1
|
Sun Q, Huang J, Tian J, Lv C, Li Y, Yu S, Liu J, Zhang J. Key Roles of Gli1 and Ihh Signaling in Craniofacial Development. Stem Cells Dev 2024; 33:251-261. [PMID: 38623785 DOI: 10.1089/scd.2024.0036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/17/2024] Open
Abstract
The Hedgehog (Hh) signaling pathway orchestrates its influence through a dynamic interplay of Hh proteins, the cell surface receptor Ptch1, Smo, and Gli transcription factors, contributing to a myriad of developmental events. Indian Hedgehog (Ihh) and Gli zinc finger transcription factor 1 (Gli1) play crucial roles in developmental regulation within the Hh signaling pathway. Ihh regulates chondrocyte proliferation, differentiation, and bone formation, impacting the development of cranial bones, cartilage, and the temporomandibular joint (TMJ). Losing Ihh results in cranial bone malformation and decreased ossification and affects the formation of cranial base cartilage unions, TMJ condyles, and joint discs. Gli1 is predominantly expressed during early craniofacial development, and Gli1+ cells are identified as the primary mesenchymal stem cells (MSCs) for craniofacial bones, crucial for cell differentiation and morphogenesis. In addition, a complex mutual regulatory mechanism exists between Gli1 and Ihh, ensuring the normal function of the Hh signaling pathway by directly or indirectly regulating each other's expression levels. And the interaction between Ihh and Gli1 significantly impacts the normal development of craniofacial tissues. This review summarizes the pivotal roles of Gli1 and Ihh in the intricate landscape of mammalian craniofacial development and outlines the molecular regulatory mechanisms and intricate interactions governing the growth of bone and cartilage exhibited by Gli1 and Ihh, which provides new insights into potential therapeutic strategies for related diseases or researches of tissue regeneration.
Collapse
Affiliation(s)
- Qi Sun
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Jie Huang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Jingjun Tian
- Department of Orthodontics, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Changhai Lv
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Yanhong Li
- Department of Preventive Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Siyuan Yu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Juan Liu
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| | - Jun Zhang
- Department of Pediatric Dentistry, School and Hospital of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
- Yunnan Key Laboratory of Stomatology, Kunming Medical University, Kunming, Yunnan, Republic of China
| |
Collapse
|
2
|
Kouchaki H, Kamyab P, Darbeheshti F, Gharezade A, Fouladseresht H, Tabrizi R. miR-939, as an important regulator in various cancers pathogenesis, has diagnostic, prognostic, and therapeutic values: a review. J Egypt Natl Canc Inst 2024; 36:16. [PMID: 38679648 DOI: 10.1186/s43046-024-00220-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 04/06/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND MicroRNAs (miRNAs or miRs) are highly conserved non-coding RNAs with a short length (18-24 nucleotides) that directly bind to a complementary sequence within 3'-untranslated regions of their target mRNAs and regulate gene expression, post-transcriptionally. They play crucial roles in diverse biological processes, including cell proliferation, apoptosis, and differentiation. In the context of cancer, miRNAs are key regulators of growth, angiogenesis, metastasis, and drug resistance. MAIN BODY This review primarily focuses on miR-939 and its expanding roles and target genes in cancer pathogenesis. It compiles findings from various investigations. MiRNAs, due to their dysregulated expression in tumor environments, hold potential as cancer biomarkers. Several studies have highlighted the dysregulation of miR-939 expression in human cancers. CONCLUSION Our study highlights the potential of miR-939 as a valuable target in cancer diagnosis, prognosis, and treatment. The aberrant expression of miR-939, along with other miRNAs, underscores their significance in advancing our understanding of cancer biology and their promise in personalized cancer care.
Collapse
Affiliation(s)
- Hosein Kouchaki
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Parnia Kamyab
- USERN Office, Fasa University of Medical Sciences, Fasa, Iran
| | - Farzaneh Darbeheshti
- Department of Radiation Oncology, Dana Farber Cancer Institute and Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Arezou Gharezade
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hamed Fouladseresht
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Reza Tabrizi
- Clinical Research Development Unit, Valiasr Hospital, Fasa University of Medical Sciences, Fasa, Iran.
- Noncommunicable Diseases Research Center, Fasa University of Medical Science, Fasa, Iran.
| |
Collapse
|
3
|
Greer SE, Haller SJ, Lee D, Dudley AT. N-cadherin and β1 integrin coordinately regulate growth plate cartilage architecture. Mol Biol Cell 2024; 35:ar49. [PMID: 38294852 PMCID: PMC11064670 DOI: 10.1091/mbc.e23-03-0101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/07/2023] [Accepted: 01/23/2024] [Indexed: 02/01/2024] Open
Abstract
Spatial and temporal regulation of chondrocyte maturation in the growth plate drives growth of many bones. One essential event to generate the ordered cell array characterizing growth plate cartilage is the formation of chondrocyte columns in the proliferative zone via 90-degree rotation of daughter cells to align with the long axis of the bone. Previous studies have suggested crucial roles for cadherins and integrin β1 in column formation. The purpose of this study was to determine the relative contributions of cadherin- and integrin-mediated cell adhesion in column formation. Here we present new mechanistic insights generated by application of live time-lapse confocal microscopy of cranial base explant cultures, robust genetic mouse models, and new quantitative methods to analyze cell behavior. We show that conditional deletion of either the cell-cell adhesion molecule Cdh2 or the cell-matrix adhesion molecule Itgb1 disrupts column formation. Compound mutants were used to determine a potential reciprocal regulatory interaction between the two adhesion surfaces and identified that defective chondrocyte rotation in a N-cadherin mutant was restored by a heterozygous loss of integrin β1. Our results support a model for which integrin β1, and not N-cadherin, drives chondrocyte rotation and for which N-cadherin is a potential negative regulator of integrin β1 function.
Collapse
Affiliation(s)
- Sydney E. Greer
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| | - Stephen J. Haller
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| | - Donghee Lee
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| | - Andrew T. Dudley
- Department of Genetics, Cell Biology, and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198
| |
Collapse
|
4
|
Le Pabic P, Dranow DB, Hoyle DJ, Schilling TF. Zebrafish endochondral growth zones as they relate to human bone size, shape and disease. Front Endocrinol (Lausanne) 2022; 13:1060187. [PMID: 36561564 PMCID: PMC9763315 DOI: 10.3389/fendo.2022.1060187] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/17/2022] [Indexed: 12/12/2022] Open
Abstract
Research on the genetic mechanisms underlying human skeletal development and disease have largely relied on studies in mice. However, recently the zebrafish has emerged as a popular model for skeletal research. Despite anatomical differences such as a lack of long bones in their limbs and no hematopoietic bone marrow, both the cell types in cartilage and bone as well as the genetic pathways that regulate their development are remarkably conserved between teleost fish and humans. Here we review recent studies that highlight this conservation, focusing specifically on the cartilaginous growth zones (GZs) of endochondral bones. GZs can be unidirectional such as the growth plates (GPs) of long bones in tetrapod limbs or bidirectional, such as in the synchondroses of the mammalian skull base. In addition to endochondral growth, GZs play key roles in cartilage maturation and replacement by bone. Recent studies in zebrafish suggest key roles for cartilage polarity in GZ function, surprisingly early establishment of signaling systems that regulate cartilage during embryonic development, and important roles for cartilage proliferation rather than hypertrophy in bone size. Despite anatomical differences, there are now many zebrafish models for human skeletal disorders including mutations in genes that cause defects in cartilage associated with endochondral GZs. These point to conserved developmental mechanisms, some of which operate both in cranial GZs and limb GPs, as well as others that act earlier or in parallel to known GP regulators. Experimental advantages of zebrafish for genetic screens, high resolution live imaging and drug screens, set the stage for many novel insights into causes and potential therapies for human endochondral bone diseases.
Collapse
Affiliation(s)
- Pierre Le Pabic
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Willmington, NC, United States
| | - Daniel B. Dranow
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Diego J. Hoyle
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
5
|
Johnson S, Heubel B, Bredesen C, Schilling T, Le Pabic P. Cellular basis of differential endochondral growth in Lake Malawi cichlids. Dev Dyn 2022; 251:2001-2014. [PMID: 36001035 PMCID: PMC9722610 DOI: 10.1002/dvdy.529] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 01/30/2023] Open
Abstract
BACKGROUND The shape and size of skeletal elements is determined by embryonic patterning mechanisms as well as localized growth and remodeling during post-embryonic development. Differential growth between endochondral growth plates underlies many aspects of morphological diversity in tetrapods but has not been investigated in ray-finned fishes. We examined endochondral growth rates in the craniofacial skeletons of two cichlid species from Lake Malawi that acquire species-specific morphological differences during postembryonic development and quantified cellular mechanisms underlying differential growth both within and between species. RESULTS Cichlid endochondral growth rates vary greatly (50%-60%) between different growth zones within a species, between different stages for the same growth zone, and between homologous growth zones in different species. Differences in cell proliferation and/or cell enlargement underlie much of this differential growth, albeit in different proportions. Strikingly, differences in extracellular matrix production do not correlate with growth rate differences. CONCLUSIONS Differential endochondral growth drives many aspects of craniofacial morphological diversity in cichlids. Cellular proliferation and enlargement, but not extracellular matrix deposition, underlie this differential growth and this appears conserved in Osteichthyes. Cell enlargement is observed in some but not all cichlid growth zones and the degree to which it occurs resembles slower growing mammalian growth plates.
Collapse
Affiliation(s)
- Savannah Johnson
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC
| | - Brian Heubel
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC
| | - Carson Bredesen
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC
| | - Thomas Schilling
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA
| | - Pierre Le Pabic
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, NC
| |
Collapse
|
6
|
Karele EN, Paze AN. Chordoma: To know means to recognize. Biochim Biophys Acta Rev Cancer 2022; 1877:188796. [PMID: 36089204 DOI: 10.1016/j.bbcan.2022.188796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/13/2022] [Accepted: 09/03/2022] [Indexed: 10/14/2022]
Abstract
Chordoma is a rare type of bone cancer characterized by its locally aggressive and destructive behavior. Chordoma is located in one of the three primary regions: skull base/clivus, sacrum or mobile spine. Chordoma grows slowly, therefore its insidious onset leads to delayed diagnosis, accounting for the low survival rates. Treatment centers around successful en bloc resection with negative margins, though, considering the anatomically constrained site of growth, it frequently requires adjuvant radiotherapy. This article analyzes the existing literature with the aim to provide a better insight in the current state of research in chordoma classification, characteristics, and management.
Collapse
Affiliation(s)
- Emija Nikola Karele
- Faculty of Medicine, Riga Stradins University, 16 Dzirciema Street, Riga LV-1007, Latvia.
| | - Anda Nikola Paze
- Faculty of Medicine, Riga Stradins University, 16 Dzirciema Street, Riga LV-1007, Latvia.
| |
Collapse
|
7
|
Hallett SA, Ono W, Franceschi RT, Ono N. Cranial Base Synchondrosis: Chondrocytes at the Hub. Int J Mol Sci 2022; 23:7817. [PMID: 35887171 PMCID: PMC9317907 DOI: 10.3390/ijms23147817] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 07/10/2022] [Accepted: 07/13/2022] [Indexed: 01/04/2023] Open
Abstract
The cranial base is formed by endochondral ossification and functions as a driver of anteroposterior cranial elongation and overall craniofacial growth. The cranial base contains the synchondroses that are composed of opposite-facing layers of resting, proliferating and hypertrophic chondrocytes with unique developmental origins, both in the neural crest and mesoderm. In humans, premature ossification of the synchondroses causes midfacial hypoplasia, which commonly presents in patients with syndromic craniosynostoses and skeletal Class III malocclusion. Major signaling pathways and transcription factors that regulate the long bone growth plate-PTHrP-Ihh, FGF, Wnt, BMP signaling and Runx2-are also involved in the cranial base synchondrosis. Here, we provide an updated overview of the cranial base synchondrosis and the cell population within, as well as its molecular regulation, and further discuss future research opportunities to understand the unique function of this craniofacial skeletal structure.
