1
|
Huang Y, Wang Z. Therapeutic potential of SOX family transcription factors in osteoarthritis. Ann Med 2025; 57:2457520. [PMID: 39887675 PMCID: PMC11789227 DOI: 10.1080/07853890.2025.2457520] [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: 06/22/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 02/01/2025] Open
Abstract
BACKGROUND As the worldwide population ages, osteoarthritis has significantly increased. This musculoskeletal condition has become a pressing global health issue and thus, prevention and treatment of osteoarthritis have become the primary focus of domestic and international research. Scholarly investigations of the molecular mechanisms that are related to the occurrence and development of osteoarthritis have shed light on the pathological causes of this condition to a certain extent, providing a foundation for its prevention and treatment. However, further research is necessary to fully understand the critical role of the transcription factor SOX9 in chondrocyte differentiation and the development of osteoarthritis. As a result, there has been widespread interest in SOX transcription factors. While SOX9 has been utilized as a biomarker to indicate the occurrence and prognosis of osteoarthritis, investigations into other members of the SOX family and the development of targeted treatments around SOX9 are still required. PURPOSE This article considers the impact of the SOX protein on the development and inhibition of osteoarthritis and highlights the need for therapeutic approaches targeting SOX9, as supported by existing research. RESULTS SOX9 can contribute to the process of osteoarthritis through acetylation and ubiquitination modifications. The regulation of the WNT signalling pathway, Nrf2/ARE signalling pathway, NF-κB signalling pathway and SOX9 is implicated in the emergence of osteoarthritis. Non-coding RNA may play a role in the onset and progression of osteoarthritis by modulating various SOX family members, including SOX2, SOX4, SOX5, SOX6, SOX8, SOX9 and SOX11. CONCLUSION SOX9 has the capability of mitigating the onset and progression of osteoarthritis through means such as medication therapy, stem cell therapy, recombinant adeno-associated virus (rAAV) vector therapy, physical therapy and other approaches.
Collapse
Affiliation(s)
- Yue Huang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| | - Zhuo Wang
- College of Exercise and Health, Shenyang Sport University, Shenyang, China
| |
Collapse
|
2
|
Tsutsumi-Arai C, Tran A, Arai Y, Ono W, Ono N. Mandibular Condylar Cartilage in Development and Diseases: A PTHrP-Centric View. Orthod Craniofac Res 2025. [PMID: 40251915 DOI: 10.1111/ocr.12936] [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: 12/25/2024] [Revised: 12/25/2024] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
The mandibular condylar cartilage (MCC) is a dual-function component of the temporomandibular joint (TMJ), acting as both articular cartilage for jaw movement and growth cartilage for vertical growth of the mandibular condyle. Parathyroid hormone-related protein (PTHrP) plays a critical role in orchestrating chondrogenesis in the long bone, and its importance is also highlighted in both MCC development and TMJ function. Here, we discuss the role of PTHrP in the development, growth and diseases of the MCC. PTHrP is a key morphogen in the MCC that regulates chondrogenesis by promoting chondrocyte proliferation and preventing premature hypertrophic differentiation. Exclusively expressed in the superficial layer, PTHrP diffuses across the MCC and targets chondrocytes in deeper layers, regulating transcription factors such as RUNX2 and SOX9. PTHrP regulates chondrocyte differentiation through two main pathways: the PTHrP-PTH1R signalling pathway, which suppresses hypertrophy and the PTHrP-Ihh negative feedback loop, which balances proliferation and hypertrophy. In the postnatal murine MCC, PTHrP levels are high early on and decrease after the onset of mastication around P21. Altered mechanical environments, such as those therapeutically induced as mandibular advancement, increase PTHrP expression, promoting chondrocyte proliferation and delaying hypertrophy. PTHrP also plays a dual role in adult TMJ diseases, particularly in osteoarthritis (OA); PTHrP expression transiently increases during the early stages of TMJ-OA to promote cell proliferation, but its eventual decrease contributes to the progression of the disease. This highlights the complex role of PTHrP in maintaining MCC homeostasis and its potential involvement in TMJ-OA pathology. The MCC combines the characteristics of growth and articular cartilage and functions distinctively in three phases: development before occlusion, growth after the occlusion is established, and maintenance after the growth is complete. While PTHrP plays a multifaceted role in all phases, further research is needed to fully understand how it regulates MCC development, growth and diseases.
Collapse
Affiliation(s)
- Chiaki Tsutsumi-Arai
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Amy Tran
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Yuki Arai
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Wanida Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| | - Noriaki Ono
- University of Texas Health Science Center at Houston School of Dentistry, Houston, Texas, USA
| |
Collapse
|
3
|
Baran K, Brzeziańska-Lasota E, Kryczka J, Boncela J, Czechowska A, Kopacz K, Padula G, Nowak K, Domżalski M. The Expression Level of SOX Family Transcription Factors' mRNA as a Diagnostic Marker for Osteoarthritis. J Clin Med 2025; 14:1176. [PMID: 40004707 PMCID: PMC11856735 DOI: 10.3390/jcm14041176] [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: 01/03/2025] [Revised: 01/25/2025] [Accepted: 01/31/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives:Osteoarthritis (OA) is the most common degenerative and chronic joint disease and is a leading cause of pain and disability in adults worldwide. The SRY-related HMG box (SOX) family transcription factors (TFs) play a crucial role during the pathogenesis of OA; however, their exact mechanisms remain unexplored. The aim of our study was to conduct a bioinformatics analysis of the common interactions of SOX-5, SOX-9, and SOX-11 with other proteins, as well as their role in OA pathogenesis. Methods:SOX5, SOX9, and SOX11 mRNA expression levels in articular cartilage with subchondral bone and synovium from knee OA patients were assessed using the qPCR method. The study group consisted of thirty-one patients (n = 31). Total RNA was isolated from the articular cartilage with subchondral bone and synovium from the affected and unaffected area of the knee joint. Results: Our results revealed a regulatory network between SOX-5, SOX-9, and SOX-11, and various proteins involved in the pathogenesis of knee OA and their collective interactions, which are involved in the regulation of cartilage extracellular matrix (ECM) organization, response to stimulus, regulation of gene expression, inflammatory response, cartilage condensation, and ossification in chondrocytes. Higher expression levels of SOX5, SOX9, and SOX11 mRNA were noted in OA-affected articular cartilage with subchondral bone compared to control tissue (p = 0.00015, p = 0.0024 and p > 0.05, respectively, Mann-Whitney U-test). All studied genes demonstrated elevated mRNA expression levels in the articular cartilage with subchondral bone from stage 4 patients than those with stage 3 (p > 0.05; Mann-Whitney U-test). Lower SOX5, SOX9, and SOX11 mRNA expression levels were found in OA-affected synovium compared to the control tissue (p = 0.0003, p > 0.05 and p = 0.0007, respectively, Mann-Whitney U-test). Decreased SOX9 mRNA expression levels in synovium were noted in patients with stage 4 disease than those with stage 3; however, SOX5 and SOX11 mRNA expression levels were higher in patients with stage 4 (p > 0.05; Mann-Whitney U-test). Conclusions: The results of our research show that the studied SOX TFs play a role in the development of OA, contributing to the formation of pathological changes not only in the articular cartilage, but also in the synovial membrane. The changes in the SOX5, SOX9, and SOX11 mRNA expression levels in the articular cartilage with subchondral bone and synovium may serve as potential molecular diagnostic biomarkers for detecting OA and could indicate the progression of this disease; however, our observations require further investigation.
Collapse
Affiliation(s)
- Kamila Baran
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Ewa Brzeziańska-Lasota
- Department of Biomedicine and Genetics, Chair of Biology and Medical Microbiology, Medical University of Lodz, 92-215 Lodz, Poland;
| | - Jakub Kryczka
- Institute of Medical Biology, Polish Academy of Sciences, 93-232 Lodz, Poland; (J.K.); (J.B.)
| | - Joanna Boncela
- Institute of Medical Biology, Polish Academy of Sciences, 93-232 Lodz, Poland; (J.K.); (J.B.)
| | - Aleksandra Czechowska
- Academic Laboratory of Movement and Human Physical Performance, Medical University of Lodz, 90-001 Lodz, Poland; (A.C.); (K.K.); (G.P.)
| | - Karolina Kopacz
- Academic Laboratory of Movement and Human Physical Performance, Medical University of Lodz, 90-001 Lodz, Poland; (A.C.); (K.K.); (G.P.)
| | - Gianluca Padula
- Academic Laboratory of Movement and Human Physical Performance, Medical University of Lodz, 90-001 Lodz, Poland; (A.C.); (K.K.); (G.P.)
| | - Krzysztof Nowak
- Department of Orthopedics and Traumatology, University Clinical Hospital No. 2 of the Medical University of Lodz, 90-549 Lodz, Poland; (K.N.); (M.D.)
| | - Marcin Domżalski
- Department of Orthopedics and Traumatology, University Clinical Hospital No. 2 of the Medical University of Lodz, 90-549 Lodz, Poland; (K.N.); (M.D.)
| |
Collapse
|
4
|
Wang L, Liu Z, Zhao S, Xu K, Aceves V, Qiu C, Feng HC, Bian F, He J, Song CJ, Troutwine B, Liu L, Ma S, Niu Y, Wang S, Yuan S, Li X, Zhao L, Liu X, Qiu G, Wu Z, Deciphering disorders Involving Scoliosis and COmorbidities (DISCO) study group, Zhang TJ, Gray RS, Wu N. Variants in the SOX9 transactivation middle domain induce axial skeleton dysplasia and scoliosis. Proc Natl Acad Sci U S A 2025; 122:e2313978121. [PMID: 39854231 PMCID: PMC11789016 DOI: 10.1073/pnas.2313978121] [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: 08/18/2023] [Accepted: 09/30/2024] [Indexed: 01/30/2025] Open
Abstract
SOX9 is a crucial transcriptional regulator of cartilage development and homeostasis. Dysregulation of SOX9 is associated with a wide spectrum of skeletal disorders, including campomelic dysplasia, acampomelic campomelic dysplasia, and scoliosis. Yet how SOX9 variants contribute to the spectrum of axial skeletal disorders is not well understood. Here, we report four pathogenic variants of SOX9 identified in a cohort of patients with congenital vertebral malformations. We report a pathogenic missense variant in the transactivation middle (TAM) domain of SOX9 associated with mild skeletal dysplasia and scoliosis. We isolated a Sox9 mutant mouse with an in-frame microdeletion in the TAM domain (Sox9Asp272del), which exhibits skeletal dysplasia including kinked tails, rib cage anomalies, and scoliosis in homozygous mutants. We find that both the human missense and the mouse microdeletion mutations resulted in reduced SOX9 protein stability in cell culture, while Sox9Asp272del mutant mice show decreased SOX9 expression in the growth plate and annulus fibrosus tissues of the spine. This reduction in SOX9 expression was correlated with the reduction of extracellular matrix components, such as tenascin-X and the Adhesion G-protein coupled receptor ADGRG6. In summary, our work identified and modeled a pathologic variant of SOX9 within the TAM domain and demonstrated its importance for SOX9 protein stability. Our work demonstrates that SOX9 stability is important for the regulation of ADGRG6 expression, which is a known regulator of postnatal spine homeostasis, underscoring the essential role of SOX9 dosage in a spectrum of axial skeleton dysplasia in humans.
Collapse
Affiliation(s)
- Lianlei Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
| | - Zhaoyang Liu
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
| | - Sen Zhao
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
| | - Kexin Xu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Valeria Aceves
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Cheng Qiu
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
| | - Hong Colleen Feng
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
| | - Fangzhou Bian
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
| | - Jingyu He
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
| | - Christina J. Song
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Benjamin Troutwine
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Lian Liu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
| | - Samuel Ma
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Yuchen Niu
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Shengru Wang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Suomao Yuan
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
| | - Xiaoxin Li
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Lina Zhao
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Xinyu Liu
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
| | - Guixing Qiu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Zhihong Wu
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Deciphering disorders Involving Scoliosis and COmorbidities (DISCO) study group
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Department of Orthopedic Surgery, Qilu Hospital of Shandong University, Jinan250012, Shandong, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, CA90033
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
- Department of Orthopaedic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA90033
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
- Medical Research Center, Peking Union Medical College Hospital, Peking Union Medical College, and Chinese Academy of Medical Sciences, Beijing100730, China
| | - Terry Jianguo Zhang
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| | - Ryan S. Gray
- Department of Nutritional Sciences, Dell Pediatric Research Institute, The University of Texas at Austin, Dell Medical School, Austin, TX78723
| | - Nan Wu
- Department of Orthopedic Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing100730, China
- Beijing Key Laboratory of Big Data Innovation and Application for Skeletal Health Medical Care, Beijing100730, China
- Key Laboratory of Big Data for Spinal Deformities, Chinese Academy of Medical Sciences, Beijing100730, China
| |
Collapse
|
5
|
Hudacova E, Abaffy P, Kaplan MM, Krausova M, Kubista M, Machon O. Single-cell transcriptomic resolution of osteogenesis during craniofacial morphogenesis. Bone 2025; 190:117297. [PMID: 39461490 DOI: 10.1016/j.bone.2024.117297] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Craniofacial morphogenesis depends on complex cell fate decisions during the differentiation of post-migratory cranial neural crest cells. Molecular mechanisms of cell differentiation of mesenchymal cells to developing bones, cartilage, teeth, tongue, and other craniofacial tissues are still poorly understood. We performed single-cell transcriptomic analysis of craniofacial mesenchymal cells derived from cranial NCCs in mouse embryo. Using FACS sorting of Wnt1-Cre2 progeny, we carefully mapped the cell heterogeneity in the craniofacial region during the initial stages of cartilage and bone formation. Transcriptomic data and in vivo validations identified molecular determinants of major cell populations involved in the development of lower and upper jaw, teeth, tongue, dermis, or periocular mesenchyme. Single-cell transcriptomic analysis of Meis2-deficient mice revealed critical gene expression differences, including increased osteogenic and cell adhesion markers. This leads to affected mesenchymal cell differentiation and increased ossification, resulting in impaired bone, cartilage, and tongue formation.
