151
|
Liu Z, Easson GWD, Zhao J, Makki N, Ahituv N, Hilton MJ, Tang SY, Gray RS. Dysregulation of STAT3 signaling is associated with endplate-oriented herniations of the intervertebral disc in Adgrg6 mutant mice. PLoS Genet 2019; 15:e1008096. [PMID: 31652254 PMCID: PMC6834287 DOI: 10.1371/journal.pgen.1008096] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 11/06/2019] [Accepted: 09/18/2019] [Indexed: 12/01/2022] Open
Abstract
Degenerative changes of the intervertebral disc (IVD) are a leading cause of disability affecting humans worldwide and has been attributed primarily to trauma and the accumulation of pathology during aging. While genetic defects have also been associated with disc degeneration, the precise mechanisms driving the initiation and progression of disease have remained elusive due to a paucity of genetic animal models. Here, we discuss a novel conditional mouse genetic model of endplate-oriented disc herniations in adult mice. Using conditional mouse genetics, we show increased mechanical stiffness and reveal dysregulation of typical gene expression profiles of the IVD in adhesion G-protein coupled receptor G6 (Adgrg6) mutant mice prior to the onset of endplate-oriented disc herniations in adult mice. We observed increased STAT3 activation prior to IVD defects and go on to demonstrate that treatment of Adgrg6 conditional mutant mice with a small molecule inhibitor of STAT3 activation ameliorates endplate-oriented herniations. These findings establish ADGRG6 and STAT3 as novel regulators of IVD endplate and growth plate integrity in the mouse, and implicate ADGRG6/STAT3 signaling as promising therapeutic targets for endplate-oriented disc degeneration. Back pain is a leading cause of disability in humans worldwide and one of the most common culprits of these issues are the consequence of degenerative changes of the intervertebral disc. Here, we demonstrate that conditional loss of the Adgrg6 gene in cartilaginous tissues of the spine results in endplate-oriented disc herniations and degenerative changes of the intervertebral disc in mice. We further establish that these obvious degenerative changes of the disc are preceded by substantial alterations in normal gene expression profiles, including upregulation of pro-inflammatory STAT3 signaling, and increased mechanical stiffness of the intervertebral disc. Increased STAT3 activation is a signal observed in other models of degenerative musculoskeletal tissues. As such, we tested whether systemic treatment with a small-molecule STAT3 inhibitor would protect against the formation of endplate-oriented disc herniations in conditional Adgrg6 mutant mice, and report a significant positive improvement of histopathology in our treatment group. Taken together, we demonstrate a novel conditional model of endplate-oriented disc herniation in mouse. We establish ADGRG6 and STAT3 as novel regulators of endplate integrity of the intervertebral disc in mouse and suggest that modulation of ADGRG6/STAT3 signaling could provide robust disease-modifying targets for endplate-oriented disc degeneration in humans.
Collapse
Affiliation(s)
- Zhaoyang Liu
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas, United States of America
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, Texas, United States of America
| | - Garrett W. D. Easson
- Department of Orthopedics, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jingjing Zhao
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Nadja Makki
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
- Department of Anatomy and Cell Biology, University of Florida, College of Medicine, Gainesville, Florida, United States of America
| | - Nadav Ahituv
- Department of Bioengineering and Therapeutic Sciences and Institute for Human Genetics, University of California San Francisco, San Francisco, California, United States of America
| | - Matthew J. Hilton
- Department of Orthopedic Surgery and Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Simon Y. Tang
- Department of Orthopedics, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Ryan S. Gray
- Department of Nutritional Sciences, University of Texas at Austin, Austin, Texas, United States of America
- Department of Pediatrics, Dell Pediatric Research Institute, University of Texas at Austin Dell Medical School, Austin, Texas, United States of America
- * E-mail:
| |
Collapse
|
152
|
Fahrner JA, Lin WY, Riddle RC, Boukas L, DeLeon VB, Chopra S, Lad SE, Luperchio TR, Hansen KD, Bjornsson HT. Precocious chondrocyte differentiation disrupts skeletal growth in Kabuki syndrome mice. JCI Insight 2019; 4:129380. [PMID: 31557133 DOI: 10.1172/jci.insight.129380] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Kabuki syndrome 1 (KS1) is a Mendelian disorder of the epigenetic machinery caused by mutations in the gene encoding KMT2D, which methylates lysine 4 on histone H3 (H3K4). KS1 is characterized by intellectual disability, postnatal growth retardation, and distinct craniofacial dysmorphisms. A mouse model (Kmt2d+/βGeo) exhibits features of the human disorder and has provided insight into other phenotypes; however, the mechanistic basis of skeletal abnormalities and growth retardation remains elusive. Using high-resolution micro-CT, we show that Kmt2d+/βGeo mice have shortened long bones and ventral bowing of skulls. In vivo expansion of growth plates within skulls and long bones suggests disrupted endochondral ossification as a common disease mechanism. Stable chondrocyte cell lines harboring inactivating mutations in Kmt2d exhibit precocious differentiation, further supporting this mechanism. A known inducer of chondrogenesis, SOX9, and its targets show markedly increased expression in Kmt2d-/- chondrocytes. By transcriptome profiling, we identify Shox2 as a putative KMT2D target. We propose that decreased KMT2D-mediated H3K4me3 at Shox2 releases Sox9 inhibition and thereby leads to enhanced chondrogenesis, providing a potentially novel and plausible explanation for precocious chondrocyte differentiation. Our findings provide insight into the pathogenesis of growth retardation in KS1 and suggest therapeutic approaches for this and related disorders.