Collapse
Affiliation(s)
- Shawn A. Hallett
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (S.A.H.); (R.T.F.)
| | - Wanida Ono
- Department of Orthodontics, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, USA;
| | - Renny T. Franceschi
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; (S.A.H.); (R.T.F.)
| | - Noriaki Ono
- Department of Diagnostic and Biomedical Sciences, University of Texas Health Science Center at Houston School of Dentistry, Houston, TX 77054, USA
| |
Collapse
|
8
|
Hsieh YL, Wei X, Wang Y, Zhang H, Qi S, Xie D, Mishina Y, Mendonça D, Hatch N, Liu F. Chondrocyte Tsc1 controls cranial base bone development by restraining the premature differentiation of synchondroses. Bone 2021; 153:116142. [PMID: 34365025 PMCID: PMC8543925 DOI: 10.1016/j.bone.2021.116142] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/30/2021] [Accepted: 08/02/2021] [Indexed: 02/05/2023]
Abstract
Cranial base bones are formed through endochondral ossification. Synchondroses are growth plates located between cranial base bones that facilitate anterior-posterior growth of the skull. Coordinated proliferation and differentiation of chondrocytes in cranial base synchondroses is essential for cranial base bone growth. Herein, we report that constitutive activation of the mechanistic target of rapamycin complex 1 (mTORC1) signaling via Tsc1 (Tuberous sclerosis 1) deletion in chondrocytes causes abnormal skull development with decreased size and rounded shape. In contrast to decreased anterior-posterior growth of the cranial base, mutant mice also exhibited significant expansion of cranial base synchondroses including the intersphenoid synchondrosis (ISS) and the spheno-occipital synchondrosis (SOS). Cranial base synchondrosis expansion in TSC1-deficient mice was accounted for by an expansion of the resting zone due to increased cell number and size without alteration in cell proliferation. Furthermore, our data showed that mTORC1 activity is inhibited in the resting and proliferating zone chondrocytes of wild type mice, and Tsc1 deletion activated mTORC1 signaling of the chondrocytes in the resting zone area. Consequently, the chondrocytes in the resting zone of TSC1-deficient mice acquired characteristics generally attributed to pre-hypertrophic chondrocytes including high mTORC1 activity, increased cell size, and increased expression level of PTH1R (Parathyroid hormone 1 receptor) and IHH (Indian hedgehog). Lastly, treatment with rapamycin, an inhibitor of mTORC1, rescued the abnormality in synchondroses. Our results established an important role for TSC1-mTORC1 signaling in regulating cranial base bone development and showed that chondrocytes in the resting zone of synchondroses are maintained in an mTORC1-inhibitory environment.
Collapse
Affiliation(s)
- Yuan-Lynn Hsieh
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Xiaoxi Wei
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; Department of Orthodontics, Hospital of Stomatology Jilin University, Changchun, Jilin 130021, China
| | - Yating Wang
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA; State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Other Research Platforms & Department of Orthodontics and Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China
| | - Honghao Zhang
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Shuqun Qi
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Di Xie
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Yuji Mishina
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Daniela Mendonça
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Nan Hatch
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA
| | - Fei Liu
- Department of Biologic & Materials Sciences and Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI 48109, USA.
| |
Collapse
|
9
|
Wan Y, Szabo-Rogers HL. Chondrocyte Polarity During Endochondral Ossification Requires Protein-Protein Interactions Between Prickle1 and Dishevelled2/3. J Bone Miner Res 2021; 36:2399-2412. [PMID: 34423861 DOI: 10.1002/jbmr.4428] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/19/2021] [Accepted: 08/18/2021] [Indexed: 12/26/2022]
Abstract
The expansion and growth of the endochondral skeleton requires organized cell behaviors that control chondrocyte maturation and oriented division. In other organs, these processes are accomplished through Wnt/planar cell polarity (Wnt/PCP) signaling pathway and require the protein-protein interactions of core components including Prickle1 (PK1) and Dishevelled (DVL). To determine the function of Wnt/PCP signaling in endochondral ossification of the cranial base and limb, we utilized the Prickle1Beetlejuice (Pk1Bj ) mouse line. The Pk1Bj allele has a missense mutation in the PK1 LIM1 domain that results in a hypomorphic protein. Similar to human patients with Robinow syndrome, the Prickle1Bj/Bj mouse mutants lack growth plate expansion resulting in shorter limbs and midfacial hypoplasia. Within the Prickle1Bj/Bj limb and cranial base growth plates we observe precocious maturation of chondrocytes and stalling of terminal differentiation. Intriguingly, we observed that the growth plate chondrocytes have randomized polarity based on the location of the primary cilia and the location of PRICKLE1, DVL2, and DVL3 localization. Importantly, mutant PK1Bj protein has decreased protein-protein interactions with both DVL2 and DVL3 in chondrocytes as revealed by in vivo co-immunoprecipitation and proximity ligation assays. Finally, we propose a model where the interaction between the Prickle1 LIM1 domain and DVL2 and DVL3 contributes to chondrocyte polarity and contributes to proximal-distal outgrowth of endochondral elements. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Yong Wan
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heather L Szabo-Rogers
- Department of Oral and Craniofacial Sciences, School of Dental Medicine, Center for Craniofacial Regeneration, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Developmental Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,McGowan Institute for Regenerative Medicine, Pittsburgh, PA, USA
| |
Collapse
|
10
|
Chandrasekaran P, Kwok B, Han B, Adams SM, Wang C, Chery DR, Mauck RL, Dyment NA, Lu XL, Frank DB, Koyama E, Birk DE, Han L. Type V Collagen Regulates the Structure and Biomechanics of TMJ Condylar Cartilage: A Fibrous-Hyaline Hybrid. Matrix Biol 2021; 102:1-19. [PMID: 34314838 DOI: 10.1016/j.matbio.2021.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 05/26/2021] [Accepted: 07/15/2021] [Indexed: 12/20/2022]
Abstract
This study queried the role of type V collagen in the post-natal growth of temporomandibular joint (TMJ) condylar cartilage, a hybrid tissue with a fibrocartilage layer covering a secondary hyaline cartilage layer. Integrating outcomes from histology, immunofluorescence imaging, electron microscopy and atomic force microscopy-based nanomechanical tests, we elucidated the impact of type V collagen reduction on TMJ condylar cartilage growth in the type V collagen haploinsufficiency and inducible knockout mice. Reduction of type V collagen led to significantly thickened collagen fibrils, decreased tissue modulus, reduced cell density and aberrant cell clustering in both the fibrous and hyaline layers. Post-natal growth of condylar cartilage involves the chondrogenesis of progenitor cells residing in the fibrous layer, which gives rise to the secondary hyaline layer. Loss of type V collagen resulted in reduced proliferation of these cells, suggesting a possible role of type V collagen in mediating the progenitor cell niche. When the knockout of type V collagen was induced in post-weaning mice after the start of physiologic TMJ loading, the hyaline layer exhibited pronounced thinning, supporting an interplay between type V collagen and occlusal loading in condylar cartilage growth. The phenotype in hyaline layer can thus be attributed to the impact of type V collagen on the mechanically regulated progenitor cell activities. In contrast, knee cartilage does not contain the progenitor cell population at post-natal stages, and develops normal structure and biomechanical properties with the loss of type V collagen. Therefore, in the TMJ, in addition to its established role in regulating the assembly of collagen I fibrils, type V collagen also impacts the mechanoregulation of progenitor cell activities in the fibrous layer. We expect such knowledge to establish a foundation for understanding condylar cartilage matrix development and regeneration, and to yield new insights into the TMJ symptoms in patients with classic Ehlers-Danlos syndrome, a genetic disease due to autosomal mutation of type V collagen.
Collapse
Affiliation(s)
- Prashant Chandrasekaran
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Bryan Kwok
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Biao Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Sheila M Adams
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Chao Wang
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Daphney R Chery
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States
| | - Robert L Mauck
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Translational Musculoskeletal Research Center, Corporal Michael J. Crescenz Veterans Administration Medical Center, Philadelphia, PA 19104, United States
| | - Nathaniel A Dyment
- McKay Orthopaedic Research Laboratory, Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - X Lucas Lu
- Department of Mechanical Engineering, University of Delaware, Newark, DE 19716, United States
| | - David B Frank
- Penn-CHOP Lung Biology Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Penn Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States; Division of Pediatric Cardiology, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - Eiki Koyama
- Translational Research Program in Pediatric Orthopaedics, Division of Orthopaedic Surgery, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, United States
| | - David E Birk
- Department of Molecular Pharmacology and Physiology, Morsani School of Medicine, University of South Florida, Tampa, FL 33612, United States
| | - Lin Han
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, United States.
| |
Collapse
|
11
|
Unger CM, Devine J, Hallgrímsson B, Rolian C. Selection for increased tibia length in mice alters skull shape through parallel changes in developmental mechanisms. eLife 2021; 10:e67612. [PMID: 33899741 PMCID: PMC8118654 DOI: 10.7554/elife.67612] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
Bones in the vertebrate cranial base and limb skeleton grow by endochondral ossification, under the control of growth plates. Mechanisms of endochondral ossification are conserved across growth plates, which increases covariation in size and shape among bones, and in turn may lead to correlated changes in skeletal traits not under direct selection. We used micro-CT and geometric morphometrics to characterize shape changes in the cranium of the Longshanks mouse, which was selectively bred for longer tibiae. We show that Longshanks skulls became longer, flatter, and narrower in a stepwise process. Moreover, we show that these morphological changes likely resulted from developmental changes in the growth plates of the Longshanks cranial base, mirroring changes observed in its tibia. Thus, indirect and non-adaptive morphological changes can occur due to developmental overlap among distant skeletal elements, with important implications for interpreting the evolutionary history of vertebrate skeletal form.