Collapse
Affiliation(s)
- Erika Hudacova
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic; Department of Cell Biology, Faculty of Science, Charles University, Vinicna 7, 12000 Prague, Czech Republic.
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, 25200 Vestec, Czech Republic.
| | - Mehmet Mahsum Kaplan
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| | - Michaela Krausova
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology, Czech Academy of Sciences, Prumyslova 595, 25200 Vestec, Czech Republic.
| | - Ondrej Machon
- Department of Developmental Biology, Institute of Experimental Medicine, Czech Academy of Sciences, Videnska 1083, 14200 Prague, Czech Republic.
| |
Collapse
|
6
|
Jia Z, Mukhopadhyay N, Yang Z, Butali A, Sun J, You Y, Yao M, Zhen Q, Ma J, He M, Pan Y, Alade A, Wang Y, Olujitan M, Qi M, Adeyemo WL, Buxó CJ, Gowans LJJ, Eshete M, Huang Y, Li C, Leslie EJ, Wang L, Bian Z, Carlson JC, Shi B, Weinberg SM, Murray JC, Sun L, Marazita ML, Freathy RM, Beaumont RN. Multi-ancestry Genome Wide Association Study Meta-analysis of Non-syndromic Orofacial Clefts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.06.24318522. [PMID: 39711721 PMCID: PMC11661332 DOI: 10.1101/2024.12.06.24318522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Non-syndromic orofacial clefts (NSOC) are common craniofacial birth defects, and result from both genetic and environmental factors. NSOC include three major sub-phenotypes: non-syndromic cleft lip with palate (NSCLP), non-syndromic cleft lip only (NSCLO) and non-syndromic cleft palate only (NSCPO), NSCLP and NSCLO are also sometimes grouped as non-syndromic cleft lip with or without cleft palate (NSCL/P) based on epidemiology. Currently known loci only explain a limited proportion of the heritability of NSOC. Further, differences in genetic susceptibility among the sub-phenotypes are poorly characterized. We performed a multi-ancestry GWAS meta-analysis on 44,094 individuals (9,381 cases, 28,510 controls, 2042 case-parent trios and 18 multiplex pedigrees) of East Asian, European, Latin and South American, and African ancestry for both NSOC and subtypes. We identified 50 loci, including 11 novel loci: four loci ( CALD1 , SHH , NRG1 and LINC00320 ) associated with both NSOC and NSCL/P, two loci ( NTRK1 and RUNX1 ) only associated with NSOC, four loci ( HMGCR , PRICKLE1 , SOX9 and MYH9 ) only associated with NSCL/P and one locus ( ALX1 ) specifically associated with NSCLO. Five of the novel loci are located in regions containing genes associated with syndromic orofacial clefts ( SHH , NTRK1, CALD1, ALX1 and SOX9 ); seven of the novel loci are located in regions containing genes-implicated in craniofacial development ( HMGCR, SHH, PRICKLE1, ALX1, SOX9, RUNX1, MYH9 ). Genetic correlation and colocalization analyses revealed an overlap between signals associated with NSCLO, NSCPO and NSCLP, but there were also notable differences, emphasizing the complexity of common and distinct genetic processes affecting lip and palate development.
Collapse
|
7
|
Weber CJ, Weitzel AJ, Liu AY, Gacasan EG, Sah RL, Cooper KL. Cellular and molecular mechanisms that shape the development and evolution of tail vertebral proportion in mice and jerboas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.25.620311. [PMID: 39484405 PMCID: PMC11527341 DOI: 10.1101/2024.10.25.620311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Despite the functional importance of the vertebral skeleton, little is known about how individual vertebrae elongate or achieve disproportionate lengths as in the giraffe neck. Rodent tails are an abundantly diverse and more tractable system to understand mechanisms of vertebral growth and proportion. In many rodents, disproportionately long mid-tail vertebrae form a 'crescendo-decrescendo' of lengths in the tail series. In bipedal jerboas, these vertebrae grow exceptionally long such that the adult tail is 1.5x the length of a mouse tail, relative to body length, with four fewer vertebrae. How do vertebrae with the same regional identity elongate differently from their neighbors to establish and diversify adult proportion? Here, we find that vertebral lengths are largely determined by differences in growth cartilage height and the number of cells progressing through endochondral ossification. Hypertrophic chondrocyte size, a major contributor to differential elongation in mammal limb bones, differs only in the longest jerboa mid-tail vertebrae where they are exceptionally large. To uncover candidate molecular mechanisms of disproportionate vertebral growth, we performed intersectional RNA-Seq of mouse and jerboa tail vertebrae with similar and disproportionate elongation rates. Many regulators of posterior axial identity and endochondral elongation are disproportionately differentially expressed in jerboa vertebrae. Among these, the inhibitory natriuretic peptide receptor C (NPR3) appears in multiple studies of rodent and human skeletal proportion suggesting it refines local growth rates broadly in the skeleton and broadly in mammals. Consistent with this hypothesis, NPR3 loss of function mice have abnormal tail and limb proportions. Therefore, in addition to genetic components of the complex process of vertebral evolution, these studies reveal fundamental mechanisms of skeletal growth and proportion.
Collapse
Affiliation(s)
- Ceri J Weber
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander J Weitzel
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Alexander Y Liu
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Erica G Gacasan
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Robert L Sah
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Kimberly L Cooper
- Department of Cell and Developmental Biology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| |
Collapse
|
8
|
Ridnik M, Abberbock E, Alipov V, Lhermann SZ, Kaufman S, Lubman M, Poulat F, Gonen N. Two redundant transcription factor binding sites in a single enhancer are essential for mammalian sex determination. Nucleic Acids Res 2024; 52:5514-5528. [PMID: 38499491 PMCID: PMC11162780 DOI: 10.1093/nar/gkae178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 02/25/2024] [Accepted: 02/29/2024] [Indexed: 03/20/2024] Open
Abstract
Male development in mammals depends on the activity of the two SOX gene: Sry and Sox9, in the embryonic testis. As deletion of Enhancer 13 (Enh13) of the Sox9 gene results in XY male-to-female sex reversal, we explored the critical elements necessary for its function and hence, for testis and male development. Here, we demonstrate that while microdeletions of individual transcription factor binding sites (TFBS) in Enh13 lead to normal testicular development, combined microdeletions of just two SRY/SOX binding motifs can alone fully abolish Enh13 activity leading to XY male-to-female sex reversal. This suggests that for proper male development to occur, these few nucleotides of non-coding DNA must be intact. Interestingly, we show that depending on the nature of these TFBS mutations, dramatically different phenotypic outcomes can occur, providing a molecular explanation for the distinct clinical outcomes observed in patients harboring different variants in the same enhancer.
Collapse
Affiliation(s)
- Meshi Ridnik
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Elisheva Abberbock
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Veronica Alipov
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Shelly Ziv Lhermann
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Shoham Kaufman
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Maor Lubman
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Francis Poulat
- Group “Development and Pathology of the Gonad”. Department of Genetics, Cell Biology and Development, Institute of Human Genetics, CNRS-University of Montpellier UMR9002, Montpellier, France
| | - Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| |
Collapse
|
9
|
Stanton E, Sheridan S, Urata M, Chai Y. From Bedside to Bench and Back: Advancing Our Understanding of the Pathophysiology of Cleft Palate and Implications for the Future. Cleft Palate Craniofac J 2024; 61:759-773. [PMID: 36457208 DOI: 10.1177/10556656221142098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024] Open
Abstract
OBJECTIVE To provide a comprehensive understanding of the pathophysiology of cleft palate (CP) and future perspectives. DESIGN Literature review. SETTING Setting varied across studies by level of care and geographical locations. INTERVENTIONS No interventions were performed. MAIN OUTCOME MEASURE(S) Primary outcome measures were to summarize our current understanding of palatogenesis in humans and animal models, the pathophysiology of CP, and potential future treatment modalities. RESULTS Animal research has provided considerable insight into the pathophysiology, molecular and cellular mechanisms of CP that have allowed for the development of novel treatment strategies. However, much work has yet to be done to connect our mouse model investigations and discoveries to CP in humans. The success of innovative strategies for tissue regeneration in mice provides promise for an exciting new avenue for improved and more targeted management of cleft care with precision medicine in patients. However, significant barriers to clinical translation remain. Among the most notable challenges include the differences in some aspects of palatogenesis and tissue repair between mice and humans, suggesting that potential therapies that have worked in animal models may not provide similar benefits to humans. CONCLUSIONS Increased translation of pathophysiological and tissue regeneration studies to clinical trials will bridge a wide gap in knowledge between animal models and human disease. By enhancing interaction between basic scientists and clinicians, and employing our animal model findings of disease mechanisms in concert with what we glean in the clinic, we can generate a more targeted and improved treatment algorithm for patients with CP.
Collapse
Affiliation(s)
- Eloise Stanton
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Samuel Sheridan
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| | - Mark Urata
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
- Division of Plastic and Maxillofacial Surgery, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Yang Chai
- Center for Craniofacial Molecular Biology, University of Southern California, Los Angeles, CA, USA
- Ostrow School of Dentistry, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
10
|
Ferrari MTM, Silva ESDN, Nishi MY, Batista RL, Mendonca BB, Domenice S. Testicular differentiation in 46,XX DSD: an overview of genetic causes. Front Endocrinol (Lausanne) 2024; 15:1385901. [PMID: 38721146 PMCID: PMC11076692 DOI: 10.3389/fendo.2024.1385901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 03/18/2024] [Indexed: 01/18/2025] Open
Abstract
In mammals, the development of male or female gonads from fetal bipotential gonads depends on intricate genetic networks. Changes in dosage or temporal expression of sex-determining genes can lead to differences of gonadal development. Two rare conditions are associated with disruptions in ovarian determination, including 46,XX testicular differences in sex development (DSD), in which the 46,XX gonads differentiate into testes, and 46,XX ovotesticular DSD, characterized by the coexistence of ovarian and testicular tissue in the same individual. Several mechanisms have been identified that may contribute to the development of testicular tissue in XX gonads. This includes translocation of SRY to the X chromosome or an autosome. In the absence of SRY, other genes associated with testis development may be overexpressed or there may be a reduction in the activity of pro-ovarian/antitesticular factors. However, it is important to note that a significant number of patients with these DSD conditions have not yet recognized a genetic diagnosis. This finding suggests that there are additional genetic pathways or epigenetic mechanisms that have yet to be identified. The text will provide an overview of the current understanding of the genetic factors contributing to 46,XX DSD, specifically focusing on testicular and ovotesticular DSD conditions. It will summarize the existing knowledge regarding the genetic causes of these differences. Furthermore, it will explore the potential involvement of other factors, such as epigenetic mechanisms, in developing these conditions.
Collapse
Affiliation(s)
- Maria Tereza Martins Ferrari
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Elinaelma Suelane do Nascimento Silva
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Mirian Yumie Nishi
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Rafael Loch Batista
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Berenice Bilharinho Mendonca
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
- Laboratório de Sequenciamento em Larga Escala (SELA), Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Sorahia Domenice
- Disciplina de Endocrinologia e Metabologia, Laboratório de Hormônios e Genética Molecular/LIM42, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
11
|
Roshan MM, Azizi H, Majelan MA, Tabar AN. Sox9 downregulation in non-obstructive azoospermia by UTF1 and mediator role of POU5F1. BMC Res Notes 2024; 17:77. [PMID: 38486279 PMCID: PMC10941352 DOI: 10.1186/s13104-024-06711-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 01/31/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Spermatogenesis is the process of producing mature sperm from Spermatogonial stem cells (SSCs) and this process requires a complex cooperation of different types of somatic and germ cells. Undifferentiated spermatogonia initiate the spermatogenesis and Sertoli cells as the only somatic cells inside of the seminiferous tubule play a key role in providing chemical and physical requirements for normal spermatogenesis, here, we investigated the dysfunction of these cells in non-obstructive azoospermia. MATERIAL AND METHOD In this study, we analyzed the expression of sox9 and UTF1 in the non-obstructive human testis by immunohistochemistry. Moreover, we used the KEGG pathway and bioinformatics analysis to reveal the connection between our object genes and protein. RESULTS The immunohistochemistry analysis of the non-obstructive human seminiferous tubule showed low expression of Sox9 and UTF1 that was detected out of the main location of the responsible cells for these expressions. Our bioinformatics analysis clearly and strongly indicated the relation between UTF1 in undifferentiated spermatogonia and Sox9 in Sertoli cells mediated by POU5F1. CONCLUSION Generally, this study showed the negative effect of POU5F1 as a mediator between Sertoli cells as the somatic cells within seminiferous tubules and undifferentiated spermatogonia as the spermatogenesis initiator germ cells in non-obstructive conditions.
