+ All Categories
Home > Documents > POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE...

POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE...

Date post: 13-May-2020
Category:
Upload: others
View: 1 times
Download: 0 times
Share this document with a friend
116
POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE REPAIR OF DIABETIC HEART FAILURE SHOAIB AKHTAR NATIONAL CENTRE OF EXCELLENCE IN MOLECULAR BIOLOGY UNIVERSITY OF THE PUNJAB LAHORE, PAKISTAN. (2011)
Transcript
Page 1: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE REPAIR OF DIABETIC

HEART FAILURE

SHOAIB AKHTAR

NATIONAL CENTRE OF EXCELLENCE IN

MOLECULAR BIOLOGY UNIVERSITY OF THE PUNJAB

LAHORE, PAKISTAN. (2011)

Page 2: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE REPAIR OF DIABETIC

HEART FAILURE

A THESIS SUBMITTED TO

THE UNIVERSITY OF THE PUNJAB

IN FULFILLMENT OF THE REQUIRMENT

FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY IN MOLECULAR BIOLOGY

BY

SHOAIB AKHTAR

SUPERVISOR: DR. SHAHEEN N. KHAN

NATIONAL CENTER OF EXCELLENCE IN MOLECULAR BIOLOGY,

UNIVERSITY OF THE PUNJAB, LAHORE, PAKISTAN

(2011)

Page 3: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure
Page 4: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

DEDICATIONS

I dedicate my thesis work

To my parents

Who always showed their trust in my

capabilities and their prayers and love

always rewarded me with success

My teachers

Who from Prep to PhD helped me in

seeing things beyond

Page 5: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CONTENTS

Chapter No. Title Page No.

LIST OF FIGURES ………………………………………………………………… v

LIST OF TABLES ………………………………………………………………….. vii

SUMMARY …………………………………………………………………………. viii

ACKNOWLEDGMENTS …………………………………………………….... xi

ABBREVIATIONS AND SYMBOLS ……………………………………….. xii

1. INTRODUCTION…………………………....................................... 1―5

2. REVIEW OF LITERATURE……………………………………………………. 6―27

2.1 AN INTRODUCTION TO STEM CELLS………………………. 6

2.2 Stem Cells…………………………………………………………….. 6

2.3 Types of Stem Cells………………………………………………. 6

2.3.1 Embryonic Stem Cells…………………………………………… 6

2.3.2 Adult Stem Cells…………………………………………………… 7

2.4 Bone Marrow……………………………………………………….. 7

2.4.1 Hematopoietic Stem Cells…………………………………….. 7

2.4.2 Mesenchymal Stem Cells………………………………………. 7

2.4.3 Endothelial Progenitor Cells…………………………………… 8

2.4.4 Multipotent Adult Progenitor Cells……………………….. 8

2.5 CARDIOVASCULAR SYSTEM………………………………….. 9

2.5.1 Heart……………………………………………………………………. 9

Page 6: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.5.2 Microstructure of the Heart………………………………….. 10

2.5.3 Blood Circulation………………………………………………….. 12

2.6 CARDIOVASCULAR DISEASES………………………………… 13

2.6.1 Diabetes and Cardiovascular Diseases…………………. 14

2.6.2 Diabetic Cardiomyopathy……………………………………… 15

2.7 TREATMENT AND MANAGEMENT OF DCM…………... 19 2.7.1 Life Style……………………………………………………………….. 19

2.7.2 Glycemic Control…………………………………………………… 19

2.7.3 Dyslipidemia Treatment………………………………………… 19

2.7.4 Antihypertensive Treatment…………………………………. 20

2.8 SOME ADDITIONAL THERAPEUTIC STRATEGIES……... 20

2.8.1 Apelin and Cardiac Output……………………………………. 20

2.8.2 Kinins……………………………………………………………………. 20

2.8.3 Sarcoplasmic Reticulum Calcium ATPase (SERCA2).. 21

2.8.4 Antioxidants…………………………………………………………. 21

2.8.5 Erythropoietin………………………………………………………. 22

2.8.6 Cellular Therapy……………………………………………………. 25

2.8.7 Combining MSCs Transplanatation and EPO Treatment 26

3. MATERIALS AND METHODS 28―38

3.1 Animals………………………………………………………………… 28

3.2 Mouse Model for Diabetes…………………………………… 28

3.3 In vitro Study……………………………………………………….. 28

Page 7: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.3.1 Cell Culture………………………………………………………….. 28

3.3.2 Flow Cytometry……………………………………………………. 28

3.3.2 Preconditioning of Diabetic MSCs…………….............. 29

3.3.3 Gene Expression Profiling of MSCs……………………….. 29

3.3.4 Hypoxic Stress………………………………………………………. 31

3.3.5 Superoxide Dismutase Assay………………………………… 32

3.3.6 Apoptosis……………………………………………………………… 32

3.3.7 In vitro Tube-forming Assay………………………………….. 32

3.3.8 Chemotactic Attraction…………………………………………. 33

3.3.9 Glucose Stress………………………………………………………. 33

3.4 In vivo Study…………………………………………………………. 34

3.4.1 Diabetic Mouse Model………………………………………….. 34

3.4.2 EPO Treatment……………………………………………………… 34

3.4.3 Transplantation and EPO Treatment…………………….. 34

3.4.4 RNA Extraction and cDNA Synthesis……………………… 35

3.4.5 Gene Expression Study…………………………………………. . 35

3.4.6 Tissue Procurement for Histological Studies………….. 36

3.4.7 Tunel Assay………………………………………………………….. 36

3.4.8 Immunostaining for eNOS…………………………………….. 37

3.4.9 Detection of Stem cells in Left Ventricular Tissue….. 37

3.4.9.1 Sirius Red Staining……………………………………………….. 37

3.4.9.2 Millar Analysis…………………………………………………….. 38

Page 8: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.5 Statistical Analysis……………………………………………….. 38

4. RESULTS…………………………………………………………………………… 39―59

4.1 In vitro Studies……………………………………………………… 39

4.1.1 Characterization of MSCs……………………………………… 39

4.1.2 Stimulation of Cell Proliferation in Preconditioned Diabetic MSCs

………………………………………………..………………………………………. 40

4.1.3 Gene Expression Profiling of MSCs………………………… 42

4.1.4 Increased SOD Activity in Preconditioned MSCs……… 44

4.1.5 Effect of IGF-1 and FGF-2 Preconditioning on Apoptosis 45

4.1.6 Chemotactic Ability of Preconditioned Diabetic MSCs 46

4.1.7 In vitro tube-forming ability in diabetic MSCs………… 47

4.1.8 Real-time PCR gene Expression after Glucose Stress 49

4.2 In vivo Studies……………………………………………… 50

4.2.1 Effect of EPO treatment on Gene expression of Diabetic Heart

………………………………………………………………………………………… 50

4.2.2 Decreased Myocardial Cell Apoptosis……………………. 51

4.2.3 Enhancement of eNOS Expression…………………………. 52

4.3 Transplantation Studies……………………………………….. 54

4.3.1 Homing of MSCs…………………………………………………… 54

4.3.2 Reduction in Tissue Fibrosis………………………………….. 56

4.3.3 Hemodynamic Parameters……………………………………. 58

5. DISCUSSIONS…………………………………………………………………… 60―67

6. REFERENCES………………………………………………………............... 68―91

Page 9: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

LIST OF FIGURES

Figure No. Page No.

CHAPTER 2

Fig. 2.1A Heart anatomy showing sulci 10

Fig. 2.1B Internal anatomy of the heart 10

Fig. 2.2 Microstructure of the heart tissue 12

Fig. 2.3 Some of the diabetes associated risk factors for HF 15

Fig. 2.4 Interacting pathways of ROS production and injury in diabetic cardiomyocytes 16

Fig. 2.5 The pathophysiological substrate of DCM 18

Fig. 2.6 Signal transduction pathways involved in EPO mediated

Cytoprotection 23

CHAPTER 4

Fig. 4.1 Mouse model of diabetes 39

Fig. 4.2 Flow cytometry of diabetic MSCs 40

Fig. 4.3A Cell proliferation after preconditioning of diabetic MSCs 41

Fig.4.3B Analysis of cell proliferation for (PCNA) expression comparing

VEGF and IGF-1/FGF-2 preconditioning of diabetic MSCs 42

Fig. 4.4A-D Gene expression profiling of MSCs 43

Fig. 4.4E Quantification of gene expression profiling of MSCs 44

Fig.4.5 Comparison of SOD activity among normal, diabetic untreated

and preconditioned MSCs under normoxia and hypoxia 45

Fig. 4.6 Flow cytometry for annexin-V-positive cells 46

Fig. 4.7 Mobilization of untreated and preconditioned MSCs to SDF-1α 47

Fig. 4.8A-D Preconditioned MSCs demonstrate better tube-forming ability

Page 10: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

than untreated cells both under hypoxia and normoxia 48

Fig.4.8E Quantification of tube forming cells under hypoxic and

normoxic conditions 49

Fig. 4.9 Real-time RT-PCR gene expression analysis under conditions

of high-glucose stress 50

Fig. 4.10 Left ventricular mRNA expression of Bax, p16ᴵᴺᴷ⁴ᵃ and IGF-1

in different treatment groups of C57BL/6 male mice 51

Fig. 4.11 TUNEL assay for apoptosis in left ventricular heart sections

(40 X) of different treatment groups of C57BL/6 male mice 52

Fig. 4.12 Expression of eNOS in the left ventricular heart sections (20X)

of different treatment groups of C57BL/6 male mice 53

Fig. 4.13 Homing of GFP-positive MSCs in Left ventricular heart sections

(40X) of different treatment groups of C57BL/6 male mice 55

Fig. 4.14 Fibrosis as seen in left ventricle heart sections (40 X) of different

treatment groups of C57BL/6 male mice by sirius red staining 57

Page 11: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

LIST OF TABLES

No. Page No

CHAPTER 3

Table.1 List of primers, their product size (bp) and sequence 5´—3´ 31

CHAPTER 4

Table.2 Cardiac parameters as determined by Millar analysis in different

treatment groups of C57BL/6 male mice 59

Page 12: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

SUMMARY

Mesenchymal stem cells (MSCs) derived from bone marrow (BM) hold multilineage

differentiation potential which can be used to treat diabetic heart failure (HF). Yet,

hyperglycemia can lead to functional impairment of MSCs and would warrant an appropriate

treatment strategy to improve its functionality. Diabetes was induced in male C57BL/6 wild-

type mice by streptozotocin and MSCs were isolated 60 days after diabetes induction.

Diabetic MSCs were characterized by flow cytometry for CD44 (97.7%), CD90 (95.4%), and

CD105 (92.3%).

In the present study two strategies, in vitro preconditioning with growth factors and in

vivo erythropoietin (EPO) treatment were used to improve the functionality of diabetic

MSCs. MSCs were preconditioned with insulin-like growth factor-1 (IGF-1) and fibroblast

growth factor-2 (FGF-2), 50 ng/mL each in combination for 1 h in serum-free Iscove’s

modified Dulbecco’s medium (IMDM). EPO was administered to 40 days diabetic mice at

1000 IU/Kg for five consecutive days through intra peritoneal route.

Since the transplanted MSCs in the diabetic heart are likely to experience both

ischemia and hyperglycemia, the preconditioned MSCs in vitro were subjected to hypoxia

and high glucose exposure in an attempt to imitate microenvironment of the diabetic heart.

This was followed by assessment for the effects of preconditioning on diabetic MSCs.

Preconditioning caused profound effects on the gene expression profile of diabetic MSCs by

downregulating p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ, p53, Bax, and Bak and upregulateing prosurvival (IGF-1,

FGF-2, Akt) and early cardiac differentiation markers (GATA-4, Nkx 2.5). In a parallel

treatment group the diabetic MSCs were treated with vascular endothelial growth factor

(VEGF) (50 ng/mL) owing to its known effects on cell survival and proliferation. The

Page 13: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

comparison of gene expression profiling for the above markers between the two treatment

groups showed IGF-1/FGF-2 treatment group to be better than the VEGF treated group. The

expression of proliferating cell nuclear antigen (PCNA) gene, a marker of cell proliferation,

was also better in the IGF-1/FGF-2 treated group than that of VEGF. So IGF-1/FGF-2 based

preconditioning of diabetic MSCs was selected for experiments of hypoxic and high glucose

insults and all subsequent experiments.

Under conditions of hypoxic stress untreated diabetic MSCs showed low superoxide

dismutase (SOD) activity (36.9+3.1%) which improved significantly in the preconditioned

diabetic MSCs (52.3+2%). Similar improvements characterized by low number of annexin-

V-positive cells, better tube-forming ability, and increased migration to stromal cell-derived

factor-1α (SDF-1α) were witnessed in the preconditioned diabetic MSCs compared with the

untreated diabetic MSCs. Preconditioning also enhanced Ang-I and VEGF expression while

reduced the expression of p16ᴵᴺᴷ⁴ᵃ in the diabetic MSCs exposed to high glucose conditions.

In view of the reported cardio-protective effects of EPO sub set of experiments were

performed to study the effects of EPO on diabetic heart. Of the two groups of diabetic mice

the diabetic EPO-treated group received injections (i.p.) of EPO while the untreated group

received equal volume of normal saline injections (i.p.). Normal mice served as control. The

gene expression analysis of the left ventricular tissue on day six showed downregulation of

p16ᴵᴺᴷ⁴ᵃ and Bax genes and an upregulation of IGF-1 in the EPO treated group compared

with the untreated group. Tunel staining results showed significant reduction in myocardial

cell apoptosis in the EPO-treated group (4±0.3) compared with the untreated group (8±0.5).

EPO-treated group also showed better left ventricular endothelial nitric oxide synthase

(eNOS) expression than untreated group.

Page 14: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Owing to the observed beneficial effects of EPO on diabetic heart a combined

strategy of transplantation of preconditioned MSCs followed by EPO treatment was

employed to treat diabetic heart. Preconditioned diabetic MSCs were transplanted into the

left ventricle of diabetic mice with subsequent EPO treatment (preconditioned EPO-treated

group) and compared with the transplantation of untreated and preconditioned diabetic MSCs

alone. After four weeks of transplantation homing was highest in the preconditioned EPO-

treated group (45±3.5) followed by the preconditioned MSCs (25±2) and the untreated MSCs

(12±1.5) group. A significant reduction in left ventricular tissue fibrosis was observed in the

preconditioned EPO-treated group (6.67%±0.25%) compared with both the preconditioned

MSCs (10.59%±1.25%) and the diabetic untreated MSCs group (13.16%±0.5%).

The functional analysis of the diabetic hearts showed marked improvements in the

diabetic EPO-treated, preconditioned and preconditioned EPO-treated groups for the assessed

cardiac parameters. In particular, values of the end-systolic pressure (77.1+6.4mmHg) and

ejection fraction (26.9+0.62%) observed in the preconditioned EPO-treated group were

closest to the normal standard base line values of 90-110 mmHg and 44-62% respectively

compared with the other groups. EPO treatment thus showed improvement of diabetic heart

at several fronts characterized by a decrease in myocardial apoptosis, senescence, tissue

fibrosis and an increase in the expression of eNOS, homing of MSCs and heart functionality.

So the preconditioning of diabetic MSCs with IGF-1/FGF-2 growth factors signifies

an innovative strategy to improve the function of diabetes-impaired MSCs and the

transplantation of the preconditioned MSCs in the diabetic heart followed by in vivo EPO

treatment represents a novel combined strategy of its type to treat diabetic HF.

Page 15: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

ACKNOWLEDGEMENTS

All the praises and than s be to ll h, the Lord of the n (mankind, jinn and all

that exists). It is because of His countless blessings that I managed to perform the research

work and write this thesis. All respects are for the Holy Prophet Muhammad on whom be

peace and blessings of the ord. He is the prophet well-informed the witness to ll h’s

commandments, the bringer of glad tidings, the warner unto those who are heedless, the

summoner of the erring to the way of ll h, the resplendent light which dispels the darkness

and shows the right path¹.

It is said that where there is a will there is a way out but in my case the will of doing

the research work was there but in the beginning I was unable to find a way out, for me it

was a herculean task but with llah’s blessings I was luc y enough to find such a supportive

clique who helped me in carrying out things pragmatically and gave me the confidence to

proceed with perseverance and commitment.

Today on the completion of my project I would like to extend my thanks to Dr.

Tayyab Husnain Director, National Centre of Excellence in Molecular Biology for his kind

cooperation. I would like to extend my profound thanks to the former Director CEMB, Dr.

Sheikh Riazuddin for his keen interest in the development of stem cells lab. and providing us

all the required research facilities. I am grateful to my research supervisor Dr. Shaheen N

Khan for her guidance and kind support during whole span of my research work. My special

gratitude to Dr. Mohsin Khan and Dr. Sadia Mohsin for their special interest in my work,

their technical expertise always helped me a lot to focus on my objectives. The cooperation

and help of my all colleagues especially Shareef Masoud, Tariq Awan, Azra, Ghazanfer,

Sulaiman, Mahmood, Ali, Fatima, Sana, Maria, and Ajaml is worth to mention. My special

thanks to staff of animal house, Mr. Nasir, Mr. Najeeb and Mr. Shahzad for giving due care

to animals during my research work.

I have no words to express my feelings for the support and encouragement of my

wife. It was her patience and hard work in taking full care of home matters during my

prolonged absence that kept the momentum of my work. My little daughters Saman and

Fatima showed immense patience while waiting for late hours. My deep sense of respect and

love to my brothers, Laiq-uz-Zaman, Saeed Akhtar and Suhail Akhtar for their every possible

help during my research. I take this opportunity to thank my all cousins for their good wishes

during the entire period of my research.

Working in CEMB was a great experience and I would always cherish my stay in this

esteemed institution and surely this experience of learning will always be a source of pride

for me.

SHOAIB AKHTAR

¹ Adopted from the book “Muhammad The Ideal Prophet” by Saiyid Sulaiman Nadvi. Translated by Mohiuddin Ahmad.p.20. Islamic Book Foundation, Lahore; Pakistan

Page 16: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

ABBREVIATIONS AND SYMBOLS

Acute Myocardial Infarction AMI

Adult Stem Cells ASCs

Advanced Glycation End Products AGEs

Alpha-Lipoic Acid α-LA

Base Pair bp

Blood Pressure B.P.

Bone marrow BM

4´, 6-Diamidino-2- Phenylindole DAPI

Diabetic Cardiomyopathy DCM

Embryonic Stem Cells ESCs

Endothelial Cells ECs

Endothelial Nitric Oxide Synthase eNOS

Endothelial Progenitor Cells EPCs

End-systolic Pressure ESP

EPO Receptor EPOR

Ejection Fraction EF

Erythropoietin EPO

Ethylene diamine tetra acetic acid EDTA

Fetal Bovine Serum FBS

Fibroblast Growth Factor-2 FGF-2

Fluorescein Isothiocyanate FITC

Page 17: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Free Fatty Acid FFA

Germ line Stem Cells GSCs

Glucose Transportation GLUT

Glyceraldehyde 3-Phosphate Dehydrogenase GAPDH

Heart Failure HF

Hematopoietic Stem Cells HSCs

High Glucose HG

Insulin-like Growth Factor-1 IGF-1

International Units IU

Intraperitoneal i.p.