Collapse
Affiliation(s)
- Colton M Unger
- Department of Biological Sciences, University of CalgaryCalgaryCanada
- McCaig Institute for Bone and Joint HealthCalgaryCanada
| | - Jay Devine
- Department of Cell Biology and Anatomy, University of CalgaryCalgaryCanada
| | - Benedikt Hallgrímsson
- McCaig Institute for Bone and Joint HealthCalgaryCanada
- Department of Cell Biology and Anatomy, University of CalgaryCalgaryCanada
- Alberta Children's Hospital Research Institute for Child and Maternal Health, University of CalgaryCalgaryCanada
| | - Campbell Rolian
- McCaig Institute for Bone and Joint HealthCalgaryCanada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of CalgaryCalgaryCanada
| |
Collapse
|
12
|
Song D, Wu ZS, Xu Q, Wang K, Xu MT, Ha CZ, Zhang C, Wang DW. LRRC17 regulates the bone metabolism of human bone marrow mesenchymal stem cells from patients with idiopathic necrosis of femoral head through Wnt signaling pathways: A preliminary report. Exp Ther Med 2021; 22:666. [PMID: 33986831 PMCID: PMC8112125 DOI: 10.3892/etm.2021.10098] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/19/2021] [Indexed: 11/06/2022] Open
Abstract
Idiopathic necrosis of the femoral head (INFH) is a common disease with unknown cause. Its successful treatment relies on the repair of the necrotic bone. The application of autologous mesenchymal stem cells (MSCs) has shown great promise in saving the patients from undergoing total hip arthroplasty. Leucine-rich repeat-containing 17 (LRRC17) is less expressed in patients with femoral head necrosis and LRRC17 can inhibit bone degradation. However, it remains unknown whether LRRC17 plays a role in the pathogenesis of INFH. The present study aimed to investigate the potential role and mechanism of LRRC17 in the pathogenesis and treatment of INFH. It was found that despite the similar cell morphology and MSC surface marker expressions of human bone marrow MSCs (BMSCs) isolated from patients with INFH (INFH-hBMSC) and femoral neck fracture (FNF) (FNF-hBMSC), INFH-hBMSC had higher percentage of apoptosis (P<0.05), as well as lower osteogenic potential and higher adipogenic potential (both P<0.05). However, there was no difference in cell proliferation between FNF-hBMSC and INFH-hBMSC (P>0.05). It was also confirmed that the expression of LRRC17 was lower in the bone tissue and hBMSCs from patients with INFH compared with patients with FNF (P<0.05). Overexpression of LRRC17 promoted osteogenesis and inhibited the adipogenesis in hBMSCs, accompanied with the increase of Wnt3a and β-catenin expressions, and the decrease of Wnt5a and receptor activator of nuclear factor κ-B ligand (Rankl) expressions (all, P<0.05). Furthermore, knockout of LRRC17 in hBMSCs inhibited the expression levels of osteogenic and promoted adipogenic markers, while decreasing Wnt3a and β-catenin expressions, and increasing Wnt5a and Rankl expressions (all, P<0.05). The present preliminary study suggested that imbalanced bone metabolism may be involved in the pathogenesis of INFH. The modulation of the LRRC17 gene may delay or even restore the balance of osteogenic and adipogenic differentiation in autologous BMSCs derived from patients with INFH, providing a new target for the treatment of INFH.
Collapse
Affiliation(s)
- Da Song
- Department of Orthopedics, Liaocheng People's Hospital, Cheeloo College of Medicine, Shandong University, Liaocheng, Shandong 252000, P.R. China.,Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Zhen-Song Wu
- Department of Joint Surgery, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, P.R. China
| | - Qi Xu
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Kai Wang
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Ming-Tao Xu
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Cheng-Zhi Ha
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Chao Zhang
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Da-Wei Wang
- Department of Orthopedics, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
13
|
Chen J, Tang W, Lin C, Hong Y, Mao C, Lai Y, Liao C, Lin M, Chen W. Defining the critical period of hedgehog pathway inhibitor-induced cranial base dysplasia in mice. Dev Dyn 2021; 250:527-541. [PMID: 33165989 DOI: 10.1002/dvdy.270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The hedgehog signaling pathway is critical for developmental patterning of the limb, craniofacial and axial skeleton. Disruption of this pathway in mice leads to a series of structural malformations, but the exact role and critical period of the Hh pathway in the early development of the cranial base have been rarely described. RESULTS Embryos exposed to vismodegib from E7.5, E9.5, and E10.5 had a higher percentage of cranial base fenestra. The peak incidence of hypoplasia in sphenoid winglets and severe craniosynostosis in cranial base synchondroses was observed when vismodegib was administered between E9.5 and E10.5. Cranial base craniosynostosis results from accelerating terminal differentiation of chondrocytes and premature osteogenesis. CONCLUSIONS We define the critical periods for the induction of cranial base deformity by vismodegib administration at a meticulous temporal resolution. Our findings suggest that the Hh pathway may play a vital role in the early development of the cranial base. This research also establishes a novel and easy-to-establish mouse model of synostosis in the cranial base using a commercially available pathway-selective inhibitor.
Collapse
Affiliation(s)
- Jiangping Chen
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Wenbing Tang
- Department of Stomatology, Central Hospital of Guangdong Nongken, Zhanjiang, Guangdong, China
| | - Chengquan Lin
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Yuhang Hong
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Chuanqing Mao
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Yongzhen Lai
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Caiyu Liao
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Minkui Lin
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Institute of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
| | - Weihui Chen
- Fujian Key Laboratory of Oral Diseases & Stomatological Key lab of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, Fujian, China
- Department of Oral and Maxillofacial Surgery, Union Hospital, Fujian Medical University, Fuzhou, Fujian, China
- Fujian Biological Materials Engineering and Technology Center of Stomatology, Fuzhou, Fujian, China
| |
Collapse
|
14
|
Wu ZL, Xie QQ, Liu TC, Yang X, Zhang GZ, Zhang HH. Role of the Wnt pathway in the formation, development, and degeneration of intervertebral discs. Pathol Res Pract 2021; 220:153366. [PMID: 33647863 DOI: 10.1016/j.prp.2021.153366] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/31/2021] [Accepted: 02/02/2021] [Indexed: 12/13/2022]
Abstract
Intervertebral disc degeneration (IVDD) is an age-related degenerative disease that is the main cause of low back pain. It seriously affects the quality of life of patients and places a heavy economic burden on families and society. The Wnt pathway plays an important role in the growth, development, and degeneration of intervertebral discs (IVDs). In the embryonic stage, the Wnt pathway participates in the growth and development of IVD by promoting the transformation of progenitor cells into notochord cells and the extension of the notochord. However, the activation of the Wnt pathway after birth promotes IVD cell senescence, apoptosis, and degradation of the extracellular matrix and induces the production of inflammatory factors, thereby accelerating the IVDD process. This article reviews the relationship between the Wnt pathway and IVD, emphasizing its influence on IVD growth, development, and degeneration. Targeting this pathway may become an effective strategy for the treatment of IVDD.
Collapse
Affiliation(s)
- Zuo-Long Wu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Qi-Qi Xie
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Tai-Cong Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Xing Yang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Guang-Zhi Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China
| | - Hai-Hong Zhang
- The Second Clinical Medical College, Lanzhou University, Lanzhou, Gansu 730000, China; Department of Orthopaedics, Second Hospital of Lanzhou University, Lanzhou, Gansu 730000, China; Key Laboratory of Orthopaedics Disease of Gansu Province, Lanzhou, Gansu 730000, China.
| |
Collapse
|
15
|
The Skull's Girder: A Brief Review of the Cranial Base. J Dev Biol 2021; 9:jdb9010003. [PMID: 33498686 PMCID: PMC7838769 DOI: 10.3390/jdb9010003] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/14/2021] [Accepted: 01/19/2021] [Indexed: 12/11/2022] Open
Abstract
The cranial base is a multifunctional bony platform within the core of the cranium, spanning rostral to caudal ends. This structure provides support for the brain and skull vault above, serves as a link between the head and the vertebral column below, and seamlessly integrates with the facial skeleton at its rostral end. Unique from the majority of the cranial skeleton, the cranial base develops from a cartilage intermediate-the chondrocranium-through the process of endochondral ossification. Owing to the intimate association of the cranial base with nearly all aspects of the head, congenital birth defects impacting these structures often coincide with anomalies of the cranial base. Despite this critical importance, studies investigating the genetic control of cranial base development and associated disorders lags in comparison to other craniofacial structures. Here, we highlight and review developmental and genetic aspects of the cranial base, including its transition from cartilage to bone, dual embryological origins, and vignettes of transcription factors controlling its formation.
Collapse
|
16
|
Bartoletti G, Dong C, Umar M, He F. Pdgfra regulates multipotent cell differentiation towards chondrocytes via inhibiting Wnt9a/beta-catenin pathway during chondrocranial cartilage development. Dev Biol 2020; 466:36-46. [PMID: 32800757 DOI: 10.1016/j.ydbio.2020.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 12/31/2022]
Abstract
The mammalian skull is composed of the calvarial bones and cartilages. Malformation of craniofacial cartilage has been identified in multiple human syndromes. However, the mechanisms of their development remain largely unknown. In the present study, we identified Pdgfra as a novel player of chondrocranial cartilage development. Our data show that Pdgfra is required for normal chondrocranial cartilage development. Using tissue-specific genetic tools, we demonstrated that Pdgfra is essential for chondrocyte progenitors formation, but not in mature chondrocytes. Further analysis revealed that Pdgfra regulates chondrocytes progenitors development at two stages: in embryonic mesenchymal stem cells (eMSCs), Pdgfra directs their differentiation toward chondrocyte progenitors; in chondrocytes progenitors, Pdgfra activation promotes cell proliferation. We also found that excessive Pdgfra activity causes ectopic cartilage formation. Our data show that Pdgfra directs eMSCs differentiation via inhibiting Wnt9a transcription and its downstream signaling, and activating Wnt signaling rescues ectopic cartilage phenotype caused by excessive Pdgfra activity. In summary, our study dissected the role of Pdgfra signaling in chondrocranial cartilage formation, and illustrated the underlying mechanisms at multiple stages.
Collapse
Affiliation(s)
- Garrett Bartoletti
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Chunmin Dong
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Meenakshi Umar
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA
| | - Fenglei He
- Department of Cell and Molecular Biology, Tulane University, New Orleans, LA, 70118, USA.
| |
Collapse
|
17
|
Heubel BP, Bredesen CA, Schilling TF, Le Pabic P. Endochondral growth zone pattern and activity in the zebrafish pharyngeal skeleton. Dev Dyn 2020; 250:74-87. [PMID: 32852849 DOI: 10.1002/dvdy.241] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Endochondral ossification is a major bone forming mechanism in vertebrates, defects in which can result in skeletal dysplasia or craniofacial anomalies in humans. The zebrafish holds great potential to advance our understanding of endochondral growth zone development and genetics, yet several important aspects of its biology remain unexplored. Here we provide a comprehensive description of endochondral growth zones in the pharyngeal skeleton, including their developmental progression, cellular activity, and adult fates. RESULTS Postembryonic growth of the pharyngeal skeleton is supported by endochondral growth zones located either at skeletal epiphyses or synchondroses. Col2a1a and col10a1a in situ hybridization and anti-PCNA immunostaining identify resting-, hypertrophic- and proliferative zones, respectively, in pharyngeal synchondroses. Cellular hypertrophy and matrix deposition contribute little, if at all, to axial growth in most skeletal elements. Zebrafish endochondral growth zones develop during metamorphosis and arrest in adults. CONCLUSIONS Two endochondral growth zone configurations in the zebrafish pharyngeal skeleton produce either unidirectional (epiphyses) or bidirectional (synchondroses) growth. Cell proliferation drives endochondral growth and its modulation, in contrast to mammalian long bones in which bone length depends more on cell enlargement during hypertrophy and intramembranous ossification is the default mechanism of bone growth in zebrafish adults.
