expression analysis of cthrc1 in the murine embryo during ...€¦ · 1.1.3 facial prominences and...

80
Expression Analysis of CTHRC1 in the Murine Embryo during Mid-facial Development by Dr. Laurene Dao-Pei Yen A thesis submitted in conformity with the requirements for the degree of Masters of Science in Dentistry (Orthodontics) Department of Orthodontics University of Toronto © Copyright by Laurene Dao-Pei Yen, 2014

Upload: others

Post on 22-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

Expression Analysis of CTHRC1 in the Murine Embryo during

Mid-facial Development

by

Dr. Laurene Dao-Pei Yen

A thesis submitted in conformity with the requirements

for the degree of Masters of Science in Dentistry (Orthodontics)

Department of Orthodontics

University of Toronto

© Copyright by Laurene Dao-Pei Yen, 2014

Page 2: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

ii

Expression Analysis of CTHRC1 in the Murine Embryo during Mid-

facial Development

Dr. Laurene Dao-Pei Yen

Masters of Science in Dentistry (Orthodontics)

Department of Orthodontics

University of Toronto

2014

Abstract

Collagen Triple Helix Repeat Containing 1 (CTHRC1) has been shown to regulate collagen

expression, cell migration and interact with the Wnt/planar cell polarity pathway. Objective:

Determine CTHRC1 expression in the midface of mouse embryos during craniofacial

development. Methods: qPCR and immunohistochemistry were used to determine the

expression of Cthrc1 at embryonic (E) days of development E8.5-E13.5. Results: CTHRC1

expression was in the notochord and neural tube at E8.5, mesenchyme of the midface at E9.5-

E10.5 and areas of cartilage formation at E11.5-E13.5. Conclusions: The expression of

CTHRC1 in the developing craniofacial region suggests a role of CTHRC1 in migration of

cranial neural crest cells and in chondrocyte proliferation and differentiation. We speculate the

functions of CTHRC1 during craniofacial development are via its interactions with the Wnt/PCP

pathway, shown previously to play a significant role during craniofacial development. Support:

Donald G. Woodside Fund and NSERC.

Page 3: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

iii

Acknowledgments

I would like to thank my supervisor, Dr. Siew-Ging Gong for your guidance and support

throughout my thesis. Thank you for the constant encouragement, especially during the writing

of my thesis.

I would also like to express my sincere thanks to the members of my committee, Dr. Bernhard

Ganss and Dr. Bryan Tompson for their scientific guidance and advice throughout my MSc

degree.

I would like to thank the Donald G. Woodside Fund for funding part of my project.

A special thanks to Mr. James Holcroft, for his help with the quantitative PCR portion of my

thesis, and Feryal Sarraf, for her technical expertise in the use of the microscopes and digital

cameras in the histology lab.

Thank you to my husband, Dr. Alexander Unterberger¸ for his constant support and

encouragement throughout my degree. I could not have done it without you!

Page 4: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

iv

Table of Contents

Abstract …ii

Acknowledgments…iii

List of Figures…viii

Chapter 1: Introduction…1

1.1 Development and morphogenesis of the vertebrate face…1

1.1.1 Role of neural crest cells in craniofacial development…1

1.1.2 Formation and migration of cranial neural crest cells…2

1.1.2.1 Induction and specification of CNCCs and NCCs…2

1.1.2.2 Delamination…3

1.1.2.3 Migration…3

1.1.2.4 NCC positional identity…5

1.1.2.5 NCC differentiation and proliferation…6

1.1.3 Facial prominences and CNCC derivatives of the face…6

1.1.4 Facial development and facial prominences…7

1.2 Craniofacial developmental anomalies…10

1.3 Proteins and genes involved in formation of craniofacial structures…11

1.3.1 Fibroblast growth factors and bone morphogenetic proteins…12

1.3.2 Sonic hedgehog…13

1.3.3 Wnt pathways…14

1.3.3.1Wnt pathways and craniofacial development…17

Page 5: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

v

1.3.3.2 Wnt5A…18

1.4 Collagen Triple Helix Repeat Containing 1(Cthrc1)…20

1.4.1 Background…20

1.4.2 CTHRC1 structure…21

1.4.3 Expression pattern of Cthrc1…22

1.4.4 Possible functions of Cthrc1…22

1.4.4.1 Cthrc1 role in collagen deposition…23

1.4.4.2 Role of Cthrc1 in osteogenesis, osteoclastic bone formation and bone

remodeling…24

1.4.4.3 Role in cell motility and tissue repair…27

1.4.5 Genes and signalling pathways involved with CTHRC1…28

1.4.5.1 Cthrc1 activation of Wnt/PCP pathway…28

1.4.5.2 Cell-specific action of Cthrc1…30

1.5 Rationale for study…31

1.6 Hypothesis/Aims…31

Chapter 2: Materials and Methods…32

2.1 Embryos…32

2.2 RT-qPCR…32

2.2.1 RNA isolation…32

2.2.2 Reverse transcription of RNA samples…33

2.2.3 Quantitative PCR…33

Page 6: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

vi

2.3 Histological processing and Paraffin Embedding…34

2.4 Immunohistochemistry…34

2.5 Documentation and analysis of Cthrc1 expression…35

Chapter 3: Results…36

3.1 Quantitative expression of Cthrc1 mRNA transcripts during midface development…36

3.2 Expression of CTHRC1 protein…37

3.2.1 CTHRC1 peptide competition assay…37

3.2.2 Spatial expression of CTHRC1 in the midface at different developmental

stages…38

3.2.3 CTHRC1 expression in the mandible and developing tooth bud…43

Chapter 4: Discussion…45

4.1 Summary of Cthrc1 expression during mouse embryo midface development…45

4.1.1 Summary of qPCR mRNA levels of Cthrc1 mRNA during E8-5-E13.5…45

4.1.2 Summary of immunolocalization experiment results of CTHRC1 protein from E8.5-

E13.5…46

4.2 Involvement of Cthrc1 in midface development…47

4.2.1 Role of Cthrc1 in cell migration…47

4.2.2 Role of Cthrc1 in epithelial/mesenchyme interactions…48

4.2.3 Role of Cthrc1 in regulation of collagen formation and deposition…50

4.2.4 Role of Cthrc1 in bone formation…53

4.2.5 Summary of roles of Cthrc1 in midface development…55

Page 7: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

vii

4.3 Future Directions for Cthrc1 studies…56

4.3.1 Midface development in Cthrc1 knockout models…56

4.3.2 Cthrc1 expression in mouse models specifically targeting effects on midface

development…57

4.3.3 Colocalization studies of CTHRC1 with other proteins with a known function in

midface development…57

4.4 Conclusions…58

References…59

Copyright Acknowledgements…68

Page 8: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

viii

List of Figures

Figure 1: Migration and skeletal fates of the CNCCs…4

Figure 2: A schematic diagram of the pharyngeal arches…7

Figure 3: The canonical Wnt signaling pathway…15

Figure 4: The non-canonical Wnt signaling pathway…17

Figure 5: Summary of WNT5A functions during cartilage development and disease….20

Figure 6: The structure of the CTHRC1 protein…21

Figure 7: A model of selective activation of the Wnt/PCP pathway by CTHRC1…30

Figure 8: Temporal expression profile of Cthrc1 mRNA across E8.5-E13.5…36

Figure 9: Peptide competition assay on consecutive coronal sections through the anterior part of

the midface in an E13.5 embryo…37

Figure 10: CTHRC1 peptide expression in E8.5…38

Figure 11: CTHRC1 protein expression in E10.5 embryos…39

Figure 12: CTHRC1 protein expression in E11.5…40

Figure 13: CTHRC1 protein expression at E12.5…41

Figure 14: CTHRC1 protein expression at E13.5…42

Figure 15: CTHRC1 protein expression in tooth bud formation…44

Page 9: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

1

Chapter 1

Introduction

Development of the craniofacial region is a complex process. It is regulated by different genes,

proteins and signalling pathways. In this chapter, the development and morphogenesis of the

vertebrate face and the involvement of neural crest cells will be discussed. These topics will be

followed by the sequence of formation of the facial structures and a discussion of craniofacial

developmental anomalies.

1.1 Development and morphogenesis of the vertebrate face

1.1.1 Role of neural crest cells in craniofacial development

Development and morphogenesis of the vertebrate face is a complex and highly orchestrated

process. A major population of cells that contribute to the craniofacial region is the neural crest

cells (NCCs) that form along the entire length of the developing embryo. NCCs are transient

migratory cells that develop at the border between the neural plate (NP) and the epidermis. NCCs

delaminate from their site of origin along the dorsal neural tube and migrate ventrally along

different pathways. NCCs contribute to many different cell types and tissues in the body, such as

the enteric nervous system, melanocytes, connective tissues, and myofibroblasts which line

blood vessels, neurons and glia of the peripheral nervous system, pigment cells, endocrine cells,

cardiac structures, smooth muscle cells and tendons (1). NCCs in the rostral region of the

developing embryo are known as cranial neural crest cells (CNCCs). In contrast to trunk NCCs,

CNCCs can give rise to osteoblasts and chondrocytes (2) and are the major contributor to

structures of the facial skeleton, cartilages and bones of the jaws, middle ear and neck, cranial

nerves, smooth muscles, dermis and facial connective tissues (3).

During the migration of the CNCCs and after their arrival, their interactions with the surrounding

ectoderm, endoderm and neuroectoderm are integral for normal development of the face (4-6).

This multi-step process is regulated both temporally and spatially and is made up of the

coordinated action of numerous genes and signalling pathways (7). Due to this high level of

regulation, craniofacial development is susceptible to perturbations, resulting in facial

dysmorphologies. Many birth defects associated with craniofacial malformations can be

attributed to the defects in the generation, proliferation, migration and differentiation of CNCCs

Page 10: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

2

(8). Understanding the complex and sequential mechanisms of craniofacial development and

how they relate to the formation of facial structures is essential to the orthodontic specialty.

1.1.2 Formation and migration of cranial neural crest cells

The development of the NCCs and CNCCs can be divided into a number of stages:

a) Induction

b) Delamination

c) Migration

d) Differentiation

1.1.2.1 Induction and Specification of CNCCs and NCCs

Induction of NCCs is a multistep process resulting in a molecular cascade of events involved in

establishing their migratory and multipotent characteristics. The origin of NCCs can be traced to

the border of the NP in a region of ectoderm situated between the NP and the non-neural

ectoderm (NNE). Immediately beneath the ectoderm there is a layer of mesoderm, and together

with the NP and NNE, these tissues are collectively believed to contribute to the induction of the

NC. Induction of NCCs has been shown as early as during gastrulation during embryonic

development (9).

Induction involves a complex set of extracellular signals which transform the fate of cells along

the medio-lateral and anterior-posterior axes of the embryo (10). The signals that position the

NCCs along the axes are released from the NP, the epidermis and the lateral mesoderm. For

complete induction of NCCs, there must be a transformation of the naive ectoderm into the

neural crest. A precise combination of extracellular signalling molecules operate to activate the

expression of transcription factors that define the neural crest territory and control subsequent

neural crest development (11). Molecules such as wingless-type MMTV integration site

(WNTs), Notch, bone morphogenetic proteins (BMPs) and fibroblast growth factors (FGFs) are

secreted from the adjacent epidermis and underlying mesoderm and separate the NNE

(epidermis) from the NP during neural induction (12-14). The induction of the expression of

regulatory transcription factors (Msx1/2, Pax3/7, Zic1, Dlx3/5, Hairy2, Id3, Ap2) specify the NP

border that subsequently trigger the expression of NC specifiers, a second set of transcription

factors (Snail2, FoxD3, Sox9/10, Twist, cMyc, and Ap2). NC specifiers are proposed to

Page 11: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

3

ultimately control neural crest behavior, from epithelial-mesenchymal transition (EMT) and

delamination to migration and differentiation (15).

One theory proposes that NCCs are specified during gastrulation (9) whereas the second, more

classical, theory postulates that NCCs are specified during the time of neural tube closure. After

specification, contact-mediated signalling between tissues in the dorsal neural tube results in

EMT in neural ectoderm cells at the neuroectoderm and NNE border (1).

1.1.2.2 Delamination

After induction and specification at the neuroepithelium, NCCs undergo EMT and leave their

site of origin through a delamination process. Delamination is the splitting of the NCCs from

their surrounding tissues whereas EMT is a series of events at the molecular level which

orchestrates a change from an epithelial to mesenchymal phenotype (11, 16). EMT is a multi-

step process in which the NCCs lose their apico-basal polarity and disassemble intercellular

adhesion complexes required for epithelial formation (16), allowing the NCCs to separate from

the neuroepithelium and ectoderm (11). Contact-mediated signaling between tissues in the dorsal

neural tube stimulates cells at the neural/non-neural border to undergo EMT(17). This results in a

highly invasive phenotype characteristic of NCCs, behaviour which is also shared with

metastatic cells (18).

In the cranial region, CNCCs delaminate all at once, whereas the trunk NCCs delaminate

progressively, leaving the neuroepithelium one by one after neural tube closure (19).

1.1.2.3 Migration

The CNCCs follow migratory pathways that are conserved among vertebrate species. CNCC

migration is directed along well-defined routes that end in the ventral part of the brain and

branchial arches (20). Interactions between the CNCCs and their local environment are critical in

CNCC directional and collective migration (21). A number of cellular mechanisms have been

shown to operate during the directed migration of the NCCs. NCCs become polarized in the

direction of their migration with a tail at the back of the cell and filopodia and lamellipodia at the

side towards their migration. A mechanism known as contact inhibition of locomotion (CIL)

exists when two migrating NCCs make contact and they retract their protrusions and change

direction (22, 23). The non-canonical WNT planar cell polarity (PCP) signalling pathway

Page 12: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

4

(section 1.3.3) and cell-cell contact are crucial in controlling the polarity of migrating NCCs (8).

Even though the NCCs experience CIL when they contact each other, they also tend to migrate

in large groups, more so than what would be predicted given the CIL phenomenon. This is a

result of a phenomenon called co-attraction, which allows the collective migration of NCCs

despite low cell adhesion and dispersion due to CIL (14). NCCs have also been shown to move

around barriers introduced into the migration path and can re-target their direction (24).

The migration of CNCCs proceeds along a well-defined route, ending in the ventral part of the

brain and the pharyngeal arches (PA) (25). NCCs from the diencephalon and anterior

mesencephalon migrate into the frontonasal process (FNP) and NCCs from the posterior

mesencephalon and hindbrain migrate to the PAs (8). CNCCs in the hindbrain region are

compartmentalized in 7 distinct regions called rhombomeres (r). At first, the CNCCs migrating

to the PAs migrate as a continuous wave but then they split into three distinct segregated streams

(8). CNCCs from the posterior mesencephalon, r1 and r2 fill the first PA (PA1) and NCCs from

r4 fill pharyngeal arch 2 (PA2) (Figure 1 ) (3, ). In the post-otic hindbrain, NCCs from r6-r8

colonize PA3-6 (21). The subpopulations of NCCs that migrate to and target the PAs migrate in

stereotypical streams, maintaining a spatial segregation. This segregation has an important

impact on craniofacial patterning and the early anteroposterior patterning of NCCs by

establishing the segmental pattern in which the pharyngeal region of the vertebrate head is

formed (26, 27).

Page 13: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

5

Figure 1: Migration and skeletal fates of the CNCCs. A) Embryo showing the colonization of the

head and pharyngeal arches by fore-, mid- and hindbrain NCCs. There are 7 segments in the

hindbrain known as rhombomeres (r) from which crest cells emigrate in three major paths to the

pharyngeal or branchial arches (BA), coded in blue, yellow and green. B) A skull drawing

showing comparative contributions of the NCC populations to cranial skeletal elements of the

human. The major bones are coded to match the origin of the contributing migratory neural crest

streams. Reproduced from Gong, 2014. (3, 25).

To allow proper positioning in the PA and the proper assembly of structures, the subpopulations

of NCCs are guided by a complex set of cues to which they respond to locally during their

migration (28). From the dorsal neural tube into the craniofacial region the NCCs follow well-

marked paths by communication with the surrounding neural, facial and pharyngeal epithelia and

cephalic mesoderm (17). Examples of signalling molecules that guide the NCCs include: FGF-2

and FGF-8, which function in a chemotactic manner (29); Ephrins, which are ligands for Eph

receptors (30); and semaphorin proteins (31).

1.1.2.4 NCC positional identity

The positional identity of NCCs is regulated by the homeodomain (HD) transcription factors of

the homeobox gene family (Hox) (32, 33). The HD expression is induced and maintained in

NCCs in later developmental stages via signals from the surrounding local environment (8).

Anteroposterior NCC positional identity is thought to be acquired pre-migration (34, 35);

however, it is not permanent and there is some degree of plasticity. For example, intrinsic

molecular programs of the NCCs can be switched to new programs if exposed to ectopic

environmental cues (34, 36, 37). The involvement of the Hox genes was first shown in a mouse

model where targeted inactivation of Hox2a resulted in homeotic transformation of PA2

elements into PA1-like skeletal elements (38). Previous experiments by Santagati et al. (28) and

Pasqualetti et al. (39) in Xenopus demonstrated that the skeletal pattern of mandibular and hyoid

crest is not irreversibly committed before migration of NCC. However, positional information

has to be maintained throughout the post-migratory states in order to provide information about

size, shape and orientation of PA2 skeletal elements.

Page 14: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

6

Dorso-ventral axis patterning information is regulated by the NCCs through the Dlx homeobox

code. The involvement of Dlx genes has been investigated mainly through loss of function

mutations in the mouse (40). Namely, the partitioning of PA1 is achieved with the two Dlx

combinations; Dlx1/2 for the maxilla and Dlx 1/2/5/6 for the mandibular process. Mutations of

Dlx1 or Dlx2 primarily affect the maxilla and mutations of Dlx 5 affect the mandibular process

(41-43).

It is clear the diversity of NCC-derived skeletal elements among vertebrates arises from

modification of the levels, timing or spatial expression of HD factors. These factors play a

crucial role in spatial identity and modifying the expression of pattern genes can result in

aberrant development of structures (8).

1.1.2.5 NCC differentiation and proliferation

Once the NCCs have arrived in the facial prominences, they begin to proliferate and form the

facial structures. Many factors regulate the proliferation of NCCs, such as the Sonic hedgehog

(SHH) protein and WNT ligands. Disruption of these factors can affect the process of

craniofacial morphogenesis and cause aberrant proliferation and morphogenesis of NCCs (4).

There are two theories as to how NCCs determine which structures to make once they migrate to

their final destination. The first theory is that the NCCs have an intrinsic program that is

activated once they leave the neural tube. This intrinsic program contains information for

molecular patterning for the formation of facial structures. The second theory is that the NCCs

acquire facial patterning information from the surrounding tissue at their destination (44). The

dominant theory is that of the latter theory, that the NCCs retain multipotency into the later

stages of embryonic development (1).