Collapse
Affiliation(s)
- Mehdi Mehdinezhad Roshan
- Faculty of Biotechnology, Amol University of Special Modern Technologies, 49767, Amol, P.O. Box: 49767, Iran
- Department of Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hossein Azizi
- Faculty of Biotechnology, Amol University of Special Modern Technologies, 49767, Amol, P.O. Box: 49767, Iran.
| | | | - Amirreza Niazi Tabar
- Faculty of Biotechnology, Amol University of Special Modern Technologies, 49767, Amol, P.O. Box: 49767, Iran
| |
Collapse
|
12
|
Molin AN, Contentin R, Angelozzi M, Karvande A, Kc R, Haseeb A, Voskamp C, de Charleroy C, Lefebvre V. Skeletal growth is enhanced by a shared role for SOX8 and SOX9 in promoting reserve chondrocyte commitment to columnar proliferation. Proc Natl Acad Sci U S A 2024; 121:e2316969121. [PMID: 38346197 PMCID: PMC10895259 DOI: 10.1073/pnas.2316969121] [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: 09/29/2023] [Accepted: 12/26/2023] [Indexed: 02/15/2024] Open
Abstract
SOX8 was linked in a genome-wide association study to human height heritability, but roles in chondrocytes for this close relative of the master chondrogenic transcription factor SOX9 remain unknown. We undertook here to fill this knowledge gap. High-throughput assays demonstrate expression of human SOX8 and mouse Sox8 in growth plate cartilage. In situ assays show that Sox8 is expressed at a similar level as Sox9 in reserve and early columnar chondrocytes and turned off when Sox9 expression peaks in late columnar and prehypertrophic chondrocytes. Sox8-/- mice and Sox8fl/flPrx1Cre and Sox9fl/+Prx1Cre mice (inactivation in limb skeletal cells) have a normal or near normal skeletal size. In contrast, juvenile and adult Sox8fl/flSox9fl/+Prx1Cre compound mutants exhibit a 15 to 20% shortening of long bones. Their growth plate reserve chondrocytes progress slowly toward the columnar stage, as witnessed by a delay in down-regulating Pthlh expression, in packing in columns and in elevating their proliferation rate. SOX8 or SOX9 overexpression in chondrocytes reveals not only that SOX8 can promote growth plate cell proliferation and differentiation, even upon inactivation of endogenous Sox9, but also that it is more efficient than SOX9, possibly due to greater protein stability. Altogether, these findings uncover a major role for SOX8 and SOX9 in promoting skeletal growth by stimulating commitment of growth plate reserve chondrocytes to actively proliferating columnar cells. Further, by showing that SOX8 is more chondrogenic than SOX9, they suggest that SOX8 could be preferred over SOX9 in therapies to promote cartilage formation or regeneration in developmental and degenerative cartilage diseases.
Collapse
Affiliation(s)
- Arnaud N. Molin
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Romain Contentin
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Marco Angelozzi
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Anirudha Karvande
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Ranjan Kc
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Abdul Haseeb
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Chantal Voskamp
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Charles de Charleroy
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedic Surgery, The Children’s Hospital of Philadelphia, Philadelphia, PA19104
| |
Collapse
|
13
|
Riquelme-Guzmán C, Sandoval-Guzmán T. The salamander limb: a perfect model to understand imperfect integration during skeletal regeneration. Biol Open 2024; 13:bio060152. [PMID: 38319134 PMCID: PMC10868587 DOI: 10.1242/bio.060152] [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] [Indexed: 02/07/2024] Open
Abstract
Limb regeneration in salamanders is achieved by a complex coordination of various biological processes and requires the proper integration of new tissue with old. Among the tissues found inside the limb, the skeleton is the most prominent component, which serves as a scaffold and provides support for locomotion in the animal. Throughout the years, researchers have studied the regeneration of the appendicular skeleton in salamanders both after limb amputation and as a result of fracture healing. The final outcome has been widely seen as a faithful re-establishment of the skeletal elements, characterised by a seamless integration into the mature tissue. The process of skeletal integration, however, is not well understood, and several works have recently provided evidence of commonly occurring flawed regenerates. In this Review, we take the reader on a journey through the course of bone formation and regeneration in salamanders, laying down a foundation for critically examining the mechanisms behind skeletal integration. Integration is a phenomenon that could be influenced at various steps of regeneration, and hence, we assess the current knowledge in the field and discuss how early events, such as tissue histolysis and patterning, influence the faithful regeneration of the appendicular skeleton.
Collapse
Affiliation(s)
- Camilo Riquelme-Guzmán
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307 Dresden, Germany
| | - Tatiana Sandoval-Guzmán
- Department of Internal Medicine 3, Center for Healthy Aging, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307 Dresden, Germany
- Paul Langerhans Institute Dresden of Helmholtz Centre Munich, University Hospital Carl Gustav Carus at the Technische Universität Dresden, 01307 Dresden, Germany
| |
Collapse
|
14
|
Koosha E, Brenna CTA, Ashique AM, Jain N, Ovens K, Koike T, Kitagawa H, Eames BF. Proteoglycan inhibition of canonical BMP-dependent cartilage maturation delays endochondral ossification. Development 2024; 151:dev201716. [PMID: 38117077 PMCID: PMC10820745 DOI: 10.1242/dev.201716] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 11/28/2023] [Indexed: 12/21/2023]
Abstract
During endochondral ossification, chondrocytes secrete a proteoglycan (PG)-rich extracellular matrix that can inhibit the process of cartilage maturation, including expression of Ihh and Col10a1. Because bone morphogenetic proteins (BMPs) can promote cartilage maturation, we hypothesized that cartilage PGs normally inhibit BMP signalling. Accordingly, BMP signalling was evaluated in chondrocytes of wild-type and PG mutant (fam20b-/-) zebrafish and inhibited with temporal control using the drug DMH1 or an inducible dominant-negative BMP receptor transgene (dnBMPR). Compared with wild type, phospho-Smad1/5/9, but not phospho-p38, was increased in fam20b-/- chondrocytes, but only after they secreted PGs. Phospho-Smad1/5/9 was decreased in DMH1-treated or dnBMPR-activated wild-type chondrocytes, and DMH1 also decreased phospho-p38 levels. ihha and col10a1a were decreased in DMH1-treated or dnBMPR-activated chondrocytes, and less perichondral bone formed. Finally, early ihha and col10a1a expression and early perichondral bone formation of fam20b mutants were rescued with DMH1 treatment or dnBMPR activation. Therefore, PG inhibition of canonical BMP-dependent cartilage maturation delays endochondral ossification, and these results offer hope for the development of growth factor therapies for skeletal defects of PG diseases.
Collapse
Affiliation(s)
- Elham Koosha
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Connor T. A. Brenna
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Amir M. Ashique
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Niteesh Jain
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Katie Ovens
- Department of Computer Science, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Toshiyasu Koike
- Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-0003, Japan
| | - Hiroshi Kitagawa
- Department of Biochemistry, Kobe Pharmaceutical University, Higashinada-ku, Kobe 658-0003, Japan
| | - B. Frank Eames
- Department of Anatomy, Physiology, and Pharmacology, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| |
Collapse
|
15
|
Papaioannou VE, Behringer RR. Phenotypic Analysis of Dominant Mutant Effects in Mice. Cold Spring Harb Protoc 2024; 2024:107978. [PMID: 37932089 DOI: 10.1101/pdb.over107978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Dominant effects of a mutation may show up at any time during a mutational analysis, including during the early stages of an embryonic stem (ES) cell gene targeting experiment. Here, we discuss the mechanisms of dominant and semidominant effects and how they might appear if they show up in heterozygous ES cells, in ES cell chimeras, or in heterozygous progeny of chimeras. Similarly, dominant effects may be seen in mice heterozygous for CRISPR-Cas-targeted, -induced, or spontaneous mutations. If the dominant effects prevent the germline transmission of ES cells or cause fertility problems in heterozygotes, they can severely limit further analysis of the mutation. Ways to circumvent such reproductive problems are presented. The special case of imprinted genes, which may be functionally hemizygous and present a different phenotype when inherited from the mother than when inherited from the father, is discussed.
Collapse
Affiliation(s)
- Virginia E Papaioannou
- Department of Genetics and Development, Columbia University Medical Center, New York, New York 10032, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030
| |
Collapse
|
16
|
Papaioannou VE, Behringer RR. Recovering a Targeted Mutation in Mice from Embryonic Stem Cell Chimeras or CRISPR-Cas Founders. Cold Spring Harb Protoc 2024; 2024:107959. [PMID: 37932094 DOI: 10.1101/pdb.over107959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Following the production of chimeras from targeted embryonic stem (ES) cells or obtaining founders from CRISPR-Cas gene editing in preimplantation embryos, the desired targeted mutation must be recovered and established in the heterozygous state in a strain or stock of mice for further study. The breeding schemes for ES chimeras and CRISPR-Cas founders differ. For ES cell chimeras, we discuss the relative benefits of breeding from male or female chimeras. We discuss the importance of genetic background and provide practical advice for putting the mutation on inbred or outbred backgrounds or producing a coisogenic strain. For CRISPR-Cas founders, which will most likely be mosaic for different mutations, initial breeding strategies are discussed to maintain a desired genetic background at the same time as producing progeny to segregate different alleles. Strategies for testing the progeny to recognize indels, missense mutations, and knock-in mutations are discussed. In the event that ES cell chimeras or CRISPR-Cas founders produce no offspring or fail to transmit the mutant allele(s), there is a troubleshooting guide to pinpoint the problem. If heterozygous offspring from the chimeras or founders are normal, fertile, and of both sexes, the analysis of homozygous effects of the mutation can now begin; if not, possible dominant effects are considered.
Collapse
Affiliation(s)
- Virginia E Papaioannou
- Department of Genetics and Development, Columbia University Medical Center, New York, New York 10032, USA
| | - Richard R Behringer
- Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| |
Collapse
|
17
|
Hossain N, Igawa T, Suzuki M, Tazawa I, Nakao Y, Hayashi T, Suzuki N, Ogino H. Phenotype-genotype relationships in Xenopus sox9 crispants provide insights into campomelic dysplasia and vertebrate jaw evolution. Dev Growth Differ 2023; 65:481-497. [PMID: 37505799 DOI: 10.1111/dgd.12884] [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: 05/18/2023] [Revised: 07/20/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Since CRISPR-based genome editing technology works effectively in the diploid frog Xenopus tropicalis, a growing number of studies have successfully modeled human genetic diseases in this species. However, most of their targets were limited to non-syndromic diseases that exhibit abnormalities in a small fraction of tissues or organs in the body. This is likely because of the complexity of interpreting the phenotypic variations resulting from somatic mosaic mutations generated in the founder animals (crispants). In this study, we attempted to model the syndromic disease campomelic dysplasia (CD) by generating sox9 crispants in X. tropicalis. The resulting crispants failed to form neural crest cells at neurula stages and exhibited various combinations of jaw, gill, ear, heart, and gut defects at tadpole stages, recapitulating part of the syndromic phenotype of CD patients. Genotyping of the crispants with a variety of allelic series of mutations suggested that the heart and gut defects depend primarily on frame-shift mutations expected to be null, whereas the jaw, gill, and ear defects could be induced not only by such mutations but also by in-frame deletion mutations expected to delete part of the jawed vertebrate-specific domain from the encoded Sox9 protein. These results demonstrate that Xenopus crispants are useful for investigating the phenotype-genotype relationships behind syndromic diseases and examining the tissue-specific role of each functional domain within a single protein, providing novel insights into vertebrate jaw evolution.
Collapse
Affiliation(s)
- Nusrat Hossain
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Takeshi Igawa
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Makoto Suzuki
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Ichiro Tazawa
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Yuta Nakao
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Toshinori Hayashi
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| | - Nanoka Suzuki
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
| | - Hajime Ogino
- Amphibian Research Center, Hiroshima University, Hiroshima, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, Japan
| |
Collapse
|
18
|
Bottasso-Arias N, Burra K, Sinner D, Riede T. Disruption of BMP4 signaling is associated with laryngeal birth defects in a mouse model. Dev Biol 2023; 500:10-21. [PMID: 37230380 PMCID: PMC10330877 DOI: 10.1016/j.ydbio.2023.04.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 04/18/2023] [Accepted: 04/24/2023] [Indexed: 05/27/2023]
Abstract
Laryngeal birth defects are considered rare, but they can be life-threatening conditions. The BMP4 gene plays an important role in organ development and tissue remodeling throughout life. Here we examined its role in laryngeal development complementing similar efforts for the lung, pharynx, and cranial base. Our goal was to determine how different imaging techniques contribute to a better understanding of the embryonic anatomy of the normal and diseased larynx in small specimens. Contrast-enhanced micro CT images of embryonic larynx tissue from a mouse model with Bmp4 deletion informed by histology and whole-mount immunofluorescence were used to reconstruct the laryngeal cartilaginous framework in three dimensions. Laryngeal defects included laryngeal cleft, laryngeal asymmetry, ankylosis and atresia. Results implicate BMP4 in laryngeal development and show that the 3D reconstruction of laryngeal elements provides a powerful approach to visualize laryngeal defects and thereby overcoming shortcomings of 2D histological sectioning and whole mount immunofluorescence.