Collapse
Affiliation(s)
- Jill A Fahrner
- McKusick-Nathans Institute of Genetic Medicine.,Department of Pediatrics
| | | | | | - Leandros Boukas
- McKusick-Nathans Institute of Genetic Medicine.,Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Valerie B DeLeon
- Department of Anthropology, University of Florida, Gainesville, Florida, USA
| | | | - Susan E Lad
- Department of Anthropology, University of Florida, Gainesville, Florida, USA
| | | | - Kasper D Hansen
- McKusick-Nathans Institute of Genetic Medicine.,Department of Biostatistics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hans T Bjornsson
- McKusick-Nathans Institute of Genetic Medicine.,Department of Pediatrics.,Landspitali University Hospital, Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
153
|
Chen J, Chen F, Bian H, Wang Q, Zhang X, Sun L, Gu J, Lu Y, Zheng Q. Hypertrophic chondrocyte-specific Col10a1 controlling elements in Cre recombinase transgenic studies. Am J Transl Res 2019; 11:6672-6679. [PMID: 31737217 PMCID: PMC6834502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2018] [Accepted: 10/11/2019] [Indexed: 06/10/2023]
Abstract
The type X collagen gene (COL10A1) is specifically expressed in chondrocytes undergoing hypertrophy, which is an essential late stage of endochondral ossification during the development of long bones. We have previously localized multiple murine Col10a1 promoter-enhancer elements and used these elements for transgenic studies with LacZ reporter gene or genes of interest. Here, we report two additional transgenic mouse lines in which Cre was driven by the 10 kb Col10a1 promoter/intron and the 300-bp enhancer elements respectively. Cre activity was assessed by breeding the transgenic founders onto the RosA26R genetic background and to examine its β-gal activity (blue staining) via Cre/Lox P recombination. Our results showed that, in addition to the Cre activity in hypertrophic chondrocytes, we also observed blue staining of the bone marrow and the surrounding digits when the 10 kb Col10a1 promoter/intron element was used, whereas the 300-bp enhancer element could drive Cre expression exclusively within the hypertrophic zone as demonstrated by the blue staining pattern. This is intriguing, as the 10 kb promoter covers the 300-bp enhancer element. We then further reanalyzed the LacZ transgenic mice. We did observe non-specific blue staining in 10 kb-LacZ mice but not the mice with the 300-bp enhancer. In addition, the Cre reporter construct was on a coat-color vector backbone, which enables direct visual genotyping of the transgenic mice in the FVB/N albino background. Together, our results support that the 300 bp Col10a1 enhancer provides a more efficient genetic tool to target the hypertrophic zone for studies of skeletal development and disease.