1.1.3 Facial prominences and CNCC derivatives of the face

During the migration towards their final destinations ventrally, extensive interactions occur

between the CNCCs and the surrounding mesoderm and overlying ectoderm and endoderm. The

CNCCs in each facial prominence have distinctive molecular signatures and interact with

different epithelia during development (1).

By the end of fourth week of human embryonic development, most of the CNCCs have reached

their final destinations. Within the pharyngeal arch is a center mass of mesodermal cells,

Page 15: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

7

surrounded by CNCCs and externally covered by ectoderm and internally bordered by endoderm

(Figure 2) (45). Continued interactions between surface ectoderm, CNCCs, mesoderm and

pharyngeal endoderm are needed for the proper development of the facial structures (46, 47). For

example, coordinated interactions between CNCCs and the mesoderm cells result in the

differentiation of myoblast and skeletal precursors to form the musculature and skeleton of the

facial region (48, 49). Signals that emanate from the CNCCs instruct and inform mesodermal

cells to differentiate into myoblast precursors and how to organize themselves around the

developing skeletal elements.

1.1.4 Facial development and facial prominences

The major portion of the face in the human is formed starting between the fourth and eighth

weeks of development. The face is derived from buds of tissue—called facial prominences—that

undergo symmetrical and asymmetrical growth. Appropriate fusion events are required to result

in mature facial structures (50). These facial primordia appear in the fourth week of development

around the primordial stomodeum through inductive influences of organizing centers located in

the prosencephalon and rhombencephalon (51).

Figure 2: A schematic diagram of the pharyngeal arches. (A) The arches are indicated by

different colors. The first arch is divided into maxillary (1a, orange) and mandibular (1b, yellow)

Page 16: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

8

component. The second arch (2, green); third arch (3, purple); fourth arch (4, pink) and the sixth

arch (6, blue). B. A coronal cut through the human embryo showing the tissue contribution to

each arch. The arches (1,2,3,4) are composed of a core of neural crest (nc, yellow) and

mesoderm (mes, green) surrounded by both surface ectoderm (se, pink) and pharyngeal

endoderm (pe, purple). Reproduced from Cordero et al. 2010 (1).

At about the 4th

to 5th

week range (E9.5 in mice) of human development, CNCCs are migrating

and arriving at the facial prominences (52). A total of seven prominences in the face will

eventually be present ventral to the forebrain the FNP, two lateral nasal processes (LNP), two

maxillary processes and two mandibular processes. The FNP surrounds the ventrolateral part of

the forebrain which give rise to the optic vesicles of the eyes and also will give rise to the

forehead, middle of the nose, upper lip, philtrum and primary palate (53). The lateral region of

the FNP, also known as the median nasal process (MNP), will fuse with the LNPs and the

maxillary processes to create the alae and columnellae of the nose. Initially, around the 5th

week

of embryonic development, a pair of placodes form ventral to the developing forebrain. Further

growth and proliferation of the placodes lead to a deepening, resulting in the formation of nasal

pits which are surrounded by the lateral nasal process (LNP) and medial nasal processes (MNP)

(51). These nasal placodes are bilateral oval thickenings of the surface ectoderm and develop on

the inferolateral parts of the FNP. Mesenchyme in the margins of the placodes proliferates to

produce horseshoe shaped elevations known as the MNP and LNP (Figure 2).

The maxillary and mandibular processes are derivatives of the first pharyngeal arch and give rise

to the upper and lower jaws, respectively. The maxillary processes enlarge and grow medially

towards each other due to proliferation of the mesenchyme within the processes. This causes the

MNPs to move towards the median plane and each other. The LNPs are separated from the

maxillary processes by the cleft of the nasolacrimal groove. Merging of the LNP with the

maxillary process starts around the 6th

week of development. Between the 7th

and 10th

weeks of

development the MNPs merge with each other and with the maxillary and lateral nasal processes

resulting in a continuity of the upper jaw, lip and separation of the nasal pits from the

stomadeum. When the MNPs merge they form the intermaxillary segment which is made up of

the philtrum of the upper lip, the premaxillary part of the maxilla and the primary palate (55).

The lateral parts of the upper lip, most of the maxilla and secondary palate arises from the

maxillary processes (51). The secondary palate begins to form in the early 6th

week from the

Page 17: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

9

mesenchymal projections of the maxillary processes. The palatal shelves are outgrowths of

NCC-derived mesenchyme covered by a layer of oral and surface epithelia on their outer surface.

Within the palatal shelves is NCC derived mesenchyme. For the palate to form, the maxillary

processes expand and undergo rotation to a horizontal position. The palatal shelves must have

outgrowth in order to fuse. As the jaws grow, the tongue becomes relatively smaller and moves

inferiorly, allowing the lateral palatine processes to elongate and ascend to a horizontal position.

These palatal processes fuse in the median plane as well as with the nasal septum and primary

palate (55).

The maxillary processes merge with the mandibular prominences and the mesenchyme from the

second pair of PA invades the primitive lips and cheeks and will later differentiate into the facial

muscles of expression which are supplied by the facial nerve, a nerve of the second PA. The

mesenchyme in the first PA differentiates into the muscles of mastication which are innervated

by the trigeminal nerve from the first PA.

At the 9th

-12th

week of development the nasal septum develops in a downward growth fashion

from the fused MNPs and begins fusing with the palatine processes. Bone will gradually develop

in the primary palate and extends from the maxillae and palatine bones into the lateral palatine

processes to form the hard palate. In the midfacial region, NCCs condense within the single

median frontonasal process to form the pre-cartilaginous nasal capsule, which undergoes

chondrogenesis to form the nasal septum. The nasal septum has been proposed to play the role of

pacemaker or growth center for the subsequent growth of the face and the skull. The nasal

septum contributes to the overall changes in morphology observed during the development of the

facial skeleton (56). To date, the fundamental mechanisms involved in the initiation, growth,

boundary setting and differentiation of the cartilaginous and skeletal structures of the frontonasal

region are not well characterized.

Tooth formation also occurs around this stage. At 37 days of embryonic development in humans,

a thickened band of epithelium forms in the upper and lower jaws around the mouth. The

primary epithelial bands give rise to the dental lamina and the vestibular lamina. Localized

thickening of the epithelium, also known as placodes, form within the epithelial bands (57).

Beneath the epithelium lies an embryonic connective tissue called ectomesenchyme into which

NCCs have migrated. Epithelial outgrowths descend into the ectomesenchyme at the sites of

Page 18: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

10

deciduous teeth and ectomesenchymal cells proliferate and accumulate around these outgrowths.

Within the first twelve days of development in mice, the epithelium possesses the potential to

induce tooth formation but after 12 days of development this potential is assumed by the

ectomesenchyme such that the ectomesenchyme can elicit tooth formation form different

epithelia. Experiments have shown that first arch ectomesenchyme combined with foot

epithelium can induce enamel organ formation from the plantar epitheliulm (58). If the epithelial

enamel organ is recombined with skin mesenchyme, the organ loses its dental characteristics and

assumes those of the epidermis. These experiments indicate that odontogenesis is initiated by

factors from the epithelium initially and then later the ectomesenchyme.

1.2 Craniofacial developmental anomalies

Due to the highly regulated nature of growth and fusion of the facial processes, craniofacial

development is extremely susceptible to genetic and environmental perturbation that results in

craniofacial malformations. There are many opportunities for congenital facial anomalies to

occur. Anomalies result mainly from maldevelopment of the neural crest tissue which give rise

to the connective tissue and skeletal primordia of the face (51).

Facial dysmorphologies are a component of 75% of birth defects (7). Additionally, perturbances

in the critical and highly regulated roles of CNCC specification, delamination, migration and

differentiation during craniofacial development is best illustrated when abnormalities occur in

craniofacial birth defects such as Treacher Collins syndrome, cleft lip and palate, 22q11.2

microdeletion syndrome and CHARGE syndromes.

One of the most common craniofacial malformations is clefting of the lip and palate and clefting

of the secondary palate alone. Cleft of the primary lip and palate has an incidence of 1/700 and

clefting of the secondary palate alone has an incidence of 1/1500. Environmental factors such as

a large tongue or ankylosis can cause clefting of the palate because the tongue will not descend,

preventing the palatal shelves from fusing (1). Genetic factors, such as mutations in transcription

factors like TBX 22 (59) and the gene Wntb9, play a role in secondary palatal clefting (60). In

mammals, Wnt signalling is critical for the proliferation of CNCCs within the maxillary

processes (61). Transforming growth factor (TGF-α and TGF-β) and epithelial growth factor

receptor (EGFR) play a role in the fusion process of the epithelial cells on the palatal processes

of the secondary palate. Egfr-/- mice have midline defects with an elongated primary palate,

Page 19: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

11

clefting of the palate and micrognathia (62). Any disruption in the timing or extent of growth of

the facial prominences can result in facial malformations.

Treacher Collins syndrome is an example of abnormal regulation and proliferation of CNCCs. It

is characterized by downward slanting eyes, micrognathia, conductive hearing loss,

underdeveloped zygoma, drooping part of the lateral lower eyelids, and malformed or absent

ears. The syndrome is caused by a mutation in TCOF1, which encodes the Treacle protein. A

decrease in the Treacle protein results in a significant reduction in the number CNCCs, due to

apoptosis and a decrease in migration and proliferation (63), and results in severe craniofacial

hyperplasia and dysplasia.

In summary, early development of the craniofacial region starts with the highly regulated process

of CNCC development and their eventual contribution to the major structures of the craniofacial

region. During the formation of the CNCCs, interactions between the surrounding surface

ectoderm, neuroectoderm and endoderm are crucial for normal development of the face (1) and

under strict molecular regulation. Many genes have been characterized to be significant players

in the regulation of CNCC development and morphogenesis.

1.3 Proteins and genes involved in formation of craniofacial structures

A number of genes, proteins and specific signalling pathways have been shown to be involved at

different stages of craniofacial formation. During early craniofacial development, epithelial-

mesenchymal interactions are especially crucial in determining the types of tissues and extent of

outgrowth of the different structures. Patterning and differentiation events later in development

are also regulated by a series of molecules with specific functions. A review of the major

signalling pathways and specific molecules with known roles in regulating some of the major

events in craniofacial development will be discussed. For example, members of the BMP family,

Wnt/β-catenin pathways and FGFs are necessary for NCC generation and survival (64-66). In the

final part of this section, a description of Cthrc1, the focus of the current study, will be

conducted.

Early on, the prosencephalic organizing center, located at the rostral end of the notochord

beneath the forebrain, induces the visual, inner ear apparatuses and the upper third of the face.

Page 20: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

12

The rhombencephalic center induces the middle and lower thirds of the face (52). The forebrain

establishes many signalling centres in the ectoderm that covers the frontonasal zone. Interactions

between CNCCs and epithelia from the forebrain and facial ectoderm are required for proper

development of the FNP (67). The forebrain provides SHH signalling which imprints on the

forebrain ectoderm, controlling differential cell proliferation, and transforming the FNP into the

nasal region. The nasal placodes provide morphogenetic information to the mesenchyme of the

lateral nasal prominence and act as a signalling center for the lateral nasal skeleton, inducing

cartilage and bone (68).

Many genes involved in signal transductions, transcription regulation, binding and catalytic

activities have been identified in the developing first PA. Examples are Msx-1 and 2, Pax8,

Bmp4, 5 and 7, Fgfr11 and Tgf-β. Reciprocal epithelial and mesenchymal signalling events in the

first PA induce transcription factors that function to differentiate oral versus aboral and

rostrocaudal surfaces and patterning of skeletal elements (68). An example of the signalling

between the endoderm, mesoderm and epithelium is that of TBX1, a T-box transcription factor.

Tbx1 is expressed in the endoderm, mesoderm and epithelium of the palatal shelves and FNP

(69). Decreased Tbx1 expression in the endoderm in PA development and epithelia of the palatal

shelves results in secondary NCC abnormalities (70). Tbx1 knockout mice show defects in the

PAs and pouches (70).

The secreted peptide, Endothelin I, signals from the facial epithelia and mesoderm to the

intervening CNCCs that form the PA1 skeleton. Mice with a homozygous mutation in EdnI have

malformed bones of the jaw and throat and defective musculature (71). Recent genetic studies in

humans have indicated that mutations in the Endothelin pathway may play a role in

Waardenburg-Shah syndrome and Hirschprung disease which are caused by defects in NCC

behaviour.

Epithelial-mesenchymal interactions have been shown to play a key role in tooth development.

Examples of genes that are expressed in the NCC-derived ectomesenchyme which initiate tooth

formation are Lim-homeobox genes, Lhx-6 and Lhx-7. It has been shown that Fgf8 signalling

from the epithelium induces the expression of the Lim-homeobox genes in the mesenchyme.

Fgf8 from the epithelium also induces Pax-9 expression in the mesenchyme (57).

Page 21: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

13

1.3.1 Fibroblast growth factors and bone morphogenetic proteins

FGFs and BMPs have many roles during craniofacial and pharyngeal skeletal morphogenesis.

The roles of FGF include serving as survival factors for CNCCs (72, 73), directing CNCCs to

adopt the ectomesenchymal fate (74), acting as chemoattractants to promote lateral migration of

endodermal cells for segmentation of the pharyngeal endoderm into pouches and the correct

patterning of the CNCC-derived skeletal elements (26). For example, a Fgf8 conditional

inactivation in the ectoderm of PA1 in a mouse model results in NCC apoptosis and absence of

most PA1 skeletal elements (73). FGF signalling is also involved in NCC spatial identity and the

establishment of anteroposterior and dorsoventral polarity of PAs (75, 76).

BMPs are osteogenic agents that induce differentiation of mesenchymal cells toward an

osteoblastic lineage and stimulate differentiation and functions of osteoblasts during bone

modeling (77, 78). BMPs are critical in regulating bone formation (79) and play an integral role

during craniofacial and pharyngeal skeletal morphogenesis, such as patterning of the maxilla and

mandible (8), mesenchyme proliferation (80) and the pathogenesis of craniofacial synostosis

(81).

At early stages of development, FGF8 and BMP4 pattern the maxilla-mandibular region and

define it from the premandibular domain (82) before the CNCCs arrive. The Fgf8 expression

domain is induced by SHH signalling from the endoderm and delimited by Bmp4 which is

expressed on both sides of adjacent Fgf-8 expressing ectoderm (82-84). BMP4 and FGF8 control

the location of incoming NCCs by activating the patterning genes Dlx1, Barx1 and Msx1. These

epithelial-mesenchymal interactions are needed for specifying the identity of the pre-mandibular

and maxilla-mandibular regions. Fgf8 also has a role in anteroposterior and dorsoventral PA

patterning and left-right symmetry of the craniofacial skeleton (85).

1.3.2 Sonic hedgehog

SHH signalling has many roles in craniofacial development. Disruption of SHH signalling in

chick, mouse and zebrafish models results in severe head skeleton abnormalities due to defects in

CNCC survival, proliferation and patterning (86, 87). If SHH is absent in the foregut endoderm,

development of Meckel’s cartilage is prevented as well as some associated PA1 structures, due

Page 22: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

14

to NCC apoptosis (88). SHH also has a late-stage role in NCCs to promote differentiation into

cartilage and establishing skeletal polarity in the mediolateral axis of the embryo (89, 90).

SHH signalling from facial ectoderm is involved in the specification of NCC spatial identity

(91). Excess SHH results in supernumerary Meckel’s cartilage. SHH also has a late-stage role in

NCCs to promote differentiation into cartilage and establishing skeletal polarity in the

mediolateral axis of the embryo (89, 90).

A signalling center called the frontonasal ectodermal zone (FEZ) exists in the ectoderm

overlying the FNP. It regulates the growth and dorsoventral polarity of the upper beak in birds

(91). Mice have two FEZ in the left and right MNP (92). When the FEZ is grafted ectopically it

can reprogram the fate of underlying NCCs (91), showing an epithelial-mediated patterning

instruction. FGF8 and SHH are both expressed in the FEZ facial epithelia and they promote

cartilage outgrowth by inducing BMP4 expression the underlying NCCs (92). It has been

proposed that the persistence of FGF8 expression in the facial ectoderm of the duck, but not the

chick, is what contributes to the distinct morphology of the duck beak by inducing it to produce

more cartilage (93).

SHH is also involved in epithelial-mesenchymal interactions during tooth initiation. SHH is

expressed in the dental ectoderm of mice at E11. Shh knockout mice have little development of

facial processes. Addition of SHH soaked beads to oral ectoderm can induce local epithelial

proliferation to produce invaginations like those of tooth bud formation, implicating Shh in tooth

initiation (57).

1.3.3 Wnt pathways

In vertebrates, Wnt proteins have roles in regulating body axis specification, patterning of germ

layers and tissues, cell adhesion, cell migration, cell proliferation, cell differentiation,

morphogenetic movements during embryogenesis and organogenesis, and growth and metastasis

of cancers/tumours (94). WNTs are secreted glycoproteins that interact with Frizzled (Fz)

proteins on the cell surface. Fz proteins are a family of seven transmembrane G protein-coupled

receptor proteins and they mediate cell signalling from the cell surface to cytoplasm (95). The

Wnt signalling pathway can be divided into two subsets: the β-catenin-dependent canonical

pathway and the β-catenin-independent non-canonical pathways.

Page 23: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

15

In the β-catenin-dependent canonical pathway, Wnt proteins combine and interact with an

Fz/low density lipoprotein receptor (LRP) complex on the surface of target cells (Fig. 3). This

interaction leads to stabilization of β-catenin proteins (96) and subsequent signalling cascades. In

the resting state β-catenin in the cytoplasm is phosphorylated. During activation, the accumulated

β-catenin translocates to the nucleus and interacts with transcriptional factors to activate

transcription of target genes (97). This canonical Wnt pathway is involved in cell fate, regulation

of body axis, patterning of neuroectoderm and amplification of neural progenitors (94).

Figure 3: The canonical Wnt signalling pathway. In resting cells, β-catenin is assembled in a

multiprotein complex with CK1α, GSK3β, APC and Axin and is primed for phosphorylation by

GSK3β. Phosphorylated β-catenin interacts with β-Trcp and is degraded by ubiquitation (A). In

Page 24: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

16

the activated state, Wnt proteins act on the Fz/LRP complex on the surfaces of the target cells.

Upon Wnt-Fz signaling, Wnt-Fz and LRP coordinate Dvl activation, which results in recruitment

of axin to the plasma membrane. Activated Dvl dissociates the multiprotein complex which leads

to the inactivation of GSK3β, which can no longer phosphorylate β-catenin. Excess free

cytoplasmic β-catenin translocates to the nucleus and binds to TCF/LEF transcription factors,

causing transcriptional activation of target genes (B). Reproduced from Miao et al., 2013 (95).