Collapse
Affiliation(s)
- N Bottasso-Arias
- Neonatology and Pulmonary Biology, Perinatal Institute Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - K Burra
- Neonatology and Pulmonary Biology, Perinatal Institute Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - D Sinner
- Neonatology and Pulmonary Biology, Perinatal Institute Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA; College of Medicine, University of Cincinnati, Cincinnati, OH, USA.
| | - T Riede
- Department of Physiology, Midwestern University, Glendale, AZ, USA.
| |
Collapse
|
19
|
Shigley C, Trivedi J, Meghani O, Owens BD, Jayasuriya CT. Suppressing Chondrocyte Hypertrophy to Build Better Cartilage. Bioengineering (Basel) 2023; 10:741. [PMID: 37370672 DOI: 10.3390/bioengineering10060741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 06/13/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Current clinical strategies for restoring cartilage defects do not adequately consider taking the necessary steps to prevent the formation of hypertrophic tissue at injury sites. Chondrocyte hypertrophy inevitably causes both macroscopic and microscopic level changes in cartilage, resulting in adverse long-term outcomes following attempted restoration. Repairing/restoring articular cartilage while minimizing the risk of hypertrophic neo tissue formation represents an unmet clinical challenge. Previous investigations have extensively identified and characterized the biological mechanisms that regulate cartilage hypertrophy with preclinical studies now beginning to leverage this knowledge to help build better cartilage. In this comprehensive article, we will provide a summary of these biological mechanisms and systematically review the most cutting-edge strategies for circumventing this pathological hallmark of osteoarthritis.
Collapse
Affiliation(s)
- Christian Shigley
- The Warren Alpert Medical School, Brown University, Providence, RI 02903, USA
| | - Jay Trivedi
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Ozair Meghani
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Brett D Owens
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
- Division of Sports Surgery, Department of Orthopaedic Surgery, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| | - Chathuraka T Jayasuriya
- Department of Orthopaedics, Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02903, USA
| |
Collapse
|
20
|
Wang L, Liu Z, Zhao S, Xu K, Aceves V, Qiu C, Troutwine B, Liu L, Ma S, Niu Y, Wang S, Yuan S, Li X, Zhao L, Liu X, Wu Z, Zhang TJ, Gray RS, Wu N. Variants in the SOX9 transactivation middle domain induce axial skeleton dysplasia and scoliosis. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.05.29.23290174. [PMID: 37398377 PMCID: PMC10312849 DOI: 10.1101/2023.05.29.23290174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
SOX9 is an essential transcriptional regulator of cartilage development and homeostasis. In humans, dysregulation of SOX9 is associated with a wide spectrum of skeletal disorders, including campomelic and acampomelic dysplasia, and scoliosis. The mechanism of how SOX9 variants contribute to the spectrum of axial skeletal disorders is not well understood. Here, we report four novel pathogenic variants of SOX9 identified in a large cohort of patients with congenital vertebral malformations. Three of these heterozygous variants are in the HMG and DIM domains, and for the first time, we report a pathogenic variant within the transactivation middle (TAM) domain of SOX9 . Probands with these variants exhibit variable skeletal dysplasia, ranging from isolated vertebral malformation to acampomelic dysplasia. We also generated a Sox9 hypomorphic mutant mouse model bearing a microdeletion within the TAM domain ( Sox9 Asp272del ). We demonstrated that disturbance of the TAM domain with missense mutation or microdeletion results in reduced protein stability but does not affect the transcriptional activity of SOX9. Homozygous Sox9 Asp272del mice exhibited axial skeletal dysplasia including kinked tails, ribcage anomalies, and scoliosis, recapitulating phenotypes observed in human, while heterozygous mutants display a milder phenotype. Analysis of primary chondrocytes and the intervertebral discs in Sox9 Asp272del mutant mice revealed dysregulation of a panel of genes with major contributions of the extracellular matrix, angiogenesis, and ossification-related processes. In summary, our work identified the first pathologic variant of SOX9 within the TAM domain and demonstrated that this variant is associated with reduced SOX9 protein stability. Our finding suggests that reduced SOX9 stability caused by variants in the TAM domain may be responsible for the milder forms of axial skeleton dysplasia in humans.
Collapse
|
21
|
Henke K, Farmer DT, Niu X, Kraus JM, Galloway JL, Youngstrom DW. Genetically engineered zebrafish as models of skeletal development and regeneration. Bone 2023; 167:116611. [PMID: 36395960 PMCID: PMC11080330 DOI: 10.1016/j.bone.2022.116611] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/01/2022] [Accepted: 11/08/2022] [Indexed: 11/16/2022]
Abstract
Zebrafish (Danio rerio) are aquatic vertebrates with significant homology to their terrestrial counterparts. While zebrafish have a centuries-long track record in developmental and regenerative biology, their utility has grown exponentially with the onset of modern genetics. This is exemplified in studies focused on skeletal development and repair. Herein, the numerous contributions of zebrafish to our understanding of the basic science of cartilage, bone, tendon/ligament, and other skeletal tissues are described, with a particular focus on applications to development and regeneration. We summarize the genetic strengths that have made the zebrafish a powerful model to understand skeletal biology. We also highlight the large body of existing tools and techniques available to understand skeletal development and repair in the zebrafish and introduce emerging methods that will aid in novel discoveries in skeletal biology. Finally, we review the unique contributions of zebrafish to our understanding of regeneration and highlight diverse routes of repair in different contexts of injury. We conclude that zebrafish will continue to fill a niche of increasing breadth and depth in the study of basic cellular mechanisms of skeletal biology.
Collapse
Affiliation(s)
- Katrin Henke
- Department of Orthopaedics, Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA.
| | - D'Juan T Farmer
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA; Department of Orthopaedic Surgery, University of California, Los Angeles, CA 90095, USA.
| | - Xubo Niu
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Jessica M Kraus
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| | - Jenna L Galloway
- Center for Regenerative Medicine, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Daniel W Youngstrom
- Department of Orthopaedic Surgery, University of Connecticut Health Center, Farmington, CT 06030, USA.
| |
Collapse
|
22
|
Brent AE, Buchholtz EA, Mansfield JH. Evolutionary assembly and disassembly of the mammalian sternum. Curr Biol 2023; 33:197-205.e2. [PMID: 36563692 DOI: 10.1016/j.cub.2022.11.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/15/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022]
Abstract
Evolutionary transitions are frequently associated with novel anatomical structures,1 but the origins of the structures themselves are often poorly known. We use developmental, genetic, and paleontological data to demonstrate that the therian sternum was assembled from pre-existing elements. Imaging of the perinatal mouse reveals two paired sternal elements, both composed primarily of cells with lateral plate mesoderm origin. Location, articulations, and development identify them as homologs of the interclavicle and the sternal bands of synapsid outgroups. The interclavicle, not previously recognized in therians,2 articulates with the clavicle and differs from the sternal bands in both embryonic HOX expression and pattern of skeletal maturation. The sternal bands articulate with the ribs in two styles, most clearly differentiated by their association with sternebrae. Evolutionary trait mapping indicates that the interclavicle and sternal bands were independent elements throughout most of synapsid history. The differentiation of rib articulation styles and the subdivision of the sternal bands into sternebrae were key innovations likely associated with transitions in locomotor and respiratory mechanics.3,4 Fusion of the interclavicle and the anterior sternal bands to form a presternum anterior to the first sternebra was a historically recent innovation unique to therians. Subsequent disassembly of the radically reduced sternum of mysticete cetaceans was element specific, reflecting the constraints that conserved developmental programs exert on composite structures.
Collapse
Affiliation(s)
- Ava E Brent
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, USA
| | - Emily A Buchholtz
- Department of Biological Sciences, Wellesley College, 106 Central Street, Wellesley, MA 02481, USA.
| | - Jennifer H Mansfield
- Department of Biology, Barnard College, Columbia University, 3009 Broadway, New York, NY 10027, USA
| |
Collapse
|
23
|
Au TYK, Yip RKH, Wynn SL, Tan TY, Fu A, Geng YH, Szeto IYY, Niu B, Yip KY, Cheung MCH, Lovell-Badge R, Cheah KSE. Hypomorphic and dominant-negative impact of truncated SOX9 dysregulates Hedgehog-Wnt signaling, causing campomelia. Proc Natl Acad Sci U S A 2023; 120:e2208623119. [PMID: 36584300 PMCID: PMC9910594 DOI: 10.1073/pnas.2208623119] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/02/2022] [Indexed: 01/01/2023] Open
Abstract
Haploinsufficiency for SOX9, the master chondrogenesis transcription factor, can underlie campomelic dysplasia (CD), an autosomal dominant skeletal malformation syndrome, because heterozygous Sox9 null mice recapitulate the bent limb (campomelia) and some other phenotypes associated with CD. However, in vitro cell assays suggest haploinsufficiency may not apply for certain mutations, notably those that truncate the protein, but in these cases in vivo evidence is lacking and underlying mechanisms are unknown. Here, using conditional mouse mutants, we compared the impact of a heterozygous Sox9 null mutation (Sox9+/-) with the Sox9+/Y440X CD mutation that truncates the C-terminal transactivation domain but spares the DNA-binding domain. While some Sox9+/Y440X mice survived, all Sox9+/- mice died perinatally. However, the skeletal defects were more severe and IHH signaling in developing limb cartilage was significantly enhanced in Sox9+/Y440X compared with Sox9+/-. Activating Sox9Y440X specifically in the chondrocyte-osteoblast lineage caused milder campomelia, and revealed cell- and noncell autonomous mechanisms acting on chondrocyte differentiation and osteogenesis in the perichondrium. Transcriptome analyses of developing Sox9+/Y440X limbs revealed dysregulated expression of genes for the extracellular matrix, as well as changes consistent with aberrant WNT and HH signaling. SOX9Y440X failed to interact with β-catenin and was unable to suppress transactivation of Ihh in cell-based assays. We propose enhanced HH signaling in the adjacent perichondrium induces asymmetrically localized excessive perichondrial osteogenesis resulting in campomelia. Our study implicates combined haploinsufficiency/hypomorphic and dominant-negative actions of SOX9Y440X, cell-autonomous and noncell autonomous mechanisms, and dysregulated WNT and HH signaling, as the cause of human campomelia.
Collapse
Affiliation(s)
- Tiffany Y. K. Au
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Raymond K. H. Yip
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Sarah L. Wynn
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Tiong Y. Tan
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Alex Fu
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, New Territories, Shatin, Hong Kong SAR, China
| | - Yu Hong Geng
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Irene Y. Y. Szeto
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Ben Niu
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Kevin Y. Yip
- Department of Computer Science and Engineering, The Chinese University of Hong Kong, New Territories, Shatin, Hong Kong SAR, China
| | - Martin C. H. Cheung
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | | | - Kathryn S. E. Cheah
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| |
Collapse
|
24
|
Szeto IYY, Chu DKH, Chen P, Chu KC, Au TYK, Leung KKH, Huang YH, Wynn SL, Mak ACY, Chan YS, Chan WY, Jauch R, Fritzsch B, Sham MH, Lovell-Badge R, Cheah KSE. SOX9 and SOX10 control fluid homeostasis in the inner ear for hearing through independent and cooperative mechanisms. Proc Natl Acad Sci U S A 2022; 119:e2122121119. [PMID: 36343245 PMCID: PMC9674217 DOI: 10.1073/pnas.2122121119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 09/10/2022] [Indexed: 11/09/2022] Open
Abstract
The in vivo mechanisms underlying dominant syndromes caused by mutations in SRY-Box Transcription Factor 9 (SOX9) and SOX10 (SOXE) transcription factors, when they either are expressed alone or are coexpressed, are ill-defined. We created a mouse model for the campomelic dysplasia SOX9Y440X mutation, which truncates the transactivation domain but leaves DNA binding and dimerization intact. Here, we find that SOX9Y440X causes deafness via distinct mechanisms in the endolymphatic sac (ES)/duct and cochlea. By contrast, conditional heterozygous Sox9-null mice are normal. During the ES development of Sox9Y440X/+ heterozygotes, Sox10 and genes important for ionic homeostasis are down-regulated, and there is developmental persistence of progenitors, resulting in fewer mature cells. Sox10 heterozygous null mutants also display persistence of ES/duct progenitors. By contrast, SOX10 retains its expression in the early Sox9Y440X/+ mutant cochlea. Later, in the postnatal stria vascularis, dominant interference by SOX9Y440X is implicated in impairing the normal cooperation of SOX9 and SOX10 in repressing the expression of the water channel Aquaporin 3, thereby contributing to endolymphatic hydrops. Our study shows that for a functioning endolymphatic system in the inner ear, SOX9 regulates Sox10, and depending on the cell type and target gene, it works either independently of or cooperatively with SOX10. SOX9Y440X can interfere with the activity of both SOXE factors, exerting effects that can be classified as haploinsufficient/hypomorphic or dominant negative depending on the cell/gene context. This model of disruption of transcription factor partnerships may be applicable to congenital deafness, which affects ∼0.3% of newborns, and other syndromic disorders.
