1
|
Liu Y, Jia F, Li K, Liang C, Lin X, Geng W, Li Y. Critical signaling molecules in the temporomandibular joint osteoarthritis under different magnitudes of mechanical stimulation. Front Pharmacol 2024; 15:1419494. [PMID: 39055494 PMCID: PMC11269110 DOI: 10.3389/fphar.2024.1419494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/14/2024] [Indexed: 07/27/2024] Open
Abstract
The mechanical stress environment in the temporomandibular joint (TMJ) is constantly changing due to daily mandibular movements. Therefore, TMJ tissues, such as condylar cartilage, the synovial membrane and discs, are influenced by different magnitudes of mechanical stimulation. Moderate mechanical stimulation is beneficial for maintaining homeostasis, whereas abnormal mechanical stimulation leads to degeneration and ultimately contributes to the development of temporomandibular joint osteoarthritis (TMJOA), which involves changes in critical signaling molecules. Under abnormal mechanical stimulation, compensatory molecules may prevent degenerative changes while decompensatory molecules aggravate. In this review, we summarize the critical signaling molecules that are stimulated by moderate or abnormal mechanical loading in TMJ tissues, mainly in condylar cartilage. Furthermore, we classify abnormal mechanical stimulation-induced molecules into compensatory or decompensatory molecules. Our aim is to understand the pathophysiological mechanism of TMJ dysfunction more deeply in the ever-changing mechanical environment, and then provide new ideas for discovering effective diagnostic and therapeutic targets in TMJOA.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Geng
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| | - Yanxi Li
- Department of Dental Implant Center, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Kamalitdinov TB, Fujino K, Keith Lang S, Jiang X, Madi R, Evans MK, Zgonis MH, Kuntz AF, Dyment NA. Targeting the hedgehog signaling pathway to improve tendon-to-bone integration. Osteoarthritis Cartilage 2023; 31:1202-1213. [PMID: 37146960 PMCID: PMC10524548 DOI: 10.1016/j.joca.2023.04.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 04/24/2023] [Accepted: 04/29/2023] [Indexed: 05/07/2023]
Abstract
OBJECTIVE While the role of hedgehog (Hh) signaling in promoting zonal fibrocartilage production during development is well-established, whether this pathway can be leveraged to improve tendon-to-bone repair in adults is unknown. Our objective was to genetically and pharmacologically stimulate the Hh pathway in cells that give rise to zonal fibrocartilaginous attachments to promote tendon-to-bone integration. DESIGN Hh signaling was stimulated genetically via constitutive Smo (SmoM2 construct) activation of bone marrow stromal cells or pharmacologically via systemic agonist delivery to mice following anterior cruciate ligament reconstruction (ACLR). To assess tunnel integration, we measured mineralized fibrocartilage (MFC) formation in these mice 28 days post-surgery and performed tunnel pullout testing. RESULTS Hh pathway-related genes increased in cells forming the zonal attachments in wild-type mice. Both genetic and pharmacologic stimulation of the Hh pathway increased MFC formation and integration strength 28 days post-surgery. We next conducted studies to define the role of Hh in specific stages of the tunnel integration process. We found Hh agonist treatment increased the proliferation of the progenitor pool in the first week post-surgery. Additionally, genetic stimulation led to continued MFC production in the later stages of the integration process. These results indicate that Hh signaling plays an important biphasic role in cell proliferation and differentiation towards fibrochondrocytes following ACLR. CONCLUSION This study reveals a biphasic role for Hh signaling during the tendon-to-bone integration process after ACLR. In addition, the Hh pathway is a promising therapeutic target to improve tendon-to-bone repair outcomes.
Collapse
Affiliation(s)
- Timur B Kamalitdinov
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Keitaro Fujino
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Osaka Medical and Pharmaceutical University, Takatsuki, Osaka Prefecture, Japan
| | - Sinaia Keith Lang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Biology, University of Pennsylvania, Philadelphia, PA, USA
| | - Xi Jiang
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Rashad Madi
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Mary Kate Evans
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Miltiadis H Zgonis
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew F Kuntz
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathaniel A Dyment
- Department of Orthopaedic Surgery, University of Pennsylvania, Philadelphia, PA, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
3
|
Zhu Q, Tan M, Wang C, Chen Y, Wang C, Zhang J, Gu Y, Guo Y, Han J, Li L, Jiang R, Fan X, Xie H, Wang L, Gu Z, Liu D, Shi J, Feng X. Single-cell RNA sequencing analysis of the temporomandibular joint condyle in 3 and 4-month-old human embryos. Cell Biosci 2023; 13:130. [PMID: 37468984 DOI: 10.1186/s13578-023-01069-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/13/2023] [Indexed: 07/21/2023] Open
Abstract
BACKGROUND The temporomandibular joint (TMJ) is a complex joint consisting of the condyle, the temporal articular surface, and the articular disc. Functions such as mastication, swallowing and articulation are accomplished by the movements of the TMJ. To date, the TMJ has been studied more extensively, but the types of TMJ cells, their differentiation, and their interrelationship during growth and development are still unclear and the study of the TMJ is limited. The aim of this study was to establish a molecular cellular atlas of the human embryonic temporomandibular joint condyle (TMJC) by single-cell RNA sequencing, which will contribute to understanding and solving clinical problems. RESULTS Human embryos at 3 and 4 months of age are an important stage of TMJC development. We performed a comprehensive transcriptome analysis of TMJC tissue from human embryos at 3 and 4 months of age using single-cell RNA sequencing. A total of 16,624 cells were captured and the gene expression profiles of 15 cell clusters in human embryonic TMJC were determined, including 14 known cell types and one previously unknown cell type, "transition state cells (TSCs)". Immunofluorescence assays confirmed that TSCs are not the same cell cluster as mesenchymal stem cells (MSCs). Pseudotime trajectory and RNA velocity analysis revealed that MSCs transformed into TSCs, which further differentiated into osteoblasts, hypertrophic chondrocytes and tenocytes. In addition, chondrocytes (CYTL1high + THBS1high) from secondary cartilage were detected only in 4-month-old human embryonic TMJC. CONCLUSIONS Our study provides an atlas of differentiation stages of human embryonic TMJC tissue cells, which will contribute to an in-depth understanding of the pathophysiology of the TMJC tissue repair process and ultimately help to solve clinical problems.
Collapse
Affiliation(s)
- Qianqi Zhu
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Miaoying Tan
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Chengniu Wang
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China
| | - Yufei Chen
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China
| | - Chenfei Wang
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Junqi Zhang
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Yijun Gu
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Yuqi Guo
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Jianpeng Han
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Lei Li
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Rongrong Jiang
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Xudong Fan
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Huimin Xie
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Liang Wang
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China
| | - Zhifeng Gu
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China.
| | - Dong Liu
- School of Life Science, Nantong Laboratory of Development and Diseases Second Affiliated Hospital Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226019, China.
| | - Jianwu Shi
- Institute of Reproductive Medicine, Medical School of Nantong University, Nantong, 226001, China.
| | - Xingmei Feng
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of, Nantong University, Nantong, 226001, China.
| |
Collapse
|
4
|
Zhang N, Barrell WB, Liu KJ. Identification of distinct subpopulations of Gli1-lineage cells in the mouse mandible. J Anat 2023; 243:90-99. [PMID: 36899483 PMCID: PMC10273353 DOI: 10.1111/joa.13858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/12/2023] Open
Abstract
The Hedgehog pathway gene Gli1 has been proposed to mark a subpopulation of skeletal stem cells (SSCs) in craniofacial bone. Skeletal stem cells (SSCs) are multi-potent cells crucial for the development and homeostasis of bone. Recent studies on long bones have suggested that skeletal stem cells in endochondral or intramembranous ossification sites have different differentiation capacities. However, this has not been well-defined in neural crest derived bones. Generally, the long bones are derived from mesoderm and follow an endochondral ossification model, while most of the cranial bones are neural crest (NC) in origin and follow an intramembranous ossification model. The mandible is unique: It is derived from the neural crest lineage but makes use of both modes of ossification. Early in fetal development, the mandibular body is generated by intramembranous ossification with subsequent endochondral ossification forming the condyle. The identities and properties for SSCs in these two sites remain unknown. Here, we use genetic lineage tracing in mouse to identify cells expressing the Hedgehog responsive gene Gli1, which is thought to mark the tissue resident SSCs. We track the Gli1+ cells, comparing cells within the perichondrium to those in the periosteum covering the mandibular body. In juvenile mice, these have distinct differentiation and proliferative potential. We also assess the presence of Sox10+ cells, thought to mark neural crest stem cells, but find no substantial population associated with the mandibular skeleton, suggesting that Sox10+ cells have limited contribution to maintaining postnatal mandibular bone. All together, our study indicates that the Gli1+ cells display distinct and limited differentiation capacity dependent on their regional associations.
Collapse
Affiliation(s)
- Nian Zhang
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial SciencesKing's College LondonLondonUK
- State Key Laboratory of Oral Disease, Department of Oral and Maxillofacial Surgery, National Clinical Research Center for Oral DiseasesWest China Hospital of Stomatogy, Sichuan UniversityChengduChina
| | - William B. Barrell
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial SciencesKing's College LondonLondonUK
| | - Karen J. Liu
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral and Craniofacial SciencesKing's College LondonLondonUK
| |
Collapse
|
5
|
Huang L, Jin M, Gu R, Xiao K, Lu M, Huo X, Sun M, Yang Z, Wang Z, Zhang W, Zhi L, Meng Z, Ma J, Ma J, Zhang R. miR-199a-5p Reduces Chondrocyte Hypertrophy and Attenuates Osteoarthritis Progression via the Indian Hedgehog Signal Pathway. J Clin Med 2023; 12:jcm12041313. [PMID: 36835852 PMCID: PMC9959662 DOI: 10.3390/jcm12041313] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Osteoarthritis (OA), the most common type of arthritis, is an age-associated disease, characterized by the progressive degradation of articular cartilage, synovial inflammation, and degeneration of subchondral bone. Chondrocyte proliferation is regulated by the Indian hedgehog (IHH in humans, Ihh in animals) signaling molecule, which regulates hypertrophy and endochondral ossification in the development of the skeletal system. microRNAs (miRNAs, miRs) are a family of about 22-nucleotide endogenous non-coding RNAs, which negatively regulate gene expression. In this study, the expression level of IHH was upregulated in the damaged articular cartilage tissues among OA patients and OA cell cultures, while that of miR-199a-5p was the opposite. Further investigations demonstrated that miR-199a-5p could directly regulate IHH expression and reduce chondrocyte hypertrophy and matrix degradation via the IHH signal pathway in the primary human chondrocytes. The intra-articular injection of synthetic miR-199a-5p agomir attenuated OA symptoms in rats, including the alleviation of articular cartilage destruction, subchondral bone degradation, and synovial inflammation. The miR-199a-5p agomir could also inhibit the Ihh signaling pathway in vivo. This study might help in understanding the role of miR-199a-5p in the pathophysiology and molecular mechanisms of OA and indicate a potential novel therapeutic strategy for OA patients.
Collapse
Affiliation(s)
- Lei Huang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Meng Jin
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ruiying Gu
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Kunlin Xiao
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Mengnan Lu
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Xinyu Huo
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Mengyao Sun
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Zhi Yang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Zhiyuan Wang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Weijie Zhang
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Liqiang Zhi
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Ziang Meng
- Department of Mathematics and Computing Science, Simon Fraser University, Vancouver, BC V6B 5K3, Canada
| | - Jie Ma
- School of Basic Medical Science, Xi’an Jiaotong University Health Science Center, Xi’an 710049, China
| | - Jianbing Ma
- Department of Joint Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: (J.M.); (R.Z.)
| | - Rui Zhang
- Translational Medicine Center, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
- Correspondence: (J.M.); (R.Z.)
| |
Collapse
|
6
|
Kikuchi K, Haneda M, Hayashi S, Maeda T, Nakano N, Kuroda Y, Tsubosaka M, Kamenaga T, Fujita M, Ikuta K, Anjiki K, Tachibana S, Onoi Y, Matsumoto T, Kuroda R. P21 deficiency exhibits delayed endochondral ossification during fracture healing. Bone 2022; 165:116572. [PMID: 36180020 DOI: 10.1016/j.bone.2022.116572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 09/21/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Endochondral ossification is a complex biological phenomenon involving a variety of factors and cells. Cyclin-dependent kinase inhibitor 1 (p21) inhibits cell cycle progression and is affected by external stress. We recently reported that embryonic endochondral ossification is unaffected by endogenous p21 deficiency. In this study, we evaluated whether p21 expression affects endochondral ossification during fracture healing. METHODS Tibial fractures were introduced into p21 knockout (p21-/-) (n = 24) and wild-type C57BL/6 (p21+/+) (n = 24) mice at age 10 weeks. Fracture healing was evaluated using radiological, histological, and immunohistochemical (IHC) analyses. The effect of p21 small interfering RNA (siRNA) on ATDC5 cells was assessed in vitro. RESULTS The Allen score for fracture healing was lower in p21-/- mice than in p21+/+ mice. In addition, p21-/- mice exhibited larger calluses and lower bone mineral density. IHC analyses showed that p21-/- mice exhibited delayed endochondral ossification via the Ihh-Runx2-Osterix pathway in vivo. Down-regulation of p21 expression in ATDC5 cells delayed endochondral ossification in vitro. CONCLUSIONS p21 deficiency leads to delayed endochondral ossification by attenuating the Ihh-Runx2-Osterix pathway in vivo, and p21 deficiency in hypertrophic chondrocytes causes delayed differentiation of hypertrophic chondrocytes in vitro. p21 plays a role in endochondral ossification during fracture healing.