Iscove’s Modified Dulbecco’s Medium IMDM

Kallikrein-Kinin System KKS

Kilogram kg.

Matrix MetalloProteinase2 MMP2

Mesenchymal Stem Cells MSCs

Metabolic Syndrome MetS

Min minute

Moloney Murine Leukemia Virus M-MLV

Multipotent Adult Progenitor Cells MAPCs

Myocardial Infarction MI

Nitric Oxide NO

Paraformaldehyde PFA

Polymerase Chain Reaction PCR

Page 18: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Proliferating Cell Nuclear Antigen PCNA

Protein Kinase C PKC

Quantitative PCR qPCR

Reactive Oxygen Species ROS

Recombinant Human EPO rHuEPO

Reverse Transcriptase-PCR RT-PCR

Sarcoplasmic Reticulum Calcium ATPase 2 SERCA2

Sodium 3-[1-{phenylaminocarbonyl}- 3,4-tetrazolium]-

-bis{4-methoxy-6-nitro} Benzene Sulfonic acid Hydrate XTT

Somatic Stem Cells SSCs

Stromal Cell-Derived Factor-1Alpha SDF-1α

Superoxide Dismutase SOD

Terminal deoxy nucleotidyl transferase-mediated TUNEL dUTP Nick End Labeling

UK Prospective Diabetes Study UKPDS

Vascular Endothelial Growth Factor VEGF

Page 19: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CHAPTER 1

INTRODUCTION

Page 20: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

The spread of heart disease is rapid around the globe (Lloyd-Jones et al., 2010) and

the consequences of heart disease have been reported to be more dangerous among diabetics.

The number of diabetes affected people in the world is around 150 million which is

anticipated to touch the figure of 299 million by the year 2025. Among other diseases

cardiovascular diseases account for a considerable increase in incidence and risk of mortality

in the diabetic patients (Francia et al., 2009). Diabetes has also been previously reported as

one of the most frequent causes of cardiovascular complications among a large section of

population (Gaede et al., 2003) and one of the leading causes of deaths in diabetics (Candido

et al., 2003; Sowers et al., 2001; Mazzone et al., 2008). The development of diabetic

cardiomyopathy in the absence of established risk factors (Sarwar et al., 2010) is among

some recent findings seen as a consequence of diabetes.

Diabetes associated increase in oxidative stress can cause exceedingly high levels of

reactive oxygen species (ROS) within cells. The rise of free radicals and radical-derived

reactive species within the cells dictate the cellular response under conditions of

hyperglycemia-induced oxidative stress. This oxidative stress can slow down cells ability to

proliferate and instigate cellular senescence and apoptosis (Martindale & Holbrook, 2002;

Burch & Heintz, 2005). Thus the main pathophysiological mechanism which associates

hyperglycemia with certain diseases like atherosclerosis, nephropathy and cardiomyopathy is

production of oxygen derived free radicals (Francia et al., 2009).

Pathology of diabetic cardiomyopathy (DCM) is characterized by myocardial injury,

hypertrophy, interstitial fibrosis, structural and physiological changes of small coronary

vasculature, disturbance of the management of the metabolic cardiovascular load, and cardiac

autonomic neuropathy (Voulgari et al., 2010). Stem cells have the potential to revive diabetic

Page 21: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

heart (Zhang, 2008). The results of the stem cells therapy however, depends on the prevailing

levels of ROS-induced oxidative stress and the microenvironment of the diabetic heart.

Proliferation and mobilization potential of stem cell is compromised due to the

development of oxidative stress (Thum et al., 2007; Sambuceti et al., 2009). In this scenario

the preservation of stem cell function to support its proliferation and differentiation is

possible through various strategies (Gallagher, 2007; Ohshima, 2009). Use of growth factors,

hypoxia, and anti-aging compounds are among some preconditioning strategies (Hahn et al.,

2008; Rosova et al., 2008) meant for enhancing potency of stem cell (Haider & Ashraf,

2008). Growth factor preconditioning has been recently documented to improve the

cytoprotective potential of stem cells and increase its effectiveness for cell therapy (Rosova

et al., 2008). Similarly previous studies have indicated that the antioxidant system can be

activated to provide protection against oxidative stress by preconditioning with antioxidants

(Orzechowski, 2003).

Among other cytokines, known to improve cellular functions, erythropoietin (EPO)

has also been documented for the improvement in different disease modalities such as

diabetes, neurodegeneration, renal and cardiovascular systems (Maiese et al., 2008). The

EPO administration has shown to decrease complications and increase cardiac functions in

diabetes (Silverberg et al., 2006). In conditions such as ischemic cardiac disease, EPO

administration improved cardiac function, inhibited cardiomyocyte apoptosis and enhanced

cardiac remodeling (Gao et al., 2007; Toma et al., 2007; Asaumi et al., 2007; Westenbrink et

al., 2007). Furthermore, studies have shown that EPO independently leads to angiogenesis

(Reinders et al., 2006; Li et al., 2007). These findings indicate the importance of EPO

Page 22: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

administration in the improvement of diabetic myocardium, which may also help

preconditioned stem cells to perform better after being transplanted in the diabetic heart.

Mesenchymal stem cells (MSCs) are regarded as an important source for

cellular therapies (Kudo et al., 2003; Orlic et al., 2001) owing to their potential to

differentiate towards multilineage (Sharif et al., 2007; Krause, 2002). Yet, the available data

do not report any study known to date that characterizes the effect of diabetes on MSCs

function. This is particularly relevant to our quest for treatment of diabetic HF.

The present study reports that diabetes impairs MSCs function by increasing cell

senescence and death and downregulating the survival factors. To improve the function of

diabetes-impaired MSCs and provide protection from potential hypoxic and high glucose

insults a preconditioning strategy based on growth factors was employed. The diabetic MSCs

were preconditioned with insulin-like growth factor-1 (IGF-1) and fibroblast growth factor-2

(FGF-2). The preconditioning enhanced proliferation of MSCs, downregulated proapoptotic

factors, upregulated survival factors, enhanced their angiogenic ability and tolerance to

hypoxia and high glucose insults. The study thus identifies IGF-1 and FGF-2 based

preconditioning to be unique of its kind for the functional improvement of diabetes affected

MSCs.

In view of the documented cardio-protective role of EPO (Asaumi et al., 2007;

Ruifrok et al., 2008; Maiese et al., 2008), a pilot study was also done to investigate the

potential of EPO treatment on functional enhancement of the diabetic heart. The effects of

EPO treatment alone on the diabetic heart were first determined. For this purpose mice at

diabetic day 40 were divided into two groups which included diabetic untreated mice and

diabetic mice administered with injections of EPO (1000 IU /kg i.p.) for five consecutive

Page 23: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

days. The normal mice group served as control. The gene expression analysis of the left

ventricular tissue on day six showed down regulation of p16ᴵᴺᴷ⁴ᵃ and Bax genes and

upregulation of IGF-1in the diabetic EPO-treated group compared with the diabetic untreated

group. Tunel staining also showed significant reduction in myocardial cell apoptosis in the

EPO-treated group compared with the untreated group. This was concomitant with a better

eNOS expression in the left ventricular tissue of the EPO-treated group compared with the

untreated group.

After the initial establishment of the beneficial effects of EPO on diabetic heart the

combined strategy of transplantation of preconditioned diabetic MSCs followed by EPO

treatment was employed. The transplanted mice groups at diabetic day 40 included mice

transplanted with untreated diabetic MSCs (diabetic untreated group), mice transplanted with

preconditioned diabetic MSCs (diabetic preconditioned group) and mice transplanted with

preconditioned diabetic MSCs and given EPO treatment (diabetic EPO-treated group). The

non-transplanted groups included normal, diabetic, diabetic sham, and diabetic EPO-treated

mice. Four weeks after transplantation Millar analysis of the various treatment groups was

done to assess heart functions. The combined strategy of transplantation of preconditioned

diabetic MSCs and EPO administration ensured significantly high homing of MSCs,

reduction in fibrosis and marked improvements in cardiac functions compared to all other

treatment groups.

The results of the pilot study employing EPO treatment thus showed better effects on

the microenvironment of the diabetic heart and when used in conjunction with transplantation

of preconditioned diabetic MSCs improved the cardiac status of the transplanted mice.

Despite these considerable improvements involving use of combined strategy, the cardiac

Page 24: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

functions of the diabetic mice were below the normal standard base line value, which was

understandable and can be associated with diabetes. It is therefore suggested that future

studies be aimed at either increasing the dose or duration of EPO treatment for up to seven

days. Further the analysis of heart function should be done after eight weeks to further

elucidate the functionality of the diabetic heart following this treatment strategy.

The present study thus identifies IGF-1/FGF-2 based preconditioning to be a novel

strategy to enhance the function of diabetes-impaired MSCs and transplantation of these

preconditioned MSCs together with EPO treatment represents a unique combined strategy of

its type not known yet to treat diabetic HF.

Page 25: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CHAPTER 2

REVIEW OF LITERATURE

Page 26: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.1 AN INTRODUCTION TO STEM CELLS

2.2 Stem Cells

Stem cells are classified as a subset of cells that have the potential of self renewal and

differentiation in to one or more mature cell types (Verfaillie, 2002; Orkin & Zon, 2002;

Anderson et al., 2001). The division of stem cells during self renewal is symmetric i.e. one

stem cell produces two daughter stem cells while it is asymmetric during course of

differentiation. In asymmetric division stem cell produces a daughter stem cell and a

progenitor cell. The progenitor cell is committed towards a particular cell type and can

develop in to a mature differentiated cell type (Gomperts & Strieter, 2007).

2.3 Types of Stem Cells

Stem cells can be classified on the basis of various parameters such as their anatomy,

functions, transcription factors and proteins. However, the two broad divisions of stem cells

are embryonic stem cells (ESCs) and adult stem cells (ASCs). ESCs are found in embryo

while ASCs are found in different adult tissues (Mathur & Martin, 2004).

2.3.1 Embryonic Stem Cells (ESCs)

Blasotcyst have inner cell mass which provide a good source to derive ESCs at

around 5 days after fertilization and these ESCs are pluripotent. This means that they are able

to differentiate into three germ layers i.e. ectoderm, mesoderm and endoderm (Chambers &

Smith, 2004; Thomson et al., 1998). The development of embryo leads to the formation of

stem cells for reproduction and organogenesis termed as germ line stem cells (GSCs) and

somatic stem cells (SSCs) respectively. Although diversified from the ESCs the GSCs and

SSCs retain the property of self renewal. They either progressively restrict in to multiple

lineages or become unipotent (Fuchs et al., 2004; Rossant, 2004; Weissman, 2000).

Page 27: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.3.2 Adult Stem Cells (ASCs)

After birth ASCs including GSCs and SSCs reside in a special microenvironment

called ―niche‖. The location and characteristics of this niche varies and depends on the tissue

types (Linheng & Ting, 2005). ASCs are also referred as progenitor cells so that they may

not be taken up as totipotent (Herzog et al., 2003; Krause et al., 2001). This naming is,

however, confusing in the sense that they might be thought of as committed progenitor cells

or transient multiplying cells, neither of which qualify the criteria for stem cells (Gomperts &

Strieter, 2007). ASCs are thus considered as multipotent or unipotent which can differentiate

in to one or more mature cell types (Fuchs & Segre, 2000; Alison et al., 2002). ASCs are

found in a variety of adult tissues including skin, muscle, intestine, testis, heart and bone

marrow.

2.4 Bone marrow (BM)

BM contains a heterogeneous set of different population of cells such as

hematopoietic stem cells (HSCs), mesenchymal stem cells (MSCs), endothelial progenitor

cells (EPCs) and multipotent adult progenitor cells (MAPCs).

2.4.1 Hematopoietic Stem Cells (HSCs)

The best characterized types among ASCs are the HSCs which are linˉ CD45⁺, Sca-

1⁺ (only in mice) and c-kit⁺ (Fuchs & Segre, 2000; Alison et al., 2002; McCulloch & Till

2005). HSCs can give rise to all types of mature blood cells.

2.4.2 Mesenchymal Stem Cells (MSCs)

MSCs are regarded as a potential tool for cellular therapy. MSCs represent a part of

multipotent adult stem cell population which if provided suitable culture conditions can

differentiate in to osteocytes, chondrocytes, adipocytes, myocytes, and cardiomyocytes i.e.

Page 28: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

cells of mesodermal origin as well as in to hepatocytes and neuronal cells of non-mesodermal

lineages (Wagner et al., 2008). MSCs are known to express CD44, CD90, CD105, CD106,

but not CD45. However, there still exists lack of consensus regarding their receptors

profiling.

2.4.3 Endothelial Progenitor Cells (EPCs)

EPCs enter the peripheral blood after they leave bone marrow. They circulate and

home at various adult tissue sites. EPCs express markers such as CD34, VEGFR, and Ties-2

but are negative for CD45 (Asahar & Kawamoto, 2004; Urbich & Dlmmeler, 2004).

Recently flow cyotmetry studies on human EPCs isolated from adult peripheral blood,

adipose tissue and liver has demonstrated expression of CD105 on EPCs from these sources

(Tarnok et al., 2009). EPCs are known to play important role in regeneration of vasculature

in adult organ.

2.4.4 Multipotent Adult Progenitor Cells (MAPCs)

BM constitutes one of the sources for in vitro culturing of MAPCs (Jiang et al.,

2002). MAPCs may occur along with MSCs in the MSCs culture due to their adherent

nature. So MAPCs may represent a side population of MSCs. However MAPCs are different

in the sense that they can be grown indefinitely in the presence of few growth factors

(Gomperts & Strieter, 2007). The remarkable plasticity seen in both MSCs and MAPCs may

be attributed to their ability of cell fusion. Transdifferentiation is the term used for change in

phenotype of one cell into another type of cell which may be the result of cell fusion (Terada

et al., 2002; Ying et al., 2002).

ASCs play an important role in tissue homeostasis by supporting tissue regeneration

and replacing the cells lost due to injury and apoptosis. So a fine balance between both is

Page 29: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

inevitable for normal functions to ta e place in an individual’s life span. The study of

mechanisms that govern this fine balance is crucial to our understanding of stem cell

regulation, cancer development and potential therapeutic use of stem cell (Linheng & Ting,

2005).

2.5 CARDIOVASCULAR SYSTEM

Cardiovascular system is comprised of heart and associated blood vessels by which

blood is pumped and distributed throughout the body. This distribution of blood occurs by

three important forms of blood circulations which are: pulmonary, systemic and coronary.

During which blood supplies nutrients, oxygen, hormones, salts, etc. vital for the functioning

of all body tissues and removes the wastes harmful for these tissues.

2.5.1 Heart

Human heart is a fibromuscular pumping organ that lies in the middle mediastinum

between the lungs. It is enclosed by a pericardium which consists of two components that

include a fibrous and a serous pericardium. Heart is somewhat conical in shape. The two

distinct parts of a heart include an apex and a base. An average adult heart is 12 cm long

from base to apex and a diameter of 8 to 9 cm at the broadest part. It weighs about 300g in

males and 250g in females (Standrings et al., 2008).

Heart comprise of four chambers. These are two atria and two ventricles. The division

of heart in to four chambers produces externally visible grooves called sulci (Fig.2.1A). The

two atria have weak contractility and empty blood in to ventricles which with their powerful

contraction pump blood to the arterial trunk. The figure 2.1B briefly describes the key

anatomical parts of the heart.

Page 30: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 2.1 Anatomy of heart A. sulci B. internal anatomy.

2.5.2 Microstructure of the Heart

Heart consists of three layers. An outer thin epicardium, middle thick myocardium

and an inner thin endocardium. The epicardium and endocardium consist of simple squamous

epithelium and simple squamous endothelium respectively overlying a thin layer of areolar

tissue. The middle layer which is thickest called myocardium. The bulk of myocardium

consists of muscle cells called cardiac cells, cardiac myocytes or cadiomyocytes (Saladin,

2005). It should be noted that the muscles of the ventricle are spiral and this is the reason that

in the microscopic sections cardiac muscle fibres never appear linear in arrangement. Linear

arrangement is only seen in sections of papillary muscles and trabeculae carneae.

Cardiac muscle cells are elongated in shape and branched at their ends containing one

or two nuclei. The average size of a normal cardiac muscle cell is 120µm long and 20-30 µm

in diameter. The arterial muscle cells are smaller in size as compared to the ventricular

muscle cells. Some 35 % of the cardiac cell volume is occupied by mitochondria indicative

of requirement for high oxidative metabolism. In this context cardiac muscle cells are

Page 31: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

supplied by a heavy anastamosing complex of capillaries. This clearly shows why the cardiac

cells are sensitive to ischemic events. This complex of capillaries also provides a potential

means to develop collateral circulation in case of ischemic attacks. The ends of cardiac

muscle cells are joined together to form a complex and the adjacent cardiac cells are

connected with each other by gap junction complex. The forces of contraction of muscle cells

are strengthen by gap junctions and their complex networking establishes contractions as

syncytium. The contractile proteins arranged in sarcomere in the cardiac muscle cells are

responsible for this contraction. The sarcoplasmic reticulum lies in close association to the

sarcomere and is concerned with the storage, release and reaccmulation of calcium (Fig.2.2).

When calcium is released in to the sarcoplasm, it causes contraction which corresponds to the

systolic phase of the heart cycle. The reuptake of calcium by sarcoplasmic reticulum

produces relaxation that corresponds to the diastolic phase of the heart cycle (Standrings et

al., 2008).

Page 32: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 2.2. Microstructure of the heart tissue.

2.5.3 Blood Circulation

Blood distributes nutrients, oxygen, hormones, etc. throughout the body as a result of

pumping action of the heart. It is this contraction of the heart which, together with the help of

great vessels and the complex network of the arteries, veins and capillaries, ensures an

efficient blood circulation. Pumping of the heart feeds a minor loop which takes blood

towards the lungs for oxygenation, constitute pulmonary circulation. While the major loop it

feeds, constitute systemic circulation for the distribution of the oxygenated blood. With few

limited exceptions the blood remains within these loops and does not usually leave the

circulation. This is what known as a closed type of blood vascular system.

During course of blood circulation the right atrium receives venous blood from

superior vena cava, inferior vena cave and the main venous inflow from the heart itself. The

Page 33: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

venous blood enters the right ventricle through the atrioventricular orifice guarded by the

tricuspid valve. The contraction of the right ventricle forces the venous blood into the

pulmonary trunk through the pulmonary valve. The contraction of the right ventricle

particularly its apical trabecular component ensures closure of the tricuspid valve and

prevents blood to re enter in to the right atrium again. The left atrium receives all the

oxygenated blood coming from the lungs together with some coronary venous in flow.

Contraction of the left atrium causes the blood to enter left ventricle through the

atrioventricular orifice guarded by the mitral valve. The contraction of the left ventricle

rapidly increases pressure on its apical trabecular component which closes mitral valve. This

strong buildup of the pressure opens the aortic valve and ejects the blood into the aortic

sinuses and the ascending aorta. The blood is thus distributed to all parts of the body through

the arterial tree. The exchange of gases, nutrients and wastes then takes place at the level of

capillaries, after which the blood is again supplied back to the heart by venous system of our

body.

The complex vascular bed in the entire body generates a peripheral resistance which

helps us to know the reason for the massive structural organization of the left ventricle.