Collapse
Affiliation(s)
- Irene Y. Y. Szeto
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Daniel K. H. Chu
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Peikai Chen
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Ka Chi Chu
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Tiffany Y. K. Au
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Keith K. H. Leung
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Yong-Heng Huang
- Genome Regulation Laboratory, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou Medical University, Guangzhou 511436, China
| | - Sarah L. Wynn
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Angel C. Y. Mak
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Ying-Shing Chan
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | - Wood Yee Chan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Ralf Jauch
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
- Genome Regulation Laboratory, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
- Guangzhou Medical University, Guangzhou 511436, China
| | - Bernd Fritzsch
- Department of Biology, College of Arts & Sciences, University of Iowa, Iowa City, IA 52242
- Department of Otolaryngology, College of Arts & Sciences, University of Iowa, Iowa City, IA 52242
| | - Mai Har Sham
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| | | | - Kathryn S. E. Cheah
- School of Biomedical Sciences, The University of Hong Kong, Li Ka Shing Faculty of Medicine, Hong Kong, China
| |
Collapse
|
25
|
Ming Z, Vining B, Bagheri-Fam S, Harley V. SOX9 in organogenesis: shared and unique transcriptional functions. Cell Mol Life Sci 2022; 79:522. [PMID: 36114905 PMCID: PMC9482574 DOI: 10.1007/s00018-022-04543-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 08/13/2022] [Accepted: 08/31/2022] [Indexed: 11/28/2022]
Abstract
The transcription factor SOX9 is essential for the development of multiple organs including bone, testis, heart, lung, pancreas, intestine and nervous system. Mutations in the human SOX9 gene led to campomelic dysplasia, a haploinsufficiency disorder with several skeletal malformations frequently accompanied by 46, XY sex reversal. The mechanisms underlying the diverse SOX9 functions during organ development including its post-translational modifications, the availability of binding partners, and tissue-specific accessibility to target gene chromatin. Here we summarize the expression, activities, and downstream target genes of SOX9 in molecular genetic pathways essential for organ development, maintenance, and function. We also provide an insight into understanding the mechanisms that regulate the versatile roles of SOX9 in different organs.
Collapse
Affiliation(s)
- Zhenhua Ming
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - Brittany Vining
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - Stefan Bagheri-Fam
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia
| | - Vincent Harley
- Sex Development Laboratory, Hudson Institute of Medical Research, PO Box 5152, Melbourne, VIC, 3168, Australia.
- Department of Molecular and Translational Science, Monash University, Melbourne, VIC, 3800, Australia.
| |
Collapse
|
26
|
Jiang T, Hu W, Zhang S, Ren C, Lin S, Zhou Z, Wu H, Yin J, Tan L. Fibroblast growth factor 10 attenuates chronic obstructive pulmonary disease by protecting against glycocalyx impairment and endothelial apoptosis. Respir Res 2022; 23:269. [PMID: 36183124 PMCID: PMC9526324 DOI: 10.1186/s12931-022-02193-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 09/22/2022] [Indexed: 11/23/2022] Open
Abstract
Background The defects and imbalance in lung repair and structural maintenance contribute to the pathogenesis of chronic obstructive pulmonary diseases (COPD), yet the molecular mechanisms that regulate lung repair process are so far incompletely understood. We hypothesized that cigarette smoking causes glycocalyx impairment and endothelial apoptosis in COPD, which could be repaired by the stimulation of fibroblast growth factor 10 (FGF10)/FGF receptor 1 (FGFR1) signaling. Methods We used immunostaining (immunohistochemical [IHC] and immunofluorescence [IF]) and enzyme-linked immunosorbent assay (ELISA) to detect the levels of glycocalyx components and endothelial apoptosis in animal models and in patients with COPD. We used the murine emphysema model and in vitro studies to determine the protective and reparative role of FGF10/FGFR1. Results Exposure to cigarette smoke caused endothelial glycocalyx impairment and emphysematous changes in murine models and human specimens. Pretreatment of FGF10 attenuated the development of emphysema and the shedding of glycocalyx components induced by CSE in vivo. However, FGF10 did not attenuate the emphysema induced by endothelial-specific killing peptide CGSPGWVRC-GG-D(KLAKLAK)2. Mechanistically, FGF10 alleviated smoke-induced endothelial apoptosis and glycocalyx repair through FGFR1/ERK/SOX9/HS6ST1 signaling in vitro. FGF10 was shown to repair pulmonary glycocalyx injury and endothelial apoptosis, and attenuate smoke-induced COPD through FGFR1 signaling. Conclusions Our results suggest that FGF10 may serve as a potential therapeutic strategy against COPD via endothelial repair and glycocalyx reconstitution. Supplementary Information The online version contains supplementary material available at 10.1186/s12931-022-02193-5. It is the first time to prove the confirm the endothelial glycocalyx impairment in COPD. FGF10 attenuates the development of emphysema and the shedding of glycocalyx induced by CSE in vivo. FGF10 alleviates smoke-induced endothelial apoptosis and glycocalyx repair through FGFR1/ERK/SOX9/HS6ST1 signaling.
Collapse
Affiliation(s)
- Tian Jiang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China
| | - Weiping Hu
- Department of Critical Care and Respiratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Shaoyuan Zhang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China.,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China
| | - Changhao Ren
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Siyun Lin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China
| | - Zhenyu Zhou
- Department of Vascular Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Hao Wu
- Department of Clinical Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Jun Yin
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China.
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, No.180 Fenglin Road, Shanghai, 200032, China. .,Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, China. .,Key Laboratory of Lung Inflammation and Injury, Shanghai, 200032, China.
| |
Collapse
|
27
|
Saito T, Nakamichi R, Yoshida A, Hiranaka T, Okazaki Y, Nezu S, Matsuhashi M, Shimamura Y, Furumatsu T, Nishida K, Ozaki T. The effect of mechanical stress on enthesis homeostasis in a rat Achilles enthesis organ culture model. J Orthop Res 2022; 40:1872-1882. [PMID: 34783068 DOI: 10.1002/jor.25210] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 10/19/2021] [Accepted: 10/30/2021] [Indexed: 02/04/2023]
Abstract
Tendons and ligaments are jointed to bones via an enthesis that is essential to the proper function of the muscular and skeletal structures. The aim of the study is to investigate the effect of mechanical stress on the enthesis. We used ex vivo models in organ cultures of rat Achilles tendons with calcaneus including the enthesis. The organ was attached to a mechanical stretching apparatus that can conduct cyclic tensile strain. We made the models of 1-mm elongation (0.5 Hz, 3% elongation), 2-mm elongation (0.5 Hz, 5% elongation), and no stress. Histological evaluation by Safranin O staining and Toluidin Blue and Picro Sirius red staining was conducted. Expression of sex-determining region Y-box 9 (Sox9), scleraxis (Scx), Runt-related transcription factor 2 (Runx2), and matrix metalloproteinase 13 (Mmp13) were examined by real-time polymerase chain reaction and immunocytochemistry. Terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate biotin nick end-labeling and live/dead staining and was conducted for evaluation of the apoptosis and cell viability. The structure of the enthesis was most maintained in the model of 1-mm elongation. The electronic microscope showed that the enthesis of the no stress model had ill-defined borders between fibrocartilage and mineralized fibrocartilage, and that calcification of mineralized fibrocartilage occurred in the model of 2-mm elongation. Sox9 and Scx was upregulated by 1-mm elongation, whereas Runx2 and Mmp13 were upregulated by 2-mm elongation. Apoptosis was inhibited by low stress. The results of this study suggested that 1-mm elongation can maintain the structure of the enthesis, while 2-mm elongation promotes degenerative changes.
Collapse
Affiliation(s)
- Taichi Saito
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Ryo Nakamichi
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Aki Yoshida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Takaaki Hiranaka
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yuki Okazaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Nezu
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Minami Matsuhashi
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yasunori Shimamura
- Department of Sports Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Furumatsu
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Keiichiro Nishida
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Toshifumi Ozaki
- Department of Orthopaedic Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
28
|
Lin Y, Wang D, Zeng Y. A Maverick Review of Common Stem/Progenitor Markers in Lung Development. Stem Cell Rev Rep 2022; 18:2629-2645. [DOI: 10.1007/s12015-022-10422-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 10/16/2022]
|
29
|
Sreenivasan R, Gonen N, Sinclair A. SOX Genes and Their Role in Disorders of Sex Development. Sex Dev 2022; 16:80-91. [PMID: 35760052 DOI: 10.1159/000524453] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 03/29/2022] [Indexed: 11/19/2022] Open
Abstract
SOX genesare master regulatory genes controlling development and are fundamental to the establishment of sex determination in a multitude of organisms. The discovery of the master sex-determining gene SRY in 1990 was pivotal for the understanding of how testis development is initiated in mammals. With this discovery, an entire family of SOX factors were uncovered that play crucial roles in cell fate decisions during development. The importance of SOX genes in human reproductive development is evident from the various disorders of sex development (DSD) upon loss or overexpression of SOX gene function. Here, we review the roles that SOX genes play in gonad development and their involvement in DSD. We start with an overview of sex determination and differentiation, DSDs, and the SOX gene family and function. We then provide detailed information and discussion on SOX genes that have been implicated in DSDs, both at the gene and regulatory level. These include SRY, SOX9, SOX3, SOX8, and SOX10. This review provides insights on the crucial balance of SOX gene expression levels needed for gonad development and maintenance and how changes in these levels can lead to DSDs.
Collapse
Affiliation(s)
- Rajini Sreenivasan
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Nitzan Gonen
- The Mina and Everard Goodman Faculty of Life Sciences, Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Andrew Sinclair
- Reproductive Development, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
30
|
Fujii Y, Liu L, Yagasaki L, Inotsume M, Chiba T, Asahara H. Cartilage Homeostasis and Osteoarthritis. Int J Mol Sci 2022; 23:6316. [PMID: 35682994 PMCID: PMC9181530 DOI: 10.3390/ijms23116316] [Citation(s) in RCA: 94] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 05/29/2022] [Accepted: 06/03/2022] [Indexed: 01/27/2023] Open
Abstract
Healthy limb joints are important for maintaining health and attaining longevity. Endochondral ossification (the replacement of cartilage with bone, occurring during skeletal development) is essential for bone formation, especially in long-axis bones. In contrast to endochondral ossification, chondrocyte populations in articular cartilage persist and maintain joint tissue into adulthood. Articular cartilage, a connective tissue consisting of chondrocytes and their surrounding extracellular matrices, plays an essential role in the mechanical cushioning of joints in postnatal locomotion. Osteoarthritis (OA) pathology relates to disruptions in the balance between anabolic and catabolic signals, that is, the loss of chondrocyte homeostasis due to aging or overuse of cartilages. The onset of OA increases with age, shortening a person's healthy life expectancy. Although many people with OA experience pain, the mainstay of treatment is symptomatic therapy, and no fundamental treatment has yet been established. To establish regenerative or preventative therapies for cartilage diseases, further understanding of the mechanisms of cartilage development, morphosis, and homeostasis is required. In this review, we describe the general development of cartilage and OA pathology, followed by a discussion on anabolic and catabolic signals in cartilage homeostasis, mainly microRNAs.
Collapse
Affiliation(s)
- Yuta Fujii
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8501, Japan; (Y.F.); (L.L.); (L.Y.); (M.I.); (T.C.)
| | - Lin Liu
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8501, Japan; (Y.F.); (L.L.); (L.Y.); (M.I.); (T.C.)
| | - Lisa Yagasaki
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8501, Japan; (Y.F.); (L.L.); (L.Y.); (M.I.); (T.C.)
- Department of Periodontology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-851, Japan
| | - Maiko Inotsume
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8501, Japan; (Y.F.); (L.L.); (L.Y.); (M.I.); (T.C.)
| | - Tomoki Chiba
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8501, Japan; (Y.F.); (L.L.); (L.Y.); (M.I.); (T.C.)
| | - Hiroshi Asahara
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo 113-8501, Japan; (Y.F.); (L.L.); (L.Y.); (M.I.); (T.C.)
