma 26(6): 889-899.

DeRidder, M. N., M. J. Simon, R. Siman, Y. P. Auberson, R. Raghupathi and D. F. Meaney (2006). "Traumatic mechanical injury to the hippocampus in vitro causes regional caspase-3 and calpain activation that is influenced by NMDA receptor subunit composition." Neurobiol Dis 22(1): 165-176.

DeWitt, D. S. and D. S. Prough (2003). "Traumatic cerebral vascular injury: the effects of concussive brain injury on the cerebral vasculature." J Neurotrauma 20(9): 795-825.

Dietrich, W. D., O. Alonso, R. Busto and S. P. Finklestein (1996). "Posttreatment with intravenous basic fibroblast growth factor reduces histopathological damage following fluid-percussion brain injury in rats." J Neurotrauma 13(6): 309-316.

Dietrich, W. D., O. Alonso, R. Busto and M. D. Ginsberg (1994). "Widespread metabolic depression and reduced somatosensory circuit activation following traumatic brain injury in rats." J Neurotrauma 11(6): 629-640.

Dixon, C. E., G. L. Clifton, J. W. Lighthall, A. A. Yaghmai and R. L. Hayes (1991). "A controlled cortical impact model of traumatic brain injury in the rat." J Neurosci Methods 39(3): 253-262.

Dixon, C. E., P. M. Kochanek, H. Q. Yan, J. K. Schiding, R. G. Griffith, E. Baum, D. W. Marion and S. T. DeKosky (1999). "One-year study of spatial memory

Page 253: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

240

performance, brain morphology, and cholinergic markers after moderate controlled cortical impact in rats." J Neurotrauma 16(2): 109-122.

Dixon, C. E., B. G. Lyeth, J. T. Povlishock, R. L. Findling, R. J. Hamm, A. Marmarou, H. F. Young and R. L. Hayes (1987). "A fluid percussion model of experimental brain injury in the rat." J Neurosurg 67(1): 110-119.

Dong, Y. and E. N. Benveniste (2001). "Immune function of astrocytes." Glia 36(2): 180-190.

Duan, C. (2002). "Specifying the cellular responses to IGF signals: roles of IGF-binding proteins." J Endocrinol 175(1): 41-54.

Dunn-Meynell, A. A. and B. E. Levin (1997). "Histological markers of neuronal, axonal and astrocytic changes after lateral rigid impact traumatic brain injury." Brain Res 761(1): 25-41.

Elkin, B. S., A. Ilankovan and B. Morrison, 3rd (2010). "Age-dependent regional mechanical properties of the rat hippocampus and cortex." J Biomech Eng 132(1): 011010.

Elkin, B. S. and B. Morrison, 3rd (2007). "Region-specific tolerance criteria for the living brain." Stapp Car Crash J 51: 127-138.

Emery, D. L., C. T. Fulp, K. E. Saatman, C. Schutz, E. Neugebauer and T. K. McIntosh (2005). "Newly born granule cells in the dentate gyrus rapidly extend axons into the hippocampal CA3 region following experimental brain injury." J Neurotrauma 22(9): 978-988.

Eriksson, P. S., E. Perfilieva, T. Bjork-Eriksson, A. M. Alborn, C. Nordborg, D. A. Peterson and F. H. Gage (1998). "Neurogenesis in the adult human hippocampus." Nat Med 4(11): 1313-1317.

Escartin, C., F. Boyer, A. P. Bemelmans, P. Hantraye and E. Brouillet (2004). "Insulin growth factor-1 protects against excitotoxicity in the rat striatum." Neuroreport 15(14): 2251-2254.

Farkas, O., J. Lifshitz and J. T. Povlishock (2006). "Mechanoporation induced by diffuse traumatic brain injury: an irreversible or reversible response to injury?" J Neurosci 26(12): 3130-3140.

Faul M, X. L., Wald MM, Coronado VG (2010). Traumatic brain injury in the United

States: emergency department visits, hospitalizations, and deaths., Atlanta (GA):

Centers for Disease Control and Prevention, National Center for Injury Prevention

and Control.

Page 254: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

241

Feeney, D. M., M. G. Boyeson, R. T. Linn, H. M. Murray and W. G. Dail (1981). "Responses to cortical injury: I. Methodology and local effects of contusions in the rat." Brain Res 211(1): 67-77.

Fernandez, A. M., A. G. de la Vega and I. Torres-Aleman (1998). "Insulin-like growth factor I restores motor coordination in a rat model of cerebellar ataxia." Proc Natl Acad Sci U S A 95(3): 1253-1258.

Fink, K. B., L. J. Andrews, W. E. Butler, V. O. Ona, M. Li, M. Bogdanov, M. Endres, S. Q. Khan, S. Namura, P. E. Stieg, M. F. Beal, M. A. Moskowitz, J. Yuan and R. M. Friedlander (1999). "Reduction of post-traumatic brain injury and free radical production by inhibition of the caspase-1 cascade." Neuroscience 94(4): 1213-1218.

Finkelstein E, C. P., Miller T (2006). The Incidence and Economic Burden of Injuries in the United States. New York (NY), Oxford University Press.

Fitch, M. T., C. Doller, C. K. Combs, G. E. Landreth and J. Silver (1999). "Cellular and molecular mechanisms of glial scarring and progressive cavitation: in vivo and in vitro analysis of inflammation-induced secondary injury after CNS trauma." J Neurosci 19(19): 8182-8198.

Fitch, M. T. and J. Silver (2008). "CNS injury, glial scars, and inflammation: Inhibitory extracellular matrices and regeneration failure." Exp Neurol 209(2): 294-301.

Flood, C., J. Akinwunmi, C. Lagord, M. Daniel, M. Berry, A. Jackowski and A. Logan (2001). "Transforming growth factor-beta1 in the cerebrospinal fluid of patients with subarachnoid hemorrhage: titers derived from exogenous and endogenous sources." J Cereb Blood Flow Metab 21(2): 157-162.

Flygt, J., A. Djupsjo, F. Lenne and N. Marklund (2013). "Myelin loss and oligodendrocyte pathology in white matter tracts following traumatic brain injury in the rat." Eur J Neurosci.

Fox, G. B., L. Fan, R. A. Levasseur and A. I. Faden (1998). "Sustained sensory/motor and cognitive deficits with neuronal apoptosis following controlled cortical impact brain injury in the mouse." J Neurotrauma 15(8): 599-614.

Fujimoto, S. T., L. Longhi, K. E. Saatman, V. Conte, N. Stocchetti and T. K. McIntosh (2004). "Motor and cognitive function evaluation following experimental traumatic brain injury." Neurosci Biobehav Rev 28(4): 365-378.

Fukunaga, K. and T. Kawano (2003). "Akt is a molecular target for signal transduction therapy in brain ischemic insult." J Pharmacol Sci 92(4): 317-327.

Galford, J. E. and J. H. McElhaney (1970). "A viscoelastic study of scalp, brain, and dura." J Biomech 3(2): 211-221.

Page 255: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

242

Gao, X. and J. Chen (2013). "Moderate traumatic brain injury promotes neural precursor proliferation without increasing neurogenesis in the adult hippocampus." Exp Neurol 239: 38-48.

Gao, X., Y. Deng-Bryant, W. Cho, K. M. Carrico, E. D. Hall and J. Chen (2008). "Selective death of newborn neurons in hippocampal dentate gyrus following moderate experimental traumatic brain injury." J Neurosci Res 86(10): 2258-2270.

Gao, X., P. Deng, Z. C. Xu and J. Chen (2011). "Moderate traumatic brain injury causes acute dendritic and synaptic degeneration in the hippocampal dentate gyrus." PLoS One 6(9): e24566.

Gao, X., G. Enikolopov and J. Chen (2009). "Moderate traumatic brain injury promotes proliferation of quiescent neural progenitors in the adult hippocampus." Exp Neurol 219(2): 516-523.

Geddes, D. M., R. S. Cargill, 2nd and M. C. LaPlaca (2003). "Mechanical stretch to neurons results in a strain rate and magnitude-dependent increase in plasma membrane permeability." J Neurotrauma 20(10): 1039-1049.

Geddes, D. M., M. C. LaPlaca and R. S. Cargill, 2nd (2003). "Susceptibility of hippocampal neurons to mechanically induced injury." Exp Neurol 184(1): 420-427.

Gehrmann, J., D. L. Yao, B. Bonetti, C. A. Bondy, M. Brenner, J. Zhou, G. W. Kreutzberg and H. D. Webster (1994). "Expression of insulin-like growth factor-I and related peptides during motoneuron regeneration." Exp Neurol 128(2): 202-210.

Giffard, R. G., H. Monyer and D. W. Choi (1990). "Selective vulnerability of cultured cortical glia to injury by extracellular acidosis." Brain Res 530(1): 138-141.

Giulian, D., J. Chen, J. E. Ingeman, J. K. George and M. Noponen (1989). "The role of mononuclear phagocytes in wound healing after traumatic injury to adult mammalian brain." J Neurosci 9(12): 4416-4429.

Gluckman, P., N. Klempt, J. Guan, C. Mallard, E. Sirimanne, M. Dragunow, M. Klempt, K. Singh, C. Williams and K. Nikolics (1992). "A role for IGF-1 in the rescue of CNS neurons following hypoxic-ischemic injury." Biochem Biophys Res Commun 182(2): 593-599.

Goodman, J. C., L. Cherian, R. M. Bryan, Jr. and C. S. Robertson (1994). "Lateral cortical impact injury in rats: pathologic effects of varying cortical compression and impact velocity." J Neurotrauma 11(5): 587-597.

Graeber, M. B. and M. J. Christie (2012). "Multiple mechanisms of microglia: a gatekeeper's contribution to pain states." Exp Neurol 234(2): 255-261.

Greve, M. W. and B. J. Zink (2009). "Pathophysiology of traumatic brain injury." Mt Sinai J Med 76(2): 97-104.

Page 256: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

243

Guan, J., L. Bennet, S. George, D. Wu, H. J. Waldvogel, P. D. Gluckman, R. L. Faull, P. S. Crosier and A. J. Gunn (2001). "Insulin-like growth factor-1 reduces postischemic white matter injury in fetal sheep." J Cereb Blood Flow Metab 21(5): 493-502.

Guan, J., T. L. Bennet, S. George, H. J. Waldvogel, R. L. Faull, P. D. Gluckman, H. Keunen and A. J. Gunn (2000). "Selective neuroprotective effects with insulin-like growth factor-1 in phenotypic striatal neurons following ischemic brain injury in fetal sheep." Neuroscience 95(3): 831-839.

Guan, J., A. J. Gunn, E. S. Sirimanne, J. Tuffin, M. I. Gunning, R. Clark and P. D. Gluckman (2000). "The window of opportunity for neuronal rescue with insulin-like growth factor-1 after hypoxia-ischemia in rats is critically modulated by cerebral temperature during recovery." J Cereb Blood Flow Metab 20(3): 513-519.

Guan, J., O. T. Miller, K. M. Waugh, D. C. McCarthy and P. D. Gluckman (2001). "Insulin-like growth factor-1 improves somatosensory function and reduces the extent of cortical infarction and ongoing neuronal loss after hypoxia-ischemia in rats." Neuroscience 105(2): 299-306.

Guan, J., S. J. Skinner, E. J. Beilharz, K. M. Hua, S. Hodgkinson, P. D. Gluckman and C. E. Williams (1996). "The movement of IGF-I into the brain parenchyma after hypoxic-ischaemic injury." Neuroreport 7(2): 632-636.