Collapse
Affiliation(s)
- Kenichi Kikuchi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiko Haneda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shinya Hayashi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan.
| | - Toshihisa Maeda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Naoki Nakano
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuichi Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masanori Tsubosaka
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Kamenaga
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Masahiro Fujita
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kenmei Ikuta
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kensuke Anjiki
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Shotaro Tachibana
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yuma Onoi
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Tomoyuki Matsumoto
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Ryosuke Kuroda
- Department of Orthopaedic Surgery, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
7
|
Mangu SR, Patel K, Sukhdeo SV, Savitha MR, Sharan K. Maternal high-cholesterol diet negatively programs offspring bone development and downregulates hedgehog signaling in osteoblasts. J Biol Chem 2022; 298:102324. [PMID: 35931113 PMCID: PMC9440389 DOI: 10.1016/j.jbc.2022.102324] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cholesterol is one of the essential intrauterine factors required for fetal growth and development. Maternal high cholesterol levels are known to be detrimental for offspring health. However, its long-term effect on offspring skeletal development remains to be elucidated. We performed our studies in two strains of mice (C57BL6/J and Swiss Albino) and human subjects (65 mother-female newborn dyads) to understand the regulation of offspring skeletal growth by maternal high cholesterol. We found that mice offspring from high-cholesterol-fed dams had low birth weight, smaller body length, and delayed skeletal ossification at the E18.5 embryonic stage. Moreover, we observed that the offspring did not recover from the reduced skeletal mass and exhibited a low bone mass phenotype throughout their life. We attributed this effect to reduced osteoblast cell activity with a concomitant increase in the osteoclast cell population. Our investigation of the molecular mechanism revealed that offspring from high-cholesterol-fed dams had a decrease in the expression of ligands and proteins involved in hedgehog signaling. Further, our cross-sectional study of human subjects showed a significant inverse correlation between maternal blood cholesterol levels and cord blood bone formation markers. Moreover, the bone formation markers were significantly lower in the female newborns of hypercholesterolemic mothers compared with mothers with normal cholesterolemic levels. Together, our results suggest that maternal high cholesterol levels deleteriously program offspring bone mass and bone quality and downregulate the hedgehog signaling pathway in their osteoblasts.
Collapse
Affiliation(s)
- Svvs Ravi Mangu
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kalpana Patel
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Shinde Vijay Sukhdeo
- Department of Meat and Marine Sciences, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - M R Savitha
- Department of Paediatrics, Mysore Medical College and Research Institute, Mysuru, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
8
|
Su Y, Xing H, Kang J, Bai L, Zhang L. Role of the hedgehog signaling pathway in rheumatic diseases: An overview. Front Immunol 2022; 13:940455. [PMID: 36105801 PMCID: PMC9466598 DOI: 10.3389/fimmu.2022.940455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/05/2022] [Indexed: 11/13/2022] Open
Abstract
Hedgehog (Hh) signaling pathway is an evolutionarily conserved signal transduction pathway that plays an important regulatory role during embryonic development, cell proliferation, and differentiation of vertebrates, and it is often inhibited in adult tissues. Recent evidence has shown that Hh signaling also plays a key role in rheumatic diseases, as alterations in their number or function have been identified in rheumatoid arthritis, osteoarthritis, ankylosing spondylitis, systemic sclerosis, and Sjogren's Syndrome. As a result, emerging studies have focused on the blockade of this pathogenic axis as a promising therapeutic target in several autoimmune disorders; nevertheless, a greater understanding of its contribution still requires further investigation. This review aims to elucidate the most recent studies and literature data on the pathogenetic role of Hh signaling in rheumatic diseases.
Collapse
Affiliation(s)
| | | | | | | | - Liyun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, Shanxi, China
| |
Collapse
|
9
|
Thielen NGM, Neefjes M, Vitters EL, van Beuningen HM, Blom AB, Koenders MI, van Lent PLEM, van de Loo FAJ, Blaney Davidson EN, van Caam APM, van der Kraan PM. Identification of Transcription Factors Responsible for a Transforming Growth Factor-β-Driven Hypertrophy-like Phenotype in Human Osteoarthritic Chondrocytes. Cells 2022; 11:cells11071232. [PMID: 35406794 PMCID: PMC8998018 DOI: 10.3390/cells11071232] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/28/2022] [Accepted: 03/31/2022] [Indexed: 12/18/2022] Open
Abstract
During osteoarthritis (OA), hypertrophy-like chondrocytes contribute to the disease process. TGF-β's signaling pathways can contribute to a hypertrophy(-like) phenotype in chondrocytes, especially at high doses of TGF-β. In this study, we examine which transcription factors (TFs) are activated and involved in TGF-β-dependent induction of a hypertrophy-like phenotype in human OA chondrocytes. We found that TGF-β, at levels found in synovial fluid in OA patients, induces hypertrophic differentiation, as characterized by increased expression of RUNX2, COL10A1, COL1A1, VEGFA and IHH. Using luciferase-based TF activity assays, we observed that the expression of these hypertrophy genes positively correlated to SMAD3:4, STAT3 and AP1 activity. Blocking these TFs using specific inhibitors for ALK-5-induced SMAD signaling (5 µM SB-505124), JAK-STAT signaling (1 µM Tofacitinib) and JNK signaling (10 µM SP-600125) led to the striking observation that only SB-505124 repressed the expression of hypertrophy factors in TGF-β-stimulated chondrocytes. Therefore, we conclude that ALK5 kinase activity is essential for TGF-β-induced expression of crucial hypertrophy factors in chondrocytes.
Collapse
|
10
|
Zhang J, Liu J, Zhang H, Wang J, Hua H, Jiang Y. The role of network-forming collagens in cancer progression. Int J Cancer 2022; 151:833-842. [PMID: 35322886 DOI: 10.1002/ijc.34004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/05/2023]
Abstract
Collagens are the main components of extracellular matrix in the tumor microenvironment. Both fibrillar and nonfibrillar collagens are involved in tumor progression. The nonfibrillar network-forming collagens such as type IV and type VIII collagens are frequently overexpressed in various types of human cancers, which promotes tumor cell proliferation, adhesion, invasion, metastasis and angiogenesis. Studies on the roles of these collagens have shed light on the mechanisms underpinning the effects of this protein family. Future research has to explicit the role of network-forming collagens with respect to cancer progression and treatment. Herein, we review the regulation of network-forming collagens expression in cancer; the roles of network-forming collagens in tumor invasion, metastasis and angiogenesis; and the clinical significance of network-forming collagens expression in cancer patients. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jin Zhang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jieya Liu
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongying Zhang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Wang
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, China
| | - Hui Hua
- Laboratory of Stem Cell Biology, West China Hospital, Sichuan University, Chengdu, China
| | - Yangfu Jiang
- Laboratory of Oncogene, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
11
|
On the horizon: Hedgehog signaling to heal broken bones. Bone Res 2022; 10:13. [PMID: 35165260 PMCID: PMC8844053 DOI: 10.1038/s41413-021-00184-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/21/2021] [Accepted: 11/25/2021] [Indexed: 12/22/2022] Open
Abstract
Uncovering the molecular pathways that drive skeletal repair has been an ongoing challenge. Initial efforts have relied on in vitro assays to identify the key signaling pathways that drive cartilage and bone differentiation. While these assays can provide some clues, assessing specific pathways in animal models is critical. Furthermore, definitive proof that a pathway is required for skeletal repair is best provided using genetic tests. Stimulating the Hh (Hedgehog) pathway can promote cartilage and bone differentiation in cell culture assays. In addition, the application of HH protein or various pathway agonists in vivo has a positive influence on bone healing. Until recently, however, genetic proof that the Hh pathway is involved in bone repair has been lacking. Here, we consider both in vitro and in vivo studies that examine the role of Hh in repair and discuss some of the challenges inherent in their interpretation. We also identify needed areas of study considering a new appreciation for the role of cartilage during repair, the variety of cell types that may have differing roles in repair, and the recent availability of powerful lineage tracing techniques. We are optimistic that emerging genetic tools will make it possible to precisely define when and in which cells promoting Hh signaling can best promote skeletal repair, and thus, the clinical potential for targeting the Hh pathway can be realized.
Collapse
|
12
|
O-fucosylation of thrombospondin type 1 repeats is essential for ECM remodeling and signaling during bone development. Matrix Biol 2022; 107:77-96. [DOI: 10.1016/j.matbio.2022.02.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 01/18/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022]
|
13
|
Chabronova A, van den Akker GGH, Meekels-Steinbusch MMF, Friedrich F, Cremers A, Surtel DAM, Peffers MJ, van Rhijn LW, Lausch E, Zabel B, Caron MMJ, Welting TJM. Uncovering pathways regulating chondrogenic differentiation of CHH fibroblasts. Noncoding RNA Res 2022; 6:211-224. [PMID: 34988338 PMCID: PMC8688813 DOI: 10.1016/j.ncrna.2021.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/08/2021] [Accepted: 12/08/2021] [Indexed: 02/08/2023] Open
Abstract
Mutations in the non-coding snoRNA component of mitochondrial RNA processing endoribonuclease (RMRP) are the cause of cartilage-hair hypoplasia (CHH). CHH is a rare form of metaphyseal chondrodysplasia characterized by disproportionate short stature and abnormal growth plate development. The process of chondrogenic differentiation within growth plates of long bones is vital for longitudinal bone growth. However, molecular mechanisms behind impaired skeletal development in CHH patients remain unclear. We employed a transdifferentiation model (FDC) combined with whole transcriptome analysis to investigate the chondrogenic transdifferentiation capacity of CHH fibroblasts and to examine pathway regulation in CHH cells during chondrogenic differentiation. We established that the FDC transdifferentiation model is a relevant in vitro model of chondrogenic differentiation, with an emphasis on the terminal differentiation phase, which is crucial for longitudinal bone growth. We demonstrated that CHH fibroblasts are capable of transdifferentiating into chondrocyte-like cells, and show a reduced commitment to terminal differentiation. We also found a number of key factors of BMP, FGF, and IGF-1 signalling axes to be significantly upregulated in CHH cells during the chondrogenic transdifferentiation. Our results support postulated conclusions that RMRP has pleiotropic functions and profoundly affects multiple aspects of cell fate and signalling. Our findings shed light on the consequences of pathological CHH mutations in snoRNA RMRP during chondrogenic differentiation and the relevance and roles of non-coding RNAs in genetic diseases in general.
Collapse
Affiliation(s)
- Alzbeta Chabronova
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Guus G H van den Akker
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Mandy M F Meekels-Steinbusch
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Franziska Friedrich
- Department of Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Mandy J Peffers
- Institute of Life Course and Medical Sciences, University of Liverpool, William Henry Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Ekkehart Lausch
- Department of Pediatrics, Medical Center, University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bernhard Zabel
- Medical Faculty, Otto van Guericke University of Magdeburg, 39106, Magdeburg, Germany
| | - Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, MUMC+, 6202, AZ, Maastricht, the Netherlands
| |
Collapse
|
14
|
Wang Z, Wang B, Zhang J, Wu Z, Yu L, Sun Z. Chemokine (C-C Motif) Ligand 2/Chemokine Receptor 2 (CCR2) Axis Blockade to Delay Chondrocyte Hypertrophy as a Therapeutic Strategy for Osteoarthritis. Med Sci Monit 2021; 27:e930053. [PMID: 34876548 PMCID: PMC8667482 DOI: 10.12659/msm.930053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Chondrocytes play a vital role in the later stages of osteoarthritis (OA). The roles of chemokine (C-C motif) ligand 2 (CCL2) and its receptor, chemokine receptor 2 (CCR2), are as yet poorly elucidated in chondrocyte hypertrophy (CH). Here, we aimed to regulate the CCL2/CCR2 axis and explore its effect on progression of CH. Material/Methods Chondrocytes isolated from patients with OA were used in the present study. In vitro experiments were conducted to test hypertrophic gene and CCL2/CCR2 expression in chondrocyte degeneration caused by interleukin (IL)-17A or CCL2 protein stimulation. In addition, inhibition of CCL2 and CCR2 was used to assess the role of CCL2 and CCR2 blockade in CH. Relative gene expression was determined with real-time polymerase chain reaction, western blot, or immunofluorescence. Hypertrophic changes were assessed with cell area measurement. Moreover, the viability of chondrocytes was analyzed using an MTT assay and flow cytometry was used to assess cell apoptosis. Results CCL2 and CCR2 were upregulated in IL-17A-treated chondrocytes. The exogenic CCL2 stimulation also promoted CH and increased the expression of Type 10 collagen, RUNX2, and IHH, which could be reversed via suppression of CCR2. Inhibition of CCL2 and CCR2 expression was sufficient to: 1) protect Type 2 collagen synthesis; 2) alleviate IL-17A-induced overexpression of Type 10 collagen, RUNX2, and IHH; and 3) improve chondrocyte proliferation and apoptosis. Conclusions Blockading the CCL2/CCR2 axis plays a role in delaying the development of CH.