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| |
Collapse
|
31
|
Yang X, Tian S, Fan L, Niu R, Yan M, Chen S, Zheng M, Zhang S. Integrated regulation of chondrogenic differentiation in mesenchymal stem cells and differentiation of cancer cells. Cancer Cell Int 2022; 22:169. [PMID: 35488254 PMCID: PMC9052535 DOI: 10.1186/s12935-022-02598-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Chondrogenesis is the formation of chondrocytes and cartilage tissues and starts with mesenchymal stem cell (MSC) recruitment and migration, condensation of progenitors, chondrocyte differentiation, and maturation. The chondrogenic differentiation of MSCs depends on co-regulation of many exogenous and endogenous factors including specific microenvironmental signals, non-coding RNAs, physical factors existed in culture condition, etc. Cancer stem cells (CSCs) exhibit self-renewal capacity, pluripotency and cellular plasticity, which have the potential to differentiate into post-mitotic and benign cells. Accumulating evidence has shown that CSCs can be induced to differentiate into various benign cells including adipocytes, fibrocytes, osteoblast, and so on. Retinoic acid has been widely used in the treatment of acute promyelocytic leukemia. Previous study confirmed that polyploid giant cancer cells, a type of cancer stem-like cells, could differentiate into adipocytes, osteocytes, and chondrocytes. In this review, we will summarize signaling pathways and cytokines in chondrogenic differentiation of MSCs. Understanding the molecular mechanism of chondrogenic differentiation of CSCs and cancer cells may provide new strategies for cancer treatment.
Collapse
Affiliation(s)
- Xiaohui Yang
- Nankai University School of Medicine, Nankai University, Tianjin, 300071 People’s Republic of China
| | - Shifeng Tian
- Graduate School, Tianjin Medical University, Tianjin, 300070 People’s Republic of China
| | - Linlin Fan
- Department of Pathology, Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617 People’s Republic of China
| | - Rui Niu
- Department of Pathology, Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617 People’s Republic of China
| | - Man Yan
- Department of Pathology, Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617 People’s Republic of China
| | - Shuo Chen
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, People’s Republic of China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071 People’s Republic of China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300071 People’s Republic of China
| |
Collapse
|
32
|
Live imaging approach of dynamic multicellular responses in ERK signaling during vertebrate tissue development. Biochem J 2022; 479:129-143. [PMID: 35050327 PMCID: PMC8883488 DOI: 10.1042/bcj20210557] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 11/17/2022]
Abstract
The chemical and mechanical responses of cells via the exchange of information during growth and development result in the formation of biological tissues. Information processing within the cells through the signaling pathways and networks inherent to the constituent cells has been well-studied. However, the cell signaling mechanisms responsible for generating dynamic multicellular responses in developing tissues remain unclear. Here, I review the dynamic multicellular response systems during the development and growth of vertebrate tissues based on the extracellular signal-regulated kinase (ERK) pathway. First, an overview of the function of the ERK signaling network in cells is provided, followed by descriptions of biosensors essential for live imaging of the quantification of ERK activity in tissues. Then adducing four examples, I highlight the contribution of live imaging techniques for studying the involvement of spatio-temporal patterns of ERK activity change in tissue development and growth. In addition, theoretical implications of ERK signaling are also discussed from the viewpoint of dynamic systems. This review might help in understanding ERK-mediated dynamic multicellular responses and tissue morphogenesis.
Collapse
|
33
|
De la Fuente-Hernandez MA, Sarabia-Sanchez MA, Melendez-Zajgla J, Maldonado-Lagunas V. Role of lncRNAs into Mesenchymal Stromal Cell Differentiation. Am J Physiol Cell Physiol 2022; 322:C421-C460. [PMID: 35080923 DOI: 10.1152/ajpcell.00364.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Currently, findings support that 75% of the human genome is actively transcribed, but only 2% is translated into a protein, according to databases such as ENCODE (Encyclopedia of DNA Elements) [1]. The development of high-throughput sequencing technologies, computational methods for genome assembly and biological models have led to the realization of the importance of the previously unconsidered non-coding fraction of the genome. Along with this, noncoding RNAs have been shown to be epigenetic, transcriptional and post-transcriptional regulators in a large number of cellular processes [2]. Within the group of non-coding RNAs, lncRNAs represent a fascinating field of study, given the functional versatility in their mode of action on their molecular targets. In recent years, there has been an interest in learning about lncRNAs in MSC differentiation. The aim of this review is to address the signaling mechanisms where lncRNAs are involved, emphasizing their role in either stimulating or inhibiting the transition to differentiated cell. Specifically, the main types of MSC differentiation are discussed: myogenesis, osteogenesis, adipogenesis and chondrogenesis. The description of increasingly new lncRNAs reinforces their role as players in the well-studied field of MSC differentiation, allowing a step towards a better understanding of their biology and their potential application in the clinic.
Collapse
Affiliation(s)
- Marcela Angelica De la Fuente-Hernandez
- Facultad de Medicina, Posgrado en Ciencias Biológicas, Universidad Nacional Autónoma de México, Mexico City, Mexico.,Laboratorio de Epigenética, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Miguel Angel Sarabia-Sanchez
- Facultad de Medicina, Posgrado en Ciencias Bioquímicas, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Jorge Melendez-Zajgla
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | | |
Collapse
|
34
|
Hoyle DJ, Dranow DB, Schilling TF. Pthlha and mechanical force control early patterning of growth zones in the zebrafish craniofacial skeleton. Development 2022; 149:dev199826. [PMID: 34919126 PMCID: PMC8917414 DOI: 10.1242/dev.199826] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 12/07/2021] [Indexed: 11/23/2022]
Abstract
Secreted signals in patterning systems often induce repressive signals that shape their distributions in space and time. In developing growth plates (GPs) of endochondral long bones, Parathyroid hormone-like hormone (Pthlh) inhibits Indian hedgehog (Ihh) to form a negative-feedback loop that controls GP progression and bone size. Whether similar systems operate in other bones and how they arise during embryogenesis remain unclear. We show that Pthlha expression in the zebrafish craniofacial skeleton precedes chondrocyte differentiation and restricts where cells undergo hypertrophy, thereby initiating a future GP. Loss of Pthlha leads to an expansion of cells expressing a novel early marker of the hypertrophic zone (HZ), entpd5a, and later HZ markers, such as ihha, whereas local Pthlha misexpression induces ectopic entpd5a expression. Formation of this early pre-HZ correlates with onset of muscle contraction and requires mechanical force; paralysis leads to loss of entpd5a and ihha expression in the pre-HZ, mislocalized pthlha expression and no subsequent ossification. These results suggest that local Pthlh sources combined with force determine HZ locations, establishing the negative-feedback loop that later maintains GPs.
Collapse
Affiliation(s)
| | | | - Thomas F. Schilling
- Department of Developmental and Cell Biology, University of California, Irvine, CA 92693, USA
| |
Collapse
|
35
|
Qiao X, Wu L, Tang J, Xiang R, Fan L, Huang H, Chen Y. Case report: A de novo Non-sense SOX9 mutation (p.Q417*) located in transactivation domain is Responsible for Campomelic Dysplasia. Front Pediatr 2022; 10:1089194. [PMID: 36741086 PMCID: PMC9890166 DOI: 10.3389/fped.2022.1089194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 12/28/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Campomelic dysplasia (CD) is an autosomal dominant skeletal dysplasia syndrome characterized by shortness and bowing of lower extremities, and often accompanied by XY sex reversal. Heterozygous pathogenic variants of SOX9 or rearrangement involving the long arm of chromosome 17 are the causes of disease. However, evidence for pathogenesis of SOX9 haploinsufficiency is insufficient. METHODS We enrolled a Chinese family where the fetus was diagnosed with CD. The affected fetus was selected for whole-exome sequencing to identify the pathogenic mutations in this family. RESULTS After data filtering, a novel non-sense SOX9 variant (NM_000346.3; c.1249C > T; p.Q417*) was identified as the pathogenic lesion in the fetus. Further co-segregation analysis using Sanger sequencing confirmed that this novel SOX9 mutation (c.1249C > T; p.Q417*) was a de novo mutation in the affected fetus. This terminated codon mutation identified by bioinformatics was located at an evolutionarily conserved site of SOX9. The bioinformatics-based analysis predicted this variant was pathogenic and affected SOX9 transactivation activity. CONCLUSION CD is a rare condition, which connected with SOX9 tightly. We identified a novel heterozygous SOX9 variant (p.Q417*) in a Chinese CD family. Our study supports the putative reduced transactivation of SOX9 variants in the pathogenicity of CD.
Collapse
Affiliation(s)
- Xingxing Qiao
- Department of Cardiology, Second Xiangya Hospital Central South University, Changsha, China
| | - Liping Wu
- Department of Medical Genetics and Prenatal Diagnosis, Longgang District Maternity and Child Healthcare Hospital, Shenzhen, China.,Obstetric Inpatient Department, Shenzhen Longgang District Maternity and Child Healthcare Hospital, Shenzhen, China
| | - Jianjun Tang
- Department of Cardiology, Second Xiangya Hospital Central South University, Changsha, China
| | - Rong Xiang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Liangliang Fan
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Hao Huang
- Department of Nephrology, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Yaqin Chen
- Department of Cardiology, Second Xiangya Hospital Central South University, Changsha, China
| |
Collapse
|
36
|
Liang F, Peng L, Ma YG, Hu W, Zhang WB, Deng M, Li YM. Bioinformatics analysis and experimental validation of differentially expressed genes in mouse articular chondrocytes treated with IL-1β using microarray data. Exp Ther Med 2022; 23:6. [PMID: 34815758 PMCID: PMC8593859 DOI: 10.3892/etm.2021.10928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 02/02/2021] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic degenerative disease that affects the health of the elderly. The present study aimed to identify significant genes involved in OA via bioinformatics analysis. A gene expression dataset (GSE104793) was downloaded from the Gene Expression Omnibus. Bioinformatics analysis was then performed in order to identify differentially expressed genes (DEGs) between untreated chondrocytes and chondrocytes cultured with interleukin-1β (IL-1β) for 24 h. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were performed using Metascape. A protein-protein interaction network of DEGs was constructed using the Search Tool for the Retrieval of Interacting Genes. Gene set enrichment analysis (GSEA) was performed using GSEA software. Furthermore, chondrocytes were extracted and treated with IL-1β (10 ng/ml) for 24 h, and reverse-transcription quantitative PCR was used to confirm differential expression of hub genes. Patient samples were also collected to verify the bioinformatic analysis results. Based on the cut-off criteria used for determination of the DEGs, a total of 844 DEGs, including 498 upregulated and 346 downregulated DEGs, were identified. The DEGs were mainly enriched in the GO terms and KEGG pathways 'inflammatory response', 'negative regulation of cell proliferation', 'ossification', 'taxis', 'blood vessel morphogenesis', 'extracellular structure organization', 'mitotic cell cycle process' and 'TNF signaling pathway'. The majority of the PCR results, namely the differential expression of kininogen 2, complement C3, cyclin B1, cell division cycle 20, cyclin A2, 1-phosphatidylinositol 4-kinase, BUB1 mitotic checkpoint serine/threonine kinase, kinesin family member 11, cyclin B2 and BUB1 mitotic checkpoint serine/threonine kinase B were consistent with the bioinformatics results. Collectively, the present observations provided a regulation network of IL-1β-stimulated chondrocytes, which may provide potential targets of OA therapy.
Collapse
Affiliation(s)
- Fan Liang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Le Peng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yong-Gang Ma
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei Hu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Wei-Bing Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ming Deng
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Ya-Ming Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
37
|
Sex Chromosomes and Master Sex-Determining Genes in Turtles and Other Reptiles. Genes (Basel) 2021; 12:genes12111822. [PMID: 34828428 PMCID: PMC8622242 DOI: 10.3390/genes12111822] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 11/12/2021] [Accepted: 11/16/2021] [Indexed: 11/24/2022] Open
Abstract
Among tetrapods, the well differentiated heteromorphic sex chromosomes of birds and mammals have been highly investigated and their master sex-determining (MSD) gene, Dmrt1 and SRY, respectively, have been identified. The homomorphic sex chromosomes of reptiles have been the least studied, but the gap with birds and mammals has begun to fill. This review describes our current knowledge of reptilian sex chromosomes at the cytogenetic and molecular level. Most of it arose recently from various studies comparing male to female gene content. This includes restriction site-associated DNA sequencing (RAD-Seq) experiments in several male and female samples, RNA sequencing and identification of Z- or X-linked genes by male/female comparative transcriptome coverage, and male/female transcriptomic or transcriptome/genome substraction approaches allowing the identification of Y- or W-linked transcripts. A few putative master sex-determining (MSD) genes have been proposed, but none has been demonstrated yet. Lastly, future directions in the field of reptilian sex chromosomes and their MSD gene studies are considered.
Collapse
|
38
|
Fukui Y, Hayano S, Kawanabe N, Wang Z, Shimada A, Saito MK, Asaka I, Kamioka H. Investigation of the molecular causes underlying physical abnormalities in Diamond-Blackfan anemia patients with RPL5 haploinsufficiency. Pathol Int 2021; 71:803-813. [PMID: 34587661 DOI: 10.1111/pin.13168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/12/2021] [Indexed: 01/05/2023]
Abstract
Diamond-Blackfan anemia (DBA) is a genetic disorder caused by mutations in genes encoding ribosomal proteins and characterized by erythroid aplasia and various physical abnormalities. Although accumulating evidence suggests that defective ribosome biogenesis leads to p53-mediated apoptosis in erythroid progenitor cells, little is known regarding the underlying causes of the physical abnormalities. In this study, we established induced pluripotent stem cells from a DBA patient with RPL5 haploinsufficiency. These cells retained the ability to differentiate into osteoblasts and chondrocytes. However, RPL5 haploinsufficiency impaired the production of mucins and increased apoptosis in differentiated chondrocytes. Increased expression of the pro-apoptotic genes BAX and CASP9 further indicated that RPL5 haploinsufficiency triggered p53-mediated apoptosis in chondrocytes. Murine double minute 2 (MDM2), the primary negative regulator of p53, plays a crucial role in erythroid aplasia in DBA patient. We found the phosphorylation level of MDM2 was significantly decreased in RPL5 haploinsufficient chondrocytes. In stark contrast, we found no evidence that RPL5 haploinsufficiency impaired osteogenesis. Collectively, our data support a model in which RPL5 haploinsufficiency specifically induces p53-mediated apoptosis in chondrocytes through MDM2 inhibition, which leads to physical abnormalities in DBA patients.