Guan, J., C. Williams, M. Gunning, C. Mallard and P. Gluckman (1993). "The effects of IGF-1 treatment after hypoxic-ischemic brain injury in adult rats." J Cereb Blood Flow Metab 13(4): 609-616.

Guler, H. P., J. Zapf and E. R. Froesch (1987). "Short-term metabolic effects of recombinant human insulin-like growth factor I in healthy adults." N Engl J Med 317(3): 137-140.

Guler, H. P., J. Zapf, C. Schmid and E. R. Froesch (1989). "Insulin-like growth factors I and II in healthy man. Estimations of half-lives and production rates." Acta Endocrinol (Copenh) 121(6): 753-758.

Hall, E. D., Y. D. Bryant, W. Cho and P. G. Sullivan (2008). "Evolution of post-traumatic neurodegeneration after controlled cortical impact traumatic brain injury in mice and rats as assessed by the de Olmos silver and fluorojade staining methods." J Neurotrauma 25(3): 235-247.

Hall, E. D., P. G. Sullivan, T. R. Gibson, K. M. Pavel, B. M. Thompson and S. W. Scheff (2005). "Spatial and temporal characteristics of neurodegeneration after controlled cortical impact in mice: more than a focal brain injury." J Neurotrauma 22(2): 252-265.

Hamm, R. J. (2001). "Neurobehavioral assessment of outcome following traumatic brain injury in rats: an evaluation of selected measures." J Neurotrauma 18(11): 1207-1216.

Page 257: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

244

Hamm, R. J., C. E. Dixon, D. M. Gbadebo, A. K. Singha, L. W. Jenkins, B. G. Lyeth and R. L. Hayes (1992). "Cognitive deficits following traumatic brain injury produced by controlled cortical impact." J Neurotrauma 9(1): 11-20.

Hannay, H. J., Z. Feldman, P. Phan, A. Keyani, N. Panwar, J. C. Goodman and C. S. Robertson (1999). "Validation of a controlled cortical impact model of head injury in mice." J Neurotrauma 16(11): 1103-1114.

Harting, M. T., F. Jimenez, S. D. Adams, D. W. Mercer and C. S. Cox, Jr. (2008). "Acute, regional inflammatory response after traumatic brain injury: Implications for cellular therapy." Surgery 144(5): 803-813.

Hatton, J., R. Kryscio, M. Ryan, L. Ott and B. Young (2006). "Systemic metabolic effects of combined insulin-like growth factor-I and growth hormone therapy in patients who have sustained acute traumatic brain injury." J Neurosurg 105(6): 843-852.

Hatton, J., R. P. Rapp, K. A. Kudsk, R. O. Brown, M. S. Luer, J. G. Bukar, S. A. Chen, C. J. McClain, N. Gesundheit, R. J. Dempsey and B. Young (1997). "Intravenous insulin-like growth factor-I (IGF-I) in moderate-to-severe head injury: a phase II safety and efficacy trial." J Neurosurg 86(5): 779-786.

Hausmann, R. and P. Betz (2000). "The time course of the vascular response to human brain injury--an immunohistochemical study." Int J Legal Med 113(5): 288-292.

Hayes, R. L., D. Stalhammar, J. T. Povlishock, A. M. Allen, B. J. Galinat, D. P. Becker and H. H. Stonnington (1987). "A new model of concussive brain injury in the cat produced by extradural fluid volume loading: II. Physiological and neuropathological observations." Brain Inj 1(1): 93-112.

Hicks, R., H. Soares, D. Smith and T. McIntosh (1996). "Temporal and spatial characterization of neuronal injury following lateral fluid-percussion brain injury in the rat." Acta Neuropathol 91(3): 236-246.

Hicks, R. R., S. A. Baldwin and S. W. Scheff (1997). "Serum extravasation and cytoskeletal alterations following traumatic brain injury in rats. Comparison of lateral fluid percussion and cortical impact models." Mol Chem Neuropathol 32(1-3): 1-16.

Hollis, E. R., 2nd, P. Lu, A. Blesch and M. H. Tuszynski (2009). "IGF-I gene delivery promotes corticospinal neuronal survival but not regeneration after adult CNS injury." Exp Neurol 215(1): 53-59.

Holly, J. M. and J. A. Wass (1989). "Insulin-like growth factors; autocrine, paracrine or endocrine? New perspectives of the somatomedin hypothesis in the light of recent developments." J Endocrinol 122(3): 611-618.

Holmin, S., T. Mathiesen, J. Shetye and P. Biberfeld (1995). "Intracerebral inflammatory response to experimental brain contusion." Acta Neurochir (Wien) 132(1-3): 110-119.

Page 258: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

245

Horwood, J. M., F. Dufour, S. Laroche and S. Davis (2006). "Signalling mechanisms mediated by the phosphoinositide 3-kinase/Akt cascade in synaptic plasticity and memory in the rat." Eur J Neurosci 23(12): 3375-3384.

Hsieh, J., J. B. Aimone, B. K. Kaspar, T. Kuwabara, K. Nakashima and F. H. Gage (2004). "IGF-I instructs multipotent adult neural progenitor cells to become oligodendrocytes." J Cell Biol 164(1): 111-122.

Huh, J. W., A. G. Widing and R. Raghupathi (2011). "Differential effects of injury severity on cognition and cellular pathology after contusive brain trauma in the immature rat." J Neurotrauma 28(2): 245-257.

Hung, K. S., S. H. Tsai, T. C. Lee, J. W. Lin, C. K. Chang and W. T. Chiu (2007). "Gene transfer of insulin-like growth factor-I providing neuroprotection after spinal cord injury in rats." J Neurosurg Spine 6(1): 35-46.

Hunt, R. F., S. W. Scheff and B. N. Smith (2009). "Posttraumatic epilepsy after controlled cortical impact injury in mice." Exp Neurol 215(2): 243-252.

Hunt, R. F., S. W. Scheff and B. N. Smith (2011). "Synaptic reorganization of inhibitory hilar interneuron circuitry after traumatic brain injury in mice." J Neurosci 31(18): 6880-6890.

Huttenlocher, A., S. P. Palecek, Q. Lu, W. Zhang, R. L. Mellgren, D. A. Lauffenburger, M. H. Ginsberg and A. F. Horwitz (1997). "Regulation of cell migration by the calcium-dependent protease calpain." J Biol Chem 272(52): 32719-32722.

Jang, S. H. (2009). "Review of motor recovery in patients with traumatic brain injury." NeuroRehabilitation 24(4): 349-353.

Jessberger, S., R. E. Clark, N. J. Broadbent, G. D. Clemenson, Jr., A. Consiglio, D. C. Lie, L. R. Squire and F. H. Gage (2009). "Dentate gyrus-specific knockdown of adult neurogenesis impairs spatial and object recognition memory in adult rats." Learn Mem 16(2): 147-154.

Jessberger, S. and G. Kempermann (2003). "Adult-born hippocampal neurons mature into activity-dependent responsiveness." Eur J Neurosci 18(10): 2707-2712.

Johanson, C. E., J. Szmydynger-Chodobska, A. Chodobski, A. Baird, P. McMillan and E. G. Stopa (1999). "Altered formation and bulk absorption of cerebrospinal fluid in FGF-2-induced hydrocephalus." Am J Physiol 277(1 Pt 2): R263-271.

Jones, N. C., M. J. Prior, E. Burden-Teh, C. A. Marsden, P. G. Morris and S. Murphy (2005). "Antagonism of the interleukin-1 receptor following traumatic brain injury in the mouse reduces the number of nitric oxide synthase-2-positive cells and improves anatomical and functional outcomes." Eur J Neurosci 22(1): 72-78.

Jones, S. P., O. Rahimi, M. P. O'Boyle, D. L. Diaz and B. J. Claiborne (2003). "Maturation of granule cell dendrites after mossy fiber arrival in hippocampal field CA3." Hippocampus 13(3): 413-427.

Page 259: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

246

Karpova, A., P. P. Sanna and T. Behnisch (2006). "Involvement of multiple phosphatidylinositol 3-kinase-dependent pathways in the persistence of late-phase long term potentiation expression." Neuroscience 137(3): 833-841.

Kazanis, I., M. Giannakopoulou, H. Philippidis and F. Stylianopoulou (2004). "Alterations in IGF-I, BDNF and NT-3 levels following experimental brain trauma and the effect of IGF-I administration." Exp Neurol 186(2): 221-234.

Kee, N., C. M. Teixeira, A. H. Wang and P. W. Frankland (2007). "Preferential incorporation of adult-generated granule cells into spatial memory networks in the dentate gyrus." Nat Neurosci 10(3): 355-362.

Kempermann, G., K. Fabel, D. Ehninger, H. Babu, P. Leal-Galicia, A. Garthe and S. A. Wolf (2010). "Why and how physical activity promotes experience-induced brain plasticity." Front Neurosci 4: 189.

Kenne, E., A. Erlandsson, L. Lindbom, L. Hillered and F. Clausen (2012). "Neutrophil depletion reduces edema formation and tissue loss following traumatic brain injury in mice." J Neuroinflammation 9: 17.

Kernie, S. G., T. M. Erwin and L. F. Parada (2001). "Brain remodeling due to neuronal and astrocytic proliferation after controlled cortical injury in mice." J Neurosci Res 66(3): 317-326.

Kilinc, D., G. Gallo and K. A. Barbee (2009). "Mechanical membrane injury induces axonal beading through localized activation of calpain." Exp Neurol 219(2): 553-561.

Kizhakke Madathil S, P. J., Carlson S, Saatman KE (2010). Insulin-like growth factor-1 overexpression favours brain remodeling and improves motor and cognitive function after traumatic brain injury, Journal of Neurotrauma. May 2010, 27(5): Poster 224. doi:10.1089/neu.2010.9950.

Kleindienst, A., M. J. McGinn, H. B. Harvey, R. J. Colello, R. J. Hamm and M. R. Bullock (2005). "Enhanced hippocampal neurogenesis by intraventricular S100B infusion is associated with improved cognitive recovery after traumatic brain injury." J Neurotrauma 22(6): 645-655.

Kochanek, P. M., D. W. Marion, W. Zhang, J. K. Schiding, M. White, A. M. Palmer, R. S. Clark, M. E. O'Malley, S. D. Styren, C. Ho and et al. (1995). "Severe controlled cortical impact in rats: assessment of cerebral edema, blood flow, and contusion volume." J Neurotrauma 12(6): 1015-1025.

Komoly, S., L. D. Hudson, H. D. Webster and C. A. Bondy (1992). "Insulin-like growth factor I gene expression is induced in astrocytes during experimental demyelination." Proc Natl Acad Sci U S A 89(5): 1894-1898.

Kreutzberg, G. W. (1996). "Microglia: a sensor for pathological events in the CNS." Trends Neurosci 19(8): 312-318.

Page 260: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

247

Krishnamurthy, S., J. Li, L. Schultz and J. P. McAllister, 2nd (2009). "Intraventricular infusion of hyperosmolar dextran induces hydrocephalus: a novel animal model of hydrocephalus." Cerebrospinal Fluid Res 6: 16.

Kurland, D., C. Hong, B. Aarabi, V. Gerzanich and J. M. Simard (2012). "Hemorrhagic progression of a contusion after traumatic brain injury: a review." J Neurotrauma 29(1): 19-31.

Lam, X. M., E. T. Duenas, A. L. Daugherty, N. Levin and J. L. Cleland (2000). "Sustained release of recombinant human insulin-like growth factor-I for treatment of diabetes." J Control Release 67(2-3): 281-292.

