+ All Categories
Home > Documents > By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88...

By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88...

Date post: 10-Jun-2020
Category:
Upload: others
View: 0 times
Download: 0 times
Share this document with a friend
115
ROLE OF ROBO GENES DURING MURINE VERTEBRAL COLUMN FORMATION By LISA Y. LAWSON A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2017
Transcript
Page 1: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

ROLE OF ROBO GENES DURING MURINE VERTEBRAL COLUMN FORMATION

By

LISA Y. LAWSON

A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT

OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY

UNIVERSITY OF FLORIDA

2017

Page 2: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

2

© 2017 Lisa Y. Lawson

Page 3: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

3

To my parents for their encouragement and support, and to my sister who kept me sane in all my endeavors

Page 4: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

4

ACKNOWLEDGMENTS

I thank members of the Cohn Lab for their support and understanding, and for

being so inclusive in their science and friendship. I thank Kendra McKee for always

being so present, even when we couldn‟t be on the same floor. I couldn‟t have asked for

a better friend and more helpful lab mom. I also thank Emily Merton for being so

generous in her understanding while I worked to finish my studies in the Harfe lab.

Finally, I thank Brian Harfe for his patience and understanding every time I found myself

flustered by a deadline.

Page 5: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

5

TABLE OF CONTENTS

page AWKNOWLEDGEMENTS………………………………………………………… 4

LIST OF TABLES……………………………………………………….…………. 7

LIST OF FIGURES……………………………………………………….…….….. 8

ABSTRACT……………………………………………………………………….… 10

CHAPTER

1 LITERATURE REVIEW: DEVELOPMENTAL MECHANISMS OF VERTEBRAL COLUMN AND INTERVERTEBRAL DISC MORPHOGENESIS………………………………………………………

12

Introduction………………………………………………………………... 12 Embryonic Origins of the Vertebral Column…………………….…….. 14 The Notochord………………………………………………….. 15 Somites………………………………………………………….. 16 Developmental Origins of the Annulus Fibrosus……………………… 18 Genes Required For Development of the Nucleus Pulposus……….. 19 Hedgehog Signaling Pathway…………………………………. 19 Foxa1 and Foxa2……………………………………………….. 21 T-Brachyury……………………………………………………… 22 Noto……………………………….………………………………. 22 Genes Required For Development of the Annulus Fibrosus……….. 23 Pax1 and Pax9………………………………………………….. 24 Bapx1……………………………….……………………………. 26 Sox Family of Genes…………………………………………... 26 Scleraxis And TGFβ Signaling………………………………… 28 Conclusions……………………………….………………………………. 29 Genes Required For Murine Vertebral Column Formation…………... 31 Anatomy Of The Vertebral Column And Intervertebral Disc…………. 34 Vertebral Column Morphogenesis In Mice…………………………….. 34 Postnatal Intervertebral Disc Development……………………………. 35 2 ROLE OF ROBO GENES DURING MURINE VERTEBRAL

COLUMN MORPHOGENESIS…………………………………………. 36

Introduction……………………………….……………………………… 36 Results……………………………….…………………………………… 39 Complementary Robo and Slit Expression Patterns in the

Vertebral Column……………………………………………….. 39

Robo expression…………………………………………. 39 Slit expression……………………………………………. 41

Page 6: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

6

Intervertebral Disc Malformations………………………………. 42 The Notochord to Nucleus Pulposus Transition...……………. 43 Rib and Sternal Development…………………………………... 48 Proliferative Deficits in the Sclerotome………………………… 49 Robo and Pax1 Expression in the Vertebral Column………… 50 Robo Gene Expression and Function in the Growth Plates…. 52 Annulus Fibrosus Cell Morphology and Gene Expression…... 54 Cartilage Matrix Protein Expression …………………………… 54 Tenascin C Upregulation………………………………………… 55 Discussion and Conclusions…………………………………………….. 56 Role of Robo1 and Robo2 in IVD Development……………… 56 Robo1 and Robo2 Function in the Sclerotome……………….. 59 Robo1 and Robo2 in Growth Plate Maintenance…………….. 60 Figures……………………………………………………………………... 62 3 WNT/β-CATENIN SIGNALING DURING INTERVERTEBRAL DISC

DEVELOPMENT………………………………………………………….. 79

Introduction…………...…………………………………………………… 79 Results……………………………………………………………………... 81 Conclusions……………………………………………………………….. 83 Figures……………………………………………………………………... 86 4 METHODS……………………………….………………………………...

90

Histology……………………………….………………………………….. 90 Skeletal Preparations…………………………………………………….. 90 Western Blot Analysis……………………………………………………. 91 ECM Analysis By Immunofluorescence………………………………... 92 Characterization Of Robo And Slit Expression………………………... 93 X-Gal Staining…………………………………………………… 93 RNA In Situ Hybridization………………………………………. 94 Lineage Tracing Analyses……………………………………………….. 96 Analysis Of Sclerotomal Proliferation…………………………………... 97 EdU Pulse And Detection………………………………………. 97 B-Gal Antibody As Proxy For Robo Expression……………... 98 Quantification……………………………….…………………… 98 Growth Plate Analysis……………………………….…………………… 99 Characterization of Wnt/β-Catenin Signaling Activity………………… 99 Immunofluorescence And Western Blot Antibodies…………………... 100 Genotyping Primers……………………………….……………………… 100

REFERENCES……………………………….…………………………………….. 102 BIOGRAPHICAL SKETCH………………………………………………………...

114

Page 7: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

7

LIST OF TABLES

Table Page

1-1 Genes Required For Murine Vertebral Column Formation……… 31

4-1 ECM Immunofluorescence And Western Blot Antibodies……….. 100

4-2 Genotyping Primers…………………………………………………. 100

Page 8: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

8

LIST OF FIGURES

Figure page

1-1 Anatomy of the Vertebral Column and Intervertebral Disc…….... 34

1-2 Vertebral Column Development In Mice…………………………... 34

1-3 Postnatal Intervertebral Disc Development……………………….. 35

2-1 Robo1 and Robo2 Expression Analysis by RNA In Situ Hybridization and Xgal Staining…………………………………….

62

2-2 Robo Gene Expression During Vertebral Column Formation....... 63

2-3 Slit Gene Expression During Vertebral Column Formation……... 64

2-4 Removal of Robo1 and Robo2 Causes Intervertebral Disc Malformations in P0 Mice……………………………………………

65

2-5 Conditional Inactivation of Robo2F on Robo1 Null Background.. 66

2-6 The Notochord to Nucleus Pulposus Transition Occurs Normally in the Absence of Robo1 and Robo2………………………………

67

2-7 Lineage Tracing Analysis of ShhCreERT2 Marked Cells............. 68

2-8 Robo1;Robo2 Null Mice Have Deficits in the Distal Ribs and Sterna at P0……………………………………………………….....

69

2-9 Impaired Proliferation in Robo1; Robo2 Null Mutants at 13.5dpc 70

2-10 Robo and Pax1 Have Overlapping Expression Patterns in the Developing Vertebral Column……………………………………….

72

2-11 Disrupted Growth Plate Marker Expression in Robo1;Robo2 Null Mutants…………………...……………………………………...

73

2-12 Robo1;Robo2 Null Annuli Fibrosi Have Aberrant Cell Morphologies And Ectopic Expression of Growth Plate Chondrocyte Markers………………………………………………..

75

2-13 Robo1 and Robo2 Loss is Associated With Reduced Expression of Cartilage Matrix Proteins…………………………...

76

Page 9: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

9

2-14 Tenascin C is Upregulated in the Intervertebral Discs of Robo1;Robo2 Null Mice…………………………………………….

77

2-15 Slit1;Slit2 Null Phenotype………...………...………...………....... 78

3-1 WNT/β-Catenin Signaling in the Notochord at 12.5 dpc………… 86

3-2 WNT Signaling in the Embryonic Intervertebral Discs…………… 87

3-3 Postnatal Wnt Signaling in the Intervertebral Discs……………… 88

Page 10: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

10

Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy

ROLE OF ROBO GENES DURING MURINE VERTEBRAL COLUMN FORMATION

By

Lisa Y. Lawson

May 2017

Chair: Brian D. Harfe Major: Medical Sciences – Genetics

The vertebral column is comprised of ossified vertebrae which are adjoined by

the fibrocartilagenous intervertebral discs. In humans, common pathologies affecting the

vertebral column include scoliosis and intervertebral disc degeneration, which is thought

to be a major cause of lower back pain in adults worldwide. The molecular mechanisms

underlying scoliosis and disc degeneration in humans are not well understood. Robo

proteins are a conserved class of transmembrane receptors which are most classically

known for their role in regulating axonal projections via a chemorepulsive mechanism

during nervous system development. The diverse roles of Robo proteins in modulating

cell proliferation, survival, migration, and adhesion in vivo during vertebrate

embryogenesis, however, has been illuminated more clearly by recent studies in Robo

knockout mice. A role for Robo proteins in sclerotome differentiation and vertebral

column development has not been reported previously. Here we show that Robo1 and

Robo2 are expressed in the embryonic sclerotome, and mice that lack both Robo1 and

Robo2 develop vertebral columns with enlarged intervertebral discs with aberrant

annulus fibrosus cell morphology. In addition, we demonstrate that in the absence of

Robo1 and Robo2, the organized expression of Col10a and Ihh in vertebral growth plate

Page 11: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

11

chondrocytes is disrupted. These changes are preceded by decreased proliferation in

the sclerotome and are accompanied by deficits in the distal ribs and sterna at birth.

Page 12: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

12

CHAPTER 1 LITERATURE REVIEW: DEVELOPMENTAL MECHANISMS OF VERTEBRAL

COLUMN AND INTERVERTEBRAL DISC MORPHOGENESIS

Introduction

The axial skeleton is comprised of the skull and ossified elements of the middle

ear, the hyoid bone at the base of the neck, the ribs, sternum, and the vertebral column

(White, et al. 2012). Within the vertebral column, the intervertebral discs are the

fibrocartilagenous joint-like structures that connect and cushion the vertebrae. In young

and healthy animals, the intervertebral discs are comprised of three functionally distinct

regions that interact synergistically to facilitate fluid and painless movement of the

spine. Each intervertebral disc is comprised of a gel-like core called the nucleus

pulposus, a fibrocartilagenous surrounding called the annulus fibrosus, and the

cartilaginous endplates (Fig. 1-1).

Within each disc, the nucleus pulposus is comprised of cells suspended in a gel-

like matrix of negatively charged proteoglycans that attract and retain water molecules

in the intervertebral disc core (Sivan, et al. 2014). Hydration in the nucleus pulposus

allows for uniform re-distribution of compressive forces generated by vertebral column

movement (Sivan, et al. 2012). Surrounding each nucleus pulposus core is the annulus

fibrosus, which is comprised of spindle-shaped chondrocyte-like cells embedded in a

highly organized matrix of fibrillar proteins. The concentric, lamellar organization and

structured orientation of fibrillar proteins in the annulus fibrosus allow it to withstand the

shear and ply tensions it is subjected to as the intervertebral discs bear loads (Cortes

and Elliot, 2012; Romgens, et al. 2013; Hayes, et al. 2011). Finally, as the intervertebral

discs are aneural and avascular in adulthood, the cartilaginous endplates are important

not only for adhering the discs to adjacent vertebrae but also for their role in nutrient

Page 13: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

13

and waste diffusion into and out of the discs (Richardson, et al., 2012; Smith and Elliot,

2011; Malandrino, et al., 2014).

As animals age, cell senescence and metabolic changes in the discs can lead to

disc degeneration and consequently, to impaired disc function and disc related lower

back pain (Colombier, et al., 2014; Gruber et al., 2007; Gruber, et al., 2009; Le Maitre,

et al., 2007; Smith, et al., 2011). Disc degeneration can be caused by metabolic

imbalances or by acute trauma. Hallmarks of disc degeneration include decreased

nucleus pulposus hydration, decreased disc height, ruptured annuli fibrosi, and calcified

endplates (Smith, et al., 2011). A major constituent of the non-collagenous extracellular

matrix in the discs is Aggrecan. Aggrecan plays an integral role in maintaining the load-

bearing properties of the discs, and in humans Aggrecan loss is associated with disc

degenration (Sivan, et al. 2014). By covalently interacting with 100 or more negatively

charged sulfated glycosaminoglycan (GAG) side chain groups, Aggrecan maintains the

osmolality required to keep the discs hydrated (Kiani, et al., 2002).

In humans, Aggrecan abundance in the discs peaks in the early twenties and

steadily declines thereafter (Sivan, et al. 2014). As a consequence of Aggrecan loss,

the ability of nuclei pulposi to uniformly redistribute compressive forces becomes

compromised, leading to uneven pressures against the surrounding annuli fibrosi. As a

result, the annulus fibrosus can tear, leading to eruption of nucleus pulposus matter

through the annulus fibrosus. These tears trigger wound-healing processes that

promote abnormal neo-vascularization and innervation in the discs, which are thought to

be a contributing factor in disc related back pain in humans (Tolofari, et al., 2010;

Freemont, et al., 2002).

Page 14: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

14

Disc degeneration and associated back pain in humans is proposed to arise from

a combination of genetic and environmental factors, which can include occupation,

pregnancy, smoking, and body mass index (Abbas, et al., 2013; Dario, et al., 2015;

Hestbaek, et al., 2004; Nasto, et al., 2014). By some estimates, the direct and indirect

economic burden associated with low back pain in the United States alone exceeds

$100 billion annually (Andersson, 1999; Katz, 2006; Buchbinder, et al., 2013). Based on

recent projections made by the Global Burden of Disease Study (GBD), low back pain is

now the leading cause of disability worldwide, superseding heart disease, diabetes, and

major depressive and anxiety disorders (Millenium, 2003). The lifetime prevalence for

low back pain has been reported to be as high as 80-85% (Buchbinder, et al. 2013).

Disc associated lower back pain can be debilitating, limiting range of motion and

quality of life. To reverse disc pathology in the clinic, a better understanding of disc

biology and development in the context of vertebral column formation is necessary. This

literature review focuses on what is known about the molecular and signaling

mechanisms underlying vertebral column formation and intervertebral disc

morphogenesis with an emphasis on genetic mouse models.

Embryonic Origins of the Vertebral Column

The vertebral column forms from cells contained in two distinct embryonic

compartments: the notochord and somites. The notochord gives rise to the nuclei

pulposi of each intervertebral disc in adult mice (Choi and Harfe, 2011; MacCann, et al.,

2012). Within each somite, a specific compartment known as the sclerotome

differentiates to form the vertebrae and annuli fibrosi. Notochord and somite

differentiation occur in unison during embryogenesis. Because signaling interactions

between the notochord and somites are required for sclerotome induction and

Page 15: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

15

subsequent development, genetic abnormalities that impair notochord formation impede

vertebral column development early in development.

The Notochord

In mice the notochord forms from the node in the axial mesoderm beginning

around 7.5 dpc (days post coitum) as a tube-like structure that elongates rostro-caudally

at the midline, ventral to the neural tube (Sulik, et al., 1994; Yamanaka, et al., 2007).

The notochord is composed of large vacuolated cells which are enveloped in an

acellular membrane known as the “notochordal sheath” (Paavola, et al. 1980). These

large vacuoles inside notochordal cells are thought to play a role in body axis elongation

by exerting a hydrostatic pressure against the basement membrane of the notochordal

sheath (Corallo, et al., 2015; Stemple, 2005). The acellular notochordal sheath is

composed of collagens, cytokeratins, laminin, fibronectin, and other glycosaminoglycan-

modified proteoglycans (Lehtonen, et al., 1995; Gotz, et al., 1995). The notochordal

sheath, which physically segregates notochordal cells from the surrounding paraxial

mesoderm, is integral to notochord cell survival and is likely synthesized by notochordal

cells. Genetic perturbations that impair notochordal sheath integrity have been linked to

aberrant notochord cell death in mice (Smits and Lefebvre, 2003; Choi and Harfe,

2011). Presently, it is unclear whether cell death occurs as a consequence of

notochordal sheath loss, or vice versa.

Recently, it was proposed that the notochordal sheath was essential for the

notochord to nucleus pulposus transition. In this model, the notochordal sheath acts as

a membranous barrier, separating the axial and paraxial mesoderms (i.e. notochord and

somites, respectively) as notochordal cells move into nucleus pulposus anlagen

Page 16: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

16

beginning around 12.5 dpc (Fig. 1-2). In these experiments, Hedgehog signaling was

conditionally removed in the notochord, resulting in disrupted notochordal sheath

integrity, followed by aberrant scattering of notochordal cells throughout the vertebral

column. As a result of Hedghog signaling removal, mice developed to 18.5 dpc with

severe loss of disc and vertebral structures in the axial skeleton (Choi and Harfe, 2011).

Similarly, inactivation of Sox5 and Sox6 together produced defects in the notochordal

sheath followed by deficits in vertebral structures (Smits et al. 2003). Deletion of the

Foxa transcription factors (Foxa1 and Foxa2), which act upstream of Shh, have also

been shown to cause notochordal sheath defects with resultant phenotypes in the

vertebral column (Maier and Harfe, 2013).

Somites

Somites are bilateral blocks of mesoderm that flank the notochord and neural

tube. During embryogenesis cells in the unsegmented pre-somitic mesoderm (PSM)

epithelize and bud off rostrocaudally to form somites, which flank the notochord and

neural tube. (Tam and Trainor, 1994). Somite formation, or somitogenesis, is regulated

by the FGF, Notch, and Wnt signaling pathways (Saga, 2012). In mice, 65 somite pairs

give rise to 65 vertebrae, while in humans 33 somite pairs form the 33 vertebrae

(Theiler, 1972; Haque, et al. 2001).

Once formed, somites acquire dorso-ventral polarity in response to signals from

the surface ectoderm and ventral midline (floor plate and notochord). Wnt signals from

the surface ectoderm induce dermamyotome specification in the dorsal somite. The

dermamyotome, which expresses Pax3, Pax7, and Sim1 (Goulding, et al. 1991; Fan, et

al. 1994), subsequently differentiates to form the myotome and dermatome, which

Page 17: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

17

develop into muscles and dermis, respectively. Myotome specification is distinguished

by the expression of Myod1 and Myf5 (Borycki, et al. 1999), which are required to

initiate FGF signaling in the myotome (Brent, et al. 2005). FGF signaling in the myotome

is required for specification of tendon progenitors in the ventral somitic mesoderm

(Brent, et al. 2003).

In the ventral somite, Shh from the notochord and floor plate induce Pax1

expression, which marks specification of the sclerotome. Following the induction of

chondroprogenitors in the sclerotome, cells in the dorsolateral edge of each sclerotome

express Scx (Sclerxis) in response to FGF signaling from the myotome (Brent, et al.

2003). The syndetome is comprised of Scx-expressing cells, which are the progenitors

that develop into tendons and ligaments (Brent, et al. 2003; Brent et al. 2005; Cserjesi,

et al. 1995). Mutations in Scx or its targets, Tenomodulin and Mohawk, produce deficits

in tendon formation or function, including those that anchor the vertebrae to each other

(Shukunami, et al. 2006; Liu, et al. 2015; Murchison, et al. 2007). Syndetome induction

in chick embryos is mediated in part by FREK and FGF effectors Pea3 and Ets (Brent,

et al. 2004).

Finally, the sclerotome gives rise to all the cartilaginous and ossified components

of the ribs and vertebral column. The sclerotome is molecularly distinguishable within

the somitic mesoderm by Pax1, Pax9, and Bapx1 expression (Herbrand, et al. 2002;

Rodrigo, et al. 2003; Peters, et al. 1999). The sclerotome forms the outer parts of the

intervertebral discs, including the annuli fibrosi and cartilaginous endplates (Bruggeman,

et al. 2012). The sclerotome also gives rise to the vertebrae, including the ventral

vertebral bodies the dorsal spinous processes that encircle and protect the spinal cord.

Page 18: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

18

The sclerotome also gives rise to the proximal and distal ribs (Aoyama, et al. 2005).

Formation of the vertebrae occurs by re-segmentation, wherein the caudal half of one

somite interacts with the rostral half of an adjacent somite to form one vertebral unit.

Re-segmentation has been observed in chicks by tracking dye-labeled cells and in mice

by observing deficits that occur when genes that regulate somite polarity and re-

segmentation, like Uncx4.1 and Tbx18, are removed (Aoyama, et al. 2000; Leitges, et

al. 2000; Bussen, et al. 2004).

Developmental Origins of the Annulus Fibrosus

The intervertebral discs form from cells present in the notochord and sclerotome

(Choi, et al. 2012; Christ, et al. 2004). All cells in nuclei pulposi are derived from the

notochord (Choi and Harfe, 2011; McCann, et al. 2012). The annulus fibrosi and

endplates form from the sclerotome (Bruggeman, et al. 2012). Cell tracing analyses of

lipophilic dye injections have demonstrated that in chickens, which do not contain nuclei

pulposi, the intervertebral discs (annulus fibrosus) are derived from rostral sclerotome

(Bruggeman, et al. 2012). Other lines of evidence suggesting a sclerotomal origin for

the annuli fibrosi include reports that Pax1 is expressed in the sclerotome and annuli

fibrosi but is absent in the notochord and nuclei pulposi (DiPaola, et al. 2005;

Senthinathan, et al. 2012).

Other genes expressed in the sclerotome include Uncx4.1 (Uncx) and Tbx18

(Leitges, et al. 2000; Mansouri, et al. 1997; Kraus, et al. 2001). Analyses of Uncx4.1-

LacZ transgenic mice showed that Uncx expressing cells in the caudal sclerotome gave

rise to the presumptive endplates and annulus fibrosi of the intervertebral discs

(Takahashi, et al. 2013). Tbx18 was shown to be expressed in the anterior mouse

Page 19: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

19

sclerotome at 10.5 dpc and fate mapping of Tbx18Cre;R26R mice showed that these

Tbx18-expressing cells later formed the annulus fibrosus (Kraus, et al. 2001). Both

Uncx and Tbx18 null mice die perinatally with severe abnormalities in the vertebral

column suggestive of defects in lateral sclerotome differentiation (Bussen, et al. 2004;

Leitges, et al. 2000).

Genes Required For Development Of The Nucleus Pulposus

Uncovering the molecular pathways responsible for formation of the

intervertebral discs has lagged compared to many other tissues. For example, the role

Shh plays in limb development was initially described >20 years ago but only recently

was Shh shown to be important for formation of the intervertebral discs (Riddle, et al.

1993; Choi and Harfe, 2011). Many of the key signaling pathways involved in vertebral

column formation and intervertebral disc morphogenesis also play critical roles in

multiple other developmental processes. (Chiang, et al. 1996). Thus, the creation of null

alleles of many of these genes results in early lethality, prior to disc formation.