Collapse
Affiliation(s)
- Brian P Heubel
- Department of Biological Sciences, University of Delaware, Newark, Delaware, USA
| | - Carson A Bredesen
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| | - Thomas F Schilling
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, USA
| | - Pierre Le Pabic
- Department of Biology and Marine Biology, University of North Carolina Wilmington, Wilmington, North Carolina, USA
| |
Collapse
|
18
|
Funato N. New Insights Into Cranial Synchondrosis Development: A Mini Review. Front Cell Dev Biol 2020; 8:706. [PMID: 32850826 PMCID: PMC7432265 DOI: 10.3389/fcell.2020.00706] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Accepted: 07/13/2020] [Indexed: 11/24/2022] Open
Abstract
The synchondroses formed via endochondral ossification in the cranial base are an important growth center for the neurocranium. Abnormalities in the synchondroses affect cranial base elongation and the development of adjacent regions, including the craniofacial bones. In the central region of the cranial base, there are two synchondroses present—the intersphenoid synchondrosis and the spheno-occipital synchondrosis. These synchondroses consist of mirror image bipolar growth plates. The cross-talk of several signaling pathways, including the parathyroid hormone-like hormone (PTHLH)/parathyroid hormone-related protein (PTHrP), Indian hedgehog (Ihh), Wnt/β-catenin, and fibroblast growth factor (FGF) pathways, as well as regulation by cilium assembly and the transcription factors encoded by the RUNX2, SIX1, SIX2, SIX4, and TBX1 genes, play critical roles in synchondrosis development. Deletions or activation of these gene products in mice causes the abnormal ossification of cranial synchondrosis and skeletal elements. Gene disruption leads to both similar and markedly different abnormalities in the development of intersphenoid synchondrosis and spheno-occipital synchondrosis, as well as in the phenotypes of synchondroses and skeletal bones. This paper reviews the development of cranial synchondroses, along with its regulation by the signaling pathways and transcription factors, highlighting the differences between intersphenoid synchondrosis and spheno-occipital synchondrosis.
Collapse
Affiliation(s)
- Noriko Funato
- Department of Signal Gene Regulation, Tokyo Medical and Dental University, Tokyo, Japan.,Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
19
|
Funato N, Srivastava D, Shibata S, Yanagisawa H. TBX1 Regulates Chondrocyte Maturation in the Spheno-occipital Synchondrosis. J Dent Res 2020; 99:1182-1191. [PMID: 32442036 DOI: 10.1177/0022034520925080] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The synchondrosis in the cranial base is an important growth center for the craniofacial region. Abnormalities in the synchondroses affect the development of adjacent regions, including the craniofacial skeleton. Here, we report that the transcription factor TBX1, the candidate gene for DiGeorge syndrome, is expressed in mesoderm-derived chondrocytes and plays an essential and specific role in spheno-occipital synchondrosis development by inhibiting the expression of genes involved in chondrocyte hypertrophy and osteogenesis. In Tbx1-deficient mice, the spheno-occipital synchondrosis was completely mineralized at birth. TBX1 interacts with RUNX2, a master molecule of osteoblastogenesis and a regulator of chondrocyte maturation, and suppresses its transcriptional activity. Indeed, deleting Tbx1 triggers accelerated mineralization due to accelerated chondrocyte differentiation, which is associated with ectopic expression of downstream targets of RUNX2 in the spheno-occipital synchondrosis. These findings reveal that TBX1 acts as a regulator of chondrocyte maturation and osteogenesis during the spheno-occipital synchondrosis development. Thus, the tight regulation of endochondral ossification by TBX1 is crucial for the normal progression of chondrocyte differentiation in the spheno-occipital synchondrosis.
Collapse
Affiliation(s)
- N Funato
- Department of Signal Gene Regulation, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| | - D Srivastava
- Gladstone Institute of Cardiovascular Disease and Department of Pediatrics, University of California, San Francisco, San Francisco, CA, USA
| | - S Shibata
- Department of Maxillofacial Anatomy, Tokyo Medical and Dental University, Tokyo, Japan
| | - H Yanagisawa
- Life Science Center for Survival Dynamics, Tsukuba Advanced Research Alliance, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
20
|
Leitch VD, Bassett JHD, Williams GR. Role of thyroid hormones in craniofacial development. Nat Rev Endocrinol 2020; 16:147-164. [PMID: 31974498 DOI: 10.1038/s41574-019-0304-5] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2019] [Indexed: 02/07/2023]
Abstract
The development of the craniofacial skeleton relies on complex temporospatial organization of diverse cell types by key signalling molecules. Even minor disruptions to these processes can result in deleterious consequences for the structure and function of the skull. Thyroid hormone deficiency causes delayed craniofacial and tooth development, dysplastic facial features and delayed development of the ossicles in the middle ear. Thyroid hormone excess, by contrast, accelerates development of the skull and, in severe cases, might lead to craniosynostosis with neurological sequelae and facial hypoplasia. The pathogenesis of these important abnormalities remains poorly understood and underinvestigated. The orchestration of craniofacial development and regulation of suture and synchondrosis growth is dependent on several critical signalling pathways. The underlying mechanisms by which these key pathways regulate craniofacial growth and maturation are largely unclear, but studies of single-gene disorders resulting in craniofacial malformations have identified a number of critical signalling molecules and receptors. The craniofacial consequences resulting from gain-of-function and loss-of-function mutations affecting insulin-like growth factor 1, fibroblast growth factor receptor and WNT signalling are similar to the effects of altered thyroid status and mutations affecting thyroid hormone action, suggesting that these critical pathways interact in the regulation of craniofacial development.
Collapse
Affiliation(s)
- Victoria D Leitch
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Royal Melbourne Institute of Technology (RMIT) Centre for Additive Manufacturing, RMIT University, Melbourne, VIC, Australia
| | - J H Duncan Bassett
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Graham R Williams
- Molecular Endocrinology Laboratory, Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
21
|
Itoyama T, Fukui M, Kawaguchi M, Kaneko S, Sugahara F, Murakami Y. FGF- and SHH-based molecular signals regulate barbel and craniofacial development in catfish. ZOOLOGICAL LETTERS 2019; 5:19. [PMID: 31223485 PMCID: PMC6570838 DOI: 10.1186/s40851-019-0135-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 05/19/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Catfish (Siluriformes) are characterized by unique morphologies, including enlarged jaws with movable barbels and taste buds covering the entire body surface. Evolution of these characteristics was a crucial step in their adaptive radiation to freshwater environments. However, the developmental processes of the catfish craniofacial region and taste buds remain to be elucidated; moreover, little is known about the molecular mechanisms underlying the morphogenesis of these structures. RESULTS In Amur catfish (Silurus asotus), three pairs of barbel primordia are formed by 2 days post-fertilization (dpf). Innervation of the peripheral nerves and formation of muscle precursors are also established during early development. Taste buds from the oral region to the body trunk are formed by 4 dpf. We then isolated catfish cognates Shh (SaShh) and Fgf8 (SaFgf8), which are expressed in maxillary barbel primordium at 1-2 dpf. Further, SHH signal inhibition induces reduction of mandibular barbels with abnormal morphology of skeletal elements, whereas it causes no apparent abnormality in the trigeminal and facial nerve morphology. We also found that mandibular barbel lengths and number of taste buds are reduced by FGF inhibition, as seen in SHH signal inhibition. However, unlike with SHH inhibition, the abnormal morphology of the trigeminal and facial nerves was observed in FGF signal-inhibited embryos. CONCLUSION The developmental processes of Amur catfish are consistent with those reported for other catfish species. Thus, developmental aspects of craniofacial structures and taste buds may be conserved in Siluriformes. Our findings also suggest that SHH signaling plays a crucial role in the formation of barbels and taste buds, without affecting nerve projection, while FGF signaling is required for the development of barbels, taste buds, and branchial nerves. Thus, SHH and FGF signaling plays key roles in the ontogenesis and evolution of some catfish-specific characteristics.
Collapse
Affiliation(s)
- Tatsuya Itoyama
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577 Japan
| | - Makiko Fukui
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577 Japan
| | - Masahumi Kawaguchi
- Department of Anatomy and Neuroscience, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama, 930-0194 Japan
| | - Saki Kaneko
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577 Japan
| | - Fumiaki Sugahara
- Division of Biology, Hyogo College of Medicine, Nishinomiya, 663-8501 Japan
| | - Yasunori Murakami
- Graduate School of Science and Engineering, Ehime University, 2-5 Bunkyo-cho, Matsuyama, 790-8577 Japan
| |
Collapse
|
22
|
Mansour TA, Lucot K, Konopelski SE, Dickinson PJ, Sturges BK, Vernau KL, Choi S, Stern JA, Thomasy SM, Döring S, Verstraete FJM, Johnson EG, York D, Rebhun RB, Ho HYH, Brown CT, Bannasch DL. Whole genome variant association across 100 dogs identifies a frame shift mutation in DISHEVELLED 2 which contributes to Robinow-like syndrome in Bulldogs and related screw tail dog breeds. PLoS Genet 2018; 14:e1007850. [PMID: 30521570 PMCID: PMC6303079 DOI: 10.1371/journal.pgen.1007850] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 12/21/2018] [Accepted: 11/24/2018] [Indexed: 12/30/2022] Open
Abstract
Domestic dog breeds exhibit remarkable morphological variations that result from centuries of artificial selection and breeding. Identifying the genetic changes that contribute to these variations could provide critical insights into the molecular basis of tissue and organismal morphogenesis. Bulldogs, French Bulldogs and Boston Terriers share many morphological and disease-predisposition traits, including brachycephalic skull morphology, widely set eyes and short stature. Unlike other brachycephalic dogs, these breeds also exhibit vertebral malformations that result in a truncated, kinked tail (screw tail). Whole genome sequencing of 100 dogs from 21 breeds identified 12.4 million bi-allelic variants that met inclusion criteria. Whole Genome Association of these variants with the breed defining phenotype of screw tail was performed using 10 cases and 84 controls and identified a frameshift mutation in the WNT pathway gene DISHEVELLED 2 (DVL2) (Chr5: 32195043_32195044del, p = 4.37 X 10-37) as the most strongly associated variant in the canine genome. This DVL2 variant was fixed in Bulldogs and French Bulldogs and had a high allele frequency (0.94) in Boston Terriers. The DVL2 variant segregated with thoracic and caudal vertebral column malformations in a recessive manner with incomplete and variable penetrance for thoracic vertebral malformations between different breeds. Importantly, analogous frameshift mutations in the human DVL1 and DVL3 genes cause Robinow syndrome, a congenital disorder characterized by similar craniofacial, limb and vertebral malformations. Analysis of the canine DVL2 variant protein showed that its ability to undergo WNT-induced phosphorylation is reduced, suggesting that altered WNT signaling may contribute to the Robinow-like syndrome in the screwtail breeds.
Collapse
Affiliation(s)
- Tamer A. Mansour
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
- Department of Clinical Pathology, School of Medicine, University of Mansoura, Mansoura Egypt
| | - Katherine Lucot
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
- Integrative Genetics and Genomics Graduate Group, University of California Davis, Davis, CA, United States of America
| | - Sara E. Konopelski
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA, United States of America
| | - Peter J. Dickinson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Beverly K. Sturges
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Karen L. Vernau
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Shannon Choi
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA, United States of America
| | - Joshua A. Stern
- Department of Medicine and Epidemiology, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Sara M. Thomasy
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Sophie Döring
- William R. Pritchard Veterinary Medical Teaching Hospital, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Frank J. M. Verstraete
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Eric G. Johnson
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Daniel York
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Robert B. Rebhun
- Department of Surgical and Radiological Sciences, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
| | - Hsin-Yi Henry Ho
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA, United States of America
| | - C. Titus Brown
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
- Genome Center, University of California Davis, Davis, CA, United States of America
| | - Danika L. Bannasch
- Department of Population Health and Reproduction, School of Veterinary Medicine, University of California Davis, Davis, CA, United States of America
- Genome Center, University of California Davis, Davis, CA, United States of America
| |
Collapse
|
23
|
Abstract
The cranial base is a central and integral component of the cranioskeleton, yet little is known about its growth. Despite the dissimilarities between human and murine cranioskeletal form, mouse models are proving instrumental in studying craniofacial growth. The objectives of this review are to summarize recent findings from numerous mouse models that display growth defects in one or more cranial base synchondroses, with accompanying changes in chondrocyte cellular zones. Many of these models also display altered growth of the cranial vault and/or the facial region. FGFR, PTHrP, Ihh, BMP and Wnt/β-catenin, as well as components of primary cilia, are the major genes and signalling pathways identified in cranial base synchondroses. Together, these models are helping to uncover specific genetic influences and signalling pathways operational at the cranial base synchondroses. Many of these genes are in common with those of importance in the cranial vault and the facial skeleton, emphasizing the molecular integration of growth between the cranial base and other cranial regions. Selected models are also being utilized in testing therapeutic agents to correct defective craniofacial and cranial base growth.