The β-catenin-independent non-canonical Wnt signalling pathways has two categories: the

Wnt/Ca2+

pathway and the PCP pathway (Figure 4) (95). These pathways are involved in cell

proliferation, cell adhesion and cell differentiation, among other biological roles (98). The

Wnt/Ca2+

pathway leads to the release of intracellular calcium through G-proteins and also

involves activation of phospholipase C and protein kinase C (PKC) (99, 100). The non-canonical

Wnt-PCP pathway regulates cytoskeleton organization via actin polymerization through

activation of small GTPases (Figure 4). It also regulates the establishment of polarity within the

plane of the epithelium and allows cells to obtain directional information during embryogenesis

(101). Fzd activates JNK and directs asymmetrical cytoskeletal organization and coordinated

polarization of cells within the plane of epithelial sheets (102, 103). The Wnt-PCP pathway also

controls convergent extension movements during gastrulation and is involved in the directional

migration of cells in the developing palatal shelves (104).

Page 25: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

17

Figure 4: The non-canonical Wnt signalling pathway. The non-canonical Wnt signalling pathway

includes the Wnt/Ca2+

signalling pathway and the planar cell polarity Wnt pathway. In Wnt/Ca2+

,

certain Wnt-Fz interactions activate the cytoplasmic protein Dvl, which increases the level of

cytosolic Ca2+

. Subsequent activation of Ca2+

-sensitive enzymes CamKII, PKC and others

induces the target gene expression and corresponding biological effects. In the Wnt/PCP

pathway, activated Dvl activates the small GTPases Rho and Rac, which leads to the activation

of Jun kinase (JNK) and rho kinase (ROK). Subsequently, transcription factors, such as the AP1

family, are activated and target gene expression is induced. Reproduced from Miao et al., 2013

(95).

1.3.3.1 Wnt pathways and craniofacial development

The Wnt/β-catenin signalling pathway components are highly conserved and are crucial for the

patterning and morphogenesis of many developmental processes, including face, limb, skeleton,

central nervous system, skin and ectodermal appendages like teeth and hair (105, 106). In

Page 26: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

18

craniofacial development, Wnt/β-catenin is crucial for NCC induction and migration and for

proper fusion of facial prominences (66, 107, 108).

Studies in the chick and mouse have shown that Wnt signalling in specific regions of the facial

prominences correlate with outgrowth and fusion of processes (61, 109). In particular, mouse

studies have demonstrated that the LNPs were the primary region of Wnt-β-catenin signalling

(61). Mice with a loss-of-function (LOF) or gain-of-function (GOF) mutation for β-catenin

resulted in aberrant Wnt signalling in the ectoderm and embryos showed facial dysmorphologies

at the crucial stage of fusion and growth of prominences of mid-face development. Between E9.5

and E12.5 (7), LOF mutants of β-catenin had a hypoplastic facial morphology, e.g.,

underdeveloped mandibular processes, and a narrow FNP which resembled a beak at birth. GOF

mutants had a lack of controlled directional growth in their facial prominences, resulting in

increased size of the maxillary and mandibular processes and improper fusion of the processes.

GOF mutants also had ectopic cartilage throughout the head region resulting in malformations of

the head. Both the GOF and LOF mutants had alterations in the shape of their face, which would

affect the underlying cranial skeleton.

The proper growth of facial prominences is needed for cartilage formation as the mesenchymal

cells beneath the ectoderm require proliferation signals to establish a critical number of

progenitor cells (110). The progenitor cells respond to localized and intrinsic patterning signals

from the ectoderm to form cartilage of the correct shape and size (110). The strictly canonical

WNTs (2B, 7A, 9B and 16) and WNT4 and WNT6 (canonical and non-canonical Wnt ligands)

are expressed in the epithelia while WNT5A, 5B and 11 are limited to mesenchyme (109). These

data demonstrate that ectodermal Wnt/β-catenin signalling plays a crucial role in the formation

and patterning of the craniofacial skeleton.

1.3.3.2 Wnt5A

Mutations in the human Wnt pathway genes have been shown to have an effect on the

craniofacial and limb skeleton (111). For example, a recessive missense mutation of WNT5A or

WNT5A receptor ROR2 results in micrognathia, clefting and rhizomelic limb shortening seen in

Robinow syndrome.

Page 27: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

19

Wnt5a was exclusively expressed in the mesenchyme in the facial region of developing chicken

embryos (109). Of the WNTs found in the chicken embryo face (WNT5A, WNT5B and

WNT11), 5A was the most abundant and was found in all facial prominences. At stage 21 of

chicken embryo development there are strong WNT5A signals throughout the mesenchyme of

the maxillary and mandibular prominences, lateral nasal processesand lateral edges of the

frontonasal mass, with the highest expression in Meckel’s cartilage.

In the non-canonical pathway, WNT5A binds to Fz receptors or can bind to the Ror2 receptor

(112). When WNT5A binds these receptors the JNK/ PCP pathway or calcium signalling

pathways are activated, leading to changes in actin cytoskeleton, cell polarity and cell movement.

In development, WNT5A is expressed in cartilage blastema, which may promote

chondrogenesis. However, in excess it induced rapid loss of the cartilage matrix due to induction

of metalloproteinase and aggrecanase enzymes. WNT5A regulates matrix stability but not the

initial steps of chondrogenesis. It keeps canonical signalling low in cartilage blastema and in this

manner, promotes chondrogenesis and cartilage differentiation (113). See Figure 5 for the roles

of WNT5A in development.

Some other Wnt ligands of significance to craniofacial development are WNT3A and WNT9B.

A deletion of Wnt3a causes death upon birth and these embryos have mandibular defects. Wnt9b

is involved in lip fusion and its targeted deletion results in cleft lip in animal models. A deletion

of Wnt5a leads to truncation of the upper and lower jaws (113) (114).

Page 28: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

20

Figure 5: Summary of WNT5A functions during cartilage development and disease. A) During

normal development, there is synthesis of WNT5A in the cartilage blastema and by the newly

differentiated chondrocytes. The secreted WNT5A acts back on the same cells to inhibit

canonical activity. The net result of reducing canonical activity is to promote chondrocyte

differentiation and matrix secretion. Reproduced from Hosseini-Farahabadi et al. (113).

1.4 Collagen Triple Helix Repeat Containing 1 (Cthrc1)

1.4.1 Background

Collagen Triple Helix Repeat Containing 1 (Cthrc1) was originally discovered in a screen for

novel sequences induced in a rat arterial injury model, being expressed in adventitial cells of

remodelling arteries and dermal fibroblasts during skin wound healing (115). Cthrc1 was also

identified in a microarray study of the midface region of the mouse embryo at E10.5 (Gong

unpublished data, 2006). Cthrc1 mRNA was upregulated in the MNP compared to the LNP,

suggesting a possible critical role in the development of the midface. It is for this reason as well

as information that will be discussed in the following section that the CTHRC1 gene and protein

was chosen as a gene of interest for a role in development of the midface.

Page 29: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

21

1.4.2 CTHRC1 Structure

CTHRC1 protein is a 30 kDa secreted N-glycoprotein containing a short collagen motif with an

NH2-terminal peptide for extracellular secretion, a short collagen triple helix repeat of 36 amino

acids and a COOH terminal globular domain (Figure 6). The helix repeat comprises 12 Gly-X-Y

repeats, a domain believed to be responsible for trimerization of the protein and protection from

cleavage (116). The normal active form of CTHRC1 is an N-glycosylated trimer anchored on the

cell surface (94).

Figure 6: The structure of the CTHRC1 protein. Adapted from Lindner (117).

CTHRC1 is exclusive to vertebrates and is highly conserved throughout evolution, showing little

homology to other currently known proteins (115). Its short collagen-like motif is similar to the

collagen domains present in the C1q/tumour necrosis factor-α related proteins, (118). CTHRC1

is thought to belong to the adiponectin/complement factor family (119) that all contain the

conserved collagen domain with the 12 GLY-x-y repeats and a globular domain in the C-

terminal half. Adiponectin is a protein hormone that modulates a number of metabolic processes,

including glucose regulation and fatty acid oxidation (120). Similar to adiponectin, CTHRC1

exists in monomeric, dimeric and trimeric forms (55). Under reducing conditions in Western blot

analysis, CTHRC1 exists as a 28 kDa protein (119). The biological activity of CTHRC1 is

restricted to the highly conserved 200 amino acids at the C-terminal region (94) which contains

an N-glycosylation site that stabilizes the CTHRC1 protein by decreasing its turnover rate.

Upregulation of CTHRC1 is seen to coincide with its increased glycosylation rate in certain cells

Page 30: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

22

such as human oral squamous cell carcinoma (121). N-glycosylation also promotes tethering of

CTHRC1 to the cell membrane, which promotes actin polymerization and cell polarity (94).

1.4.3 Expression Pattern of Cthrc1

During development, Cthrc1 mRNA expression has been identified in visceral endoderm,

developing kidney and the heart of mouse embryos, with specific abundance in the cartilage

primordia, growth plate cartilage (excluding the hypertrophic zone), bone matrix and periosteum

(122). Prior to neural crest migration (E8.5 in mice), Cthrc1 mRNA was seen in the notochord

and the floor plate of the anterior ventral neural tube. By E9.5 transcripts were seen in somites,

branchial arches, otic placode and the hindbrain-midbrain junction with the somatic expression

becoming more pronounced by E10.5. Expression of the gene was observed in developing bone

formed via endochondral and intramembranous ossification, such as skull bones, ribs, vertebrae

and cartilage primordia (122). Cthrc1 transcripts have been shown to be expressed prominently

in chondrocytes during E14.5 of mouse development (119). Although chondrocytes from

condensing mesenchyme contained high levels of CTHRC1 protein and mRNA, hypertrophic

chondrocytes no longer expressed the mRNA. At much later time points (E18.5) developing

incisors show CTHRC1 in the dentin line and at the interface of epithelial ameloblasts and

underlying mesenchyme.

Postnatally, the expression of Cthrc1 in adult tissues has been shown at low levels and restricted

to basal expression in bone, brain and mature bone-resorbing osteoclasts. However, Cthrc1

expression surges in pathological states such as arterial injury, skin wounds or cancer (115, 122,

123). During development, Cthrc1 transcripts are prominently expressed in chondrocytes (119).

In mouse pups, high levels of CTHRC1 protein expression were seen in chondrocytes of the

resting and proliferating zone of the growth plate (122). Conversely, non-proliferating

chondrocytes in adult articular cartilage and fibrous cartilage of the meniscus did not express

CTHRC1.

1.4.4 Possible Functions of Cthrc1

Several studies have suggested that Cthrc1 plays important roles in collagen regulation, postnatal

bone remodeling, and cell migration. The following section will review the literature on the

Page 31: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

23

involvement of Cthrc1 in these processes and discuss the possible role of Cthrc1 in the processes

in development.

1.4.4.1 Cthrc1 Role in collagen deposition

The localization of CTHRC1 to tissues rich in collagen, such as cartilage (collagen type II), bone

matrix (collagen type I) and skin (collagen type I and III), and to active sites of collagenous

matrix deposition suggests a role for Cthrc1 in modulating collagen matrix synthesis. The role of

Cthrc1 in collagen deposition has mainly been seen in studies in vascular remodelling after

injury. Cthrc1 was first discovered in a screen for differentially expressed sequences in balloon-

injured versus normal arteries where it was expressed by fibroblasts of the remodeling adventitia

and by smooth muscle cells of the neointima and was shown to decrease or inhibit collagen

matrix deposition (115). In vitro overexpression of CTHRC1 caused a dramatic reduction in

collagen type I mRNA, procollagen protein levels and collagen deposition (115, 124).

Transgenic mice that constitutively overexpress Cthrc1 under a CMV promoter have brittle

bones, caused by a reduction in collagenous bone matrix (124). The blood vessels of Cthrc1

transgenic mice had widespread cartilaginous metaplasia of the tunica media which suggests that

Cthrc1 causes the shifting of type I collagen expression profile typical of bone to a collagen type

II pattern typically found in cartilage. This shift to the subtype of collagen found in cartilage

would further support findings showing chondrocytes of the growth plate of developing bones

with abundant expression of Cthrc1 mRNA (122).

Vascular remodeling after injury is controlled by TGF-β signalling that mediates negative

aspects of vessel repair such as neointimal lesion formation, smooth muscle cell proliferation,

increased collagen deposition and lumen narrowing (125). Cthrc1 expression patterns overlap

significantly with those of TGF-β family members in calcified tissues and cartilaginous matrix,

as well as developing bone and cartilage (122, 124), periosteum, osteocytes (126), developing

skull bones, ribs, vertebrae, cartilage primordia, hyptertrophic and proliferative zones of growth

plate chondrocytes and all zones of endochondral ossification (122). CTHRC1 interacts with the

TGF-β signalling pathway to modulate collagen regulation (59). CTHRC1 can be regulated by

TGF-β and conversely, may also regulate TGF-β responsiveness and affect TGF–β target genes

(124). For example, Cthrc1 mRNA levels are increased in response to TGF-β but CTHRC1

protein is also a cell type-specific inhibitor of TGF-β which impacts collagen type I and III

Page 32: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

24

deposition (61), neointimal formation and differentiation of smooth muscle cells (122, 124). In

transgenic mice that have upregulated Cthrc1 gene expression, TGF–β signalling is reduced in

smooth muscle cells (124).

CTHRC1’s signalling interactions with TGF-β have been shown to be via activation of Smad 2/3

complexes (127). The promoter region of Cthrc1 contains a putative Smad binding site (128) . It

has been suggested that the activation of Cthrc1 transcription could be regulated by TGF-β

signalling through Smad proteins (123). Different studies have shown that CTHRC1 regulates

extracellular collagen deposition by inhibiting phosphorylation of Smad2/3 activation via

inhibition of TGF-β signalling. Phosphorylated Smad 2/3 increases expression of CTHRC1 that

in turn inhibits the deposition of extracellular collagen controlled by Smad 2/3 phosphorylation

(123). Smooth muscle cells that overexpressed CTHRC1 protein had reduced levels of phospho-

Smad 2/3 (122). Cthrc1 transcript induction by TGF-β, the CTHRC1 inhibition of TGF-β

sensitive reporters and the reduction of phospho-Smad 2/3 levels in vivo provide evidence that

these two proteins are working as antagonists to maintain balance in extracellular matrix

components. The mechanism by which CTHRC1 disrupts TGF-β signalling, leading to a

reduction in collagen is not clear. Since CTHRC1 is a secreted protein, it is likely to function as a

ligand in a signalling pathway with downstream effects on collagen promoter activity (124).

1.4.4.2 Role of Cthrc1 in osteogenesis, osteoclastic bone formation and bone remodelling

The skeletal system fulfills mechanical, supportive, and protective roles in animals (129). Its

formation is controlled by tightly regulated programs of cell proliferation, differentiation,

survival, and organization (130). The initiation of the skeletal system is from mesenchymal

condensations, in which skeletal precursor cells, also known as osteochondral progenitors, give

rise to either chondrocytes to form the cartilage or osteoblasts to form the bone.

Bone is a mineralized connective tissue consisting by weight of 28% type 1 collagen and 5%

noncollagenous structural matrix proteins, synthesized by osteoblasts. These constituents

accumulate as the uncalficied matrix, osteoid, that acts as a scaffold for the deposition of apatite

crystals of bone to make up the remaining 67% of bone. This mineral is in the form of small

plates which lodge in the holes and pores of collagen fibrils (57). Some osteoblasts become

trapped in the bone matrix and are then referred to as osteocytes. The osteoclast is a

Page 33: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

25

multinucleated cell which resides against the bone surface where they can bind and secrete

enzymes to demineralize the bone matrix and degrade the organic matrix.

The difference in the initial fate choices of osteochondral progenitors to either chondrocytes or

osteoblasts determines whether ossification is endochondral or intramembranous (57).

Intramembranous ossification is the direct laying down of bone into the primitive connective

tissue (mesenchyme). During intramembranous ossification, bone develops directly within the

soft connective tissue whereby osteoblasts differentiate from the mesenchyme and begin to

produce bone matrix (57). In contrast, the initiation of endochondral ossification requires the

presence of cartilage as a precursor (131). During endochondral ossification, condensation of

mesenchymal cells gives rise to cartilage cells. This cartilage will eventually be replaced by

bone. Cartilage consists of collagen of which the main type is collagen type II, in contrast to

bone which is primarily collagen type I. Perichondrium forms around the periphery of the

cartilage which gives rise to a cartilage model that will eventually be replaced by bone through

the action of osteoblasts. Prechondrogenic mesenchymal condensations in the developing mouse

face are seen as early as E8.5 and 9.5 and cartilage primordia forms at E11.5-14.5 (132, 133).

Overall, collagen deposition is critical in both endochondral and intramembranous bone

formation as the collagen matrix acts as a scaffold in endochondral bone formation and is a

component of the bone material in intramembranous bone formation and makes up the

components of cartilage.

Bone mass is regulated by a process of continual remodelling, which is based on the action

between osteoblastic bone formation and bone resorption, and is coordinated by mediation of

many signalling pathways such as parathyroid hormone, TGF-β and BMPs (134). Bone turnover

rates of 30%-100% in childhood are common and slow down in adulthood (57).

CTHRC1 has been shown to be involved in postnatal bone formation mainly through effects on

osteoblasts. CTHRC1 is expressed in bone tissue in vivo, and is a gene identified as a

downstream target of bone morphogenetic protein-2 (BMP-2) in osteochondroprogenitor-like

cells (119). Micro-computed tomography and bone histomorphometry analyses showed that

Cthrc1-null mice have low bone mass due to decreased osteoblastic formation and conversely,

transgenic mice overexpressing Cthrc1 under an osteoblastic promoter displayed high bone mass

due to an increase in osteoblastic bone formation (135). Cthrc1 osteoblast-specific

Page 34: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

26

overexpressing and null mice also had increased and decreased levels of osteoblast specific

genes, ALP, Col1a1 and Osteocalcin, respectively, with no change in osteoclast numbers. In

vitro, CTHRC1 stimulated differentiation and mineralization of osteoprogenitor cells with a

particular acceleration of osteoblast proliferation (135). CTHRC1 has been shown to be induced

by BMP2 and is not required for skeletal development, though it is required for bone

maintenance homeostasis (135, 136). In addition to TGF-β, CTHRC1 has also been shown to be

regulated by BMP-4 (115, 128), suggesting that CTHRC1 may act as one of the downstream

targets for BMP-Smad signalling and that its expression in growth plate cartilage and bone

matrix controls collagen deposition though regulation of Smad2/3 and TGF-β signaling (122,

124).

In addition to transgenic and knockout studies, in vitro studies have further supported a role of

CTHRC1 in bone formation through its effects on osteoblasts via osteoclasts. Osteoclasts placed

on a substrate containing hydroxyapatite had upregulated expression of Cthrc1 mRNA compared

to the dentin slices, suggesting the CTHRC1 protein production is closely linked with osteoclast

attachment to calcified tissue (119). The same result was observed when osteoclasts were placed

in an environment with high extracellular calcium and phosphate. Cthrc1 expression increased in

a high-turnover state (RANKL injections in vivo) and decreased in conditions associated with

suppressed bone turnover (aging and after alendronate treatment) (119). BrdU incorporation

assays showed that CTHRC1 stimulates osteoblast proliferation in vitro and in vivo. Colony-

forming unit (CFU) assays in bone marrow cells showed CTHRC1 stimulates differentiation of

osteoprogenitor cells and osteoblasts (136). Experiments using ST2 cells, a stromal line derived

from mouse bone marrow showed that CTHRC1 targets stromal cells to stimulate osteogenesis

by binding to a putative cell surface receptor on ST2 cells to stimulate osteoblastic differentiation

as well as recruitment in order to promote bone formation (119).