Collapse
Affiliation(s)
- Zidong Wang
- Department of Orthopedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Bei Wang
- Department of Imaging, Liaocheng Infectious Disease Hospital, Liaocheng, Shandong, China (mainland)
| | - Jian Zhang
- Department of Orthopedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Zhensong Wu
- Department of Joint Sports Medicine, Zaozhuang Municipal Hospital, Zaozhuang, Shandong, China (mainland)
| | - Liankui Yu
- Department of Orthopedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| | - Zhongye Sun
- Department of Orthopedic Surgery, Liaocheng People's Hospital, Liaocheng, Shandong, China (mainland)
| |
Collapse
|
15
|
Li B, Guan G, Mei L, Jiao K, Li H. Pathological mechanism of chondrocytes and the surrounding environment during osteoarthritis of temporomandibular joint. J Cell Mol Med 2021; 25:4902-4911. [PMID: 33949768 PMCID: PMC8178251 DOI: 10.1111/jcmm.16514] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
Temporomandibular joint (TMJ) osteoarthritis is a common chronic degenerative disease of the TMJ. In order to explore its aetiology and pathological mechanism, many animal models and cell models have been constructed to simulate the pathological process of TMJ osteoarthritis. The main pathological features of TMJ osteoarthritis include chondrocyte death, extracellular matrix (ECM) degradation and subchondral bone remodelling. Chondrocyte apoptosis accelerates the destruction of cartilage. However, autophagy has a protective effect on condylar chondrocytes. Degradation of ECM not only changes the properties of cartilage but also affects the phenotype of chondrocytes. The loss of subchondral bone in the early stages of TMJ osteoarthritis plays an aetiological role in the onset of osteoarthritis. In recent years, increasing evidence has suggested that chondrocyte hypertrophy and endochondral angiogenesis promote TMJ osteoarthritis. Hypertrophic chondrocytes secrete many factors that promote cartilage degeneration. These chondrocytes can further differentiate into osteoblasts and osteocytes and accelerate cartilage ossification. Intrachondral angiogenesis and neoneurogenesis are considered to be important triggers of arthralgia in TMJ osteoarthritis. Many molecular signalling pathways in endochondral osteogenesis are responsible for TMJ osteoarthritis. These latest discoveries in TMJ osteoarthritis have further enhanced the understanding of this disease and contributed to the development of molecular therapies. This paper summarizes recent cognition on the pathogenesis of TMJ osteoarthritis, focusing on the role of chondrocyte hypertrophy degeneration and cartilage angiogenesis.
Collapse
Affiliation(s)
- Baochao Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Guangzhao Guan
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Li Mei
- Department of Oral Sciences, Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Kai Jiao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Huang Li
- Department of Orthodontics, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
16
|
Ono K, Hata K, Nakamura E, Ishihara S, Kobayashi S, Nakanishi M, Yoshida M, Takahata Y, Murakami T, Takenoshita S, Komori T, Nishimura R, Yoneda T. Dmrt2 promotes transition of endochondral bone formation by linking Sox9 and Runx2. Commun Biol 2021; 4:326. [PMID: 33707608 PMCID: PMC7952723 DOI: 10.1038/s42003-021-01848-1] [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: 08/20/2020] [Accepted: 02/12/2021] [Indexed: 11/23/2022] Open
Abstract
Endochondral bone formation is fundamental for skeletal development. During this process, chondrocytes undergo multiple steps of differentiation and coordinated transition from a proliferating to a hypertrophic stage, which is critical to advance skeletal development. Here, we identified the transcription factor Dmrt2 (double-sex and mab-3 related transcription factor 2) as a Sox9-inducible gene that promotes chondrocyte hypertrophy in pre-hypertrophic chondrocytes. Epigenetic analysis further demonstrated that Sox9 regulates Dmrt2 expression through an active enhancer located 18 kb upstream of the Dmrt2 gene and that this enhancer's chromatin status is progressively activated through chondrocyte differentiation. Dmrt2-knockout mice exhibited a dwarf phenotype with delayed initiation of chondrocyte hypertrophy. Dmrt2 augmented hypertrophic chondrocyte gene expression including Ihh through physical and functional interaction with Runx2. Furthermore, Dmrt2 deficiency reduced Runx2-dependent Ihh expression. Our findings suggest that Dmrt2 is critical for sequential chondrocyte differentiation during endochondral bone formation and coordinates the transcriptional network between Sox9 and Runx2.
Collapse
Affiliation(s)
- Koichiro Ono
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Orthopedics, Nippon Medical School, Tokyo, Japan
| | - Kenji Hata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan.
| | - Eriko Nakamura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Shota Ishihara
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Sachi Kobayashi
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Masako Nakanishi
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
- Department of Pathology, Wakayama Medical University, Wakayama, Japan
| | - Michiko Yoshida
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Yoshifumi Takahata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Tomohiko Murakami
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| | - Seiichi Takenoshita
- Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| | - Toshihisa Komori
- Department of Cell Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan.
| | - Toshiyuki Yoneda
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, Osaka, Japan
| |
Collapse
|
17
|
Caron MMJ, van Rietbergen B, Castermans TMR, Haartmans MJJ, van Rhijn LW, Welting TJM, Witlox AMA. Evaluation of impaired growth plate development of long bones in skeletally immature mice by antirheumatic agents. J Orthop Res 2021; 39:553-564. [PMID: 32740982 PMCID: PMC7984053 DOI: 10.1002/jor.24819] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 06/05/2020] [Accepted: 07/13/2020] [Indexed: 02/04/2023]
Abstract
Restriction of physical growth and development is a known problem in patients with juvenile idiopathic arthritis (JIA). However, the effect of medical treatment for JIA on skeletal growth in affected children has not been properly investigated. We, therefore, hypothesize that naproxen and methotrexate (MTX) affect endochondral ossification and will lead to reduced skeletal development. Treatment of ATDC5 cells, an in vitro model for endochondral ossification, with naproxen or MTX resulted in increased chondrogenic but decreased hypertrophic differentiation. In vivo, healthy growing C57BL/6 mice were treated with naproxen, MTX, or placebo for 10 weeks. At 15 weeks postnatal, both the length of the tibia and the length of the femur were significantly reduced in the naproxen- and MTX-treated mice compared to their controls. Growth plate analysis revealed a significantly thicker proliferative zone, while the hypertrophic zone was significantly thinner in both experimental groups compared to their controls, comparable to the in vitro results. Micro-computed tomography analysis of the subchondral bone region directly below the growth disc showed significantly altered bone microarchitecture in naproxen and MTX groups. In addition, the involvement of the PTHrP-Ihh loop in naproxen- and MTX-treated cells was shown. Overall, these results demonstrate that naproxen and MTX have a profound effect on endochondral ossification during growth plate development, abnormal subchondral bone morphology, and reduced bone length. A better understanding of how medication influences the development of the growth plate will improve understanding of endochondral ossification and reveal possibilities to improve the treatment of pediatric patients.
Collapse
Affiliation(s)
- Marjolein M. J. Caron
- Department of Orthopaedic Surgery, CAPHRI Care and Public Health Research InstituteMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Bert van Rietbergen
- Department of Orthopaedic Surgery, CAPHRI Care and Public Health Research InstituteMaastricht University Medical CenterMaastrichtThe Netherlands
- Orthopaedic Biomechanics, Department of Biomedical EngineeringEindhoven University of TechnologyEindhovenThe Netherlands
| | | | - Mirella J. J. Haartmans
- Department of Orthopaedic Surgery, CAPHRI Care and Public Health Research InstituteMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Lodewijk W. van Rhijn
- Department of Orthopaedic Surgery, CAPHRI Care and Public Health Research InstituteMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Tim J. M. Welting
- Department of Orthopaedic Surgery, CAPHRI Care and Public Health Research InstituteMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Adhiambo M. A. Witlox
- Department of Orthopaedic Surgery, CAPHRI Care and Public Health Research InstituteMaastricht University Medical CenterMaastrichtThe Netherlands
| |
Collapse
|
18
|
Dong X, Xu X, Yang C, Luo Y, Wu Y, Wang J. USP7 regulates the proliferation and differentiation of ATDC5 cells through the Sox9-PTHrP-PTH1R axis. Bone 2021; 143:115714. [PMID: 33127578 DOI: 10.1016/j.bone.2020.115714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/25/2020] [Accepted: 10/26/2020] [Indexed: 10/23/2022]
Abstract
This study aimed to examine the effect of ubiquitin-specific peptidase 7 (USP7) on the proliferation and differentiation of ATDC5 cells and explore the underlying mechanisms. PCR, western blot, and immunofluorescence staining were used to observe the expression of USP7 after chondrogenic induction. The expressions of markers of chondrogenic and hypertrophic differentiation, and parathyroid hormone-related protein (PTHrP)/parathyroid hormone 1 receptor (PTH1R) signalling, were assessed by PCR, western blot, and histological staining under USP7 knockdown or its inhibitor. Cell proliferation was assessed by the CCK-8 assay and crystal violet staining. An in vivo experiment was performed to verify the functions of USP7 through histological and immunohistochemistry staining. Cyclopamine and abaloparatide were used to verify the signalling pathway. The interactions between USP7 and both PTHrP and sex-determining region Y-box 9 (Sox9) were tested by co-immunoprecipitation. The relationship between Sox9 and PTHrP was tested by chromatin immunoprecipitation and siRNA. USP7 knockdown or its inhibitor suppressed cell proliferation and chondrogenic differentiation but improved hypertrophic differentiation. The in vivo study obtained the same results. USP7 knockdown or its inhibitor inhibited PTHrP/PTH1R signalling to exert its function. Supplementation with cyclopamine suppressed PTHrP/PTH1R signalling and inhibited ATDC5 cell proliferation and differentiation. Supplementation with abaloparatide activated PTH1R to upregulate proliferation and chondrogenic differentiation but downregulated hypertrophic differentiation. Furthermore, USP7 interacted with Sox9 and Sox9 bound to PTTHrP to promote its expression. In conclusion, USP7 modulates the proliferation and differentiation of ATDC5 cells via the Sox9-PTHrP-PTH1R axis.
Collapse
Affiliation(s)
- Xiaofei Dong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Xiaoxiao Xu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Chang Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Yao Luo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Yanru Wu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China
| | - Jiawei Wang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Wuhan, Hubei 430079, China.
| |
Collapse
|
19
|
Zhang FY, Zhen YF, Guo ZX, Dai J, Zhu LQ, Liang PR, Su GH, Zhang WY, Fang JF, Yuan QW, Yao F, Liu Y, Qiao Y, Zhang Y, Guo WL, Liu Y, Wang XD. miR-143 is implicated in growth plate injury by targeting IHH in precartilaginous stem cells. Int J Med Sci 2021; 18:1999-2007. [PMID: 33850470 PMCID: PMC8040405 DOI: 10.7150/ijms.46474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 02/02/2021] [Indexed: 11/24/2022] Open
Abstract
Precartilaginous stem cells (PCSCs) are able to initiate chondrocyte and bone development. The present study aimed to investigate the role of miR-143 and the underlying mechanisms involved in PCSC proliferation. In a rat growth plate injury model, tissue from the injury site was collected and the expression of miR-143 and its potential targets was determined. PCSCs were isolated from the rabbits' distal epiphyseal growth plate. Cell viability, DNA synthesis, and apoptosis were determined with MTT, BrdU, and flow cytometric analysis, respectively. Real time PCR and western blot were performed to detect the mRNA and protein expression of the indicated genes. Indian hedgehog (IHH) was identified as a target gene for miR-143 with luciferase reporter assay. Decreased expression of miR-143 and increased expression of IHH gene were observed in the growth plate after injury. miR-143 mimics decreased cell viability and DNA synthesis and promoted apoptosis of PCSCs. Conversely, siRNA-mediated inhibition of miR-143 led to increased growth and suppressed apoptosis of PCSCs. Transfection of miR-143 decreased luciferase activity of wild-type IHH but had no effect when the 3'-UTR of IHH was mutated. Furthermore, the effect of miR-143 overexpression was neutralized by overexpression of IHH. Our study showed that miR-143 is involved in growth plate behavior and regulates PCSC growth by targeting IHH, suggesting that miR-143 may serve as a novel target for PCSC-related diseases.