Collapse
Affiliation(s)
- S R Vora
- Oral Health Sciences, Orthodontics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Jing Y, Jing J, Wang K, Chan K, Harris SE, Hinton RJ, Feng JQ. Vital Roles of β-catenin in Trans-differentiation of Chondrocytes to Bone Cells. Int J Biol Sci 2018; 14:1-9. [PMID: 29483820 PMCID: PMC5821044 DOI: 10.7150/ijbs.23165] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 10/14/2017] [Indexed: 02/05/2023] Open
Abstract
A recent breakthrough showing that direct trans-differentiation of chondrocytes into bone cells commonly occurs during endochondral bone formation in the growth plate, articular cartilage, and mandibular condylar cartilage suggests that chondrogenesis and osteogenesis are likely one continuous biological process instead of two separate processes. Yet, gene regulation of this cell transformation is largely unclear. Here, we employed cartilage-specific β-catenin loss-of-function (β-catenin fx/fx ) and gain-of-function (β-catenin fx(exon3)/ fx(exon3) ) models in the R26RTomato background (for better tracing the cell fate of chondrocytes) to study the role of β-catenin in cell trans-differentiation. Using histological, immunohistochemical, and radiological methods combined with cell lineage tracing techniques, we showed that deletion of β-catenin by either Acan-CreERT2 or Col10a1-Cre resulted in greatly reduced cell trans-differentiation with a significant decrease in subchondral bone volume during mandibular condylar growth. Molecular studies demonstrated severe defects in cell proliferation and differentiation in both chondrocytes and bone cells. The gain of function studies (constitutive activation of β-catenin with Acan-CreERT2 at ages of postnatal day 7, 4-weeks and 6-months) led to more bone cell trans-differentiation of chondrocytes in the mandibular condyle due to increased proliferation and accelerated chondrocyte differentiation with incipient osteogenic changes within the cartilage matrix, resulting in an increased volume of poorly-formed immature subchondral bone. These results support the notion that chondrogenesis and osteogenesis are one continuous process, in which β-catenin signaling plays an essential role in the cell trans-differentiation of chondrocytes into bone cells during mandibular condylar development and growth.
Collapse
Affiliation(s)
- Yan Jing
- Department of Orthodontics, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Junjun Jing
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China, 610041
| | - Ke Wang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Kevin Chan
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Stephen E Harris
- Department of Periodontics, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229, USA
| | - Robert J Hinton
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| |
Collapse
|
25
|
Kaucka M, Zikmund T, Tesarova M, Gyllborg D, Hellander A, Jaros J, Kaiser J, Petersen J, Szarowska B, Newton PT, Dyachuk V, Li L, Qian H, Johansson AS, Mishina Y, Currie JD, Tanaka EM, Erickson A, Dudley A, Brismar H, Southam P, Coen E, Chen M, Weinstein LS, Hampl A, Arenas E, Chagin AS, Fried K, Adameyko I. Oriented clonal cell dynamics enables accurate growth and shaping of vertebrate cartilage. eLife 2017; 6. [PMID: 28414273 PMCID: PMC5417851 DOI: 10.7554/elife.25902] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/16/2017] [Indexed: 11/30/2022] Open
Abstract
Cartilaginous structures are at the core of embryo growth and shaping before the bone forms. Here we report a novel principle of vertebrate cartilage growth that is based on introducing transversally-oriented clones into pre-existing cartilage. This mechanism of growth uncouples the lateral expansion of curved cartilaginous sheets from the control of cartilage thickness, a process which might be the evolutionary mechanism underlying adaptations of facial shape. In rod-shaped cartilage structures (Meckel, ribs and skeletal elements in developing limbs), the transverse integration of clonal columns determines the well-defined diameter and resulting rod-like morphology. We were able to alter cartilage shape by experimentally manipulating clonal geometries. Using in silico modeling, we discovered that anisotropic proliferation might explain cartilage bending and groove formation at the macro-scale. DOI:http://dx.doi.org/10.7554/eLife.25902.001
Collapse
Affiliation(s)
- Marketa Kaucka
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Tomas Zikmund
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Marketa Tesarova
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Daniel Gyllborg
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Andreas Hellander
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| | - Josef Jaros
- Department of Histology and Embryology, Medical Faculty, Masaryk University, Brno, Czech Republic
| | - Jozef Kaiser
- Central European Institute of Technology, Brno University of Technology, Brno, Czech Republic
| | - Julian Petersen
- Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Bara Szarowska
- Center for Brain Research, Medical University Vienna, Vienna, Austria
| | - Phillip T Newton
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | | - Lei Li
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Hong Qian
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Yuji Mishina
- Department of Biologic and Materials Sciences, University of Michigan School of Dentistry, Ann Arbor, United States
| | - Joshua D Currie
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Elly M Tanaka
- Center for Regenerative Therapies, Technische Universität Dresden, Dresden, Germany
| | - Alek Erickson
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, United States
| | - Andrew Dudley
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, United States
| | - Hjalmar Brismar
- Science for Life Laboratory, Royal Institute of Technology, Solna, Sweden
| | | | | | - Min Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Lee S Weinstein
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Ales Hampl
- Department of Histology and Embryology, Medical Faculty, Masaryk University, Brno, Czech Republic
| | - Ernest Arenas
- Unit of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Andrei S Chagin
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Kaj Fried
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.,Center for Brain Research, Medical University Vienna, Vienna, Austria
| |
Collapse
|
26
|
Wei X, Hu M, Mishina Y, Liu F. Developmental Regulation of the Growth Plate and Cranial Synchondrosis. J Dent Res 2016; 95:1221-9. [PMID: 27250655 DOI: 10.1177/0022034516651823] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Long bones and the cranial base are both formed through endochondral ossification. Elongation of long bones is primarily through the growth plate, which is a cartilaginous structure at the end of long bones made up of chondrocytes. Growth plate chondrocytes are organized in columns along the longitudinal axis of bone growth. The cranial base is the growth center of the neurocranium. Synchondroses, consisting of mirror-image growth plates, are critical for cranial base elongation and development. Over the last decade, considerable progress has been made in determining the roles of the parathyroid hormone-related protein, Indian hedgehog, fibroblast growth factor, bone morphogenetic protein, and Wnt signaling pathways in various aspects of skeletal development. Furthermore, recent evidence indicates the important role of the primary cilia signaling pathway in bone elongation. Here, we review the development of the growth plate and cranial synchondrosis and the regulation by the above-mentioned signaling pathways, highlighting the similarities and differences between these 2 structures.
Collapse
Affiliation(s)
- X Wei
- Department of Biologic and Materials Sciences and Division of Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA Department of Orthodontics, Jilin University School and Hospital of Stomatology, Changchun, Jilin, China
| | - M Hu
- Department of Orthodontics, Jilin University School and Hospital of Stomatology, Changchun, Jilin, China
| | - Y Mishina
- Department of Biologic and Materials Sciences and Division of Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| | - F Liu
- Department of Biologic and Materials Sciences and Division of Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, MI, USA
| |
Collapse
|
27
|
Staines KA, Madi K, Mirczuk SM, Parker S, Burleigh A, Poulet B, Hopkinson M, Bodey AJ, Fowkes RC, Farquharson C, Lee PD, Pitsillides AA. Endochondral Growth Defect and Deployment of Transient Chondrocyte Behaviors Underlie Osteoarthritis Onset in a Natural Murine Model. Arthritis Rheumatol 2016; 68:880-91. [PMID: 26605758 PMCID: PMC4832379 DOI: 10.1002/art.39508] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 11/05/2015] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To explore whether aberrant transient chondrocyte behaviors occur in the joints of STR/Ort mice (which spontaneously develop osteoarthritis [OA]) and whether they are attributable to an endochondral growth defect. METHODS Knee joints from STR/Ort mice with advanced OA and age-matched CBA (control) mice were examined by Affymetrix microarray profiling, multiplex polymerase chain reaction (PCR) analysis, and immunohistochemical labeling of endochondral markers, including sclerostin and MEPE. The endochondral phenotype of STR/Ort mice was analyzed by histologic examination, micro-computed tomography, and ex vivo organ culture. A novel protocol for quantifying bony bridges across the murine epiphysis (growth plate fusion) using synchrotron x-ray computed microtomography was developed and applied. RESULTS Meta-analysis of transcription profiles showed significant elevation in functions linked with endochondral ossification in STR/Ort mice (compared to CBA mice; P < 0.05). Consistent with this, immunolabeling revealed increased matrix metalloproteinase 13 (MMP-13) and type X collagen expression in STR/Ort mouse joints, and multiplex quantitative reverse transcriptase-PCR showed differential expression of known mineralization regulators, suggesting an inherent chondrocyte defect. Support for the notion of an endochondral defect included accelerated growth, increased zone of growth plate proliferative chondrocytes (P < 0.05), and widespread type X collagen/MMP-13 labeling beyond the expected hypertrophic zone distribution. OA development involved concomitant focal suppression of sclerostin/MEPE in STR/Ort mice. Our novel synchrotron radiation microtomography method showed increased numbers (P < 0.001) and mean areal growth plate bridge densities (P < 0.01) in young and aged STR/Ort mice compared to age-matched CBA mice. CONCLUSION Taken together, our data support the notion of an inherent endochondral defect that is linked to growth dynamics and subject to regulation by the MEPE/sclerostin axis and may represent an underlying mechanism of pathologic ossification in OA.