Collapse
Affiliation(s)
- Jinnan Chen
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
- Department of Internal Medicine, Rush University Medical CenterChicago, IL 60612, USA
| | - Fangzhou Chen
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
| | - Huiqin Bian
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
| | - Qian Wang
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
| | - Xiaojing Zhang
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
| | - Lichun Sun
- Department of Medicine, School of Medicine, Tulane Health Sciences CenterNew Orleans, LA 70112-2699, USA
- Shenzhen Academy of Peptide Targeting Technology at Pingshan, Shenzhen Tyercan Bio-pharm Co., Ltd.Shenzhen 518118, Guangdong, China
| | - Junxia Gu
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
| | - Yaojuan Lu
- Shenzhen Academy of Peptide Targeting Technology at Pingshan, Shenzhen Tyercan Bio-pharm Co., Ltd.Shenzhen 518118, Guangdong, China
| | - Qiping Zheng
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu UniversityZhenjiang 212013, Jiangsu, China
- Shenzhen Academy of Peptide Targeting Technology at Pingshan, Shenzhen Tyercan Bio-pharm Co., Ltd.Shenzhen 518118, Guangdong, China
| |
Collapse
|
154
|
Marín-Llera JC, Garciadiego-Cázares D, Chimal-Monroy J. Understanding the Cellular and Molecular Mechanisms That Control Early Cell Fate Decisions During Appendicular Skeletogenesis. Front Genet 2019; 10:977. [PMID: 31681419 PMCID: PMC6797607 DOI: 10.3389/fgene.2019.00977] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 09/13/2019] [Indexed: 12/02/2022] Open
Abstract
The formation of the vertebrate skeleton is orchestrated in time and space by a number of gene regulatory networks that specify and position all skeletal tissues. During embryonic development, bones have two distinct origins: bone tissue differentiates directly from mesenchymal progenitors, whereas most long bones arise from cartilaginous templates through a process known as endochondral ossification. Before endochondral bone development takes place, chondrocytes form a cartilage analgen that will be sequentially segmented to form joints; thus, in the cartilage template, either the cartilage maturation programme or the joint formation programme is activated. Once the cartilage differentiation programme starts, the growth plate begins to form. In contrast, when the joint formation programme is activated, a capsule begins to form that contains special articular cartilage and synovium to generate a functional joint. In this review, we will discuss the mechanisms controlling the earliest molecular events that regulate cell fate during skeletogenesis in long bones. We will explore the initial processes that lead to the recruitment of mesenchymal stem/progenitor cells, the commitment of chondrocyte lineages, and the formation of skeletal elements during morphogenesis. Thereafter, we will review the process of joint specification and joint morphogenesis. We will discuss the links between transcription factor activity, cell–cell interactions, cell–extracellular matrix interactions, growth factor signalling, and other molecular interactions that control mesenchymal stem/progenitor cell fate during embryonic skeletogenesis.
Collapse
Affiliation(s)
- Jessica Cristina Marín-Llera
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | | | - Jesús Chimal-Monroy
- Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
155
|
Diederichs S, Tonnier V, März M, Dreher SI, Geisbüsch A, Richter W. Regulation of WNT5A and WNT11 during MSC in vitro chondrogenesis: WNT inhibition lowers BMP and hedgehog activity, and reduces hypertrophy. Cell Mol Life Sci 2019; 76:3875-3889. [PMID: 30980110 PMCID: PMC11105731 DOI: 10.1007/s00018-019-03099-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022]
Abstract
Re-directing mesenchymal stromal cell (MSC) chondrogenesis towards a non-hypertrophic articular chondrocyte-(AC)-like phenotype is important for improving articular cartilage neogenesis to enhance clinical cartilage repair strategies. This study is the first to demonstrate that high levels of non-canonical WNT5A followed by WNT11 and LEF1 discriminated MSC chondrogenesis from AC re-differentiation. Moreover, β-catenin seemed incompletely silenced in differentiating MSCs, which altogether suggested a role for WNT signaling in hypertrophic MSC differentiation. WNT inhibition with the small molecule IWP-2 supported MSC chondrogenesis according to elevated proteoglycan deposition and reduced the characteristic upregulation of BMP4, BMP7 and their target ID1, as well as IHH and its target GLI1 observed during endochondral differentiation. Along with the pro-hypertrophic transcription factor MEF2C, multiple hypertrophic downstream targets including IBSP and alkaline phosphatase activity were reduced by IWP-2, demonstrating that WNT activity drives BMP and hedgehog upregulation, and MSC hypertrophy. WNT inhibition almost matched the strong anti-hypertrophic capacity of pulsed parathyroid hormone-related protein application, and both outperformed suppression of BMP signaling with dorsomorphin, which also reduced cartilage matrix deposition. Yet, hypertrophic marker expression under IWP-2 remained above AC level, and in vivo mineralization and ectopic bone formation were reduced but not eliminated. Overall, the strong anti-hypertrophic effects of IWP-2 involved inhibition but not silencing of pro-hypertrophic BMP and IHH pathways, and more advanced silencing of WNT activity as well as combined application of IHH or BMP antagonists should next be considered to install articular cartilage neogenesis from human MSCs.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Veronika Tonnier
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie März
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Simon I Dreher
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Geisbüsch
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
156
|
Tsang KY, Cheah KS. The extended chondrocyte lineage: implications for skeletal homeostasis and disorders. Curr Opin Cell Biol 2019; 61:132-140. [PMID: 31541943 DOI: 10.1016/j.ceb.2019.07.011] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/19/2019] [Accepted: 07/30/2019] [Indexed: 01/14/2023]
Abstract
Endochondral bone formation relies on a finely controlled sequence of chondrocyte proliferation, maturation and hypertrophy that establishes the growth plate which, combined with the deposition of bone upon the cartilage template, mediates longitudinal skeletal growth. Recent lineage studies support a chondrocyte-osteoblast differentiation continuum and the presence of skeletal stem cells within cartilage. The biological significance of the lineage extension and the mechanisms controlling the process are unclear. In this review, we describe recent work on the extended chondrocyte-osteoblast lineage and its contribution to the development, growth and repair of bone and to bone disorders that provides insight into the process and the molecular controls involved. The implications for skeletal homeostasis are discussed.