Langlois, J. A., W. Rutland-Brown and M. M. Wald (2006). "The epidemiology and impact of traumatic brain injury: a brief overview." J Head Trauma Rehabil 21(5): 375-378.

LaPlaca, M. C., V. M. Lee and L. E. Thibault (1997). "An in vitro model of traumatic neuronal injury: loading rate-dependent changes in acute cytosolic calcium and lactate dehydrogenase release." J Neurotrauma 14(6): 355-368.

Laviola, L., A. Natalicchio and F. Giorgino (2007). "The IGF-I signaling pathway." Curr Pharm Des 13(7): 663-669.

Lee, W. H., J. A. Clemens and C. A. Bondy (1992). "Insulin-like growth factors in the response to cerebral ischemia." Mol Cell Neurosci 3(1): 36-43.

Levin, H. S. (1998). "Cognitive function outcomes after traumatic brain injury." Curr Opin Neurol 11(6): 643-646.

Levin, H. S., E. A. Wilde, Z. Chu, R. Yallampalli, G. R. Hanten, X. Li, J. Chia, A. C. Vasquez and J. V. Hunter (2008). "Diffusion tensor imaging in relation to cognitive and functional outcome of traumatic brain injury in children." J Head Trauma Rehabil 23(4): 197-208.

Lichtenwalner, R. J., M. E. Forbes, S. A. Bennett, C. D. Lynch, W. E. Sonntag and D. R. Riddle (2001). "Intracerebroventricular infusion of insulin-like growth factor-I ameliorates the age-related decline in hippocampal neurogenesis." Neuroscience 107(4): 603-613.

Lighthall, J. W. (1988). "Controlled cortical impact: a new experimental brain injury model." J Neurotrauma 5(1): 1-15.

Lighthall, J. W., H. G. Goshgarian and C. R. Pinderski (1990). "Characterization of axonal injury produced by controlled cortical impact." J Neurotrauma 7(2): 65-76.

Lin, S., L. W. Fan, P. G. Rhodes and Z. Cai (2009). "Intranasal administration of IGF-1 attenuates hypoxic-ischemic brain injury in neonatal rats." Exp Neurol 217(2): 361-370.

Page 261: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

248

Liu, X. F., J. R. Fawcett, R. G. Thorne and W. H. Frey, 2nd (2001). "Non-invasive intranasal insulin-like growth factor-I reduces infarct volume and improves neurologic function in rats following middle cerebral artery occlusion." Neurosci Lett 308(2): 91-94.

Llorens-Martin, M., I. Torres-Aleman and J. L. Trejo (2010). "Exercise modulates insulin-like growth factor 1-dependent and -independent effects on adult hippocampal neurogenesis and behaviour." Mol Cell Neurosci 44(2): 109-117.

Logan, A., S. A. Frautschy, A. M. Gonzalez and A. Baird (1992). "A time course for the focal elevation of synthesis of basic fibroblast growth factor and one of its high-affinity receptors (flg) following a localized cortical brain injury." J Neurosci 12(10): 3828-3837.

Longhi, L., C. Perego, F. Ortolano, E. R. Zanier, P. Bianchi, N. Stocchetti, T. K. McIntosh and M. G. De Simoni (2009). "C1-inhibitor attenuates neurobehavioral deficits and reduces contusion volume after controlled cortical impact brain injury in mice." Crit Care Med 37(2): 659-665.

Lotocki, G., J. P. de Rivero Vaccari, O. Alonso, J. S. Molano, R. Nixon, P. Safavi, W. D. Dietrich and H. M. Bramlett (2011). "Oligodendrocyte vulnerability following traumatic brain injury in rats." Neurosci Lett 499(3): 143-148.

Lu, D., A. Mahmood, C. Qu, A. Goussev, T. Schallert and M. Chopp (2005). "Erythropoietin enhances neurogenesis and restores spatial memory in rats after traumatic brain injury." J Neurotrauma 22(9): 1011-1017.

Ludwig, T., J. Eggenschwiler, P. Fisher, A. J. D'Ercole, M. L. Davenport and A. Efstratiadis (1996). "Mouse mutants lacking the type 2 IGF receptor (IGF2R) are rescued from perinatal lethality in Igf2 and Igf1r null backgrounds." Dev Biol 177(2): 517-535.

Lugert, S., O. Basak, P. Knuckles, U. Haussler, K. Fabel, M. Gotz, C. A. Haas, G. Kempermann, V. Taylor and C. Giachino (2010). "Quiescent and active hippocampal neural stem cells with distinct morphologies respond selectively to physiological and pathological stimuli and aging." Cell Stem Cell 6(5): 445-456.

Lundin, A., C. de Boussard, G. Edman and J. Borg (2006). "Symptoms and disability until 3 months after mild TBI." Brain Inj 20(8): 799-806.

Lyeth, B. G., L. W. Jenkins, R. J. Hamm, C. E. Dixon, L. L. Phillips, G. L. Clifton, H. F. Young and R. L. Hayes (1990). "Prolonged memory impairment in the absence of hippocampal cell death following traumatic brain injury in the rat." Brain Res 526(2): 249-258.

Lynch, D. R. and T. M. Dawson (1994). "Secondary mechanisms in neuronal trauma." Curr Opin Neurol 7(6): 510-516.

M.F, S. K. E. S. G. B. (2009). Axonal damage due to traumatic brain injury. NY, Springer.

Page 262: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

249

Ma, Y., P. R. Hof, S. C. Grant, S. J. Blackband, R. Bennett, L. Slatest, M. D. McGuigan and H. Benveniste (2005). "A three-dimensional digital atlas database of the adult C57BL/6J mouse brain by magnetic resonance microscopy." Neuroscience 135(4): 1203-1215.

Maas, A. I., C. W. Hukkelhoven, L. F. Marshall and E. W. Steyerberg (2005). "Prediction of outcome in traumatic brain injury with computed tomographic characteristics: a comparison between the computed tomographic classification and combinations of computed tomographic predictors." Neurosurgery 57(6): 1173-1182; discussion 1173-1182.

Maas, A. I., A. Marmarou, G. D. Murray, S. G. Teasdale and E. W. Steyerberg (2007). "Prognosis and clinical trial design in traumatic brain injury: the IMPACT study." J Neurotrauma 24(2): 232-238.

Maas, A. I., B. Roozenbeek and G. T. Manley (2010). "Clinical trials in traumatic brain injury: past experience and current developments." Neurotherapeutics 7(1): 115-126.

Maas, A. I., E. W. Steyerberg, G. D. Murray, R. Bullock, A. Baethmann, L. F. Marshall and G. M. Teasdale (1999). "Why have recent trials of neuroprotective agents in head injury failed to show convincing efficacy? A pragmatic analysis and theoretical considerations." Neurosurgery 44(6): 1286-1298.

Maas, A. I., N. Stocchetti and R. Bullock (2008). "Moderate and severe traumatic brain injury in adults." Lancet Neurol 7(8): 728-741.

Mac Donald, C. L., K. Dikranian, S. K. Song, P. V. Bayly, D. M. Holtzman and D. L. Brody (2007). "Detection of traumatic axonal injury with diffusion tensor imaging in a mouse model of traumatic brain injury." Exp Neurol 205(1): 116-131.

Madathil, S. K., Carlson S. W., Brelsfoard J.M., Ping, Y., D`Ercole A.J., Saatman, K.E. (2013). "Astrocyte-specific overexpression of insulin-like growth factor-1 protects hippocampal

neurons and reduces behavioral deficits following traumatic brain injury in mice." PLOS ONE.

Madathil, S. K., H. N. Evans and K. E. Saatman (2010). "Temporal and regional changes in IGF-1/IGF-1R signaling in the mouse brain after traumatic brain injury." J Neurotrauma 27(1): 95-107.

Mahmood, A., D. Lu and M. Chopp (2004). "Intravenous administration of marrow stromal cells (MSCs) increases the expression of growth factors in rat brain after traumatic brain injury." J Neurotrauma 21(1): 33-39.

Manning, B. D. and L. C. Cantley (2007). "AKT/PKB signaling: navigating downstream." Cell 129(7): 1261-1274.

Page 263: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

250

Mao, H., F. Guan, X. Han and K. H. Yang (2011). "Strain-based regional traumatic brain injury intensity in controlled cortical impact: a systematic numerical analysis." J Neurotrauma 28(11): 2263-2276.

Mao, H., X. Jin, L. Zhang, K. H. Yang, T. Igarashi, L. J. Noble-Haeusslein and A. I. King (2010). "Finite element analysis of controlled cortical impact-induced cell loss." J Neurotrauma 27(5): 877-888.

Mao, H., K. H. Yang, A. I. King and K. Yang (2010). "Computational neurotrauma-design, simulation, and analysis of controlled cortical impact model." Biomech Model Mechanobiol.

Mao, H., L. Zhang, K. H. Yang and A. I. King (2006). "Application of a finite element model of the brain to study traumatic brain injury mechanisms in the rat." Stapp Car Crash J 50: 583-600.

Margulies, S. and R. Hicks (2009). "Combination therapies for traumatic brain injury: prospective considerations." J Neurotrauma 26(6): 925-939.

Marklund, N., A. Bakshi, D. J. Castelbuono, V. Conte and T. K. McIntosh (2006). "Evaluation of pharmacological treatment strategies in traumatic brain injury." Curr Pharm Des 12(13): 1645-1680.

Marklund, N., C. Keck, R. Hoover, K. Soltesz, M. Millard, D. LeBold, Z. Spangler, A. Banning, J. Benson and T. K. McIntosh (2005). "Administration of monoclonal antibodies neutralizing the inflammatory mediators tumor necrosis factor alpha and interleukin -6 does not attenuate acute behavioral deficits following experimental traumatic brain injury in the rat." Restor Neurol Neurosci 23(1): 31-42.

Marmarou, A., M. A. Foda, W. van den Brink, J. Campbell, H. Kita and K. Demetriadou (1994). "A new model of diffuse brain injury in rats. Part I: Pathophysiology and biomechanics." J Neurosurg 80(2): 291-300.

Marmarou, A., K. Shulman and R. M. Rosende (1978). "A nonlinear analysis of the cerebrospinal fluid system and intracranial pressure dynamics." J Neurosurg 48(3): 332-344.

Marshall, L. F., S. B. Marshall, M. R. Klauber, M. Van Berkum Clark, H. Eisenberg, J. A. Jane, T. G. Luerssen, A. Marmarou and M. A. Foulkes (1992). "The diagnosis of head injury requires a classification based on computed axial tomography." J Neurotrauma 9 Suppl 1: S287-292.

Martin, N. A., R. V. Patwardhan, M. J. Alexander, C. Z. Africk, J. H. Lee, E. Shalmon, D. A. Hovda and D. P. Becker (1997). "Characterization of cerebral hemodynamic phases following severe head trauma: hypoperfusion, hyperemia, and vasospasm." J Neurosurg 87(1): 9-19.

Page 264: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

251

Mason, J. L., P. Ye, K. Suzuki, A. J. D'Ercole and G. K. Matsushima (2000). "Insulin-like growth factor-1 inhibits mature oligodendrocyte apoptosis during primary demyelination." J Neurosci 20(15): 5703-5708.

Mbye, L. H., I. N. Singh, K. M. Carrico, K. E. Saatman and E. D. Hall (2009). "Comparative neuroprotective effects of cyclosporin A and NIM811, a nonimmunosuppressive cyclosporin A analog, following traumatic brain injury." J Cereb Blood Flow Metab 29(1): 87-97.