Although the ejection phase of the left ventricle is shorter than the right ventricle but the

pressure variation in the left ventricle is more than the right ventricle. So the heart be viewed

as far from a simple pumping organ (Standrings et al., 2008).

2.6 CARDIOVASCULAR DISEASES

The spread of heart disease is excessive in the developed world and spread of this

epidemic around the globe is also rapid (Lloyd-Jones et al., 2010). Among various factors

contributing to an increase in the prevalence of heart disease, diabetes is one of the major

Page 34: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

contributors. Aging, obesity, and sedentary life style are some of the main factors that

contribute to a significant increase in the prevalence of diabetes.

2.6.1 Diabetes and Cardiovascular Diseases

In late 1800s it was recognized for the first time that diabetes is associated with

cardiac diseases (Gaede et al., 2003). Later, studies provided definite evidence of the role of

diabetes in HF and indicated the increased risk of HF in diabetics compared with the subjects

without diabetes (Kannel et al., 1974; Kannel & McGee, 1979). The cardiovascular

complications of diabetes have been reported to account for 80% of mortality in diabetics

(Amos et al., 1997) and are considered as one of the leading causes of deaths in diabetics

(Candido et al., 2003; Sowers et al., 2001; Mazzone et al., 2008). Hyperglycemia has also

been implicated to increase the mortality rate in patients of acute myocardial infarction. In

animal model diabetes has shown to enhance severity of acute myocardial infarction (AMI)

by causing an increase in infarct size and development of HF (Di Filippo et al., 2005; Frantz

et al., 2005; Liu et al., 2005; Marfella et al., 2004; Verma et al., 2002).

Diabetes induced damage to the cardiac muscle has been reported by both animal and

clinical data as mentioned above. The association of diabetes with some of the most known

risk factors for HF is established now. Among such risk factors include obesity,

hypercholestoremia, hyperlipidemia, hypertension, infarction, endothelial dysfunction,

coronary artery disease and activation of number of cytokine and cytokine system (Wang et

al., 2006) as depicted in Figure 2.3.

Although hypertension and coronary artery disease are mainly responsible for the

most of the myocardial abnormality such as LV hypertrophy and impaired contractility seen

in diabetics, the evidence of cardiomyopathy has been reported long ago to contribute to

Page 35: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

myocardial dysfunction in absence of coronary artery atheroma (Fisher et al., 1986). The

development of DCM in the absence of established risk factors has also been recently

documented (Sarwar et al., 2010).

Figure 2.3. Some of the diabetes associated risk factors for HF. AGE: advanced glycation

end product. (Adopted from Wang et al., 2006).

2.6.2 Diabetic cardiomyopathy (DCM)

Rubler et al coined the phrase ― diabetic cardiomyopathy‖ and described diabetes as

the key factor that elicits changes at the cellular and molecular level and results in structural

and functional abnormalities of the heart (Rubler et al., 1972). Cardiomyopathy in type 1 or

type 2 diabetic patients is associated with a cluster of common features (Wang et al., 2006)

shown in Figure 2.4.

Page 36: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 2.4. Interacting pathways of ROS production and injury in diabetic cardiomyocytes.

FFA: free fatty acid; FATP: free fatty acid transport protein; CPT-1: carnitine

palmitoyl transferase-1; AGE: advanced glycation end products; AT1R:

angiotensin 2 type 1 receptor; Glut: glucose transporter; SR: sarcoplasmic

reticulum. SRCA: sarcoplasmic reticulum Ca 2+-ATPase.(Adopted from Wang

et al., 2006).

The mechanisms at the molecular level which cause diabetic cardiomyopathy are

poorly known. The previous findings indicate that enhanced cardiac fibrosis (Tschope et al.,

2004; Westermann et al., 2007; Zhang et al., 2008) generation of ROS and cardiac

inflammation (Li et al., 2007; Tschope et al., 2005; Westermann et al., 2007) are some

important contributors leading to DCM. Enhanced dependence of diabetic cardiomyocyte on

fatty acid metablolism has been previously reported to be an uncertain but a suspect cause of

Page 37: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

cardiomyopathy (Yagyu et al., 2003). This excessive dependence on fatty acid metabolism

can produce harmful effects on mitochondria (Di Paola & Lorusso, 2006) and one of the fatty

acid, palmitate has been described to induce apoptosis in cardiomyocytes (Sparagna et al.,

2004) and damage the contractile apparatus (Dyntar et al., 2001).

Hyperglycemia has been implicated in cardiomyopathy based on vast evidence

reported from diabetes type 1 and type 2 experimental models(An & Rodrigues, 2006). ROS

and AGEs comprise important components known to cause cellular injury as a result of

exposure to high glucose. The production of stable covalent modifications of proteins by

glucose or glucose metabolites leads to formation of AGEs. The protein adducts not only

contribute to ROS generation but can cause direct damages as well (Wang et al., 2006).

The recent studies also indicate hyperglycemia induced mitochondrial ROS to be a

major contributor in the development of DCM (Hayat et al., 2004 ; An & Rodrigues, 2006;

Boudina & Abel, 2010; Dobrin & Lebeche, 2010).

Pathology of DCM is characterized by myocardial damage, hypertrophy, tissue

fibrosis, structural and physiological changes of small coronary vessels, cardiac autonomic

neuropathy and disorder of the metabolic cardiovascular load. These modifications make it

difficult for the diabetic heart to recover from ischemic attack and increase its susceptibility

to ischemia. Arterial hypertension frequently coexists with and exacerbates cardiac

functioning, leading to the premature appearance of HF. (Voulgari et al., 2010).

In conclusion, the pathogenic contribution to the development of DCM includes

insulin resistance, increased formation of AGEs, Metabolic syndrome (MetS),

hyperglycemia, dyslipidemia, obesity, and presence of microangiopathy. Their interplay is

complex, since they can act in parallel and synergically and, at the same time, have a cause

Page 38: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

and effect result. Thus, they should be equally predicted and effectively treated (Voulgari et

al., 2010). The contribution of the various factors in the development of DCM is depicted in

Figure 2.5.

Figure 2.5. The pathophysiological substrate of DCM: in diabetes, hyperglycemia, excess

FFA release, and insulin resistance, cause adverse metabolic events that affect

the cardiac myocytes. Hyperglycemia is associated with decreased glucose

transportation (GLUT), uptake, and oxidation, as well as increased formation of

AGEs and increased activation of protein kinase C (PKC). Excess FFA release is

followed by cardiac lipotoxicity, i.e. increased cardiac lipid accumulation and

increased generation of reduced ROS at the level of the electron transport chain.

Together with insulin resistance and impaired insulin action and signaling, these

metabolic paths augment vasoconstriction, produce and further aggravate arterial

hypertension, increase inflammation with liberation of leukocyte-attracting

chemokines, increase production of inflammatory cytokines, and augment

expression of cellular adhesion molecules. Thrombosis is further promoted,

together with platelet activation. (Adopted from Voulgari et al., 2010).

Page 39: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.7 TREATMENT AND MANAGEMENT OF DCM

2.7.1 Life style

Obesity in both type 1 and type 2 diabetics is an important issue in the management

of DCM. Important management measures, therefore, include increase in physical activity,

reduction in fat consumption and total energy intake. These changes in life styles have been

shown to significantly reduce the incidence and magnitude of DCM (Guang et al., 2002; Hu

et al., 2004). Reduction in magnitude of DCM has also been reported following cessation of

smoking, which also reduces absolute risk of cardiovascular deaths in the diabetic subjects

compared with the non diabetics (Stamler et al., 1993; Kempler, 2005).

2.7.2 Glycemic Control

The UK Prospective Diabetes Study (UKPDS) indicates a reduction in risk for MI

and HF in type 2 diabetic patients following exposure to glycemia over time (UKPDSG,

2000). Good glycemic control thus represents another important approach towards

management of DCM. An analysis of multicenter, population based prospective study in

newly diagnosed type 2 diabetes patients, also indicates decrease in incidence of both risk for

myocardial infarction (MI) and DCM (Hanefeld et al., 1996; Meier & Hummel, 2009).

2.7.3 Dyslipidemia Treatment

Use of cholesterol lowering agents in diabetic patients is associated with a decrease in

incidence of coronary heart disease as indicated by a double-blind, randomized, placebo-

controlled, multicenter clinical trial study (Haffner, 1997). A similar study using gemfibrozil

also reported a decrease in incidence of DCM and major cardiovascular events in diabetic

patients (Robins et al., 2001).

Page 40: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.7.4 Antihypertensive Treatment

The use of antihypertensive medicines to keep tight control of blood pressure (B.P.)

(<155/85mm Hg) in patients of hypertension has been reported to be associated with

decrease in cardiovascular risk and HF. A randomized control study indicates some 24%

reduction in cardiovascular risk related to diabetes and 56% reduction in risk of HF following

tight control of hypertension (UKPDSG, 1998). Another study employing strategy of

lowering B.P. in diabetic patients indicates reduction in fatal and nonfatal macrovascular and

microvascular complications (Holman, 1998).

2.8 SOME ADDITIONAL THERAPEUTIC STRATEGIES:

2.8.1 Apelin and Cardiac Output

In conditions of pressure overload such as DCM, apelin plasma levels have been

associated with compensatory mechanism to maintain cardiac output. In one such study

conducted in streptozotcin-induced diabetic rats the myocardial expression was found to be

down regulated as against its plasma levels after increase of pressure over load. Similar

findings were seen in diabetic patients in another parallel study (Falcão-Pires et al., 2010). So

apelin may represent a possible target for treatment of conditions due to pressure overload.

2.8.2 Kinins

The Kallikrein-kinin system (KKS) has been shown to protect from inflammation,

fibrosis, and apoptosis in myocardial disease. Kinins which are vasoactive peptides are part

of KKS and recent observations indicate their role in different aspects of remodeling,

inflammation and angiogenesis. Formation of new blood vessels, recruitment of EPCs in the

ischemic areas and endothelial dysfunction are some of the novel functions of KKS. The

KKS therefore has potential to treat DCM and myocardial ischemia (Savvatis et al., 2010).

Page 41: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.8.3 Sarcoplasmic Reticulum Calcium ATPase (SERCA2)

It has been previously indicated that the reduction of SERCA2 expression plays a

significant role in the myocardial dysfunction of DCM (Rodrigues et al., 1998). A recent

study carried out in this perspective on streptozotocin induced male diabetic mice focused on

the activation of SERCA2 expression by administring resveratrol. The data demonstrate that

in DCM, where the expression of SERCA2 is reduced, resveratrol enhances the expression of

the SERCA2 and improves cardiac function (Sulaiman et al., 2010).

2.8.4 Antioxidants

Role of ROS in the development of DCM has been previously documented (Devereux

et al., 2000; Singh et al., 2001). ROS induced matrix metalloproteinase2 (MMP2) activation

has been implicated in conditions of acute loss of myocardial contractile functions. In a

recent in vivo study in STZ-induced diabetic rats the use of sodium selenate or pue Omega 3

fish oil with antioxidant vitamin E has demonstrated prevention of diabetes induced

functional changes. These agents could have therapeutic benefits in DCM (Aydemir-Koksoy

et al., 2009).

The suppression of mitochondrial oxidative stress and mitochondrion-dependent

myocardial apoptosis was aimed in another study with the administration of antioxidant α-

lipoic acid (α-LA) to check progression of DCM. In a streptozotocin-induced diabetic rat

model α-LA was administered (100 mg/ g i.p. per day) for 12 wee s. ntioxidant α-LA

effectively attenuated mitochondrion dependent cardiac apoptosis and exerted a protective

role against the progress of DCM. This activity of α-LA was concomitant with an increase in

manganese SOD activity and glutathione content of myocardial mitochondria (Li et al.,

2009).

Page 42: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

2.8.5 Erythropoietin (EPO)

EPO is a hematopoietic cyto ine which was initially termed as ―hemopoietine‖. The

ability of EPO to stimulate erythropoiesis was first demonstrated in 1906 by the

revolutionary study of Carnot and DeFlandre. Recent studies indicate the presence of EPO

receptors (EPOR) on nonhematopoietic organs including heart (Parsa et al., 2003; Calvillo et

al., 2003). The direct involvement of EPO on cardiomyocytes has been strengthened by the

findings of EPOR on myocardial tissues of various species including man (Wright et al.,

2004; Deeping et al., 2005). The discovery prompted a keen interest of the researchers to

discern nonhematopoietic functions of EPO. This is evident from some recent studies which

demonstrate tissue protective effects of EPO and its role in prevention of vascular and tissue

damage in conditions of acute ischemia in different organs including heart (Ruschitzka et al.,

2000; Fliser & Haller, 2007; Lipsic et al., 2006). The protective role of EPO at the cellular

level involves multiple signal transduction pathways and is depicted in the figure 2.6. Recent

insights in to mechanism involved in cytoprotection of H9C2 cells, both perfused and in situ

rat hearts, reveal an upregulation of Akt, ERK1/2 and JAK1-STAT signal pathways (Parsa et

al., 2003; Bullard et al., 2005). Besides this EPO has been implicated in enhancing release of

nitric oxide (NO) which carries significance in prevention of cardiovascular diseases. NO

released, eNOS being the predominant form in the vascular system, from the intact

endothelial cells is known to cause vascular relaxtion and provide antiproliferative and

antithrombotic functions. Ruschitzka et al. (2000) generated transgenic EPO overexpressing

polyglobulic mice. These mice were observed for survival and cardiovascular functions to

determine role of NO. It was found that blood pressure, heart rate and cardiac output of

transgenic mice remained unaltered despite high hematocrit values as compared to the non-

Page 43: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

transgenic littermates. The EPO overexpressing mice had a six fold increase in levels of

eNOS. The transgenic mice did not develop hypertension, MI, stroke or thromboembolism

apparently because of the constitutive expression of eNOS associated with high NO

bioavailability.

Figure 2.6. Signal transduction pathways involved in EPO mediated cytoprotection.

(Adopted from Maiese et al., 2008).

Initial findings in late 90s demonstrated protective role of EPO in ischemic brain

injuries (Sadamoto et al., 1998; Sakanaka et al., 1998). Protective role of EPO against

ischemia-reperfusion injuries of heart tissue was elucidated in number of subsequent studies

such as those carriedout by Calvillo et al. (2003) and Tramontano et al. (2003). Similarly

Moon et al. (2003) demonstrated reduction in infarct size following intravenous

administration of EPO (3000 I.U. /kg) at the time of coronary ligation in a myocardial

Page 44: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

ischemia model. In an experimental setup of isolated perfused rat hearts, Cai et al. (2003)

reported improvement in left ventricular function and reduction in apoptosis of

cardiomyocytes 24 h after EPO administration. Four independent studies evaluating role of

EPO in post MI left ventricular dysfunction also confirm the useful effects of EPO on

microvascularization and cardiac function (Li et al., 2006; Hirata et al., 2006; Toma et al.,

2007; Prunier et al., 2007). EPO has been described to preserve cardiac function in cases of

chronic HF and myocardial ischemia largely by neovascularization and inhibition of

apoptosis (Ruifrok et al., 2008). Recently in a mice model of coronary ligation, EPO has

been shown to decrease infarct size, inhibit left ventricular remodeling, enhance angiogenesis

and improve cardiac function (Ueda et al., 2010).

Although the studies investigating role of EPO on heart under diabetic conditions are

limited, yet these indicate beneficial effects of EPO administration. In diabetic and non

diabetic subjects with severe and resistanat congestive HF, EPO administration has been

reported to decrease fatigue and increase LV ejection fraction. This improved the health

status of the patients and significantly increased their early discharge from hospital

(Silverberg et al., 2006). In vitro studies of Chong et al. (2007) report strong cytoprotective

effects of EPO which enhanced survival of endothelial cells (ECs) exposed to high glucose.

A recent study has focused on evaluating the role of EPO treatment in DCM and as claimed

represents first of its kind in demonstrating beneficial effects of a recombinant human EPO

(rHuEPO) treatment on chronic non-ischaemic cardiac tissue injury in mice diabeitic model.

Shushakova et al. (2009) have demonstrated that chronic rHuEPO treatment, in a mouse

model of diabetes exhibiting clinical features of DCM characterized by fibrosis and

contractile dysfunction inhibits cardiac fibrosis and protects cardiac tissue.

Page 45: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

The findings mentioned above are clearly suggestive of beneficial effects of EPO in

different heart ailments and thus holds significance to treat diabetic HF.

2.8.6 Cellular Therapy

Reduction in number of cardiomyocytes is evident in many forms of both paediatric

and adult heart diseases. Repopulating such hearts with new cardiomyocytes can potentially

reverse cardiac diseases. Different approaches have been adopted in this respect. These

involve transplantation of skeletal myoblasts, cardiomyocytes, mobilization and

transplantation of stem cells (Rubart & Field, 2006). Recently the discovery of resident

cardiac stem cells has given a new insight into our understandings regarding regenerative

capability of heart (Bearzi et al., 2007).

Previous data indicate that bone marrow cells constitute a collection of stem and

progenitor cells and are most extensively studied in animal and clinical studies designed for

cardiac repair (Fazel et al., 2006; Haider & Ashraf, 2005). In addition endometrial stem cells,

circulating blood-derived progenitors, induced pluripotent stem cells, umbilical cord cells

and mononuclear cells are some of the diverse cell types which have also been proposed for

heart repair (Voulgari et al., 2010).

In the repertoire of bone marrow cells, MSCs represents a common source of adult

stem cells. Cardiomyognic differentiation of MSCs has been demonstrated in both in vivo

and in vitro studies (Shake et al., 2002; Toma et al., 2002; Wang et al., 2000). MSCs have

also been documented for their paracrine effects. The effects of media of MSCs culture has

been shown to be similar to MSCs therapy. Similarly in a murine model of hind limb

ischemia, enhancement of tissue repair was attributed to secretion of multiple cytokines such

as VEGF, FGF-2 and placental growth factor (PIGF) by MSCs (Kinnaird et al., 2004). Basic

Page 46: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

characteristics of MSCs such as ease in acquisition, isolation, and expansion together with

low immunogenicity make them suitable for potential clinical applications (Mishra, 2008).

Extensive studies in animals and clinical studies conducted during the last decade

indicate the feasibility and potential of cell based therapy for cardiac repair (Anversa et al.,

2007; Sanchez & Garcia-Sancho, 2007) which can be potentially used in the treatment of

DCM to restrict consequences of decrease contractile functions and compliance of damaged

ventricles. Patients receiving stem cell therapy should be in early stage of HF. The therapy,

however, is not an alternate to heart transplantation and foremost objective is to avoid or

delay organ transplantation (Voulgari et al., 2010). Besides these limitations and the low

regenerative potential of human heart, the efforts to remuscularize the damaged heart are in

progress. Some of the main strategies employed for heart repair use adult stem cells and

pluriipotent stem cells, cellular reprogramming and tissue engineering which are expected to

treat or prevent HF in a better way (Laflamme & Murry, 2011).

2.8.7 Combining MSCs Transplantation and EPO Treatment

The ischemic myocardium has been described to be hostile environment for the

transplantation of MSCs and warrants strategies to improve its survival and engraftment

(Haider & Ashraf, 2008). Among various strategies include transplantation of MSCs

combined with EPO treatment. Previous studies have shown that the combined strategy

enhances efficiency of stem cells in different disease conditions. A study describes that EPO

can help in synergistic way with MSCs to enhance post ischemic neurogenesis (Esneault et

al., 2008). Similarly Zhang et al. (2006) demonstrated that EPO increases the therapeutic

potential of MSCs and improved angiogenesis and cardiac function. Recently Zhang et al.