Collapse
Affiliation(s)
- Fu-Yong Zhang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yun-Fang Zhen
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Zhi-Xiong Guo
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Jin Dai
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Lun-Qing Zhu
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Pei-Rong Liang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Guang-Hao Su
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Wen-Yan Zhang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Jian-Feng Fang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Quan-Wen Yuan
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Feng Yao
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Ya Liu
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yi Qiao
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Ya Zhang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Wan-Liang Guo
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Yao Liu
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| | - Xiao-Dong Wang
- Department of Orthopaedics, Children's Hospital of Soochow University, Suzhou, 215000, China
| |
Collapse
|
20
|
Masutani T, Yamada S, Hara A, Takahashi T, Green PG, Niwa M. Exogenous Application of Proteoglycan to the Cell Surface Microenvironment Facilitates to Chondrogenic Differentiation and Maintenance. Int J Mol Sci 2020; 21:ijms21207744. [PMID: 33086766 PMCID: PMC7589071 DOI: 10.3390/ijms21207744] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/10/2020] [Accepted: 10/14/2020] [Indexed: 11/16/2022] Open
Abstract
Osteoarthritis (OA), a disease that greatly impacts quality of life, has increasing worldwide prevalence as the population ages. However, its pathogenic mechanisms have not been fully elucidated and current therapeutic treatment strategies are inadequate. In recent years, abnormal endochondral ossification in articular cartilage has received attention as a pathophysiological mechanism in OA. Cartilage is composed of abundant extracellular matrix components, which are involved in tissue maintenance and regeneration, but how these factors affect endochondral ossification is not clear. Here, we show that the application of aggrecan-type proteoglycan from salmon nasal cartilage (sPG) exhibited marked proliferative capacity through receptor tyrosine kinases in chondroprogenitor cells, and also exhibited differentiation and three-dimensional structure formation via phosphorylation of Insulin-like Growth Factor-1 Receptor and Growth Differentiation Factor 5 expression. Furthermore, sPG inhibited calcification via expression of Runx2 and Col10 (factors related to induction of calcification), while increasing Mgp, a mineralization inhibitory factor. As a result of analyzing the localization of sPG applied to the cells, it was localized on the surface of the cell membrane. In this study, we found that sPG, as a biomaterial, could regulate cell proliferation, differentiation and calcification inhibition by acting on the cell surface microenvironment. Therefore, sPG may be the foundation for a novel therapeutic approach for cartilage maintenance and for improved symptoms in OA.
Collapse
Affiliation(s)
- Teruaki Masutani
- Medical Education Development Center, Gifu University School of Medicine, 1-1 Yanagido, Gifu City 501-1194, Japan;
- Research & Development Dept., Ichimaru Pharcos Co., Ltd., 318-1 Asagi, Motosu City 501-0475, Japan;
| | - Shuhei Yamada
- Department of Pathobiochemistry, Faculty of Pharmacy, Meijo University, 150 Yagotoyama, Tempaku-ku, Nagoya 468-8503, Japan;
| | - Akira Hara
- Department of Tumor Pathology, Gifu University Graduate School of Medicine, Gifu City 501-1194, Japan;
| | - Tatsuji Takahashi
- Research & Development Dept., Ichimaru Pharcos Co., Ltd., 318-1 Asagi, Motosu City 501-0475, Japan;
| | - Paul G Green
- Department of Oral and Maxillofacial Surgery, UCSF, San Francisco, CA 94143, USA;
| | - Masayuki Niwa
- Medical Education Development Center, Gifu University School of Medicine, 1-1 Yanagido, Gifu City 501-1194, Japan;
- Correspondence: ; Tel.: +81-58-230-6470
| |
Collapse
|
21
|
Neefjes M, van Caam APM, van der Kraan PM. Transcription Factors in Cartilage Homeostasis and Osteoarthritis. BIOLOGY 2020; 9:biology9090290. [PMID: 32937960 PMCID: PMC7563835 DOI: 10.3390/biology9090290] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/07/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease, and it is characterized by articular cartilage loss. In part, OA is caused by aberrant anabolic and catabolic activities of the chondrocyte, the only cell type present in cartilage. These chondrocyte activities depend on the intra- and extracellular signals that the cell receives and integrates into gene expression. The key proteins for this integration are transcription factors. A large number of transcription factors exist, and a better understanding of the transcription factors activated by the various signaling pathways active during OA can help us to better understand the complex etiology of OA. In addition, establishing such a profile can help to stratify patients in different subtypes, which can be a very useful approach towards personalized therapy. In this review, we discuss crucial transcription factors for extracellular matrix metabolism, chondrocyte hypertrophy, chondrocyte senescence, and autophagy in chondrocytes. In addition, we discuss how insight into these factors can be used for treatment purposes.
Collapse
|
22
|
Salhotra A, Shah HN, Levi B, Longaker MT. Mechanisms of bone development and repair. Nat Rev Mol Cell Biol 2020; 21:696-711. [PMID: 32901139 DOI: 10.1038/s41580-020-00279-w] [Citation(s) in RCA: 467] [Impact Index Per Article: 116.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/23/2020] [Indexed: 12/19/2022]
Abstract
Bone development occurs through a series of synchronous events that result in the formation of the body scaffold. The repair potential of bone and its surrounding microenvironment - including inflammatory, endothelial and Schwann cells - persists throughout adulthood, enabling restoration of tissue to its homeostatic functional state. The isolation of a single skeletal stem cell population through cell surface markers and the development of single-cell technologies are enabling precise elucidation of cellular activity and fate during bone repair by providing key insights into the mechanisms that maintain and regenerate bone during homeostasis and repair. Increased understanding of bone development, as well as normal and aberrant bone repair, has important therapeutic implications for the treatment of bone disease and ageing-related degeneration.
Collapse
Affiliation(s)
- Ankit Salhotra
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Harsh N Shah
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Benjamin Levi
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA.
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA. .,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
23
|
Svandova E, Anthwal N, Tucker AS, Matalova E. Diverse Fate of an Enigmatic Structure: 200 Years of Meckel's Cartilage. Front Cell Dev Biol 2020; 8:821. [PMID: 32984323 PMCID: PMC7484903 DOI: 10.3389/fcell.2020.00821] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Meckel's cartilage was first described by the German anatomist Johann Friedrich Meckel the Younger in 1820 from his analysis of human embryos. Two hundred years after its discovery this paper follows the development and largely transient nature of the mammalian Meckel's cartilage, and its role in jaw development. Meckel's cartilage acts as a jaw support during early development, and a template for the later forming jaw bones. In mammals, its anterior domain links the two arms of the dentary together at the symphysis while the posterior domain ossifies to form two of the three ear ossicles of the middle ear. In between, Meckel's cartilage transforms to a ligament or disappears, subsumed by the growing dentary bone. Several human syndromes have been linked, directly or indirectly, to abnormal Meckel's cartilage formation. Herein, the evolution, development and fate of the cartilage and its impact on jaw development is mapped. The review focuses on developmental and cellular processes that shed light on the mechanisms behind the different fates of this cartilage, examining the control of Meckel's cartilage patterning, initiation and maturation. Importantly, human disorders and mouse models with disrupted Meckel's cartilage development are highlighted, in order to understand how changes in this cartilage impact on later development of the dentary and the craniofacial complex as a whole. Finally, the relative roles of tissue interactions, apoptosis, autophagy, macrophages and clast cells in the removal process are discussed. Meckel's cartilage is a unique and enigmatic structure, the development and function of which is starting to be understood but many interesting questions still remain.
Collapse
Affiliation(s)
- Eva Svandova
- Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia
| | - Neal Anthwal
- Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Abigail S. Tucker
- Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Eva Matalova
- Institute of Animal Physiology and Genetics, Academy of Sciences, Brno, Czechia
- Department of Physiology, University of Veterinary and Pharmaceutical Sciences, Brno, Czechia
| |
Collapse
|
24
|
Amano K, Okuzaki D, Aikawa T, Kogo M. Indian hedgehog in craniofacial neural crest cells links to skeletal malocclusion by regulating associated cartilage formation and gene expression. FASEB J 2020; 34:6791-6807. [PMID: 32223017 DOI: 10.1096/fj.201903269r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 12/29/2022]
Abstract
The frontal craniofacial skeleton derived from neural crest cells is vital for facial structure and masticatory functions. The exact role of Indian hedgehog (Ihh) in facial and masticatory development has not been fully explored. In this study, we generated craniofacial neural crest cells-specific Ihh deletion mice (Wnt1-Cre;Ihhfl/fl ;Tomatofl/+ ) and found the gradual dwarfism without perinatal lethality. Morphological and histological analyses revealed unambiguous craniofacial phenotypes in mutants, where we observed skeletal malocclusion accompanied by markedly hypoplastic nasomaxillary complex and reversed incisor occlusion. Both the replacement of nasal concha cartilage by turbinate bones and the endochondral ossification of nasal septum ethmoid bone were substantially delayed. We also observed hypoplastic mandibles in mutants where the mandibular ramus was unexpectedly the most affected. Both the condylar process and mandibular angle cartilages were distorted. However, dental examination showed no significant changes in teeth and dentition. Finally, a comprehensive RNA sequence analysis utilizing condylar cartilage identified Ihh-associated gene network including several cell cycle genes and 16 genes related to the extracellular matrix, sulfate transporters, transcription factors, receptors, a ciliogenesis factor, and an adhesion molecule. Our data provide direct in vivo evidence that Ihh plays crucial roles in midface and masticatory system formation, likely by activating key genes.