McCurdy, R. D., F. Feron, J. J. McGrath and A. Mackay-Sim (2005). "Regulation of adult olfactory neurogenesis by insulin-like growth factor-I." Eur J Neurosci 22(7): 1581-1588.

McDermott, K. L., R. Raghupathi, S. C. Fernandez, K. E. Saatman, A. A. Protter, S. P. Finklestein, G. Sinson, D. H. Smith and T. K. McIntosh (1997). "Delayed administration of basic fibroblast growth factor (bFGF) attenuates cognitive dysfunction following parasagittal fluid percussion brain injury in the rat." J Neurotrauma 14(4): 191-200.

McIntosh, T. K., L. Noble, B. Andrews and A. I. Faden (1987). "Traumatic brain injury in the rat: characterization of a midline fluid-percussion model." Cent Nerv Syst Trauma 4(2): 119-134.

McIntosh, T. K., K. E. Saatman, R. Raghupathi, D. I. Graham, D. H. Smith, V. M. Lee and J. Q. Trojanowski (1998). "The Dorothy Russell Memorial Lecture. The molecular and cellular sequelae of experimental traumatic brain injury: pathogenetic mechanisms." Neuropathol Appl Neurobiol 24(4): 251-267.

McIntosh, T. K., R. Vink, L. Noble, I. Yamakami, S. Fernyak, H. Soares and A. L. Faden (1989). "Traumatic brain injury in the rat: characterization of a lateral fluid-percussion model." Neuroscience 28(1): 233-244.

McKeon, R. J., R. C. Schreiber, J. S. Rudge and J. Silver (1991). "Reduction of neurite outgrowth in a model of glial scarring following CNS injury is correlated with the expression of inhibitory molecules on reactive astrocytes." J Neurosci 11(11): 3398-3411.

Meaney, D. F., S. S. Margulies and D. H. Smith (2001). "Diffuse axonal injury." J Neurosurg 95(6): 1108-1110.

Meaney, D. F., D. T. Ross, B. A. Winkelstein, J. Brasko, D. Goldstein, L. B. Bilston, L. E. Thibault and T. A. Gennarelli (1994). "Modification of the cortical impact model to produce axonal injury in the rat cerebral cortex." J Neurotrauma 11(5): 599-612.

Meinel, L., O. E. Illi, J. Zapf, M. Malfanti, H. Peter Merkle and B. Gander (2001). "Stabilizing insulin-like growth factor-I in poly(D,L-lactide-co-glycolide) microspheres." J Control Release 70(1-2): 193-202.

Metzger, F., W. Sajid, S. Saenger, C. Staudenmaier, C. van der Poel, B. Sobottka, A. Schuler, M. Sawitzky, R. Poirier, D. Tuerck, E. Schick, A. Schaubmar, F. Hesse,

Page 265: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

252

K. Amrein, H. Loetscher, G. S. Lynch, A. Hoeflich, P. De Meyts and H. J. Schoenfeld (2011). "Separation of fast from slow anabolism by site-specific PEGylation of insulin-like growth factor I (IGF-I)." J Biol Chem 286(22): 19501-19510.

Milla, P., F. Dosio and L. Cattel (2012). "PEGylation of proteins and liposomes: a powerful and flexible strategy to improve the drug delivery." Curr Drug Metab 13(1): 105-119.

Ming, G. L. and H. Song (2011). "Adult neurogenesis in the mammalian brain: significant answers and significant questions." Neuron 70(4): 687-702.

Miyazaki, S., Y. Katayama, B. G. Lyeth, L. W. Jenkins, D. S. DeWitt, S. J. Goldberg, P. G. Newlon and R. L. Hayes (1992). "Enduring suppression of hippocampal long-term potentiation following traumatic brain injury in rat." Brain Res 585(1-2): 335-339.

Monson, K. L., W. Goldsmith, N. M. Barbaro and G. T. Manley (2003). "Axial mechanical properties of fresh human cerebral blood vessels." J Biomech Eng 125(2): 288-294.

Morales, D. M., N. Marklund, D. Lebold, H. J. Thompson, A. Pitkanen, W. L. Maxwell, L. Longhi, H. Laurer, M. Maegele, E. Neugebauer, D. I. Graham, N. Stocchetti and T. K. McIntosh (2005). "Experimental models of traumatic brain injury: do we really need to build a better mousetrap?" Neuroscience 136(4): 971-989.

Morganti-Kossman, M. C., P. M. Lenzlinger, V. Hans, P. Stahel, E. Csuka, E. Ammann, R. Stocker, O. Trentz and T. Kossmann (1997). "Production of cytokines following brain injury: beneficial and deleterious for the damaged tissue." Mol Psychiatry 2(2): 133-136.

Morganti-Kossmann, M. C., M. Rancan, P. F. Stahel and T. Kossmann (2002). "Inflammatory response in acute traumatic brain injury: a double-edged sword." Curr Opin Crit Care 8(2): 101-105.

Muizelaar, J. P., E. P. Wei, H. A. Kontos and D. P. Becker (1983). "Mannitol causes compensatory cerebral vasoconstriction and vasodilation in response to blood viscosity changes." J Neurosurg 59(5): 822-828.

Munch, E., P. Horn, L. Schurer, A. Piepgras, T. Paul and P. Schmiedek (2000). "Management of severe traumatic brain injury by decompressive craniectomy." Neurosurgery 47(2): 315-322; discussion 322-313.

Myer, D. J., G. G. Gurkoff, S. M. Lee, D. A. Hovda and M. V. Sofroniew (2006). "Essential protective roles of reactive astrocytes in traumatic brain injury." Brain 129(Pt 10): 2761-2772.

Nagaraja, T. N., P. Patel, M. Gorski, P. D. Gorevic, C. S. Patlak and J. D. Fenstermacher (2005). "In normal rat, intraventricularly administered insulin-like

Page 266: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

253

growth factor-1 is rapidly cleared from CSF with limited distribution into brain." Cerebrospinal Fluid Res 2: 5.

Nakamura, M., K. E. Saatman, J. E. Galvin, U. Scherbel, R. Raghupathi, J. Q. Trojanowski and T. K. McIntosh (1999). "Increased vulnerability of NFH-LacZ transgenic mouse to traumatic brain injury-induced behavioral deficits and cortical damage." J Cereb Blood Flow Metab 19(7): 762-770.

Narayan, R. K., M. E. Michel, B. Ansell, A. Baethmann, A. Biegon, M. B. Bracken, M. R. Bullock, S. C. Choi, G. L. Clifton, C. F. Contant, W. M. Coplin, W. D. Dietrich, J. Ghajar, S. M. Grady, R. G. Grossman, E. D. Hall, W. Heetderks, D. A. Hovda, J. Jallo, R. L. Katz, N. Knoller, P. M. Kochanek, A. I. Maas, J. Majde, D. W. Marion, A. Marmarou, L. F. Marshall, T. K. McIntosh, E. Miller, N. Mohberg, J. P. Muizelaar, L. H. Pitts, P. Quinn, G. Riesenfeld, C. S. Robertson, K. I. Strauss, G. Teasdale, N. Temkin, R. Tuma, C. Wade, M. D. Walker, M. Weinrich, J. Whyte, J. Wilberger, A. B. Young and L. Yurkewicz (2002). "Clinical trials in head injury." J Neurotrauma 19(5): 503-557.

Narhi, L. O., Q. X. Hua, T. Arakawa, G. M. Fox, L. Tsai, R. Rosenfeld, P. Holst, J. A. Miller and M. A. Weiss (1993). "Role of native disulfide bonds in the structure and activity of insulin-like growth factor 1: genetic models of protein-folding intermediates." Biochemistry 32(19): 5214-5221.

Newcomb, J. K., X. Zhao, B. R. Pike and R. L. Hayes (1999). "Temporal profile of apoptotic-like changes in neurons and astrocytes following controlled cortical impact injury in the rat." Exp Neurol 158(1): 76-88.

Niblock, M. M., J. K. Brunso-Bechtold and D. R. Riddle (2000). "Insulin-like growth factor I stimulates dendritic growth in primary somatosensory cortex." J Neurosci 20(11): 4165-4176.

Nishibe, M., S. Barbay, D. Guggenmos and R. J. Nudo (2010). "Reorganization of motor cortex after controlled cortical impact in rats and implications for functional recovery." J Neurotrauma 27(12): 2221-2232.

Nishijima, T., J. Piriz, S. Duflot, A. M. Fernandez, G. Gaitan, U. Gomez-Pinedo, J. M. Verdugo, F. Leroy, H. Soya, A. Nunez and I. Torres-Aleman (2010). "Neuronal activity drives localized blood-brain-barrier transport of serum insulin-like growth factor-I into the CNS." Neuron 67(5): 834-846.

Norris, C. M. and S. W. Scheff (2009). "Recovery of afferent function and synaptic strength in hippocampal CA1 following traumatic brain injury." J Neurotrauma 26(12): 2269-2278.

Noshita, N., A. Lewen, T. Sugawara and P. H. Chan (2001). "Evidence of phosphorylation of Akt and neuronal survival after transient focal cerebral ischemia in mice." J Cereb Blood Flow Metab 21(12): 1442-1450.

Page 267: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

254

Noshita, N., A. Lewen, T. Sugawara and P. H. Chan (2002). "Akt phosphorylation and neuronal survival after traumatic brain injury in mice." Neurobiol Dis 9(3): 294-304.

O'Kusky, J. R., P. Ye and A. J. D'Ercole (2000). "Insulin-like growth factor-I promotes neurogenesis and synaptogenesis in the hippocampal dentate gyrus during postnatal development." J Neurosci 20(22): 8435-8442.

Ozdinler, P. H. and J. D. Macklis (2006). "IGF-I specifically enhances axon outgrowth of corticospinal motor neurons." Nat Neurosci 9(11): 1371-1381.

Pardridge, W. M. (1991). Transnasal and intraventricular delivery. New York, Raven Press.

Pardridge, W. M. (1993). "Transport of insulin-related peptides and glucose across the blood-brain barrier." Ann N Y Acad Sci 692: 126-137.

Parent, J. M., T. W. Yu, R. T. Leibowitz, D. H. Geschwind, R. S. Sloviter and D. H. Lowenstein (1997). "Dentate granule cell neurogenesis is increased by seizures and contributes to aberrant network reorganization in the adult rat hippocampus." J Neurosci 17(10): 3727-3738.

Paxinos, G. and K. B. J. Franklin (2001). The mouse brain in stereotaxic coordinates. San Diego, CA, Academic Press.

Peruzzi, F., M. Prisco, M. Dews, P. Salomoni, E. Grassilli, G. Romano, B. Calabretta and R. Baserga (1999). "Multiple signaling pathways of the insulin-like growth factor 1 receptor in protection from apoptosis." Mol Cell Biol 19(10): 7203-7215.

Philips, M. F., G. Mattiasson, T. Wieloch, A. Bjorklund, B. B. Johansson, G. Tomasevic, A. Martinez-Serrano, P. M. Lenzlinger, G. Sinson, M. S. Grady and T. K. McIntosh (2001). "Neuroprotective and behavioral efficacy of nerve growth factor-transfected hippocampal progenitor cell transplants after experimental traumatic brain injury." J Neurosurg 94(5): 765-774.