Advances in mouse genes in the past decade, however, including the creation of large

numbers of conditional mouse alleles and the characterization of Cre alleles, including

ShhCre and NotoCre, have allowed researchers in the field to study the roles of specific

genes in the relevant tissues that give rise to the discs (Harfe, et al. 2004; Choi and

Harfe, 2011; McCann, et al. 2012).

Hedgehog Signaling Pathway

Shh (Sonic Hedgehog) is a secreted ligand that is responsible for activating

Hedgehog signaling within many cell types. It is expressed in the node, notochord and

nucleus pulposus (Choi and Harfe, 2011; Jeong, et al. 2003; Dahia, et al. 2012).

Page 20: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

20

Mutations in Shh affect vertebral column formation, both directly and indirectly (Choi

and Harfe, 2011; Chiang, et al. 1996; Choi, et al. 2012). Shh null embryos developed a

node and caudal notochord, as evidenced by the presence of T-brachyury mRNA, but

the notochord quickly disintegrated following down-regulation in Foxa2 in the

rudimentary notochord (Chiang, et al. 1996). These findings suggested that Shh was

dispensable for node formation and initiation of the notochord from the node, but was

required for notochord growth and elongation in a Foxa2-dependent manner (Chiang, et

al. 1996). Shh null embryos were embryonic lethal with severe deficits in multiple organ

systems and had severely reduced axial skeletons with completely absent intervertebral

discs and vertebrae (Chiang, et al. 1996).

Subsequent studies in conditional Hedgehog mutants further illuminated the role

of Shh in notochord and intervertebral disc biology. Hedgehog signaling is mediated by

the Smo (Smoothened) cell surface receptor. Smo removal has been shown to abolish

Hedgehog signaling in targeted cells (Zhang, et al. 2001). When Hedgehog signaling

was conditionally inactivated in the notochord using a floxed allele of Smo, the vertebral

column failed to form (Choi and Harfe, 2011). Hedgehog inactivation in the notochord

resulted in loss of the notochordal sheath, proliferative deficits in the notochord, and

aberrant scattering of notochordal cells throughout the paraxial mesenchyme (Choi and

Harfe, 2011). Hedgehog signaling has also been documented in the postnatal discs and

has been suggested to play a role in re-activating Wnt signaling for intervertebral disc

maintenance in aged animals (Dahia, et al. 2009; Dahia, et al. 2012; Winkler, et al.

2014).

Page 21: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

21

Foxa1 and Foxa2

Foxa2 (also called HNF-3b or hepatocyte nuclear factor 3b) is a DNA-binding

protein in the Forkhead box family of transcription factors that is critical for notochord

formation and maintenance (Weinstein, et al. 1994; Ang and Rossant, 1994). Foxa2 null

embryos were described as having defects in primitive streak elongation which resulted

in notochord agenesis followed by embryonic lethality by 11.5 dpc (Ang and Rossant,

1994). Expression of another Foxa family transcription factor, Foxa1 (also called HNF-

3a) has also been described in the notochord (Kaestner, et al. 1994). Homozygous

deletion of Foxa1 did not affect notochord formation and intervertebral disc

morphogenesis (Kaestner, et al. 1994).

A subsequent study illustrated the functional redundancy between Foxa1 and

Foxa2 in the notochord. ShhCre was used to conditionally inactivate a floxed allele of

Foxa2 on a Foxa1 null background in mice (Maier et al. 2013). Using this approach, it

was shown that while removal of Foxa1 or Foxa2 resulted in normal development,

removal of both Foxa1 and Foxa2 in the notochord severely impaired the ability of the

notochord to transition into nuclei pulposi (Maier, et al. 2013). Perinatal Foxa1;Foxa2

mutants had underdeveloped vertebral columns with severely reduced and misshapen

intervertebral discs (Maier, et al. 2013). These results make sense in light of other

studies that showed direct binding of Foxa transcription factors to the regulatory

sequences of many notochord-specific genes (Tamplin, et al. 2011).

Notably, the Foxa1;Foxa2 mutant phenotype was similar to the phenotype

observed in the vertebral column when Hedgehog signaling was inactivated in the

notochord (Maier, et al. 2013; Choi and Harfe, 2011). These results were congruent with

Page 22: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

22

what is known about the genetic interaction between Shh and the Foxa transcription

factors. Foxa1 protein has been shown to directly bind the Shh notochord-specific

enhancer (Jeong and Epstein, 2003). Consistent with these observations, Hedgehog

signaling was down-regulated in Foxa1;Foxa2 mutants (Maier, et al. 2013). Based on

these studies, it is likely that the Foxa family of transcription factors directly regulate Shh

expression in the notochord. Curiously, Foxa2 expression was observed in Shh null

embryos. However, in the absence of Shh, Foxa2 was subsequently down-regulated in

the notochord (Chiang, et al. 1996). These results suggest that maintenance of Foxa2

expression is regulated by a Shh-dependent feedback loop in the notochord.

T-Brachyury

T-Brachyury is a conserved T-box transcription factor that is expressed in the

node, notochord, and nucleus pulposus (Wilkinson, et al. 1990; Hermann, 1992; Dahia,

et al. 2012). Compared to Foxa2 null embryos, which failed to form any notochord, T-

brachyury null embryos formed a caudal notochord but died by 10.5 dpc with a

truncated notochord at the forelimb level (Weinstein, et al. 1994; Ang and Rossant,

1994; Beddington, et al. 1992; Rashbass, et al. 1994). During normal development, T-

brachyury is expressed in the notochord and its expression is maintained in notochord-

derived nucleus pulposus cells postnatally (Dahia, et al. 2012). The role of T-brachyury

in postnatal intervertebral discs is unknown. Gene duplications in T-brachyury have

been linked to risk for familial chordomas in humans (Yang, et al. 2009).

Noto

The homeobox transcription factor Noto is expressed in the node and notochord

between 7.5dpc and 12.5 dpc. (Zizic-Mitrecic, et al. 2010; Abdelkhalek, et al. 2004). In

Page 23: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

23

mice, Noto is required for tail bud morphogenesis and Noto deletion resulted in

notochord truncation at the tail level (Zizic-Mitrecic, et al. 2010; Abdelkhalek, et al.

2004). Mutations in Noto underlie the floating head (flh) phenotype in zebrafish, which is

characterized by complete absence of a notochord (Talbot, et al. 1995).

In mice Noto is thought to be an important regulator of axial mesoderm

(notochord) identity (McCann, et al. 2012; Zizic-Mitrecic, et al. 2010; Abdelkhalek, et al.

2004; Yamanaka, et al. 2007). In mice where notochord cells had been GFP-labeled,

Noto inactivation resulted in ectopic localization of GFP-labeled cells in the paraxial

mesoderm rather than at the midline, as in control mice. The authors‟ interpretation was

that in the absence of Noto, axial mesoderm (notochord cells) had acquired somitic

mesoderm cell fate and were thus found in the paraxial mesoderm (McCann, et al.

2012). Additional studies, perhaps by probing for Shh, T-brachyury, or Foxa2

expression in these ectopic GFP-positive cells, would enhance our understanding about

the role of Noto in axial mesoderm maintenance. Absence of Shh, T-brachyury, and

Foxa2 in ectopic GFP-positive cells would conclusively demonstrate that a true cell fate

re-specification of notochord cells had occurred in Noto null mutants.

Genes Required For Development Of The Annulus Fibrosis

Following somitogenesis, cells in the ventral somite undergo an epithelial to

mesenchymal transition to form the sclerotome (Hay, 2005; Yusuf and Brand-Saberi,

2006; Christ and Scaal, 2008). In addition to producing the ossified vertebral

components in the axial skeleton, progenitors contained in the embryonic sclerotome

give rise to the cartilaginous annuli fibrosi of each intervertebral disc.

Page 24: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

24

Pax1 and Pax9

One of the earliest markers of sclerotome specification is Pax1, which requires

Shh and Noggin expression in the ventral midline for its induction (Wallin, et al. 1994;

McMahon, et al. 1998; Koseki, et al. 1993). Soon after Pax1 induction, Pax9 expression

is activated in the sclerotome (Neubuser, et al. 1995). Pax1 and Pax9 have overlapping

and spatially distinct expression patterns in the sclerotome (Neubuser, et al. 1995).

Following induction of both Pax1 and Pax9, Pax1 expression becomes enriched in the

ventral sclerotome while Pax9 expression becomes relegated to the dorsal sclerotome

(Neubuser, et al. 1995). In the absence of Pax1, the Pax9 expression domain becomes

upregulated and spatially expands in the sclerotome (Peters, et al. 1999).

The role of Pax1 in vertebral column formation is well documented (Table 1-1).

The characterization of multiple Pax1 mutants have illustrated its importance in

sclerotome differentiation. In the Undulated short-tail (Pax1un-s) mutant, the complete

absence of the Pax1 locus resulted in persistent notochord, loss of all vertebrae and

intervertebral discs, and severe malformations of the ribs and sternum (Wallin, et al.

1994). The Pax1un-s mouse, which was generated by gene targeting to produce a

defined Pax1 null allele, had similar deficits in the vertebral column including scoliosis,

split vertebrae, abnormalities in the lateral vertebral processes, and loss of

intervertebral disc structures (Wilm, et al. 1998). Pax1 null mice had a mild but fully

penetrant phenotype in the vertebral column as well as deficits in the sternum and

scapula (Wilm, et al. 1998). Additionally, 88% of Pax1 heterozygotes were observed to

have some form of defect in sclerotome-derived structures (Wilm, et al. 1998). Despite

these deficits, Pax1 null mutants were viable (Wilm, et al. 1998).

Page 25: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

25

Pax1 and Pax9 have partially redundant roles in sclerotome differentiation with

Pax1 appearing to be more important for vertebral column formation (Peters, et al.

1999). Deletion of Pax9 did not result in major deficits in the vertebral column (Peters, et

al. 1999). While removal of Pax1 resulted in some malformations in the vertebrae and

intervertebral discs, removal of both Pax1 and Pax9 resulted in major deficiencies in the

vertebral column including completely absent vertebral bodies and intervertebral discs

(Peters, et al. 1999). Additionally, Pax1;Pax9 null mutants had deficits in the proximal

ribs (Peters, et al. 1999). Interestingly, while Pax1 and Pax9 removal resulted in

agenesis of the ventral vertebrae (vertebral bodies), the lateral vertebrae formed in the

absence of Pax1 and Pax9. Together, these results demonstrated that the ventral and

lateral vertebral processes have distinct genetic requirements for development. Studies

have shown that the ventromedial sclerotome region gives rise to the vertebral bodies

and annuli fibrosi while the dorsomedial and ventrolateral regions form the spinous

processes and neural arches that form the dorsal enclosure for the spinal cord

(Bruggeman, et al. 2012; Christ, et al. 2004; Wallin, et al. 1994; Leitges, et al. 2000;

Adham, et al. 2005).

Mutations in genes known to function upstream of Pax1 and Pax9 also impair

sclerotome differentiation. Deletion of Pbx1/Pbx2, which regulate Pax1 and Pax9 in the

sclerotome, resulted in vertebral defects (Capellini, et al. 2008). Pbx1 and Pbx2 are

expressed in the notochord and surrounding mesenchyme at E12.5 and are important

for chondrocyte differentiation and axial skeleton patterning (Capellini, et al. 2008;

Selleri, et al. 2001). On a Pbx1 null background, removal of one Pbx2 produced thinner

transverse processes and flattened vertebrae at e13.5 (Capellini, et al. 2008). Impaired

Page 26: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

26

Shh signaling can also impact sclerotome maturation as hedgehog signaling is required

for Pax1 induction in the sclerotome (McMahon, et al. 1998; Koseki, et al. 1993; Mo, et

al. 1997; Buttitta, et al. 2003).

Bapx1

After the tendon-producing syndetome splits off from the sclerotome, sustained

expression of Pax genes in the remaining sclerotome ensures the continued

specification of chondroprogenitors. Bapx1 is an important mediator of chondrogenic

differentiation that functions downstream of Pax1 and Pax9 in the sclerotome (Rodrigo,

et al. 2003; Tribioli, et al. 1999). Targeted deletion of Bapx1 resulted in impaired

sclerotome differentiation, dysplastic vertebral columns at birth, and perinatal lethality

(Akazawa, et al. 2000). Bapx1 null mutants had unossified vertebrae with a residual

notochord that failed differentiate into nuclei pulposi (Akazawa, et al. 2000).

As differentiation proceeds, Pax1 downregulation in the sclerotome is

accompanied by up-regulated expression in Sox9 (Takimoto, et al. 2013). Sox9 is a

critical regulator of chondrogenic differentiation. Following Pax1 downregulation, Bapx1

expression is maintained in sclerotome-derivatives by Sox9 to suppress osteogenic

differentiation in favor of a chondrogenic pathway (Yamashita, et al. 2009).

Sox Family of Genes

The intervertebral discs are anchored to adjacent vertebrae by the cartilaginous

endplates, which are structurally contiguous with the fibrocartilaginous annulus fibrosus.

Sox9, which is regarded as the master regulator of chondrogenesis, is a SRY-related

HMG box containing transcription factor expressed in the notochord, sclerotome, and

their derivatives (Bi, et al. 1999; Sugimoto, et al. 2013; Barrionuevo, et al. 2006). Sox9

Page 27: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

27

and its homologs Sox5 and Sox6 play critical roles in development and maintenance of

cartilage in the vertebral column. Sox9 is expressed in the notochord and is required for

its maintenance and differentiation (Barrionuevo, et al. 2006).

Sox9 is a critical regulator of many of the steps involved in cartilage

differentiation, including the earliest stages of mesenchymal condensation that occurs in

the somitic mesoderm (Bi, et al. 1999; Akiyama, et al. 2002). Mutations in Sox9 underlie

chondrodysplasias in animals and humans, and homozygous inactivation of Sox9 is

known to result in complete absence of cartilage while Sox9 mis-expression induces

ectopic cartilage formation (Akiyama, et al. 2002; Hargus, et al. 2008; Henry, et al.

2012; Bi, et al. 1999; Takimoto, et al. 2012). In the absence of Sox9, loose

undifferentiated sclerotome fails to condense to form chondrogenic nodules, which

usually occurs between 11.5 and 12.5 dpc in mice (Bi, et al. 1999). In chimeric mice

generated with Sox9 null embryonic stem cells, Sox9 null cells failed to express

chondrogenic markers, were excluded from the vertebral cartilage, and were instead

relegated to the supporting mesenchyme (Bi, et al. 1999).

Sox9 is also a critical regulator of postnatal cartilage maintenance. Conditional

inactivation of Sox9 in postnatal mice resulted in arrested development of growth plate

chondrocytes, proteoglycan loss, and down-regulated expression of many cartilage-

specific genes (Henry, et al. 2012). Sox9 inactivation resulted in kyphosis of the

vertebral column and compressed intervertebral discs, mimicking degenerative changes

seen in humans (Henry, et al. 2012).

Sox9 is also critical for its role in transcriptionally activating Sox5 and Sox6 in the

cartilage (Akiyama, et al. 2002). Sox5 and Sox6 cooperate to stabilize Sox9 binding at

Page 28: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

28

its target regulatory sequences, thus enhancing transcription of chondrogenic mediators

(Lefebvre, et al. 1998). Sox9, Sox5, and Sox6 have overlapping and distinct roles in

chondrogenic differentiation (Ikeda, et al. 2005; Smits, et al. 2001; Smits, et al. 2003; de

Crombrugghe, et al. 2000). Like Sox9, Sox5 and Sox6 are expressed in the notochord,

sclerotome, and intervertebral discs. Mice that lacked both Sox5 and Sox6 lacked

notochordal sheaths and showed signs of aberrant cell death in the notochord. As a

result, Sox5;Sox6 null embryos developed vertebral columns that lacked nuclei pulposi

(Smits, et al. 2003).

Scleraxis And TGFβ Signaling

Tendons and ligaments are force-transmitting connective tissues that adjoin bone

to muscle and bone to bone, respectively. Within the vertebral column, tendons and

ligaments anchor the vertebrae to each other and to the musculature. Scleraxis (Scx) is

expressed in embryonic and postnatal tendons and ligaments, and is presently the

earliest known marker of tendon/ligament specification in the somitic mesoderm

(Cserjesi, et al. 1995; Brent, et al. 2003). Scx regulates expression of downstream

tenogenic genes including type I collagen, fibromodulin, decorin, tenomodulin, and

tenascin C (Takimoto, et al. 2012; Shukunami, et al. 2006; Murchison, et al. 2007;

Schneider, et al. 2011). In addition, Scx is expressed in the annulus fibrosis, although its

role in the annulus fibrosus is unknown. The annuli fibrosi develop normally in Scx null

mice (Murchison, et al. 2007).

Mice with targeted deletion of Scx were viable but had acute dorsal flexure of the

forelimbs at birth, indicating defects in the limb tendons (Murchison, et al. 2007). In the

vertebral column, Scx loss affected only a subset of tendons, and did not affect the

Page 29: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

29

annuli fibrosi (Murchison, et al. 2007). Although Scx null mice lacked most

tendons/ligaments, including the long tendons that anchor the tail vertebrae to the base

of the tail, some short tendons remained unaffected despite Scx loss (Murchison, et al.

2007). Thus, although Scx is presently the earliest known marker of syndetome

specification in the somitic mesoderm, another unidentified regulator is likely

responsible for activating the tenogenic developmental program in the somitic

mesoderm, upstream of Scx.

Another important mediator of tenogenic differentiation includes the TGFβ

signaling pathway. TGFβ2 and TGFβ3 are expressed in cartilage and tendon

progenitors early in development, and TGFβ signaling is known to regulate the

expression of many tendon-specific genes, including Scleraxis, Tenomodulin, and

Mohawk (Schneider, et al. 2011; Liu, et al. 2015; Pryce, et al. 2009). Deletions and

mutations in TGFβ or its signaling constituents affect vertebral column development and

intervertebral disc morphogenesis (Pryce, et al. 2009; Baffi, et al. 2004; Baffi, et al.

2006). In mice, TGFβ signaling disruption resulted in absent trunk tendons and

ligaments, Scx downregulation, and small, abnormally shaped vertebrae and reduced or

absent intervertebral discs (Pryce, et al. 2009; Baffi, et al. 2004).

Conclusions

A balanced synergism between the vertebrae, discs, and surrounding

tendons/ligaments and musculature contributes to the fluid and painless mobility of the

axial skeleton. The timely orchestration of activating and antagonizing signals between

the notochord, somites, neural tube, and surface ectoderm is critical for the formation of

the vertebral column. Any perturbation to this complex interplay of signals is liable to

Page 30: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

30

produce defects with functional consequences. In adulthood, the avascular, hypoxic

environment of the intervertebral discs limits its regenerative potential, rendering the

discs vulnerable to age-related degeneration.

Discogenic back pain can be a chronic and debilitating condition. The lifetime

prevalence for back pain is greater than 80% (Andersson, 1999; Katz, 2006). The

current standard of care for back pain largely focuses on pain management through

analgesics and exercise therapy rather than the reversal of underlying pathology. Even

more aggressive treatments, including disc arthroplasty and lumbar spinal fusion, fail to

address the underlying pathology and are limited in efficacy. A more thorough

appreciation of the genetic mechanisms underlying vertebral column formation, and

degeneration, will be requisite for the development of more effective therapies for the

treatment of disc degeneration and back pain.

Page 31: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

31

Table 1-1. Genes Required For Murine Vertebral Column Formation

Gene Mutation Phenotype, References

Foxa1 Null No vertebral column phenotype (Maier, et al. 2013)

Foxa2 Null Early embryonic lethality; no notochord forms, precluding vertebral column formation (Ang and Rossant, 1994)

Foxa2 Conditional inactivation in the notochord

Foxa2 inactivation at e7.5 using ShhCreERT2 produces no vertebral column phenotype (Maier, et al. 2013)

Foxa1/Foxa2 Foxa1 null; Foxa2 conditional inactivation in the notochord

Foxa2 inactivation at e7.5 using ShhCreERT2 on a Foxa1 null background results in abnormal vertebral columns with small, misshapen nuclei pulposi (Maier, et al. 2013)

Gdf-5 Null No vertebral column phenotype (Maier and Harfe, 2011)

Gli2 Gli2zfd Reduced vertebral ossification; reduced or absent discs (Mo, et al. 1997)

Gli3 Gli3XtJ Mild neural arch defects; Gli3XtJ/XtJ phenotype less severe than that in Gli2zfd (Hui and Joyner, 1993; Mo, et al. 1997)

Gli2/Gli3 Gli2zfd; Gli3XtJ

Gli2zfd/zfd; Gli3XtJ/XtJ mice are embryonic lethal by E10.5. Gli2zfd/zfd; Gli3XtJ/+ mice have severe sternal defects and vertebral columns with malformed discs and vertebrae (Mo, et al. 1997)

HIF-1α Conditional inactivation in the notochord

HIF-1α inactivation using Foxa2cre resulted in smaller nuclei pulposi at birth. Postnatally, notochord-derived nuclei pulposi cells undergo massive cell death and are replaced by chondrocyte-like cells of non-notochordal origin (Merceron, et al. 2014)

Nkx3.1 Null No vertebral column phenotype (Tanaka, et al. 2000; Herbrand, et al. 2002)

Page 32: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

32

Table 1-1. Continued

Gene Mutation Phenotype, References

Nkx3.2 (Bapx1) Null Mild phenotype; ventromedial vertebral elements (vertebral bodies) affected; lack of ossification; abnormal discs (Akazawa, et al. 2000; Lettice, et al. 2001)

Nkx3.1/Nkx3.2 Nkx3.1 null; Nkx3.2 null

Embryonic lethal by e17.5. Reduced ossification and defects in dorsal vertebral elements (transverse processes); anterior vertebral column more severely affected (Herbrand, et al. 2002)

Noggin Null Perinatal lethality; aberrant presence of non-notochordal cells in the notochord; loss of caudal sclerotome and vertebrae; kinked and shortened tails (McMahon, et al. 1998; Tylzanowski, et al. 2006; Li, et al. 2007)

Noto88, 89, 133 Nottc

NoteGFP Reduced viability. Truncation in tail notochord results in loss of tail vertebrae (Theiler, 1959; Abdelkhalek, et al. 2004; Zizic-Mitrecic, et al. 2010)

Pax1 Pax1un Pax1un-s Pax1un-ex Pax1un-i Pax1null

Vertebral column phenotypes of variable severity. Defects include shortened or kinky tails; scoliosis; loss of vertebrae and intervertebral discs. Pax1un-s produces the most severe phenotype and is regarded as semi-dominant as heterozygotes also exhibit vertebral column defects (Wallin, et al. 1994; Wilm, et al. 1998; Adham, et al. 2005)

Pax9 Null No vertebral column phenotype (Peters, et al. 1998; Peters, et al. 1999)

Pax1/Pax9 Pax1 null; Pax9 null Complete loss of ventral vertebral elements (vertebral bodies) and intervertebral discs (Peters, et al. 1999)

Shh Null Cyclopia; complete agenesis of the vertebral column; rudimentary ribs (Chiang, et al. 1996)

Shh Conditional inactivation in the notochord

Inactivation at E7.5 using ShhCreERT2 resulted in complete absence of discs and vertebrae. Shh inactivation at e11.5 produced a normal vertebral column (Choi and Harfe, 2011)

Page 33: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

33

Table 1-1. Continued

Gene Mutation Phenotype, References

Sox5 Null Perinatal lethality; vertebral column formed with mild vertebral ossification delay (Smits, et al. 2001)

Sox6 Null Perinatal lethality; vertebral column formed with mild vertebral ossification delay (Smits, et al. 2001)

Sox5/6 Sox5 null; Sox6 null Embryonic lethal at E16.5 with severe defects in the vertebral column including chondrodysplasia; cartilage deficits; small and malformed or absent nuclei pulposi (Smits, et al. 2001; Smits, et al. 2003)

T-brachyury Null Embryonic lethal by E10.5; loss of trunk and tail level notochord precludes vertebral column formation (Gruneberg, 1958; Wilkinson, et al. 1990; Hermann, 1992; Beddington, et al. 1992)

Tbx18 Null Perinatal lethality with shortened axial skeletons. Defects included kinks in the thoracic vertebral column; malformed, flattened discs and vertebral bodies; expanded pedicles and transverse processes (Bussen, et al. 2004)

Tgfbr2 Conditional inactivation in cartilage

Inactivation in chondrocytes using Col2a-cre produced vertebral column defects including missing or malformed discs; neural arch and transverse process defects; loss of spindle cell shape in the annulus fibrosus (Baffi, et al. 2004; Baffi, et al. 2006)

Uncx4.1 Null Perinatal lethality with defects in the lateral vertebral elements (neural arches; pedicles; transverse process); kink in the thoracic vertebral column (Leitges, et al. 2000)

Page 34: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

34

Figure 1-1. Anatomy of the Vertebral Column and Intervertebral Disc. A) The fibrocartilagenous intervertebral discs (IVD) separate and cushion the vertebral bodies (VB) along the length of the vertebral column. B) Each intervertebral disc is comprised of a central, hydrated core called the nucleus pulposus, which is encapsulated by the annulus fibrosus. Each disc is adjoined to the vertebrae by the cartilaginous endplates, which serve as the interface through which nutrient diffusion to the discs occurs.