The impact of CTHRC1 on bone mass was examined in mice with ovariectomies.

Ovariectomised mice are estrogen deficient and develop bone loss secondary to bone resorption.

(135). Wild-type mice showed a 47% trabecular bone loss after ovariectomy while transgenic

mice overexpressing Cthrc1 showed only 38% trabecular bone loss, suggesting a stimulatory

effect of CTHRC1 on bone formation.

Page 35: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

27

More recently it has been shown that CTHRC1 is secreted by resorbing active osteoclasts and

acts as a coupling agent between bone resorption and formation. In vitro experiments showed

that Cthrc1 expression stimulated osteogenic differentiation of osteoblasts, measurable by

increased alkaline phosphatase and osteocalcin and had bone formation stimulating activity

comparable to that of BMP-2 and FGF-1(119). Mice containing a systemic or osteoclast specific

knockout of Cthrc1 were born appearing grossly normal but with absence of Cthrc1 mRNA.

There was no difference in mRNA levels between the two types of knockout mice, indicating

that mature osteoclasts are the major source of CTHRC1 in vivo. Both types of knockout mice

had low bone mass and abnormalities similar to osteoporosis. Histomorphometirc analysis

showed decreased osteoid surface and bone formation rate compared to control mice. These

results concluded that CTHRC1 protein is produced and secreted by mature osteoclasts and acts

on osteoblastic cells to stimulate osteoblastic differentiation and recruitment which promotes

bone formation and maintains bone mass and trabecular structure through regulation of bone

formation.

From these studies the common theme is that Cthrc1 plays a role in postnatal bone formation

due to regulation of collagen deposition as well as having an effect on osteoblasts. Taken with

results from Cthrc1 transgenic and knock out mouse experiments, evidence of a role in collagen

deposition and in vitro studies on osteoblast proliferation, CTHRC1 seems to have an anabolic

effect on bone formation though collagen matrix deposition and regulation of osteoclasts and

osteoblasts.

1.4.4.3 Role in cell motility and tissue repair

CTHRC1 is also shown to be involved in cell migration (137). Inhibition of Cthrc1 in vitro

decreases cell migration, while overexpression increases cell migration (115, 128). CTHRC1

enhances migration of smooth muscle cells and fibroblasts in a scratch wound assay, enabling

CTHRC1 overexpressing cells to migrate into the wound area significantly faster than control

cells (115, 122). Enhanced expression of CTHRC1 is involved in vascular remodelling; in rat

fibroblasts it promotes cell migration and inhibits collagen I synthesis in these cells (124). Under

normal conditions in blood vessels, CTHRC1 resides in the cytoplasm of smooth muscle cells in

an unprocessed form, despite the presence of a signal peptide. Under conditions of vascular

injury, CTHRC1 is released from the smooth muscle cells and cleavage of the N-terminal

Page 36: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

28

propeptide results in a molecule with an increased ability to inhibit collagen matrix deposition

(138). The ability of CTHRC1 to reduce collagen matrix deposition correlates with increased cell

migration.(139). This limitation of collagen matrix deposition has been suggested as one

mechanism by which CTHRC1 promotes cell migration (128). CTHRC1 expression is also

linked to cellular proliferation, such as Schwann cell proliferation (140) and enhanced

proliferation of osteoblasts (135).

CTHRC1 has the same positive effect on migration in tumour cell lines and has been found in

increased levels in highly metastatic tumours, which suggests it plays a role in cancer

progression by promoting cell migration (128). Cthrc1 transcripts and protein are increased in

malignant melanoma, cancers of the GI tract, lung, breast, liver and pancreatic cancers when

compared to normal tissues (141) and are highly active in degrading extracellular matrix proteins

in several of these malignant tumours (128, 142, 143). A significant increase of CTHRC1 has

been observed in patients with metastatic bone cancer compared to non-metastatic tumours

(121). A germline mutation in Cthrc1 was identified in patients with Barrett’s esophagus and

esophageal adenocarcinoma (143). Cthrc1 was among 48 genes that are significantly associated

with risk of recurrence of patients with stage II/III colon cancer (144).

CTHRC1 expression has been used as a cancer prognostic indicator. Lack of CTHRC1 has been

demonstrated in non-invasive stages of melanoma in contrast to enhanced expression in primary

invasive melanomas and metastatic melanomas (142). Inhibition of Cthrc1 using short

interfering RNA showed decreased invasion of melanoma cell lines and patients with bone

metastases had a significant increase in CTHRC1 stromal expression compared to patients

without.

In conclusion, the current literature supports a role for Cthrc1 in cell migration, collagen

deposition and bone formation.

1.4.5 Genes and signalling pathways involved with CTHRC1

1.4.5.1 Cthrc1 Activation of Wnt/PCP pathway

Cthrc1 is also suggested to interact with components of the Wnt/PCP pathway in the non-

canonical Wnt pathway by forming a CTHRC1-Wnt-FZD/Ror2 complex to selectively activate

the Wnt/PCP pathway (94). The normal active form of CTHRC1 is an N-glycosylated trimer

Page 37: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

29

anchored on the cell surface. CTHRC1 is a positive regulator of the non-canonical Wnt

signalling and simultaneously inhibits the canonical branch (94). Results from Yamamoto et al.

(94) suggest that CTHRC1 is a Wnt cofactor protein that selectively activates the Wnt/PCP

pathway by stabilizing an extracellular ligand-receptor interaction. In vitro, CTHRC1 has been

shown to interact with multiple extracellular components of Wnt signalling including both

canonical and non-canonical Wnt proteins, Fzd proteins and the Wnt/PCP co-receptor Ror2 but

not with the canonical Wnt co-receptor LRP6 or the PC component Vangl2 (94, 139) (Figure 7).

The CTHRC1 protein is thought to positively regulate the Wnt/PCP pathway by promoting

interaction between the Wnt ligand and the Fz receptor complex (94). In conditions where

appropriate receptors are available, Wnt proteins may activate the canonical or non-canonical

pathways but when CTHRC1 is present the interaction of Wnt proteins with the Fzd/Ror2

complex is enhanced and the selective activation of the Wnt/PCP pathway in this environment

suppresses the canonical pathway (145).

Through the Wnt/PCP pathway, CTHRC1 may regulate cell motility in embryogenesis and

cancer cell migration and invasiveness in adult tissues (128). It has also been speculated that the

Wnt/PCP pathway activation by CTHRC1 contributes to the promotion of cell motility because

the PCP pathway regulates actin polymerization through GTPase signalling, which alters cellular

morphology and increase cellular motility (94). This Wnt/PCP pathway plays an important role

in controlling cell polarity and movement and has been implicated in chondrocyte maturation

and cartilage formation during ontogenesis (146). The Wnt/PCP pathway regulates chondrocyte

maturation and cartilage formation as genetic alterations in the pathway are associated with

chondroplasia, dysregulation of collagen deposition and changes in cartilage morphology (94,

139, 147), thus further supporting a role for Cthrc1 in collagen deposition and cartilage

formation (139).

Page 38: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

30

Figure 7: A model of selective activation of the Wnt/PCP pathway by CTHRC1. CTHRC1 is

cell-surface anchored and enhances the interaction of Wnt proteins, Fzd proteins and Ror2

complex to activate the Wnt/PCP pathway (94). The interaction of Wnt proteins with Fzd/Ror2 is

selectively enhanced in the presence of Cthrc1. Adapted from Yamamoto et al.(94)

1.4.5.2 Cell-specific action of Cthrc1

Cthrc1’s cell-specific behaviour is well characterized, with different actions or effects depending

on the tissue it is expressed in. CTHRC1 inhibits TGF-β signalling in smooth muscle cells but

not in endothelial cells (124). There are differing results on the effect of CTHRC1 on Col1a1

expression—upregulation of CTHRC1 in osteoblasts results in increased expression of Col1a1

whereas in PAC1 cells, a smooth muscle cell line, upregulated CTHRC1 produced reduced

mRNA levels of Col1a1 and collagen deposition (115, 124). High levels of CTHRC1 were also

associated with reduced collagen deposition in smooth muscle cells but increased collagen

deposition in osteoblasts (123, 135). These results suggest CTHRC1 function may differ between

cell types and that it may work through its own signalling pathway that is functional in a cell-

type specific manner (135).

In summary, many roles of CTHRC1 are supported by various studies in the literature.

Transgenic mouse studies have suggested CTHRC1 has a possible role in bone formation,

specifically in osteoblast regulation and collagen matrix deposition. In disease states, CTHRC1 is

associated with increased cell motility and migration and alterations in collagen matrix

deposition. These roles in cell migration, collagen matrix deposition and regulation of bone

formation have been demonstrated in vitro as well as in postnatal animals. Cell migration and

motility, bone formation and regulation of collagen expression are all important steps in

Page 39: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

31

embryogenesis and therefore the involvement of CTHRC1 in these processes during

development is highly probable. The role of CTHRC1 in cell migration, collagen matrix

deposition and bone formation makes it a gene of interest to study in craniofacial formation as

these processes are crucial in midface development.

1.5 Rationale for Study

Many craniofacial abnormalities can be attributed to defects in the generation, proliferation,

migration and differentiation of cranial NCCs and epithelial-mesenchymal induction interactions.

Any perturbation in these processes can result in craniofacial malformations and developmental

anomalies of the facial structures. Although existing studies describe the involvement of a

number of well-known genes in midfacial development, there still remain a great number of

candidate genes whose role in development remains unexplored.

Given the evidence that the CTHRC1 protein and mRNA play roles in cell migration, collagen

deposition, and bone formation, we speculate that it plays such roles in the early and later events

during the development of the midface.

1.6 Hypothesis/Aims

The hypothesis is that CTHRC1 protein has an expression pattern in the developing mouse

embryo midface indicative of a role in craniofacial development of the mouse with respect to cell

migration, bone formation and collagen deposition.

The aims of this study are to perform a temporal and spatial analysis of CTHRC1 expression in

at crucial time points of the developing midface in mouse embryos.

Page 40: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

32

Chapter 2

Materials and Methods

2.1 Embryos

Timed pregnant wild-type CD-1 mice were obtained from the Toronto Centre for

Phenogenomics. Embryos were removed from the uteri of mice at embryonic (E) stages E8.5,

E9.5, E10.5, E11.5, E12.5 and E13.5. Embryos were isolated from the uteri with the aid of a

Nikon SMZ800 dissecting microscope, at a temperature of 4°C. Whole embryos were dissected

for time points E8.5 and E9.5 and the head was dissected from time points E11.5, E12.5 and

E13.5. Approximately five embryos were used for each time point of immunohistochemical

staining with the CTHRC1 antibody.

Another group of eight embryos at stages E8.5, E9.5, E10.5, E11.5 and E13.5 were used for

reverse transcriptase quantitative polymerase chain reactions (RT-qPCR). The whole embryo

was collected for stage E8.5. The head was dissected from the body of the embryos of stage

E9.5, E10.5, E11.5 and E13.5. Tissues were stored at -80°C.

2.2 RT-qPCR

To obtain a temporal profile of the expression of the Cthrc1 gene mRNA, RT-qPCR was

performed on RNA samples from embryos at stages E8.5, E9.5, E10.5, E11.5 and E13.5.

2.2.1 RNA isolation

Tissues of embryos harvested at specific embryonic stages, as described above, were

homogenized in TRI Reagent (Sigma Aldrich) for 5 minutes. Chloroform was added to samples,

which were shaken for 15 seconds. Tissue samples were centrifuged at 12,000g for 15 minutes at

4°C. The aqueous phase was removed and mixed with isopropanol, incubated for 10 minutes at

room temperature and then centrifuged at 12,000g for 8 minutes at 4 °C. The supernatant was

removed to isolate the RNA pellet, which was washed with 75% ethyl alcohol (ETOH) and

centrifuged at 7500 g for 5 minutes at 4 °C. The ETOH supernatant was removed and the RNA

pellet was resuspended in distilled H20. Samples were run on a 2% agarose gel and stained with

Page 41: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

33

ethidium bromide to determine the integrity of the RNA; specifically, to determine the presence

of the small (2 kb) and large (5 kb) ribosomal RNA.

2.2.2 Reverse transcription of RNA samples

Reverse transcription of the RNA from each time point sample was performed using the Maxima

First Strand cDNA Synthesis Kit for RT-qPCR (#K1641; Thermo Fischer Scientific). A reaction

mix was made with the sample template RNA, H20, Maxima Enzyme Mix and 5X reaction mix.

Reaction samples were incubated for one cycle of 10 minutes 25°C, 30 minutes 72°C, 5 minutes

85°C and then stored at 4°C.

2.2.3 Quantitative PCR

Primers for Cthrc-1 were designed using Ensembl software on the Cthrc1 gene

(http://useast.ensembl.org/Mus_musculus/Gene/Sequence?g=ENSMUSG00000054196;r=15:390

76932-39087121). Primers were selected that skipped the intron between exons 1 and 2 (3,323

bases). The primers spanned basepairs 62-279 of the Cthrc1 gene, producing an amplified

fragment of 218 basepairs. The primer sequences were:

Forward primer: 5’-T G C T G C T G C T A C A G T T G T C C-3’

Reverse primer: 5-’T C C C T T T T C C C C T T T G A A T C-3’.

All reactions were performed in a 96-well plate using the iTaq™ Universal SYBR® Green

Supermix (Biorad). GAPDH was used as the reference gene.

PCR reactions of cDNAs from tissue samples at each time point sample were run in duplicate at

two different dilutions (1:5 and 1:10), for quality control. PCR conditions were as follows: 40

cycles of 94°C, 58°C and 72°C to allow for denaturing of the cDNA, binding of the primer and

extension of the primer, respectively. cDNA quantities were determined using the CFX96

Touch™ Real-Time PCR Detection System and CFX manager software 3.0 (Biorad) and plotted

on a graph using the formula: 2∆CT(target)

/ 2∆CT(reference)

. “∆CT target” was the change in cycle

threshold value for the tissues at each time point compared to E8.5 and “∆CT reference” was the

change in cycle threshold values of GAPDH at the corresponding time points compared to E8.5.

The values from this ratio were plotted using Excel software (Microsoft Office 2007).

Page 42: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

34

2.3 Histological Processing and Paraffin Embedding

Embryos were transferred to Bouin’s fixative (Polysciences, Inc.) to incubate for 24 hours,

followed by storage in 70% ETOH at room temperature (20°C). Prior to paraffin embedding the

embryos were stored in 80% ETOH for 1 hour, 100% ETOH for 1 hour, followed by incubation

in methyl benzoate overnight. Afterwards, embryos were transferred to toluene for 2 hours.

All embryos were processed for paraffin sectioning according to Gong (2001)(148). Sections

were mounted on Superfrost slides (Fisherbrand). Embryos were oriented such that they were cut

in a coronal, transverse or sagittal plane. The choice of orientation for different time points

depended on the need to capture the pattern of CTHRC1 expression in specific midfacial

structures at different stages of development in the midface. Therefore, embryos at E8.5 and E9.5

were oriented only in sagittal or coronal planes and E10.5-13.5 were oriented in the coronal or

transverse planes.

2.4 Immunohistochemistry

Immunohistochemical reactions using a rabbit polyclonal antibody to Cthrc1 (ab85739; Abcam)

were conducted on tissue sections at different embryonic stages according to Gong (2001)(148).

To ensure specificity of the α-CTHRC1 antibody, a peptide competition assay was performed to

rule out false positive results. Transverse sections of embryos at E13.5 were selected based on a

distinct pattern of expression. The Cthrc1 peptide (ab101727; Abcam) was incubated with the

primary antibody at 5x the primary antibody concentration used for the immunohistochemistry at

4°C for 24 hours prior to the staining. The staining was then carried out as described in Gong

(2001)(148), with additional slides receiving the peptide-blocked primary antibody instead of the

primary antibody alone. Subsequent steps for the staining of these slides were as described in

Gong (2001)(148). The signal detection was compared in the sections stained with the peptide-

blocked primary antibody and the primary antibody alone.

The sections on the slides were deparaffinised and rehydrated with washes of 100% ETOH, 95%

ETOH and 70% ETOH. Tissue sections were washed for 30 minutes in phosphate buffered

saline (PBS). Sections were treated with 0.3% H202 for 20 minutes to quench endogenous

peroxidase activity and washed for 10 minutes with PBS. Antigen retrieval was conducted by

treating sections with 0.01 NaCitrate buffer (pH 6.0) for 10 minutes at 90°C, followed by washes

Page 43: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

35

for 10 minutes in PBS. Tissues were then incubated in a blocking solution of 1.5X normal goat

serum (Zymed), 0.01% Saponin and 0.1% Bovine Serum Albumin (Sigma) to prevent non-

specific binding of the secondary antibody. The tissue sections were subsequently incubated with

the primary antibody at a 1:100 dilution in PBS overnight at 4°C.

The next day, tissue sections were washed with PBS for 30 minutes and incubated with a

polyclonal goat-anti-rabbit biotinylated IgG (R&D Systems) at a dilution of 3:100 for 40 minutes

at room temperature. The tissues were washed for 30 minutes with PBS and then incubated with

Vectastain Elite ABC kit (Vector Laboratories) for 1 hour. Lastly, sections were washed in PBS

for 30 minutes and the antibody signal was detected using a DAB Peroxidase Substrate Kit

(Vector Labs). The sections were then rehydrated and coverslips were placed using Permount

mounting media (Fischer Scientific). Negative controls were performed concurrently, without the

presence of the primary antibody.

2.5 Documentation and analysis of Cthrc1 expression

Images of immunohistochemically stained histological sections were taken using Spot Advanced

software and the Spot Diagnostic Camera attached to an Olympus BX51 camera at 10x and 4x

magnification. Signal localization was determined by images taken with the digital camera and

observations were recorded for each developmental stage. Location as well as spatial distribution

of the signal was recorded.

Page 44: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

36

Chapter 3

Results

3.1 Quantitative expression of Cthrc1 mRNA transcripts during midface development

Analysis of Cthrc1 expression in the midface tissues of embryos at E8.5, E9.5, E10.5, E11.5 and

E13.5 developmental stages by quantitative polymerase chain reaction (qPCR) revealed a

specific temporal pattern. There was an increase of Cthrc1 mRNA from time points E8.5 to E9.5.

At time point E10.5 there was a decrease in mRNA levels from the E9.5 time point. At E11.5

there was an increase of the Cthrc1 transcripts back to a level similar to what was seen at E9.5.

At the last time point of E13.5 the highest level of Cthrc1 transcript increase was seen, as

compared to E8.5 (Figure 8).