Pierce, J. E., D. H. Smith, J. Q. Trojanowski and T. K. McIntosh (1998). "Enduring cognitive, neurobehavioral and histopathological changes persist for up to one year following severe experimental brain injury in rats." Neuroscience 87(2): 359-369.

Pike, B. R., X. Zhao, J. K. Newcomb, C. C. Glenn, D. K. Anderson and R. L. Hayes (2000). "Stretch injury causes calpain and caspase-3 activation and necrotic and apoptotic cell death in septo-hippocampal cell cultures." J Neurotrauma 17(4): 283-298.

Pleasant, J. M., S. W. Carlson, H. Mao, S. W. Scheff, K. H. Yang and K. E. Saatman (2011). "Rate of neurodegeneration in the mouse controlled cortical impact model is influenced by impactor tip shape: implications for mechanistic and therapeutic studies." J Neurotrauma 28(11): 2245-2262.

Page 268: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

255

Popken, G. J., R. D. Hodge, P. Ye, J. Zhang, W. Ng, J. R. O'Kusky and A. J. D'Ercole (2004). "In vivo effects of insulin-like growth factor-I (IGF-I) on prenatal and early postnatal development of the central nervous system." Eur J Neurosci 19(8): 2056-2068.

Posmantur, R., R. L. Hayes, C. E. Dixon and W. C. Taft (1994). "Neurofilament 68 and neurofilament 200 protein levels decrease after traumatic brain injury." J Neurotrauma 11(5): 533-545.

Povlishock, J. T. (1992). "Traumatically induced axonal injury: pathogenesis and pathobiological implications." Brain Pathol 2(1): 1-12.

Povlishock, J. T. and D. I. Katz (2005). "Update of neuropathology and neurological recovery after traumatic brain injury." J Head Trauma Rehabil 20(1): 76-94.

Prins, M. L., D. Alexander, C. C. Giza and D. A. Hovda (2013). "Repeated mild traumatic brain injury: mechanisms of cerebral vulnerability." J Neurotrauma 30(1): 30-38.

Prins, M. L. and D. A. Hovda (2001). "Mapping cerebral glucose metabolism during spatial learning: interactions of development and traumatic brain injury." J Neurotrauma 18(1): 31-46.

Pullela, R., J. Raber, T. Pfankuch, D. M. Ferriero, C. P. Claus, S. E. Koh, T. Yamauchi, R. Rola, J. R. Fike and L. J. Noble-Haeusslein (2006). "Traumatic injury to the immature brain results in progressive neuronal loss, hyperactivity and delayed cognitive impairments." Dev Neurosci 28(4-5): 396-409.

Ramaswamy, S., G. E. Goings, K. E. Soderstrom, F. G. Szele and D. A. Kozlowski (2005). "Cellular proliferation and migration following a controlled cortical impact in the mouse." Brain Res 1053(1-2): 38-53.

Reeves, T. M., B. G. Lyeth and J. T. Povlishock (1995). "Long-term potentiation deficits and excitability changes following traumatic brain injury." Exp Brain Res 106(2): 248-256.

Reinhardt, R. R. and C. A. Bondy (1994). "Insulin-like growth factors cross the blood-brain barrier." Endocrinology 135(5): 1753-1761.

Ribak, C. E., M. J. Korn, Z. Shan and A. Obenaus (2004). "Dendritic growth cones and recurrent basal dendrites are typical features of newly generated dentate granule cells in the adult hippocampus." Brain Res 1000(1-2): 195-199.

Rice, A. C., A. Khaldi, H. B. Harvey, N. J. Salman, F. White, H. Fillmore and M. R. Bullock (2003). "Proliferation and neuronal differentiation of mitotically active cells following traumatic brain injury." Exp Neurol 183(2): 406-417.

Ridet, J. L., S. K. Malhotra, A. Privat and F. H. Gage (1997). "Reactive astrocytes: cellular and molecular cues to biological function." Trends Neurosci 20(12): 570-577.

Page 269: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

256

Rinderknecht, E. and R. E. Humbel (1978). "The amino acid sequence of human insulin-like growth factor I and its structural homology with proinsulin." J Biol Chem 253(8): 2769-2776.

Rink, A., K. M. Fung, J. Q. Trojanowski, V. M. Lee, E. Neugebauer and T. K. McIntosh (1995). "Evidence of apoptotic cell death after experimental traumatic brain injury in the rat." Am J Pathol 147(6): 1575-1583.

Rockich, K. T., J. C. Hatton, R. J. Kryscio, B. A. Young and R. A. Blouin (1999). "Effect of recombinant human growth hormone and insulin-like growth factor-1 administration on IGF-1 and IGF-binding protein-3 levels in brain injury." Pharmacotherapy 19(12): 1432-1436.

Rola, R., S. Mizumatsu, S. Otsuka, D. R. Morhardt, L. J. Noble-Haeusslein, K. Fishman, M. B. Potts and J. R. Fike (2006). "Alterations in hippocampal neurogenesis following traumatic brain injury in mice." Exp Neurol 202(1): 189-199.

Royo, N. C., V. Conte, K. E. Saatman, S. Shimizu, C. M. Belfield, K. M. Soltesz, J. E. Davis, S. T. Fujimoto and T. K. McIntosh (2006). "Hippocampal vulnerability following traumatic brain injury: a potential role for neurotrophin-4/5 in pyramidal cell neuroprotection." Eur J Neurosci 23(5): 1089-1102.

Rubovitch, V., S. Edut, R. Sarfstein, H. Werner and C. G. Pick (2010). "The intricate involvement of the Insulin-like growth factor receptor signaling in mild traumatic brain injury in mice." Neurobiol Dis 38(2): 299-303.

Russell, J. W., A. J. Windebank, A. Schenone and E. L. Feldman (1998). "Insulin-like growth factor-I prevents apoptosis in neurons after nerve growth factor withdrawal." J Neurobiol 36(4): 455-467.

Rutgers, D. R., F. Toulgoat, J. Cazejust, P. Fillard, P. Lasjaunias and D. Ducreux (2008). "White matter abnormalities in mild traumatic brain injury: a diffusion tensor imaging study." AJNR Am J Neuroradiol 29(3): 514-519.

Saatman, K. E., D. Bozyczko-Coyne, V. Marcy, R. Siman and T. K. McIntosh (1996). "Prolonged calpain-mediated spectrin breakdown occurs regionally following experimental brain injury in the rat." J Neuropathol Exp Neurol 55(7): 850-860.

Saatman, K. E., P. C. Contreras, D. H. Smith, R. Raghupathi, K. L. McDermott, S. C. Fernandez, K. L. Sanderson, M. Voddi and T. K. McIntosh (1997). "Insulin-like growth factor-1 (IGF-1) improves both neurological motor and cognitive outcome following experimental brain injury." Exp Neurol 147(2): 418-427.

Saatman, K. E., A. C. Duhaime, R. Bullock, A. I. Maas, A. Valadka and G. T. Manley (2008). "Classification of traumatic brain injury for targeted therapies." J Neurotrauma 25(7): 719-738.

Saatman, K. E., K. J. Feeko, R. L. Pape and R. Raghupathi (2006). "Differential behavioral and histopathological responses to graded cortical impact injury in mice." J Neurotrauma 23(8): 1241-1253.

Page 270: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

257

Saatman, K. E., D. I. Graham and T. K. McIntosh (1998). "The neuronal cytoskeleton is at risk after mild and moderate brain injury." J Neurotrauma 15(12): 1047-1058.

Saatman, K. E., C. Zhang, R. T. Bartus and T. K. McIntosh (2000). "Behavioral efficacy of posttraumatic calpain inhibition is not accompanied by reduced spectrin proteolysis, cortical lesion, or apoptosis." J Cereb Blood Flow Metab 20(1): 66-73.

Saenger, S., B. Holtmann, M. R. Nilges, S. Schroeder, A. Hoeflich, H. Kletzl, W. Spooren, S. Ostrowitzki, T. Hanania, M. Sendtner and F. Metzger (2012). "Functional improvement in mouse models of familial amyotrophic lateral sclerosis by PEGylated insulin-like growth factor I treatment depends on disease severity." Amyotroph Lateral Scler 13(5): 418-429.

Sahay, A., K. N. Scobie, A. S. Hill, C. M. O'Carroll, M. A. Kheirbek, N. S. Burghardt, A. A. Fenton, A. Dranovsky and R. Hen (2011). "Increasing adult hippocampal neurogenesis is sufficient to improve pattern separation." Nature 472(7344): 466-470.

Sandberg Nordqvist, A. C., H. von Holst, S. Holmin, V. R. Sara, B. M. Bellander and M. Schalling (1996). "Increase of insulin-like growth factor (IGF)-1, IGF binding protein-2 and -4 mRNAs following cerebral contusion." Brain Res Mol Brain Res 38(2): 285-293.

Sandhir, R. and N. E. Berman (2010). "Age-dependent response of CCAAT/enhancer binding proteins following traumatic brain injury in mice." Neurochem Int 56(1): 188-193.

Sandhir, R., G. Onyszchuk and N. E. Berman (2008). "Exacerbated glial response in the aged mouse hippocampus following controlled cortical impact injury." Exp Neurol 213(2): 372-380.

Santos, D. M., J. M. Xavier, A. L. Morgado, S. Sola and C. M. Rodrigues (2012). "Distinct regulatory functions of calpain 1 and 2 during neural stem cell self-renewal and differentiation." PLoS One 7(3): e33468.

Scafidi, S., J. Racz, J. Hazelton, M. C. McKenna and G. Fiskum (2010). "Neuroprotection by acetyl-L-carnitine after traumatic injury to the immature rat brain." Dev Neurosci 32(5-6): 480-487.

Schabitz, W. R., T. T. Hoffmann, S. Heiland, R. Kollmar, J. Bardutzky, C. Sommer and S. Schwab (2001). "Delayed neuroprotective effect of insulin-like growth factor-i after experimental transient focal cerebral ischemia monitored with mri." Stroke 32(5): 1226-1233.

Scharfman, H. E., J. H. Goodman and A. L. Sollas (2000). "Granule-like neurons at the hilar/CA3 border after status epilepticus and their synchrony with area CA3 pyramidal cells: functional implications of seizure-induced neurogenesis." J Neurosci 20(16): 6144-6158.

Page 271: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

258

Scheff, S. W., S. A. Baldwin, R. W. Brown and P. J. Kraemer (1997). "Morris water maze deficits in rats following traumatic brain injury: lateral controlled cortical impact." J Neurotrauma 14(9): 615-627.

Scheff, S. W., D. A. Price, R. R. Hicks, S. A. Baldwin, S. Robinson and C. Brackney (2005). "Synaptogenesis in the hippocampal CA1 field following traumatic brain injury." J Neurotrauma 22(7): 719-732.

Scherbel, U., R. Raghupathi, M. Nakamura, K. E. Saatman, J. Q. Trojanowski, E. Neugebauer, M. W. Marino and T. K. McIntosh (1999). "Differential acute and chronic responses of tumor necrosis factor-deficient mice to experimental brain injury." Proc Natl Acad Sci U S A 96(15): 8721-8726.

Schmued, L. C., C. Albertson and W. Slikker, Jr. (1997). "Fluoro-Jade: a novel fluorochrome for the sensitive and reliable histochemical localization of neuronal degeneration." Brain Res 751(1): 37-46.

Schoch, K. M., H. N. Evans, J. M. Brelsfoard, S. K. Madathil, J. Takano, T. C. Saido and K. E. Saatman (2012). "Calpastatin overexpression limits calpain-mediated proteolysis and behavioral deficits following traumatic brain injury." Exp Neurol 236(2): 371-382.