Collapse
Affiliation(s)
- Yuko Fukui
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Satoru Hayano
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Department of Orthodontics, Okayama University Hospital, Okayama, Japan
| | - Noriaki Kawanabe
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Ziyi Wang
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Akira Shimada
- Department of Pediatric Hematology/Oncology, Okayama University Hospital, Okayama, Japan.,Department of Pediatrics, Jichi Medical University, Tochigi, Japan
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Isao Asaka
- Department of Fundamental Cell Technology, Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Hiroshi Kamioka
- Department of Orthodontics, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
39
|
Bao J, Närhi K, Teodòsio A, Hemmes A, Linnavirta NM, Mäyränpää MI, Salmenkivi K, Le Quesne J, Verschuren EW. SOX9 has distinct roles in the formation and progression of different non-small cell lung cancer histotypes. J Pathol 2021; 255:16-29. [PMID: 34021911 PMCID: PMC11497254 DOI: 10.1002/path.5733] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 04/25/2021] [Accepted: 05/19/2021] [Indexed: 12/16/2022]
Abstract
The transcription factor SOX9 is a key regulator of multiple developmental processes and is frequently re-expressed in non-small cell lung cancer (NSCLC). Its precise role in the progression of NSCLC histotypes has, however, remained elusive. We show that SOX9 expression relates to poor overall survival and invasive histopathology in human non-mucinous adenocarcinoma and is absent in murine early minimally invasive and low in human in situ adenocarcinoma. Interestingly, despite wide SOX9 expression across advanced NSCLC histotypes, its genetic deletion in the murine KrasG12D ;Lkb1fl/fl model selectively disrupted only the growth of papillary NSCLC, without affecting the initiation of precursor lesions or growth of mucinous or squamous tissue. Spatial tissue phenotyping indicated a requirement of SOX9 expression for the progression of surfactant protein C-expressing progenitor cells, which gave rise to papillary tumours. Intriguingly, while SOX9 expression was dispensable for squamous tissue formation, its loss in fact led to enhanced squamous tumour metastasis, which was associated with altered collagen IV deposition in the basement membrane. Our work therefore demonstrates histopathology-selective roles for SOX9 in NSCLC progression, namely as a promoter for papillary adenocarcinoma progression, but an opposing metastasis-suppressing role in squamous histotype tissue. This attests to a pleiotropic SOX9 function, linked to the cell of origin and microenvironmental tissue contexts. © 2021 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jie Bao
- Institute for Molecular Medicine Finland, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Katja Närhi
- Institute for Molecular Medicine Finland, HiLIFEUniversity of HelsinkiHelsinkiFinland
- GlaxoSmithKlineEspooFinland
| | - Ana Teodòsio
- MRC Toxicology UnitUniversity of CambridgeCambridgeUK
| | - Annabrita Hemmes
- Institute for Molecular Medicine Finland, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Nora M Linnavirta
- Institute for Molecular Medicine Finland, HiLIFEUniversity of HelsinkiHelsinkiFinland
| | - Mikko I Mäyränpää
- HUSLAB, Division of PathologyHelsinki University HospitalHelsinkiFinland
- Department of PathologyUniversity of HelsinkiHelsinkiFinland
| | | | - John Le Quesne
- MRC Toxicology UnitUniversity of CambridgeCambridgeUK
- Leicester Cancer Research CentreUniversity of LeicesterLeicesterUK
| | - Emmy W Verschuren
- Institute for Molecular Medicine Finland, HiLIFEUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
40
|
Machado RA, Martelli-Junior H, Reis SRDA, Küchler EC, Scariot R, das Neves LT, Coletta RD. Identification of Novel Variants in Cleft Palate-Associated Genes in Brazilian Patients With Non-syndromic Cleft Palate Only. Front Cell Dev Biol 2021; 9:638522. [PMID: 34307341 PMCID: PMC8297955 DOI: 10.3389/fcell.2021.638522] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 04/30/2021] [Indexed: 12/17/2022] Open
Abstract
The identification of genetic risk factors for non-syndromic oral clefts is of great importance for better understanding the biological processes related to this heterogeneous and complex group of diseases. Herein we applied whole-exome sequencing to identify potential variants related to non-syndromic cleft palate only (NSCPO) in the multiethnic Brazilian population. Thirty NSCPO samples and 30 sex- and genetic ancestry-matched healthy controls were pooled (3 pools with 10 samples for each group) and subjected to whole-exome sequencing. After filtering, the functional affects, individually and through interactions, of the selected variants and genes were assessed by bioinformatic analyses. As a group, 399 variants in 216 genes related to palatogenesis/cleft palate, corresponding to 6.43%, were exclusively identified in the NSCPO pools. Among those genes are 99 associated with syndromes displaying cleft palate in their clinical spectrum and 92 previously related to cleft lip palate. The most significantly biological processes and pathways overrepresented in the NSCPO-identified genes were associated with the folic acid metabolism, highlighting the interaction between LDL receptor-related protein 6 (LRP6) and 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR) that interconnect two large networks. This study yields novel data on characterization of specific variants and complex processes and pathways related to NSCPO, including many variants in genes of the folate/homocysteine pathway, and confirms that variants in genes related to syndromic cleft palate and cleft lip-palate may cause NSCPO.
Collapse
Affiliation(s)
- Renato Assis Machado
- Department of Oral Diagnosis, School of Dentistry, University of Campinas (FOP), Piracicaba, Brazil.,Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo, Bauru, Brazil
| | - Hercílio Martelli-Junior
- Stomatology Clinic, School of Dental, State University of Montes Claros, Montes Claros, Brazil.,Center for Rehabilitation of Craniofacial Anomalies, School of Dental, UNIFENAS - Universidade José do Rosario Vellano, Alfenas, Brazil
| | | | | | - Rafaela Scariot
- Department of Oral and Maxillofacial Surgery, School of Health Science, Federal University of Paraná, Curitiba, Brazil
| | - Lucimara Teixeira das Neves
- Hospital for Rehabilitation of Craniofacial Anomalies, University of São Paulo, Bauru, Brazil.,Department of Biological Sciences, Bauru School of Dentistry, University of São Paulo (FOB), Bauru, Brazil
| | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas (FOP), Piracicaba, Brazil
| |
Collapse
|
41
|
Yadav PS, Feng S, Cong Q, Kim H, Liu Y, Yang Y. Stat3 loss in mesenchymal progenitors causes Job syndrome-like skeletal defects by reducing Wnt/β-catenin signaling. Proc Natl Acad Sci U S A 2021; 118:e2020100118. [PMID: 34172578 PMCID: PMC8256036 DOI: 10.1073/pnas.2020100118] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Job syndrome is a rare genetic disorder caused by STAT3 mutations and primarily characterized by immune dysfunction along with comorbid skeleton developmental abnormalities including osteopenia, recurrent fracture of long bones, and scoliosis. So far, there is no definitive cure for the skeletal defects in Job syndrome, and treatments are limited to management of clinical symptoms only. Here, we have investigated the molecular mechanism whereby Stat3 regulates skeletal development and osteoblast differentiation. We showed that removing Stat3 function in the developing limb mesenchyme or osteoprogenitor cells in mice resulted in shortened and bow limbs with multiple fractures in long bones that resembled the skeleton symptoms in the Job Syndrome. However, Stat3 loss did not alter chondrocyte differentiation and hypertrophy in embryonic development, while osteoblast differentiation was severely reduced. Genome-wide transcriptome analyses as well as biochemical and histological studies showed that Stat3 loss resulted in down-regulation of Wnt/β-catenin signaling. Restoration of Wnt/β-catenin signaling by injecting BIO, a small molecule inhibitor of GSK3, or crossing with a Lrp5 gain of function (GOF) allele, rescued the bone reduction phenotypes due to Stat3 loss to a great extent. These studies uncover the essential functions of Stat3 in maintaining Wnt/β-catenin signaling in early mesenchymal or osteoprogenitor cells and provide evidence that bone defects in the Job Syndrome are likely caused by Wnt/β-catenin signaling reduction due to reduced STAT3 activities in bone development. Enhancing Wnt/β-catenin signaling could be a therapeutic approach to reduce bone symptoms of Job syndrome patients.
Collapse
Affiliation(s)
- Prem Swaroop Yadav
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Shuhao Feng
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Hanjun Kim
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yuchen Liu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA 02115;
- Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
42
|
Lindeman RE, Murphy MW, Agrimson KS, Gewiss R, Bardwell V, Gearhart M, Zarkower D. The conserved sex regulator DMRT1 recruits SOX9 in sexual cell fate reprogramming. Nucleic Acids Res 2021; 49:6144-6164. [PMID: 34096593 PMCID: PMC8216462 DOI: 10.1093/nar/gkab448] [Citation(s) in RCA: 31] [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: 03/29/2021] [Revised: 04/30/2021] [Accepted: 05/17/2021] [Indexed: 01/16/2023] Open
Abstract
Mammalian sexual development commences when fetal bipotential progenitor cells adopt male Sertoli (in XY) or female granulosa (in XX) gonadal cell fates. Differentiation of these cells involves extensive divergence in chromatin state and gene expression, reflecting distinct roles in sexual differentiation and gametogenesis. Surprisingly, differentiated gonadal cell fates require active maintenance through postnatal life to prevent sexual transdifferentiation and female cell fate can be reprogrammed by ectopic expression of the sex regulator DMRT1. Here we examine how DMRT1 reprograms granulosa cells to Sertoli-like cells in vivo and in culture. We define postnatal sex-biased gene expression programs and identify three-dimensional chromatin contacts and differentially accessible chromatin regions (DARs) associated with differentially expressed genes. Using a conditional transgene we find DMRT1 only partially reprograms the ovarian transcriptome in the absence of SOX9 and its paralog SOX8, indicating that these factors functionally cooperate with DMRT1. ATAC-seq and ChIP-seq show that DMRT1 induces formation of many DARs that it binds with SOX9, and DMRT1 is required for binding of SOX9 at most of these. We suggest that DMRT1 can act as a pioneer factor to open chromatin and allow binding of SOX9, which then cooperates with DMRT1 to reprogram sexual cell fate.
Collapse
Affiliation(s)
- Robin E Lindeman
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Mark W Murphy
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Kellie S Agrimson
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Rachel L Gewiss
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - Vivian J Bardwell
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
- University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| | - Micah D Gearhart
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
| | - David Zarkower
- Developmental Biology Center and Department of Genetics, Cell Biology, and Development, University of Minnesota, Minneapolis, MN 55455 USA
- University of Minnesota Masonic Cancer Center, Minneapolis, MN 55455, USA
| |
Collapse
|
43
|
Shao R, Zhang Z, Xu Z, Ouyang H, Wang L, Ouyang H, Greenblatt M, Chen X, Zou W. H3K36 methyltransferase NSD1 regulates chondrocyte differentiation for skeletal development and fracture repair. Bone Res 2021; 9:30. [PMID: 34099628 PMCID: PMC8185073 DOI: 10.1038/s41413-021-00148-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 01/22/2021] [Accepted: 02/19/2021] [Indexed: 12/17/2022] Open
Abstract
Chondrocyte differentiation is a critical process for endochondral ossification, which is responsible for long bone development and fracture repair. Considerable progress has been made in understanding the transcriptional control of chondrocyte differentiation; however, epigenetic regulation of chondrocyte differentiation remains to be further studied. NSD1 is a H3K36 (histone H3 at lysine 36) methyltransferase. Here, we showed that mice with Nsd1 deficiency in Prx1+ mesenchymal progenitors but not in Col2+ chondrocytes showed impaired skeletal growth and fracture healing accompanied by decreased chondrogenic differentiation. Via combined RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) analysis, we identified sex determining region Y box 9 (Sox9), the key transcription factor of chondrogenic differentiation, as a functional target gene of NSD1. Mechanistically, NSD1 regulates Sox9 expression by modulating H3K36me1 and H3K36me2 levels in the Sox9 promoter region, constituting a novel epigenetic regulatory mechanism of chondrogenesis. Moreover, we found that NSD1 can directly activate the expression of hypoxia-inducible factor 1α (HIF1α), which plays a vital role in chondrogenic differentiation through its regulation of Sox9 expression. Collectively, the results of our study reveal crucial roles of NSD1 in regulating chondrogenic differentiation, skeletal growth, and fracture repair and expand our understanding of the function of epigenetic regulation in chondrogenesis and skeletal biology.