Figure 8: Temporal expression profile of Cthrc1 mRNA across E8.5-E13.5. RNAs were

extracted from tissues of embryos from each time point, reverse transcribed to produce the

corresponding cDNA and amplified by qPCR. Cthrc1 mRNA levels were compared to levels at

time point E8.5.

Overall, the results from the qPCR revealed that expression of Cthrc1 mRNA peaked at E9.5

relative to E8.5, followed by a decrease in expression at E10.5 and then a steady increase after

Page 45: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

37

E11.5. The results confirmed the presence of Cthrc1 mRNA in the midface during these time

points of embryonic development.

3.2 Expression of CTHRC1 protein

The spatial expression of CTHRC1 protein was assayed at six different time points of embryonic

development: E8.5, E9.5, E10.5, E11.5, E12.5 and E13.5. Tissue sections of the sagittal,

transverse and coronal planes through the developing craniofacial region were used for the

expression analysis. A distinct spatial pattern of expression of CTHRC1 was clearly observed at

each time point (Figures 10-15).

3.2.1 CTHRC1 peptide competition assay at E13.5 to confirm specificity of primary CTHRC1 antibody

A peptide competition assay of the CTHRC1 antibody revealed the specificity of the antibody for

its antigen. Compared to the distinct expression of CTHRC1 in the nasal septum and nasal

capsule (Figure 9C; see below for more details of expression), no staining was observed in the

areas of CTHRC1 expression on a consecutive tissue section of the nasal capsule and septum

where the antibody was blocked with the CTHRC1 peptide (Figure 9A). The negative control

(absence of primary antibody) also showed no staining (Figure 9B), similar to that of the blocked

primary antibody. These results indicate that the primary antibody, raised against the CTHRC1

peptide, was specific to the protein with little cross-reactivity or non-specific binding.

Figure 9: Peptide competition assay on consecutive coronal sections through the anterior part of

the midface in an E13.5 embryo. A) Peptide block at 5X antibody concentration showing no

signal. B) Negative control staining with goat anti-rabbit secondary antibody showing minimal

Page 46: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

38

background signal. C) CTHRC1 primary antibody (1:100) and secondary goat anti-rabbit

antibody showing a distinct pattern of CTHRC1 expression in the nasal septum (asterisks) and

nasal capsule (arrow).

3.2.2 Spatial expression analysis of CTHRC1 in the midface at different developmental stages

At the earliest observed time point of E8.5, CTHRC1 expression was limited to the midline of

the embryo, most strikingly in the notochord and neural tube (Figure 10). In the midline,

expression was observed in the notochord, extending from the rostral end and along the length of

the dorsal side of the embryo (asterisks, Figure 10A and C). In addition, CTHRC1 expression

was also seen in the neuroectoderm in the area of the ventral brain vesicle and at the rostral end

of the embryo (arrows, Figure 10B and D). Expression of the protein was observed in a small

area of the oral ectoderm (open arrowhead, Figure 10D).

Figure 10: CTHRC1 protein expression in E8.5 of coronal (A, B and D) and sagittal (C) sections.

A) Asterisk shows CTHRC1 localized to the notochord (nc) and arrow shows CTHRC1

expression in neuroectoderm (ne). B) Arrow shows CTHRC1 expression in neuroectoderm (ne).

C) Sagittal section showing CTHRC1 localized (asterisks) to notochord (nc) along rostral-caudal

length of the embryo. D) CTHRC1 expression in the neuroectoderm (arrow) and oral ectoderm

(open arrowhead).

Page 47: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

39

By E9.5 and E10.5, CTHRC1 protein was exclusively expressed in the mesenchyme (Figure 11).

Expression was present in the facial prominences, specifically the mesenchyme of the

frontonasal and medial nasal processes and the maxillary and mandibular processes (Figure 11 A

and B). In the more anterior or ventral region of the medial portion of the midface, e.g., the

developing frontonasal region, CTHRC1 was widely expressed throughout the whole area,

including the medial-most or central portion of the FNP (Figure 11B and C).

Another consistent pattern of CTHRC1 expression in the craniofacial region was the

mesenchymal expression of CTHRC1 adjacent to the ectoderm and neuroectoderm. CTHRC1

mesenchymal expression in the frontonasal and medial nasal processes was adjacent to the

ectoderm of the respective facial processes (Figure 11A and C). Also, in certain areas, CTHRC1

protein was strongly expressed in mesenchymal tissues immediately adjacent to the

neuroectoderm of the telencephalic brain vesicles (arrowhead, Figure 11D).

Figure 11: CTHRC1 protein expression in E10.5 embryos, in coronal (A, B and D) and sagittal

(C) sections. A) CTHRC1 protein expression in mesenchyme of the frontonasal process (FNP)

Page 48: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

40

(arrows). B) CTHRC1 expression in the frontonasal process (FNP, arrow), maxillary process

(Mx, tailed arrow) and mandibular process (Md, open arrow). C) Expression in FNP adjacent to

ectoderm (arrow) and in ventricle of heart (H). D) Distinct expression in mesenchyme next to

neuroectoderm (MeN, arrowhead) and mesenchyme in Md process (open arrow). Figures A, B

and D are sections from three E10.5 representative embryos, with A and B sections being more

ventral and D located more dorsally.

By stage E11.5 of embryonic craniofacial development, expression of the CTHRC1 protein was

observed in a distinctive pattern in areas of cartilage formation and deposition. As cellular

condensations are initiated in different parts of the developing craniofacial region, CTHRC1

expression was clearly expressed in many of these structures. The maxilla at this stage presents

with a nasal septal cartilage analgen, a rod like structure that is flanked with two wing-like

cartilaginous processes in the developing nasal capsule. In the maxilla and forming snout of the

mouse, distinct expression was observed to localize at the forming nasal septum and nasal

capsule (Figure 12 A and B).

Figure 12: CTHRC1 protein expression in E11.5 through transverse sections of the maxilla nasal

septum and nasal capsule. A) CTHRC1 expressed in areas of the nasal capsule (nc) and nasal

septum (ns) (arrows). B) CTHRC1 in nasal capsule (nc) and nasal septum (ns). A, B = 4x and

10X magnifications, respectively.

ns

nc

Page 49: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

41

CTHRC1 was still strongly expressed in the nasal septum and other areas of cartilage formation

of the midface at the later time points of E12.5 and E13.5. With further development of the nasal

cartilaginous areas, the staining of CTHRC1 appeared as an almost complete ring-like pattern

with a central rod staining, within which contains the bilateral nasal cavities (Figures 13A, 13B

and 14).

Figure 13: CTHRC1 protein expression at E12.5 in coronal sections of the nasal capsule and a

transverse section of the mandible. A) A coronal section of the snout showing CTHRC1

expression in the nasal capsule (nc). B) A coronal section showing CTHRC1 expression in the

Page 50: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

42

nasal septum (ns). C) A transverse section showing CTHRC1 expression in Meckel’s cartilage

(mc).

At E13.5, the same pattern of expression of the CTHRC1 protein was seen in the maxilla and

mandible, but the signal became more distinct and covered a larger surface area as the regions of

cartilage formation and deposition increased with further development of the midface.

Expression was also seen in other areas of cartilage formation in the mouse embryo head, such as

in the developing bones of the calvarium (Figure 14A).

Figure 14: CTHRC1 protein expression at E13.5 in sections of head (A), the maxilla (C and D)

and midface including mandible (D). A) CTHRC1 expression in Meckel’s cartilage (arrow, mc)

and bones of skull (arrow, c). B) More dorsal transverse section of midface and mandible

showing CTHRC1 expression in nasal septum (ns) and nasal capsule bones (nc). C) More

anterior section of CTHRC1 expression in nasal septum (ns) and nasal capsule bones (nc). D)

Arrows showing Cthrc1 expression in nasal capsule (arrow, nc) and bones of maxilla (arrow,

Mx).

mc

nc

M

x

c

Page 51: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

43

3.2.3 CTHRC1 expression in the mandible and developing tooth bud

In the developing mandible of embryos at stages E9.5, E10.5 and E11.5, the pattern of CTHRC1

expression was consistent with the pattern seen at E10.5 in the maxillary process and frontal

nasal process. CTHRC1 expression was localized to the mesenchyme and was always adjacent to

areas of the ectoderm. For instance, the protein was expressed in embryos at stages E9.5 and 10.5

in areas of outgrowth, with a higher expression in the anterior and ventral portion of the facial

processes (Figure 11 A and C) and the oral half of the mandibular process (open arrow, Figure

11B). As cartilaginous structures are laid down in the mandible, the expression of CTHRC1 was

clearly evident. One structure is Meckel’s cartilage, a bilateral cartilaginous bar across the

mandibular process. A strong CTHRC1 signal was localized in the area corresponding to

Meckel’s cartilage (Figure 13C and 14A). The distinct pattern of CTHRC1 expression in

Meckel’s cartilage was only observed at later time points, specifically E12.5 (Figure 13C) and

E13.5 (Figure 14A). CTHRC1 expression was also observed at time point E12.5 lateral to the

Meckel’s cartilage in an area that most likely represents the membranous ossification of the body

of the mandible (arrow, Figure 13C).

CTHRC1 also appeared to be expressed in areas of tooth formation. Initially, its expression was

generally throughout the mesenchyme with a stronger signal near the epithelial thickenings on

the margins of the stomodeum, which corresponds to the formation of the dental placode (Figure

11B, 11D and 15A). At E11.5, the formation of what is equivalent to the cap stage of tooth

development has started in the mandible. CTHRC1 expression at this time was seen in the

mesenchyme directly below areas of epithelia corresponding to the enamel organ (Figure 15B

and C). This distinct pattern of mesenchymal expression in the area corresponding to the dental

papilla continued until the last observed time of E13.5 (Figure 15D).

Page 52: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

44

Figure 15: CTHRC1 protein expression in tooth bud formation. A) E10.5: Mandible, arrows

showing CTHRC1 expression in mesenchyme of the mandibular process (Md) near the

ectoderm. B) E11.5: Tongue (t) and Mandibular process (Md), arrow indicating CTHRC1

expression in mesenchyme near tooth buds. C). E11.5: Arrows pointing to CTHRC1 expression

in mesenchyme near tooth bud. D) E13.5: Arrows showing expression in mesenchyme adjacent

to developing tooth.

Page 53: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

45

Chapter 4

Discussion

This chapter of the thesis will outline the possible roles for Cthrc1 in midface development by

drawing conclusions based on the data from the Results section in combination with evidence

from the literature. The results will be briefly summarized followed by a discussion of proposed

roles for Cthrc1 in the development of the midface, future directions and concluding remarks.

4.1 Summary of Cthrc1 expression during mouse embryo midface development

4.1.1 Summary of qPCR mRNA levels of Cthrc1 mRNA during E8.5–E13.5

Overall, our data revealed that Cthrc1 has a quantitative expression profile that changed across

development, with these changes correlating closely with some of the major changes during early

midfacial development. There was an increase in Cthrc1 mRNA levels from E8.5–E9.5, a stage

when the CNCC begin to migrate from the neural tube (149) towards the facial prominences

located ventrally (150). At E10.5 there was a decrease from the E9.5 mRNA levels with levels

still about that of E8.5. This interval corresponds to CNCCs arriving at the locations where they

will begin to interact with surrounding tissue to begin the process of condensation of cell types

(57). At E11.5 there was an increase of the Cthrc1 transcripts back to a level similar to E9.5. At

E11.5 there has been condensation of cell types, such as mesenchyme, that will start developing

into chondrocytes or other specific cell types. At the last time point of E13.5 the highest level of

Cthrc1 mRNA transcript increase was seen compared to E8.5. At this time, proliferation and

differentiation of tissue such as cartilage is starting in the midface of the embryo. Overall,

increases in Cthrc1 mRNA were seen at E9.5, with a relative decrease at E10.5 and then a steady

increase from E11.5 onwards.

It is important to note that for the time point of E8.5, mRNA was extracted from the whole

embryo due to difficulty in obtaining enough materials from only the heads of the small embryo.

For the remaining time points the head of the embryo was dissected from the body and processed

for qPCR. The levels of mRNA at E8.5 may not reflect a true comparison to the other time points

Page 54: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

46

because it was measured from the whole embryo rather than the head. This would most likely

lead to an overestimation of the mRNA level at E8.5 in the head as all tissues in the embryo were

processed.

4.1.2 Summary of immunolocalization experiments results of CTHRC1 protein from E8.5-13.5

The immunolocalization experiments, performed by immunohistochemistry (IHC), in this study

provided information regarding the spatial expression of the CTHRC1 protein at the time points

E8.5, E9.5, E10.5, E11.5, E12.5 and E13.5, allowing visualization of the type of tissue and

boundaries of CTHRC1 expression. The spatial expression patterns of CTHRC1 suggest that it

has roles in cell migration, epithelial-mesenchyme interactions, collagen formation and bone

formation. Expression at E8.5 of CTHRC1 at the notochord, oral ectoderm and neuroectoderm

suggest a role in cell migration and signalling of NCCs towards the structures or prominences to

which they will migrate. This corresponds to previous results from Durmus et al. (122) who

showed Cthrc1 mRNA expression in the notochord at E8.5. At E9.5 and 10.5, the close

association of CTHRC1 protein in the epithelial-mesenchyme border supports a role of CTHRC1

in epithelial-mesenchymal interactions and signalling between the two tissues during formation

of midface structures. The presence of CTHRC1 in the medial nasal process of the FNP supports

results from the microarray experiment screening genes expressed in the medial nasal and lateral

nasal process (Gong, unpublished results 2006). CTHRC1 protein expression was also seen in

the heart which corresponds to previous mRNA expression studies of Cthrc1 mRNA in the

mouse embryo (122). CTHRC1 expression patterns at E11.5 in areas of condensation of

mesenchyme for future cartilage and bone formation such as the area of the future nasal septum,

nasal capsule suggest a role of CTHRC1 in cell condensations. Expression was also seen in the

mandibular process in the mesenchyme but was concentrated to areas adjacent to the epithelium

of developing tooth buds at the placode and bud stage of tooth development, suggesting a role in

epithelial mesenchyme interactions. At E12.5 and E13.5 CTHRC1 expression suggests a role in

chondrocytic differentiation, as it was present in the nasal septum and capsule, a role in

intramembranous ossification due to its expression in developing bones of the calvarium and the

area adjacent to Meckel’s cartilage, and a role in epithelial-mesenchyme interactions due to its

expression in mesenchyme corresponding to the dental papilla.

Page 55: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

47

4.2 Involvement of Cthrc1 in midface development

This section of the discussion will examine the possible roles for Cthrc1 in the development of

the midface in the mouse embryo based on previous studies from the literature as well as the

results presented in this thesis. Known events of craniofacial development will be correlated to

the expression patterns seen with the qPCR and IHC results and from this, possible roles for

Cthrc1 will be discussed.

4.2.1 Role of Cthrc1 in cell migration

The previously documented role of Cthrc1 in cell migration (128, 137), cancer progression and

vessel injury (83, 115) via its ability to reduce collagen matrix deposition supports a possible role

for it in cell migration during embryogenesis. NCCs need to migrate in order to reach their target

tissues from the neural tube (8).

Overall, a role for CTHRC1 in cell migration is supported by the spatial expression pattern in the

midface during early embryonic development when CNCCs are known to migrate from the

neural crest to the facial prominences. The upregulation of Cthrc1 mRNA as assayed by RT-

qPCR during this time point also supports an increase in CTHRC1 protein in preparation for its

role in cell migration. During early embryonic development, around E8.5 (day 17-19 in human

embryonic development), NCCs are still located in the neural tube, but about to begin migration

to various structures in the body or head (68). Our spatial analysis of CTHRC1 showed a change

in expression in the embryonic midline at E8.5, to the mesenchyme in the facial prominences by

E9.5 (approximately day 22 in human embryo development) (50, 149). During this stage in the

embryonic mouse development the CNCCs are beginning to migrate (149) ventrally towards the

facial prominences (52, 150). The increase in Cthrc1 mRNA levels from E8.5–9.5 in

combination with this dramatic change in expression pattern from the embryo midline to facial

prominences suggest that CTHRC1 protein may play a role in the promotion and emigration of

the CNCCs.

A possible role for Cthrc1 in mediating migration of CNCC is corroborated by other studies

showing the involvement of CTHRC1 with Wnt ligands. Recent studies have shown that the

non-canonical Wnt–PCP pathway plays a major role in neural crest migration (151). PCP

signalling controls CIL between NCCs by localizing different PCP proteins at the site of cell

Page 56: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

48

contact during collision and locally regulating the activity of Rho GTPases (151). The PCP

pathway regulates actin polymerization which can alter cellular morphology and cellular

motility. Vertebrate PCP signalling is regulated by Wnt proteins and CTHRC1 has been shown

to co-precipitate in vitro with Ror2, the receptor for WNT5A, and non-canonical Wnt proteins

(94). One study supporting a possible role in cell migration for CTHRC1 during development

was Yamamoto et al. (94) where a knockout of Cthrc1 alone did not produce a phenotype;

however, upon introduction of a heterozygous Vangl2 mutation, a gene involved in the migration

of groups of cells during vertebrate embryogenesis, abnormalities characteristic of a PCP mutant

were expressed. Abnormalities included a shortened body axis, open neural tube and

misorientation of sensory hair cells of the cochlea. These defects may be due to failure of NCC

migration, indicating a possible role for Cthrc1 in the migration of these cells. Based on both in

vivo and in vitro results (94) it is possible that CTHRC1 promotes cell migration through

activation of the Wnt/PCP pathway.

Another interesting finding from our study was the localization of CTHRC1 to midline structures

such as the notochord at E8.5. The notochord is located in the embryonic midline, ventral to the

neural tube, exists transiently in the vertebrate and has a role in patterning of the neural tube

(152). The proximity of the CTHRC1 protein expression in the notochord to the location of

CNCCs in the neural crest in our immunolocalization experiments suggests an association and

possible interaction between the CTHRC1 protein and CNCCs at this time point. If there is an

association of CTHRC1 with NCCs with respect to migration or guidance, this colocalization to

the midline adjacent to the area where NCCs reside would support this.

4.2.2 Role of Cthrc1 in epithelial/mesenchyme interactions

Epithelial-mesenchymal tissue interactions (EMI) are critical in the initiation of cell

differentiation and morphogenesis in virtually every organ in the vertebrate body (153). During

development, the mesenchymal cells beneath the facial ectoderm receive proliferation signals for

the appropriate amount of time to establish a critical number of progenitor cells (110). EMI

immediately precede condensation of either prechondrogenic mesenchyme during

chondrogenesis or they initiate transition from osteogenic mesenchyme to preosteoblasts in

osteogenesis in craniofacial skeletal development (153). Several molecules such as BMP-4,

BMP-2, Msx-1 and TGFβ are known to be involved in EMI which form the structures of the

Page 57: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

49

developing face (8, 80, 153). CTHRC1 is a downstream target of BMP-2 and is known to be

regulated by TGF-β and BMP-4 (115, 124, 128, 135). The association of CTHRC1 with

signalling molecules known to participate in epithelial-mesenchymal interactions, in addition to

the spatial expression seen in the mouse midface in this study supports a role for CTHRC1 in the

EMI process. There was a consistent pattern of expression in the mesenchyme of the FNP,

maxillary and mandibular processes concentrated in areas adjacent to the epithelia and

neuroectoderm at E10.5 and E11.5. EMIs are crucial for specifying the identity of the pre-

mandibular and maxilla-mandibular regions (82). These signalling interactions co-ordinate the

outgrowth of the facial primordia from buds of undifferentiated mesenchyme into the intricate

series of bones and cartilage structures that, together with muscle and other tissues, form the

adult face (154). SHH signalling promotes the differentiation of NCCs into cartilages (89, 90)

and based on our results showing CTHRC1 colocalization to areas of cartilage formation there is

a possibility of interactions between the SHH in the epithelium and CTHRC1 in the underlying

mesenchyme during differentiation of chondrocytes.