Schoch, K. M., S. K. Madathil and K. E. Saatman (2012). "Genetic manipulation of cell death and neuroplasticity pathways in traumatic brain injury." Neurotherapeutics 9(2): 323-337.

Schwarzbach, E., D. P. Bonislawski, G. Xiong and A. S. Cohen (2006). "Mechanisms underlying the inability to induce area CA1 LTP in the mouse after traumatic brain injury." Hippocampus 16(6): 541-550.

Seger, R. and E. G. Krebs (1995). "The MAPK signaling cascade." FASEB J 9(9): 726-735.

Selassie, A. W., E. Zaloshnja, J. A. Langlois, T. Miller, P. Jones and C. Steiner (2008). "Incidence of long-term disability following traumatic brain injury hospitalization, United States, 2003." J Head Trauma Rehabil 23(2): 123-131.

Selvamani, A. and F. Sohrabji (2010). "The neurotoxic effects of estrogen on ischemic stroke in older female rats is associated with age-dependent loss of insulin-like growth factor-1." J Neurosci 30(20): 6852-6861.

Serra-Grabulosa, J. M., C. Junque, K. Verger, P. Salgado-Pineda, C. Maneru and J. M. Mercader (2005). "Cerebral correlates of declarative memory dysfunctions in early traumatic brain injury." J Neurol Neurosurg Psychiatry 76(1): 129-131.

Singh, I. N., P. G. Sullivan, Y. Deng, L. H. Mbye and E. D. Hall (2006). "Time course of post-traumatic mitochondrial oxidative damage and dysfunction in a mouse model of focal traumatic brain injury: implications for neuroprotective therapy." J Cereb Blood Flow Metab 26(11): 1407-1418.

Page 272: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

259

Singh, M., B. Shirley, K. Bajwa, E. Samara, M. Hora and D. O'Hagan (2001). "Controlled release of recombinant insulin-like growth factor from a novel formulation of polylactide-co-glycolide microparticles." J Control Release 70(1-2): 21-28.

Smith, D. H., X. H. Chen, J. E. Pierce, J. A. Wolf, J. Q. Trojanowski, D. I. Graham and T. K. McIntosh (1997). "Progressive atrophy and neuron death for one year following brain trauma in the rat." J Neurotrauma 14(10): 715-727.

Smith, D. H., K. Okiyama, M. J. Thomas, B. Claussen and T. K. McIntosh (1991). "Evaluation of memory dysfunction following experimental brain injury using the Morris water maze." J Neurotrauma 8(4): 259-269.

Smith, D. H., H. D. Soares, J. S. Pierce, K. G. Perlman, K. E. Saatman, D. F. Meaney, C. E. Dixon and T. K. McIntosh (1995). "A model of parasagittal controlled cortical impact in the mouse: cognitive and histopathologic effects." J Neurotrauma 12(2): 169-178.

Smith, G. M., R. H. Miller and J. Silver (1986). "Changing role of forebrain astrocytes during development, regenerative failure, and induced regeneration upon transplantation." J Comp Neurol 251(1): 23-43.

Snow, D. M., D. A. Steindler and J. Silver (1990). "Molecular and cellular characterization of the glial roof plate of the spinal cord and optic tectum: a possible role for a proteoglycan in the development of an axon barrier." Dev Biol 138(2): 359-376.

Soares, H. D., R. R. Hicks, D. Smith and T. K. McIntosh (1995). "Inflammatory leukocytic recruitment and diffuse neuronal degeneration are separate pathological processes resulting from traumatic brain injury." J Neurosci 15(12): 8223-8233.

Sofroniew, M. V. and H. V. Vinters (2010). "Astrocytes: biology and pathology." Acta Neuropathol 119(1): 7-35.

Spain, A., S. Daumas, J. Lifshitz, J. Rhodes, P. J. Andrews, K. Horsburgh and J. H. Fowler (2010). "Mild fluid percussion injury in mice produces evolving selective axonal pathology and cognitive deficits relevant to human brain injury." J Neurotrauma 27(8): 1429-1438.

Spigelman, I., X. X. Yan, A. Obenaus, E. Y. Lee, C. G. Wasterlain and C. E. Ribak (1998). "Dentate granule cells form novel basal dendrites in a rat model of temporal lobe epilepsy." Neuroscience 86(1): 109-120.

Summers, C. R., B. Ivins and K. A. Schwab (2009). "Traumatic brain injury in the United States: an epidemiologic overview." Mt Sinai J Med 76(2): 105-110.

Sun, D., M. R. Bullock, N. Altememi, Z. Zhou, S. Hagood, A. Rolfe, M. J. McGinn, R. Hamm and R. J. Colello (2010). "The effect of epidermal growth factor in the injured brain after trauma in rats." J Neurotrauma 27(5): 923-938.

Page 273: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

260

Sun, D., M. R. Bullock, M. J. McGinn, Z. Zhou, N. Altememi, S. Hagood, R. Hamm and R. J. Colello (2009). "Basic fibroblast growth factor-enhanced neurogenesis contributes to cognitive recovery in rats following traumatic brain injury." Exp Neurol 216(1): 56-65.

Sun, D., R. J. Colello, W. P. Daugherty, T. H. Kwon, M. J. McGinn, H. B. Harvey and M. R. Bullock (2005). "Cell proliferation and neuronal differentiation in the dentate gyrus in juvenile and adult rats following traumatic brain injury." J Neurotrauma 22(1): 95-105.

Sun, D., M. J. McGinn, Z. Zhou, H. B. Harvey, M. R. Bullock and R. J. Colello (2007). "Anatomical integration of newly generated dentate granule neurons following traumatic brain injury in adult rats and its association to cognitive recovery." Exp Neurol 204(1): 264-272.

Sutton, R. L., L. Lescaudron and D. G. Stein (1993). "Unilateral cortical contusion injury in the rat: vascular disruption and temporal development of cortical necrosis." J Neurotrauma 10(2): 135-149.

Swanson, R. A., K. Farrell and B. A. Stein (1997). "Astrocyte energetics, function, and death under conditions of incomplete ischemia: a mechanism of glial death in the penumbra." Glia 21(1): 142-153.

Takadera, T., I. Matsuda and T. Ohyashiki (1999). "Apoptotic cell death and caspase-3 activation induced by N-methyl-D-aspartate receptor antagonists and their prevention by insulin-like growth factor I." J Neurochem 73(2): 548-556.

Takala, J., E. Ruokonen, N. R. Webster, M. S. Nielsen, D. F. Zandstra, G. Vundelinckx and C. J. Hinds (1999). "Increased mortality associated with growth hormone treatment in critically ill adults." N Engl J Med 341(11): 785-792.

Tang, Y. P., Y. Noda, T. Hasegawa and T. Nabeshima (1997). "A concussive-like brain injury model in mice (II): selective neuronal loss in the cortex and hippocampus." J Neurotrauma 14(11): 863-873.

Tashiro, A., H. Makino and F. H. Gage (2007). "Experience-specific functional modification of the dentate gyrus through adult neurogenesis: a critical period during an immature stage." J Neurosci 27(12): 3252-3259.

Taupin, P. (2007). "BrdU immunohistochemistry for studying adult neurogenesis: paradigms, pitfalls, limitations, and validation." Brain Res Rev 53(1): 198-214.

Teasdale, G. and B. Jennett (1976). "Assessment and prognosis of coma after head injury." Acta Neurochir (Wien) 34(1-4): 45-55.

Terpolilli, N. A., S. W. Kim, S. C. Thal, W. M. Kuebler and N. Plesnila (2013). "Inhaled nitric oxide reduces secondary brain damage after traumatic brain injury in mice." J Cereb Blood Flow Metab 33(2): 311-318.

Page 274: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

261

Thau-Zuchman, O., E. Shohami, A. G. Alexandrovich and R. R. Leker (2010). "Vascular endothelial growth factor increases neurogenesis after traumatic brain injury." J Cereb Blood Flow Metab 30(5): 1008-1016.

Thau-Zuchman, O., E. Shohami, A. G. Alexandrovich and R. R. Leker (2012). "Combination of vascular endothelial and fibroblast growth factor 2 for induction of neurogenesis and angiogenesis after traumatic brain injury." J Mol Neurosci 47(1): 166-172.

Thompson, H. J., W. C. McCormick and S. H. Kagan (2006). "Traumatic brain injury in older adults: epidemiology, outcomes, and future implications." J Am Geriatr Soc 54(10): 1590-1595.

Thompson, S. N., T. R. Gibson, B. M. Thompson, Y. Deng and E. D. Hall (2006). "Relationship of calpain-mediated proteolysis to the expression of axonal and synaptic plasticity markers following traumatic brain injury in mice." Exp Neurol 201(1): 253-265.

Thorne, R. G., G. J. Pronk, V. Padmanabhan and W. H. Frey, 2nd (2004). "Delivery of insulin-like growth factor-I to the rat brain and spinal cord along olfactory and trigeminal pathways following intranasal administration." Neuroscience 127(2): 481-496.

Thurman, D. J., C. Alverson, D. Browne, K. A. Dunn, J. Guerrero, R. Johnson, V. Johnson, J. Langlois, D. Pilkey, J. E. Sniezek and S. Toal (1999). Traumatic Brain Injury In the United States: A Report to Congress.

Tolbert, D. L. and B. R. Clark (2003). "GDNF and IGF-I trophic factors delay hereditary Purkinje cell degeneration and the progression of gait ataxia." Exp Neurol 183(1): 205-219.

Torres-Aleman, I., F. Naftolin and R. J. Robbins (1989). "Growth promoting effects of IGF-I on fetal hypothalamic cell lines under serum-free culture conditions." Int J Dev Neurosci 7(2): 195-202.

Toulmond, S. and N. J. Rothwell (1995). "Interleukin-1 receptor antagonist inhibits neuronal damage caused by fluid percussion injury in the rat." Brain Res 671(2): 261-266.

Trabold, R., S. Krieg, K. Scholler and N. Plesnila (2008). "Role of vasopressin V(1a) and V2 receptors for the development of secondary brain damage after traumatic brain injury in mice." J Neurotrauma 25(12): 1459-1465.

Trejo, J. L., E. Carro and I. Torres-Aleman (2001). "Circulating insulin-like growth factor I mediates exercise-induced increases in the number of new neurons in the adult hippocampus." J Neurosci 21(5): 1628-1634.

Trejo, J. L., M. V. Llorens-Martin and I. Torres-Aleman (2008). "The effects of exercise on spatial learning and anxiety-like behavior are mediated by an IGF-I-dependent

Page 275: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

262

mechanism related to hippocampal neurogenesis." Mol Cell Neurosci 37(2): 402-411.

Truelle, J. L., S. Koskinen, G. Hawthorne, J. Sarajuuri, R. Formisano, K. Von Wild, E. Neugebauer, L. Wilson, H. Gibbons, J. Powell, M. Bullinger, S. Hofer, A. Maas, G. Zitnay and N. Von Steinbuechel (2010). "Quality of life after traumatic brain injury: the clinical use of the QOLIBRI, a novel disease-specific instrument." Brain Inj 24(11): 1272-1291.

Truettner, J., R. Schmidt-Kastner, R. Busto, O. F. Alonso, J. Y. Loor, W. D. Dietrich and M. D. Ginsberg (1999). "Expression of brain-derived neurotrophic factor, nerve growth factor, and heat shock protein HSP70 following fluid percussion brain injury in rats." J Neurotrauma 16(6): 471-486.