Collapse
Affiliation(s)
- K. A. Staines
- Royal Veterinary College, University of London, London, UK, and Roslin Institute and Royal (Dick) School of Veterinary StudiesUniversity of Edinburgh, Easter Bush, UK
| | - K. Madi
- Manchester X‐Ray Imaging Facility, University of Manchester, Manchester, UK
| | - S. M. Mirczuk
- Royal Veterinary College, University of London, London, UK;
| | - S. Parker
- Royal Veterinary College, University of London, London, UK;
| | - A. Burleigh
- Royal Veterinary College, University of London, London, UK;
| | - B. Poulet
- University College London Medical School, London, UK
| | - M. Hopkinson
- Royal Veterinary College, University of London, London, UK;
| | - A. J. Bodey
- Diamond Light Source, Harwell Science and Innovation Campus, Didcot, UK
| | - R. C. Fowkes
- Royal Veterinary College, University of London, London, UK;
| | - C. Farquharson
- Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush, UK
| | - P. D. Lee
- Manchester X‐Ray Imaging Facility, University of Manchester, Manchester, UK
| | | |
Collapse
|
28
|
XU JIAJUN, SUN TAO, HU XUEBING. microRNA-513c suppresses the proliferation of human glioblastoma cells by repressing low-density lipoprotein receptor-related protein 6. Mol Med Rep 2015; 12:4403-4409. [DOI: 10.3892/mmr.2015.3913] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 04/14/2015] [Indexed: 11/06/2022] Open
|
29
|
MiR-939 promotes the proliferation of human ovarian cancer cells by repressing APC2 expression. Biomed Pharmacother 2015; 71:64-9. [DOI: 10.1016/j.biopha.2015.02.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2015] [Accepted: 02/15/2015] [Indexed: 12/30/2022] Open
|
30
|
Xu L, Liu Y, Hou Y, Wang K, Wong Y, Lin S, Li G. U0126 promotes osteogenesis of rat bone-marrow-derived mesenchymal stem cells by activating BMP/Smad signaling pathway. Cell Tissue Res 2015; 359:537-545. [PMID: 25363751 DOI: 10.1007/s00441-014-2025-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 10/08/2014] [Indexed: 12/26/2022]
Abstract
U0126 has been reported as a specific inhibitor of the ERK1/2 signaling pathway, which plays a vital role during the osteogenic differentiation of mesenchymal stem cells (MSCs). We report the positive effect of U0126 on the osteogenesis of rat MSCs. We find that U0126 promotes the osteogenic differentiation of rat MSCs as demonstrated by the quantitative real-time polymerase chain reaction for osteogenic markers, alkaline phosphatase activity and calcium nodule formation. Our data indicate that U0126 enhances the BMP/Smad signaling pathway in rat MSCs, while inhibiting the ERK1/2 signaling pathway. Furthermore, Western blot results demonstrate that U0126 increases Smad1/5/8 phosphorylation synergistically with β-glycerophosphate. In addition, U0126 significantly increases the expression of BMP2 during the process of osteogenesis in rat MSCs and the level of phosphorylated Smad1/5/8 is significantly reduced by BMP2 antibody, suggesting that U0126 also promotes the expression of BMP2 to enhance Smad proteins phosphorylation. Thus, we demonstrate a novel function for U0126 in promoting osteogenic differentiation of rat MSCs by the activation of the BMP/Smad signaling pathway.
Collapse
Affiliation(s)
- Liangliang Xu
- Department of Orthopaedics & Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yang Liu
- Department of Orthopaedics & Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yonghui Hou
- School of Biomedical Sciences, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Kuixing Wang
- Department of Orthopaedics & Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Yinmei Wong
- Department of Orthopaedics & Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Sien Lin
- Department of Orthopaedics & Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China
| | - Gang Li
- Department of Orthopaedics & Traumatology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, People's Republic of China.
- MOE Key Laboratory of Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, People's Republic of China.
- The CUHK-ACC Space Medicine Centre on Health Maintenance of Musculoskeletal System, Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, People's Republic of China.
- Li Ka Shing Institute of Health Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Room 904, 9/F, Shatin, Hong Kong, SAR, People's Republic of China.
| |
Collapse
|
31
|
Tanaka M, Watanabe M, Yokomi I, Matsumoto N, Sudo K, Satoh H, Igarashi T, Seki A, Amano H, Ohura K, Ryu K, Shibata S, Nagayama M, Tanuma JI. Establishment of a novel dwarf rat strain: cartilage calcification insufficient (CCI) rats. Exp Anim 2014; 64:121-8. [PMID: 25736479 PMCID: PMC4427726 DOI: 10.1538/expanim.14-0072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Rats with dwarfism accompanied by skeletal abnormalities, such as shortness of the limbs, tail, and body (dwarf rats), emerged in a Jcl-derived Sprague-Dawley rat colony maintained at the Institute for Animal Experimentation, St. Marianna University Graduate School of Medicine. Since the dwarfism was assumed to be due to a genetic mutation based on its frequency, we bred the dwarf rats and investigated their characteristics in order to identify the causative factors of their phenotypes and whether they could be used as a human disease model. One male and female that produced dwarf progeny were selected, and reproduction was initiated by mating the pair. The incidence of dwarfism was 25.8% among the resultant litter, and dwarfism occurred in both genders, suggesting that it was inherited in an autosomal recessive manner. At 12 weeks of age, the body weights of the male and female dwarf rats were 40% and 57% of those of the normal rats, respectively. In soft X-ray radiographic and histological examinations, shortening and hypoplasia of the long bones, such as the tibia and femur, were observed, which were suggestive of endochondral ossification abnormalities. An immunohistochemical examination detected an aggrecan synthesis disorder, which might have led to delayed calcification and increased growth plate thickening in the dwarf rats. We hypothesized that the principal characteristics of the dwarf rats were systemically induced by insufficient cartilage calcification in their long bones; thus, we named them cartilage calcification insufficient (CCI) rats.
Collapse
Affiliation(s)
- Masami Tanaka
- Department of Food and Nutrition, Junior College Division, The University of Aizu, Aizu-Wakamatsu, Fukushima 965-8570, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Yakkioui Y, van Overbeeke JJ, Santegoeds R, van Engeland M, Temel Y. Chordoma: the entity. Biochim Biophys Acta Rev Cancer 2014; 1846:655-69. [PMID: 25193090 DOI: 10.1016/j.bbcan.2014.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 01/08/2023]
Abstract
Chordomas are malignant tumors of the axial skeleton, characterized by their locally invasive and slow but aggressive growth. These neoplasms are presumed to be derived from notochordal remnants with a molecular alteration preceding their malignant transformation. As these tumors are most frequently observed on the skull base and sacrum, patients suffering from a chordoma present with debilitating neurological disease, and have an overall 5-year survival rate of 65%. Surgical resection with adjuvant radiotherapy is the first-choice treatment modality in these patients, since chordomas are resistant to conventional chemotherapy. Even so, management of chordomas can be challenging, as chordoma patients often present with recurrent disease. Recent advances in the understanding of the molecular events that contribute to the development of chordomas are promising; the most novel finding being the identification of brachyury in the disease process. Here we present an overview of the current paradigms and summarize relevant research findings.
Collapse
Affiliation(s)
- Youssef Yakkioui
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - Jacobus J van Overbeeke
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Remco Santegoeds
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Manon van Engeland
- Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Yasin Temel
- Department of Neurosurgery, Maastricht University Medical Center, Maastricht, The Netherlands; Department of Neuroscience, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
33
|
Romereim SM, Conoan NH, Chen B, Dudley AT. A dynamic cell adhesion surface regulates tissue architecture in growth plate cartilage. Development 2014; 141:2085-95. [PMID: 24764078 DOI: 10.1242/dev.105452] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The architecture and morphogenetic properties of tissues are founded in the tissue-specific regulation of cell behaviors. In endochondral bones, the growth plate cartilage promotes bone elongation via regulated chondrocyte maturation within an ordered, three-dimensional cell array. A key event in the process that generates this cell array is the transformation of disordered resting chondrocytes into clonal columns of discoid proliferative cells aligned with the primary growth vector. Previous analysis showed that column-forming chondrocytes display planar cell divisions, and the resulting daughter cells rearrange by ∼90° to align with the lengthening column. However, these previous studies provided limited information about the mechanisms underlying this dynamic process. Here we present new mechanistic insights generated by application of a novel time-lapse confocal microscopy method along with immunofluorescence and electron microscopy. We show that, during cell division, daughter chondrocytes establish a cell-cell adhesion surface enriched in cadherins and β-catenin. Rearrangement into columns occurs concomitant with expansion of this adhesion surface in a process more similar to cell spreading than to migration. Column formation requires cell-cell adhesion, as reducing cadherin binding via chelation of extracellular calcium inhibits chondrocyte rearrangement. Importantly, physical indicators of cell polarity, such as cell body alignment, are not prerequisites for oriented cell behavior. Our results support a model in which regulation of adhesive surface dynamics and cortical tension by extrinsic signaling modifies the thermodynamic landscape to promote organization of daughter cells in the context of the three-dimensional growth plate tissue.
Collapse
Affiliation(s)
- Sarah M Romereim
- Department of Genetics, Cell Biology, and Anatomy and the Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, 985965 Nebraska Medical Center, Omaha, NE 68198-5965, USA
| | | | | | | |
Collapse
|
34
|
Kessels MY, Huitema LFA, Boeren S, Kranenbarg S, Schulte-Merker S, van Leeuwen JL, de Vries SC. Proteomics analysis of the zebrafish skeletal extracellular matrix. PLoS One 2014; 9:e90568. [PMID: 24608635 PMCID: PMC3946537 DOI: 10.1371/journal.pone.0090568] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/03/2014] [Indexed: 11/30/2022] Open
Abstract
The extracellular matrix of the immature and mature skeleton is key to the development and function of the skeletal system. Notwithstanding its importance, it has been technically challenging to obtain a comprehensive picture of the changes in skeletal composition throughout the development of bone and cartilage. In this study, we analyzed the extracellular protein composition of the zebrafish skeleton using a mass spectrometry-based approach, resulting in the identification of 262 extracellular proteins, including most of the bone and cartilage specific proteins previously reported in mammalian species. By comparing these extracellular proteins at larval, juvenile, and adult developmental stages, 123 proteins were found that differed significantly in abundance during development. Proteins with a reported function in bone formation increased in abundance during zebrafish development, while analysis of the cartilage matrix revealed major compositional changes during development. The protein list includes ligands and inhibitors of various signaling pathways implicated in skeletogenesis such as the Int/Wingless as well as the insulin-like growth factor signaling pathways. This first proteomic analysis of zebrafish skeletal development reveals that the zebrafish skeleton is comparable with the skeleton of other vertebrate species including mammals. In addition, our study reveals 6 novel proteins that have never been related to vertebrate skeletogenesis and shows a surprisingly large number of differences in the cartilage and bone proteome between the head, axis and caudal fin regions. Our study provides the first systematic assessment of bone and cartilage protein composition in an entire vertebrate at different stages of development.
Collapse
Affiliation(s)
- Maurijn Y. Kessels
- Laboratory of Biochemistry, Wageningen University, Wageningen, the Netherlands
- Experimental Zoology Group, Wageningen University, Wageningen, the Netherlands
| | - Leonie F. A. Huitema
- Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University, Wageningen, the Netherlands
| | - Sander Kranenbarg
- Experimental Zoology Group, Wageningen University, Wageningen, the Netherlands
| | - Stefan Schulte-Merker
- Experimental Zoology Group, Wageningen University, Wageningen, the Netherlands
- Hubrecht Institute-KNAW and University Medical Centre Utrecht, Utrecht, the Netherlands
| | | | - Sacco C. de Vries
- Laboratory of Biochemistry, Wageningen University, Wageningen, the Netherlands
- * E-mail:
| |
Collapse
|
35
|
Feng W, Choi I, Clouthier DE, Niswander L, Williams T. The Ptch1(DL) mouse: a new model to study lambdoid craniosynostosis and basal cell nevus syndrome-associated skeletal defects. Genesis 2013; 51:677-89. [PMID: 23897749 DOI: 10.1002/dvg.22416] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Accepted: 07/16/2013] [Indexed: 12/19/2022]
Abstract
Mouse models provide valuable opportunities for probing the underlying pathology of human birth defects. By using an N-ethyl-N-nitrosourea-based screen for recessive mutations affecting craniofacial anatomy, we isolated a mouse strain, Dogface-like (DL), with abnormal skull and snout morphology. Examination of the skull indicated that these mice developed craniosynostosis of the lambdoid suture. Further analysis revealed skeletal defects related to the pathology of basal cell nevus syndrome (BCNS) including defects in development of the limbs, scapula, ribcage, secondary palate, cranial base, and cranial vault. In humans, BCNS is often associated with mutations in the Hedgehog receptor PTCH1 and genetic mapping in DL identified a point mutation at a splice donor site in Ptch1. By using genetic complementation analysis we determined that DL is a hypomorphic allele of Ptch1, leading to increased Hedgehog signaling. Two aberrant transcripts are generated by the mutated Ptch1(DL) gene, which would be predicted to reduce significantly the levels of functional Patched1 protein. This new Ptch1 allele broadens the mouse genetic reagents available to study the Hedgehog pathway and provides a valuable means to study the underlying skeletal abnormalities in BCNS. In addition, these results strengthen the connection between elevated Hedgehog signaling and craniosynostosis.