Collapse
Affiliation(s)
- Katsuhiko Amano
- The First Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Daisuke Okuzaki
- Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | - Tomonao Aikawa
- The First Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Japan
| | - Mikihiko Kogo
- The First Department of Oral and Maxillofacial Surgery, Osaka University Graduate School of Dentistry, Suita, Japan
| |
Collapse
|
25
|
Chou L, Chen C, Lin Y, Chuang S, Chou H, Lin S, Fu Y, Chang J, Ho M, Wang C. Discoidin domain receptor 1 regulates endochondral ossification through terminal differentiation of chondrocytes. FASEB J 2020; 34:5767-5781. [DOI: 10.1096/fj.201901852rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 02/12/2020] [Accepted: 02/18/2020] [Indexed: 01/27/2023]
Affiliation(s)
- Liang‐Yin Chou
- Graduate Institute of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
| | - Chung‐Hwan Chen
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics Kaohsiung Municipal Ta‐Tung Hospital Kaohsiung Medical University Kaohsiung Taiwan
- Institute of Medical Science and Technology National Sun Yat‐Sen University Kaohsiung Taiwan
| | - Yi‐Hsiung Lin
- Department of Biotechnology Kaohsiung Medical University Kaohsiung Taiwan
- Division of Cardiology Department of Internal Medicine Kaohsiung Medical University Hospital Kaohsiung Taiwan
- Lipid Science and Aging Research Center Kaohsiung Medical University Kaohsiung Taiwan
| | - Shu‐Chun Chuang
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
| | - Hsin‐Chiao Chou
- Graduate Institute of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
| | - Sung‐Yen Lin
- Graduate Institute of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics Kaohsiung Municipal Ta‐Tung Hospital Kaohsiung Medical University Kaohsiung Taiwan
| | - Yin‐Chi Fu
- Graduate Institute of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
| | - Je‐Ken Chang
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics Kaohsiung Municipal Ta‐Tung Hospital Kaohsiung Medical University Kaohsiung Taiwan
- Division of Adult Reconstruction Surgery Department of Orthopedics Kaohsiung Medical University Hospital Kaohsiung Medical University Kaohsiung Taiwan
| | - Mei‐Ling Ho
- Graduate Institute of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
- Department of Orthopedics College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Department of Physiology College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
| | - Chau‐Zen Wang
- Graduate Institute of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Orthopaedic Research Centre Kaohsiung Medical University Kaohsiung Taiwan
- Regeneration Medicine and Cell Therapy Research Center Kaohsiung Medical University Kaohsiung Taiwan
- Department of Physiology College of Medicine Kaohsiung Medical University Kaohsiung Taiwan
- Department of Medical Research Kaohsiung Medical University Hospital Kaohsiung Taiwan
| |
Collapse
|
26
|
Liu Z, Ramachandran J, Vokes SA, Gray RS. Regulation of terminal hypertrophic chondrocyte differentiation in Prmt5 mutant mice modeling infantile idiopathic scoliosis. Dis Model Mech 2019; 12:dmm.041251. [PMID: 31848143 PMCID: PMC6955203 DOI: 10.1242/dmm.041251] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/18/2019] [Indexed: 12/20/2022] Open
Abstract
Idiopathic scoliosis (IS) is the most common type of musculoskeletal defect affecting children worldwide, and is classified by age of onset, location and degree of spine curvature. Although rare, IS with onset during infancy is the more severe and rapidly progressive form of the disease, associated with increased mortality due to significant respiratory compromise. The pathophysiology of IS, in particular for infantile IS, remains elusive. Here, we demonstrate the role of PRMT5 in the infantile IS phenotype in mouse. Conditional genetic ablation of PRMT5 in osteochondral progenitors results in impaired terminal hypertrophic chondrocyte differentiation and asymmetric defects of endochondral bone formation in the perinatal spine. Analysis of these several markers of endochondral ossification revealed increased type X collagen (COLX) and Ihh expression, coupled with a dramatic reduction in Mmp13 and RUNX2 expression, in the vertebral growth plate and in regions of the intervertebral disc in the Prmt5 conditional mutant mice. We also demonstrate that PRMT5 has a continuous role in the intervertebral disc and vertebral growth plate in adult mice. Altogether, our results establish PRMT5 as a critical promoter of terminal hypertrophic chondrocyte differentiation and endochondral bone formation during spine development and homeostasis. This article has an associated First Person interview with the first author of the paper. Summary: Loss of Prmt5 in osteochondral progenitors impairs terminal hypertrophic chondrocyte differentiation, leading to defects in endochondral bone formation and models infantile idiopathic scoliosis in mouse.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, TX 78723, USA
| | - Janani Ramachandran
- Department of Molecular Biosciences, 2500 Speedway, The University of Texas at Austin, Austin, TX 78712, USA
| | - Steven A Vokes
- Department of Molecular Biosciences, 2500 Speedway, The University of Texas at Austin, Austin, TX 78712, USA
| | - Ryan S Gray
- Department of Pediatrics, Dell Pediatric Research Institute, 1400 Barbara Jordan Blvd, The University of Texas at Austin, Dell Medical School, Austin, TX 78723, USA .,Department of Nutritional Sciences, 200 W 24th Street, The University of Texas at Austin, Austin, TX 78712, USA
| |
Collapse
|
27
|
Long HQ, Tian PF, Guan YX, Liu LX, Wu XP, Li B. Expression of Ihh signaling pathway in condylar cartilage after bite-raising in adult rats. J Mol Histol 2019; 50:459-470. [PMID: 31302828 DOI: 10.1007/s10735-019-09840-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/09/2019] [Indexed: 01/09/2023]
Abstract
Temporomandibular joint osteoarthritis (TMJOA) is a complex inflammatory condition with multiple factors and degenerative processes co-occurring. However, its pathogenesis remains uncertain. The purpose of the study was to observe the expression of Indian hedgehog (Ihh) signal related molecules in TMJOA induced by bite-raising and to study the effect and mechanism of Ihh signaling. Our research indicated that Ihh signaling pathway can be activated in condylar cartilage induced by bite-raising. The histological analysis showed TMJOA-like structural changes of condylar cartilage in experiment groups. Ihh, Smoothened (Smo), and Gli zinc finger transcription factors-1 (Gli-1) were activated in the experimental groups, and the expression levels increased significantly over time, whereas the sham control groups showed no fluctuation. Additionally, the expression levels of matrix metalloproteinase-13 (MMP-13) and cysteinyl aspartate specific proteinase-3 (Caspase-3) in the experiment groups increased in a time-dependent manner compared with the matched sham control groups. In conclusion, our results indicated that the Ihh signaling pathway may activate the occurrence of TMJOA by mediating the hypertrophy of chondrocytes, which may be an important regulatory mechanism and potential therapeutic target in the repair of condylar cartilage.
Collapse
Affiliation(s)
- Hui-Qing Long
- Department of Orthodontics, School of Dentistry, Shanxi Medical University, Taiyuan, China
| | - Peng-Fei Tian
- Department of Orthodontics, School of Dentistry, Shanxi Medical University, Taiyuan, China
| | - Yu-Xin Guan
- Department of Orthodontics, School of Dentistry, Shanxi Medical University, Taiyuan, China
| | - Ling-Xia Liu
- Department of Orthodontics, School of Dentistry, Shanxi Medical University, Taiyuan, China
| | - Xiu-Ping Wu
- Department of Orthodontics, School of Dentistry, Shanxi Medical University, Taiyuan, China.
| | - Bing Li
- Department of Orthodontics, School of Dentistry, Shanxi Medical University, Taiyuan, China
| |
Collapse
|
28
|
Zhang B, Xu H, Zhang Y, Yi X, Zhang G, Zhang X, Xu D, Gao X, Li S, Zhu Y, Zhang H, Wei Z, Li S, Zhang L, Wang R, Yang F. Targeting the RAS axis alleviates silicotic fibrosis and Ang II-induced myofibroblast differentiation via inhibition of the hedgehog signaling pathway. Toxicol Lett 2019; 313:30-41. [PMID: 31181250 DOI: 10.1016/j.toxlet.2019.05.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/22/2019] [Accepted: 05/31/2019] [Indexed: 01/01/2023]
Abstract
The hedgehog (HH) signaling pathway plays an important role in lung development, but its significance in silicosis is unclear. We showed that in human coal pneumoconiosis autopsy specimens, Sonic Hedgehog (SHH) and the Glioma-associated oncogene homolog transcription factors family (GLI) 1 proteins were up-regulated, whereas Patch-1 (PTC) was down-regulated. The protein levels of SHH, smoothened (SMO), GLI1, GLI2, α-smooth muscle actin (α-SMA) and collagen type Ⅰ (Col Ⅰ) were also elevated gradually in the bronchoalveolar lavage fluid (BALF) of different stages of coal pneumoconiosis patients, dynamic silica-inhalation rat lung tissue and MRC-5 cells induced by Ang II at different time points, whereas the PTC and GLI3 levels were diminished gradually. Ac-SDKP, an active peptide of renin-angiotensin system (RAS), is an anti-fibrotic tetrapeptide. Targeting RAS axis also has anti-silicotic fibrosis effects. However, their roles on the HH pathway are still unknown. Here, we reported that Ac-SDKP + Captopril, Ac-SDKP, Captopril, or Ang (1-7) could alleviate silicotic fibrosis and collagen deposition, as well as improve the lung functions of silicotic rat. These treatments decreased the expression of SHH, SMO, GLI1, GLI2, α-SMA, and Col Ⅰ and increased the expression of PTC and GLI3 on both the silicotic rat lung tissue and MRC-5 cells induced by Ang II. We also reported that Ang II may promote myofibroblast differentiation via the GLI1 transcription factor and independently of the SMO receptor.
Collapse
Affiliation(s)
- Bonan Zhang
- School of Public Health, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Clinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Hong Xu
- Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Yi Zhang
- Clinical Medical College, North China University of Science and Technology, Tangshan, China
| | - Xue Yi
- Key Laboratory of Functional and Clinical Translational Medicine, Fujian Province University, Department of Basic Medicine, Xiamen Medical College, Xiamen, China
| | - Guizhen Zhang
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Clinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Xin Zhang
- Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Dingjie Xu
- College of Traditional Chinese Medicine, North China University of Science and Technology, Tangshan, China
| | - Xuemin Gao
- Basic Medical College, Hebei Medical University, Shijiazhuang, China
| | - Shifeng Li
- Basic Medical College, Hebei Medical University, Shijiazhuang, China
| | - Ying Zhu
- School of Public Health, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Hui Zhang
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Clinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Zhongqiu Wei
- Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Clinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Shumin Li
- School of Public Health, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Chronic Diseases, Tangshan Key Laboratory for Clinical and Basic Research on Chronic Diseases, School of Basic Medical Sciences, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Lijuan Zhang
- Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Ruimin Wang
- School of Public Health, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China
| | - Fang Yang
- School of Public Health, North China University of Science and Technology, Tangshan, China; Hebei Key Laboratory for Organ Fibrosis, Medical Research Center, North China University of Science and Technology, Tangshan, China.
| |
Collapse
|
29
|
Lima-Fernandes E, Murison A, da Silva Medina T, Wang Y, Ma A, Leung C, Luciani GM, Haynes J, Pollett A, Zeller C, Duan S, Kreso A, Barsyte-Lovejoy D, Wouters BG, Jin J, Carvalho DDD, Lupien M, Arrowsmith CH, O'Brien CA. Targeting bivalency de-represses Indian Hedgehog and inhibits self-renewal of colorectal cancer-initiating cells. Nat Commun 2019; 10:1436. [PMID: 30926792 PMCID: PMC6441108 DOI: 10.1038/s41467-019-09309-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 02/27/2019] [Indexed: 12/21/2022] Open
Abstract
In embryonic stem cells, promoters of key lineage-specific differentiation genes are found in a bivalent state, having both activating H3K4me3 and repressive H3K27me3 histone marks, making them poised for transcription upon loss of H3K27me3. Whether cancer-initiating cells (C-ICs) have similar epigenetic mechanisms that prevent lineage commitment is unknown. Here we show that colorectal C-ICs (CC-ICs) are maintained in a stem-like state through a bivalent epigenetic mechanism. Disruption of the bivalent state through inhibition of the H3K27 methyltransferase EZH2, resulted in decreased self-renewal of patient-derived C-ICs. Epigenomic analyses revealed that the promoter of Indian Hedgehog (IHH), a canonical driver of normal colonocyte differentiation, exists in a bivalent chromatin state. Inhibition of EZH2 resulted in de-repression of IHH, decreased self-renewal, and increased sensitivity to chemotherapy in vivo. Our results reveal an epigenetic block to differentiation in CC-ICs and demonstrate the potential for epigenetic differentiation therapy of a solid tumour through EZH2 inhibition.
Collapse
Affiliation(s)
- Evelyne Lima-Fernandes
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G1L7, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Tiago da Silva Medina
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Yadong Wang
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Anqi Ma
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Cherry Leung
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Genna M Luciani
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G1L7, Canada.,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Jennifer Haynes
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Aaron Pollett
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S1A8, Canada.,Lunenfeld-Tanenbaum Research Institute Toronto, Toronto, ON, M5G1X5, Canada
| | - Constanze Zeller
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | - Shili Duan
- Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada
| | - Antonija Kreso
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada
| | | | - Bradly G Wouters
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada.,Department of Radiation Oncology, University of Toronto, Toronto, ON, M5G1L7, Canada
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Daniel D De Carvalho
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada
| | - Mathieu Lupien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada.,Ontario Institute for Cancer Research, Toronto, ON, M5G1L7, Canada
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto, Toronto, ON, M5G1L7, Canada. .,Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada.
| | - Catherine A O'Brien
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, M5G1L7, Canada. .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S1A8, Canada. .,Department of Medical Biophysics, University of Toronto, Toronto, ON, M5G1L7, Canada. .,Department of Physiology, University of Toronto, Toronto, ON, M5G1L7, Canada. .,Department of Surgery, Toronto General Hospital, Toronto, ON, M5G2C4, Canada.
| |
Collapse
|
30
|
Ali SA, Niu B, Cheah KSE, Alman B. Unique and overlapping GLI1 and GLI2 transcriptional targets in neoplastic chondrocytes. PLoS One 2019; 14:e0211333. [PMID: 30695055 PMCID: PMC6350985 DOI: 10.1371/journal.pone.0211333] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 01/11/2019] [Indexed: 01/11/2023] Open
Abstract
Excessive Hedgehog (Hh) signaling in chondrocytes is sufficient to cause formation of enchondroma-like lesions which can progress to chondrosarcoma. To elucidate potential underlying mechanisms, we identified GLI1 and GLI2 target genes in human chondrosarcoma. Using chromatin immunoprecipitation (ChIP) sequencing and microarray data, in silico analyses were conducted to identify and characterize unique and overlapping GLI1 and GLI2 binding regions in neoplastic chondrocytes. After overlaying microarray data from human chondrosarcoma, 204 upregulated and 106 downregulated genes were identified as Hh-responsive Gli binding targets. After overlaying published Gli ChIP-on-chip data from mouse, 48 genes were identified as potential direct downstream targets of Hedgehog signaling with shared GLI binding regions in evolutionarily conserved DNA elements. Among these was BMP2, pointing to potential cross-talk between TGF beta signaling and Hh signaling. Our identification of potential target genes that are unique and common to GLI1 and GLI2 in neoplastic chondrocytes contributes to elucidating potential pathways through which Hh signaling impacts cartilage tumor biology.