(2007) have reported that combinating EPO and Intramyocardial delivery of bone marrow

Page 47: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

cells effectively enhanced cardiac repair by reducing fibrotic area and enhancing capillary

density. Apart from its role in wide range of processes in cardiovascular pathophysiology, the

clinical trials involving use of EPO for the HF and renal failure reveal contradictory results

(Mastromarino et al., 2011). Moreover the literature survey indicates that studies which

could explore potential of EPO treatment for diabetic heart combined with MSCs

transplantation are missing.

The present study, therefore, aims to characterize the effects of preconditioning on

diabetes-impaired MSCs and evaluate the potential of EPO treatment for improvement in

cardiac status of the diabetic mice after transplantation of preconditioned MSCs. The

application of this combined approach for improvement in cardiac status of the diabetic mice

is viewed as a pioneer study of its type known to date and holds significance for future

strategies to treat diabetic HF.

Page 48: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CHAPTER 3

MATERIALS AND METHODS

Page 49: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.1 Animals

The recommendations and procedural guide lines for the use and care of laboratory

animals were carefully observed throughout the experimental period as approved by the

Institutional Committee of the National Center of Excellence in Molecular Biology, Pakistan.

3.2 Mouse Model for Diabetes

Six to 8 weeks old C57BL/6 male wild-type mice were treated with intra peritoneal

injections of streptozotocin (55mg/kg) for five consecutive days to induce diabetes as

previously described (Leiter & McNeill, 1999). Owing to the rapid metabolization of

streptozotocin within 24 h mice were housed with absorbent bedding and given free access to

water and food. Blood glucose levels of mice were measured at day 16 after the first injection

and only those mice were included in the study that had blood glucose levels > 300mg/dL.

3.3 In vitro Study

3.3.1 Cell Culture

Isolation of MSCs was done from bone marrow as described earlier (Khan et al.,

2009). Cells were isolated from femur and tibia bones of 60-day diabetic C57BL/6 mice and

cultured in Iscove’s modified Dulbecco’s medium (IMDM) at 37°C in humidified air with

5% CO₂. IMDM used for cell culture was supplemented with streptomycin (100 µg/mL),

penicillin (100U/mL) and 20% fetal bovine serum (FBS). Non-adherent cells were removed

by changing the medium at 48 h post plating and then every 3 day afterward.

3.3.2 Flow Cytometry

MSCs of diabetic mice (N=6) were analyzed for CD34, CD45, CD44, CD90 and CD

105 by flow cytometry. MSCs were incubated with CD34PE, CD45FITC, CD44PE,

CD90FITC, and CD105FITC antibodies (BD Biosciences) for 30 min in dark at room

Page 50: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

temperature. Specific fluorescence of 10,000 cells was examined by using Cell Quest Pro

software on FACScalibur (Becton Dickinson).

3.3.2 Preconditioning of Diabetic MSCs

IGF-1 and FGF-2 growth factors (Santa Cruz Biotechnologies) were selected for

preconditioning. The two growth factors were used in different concentrations alone as well

as in combination. This was followed by selection of an optimum preconditioning regimen

based on cell proliferation assay using XTT as per manufacturer’s instruction (Roche).

Briefly, the assay was carriedout in a 96-well plate. Some 4×10³ diabetic MSCs were plated

in each well containing IMDM supplemented medium in a humidified incubator at 37°C with

5% CO₂ and left overnight. Cells were preconditioned under serum free conditions with IGF-

1 and FGF-2 in different concentrations (ng/mL) for 1 h and cell proliferation was quantified

at 48 h by measuring absorbance values at 450 nm using Spectra max PLUS 384 (Molecular

Devices). Absorbance values at 650 nm were also taken as reference wavelength.

Corresponding experiments involved preconditioning of diabetic MSCs with vascular

endothelial growth factor (VEGF) (Santa Cruz Biotechnologies) which is also known for its

potential to protect from hyperglycemia. For this purpose cell proliferation with VEGF

preconditioning was quantified for proliferating cell nuclear antigen (PCNA) by real time

polymerase chain reaction on a 7500 Real-Time PCR system (Applied Biosystems).

3.3.3 Gene Expression Profiling of MSCs

The three groups of MSCs that included preconditioned diabetic MSCs, untreated

diabetic MSCs and normal MSCs were used for RNA extraction using trizole reagent

(Invitrogen Corporation). RNA samples were subsequently quantified by ND-1000

spectrophotometer (NanoDrop Technologies). Moloney Murine Leukemia Virus (M-MLV)

Page 51: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

reverse transcriptase (Invitrogen Corporation) was used to synthesize cDNA from 1 µg of

RNA sample. cDNA samples were then used for reverse transcriptase-PCR (RT-PCR)

analysis for different proapoptotic, prosurvival, and cardiac markers which included IGF-1,

FGF-2, Akt, p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ, p53, Bax, Bak, GATA-4, and Nkx 2.5. RT-PCR analysis was

carriedout on a GeneAmp PCR system 9700 (Applied Biosystem) and glyceraldehydes 3-

phosphate dehydrogenase (GAPDH) was used as an internal control. The details of different

primers used with the description of their product size (bp) and sequences (5ˊ–3ˊ) have been

mentioned in Table 1. The RT-PCR products run on 2.5% agarose gel were quantified using

Quantity One ® 1-D Analysis Software, version 4.4 (Bio-Rad Laboratories, Inc.) as per

recommended guidelines with the quantity of the ladder (Fermentas) used as standard.

Page 52: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Table 1. List of primers, their product size (bp) and sequence 5´—3´.

Genes Product Size(bp)

Sequence (5´—3´)

GAPDH (f) 370 CTCTTGCTCTCAGTATCCTTG

GAPDH (r) GCTCACTGGCATGGCCTTCCG

β-Actin (f) 106 GCTGTGTTGTCCCTGTATGC

β-Actin (r) GAGCGCGTAACCCTCATAGA

IGF-1 (f) 166 AGGCTATGGCTCCAGCATTC

IGF-1 (r) AGTCTTGGGCATGTCAGTGTC

FGF-2 (f) 154 TGTCTATCAAGGGAGTGTGTGC

FGF-2 (r) CAACTGGAGTATTTCCGTGACC

Akt (f) 151 CCTCAAGAACGATGGCACCT

Akt (r) CAGGCAGCGGATGATAAAGG

p16ᴵᴺᴷ⁴ᵃ (f) 196 GCTCAACTACGGTGCAGATTC

p16ᴵᴺᴷ⁴ᵃ (r) TCGCACGATGTCTTGATGTC

p66ˢʱᶜ (f) 194 TGACTTCAATACCCGGACTCAG

p66ˢʱᶜ (r) TGAGGTTAAGGCTGCTGGTAGA

p53 (f) 157 AGCATCTTATCCGGGTGGAAG

p53 (r) CCCATGCAGGAGCTATTACACA

Bax (f) 152 TGGAGATGAACTGGACAGCA

Bax (r) CAAAGTAGAAGAGGGCAACCAC

Bak (f) 182 CAGGACACAGAGGAGGTCTTTC

Bak (r) TAGCGCCGGTTAATATCATCTC

Ang I (f) 143 GACACCTTGAAGGAGGAGAAAG

Ang I (r) GTGTCCATGAGCTCCAGTTGT

VEGF (f) 200 ACCCCGACGAGATAGAGTACAT

VEGF (r) CTTCTAATGCCCCTCCTTGT

PCNA (f) 720 GGTTGGTAGTTGTCGCTGTA

PCNA (r) CAGGCTCATTCATCTCTATCG

GATA-4 (f) 197 CCTCTCCCAGGAACATCAAA

GATA-4 (r) ACCCATAGTCACCAAGGCTG

Nkx 2.5 (f) 147 GGTCTCAATGCCTATGGCTAC

Nkx 2.5 (r) GTTCACGAAGTTGCTGTTGG

3.3.4 Hypoxic Stress

Diabetic MSCs (both untreated and preconditioned) were exposed to hypoxic stress

with H₂O₂ (100 µM) in serum free medium for 90 min followed by 1 h recovery in serum-

supplemented medium. After the recovery period the two sets of diabetic MSCs were

Page 53: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

analyzed for parameters such as superoxide dismutase (SOD) activity, apoptosis, in vitro tube

forming ability, and chemotactic attraction to evaluate the effects of preconditioning.

3.3.5 Superoxide Dismutase (SOD) Assay

SOD activity helps cells to cope with the oxidative stress. The two sets of MSCs

(both untreated and preconditioned) were analyzed for SOD activity using SOD activity

colorimetric assay kit (Abcam) as per recommended protocol. Briefly, after 24 and 48 h of

recovery period the cells of the two sets of MSCs were lysed with lysis buffer. SOD activity

from 10 µg samples of the total protein extract was measured by taking absorbance values at

450 nm using Spectra max PLUS 384 (Molecular Devices).

3.3.6 Apoptosis

Flow cytometric analysis for number of apoptotic cells in the two sets of diabetic

MSCs (untreated and preconditioned) was carriedout on FACScalibur (Becton Dickinson)

using fluorescein isothiocyanate (FITC) Annexin-V it as per manufacturer’s instructions

(Abcam).

3.3.7 In vitro Tube-forming Assay

Matrigel was first thawed on ice to avoid premature polymerization before plating.

Then to a pre-cooled 48-well tissue culture plate (Corning), aliquots of 50 µL matrigel were

plated into each well. The culture plate was then placed in an incubator at 37°C for about 30

minutes for polymerization of the matrigel. The two sets of diabetic MSCs (untreated and

preconditioned) were trypsinized with trypsin / Ethylene diamine tetra acetic acid (EDTA)

and plated on matrigel-coated wells at 1.5×10⁴ cells/well. The cells were incubated at 37°C

in 5% CO₂ and examined under a phase-contrast microscope (IX-51 Olympus) at 6, 24, and

48 h to evaluate the effect of preconditioning on development of tube like structures.

Page 54: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.3.8 Chemotactic Attraction

Preconditioning effect on mobilization of diabetic MSCs towards chemotactic signals

was studied as described earlier (Kucia et al., 2004). Briefly, to a 6-well transmembrane

culture plate (Costar) 2 mL of serum free medium was added in the lower chamber and

supplemented with 50ng/mL of stromal cell-derived factor-1α (SDF-1α; Upstate

Biotechnology). The wells without SDF-1α supplementation containing serum free medium

alone served as control. Diabetic MSCs (both untreated and preconditioned) in concentration

of 5×10⁴ cells/well were plated on the upper chamber after hypoxia treatment and examined

at 24 h for migration towards SDF-1α. The migrated cells were stained with 4´, 6-diamidino-

2- phenylindole (DAPI) and counted in 10 high power fields using an inverted microscope

IX-51 (Olympus).

3.3.9 Glucose Stress

To analyze the effect of preconditioning on the ability of diabetic MSCs to tolerate

hyperglycemia, diabetic MSCs (untreated and preconditioned) were given exposure to high

glucose stress (30 mmol/mL) for 1 h under serum free conditions followed by 1 h recovery in

serum supplemented medium. RNA extracted from the representative groups were used for

Quantitative PCR (qPCR) analysis of mRNA expression of Ang-I, VEGF, and p16ᴵᴺᴷ⁴ᵃ in a

reaction volume of 25 µL using Maxima SYBR Green qPCR Master Mix (Fermentas) on

IQ5 Multi color Real-Time PCR Detection System (Bio-Rad).

Page 55: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.4 In vivo Study

3.4.1 Diabetic Mouse Model

Diabetes was induced in C57BL/6 male mice using streptozotocin as mentioned

previously. Mice at 40 days being diabetic were included in the study for EPO treatment and

transplantation experiments with age and sex match animals as control were carriedout.

3.4.2 EPO Treatment

In the first phase of the experiments recombinant human EPO (Cheil Jedang

Corporation) alone was used to assess its effects on diabetic myocardium. The mice were

divided into three groups (N=6). These included normal control (group -1), diabetic untreated

(Group-2) and diabetic EPO-treated (Group-3). The mice in the treated group were given

injections (1000 IU/Kg i.p.) of EPO for five consecutive days while control and untreated

groups received injections of normal saline. On day six mice were euthanized and tissue

procurement carried out for subsequent RNA extraction and histological studies.

3.4.3 Transplantation and EPO Treatment

In the second phase of experiments, transplantation of MSCs was done. The mice

were divided into six groups (N=6). Four non-transplanted groups of mice included normal

mice (Group-1), sham operated diabetic mice (Group-2), the diabetic mice injected with

normal saline (Group-3) and the diabetic-EPO treated mice (Group-4). The remaining three

groups included diabetic mice transplanted with untreated MSCs (Group-5), diabetic mice

transplanted with preconditioned MSCs (Group-6) and the diabetic mice transplanted with

preconditioned MSCs and given EPO treatment (Group-7). Some 20,000 MSCs (both

untreated and preconditioned) at passage-1 from sixty days diabetic GFP transgenic male

mice were transplanted at two random sites in the left ventricles of the respective non GFP

Page 56: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

groups of diabetic mice (Group-5, 6 & 7) using a 30G syringe. The group-3 mice received

injections of normal saline only while EPO treatment for five consecutive days was given to

the mice of group-4 and group-7. Four weeks following transplantations functional analysis

of the heart was done using Millar apparatus and hearts of the euthanized mice from different

groups examined for MSCs homing and tissue fibrosis.

3.4.4 RNA Extraction and cDNA Synthesis

Freshly isolated hearts of the euthanized mice were dissected to separate left

ventricles of different treatment groups. RNA extraction was then carried out from 50mg of

LV heart tissues using Trizole reagent and quantified using ND-1000 spectrophotometer. 1

µg RNA from respective samples was used to synthesize cDNA using M-MLV reverse

transcriptase.

3.4.5 Gene Expression Study

RT-PCR was carried out for IGF-1 using GeneAmp PCR system 9700 (Applied

Biosystem) with β-actin as internal control. The gene specific primers (Table 1) were used

under PCR conditions of initial denaturation at 95°C for 5 min, 35 cycles of denaturation at

95°C for 45 sec, annealing at 60°C (IGF-1) and 58°C (β-actin) for 45 sec, extension at 72°C

for 45 sec, and final extension for 10 min at 72°C.

Quantitative RT-PCR was done for mRNA expressions of Bax and p16INK4a

using

7500 thermal cycler (Applied Biosystem). The 20uL reaction mixture (triplicate) was made

using maxima sybr green (Fermentas) and cDNA amplified using gene specific primers

(Table.1). Amplification conditions were same as described for RT-PCR with annealing

conditions at 58°C and 60°C for Bax and p16INK4a

respectively. β-actin was kept as internal

control and normalized gene expression analysis carried out using 7500 System SDS

software (version 1.3.1).

Page 57: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.4.6 Tissue Procurement for Histological Studies

For paraffin sections the hearts of the euthanized mice were first perfused while for

cryo-sections the hearts were freshly isolated and rapidly frozen in liquid nitrogen. The

isolated hearts were cut into three sections representing its apex, middle and base. The

middle sections were then undertaken for the respective processing procedures.

Briefly, for paraffin sections, the middle sections were fixed in 4% paraformaldehyde

(PFA) and then dehydrated in increasing ethanol grades (70%, 80%, 95% and 100%). This

was followed by clearing step in xylene. The sections were then placed in liquid paraffin at

70°C for 30 minutes twice and then embedded. 5µM thick sections were made using

microtom machine (HM-340E, Microm Inc.USA). For cryo-sections, sections were first

embedded in Tissue-Teck OCT (Sakura Torrance, CA, USA) then rapidly frozen in liquid

nitrogen. The tissue sections were kept frozen at -20°C until sectioning was done using

cryostat (Microm) at -20°C on tissue treated slides. 5µM thick sections were then processed

for tunel assay and immunostaining.

3.4.7 Tunel Assay

To assess LV myocardial cell apoptosis frozen serial sections 5µm thick from group-

1, 2&3 of EPO treatment experiments were subjected to tunel staining. Briefly sections were

first washed in PBS for thirty minutes and fixed for 10 minutes by 4% PFA. The sections

were labeled with TUNEL as per protocol provided by the manufacturer (Millipore). After

incubation with Avidin-FITC for 30 minutes in dark, the sections were counterstained with

DAPI (Tuo et al., 2008). Apoptosis was quantified by counting tunel positive cells in the left

ventricle at six random sites per slide in sets of three slides per treatment group and average

taken.

Page 58: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

3.4.8 Immunostaining for eNOS

The frozen ventricular tissue sections of the above mentioned treatment groups of the

EPO experiment were air dried and washed with PBS for 5 minutes followed by fixation in

ice cold methanol for 15 minutes. The sections were then treated with normal serum specific

to secondary antibody at room temperature for 30 minutes. Sections were incubated

overnight at 4°C with a rabbit polyclonal anti eNOS antibody (1: 100 dilution, Abcam plc.

Cambridge, UK) in blocking serum. Sections were then incubated with a rhodamine

conjugated anti-rabbit IgG for 1 h. This was followed by counterstaining with DAPI and

mounting using Vectashield (Vector Labs). Images of LV sections of the respective treatment

groups at six random fields in triplicate per treatment groups were taken using an inverted

microscope IX-51 (Olympus).

3.4.9 Detection of Stem cells in LV Tissue

The frozen heart sections of the different transplanted groups (Group-5,6&7) were

analyzed for homing of transplanted stem cells. The frozen sections were stained with DAPI

and visualized for homing of GFP⁺ stem cells in the left ventricular tissue using inverted

microscope IX-51 (Olympus). Six random fields in set of three per treatment groups were

selected and an average count of GFP⁺ stem cells made.

3.4.9.1 Sirius Red Staining

Paraffin embedded serial sections of the two control non-transplanted (Group-1 & 2)

and the three transplanted (Group-5, 6&7) groups in the transplantation experiments were

analyzed for fibrosis by Sirius red staining. Before rehydration step the slides were baked at

60°C for 1 h followed by the xylene clearing step. The slides were then passed through

graded ethanol (100%, 95%, 85%, 75%, 60%, and 50%) into distilled water. A saturated

Page 59: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

picric acid solution with 0.1% Sirius Red (Aldrich Chemicals) was used to stain the slides for

overnight. Slides were then washed for two minutes in 0.01 N HCl and quickly dehydrated

in graded ethanol. After passing through xylene slides were coverslipped in mounting

medium (Grimm et al., 2003). The slides were visualized for collagen accumulation and

images taken using inverted microscope IX-51 (Olympus) equipped with a digital camera

DP-71 (Olympus). Images from six random fields in set of three per treatment groups were

then analyzed for percentage of fibrosis in left ventricular tissue by image J software.

3.4.9.2 Millar Analysis

Hemodynamic parameters of the mice in the different treatment groups (Group 1―6)

of transplantation experiment were taken using a Millar microtip pressure transducer. Briefly,

mice were intubated after anesthetizing with Pentobarbital sodium (55 mg g−1) and

ventilated with room air by using MiniVent (type 845, Harvard Apparatus). The breathing

rate of mice was maintained at 95 breaths per minute and body temperature kept at 37°C

during the procedure. The catheter was advanced into the left ventricle through the right

carotid artery. Pressure volume loops were observed on the chart recorder and the data

recorded in the closed chest preparation (Limana et al., 2002; Leri et al., 2003; Anversa et

al., 2002; Pacher et al., 2008). The parameters assessed included maximum pressure, end

systolic pressure, stroke volume and ejection fraction.