Collapse
Affiliation(s)
- Weiguo Feng
- Department of Craniofacial Biology and Cell and Developmental Biology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | | | | | | |
Collapse
|
36
|
Pan A, Chang L, Nguyen A, James AW. A review of hedgehog signaling in cranial bone development. Front Physiol 2013; 4:61. [PMID: 23565096 PMCID: PMC3613593 DOI: 10.3389/fphys.2013.00061] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 03/13/2013] [Indexed: 12/20/2022] Open
Abstract
During craniofacial development, the Hedgehog (HH) signaling pathway is essential for mesodermal tissue patterning and differentiation. The HH family consists of three protein ligands: Sonic Hedgehog (SHH), Indian Hedgehog (IHH), and Desert Hedgehog (DHH), of which two are expressed in the craniofacial complex (IHH and SHH). Dysregulations in HH signaling are well documented to result in a wide range of craniofacial abnormalities, including holoprosencephaly (HPE), hypotelorism, and cleft lip/palate. Furthermore, mutations in HH effectors, co-receptors, and ciliary proteins result in skeletal and craniofacial deformities. Cranial suture morphogenesis is a delicate developmental process that requires control of cell commitment, proliferation and differentiation. This review focuses on both what is known and what remains unknown regarding HH signaling in cranial suture morphogenesis and intramembranous ossification. As demonstrated from murine studies, expression of both SHH and IHH is critical to the formation and fusion of the cranial sutures and calvarial ossification. SHH expression has been observed in the cranial suture mesenchyme and its precise function is not fully defined, although some postulate SHH to delay cranial suture fusion. IHH expression is mainly found on the osteogenic fronts of the calvarial bones, and functions to induce cell proliferation and differentiation. Unfortunately, neonatal lethality of IHH deficient mice precludes a detailed examination of their postnatal calvarial phenotype. In summary, a number of basic questions are yet to be answered regarding domains of expression, developmental role, and functional overlap of HH morphogens in the calvaria. Nevertheless, SHH and IHH ligands are integral to cranial suture development and regulation of calvarial ossification. When HH signaling goes awry, the resultant suite of morphologic abnormalities highlights the important roles of HH signaling in cranial development.
Collapse
Affiliation(s)
- Angel Pan
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | | |
Collapse
|
37
|
Xu L, Meng F, Ni M, Lee Y, Li G. N-cadherin regulates osteogenesis and migration of bone marrow-derived mesenchymal stem cells. Mol Biol Rep 2012. [PMID: 23187741 DOI: 10.1007/s11033-012-2334-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
N-cadherin, a calcium-dependent cellular adhesive protein, plays important roles during embryonic development and bone formation. The potential of mesenchymal stem cells (MSCs) in osteoblast differentiation and homing to the sites of injury make it a promising cell resource for tissue engineering. However, the role of N-cadherin in MSCs osteoblast differentiation and migration remains still obscure. In the present study, our results showed that prolonged N-cadherin overexpression inhibited osteogenic differentiation of MSCs through negatively regulating β-catenin and ERK1/2 signaling pathways. The mRNA expression levels of osteogenesis-related genes (Osteopontin, Osteocalcin, runt-related transcription factor 2 (Runx2), alkaline phosphatase (ALP) and bone morphogenetic protein 2) were significantly inhibited by N-cadherin, as well as the ALP activity and calcium deposit as stained by Alizarin Red S. While, silencing N-cadherin using shRNA reversed this effect. Furthermore, ectopic bone formation conducted in nude mice verified that N-cadherin significantly inhibited ectopic bone formation of MSCs in vivo. In addition, we also found that the N-cadherin overexpression could promote the migration potential of MSCs. These findings reveal that N-cadherin inhibits osteogenesis but promotes migration of MSCs. The underlying mechanism of N-cadherin inhibiting osteogenesis may through suppressing β-catenin and ERK1/2 signaling pathways.
Collapse
Affiliation(s)
- Liangliang Xu
- Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, People's Republic of China
| | | | | | | | | |
Collapse
|
38
|
Gurkan UA, Golden R, Kishore V, Riley CP, Adamec J, Akkus O. Immune and inflammatory pathways are involved in inherent bone marrow ossification. Clin Orthop Relat Res 2012; 470:2528-40. [PMID: 22798134 PMCID: PMC3830098 DOI: 10.1007/s11999-012-2459-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND Bone marrow plays a key role in bone formation and healing. Although a subset of marrow explants ossifies in vitro without excipient osteoinductive factors, some explants do not undergo ossification. The disparity of outcome suggests a significant heterogeneity in marrow tissue in terms of its capacity to undergo osteogenesis. QUESTIONS/PURPOSES We sought to identify: (1) proteins and signaling pathways associated with osteogenesis by contrasting the proteomes of ossified and poorly ossified marrow explants; and (2) temporal changes in proteome and signaling pathways of marrow ossification in the early and late phases of bone formation. METHODS Explants of marrow were cultured. Media conditioned by ossified (n = 4) and poorly ossified (n = 4) subsets were collected and proteins unique to each group were identified by proteomic analysis. Proteomic data were processed to assess proteins specific to the early phase (Days 1-14) and late phase (Days 15-28) of the culture period. Pathways involved in bone marrow ossification were identified through bioinformatics. RESULTS Twenty-eight proteins were unique to ossified samples and eight were unique to poorly ossified ones. Twelve proteins were expressed during the early phase and 15 proteins were specific to the late phase. Several identified pathways corroborated those reported for bone formation in the literature. Immune and inflammatory pathways were specific to ossified samples. CONCLUSIONS The marrow explant model indicates the inflammatory and immune pathways to be an integral part of the osteogenesis process.
Collapse
Affiliation(s)
- Umut Atakan Gurkan
- />Harvard-MIT Division of Health Sciences and Technology, Brigham and Women’s Hospital, Harvard Medical School, 65 Landsdowne Street, PRB 252, Cambridge, MA 02139 USA
| | - Ryan Golden
- />Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN USA
| | - Vipuil Kishore
- />Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
| | - Catherine P. Riley
- />Department of Research and Development Pathology Associates, Medical Laboratories, Spokane, WA 99204 USA
| | - Jiri Adamec
- />Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE USA
| | - Ozan Akkus
- />Department of Mechanical and Aerospace Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- />Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106 USA
- />Department of Orthopaedics, University Hospitals of Cleveland, Cleveland, OH USA
| |
Collapse
|
39
|
Paik S, Jung HS, Lee S, Yoon DS, Park MS, Lee JW. miR-449a regulates the chondrogenesis of human mesenchymal stem cells through direct targeting of lymphoid enhancer-binding factor-1. Stem Cells Dev 2012; 21:3298-308. [PMID: 22769578 DOI: 10.1089/scd.2011.0732] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
microRNAs are small molecules, about 17-23 nucleotides in length, that act as translational regulators of their target gene. By binding to a target, microRNAs are known to either inhibit translation or induce degradation of the target. Despite the great interest in microRNAs, however, the exact targets of each individual microRNA in different processes remain largely unknown. In this study, we determined that the lymphoid enhancer-binding factor-1 (LEF-1) was expressed during the chondrogenesis of human bone marrow-derived mesenchymal stem cells (hBM-MSCs) and sought to identify a novel microRNA targeting this gene. Through subsequent studies, we have identified, for the first time, one particular microRNA, miR-449a, that recognizes and regulates the expression of LEF-1 in a dose-dependent and sequence-specific manner. In addition, we observed that the inhibition of LEF-1 via miR-449a led to the subsequent repression of Sox 9, which is a well-established regulator of chondrogenesis. Collectively, this study demonstrated that miR-449a directly targets LEF-1, which in turn affects the expression of Sox 9, ultimately leading to the proper regulation of the differentiation and chondrogenesis of human MSCs (hBM-MSCs).
Collapse
Affiliation(s)
- Seungil Paik
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | | | | | | | | | | |
Collapse
|
40
|
Elshawi A, Wakamatsu N, Iinuma M, Nagayama M, Tamura Y. TMJ Degenerative Changes in SAMP3 Mice by Occlusal Disharmony and Aging. J HARD TISSUE BIOL 2012. [DOI: 10.2485/jhtb.21.399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
41
|
Kinumatsu T, Shibukawa Y, Yasuda T, Nagayama M, Yamada S, Serra R, Pacifici M, Koyama E. TMJ development and growth require primary cilia function. J Dent Res 2011; 90:988-94. [PMID: 21566205 DOI: 10.1177/0022034511409407] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Primary cilia regulate limb and axial skeletal formation and hedgehog signaling, but their roles in temporomandibular joint (TMJ) development are unknown. Thus, we created conditional mouse mutants deficient in ciliary transport protein Kif3a in cartilage. In post-natal wild-type mice, primary cilia were occasionally observed on the superior, inferior, or lateral side of condylar cells. Cilia were barely detectable in mutant chondrocytes but were evident in surrounding tissues, attesting to the specificity of chondrocyte Kif3a ablation. Mutant condyles from 3-month-old mice were narrow and flat along their antero-posterior and medio-lateral axes, were often fused with the articular disc, and displayed an irregular bony surface. The polymorphic layer in P15 mutants contained fewer Sox9-expressing chondroprogenitor cells because of reduced mitotic activity, and newly differentiated chondrocytes underwent precocious hypertrophic enlargement accompanied by early activation of Indian hedgehog (Ihh). Interestingly, there was excessive intramembranous ossification along the perichondrium, accompanied by local expression of the hedgehog receptor Patched-1 and up-regulation of Osterix and Collagen I. In summary, Kif3a and primary cilia are required for coordination of chondrocyte maturation, intramembranous bone formation, and chondrogenic condylar growth. Defects in these processes in Kif3a condylar cartilage are likely to reflect abnormal hedgehog signaling topography and dysfunction.
Collapse
Affiliation(s)
- T Kinumatsu
- Department of Surgery, The Children's Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center, 902 Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Sugito H, Shibukawa Y, Kinumatsu T, Yasuda T, Nagayama M, Yamada S, Minugh-Purvis N, Pacifici M, Koyama E. Ihh signaling regulates mandibular symphysis development and growth. J Dent Res 2011; 90:625-31. [PMID: 21297010 DOI: 10.1177/0022034510397836] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Symphyseal secondary cartilage is important for mandibular development, but the molecular mechanisms underlying its formation remain largely unknown. Here we asked whether Indian hedgehog (Ihh) regulates symphyseal cartilage development and growth. By embryonic days 16.5 to 18.5, Sox9-expressing chondrocytes formed within condensed Tgfβ-1/Runx2-expressing mesenchymal cells at the prospective symphyseal joint site, and established a growth-plate-like structure with distinct Ihh, collagen X, and osteopontin expression patterns. In post-natal life, mesenchymal cells expressing the Ihh receptor Patched1 were present anterior to the Ihh-expressing secondary cartilage, proliferated, differentiated into chondrocytes, and contributed to anterior growth of alveolar bone. In Ihh-null mice, however, symphyseal development was defective, mainly because of enhanced chondrocyte maturation and reduced proliferation of chondroprogenitor cells. Proliferation was partially restored in dual Ihh;Gli3 mutants, suggesting that Gli3 is normally a negative regulator of symphyseal development. Thus, Ihh signaling is essential for symphyseal cartilage development and anterior mandibular growth.