Figure 1-2. Vertebral Column Development In Mice. The vertebral column develops rostrocaudally with overt chondrogenesis, defined as the accumulation of cartilage matrix molecules in the sclerotome, beginning at 12.5dpc in mice. These chondrogenic condensations can be observed as Alcian blue staining in the vertebral column surrounding the notochord, N, at 12.5 dpc. As development progresses, these chondrogenic condensations exert an inward pressure against the notochordal sheath at repeating intervals. These repeating intervals of compression promote the notochord to disc transition, which can be observed by 14.5 dpc. By 15.5 dpc in the rostral embryo, the discs have formed as distinct, repeating units in the vertebral column. By P0 the discs have acquired their characteristic flattened form. Postnatal vertebral column

Page 35: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

35

development entails growth and elongation of the vertebral bones by endochondral ossification and proteoglycan accumulation and eventual cell senescence in the discs.

Figure 1-3. Postnatal Intervertebral Disc Development. By birth, the nucleus pulposus is distinct from the annulus fibrosus, which encapsulates the water-rich disc core. Postnatally as the vertebral column grows in size, the discs enlarge largely by extracellular matrix deposition. By three weeks old, proliferation has largely ceased in the intervertebral discs (Dahia 2006). Postnatal disc development is characterized by deposition of additional annulus fibrosus layers and the emergence of the cartilaginous endplates (EP) as a distinct structure at the disc-vertebra interface. As can be seen in postnatal day 21 (P21) discs, the endplates are contiguous with the inner annuli fibrosi. At P21 the vertebrae continue to grow by endochondral ossification, whereby vertebral growth plate chondrocytes (VG) undergo a sequential differentiation process to form new bone. Adult discs, by comparison, are much less cellular than perinatal discs and P21 discs. As can be seen in discs from 3 month old mice, the discs are much less cellular and rich in extracellular proteoglycans. All images are of frontally sectioned, Alcian blue and Picrosirius red stained discs from wildtype mice.

Page 36: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

36

CHAPTER 2 ROLE OF ROBO GENES DURING MURINE VERTEBRAL COLUMN DEVELOPMENT

Introduction

The vertebral column is comprised of ossified vertebrae which are adjoined by

the cartilaginous intervertebral discs. In adult animals, the intervertebral discs can be

characterized as having three functionally distinct domains. Each intervertebral disc is

comprised of a gel-like inner core called the nucleus pulposus (Choi and Harfe, 2011).

The nuclei pulposi are encapsulated by the fibrocartilagenous annuli fibrosi, which can

be further divided/specified/distinguished as inner and outer annuli fibrosi based on

extracellular matrix (ECM) composition (Cortes, et al., 2012). Contiguous with the annuli

fibrosi are the cartilaginous endplates, which serve as the interface between the discs

and the vertebral growth plates (Maladrino, et al., 2014). In adulthood, the discs are

aneural and avascular, leading to a hypoxic environment for nucleus pulposus cells

(Richardson, et al. 2012). Additionally, as growth occurs and the vertebral column

enlarges, nutrient supply to the discs becomes limited to diffusion through the

endplates, which can become calcified and pathologic with age. Altogether, these

changes are thought to contribute to intervertebral disc pathology and back pain in

humans (Tolofari, et al., 2010; Smith, et al., 2011).

The intervertebral discs form from the axial mesoderm (notochord) and paraxial

mesoderm (somites) while the vertebrae form from the paraxial mesoderm (Christ and

Scaal, 2008; Christ, et al., 2004; Choi and Harfe, 2011). Within the paraxial mesoderm,

progenitor cells in the sclerotome give rise to the vertebrae and fibrocartilagenous

components of the discs (endplates and annuli fibrosi) (Bruggeman, et al., 2012; Christ

Page 37: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

37

and Scaal, 2008). The nuclei pulposi are known to derive exclusively from the

embryonic notochord (Choi and Harfe, 2011; McCann, et al. 2012).

During vertebral column morphogenesis, cells in the sclerotome differentiate and

form chondrogenic condensations. In mice overt chondrogenesis, defined as the

accumulation of Col2a1 in these condensed regions, beings at approximately 12.5 dpc

in the rostral embryo. As these mesenchymal nodules condense against the

notochordal sheath, notochordal cells are pushed into nucleus pulposus anlagen,

resulting in the formation of vertebrae and intervertebral discs in tandem (Choi and

Harfe, 2011).

Robo proteins are transmembrane cell surface receptors which belong to the

immunoglobulin class of cell adhesion molecules (Blockus and Chedotal, 2016). Four

Robo receptors have been identified and characterized in mice. Robo receptors have

extracellular domains consisting of three fibronectin type III domains and five Ig-like

domains, which interact with secreted glyocoprotein ligands (Hohenester, 2008). The

primary ligands for Robo receptors are Slit proteins (Hohenester, 2008). Heparin

sulfates serve as co-receptors to stabilize Robo-Slit interactions. Three Slit genes have

been identified in mice. Slit proteins undergo proteolytic processing to produce cleavage

fragments that have unique functional properties (Ordan, et al., 2015). Robo receptors

also have conserved intracellular domains that are important for transducing Slit-Robo

signals to intracellular proteins (Ypsilanti, et al., 2010). These conserved domains

interact with cytoplasmic adaptor proteins, including srGAP proteins, or Slit-Robo

GTPase activating proteins, which mediate the effects of Slit-Robo signaling (Ypsilanti,

et al., 2010).

Page 38: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

38

The term „promiscuous binding‟ has been used to describe Slit-Robo interactions,

reflecting the ability of any Slit protein to bind to any of the four Robo receptors

(Ypsilanti, et al., 2010). Additionally, Robo receptors are predicted to interact with other

proteins belonging to the LRR family of proteins, which includes the Slit proteins (Howitt,

et al., 2004). Examples of other small LRR proteins include fibromodulin, decorin, and

biglycan, which are expressed in cartilaginous tissues including the intervertebral discs

(Chen, et al., 2015).

In vitro and in vivo studies have shown that Slit-Robo interactions mediate a

chemorepulsive response in Robo-expressing cells (Domyan, et al., 2014; Kim, et al.,

2015). Unlike other signaling pairs like Ephrin-Eph receptor interactions, which result in

bi-directional signaling, Slit-Robo interactions are not known to elicit a response in Slit-

expressing cells. Thus, Slit-Robo signaling is unidirectional. Additionally, Slit-Robo

signaling is known to stimulate changes in cell polarity, adhesion, and migration by

modulating the localization of microtubule organizing centers (MTOC). These changes

are mediated through modulation of cytoskeletal dynamics in a RhoA-dependent

manner (Ypsilanti, et al., 2010).

A role for Robo and Slit in chondrocyte differentiation and function can be

inferred from reports of Robo and Slit expression in chondrocytes. In chicks and rats, for

example, Robo and Slit mRNA are expressed by limb bud chondrocytes in the

perichondrium and appendicular growth plates (Noel, et al., 1998; Holmes and

Niswander, 2001). In rat tibial bones, for example, Robo genes are expressed in the

growth plates (Noel, et al., 1998). Additionally, Robo mRNA expression can be induced

Page 39: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

39

by mechanical loading, suggesting a role for Robo receptors in cartilage and bone

remodeling (Noel, et al., 1998)

At present, few studies have investigated the role of Robo and Slit genes in the

sclerotome or its derivatives. We found that in mice, Robo1 and Robo2 were expressed

in the sclerotome and had complementary expression patterns to Slit mRNA in the

notochord. In the absence of Robo1 and Robo2, proliferation in the sclerotome was

impaired and resulted in malformed intervertebral discs. Additionally, Robo1 and Robo2

loss in the sclerotome resulted in deficits in the distal ribs with accompanying defects in

the sternum.

Results

Complementary Robo and Slit Expression Patterns in the Vertebral Column Robo Expression

To determine where and when Robo and Slit are expressed during formation of

the vertebral column we used RNA in situ hybridization, antibodies, and Xgal staining.

LacZ knockin into the Robo1 or Robo2 loci was used to generate the Robo1 and Robo2

null alleles. Robo expression was tracked by performing Xgal staining in Robo1+/-

;Robo2+/- mice (also referred to as Robo1;Robo2 heterozygotes in this manuscript). For

Xgal staining, embryos were whole mount stained and then sectioned for imaging by

bright field microscopy. For RNA in situ hybridization, probe templates for Robo1 and

Robo2 were generously provided by Dr. Xin Sun.

Robo expression in the mouse neural tube has been reported previously

(Camurri, Sundaresan, et al, 2004). To confirm specificity of our Robo probes, wildtype

10.5dpc embryos were used for in situ hybridizations. Robo2 mRNA was identified in

Page 40: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

40

the neural tube by RNA in situ hybridization (Fig. 2-1A). To confirm these results, stage-

matched Robo1;Robo2 heterozygous embryos were Xgal stained. Robo expression in

the neural tube at 10.5 dpc was confirmed by Xgal staining (Fig. 2-1C). Similarly, Robo2

in situ hybridization and Xgal staining yielded congruent results for Robo expression in

the vertebral column at 12.5 dpc (Fig. 2-1B and Fig. 2-1D). Thus, Xgal staining is a

reliable method to show Robo1 and Robo2 gene expression in the vertebral column.

At 12.5dpc Robo expression was identified in the somitic mesoderm, both lateral

and ventral to the notochord (Fig. 2-2A-B). By RNA in situ hybridization, Robo1 and

Robo2 were detected in the somitic mesoderm of the vertebral column (Fig. 2-2C-D).

Notably, while Robo2 is robustly expressed at the midline, dorsal and ventral to the

notochord (Fig. 2-2D), Robo1 mRNA was not detected at the midline (not shown).

Instead, a repeating pattern of Robo1 expression was observed in the lateral vertebral

column mesenchyme, laterally adjacent to the notochord (Fig. 2-2C). At 14.5 dpc,

Robo1 and Robo2 were expressed in the presumptive annuli fibrosi, which

encapsulated the developing nuclei pulposi (Fig. 2-2E-F). At 18.5 dpc, Robo2 mRNA

was expressed in the spinal cord and in the vertebral column, albeit more diffusely than

at earlier stages (Fig. 2-2G).

Following whole mount Xgal staining of Robo1;Robo2 heterozygotes, a

microtome was used to generate 10um cross-sections of the vertebral column. Using

this method, Robo1 and Robo2 expression was observed in the vertebral trabeculae

(Fig. 2-2I) and in isolated annulus fibrosus cells (Fig. 2-2J) at P0. We did not detect

Robo expression in the nucleus pulposus at any time. To generate a more global

impression of Robo expression in the vertebral column, we used agarose-embedded,

Page 41: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

41

Xgal-stained tissues to generate 100um sections on a vibratome. Using this method

Robo1 and Robo2 expression was found to be enriched in the vertebral growth plates

(Fig. 2-2H).

Slit Expression

A number of studies have reported Slit expression in the embryonic floor plate

and notochord3. RNA in situ hybridization confirmed Slit1 and Slit2 expression in the

mouse floor plate and notochord at 10.5 dpc (Fig. 2-3A-B) and 12.5 dpc (Fig. 2-3D-E).

Slit3 mRNA was also detected in the floor plate at 10.5 dpc and 12.5 dpc (Fig. 2-3C and

Fig. 2-3F). At 15.5 dpc, Slit2 expression was maintained and enriched in the nuclei

pulposi (Fig. 2-3H). Slit1 and Slit3 were not expressed at appreciable levels in the nuclei

pulposi of wildtype 15.5 dpc embryos (Fig. 2-3G and Fig. 2-3I).

Robo and Slit maintained complementary expression patterns during vertebral

column morphogenesis. At 12.5 dpc, for example, Robo1 and Robo2 were expressed in

the somitic mesoderm lateral and ventral to the notochord, but were not expressed in

the notochord (Fig. 2-2A-B). By comparison, Slit2 mRNA was expressed in the

notochord but not in the surrounding mesenchyme (Fig. 2-3E). This complementarity

was maintained until at least 14.5 dpc, after initiation of the notochord to nucleus

pulposus transition. At 14.5 dpc Robo1 and Robo2 were expressed in the presumptive

annuli fibrosi (Fig. 2-2E-F) while in comparably aged embryos, Slit2 mRNA was

enriched in the nuclei pulposi anlagen (Fig. 2-3H). Thus, Robo and Slit exhibit

complementary expression patterns during vertebral column morphogenesis in mice.

Page 42: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

42

Intervertebral Disc Malformations

The Robo1 and Robo2 null alleles, which reside 1.1 cM apart on chromosome 16

in mice, were previously generated by LacZ knockin to investigate their role in kidney

development (Long, et al, 2004; Grieshammer, et al, 2004). Robo1;Robo2 null mice

were then generated by homologous recombination of the Robo1 and Robo2 null alleles

to produce linked mutant alleles (Domyan, et al, 2013). As described previously,

Robo1;Robo2 null mice die immediately after birth due to respiratory defects but are

comparable in size to control littermates at birth (Domyan et al, 2013).

Alcian blue and Picrosirius red were used to histologically examine the effects of

Robo1 and Robo2 removal vertebral column morphogenesis at P0. In control mice the

vertebral column was comprised of repeating units of ossified vertebrae separated by

characteristically flattened intervertebral discs. By comparison, the intervertebral discs

in Robo1;Robo2 null mice were bulbous in appearance and had reduced Alcian blue

staining (Fig. 2-4A).

Homozygous removal of Robo1 alone did not produce a defect in the

intervertebral discs (Fig. 2-4B). To assess the potential role of Slit proteins in mediating

the Robo-dependent phenotype, Slit1;Slit2 null mutants were examined. Intervertebral

discs from Slit1;Slit2 null mice appeared comparable to those from control littermates at

17.5 dpc (Fig. 2-4C). Together, these data suggest a functional redundancy between

Robo1 and Robo2 in intervertebral disc development and show that removal of Slit1 and

Slit2 does not recapitulate the Robo1;Robo2 null phenotype in the vertebral column.

To confirm that Robo1 and Robo2 loss in the somitic mesoderm was the cause

of the chondrogenic deficits and intervertebral disc malformations, Robo2 was

Page 43: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

43

conditionally inactivated on a Robo1 null background using a Robo2Floxed allele.

Inactivation of Robo2Floxed using ShhCre resulted in a normal vertebral column at P0

(Fig. 2-5C). Inactivation of Robo2Floxed using Dermo1Cre partially recapitulated the

Robo1;Robo2 null phenotype (Fig. 2-5B). Compared to control littermates, Robo1-/-

;Robo2F/F;Dermo1Cre mice had reduced Alcian blue staining, suggesting a deficit in

cartilage formation or maintenance following formation of the vertebrae and discs (Fig.

2-5B).

The Notochord to Nucleus Pulposus Transition

One of the hallmarks of disc degeneration is fibrogenesis and eventual loss of

„notochord-like‟ cells in the nucleus pulposus (Lv, Peng, et al, 2016). In mice, for

example, removal of HIF1a in the discs resulted in complete loss of normal nucleus

pulposus cells by apoptosis, and eventual replacement of those cells by fibroblast-like

cells of unknown origin. These changes were accompanied by deficits in vertebral

column mechanical function. Moreover, one of the characteristics of aging discs in

humans is the gradual loss or effacement of the nucleus pulposus-annulus fibrosus

boundary (Urban and Roberts, 2003). Thus, normal disc biology entails strict

segregation of nucleus pulposus cells from annulus fibrosus cells during development

and loss of this separation is associated with disc degeneration.

Fate mapping studies in mice have shown that adult nuclei pulposi are comprised

of cells exclusively of notochordal origin. Equally, cells of non-notochordal origin are

excluded from (or undergo apoptosis) the inner nucleus pulposus in adult mice,

although the mechanism underlying this phenomenon is unknown.

Page 44: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

44

In other developing tissues, Robo-Slit signaling is essential for modulating cell

migration and organ positioning via a chemorepulsive mechanism. In the developing

central nervous system (CNS), for example, Robo-expressing commissural axons are

repelled from the midline by responding to chemorepulsive Slit cues secreted from the

neural tube floor plate. In the CNS, Slit-mediated repulsion of Robo-expressing

commissural axons prevents aberrant re-crossing of axons across the midline. In the

developing foregut, Robo-expressing foregut mesenchyme responds to secreted Slit

ligands from the body wall to ensure that the developing gut becomes properly

positioned in the abdominal cavity.

The observation that Slit and Robo have complementary expression patterns

during vertebral column formation (Fig. 2-2 and Fig. 2-3) suggested a potential role for

Robo-Slit signaling in mediating the notochord to nucleus pulposus transition in mice.

The expression patterns of Slit and Robo and their known functions in other tissues

suggested a model in which Robo-expressing cells were repelled by Slit expression in

the notochord and nuclei pulposi (Fig. 2-6A).

In this model, Slit-mediated repulsion of Robo-expressing cells would prevent

mixing of nucleus pulposus and annulus fibrosus cells during vertebral column

morphogenesis (Fig. 2-6A). Based on this model, removal of Robo1 and Robo2 would

impair the inhibitory effect of Slit expression in the nucleus pulposus on annulus fibrosus

cells, resulting in a mixed population of cells in the nuclei pulposi at birth (Fig. 2-6A).

Specifically, this mixture of cells would include cells of notochordal origin and cells of

non-notochordal (sclerotomal) origin. To address this possibility, and to determine the

Page 45: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

45

developmental origin of nucleus pulposus cells at birth, the Rosa-mT/mG dual

fluorescence reporter was used.

The Rosa-mT/mG reporter was generated by insertion of the mT/mG construct at

the constitutively active Rosa26 locus (Muzumdar, et al., 2007). In the absence of Cre,

mice that carry the Rosa-mT/mG allele have membrane-targeted tdTomato

fluorescence in all cells of the intervertebral discs. In the presence of Cre, excision of

the tdTomato cassette via flanking loxP sites results in expression of membrane-

targeted GFP in lieu of membrane-targeted tdTomato. Lineage tracing analyses by Cre-

mediated homologous recombination results in the „genetic marking‟ of all Cre-

expressing cells as well as all daughter cells derived from the Cre-expressing cell

population. In these experiments activation of GFP is permanent such that all daughter

cells are irreversibly GFP-marked, even in the absence of active Cre expression.

Shh is expressed in the nascent notochord and continues to be expressed as the

notochord begins to transition into the nuclei pulposi (Choi and Harfe, 2011). A ShhCre

allele was used to genetically mark the notochord and its derivatives, the nucleus

pulposus. Previous studies have reported a small number of notochord-derived cells

scattered throughout the vertebral column outside of the nuclei pulposi. These cells are

“notochordal remnants” and are potentially the cell population that gives rise to

chordomas. The majority of notochord cells form nucleus pulposus cells with only a few

notochord-derived cells residing outside the nuclei pulposi. Additionally, under normal

circumstances, sclerotome-derived cells are strictly excluded from the nucleus pulposus

(Harfe and Choi, 2011).

Page 46: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

46

To assess if Robo-Slit signaling was responsible for maintaining separation of

nucleus pulposus (NP) and annulus fibrosus (AF) cells in the discs, Robo1-/-; Robo2-/-;

ShhCre; Rosa-mT/mG mice were generated and harvested at 18.5 dpc for lineage

tracing analyses (Fig. 2-6B). In the absence of Robo1 and Robo2, the notochord to

nucleus pulposus transition occurred normally (Fig. 2-6C). Notochord-derived cells were

found largely within the nuclei pulposi of Robo1;Robo2 null mice (Fig. 2-6C). Both

controls and Robo1;Robo2 null mice had isolated GFP+ cells in the vertebral column

outside of the nuclei pulposi (not shown). In addition, non-notochord-derived tdTomato+

cells were completely excluded from the nuclei pulposi in both control and

Robo1;Robo2 null mutants (Fig. 2-6C). These data indicate that in the absence of

Robo1 and Robo2, the notochord to nucleus pulposus transition occurs normally.