3.5 Statistical Analysis

Analysis of data for different experiments such as cell proliferation, densitometry,

SOD activity, apoptosis, chemotactic ability, real time PCR, homing of GFP positive MSCs

and tissue fibrosis, was performed using Student’s unpaired t-test (p value of <0.05 was

considered statistically significant) and all the data are expressed as mean ± standard error of

the mean except for homing studies where expressed as +SD.

Page 60: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CHAPTER 4

RESULTS

Page 61: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

4.1 In vitro Studies 4.1.1 Characterization of MSCs

The study included mice at diabetic day 60 with blood glucose levels above

300mg/dL (Fig. 4.1).

Figure 4.1. Mouse model of diabetes. Streptozotocin induced changes in blood glucose levels

of C57BL/6 wild-type mice till 60 days. (STZ: streptozotocin).

MSCs from mice at diabetic day 60 were isolated, cultured and analyzed for stem cell

markers by flow cytometry. The analysis revealed that MSCs were positive for CD44

(97.7%), CD90 (95.4%), and CD105 (92.3%) and negative for CD34 (1.4%) and CD45

(0.8%) (Fig. 4.2 ―E).

Page 62: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

FLOW CYTOMETRY

Figure 4.2. Flow cytometry of diabetic MSCs: (A–C) diabetic MSCs positive for CD44,

CD90, and CD105 and (D-E) negative for CD45 and CD34 (hematopoietic

markers).

4.1.2 Stimulation of Cell Proliferation in Preconditioned Diabetic MSCs

IGF-1 & FGF-2 were used alone and in combination in different concentrations to

precondition diabetic MSCs. The best cytokine regimen was selected on the basis of cell

proliferation measured after 48 h of preconditioning. The preconditioning of diabetic MSCs

with IGF-1/FGF-2 in combination at 50ng/mL each showed the highest cell proliferation

(1.61±0.11) as against of untreated cells (0.92±0.08). Cell proliferation figures of 1.10±0.11,

1.35±0.12, 1.37±0.05, 1.44±0.07 and 1.38±0.09 were observed for preconditioning with 50

ng/mL IGF-1, 50 ng/mL FGF-2, 100 ng/mL IGF-1, 100 ng/mL FGF-2, and combination of 1

ng/mL FGF-2, and 50 ng/mL IGF-1 respectively (Fig. 4.3A). The growth factors regimen

Page 63: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

showing highest cell proliferation was thus represented by 50ng/mL each of IGF-1 & FGF-2

in combination and used as the preconditioning strategy for subsequent experiments.

Figure 4.3A. Cell proliferation after preconditioning of diabetic MSCs. All values were

expressed as mean±standard error of the mean; *P<0.05 versus untreated

diabetic cells.

Cell proliferation of diabetic MSCs based on PCNA expression was also determined

after preconditioning with VEGF (50ng/mL) and compared with IGF-1/FGF-2 (50ng/mL

each) preconditioning. The PCNA expression in the two preconditioned groups as quantified

by qRT-PCR showed a significantly higher PCNA expression in the IGF-1/FGF-2 (50ng/mL

each) group compared to that of VEGF (50ng/mL) (Fig. 4.3B).

Page 64: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.3B. Analysis of cell proliferation for (PCNA) expression comparing VEGF and

IGF-1/FGF-2 preconditioning of diabetic MSCs.

4.1.3 Gene Expression Profiling of MSCs

The two sets of diabetic MSCs, one preconditioned with VEGF and the other with

IGF-1/FGF-2 were compared with the normal and diabetic untreated MSCs for the gene

expression profiling. The analysis revealed a down regulation of prosurvival markers (IGF-1,

FGF-2, and Akt) and an upregulation of senescent (p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ, and p53), and apoptotic

markers (Bax and Bak) in the diabetic untreated MSCs compared with MSCs from normal

animals (Fig. 4.4A&B). Improvement in gene expression profiling of diabetic MSCs was

observed after preconditioning with both VEGF (50ng/mL) and IGF-1/FGF-2 (50ng/mL

each). Low levels of prosurvival (IGF-1, FGF-2, and Akt) and differentiation markers

(GATA-4 and Nkx 2.5) observed in the diabetic untreated MSCs significantly increased after

preconditioning. Whereas high levels of apoptotic markers (Bax and Bak) in the diabetic

untreated MSCs significantly reduced after preconditioning. A similar reduction in the

Page 65: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

expression levels of senescent markers (p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ and p53) was also observed in the

preconditioned diabetic MSCs compared with the diabetic untreated MSCs. Gene expression

results after VEGF and IGF-1/FGF-2 based preconditioning of diabetic MSCs differed

significantly and IGF-1/FGF-2 preconditioning appeared better than VEGF. (Fig. 4.4C&D).

Densitometry of the gel was performed for quantification of bands and analysis made

accordingly (Fig. 4.4E).

Figure 4.4A-D. Gene expression profiling of MSCs. (A) Normal MSCs; (B) diabetic

untreated MSCs; (C) diabetic MSCs preconditioned with VEGF; (D)

diabetic MSCs preconditioned with IGF-1/FGF-2.

Page 66: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.4E. Quantification of gene expression profiling of MSCs. The difference between

gene expression in IGF-1/FGF-2-preconditioned versus VEGF-preconditioned

MSCs was significant; *P<0.05. Quantification of gels was done as per

recommended guidelines in the user manual using Quantity One-1-D Analysis

Software, version 4.4 (Bio-Rad Laboratories, Inc.). Ladder band was used as a

standard. The results of PCNA expression and gene expression profiling data clearly indicated

IGF-1/FGF-2 combination to be better than VEGF treatment. So this treatment regimen was

selected for preconditioning of diabetic MSCs in all subsequent experiments. The

preconditioning of MSCs, therefore, would refer to treatment of MSCs with 50 ng/mL each

in combination.

4.1.4 Increased SOD Activity in Preconditioned MSCs

SOD activity increased significantly in the preconditioned MSCs (52.3% ±2.0%,

P<0.05) after 24 h of hypoxia treatment compared with the untreated diabetic MSCs

(36.9%±3.1%). Although SOD activity showed a decline after 48 h of hypoxia treatment in

Page 67: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

both sets of MSCs, nevertheless the SOD activity was significantly higher in the

preconditioned MSCs (32.2% ±2.5%) compared with the untreated diabetic MSCs (23.1%

±2.2%). SOD activity measured under normoxic conditions in the MSCs from normal and

diabetic animals was used as a standard (Fig. 4.5).

Figure 4.5. Comparison of SOD activity among normal, diabetic untreated and diabetic

preconditioned MSCs under normoxia and hypoxia. Under normoxia, a high

SOD activity was observed in MSCs from normal animals compared with

MSCs from diabetic. Under hypoxia, a significant increase in SOD activity was

seen in the preconditioned diabetic MSCs compared with the untreated diabetic

MSCs at 24 h followed by a decline in both sets of MSCs at 48 h. * and **

represents comparison between diabetic preconditioned and diabetic untreated

MSCs at 24 and 48 h, (P<0.05 considered significant). 4.1.5 Effect of IGF-1 and FGF-2 Preconditioning on Apoptosis

A significant reduction in number of apoptotic cells was observed in the

preconditioned diabetic MSCs (51.8% ± 2.3%) compared with the untreated diabetic MSCs

(76.2% ± 1.8%) after hypoxia treatment as determined by flow cytometry for annexin-V (Fig.

4.6). The figure under normoxic conditions in both sets of diabetic MSCs was 17.2% ± 3.3%

and 31.5% ± 2.4% respectively.

Page 68: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.6. Flow cytometry for annexin-V-positive cells. Preconditioned MSCs showed a

reduction in annexin-V positive cells compared with untreated cells. All values

were expressed as mean±SEM and (*P<0.05).

4.1.6 Chemotactic Ability of Preconditioned Diabetic MSCs

The mobilization of diabetic MSCs (both untreated and preconditioned) towards

chemotactic signals from SDF-1α under hypoxic stress was analyzed. The analysis revealed

that preconditioned diabetic MSCs showed a significantly high mobilization number

(73.9±5.3, P<0.05) towards SDF-1α compared with the untreated diabetic MSCs (31.9±5.9)

(Fig. 4.7). Wells with serum free medium alone without SDF1-α supplementation and

containing both sets of MSCs were considered as control.

Page 69: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.7. Mobilization of untreated and preconditioned MSCs to SDF-1α.

A significantly high mobilization of preconditioned MSCs to SDF-1α seen as

compared to untreated MSCs. All values were expressed as mean±SEM. *P<0.05.

4.1.7 In vitro Tube-forming Ability in Diabetic MSCs

In vitro tube forming ability of both sets of diabetic MSCs (untreated and

preconditioned) was studied under both normoxia and hypoxia. It was evident from the

results that the preconditioned diabetic MSCs exhibited better tube forming ability under

both hypoxia and normoxia compared with the untreated diabetic MSCs (Fig. 4.8). In

addition, the ability of preconditioned diabetic MSCs to form tubular structure was much

better under hypoxia compared to normoxia (Fig. 4.8A&C).

Page 70: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.8A-D. Preconditioned MSCs (A&C) demonstrate better tube-forming ability than

untreated cells (B&D) under conditions of both normoxia and hypoxia

(100×; Scale bar: ˜20 µm).

The quantification of tube formation in both sets of MSCs further confirmed the

enhanced angiogenic ability of the preconditioned diabetic MSCs compared with the

untreated diabetic MSCs (Fig. 4.8E).

Page 71: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.8E. Quantification of tube forming cells under hypoxic and normoxic conditions.

MSC, mesenchymal stem cell. 4.1.8 Real-time RT-PCR Gene Expression after Glucose Stress

A real-time RT-PCR analysis of two angiogenic markers (Ang-1 and VEGF) and a

senescent marker (p16ᴵᴺᴷ⁴ᵃ) in two sets of diabetic MSCs (untreated and preconditioned) was

performed under conditions of high glucose (HG) stress. The preconditioned diabetic MSCs

showed an increase in gene expression of Ang-1 and VEGF and a decline in the expression of

p16ᴵᴺᴷ⁴ᵃ gene compared with the untreated diabetic MSCs (Fig. 4.9).

Page 72: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.9. Real-time RT-PCR gene expression analysis under conditions of HG stress.

4.2 In vivo Studies

4.2.1 Effect of EPO Treatment on Gene Expression of Diabetic Heart

In EPO treatment experiments without transplantation of MSCs, the quantitative RT-

PCR results showed a significant decrease in the left ventricular mRNA expressions of Bax

and p16ᴵᴺᴷ⁴ᵃ in the diabetic EPO treated group (Group-3) as compared to the diabetic

untreated group (Group-2). Bax and p16ᴵᴺᴷ⁴ᵃ mRNA expressions of the control normal group

(Group-1) were less than both the previous groups (Fig.4.10A).

RT-PCR based Left ventricular mRNA expression of IGF-1 was better in the EPO

treated group as compared to the untreated diabetic group. The control normal group showed

high expression than both of these groups (Fig. 4.10B).

Page 73: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.10. Left ventricular mRNA expression of Bax, p16ᴵᴺᴷ⁴ᵃ and IGF-1 in different

treatment groups of C57BL/6 male mice. A. Real time RT-PCR and B. RT-

PCR. (*p<0.05 vs. diabetic untreated).

4.2.2 Decreased Myocardial cell Apoptosis

Concomitant to the real time RT-PCR results for expression of bax, tunel assay for

apoptosis performed on LV cryosections revealed a significant decrease in myocardial cell

apoptosis in the EPO treated group (4±0.3) compared with the untreated group (8±0.5).

Apoptosis was significantly less in the normal heart sections (1±0.3) compared with both the

untreated and EPO treated groups (Fig.4.11).

Page 74: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.11. A.TUNEL assay for apoptosis in left ventricular heart sections (40 X) of

different treatment groups of C57BL/6 male mice. B. Number of apoptotic cells

per random field in respective heart sections. (*p<0.05).

4.2.3 Enhancement of eNOS Expression

Expression of eNOS, a constitutive form of nitric oxide synthase expressed in the

endothelial cells, was found to be decreased in the diabetic group compared with the control

normal group. The expression of eNOS in the diabetic myocardium was found to be

increased after EPO treatment (Fig. 4.12).

Page 75: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.12. Expression of eNOS in the left ventricular heart sections (20 X) of different

treatment groups of C57BL/6 male mice.

The treatment of diabetic mice with EPO alone indicated an improvement in the

microenvironment of the diabetic heart. This was shown by a decrease in myocardial cell

apoptosis and an increase in expression of prosurvival marker (IGF-1) and eNOS. In the light

of the beneficial effects of preconditioning on diabetic MSCs mentioned earlier,

transplantation studies were performed which employed a combined strategy of transplanting

preconditioned diabetic MSCs in to the left ventricle of diabetic heart followed by EPO

treatment in vivo.

Page 76: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

4.3 Transplantation Studies

4.3.1 Homing of MSCs

GFP positive (GFP⁺) MSCs (Both untreated and preconditioned) transplanted in left

ventricle of various transplantation groups of diabetic mice were studied for homing. A

significant number of GFP⁺ cells were observed in the mice transplanted with preconditioned

MSCs (Group-6) compared with that of diabetic untreated MSCs (Group-5) (Fig. 4.13 A, B).

Additional improvement in homing was observed in the diabetic mice transplanted with the

preconditioned MSCs and given EPO treatment (Group-7) compared with the previous two

groups (Fig. 4.13C). The diabetic myocardium transplanted with untreated diabetic MSCs

contained 12±1.5 GFP positive cells, followed by 25±2.0 in the preconditioned MSCs and

45±3.5 in the diabetic preconditioned MSCs and EPO-treatment group (Fig. 4.13 D).

Page 77: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.13. A-C. Homing of GFP-positive MSCs in Left ventricular heart sections (40 X) of

different treatment groups of C57BL/6 male mice. D. Number of GFP positive

cells, all values were expressed as mean standard deviation of the mean;

*P<0.05 versus untreated diabetic group.

Page 78: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

4.3.2 Reduction in Tissue Fibrosis

Paraffin embedded sections (LV) of various transplanted groups stained with Sirius

red staining showed reduction in fibrosis percentage (Fig.4.14 A-E). Maximum fibrosis

(14.75%±1.75%) was noted in the diabetic heart without transplantation. This was followed

by the group transplanted with untreated MSCs (13.16±0.5%), diabetic group transplanted

with the preconditioned MSCs (10.59%±1.25%) and the diabetic group transplanted with

preconditioned MSCs and given EPO treatment (6.67%±0.25%).

Page 79: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Figure 4.14. A-E. Fibrosis as seen in left ventricle heart sections(40 X) of different treatment

groups of C57BL/6 male mice by sirius red staining. F. Percentage of collagen

accumulation in LV sections (A-E) of different treatment groups (* p<0.05).

Page 80: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

4.3.3 Hemodynamic Parameters

Hemodynamic parameters of diabetic mice of various treatment groups in the

transplantation experiments were analyzed by Millar analysis. Millar analysis of normal

(group-1) and diabetic mice (group-2) without transplantation of MSCs were performed and

considered as standard. The analysis between the two groups (group-1 & group-2) for

parameters such as maximum pressure, end systolic pressure, stroke volume, and ejection

fraction showed an obvious impairments in the diabetic group compared with the normal

mice group. The heart functions of diabetic mice however increased significantly after EPO

treatment alone (Group-4) (Table 2).

The Millar analysis of the transplanted groups for the aforesaid cardiac parameters

revealed that the group-7 (Diabetic mice transplanted with the preconditioned MSCs and

given EPO treatment) showed the highest improvement in the heart functioning of diabetic

mice followed by group-6 (diabetic mice transplanted with the preconditioned MSCs)

compared with the group of diabetic mice transplanted with untreated MSCs (Group-5)

(Table 2).

Page 81: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Table 2. Cardiac parameters as determined by Millar analysis in different treatment groups of

C57BL/6 male mice. All values were expressed as mean±standard error of the mean;

NS; Normal saline group. UT; Untreated. Pre; Preconditioned.

Page 82: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CHAPTER 5

DISCUSSIONS

Page 83: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Among various types of stem cells used to repair injured myocardium, MSCs derived

from BM represent the most common type used for this purpose (Ohnishi et al., 2007; Song

et al., 2009). The potential of MSCs to repair damaged heart has been highlighted in some

recent studies and constitute an important area of research for the treatment of diabetic HF

(Zhang et al., 2008; Abdel Aziz et al., 2008; Dong et al., 2008; Paul et al., 2009; Li et al.,

2010; Jin et al., 2011). However, hyperglycemia can affect their reparability by forcing stem

cells into early aging (Rota et al., 2006) and could severely undermine the outcome of stem

cell based therapies for HF. So the present study intended to delineate the effects of diabetes

on MSCs derived from bone marrow and augment the function of diabetes-impaired MSCs

using a preconditioning strategy.

The RT-PCR results indicated a downregulation of survival markers (IGF-1, FGF-2,

Akt) and a high expression of senescent (p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ, p53) and apoptotic (Bax, Bak)

genes in the diabetic MSCs compared with the MSCs form normal animals. These findings

highlight diabetes-induced impairment of MSCs and are in agreement with the previous

reports which also describe that hyperglycemia can cause stem cell dysfunction (Awad et al.,

2005; Ohnishi et al., 2007). Rota et al. (2006) for instance, reported that diabetes increases

the expression of p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ, and p53 and can promote apoptosis in stem cells. A

negative correlation between senescent and prosurvival markers as documented previously

(Pons et al., 2008) also helps explain our present findings in the diabetes affected MSCs.

A preconditioning strategy employing growth factors was adopted to enhance the

functioning of diabetic MSCs. The IGF-1/FGF-2 growth factors in combination (50ng/mL

each) represented the best preconditioning regimen as determined by the cell proliferation

assay (Fig. 4.3A). This was also reflected in the results of gene expression profiling where

Page 84: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

preconditioning of diabetic MSCs with IGF-1/FGF-2 significantly upregulated the

prosurvival markers (IGF-1, FGF-2, Akt), and downregulated the senescent (p16ᴵᴺᴷ⁴ᵃ, p66ˢʱᶜ,

p53) and apoptotic markers (Bax, Bak). The preconditioning also resulted in a significant

increase in the expression of early cardiac differentiation markers (GATA-4, Nkx2.5) in the

preconditioned diabetic MSCs compared with the untreated diabetic MSCs (Fig. 4.4). The

selection of IGF-1/FGF-2 growth factors to precondition diabetic MSCs was a key factor for

this remarkable cellular response. Owing to the known role of IGF-1 in survival pathways

(Anversa, 2005) IGF-1 has been in extensive use to enhance stem cell function (Li et al.,

2007; Lu et al., 2009). The previous data on FGF-2 indicate its role in promoting expression

of early cardiac differentiation markers (Degeorge et al., 2008), cell proliferation (Choi et al.,

2008) and suppressing cellular senescence (Ito et al., 2007). Therefore, the use of this novel

combination of IGF-1/FGF-2 resulted in an increase in cell proliferation, the expression of

survival genes and more importantly caused a decline in expression of the senescent genes.