Before the CNCCs arrive at their destination in the facial prominences, ectodermal Fgf8

prefigures the prospective oral cavity. This FGF8 expression is induced by SHH signalling from

the endoderm and is delimited by BMP-4 expressed on both sides of the adjacent Fgf8-

expressing endoderm (82-84). The ectodermally derived FGF8 and BMP4 control the

regionalization of incoming NCCs through the activation of specific patterning genes such as

Dlx1in the cases of FGF8 while BMP-4 induces Msx1 expression in the underlying mesenchyme

(8, 82). CTHRC1 has been shown to be regulated by BMP-4 (115, 128). Bmp4 mediates the

activity of proliferation zones in the FNP and between the FNP, maxillary and nasal prominences

(93) and its expression in the FNP is crucial for the final morphology of the midface in chick

embryos (155, 156). BMP-4 expression in NCCs of the mesenchyme in the FEZ is induced by

Shh and Fgf8 and promotes cartilage outgrowth (92, 157). Therefore the evidence from our study

showing mesenchymal spatial expression of CTHRC1 in proximity to the epithelia in both

maxilla and mandible prominences in addition to evidence that BMP-4 is already involved in

epithelial-mesenchymal supports a role for CTHRC1 in epithelial-mesenchyme interactions.

CTHRC1 expression was first seen in the mesenchyme adjacent to the epithelium in the facial

processes of E9.5 and E10.5 embryos in our study. β-catenin mouse mutants with upregulated

Wnt/β-catenin in the ectoderm during facial morphogenesis showed normal facial development

Page 58: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

50

at E9.0. Shortly after, when the facial prominences began to enlarge, the nasal processes,

maxillary and mandibular processes of these mice also had an increase in overall size compared

to wild-type and altered expression of fgf8 and Shh, Bmp2 (7). Interactions of CTHRC1 with

members of the Wnt signalling pathway, CTHRC1 as a downstream target of BMP-2 and FGF8,

and BMP4 having a possible connection with CTHRC1 signalling, is further evidence that

CTHRC1 plays a critical role in mediating critical interactions between the ectoderm and

mesenchyme that leads to the proper shape and size of the face.

The proximity of CTHRC1 expression to the epithelium destined to become the enamel organ

supports a role in induction of this tissue which will go on to form the enamel and dentin of the

tooth. This concentration and proximity of the CTHRC1 expression in the mesenchyme of the

facial prominences to the epithelium and ectoderm in addition to the fact that CTHRC1 is known

to interact with other molecules involved in epithelial mesenchyme interaction would support its

role in this process. In the case of tooth development, BMP-4 is a regulator of CTHRC1 and

when it is expressed in the ectoderm of the facial processes it induces the expression of Msx1 in

the underlying mesenchyme (8) which is important in tooth formation. Bmp4 and Msx1 act in a

positive feedback loop to drive sequential tooth formation. BMPs, FGFs, SHH and Wnt

pathways are all repeatedly used throughout tooth development (158). Our results show a

localization of CTHRC1 in the mesenchyme starting at E9.5 and remains until our last time point

of E13.5. Therefore at these time points there may be epithelial-mesenchyme interactions driving

tooth formation from the placode stage to the tooth bud stage.

4.2.3 Role of Cthrc1 in regulation of collagen formation and deposition

The stages of chondrogenesis in the craniofacial area have been divided into a) epithelial-

mesenchymal interactions preceding condensation of prechondrogenic mesenchyme, b)

condensation c) prechondroblasts differentiating into chondroblasts, d) deposition of

extracellular matrix and e) terminal differentiation and mineralization depending on the cartilage

type (153). Condensations are initiated by alteration of the mitotic activity and aggregation of

cells and are a product of the preceding stage of EMI (153). Our spatial expression analyses of

embryos at later developmental stages support important roles for CTHRC1 in a number of these

critical stages of chondrocytic condensation and differentiation. By E11.5 (approximately day 33

of human development), sites of mesenchymal condensations in the facial prominences are

Page 59: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

51

initiated and these areas will eventually differentiate into chondrocytes or other specific cell

types. The CTHRC1 pattern of expression in our experiments at this time most likely represents

the beginning of condensations of chondrogenic mesenchyme which is verified in our results by

CTHRC1 expression in areas of future cartilage formation. The qPCR experiment shows a

dramatic increase in the levels of Cthrc1 mRNA between E10.5 and E11.5. The increase may

reflect an increased production of CTHRC1 protein for its upcoming role in condensation of

chondrogenic cells. Our results did not show expression in the area of the future nasal septum at

E9.5 or E10.5 but a distinct pattern was seen at E11.5. This may be interpreted as condensation

not having started yet or if it has, CTHRC1 protein expression may not be strong enough to

produce a visible signal with IHC or its expression has not started yet in this area of

condensation. These areas will form cartilage and may go on to ossify via endochondral

ossification. This is the first time point in the IHC experiments to show expression of CTHRC1

in the exact position of future cartilage formation. In our experiments, the CTHRC1 expression

at E11.5 in the nasal septum and capsule marks chondrogenic condensation for the maxilla,

preceding the cell differentiation into chondrocytes prior to cartilage formation. At E12.5 and

E13.5 there is also a significant expression of CTHRC1 in the area of future Meckel’s cartilage.

Meckel’s cartilage does not form the bones of the mandible, which form from intramembranous

ossification, but will go on to form the sphenomandibular ligament, and malleus and incus bones

of the ear via endochondral ossification. Meckel’s cartilage forms in the 5th

week of human

development which corresponds to E14.5 in the mouse. Therefore at E12.5 and 13.5

condensations of chondrogenic precursors cells would be taking place in Meckel’s cartilage,

slightly later than the condensations in the maxilla.

BMP-4 is another growth factor that has been shown to be upregualted during chondrogenic

condensation, acting as an inducer of condensations (159). It has been shown to be present in

condensing mesenchyme in facial processes of mice (160). BMP-2 is also present in condensing

chondrogenic and osteogenic mesenchyme in the embryonic chick (161). CTHRC1 is a

downstream target of BMP-2 and is regulated by BMP-4 (115, 128) therefore one mechanism by

which CTHRC1 regulates chondrogenic condensation may be through these two molecules.

These condensations mark the onset of selective gene activity that will enable cell differentiation,

for example in prechondrogenic condensations there is a 7-fold increase in mRNA for type II

collagen, illustrating the importance of condensation for the initiation of cell differentiation (153,

Page 60: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

52

162). Similarly in our results we see a near 6-fold increase in Cthrc1 mRNA at E11.5 relative to

E8.5. This increase in mRNA indirectly reflects a future increase in the protein. In the case of

CTHRC1, the amount of mRNA may be increasing at this time in preparation for differentiation

of chondrocytes or to start producing collagen. Since CTHRC1 is known to promote collagen

type 2 deposition, the protein expression in the cartilage anlagen of the nasal capsule and nasal

septum as well as Meckel’s cartilage in addition to the increase in mRNA transcripts supports a

role for CTHRC1 in chondrocyte differentiation. The duration of condensation can vary as it is a

transient stage on the way to overt cell differentiation.

Our results also support a role for CTHRC1 in collagen deposition. At E12.5 and 13.5, a strong

pattern of CTHRC1 protein expression was seen in the cartilage anlagen of the nasal septum and

nasal capsule. Like E11.5, this distinct pattern mirrors exactly the future structure of the nasal

septum and capsule, supporting a role for CTHRC1 in collagen deposition. The fact that

CTHRC1 shifts the collagen expression pattern to that of collagen type II supports a role for

CTHRC1 in collagen deposition for the cartilage of this area. The situation would not be the

same for bone deposition since CTHRC1 inhibits expression of type I collagen mRNA and

matrix deposition (122, 123).

The exact mechanism by which CTHRC1 regulates collagen deposition is not known but there

are several possibilities based on current studies in the literature. CTHRC1 inhibits type I

collagen deposition via inhibition of TGF-β (138) (124) and collagen deposition is regulated by

TGF-β activation of Smad 2/3 complexes. Therefore, a role of CTHRC1 in collagen matrix

deposition for bone formation would likely be inhibitory. Transgenic studies show that CTHRC1

overexpression resulted in brittle bones when overexpressed in all cell types (124) but when only

overexpressed in osteoblasts, these mice had increased bone mass. Although CTHRC1 inhibits

TGF-β, certain members of the TGF-β family such as Activin are known to enhance

condensation size and stimulate chondrogenesis (163). This suggests that CTHRC1 may have a

positive role in bone formation, just not via collagen deposition. In the developing face the TGF-

βs and BMPs regulate maxillary mesenchymal cell proliferation and extracellular matrix

synthesis (164). However, the exact mechanism by which CTHRC1 regulates collagen matrix

deposition through TGF-β is not entirely clear

Page 61: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

53

Another mechanism by which CTHRC1 may regulate collagen deposition and chondrogenesis is

by its interaction with Wnt ligands. It has been suggested that CTHRC1 interacts with

components of the Wnt/PCP pathway by forming a CTHRC1-Wnt-Fzd/Ror2 complex to

promote activation of this pathway (94). The WNT5A ligand also binds the Ror2 receptor and

activates PCP pathway (165). WNT5A is expressed at high levels in mandibular chondrogenesis

in the chicken embryo in Meckel’s cartilage (113). Results from this thesis also show that

CTHRC1 is expressed in Meckel’s cartilage. WNT5A regulates collagen matrix stability, thereby

promoting chondrogenesis via suppression of the canonical Wnt pathway and promoting the PCP

pathway in cartilage blastema which is needed for chondrogenesis (113). The spatial expression

pattern of WNT5A in the chicken embryo at stage 24 in the mesenchyme of the frontonasal

mass, lateral nasal and maxillary process has a similar pattern as what was seen for CTHRC1 at

stage E10.5 in the mouse embryo with respect to distribution near the epithelia (109). Based on

the similarities in spatial expression of WNT5A and CTHRC1 in Meckel’s cartilage and the

mesenchyme of the midface, the role of WNT5A in promoting collagen matrix stability and the

proposed role of CTHRC1 stabilizing the Wnt/Ror2 interaction, it is possible that CTHRC1

regulates collagen deposition through an interaction with WNT5A.

Overall the results from the IHC and qPCR data of this study support a role for CTHRC1 in

chondrogenic condensation, as seen at E10.5 and 11.5 in the maxilla and E12.5 in Meckel’s

cartilage as well as differentiation of chondrocytes. The results also support a positive role for

deposition of collagen type II since the CTHRCI expression is seen in areas of future cartilage

formation. However, since past studies show that CTHRC1 actually downregulates expression of

type I collagen mRNA and inhibits its deposition, it may have a negative role for this type of

collagen. The exact mechanism by which CTHRC1 regulates collagen deposition and

chondrogenic condensation is not known but several possibilities have been put forth in the

preceding paragraphs.

4.2.4 Role of Cthrc1 in bone formation

Our data of a possible function for CTHRC1 in cartilage formation would support a role in

endochondral bone formation, where cartilage is replaced by bone. We speculate that CTHRC1,

being highly expressed at sites of intramembranous ossification in the craniofacial region, e.g.,

Page 62: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

54

body of the mandible and calvarium, plays a direct role in osteogenesis in the developing

craniofacial region.

Studies have shown via transgenic and Cthrc1 null mice that CTHRC1 is a positive regulator of

osteoblastic bone formation postnatally (135). Even though these studies were done on postnatal

mice, the positive effect on osteoblasts would warrant further investigation into the effects of

CTHRC1 in osteoblasts during development. Our results show the presence of CTHRC1 protein

in areas of intramembranous ossification in the midface, mandible and skull of E12.5 and E13.5

mouse embryos. At E13.5, CTHRC1 protein expression was seen in the forming bones of the

skull in the posterior calvarium which forms by intramembranous ossification. This presence of

CTHRC1 in areas of intramembranous ossification in addition to previous studies in postnatal

mice supports a role for CTHRC1 in osteoblastic bone formation and regulation in the embryo.

Another function of CTHRC1 in bone formation in development may be coupling of bone

formation and resorption. CTHRC1 has been shown to couple bone formation and resorption. It

is secreted by osteoclasts in postnatal mice (119). CTHRC1 secreted by osteoclasts targets

stromal cells to stimulate osteogenesis and is increased in high bone turnover states and

decreased in conditions associated with suppressed bone turnover. An osteoclast specific deletion

of Cthrc1 induced osteopenia due to reduced bone formation (166). These studies as well as the

expression of CTHRC1 from the experiments in this thesis, support a role for CTHRC1 as a

stimulatory signal for stromal cells to stimulate osteogenesis at E12.5 and E13.5 in areas of

intramembranous ossification.

Again, the mechanism by which CTHRC1 is involved in bone formation is unclear and can only

be speculated on based on the current studies and the results of this thesis. It is known that

CTHRC1 is regulated by BMP-4, BMP-2 and TGF-β, all of which have known roles in bone

formation as well as midface development. CTHRC1 is a downstream target of BMP-2 in

osteochondroprogenitor-like cells and is also present in bone cells in vivo. Other candidates in

the mechanism of CTHRC1 regulation of bone formation are Wnt ligands. We previously

discussed the involvement of WNT5A in chondrogenesis and its possible interaction with

CTHRC1. In vitro studies have shown that when combined with WNT3A, a bone anabolic

protein, CTHRC1 was capable of stimulating chemotaxis of bone-marrow derived stromal cells

(119), which is consistent with previous studies that show cross-talk between the Wnt pathway

Page 63: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

55

and CTHRC1 (94). Therefore, CTHRC1 may interact with WNT3A to stimulate chemotaxis of

stromal cells involved in bone formation. Wnt3a is known to control the fate of both

mesenchymal cells and NCCs in the craniofacial processes (167) and regulates palatal fusion in

animal models (168). Also, targeted deletions of Wnt3a in mouse models caused death up birth

due to mandibular defects (109) however at this time no studies have shown WNT3A protein

expression in the developing midface.

During craniofacial growth, CTHRC1 may play a role in bone formation by being directly

involved in regulating the differentiation of mesenchyme into preosteoblast cells. In vitro studies

showed that forced Cthrc1 expression in osteoblastic cells stimulated osteoblast differentiation

(166) and CTHRC1 protein stimulated differentiation and mineralization of osteoprogenitor cells

and osteoblasts (135). Interestingly, differentiation of preosteoblasts precedes condensation

which amplifies the number of osteogenic cells in contrast to chondrogenesis where

condensation precedes the appearance of chondrogenic cells (153). It is thus logical to infer that

activation of differentiation to the osteogenic pathway is occurring in osteoblasts just prior to and

around the time of osteogenic condensation in the areas lateral to Meckel’s cartilage and in the

bones of the maxilla and skull.

Although there are few studies on the effect of CTHRC1 on bone formation, the present

literature and results from this thesis support a role for CTHRC1 in osteogenic condensation,

osteoblastic bone formation, coupling of bone resorption and formation and differentiation of

osteoblasts in the developing midface. No studies to date have looked at differences in

differentiation and mineralization of theses cell types in the developing midface during

craniofacial formation in the mouse embryo in the absence or presence of CTHRC1. Most

transgenic and knockout studies have focused on the absence or presence of gross abnormalities

in Cthrc1 transgenic and knockout mice at birth, without looking at changes in osteogenesis

during development. Further studies need to be done of mice embryos during development

especially in the area of the midface to determine the exact role of CTHRC1 in bone formation

during this crucial time of craniofacial formation.

4.2.5 Summary of role of Cthrc1 in midface development

In summary, the results from this thesis support four main roles for CTHRC1 in midface

development of the mouse embryo at critical stages in midface development; a role in collagen

Page 64: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

56

formation of the midface, a role in bone formation of the midface, a role in epithelial-

mesenchyme interactions and lastly a role in cell migration of CNCCs to the facial prominences.

Measurement of the Cthrc1 mRNA transcripts and analysis of spatial expression of the CTHRC1

protein in the developing midface allowed us to draw these conclusions in addition to previous

studies supporting these roles.

4.3 Future Directions for Cthrc1 studies

The results from this thesis show the temporal and spatial patterns of expression of Cthrc1

mRNA transcripts and CTHRC1 protein at critical stages of midface development in the murine

embryo. These results show changes in the CD1 wild-type mouse during normal development.

4.3.1 Midface development in Cthrc1 knockout models

Several knockout mouse models for Cthrc1 have been generated (94, 119, 122, 135, 169). Most

of these studies have not looked at changes in midface development during embryogenesis.

Therefore, future directions of this project would be to study in greater detail the formation of the

midface in Cthrc1 knockout mice compared to wild type mice. Future studies involving the use

of Cthrc1 knockout models can address the effects of an absent Cthrc1 gene specifically on

craniofacial growth and development. For example, studies looking at the amount and quality of

collagen formation in the midface in knockout mice compared to wild-type animals could

evaluate the role of Cthrc1 in chondrogenic condensation and collagen deposition. An

examination of the architecture of midfacical structures such as snout length, size of nasal

capsule and presence of abnormalities such as clefting, and changes in structures requiring

epithelial-mesenchyme interactions such as teeth would give more information of the role of

Cthrc1 during craniofacial development. The focus until now has been on postnatal changes after

Cthrc1 knockout or upregulation in transgenic mice, with any observations on the effects in

embryos being on a gross-anatomical level. Studies on changes in the midface of mice embryos

at the histological and molecular level as a result of Cthrc1 knockout will give more detailed

information as to its role in midface development.

Page 65: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

57

4.3.2 Cthrc1 expression in mouse models specifically targeting effects on midface development

The expression of Cthrc1 mRNA and protein during development (122) and postnatally (119,

124, 135) has been shown in several studies, including this thesis. Numerous mouse models with

deletions, mutations or overexpression of specific genes resulting in craniofacial malformations

have been mentioned in the literature (170-173). For example, deletions of the Wnt3a gene

results in mandibular defects, deletion of Wnt9b results in cleft lip and deletion of Wnt5a results

in the truncation of upper and lower jaws. Deletions of the Tcof1 gene produce a mouse model of

Treacher-Collins syndrome (63, 174). IHC and qPCR of the midface region for Cthrc1 mRNA

and protein in these mouse models compared with what is seen in wild-type mice would show

any changes in Cthrc1 expression during specific syndromes. This may help further support or

disprove already known roles of Cthrc1 during midface development.