Using the ShhCre allele, it is impossible to determine if an annulus fibrosus

progenitor cell aberrantly migrated into the nucleus pulposus and began to express Shh

as a consequence of its ectopic location in the notochord. If this occurred, the ectopic

cell would express Shh and activate GFP, making it indistinguishable from Shh-

expressing notochord-derived cells located in the nucleus pulposus. Notochordal cell-

conditioned media has been shown to modulate annulus fibrosus cell gene expression

in vitro. In addition, pluripotent cells are known to acquire notochord-like cell phenotypes

when cultured on nucleus pulposus tissue matrix. These data suggest that other cell

types, if located in the nucleus pulposus, could assume a nucleus pulposus cell-like

expression profile.

To address the limitations of using an approach that could not discriminate

nucleus pulposus cells from cells that had inappropriately migrated and assumed a

Page 47: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

47

nucleus pulposus-like expression profile, the lineage tracing analyses were repeated

using the Tamoxifen-inducible ShhCreERT2 allele. In ShhCreERT2 mice, Cre remains

cytoplasmic and inactive in the absence of Tamoxifen. Upon Tamoxifen treatment, Cre

translocates into the nucleus and mediates homologous recombination of DNA flanked

by loxP sites.

Previous data indicates that Tamoxifen administered by oral gavage genetically

marks cells over a 24 hour window in most tissues including the discs (Harfe and Choi,

2011). Using a single dose of Tamoxifen administered to pregnant dams at 10.5 dpc

specifically marks Shh-expressing cells between 10.5 dpc and 11.5 dpc. In the

developing vertebral column, these experiments labeled all cells of the notochord (Harfe

Choi, 2011). By the time the nucleus pulposus formed at ~13.5 dpc, any non-

notochordal cells located in the nucleus pulposus that expressed Shh would not be

marked (ie. GFP-positive). For example, ectopic annulus fibrosus or end plate cells that

inappropriately migrated into the notochord after 11.5 dpc would not be expected to be

GFP-positive even if they were located in the nucleus pulposus and expressed Shh.

Using the above strategy, no mixing of cells in the nucleus pulposus was

observed in Robo1;Robo2 null mutants. All nucleus pulposus cells were GFP-positive

and all tdTomato-positive cells were strictly excluded from the nucleus pulposus region

in both control and Robo1;Robo2 null animals (Fig. 2-7B). A notable difference between

control and Robo1;Robo2 null mice was that a greater number of GFP+ cells were

observed outside the nuclei pulposi in Robo1;Robo2 mutants (Fig. 2-7B). Although a

few isolated GFP+ cells were observed outside the nuclei pulposi in controls (not

Page 48: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

48

shown), Robo1;Robo2 null mice had a greater number of GFP+ cells in the annuli fibrosi

and vertebrae (Fig. 2-7B).

These data can be interpreted in at least two ways. First, it is possible that GFP+

cells located outside the nuclei pulposi in Robo1;Robo2 null mice are notochordal in

origin. It is possible that in the absence of Robo1 and Robo2, the notochord to disc

transition is affected and notochord-derived cells become ectopically scattered

throughout the vertebral column.

Alternatively, it is possible that the GFP+ cells outside the nuclei pulposi are

sclerotomal in origin. A Tamoxifen pulse at 10.5dpc would mark all cells expressing Shh

between 10.5 and 11.5dpc, including cells in the notochord. This would also mark

isolated progenitors in the sclerotome that may have transiently expressed Shh during

the same window. Shh expression and function in the sclerotome has not been reported

previously.

One way to resolve this uncertainty would be to co-label GFP+ cells from Rosa-

mT/mG mice with an anti-T-brachyury antibody. T-brachyury is expressed in the

notochord and its expression, as determined by T-brachyury protein

immunofluorescence, is maintained in the nucleus pulposus at P0. If GFP+ cells outside

the nuclei pulposi co-labeled with T-brachyury, it would suggest that these GFP+ cells

were notochord-derived.

Rib and Sternal Development

In addition to abnormalities in the vertebral column, deficits in the distal ribs and

sterna were observed in Robo1;Robo2 null mice. Rib cages from Robo1;Robo2 null

mice were smaller in comparison to those from control littermates (Fig. 2-8A). Closer

Page 49: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

49

examination revealed that in addition to having smaller rib cages, Robo1;Robo2 null

mice had deficits in the distal ribs and sterna (Fig. 2-8B). Specifically, compared to

controls which had attachment of the cartilaginous distal ribs at the sternum, (Fig. 2-8B,

a-b), the distal ribs in Robo1;Robo2 null mice failed to meet at the midline (Fig. 2-8B, b-

f). Additionally, the distal ribs showed reduced Alcian blue staining, suggesting a

decrease in chondrogenesis in the distal ribs (Fig. 2-8B, e-f). Moreover, while the

sterna in control animals was always completely fused at birth (Fig. 2-8B, a-b),

Robo1;Robo2 null mice had incomplete fusion of the sternal bars at the midline (Fig. 2-

8B, c-f). To quantify these deficits, sternal length was measured. At P0, Robo1;Robo2

null mice had a 30% reduction in sternal length compared to controls (Fig. 2-8C).

Proliferative Deficits in the Sclerotome

Based on observations that the ribs and sterna were reduced in size in

Robo1;Robo2 null mice, we speculated that Robo1;Robo2 null mice had a proliferative

deficit earlier in development. The ribs and sternum, as well as the vertebrae and annuli

fibrosi, are derived from the sclerotome.

To address the possibility that the ribs and sterna in Robo1;Robo2 null mice were

smaller due to proliferative differences in the sclerotome, an EdU-based assay was

used to assess in vivo proliferation at 13.5 dpc. EdU is a thymidine analogue that

becomes incorporated into newly synthesized DNA as cells undergo mitosis. Using a

fluorescent antibody to detect incorporated EdU, the effect of Robo1 and Robo2 loss on

proliferation was quantified.

Although Robo1 and Robo2 antibodies are commercially available, we were

unable to detect Robo using these antibodies, even in positive control brain tissues. In

Page 50: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

50

lieu of direct detection of Robo protein, an antibody against β-Galactosidase (bGal)

protein, which is produced from the lacZ transcript transcribed from the Robo1 and

Robo2 loci, was used. bGal immunofluorescence recapitulated the Robo expression

pattern as visualized by Xgal staining (Fig. 2-9A).

To assess differences in proliferation, we used Robo1;Robo2 null mutants and

Robo1+/-;Robo2+/- heterozygous control littermates. Following the manufacturer‟s

protocol for EdU detection (Fig. 2-9B, d), tissues were incubated with an anti-bGal

primary antibody, followed by incubation with a 488-conjugated secondary, to mark

Robo expression domains in the vertebral column (Fig. 2-9B, b).

Compared to control littermates, Robo1;Robo2 null embryos exhibited decreased

proliferation. Removal of Robo1 and Robo2 resulted in a greater than 50% reduction in

sclerotomal proliferation at 13.5dpc (Fig. 2-9C). Approximately 26% of cells in the

sclerotome were found to be proliferative in controls while only 11% of cells in the

sclerotome were proliferative in Robo1;Robo2 mutants (Fig. 2-9C, d). To determine if

the proliferative deficit was due to an overall decrease in mitotic index caused by Robo

loss, proliferation in the notochord, which does not express Robo1 or Robo2, was

quantified. No difference in proliferation was observed in the notochord (Fig. 2-9C, d).

Robo and Pax1 Expression in the Vertebral Column

Many of the molecular regulators of sclerotome induction and differentiation have

been identified, including proteins belonging to the Meox, Bapx, and Pax family of

transcription factors. One of the earliest markers of sclerotome specification in the

paraxial mesoderm is Pax1. Pax1 is required for sclerotome specification and

differentiation, and in its absence rib and vertebral defects occur. Midline-derived

Page 51: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

51

Hedgehog signals are required for Pax1 induction in the paraxial mesoderm. Once

activated, Pax1 directly regulates Bapx1 expression, which is required for subsequent

chondrogenic differentiation of sclerotome-derived tissues.

Pax1 mutants have been reported to have rib and vertebral defects which are

reminiscent of the deficits observed in P0 Robo1;Robo2 null mice (Fig. 2-8A).

Additionally, like Robo1;Robo2 null mice which showed decreased proliferation (Fig. 2-

9C), Pax1 removal has been shown to cause decreased cell proliferation in the

sclerotome (Peters, et al., 1999). To investigate potential interactions between Pax1,

Robo1, and Robo2, we characterized Pax1, Robo1, and Robo2 expression during the

early stages of vertebral column morphogenesis.

At 12.5dpc, Pax1 protein and Robo1 and Robo2 were expressed in the somitic

mesoderm in spatially similar but not identical domains (Fig. 2-10A, a-b). Specifically,

Robo1 and Robo2 expression appeared to be confined to the more medial

mesenchyme closer to the notochord (Fig. 2-10A, a), while Pax1 expression extended

to the lateral mesenchyme (Fig. 2-10A, b). Pax1 expression also appeared broader in

the lateral mesenchyme (Fig. 2-10A, b). Similarly, in the caudal region of 13.5 dpc

embryos Robo2 mRNA and Pax1 protein were expressed in a similar metameric pattern

in the dorsal and ventral mesenchyme surrounding the notochord (Fig. 2-10A, c-d).

Finally, at 14.5 dpc as the notochord to nucleus pulposus transition occurred, Robo1

and Robo2 and Pax1 shared a similar fan-shaped expression pattern in the presumptive

annuli fibrosi (Fig. 2-10A, e-f).

These data suggest that Robo1 and Robo2 are expressed in the embryonic

sclerotome, which is molecularly distinguishable within the somitic mesoderm by Pax1

Page 52: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

52

expression. To establish whether Robo1 and Robo2 were co-expressed with Pax1 in

the same cells, vertebral columns from 13.5 dpc embryos were incubated with

antibodies to Pax1 and bGal for confocal microscopy analysis. Confocal microscopy

revealed that although bGal and Pax1 had similar expression domains in the vertebral

column, Pax1-bGal co-localization, as defined as yellow fluorescence that occurs when

Pax1-555 and bGal-488 are in close proximity, occurred in only a subset of cells (Fig. 2-

10B).

Robo Gene Expression and Function in the Growth Plates

The bony vertebrae develop by endochondral ossification, wherein an avascular

cartilaginous template is first established by progenitors in the sclerotome. Following

formation of a cartilaginous skeletal element, cartilage-forming chondrocytes are

eventually replaced by bone forming osteoblasts as the cartilage template undergoes

remodeling and vascularization in preparation for ossification. By P0, the vertebrae have

begun to ossify and chondrocytes become organized at the vertebral growth plates, as

evidenced by the expression of distinct molecular markers. Bone elongation via

endochondral ossification occurs as growth plate chondrocytes, organized into distinct

layers, undergo stages of chondrocyte maturation. The coordinated differentiation of

chondrocytes at the rostal and caudal ends of the vertebra results in appositional growth

and elongation of the vertebrae.

Chondrocytes in the vertebral growth plates can be distinguished as being part of

the quiescent, resting zone (RZ); the proliferative, pre-hypertrophic zone (PZ); the

hypertrophic zone (HZ); and finally, as terminally differentiated, apoptotic cells that give

way to ossification (Fig. 2-11A). Growth plate chondrocytes can be distinguished based

Page 53: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

53

on cell morphology (ie. enlarged, hypertrophic cells) and by expression of specific

molecular markers. Chondrocyte differentiation and progression through these growth

plate zones is tightly regulated by a complex interaction of many regulatory factors,

including Bapx1, Pax1, Sox9, Runx2, PTHrP, and Ihh.

During bone growth, quiescent cells in the resting zone (RZ) express PTHrP.

PTHrP is a secreted signaling protein that promotes proliferation and inhibits terminal

differentiation via interaction with the PTH receptor. In response to PTHrP, cells

adjacent to the resting zone become proliferative and begin to express Ihh. Ihh, which is

expressed by pre-hypertrophic cells, feeds back to resting zone cells to stimulate PTHrP

secretion. Thus, the Ihh-PTHrP feedback loop is important for the maintenance of

chondrocytes in an immature state of differentiation. As bone elongation occurs and the

available amount of PTHrP diminishes, cells further away from the resting zone

differentiate further and become hypertrophic. Hypertrophic chondrocytes express

Col10a.

Chondrocytes at the disc-vertebra junction of control animals appeared

organized with distinct layers of small, flattened chondrocytes near the discs, and layers

of larger hypertrophic chondrocytes nearer to the trabecular centers of the vertebrae

(Fig. 2-11A, b) In contrast, chondrocytes at the disc-vertebra junction in Robo1;Robo2

null mice appeared disorganized (Fig. 2-11A, d). In P0 vertebral columns Robo1 and

Robo2 are expressed in the vertebral growth plates (Fig. 2-11B). Robo1 and Robo2

removal resulted in decreased Ihh and Col10a expression (Fig. 2-11C). While Ihh and

Col10a were expressed as distinct, tight bands along the vertebral growth plates in

controls, Ihh in Robo1;Robo2 null growth plates appeared diffuse (Fig. 2-11C, b) and

Page 54: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

54

Col10a expression was markedly diminished in mutants (Fig. 2-11C, d). Together, these

data suggest a role for Robo1 and Robo2 in regulating growth plate chondrocyte

differentiation.

Annulus Fibrosus Cell Morphology and Gene Expression

Under normal circumstances, Ihh and Col10a1 expression is restricted to growth

plate chondrocytes and is not expressed in the intervertebral discs (Fig. 2-12B, a, c). In

Robo1;Robo2 null mice, however, ectopic Ihh and Col10a expression was observed in

annulus fibrosus cells (Fig. 2-12B, b and d). These changes were accompanied by cell

shape changes in Robo1;Robo2 null annuli fibrosi, which can be observed by histology

(Fig. 2-12A) as well as by membrane-targeted fluorescence in Rosa-mT/mG mice (Fig.

2-6 C).

Cartilage Matrix Protein Expression

Based on observations that Robo1;Robo2 null mice had decreased Alcian blue

staining in the intervertebral discs (Fig. 2-4), antibodies specific for cartilage matrix

proteins, including Col6a1, Col2a1, Tenascin C, and Aggrecan, were used to assess

potential differences in matrix composition by immunofluorescence. Col2a1, Col6a1,

and Aggrecan are critically important components of cartilage. Col2a1 and Aggrecan

loss is associated with intervertebral disc pathology in humans.

By immunofluorescence a qualitative decrease in Col6a1 expression was

observed in the annuli fibrosi of Robo1;Robo2 null discs (Fig. 2-13A). Col2a1

expression appeared comparable, and was enriched in the inner annuli fibrosi of both

controls and mutants (Fig. 2-13B). Aggrecan immunofluorescence also appeared

diminished in Robo1;Robo2 null tissues (Fig. 2-13C). Aggrecan appeared reduced in

Page 55: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

55

both the annuli fibrosi and nuclei pulposi in Robo1;Robo2 null mice (Fig. 2-13C). A

decrease in Aggrecan in the vertebral column was observed in Robo1;Robo2 null mice

as early as 16.5 dpc (not shown).

Tenascin C Upregulation

Tenascin C (TNC) is associated with cartilaginous, ligamentous, and tendinous

tissues and its expression can be induced by mechanical stimulation. In the

intervertebral discs TNC is enriched in the annulus fibrosus. TNC is transcriptionally

regulated by PRX1, and is downstream of the PDGF and TGFbeta signaling pathways.

TNC null mice exhibit deficits in wound healing, but the in vivo consequence of TNC

knockout on chondrogenic differentiation is unknown. TNC belongs to a class of matrix

molecules known as adhesion modulatory proteins. In vitro, TNC has been shown to

modulate cell shape and adhesion by interfering with cell interactions with fibronectin.

Compared to cells plated on fibronectin alone, which were elongated and adherent,

cells plated on fibronectin and tenascin C detached from the substratum and showed

cell rounding.

Based on immunofluorescence, a decrease in TNC protein was observed in

Robo1;Robo2 null discs (Fig. 2-14A). Quantification of these differences by western blot

analysis revealed that Robo1;Robo2 null discs had a 60% increase in 170kD and 250kD

TNC fragments compared to control discs (Fig. 2-14B-C). An increase in the abundance

of 170kD and 250kD TNC fragments has been linked to joint pathology in humans. In

the same report it was demonstrated that recombinant 170kD and 250kD sized TNC

induced cartilage matrix degradation. These data suggest that Robo receptor proteins

Page 56: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

56

may play a role in cartilage matrix homeostasis by a Tenascin C-dependent

mechanism.

Discussion and Conclusions

In our studies the combined role of Robo1 and Robo2 in vertebral column

development was investigated using mice that lacked Robo1 and Robo2. The Robo1

and Robo2 individual null alleles were previously generated by LacZ knock-in and the

Robo1;Robo2 null mouse was subsequently generated by homologous recombination

to produce linked mutant alleles (Grieshammer, et al, 2004; Long, et al, 2004; Domyan,

et al, 2013). Robo1 and Robo2 are located 1.1 cM apart on chromosome 16 in mice. As

described previously, removal of Robo1 and Robo2 resulted in perinatal lethality caused

by respiratory defects (Domyan, et al, 2013). As such, previous studies were limited to

investigation of Robo1 and Robo2 expression and function only during embryonic

stages.

Role of Robo1 and Robo2 in IVD Development

Mice that lacked both Robo1 and Robo2 were observed to have intervertebral

disc defects (IVD). Macroscopically, Robo1;Robo2 null discs were bulbous rather than

flattened and disc-shaped as in controls (Fig. 2-4). Additionally, cells in the annulus

fibrosus lacked their characteristic spindle-shaped morphology and lamellar

organization around the nucleus pulposus (Fig. 2-4; Fig. 2-12). Robo1 and Robo2 were

expressed in the somitic mesoderm at 12.5 dpc and were distinctly absent in the

notochord (Fig. 2-2). More specifically, co-localization experiments showed that Robo1

and Robo2 expression domains overlapped with that of Pax1, which is a molecular

marker for sclerotome specification (Fig. 2-10). These data suggest that Robo1 and

Page 57: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

57

Robo2 were expressed in the sclerotome, which contains annulus fibrosus progenitors.

Robo1 and Robo2 were also observed in the presumptive annuli fibrosi surrounding the

rudimentary nuclei pulposi at 14.5 dpc, suggesting a potential role for Robo receptors in

annulus fibrosus differentiation from the sclerotome (Fig. 2-2).

Lineage tracing studies using genetic methods have shown that in mice, cells in

the notochord and sclerotome remain separate during morphogenesis of the

intervertebral discs. Nuclei pulposi in adult mice were exclusively of notochordal origin.

Equally, almost all notochord-derived cells were excluded or removed from the

vertebrae and annuli fibrosi (Choi and Harfe, 2011). Notochordal cells that remained in

the vertebrae were postulated to be notochordal remnants, which are thought to be the

cell reservoir responsible for chordoma formation in humans. The mechanism that

ensures proper separation of notochordal and sclerotomal cell populations during the

formation of the intervertebral discs is unknown.

In mice it has been shown that removal of Sox5 and Sox6, or inactivation of the

Hedgehog signaling pathway, results in aberrant scattering of notochord-derived cells in

the vertebrae and annuli fibrosi. Consequently, Sox5;Sox6 null and Hedgehog signaling

mutants had incompletely formed vertebral columns with absent or rudimentary

intervertebral discs (Smits and Lefebvre, 2003; Choi and Harfe, 2011). Thus,

maintenance of cell separation between notochord and non-notochord derived cells is

integral to normal intervertebral disc morphogenesis and vertebral column formation.

At 14.5 dpc, Robo1 and Robo2 expression were observed in the presumptive

annuli fibrosi (Fig. 2-2). At a similar stage, Slit2 mRNA was found to be enriched in the

nuclei pulposi (Fig. 2-3). Based on these complementary expression patterns, it was

Page 58: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

58

hypothesized that Robo-Slit signaling played a role in maintaining cell boundaries during

intervertebral disc development. Specifically, it was hypothesized that Slit secretion from

the nucleus pulposus repelled Robo-expressing annulus fibrosus cells from aberrantly

migrating into the nucleus pulposus (Fig. 2-8). Thus, in the absence of Robo1 and

Robo2 it was predicted that intervertebral discs in P0 mice would show signs of aberrant

cell mixing in the nuclei pulposi.

To determine if Robo1 and Robo2 in the presumptive annuli fibrosi played a role

in preventing migration of annulus fibrosus cells into the nucleus pulposus, ShhCre and

Rosa-mT/mG alleles were crossed into Robo1-/+;Robo2+/- mice (Fig. 2-6). ShhCre was

used to GFP-mark the notochord and all cells derived from the notochord. As in control

littermates, all cells in the nuclei pulposi of P0 Robo1;Robo2 null mice were GFP-

positive, indicating notochordal origin. Absence of tdTomato-positive cells in the nuclei

pulposi indicated that annulus fibrosus cells had not aberrantly migrated into the nuclei

pulposi during development. Additionally, as in controls, the vertebrae and annuli fibrosi

in Robo1;Robo2 null mice lacked GFP-positive cells, demonstrating that the notochord

had properly transitioned into nuclei pulposi anlagen during development (Fig. 2-6).

Together, these data show that the intervertebral disc defect seen in Robo1;Robo2 null

mutants was not caused by aberrant cell mixing in the nuclei pulposi as a result of

Robo1 and Robo2 removal.

Robo1;Robo2 null annuli fibrosi had aberrantly shaped cells that lacked the

distinct lamellar organization seen in control mice (Fig. 2-4). These changes were

accompanied by changes in the expression and distribution of extracellular matrix

proteins including Col2a1, Col6a1, and Aggrecan (Fig. 2-13). Col2a1, Col6a1, and

Page 59: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

59

Aggrecan are important for cartilage function and reduced expression of Col2a1,

Col6a1, and Aggrecan are linked to intervertebral disc degeneration in humans and

animals (Smith, et al, 2011; Urban and Roberts, 2003; Sivan, et al, 2014).

Notably, one matrix protein that was found to be upregulated in Robo1;Robo2

null intervertebral discs was Tenascin C (Fig. 2-14) Tenascin C upregulation in the

intervertebral discs was accompanied by abnormal cell rounding in the annuli fibrosi of

Robo1;Robo2 null mice (Fig. 2-12). Tenascin C is a matricellular adhesion modulatory

protein that has been shown to impede cell-matrix adhesion by interfering with cell

interactions with fibronectin (Chiquet-Ehrismann, et al, 1988). Tenascin C has been

shown to cause cell rounding in in vitro studies (Huang, et al, 2001). Western blot

analyses of control and mutant littermate tissues showed that removal of Robo1 and

Robo2 resulted in an increase in Tenascin C protein expression in the discs (Fig. 2-14).