Collapse
Affiliation(s)
- Shabana Amanda Ali
- Genetics and Development, Krembil Research Institute, Toronto, Ontario, Canada
| | - Ben Niu
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Kathryn S. E. Cheah
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Benjamin Alman
- Department of Orthopaedic Surgery, Duke University, Durham, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
31
|
Moeinzadeh S, Monavarian M, Kader S, Jabbari E. Sequential Zonal Chondrogenic Differentiation of Mesenchymal Stem Cells in Cartilage Matrices. Tissue Eng Part A 2018; 25:234-247. [PMID: 30146939 DOI: 10.1089/ten.tea.2018.0083] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
IMPACT STATEMENT The higher regenerative capacity of fetal articular cartilage compared with the adult is rooted in differences in cell density and matrix composition. We hypothesized that the zonal organization of articular cartilage can be engineered by encapsulation of mesenchymal stem cells in a single superficial zone-like matrix followed by sequential addition of zone-specific growth factors within the matrix, similar to the process of fetal cartilage development. The results demonstrate that the zonal organization of articular cartilage can potentially be regenerated using an injectable, monolayer cell-laden hydrogel with sequential release of growth factors.
Collapse
Affiliation(s)
- Seyedsina Moeinzadeh
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| | - Mehri Monavarian
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| | - Safaa Kader
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina.,2 Department of Chemistry and Biochemistry, University of South Carolina, Columbia, South Carolina
| | - Esmaiel Jabbari
- 1 Biomimetic Materials and Tissue Engineering Laboratory, Department of Chemical Engineering, University of South Carolina, Columbia, South Carolina
| |
Collapse
|
32
|
Kitami M, Yamaguchi H, Ebina M, Kaku M, Chen D, Komatsu Y. IFT20 is required for the maintenance of cartilaginous matrix in condylar cartilage. Biochem Biophys Res Commun 2018; 509:222-226. [PMID: 30587338 DOI: 10.1016/j.bbrc.2018.12.107] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/16/2022]
Abstract
Condylar cartilage is a joint cartilage essential for smooth jaw movement. The importance of ciliary proteins in condylar cartilage development has been reported. However, little is known about how ciliary proteins control the homeostasis of condylar cartilage. Here we show that intraflagellar transport 20 (IFT20), a ciliary protein, is required for the maintenance of cartilaginous matrix in condylar cartilage. Utilizing NG2-CreER mice expressed in condylar cartilage, we deleted Ift20 by tamoxifen treatment at juvenile-to-adult stages. In wild-type condylar cartilage, IFT20 was robustly produced in the cis-Golgi, but deletion of Ift20 by tamoxifen induction of NG2-CreER (Ift20:NG2-CreER) resulted in reduced cell proliferation and decreased Golgi size in condylar cartilage. Importantly, while the primary cilia were present in cartilage cells in the condylar layers of wild-type mice, no primary cilia were present in the Ift20:NG2-CreER condylar layers. Consistent with this finding, ciliary-mediated Hedgehog signaling was severely attenuated in Ift20 mutant chondrocytes, and thus the production levels of type X collagen were significantly reduced in Ift20:NG2-CreER mice. These results suggest that IFT20 is required for Golgi size and Hedgehog signaling to maintain cartilaginous matrix in condylar cartilage. Our study highlights the unique function of IFT20 in the homeostasis of condylar cartilage.
Collapse
Affiliation(s)
- Megumi Kitami
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Hiroyuki Yamaguchi
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Masayuki Ebina
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA
| | - Masaru Kaku
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA; Division of Bioprosthodontics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 951-8514, Japan
| | - Di Chen
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yoshihiro Komatsu
- Department of Pediatrics, The University of Texas Medical School at Houston, Houston, TX, 77030, USA; Graduate Program in Genetics and Epigenetics, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, TX, 77030, USA.
| |
Collapse
|
33
|
Izawa T, Hutami IR, Tanaka E. Potential Role of Rebamipide in Osteoclast Differentiation and Mandibular Condylar Cartilage Homeostasis. Curr Rheumatol Rev 2018; 14:62-69. [PMID: 29046162 PMCID: PMC5925868 DOI: 10.2174/1573397113666171017113441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 09/23/2017] [Accepted: 09/28/2017] [Indexed: 12/27/2022]
Abstract
Background: Temporomandibular joint osteoarthritis (TMJ-OA) is a degenerative disease that involves changes in subchondral bone and progressive degradation of cartilage. Currently, rebamipide, a gastroprotective drug, is administered to protect gastric mucosa and accelerate ulcer healing. Objectives: Recent studies have shown that rebamipide also attenuates cartilage degeneration by suppressing oxidative damage and inducing homeostasis of the extracellular matrix of articular chondrocytes. Regarding the latter, reduced expression of cathepsin K, NFATc1, c-Src, and integrin β3, and increased expression of nuclear factor-kappa B, have been found to be mediated by the transcription factor, receptor activator of nuclear factor kappa-B ligand (RANKL). Methods: Treatment with rebamipide was also found to activate, mitogen-activated protein kinases such as p38, ERK, and JNK to reduce osteoclast differentiation. Taken together, these results strongly indicate that rebamipide mediates inhibitory effects on cartilage degradation and osteoclastogenesis in TMJ-OA. Results and Conclusion: Here, we highlight recent evidence regarding the potential for rebamipide to affect osteoclast differentiation and TMJ-OA pathogenesis. We also discuss the potential role of rebamipide to serve as a new strategy for the treatment of TMJ-OA.
Collapse
Affiliation(s)
- Takashi Izawa
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima 7708504, Japan
| | - Islamy Rahma Hutami
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima 7708504, Japan
| | - Eiji Tanaka
- Department of Orthodontics and Dentofacial Orthopedics, Tokushima University Graduate School of Biomedical Sciences, 3-18-15 Kuramoto-cho, Tokushima 7708504, Japan
| |
Collapse
|
34
|
Tan Z, Niu B, Tsang KY, Melhado IG, Ohba S, He X, Huang Y, Wang C, McMahon AP, Jauch R, Chan D, Zhang MQ, Cheah KSE. Synergistic co-regulation and competition by a SOX9-GLI-FOXA phasic transcriptional network coordinate chondrocyte differentiation transitions. PLoS Genet 2018; 14:e1007346. [PMID: 29659575 PMCID: PMC5919691 DOI: 10.1371/journal.pgen.1007346] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/26/2018] [Accepted: 03/29/2018] [Indexed: 11/18/2022] Open
Abstract
The growth plate mediates bone growth where SOX9 and GLI factors control chondrocyte proliferation, differentiation and entry into hypertrophy. FOXA factors regulate hypertrophic chondrocyte maturation. How these factors integrate into a Gene Regulatory Network (GRN) controlling these differentiation transitions is incompletely understood. We adopted a genome-wide whole tissue approach to establish a Growth Plate Differential Gene Expression Library (GP-DGEL) for fractionated proliferating, pre-hypertrophic, early and late hypertrophic chondrocytes, as an overarching resource for discovery of pathways and disease candidates. De novo motif discovery revealed the enrichment of SOX9 and GLI binding sites in the genes preferentially expressed in proliferating and prehypertrophic chondrocytes, suggesting the potential cooperation between SOX9 and GLI proteins. We integrated the analyses of the transcriptome, SOX9, GLI1 and GLI3 ChIP-seq datasets, with functional validation by transactivation assays and mouse mutants. We identified new SOX9 targets and showed SOX9-GLI directly and cooperatively regulate many genes such as Trps1, Sox9, Sox5, Sox6, Col2a1, Ptch1, Gli1 and Gli2. Further, FOXA2 competes with SOX9 for the transactivation of target genes. The data support a model of SOX9-GLI-FOXA phasic GRN in chondrocyte development. Together, SOX9-GLI auto-regulate and cooperate to activate and repress genes in proliferating chondrocytes. Upon hypertrophy, FOXA competes with SOX9, and control toward terminal differentiation passes to FOXA, RUNX, AP1 and MEF2 factors. In the development of the mammalian growth plate, while several transcription factors are individually well known for their key roles in regulating phases of chondrocyte differentiation, there is little information on how they interact and cooperate with each other. We took an unbiased genome wide approach to identify the transcription factors and signaling pathways that play dominant roles in the chondrocyte differentiation cascade. We developed a searchable library of differentially expressed genes, GP-DGEL, which has fine spatial resolution and global transcriptomic coverage for discovery of processes, pathways and disease candidates. Our work identifies a novel regulatory mechanism that integrates the action of three transcription factors, SOX9, GLI and FOXA. SOX9-GLI auto-regulate and cooperate to activate and repress genes in proliferating chondrocytes. Upon entry into prehypertrophy, FOXA competes with SOX9, and control of hypertrophy passes to FOXA, RUNX, AP1 and MEF2 factors.
Collapse
Affiliation(s)
- Zhijia Tan
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Ben Niu
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Kwok Yeung Tsang
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Ian G. Melhado
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Shinsuke Ohba
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Xinjun He
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Yongheng Huang
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Cheng Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Ralf Jauch
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Danny Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Michael Q. Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Dallas, Texas, United States of America
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, TNLIST, Tsinghua University, Beijing, China
| | - Kathryn S. E. Cheah
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
- * E-mail:
| |
Collapse
|
35
|
Jensen PT, Lambertsen KL, Frich LH. Assembly, maturation, and degradation of the supraspinatus enthesis. J Shoulder Elbow Surg 2018; 27:739-750. [PMID: 29329904 DOI: 10.1016/j.jse.2017.10.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 10/22/2017] [Accepted: 10/27/2017] [Indexed: 02/01/2023]
Abstract
The development of the rotator cuff enthesis is still poorly understood. The processes in the early and late developmental steps are gradually elucidated, but it is still unclear how cell activities are coordinated during development and maturation of the structured enthesis. This review summarizes current knowledge about development and age-related degradation of the supraspinatus enthesis. Healing and repair of an injured and degenerated supraspinatus enthesis also remain a challenge, as the original graded transitional tissue of the fibrocartilaginous insertion is not re-created after the tendon is surgically reattached to bone. Instead, mechanically inferior and disorganized tissue forms at the healing site because of scar tissue formation. Consequently, the enthesis never reaches mechanical properties comparable to those of the native enthesis. So far, no novel biologic healing approach has been successful in enhancing healing of the injured enthesis. The results revealed in this review imply the need for further research to pave the way for better treatment of patients with rotator cuff disorder.
Collapse
Affiliation(s)
- Peter T Jensen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kate L Lambertsen
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lars H Frich
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Orthopaedics and Traumatology, Odense University Hospital, Odense, Denmark.
| |
Collapse
|
36
|
Thyroid Hormone Signaling in the Development of the Endochondral Skeleton. VITAMINS AND HORMONES 2018; 106:351-381. [PMID: 29407442 PMCID: PMC9830754 DOI: 10.1016/bs.vh.2017.06.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Thyroid hormone (TH) is an established regulator of skeletal growth and maintenance both in clinical studies and in laboratory models. The clinical consequences of altered thyroid status on the skeleton during development and in adulthood are well known, and genetic mouse models in which elements of the TH signaling axis have been manipulated illuminate the mechanisms which underlie TH regulation of the skeleton. TH is involved in the regulation of the balance between proliferation and differentiation in several skeletal cell types including chondrocytes, osteoblasts, and osteoclasts. The effects of TH are mediated primarily via the thyroid hormone receptors (TRs) α and β, ligand-inducible nuclear receptors which act as transcription factors to regulate target gene expression. Both TRα and TRβ signaling are important for different stages of skeletal development. The molecular mechanisms of TH action in bone are complex and include interaction with a number of growth factor signaling pathways. This review provides an overview of the regulation and mechanisms of TH action in bone, focusing particularly on the role of TH in endochondral bone formation during postnatal growth.
Collapse
|
37
|
Ren J, Ma J, Zhang X, Aimaiti A, Saiyiti M, Chen Y, Cao L. Diagnostic value of combined serum marker changes and quantitative MRI evaluation of cartilage volume of tibial plateau in a surgically-induced osteoarthritis dog model. J Int Med Res 2017; 45:2023-2035. [PMID: 29125013 PMCID: PMC5805226 DOI: 10.1177/0300060517730452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Objective To evaluate the combined diagnostic value of two serum osteoarthritis (OA)
markers and quantitative magnetic resonance imaging (MRI) evaluation of the
cartilage volume of the tibial plateau in a canine model of experimental
OA. Methods A total of 18 male Beagle dogs were used in this longitudinal study. OA was
surgically induced via anterior cruciate ligament transection (ACLT) of the
right knee in 10 dogs. The remaining eight dogs formed the sham operation
control group and underwent the same procedure without ACLT. At various
times after surgery, enzyme-linked immunosorbent assay was used to measure
serum C-telopeptide of type II collagen (CTX-II) and type X collagen (ColX)
levels. Quantitative evaluation of the tibial plateau volume was undertaken
using MRI and ImageJ software. Results The serum CTX-II levels were significantly higher in the OA group at weeks 8,
12 and 16 after surgery, but not at week 4, compared with the control group.