Owing to the multifunctional attributes ascribed to VEGF (Tong et al., 2006), a

parallel treatment option of preconditioning diabetic MSCs with VEGF (50ng/mL) was also

used. The rationale behind this strategy was to compare the two treatment options (IFG-

1/FGF-2 vs. VEGF preconditioning) and select the better one. The diabetic MSCs did show

improvement in cell senescent state, cell survival signaling and proliferation after VEGF

preconditioning but these effects were less than the overall effects of IFG-1/FGF-2 based

preconditioning (Fig. 4.4). IGF-1/FGF-2 preconditioning was hence identified as a better

treatment option to enhance the functionality of MSCs under hyperglycemia and used for

further analysis in subsequent sets of experiments.

Page 85: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Hypoxic and high glucose insults given to the preconditioned MSCs formed the basis

of these subsets of experiments in a way to imitate microenvironment of the diabetic heart.

Preconditioned diabetic MSCs showed a significant improvement in SOD activity under

conditions of hypoxic stress compared with the untreated diabetic MSCs exhibiting low SOD

activity (Fig. 4.5). In a recent study Stolzing et al. (2008) reported that SOD activity depends

on ROS levels and is affected by the donor age. ROS levels tend to become high in diabetes

(Ohshima et al., 2009) and this may have caused the low SOD activity observed in the

diabetic MSCs. ROS generation in hyperglycemia is also well documented to cause stem

cells susceptibility to apoptosis, and necrosis (Fiers et al., 1999) and impairs the angiogenic

ability of cells (Ebrahimian et al., 2006). In concordant with these findings diabetic MSCs

showed a high number of apoptotic cells and reduced tube-forming ability. After

preconditioning of diabetic MSCs a significant decline in number of apoptotic cells (Fig. 4.6)

concomitant with enhanced in vitro tube-forming ability was observed (Fig. 4.8).

The mobilization of diabetic MSCs to chemotactic signals in response to SDF-1α also

significantly improved after preconditioning (Fig. 4.7). De Falco et al. (2009) described

hyperglycemia-induced impairment of the SDF-1α–CXCR-4 pathway to affect recruitment of

EPC in patients with coronary artery disease. The importance of SDF-1α–CXCR-4 axis for

mobilization and recruitment of stem cells has been previously reported by Wang et al.

(2006) and it has been shown that release of SDF-1α in conditions such as ischemia causes

upregulation of CXCR-4 receptor in different types of stem cells to ensue mobilization (Zhou

et al., 2002; Kucia et al., 2004). So a low chemotactic response by the untreated diabetic

MSCs reported here is in agreement with the previous findings which improved significantly

after preconditioning with IGF-1/FGF-2 in combination.

Page 86: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

The tolerance of diabetic MSCs to high glucose stress was evaluated by emulating a

clinical hyperglycemic condition in vitro. For this purpose diabetic MSCs (both untreated

and preconditioned) were exposed to high glucose (30 mmol/L) treatment to roughly mimic

blood glucose levels in patients with inadequately controlled diabetes or as observed under

conditions of stress like inflammation, infection or hospitalization (Chen et al., 2007). The

real time-PCR results showed a downregulation in the expression of p16ᴵᴺᴷ⁴ᵃ coupled with

an upregulation of Ang-I and VEGF in preconditioned diabetic MSCs compared with the

untreated diabetic MSCs (Fig. 4.9). This exciting finding indicates that preconditioning of

diabetic MSCs not only improves cell survival and its angiogenic ability but also helps

withstand cellular senescence under conditions of high glucose stress.

Thus IGF-1/FGF-2 combination of growth factors has the potential to improve the

functionality of diabetes-impaired MSCs by enhancing cell proliferation, downregulating

senescent and apoptotic genes and upregulating prosurvival genes. This novel combination

also offers protection from hypoxic and high glucose insults and will be of great use in the

study of diabetes affected MSCs in future.

In view of the documented role of EPO in enhancing cellular response in different

disease modalities such as diabetes, neurodegeneration, cardiovascular system and renal

systems (Maiese et al., 2008), a subset of experiments were also carriedout in this respect.

The study intended to evaluate the effects of EPO administration on improvement of diabetic

myocardium and its functional status after transplantation of MSCs.

The gene expression studies showed a down regulation of p16ᴵᴺᴷ⁴ᵃ and Bax genes

and an upregulation of IGF-1 in the left ventricular tissue of the diabetic heart following EPO

administration compared with the untreated group (Fig. 4.10). The indication of an increase

Page 87: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

in apoptosis in the diabetic heart tissue was further evident in the results of tunel staining

(Fig. 4.11A). The EPO treated group on the other hand showed less number of apoptotic cells

(Fig. 4.11B). These findings are in agreement with the previous studies which support that

EPO administration reduces complications in diabetes (Silverberg et al., 2006) and decreases

cardiomyocyte apoptosis in ischemic cardiac diseases (Tramontano et al., 2003; Gao et al.,

2007; Ueda et al., 2010). This relevance is further supported by the findings that one of the

most common reason of cardiovascular complications is diabetes and the incidence of and

risk of mortality from cardiovascular disease is two to eight fold higher in the diabetic

patients (Gaede, et al., 2003; Francia et al., 2009).

Although apoptosis prevention by EPO is considered to be a key mechanism in acute

cardioprotection (Paschos et al., 2008) the role of nitric oxide (NO) is also important

(Vanhoutte, 1988; De Caterina et al., 1995; Tharaux et al., 1999). The endothelial isoform of

NO is the prevailing form in the cardiovascular system and depends on eNOS expression.

eNOS has been documented to protect from hypertension and myocardial infarction in

transgenic mice model overexpressing recombinant human EPO through up regulation of

eNOS expression (Paschos et al., 2008). EPO has also been reported to enhance production

of coronary NO and provide protection from reperfusion-induced myocardial injury (Mihov

et al., 2009). Better eNOS expression was observed in the left ventricular tissue of the

diabetic mice following EPO administration compared with the untreated group (Fig. 4.12).

The site of eNOS upregulation in the left ventricular tissue most likely seems to be the

coronary endothelium, since no difference in eNOS expression of cardiomyocytes was

reported in a recent study employing EPO administration in rat model of ischemia /

reperfusion injury (Mihov et al., 2009). EPO together with its benefit of cardioprotection

Page 88: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

may be an indicator of enhanced angiogenesis, since studies have shown that EPO

independently leads to angiogenesis (Reinders et al., 2006; Li et al., 2007) and promotes

mobilization of EPC that is associated with neovascularization of ischemic tissue (Heeschen

et al., 2003).

In continuation of these findings, transplantation of MSCs in the left ventricular

tissue of diabetic mice was done followed by EPO administration to evaluate improvement of

diabetic myocardium. The study involved assessment for homing of MSCs and reduction in

fibrosis in various transplantation groups which was followed by Millar analysis of cardiac

parameters.

The results indicated a significant increase in homing of the preconditioned MSCs

and reduction in left ventricular tissue fibrosis after EPO treatment compared with the

preconditioned MSCs group (Fig. 4.13 & 4.14) without subsequent EPO treatment. The

preconditioned MSCs group without EPO treatment in turn showed a significant homing and

reduction in fibrosis compared with the untreated group (Fig. 4.13 & 4.14). Evidence of

increase in cardiac fibrosis due to diabetic cardiomyopathy has previously been documented

(Tschope et al., 2004; Westermann, et al., 2007; Zhang, et al., 2008). The reduction in

fibrosis reported in the present study is in concordant with the previous findings which also

implicate EPO in reducing interstitial fibrosis and cardiac remodeling in cases of ischemic

heart diseases and MI (Gao et al., 2007; Ueda et al., 2010).

The functionality of the diabetic heart based on maximum pressure, end systolic

pressure, stroke volume, and ejection fraction was assessed in different transplanted and non

transplanted groups (Table 2). Heart functionality of diabetic mice (Group-2&3) was

considerably impaired as compared to that of control normal mice (Group-1). However, the

Page 89: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

heart functionality of the diabetic mice improved significantly following administration of

EPO alone (Group-4). A more significant increase in cardiac functions was observed in the

diabetic mice transplanted with the preconditioned MSCs (Group-6) compared with the

diabetic mice transplanted with untreated MSCs (Group-5) and the group-4 given EPO

treatment alone. The diabetic mice transplanted with preconditioned MSCs and given EPO

treatment (Group-7) showed maximum improvement in the cardiac parameters assessed

compared with all other diabetic groups. In particular, values of the end-systolic pressure

(ESP) (77.1+6.4mmHg) and ejection fraction (EF) (26.9+0.62%) observed in the

preconditioned EPO-treated group were closest to the normal standard base line values of

ESP (90-110 mmHg) and EF (44-62%) as reported by Pacher et al. ( 2008) compared with all

other diabetic groups. To date no known study has been carried out that characterizes the

effects of EPO on heart of type-1 diabetic mice and its outcome after transplantation of

MSCs preconditioned with IGF-1/FGF-2. Although a limited study done on a human atrial

model of hypoxia/reoxygenation in diabetic patients do indicates reduction in myocardial

apoptosis and improvement in contractility of the cardiac tissues (Mudalagiri et al., 2008).

Other studies, which document improvement of heart function after EPO administration,

mostly involved model of ischemia-reperfusion injury or MI in different animals and human

subjects (Calvillo et al., 2003; Parsa et al., 2003; Moon et al., 2003; Van der Meer et al.,

2005; Hirata et al., 2006; Westenbrink et al., 2007; Lipsic et al., 2008). So the findings about

the functionality of the diabetic heart shown in the present study should be viewed as a

pioneer study of its kind and indicates potential for the future studies.

In the present study EPO (1000 I.U/kg) was administered for five consecutive days

after MSCs transplantation. Future studies should be designed to evaluate the functionality of

Page 90: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

diabetic heart by employing different doses and increasing duration of EPO administration.

Moreover, different local and systemic factors such as VEGF, endothelin 1 and transformimg

growth factors known to be modulated by EPO should also be investigated to determine the

role of EPO in the acute and late cardioprotection (Arcasoy, 2008; Piuhola et al., 2008). The

study should also address some important safety issues such as rise in levels of serum EPO

(den Elzen et al., 2010), hemoglobin, blood pressure and risk of thrombosis (Besarab et al.,

1998).

Although the present EPO based transplantation study indicates improvement of

diabetic heart at several fronts such as a decrease in myocardial apoptosis, senescence, left

ventricular tissue fibrosis, and an increase in homing of preconditioned MSCs, expression of

eNOS and heart functionality, the inclusion of above mentioned considerations for future

studies would further illuminate the potential of this novel combination to treat diabetic HF.

Page 91: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

CHAPTER 6

REFERENCES

Page 92: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Abdel Aziz, M.T., El-Asmar, M.F., Haidara, M., Atta, H.M., Roshdy, N.K., Rashed, L.A.,

Sabry, D., Youssef, M.A., Abdel Aziz, A.T., Moustafa, M. (2008). Effect of bone

marrow-derived mesenchymal stem cells on cardiovascular complications in diabetic

rats. Med Sci Monit 14:249–255.

Alison, M.R., Poulsom, R., Forbes, S. et al. (2002). An introduction to stem cells. J Pathol

197:419–23.

Amos, A.F., McCarty, D.J., Zimmet, P. (1997). The rising global burden of diabetes and its

complications: estimates and projections to the year 2010. Diabet Med 14 Suppl

5:S1–S85.

An, D., Rodrigues, B. (2006). Role of changes in cardiac metabolism in development of

diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 291(4):H1489–506.

An, D., Rodrigues, B. (2006). Role of changes in cardiac metabolism in development of

diabetic cardiomyopathy. Am J Physiol Heart Circ Physiol 291(4):H1489–506.

Anderson, D.J., Gage, F.H., Weissman, I.L. (2001). Can stem cells cross lineage boundaries?

Nat Med 7:393–95.

Anversa, P. (2005). Aging and longevity: the IGF-1 enigma. Circ Res 97:411–414.

Anversa, P., Leri, A., Rota, M., Hosoda, T., Bearzi, C., Urbanek, K., Urbanek, J., Bolli, R.

(2007).Concise review: stem cells, myocardial regeneration, and methodological

artifacts. Stem Cells 25:589–601.

Anversa, P., Olivetti, G. (2002). Cellular basis of physiological and pathological myocardial

growth. In: Page E, Fozzard, H.A., Solaro, R.J., eds. Handbook of Physiology. Section

2. The Cardiovascular System. Volume I. The Heart 1:75–144.

Page 93: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Arcasoy, M.O. (2008).The non-haematopoietic biological effects of erythropoietin. Br J

Haematol 141:14–31.

Asahar, T., Kawamoto, A. (2004). Endothelial progenitor cells for postnatal vasculogenesis.

Am J Physiol cell Physiol 1287:C572–C579.

Asaumi, Y., Kagaya, Y., Takeda, M., et al. (2007). Protective Role of Endogenous

Erythropoietin System in Nonhematopoietic Cells Against Pressure Overload–

Induced Left Ventricular Dysfunction in Mice. Circulation 115:2022–2032.

Awad, O., Jiao, C., Ma, N., Dunnwald, M., Schatterman, G.C. (2005). Obese diabetic mouse

environment differentially affects primitive and monocytic endothelial cell

progenitors. Stem cells 23:575–583.

Aydemir-Koksoy, A., Bilginoglu, A., Sariahmetoglu, M., Schulz, R., Turan, B. (2010).

Antioxidant treatment protects diabetic rats from cardiac dysfunction by preserving

contractile protein targets of oxidative stress. J Nutr Biochem 21(9):827–33.

Bearzi, C., Rota, M., Hosoda, T., Tillmanns, J., Nascimbene, A., De Angelis, A., Yasuzawa-

Amano, S., Trofimova, I., Siggins, R.W., Lecapitaine, N., Cascapera, S., Beltrami,

A.P., D'Alessandro, D.A., Zias, E., Quaini, F., Urbanek, K., Michler, R.E., Bolli, R.,

Kajstura, J., Leri, A., Anversa, P. (2007). Human cardiac stem cells. Proc Natl Acad

Sci USA 104:14068–14073.

Besarab, A., Bolton, W.K., Browne, J.K., et al. (1998). The effects of normal as compared

with low hematocrit values in patients with cardiac disease who are receiving

hemodialysis and epoetin. N Engl J Med 339:584–90.

Boudina, S., Abel, E.D. (2010). Diabetic cardiomyopathy, causes and effects. Rev Endocr

Metab Disord 11(1):31–9.

Page 94: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Bullard, A.J., Govewalla, P., Yellon, D.M. (2005). Erythropoietin protects the myocardium

against reperfusion injury in vitro and in vivo. Basic Res Cardiol 100: 397–403.

Burch, P., Heintz, N. (2005). Redox regulation of cell cycle re-entry: cyclin D1 as a primary

target for the mitogenic effects of reactive oxygen and nitrogen species. Antioxid

Redox Signal 7:741–751.

Cai, Z., Manalo, D.J., Wei, G., Rodriguez, E.R., Fox-Talbot, K., Lu, H., et al. (2003). Hearts

from rodents exposed to intermittent hypoxia or erythropoietin are protected against

ischemia–reperfusion injury. Circulation 108:79–85.

Calvillo, L., Latini, R., Kajstura, J., Leri, A., Anversa, P., Ghezzi, P., Salio, M., Cerami, A.,

Brines, M. (2003). Recombinant human erythropoietin protects the myocardium from

ischemia-reperfusion injury and promotes beneficial remodeling. Proc Natl Acad Sci

USA. 100:4802– 4806.

Candido, R., Srivastava, P., Cooper, M.E., Burrell, L.M. (2003). Diabetes mellitus: a

cardiovascular disease. Curr Opin Investig Drugs 4(9):1088–1094.

Carnot P. DeFlandre C. (1906). Sur l’activite hemopoietique de serum au cours de la

regeneration du sang. C R Acad Sci (Paris) 143:384–6.

Chambers, I., Smith, A. (2004). Self-renewal of teratocarcinoma and embryonic stem cells.

Oncogene 23:7150–60.

Chen, Y.H., Lin, S.J., Lin, F.Y., Wu, T.C., Tsao, C.R., Huang, P.H., Liu, P.L., Chen, Y.L.,

Chen, J.W. (2007). High glucose impairs early and late endothelial progenitor cells by

modifying nitric oxide-related but not oxidative stress-mediated mechanisms.

Diabetes 56:1559–1568.

Page 95: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Choi, S.C., Kim, S.J., Choi, J.H., Park, C.Y., Shim, W.J., Lim, D.S. (2008). Fibroblast

growth factor-2 and -4 promote the proliferation of bone marrow mesenchymal stem

cells by the activation of the P13K-Akt and ERK1/2 signaling pathways. Stem Cells

Dev 17:725–736.

Chong, Z.Z., Shang, Y.C., Maiese, K. (2007). Vascular injury during elevated glucose can be

mitigated by erythropoietin and Wnt signaling. Curr Neurovasc Res 4:194–204.

De Caterina, R., Libby, P., Peng, H.B., et al. (1995). Nitric oxide decreases cytokine-

induced endothelial activation. Nitric oxide selectively reduces endothelial expression

of adhesion molecules and proinflammatory cytokines. J Clin Invest 96:60–8.

De Falco, E., D Avitabile, D., P Totta, P., S Straino, S., F Spallotta, F., C Cencioni, C., AR

Torella, A.R., Rizzi, R., Porcelli, D., Zacheo, A., Di Vito, L., Pompilio, G.,

Napolitano, M., Melillo, G., Capogrossi, M.C., Pesce, M. (2009). Altered SDF-1

mediated differentiation of bone marrow-derived endothelial progenitor cells in

diabetes mellitus. J Cell Mol Med 13: 3405–3414.

Deeping, R., Kawakami, K., Ocker, H., Wagner, J.M., Heringlade, M., Noetzold, A., et al.

(2005). Expression of the erythropoietin receptor in human heart. J Thorac

Cardiovasc Surg 130:877.

den Elzen, W.P., Willems, J.M., Westendorp, R.G., de Craen, A.J., Blauw, G.J., Ferrucci, L.,

Assendelft, W.J., Gussekloo, J. (2010). Effect of erythropoietin levels on mortality in

old age: the Leiden 85-plus Study. CMAJ 182(18):1953–8.

Degeorge, B.R.Jr., Rosenberg, M., Eckstein, V., Gao, E., Herzog, N., Katus, H.A., Koch,

W.J., Frey, N., Most, P. (2008). BMP-2 and FGF-2 synergistically facilitate adoption

Page 96: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

of a cardiac phenotype in somatic bone marrow c-kit+/Sca-1+ stem cells. Clin Transl

Sci 1(2):116-25.

Devereux, R.B., Roman, M.J., Paranicas, M. (2000). Impact of diabetes on cardiac structure

and function: The Strong Heart Study. Circulation 101: 2271–2276.

Di Filippo, C., Marfella, R., Cuzzocrea, S., Piegari, E., Petronella, P., Giugliano, D., Rossi,

F. D’ mico M. (2005). Hyperglycemia in streptozotocin induced diabetic rat

increases infarct size associated with low levels of myocardial HO-1 during

ischemia/reperfusion. Diabetes 54:803–810.