In these experiments, biological triplicates from control and mutant littermates were

used to quantify 170kD and 250kD Tenascin C fragments in the discs. Upregulation in

170kD and 250kD Tenascin C fragments has previously been linked to joint pathology

in humans (Sofat, et al, 2012). Additionally, recombinant 170kD and 250kD Tenascin C

fragments were previously shown to induce cartilage matrix degradation (Sofat, et al,

2012). Based on these results, the observation that Robo1;Robo2 null mice had

cartilage matrix protein deficiencies and aberrant cell rounding in the annulus fibrosus is

consistent with the observation that Tenascin C was upregulated in Robo1;Robo2 null

mutants.

Page 60: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

60

Robo1 and Robo2 Function in the Sclerotome

Robo genes have been reported to regulate cell proliferation in vivo (Borrell, et al,

2012). Based on Robo1 and Robo2 expression in the sclerotome and the observation

that sclerotome-derived tissues (ribs) were hypoplastic at P0 (Fig. 2-8), 13.5 dpc

embryos were EdU pulsed for one hour and then examined for changes in cell

proliferation. Based on these experiments, removal of Robo1 and Robo2 was found to

impair proliferation in the sclerotome at 13.5 dpc (Fig. 2-9).

The proximal and distal ribs form from the sclerotome (Aoyama, et al, 2005).

Robo1;Robo2 null mutants had deficits in the ribs. Specifically, the distal ribs failed to

meet at the midline where the ribs fuse to the sternum (Fig. 2-8). The sternum derives

from the lateral plate mesoderm, which has been shown to require interactions with the

sclerotome for proper differentiation (Sudo, et al, 2001). In addition to deficits in the

distal ribs, Robo1;Robo2 null mutants sternal defects which included incomplete sternal

bar fusion and decreased sternal length (Fig. 2-8). It is not clear from these results if the

sternal defects were indirectly caused by Robo1 and Robo2 loss in the sclerotome, or if

Robo1 and Robo2 are expressed in and required for lateral plate mesoderm

differentiation.

Robo1 and Robo2 in Growth Plate Maintenance

Robo gene expression in the growth plate has been reported previously (Noel, et

al). Mechanical loading was found to increase Robo mRNA expression in the growth

plates, suggesting a role for Robo receptors in regulating growth plate chondrocyte

differentiation or homeostasis (Noel, et al, 1998). Xgal staining of vertebral columns

from Robo1+/-;Robo2+/- mice showed that Robo1 and Robo2 were expressed in the

Page 61: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

61

vertebral growth plates at P0 (Fig. 2-11). To determine if Robo1 and Robo2 played a

role in regulating chondrocyte differentiation or function in the growth plates, RNA in situ

hybridization was used to evaluate expression of chondrocyte differentiation markers in

the growth plates.

Compared to control littermates which had distinct bands of Ihh and Col10a

expression in the vertebral growth plates, Robo1;Robo2 null growth plates had

markedly reduced Ihh and Col10a expression (Fig. 2-11). In the growth plates, Ihh and

Col10a are expressed by pre-hypertrophic and hypertrophic chondrocytes, respectively

(de Crombrugghe, et al, 2000). These gene expression changes were accompanied by

chondrocyte disorganization in the growth plates of Robo1;Robo2 null mice (Fig. 2-11).

While examining Ihh and Col10a expression in the growth plates, ectopic Ihh and

Col10a was observed in the annuli fibrosi of P0 Robo1;Robo2 null mice (Fig. 2-12).

Together, these data support a role for Robo receptors in regulating chondrocyte

maturation in the vertebral growth plates and suggest that the cell rounding phenotype

seen in Robo1;Robo2 null mutants may be caused by ectopic expression of

chondrocyte hypertrophy markers in the annuli fibrosi.

Page 62: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

62

Figure 2-1. Robo1 and Robo2 Expression Analysis by RNA In Situ Hybridization and Xgal Staining. A) Robo expression analysis by RNA in situ hybridization and Xgal staining produces congruent results. Robo2 mRNA was detected in the neural tubes of wildtype 10.5 dpc embryos. B) Robo2 expression in the neural tube was confirmed by Xgal staining in stage matched Robo1;Robo2 heterozygotes. C) At 12.5 dpc, Robo2 mRNA appeared as repeating units in the somitic mesoderm along the rostrocaudal axis. D) Expression analysis by Xgal staining confirmed Robo2 expression in the vertebral column at 12.5 dpc. Wildtype embryos were used for RNA in situ hybridizations and Robo1;Robo2 heterozygotes were used for Xgal staining experiments. N=notochord; FP=floor plate

Page 63: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

63

Figure 2-2. Robo Gene Expression During Vertebral Column Formation. A) Robo1 and Robo2 expression was assessed by Xgal staining and RNA in situ hybridization. At 12.5 dpc, Robo1 and Robo2 expression was observed as repeating segments in the somitic mesoderm lateral to the notochord. B) At 12.5 dpc Robo1 and Robo2 expression was observed in the somitic mesenchyme ventral to the notochord (red arrow). C) In wildtype 13.5 dpc embryos, Robo1 mRNA was found in the vertebral column just lateral to the midline. D) At 13.5 dpc, Robo2 mRNA was expressed at the midline, dorsal and ventral to the notochord. E) As assayed by Xgal staining, Robo1 and Robo2 were expressed in the spinal cord and vertebral column at 14.5 dpc. F) At 14.5 dpc Robo1 and Robo2 were expressed in the presumptive annuli fibrosi but were absent in the nuclei pulposi anlagen. G) At 18.5 dpc, Robo2 mRNA was expressed in the spinal cord, and to a lesser degree, in the vertebral column. H) 100um sections through the vertebral column showed that Robo1 and Robo2 were absent in the discs and were enriched in the vertebral growth plates (red arrows). I) In P0 vertebral columns, Robo1 and Robo2 were expressed in the trabecular centers of the vertebrae (red arrow), in the perichondrium (white arrowheads), and in isolated annulus fibrosus cells (red arrows). SpC=Spinal cord; VC=Vertebral column; NP=Nucleus pulposus; AF=Annulus fibrosus; V=Vertebra. Notochord denoted by black arrowheads in A, B, and D.

Page 64: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

64

Figure 2-3. Slit Gene Expression During Vertebral Column Formation. A) RNA in situ hybridization using DIG-labeled antisense probes showed Slit1 gene expression in the notochord and floor plate at 10.5 dpc. B) At 10.5 dpc Slit2 mRNA was expressed in the floor plate and notochord. C) At 10.5 dpc Slit3 mRNA was observed in the floor plate. D) At 12.5 dpc Slit1 mRNA was expressed in the floor plate and notochord (black arrowhead). E) At 12.5 dpc Slit2 mRNA was expressed in the floor plate and in the notochord but was absent in the surrounding somitic mesoderm. F) At 12.5 dpc Slit3 was expressed in the floor plate but was not detected in the notochord. G) Slit1 mRNA was not detected in the intervertebral discs at 15.5 dpc. H) At 15.5 dpc Slit2 mRNA was enriched in the nuclei pulposi anlagen. I) At 15.5 dpc Slit3 mRNA was not observed in the intervertebral discs. FP=Floor plate; NP=Nucleus pulposus. Notochord denoted by black arrowheads in A-F.

Page 65: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

65

Figure 2-4. Removal of Robo1 and Robo2 Causes Intervertebral Disc Malformations in P0 Mice. A) Removal of Robo1 and Robo2 resulted in enlarged, abnormally shaped discs. Compared to intervertebral discs from control littermates (a), mice null for Robo1 and Robo2 had enlarged, bulbous intervertebral discs (b). Disc malformations were apparent along the length of the thoracic and lumbar level regions (c, d). Robo1+/- and Robo1+/-;Robo2+/- controls were indistinguishable from wildtype littermates (not shown). B) Homozygous removal of Robo1 resulted in less severe disc malformations at P0. Robo1+/- control littermates had characteristically flattened discs (a) while Robo1 null mice had slight malformations in disc shape (b). C) Vertebral columns from 17.5 dpc Slit1-/-;Slit2-/- mice appeared comparable to those from control littermates. All images are of frontally sectioned vertebral columns that were stained with Alcian blue and Picrosirius red to show cartilage and fibrillar collagens, respectively. IVD=Intervertebral discs; NP=Nucleus pulposus.

Page 66: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

66

Fig 2-5. Conditional Inactivation of Robo2F on Robo1 Null Background. A) X-gal stained Dermo1Cre;R26R tissues. At 9.5dpc Dermo1 was expressed in the notochord and surrounding mesoderm (a, b). At P0, Dermo1 showed mosaic expression in the vertebrae, annulus fibrosus, and nucleus pulposus (c). Embryos were sectioned across the transverse plane (a, b) or frontal plane (c). Tissues were Nuclear Red counterstained to show tissue architecture. B) Robo2F was inactivated in the somitic mesoderm using Dermo1Cre. Homozygous inactivation of Robo2F on a Robo1 null background partially recapitulated the Robo1;Robo2 null phenotype at P0. Vertebral columns from mutant and control animals were frontally sectioned and stained with Alcian blue and Picrosirius Red. Compared to controls (a), homozygous inactivation of Robo2F with Dermo1Cre on a Robo1 null background resulted in reduced Alcian blue staining in the vertebrae and intervertebral discs (b), signifying deficits in chondrogenesis following vertebral column formation. C) Robo2F inactivation with ShhCre on a Robo1 null background did not recapitulate the Robo1;Robo2 null phenotype. ShhCre is expressed in the notochord and is not expressed in the somitic mesoderm. NT=Neural tube; LB=Limb bud; FP=Floor plate; N=Notochord; NP=Nucleus pulposus; AF=Annulus fibrosus; V=Vertebrae.

Page 67: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

67

Figure 2-6. The Notochord to Nucleus Pulposus Transition Occurs Normally in the Absence of Robo1 and Robo2. A) Model for Robo-Slit interaction during intervertebral disc morphogenesis. Slit and Robo exhibited complementary expression patterns in the vertebral column at 15.5 dpc with Robo1 and Robo2 expression in the presumptive annuli fibrosi (blue region) and Slit2 mRNA expression in the nuclei pulposi anlagen (red region) (a). Based on this model, in wildtype embryos Slit glycoprotein secretion from the nucleus pulposus would inhibit annulus fibrosus cells from migrating into nuclei pulposi anlagen via Slit protein interactions with Robo1 and Robo2 receptors expressed on annulus fibrosus cells (b). Removal of Robo1 and Robo2 would result in loss of Slit-mediated repulsion leading to a mixed population of nucleus pulposus and annulus fibrosus cells at birth. NP=Nucleus pulposus; AF=Annulus fibrosus. B) Mating scheme for lineage tracing analysis. The Rosa-mT/mG dual fluorescence reporter was used to assess the lineage of disc cells in perinatal vertebral columns. In the absence of Cre protein, all cells in the Rosa-mT/mG mouse fluoresce red (tdTomato). In the presence of Cre, Cre-mediated excision of the tdTomato cassette results in the production of membrane-targeted GFP protein. ShhCre was used to mark all cells in the embryonic notochord. All cells that derive from the notochord fluoresce green (GFP) in the perinatal vertebral column. Robo1+/-;Robo2+/-; ShhCre males were mated to Robo1+/-;Robo2+/-; Rosa-mT/mG females. Embryos were collected at 18.5 dpc and mice that were positive for both ShhCre and Rosa-mT/mG were selected for lineage analysis. C) Representative images from controls (a-h) and Robo1;Robo2 null mutants (i-p) showing localization of GFP+ cells within the nuclei pulposi (b, j) and exclusion of tdTomato+ non-notochord derived cells from the nucleus pulposus (a, i). Higher magnification of the nucleus pulposus – annulus fibrosus junction from a-d and i-l are shown in e-h and m-p, respectively. Scale bars = 100um (a-d, i-l); Scale bars = 50um (e-h, m-p). VB=vertebral body; AF=annulus fibrosus; NP=nucleus pulposus.

Page 68: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

68

Figure 2-7. Lineage Tracing Analysis of ShhCreERT2 Marked Cells. A) Mating scheme for lineage tracing analysis, using Tamoxifen-inducible ShhCreERT2. Robo1+/-; Robo2+/-; Rosa-mT/mG females were mated to Robo1+/-; Robo2+/-; ShhCreERT2 males. Pregnant dams were administered a pulse of Tamoxifen by oral gavage on 10.5 dpc, and embryos were harvested for lineage analysis at 18.5 dpc. Using this scheme, all cells expressing Shh between 10.5dpc and approximately 12.0 dpc undergo Cre-mediated excision of the tdTomato cassette. B) Lineage tracing analyses by fluorescent microscopy. Derivatives of Shh-expressing cells reside in the nucleus pulposus at 18.5 dpc in controls (b, f) and Robo1;Robo2 null mutants (j, n). No ectopic tdTomato+ cells were found in the nuclei pulposi or either controls (a, e) or Robo1;Robo2 null mice (i, m). Higher magnification of the nucleus pulposus – annulus fibrosus junction shown in (e-h) and (m-p). Scale bars = 100um (a-h); Scale bars = 50um in (a‟-h)‟. VB=vertebral body; AF=annulus fibrosus; NP=nucleus pulposus.

Page 69: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

69

Figure 2-8. Robo1;Robo2 Null Mice Have Deficits in the Distal Ribs and Sterna at P0. A) Robo1;Robo2 null mice have smaller rib cages at birth. Compared to controls (a-c), loss of Robo1 and Robo2 results in smaller ribs with underdeveloped distal ribs that are unfused to the sternum (d-g). All skeletal images were imaged at the same magnification (8x). B) Robo1 and Robo2 loss caused sternal defects. Representative skeletal preparations from one control (a) and two Robo1;2 null mutants (c, e) depict sternal fusion deficits caused by Robo1 and Robo2 removal. Higher magnification of the sterna in a, c, and e are shown in b, d, and f, respectively. Compared to control mice, which had distal ribs that attached at fused, segmented sterna (a, b), Robo1;Robo2 null mice had shorter sterna (c) that were unfused at the midline (d) and distal ribs that failed to stain with Alcian blue (e, f). C) Robo1;2 null mutants had a 30% decrease in sternal length at P0 compared to controls. n=2 controls; n=5 Robo1;Robo2 null mutants.

Page 70: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

70

Figure 2-9. Impaired Proliferation in Robo1; Robo2 Null Mutants at 13.5 dpc. A) Anti-β-Galactosidase (bGal) antibody is a reliable proxy for Robo expression in the vertebral column. Sagittal cross sections from a 14.5 dpc Robo1+/-;Robo2+/- embryo shows Robo expression by Xgal staining in the vertebral column (a). Sagittal sections from 13.5 dpc Robo1+/-;Robo2+/- mice were incubated with anti-bGal primary antibody and 555-conjugated secondary antibody to show bGal protein in the vertebral column (b). Both images are shown at 20x. B) Schematic illustrating how percent proliferation in the sclerotome was quantified. Sagittal cross sections through the midline from the trunk level of 13.5 dpc embryos were used to fluorescently label nuclei (c). An antibody against β-galactosidase was used as a proxy for Robo1/2 expression in the vertebral column (b). Percent proliferation was calculated as the total number of EdU positive

Page 71: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

71

cells within β-Gal-488 positive domains (d) divided by total number of DAPI nuclei within the same β-Gal-488 domains (c). SpC = Spinal cord; Noto = Notochord. C) Representative images from control (a-d) and Robo1;2 mutants (e-h) showing proliferation in trunk region at 13.5 dpc. Vertebral columns were sectioned sagittally and are shown in the same orientation, with spinal cord on the left. Midline sections in the trunk region between the fore- and hindlimbs were selected for analysis. Scalebar = 100um. D) Compared to controls, which showed 23.6 % proliferation in the Robo-expressing sclerotome, Robo1/2 null embryos showed 11.5% proliferation in the sclerotome. Percent proliferation in the notochord, which does not express Robo, was used as a control for overall mitotic index. No change in proliferation in the notochord was observed. Pregnant dams were EdU pulsed for one hour by IP injection. Robo1+/-;Robo2+/- littermates were used as controls to evaluate the effect of Robo loss on proliferation. n=3 controls, n=3 mutants.

Page 72: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

72

Figure 2-10. Robo and Pax1 Have Overlapping Expression Patterns in the Developing Vertebral Column. A) Pax1 and Robo1;Robo2 have similar but not identical expression patterns in the developing vertebral column. Robo1+/-;Robo2+/- embryos were Xgal stained to show Robo1 and Robo2 in the vertebral column at 12.5 dpc and 14.5 dpc (a, e). An antisense probe to Robo2 mRNA was used to show Robo2 expression in wildtype 13.5 dpc embryos (c). Anti-Pax1 antibody was used to show Pax1 expression in stage-matched wildtype or Robo1+/-;Robo2+/- embryos (b, d, f). SpC=spinal cord. Merged Pax1 and DAPI immunofluorescence shown in b, d, f. B) Pax1 and Robo are co-expressed in a small subset of cells in the sclerotome. Sagittally sectioned tissue from a 13.5 dpc Robo1+/-; Robo2+/- embryo was fluorescently labeled for Pax1 (a) and bGal (b). bGal was used as a proxy for Robo expression. Z-stacked images taken by confocal microscopy show that Pax1 and Robo have similar expression patterns in the vertebral column (a, b), and that a subset of cells in the ventral sclerotome co-express Pax1 and Robo at this stage (c).

Page 73: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

73

Figure 2-11. Disrupted Growth Plate Marker Expression in Robo1;Robo2 Null Mutants. A) Chondrocytes in the vertebral body (VB) growth plates appear disorganized in Robo1;Robo2 null mutants at P0 (d) compared to control littermate growth plates, which have distinct layers of small, quiescent cells and larger, more differentiated cells nearer to the VB ossification centers (b). RZ=Resting zone; PZ=Proliferative zone;

Page 74: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

74

HZ=Hypertrophic zone; TB=Trabecular bone. All images are of frontally sectioned, Alcian blue and Picrosirius red stained vertebral columns from P0 mice. (B) In perinatal vertebral columns, Robo1;Robo2 expression, as assayed by Xgal, is enriched in the VB growth plates. C) Expression of chondrocyte differentiation markers is disrupted in the vertebral growth plates of Robo1;Robo2 null mice at P0. Ihh expression appeared reduced in Robo1;Robo2 null mutants at P0 (b) compared to control littermates (a) which had Ihh mRNA enriched at the growth plates. Col10a, which is expressed by hypertrophic chondrocytes, was barely detectable in Robo1;Robo2 null vertebral columns (d). By comparison Col10a expression was specific to and enriched in the vertebral growth plates (c). VB=Vertebral body; NP=Nucleus pulposus. Growth plates denoted by white arrows.

Figure 2-12. Robo1;Robo2 Null Annuli Fibrosi Have Aberrant Cell Morphologies And Ectopic Expression of Growth Plate Chondrocyte Markers. A) Robo1;Robo2 null

Page 75: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

75

mutants show aberrant cell morphology in the annuli fibrosi at P0. Compared to annulus fibrosus cells in control mice which are spindle shaped and organized into concentric rings around the nucleus pulposus (a), annulus fibrosus cells in Robo1;Robo2 null mice appear rounded and enlarged (b). B) These cell shape changes are accompanied by ectopic expression of chondrocyte hypertrophy and pre-hypertrophy markers in the discs. While Ihh expression is confined to the vertebral growth plates in controls (a), Ihh is ectopically expressed by annulus fibrosus cells in Robo1;Robo2 null mice (b, red arrow). Col10a1 expression is restricted to hypertrophic growth plate chondrocytes in controls (c). Robo1;Robo2 null mice show loss of an organized hypertrophic zone and express Col10a1 ectopically in the annulus fibrosus (d, red arrow). All images (A, B) are frontal cross-sections from P0 mice.

Figure 2-13. Robo1 and Robo2 Loss is Associated With Reduced Expression of Cartilage Matrix Proteins. A) Col6a1 is decreased in the intervertebral discs of Robo1;Robo2 null mice. Compared to discs from control littermates (a), discs from Robo1;Robo2 null mice show decreased staining for Col6a1 (b). B) Col2a1 expression is enriched in the inner annuli fibrosi. At P0, Col2a1 immunofluorescence in control and Robo1;Robo2 null littermates appears comparable. In control (a) and Robo1;Robo2 mutant discs (b) Col2a1 is enriched in the inner annuli fibrosi. C) Aggrecan is decreased in Robo1;Robo2 null mutants. Compared to discs from control littermates which had robust staining throughout the nucleus pulposus and annulus fibrosus (a), Robo1;Robo2 null discs had reduced staining in the annulus fibrosus (b). All images are of frontally sectioned vertebral columns from P0 mice and are shown at 20x. af=annulus fibrosus; np=nucleus pulposus. DAPI shown in c, d; merged images shown in e, f.

Page 76: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

76

Figure 2-14. Tenascin C is Upregulated in the Intervertebral Discs of Robo1;Robo2 Null mice. A) Tenascin C upregulation in Robo1;Robo2 null mutant discs can be seen by immunofluorescence. Frontally sectioned discs from P0 control and Robo1;Robo2 null littermates were incubated with anti-Tenascin C antibody. In both control and Robo1;Robo2 null discs, Tenascin C is enriched in the annuli fibrosi (a, b). DAPI shown in c, d. Tenascin C/DAPI merged images shown in e, f. All images are 20x. B) Western blot analysis of Tenascin C in P0 discs. Thoracic and lumbar level discs from P0 mice were pooled for protein quantification by western blot. Each lane represents one biological replicate (10-14 discs pooled from the same animal). Compared to discs from wildtype littermates (lanes 1-3), discs from Robo1;Robo2 null mice showed an increase in high molecular weight Tenascin C (lanes 4-6). C) Western blot quantification. Robo1;Robo2 null mutants had a 60% increase in Tenascin C in the discs at P0. GAPDH was used as a loading control to calculate fold change based on densitometry of the 170kD and 250kD Tenascin C bands.

Page 77: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

77

Figure 2-15. Slit1;Slit2 Null Phenotype. Deletion of Slit1 and Slit2 did not recapitulate the Robo1;Robo2 null intervertebral disc phenotype. Vertebral columns from 17.5 dpc Slit1+/-;Slit2+/- control littermates (A, C) and Slit1;Slit2 null mutants (B, D) were frontally sectioned and stained with Alcian blue and Picrosirius Red. Annulus fibrosus cell morphologies were normal in Slit1;Slit2 null mice at 17.5 dpc (C-D). NP=Nucleus pulposus; AF=Annulus fibrosus; V=Vertebra

Page 78: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

78

CHAPTER 3 WNT/β-CATENIN SIGNALING DURING INTERVERTEBRAL DISC DEVELOPMENT

Introduction

The Wnt/β-Catenin signaling pathway is an evolutionarily conserved signal

transduction network that mediates many crucial developmental and disease processes,

including intervertebral disc formation, maintenance, and intervertebral disc disease as

well as cartilage and long bone development (Dahia, Mahoney, et al. 2009; Winkler,

Mahoney, et al. 2014; Usami, Gunawardena, et al. 2016). In the canonical Wnt/β-

Catenin signaling pathway, secreted Wnt glycoproteins bind to Frizzled (Fz)

transmembrane G protein-coupled receptor proteins, resulting in β-Catenin

accumulation in the cytoplasm followed by β-Catenin translocation into the nucleus. In

the nucleus, β-Catenin proteins interact with TCF/Lef family of transcription factors to

regulate expression of downstream targets (Cadigan and Nusse).