The serum ColX levels in the OA group were significantly higher than in the
control group at weeks 8 and 12. The tibial plateau cartilage volumes in the
OA group were significantly lower than in the control group at weeks 8 and
16. Conclusion Serum CTX-II and ColX levels combined with quantitative MRI evaluation of the
tibial plateau cartilage volume in a canine model of OA demonstrated the
potential to detect and monitor OA progression.
Collapse
Affiliation(s)
- Jiangdong Ren
- 1 Department of Joint Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jinsai Ma
- 3 Department of Orthopaedics, Changji Branch of the First Affiliated Hospital of Xinjiang Medical University, Changji, China
| | - Xiaogang Zhang
- 1 Department of Joint Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Abdusami Aimaiti
- 1 Department of Joint Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Maimaitiming Saiyiti
- 2 Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yuming Chen
- 4 Department of Pain Management, Changji Branch of the First Affiliated Hospital of Xinjiang Medical University, Changji, China
| | - Li Cao
- 1 Department of Joint Surgery, First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
38
|
Deng A, Zhang H, Hu M, Liu S, Wang Y, Gao Q, Guo C. The inhibitory roles of Ihh downregulation on chondrocyte growth and differentiation. Exp Ther Med 2017; 15:789-794. [PMID: 29434683 PMCID: PMC5772930 DOI: 10.3892/etm.2017.5458] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 11/03/2017] [Indexed: 01/05/2023] Open
Abstract
The proliferative rate of chondrocytes affects bone elongation. Chondrocyte hypertrophy is required for endochondral bone formation as chondrocytes secrete factors required for osteoblast differentiation and maturation. Previous studies have demonstrated that the Indian hedgehog (Ihh) signaling pathway is a key regulator of skeletal development and homeostasis. The aim of the present study was to investigate the function of Ihh in chondrocyte proliferation and differentiation, as well as the underlying mechanisms. Ihh was knocked down in mouse chondrocyte cells using short hairpin RNA. Chondrocyte apoptosis and cell cycle arrest were assessed using flow cytometry and the results indicated that knockdown of Ihh significantly inhibited cell growth (P<0.05) and increased apoptosis (P<0.001) compared with negative control cells. Downregulation of Ihh also resulted in cell cycle arrest at G1 to S phase in chondrocytes. It was also observed that knockdown of Ihh decreased alkaline phosphatase activity and mineral deposition of chondrocytes. The inhibitory roles of Ihh downregulation on chondrocyte growth and differentiation may be associated with the transforming growth factor-β/mothers against decapentaplegic and osteoprotegerin/receptor activator of nuclear factor κB ligand signaling pathway. The results of the present study suggest that chondrocyte-derived Ihh is essential for maintaining bone growth plates and that manipulation of Ihh expression or its signaling components may be a novel therapeutic technique for the treatment of skeletal diseases, including achondroplasia.
Collapse
Affiliation(s)
- Ang Deng
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Hongqi Zhang
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Minyu Hu
- Department of Nutrition and Food Hygiene, School of Public Health, Central South University, Changsha, Hunan 410078, P.R. China
| | - Shaohua Liu
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Yuxiang Wang
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Qile Gao
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| | - Chaofeng Guo
- Department of Spine Surgery, Xiangya Hospital of Central South University, Changsha, Hunan 410008, P.R. China
| |
Collapse
|
39
|
Aghajanian P, Xing W, Cheng S, Mohan S. Epiphyseal bone formation occurs via thyroid hormone regulation of chondrocyte to osteoblast transdifferentiation. Sci Rep 2017; 7:10432. [PMID: 28874841 PMCID: PMC5585223 DOI: 10.1038/s41598-017-11050-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 08/18/2017] [Indexed: 01/08/2023] Open
Abstract
Endochondral ossification in the diaphysis of long bones has been studied in-depth during fetal development but not postnatally in the epiphysis. Immunohistochemical studies revealed that Sox9 and Col2 expressing immature chondrocytes in the epiphysis transition into prehypertrophic and hypetrophic chondrocytes and finally into osteoblasts expressing Col1 and BSP during postnatal day 7–10, when serum levels of thyroid hormone (TH) rise. Lineage tracing using Rosa-td tomatoCol2-Cre-ERT2 mice treated with tamoxifen indicated that the same Col2 expressing chondrocytes expressed prehypertrophic, hypertrophic, and subsequently bone formation markers in a sequential manner in euthyroid but not hypothyroid mice, thus providing evidence that chondrocyte to osteoblast transdifferentiation is TH-dependent. Vascular invasion was apparent at the time of bone formation but not earlier. In vitro studies revealed that TH acting via TRα1 promoted expression of SHH while TRβ1 activation increased IHH but inhibited SHH expression. SHH promoted expression of markers of immature chondrocytes but inhibited chondrocyte hypertrophy while IHH promoted chondrocyte hypertrophy. Based on our data, we propose a model in which TH acting through TRα1 and TRβ1, respectively, fine tune levels of SHH and IHH and, thereby control the transit of proliferating immature chondrocytes into mature hypertrophic chondrocytes to become osteoblasts at the epiphysis.
Collapse
Affiliation(s)
- Patrick Aghajanian
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California, United States
| | - Weirong Xing
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California, United States.,Department of Medicine, Loma Linda University, Loma Linda, California, United States
| | - Shaohong Cheng
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California, United States
| | - Subburaman Mohan
- Musculoskeletal Disease Center, Veterans Affairs Loma Linda Healthcare System, Loma Linda, California, United States. .,Department of Medicine, Loma Linda University, Loma Linda, California, United States. .,Department of Orthopedics, Loma Linda University, Loma Linda, California, United States. .,Department of Biochemistry, Loma Linda University, Loma Linda, California, United States.
| |
Collapse
|
40
|
Li J, Cui Y, Xu J, Wang Q, Yang X, Li Y, Zhang X, Qiu M, Zhang Z, Zhang Z. Suppressor of Fused restraint of Hedgehog activity level is critical for osteogenic proliferation and differentiation during calvarial bone development. J Biol Chem 2017; 292:15814-15825. [PMID: 28794157 DOI: 10.1074/jbc.m117.777532] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 07/04/2017] [Indexed: 12/31/2022] Open
Abstract
Hedgehog signaling plays crucial roles in the development of calvarial bone, relying on the activation of Gli transcription factors. However, the molecular mechanism of the role of regulated Gli protein level in osteogenic specification of mesenchyme still remains elusive. Here, we show by conditionally inactivating Suppressor of Fused (Sufu), a critical repressor of Hedgehog signaling, in Wnt1-Cre-mediated cranial neural crest (CNC) or Dermo1-Cre-mediated mesodermal lineages that Sufu restraint of Hedgehog activity level is critical for differentiation of preosteogenic mesenchyme. Ablation of Sufu results in failure of calvarial bone formation, including CNC-derived bones and mesoderm-derived bones, depending on the Cre line being used. Although mesenchymal cells populate to frontonasal destinations, where they are then condensed, Sufu deletion significantly inhibits the proliferation of osteoprogenitor cells, and these cells no longer differentiate into osteoblasts. We show that there is suppression of Runx2 and Osterix, the osteogenic regulators, in calvarial mesenchyme in the Sufu mutant. We show that down-regulation of several genes upstream to Runx2 and Osterix is manifested within the calvarial primordia, including Bmp2 and its downstream genes Msx1/2 and Dlx5 By contrast, we find that Gli1, the Hedgehog activity readout gene, is excessively activated in mesenchyme. Deletion of Sufu in CNC leads to a discernible decrease in the repressive Gli3 form and an increase in the full-length Gli2. Finally, we demonstrate that simultaneous deletion of Gli2 and Sufu in CNC completely restores calvarial bone formation, suggesting that a sustained level of Hedgehog activity is critical in specification of the osteogenic mesenchymal cells.
Collapse
Affiliation(s)
- Jianying Li
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Ying Cui
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Jie Xu
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Qihui Wang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Xueqin Yang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Yan Li
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Xiaoyun Zhang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Mengsheng Qiu
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| | - Ze Zhang
- the Department of Ophthamology, Tulane Medical Center, Tulane University, New Orleans, Louisiana 70112
| | - Zunyi Zhang
- From the Zhejiang Key Laboratory for Organogenesis and Regenerative Technology, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 310036, China and
| |
Collapse
|
41
|
Rothrauff BB, Pauyo T, Debski RE, Rodosky MW, Tuan RS, Musahl V. The Rotator Cuff Organ: Integrating Developmental Biology, Tissue Engineering, and Surgical Considerations to Treat Chronic Massive Rotator Cuff Tears. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:318-335. [PMID: 28084902 DOI: 10.1089/ten.teb.2016.0446] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The torn rotator cuff remains a persistent orthopedic challenge, with poor outcomes disproportionately associated with chronic, massive tears. Degenerative changes in the tissues that comprise the rotator cuff organ, including muscle, tendon, and bone, contribute to the poor healing capacity of chronic tears, resulting in poor function and an increased risk for repair failure. Tissue engineering strategies to augment rotator cuff repair have been developed in an effort to improve rotator cuff healing and have focused on three principal aims: (1) immediate mechanical augmentation of the surgical repair, (2) restoration of muscle quality and contractility, and (3) regeneration of native enthesis structure. Work in these areas will be reviewed in sequence, highlighting the relevant pathophysiology, developmental biology, and biomechanics, which must be considered when designing therapeutic applications. While the independent use of these strategies has shown promise, synergistic benefits may emerge from their combined application given the interdependence of the tissues that constitute the rotator cuff organ. Furthermore, controlled mobilization of augmented rotator cuff repairs during postoperative rehabilitation may provide mechanotransductive cues capable of guiding tissue regeneration and restoration of rotator cuff function. Present challenges and future possibilities will be identified, which if realized, may provide solutions to the vexing condition of chronic massive rotator cuff tears.
Collapse
Affiliation(s)
- Benjamin B Rothrauff
- 1 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Thierry Pauyo
- 3 Division of Sports Medicine, Department of Orthopaedic Surgery, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Richard E Debski
- 2 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Mark W Rodosky
- 3 Division of Sports Medicine, Department of Orthopaedic Surgery, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Rocky S Tuan
- 1 Department of Orthopaedic Surgery, Center for Cellular and Molecular Engineering, University of Pittsburgh , Pittsburgh, Pennsylvania.,2 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania
| | - Volker Musahl
- 2 McGowan Institute for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania.,3 Division of Sports Medicine, Department of Orthopaedic Surgery, University of Pittsburgh , Pittsburgh, Pennsylvania.,4 Orthopaedic Robotics Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh , Pittsburgh, Pennsylvania
| |
Collapse
|
42
|
Didiasova M, Singh R, Wilhelm J, Kwapiszewska G, Wujak L, Zakrzewicz D, Schaefer L, Markart P, Seeger W, Lauth M, Wygrecka M. Pirfenidone exerts antifibrotic effects through inhibition of GLI transcription factors. FASEB J 2017; 31:1916-1928. [DOI: 10.1096/fj.201600892rr] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 01/09/2017] [Indexed: 01/22/2023]
Affiliation(s)
- Miroslava Didiasova
- Department of BiochemistryUniversities of Giessen and Marburg Lung CenterGiessenGermany
| | - Rajeev Singh
- Institute of Molecular Biology and Tumor Research (IMT)Center for Tumor Biology and ImmunologyPhilipps UniversityMarburgGermany
| | - Jochen Wilhelm
- Department of Internal MedicineUniversities of Giessen and Marburg Lung CenterGiessenGermany
- German Center for Lung ResearchJustus‐Liebig UniversityGiessenGermany
| | | | - Lukasz Wujak
- Department of BiochemistryUniversities of Giessen and Marburg Lung CenterGiessenGermany
| | - Dariusz Zakrzewicz
- Department of BiochemistryUniversities of Giessen and Marburg Lung CenterGiessenGermany
| | - Liliana Schaefer
- Institute of Pharmacology and ToxicologyGoethe University School of MedicineFrankfurt am MainGermany
| | - Philipp Markart
- Department of Internal MedicineUniversities of Giessen and Marburg Lung CenterGiessenGermany
- German Center for Lung ResearchJustus‐Liebig UniversityGiessenGermany
| | - Werner Seeger
- Department of Internal MedicineUniversities of Giessen and Marburg Lung CenterGiessenGermany
- German Center for Lung ResearchJustus‐Liebig UniversityGiessenGermany
| | - Matthias Lauth
- Institute of Molecular Biology and Tumor Research (IMT)Center for Tumor Biology and ImmunologyPhilipps UniversityMarburgGermany
| | - Malgorzata Wygrecka
- Department of BiochemistryUniversities of Giessen and Marburg Lung CenterGiessenGermany
- German Center for Lung ResearchJustus‐Liebig UniversityGiessenGermany
| |
Collapse
|
43
|
Frisch J, Cucchiarini M. Gene- and Stem Cell-Based Approaches to Regulate Hypertrophic Differentiation in Articular Cartilage Disorders. Stem Cells Dev 2016; 25:1495-1512. [DOI: 10.1089/scd.2016.0106] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Janina Frisch
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg, Germany
| | - Magali Cucchiarini
- Center of Experimental Orthopaedics, Saarland University and Saarland University Medical Center, Homburg, Germany
| |
Collapse
|
44
|
Rivadeneira F, Mäkitie O. Osteoporosis and Bone Mass Disorders: From Gene Pathways to Treatments. Trends Endocrinol Metab 2016; 27:262-281. [PMID: 27079517 DOI: 10.1016/j.tem.2016.03.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 03/04/2016] [Accepted: 03/06/2016] [Indexed: 12/28/2022]
Abstract
Genomic technologies have evolved rapidly contributing to the understanding of diseases. Genome-wide association studies (GWAS) and whole-exome sequencing have aided the identification of the genetic determinants of monogenic and complex conditions including osteoporosis and bone mass disorders. Overlap exists between the genes implicated in monogenic and complex forms of bone mass disorders, largely explained by the clustering of genes encoding factors in signaling pathways crucial for mesenchymal cell differentiation, skeletal development, and bone remodeling and metabolism. Numerous of the remaining discovered genes merit functional follow-up studies to elucidate their role in bone biology. The insight provided by genetic studies is serving the identification of biomarkers predictive of disease, redefining disease, response to treatment, and discovery of novel drug targets for skeletal disorders.