Collapse
Affiliation(s)
- H Sugito
- Department of Surgery, The Children's Hospital of Philadelphia Research Institute, 3615 Civic Center Boulevard, Abramson Research Center, 902 Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Yasuda T, Mundy C, Kinumatsu T, Shibukawa Y, Shibutani T, Grobe K, Minugh-Purvis N, Pacifici M, Koyama E. Sulfotransferase Ndst1 is needed for mandibular and TMJ development. J Dent Res 2010; 89:1111-6. [PMID: 20554886 DOI: 10.1177/0022034510373766] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Heparan sulfate proteoglycans (HS-PGs) regulate several developmental processes, but their possible roles in mandibular and TMJ formation are largely unclear. To uncover such roles, we generated mice lacking Golgi-associated N-sulfotransferase 1 (Ndst1) that catalyzes sulfation of HS-PG glycosaminoglycan chains. Ndst1-null mouse embryos exhibited different degrees of phenotypic penetrance. Severely affected mutants lacked the temporomandibular joint and condyle, but had a mandibular remnant that displayed abnormal tooth germs, substandard angiogenesis, and enhanced apoptosis. In mildly affected mutants, the condylar growth plate was dysfunctional and exhibited thicker superficial and polymorphic cell zones, a much wider distribution of Indian hedgehog signaling activity, and ectopic ossification along its lateral border. Interestingly, mildly affected mutants also exhibited facial asymmetry resembling that seen in individuals with hemifacial microsomia. Our findings indicate that Ndst1-dependent HS sulfation is critical for mandibular and TMJ development and allows HS-PGs to exert their roles via regulation of Ihh signaling topography and action.
Collapse
Affiliation(s)
- T Yasuda
- Department of Orthopaedic Surgery, Thomas Jefferson University College of Medicine, 1015 Walnut Street, Curtis Building Room 501, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ochiai T, Shibukawa Y, Nagayama M, Mundy C, Yasuda T, Okabe T, Shimono K, Kanyama M, Hasegawa H, Maeda Y, Lanske B, Pacifici M, Koyama E. Indian hedgehog roles in post-natal TMJ development and organization. J Dent Res 2010; 89:349-54. [PMID: 20200412 DOI: 10.1177/0022034510363078] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Indian hedgehog (Ihh) is essential for embryonic mandibular condylar growth and disc primordium formation. To determine whether it regulates those processes during post-natal life, we ablated Ihh in cartilage of neonatal mice and assessed the consequences on temporomandibular joint (TMJ) growth and organization over age. Ihh deficiency caused condylar disorganization and growth retardation and reduced polymorphic cell layer proliferation. Expression of Sox9, Runx2, and Osterix was low, as was that of collagen II, collagen I, and aggrecan, thus altering the fibrocartilaginous nature of the condyle. Though a disc formed, it exhibited morphological defects, partial fusion with the glenoid bone surface, reduced synovial cavity space, and, unexpectedly, higher lubricin expression. Analysis of the data shows, for the first time, that continuous Ihh action is required for completion of post-natal TMJ growth and organization. Lubricin overexpression in mutants may represent a compensatory response to sustain TMJ movement and function.
Collapse
Affiliation(s)
- T Ochiai
- Department of Orthopaedic Surgery, Thomas Jefferson University College of Medicine, 1015 Walnut Street, Curtis Building Room 501, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Choi HY, Dieckmann M, Herz J, Niemeier A. Lrp4, a novel receptor for Dickkopf 1 and sclerostin, is expressed by osteoblasts and regulates bone growth and turnover in vivo. PLoS One 2009; 4:e7930. [PMID: 19936252 PMCID: PMC2775917 DOI: 10.1371/journal.pone.0007930] [Citation(s) in RCA: 168] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2009] [Accepted: 10/27/2009] [Indexed: 12/14/2022] Open
Abstract
Lrp4 is a multifunctional member of the low density lipoprotein-receptor gene family and a modulator of extracellular cell signaling pathways in development. For example, Lrp4 binds Wise, a secreted Wnt modulator and BMP antagonist. Lrp4 shares structural elements within the extracellular ligand binding domain with Lrp5 and Lrp6, two established Wnt co-receptors with important roles in osteogenesis. Sclerostin is a potent osteocyte secreted inhibitor of bone formation that directly binds Lrp5 and Lrp6 and modulates both BMP and Wnt signaling. The anti-osteogenic effect of sclerostin is thought to be mediated mainly by inhibition of Wnt signaling through Lrp5/6 within osteoblasts. Dickkopf1 (Dkk1) is another potent soluble Wnt inhibitor that binds to Lrp5 and Lrp6, can displace Lrp5-bound sclerostin and is itself regulated by BMPs. In a recent genome-wide association study of bone mineral density a significant modifier locus was detected near the SOST gene at 17q21, which encodes sclerostin. In addition, nonsynonymous SNPs in the LRP4 gene were suggestively associated with bone mineral density. Here we show that Lrp4 is expressed in bone and cultured osteoblasts and binds Dkk1 and sclerostin in vitro. MicroCT analysis of Lrp4 deficient mutant mice revealed shortened total femur length, reduced cortical femoral perimeter, and reduced total femur bone mineral content (BMC) and bone mineral density (BMD). Lumbar spine trabecular bone volume per total volume (BV/TV) was significantly reduced in the mutants and the serum and urinary bone turnover markers alkaline phosphatase, osteocalcin and desoxypyridinoline were increased. We conclude that Lrp4 is a novel osteoblast expressed Dkk1 and sclerostin receptor with a physiological role in the regulation of bone growth and turnover, which is likely mediated through its function as an integrator of Wnt and BMP signaling pathways.
Collapse
Affiliation(s)
- Hong Y. Choi
- Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
| | - Marco Dieckmann
- Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- Institut für Physiologische Chemie und Pathobiochemie, Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, United States of America
- * E-mail:
| | - Andreas Niemeier
- Department of Orthopaedics and IBMII: Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Ochiai T, Nagayama M, Nakamura T, Morrison T, Pilchak D, Kondo N, Hasegawa H, Song B, Serra R, Pacifici M, Koyama E. Roles of the primary cilium component Polaris in synchondrosis development. J Dent Res 2009; 88:545-50. [PMID: 19587160 DOI: 10.1177/0022034509337775] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Primary cilia regulate several developmental processes and mediate hedgehog signaling. To study their roles in cranial base development, we created conditional mouse mutants deficient in Polaris, a critical primary cilium component, in cartilage. Mutant post-natal cranial bases were deformed, and their synchondrosis growth plates were disorganized. Expression of Indian hedgehog, Patched-1, collagen X, and MMP-13 was reduced and accompanied by decreases in endochondral bone. Interestingly, there was excessive intramembranous ossification along the perichondrium, accompanied by excessive Patched-1 expression, suggesting that Ihh distribution was wider and responsible for such excessive response. Indeed, expression of heparan sulfate proteoglycans (HS-PGs), normally involved in restricting hedgehog distribution, was barely detectable in mutant synchondroses. Analyses of the data provides further evidence for the essential roles of primary cilia and hedgehog signaling in cranial base development and chondrocyte maturation, and point to a close interdependence between cilia and HS-PGs to delimit targets of hedgehog action in synchondroses.
Collapse
Affiliation(s)
- T Ochiai
- Department of Orthopaedic Surgery, Thomas Jefferson University College of Medicine, 1015 Walnut Street, Curtis Building, Room 501, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wealthall RJ. In vitro regulation of proliferation and differentiation within a postnatal growth plate of the cranial base by parathyroid hormone-related peptide (PTHrP). J Cell Physiol 2009; 219:688-97. [PMID: 19229881 DOI: 10.1002/jcp.21716] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Parathyroid hormone-related peptide (PTHrP) is known to be an important regulator of chondrocyte differentiation in embryonic growth plates, but little is known of its role in postnatal growth plates. The present study explores the role of PTHrP in regulating postnatal chondrocyte differentiation using a novel in vitro organ culture model based on the ethmoidal growth plate of the cranial base taken from the postnatal day 10 mouse. In vitro the ethmoidal growth plate continued to mineralize and the chondrocytes progressed to hypertrophy, as observed in vivo, but the proliferative zone was not maintained. Treatment with PTHrP inhibited mineralization and reduced alkaline phosphatase (ALP) activity in the hypertrophic zone in the ethmoidal growth plates grown ex vivo, and also increased the proliferation of non-hypertrophic chondrocytes. In addition, exogenous PTHrP reduced the expression of genes associated with terminal differentiation: type X collagen, Runx2, and ALP, as well as the PTH/PTHrP receptor (PPR). Activation of the protein kinase A pathway using 8-Br-cAMP mimicked some of these pro-proliferative/anti-differentiative effects of PTHrP. PTHrP and PPR were found to be expressed within the ethmoidal growth plate using semi-quantitative PCR, and in other cranial growth plates such as the spheno-occipital and pre-sphenoidal synchondroses. These results provide the first functional evidence that PTHrP regulates proliferation and differentiation within the postnatal, cranial growth plate. J. Cell. Physiol. 219: 688-697, 2009. (c) 2009 Wiley-Liss, Inc.
Collapse
|
48
|
McBratney-Owen B, Iseki S, Bamforth SD, Olsen BR, Morriss-Kay GM. Development and tissue origins of the mammalian cranial base. Dev Biol 2008; 322:121-32. [PMID: 18680740 PMCID: PMC2847450 DOI: 10.1016/j.ydbio.2008.07.016] [Citation(s) in RCA: 186] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 07/10/2008] [Accepted: 07/10/2008] [Indexed: 10/21/2022]
Abstract
The vertebrate cranial base is a complex structure composed of bone, cartilage and other connective tissues underlying the brain; it is intimately connected with development of the face and cranial vault. Despite its central importance in craniofacial development, morphogenesis and tissue origins of the cranial base have not been studied in detail in the mouse, an important model organism. We describe here the location and time of appearance of the cartilages of the chondrocranium. We also examine the tissue origins of the mouse cranial base using a neural crest cell lineage cell marker, Wnt1-Cre/R26R, and a mesoderm lineage cell marker, Mesp1-Cre/R26R. The chondrocranium develops between E11 and E16 in the mouse, beginning with development of the caudal (occipital) chondrocranium, followed by chondrogenesis rostrally to form the nasal capsule, and finally fusion of these two parts via the midline central stem and the lateral struts of the vault cartilages. X-Gal staining of transgenic mice from E8.0 to 10 days post-natal showed that neural crest cells contribute to all of the cartilages that form the ethmoid, presphenoid, and basisphenoid bones with the exception of the hypochiasmatic cartilages. The basioccipital bone and non-squamous parts of the temporal bones are mesoderm derived. Therefore the prechordal head is mostly composed of neural crest-derived tissues, as predicted by the New Head Hypothesis. However, the anterior location of the mesoderm-derived hypochiasmatic cartilages, which are closely linked with the extra-ocular muscles, suggests that some tissues associated with the visual apparatus may have evolved independently of the rest of the "New Head".
Collapse
Affiliation(s)
- B McBratney-Owen
- Harvard School of Dental Medicine, Department of Developmental Biology, 190 Longwood Avenue, Boston, MA, 02115, USA.
| | | | | | | | | |
Collapse
|