4.3.3 Colocalization studies of CTHRC1 with other proteins with a known function in midface development

Many studies exist showing the role of certain genes and proteins in craniofacial development.

Examples are Wnt ligands (109) and Flrt 2 and Flrt3 (175). A colocalization study of CTHRC1

protein expression with other well-known proteins in craniofacial development would allow

comparisons between expression patterns and may help elucidate further roles of CTHRC1 in

midface development by similarities or dissimilarities in the expression patterns. For example,

our results of CTHRC1 expression at E10.5 in the mesenchyme of the facial processes show a

markedly similar expression as Wnt5A mRNA in the mesenchyme of maxillary and mandibular

processes of stage 24 chicken embryos (109). Wnt5A has a role in promoting chondrogenesis

and also has a role in outgrowth of the facial prominences and palate formation (104, 176). The

same comparison could be done at the level of mRNA transcript expression. This would show

any similarities in gene expression patterns with other well-known players in craniofacial

development such as Bmps or Fgf genes.

Page 66: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

58

4.4 Conclusions

The results obtained so far are consistent with the original hypothesis of this thesis that Cthrc1

plays a regulatory role in cell migration, collagen formation and bone formation of the midface.

Based on our results and studies in the literature there is also evidence for Cthrc1 having a role in

epithelial-mesenchyme interactions during midface formation in the developing craniofacial

region.

Page 67: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

59

References

1. Cordero DR, Brugmann S, Chu Y, Bajpai R, Jame M, Helms JA. Cranial neural crest cells on the move: their roles in craniofacial development. Am J Med Genet A. 2011;155a(2):270-9. 2. Kuratani S. Cephalic neural crest cells and the evolution of craniofacial structures in vertebrates: morphological and embryological significance of the premandibular-mandibular boundary. Zoology (Jena). 2005;108(1):13-25. 3. Gong SG. Cranial neural crest: Migratory cell behavior and regulatory networks. Exp Cell Res. 2014. 4. Cordero D, Marcucio R, Hu D, Gaffield W, Tapadia M, Helms JA. Temporal perturbations in sonic hedgehog signaling elicit the spectrum of holoprosencephaly phenotypes. J Clin Invest. 2004;114(4):485-94. 5. Creuzet S, Couly G, Le Douarin NM. Patterning the neural crest derivatives during development of the vertebrate head: insights from avian studies. J Anat. 2005;207(5):447-59. 6. Sandell LL, Trainor PA. Neural crest cell plasticity. size matters. Adv Exp Med Biol. 2006;589:78-95. 7. Reid BS, Yang H, Melvin VS, Taketo MM, Williams T. Ectodermal Wnt/beta-catenin signaling shapes the mouse face. Dev Biol. 2011;349(2):261-9. 8. Minoux M, Rijli FM. Molecular mechanisms of cranial neural crest cell migration and patterning in craniofacial development. Development. 2010;137(16):2605-21. 9. Basch ML, Bronner-Fraser M. Neural crest inducing signals. Adv Exp Med Biol. 2006;589:24-31. 10. Steventon B, Carmona-Fontaine C, Mayor R. Genetic network during neural crest induction: from cell specification to cell survival. Semin Cell Dev Biol. 2005;16(6):647-54. 11. Theveneau E, Mayor R. Neural crest delamination and migration: from epithelium-to-mesenchyme transition to collective cell migration. Dev Biol. 2012;366(1):34-54. 12. Meulemans D, Bronner-Fraser M. Gene-regulatory interactions in neural crest evolution and development. Dev Cell. 2004;7(3):291-9. 13. Huang X, Saint-Jeannet JP. Induction of the neural crest and the opportunities of life on the edge. Dev Biol. 2004;275(1):1-11. 14. Mayor R, Theveneau E. The neural crest. Development. 2013;140(11):2247-51. 15. Stuhlmiller TJ, Garcia-Castro MI. Current perspectives of the signaling pathways directing neural crest induction. Cell Mol Life Sci. 2012;69(22):3715-37. 16. Thiery JP, Sleeman JP. Complex networks orchestrate epithelial-mesenchymal transitions. Nat Rev Mol Cell Biol. 2006;7(2):131-42. 17. Noden DM, Trainor PA. Relations and interactions between cranial mesoderm and neural crest populations. J Anat. 2005;207(5):575-601. 18. Radisky DC, LaBarge MA. Epithelial-mesenchymal transition and the stem cell phenotype. Cell Stem Cell. 2008;2(6):511-2. 19. Ahlstrom JD, Erickson CA. New views on the neural crest epithelial-mesenchymal transition and neuroepithelial interkinetic nuclear migration. Commun Integr Biol. 2009;2(6):489-93. 20. Trainor PA. Craniofacial birth defects: The role of neural crest cells in the etiology and pathogenesis of Treacher Collins syndrome and the potential for prevention. Am J Med Genet A. 2010;152a(12):2984-94. 21. Kulesa PM, Fraser SE. In ovo time-lapse analysis of chick hindbrain neural crest cell migration shows cell interactions during migration to the branchial arches. Development. 2000;127(6):1161-72. 22. Carmona-Fontaine C, Matthews HK, Kuriyama S, Moreno M, Dunn GA, Parsons M, et al. Contact inhibition of locomotion in vivo controls neural crest directional migration. Nature. 2008;456(7224):957-61.

Page 68: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

60

23. Teddy JM, Kulesa PM. In vivo evidence for short- and long-range cell communication in cranial neural crest cells. Development. 2004;131(24):6141-51. 24. Kulesa PM, Lu CC, Fraser SE. Time-lapse analysis reveals a series of events by which cranial neural crest cells reroute around physical barriers. Brain Behav Evol. 2005;66(4):255-65. 25. Le Douarin NM, Dupin E. The neural crest in vertebrate evolution. Curr Opin Genet Dev. 2012;22(4):381-9. 26. Graham A. Deconstructing the pharyngeal metamere. J Exp Zool B Mol Dev Evol. 2008;310(4):336-44. 27. Kuratani S, Matsuo I, Aizawa S. Developmental patterning and evolution of the mammalian viscerocranium: genetic insights into comparative morphology. Dev Dyn. 1997;209(2):139-55. 28. Santagati F, Minoux M, Ren SY, Rijli FM. Temporal requirement of Hoxa2 in cranial neural crest skeletal morphogenesis. Development. 2005;132(22):4927-36. 29. Kubota Y, Ito K. Chemotactic migration of mesencephalic neural crest cells in the mouse. Dev Dyn. 2000;217(2):170-9. 30. Davy A, Soriano P. Ephrin-B2 forward signaling regulates somite patterning and neural crest cell development. Dev Biol. 2007;304(1):182-93. 31. Lalani SR, Safiullah AM, Molinari LM, Fernbach SD, Martin DM, Belmont JW. SEMA3E mutation in a patient with CHARGE syndrome. J Med Genet. 2004;41(7):e94. 32. Hunt P, Whiting J, Nonchev S, Sham MH, Marshall H, Graham A, et al. The branchial Hox code and its implications for gene regulation, patterning of the nervous system and head evolution. Development. 1991;Suppl 2:63-77. 33. Lumsden A, Krumlauf R. Patterning the vertebrate neuraxis. Science. 1996;274(5290):1109-15. 34. Couly G, Grapin-Botton A, Coltey P, Ruhin B, Le Douarin NM. Determination of the identity of the derivatives of the cephalic neural crest: incompatibility between Hox gene expression and lower jaw development. Development. 1998;125(17):3445-59. 35. Hunt P, Clarke JD, Buxton P, Ferretti P, Thorogood P. Segmentation, crest prespecification and the control of facial form. Eur J Oral Sci. 1998;106 Suppl 1:12-8. 36. Trainor PA, Ariza-McNaughton L, Krumlauf R. Role of the isthmus and FGFs in resolving the paradox of neural crest plasticity and prepatterning. Science. 2002;295(5558):1288-91. 37. Trainor P, Krumlauf R. Plasticity in mouse neural crest cells reveals a new patterning role for cranial mesoderm. Nat Cell Biol. 2000;2(2):96-102. 38. Gendron-Maguire M, Mallo M, Zhang M, Gridley T. Hoxa-2 mutant mice exhibit homeotic transformation of skeletal elements derived from cranial neural crest. Cell. 1993;75(7):1317-31. 39. Pasqualetti M, Ori M, Nardi I, Rijli FM. Ectopic Hoxa2 induction after neural crest migration results in homeosis of jaw elements in Xenopus. Development. 2000;127(24):5367-78. 40. Depew MJ, Simpson CA, Morasso M, Rubenstein JL. Reassessing the Dlx code: the genetic regulation of branchial arch skeletal pattern and development. J Anat. 2005;207(5):501-61. 41. Qiu M, Bulfone A, Martinez S, Meneses JJ, Shimamura K, Pedersen RA, et al. Null mutation of Dlx-2 results in abnormal morphogenesis of proximal first and second branchial arch derivatives and abnormal differentiation in the forebrain. Genes Dev. 1995;9(20):2523-38. 42. Depew MJ, Liu JK, Long JE, Presley R, Meneses JJ, Pedersen RA, et al. Dlx5 regulates regional development of the branchial arches and sensory capsules. Development. 1999;126(17):3831-46. 43. Acampora D, Merlo GR, Paleari L, Zerega B, Postiglione MP, Mantero S, et al. Craniofacial, vestibular and bone defects in mice lacking the Distal-less-related gene Dlx5. Development. 1999;126(17):3795-809. 44. del Barrio MG, Nieto MA. Overexpression of Snail family members highlights their ability to promote chick neural crest formation. Development. 2002;129(7):1583-93. 45. Graham A. Development of the pharyngeal arches. Am J Med Genet A. 2003;119a(3):251-6.

Page 69: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

61

46. Couly G, Creuzet S, Bennaceur S, Vincent C, Le Douarin NM. Interactions between Hox-negative cephalic neural crest cells and the foregut endoderm in patterning the facial skeleton in the vertebrate head. Development. 2002;129(4):1061-73. 47. Ruhin B, Creuzet S, Vincent C, Benouaiche L, Le Douarin NM, Couly G. Patterning of the hyoid cartilage depends upon signals arising from the ventral foregut endoderm. Dev Dyn. 2003;228(2):239-46. 48. Rinon A, Lazar S, Marshall H, Buchmann-Moller S, Neufeld A, Elhanany-Tamir H, et al. Cranial neural crest cells regulate head muscle patterning and differentiation during vertebrate embryogenesis. Development. 2007;134(17):3065-75. 49. Grenier J, Teillet MA, Grifone R, Kelly RG, Duprez D. Relationship between neural crest cells and cranial mesoderm during head muscle development. PLoS One. 2009;4(2):e4381. 50. Rossant J, Tam PPL. Mouse Development

Patterning, Morphogenesis, and Organogenesis. San Diego: Academic Press [Imprint]

Elsevier Science & Technology Books; 2002. Available from: http://myaccess.library.utoronto.ca/login?url=http://books.scholarsportal.info/viewdoc.html?id=/ebooks/ebooks0/elsevier/2009-12-02/2/9780125979511Available from: http://myaccess.library.utoronto.ca/login?url=http://www.sciencedirect.com/science/book/9780125979511. 51. Moore KL, Persaud TVN. Before we are born : essentials of embryology and birth defects. 6th ed. Philadelphia: Saunders; 2003. xv, 448 p. p. 52. Kaufman M. The Atlas of Mouse Development. Revised Edition ed. London: Academic Press Limited; 1992. 53. Helms JA, Cordero D, Tapadia MD. New insights into craniofacial morphogenesis. Development. 2005;132(5):851-61. 54. Proffit WR, Fields HW, Sarver DM. Contemporary orthodontics. 5th ed. St. Louis, Mo.: Elsevier/Mosby; 2013. xiii, 754 p. p. 55. Dixon A, Hoyte D, Ronning O. Fundamentals of Craniofacial Growth. Didier D, editor. New York: CRC Press; 1997. 56. Pavlov MI, Sautier JM, Oboeuf M, Asselin A, Berdal A. Chondrogenic differentiation during midfacial development in the mouse: in vivo and in vitro studies. Biol Cell. 2003;95(2):75-86. 57. Nanci A, Ten Cate AR. Ten Cate's oral histology : development, structure, and function. 8th ed. St. Louis, Mo.: Elsevier; 2013. xiii, 379 p. p. 58. Kollar EJ, Baird GR. Tissue interactions in embryonic mouse tooth germs. II. The inductive role of the dental papilla. J Embryol Exp Morphol. 1970;24(1):173-86. 59. Braybrook C, Warry G, Howell G, Mandryko V, Arnason A, Bjornsson A, et al. Physical and transcriptional mapping of the X-linked cleft palate and ankyloglossia (CPX) critical region. Hum Genet. 2001;108(6):537-45. 60. Lan Y, Ryan RC, Zhang Z, Bullard SA, Bush JO, Maltby KM, et al. Expression of Wnt9b and activation of canonical Wnt signaling during midfacial morphogenesis in mice. Dev Dyn. 2006;235(5):1448-54. 61. Brugmann SA, Tapadia MD, Helms JA. The molecular origins of species-specific facial pattern. Curr Top Dev Biol. 2006;73:1-42. 62. Miettinen PJ, Chin JR, Shum L, Slavkin HC, Shuler CF, Derynck R, et al. Epidermal growth factor receptor function is necessary for normal craniofacial development and palate closure. Nat Genet. 1999;22(1):69-73.

Page 70: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

62

63. Dixon J, Jones NC, Sandell LL, Jayasinghe SM, Crane J, Rey JP, et al. Tcof1/Treacle is required for neural crest cell formation and proliferation deficiencies that cause craniofacial abnormalities. Proc Natl Acad Sci U S A. 2006;103(36):13403-8. 64. Anderson RM, Stottmann RW, Choi M, Klingensmith J. Endogenous bone morphogenetic protein antagonists regulate mammalian neural crest generation and survival. Dev Dyn. 2006;235(9):2507-20. 65. Mayor R, Guerrero N, Martinez C. Role of FGF and noggin in neural crest induction. Dev Biol. 1997;189(1):1-12. 66. Garcia-Castro MI, Marcelle C, Bronner-Fraser M. Ectodermal Wnt function as a neural crest inducer. Science. 2002;297(5582):848-51. 67. Hu D, Helms JA. The role of sonic hedgehog in normal and abnormal craniofacial morphogenesis. Development. 1999;126(21):4873-84. 68. Sperber GH, Guttmann GD, Sperber SM. Craniofacial embryogenetics and development. 2nd ed. Shelton, CT: People's Medical Pub. House USA; 2010. 250 p. p. 69. Zoupa M, Seppala M, Mitsiadis T, Cobourne MT. Tbx1 is expressed at multiple sites of epithelial-mesenchymal interaction during early development of the facial complex. Int J Dev Biol. 2006;50(5):504-10. 70. Walker MB, Trainor PA. Craniofacial malformations: intrinsic vs extrinsic neural crest cell defects in Treacher Collins and 22q11 deletion syndromes. Clin Genet. 2006;69(6):471-9. 71. Kimmel CB, Ullmann B, Walker M, Miller CT, Crump JG. Endothelin 1-mediated regulation of pharyngeal bone development in zebrafish. Development. 2003;130(7):1339-51. 72. Szabo-Rogers HL, Geetha-Loganathan P, Nimmagadda S, Fu KK, Richman JM. FGF signals from the nasal pit are necessary for normal facial morphogenesis. Dev Biol. 2008;318(2):289-302. 73. Trumpp A, Depew MJ, Rubenstein JL, Bishop JM, Martin GR. Cre-mediated gene inactivation demonstrates that FGF8 is required for cell survival and patterning of the first branchial arch. Genes Dev. 1999;13(23):3136-48. 74. Blentic A, Tandon P, Payton S, Walshe J, Carney T, Kelsh RN, et al. The emergence of ectomesenchyme. Dev Dyn. 2008;237(3):592-601. 75. Grigoriou M, Tucker AS, Sharpe PT, Pachnis V. Expression and regulation of Lhx6 and Lhx7, a novel subfamily of LIM homeodomain encoding genes, suggests a role in mammalian head development. Development. 1998;125(11):2063-74. 76. Tucker AS, Yamada G, Grigoriou M, Pachnis V, Sharpe PT. Fgf-8 determines rostral-caudal polarity in the first branchial arch. Development. 1999;126(1):51-61. 77. Abe E, Yamamoto M, Taguchi Y, Lecka-Czernik B, O'Brien CA, Economides AN, et al. Essential requirement of BMPs-2/4 for both osteoblast and osteoclast formation in murine bone marrow cultures from adult mice: antagonism by noggin. J Bone Miner Res. 2000;15(4):663-73. 78. Hughes FJ, Collyer J, Stanfield M, Goodman SA. The effects of bone morphogenetic protein-2, -4, and -6 on differentiation of rat osteoblast cells in vitro. Endocrinology. 1995;136(6):2671-7. 79. Bandyopadhyay A, Yadav PS, Prashar P. BMP signaling in development and diseases: a pharmacological perspective. Biochem Pharmacol. 2013;85(7):857-64. 80. Lan Y, Jiang R. Sonic hedgehog signaling regulates reciprocal epithelial-mesenchymal interactions controlling palatal outgrowth. Development. 2009;136(8):1387-96. 81. Dudas M, Sridurongrit S, Nagy A, Okazaki K, Kaartinen V. Craniofacial defects in mice lacking BMP type I receptor Alk2 in neural crest cells. Mech Dev. 2004;121(2):173-82. 82. Shigetani Y, Nobusada Y, Kuratani S. Ectodermally derived FGF8 defines the maxillomandibular region in the early chick embryo: epithelial-mesenchymal interactions in the specification of the craniofacial ectomesenchyme. Dev Biol. 2000;228(1):73-85. 83. Haworth KE, Healy C, Morgan P, Sharpe PT. Regionalisation of early head ectoderm is regulated by endoderm and prepatterns the orofacial epithelium. Development. 2004;131(19):4797-806.