In the early embryo within the axial mesoderm, Wnt/β-Catenin signaling is

required for notochord formation and extension, and in mice inactivation of the Wnt/β-

Catenin signaling pathway is known to cause deficits in notochord elongation (Cheyette,

Waxman, et al. 2002; Ukita, et al. 2009). Following notochord formation, opposing

gradients of Shh and Wnt signaling in the somitic mesoderm confer dorsal-ventral

polarity to the somitic mesoderm for induction of the dermamyotome and sclerotome,

respectively (Fan, Lee, et al. 1997; Fan, Tessier-Lavigne, 1994). In vitro, pluripotent

cells can be induced to form sclerotome-like chondroprogenitors when treated with

drugs that modulate the Wnt/β-Catenin signaling pathway (Zhao, Li, et al. 2014).

Recent studies in vertebrate models implicate Wnt/β-Catenin signaling in

intervertebral disc development and disease. In 18.5dpc mice Wnt signaling has been

Page 79: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

79

observed in and near the intervertebral discs, including the annulus fibrosus,

cartilaginous endplates, and vertebral growth plates (Kondo, et al. 2012). In these

studies, Wnt/β-Catenin signaling was not observed in the nuclei pulposi during

embryonic stages, but was found to be up-regulated in the nucleus pulposus

postnatally, concomitant with Wnt/β-Catenin down-regulation in the annulus fibrosus

and complete loss of Wnt/β-Catenin signaling in the growth plates and endplates

(Kondo, et al. 2012). Another study that used immunohistological methods similarly

found evidence of Wnt signaling activity in the intervertebral discs of prenatal and

postnatal mice (Dahia, Mahoney, et al. 2009).

Changes in Wnt/β-Catenin signaling activity have also been linked to

intervertebral disc degeneration in vertebrate animal models. In rats, chemically induced

nuclear accumulation of β-Catenin in the intervertebral discs has been shown to induce

cell senescence and apoptosis in the nucleus pulposus (Hiyama, Sakai, et al. 2010). A

decrease in canonical Wnt signaling and caveolin-1 expression has also been linked to

intervertebral disc disease in chondrodysplastic dogs (Smolders, et al. 2013). In mice,

Wnt/β-Catenin signaling activity was found to be reduced in the intervertebral discs of

aged animals and was linked to degenerative changes in the intervertebral discs,

including decreases in cell proliferation and reduced expression of chondrogenic

markers, including Sox9, Aggrecan, Col2a1, and Col1a1 (Winkler, Mahoney, et al;

Hiyama, Sakai, et al). Additionally, activation of the Wnt/β-Catenin signaling pathway

has been shown to promote intervertebral disc degeneration by inducing expression of

matrix degrading enzymes (Hiyama, Sakai, et al).

Page 80: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

80

Previous studies that evaluated Wnt/β-Catenin signaling in the intervertebral

discs have produced variable and conflicting results. These results were produced using

immunohistological staining methods and Xgal staining of TOPGAL reporter mice. To

evaluate Wnt/β-Catenin signaling activity in the intervertebral discs using a novel

method, TCF/Lef-H2B-GFP reporter mice were used to characterize Wnt/β-Catenin

signaling activity in the vertebral column and intervertebral discs during embryonic and

postnatal development.

Results

TCF/Lef:H2B-GFP reporter mice (Ferrer-Vaquer, et al.) were used to investigate

Wnt/β-Catenin signaling activity in the developing vertebral column. In the canonical

Wnt signaling pathway, Wnt activation of the Fz G protein-coupled cell surface receptor

results in stabilization of cytoplasmic β-Catenin and translocation of β-Catenin to the

nucleus where it interacts with T cell-specific transcription factor/lymphoid enhancer-

binding factor 1 (TCF/Lef) family of transcription factors to drive expression of Wnt-

responsive genes (MacDonald, et al. 2009). The TCF/Lef:H2B-GFP reporter used in

these experiments were generated using a construct consisting of H2B-GFP under the

control of six TCF/Lef response elements and the hsp68 minimal promoter. Nuclear

localization of the chromatin-bound H2B-GFP fusion protein was designed to enable

GFP readout at a single-cell resolution (Ferrer-Vaquer, et al. 2010).

At 12.5 dpc, Wnt/β-Catenin signaling activity was observed in the tail-level

notochord in a region caudal to the external genitalia (Fig. 3-1B). Wnt signaling was also

observed in the more anterior trunk-level notochord, between the fore- and hindlimbs

(Fig. 3-1E). These data were consistent with published results that reported Wnt

Page 81: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

81

signaling activity in the notochord (Maretto, Cordenonsi, et al. 2003) and with reports

that demonstrated a role for Wnt/β-Catenin signaling in notochord extension and tail

formation (Ukita, Hirahara, et al. 2009).

Following the notochord to nucleus pulposus transition at 15.5 dpc, Wnt/β-

Catenin signaling activity was detected in the subset of nucleus pulposus cells in the

vertebral column (Fig. 3-2B and Fig. 3-2D). Wnt/β-Catenin signaling was not observed

in annulus fibrosus cells at this stage (Fig. 3-2A-D). At 18.5dpc, Wnt signaling was

maintained in the spinal cord, as reported previously (Ferrer-Vaquer, et al. 2010) and in

a small number of isolated chondrocytes located in the vertebrae (Fig. 3-2F). Wnt

signaling was also observed in isolated nucleus pulposus cells at 18.5 dpc (Fig. 3-2F

and Fig. 3-2H). These data are in contrast to published studies that showed absence of

Wnt/β-Catenin signaling activity in the nucleus pulposus at 18.5dpc (Kondo, et al). At

18.5dpc Wnt/β-Catenin signaling activity was reported in the annulus fibrosus and

endplates (Kondo, et al, 2011).

In postnatal mice, Wnt/β-Catenin signaling was maintained in the spinal cord

(Fig. 3-3C, Fig. 3-3L). In one-month-old postnatal mice, Wnt/β-Catenin signaling was

detected in a subset of nucleus pulposus cells (Fig. 3-3E). These data are in agreement

with a previously published study that reported Wnt/β-Catenin signaling in the nucleus

pulposus at five weeks old (Kondo, et al, 2011). At one month old, very little Wnt

signaling activity was observed in the annulus fibrosus (representative images of the

annulus fibrosus at one month shown in Fig. 3-3G-I). A few isolated GFP-positive cells

were detected in the annulus fibrosus at one month old (not shown) and in perichondrial

mesenchymal cells located at the periphery of the vertebral column (Fig. 3-3H). These

Page 82: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

82

data are also in agreement with published data that showed Wnt signaling down-

regulation in the annulus fibrosus in comparably aged postnatal mice (Kondo, et al.

2011). Finally, at one year old, Wnt signaling was markedly decreased in all parts of the

intervertebral discs but was observed in trabecular osteoblasts located in the vertebral

bodies (Fig. 3-3J-L).

Conclusions

Wnt/β-Catenin signaling has been implicated in intervertebral disc development

in disease in humans and animals. The status of Wnt/β-Catenin signaling in

intervertebral disc and disc progenitor cells have been investigated using a variety of

methods, including evaluation of β-Catenin mRNA and protein and LacZ-based reporter

methods (Hiyama, et al. 2010; Dahia, et al. 2009; Winkler, et al. 2014). These studies

have often produced inconsistent results. To assess the status of Wnt signaling using a

novel method, TCF/Lef-H2B:GFP reporter mice were used to monitor Wnt/β-Catenin

signaling in the notochord and intervertebral discs at a single-cell resolution at

embryonic and postnatal stages.

Robust GFP fluorescence, indicative of high levels of Wnt signaling activity, was

detected throughout the notochord at 12.5 dpc, congruent with published results that

demonstrated a role for Wnt/β-Catenin signaling in notochord (Ukita, Hirahara, et al.). At

15.5 dpc, after the notochord had transitioned into nuclei pulposi, Wnt/β-Catenin activity

was detected in approximately 50% of cells located in the nuclei pulposi. These data

were congruent with a published study that showed nuclear β-Catenin protein staining in

the nucleus pulposus of 15.0dpc mouse embryos (Hiyama, Sakai, et al).

Page 83: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

83

In 18.5 dpc and in one month old postnatal mice, Wnt signaling was observed in

the nuclei pulposi, albeit at lower levels than at 15.5 dpc (Fig. 3-2 and Fig. 3-3).

Compared to intervertebral discs at 18.5 dpc, tissues from one month old postnatal mice

showed GFP fluorescence in the nuclei pulposi, albeit in a smaller percentage of cells

(Fig. 3-3). These data are in contrast to a published report that showed upregulation in

Wnt/β-Catenin signaling in the nuclei pulposi between 18.5 dpc and five weeks old

(Kondo, et al. 2011). The reporter used in those studies was the TOPGAL reporter

strain, in which LacZ transgene expression is driven by three TCF/LEF binding motifs

upstream of a minimal c-fos promoter (DasGupta and Fuchs, 1999). Another study,

which also used the TOPGAL Wnt reporter, reported that in postnatal intervertebral

discs (one week and three weeks old) Wnt signaling activity was present in the annuli

fibrosi but not in the nuclei pulposi (Dahia et al. 2009). These results are inconsistent

with the observations made using the TCF/Lef:H2B-GFP reporter.

Postnatal intervertebral discs from one month old mice showed Wnt/β-Catenin

signaling in the nucleus pulposus. Intervertebral discs from one year old mice showed

markedly reduced or absent Wnt/β-Catenin signaling in the nuclei pulposi and annuli

fibrosi (Fig. 3-3). Thus, Wnt/β-Catenin signaling is down-regulated in the intervertebral

discs with age. These results are congruent with published data that suggested that

Wnt/β-Catenin signaling is down-regulated in the intervertebral discs as animals age

(Winkler, et al. 2014). Down-regulation in the Wnt/β-Catenin signaling pathway may be

important for postnatal intervertebral disc homeostasis. Mice that had conditional re-

activation of the Wnt/β-Catenin signaling pathway showed signs of degenerative

changes in the discs (Wang, et al. 2013) and postnatal activation of the Wnt/β-Catenin

Page 84: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

84

signaling pathway was shown to enhance intervertebral disc cell senescence in rats

(Hiyama, et al. 2013).

In sum, these data confirmed the presence of Wnt/β-Catenin signaling in the

embryonic notochord, where it has been shown to regulate notochord extension (Ukita,

et al 2009). These data also suggest that Wnt signaling may be more integral to nucleus

pulposus development than annulus fibrosus development, and show that Wnt signaling

is down-regulated in the discs with age. Dissimilarities between our data and published

results may be explained by the use of different methods to assess Wnt signaling

activity. Previous studies relied on the assessment of β-Catenin protein or mRNA

localization or on the use of TOPGAL reporter mice, which relied on LacZ driven by

three TCF/Lef binding motifs and c-fos promoter.

Collectively these data highlight the utility of the TCF/Lef-H2B-GFP mouse for

reporting Wnt signaling activity in postnatal intervertebral discs, when Wnt/β-Catenin

signaling activity may be too low to detect by other methods. These data support a role

for Wnt/β-Catenin signaling in intervertebral disc development, and suggest that Wnt/β-

Catenin signaling down-regulation at postnatal stages may be a normal facet of disc

biology and homeostasis.

Page 85: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

85

Figure 3-1. WNT/β-Catenin Signaling in the Notochord at 12.5 dpc. Wnt signaling was observed in the notochord at 12.5 dpc in the caudal notochord located in the tail (A-C white arrows) and in the trunk-level notochord between the fore- and hindlimb buds (D-F white arrows). Wnt signaling was also observed in neuronal progenitors in the neural tube (NT). Embryos were sectioned through the sagittal plane, counterstained with DAPI, and imaged at 20x. NT=Neural tube; N=Notochord; EG=External genitalia.

Page 86: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

86

Figure 3-2. Wnt Signaling in the Embryonic Intervertebral Discs. WNT/β-Catenin Signaling was observed in the intervertebral discs at 15.5 dpc (A-D white arrows) and 18.5 dpc (E-H white arrows). At 15.5 dpc, WNT/β-Catenin signaling was observed in the neural tube and in the nuclei pulposi of the intervertebral discs (B, white arrows). Higher magnification of the panel C inset shows WNT signaling in a subset of nucleus pulposus cells, which are circumscribed in white dotted lines. No Wnt signaling was observed in the presumptive annulus fibrosus at 15.5 dpc. At 18.5 dpc, WNT signaling was observed in the vertebral column (E-H white and red arrows). Specifically, WNT signaling was detected in intervertebral disc cells (F, white arrow) and in chondrocytes located in the vertebrae (F, red arrow). A higher magnification GFP/DAPI image shows that WNT signaling was specific to nucleus pulposus cells (H, white arrows). No WNT signaling was observed in the annulus fibrosus at 18.5 dpc. The nucleus pulposus-annulus fibrosus boundary is denoted by the dashed white line in H. Vertebral columns were sectioned through the sagittal plane, counterstained with DAPI, and imaged at 20x (A-C, E-G).

Page 87: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

87

Figure 3-3. Postnatal Wnt Signaling in the Intervertebral Discs. Sagittal cross sections from one month old postnatal TCF/Lef:H2B-GFP mice showed Wnt signaling in the nuclei pulposi of the intervertebral discs, denoted by white dashed lines in A-C. The nucleus pulposus and annulus fibrosus are denoted by the white and red boxes,

Page 88: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

88

respectively, in B-C. Higher magnification images of the nucleus pulposus, denoted by the white box, are shown in D-F. The annulus fibrosus, denoted by the red box, are shown at higher magnification in G-I. Wnt signaling was observed in isolated cells located in the nucleus pulposus (E, white arrows). Very little Wnt signaling was observed in the annulus fibrosus at one month (G-I). GFP-positive cells were observed in perichondrial mesenchyme in one month old mice (H, red arrows). At one year postnatal, Wnt signaling was undetectable in the intervertebral discs but was observed in vertebral osteoblasts (J-L, red arrows). SpC=Spinal cord; V=Vertebrae; GP=Growth plate; NP=Nucleus pulposus; AF=Annulus fibrosus. Scalebar=100um (A-F, J-L) and 50um (G-I).

Page 89: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

89

CHAPTER 4 METHODS

Histology

Vertebral column tissues were stained with Alcian blue and Picrosirius red as

described previously1, 2 (Harfe, Choi PNAS1; Gruber, et al 2002). Briefly, 4% PFA-fixed

tissues were washed in PBS and dehydrated through a series of graded ethanol

washes (water, 25% EtOH, 50% EtOH, 70% EtOH, 95% EtOH, 100% EtOH) before

being immersed in xylenes in preparation for paraffin embedding. Tissues were

perfused with Blue Ribbon paraffin and embedded in paraffin at 65°. A microtome was

used to section vertebral columns at 10um onto SuperFrost Plus Gold glass slides.

Slides were left to dry 24-72 hours on a slide warmer.

Tissues were re-hydrated through xylenes and ethanol washes and then

immersed in Alcian blue pH 2.5 for 15 minutes. Following Alcian blue, slides were

washed in running tap water to prepare for Picrosirius red. Slides were immersed in

Picrosirius red for 45 minutes, washed in acidified water (0.025% acetic acid). Slides

were then put through a series of ethanol washes again in preparation for mounting with

Permount.

Skeletal Preparations

Alcian blue and alizarin red were used to stain cartilage and ossified skeletal

elements as described previously3. Embryos were skinned, eviscerated, and fixed

overnight in 4% PFA/PBS. After fixation, tissues were PBS washed and dehydrated

through a series of ethanol washes in preparation for Alcian blue. Skeletons were

submersed in Alcian blue overnight at room temperature. The next day excess Alcian

blue was removed with multiple washes through 30% acetic acid/70%ethanol. Tissues

Page 90: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

90

were then rehydrated and stained overnight in alizarin red in 1% KOH. Following alizarin

red, skeletons were cleared for several days in 1% KOH and then stored and imaged in

80% glycerol.

Western Blot Analysis

Vertebral columns were dissected into cold PBS, and then thoracic and lumbar

level discs were microdissected out and pooled into 200 uL of RIPA

(radioimmunoprecipitation assay buffer) lysis buffer supplemented with PMSF (final

concentration of 1mM) and EDTA (0.5M) for each animal. Discs were homogenized with

an electric homogenizer on ice. Samples were centrifuged at 13,000rpm for 12 minutes

and supernatant was placed in a clean tube. BCA assay (BioRad) was used to

determine protein concentration and all samples were brought to equimolar

concentration with RIPA buffer. Samples were prepared with 6x Laemmli loading buffer

and heated to 95° for five minutes.

20ug total lysate from each sample was loaded into 4-20% Mini-PROTEAN TGX

Precast Protein Gels purchased from BioRad and standard running buffer (1x Tris

glycine) was used for gel electrophoresis. Transfer buffer (20% final concentration

MeOH) was used for wet transfer proteins onto PVDF membrane at 100V for two hours

at 4°. Blots were blocked with 3% BSA for one hour at room temperature before

incubation with primary antibody. Tenascin C blots were incubated with 1:1000 anti-

Tenascin C antibody from Santa Cruz (H-300), diluted in 3% BSA, overnight at 4°.

PBST (0.1% Tween) was used to wash blots 3x at room temperature. Blots were then

incubated at room temperature in secondary antibody, which was Anti-Rabbit-HRP

secondary antibody diluted 1:1000 in 3% BSA. Blots were left in secondary antibody at

Page 91: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

91

room temperature for one hour, or overnight at 4°. After Tenascin C detection, blots

were stripped with mild stripping buffer and GAPDH (1:2000) was applied next. GAPDH

(6C5) antibody was purchased from Abcam.

Protein bands were detected using ECL chemiluminescence on film, and ImageJ

software was used to determine, based on densitometry, changes in Tenascin C

relative to GAPDH bands. Each lane represents one biological replicate, n=3 for each

genotype. Each biological replicate is defined as 10-14 pooled discs from the thoracic

and lumbar regions.

ECM Analysis By Immunofluorescence

The relative quantity and localization of matrix proteins was examined by

immunofluorescence using antibodies against proteins that are enriched in cartilaginous

tissues9-11. Vertebral columns were dissected in cold PBS and fixed overnight in 4%

PFA/PBS at 4°. The next day, tissues were washed in PBS and cryopreserved by

immersing in 30% sucrose/PBS overnight at 4°. Tissues were embedded in Tissue-Tek

OCT (Optimal Cutting Temperature) embedding compound on dry ice and then stored

at -80°. A cryostat was used to section embryos and vertebral columns onto Superfrost

Plus Gold Slides. All sections were 20um unless otherwise specified.

Slides were retrieved from -80° and allowed to thaw to room temperature. A

PAP-Pen was used to draw a hydrophobic barrier around the tissues. Tissues were

permeabilized with three washes of PBST and then blocked for one hour at room

temperature in 3% BSA dissolved in PBST. Unless otherwise specified, all primary

antibodies were diluted 1:200 in 3% BSA/PBST. Tissues were incubated with primary

antibody overnight at 4° in a humidified slide box. The next day, tissues were washed

Page 92: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

92

three times with PBST and then incubated with secondary antibody at room

temperature for one hour, or overnight at 4°, protected from light. Secondary antibodies

were diluted 1:200 in 3% BSA/PBST. After secondary antibody incubation, slides were

counterstained with DAPI nuclear stain. After a final round of PBST washes, slides were

coverslipped using Dako fluorescent mounting media and #1.5 glass coverslips. All

slides were stored at 4°, protected from light, and imaged on a Leica confocal

microscope. Images presented in figures are representative of multiple discs from the

thoracic and lumbar regions from at least two controls and two Robo1;2 null mice.

Characterization Of Robo And Slit Expression

X-Gal Staining

LacZ knockin into the Robo1 and Robo2 loci was used to generate the Robo1

and Robo2 null alleles. This allowed us to use Xgal staining to show Robo expression in

the vertebral column. Importantly, because both Robo1 and Robo2 were replaced by

LacZ, X-gal staining is a proxy for Robo1 and/or Robo2 expression. Vertebral columns

from Robo1+/-;Robo2+/- mice were indistinguishable from vertebral columns from

wildtype (Robo1+/+;Robo2+/+) mice. Thus, Robo1+/-:Robo2+/- embryos were used for

Xgal staining experiments. Staining was done as previously described4 (Harfe et al.

2004).

Skinned embryos (14.5 dpc and younger) were fixed overnight in 0.2% PFA.

Mice older than 14.5 dpc were additionally eviscerated before fixation. The next day,

embryos were prepared for X-gal staining with three washes in PBS. Tissues were

stained overnight at room temperature, protected from light. Where applicable, wildtype

embryos and tissues were used as negative controls for endogenous b-galactosidase

Page 93: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

93

activity in the skeleton. After staining, tissues were washed in PBS and then fixed in 4%

PFA/PBS. X-gal substrate was prepared as 20mg/ml in DMF and used at a final

concentration of 1mg/ml.

Perinatal vertebral columns were additionally decalcified in Cal-Ex Decalcifier

overnight at room temperature and then washed in PBS. For sectioning, tissues were

either prepared for vibatome sectioning (100um) or microtome sectioning (10um).

Tissues that were vibratome sectioned were embedded in 6% agarose on ice after

overnight incubation in 30% sucrose. Tissues that were microtome sectioned were

prepared for paraffin embedding as described under Histology, above.

RNA In Situ Hybridization

All harvests and dissections for in situ hybridization experiments were done with

RNase Zap cleaned tools and work surfaces to minimize RNA degradation. Buffers

were DEPC-treated and kept on ice during dissections and incubations. Embryos were

harvested and dissected in cold DEPC-PBS and fixed overnight in 4% PFA prepared in

DEPC-PBS. Tissues were cryopreserved in 30% sucrose prepared in DEPC-PBS and

embedded on dry ice using OCT embedding media. Unless otherwise specified, tissues

were sectioned on a cryostat at 20um onto Superfrost Plus Gold glass slides and stored

at -80°. Following in situ hybridization, tissues were fixed in 4% PFA, PBS washed, and

mounted with Glycergel (Dako).