Collapse
Affiliation(s)
- Rui Shao
- Shanghai Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhong Zhang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Zhan Xu
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huiling Ouyang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Lijun Wang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hongwei Ouyang
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University-University of Edinburgh Institute, Zhejiang University, Hangzhou, Zhejiang, China.,China Orthopedic Regenerative Medicine Group, Hangzhou, Zhejiang, China
| | - Matthew Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Xi Chen
- Department of Molecular and Cellular Biology, Lester and Sue Smith Breast Center, Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Weiguo Zou
- Shanghai Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China. .,State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
44
|
Sox9a, not sox9b is required for normal cartilage development in zebrafish. AQUACULTURE AND FISHERIES 2021. [DOI: 10.1016/j.aaf.2019.12.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
45
|
Goodarzi N, Akbari Bazm M, Poladi S, Rashidi F, Mahmoudi B, Abumandour MMA. Histology of the small intestine in the common pheasant (Phasianus colchicus): A scanning electron microscopy, histochemical, immunohistochemical, and stereological study. Microsc Res Tech 2021; 84:2388-2398. [PMID: 33908129 DOI: 10.1002/jemt.23794] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/29/2021] [Accepted: 04/13/2021] [Indexed: 12/16/2022]
Abstract
The present investigation was conducted to investigate the histology, immunohistochemistry, stereology, and ultrastructure of the small intestine in the common pheasant (Phasianus colchicus) using light and scanning electron microscopy (SEM). Ten birds were included in the study. The obtained findings revealed that three parts in the small intestine namely duodenum, jejunum, and ilium constituted of four layers from innermost to outermost including tunica mucosa, tunica submucosa, tunica muscularis, and tunica serosa. All parts of the small intestine had simple columnar epithelium with goblet cells reacted with Periodic Acid-Schiff and Alcian Blue stains especially in the jejunum and ileum. The cells on the tip of the duodenal villi showed immuno-positive staining for Sox9 protein, while the jejunum and ileum were negative. The jejunum had longest villi; however the duodenum had deepest crypt (p < .05). The villus surface of jejunum was significantly higher than duodenum and ileum (p < .05). SEM images revealed that the duodenum had cauliflower and leaf-like villi with plicas and recess between them. Jejunum had finger-like villi with a velvety view. The shape of the ileal villi was like that observed in the jejunum with slight differences. In conclusion, the jejunum was the most prominent region in the small intestine in terms of morphologic and morphometric features, which could be attributed to the absorption of more nutrients in this area. These obtained findings would be useful to improve the knowledge in the field of histophysiology properties of the bird's digestive system.
Collapse
Affiliation(s)
- Nader Goodarzi
- Department of Basic Sciences and Pathobiology, Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Mohsen Akbari Bazm
- Department of Anatomical Sciences, Medical School, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sadra Poladi
- Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Fatemeh Rashidi
- Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Bahareh Mahmoudi
- Faculty of Veterinary Medicine, Razi University, Kermanshah, Iran
| | - Mohamed M A Abumandour
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
46
|
Enhancing the chondrogenic potential of chondrogenic progenitor cells by deleting RAB5C. iScience 2021; 24:102464. [PMID: 34013174 PMCID: PMC8113995 DOI: 10.1016/j.isci.2021.102464] [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] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/24/2021] [Accepted: 04/21/2021] [Indexed: 11/21/2022] Open
Abstract
Osteoarthritis (OA) is the most prevalent chronic joint disease that affects a large proportion of the elderly population. Chondrogenic progenitor cells (CPCs) reside in late-stage OA cartilage tissue, producing a fibrocartilaginous extracellular matrix; these cells can be manipulated in vitro to deposit proteins of healthy articular cartilage. CPCs are under the control of SOX9 and RUNX2. In our earlier studies, we showed that a knockdown of RUNX2 enhanced the chondrogenic potential of CPCs. Here we demonstrate that CPCs carrying a knockout of RAB5C, a protein involved in endosomal trafficking, exhibited elevated expression of multiple chondrogenic markers, including the SOX trio, and increased COL2 deposition, whereas no changes in COL1 deposition were observed. We report RAB5C as an attractive target for future therapeutic approaches designed to increase the COL2 content in the diseased joint.
Collapse
|
47
|
Li R, Sun Y, Chen Z, Zheng M, Shan Y, Ying X, Weng M, Chen Z. The Fibroblast Growth Factor 9 (Fgf9) Participates in Palatogenesis by Promoting Palatal Growth and Elevation. Front Physiol 2021; 12:653040. [PMID: 33959039 PMCID: PMC8093392 DOI: 10.3389/fphys.2021.653040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 03/11/2021] [Indexed: 11/28/2022] Open
Abstract
Cleft palate, a common global congenital malformation, occurs due to disturbances in palatal growth, elevation, contact, and fusion during palatogenesis. The Fibroblast growth factor 9 (FGF9) mutation has been discovered in humans with cleft lip and palate. Fgf9 is expressed in both the epithelium and mesenchyme, with temporospatial diversity during palatogenesis. However, the specific role of Fgf9 in palatogenesis has not been extensively discussed. Herein, we used Ddx4-Cre mice to generate an Fgf9–/– mouse model (with an Fgf9 exon 2 deletion) that exhibited a craniofacial syndrome involving a cleft palate and deficient mandibular size with 100% penetrance. A smaller palatal shelf size, delayed palatal elevation, and contact failure were investigated to be the intrinsic causes for cleft palate. Hyaluronic acid accumulation in the extracellular matrix (ECM) sharply decreased, while the cell density correspondingly increased in Fgf9–/– mice. Additionally, significant decreases in cell proliferation were discovered in not only the palatal epithelium and mesenchyme but also among cells in Meckel’s cartilage and around the mandibular bone in Fgf9–/– mice. Serial sections of embryonic heads dissected at embryonic day 14.5 (E14.5) were subjected to craniofacial morphometric measurement. This highlighted the reduced oral volume owing to abnormal tongue size and descent, and insufficient mandibular size, which disturbed palatal elevation in Fgf9–/– mice. These results indicate that Fgf9 facilitates palatal growth and timely elevation by regulating cell proliferation and hyaluronic acid accumulation. Moreover, Fgf9 ensures that the palatal elevation process has adequate space by influencing tongue descent, tongue morphology, and mandibular growth.
Collapse
Affiliation(s)
- Ruomei Li
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yidan Sun
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhengxi Chen
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Resident, Department of General Dentistry, Henry M. Goldman School of Dental Medicine, Boston University, Boston, MA, United States
| | - Mengting Zheng
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuhua Shan
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiyu Ying
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Mengjia Weng
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenqi Chen
- Department of Orthodontics, Shanghai Key Laboratory of Stomatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
48
|
Kitajima K, Kawahira N, Lee SW, Tamura K, Morishita Y, Ohtsuka D. Light-induced local gene expression in primary chick cell culture system. Dev Growth Differ 2021; 63:189-198. [PMID: 33733477 PMCID: PMC8252662 DOI: 10.1111/dgd.12721] [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] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/15/2021] [Accepted: 02/17/2021] [Indexed: 12/28/2022]
Abstract
The ability to manipulate gene expression at a specific region in a tissue or cell culture system is critical for analysis of target gene function. For chick embryos/cells, several gene introduction/induction methods have been established such as those involving retrovirus, electroporation, sonoporation, and lipofection. However, these methods have limitations in the accurate induction of localized gene expression. Here we demonstrate the effective application of a recently developed light‐dependent gene expression induction system (LightOn system) using the Neurospora crassa photoreceptor Vivid fused with a Gal4 DNA binding domain and p65 activation domain (GAVPO) that alters its activity in response to light stimulus in a primary chicken cell culture system. We show that the gene expression level and induction specificity in this system are strongly dependent on the light irradiation conditions. Especially, the irradiation interval is an important parameter for modulating gene expression; for shorter time intervals, higher induction specificity can be achieved. Further, by adjusting light irradiation conditions, the expression level in primary chicken cells can be regulated in a multiple step manner, in contrast to the binary expression seen for gene disruption or introduction (i.e., null or overexpression). This result indicates that the light‐dependent expression control method can be a useful technique in chick models to examine how gene function is affected by gradual changes in gene expression levels. We applied this light induction system to regulate Sox9 expression in cultures of chick limb mesenchyme cells and showed that induced SOX9 protein could modulate expression of downstream genes.
Collapse
Affiliation(s)
- Keiichi Kitajima
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan.,Research Fellow (DC2) of Japan Society for the Promotion of Science, Tokyo, Japan.,Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Naofumi Kawahira
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sang-Woo Lee
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Koji Tamura
- Department of Ecological Developmental Adaptability Life Sciences, Graduate School of Life Sciences, Tohoku University, Sendai, Japan
| | - Yoshihiro Morishita
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan.,Precursory Research for Embryonic Science and Technology (PRESTO) Program, Japan Science and Technology Agency, Kawaguchi, Japan
| | - Daisuke Ohtsuka
- Laboratory for Developmental Morphogeometry, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| |
Collapse
|
49
|
Raza S, Jokl E, Pritchett J, Martin K, Su K, Simpson K, Birchall L, Mullan AF, Athwal VS, Doherty DT, Zeef L, Henderson NC, Kalra PA, Hanley NA, Piper Hanley K. SOX9 is required for kidney fibrosis and activates NAV3 to drive renal myofibroblast function. Sci Signal 2021; 14:14/672/eabb4282. [PMID: 33653921 DOI: 10.1126/scisignal.abb4282] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Renal fibrosis is a common end point for kidney injury and many chronic kidney diseases. Fibrogenesis depends on the sustained activation of myofibroblasts, which deposit the extracellular matrix that causes progressive scarring and organ failure. Here, we showed that the transcription factor SOX9 was associated with kidney fibrosis in humans and required for experimentally induced kidney fibrosis in mice. From genome-wide analysis, we identified Neuron navigator 3 (NAV3) as acting downstream of SOX9 in kidney fibrosis. NAV3 increased in abundance and colocalized with SOX9 after renal injury in mice, and both SOX9 and NAV3 were present in diseased human kidneys. In an in vitro model of renal pericyte transdifferentiation into myofibroblasts, we demonstrated that NAV3 was required for multiple aspects of fibrogenesis, including actin polymerization linked to cell migration and sustained activation of the mechanosensitive transcription factor YAP1. In summary, our work identifies a SOX9-NAV3-YAP1 axis involved in the progression of kidney fibrosis and points to NAV3 as a potential target for pharmacological intervention.
Collapse
Affiliation(s)
- Sayyid Raza
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Elliot Jokl
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - James Pritchett
- School of Healthcare Science, Manchester Metropolitan University, Manchester M1 5GD, UK
| | - Katherine Martin
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Kim Su
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Kara Simpson
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Lindsay Birchall
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Aoibheann F Mullan
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Varinder S Athwal
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK.,Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9PT, UK
| | - Daniel T Doherty
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| | - Leo Zeef
- Bioinformatics Core Facility, Faculty of Life Sciences, University of Manchester, Manchester, UK
| | - Neil C Henderson
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
| | - Philip A Kalra
- Salford Royal NHS Foundation Trust, Stott Lane, Salford, UK
| | - Neil A Hanley
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK.,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK.,Manchester University NHS Foundation Trust, Oxford Road, Manchester M13 9PT, UK
| | - Karen Piper Hanley
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health and Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK. .,Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Oxford Road, Manchester, UK
| |
Collapse
|
50
|
Anti-epileptic drug topiramate upregulates TGFβ1 and SOX9 expression in primary embryonic palatal mesenchyme cells: Implications for teratogenicity. PLoS One 2021; 16:e0246989. [PMID: 33577554 PMCID: PMC7880431 DOI: 10.1371/journal.pone.0246989] [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] [Received: 09/01/2020] [Accepted: 01/28/2021] [Indexed: 12/27/2022] Open
Abstract
Topiramate is an anti-epileptic drug that is commonly prescribed not just to prevent seizures but also migraine headaches, with over 8 million prescriptions dispensed annually. Topiramate use during pregnancy has been linked to significantly increased risk of babies born with orofacial clefts (OFCs). However, the exact molecular mechanism of topiramate teratogenicity is unknown. In this study, we first used an unbiased antibody array analysis to test the effect of topiramate on human embryonic palatal mesenchyme (HEPM) cells. This analysis identified 40 differentially expressed proteins, showing strong connectivity to known genes associated with orofacial clefts. However, among known OFC genes, only TGFβ1 was significantly upregulated in the antibody array analysis. Next, we validated that topiramate could increase expression of TGFβ1 and of downstream target phospho-SMAD2 in primary mouse embryonic palatal mesenchyme (MEPM) cells. Furthermore, we showed that topiramate treatment of primary MEPM cells increased expression of SOX9. SOX9 overexpression in chondrocytes is known to cause cleft palate in mouse. We propose that topiramate mediates upregulation of TGFβ1 signaling through activation of γ-aminobutyric acid (GABA) receptors in the palate. TGFβ1 and SOX9 play critical roles in orofacial morphogenesis, and their abnormal overexpression provides a plausible etiologic molecular mechanism for the teratogenic effects of topiramate.
Collapse
|