Tsenter, J., L. Beni-Adani, Y. Assaf, A. G. Alexandrovich, V. Trembovler and E. Shohami (2008). "Dynamic changes in the recovery after traumatic brain injury in mice: effect of injury severity on T2-weighted MRI abnormalities, and motor and cognitive functions." J Neurotrauma 25(4): 324-333.

Underwood, L. E., A. J. D'Ercole, D. R. Clemmons and J. J. Van Wyk (1986). "Paracrine functions of somatomedins." Clin Endocrinol Metab 15(1): 59-77.

Urrea, C., D. A. Castellanos, J. Sagen, P. Tsoulfas, H. M. Bramlett and W. D. Dietrich (2007). "Widespread cellular proliferation and focal neurogenesis after traumatic brain injury in the rat." Restor Neurol Neurosci 25(1): 65-76.

van Praag, H., A. F. Schinder, B. R. Christie, N. Toni, T. D. Palmer and F. H. Gage (2002). "Functional neurogenesis in the adult hippocampus." Nature 415(6875): 1030-1034.

Vehring, R. (2008). "Pharmaceutical particle engineering via spray drying." Pharm Res 25(5): 999-1022.

Walter, C., B. L. Murphy, R. Y. Pun, A. L. Spieles-Engemann and S. C. Danzer (2007). "Pilocarpine-induced seizures cause selective time-dependent changes to adult-generated hippocampal dentate granule cells." J Neurosci 27(28): 7541-7552.

Wang, J. M., T. Hayashi, W. R. Zhang, K. Sakai, Y. Shiro and K. Abe (2000). "Reduction of ischemic brain injury by topical application of insulin-like growth factor-I after transient middle cerebral artery occlusion in rats." Brain Res 859(2): 381-385.

Wang, Y. Q., L. Wang, M. Y. Zhang, T. Wang, H. J. Bao, W. L. Liu, D. K. Dai, L. Zhang, P. Chang, W. W. Dong, X. P. Chen and L. Y. Tao (2012). "Necrostatin-1 suppresses autophagy and apoptosis in mice traumatic brain injury model." Neurochem Res 37(9): 1849-1858.

Werther, G. A., M. Abate, A. Hogg, H. Cheesman, B. Oldfield, D. Hards, P. Hudson, B. Power, K. Freed and A. C. Herington (1990). "Localization of insulin-like growth factor-I mRNA in rat brain by in situ hybridization--relationship to IGF-I receptors." Mol Endocrinol 4(5): 773-778.

Page 276: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

263

Whalen, M. J., R. S. Clark, C. E. Dixon, P. Robichaud, D. W. Marion, V. Vagni, S. H. Graham, L. Virag, G. Hasko, R. Stachlewitz, C. Szabo and P. M. Kochanek (1999). "Reduction of cognitive and motor deficits after traumatic brain injury in mice deficient in poly(ADP-ribose) polymerase." J Cereb Blood Flow Metab 19(8): 835-842.

Whalen, M. J., T. Dalkara, Z. You, J. Qiu, D. Bermpohl, N. Mehta, B. Suter, P. G. Bhide, E. H. Lo, M. Ericsson and M. A. Moskowitz (2008). "Acute plasmalemma permeability and protracted clearance of injured cells after controlled cortical impact in mice." J Cereb Blood Flow Metab 28(3): 490-505.

Whitelaw, A., S. Christie and I. Pople (1999). "Transforming growth factor-beta1: a possible signal molecule for posthemorrhagic hydrocephalus?" Pediatr Res 46(5): 576-580.

Wilkins, A., S. Chandran and A. Compston (2001). "A role for oligodendrocyte-derived IGF-1 in trophic support of cortical neurons." Glia 36(1): 48-57.

Wojtowicz, J. M. (2012). "Adult neurogenesis. From circuits to models." Behav Brain Res 227(2): 490-496.

Wu, H., D. Lu, H. Jiang, Y. Xiong, C. Qu, B. Li, A. Mahmood, D. Zhou and M. Chopp (2008). "Simvastatin-mediated upregulation of VEGF and BDNF, activation of the PI3K/Akt pathway, and increase of neurogenesis are associated with therapeutic improvement after traumatic brain injury." J Neurotrauma 25(2): 130-139.

Xiong, Y., A. Mahmood and M. Chopp (2010). "Angiogenesis, neurogenesis and brain recovery of function following injury." Curr Opin Investig Drugs 11(3): 298-308.

Xiong, Y., A. Mahmood and M. Chopp (2013). "Animal models of traumatic brain injury." Nat Rev Neurosci 14(2): 128-142.

Xiong, Y., F. S. Shie, J. Zhang, C. P. Lee and Y. S. Ho (2005). "Prevention of mitochondrial dysfunction in post-traumatic mouse brain by superoxide dismutase." J Neurochem 95(3): 732-744.

Yan, Y. P., K. A. Sailor, R. Vemuganti and R. J. Dempsey (2006). "Insulin-like growth factor-1 is an endogenous mediator of focal ischemia-induced neural progenitor proliferation." Eur J Neurosci 24(1): 45-54.

Ye, P., J. Carson and A. J. D'Ercole (1995). "In vivo actions of insulin-like growth factor-I (IGF-I) on brain myelination: studies of IGF-I and IGF binding protein-1 (IGFBP-1) transgenic mice." J Neurosci 15(11): 7344-7356.

Ye, P., J. Carson and A. J. D'Ercole (1995). "Insulin-like growth factor-I influences the initiation of myelination: studies of the anterior commissure of transgenic mice." Neurosci Lett 201(3): 235-238.

Ye, P., G. J. Popken, A. Kemper, K. McCarthy, B. Popko and A. J. D'Ercole (2004). "Astrocyte-specific overexpression of insulin-like growth factor-I promotes brain

Page 277: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

264

overgrowth and glial fibrillary acidic protein expression." J Neurosci Res 78(4): 472-484.

Ye, P., Y. Xing, Z. Dai and A. J. D'Ercole (1996). "In vivo actions of insulin-like growth factor-I (IGF-I) on cerebellum development in transgenic mice: evidence that IGF-I increases proliferation of granule cell progenitors." Brain Res Dev Brain Res 95(1): 44-54.

Yoshimura, S., T. Teramoto, M. J. Whalen, M. C. Irizarry, Y. Takagi, J. Qiu, J. Harada, C. Waeber, X. O. Breakefield and M. A. Moskowitz (2003). "FGF-2 regulates neurogenesis and degeneration in the dentate gyrus after traumatic brain injury in mice." J Clin Invest 112(8): 1202-1210.

Yoshino, A., D. A. Hovda, T. Kawamata, Y. Katayama and D. P. Becker (1991). "Dynamic changes in local cerebral glucose utilization following cerebral conclusion in rats: evidence of a hyper- and subsequent hypometabolic state." Brain Res 561(1): 106-119.

You, Z., S. I. Savitz, J. Yang, A. Degterev, J. Yuan, G. D. Cuny, M. A. Moskowitz and M. J. Whalen (2008). "Necrostatin-1 reduces histopathology and improves functional outcome after controlled cortical impact in mice." J Cereb Blood Flow Metab 28(9): 1564-1573.

Yu, S., Y. Kaneko, E. Bae, C. E. Stahl, Y. Wang, H. van Loveren, P. R. Sanberg and C. V. Borlongan (2009). "Severity of controlled cortical impact traumatic brain injury in rats and mice dictates degree of behavioral deficits." Brain Res 1287: 157-163.

Yu, T. S., G. Zhang, D. J. Liebl and S. G. Kernie (2008). "Traumatic brain injury-induced hippocampal neurogenesis requires activation of early nestin-expressing progenitors." J Neurosci 28(48): 12901-12912.

Yu, Y., A. J. Kastin and W. Pan (2006). "Reciprocal interactions of insulin and insulin-like growth factor I in receptor-mediated transport across the blood-brain barrier." Endocrinology 147(6): 2611-2615.

Yuan R, R. C., Beamer WG (2013). Aging study: IGF-1 and body weight for 33 inbred strains of mice. Mouse Phenome Database web site, The Jackson Laboratory. Bar Harbor, Maine USA.

Yuan, W., F. Wu, M. Guo and T. Jin (2009). "Development of protein delivery microsphere system by a novel S/O/O/W multi-emulsion." Eur J Pharm Sci 36(2-3): 212-218.

Zadran, S., X. Bi and M. Baudry (2010). "Regulation of calpain-2 in neurons: implications for synaptic plasticity." Mol Neurobiol 42(2): 143-150.

Zhang, X., Y. Chen, M. D. Ikonomovic, P. D. Nathaniel, P. M. Kochanek, D. W. Marion, S. T. DeKosky, L. W. Jenkins and R. S. Clark (2006). "Increased phosphorylation of protein kinase B and related substrates after traumatic brain injury in humans and rats." J Cereb Blood Flow Metab 26(7): 915-926.

Page 278: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

265

Zhang, Z., M. Artelt, M. Burnet, K. Trautmann and H. J. Schluesener (2006). "Early infiltration of CD8+ macrophages/microglia to lesions of rat traumatic brain injury." Neuroscience 141(2): 637-644.

Zhao, C., W. Deng and F. H. Gage (2008). "Mechanisms and functional implications of adult neurogenesis." Cell 132(4): 645-660.

Zhao, C., E. M. Teng, R. G. Summers, Jr., G. L. Ming and F. H. Gage (2006). "Distinct morphological stages of dentate granule neuron maturation in the adult mouse hippocampus." J Neurosci 26(1): 3-11.

Zhao, X., A. Ahram, R. F. Berman, J. P. Muizelaar and B. G. Lyeth (2003). "Early loss of astrocytes after experimental traumatic brain injury." Glia 44(2): 140-152.

Zheng, W. H., S. Kar and R. Quirion (2002). "Insulin-like growth factor-1-induced phosphorylation of transcription factor FKHRL1 is mediated by phosphatidylinositol 3-kinase/Akt kinase and role of this pathway in insulin-like growth factor-1-induced survival of cultured hippocampal neurons." Mol Pharmacol 62(2): 225-233.

Zhou, H., L. Chen, X. Gao, B. Luo and J. Chen (2012). "Moderate traumatic brain injury triggers rapid necrotic death of immature neurons in the hippocampus." J Neuropathol Exp Neurol 71(4): 348-359.

Zhu, C. Z. and R. N. Auer (1994). "Intraventricular administration of insulin and IGF-1 in transient forebrain ischemia." J Cereb Blood Flow Metab 14(2): 237-242.

Zhu, W., Y. Fan, T. Frenzel, M. Gasmi, R. T. Bartus, W. L. Young, G. Y. Yang and Y. Chen (2008). "Insulin growth factor-1 gene transfer enhances neurovascular remodeling and improves long-term stroke outcome in mice." Stroke 39(4): 1254-1261.

Zweckberger, K., M. Stoffel, A. Baethmann and N. Plesnila (2003). "Effect of decompression craniotomy on increase of contusion volume and functional outcome after controlled cortical impact in mice." J Neurotrauma 20(12): 1307-1314.

Page 279: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

266

Curriculum Vitae

Shaun William Carlson, B.S. Spinal Cord and Brain Injury Research Center Department of Physiology College of Medicine, University of Kentucky Citizenship United States of America Education Aug 2003- May 2007 University of Kansas

Bachelors of Science in Cellular Biology

Aug 2007-July 2013 University of Kentucky: Ph.D. in Physiology Kathryn E. Saatman, mentor

Research Experience May 2005- Apr 2006 Independent Study Research in the lab of Truman Christopher

Gamblin Ph.D. (Department of Molecular Biosciences, University of Kansas).