RNA probe templates for Robo1, Robo2, and Slit1-3 were generously provided

by Dr. Xin Sun5. DIG labeled probes were made from cDNA templates as previously

described6 with minor modifications. Briefly, DIG labeled RNA probes were synthesized

Page 94: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

94

by in vitro transcription using DIG RNA Labeling kit (Roche). Probes were purified using

Mini Quick Spin RNA columns (Roche) and then stored at -80°.

RNA in situ hybridization experiments were conducted as previously described7

with minor modifications. Briefly, on day 1 tissues mounted on slides were retrieved

from -80° and allowed to come to room temperature. A PAP pen was used to

circumscribe tissues with a hydrophobic barrier before tissues were put through a series

DEPC-PBST washes in preparation for probe incubation. Tissues were postfixed in 4%

PFA for 20 minutes at room temperature and incubated with prehybe (10% dextran

sulfate) for one hour at 65°. Probes were diluted into 100ul fresh, pre-warmed prehybe

before being added to tissues for overnight incubation at 65°. Tissues were carefully

covered with parafilm and incubated in clean, humidified slide boxes to prevent

evaporation during overnight hybridization. On day 2, slides were put through a series

SSC/formamide washes at 65° to remove any excess, unhybridized probe. In

preparation for antibody incubation, slides were brought to room temperature in KTBT

and then blocked for one hour at room temperature in 20% HINGS (heat-inactivated

goat serum, 20% by volume in KTBT). After blocking, tissues were incubated at 4°

overnight with an AP-conjugated anti-DIG antibody (Roche) diluted 1:2000 in 20%

HINGS. On day 3, slides were washed 6x in KTBT at room temperature, and then left in

KTBT overnight at 4°. Hybridized tissues were developed with either BM Purple (Roche)

or with BCIP/NBT (final concentration 0.175mg/ml for BCIP and 0.225mg/ml for NBT).

Both reagents are substrates for alkaline phosphate, which produces a purple (BM

Purple) or blue (BCIP/NBT) precipitate that can be visualized by light microscopy. Slides

Page 95: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

95

were PBS washed and postfixed in 4% PFA before being mounted with Glycergel

(Dako) for imaging.

Lineage Tracing Analyses

The Rosa-mT/mG dual fluorescent reporter8 allele was used to lineage trace the

origin of cells in the P0 vertebral column. ShhCre4 was used to mark notochord-derived

cells in the nucleus pulposus. For lineage tracing studies, P0 mice were collected from

Robo1+/-;Robo2+/-;Rosa-mT/mG females mated to Robo1+/-;Robo2+/-;ShhCre males

and prepared for cryostat sectioning. Briefly, vertebral columns were dissected into cold

PBS and fixed overnight in 4% PFA/PBS. Tissues were then washed in PBS and

submersed in 30% sucrose overnight. The next day, vertebral columns were embedded

on dry ice using OCT embedding media. Tissues were sectioned at 20um on a cryostat.

For imaging, tissues were counterstained with DAPI nuclear stain and mounted using

Dako fluorescent mounting media. Images were taken on a Leica confocal microscope.

For lineage tracing of notochord cells at 10.5dpc, the ShhCreERT24 allele was

used to selectively mark Shh expressing cells between 10.5dpc and 12dpc. In these

experiments, Robo1+/-;Robo2+/-;ShhCreERT2 males were mated to Robo1+/-

;Robo2+/-;Rosa-mT/mG females. The appearance of a vaginal plug was designated as

embryonic day 0.5 (0.5dpc). Tamoxifen dissolved in corn oil was administered by oral

gavage at 10.5dpc and then pups were collected at P0. Tissues were prepared for

analysis as described above.

Page 96: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

96

Analysis Of Sclerotomal Proliferation

EdU Pulse And Detection

Time mated Robo1+/-;Robo2+/- females crossed to Robo1+/-;Robo2+/- males were

used to evaluate the effect of Robo loss on sclerotome proliferation. Analysis of EdU

proliferation in skeletal tissues was performed as described previously12. Briefly,

pregnant dams were weight at 13.5dpc for EdU injection by IP. Embryos were

harvested after one hour. For analysis of 18.5dpc tissues, EdU pulse was extended to

two hours.

Embryos and vertebral columns were dissected in PBS and fixed overnight in 4%

PFA/PBS at 4°. Robo1+/-;Robo2+/- control and Robo1-/-;Robo2-/- mutant littermates were

selected for further processing. Embryos were cryopreserved in 30% sucrose/PBS

overnight and then embedded in OCT embedding medium on dry ice. Sagittal sections

from 13.5 dpc embryos were sectioned at 20um on a cryostat, and sections through the

medial vertebral column containing notochord and/or discs were selected for further

analysis.

For detection of EdU incoporation, Click-iT EdU Alexa Fluor 555 Imaging Kit was

purchased from Thermo Fisher Scientific and manufacturer‟s protocol was followed with

minor modifications. Briefly, tissues were permeabilized in PBST for 20 minutes, post-

fixed in 4% PFA/PBST for ten minutes at room temperature, and then washed in PBST.

Tissues were incubated with EdU reagent for ten minutes, protected from light, at room

temperature. Thereafter, the tissues were washed thoroughly in PBST and prepared for

immunofluorescence.

Page 97: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

97

β -Gal Antibody As Proxy For Robo Expression

After EdU labeling with AlexaFluor-555, slides were blocked with 3%BSA/PBST

for one hour at room temperature. Because Robo1 and Robo2 were replaced with LacZ

by knockin, lacZ transcript is transcribed from the Robo1 and Robo2 loci. The presence

of bGalactosidase protein, which is produced from the lacZ transcript, is accordingly an

appropriate proxy to show Robo expression domains. Thus, an antibody against β-

galactosidase was used as a proxy to mark Robo expression domains in the vertebral

column.

Primary antibody was diluted 1:200 in 3% BSA/PBST and tissues were incubated

overnight at 4° in a humidified slide box. A 488-conjugated secondary antibody was

selected for EdU quantification experiments. Secondary was incubated at one hour at

room temperature, protected from light. DAPI was used to counterstain nuclei and slides

were mounted using #1.5 glass coverslips and Dako fluorescent mounting medium.

Antibody information can be found in Table 1.

Quantification

Images were captured on a Leica confocal microscope. Effort was made to select

comparable regions from the trunk region containing Robo expressing tissues adjacent

to the notochord/disc. This was to ensure that quantification was based on proliferation

occurring in developmentally congruent regions since the vertebral column develops

rostro-caudally and rostral tissue is developmentally more advanced than caudal tissue.

Leica software was used to separate fluorescent images into separate channels

to quantify EdU-labeled and DAPI-labeled nuclei. Only nuclei within b-galactosidase-

488 labeled regions were counted. Quantification was done in a double-blind fashion. A

Page 98: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

98

numerical code was assigned to all images to conceal genotype information and a

research volunteer in the lab was asked to manually count DAPI and EdU labeled

nuclei.

One midline sagittal image from each embryo was used for quantification. Each

section contained 2-5 well-defined Robo expressing domains (by bGalatosidase-488

proxy), and these were used to define regions for quantification. Total number of EdU-

555 positive nuclei within the 488-positive domains was compared to total number of

DAPI labeled nuclei within the same 488-positive domains to calculate % proliferation in

each embryo. Three Robo1+/-;Robo2+/- controls and three Robo1-/-;Robo2-/- embryos

were used.

Percent proliferation in the notochord/discs, which do not express Robo and are

thus negative for bGalatosidase-488, was also calculated as a control for any potential

changes in overall mitotic index.

Growth Plate Analysis

Markers for growth plate chondrocyte organization were evaluated using

antisense RNA probes to Col10a1 and Ihh, which are expressed by hypertrophic and

pre-hypertrophic chondrocytes, respectively13. In situ hybridizations were performed as

described above.

Characterization of Wnt/β-Catenin Signaling Activity

TCF/Lef:H2B-GFP reporter mice were used to evaluate Wnt/β-Catenin signaling

activity in the notochord, somites, and postnatal intervertebral discs and vertebrae.

Tissues were fixed overnight 4%PFA/PBS at 4°, cryopreserved in 30% sucrose

overnight, embedded in OCT embedding medium, and sectioned on a cryostat.

Page 99: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

99

Postnatal tissues were additionally de-calcified overnight in 0.5M EDTA (pH 8) at 4°

before cryopreservation in 30% sucrose and embedding in OCT embedding medium. All

vertebral columns were sectioned through the sagittal plane at 20um or 40um. To

visualize GFP reporter activity, tissue sections were permeabilized in 0.1% PBST (0.1%

Tween-20 in PBS) and then counterstained with DAPI to show tissue architecture.

Tissues were mounted with DAKO fluorescent mounting media, and imaged on a Leica

confocal microscope.

Table 4-1. ECM Immunofluorscence And Western Blot Antibodies

Immunofluorescence Primary Antibodies

Aggrecan Millipore AB1031: polyclonal antibody against mouse aggrecan AA1177-1326

β-galactosidase Abcam: ab9361

Col1a1 Santa Cruz (D-13): sc-25974

Col2a1 Santa Cruz (C-19): sc-7763

Pax1 Abcam: ab95227

Slit2 GeneTex (FLJ14420): GTX118220

Tenascin C Santa Cruz (H-300): sc-20932

Tenascin C GeneTex (EPR4219): GTX62552

Tenascin C GeneTex (MTn-12): GTX26346)

T-brachyury Santa Cruz (N-19): sc-17743

Western blot primary antibodies

Tenascin C Santa Cruz (H-300): sc-20932

GAPDH Abcam (6C5): ab8245

Table 4-2. Genotyping Primers

Primers References

Robo1Null FwdCommon: TGGCACGAAGGTATATGTGC RevWT: GAAGGACTGGTGGTTTTGAG RevNull: CCTCCGCAAACTCCTATTTC

Long, et al.

Robo2Null FwdCommon: AAGTGCAACGTCTCTGAAGTCCC RevWT: GGCGGAATTCTTAATTAAGGCGCG RevNull: TTCTTTAGAAGGCACAACAATCTCAGAG

Grieshammer, et al.

Page 100: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

100

Robo2FLOXED FwdFloxed: CCAATCATAGTCTCTCCACG RevFloxed: CCTCTGATTCAATGAGATGC

Lu, et al.

Rosa-mT/mG Fwd: CTCTGCTGCCTCCTGGCTTCT RevWT: CGAGGCGGATCACAAGCAATA RevMTMG: TCAATGGGCGGGGGTCGTT

Mouse generated by Muzumdar, et al.

Allele Primers References

TCF4-GFP FwdGFP: ACAACAAGCGCTCGACCATCAC RevGFP: AGTCGATGCCCTTCAGCTCGAT

Mouse generated by Ferrer-Vaquer, et al.

Page 101: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

101

REFERENCES

Abbas J, Hamoud K, May H, Peled N, Sarig R, Stein D, Alperovitch-Najemson D, Hershkovitz I. Socioeconomic and physical characteristics of individuals with degenerative lumbar spinal stenosis. Spine (Phila Pa 1976) 2013, 38:E554-561.

Abdelkhalek HB, Beckers A, Schuster-Gossler K, Pavlova MN, Burkhardt H, Lickert H, Rossant J, Reinhardt R, Schalkwyk LC, Muller I, et al. The mouse homeobox gene Not is required for caudal notochord development and affected by the truncate mutation. Genes Dev 2004, 18:1725-1736.

Adham IM, Gille M, Gamel AJ, Reis A, Dressel R, Steding G, Brand-Saberi B, Engel W. The scoliosis (sco) mouse: a new allele of Pax1. Cytogenet Genome Res 2005, 111:16-26.

Akazawa H, Komuro I, Sugitani Y, Yazaki Y, Nagai R, Noda T. Targeted disruption of the homeobox transcription factor Bapx1 results in lethal skeletal dysplasia with asplenia and gastroduodenal malformation. Genes Cells 2000, 5:499-513.

Akiyama H, Chaboissier MC, Martin JF, Schedl A, de Crombrugghe B. The transcription factor Sox9 has essential roles in successive steps of the chondrocyte differentiation pathway and is required for expression of Sox5 and Sox6. Genes Dev 2002, 16:2813-2828.

Andersson GB. Epidemiological features of chronic low-back pain. Lancet 1999, 354:581-585.

Ang SL, Rossant J. HNF-3 beta is essential for node and notochord formation in mouse development. Cell 1994, 78:561-574.

Aoyama H, Asamoto K. The developmental fate of the rostral/caudal half of a somite for vertebra and rib formation: experimental confirmation of the resegmentation theory using chick-quail chimeras. Mech Dev 2000, 99:71-82.

Aoyama, H., Mizutani-koseki, S., Koseki, H. (2005) Three developmental compartments involved in rib formation. Int J Dev Biol. 49(2-3): 325-33.

Baffi MO, Moran MA, Serra R. Tgfbr2 regulates the maintenance of boundaries in the axial skeleton. Dev Biol 2006, 296:363-374.

Baffi MO, Slattery E, Sohn P, Moses HL, Chytil A, Serra R. Conditional deletion of the TGF-beta type II receptor in Col2a expressing cells results in defects in the axial skeleton without alterations in chondrocyte differentiation or embryonic development of long bones. Dev Biol 2004, 276:124-142.

Page 102: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

102

Bagnall KM. The migration and distribution of somite cells after labelling with the carbocyanine dye, Dil: the relationship of this distribution to segmentation in the vertebrate body. Anat Embryol (Berl) 1992, 185:317-324.

Barrionuevo F, Taketo MM, Scherer G, Kispert A. Sox9 is required for notochord maintenance in mice. Dev Biol 2006, 295:128-140.

Beddington RS, Rashbass P, Wilson V. Brachyury--a gene affecting mouse gastrulation and early organogenesis. Dev Suppl 1992:157-165.

Bi W, Deng JM, Zhang Z, Behringer RR, de Crombrugghe B. Sox9 is required for cartilage formation. Nat Genet 1999, 22:85-89.

Blockus, H, Chedotal, A. Slit-Robo signaling. Development, 2016, 143(17):3037-44.

Borycki AG, Brunk B, Tajbakhsh S, Buckingham M, Chiang C, Emerson CP, Jr. Sonic hedgehog controls epaxial muscle determination through Myf5 activation. Development 1999, 126:4053-4063.

Brent AE, Braun T, Tabin CJ. Genetic analysis of interactions between the somitic muscle, cartilage and tendon cell lineages during mouse development. Development 2005, 132:515-528.

Brent AE, Schweitzer R, Tabin CJ. A somitic compartment of tendon progenitors. Cell 2003, 113:235-248.

Brent AE, Tabin CJ. FGF acts directly on the somitic tendon progenitors through the Ets transcription factors Pea3 and Erm to regulate scleraxis expression. Development 2004, 131:3885-3896.

Bruggeman BJ, Maier JA, Mohiuddin YS, Powers R, Lo Y, Guimaraes-Camboa N, Evans SM, Harfe BD. Avian intervertebral disc arises from rostral sclerotome and lacks a nucleus pulposus: implications for evolution of the vertebrate disc. Dev Dyn 2012, 241:675-683.

Buchbinder R, Blyth FM, March LM, Brooks P, Woolf AD, Hoy DG. Placing the global burden of low back pain in context. Best Pract Res Clin Rheumatol 2013, 27:575-589.

Bussen M, Petry M, Schuster-Gossler K, Leitges M, Gossler A, Kispert A. The T-box transcription factor Tbx18 maintains the separation of anterior and posterior somite compartments. Genes Dev 2004, 18:1209-1221.

Buttitta L, Mo R, Hui CC, Fan CM. Interplays of Gli2 and Gli3 and their requirement in mediating Shh-dependent sclerotome induction. Development 2003, 130:6233-6243.

Page 103: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

103

Cadigan, K.M., Nusse, R. (1997) Wnt signaling: a common theme in animal development. Genes Dev. 11(24):3286-305.

Capellini TD, Zewdu R, Di Giacomo G, Asciutti S, Kugler JE, Di Gregorio A, Selleri L. Pbx1/Pbx2 govern axial skeletal development by controlling Polycomb and Hox in mesoderm and Pax1/Pax9 in sclerotome. Dev Biol 2008, 321:500-514.

Cerda J, Grund C, Franke WW, Brand M. Molecular characterization of Calymmin, a novel notochord sheath-associated extracellular matrix protein in the zebrafish embryo. Dev Dyn 2002, 224:200-209.

Chen, L., Liao, J., Klineberg, E., et al. Small leucine-rich proteoglycans (SLRPs): characteristics and function in the intervertebral disc. J Tissue Eng Regen Med, 2015.

Cheyette, B.N., Waxman, J.S., Miller, J.R., Takemaru, K., et al. (2002) Dapper, a Dishevelled-associated antagonist of beta-catenin and JNK signaling, is required for notochord formation. Dev Cell. 2(4): 449-61.

Chiang C, Litingtung Y, Lee E, Young KE, Corden JL, Westphal H, Beachy PA. Cyclopia and defective axial patterning in mice lacking Sonic hedgehog gene function. Nature 1996, 383:407-413.

Chiquet-Ehrismann, R., Kalla, P., Pearson, C., et al. (1988) Tenascin interferes with fibronectin action. Cell. 53(3): 383-390.

Choi KS, Harfe BD. Hedgehog signaling is required for formation of the notochord sheath and patterning of nuclei pulposi within the intervertebral discs. Proc Natl Acad Sci U S A 2011, 108:9484-9489.

Choi KS, Lee C, Harfe BD. Sonic hedgehog in the notochord is sufficient for patterning of the intervertebral discs. Mech Dev 2012, 129:255-262.

Christ B, Huang R, Scaal M. Formation and differentiation of the avian sclerotome. Anat Embryol (Berl) 2004, 208:333-350.

Christ B, Scaal M. Formation and differentiation of avian somite derivatives. Adv Exp Med Biol 2008, 638:1-41.

Cleaver O, Krieg PA. Notochord patterning of the endoderm. Dev Biol 2001, 234:1-12.

Colombier P, Clouet J, Hamel O, Lescaudron L, Guicheux J. The lumbar intervertebral disc: from embryonic development to degeneration. Joint Bone Spine 2014, 81:125-129.

Page 104: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

104

Corallo D, Trapani V, Bonaldo P. The notochord: structure and functions. Cell Mol Life Sci 2015.

Cortes DH, Elliott DM. Extra-fibrillar matrix mechanics of annulus fibrosus in tension and compression. Biomech Model Mechanobiol 2012, 11:781-790.

Cserjesi P, Brown D, Ligon KL, Lyons GE, Copeland NG, Gilbert DJ, Jenkins NA, Olson EN. Scleraxis: a basic helix-loop-helix protein that prefigures skeletal formation during mouse embryogenesis. Development 1995, 121:1099-1110.

Dahia CL, Mahoney E, Wylie C. Shh signaling from the nucleus pulposus is required for the postnatal growth and differentiation of the mouse intervertebral disc. PLoS One 2012, 7:e35944.

Dahia CL, Mahoney EJ, Durrani AA, C. Intercellular signaling pathways active during intervertebral disc growth, differentiation, and aging. Spine (Phila Pa 1976) 2009, 34:456-462.

Dario AB, Ferreira ML, Refshauge KM, Lima TS, Ordonana JR, Ferreira PH. The relationship between obesity, low back pain, and lumbar disc degeneration when genetics and the environment are considered: a systematic review of twin studies. Spine J 2015, 15:1106-1117.

DasGupta, R., Fuchs, E. (1999) Multiple roles for activated LEF/TCF transcription complexes during hair follicle development and differentiation. Development. 126(20):4557-68.

Dawes B. The development of the vertebral column in mammals, as illustrated by its development in Mus musculus. Philosophical Transactions of the Royal Society of London Series B-Containing Papers of a Biological Character 1930, 218:115-U122.

de Crombrugghe B, Lefebvre V, Behringer RR, Bi W, Murakami S, Huang W. Transcriptional mechanisms of chondrocyte differentiation. Matrix Biol 2000, 19:389-394.

Deries M, Schweitzer R, Duxson MJ. Developmental fate of the mammalian myotome. Dev Dyn 2010, 239:2898-2910.

Deutsch U, Dressler GR, Gruss P. Pax 1, a member of a paired box homologous murine gene family, is expressed in segmented structures during development. Cell 1988, 53:617-625.

DiPaola CP, Farmer JC, Manova K, Niswander LA. Molecular signaling in intervertebral disk development. J Orthop Res 2005, 23:1112-1119.

Page 105: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

105

Ewan KB, Everett AW. Evidence for resegmentation in the formation of the vertebral column using the novel approach of retroviral-mediated gene transfer. Exp Cell Res 1992, 198:315-320.

Fan CM, Lee CS, Tessier-Lavigne M. A role for WNT proteins in induction of dermomyotome. Dev Biol 1997, 191:160-165.

Fan CM, Tessier-Lavigne M. Patterning of mammalian somites by surface ectoderm and notochord: evidence for sclerotome induction by a hedgehog homolog. Cell 1994, 79:1175-1186.

Freemont AJ, Watkins A, Le Maitre C, Baird P, Jeziorska M, Knight MT, Ross ER, O'Brien JP, Hoyland JA. Nerve growth factor expression and innervation of the painful intervertebral disc. J Pathol 2002, 197:286-292.

Gotz W, Osmers R, Herken R. Localisation of extracellular matrix components in the embryonic human notochord and axial mesenchyme. J Anat 1995, 186 ( Pt 1):111-121.

Goulding MD, Chalepakis G, Deutsch U, Erselius JR, Gruss P. Pax-3, a novel murine DNA binding protein expressed during early neurogenesis. EMBO J 1991, 10:1135-1147.

Gruber HE, Ingram JA, Davis DE, Hanley EN, Jr. Increased cell senescence is associated with decreased cell proliferation in vivo in the degenerating human annulus. Spine J 2009, 9:210-215.

Gruber HE, Ingram JA, Norton HJ, Hanley EN, Jr. Senescence in cells of the aging and degenerating intervertebral disc: immunolocalization of senescence-associated beta-galactosidase in human and sand rat discs. Spine (Phila Pa 1976) 2007, 32:321-327.

Gruneberg H. Genetical studies on the skeleton of the mouse. XXIII. The development of brachyury and anury. J Embryol Exp Morphol 1958, 6:424-443.

Haque M, Ohata K, Takami T, Soares SB, Jr., Aree SN, Hakuba A, Hara M. Development of lumbosacral spina bifida: three-dimensional computer graphic study of human embryos at Carnegie stage twelve. Pediatr Neurosurg 2001, 35:247-252.

Harfe BD, Scherz PJ, Nissim S, Tian H, McMahon AP, Tabin CJ. Evidence for an expansion-based temporal Shh gradient in specifying vertebrate digit identities. Cell 2004, 118:517-528.