Page 71: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

63

84. Haworth KE, Wilson JM, Grevellec A, Cobourne MT, Healy C, Helms JA, et al. Sonic hedgehog in the pharyngeal endoderm controls arch pattern via regulation of Fgf8 in head ectoderm. Dev Biol. 2007;303(1):244-58. 85. Albertson RC, Yelick PC. Fgf8 haploinsufficiency results in distinct craniofacial defects in adult zebrafish. Dev Biol. 2007;306(2):505-15. 86. Ahlgren SC, Bronner-Fraser M. Inhibition of sonic hedgehog signaling in vivo results in craniofacial neural crest cell death. Curr Biol. 1999;9(22):1304-14. 87. Washington Smoak I, Byrd NA, Abu-Issa R, Goddeeris MM, Anderson R, Morris J, et al. Sonic hedgehog is required for cardiac outflow tract and neural crest cell development. Dev Biol. 2005;283(2):357-72. 88. Brito JM, Teillet MA, Le Douarin NM. An early role for sonic hedgehog from foregut endoderm in jaw development: ensuring neural crest cell survival. Proc Natl Acad Sci U S A. 2006;103(31):11607-12. 89. Eberhart JK, Swartz ME, Crump JG, Kimmel CB. Early Hedgehog signaling from neural to oral epithelium organizes anterior craniofacial development. Development. 2006;133(6):1069-77. 90. Wada N, Javidan Y, Nelson S, Carney TJ, Kelsh RN, Schilling TF. Hedgehog signaling is required for cranial neural crest morphogenesis and chondrogenesis at the midline in the zebrafish skull. Development. 2005;132(17):3977-88. 91. Hu D, Marcucio RS, Helms JA. A zone of frontonasal ectoderm regulates patterning and growth in the face. Development. 2003;130(9):1749-58. 92. Hu D, Marcucio RS. Unique organization of the frontonasal ectodermal zone in birds and mammals. Dev Biol. 2009;325(1):200-10. 93. Wu P, Jiang TX, Shen JY, Widelitz RB, Chuong CM. Morphoregulation of avian beaks: comparative mapping of growth zone activities and morphological evolution. Dev Dyn. 2006;235(5):1400-12. 94. Yamamoto S, Nishimura O, Misaki K, Nishita M, Minami Y, Yonemura S, et al. Cthrc1 Selectively Activates the Planar Cell Polarity Pathway of Wnt Signaling by Stabilizing the Wnt-Receptor Complex. Developmental Cell. 2008;15(1):23-36. 95. Miao CG, Yang YY, He X, Li XF, Huang C, Huang Y, et al. Wnt signaling pathway in rheumatoid arthritis, with special emphasis on the different roles in synovial inflammation and bone remodeling. Cell Signal. 2013;25(10):2069-78. 96. Logan CY, Nusse R. The Wnt signaling pathway in development and disease. Annu Rev Cell Dev Biol. 2004;20:781-810. 97. Krause U, Gregory CA. Potential of modulating Wnt signaling pathway toward the development of bone anabolic agent. Curr Mol Pharmacol. 2012;5(2):164-73. 98. De A. Wnt/Ca2+ signaling pathway: a brief overview. Acta Biochim Biophys Sin (Shanghai). 2011;43(10):745-56. 99. Sheldahl LC, Park M, Malbon CC, Moon RT. Protein kinase C is differentially stimulated by Wnt and Frizzled homologs in a G-protein-dependent manner. Curr Biol. 1999;9(13):695-8. 100. Kuhl M, Sheldahl LC, Park M, Miller JR, Moon RT. The Wnt/Ca2+ pathway: a new vertebrate Wnt signaling pathway takes shape. Trends Genet. 2000;16(7):279-83. 101. Dale RM, Sisson BE, Topczewski J. The emerging role of Wnt/PCP signaling in organ formation. Zebrafish. 2009;6(1):9-14. 102. Karner C, Wharton KA, Carroll TJ. Apical-basal polarity, Wnt signaling and vertebrate organogenesis. Semin Cell Dev Biol. 2006;17(2):214-22. 103. Karner C, Wharton KA, Jr., Carroll TJ. Planar cell polarity and vertebrate organogenesis. Semin Cell Dev Biol. 2006;17(2):194-203. 104. He F, Xiong W, Yu X, Espinoza-Lewis R, Liu C, Gu S, et al. Wnt5a regulates directional cell migration and cell proliferation via Ror2-mediated noncanonical pathway in mammalian palate development. Development. 2008;135(23):3871-9.

Page 72: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

64

105. Cadigan KM, Nusse R. Wnt signaling: a common theme in animal development. Genes Dev. 1997;11(24):3286-305. 106. Grigoryan T, Wend P, Klaus A, Birchmeier W. Deciphering the function of canonical Wnt signals in development and disease: conditional loss- and gain-of-function mutations of beta-catenin in mice. Genes Dev. 2008;22(17):2308-41. 107. Brault V, Moore R, Kutsch S, Ishibashi M, Rowitch DH, McMahon AP, et al. Inactivation of the beta-catenin gene by Wnt1-Cre-mediated deletion results in dramatic brain malformation and failure of craniofacial development. Development. 2001;128(8):1253-64. 108. Niemann S, Zhao C, Pascu F, Stahl U, Aulepp U, Niswander L, et al. Homozygous WNT3 mutation causes tetra-amelia in a large consanguineous family. Am J Hum Genet. 2004;74(3):558-63. 109. Geetha-Loganathan P, Nimmagadda S, Antoni L, Fu K, Whiting CJ, Francis-West P, et al. Expression of WNT signalling pathway genes during chicken craniofacial development. Dev Dyn. 2009;238(5):1150-65. 110. Chai Y, Maxson RE, Jr. Recent advances in craniofacial morphogenesis. Dev Dyn. 2006;235(9):2353-75. 111. Koay MA, Brown MA. Genetic disorders of the LRP5-Wnt signalling pathway affecting the skeleton. Trends Mol Med. 2005;11(3):129-37. 112. Kikuchi A, Yamamoto H, Sato A, Matsumoto S. New insights into the mechanism of Wnt signaling pathway activation. Int Rev Cell Mol Biol. 2011;291:21-71. 113. Hosseini-Farahabadi S, Geetha-Loganathan P, Fu K, Nimmagadda S, Yang HJ, Richman JM. Dual functions for WNT5A during cartilage development and in disease. Matrix Biol. 2013;32(5):252-64. 114. Person AD, Beiraghi S, Sieben CM, Hermanson S, Neumann AN, Robu ME, et al. WNT5A mutations in patients with autosomal dominant Robinow syndrome. Dev Dyn. 2010;239(1):327-37. 115. Pyagay P, Heroult M, Wang Q, Lehnert W, Belden J, Liaw L, et al. Collagen triple helix repeat containing 1, a novel secreted protein in injured and diseased arteries, inhibits collagen expression and promotes cell migration. Circulation Research. 2005;96(2):261-8. 116. Chen YL, Wang TH, Hsu HC, Yuan RH, Jeng YM. Overexpression of CTHRC1 in hepatocellular carcinoma promotes tumor invasion and predicts poor prognosis. PLoS One. 2013;8(7):e70324. 117. Lindner V. [cited 2014 May 4]. Available from: http://sackler.tufts.edu/Faculty-and-Research/Faculty-Research-Pages/~/media/Sackler/Page%20Images/Faculty%20Research%20Page%20Images/Lindner%20Fig%202.jpg. 118. Wang Y, Xu A, Knight C, Xu LY, Cooper GJ. Hydroxylation and glycosylation of the four conserved lysine residues in the collagenous domain of adiponectin. Potential role in the modulation of its insulin-sensitizing activity. J Biol Chem. 2002;277(22):19521-9. 119. Takeshita S, Fumoto T, Matsuoka K, Park KA, Aburatani H, Kato S, et al. Osteoclast-secreted CTHRC1 in the coupling of bone resorption to formation. J Clin Invest. 2013;123(9):3914-24. 120. Diez JJ, Iglesias P. The role of the novel adipocyte-derived hormone adiponectin in human disease. Eur J Endocrinol. 2003;148(3):293-300. 121. Liu G, Sengupta PK, Jamal B, Yang HY, Bouchie MP, Lindner V, et al. N-glycosylation induces the CTHRC1 protein and drives oral cancer cell migration. J Biol Chem. 2013;288(28):20217-27. 122. Durmus T, LeClair RJ, Park KS, Terzic A, Yoon JK, Lindner V. Expression analysis of the novel gene collagen triple helix repeat containing-1 (Cthrc1). Gene Expression Patterns. 2006;6(8):935-40. 123. LeClair R, Lindner V. The Role of Collagen Triple Helix Repeat Containing 1 in Injured Arteries, Collagen Expression, and Transforming Growth Factor beta Signaling. Trends in Cardiovascular Medicine. 2007;17(6):202-5. 124. LeClair RJ, Durmus T, Wang Q, Pyagay P, Terzic A, Lindner V. Cthrc1 is a novel inhibitor of transforming growth factor-beta signaling and neointimal lesion formation. Circulation Research. 2007;100(6):826-33.

Page 73: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

65

125. Lindner V. Vascular repair processes mediated by transforming growth factor-beta. Z Kardiol. 2001;90 Suppl 3:17-22. 126. Janssens K, ten Dijke P, Janssens S, Van Hul W. Transforming growth factor-beta1 to the bone. Endocr Rev. 2005;26(6):743-74. 127. Ghosh AK. Factors involved in the regulation of type I collagen gene expression: implication in fibrosis. Exp Biol Med (Maywood). 2002;227(5):301-14. 128. Tang L, Dai DL, Su M, Martinka M, Li G, Zhou Y. Aberrant expression of collagen triple helix repeat containing 1 in human solid cancers. Clinical Cancer Research. 2006;12(12):3716-22. 129. Yang Y. Skeletal morphogenesis during embryonic development. Crit Rev Eukaryot Gene Expr. 2009;19(3):197-218. 130. Toy J, Yang JM, Leppert GS, Sundin OH. The optx2 homeobox gene is expressed in early precursors of the eye and activates retina-specific genes. Proc Natl Acad Sci U S A. 1998;95(18):10643-8. 131. Caetano-Lopes J, Canhao H, Fonseca JE. Osteoblasts and bone formation. Acta Reumatol Port. 2007;32(2):103-10. 132. Olsen BR, Reginato AM, Wang W. Bone development. Annu Rev Cell Dev Biol. 2000;16:191-220. 133. Wright E, Hargrave MR, Christiansen J, Cooper L, Kun J, Evans T, et al. The Sry-related gene Sox9 is expressed during chondrogenesis in mouse embryos. Nat Genet. 1995;9(1):15-20. 134. Harada S, Rodan GA. Control of osteoblast function and regulation of bone mass. Nature. 2003;423(6937):349-55. 135. Kimura H, Kwan KM, Zhang Z, Deng JM, Darnay BG, Behringer RR, et al. Cthrc1 is a positive regulator of osteoblastic bone formation. PLoS One. 2008;3(9):e3174. 136. Kimura H, Kwan KM, Zhang Z, Deng JM, Darnay BG, Behringer RR, et al. Cthrci is a positive regulator of osteoblastic bone formation. PLoS ONE. 2008;3(9). 137. Kim JH, Baek TH, Yim HS, Kim KH, Jeong SH, Kang HB, et al. Collagen triple helix repeat containing-1 (CTHRC1) expression in invasive ductal carcinoma of the breast: the impact on prognosis and correlation to clinicopathologic features. Pathol Oncol Res. 2013;19(4):731-7. 138. Leclair RJ, Wang Q, Benson MA, Prudovsky I, Lindner V. Intracellular localization of Cthrc1 characterizes differentiated smooth muscle. Arteriosclerosis, Thrombosis, and Vascular Biology. 2008;28(7):1332-8. 139. Kelley MW. Leading Wnt down a PCP Path: Cthrc1 Acts as a Coreceptor in the Wnt-PCP Pathway. Developmental Cell. 2008;15(1):7-8. 140. Apra C, Richard L, Coulpier F, Blugeon C, Gilardi-Hebenstreit P, Vallat JM, et al. Cthrc1 is a negative regulator of myelination in schwann cells. Glia. 2012;60(3):393-403. 141. Kharaishvili G, Cizkova M, Bouchalova K, Mgebrishvili G, Kolar Z, Bouchal J. Collagen triple helix repeat containing 1 protein, periostin and versican in primary and metastatic breast cancer: An immunohistochemical study. Journal of Clinical Pathology. 2011;64(11):977-82. 142. Turashvili G, Bouchal J, Baumforth K, Wei W, Dziechciarkova M, Ehrmann J, et al. Novel markers for differentiation of lobular and ductal invasive breast carcinomas by laser microdissection and microarray analysis. BMC Cancer. 2007;7(55). 143. Orloff M, Peterson C, He X, Ganapathi S, Heald B, Yang YR, et al. Germline mutations in MSR1, ASCC1, and CTHRC1 in patients with Barrett esophagus and esophageal adenocarcinoma. JAMA - Journal of the American Medical Association. 2011;306(4):410-9. 144. O'Connell MJ, Lavery I, Yothers G, Paik S, Clark-Langone KM, Lopatin M, et al. Relationship between tumor gene expression and recurrence in four independent studies of patients with stage II/III colon cancer treated with surgery alone or surgery plus adjuvant fluorouracil plus leucovorin. J Clin Oncol. 2010;28(25):3937-44. 145. Park M, Moon RT. The planar cell-polarity gene stbm regulates cell behaviour and cell fate in vertebrate embryos. Nat Cell Biol. 2002;4(1):20-5.

Page 74: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

66

146. Li Y, Dudley AT. Noncanonical frizzled signaling regulates cell polarity of growth plate chondrocytes. Development. 2009;136(7):1083-92. 147. Kudryavtseva E, Forde TS, Pucker AD, Adarichev VA. Wnt signaling genes of murine chromosome 15 are involved in sex-affected pathways of inflammatory arthritis. Arthritis and rheumatism. 2012;64(4):1057-68. 148. Gong SG. Characterization of olfactory nerve abnormalities in Twirler mice. Differentiation. 2001;69(1):58-65. 149. Mort RL, Hay L, Jackson IJ. Ex vivo live imaging of melanoblast migration in embryonic mouse skin. Pigment Cell Melanoma Res. 23. England2010. p. 299-301. 150. Avery JK. Oral development and histology. 2nd ed. New York Stuttgart ; New York: Thieme Medical Publishers ; Georg Thieme Verlag; 1994. x, 422 p. p.

151. Mayor R, Theveneau E. The role of the non-canonical Wnt-planar cell polarity pathway in neural crest migration. Biochem J. 2014;457(1):19-26. 152. Stemple DL. Structure and function of the notochord: an essential organ for chordate development. Development. 2005;132(11):2503-12. 153. Hall BK, Miyake T. Divide, accumulate, differentiate: cell condensation in skeletal development revisited. Int J Dev Biol. 1995;39(6):881-93. 154. Francis-West P, Ladher R, Barlow A, Graveson A. Signalling interactions during facial development. Mech Dev. 1998;75(1-2):3-28. 155. Abzhanov A, Protas M, Grant BR, Grant PR, Tabin CJ. Bmp4 and morphological variation of beaks in Darwin's finches. Science. 2004;305(5689):1462-5. 156. Wu P, Jiang TX, Suksaweang S, Widelitz RB, Chuong CM. Molecular shaping of the beak. Science. 2004;305(5689):1465-6. 157. Abzhanov A, Tabin CJ. Shh and Fgf8 act synergistically to drive cartilage outgrowth during cranial development. Dev Biol. 2004;273(1):134-48. 158. Lan Y, Jia S, Jiang R. Molecular patterning of the mammalian dentition. Semin Cell Dev Biol. 2014;25-26:61-70. 159. King JA, Marker PC, Seung KJ, Kingsley DM. BMP5 and the molecular, skeletal, and soft-tissue alterations in short ear mice. Dev Biol. 1994;166(1):112-22. 160. Jones CM, Lyons KM, Hogan BL. Involvement of Bone Morphogenetic Protein-4 (BMP-4) and Vgr-1 in morphogenesis and neurogenesis in the mouse. Development. 1991;111(2):531-42. 161. Thesleff I, Vaahtokari A, Vainio S, Jowett A. Molecular mechanisms of cell and tissue interactions during early tooth development. Anat Rec. 1996;245(2):151-61. 162. Hall BK, Miyake T. The membranous skeleton: the role of cell condensations in vertebrate skeletogenesis. Anat Embryol (Berl). 1992;186(2):107-24. 163. Jiang TX, Yi JR, Ying SY, Chuong CM. Activin enhances chondrogenesis of limb bud cells: stimulation of precartilaginous mesenchymal condensations and expression of NCAM. Dev Biol. 1993;155(2):545-57. 164. Mukhopadhyay P, Greene RM, Pisano MM. Expression profiling of transforming growth factor beta superfamily genes in developing orofacial tissue. Birth Defects Res A Clin Mol Teratol. 2006;76(7):528-43. 165. Kikuchi A, Yamamoto H, Sato A, Matsumoto S. Wnt5a: its signalling, functions and implication in diseases. Acta Physiol (Oxf). 2012;204(1):17-33. 166. Takeshita S. An osteoclast-derived coupling factor. Osteoporosis International. 2011;22:S516-S7. 167. Yamaguchi TP, Takada S, Yoshikawa Y, Wu N, McMahon AP. T (Brachyury) is a direct target of Wnt3a during paraxial mesoderm specification. Genes Dev. 1999;13(24):3185-90.

Page 75: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

67

168. Wan XZ, Qin WX, Tan N, Yao GF, Tan YT, Kuang WX, et al. CTHRC1 gene is overexpressed in human hepatocellular carcinomas and promotes the metastasis of human hepatocellular carcinoma cell MHCC97L. [Chinese]. Tumor. 2007;27(6):476-9+83. 169. Stohn JP, Perreault NG, Wang Q, Liaw L, Lindner V. Cthrc1, a novel hormone involved in regulation of metabolism. FASEB Journal. 2013;27. 170. Mishina Y, Snider TN. Neural crest cell signaling pathways critical to cranial bone development and pathology. Exp Cell Res. 2014. 171. Shukla V, Coumoul X, Wang RH, Kim HS, Deng CX. RNA interference and inhibition of MEK-ERK signaling prevent abnormal skeletal phenotypes in a mouse model of craniosynostosis. Nat Genet. 2007;39(9):1145-50. 172. Dab S, Sokhi R, Lee JC, Sessle BJ, Aubin JE, Gong SG. Characterization of esophageal defects in the Crouzon mouse model. Birth Defects Res A Clin Mol Teratol. 2013;97(9):578-86. 173. Kamiya N, Ye L, Kobayashi T, Mochida Y, Yamauchi M, Kronenberg HM, et al. BMP signaling negatively regulates bone mass through sclerostin by inhibiting the canonical Wnt pathway. Development. 2008;135(22):3801-11. 174. Dixon J, Brakebusch C, Fassler R, Dixon MJ. Increased levels of apoptosis in the prefusion neural folds underlie the craniofacial disorder, Treacher Collins syndrome. Hum Mol Genet. 2000;9(10):1473-80. 175. Gong SG, Mai S, Chung K, Wei K. Flrt2 and Flrt3 have overlapping and non-overlapping expression during craniofacial development. Gene Expr Patterns. 2009;9(7):497-502. 176. Yamaguchi TP, Bradley A, McMahon AP, Jones S. A Wnt5a pathway underlies outgrowth of multiple structures in the vertebrate embryo. Development. 1999;126(6):1211-23.

Page 76: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

68

Copyright Acknowledgements

Page 77: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

69

Page 78: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

70

Page 79: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

71

Page 80: Expression Analysis of CTHRC1 in the Murine Embryo during ...€¦ · 1.1.3 Facial prominences and CNCC derivatives of the face…6 1.1.4 Facial development and facial prominences…7

72