Collapse
Affiliation(s)
- Fernando Rivadeneira
- Musculoskeletal Genomics, Health and Metabolism, Departments of Internal Medicine and Epidemiology, Erasmus University Medical Center, 3000CA Rotterdam, The Netherlands.
| | - Outi Mäkitie
- Children's Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Molecular Medicine and Surgery, Karolinska Institutet, and Clinical Genetics, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
45
|
Lei R, Zhang K, Wei Y, Chen M, Weinstein LS, Hong Y, Zhu M, Li H, Li H. G-Protein α-Subunit Gsα Is Required for Craniofacial Morphogenesis. PLoS One 2016; 11:e0147535. [PMID: 26859889 PMCID: PMC4747491 DOI: 10.1371/journal.pone.0147535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 01/05/2016] [Indexed: 02/05/2023] Open
Abstract
The heterotrimeric G protein subunit Gsα couples receptors to activate adenylyl cyclase and is required for the intracellular cAMP response and protein kinase A (PKA) activation. Gsα is ubiquitously expressed in many cell types; however, the role of Gsα in neural crest cells (NCCs) remains unclear. Here we report that NCCs-specific Gsα knockout mice die within hours after birth and exhibit dramatic craniofacial malformations, including hypoplastic maxilla and mandible, cleft palate and craniofacial skeleton defects. Histological and anatomical analysis reveal that the cleft palate in Gsα knockout mice is a secondary defect resulting from craniofacial skeleton deficiencies. In Gsα knockout mice, the morphologies of NCCs-derived cranial nerves are normal, but the development of dorsal root and sympathetic ganglia are impaired. Furthermore, loss of Gsα in NCCs does not affect cranial NCCs migration or cell proliferation, but significantly accelerate osteochondrogenic differentiation. Taken together, our study suggests that Gsα is required for neural crest cells-derived craniofacial development.
Collapse
Affiliation(s)
- Run Lei
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Laboratory of Developmental and Regenerative biology, Institute of Biomedicine & Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
| | - Ke Zhang
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Laboratory of Developmental and Regenerative biology, Institute of Biomedicine & Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
| | - Yanxia Wei
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lee S. Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yang Hong
- Department of Cell Biology & Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Minyan Zhu
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
| | - Hongchang Li
- Shenzhen Key Laboratory for Molecular Biology of Neural Development, Laboratory of Developmental and Regenerative biology, Institute of Biomedicine & Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
- * E-mail: Hongchang Li (HCL); Huashun Li (HSL)
| | - Huashun Li
- West China Developmental & Stem Cell Institute, West China Second Hospital, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- SARITEX Center for Stem Cell Engineering Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Chinese Academy of Sciences, Shanghai, China
- * E-mail: Hongchang Li (HCL); Huashun Li (HSL)
| |
Collapse
|
46
|
Duan L, Liang Y, Ma B, Zhu W, Wang D. Epigenetic regulation in chondrocyte phenotype maintenance for cell-based cartilage repair. Am J Transl Res 2015; 7:2127-2140. [PMID: 26807163 PMCID: PMC4697695] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/11/2015] [Indexed: 06/05/2023]
Abstract
Loss of hyaline chondrocyte phenotype during the monolayer culture in vitro is a major obstacle for cell-based articular cartilage repair. Increasing evidence implicates an important role of the epigenetic regulation in maintaining the chondrocyte phenotype. DNA methylation, histone modifications and microRNAs have all been shown to contribute to chondrocyte dedifferentiation and hypertrophy. Moreover, the interplay among epigenetic regulators forms a complicated epigenetic network in regulating chondrocyte dedifferentiation. This review provides a detailed overview of the epigenetic regulation in maintaining the chondrocyte phenotype for chondrocyte-based cartilage repair.
Collapse
Affiliation(s)
- Li Duan
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Yujie Liang
- School of Chemical Biology & Biotechnology, Peking University Shenzhen Graduate SchoolShenzhen 518000, Guangdong Province, China
| | - Bin Ma
- Division of Immunology, University Children’s Hospital ZurichZurich 8032, Switzerland
| | - Weimin Zhu
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| | - Daping Wang
- Shenzhen Key Laboratory of Tissue Engineering, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
- Department of Orthopedics, Shenzhen Second People’s HospitalShenzhen 518035, Guangdong Province, China
| |
Collapse
|
47
|
Zhong L, Huang X, Karperien M, Post JN. The Regulatory Role of Signaling Crosstalk in Hypertrophy of MSCs and Human Articular Chondrocytes. Int J Mol Sci 2015; 16:19225-47. [PMID: 26287176 PMCID: PMC4581295 DOI: 10.3390/ijms160819225] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Accepted: 08/07/2015] [Indexed: 12/26/2022] Open
Abstract
Hypertrophic differentiation of chondrocytes is a main barrier in application of mesenchymal stem cells (MSCs) for cartilage repair. In addition, hypertrophy occurs occasionally in osteoarthritis (OA). Here we provide a comprehensive review on recent literature describing signal pathways in the hypertrophy of MSCs-derived in vitro differentiated chondrocytes and chondrocytes, with an emphasis on the crosstalk between these pathways. Insight into the exact regulation of hypertrophy by the signaling network is necessary for the efficient application of MSCs for articular cartilage repair and for developing novel strategies for curing OA. We focus on articles describing the role of the main signaling pathways in regulating chondrocyte hypertrophy-like changes. Most studies report hypertrophic differentiation in chondrogenesis of MSCs, in both human OA and experimental OA. Chondrocyte hypertrophy is not under the strict control of a single pathway but appears to be regulated by an intricately regulated network of multiple signaling pathways, such as WNT, Bone morphogenetic protein (BMP)/Transforming growth factor-β (TGFβ), Parathyroid hormone-related peptide (PTHrP), Indian hedgehog (IHH), Fibroblast growth factor (FGF), Insulin like growth factor (IGF) and Hypoxia-inducible factor (HIF). This comprehensive review describes how this intricate signaling network influences tissue-engineering applications of MSCs in articular cartilage (AC) repair, and improves understanding of the disease stages and cellular responses within an OA articular joint.
Collapse
Affiliation(s)
- Leilei Zhong
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Xiaobin Huang
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
- School of Life Sciences, Chongqing University, Chongqing 400030, China.
| | - Marcel Karperien
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| | - Janine N Post
- Developmental BioEngineering, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede 7500 AE, The Netherlands.
| |
Collapse
|
48
|
Abstract
The postnatal skeleton undergoes growth, remodeling, and repair. We hypothesized that skeletal progenitor cells active during these disparate phases are genetically and phenotypically distinct. We identified a highly potent regenerative cell type that we term the fracture-induced bone, cartilage, stromal progenitor (f-BCSP) in the fracture callus of adult mice. The f-BCSP possesses significantly enhanced skeletogenic potential compared with BCSPs harvested from uninjured bone. It also recapitulates many gene expression patterns involved in perinatal skeletogenesis. Our results indicate that the skeletal progenitor population is functionally stratified, containing distinct subsets responsible for growth, regeneration, and repair. Furthermore, our findings suggest that injury-induced changes to the skeletal stem and progenitor microenvironments could activate these cells and enhance their regenerative potential.
Collapse
|
49
|
Dyment NA, Breidenbach AP, Schwartz AG, Russell RP, Aschbacher-Smith L, Liu H, Hagiwara Y, Jiang R, Thomopoulos S, Butler DL, Rowe DW. Gdf5 progenitors give rise to fibrocartilage cells that mineralize via hedgehog signaling to form the zonal enthesis. Dev Biol 2015; 405:96-107. [PMID: 26141957 DOI: 10.1016/j.ydbio.2015.06.020] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 06/03/2015] [Accepted: 06/24/2015] [Indexed: 02/01/2023]
Abstract
The sequence of events that leads to the formation of a functionally graded enthesis is not clearly defined. The current study demonstrates that clonal expansion of Gdf5 progenitors contributes to linear growth of the enthesis. Prior to mineralization, Col1+ cells in the enthesis appose Col2+ cells of the underlying primary cartilage. At the onset of enthesis mineralization, cells at the base of the enthesis express alkaline phosphatase, Indian hedgehog, and ColX as they mineralize. The mineralization front then extends towards the tendon midsubstance as cells above the front become encapsulated in mineralized fibrocartilage over time. The hedgehog (Hh) pathway regulates this process, as Hh-responsive Gli1+ cells within the developing enthesis mature from unmineralized to mineralized fibrochondrocytes in response to activated signaling. Hh signaling is required for mineralization, as tissue-specific deletion of its obligate transducer Smoothened in the developing tendon and enthesis cells leads to significant reductions in the apposition of mineralized fibrocartilage. Together, these findings provide a spatiotemporal map of events - from expansion of the embryonic progenitor pool to synthesis of the collagen template and finally mineralization of this template - that leads to the formation of the mature zonal enthesis. These results can inform future tendon-to-bone repair strategies to create a mechanically functional enthesis in which tendon collagen fibers are anchored to bone through mineralized fibrocartilage.
Collapse
Affiliation(s)
- Nathaniel A Dyment
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, United States.
| | - Andrew P Breidenbach
- Biomedical Engineering Program, College of Engineering and Applied Science, University of Cincinnati, United States
| | - Andrea G Schwartz
- Department of Orthopaedic Surgery, Washington University in St. Louis, United States
| | - Ryan P Russell
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, United States
| | | | - Han Liu
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, United States
| | - Yusuke Hagiwara
- Department of Orthopaedic Surgery, Nippon Medical School Hospital, Tokyo, Japan
| | - Rulang Jiang
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, United States
| | - Stavros Thomopoulos
- Department of Orthopaedic Surgery, Washington University in St. Louis, United States
| | - David L Butler
- Biomedical Engineering Program, College of Engineering and Applied Science, University of Cincinnati, United States
| | - David W Rowe
- Center for Regenerative Medicine and Skeletal Development, School of Dental Medicine, University of Connecticut Health Center, United States
| |
Collapse
|
50
|
Abstract
The Hedgehog (Hh) signalling pathway plays many important roles in development, homeostasis and tumorigenesis. The critical function of Hh signalling in bone formation has been identified in the past two decades. Here, we review the evolutionarily conserved Hh signalling mechanisms with an emphasis on the functions of the Hh signalling pathway in bone development, homeostasis and diseases. In the early stages of embryonic limb development, Sonic Hedgehog (Shh) acts as a major morphogen in patterning the limb buds. Indian Hedgehog (Ihh) has an essential function in endochondral ossification and induces osteoblast differentiation in the perichondrium. Hh signalling is also involved intramembrane ossification. Interactions between Hh and Wnt signalling regulate cartilage development, endochondral bone formation and synovial joint formation. Hh also plays an important role in bone homeostasis, and reducing Hh signalling protects against age-related bone loss. Disruption of Hh signalling regulation leads to multiple bone diseases, such as progressive osseous heteroplasia. Therefore, understanding the signalling mechanisms and functions of Hh signalling in bone development, homeostasis and diseases will provide important insights into bone disease prevention, diagnoses and therapeutics.
Collapse
|