Di Paola, M., Lorusso, M. (2006). Interaction of free fatty acids with mitochondria:

Coupling, uncoupling and permeability transition. Biochim Biophys Acta 1757(9-

10):1330–1337.

Dobrin, J.S., Lebeche, D. (2010). Diabetic cardiomyopathy: signaling defects and therapeutic

approaches. Expert Rev Cardiovasc Ther 8(3):373–91.

Dong, Q.Y., Chen, L., Gao, G.Q., Wang, L., Song, J., Chen, B.,Xu, Y.X., Sun,L. (2008).

Allogeneic diabetic mesenchymal stem cells transplantation in streptozotocin-induced

diabetic rat. Clin Invest Med 31:328–337.

Dyntar, D., Eppenberger-Eberhardt, M., Maedler, K., Pruschy, M., Eppenberger, H.M.,

Spinas, G.A., Donath, M.Y. (2001). Glucose and palmitic acid induce degeneration of

myofibrils and modulate apoptosis in rat adult cardiomyocytes. Diabetes 50(9):2105–

2113.

Ebrahimian, T.G., C Heymes, C., D You, D., O Blanc-Brude, O., B Mees, B., L Waeckel, L.,

M Duriez, M., Vilar, J., Brandes, R.P., Levy, B.I., Shah, A,M., Silvestre, J.S. (2006).

NADPH oxidase-derived overproduction of reactive oxygen species impairs

Page 97: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

postischemic neovascularization in mice with type I diabetes. Am J Pathol 169:719–

728.

Esneault, E., Pacary, E., Eddi, D., Freret, T., Tixier, E., Toutain, J., Touzani, O., Schumann-

Bard, P., Petit, E., Roussel, S., Bernaudin, M. (2008). Combined therapeutic strategy

using erythropoietin and mesenchymal stem cells potentiates neurogenesis after

transient focal cerebral ischemia in rats. J Cereb Blood Flow Metab 28(9):1552–63.

Falcão-Pires, I., Gonçalves, N., Gavina, C. et. al. (2010). Correlation between plasma levels

of apelin and myocardial hypertrophy in rats and humans: possible target for

treatment? Expert Opin Ther Targets 14:231–241.

Fazel, S., Cimini, M., Chen, L., Li, S., Angoulvant, D., Fedak, P., Verma, S., Weisel, R.D.,

Keating, A., Li, R.K. (2006). Cardioprotective c-kit+ cells are from the bone marrow

and regulate the myocardial balance of angiogenic cytokines. J Clin Invest 116:1865–

1877.

Fiers, W., Beyaert, R., Declercq, W., Vandenabeele, P. (1999). More than one way to die;

apoptosis, necrosis and reactive oxygen damage. Oncogene 18:7719–7730.

Fisher, B.M., Gillen, G., Lindop, G.B., Dargie, H.J., Frier, B.M. (1986). Cardiac function and

coronary arteriography in asymptomatic type-1 (insulin-dependent) diabetic patients:

evidence for a specific diabetic heart disease. Diabetologia 29(10):706–712.

Fliser, D., Haller, H. (2007). Erythropoietin and treatment of non-anemic conditions –

cardiovascular protection. Semin Hematol 44:212–7.

Francia, P., Cosentino, F., Schiavoni, M., Huang, Y., Perna, E., Camici, G.G., Lüscher, T.F.,

Volpe, M. (2009). p66ˢʱᶜ protein, oxidative stress, and cardiovascular complications of

diabetes: the missing link. J Mol Med 87:885–891.

Page 98: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Frantz, S., Calvillo, L., Tillmanns, J., Elbing, I., Dienesch, C., Bischoff, H., Ertl, G.,

Bauersachs, J. (2005). Repetitive postprandial hyperglycemia increases cardiac

ischemia / reperfusion injury: prevention by the alpha-glucosidase inhibitor acarbose.

FASEB J 19:591–593.

Fuchs, E., Segre, J.A. (2000). Stem cells: a new lease on life. Cell 100:143–55.

Fuchs, E., Tumbar, T., Guasch, G. (2004). Socializing with the neighbors: stem cells and

their niche. Cell 116:769–78.

Gaede, P., Vedel, P., Larsen, N., Jensen, G.V., Parving, H.H., Pedersen, O. (2003).

Multifactorial intervention and cardiovascular disease in patients with type 2 diabetes.

N Engl J Med 348:383–393.

Gallagher, K.A., Liu, Z.J., Xiao, M., Chen, H., Goldstein, L.J., Buerk, D.G., Nedeau, A., SR

Thom, S.R., Velazquez, O.C. (2007). Diabetic impairments in NO-mediated

endothelial progenitor cell mobilization and homing are reversed by hyperoxia and

SDF-1a. J Clin Invest 117:1249–1259.

Gao, E., Boucher, M., Chuprun, J.K., Zhou, R.H., Eckhart, A.D., Koch, W.J. (2007).

Darbepoetin alfa, a long-acting erythropoietin analog, offers novel and delayed

cardioprotection for the ischemic heart. Am J Physiol Heart Circ Physiol 293:H60–8.

Gomperts, B.N., Strieter, R.M. (2007). Stem Cells and Chronic Lung Disease. Annu Rev Med

58:285-298.

Grimm, P.C., Nickerson, P., Gough, J., Mckenna, R., Stern, E., Jeffery, J., Rush, D.N.

(2003). Computerized Image Analysis of Sirius Red–Stained Renal Allograft

Biopsies as a Surrogate Marker to Predict Long- Term Allograft Function. J Am Soc

Nephrol 14:1662–1668.

Page 99: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Guang-wei, L., Ying-hua, H., Wen-ying, Y., et al. (2002). Effects of insulin resistance and

insulin secretion on the efficacy of interventions to retard development of type 2

diabetes complications: the Da Qing IGT and diabetes study. Diabetes Res Clin

Pract. 58:193–200.

Guo, J., Lin, G., Bao, C., Hu, Z., Chu, H., Hu, M. (2008). Insulinlike growth factor 1

improves the efficacy of mesenchymal stem cells transplantation in a rat model of

myocardial infarction. J Biomed Sci 15:89–97.

Haffner, S.M. (1997). The Scandinavian Simvastatin Survival Study (4S) subgroup analysis

of diabetic subjects: implications for the prevention of coronary heart disease.

Diabetes Care 20:469–471.

Hahn J,Y., Cho, H.J., Kang, H.J., Kim, T.S., Kim, M.H., Chung, j.H., Bae, J.W., Oh, B.H.,

YB Park, Y.B., Kim, H.S. (2008). Pre-treatment of mesenchymal stem cells with a

combination of growth factors enhances gap junction formation, cytoprotective effect

on cardiomyocytes, and therapeutic efficacy for myocardial infarction. J Am Coll

Cardiol 51:933–943.

Haider, H., Asjraf, M. (2005). Bone marrow cell transplantation in clinical perspective. J Mol

Cell Cardiol 38:225–235.

Haider, H.Kh., Ashraf, M. (2008). Strategies to promote donor cell survival: combining

preconditioning approach with stem cell transplantation. J Mol Cell Cardiol 45: 554–

566.

Hanefeld, M., Fischer, S., Julius, U., et al. (1996). Risk factors for myocardial infarction and

death in newly detected NIDDM: the Diabetes Intervention Study, 11-year follow-up.

Diabetologia 39:1577–1583.

Page 100: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Hayat, S.A., et al. (2004). Diabetic cardiomyopathy: mechanisms, diagnosis and treatment.

Clin Sci (Lond) 107(6):539–57.

Heeschen, C., Aicher, A., Lehmann, R., et al. (2003). Erythropoietin is a potent physiologic

stimulus for endothelial progenitor cell mobilization. Blood 102:1340–6

Herzog, E.L., Chai, L., Krause, D.S. (2003). Plasticity of marrow-derived stem cells. Blood

102:3483–93.

Hirata, A., Minamino, T., Asanuma, H., Fujita, M., Wakeno, M., Myoishi, M., Tsukamoto,

O., Okada, K., Koyama, H., Komamura, K., Takashima, S., Shinozaki, Y., Mori, H.,

Shiraga, M., Kitakaze, M., Hori, M. (2006). Erythropoietin enhances

neovascularization of ischemic myocardium and improves left ventricular dysfunction

after myocardial infarction in dogs. J Am Coll Cardiol 48:176–184.

Holman, R., Turner, R., Stratton, I., et. al. (1998). UK Prospective Diabetes Study Group.

Efficacy of atenolol and captopril in reducing risk of macrovascular and

microvascular complications in type 2 diabetes: UKPDS 39. BMJ 317:713–720.

Hu, G., Tuomilehto, J., Silventoinen, K., Barengo, N., Jousilahti, P. (2004). Joint effects of

physical activity, body mass index, waist circumference, and waist-to-hip ratio with

the risk of cardiovascular disease among middleaged Finnish men and women. Eur

Heart J 25:2212–2219.

Ito, T., Sawada, R., Fujiwara, Y., Seyama, Y., Tsuchiya, T. (2007). FGF-2 suppresses

cellular senescence of human mesenchymal stem cells by down-regulation of TGF-

β2. Biochem Biophy Res Commun 359:108–114.

Jiang, Y., Jahagirdar, B.N., Reinhardt, R.L., et al. (2002). Pluripotency of mesenchymal stem

cells derived from adult marrow. Nature 418:41–49.

Page 101: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Jin, J., Zhao, Y., Tan, X., Guo, C., Yang, Z., Miao, D. (2011). An Improved Transplantation

Strategy for Mouse Mesenchymal Stem Cells in an Acute Myocardial Infarction

Model. PLoS One 6(6): e21005.

Kannel, W.B., Hjortland, M., Castelli, W.P. (1974). Role of diabetes in congestive heart

failure: the Framingham Study. Am J Cardiol 34(1):29–34.

Kannel, W.B., McGee, D.L. (1979). Diabetes and cardiovascular disease: the Framingham

Study. JAMA 241:2035–2038.

Khan, M., Manzoor, S., Mohsin, S., Khan, S.N., Ahmad, F.J. (2009). IGF-1 and G-CSF

complement each other in BMSCs migration towards infarcted myocardium in a

novel in vitro model. Cell Biol Int 33:650–657.

Kinnaird, T., Stabile ,E., Burnett, M.S., Shou, M., Lee, C.W., Barr, S., Fuchs, S., and

Epstein, S.E. (2004). Local delivery of marrow-derived stromal cells augments

collateral perfusion through paracrine mechanisms. Circulation 109:1543–1549.

Kinnaird, T., Stabile, E., Burnett, M.S., and Epstein, S.E. (2004). Bonemarrow- derived cells

for enhancing collateral development: mechanisms, animal data, and initial clinical

experiences. Circ Res 95:354–363.

Kinnaird, T., Stabile, E., Burnett, M.S., Lee, C.W., Barr, S., Fuchs, S., and Epstein, S.E.

(2004). Marrow-derived stromal cells express genes encoding a broad spectrum of

arteriogenic cytokines and promote in vitro and in vivo arteriogenesis through

paracrine mechanisms. Circ Res 94:678–685.

Krause, D.S. (2002). Plasticity of marrow-derived stem cells. Gene Ther 9:754–758.

Krause, D.S., Theise, N.D., Collector, M.I., et al. (2001). Multi-organ, multi-lineage

engraftment by a single bone marrow-derived stem cell. Cell 105:369–77.

Page 102: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Kucia, M., Dawn, B., Hunt, G., Wysoczynski, M., Majka, M., Ratajczak, J., Rezzoug, F.,

Ildstad, S.T.,Bolli, R., and MZ Ratajczak, M. Z. (2004). Cells expressing early

cardiac markers reside in the bone marrow and are mobilized into the peripheral

blood after myocardial infarction. Circ Res 95:1191–1199.

Kucia, M., Jankowski, K., Reca, R., Wysoczynski, M., Bandura, L., Allendorf, D.J., Zhang,

J., Ratajczak, J., Ratajczak, M.Z. (2004). CXCR4-SDF-1 signalling, locomotion,

chemotaxis and adhesion. J Mol Histol 35:233–245.

Kudo, M., Wang, Y., Wani, M.A., Xu, M., Ayub, A., M Ashraf, M. (2003). Implantation of

bone marrow stem cells reduces the infarction and fibrosis in ischemic mouse heart. J

Mol Cell Cardiol 35:1113–1119.

Laflamme, M.A. Murry, C.E. (2011). Heart regeneration. Nature 473(7347):326–35.

Leiter, E.L. 1999. In McNeill JH ed. Experimental models of diabetes. CRC Press. Boca

Raton, Fla. 418pp.

Leri, A., Franco, S., Zacheo, A., Barlucchi, L., Chimenti, S., Limana, F., Nadal-Ginard, B.,

Kajstura, J., Anversa, P., Blasco, M.A. (2003). Ablation of telomerase and telomere

loss leads to cardiac dilation and heart failure associated with p53 upregulation.

EMBO J 22:131–139.

Li, C.J., Zhang, Q.M., Li, M.Z., Zhang, J.Y., Yu, P., Yu, D.M. (2009). Attenuation of

myocardial apoptosis by alpha-lipoic acid through suppression of mitochondrial

oxidative stress to reduce diabetic cardiomyopathy. Chin Med J (Engl).122:2580–

2586.

Li, J., Leschka, S., Rutschow, S., Schwimmbeck, P.L., Husmann, L., Noutsias, M.,

Westermann, D., Poller, W., Zeichhardt, H., Klingel, K., et al. (2007).

Page 103: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Immunomodulation by interleukin-4 suppresses matrix metalloproteinases and

improves cardiac function in murine myocarditis. European journal of pharmacology

554(1):60–68.

Li, Q., Turdi, S., Thomas, D., Zhou, T., Ren, J. (2010). Intra-myocardial Delivery of

Mesenchymal Stem Cells Ameliorates Left Ventricular and Cardiomyocyte

Contractile Dysfunction Following Myocardial Infarction. Toxicol Lett 195(2-3):

119–126.

Li, Y., Takemura, G., Okada, H., Miyata, S., Maruyama, R., Li, L., Higuchi,

M.,Minatoguchi, S., Fujiwara, T., Fujiwara, H. (2006). Reduction of inflammatory

cytokine expression and oxidative damage by erythropoietin in chronic heart failure.

Cardiovasc Res 71:684–694.

Li, Y., Yu, X., Lin, S., Li, X., Zhang, S., Song, Y.H. (2007). Insulin- like growth factor 1

enhances the migratory capacity of mesenchymal stem cells. Biochem Biophys Res

Commun 356:780–784.

Limana, F., Urbanek, K., Chimenti, S., Quaini, F., Leri, A., Kajstura, J., Nadal-Ginard, B.,

Izumo, S., Anversa, P. (2002). Bcl-2 overexpression promotes myocyte proliferation.

Proc Natl Acad Sci USA 99:6257–6262.

Linheng, Li., Ting, Xie. (2005). Stem Cell Niche: Structure and Function. Annu Rev Cell Dev

Biol 21:605–631.

Lipsic, E., Schoemaker, R.G., Van Der Meer, P., Voors, A.A., Van Veldhuisen, D.J., Van

Gilst, W.H. (2006). Protective effects of erythropoietin in cardiac ischemia: from

bench to bedside. J Am Coll Cardiol 48:2161–7.

Page 104: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Liu, X., Wei, J., Peng, D.H., Layne, M.D., Yet, S.F. (2005). Absence of heme oxygenase-1

exacerbates myocardial ischemia/reperfusion injury in diabetic mice. Diabetes

54:778–784.

Lloyd-Jones, D. et al. (2010). Executive summary: heart disease and stroke statistics--2010

update: a report from the American Heart Association. Circulation 121(7):948–54.

Lu, G., Haider, H.K., Jiang, S., Ashraf, M. (2009). Sca-1þ stem cell survival and engraftment

in the infarcted heart: dual role for preconditioning induced Connexin-43. Circulation

119:2587–2596.

Maiese, K., Chong, Z.Z., Shang, Y.C. (2008). Raves and risks for erythropoietin. Cytokine &

Growth Factor Reviews 19:145–155.

Marfella, R., Di Filippo, C., Esposito, K., Nappo, F., Piegari, E., Cuzzocrea, S., Berrino, L.,

Rossi F. Giugliano D. D’ mico M. (2004). Absence of inducible nitric oxide

synthase reduces myocardial damage during ischemia reperfusion in streptozotocin-

induced hyperglycemic mice. Diabetes 53:454–462.

Martindale, J.L., Holbrook, N.J. (2002). Cellular response to oxidative stress: signaling for

suicide and survival. J Cell Physiol 192:1–15.

Mastromarino, V., Volpe, M., Musumeci, M.B., Autore, C., Conti, E. (2011). Erythropoietin

and the heart: facts and perspectives. Clin Sci 120(2):51–63.

Mathur, A., Martin, J.F. (2004). Stem cells and repair of the heart. Lancet 364:183-92.

Mazzone, T, Chait, A., Plutzky, J. (2008). Cardiovascular disease risk in type 2 diabetes

mellitus: insights from mechanistic studies. Lancet 371(9626):1800–1809.

McCulloch, E.A., Till, J.E. (2005). Perspectives on the properties of stem cells. Nat Med

11:1026–28

Page 105: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Meier, M., Hummel, M. (2009). Cardiovascular disease and intensive glucose control in type

2 diabetes mellitus: moving practice toward evidencebased strategies. Vasc Health

Risk Manag 5:859–871.

Mihov, D., Bogdanov, N., Grenacher, B., Gassmann, M., Zund, G., Bogdanova, A.,

Tavakoli, R. (2009). Erythropoietin protects from reperfusion-induced myocardial

injury by enhancing coronary endothelial nitric oxide production. European Journal

of Cardio-thoracic Surgery 35:839–846.

Mishra, P.K. (2008). Bone marrow-derived mesenchymal stem cells for treatment of heart

failure: is it all paracrine actions and immunomodulation? J Cardiovasc Med

(Hagerstown) 9:122–128.

Moon, C., Krawczyk, M., Ahn, D., Ahmet, I., Paik, D., Lakatta, E.G., et al. (2003).

Erythropoietin reduces myocardial infarction and left ventricular functional decline

after coronary artery ligation in rats. Proc Natl Acad Sci USA 100: 11612–11617.

Ohnishi, S., Yanagawa, B., Tanaka, K., Miyahara, Y., Obata, H., Kataoka, M., Kodama, M.,

Ishibashi-Ueda, H., Kangawa, K., Kitamura, S., Nagaya, N. (2007). Transplantation

of mesenchymal stem cells attenuates myocardial injury and dysfunction in a rat

model of acute myocarditis. J Mol Cell Cardiol 42:88–97.

Ohshima, M., Li, T.S., Kubo, M., Qin, S.H., Hamano, K. (2009). Antioxidant therapy

attenuates diabetes-related impairment of bone marrow stem cells. Circ J 73:162–

166.

Orkin, S.H., Zon, L.I. (2002). Hematopoiesis and stem cells: plasticity versus developmental

heterogeneity. Nat Immunol 3:323–28.

Page 106: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Orlic, D., Kajstura, J., Chimenti, S., Jakoniuk, I., Anderson, S.M., Li, B., Pickel, J., McKay,

R., Nadal-Ginard, B., Bodine, D.M., A Leri, A., Anversa, P. (2001). Bone marrow

cells regenerate infracted myocardium. Nature 410:701–705.