Page 106: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

106

Hargus G, Kist R, Kramer J, Gerstel D, Neitz A, Scherer G, Rohwedel J. Loss of Sox9 function results in defective chondrocyte differentiation of mouse embryonic stem cells in vitro. Int J Dev Biol 2008, 52:323-332.

Hay ED. The mesenchymal cell, its role in the embryo, and the remarkable signaling mechanisms that create it. Dev Dyn 2005, 233:706-720.

Hayes AJ, Isaacs MD, Hughes C, Caterson B, Ralphs JR. Collagen fibrillogenesis in the development of the annulus fibrosus of the intervertebral disc. Eur Cell Mater 2011, 22:226-241.

Henry SP, Liang S, Akdemir KC, de Crombrugghe B. The postnatal role of Sox9 in cartilage. J Bone Miner Res 2012, 27:2511-2525.

Herbrand H, Pabst O, Hill R, Arnold HH. Transcription factors Nkx3.1 and Nkx3.2 (Bapx1) play an overlapping role in sclerotomal development of the mouse. Mech Dev 2002, 117:217-224.

Herrmann BG. Action of the Brachyury gene in mouse embryogenesis. Ciba Found Symp 1992, 165:78-86; discussion 86-91.

Hestbaek L, Iachine IA, Leboeuf-Yde C, Kyvik KO, Manniche C. Heredity of low back pain in a young population: a classical twin study. Twin Res 2004, 7:16-26.

Hiyama, A., Sakai, D., Risbud, M.V., et al. (2013) Enhancement of Intervertebral Disc Cell Senescence by Wnt/β-Catenin Signaling-Induced Matrix Metalloproteinase Expression. Arthritis Rheum. 62(10):3036-3047.

Hohenester, E. Structural insight into Slit-Robo signaling. Biochem Soc Trans, 2008,

(36):251-6. Holmes, G., Niswander, L. Expression of slit-2 and slit-3 during chick development. Dev

Dyn, 2001, 222(2):301-7. Howitt, J.A., Clout, N.J., Hohenester, E. Binding site for Robo receptors revealed by

dissection of the leucine-rich repeat region of Slit. EMBO J, 2004, (22):4406-12.

Hui CC, Joyner AL. A mouse model of greig cephalopolysyndactyly syndrome: the extra-toesJ mutation contains an intragenic deletion of the Gli3 gene. Nat Genet 1993, 3:241-246.

Huang, W., Chiquet-Ehrismann, R., Moyano, J.V., et al. (2001). Interference of Tenascin-C with Syndecan-4 Binding to Fibronectin Blocks Cell Adhesion and Stimulates Tumor Cell Proliferation. Cancer Res. 61(23): 8586-94.

Page 107: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

107

Ikeda T, Kawaguchi H, Kamekura S, Ogata N, Mori Y, Nakamura K, Ikegawa S, Chung UI. Distinct roles of Sox5, Sox6, and Sox9 in different stages of chondrogenic differentiation. J Bone Miner Metab 2005, 23:337-340.

Jeong Y, Epstein DJ. Distinct regulators of Shh transcription in the floor plate and notochord indicate separate origins for these tissues in the mouse node. Development 2003, 130:3891-3902.

Johnson ZI, Shapiro IM, Risbud MV. Extracellular osmolarity regulates matrix homeostasis in the intervertebral disc and articular cartilage: evolving role of TonEBP. Matrix Biol 2014, 40:10-16.

Jostes B, Walther C, Gruss P. The murine paired box gene, Pax7, is expressed specifically during the development of the nervous and muscular system. Mech Dev 1990, 33:27-37.

Kaestner KH, Hiemisch H, Luckow B, Schutz G. The HNF-3 gene family of transcription factors in mice: gene structure, cDNA sequence, and mRNA distribution. Genomics 1994, 20:377-385.

Katz JN. Lumbar disc disorders and low-back pain: socioeconomic factors and consequences. J Bone Joint Surg Am 2006, 88 Suppl 2:21-24.

Kiani C, Chen L, Wu YJ, Yee AJ, Yang BB. Structure and function of aggrecan. Cell Res 2002, 12:19-32.

Kim, M., Fontelonga, T., Roesener, A.P., Lee, H., Gurung, S., et al. Motor neuron cell bodies are actively positioned by Slit/Robo repulsion and Netrin/DCC attraction. Dev Biol, 2015. 399(1):68-79.

Kimmel CB, Ballard WW, Kimmel SR, Ullmann B, Schilling TF. Stages of embryonic development of the zebrafish. Dev Dyn 1995, 203:253-310.

Koseki H, Wallin J, Wilting J, Mizutani Y, Kispert A, Ebensperger C, Herrmann BG, Christ B, Balling R. A role for Pax-1 as a mediator of notochordal signals during the dorsoventral specification of vertebrae. Development 1993, 119:649-660.

Kraus F, Haenig B, Kispert A. Cloning and expression analysis of the mouse T-box gene Tbx18. Mech Dev 2001, 100:83-86.

Lefebvre, V., Li, P., de Crombrugghe, B. (1998) A new long form of Sox5 (L-Sox5), Sox6 and Sox9 are coexpressed in chondrogenesis and cooperatively activate the type II collagen gene. EMBO J. 17(19):5718-5733.

Page 108: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

108

Le Maitre CL, Freemont AJ, Hoyland JA. Accelerated cellular senescence in degenerate intervertebral discs: a possible role in the pathogenesis of intervertebral disc degeneration. Arthritis Res Ther 2007, 9:R45.

Lehtonen E, Stefanovic V, Saraga-Babic M. Changes in the expression of intermediate filaments and desmoplakins during development of human notochord. Differentiation 1995, 59:43-49.

Leitges M, Neidhardt L, Haenig B, Herrmann BG, Kispert A. The paired homeobox gene Uncx4.1 specifies pedicles, transverse processes and proximal ribs of the vertebral column. Development 2000, 127:2259-2267.

Lettice L, Hecksher-Sorensen J, Hill R. The role of Bapx1 (Nkx3.2) in the development and evolution of the axial skeleton. J Anat 2001, 199:181-187.

Li Y, Litingtung Y, Ten Dijke P, Chiang C. Aberrant Bmp signaling and notochord delamination in the pathogenesis of esophageal atresia. Dev Dyn 2007, 236:746-754.

Liu H, Zhang C, Zhu S, Lu P, Zhu T, Gong X, Zhang Z, Hu J, Yin Z, Heng BC, et al. Mohawk promotes the tenogenesis of mesenchymal stem cells through activation of the TGFbeta signaling pathway. Stem Cells 2015, 33:443-455.

Lv, F.J., Peng, Y., Sun, Y., et al. (2016) Matrix metalloproteinase 12 is an indicator of intervertebral disc degeneration co-expressed with fibrotic markers. Osteoarthritis Cartilage. 24(10):1826-36.

Maier JA, Harfe BD. Nuclei pulposi formation from the embryonic notochord occurs normally in GDF-5-deficient mice. Spine (Phila Pa 1976) 2011, 36:E1555-1561.

Maier JA, Lo Y, Harfe BD. Foxa1 and Foxa2 are required for formation of the intervertebral discs. PLoS One 2013, 8:e55528.

Malandrino A, Lacroix D, Hellmich C, Ito K, Ferguson SJ, Noailly J. The role of endplate poromechanical properties on the nutrient availability in the intervertebral disc. Osteoarthritis Cartilage 2014, 22:1053-1060.

Mansouri A, Yokota Y, Wehr R, Copeland NG, Jenkins NA, Gruss P. Paired-related murine homeobox gene expressed in the developing sclerotome, kidney, and nervous system. Dev Dyn 1997, 210:53-65.

MacDonald, B.T., Tamai, K., He, X. (2009) Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev Cell. 17(1): 9-26.

Page 109: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

109

McCann MR, Tamplin OJ, Rossant J, Seguin CA. Tracing notochord-derived cells using a Noto-cre mouse: implications for intervertebral disc development. Dis Model Mech 2012, 5:73-82.

McMahon JA, Takada S, Zimmerman LB, Fan CM, Harland RM, McMahon AP. Noggin-mediated antagonism of BMP signaling is required for growth and patterning of the neural tube and somite. Genes Dev 1998, 12:1438-1452.

Merceron C, Mangiavini L, Robling A, Wilson TL, Giaccia AJ, Shapiro IM, Schipani E, Risbud MV. Loss of HIF-1alpha in the notochord results in cell death and complete disappearance of the nucleus pulposus. PLoS One 2014, 9:e110768.

Millennium WHOSGotBoMCatSotN. The burden of musculoskeletal conditions at the start of the new millennium. World Health Organ Tech Rep Ser 2003, 919:i-x, 1-218, back cover.

Mo R, Freer AM, Zinyk DL, Crackower MA, Michaud J, Heng HH, Chik KW, Shi XM, Tsui LC, Cheng SH, et al. Specific and redundant functions of Gli2 and Gli3 zinc finger genes in skeletal patterning and development. Development 1997, 124:113-123.

Murchison ND, Price BA, Conner DA, Keene DR, Olson EN, Tabin CJ, Schweitzer R. Regulation of tendon differentiation by scleraxis distinguishes force-transmitting tendons from muscle-anchoring tendons. Development 2007, 134:2697-2708.

Nasto LA, Ngo K, Leme AS, Robinson AR, Dong Q, Roughley P, Usas A, Sowa GA, Pola E, Kang J, et al. Investigating the role of DNA damage in tobacco smoking-induced spine degeneration. Spine J 2014, 14:416-423.

Neubuser A, Koseki H, Balling R. Characterization and developmental expression of Pax9, a paired-box-containing gene related to Pax1. Dev Biol 1995, 170:701-716.

Noel, L.S., Champion, B.R., Holley, C.L., et al. (1998) RoBo-1, a novel member of the urokinase plasminogen activator receptor/CD59/Ly-6/snake toxin family selectively expressed in rat bone and growth plate cartilage. J Biol Chem. 273(7):3878-83.

Olivera-Martinez I, Thelu J, Teillet MA, Dhouailly D. Dorsal dermis development depends on a signal from the dorsal neural tube, which can be substituted by Wnt-1. Mech Dev 2001, 100:233-244.

Ordan, E., Brankatschk, M., Dickson, B., et al. Slit cleavage is essential for producing an active, stable, non-diffusible short-range signal that guides muscle migration. Development, 2015. 142(8):1431-6.

Page 110: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

110

Paavola LG, Wilson DB, Center EM. Histochemistry of the developing notochord, perichordal sheath and vertebrae in Danforth's short-tail (sd) and normal C57BL/6 mice. J Embryol Exp Morphol 1980, 55:227-245.

Peters H, Neubuser A, Kratochwil K, Balling R. Pax9-deficient mice lack pharyngeal pouch derivatives and teeth and exhibit craniofacial and limb abnormalities. Genes Dev 1998, 12:2735-2747.

Peters H, Wilm B, Sakai N, Imai K, Maas R, Balling R. Pax1 and Pax9 synergistically regulate vertebral column development. Development 1999, 126:5399-5408.

Pryce BA, Watson SS, Murchison ND, Staverosky JA, Dunker N, Schweitzer R. Recruitment and maintenance of tendon progenitors by TGFbeta signaling are essential for tendon formation. Development 2009, 136:1351-1361.

Rashbass P, Wilson V, Rosen B, Beddington RS. Alterations in gene expression during mesoderm formation and axial patterning in Brachyury (T) embryos. Int J Dev Biol 1994, 38:35-44.

Richardson SM, Purmessur D, Baird P, Probyn B, Freemont AJ, Hoyland JA. Degenerate human nucleus pulposus cells promote neurite outgrowth in neural cells. PLoS One 2012, 7:e47735.

Riddle RD, Johnson RL, Laufer E, Tabin C. Sonic hedgehog mediates the polarizing activity of the ZPA. Cell 1993, 75:1401-1416.

Rodrigo I, Hill RE, Balling R, Munsterberg A, Imai K. Pax1 and Pax9 activate Bapx1 to induce chondrogenic differentiation in the sclerotome. Development 2003, 130:473-482.

Romgens AM, van Donkelaar CC, Ito K. Contribution of collagen fibers to the compressive stiffness of cartilaginous tissues. Biomech Model Mechanobiol 2013, 12:1221-1231.

Roussa E, Krieglstein K. Induction and specification of midbrain dopaminergic cells: focus on SHH, FGF8, and TGF-beta. Cell Tissue Res 2004, 318:23-33.

Saga, Y. The mechanism of somite formation in mice. Curr Opin Genet Dev 2012, 22(4):331-8.

Saraga-Babic M, Lehtonen E, Svajger A, Wartiovaara J. Morphological and immunohistochemical characteristics of axial structures in the transitory human tail. Ann Anat 1994, 176:277-286.

Page 111: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

111

Schneider PR, Buhrmann C, Mobasheri A, Matis U, Shakibaei M. Three-dimensional high-density co-culture with primary tenocytes induces tenogenic differentiation in mesenchymal stem cells. J Orthop Res 2011, 29:1351-1360.

Selleri L, Depew MJ, Jacobs Y, Chanda SK, Tsang KY, Cheah KS, Rubenstein JL, O'Gorman S, Cleary ML. Requirement for Pbx1 in skeletal patterning and programming chondrocyte proliferation and differentiation. Development 2001, 128:3543-3557.

Senthinathan B, Sousa C, Tannahill D, Keynes R. The generation of vertebral segmental patterning in the chick embryo. J Anat 2012, 220:591-602.

Shukunami C, Takimoto A, Oro M, Hiraki Y. Scleraxis positively regulates the expression of tenomodulin, a differentiation marker of tenocytes. Dev Biol 2006, 298:234-247.

Sivan SS, Hayes AJ, Wachtel E, Caterson B, Merkher Y, Maroudas A, Brown S, Roberts S. Biochemical composition and turnover of the extracellular matrix of the normal and degenerate intervertebral disc. Eur Spine J 2014, 23 Suppl 3:S344-353.

Sivan SS, Wachtel E, Roughley P. Structure, function, aging and turnover of aggrecan in the intervertebral disc. Biochim Biophys Acta 2014, 1840:3181-3189.

Smith LJ, Elliott DM. Formation of lamellar cross bridges in the annulus fibrosus of the intervertebral disc is a consequence of vascular regression. Matrix Biol 2011, 30:267-274.

Smith LJ, Nerurkar NL, Choi KS, Harfe BD, Elliott DM. Degeneration and regeneration of the intervertebral disc: lessons from development. Dis Model Mech 2011, 4:31-41.

Smits P, Lefebvre V. Sox5 and Sox6 are required for notochord extracellular matrix sheath formation, notochord cell survival and development of the nucleus pulposus of intervertebral discs. Development 2003, 130:1135-1148.

Smits P, Li P, Mandel J, Zhang Z, Deng JM, Behringer RR, de Crombrugghe B, Lefebvre V. The transcription factors L-Sox5 and Sox6 are essential for cartilage formation. Dev Cell 2001, 1:277-290.

Smolders, L. A., Meij, B. P., Onis, D., Riemers, F. M., et al. (2013) Gene expression profiling of early intervertebral disc degeneration reveals a down-regulation of canonical Wnt signaling and caveolin-1 expression: implications for development of regenerative strategies. Arthritis Res Ther. 15(1):R23.

Page 112: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

112

Sofat, N., Robertson, S.D., Hermansson, M., et al. (2012) Tenascin-C fragments are endogenous inducers of cartilage matrix degradation. Rheumatol Int. 32(9):2809-2817.

Stemple DL. Structure and function of the notochord: an essential organ for chordate development. Development 2005, 132:2503-2512.

Sugimoto Y, Takimoto A, Akiyama H, Kist R, Scherer G, Nakamura T, Hiraki Y, Shukunami C. Scx+/Sox9+ progenitors contribute to the establishment of the junction between cartilage and tendon/ligament. Development 2013, 140:2280-2288.

Sulik K, Dehart DB, Iangaki T, Carson JL, Vrablic T, Gesteland K, Schoenwolf GC. Morphogenesis of the murine node and notochordal plate. Dev Dyn 1994, 201:260-278.

Takahashi Y, Yasuhiko Y, Takahashi J, Takada S, Johnson RL, Saga Y, Kanno J. Metameric pattern of intervertebral disc/vertebral body is generated independently of Mesp2/Ripply-mediated rostro-caudal patterning of somites in the mouse embryo. Dev Biol 2013, 380:172-184.

Takimoto A, Mohri H, Kokubu C, Hiraki Y, Shukunami C. Pax1 acts as a negative regulator of chondrocyte maturation. Exp Cell Res 2013, 319:3128-3139.

Takimoto A, Oro M, Hiraki Y, Shukunami C. Direct conversion of tenocytes into chondrocytes by Sox9. Exp Cell Res 2012, 318:1492-1507.

Talbot, W.S., Trevarrow, B., Halpern, M.E., et al. (1995) A homeobox gene essential for zebrafish notochord development. Nature. 378(6553):150-7.

Tam PP, Trainor PA. Specification and segmentation of the paraxial mesoderm. Anat Embryol (Berl) 1994, 189:275-305.

Tamplin OJ, Cox BJ, Rossant J. Integrated microarray and ChIP analysis identifies multiple Foxa2 dependent target genes in the notochord. Dev Biol 2011, 360:415-425.

Tanaka M, Komuro I, Inagaki H, Jenkins NA, Copeland NG, Izumo S. Nkx3.1, a murine homolog of Ddrosophila bagpipe, regulates epithelial ductal branching and proliferation of the prostate and palatine glands. Dev Dyn 2000, 219:248-260.

Theiler K. Anatomy and development of the "truncate" (boneless) mutation in the mouse. Am J Anat 1959, 104:319-343.

Theiler K. The house mouse; development and normal stages from fertilization to 4 weeks of age. Berlin, New York,: Springer-Verlag; 1972.

Page 113: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

113

Tolofari SK, Richardson SM, Freemont AJ, Hoyland JA. Expression of semaphorin 3A and its receptors in the human intervertebral disc: potential role in regulating neural ingrowth in the degenerate intervertebral disc. Arthritis Res Ther 2010, 12:R1.

Tribioli C, Lufkin T. The murine Bapx1 homeobox gene plays a critical role in embryonic development of the axial skeleton and spleen. Development 1999, 126:5699-5711.

Tylzanowski P, Mebis L, Luyten FP. The Noggin null mouse phenotype is strain dependent and haploinsufficiency leads to skeletal defects. Dev Dyn 2006, 235:1599-1607.

Ukita, K., Hirahara, S., Oshima, N., Imuta, Y., et al. (2009) Wnt signaling maintains the notochord fate for progenitor cells and supports the posterior extension of the notochord. Mech Dev. 126(10):791-803.

Urban, J., Roberts, S. (2003) Degeneration of the intervertebral disc. Arthritis Res Ther.

5(3):120-130. Usami, Y., Gunawardena, A.T., Iwamoto, M., Enomoto-Iwamoto, M. (2016). Wnt

signaling in cartilage development and diseases: lessons from animal studies. Lab Invest. 96(2): 186-96.

Wallin J, Wilting J, Koseki H, Fritsch R, Christ B, Balling R. The role of Pax-1 in axial skeleton development. Development 1994, 120:1109-1121.

Weinstein DC, Ruiz i Altaba A, Chen WS, Hoodless P, Prezioso VR, Jessell TM, Darnell JE, Jr. The winged-helix transcription factor HNF-3 beta is required for notochord development in the mouse embryo. Cell 1994, 78:575-588.

White TD, Black MT, Folkens PA. Human osteology. 3rd ed. San Diego, Calif.: Academic Press; 2012.

Wilkinson DG, Bhatt S, Herrmann BG. Expression pattern of the mouse T gene and its role in mesoderm formation. Nature 1990, 343:657-659.

Wilm B, Dahl E, Peters H, Balling R, Imai K. Targeted disruption of Pax1 defines its null phenotype and proves haploinsufficiency. Proc Natl Acad Sci U S A 1998, 95:8692-8697.

Winkler, T., Mahoney, E.J., Sinner, D., Wylie, C.C., Dahia, C.L. (2014) Wnt Signaling Activates Shh Signaling in Early Postnatal Intervertebral Discs, and Re-Activates Shh Signaling in Old Discs in the Mouse. PLoS One. 9(6):e98444.

Page 114: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

114

Yamanaka Y, Tamplin OJ, Beckers A, Gossler A, Rossant J. Live imaging and genetic analysis of mouse notochord formation reveals regional morphogenetic mechanisms. Dev Cell 2007, 13:884-896.

Yamashita S, Andoh M, Ueno-Kudoh H, Sato T, Miyaki S, Asahara H. Sox9 directly promotes Bapx1 gene expression to repress Runx2 in chondrocytes. Exp Cell Res 2009, 315:2231-2240.

Yang, X.R., Ng, D., Alcorta, D.A., et al. (2009) T (Brachyury) gene duplication confers major susceptibility to familial chordoma. Nat Genet. 41(11):1176-8.

Ypsilanti, A.R., Zagar, Y., Chedotal, A. Moving away from the midline: new developments for Slit and Robo. Development, 2010. 137(12):1939-52.

Yusuf F, Brand-Saberi B. The eventful somite: patterning, fate determination and cell division in the somite. Anat Embryol (Berl) 2006, 211 Suppl 1:21-30.

Zhang XM, Ramalho-Santos M, McMahon AP. Smoothened mutants reveal redundant roles for Shh and Ihh signaling including regulation of L/R symmetry by the mouse node. Cell 2001, 106:781-792.

Zhao, J., Li, S., Trilok, S., et al. (2014) Small molecule-directed specification of sclerotome-like chondroprogenitors and induction of a somitic chondrogenesis program from embryonic stem cells. Development. 141(20):3848-58.

Zizic Mitrecic M, Mitrecic D, Pochet R, Kostovic-Knezevic L, Gajovic S. The mouse gene Noto is expressed in the tail bud and essential for its morphogenesis. Cells Tissues Organs 2010, 192:85-92.

Page 115: By LISA Y. LAWSON - University of Floridaufdcimages.uflib.ufl.edu › UF › E0 › 05 › 06 › 88 › 00001 › LAWSON_L.pdf · economic burden associated with low back pain in

115

BIOGRAPHICAL SKETCH

Lisa Lawson moved to the United States in 1990 from South Korea. She grew up

in Central Florida and graduated from Winter Park High School in 2003. She completed

her undergraduate studies at the University of Notre Dame from 2003-2007. After

completing her Bachelor of Science, she received a Master of Medical Sciences from

the University of South Florida. In 2009 she joined the Dickey lab at the University of

South Florida to work on Alzheimer‟s research, assisting in research on how autophagy

pathways can be manipulated to ameliorate tauopathies.In 2011, Lisa enrolled at the

University of Florida as a doctoral student to study vertebrate embryogenesis under the

tutelage of Brian Harfe.


Recommended