May 2006- July 2007 Laboratory Assistant, supervisor Truman Christopher Gamblin

Ph.D. (Department of Molecular Biosciences, University of Kansas).

Aug 2007- Oct 2007 IBS Research Rotation in the laboratory of Diane Snow Ph.D.

(Department of Anatomy and Neurobiology, University of Kentucky).

Oct 2007- Dec 2007 IBS Research Rotation in the laboratory of Steve Estus Ph.D.

(Department of Physiology, University of Kentucky). Jan 2008- May 2008 IBS Research Rotation in the laboratory of Kathryn Saatman

Ph.D. (Department of Physiology, University of Kentucky). May 2008- Present Graduate Student in the laboratory of Kathryn Saatman Ph.D.

(Department of Physiology, University of Kentucky) Awards and Achievements Aug–Dec 2003 Academic Scholarship to the University of Kansas. Aug 2005-May 2007 University of Kansas Honor Roll. Jun 2006 Northwestern Summer Research Program (Accepted but not

matched). Dec 2008 Brian J. Hardin Award for Research, Department of Physiology,

University of Kentucky. Oct 2010 Graduate School Student Support Funding recipient, (Funding for

travel) University of Kentucky. July 2011 Graduate School Student Support Funding recipient, (Funding for

travel) University of Kentucky.

Page 280: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

267

Jan 2010-Jan 2012 NIH T32 Graduate Student Fellowship through the Therapeutic Strategies for Neurodegeneration Training Grant. PI: Edward Hall. Grant 1T32 DA022738.

April 2012 Bluegrass Society for Neuroscience Poster Presentation Award Recipient.

July 2012 Graduate School Student Support Funding recipient, (Funding for travel) University of Kentucky.

July 2012-June 2013 Dissertation Year Fellowship, University of Kentucky Graduate School. This fellowship is a competitively based merit award to assist in the completion of the dissertation research in the final year of study.

July 2012 Top Student Poster Competition Finalist for the 2012 National Neurotrauma Symposium.

July 2012 Anthony Marmarou Award of Excellence, National Neurotrauma Symposium, Phoenix, AZ.

April 2013 Bluegrass Society for Neuroscience Poster Presentation Award Recipient.

April 2013 Graduate School Student Support Funding recipient, (Funding for travel) University of Kentucky.

August 2013 Top Student Poster Competition Finalist for the 2013 National Neurotrauma Symposium.

August 2013 Travel Grant Award for 2013 National Neurotrauma Symposium. Grant Funding Jan 2010-Jan 2012 NIH T32 Graduate Student Fellowship through the Therapeutic

Strategies for Neurodegeneration Training Grant. PI: Edward Hall. Grant 1T32 DA022738.

Elected and Appointed Positions 2009-2011 Member of Brain Hardin Award for Research Selection

Committee, Department of Physiology, University of Kentucky. 2010-2011 Graduate Student Representative for the Department of

Physiology, University of Kentucky. 2011-2012 College of Medicine Strategic Planning Committee, University of

Kentucky (Appointed by Michael Reid Ph.D., Senior Associate Dean for Biomedical Science).

2011-present Member of the Committee for Center Initiatives (CCI), Spinal Cord and Brain Injury Research Center.

Publications

Carlson SW, Branden M, Voss K, Sun Q, Rankin CA, Gamblin TC, 2007. A complex mechanism for inducer mediated tau polymerization. Biochemistry. 46(30):8838-49. Pleasant, JM*, SW Carlson*, H Mao, SW Scheff, K Yang, KE Saatman, 2011, “Rate of neurodegeneration in the mouse controlled cortical impact model is influenced by impactor tip shape: Implications for mechanistic and therapeutic studies.” J Neurotrauma, November 2011, 28(11)2245-62. Selected for Special Issue and Cover: Engineering Approaches to Studying and Repairing the Injured Nervous System. (*co-first authors).

Page 281: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

268

Cai W, Carlson SW, Brelsfoard JM, Mannon CE, Moncman CL, Saatman KE, Andres DE. 2012. “Rit GTPase signaling promotes immature hippocampal neuronal survival.” J Neurosci, July 2012, 32(29):9887-9897. Madathil SK, Carlson SW, Brelsfoard JM, Ye P, D’Ercole JD, Saatman KE, “Astrocyte-specific overexpression of insulin-like growth factor-1 protects hippocampal neurons following traumatic brain injury in mice.” PLoS ONE (in press).

Oral Presentations

Department of Physiology Graduate Student Seminar Series (Oct 2008, Feb 2009)

Spinal Cord and Brain Injury Research Center Journal Club (Nov 2009, Nov

2010, Oct 2011) Neuroprotective and Neuroreparative Potential of Insulin-like Growth Factor-1 after Traumatic Brain Injury, Department of Physiology Seminar Series, April 2010. IGF-1 Trials and Tribulations. Works in Progress Seminar Series, Spinal Cord and Brain Injury Research Center, October 2010. Evaluating the Neurogenic Potential of Insulin-like Growth Factor-1 after Traumatic Brain Injury,. Invited speaker at the Department of Physiology Graduate Communication Skills Workshop, Aug 2012.

Abstracts and Poster Presentations (International, National, and selected Local)

Madathil SK, Carlson SW*, Foozer H, Saatman KE. Transient induction of IGF-1/IGF-1R signaling in the mouse brain following traumatic brain injury. J Neurotrauma. 25:898. Presented at National Neurotrauma Society Symposium Orlando, FL July 2008. Carlson SW*, Madathil SK, Gao X, Chen J, Saatman KE. Proliferation in the hippocampal subgranular zone after traumatic brain injury of IGF-1 overexpressing mice. Presented at the Kentucky Spinal Cord and Head Injury Research Trust Symposium, Louisville, KY May 2009. Carlson SW*, Pleasant J, Scheff SW, Saatman KE. “Effect of Impactor Tip Geometry on Cell Death Progression Following Controlled Cortical Impact: Implications for Therapeutic Interventions.” J Neurotrauma, 26:A-50. Presented at the 27th annual International/National Neurotrauma Symposium, Santa Barbara, CA Sept 2009. Carlson SW*, S.K. Madathil, X. Gao, J. Chen, K.E. Saatman, "Proliferation in the Hippocampal Subgranular Zone Following Traumatic Brain Injury of IGF-1 Overexpressing Mice." Program Number 151.22. 2009 Neuroscience Meeting Planner Online: Society for Neuroscience, Chicago, IL Oct 2009.

Page 282: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

269

Carlson SW*, S.K. Madathil, X. Gao, J. Chen, K.E. Saatman, “Proliferation in the Hippocampus of IGF-1 Overexpressing Mice following Traumatic Brain Injury.” Presented at the Adult Neurogenesis: Structure and Function Conference, Frauenchiemsee, Germany May 2010. Carlson SW*, S.K. Madathil, X. Gao, J. Chen, K.E. Saatman, “Cell Proliferation and Survival in the Dentate Gyrus following Controlled Cortical Impact of IGF-1 Overexpressing Mice.” Presented at the Kentucky Spinal Cord and Head Injury Research Trust Symposium, Lexington, KY June 2010. Madathil S.K.*, Pleasant J, Carlson SW, K.E. Saatman, “Insulin-like Growth Factor-1 Overexpression Favours Brain Remodeling and Improves Motor and Cognitive Function after Traumatic Brain Injury.” J Neurotrauma, 26:A-65. Presented at the 28th Annual National Neurotrauma Symposium, Las Vegas, NV, June 2010. Carlson SW*, S.K. Madathil, X. Gao, J. Chen, K.E. Saatman, “Cell Proliferation and Survival in the Hippocampus after Controlled Cortical Impact of IGF-1 Overexpressing Mice.” J Neurotrauma, 26:A-80. Presented at the 28th Annual National Neurotrauma Symposium, Las Vegas, NV, June 2010. Ma, J*, Carlson SW, Saatman K.E., HM Mansour, “Advanced Spray Dried PLGA Particles for Subcutaneous Delivery.” Presented at the Pharmacy Symposium, College of Pharmacy, University of Kentucky, October 2010. Carlson SW*, Brelsfoard JM, Saatman KE, “Assessment of systemic and central infusion of insulin-like growth factor-1 following traumatic brain injury.” Presented at the Kentucky Spinal Cord and Head Injury Research Trust Symposium, Louisville, KY, May 2011. Cai W*, Carlson SW, Rudolph JL, Brelsfoard JM, Saatman KE, DA Andres, “Small GTPase Rit promotes immature hippocampal neuron survival in response to oxidative stress.” Presented at FASEB GTPase Signaling Meeting, June 2011. Carlson, SW*, Brelsfoard JM, Saatman KE, “Evaluation of systemic and central infusion of insulin-like growth factor-1 following controlled cortical impact.” Presented at the 29th Annual National Neurotrauma Symposium, Fort Lauderdale, FL, July 2011. Carlson, SW*, Brelsfoard JM, Saatman KE, “Evaluation of systemic and central infusion of insulin-like growth factor-1 following traumatic brain injury.” Program number 159.21/BB16 Neuroscience meeting Planner Online: Society for Neuroscience Symposium, Washington D.C, 2011. Carlson, SW*, Brelsfoard JM, Saatman KE, “Assessment of intracerebroventricular infusion of Insulin-like growth factor-1 after controlled cortical impact in mice.” Presented at the 30th Annual National Neurotrauma Symposium, Phoenix, AZ, July 2012.

Page 283: EVALUATION OF INSULIN-LIKE GROWTH FACTOR-1 AS A THERAPEUTIC APPRO page33+34

270

Carlson, SW*, Madathil SK, Chen J, Saatman KE, “Insulin-like growth factor-1 overexpression enhances neurogenesis after traumatic brain injury by promoting neuronal differentiation.” Poster 658.25. Presented at the Annual Society for Neuroscience Conference, New Orleans, LA, Oct 2012.

Carlson, SW*, Madathil SK, Chen J, Saatman KE, “Insulin-like growth factor-1 overexpression enhances neurogenesis after traumatic brain injury by promoting neuronal differentiation.” Presented at Bluegrass Chapter for Society for Neuroscience, Lexington, KY, April 2013. *denotes presenter

Teaching Experience July-Aug 2012 Adjunct Professor, Bluegrass Community & Technical College Lexington, KY Aug-Dec 2012 Adjunct Professor, Bluegrass Community & Technical College Lexington, KY Mentoring Jan-March 2010 Daniel Bartos

Integrated Biomedical Sciences rotation student June-Aug 2010 Emily Hall

Undergraduate student June-Aug 2010 Tina Stottman

Undergraduate student Aug-Sept 2010 Amanda Bolton

Graduate student of Kathryn E. Saatman, Ph.D. Integrated Biomedical Sciences rotation student

June-Aug 2011 Justin Jatczak Undergraduate student

June-Aug 2011 Alex Gutierrez Undergraduate student Aug-Sept 2011 Erica Littlejohn

Graduate student of Kathryn E. Saatman, Ph.D. Integrated Biomedical Sciences rotation student

Memberships 2008-present National Neurotrauma Society 2009-present Society for Neuroscience 2009-present Bluegrass Chapter of the Society for Neuroscience 2010-present Member of Teaching, Education and Mentoring (TEaM),

Department of Physiology 2012-present Member of the Graduate Women in Science (GWIS) Society.

http://www.gwis.org/


Recommended