Orzechowski, A. (2003). Justification for antioxidant preconditioning (or how to protect

insulin-mediated actions under oxidative stress). J Biosci 28:39–49.

other cells by spontaneous cell fusion. Nature 416:542–45.

Pacher, P., Nagayama, T., Mukhopadhyay, P., Batkai, S., Kass, D.A. (2008). Measurement of

cardiac function using pressure–volume conductance catheter technique in mice and

rats. Nat Protoc 3(9):1422–1434.

Parsa, C.J., Matsumoto, A., Kim, J., Riel, R.U., Pascal, L.S., Walton, G.B., Thompson, R.B.,

Petrofski, J.A., Annex, B.H., Stamler, J.S., Koch, W.J. (2003). A novel protective

effect of erythropoietin in the infarcted heart. J Clin Invest 112:999 –1007.

Paschos, N., Lykissas, M.G., Beris, A.E. (2008). The Role of Erythropoietin as an Inhibitor

of Tissue Ischemia. Int J Biol Sci 4(3):161–168.

Paul, D., Samuel, S.M., Maulik, N. (2009). Mesenchymal Stem Cell: Present Challenges and

Prospective Cellular Cardiomyoplasty Approaches for Myocardial Regeneration.

Antioxid Redox Signal 11(8): 1841–1855.

Piuhola, J., Kerkela, R., Keenan, J.I., Hampton, M.B., Richards, A.M., Pemberton, C.J.

(2008). Direct cardiac actions of erythropoietin (EPO): effects on cardiac

contractility. BNP secretion and ischaemia/reperfusion injury. Clin Sci 114:293–304.

Pons, J., Huang, Y., Arakawa-Hoyt, J., Washko, D., Takagawa, J., Ye, J., Grossman, W., Su,

H. (2008). VEGF improves survival of mesenchymal stem cells in infarcted hearts.

Biochem Biophys Res Commun 376(2):419-22.

Page 107: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Prunier, F., Pfister, O., Hadri, L., Liang, L., Del, M.F., Liao, R., Hajjar, R.J. (2007). Delayed

erythropoietin therapy reduces post-MI cardiac remodeling only at a dose that

mobilizes endothelial progenitor cells. Am J Physiol Heart Circ Physiol 292:H522–

H529.

Robins, S.J., Collins, D., Wittes, J.T., et al. (2001). Relation of gemfibrozil treatment and

lipid levels with major coronary events: VA-HIT: a randomized controlled trial.

JAMA 285:1585–1591.

Rodrigues, B., Cam, M.C., McNeill, J.H. (1998). Metabolic disturbances in diabetic

cardiomyopathy. Mol Cell Biochem 180:53–57.

Rosova, I., Dao, M., Capoccia, B., D Link, D., Nolta, J.A. (2008). Hypoxic preconditioning

results in increased motility and improved therapeutic potential of human

mesenchymal stem cells. Stem cells 26:2173–2182.

Rossant, J. (2004). Embryonic stem cells in prospective. In Handbook of Stem Cells, ed. R

Lanza. Gearhart, J., Hogan, B.L., Melton, D., Pedersen, R. et al. London: Elsevier

Acad.

Rota, M., LeCapitaine, N., Hosoda, T., Boni, A., De Angelis, A., Padin-Iruegas, M.E.,

Esposito, G., Vitale, S., Urbanek, K., Casarsa, C., Giorgio, M., Lu¨ scher, T.F.,

Pelicci, P.G., Anversa, P., Leri, A., Kajstura, J. (2006). Diabetes promotes cardiac

stem cell aging and heart failure, which are prevented by the deletion of the p66ˢʱᶜ

gene. Circ Res 99:42–52.

Rubart, M., Field, L.J. (2006). Cardiac regeneration: repopulating the heart. Annu Rev

Physiol 68:29–49.

Page 108: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Rubler, S., et al. (1972). New type of cardiomyopathy associated with diabetic

glomerulosclerosis. Am J Cardiol 30(6):595–602.

Ruifrok, W.P., de Boer, R.A., Westenbrink, B.D., van Veldhuisen, D.J., van Gilst, W.H.

(2008). Erythropoietin in cardiac disease: new features of an old drug. Eur J

Pharmacol 585(2-3):270–7.

Ruschitzka, R.T., Wenger, R.H., Stallmach, T., Quaschning, T., de Wit C., Wagner, C.K.,

Labugger, R., Kelm, M., Noll, G., Rülicke, T., Shaw, S., Lindberg, R.L.P.,

Rodenwaldt, B., Lutz, H., Bauer, C., Lüscher, T.F., , and Gassmann, M. (2000).

Nitric oxide prevents cardiovascular disease and determines survival in polyglobulic

mice overexpressing erythropoietin. Proc Natl Acad Sci USA 97(21):11609–13.

Sadamoto, Y., Igase, K., Sakanaka, M., Sato, K., Otsuka, H., Sakaki, S., et al. (1998).

Erythropoietin prevents place navigation disability and cortical infarction in rats with

permanent occlusion of the middle cerebral artery. Biochem Biophys Res Commun

253: 26–32.

Sakanaka, M., Wen, T.C., Matsuda, S., Masuda, S., Morishita, E., Nagao, M., et al. (1998).

In vivo evidence that erythropoietin protects neurons from ischemic damage. Proc

Natl Acad Sci USA 95:4635–4640.

Saladin, K.S. (2005). Human Anatomy. 1st Edition. Mc Graw Hill Higher Eduaction.

Sambuceti, G., Morbelli, S., Vanella, L., Kusmic, C., Marini, C., Massollo, M., Augeri, C.,

Corselli M. Ghersi C. Chiavarina B. Rodella .F. ’ bbate . Drummond, G.,

NG Abraham, N.G., Frassoni, F. (2009). Diabetes impairs the vascular recruitment of

normal stem cells by oxidant damage; reversed by increases in pAMPK, Heme

Oxygenase-1 and Adiponectin. Stem Cells 27:399–407.

Page 109: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Sanchez, A., Garcia-Sancho, J. (2007). Cardiac repair by stem cells. Cell Death Differ

14:1258–1261.

Sarwar, N. et. al. (2010). Diabetes mellitus, fasting blood glucose concentration, and risk of

vascular disease: a collaborative meta-analysis of 102 prospective studies. Lancet

375(9733):2215–22.

Shake, J.G., Gruber, P.J., Baumgartner, W.A., Senechal, G., Meyers, J., Redmond, J.M.,

Pittenger, M, F., and Martin, B.J. (2002). Mesenchymal stem cell implantation in a

swine myocardial infarct model: engraftment and functional effects. Ann Thorac Surg

73:1919–1925.

Sharif, S., Nakagawa, T., Ohno, T., Matsumoto, M., Kita, T., Riazuddin, S., Ito, J.,

Nakagawa, T. (2007). The potential use of bone marrow stromal cells for cochlear

cell therapy. Neuroreport 18:351–354.

Shushakova, N., Park, J.K., Menne, J., Fliser, D. (2009). Chronic erythropoietin treatment

affects different molecular pathways of diabetic cardiomyopathy in mouse. Eur J Clin

Invest 39(9):755–60.

Silverberg, D.S., Wexler, D., Iaina, A., Schwartz, D. (2006). The interaction between heart

failure and other heart diseases, renal failure, and anemia. Semin Nephrol 26:296–

306.

Singh, R., Barden, A., Mori, T., Beilin, L. (2001). Advanced glycation end products: a

review. Diabetologia 44:129–146.

Song, S.W., Chang, W., Song, B.W., Song, H., Lim, S., Kim, H.J., Cha, M.J., Choi, E., Im,

S.H., Chang, B.C., Chung, N., Jang, Y., Hwang, K.C. (2009). Integrin-linked kinase

Page 110: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

is required in hypoxic mesenchymal stem cell for strengthening cell adhesion to

ischemic myocardium. Stem Cells 27:1358–1365.

Sowers, J.R., Epstein, M., Frohlich, E.D. (2001). Diabetes, hypertension, and cardiovascular

disease: an update. Hypertension 37(4):1053–1059.

Sparagna, G.C., Jones, C.E., Hickson-Bick, D.L. (2004). Attenuation of fatty acid-induced

apoptosis by low-dose alcohol in neonatal rat cardiomyocytes. Am J Physiol Heart

Circ Physiol 287(5):H2209–H2215.

Standrings, S., Borley, N.R., Collins, P., Crossman, A.R., Gatzoulis, M.A., Healy, J.,

Johnson D. Mahadevan V. Newell R. .M. Wigley C.B. (2008). Gray’s natomy:

The Anatomical Basis of Clinical Practice.4th

Edition. CHURCHILL LIVINGSTONE

ELSEVIER.

Stolzing, A., Jones, E., McGonagle, D., Scutt, A. (2008). Age related changes in human bone

marrow-derived mesenchymal stem cells: consequences for cell therapies. Mech

Ageing Dev 129:163–173.

Sulaiman, M., Matta, M.J., Sundaresan, N.R. et al. (2010). Resveratrol, an activator of SIRT1

up-regulates sarcoplasmic calcium ATPase and improves cardiac function in diabetic

cardiomyopathy. Am J Physiol Heart Circ Physiol 298:H833–843.

Tarnok, A., Ulrich, H., Bocsi, J. (2009). Phenotypes of Stem Cells from Diverse Origin.

Cytometry 77A:6–10.

Terada, N., Hamazaki, T., Oka, M., Hoki, M., Mastalerz, D.M., Nakano, Y., Meyer, E.M.,

Morel, L., Petersen, B.E., Scott ,E.W. (2002). Bone marrow cells adopt the phenotype

of other cells by spontaneous cell fusion. Nature 416(6880):542-5.

Page 111: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Tharaux, P.L., Chatziantoniou, C., Casellas, D., et al. (1999). Vascular endothelin-1 gene

expression and synthesis and effect on renal type I collagen synthesis and

nephroangiosclerosis during nitric oxide synthase inhibition in rats. Circulation

99:2185–91.

Thomson, J.A., Itskovitz-Eldor, J., Shapiro, S.S., Waknitz, M.A., Swiergiel, J.J., et al.

(1998). Embryonic stem cell lines derived from human blastocysts. Science

282:1145–47

Thum, T., Fraccarolla, D., Schultheiss, M., Froese, S., Galuppo, P., Widder, J.D., Tsikas,

D., G Ertl, G., Bauersachs, J. (2007). Endothelial nitric oxide synthase uncoupling

impairs endothelial progenitor cell mobilization and function in diabetes. Diabetes

56:666–674.

Toma, C., Letts, D.P., Tanabe, M., Gorcsan III, J., Counihan, P.J. (2007). Positive effect of

darbepoetin on peri-infarction remodeling in a porcine model of myocardial

ischemia–reperfusion. J Mol Cell Cardiol 43:130–136.

Toma, C., Pittenger, M.F., Cahill, K.S., Byrne, B.J., and Kessler, P.D. (2002). Human

mesenchymal stem cells differentiate to a cardiomyocyte phenotype in the adult

murine heart. Circulation 105:93–98.

Tong, Q., Zheng, L., Lin, L., Li, B., Wang, D., Huang, C., Li, D. (2006). VEGF is

upregulated by hypoxia-induced mitogenic factor via the PI-3K/Akt-NF-kappaB

signaling pathway. Respir Res 7(1):37.

Tramontano, A.F., Muniyappa, R., Black, A.D., Blendea, M.C., Cohen, I., Deng, L., Sowers,

J.R., Cutaia, M.V., El-Sherif, N. (2003) Erythropoietin protects cardiac myocytes

Page 112: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

from hypoxia-induced apoptosis through an Akt-dependent pathway. Biochem

Biophys Res Commun 308:990–4.

Tschope, C., Walther, T., Escher, F., Spillmann, F., Du, J., Altmann, C., Schimke, I., Bader,

M., Sanchez-Ferrer, C.F., Schultheiss, H.P., et al. (2005). Transgenic activation of the

kallikrein-kinin system inhibits intramyocardial inflammation, endothelial

dysfunction and oxidative stress in experimental diabetic cardiomyopathy. Faseb J

19(14):2057–2059.

Tschope, C., Walther, T., Koniger, J., Spillmann, F., Westermann, D., Escher, F.,

Pauschinger, M., Pesquero, J.B., Bader, M., Schultheiss, H.P., Noutsias, M. (2004).

Prevention of cardiac fibrosis and left ventricular dysfunction in diabetic

cardiomyopathy in rats by transgenic expression of the human tissue kallikrein gene.

Faseb J 18(7):828–835.

Tuo, Q., Zeng, H., Stinnett, A., Yu, H., Aschner, J.L., Liao, D. (2008). Critical role of

angiopoietins/Tie-2 in hyperglycemic exacerbation of myocardial infarction and

impaired angiogenesis. Am J Physiol Heart Circ Physiol 294:H2547–H2557.

Ueda, K., Takano, H., Niitsuma, Y., Hasegawa, H., Uchiyama, R., Oka, T., Miyazaki, M.,

Nakaya, H., Komuro, I. (2010). Sonic hedgehog is a critical mediator of

erythropoietin-induced cardiac protection in mice. J Clin Invest 120(6):216–229.

UK Prospective Diabetes Study Group. (1998). Tight blood pressure control and risk of

macrovascular complications in type 2 diabetes: UKPDS 38. BMJ 317:703–711.

UK Prospective Diabetes Study Group. (2000). Association of glycemia with macrovascular

and microvascular complications of type 2 diabetes (UKPDS 35): prospective

observational study. BMJ 321:405–412.

Page 113: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Urbich, C., DImmeler, S. (2004). Endothelial progenitor cells: characterization and role in

vascular biology. Circ Res 95:343–353.

Vanhoutte, P.M. (1988).The endothelium--modulator of vascular smooth-muscle tone. N

Engl J Med 319:512–3.

Verfaillie, C.M. (2002). Adult stem cells: assessing the case for pluripotency. Trends Cell

Biol 12:502–08.

Verma, S., Maitland, A., Weisel, R.D., Li, S.H., Fedak, P.W., Pomroy, N.C., Mickle, D.A.,

Li, R.K., Ko, L., Rao, V. (2002). Hyperglycemia exaggerates ischemia reperfusion-

induced cardiomyocyte injury: reversal with endothelin antagonism. J Thorac

Cardiovasc Surg 123:1120–1124.

Voulgari, C., Papadogiannis, D., Tentolouris, N. 2010. Diabetic cardiomyopathy: from the

pathophysiology of the cardiac myocytes to current diagnosis and management

strategies. Vasc Health Risk Manag 6:883–903.

Wagner, W., Horn, P., Castoldi, M., Diehlmann, A., Bork, S., et al. (2008). Replicative

Senescence of Mesenchymal Stem Cells: A Continuous and Organized Process. PLoS

ONE 3(5):e2213.

Wang, J., Song, Y., Wang, Q., Kralik, P.M., Epstein, P.N. (2006). Causes and Characteristics

of Diabetic Cardiomyopathy. Rev Diabetic Stud 3(3):108–117.

Wang, J.S., Shum-Tim, D., Galipeau, J., Chedrawy, E., Eliopoulos, N., and Chiu, R.C.

(2000). Marrow stromal cells for cellular cardiomyoplasty: feasibility and potential

clinical advantages. J Thorac Cardiovasc Surg 120:999–1005.

Page 114: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Wang, Y., Haider, H.Kh., Ahmad, N., Zhang, D., Ashraf, M. (2006). Evidence for ischemia

induced host-derived bone marrow cell mobilization into cardiac allografts. J Mol

Cell Cardiol 41:478–487.

Weissman, I.L. (2000). Translating stem and progenitor cell biology to the clinic: barriers

and opportunities. Science 287:1442–46.

Westermann, D., Rutschow, S., Jager, S., Linderer, A., Anker, S., Riad, A., Unger, T.,

Schultheiss, H.P., Pauschinger, M., Tschope, C. (2007). Contributions of

inflammation and cardiac matrix metalloproteinase activity to cardiac failure in

diabetic cardiomyopathy: the role of angiotensin type 1 receptor antagonism.

Diabetes 56(3):641–646.

Westermann, D., Van Linthout, S., Dhayat, S., Dhayat, N., Schmidt, A., Noutsias, M., Song,

X.Y., Spillmann, F., Riad, A., Schultheiss, H.P., et al. (2007). Tumor necrosis

factoralpha antagonism protects from myocardial inflammation and fibrosis in

experimental diabetic cardiomyopathy. Basic research in cardiology 102(6):500–507.

Wright ,G.L., Hanlon, P., Amin, K., Steenbergen, C., Murphy, E., Arcasoy, M.O. (2004).

Erythropoietin receptor expression in adult rat cardiomyocytes is associated with an

acute cardioprotective effect for recombinant erythropoietin during ischemia–

reperfusion injury. FASEB J 18:1031–1033.

Yagyu, H., Chen, G., Yokoyama, M., Hirata, K., Augustus, A., Kako, Y., Seo, T., Hu, Y.,

Lutz, E.P., Merkel, M., Bensadoun, A., Homma, S., Goldberg, I.J. (2003).

Lipoprotein lipase (LpL) on the surface of cardiomyocytes increases lipid uptake and

produces a cardiomyopathy. J Clin Invest 111(3):419–426.

Page 115: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

Ying, Q.L., Nichols, J., Evans, E.P., et al. (2002). Changing potency by spontaneous fusion.

Nature 416:545–48.

Zhang, D., Zhang, F., Zhang, Y., Gao, X., Li, C., Yang, N., Cao, K. (2007). Combining

erythropoietin infusion with intramyocardial delivery of bone marrow cells is more

effective for cardiac repair. Transpl Int 20(2):174–83.

Zhang, D.G., Zhang, F.M., Zhang, Y.Q., Zhou, F., Gao, X., Li, C.F., Cao, K.J. (2006).

Erythropoietin enhances the therapy potency of autologous bone marrow stromal cells

in a rat heart infarction model via phosphatidylinositol-3-kinase/Akt pathway.

Zhonghua Xin Xue Guan Bing Za Zhi 34(10):912–6.

Zhang, L., Cannell, M.B., Phillips, A.R., Cooper, G.J., Ward, M.L. (2008). Altered calcium

homeostasis does not explain the contractile deficit of diabetic cardiomyopathy.

Diabetes 57(8):2158–2166.

Zhang, N., Li, J., Luo, R., Jiang, J., Wang, J.A. (2008). Bone marrow mesenchymal stem

cells induce angiogenesis and attenuate the remodeling of diabetic cardiomyopathy.

Exp Clin Endocrinol Diabetes 116:104–111.

Zhou, Y., Larsen, P.H., Hao, C., Yong, V.W. (2002). CXCR4 is a major chemokine receptor

on glioma cells and mediates their survival. J Biol Chem 277:49481–49487.

Page 116: POTENTIAL OF MESENCHYMAL STEM CELLS FOR THE …prr.hec.gov.pk/Jspui/Bitstream/123456789/2202/2/1644S.pdfpotential of mesenchymal stem cells for the repair of diabetic heart failure

PUBLICATION

Growth Factor Preconditioning Increases the Functionof Diabetes-Impaired Mesenchymal Stem Cells Mohsin Khan,* Shoaib Akhtar,* Sadia Mohsin, Shaheen N. Khan, and Sheikh Riazuddin

STEM CELLS AND DEVELOPMENT Volume 20, Number 1, 2011

*Both authors equally contributed


Recommended