Collapse
Affiliation(s)
- Kwok Yeung Tsang
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | - Kathryn Se Cheah
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China.
| |
Collapse
|
157
|
Huang MJ, Zhao JY, Xu JJ, Li J, Zhuang YF, Zhang XL. lncRNA ADAMTS9-AS2 Controls Human Mesenchymal Stem Cell Chondrogenic Differentiation and Functions as a ceRNA. MOLECULAR THERAPY-NUCLEIC ACIDS 2019; 18:533-545. [PMID: 31671346 PMCID: PMC6838486 DOI: 10.1016/j.omtn.2019.08.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/17/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as key regulators of cell differentiation and development. However, potential roles for lncRNAs in chondrogenic differentiation have remained poorly understood. Here we identify lncRNA ADAMTS9 antisense RNA 2, ADAMTS9-AS2, which controls the chondrogenic differentiation by acting as a competing endogenous RNA (ceRNA) in human mesenchymal stem cells (hMSCs). We screen out ADAMTS9-AS2 of undifferentiated and differentiated cells during chondrogenic differentiation by microarrays. Suppression or overexpression of lncRNA ADAMTS9-AS2 correlates with inhibition and promotion of hMSC chondrogenic differentiation, respectively. We find that ADAMTS9-AS2 can sponge miR-942-5p to regulate the expression of Scrg1, a transcription factor promoting chondrogenic gene expression. Finally, we confirm the function of ADAMTS9-AS2 to cartilage repair in the absence of transforming growth factor β (TGF-β) in vivo. In conclusion, ADAMTS9-AS2 plays an important role in chondrogenic differentiation as a ceRNA, so that it can be regarded as a therapy target for cartilage repair.
Collapse
Affiliation(s)
- Ming-Jian Huang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Jing-Yu Zhao
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Jia-Jia Xu
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Jing Li
- The Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Jiao Tong University School of Medicine (SJTUSM) & Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Yi-Fu Zhuang
- Department of Orthopedic Surgery, Shanghai Ninth People's Hospital Affiliated with Shanghai Jiao Tong University School of Medicine (SJTUSM), Shanghai 201999, China
| | - Xiao-Ling Zhang
- Department of Orthopedic Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
158
|
Marofi F, Hassanzadeh A, Solali S, Vahedi G, Mousavi Ardehaie R, Salarinasab S, Aliparasti MR, Ghaebi M, Farshdousti Hagh M. Epigenetic mechanisms are behind the regulation of the key genes associated with the osteoblastic differentiation of the mesenchymal stem cells: The role of zoledronic acid on tuning the epigenetic changes. J Cell Physiol 2019; 234:15108-15122. [PMID: 30652308 DOI: 10.1002/jcp.28152] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Accepted: 01/08/2019] [Indexed: 01/24/2023]
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells and show distinct features such as capability for self-renewal and differentiation into several lineages of cells including osteoblasts, chondrocytes, and adipocytes. In this study, the methylation status of the promoter region of zinc finger and BTB domain containing 16 (ZBTB16), twist-related protein 1(Twist1), de novo DNA methyltransferases 3A (DNMT3A), SRY-box 9 (Sox9), osteocalcin (OCN), and peroxisome proliferator-activated receptor γ2 (PPARγ2) genes and their messenger RNA (mRNA) expression levels were evaluated during the osteoblastic differentiation of MSCs (ODMSCs). We planned two experimental groups including zoledronic acid (ZA)-treated and nontreated cells (negative control) which both were differentiated into the osteoblasts. Methylation level of DNA in the promoter regions was assayed by methylation-specific-quantitative polymerase chain reaction (MS-qPCR), and mRNA levels of the target inhibitory/stimulatory genes during osteoblastic differentiation of MSCs were measured using real-time PCR. During the experimental induction of ODMSCs, the mRNA expression of the OCN gene was upregulated and methylation level of its promoter region was decreased. Moreover, Sox9 and PPARγ2 mRNA levels were attenuated and their promoter regions methylation levels were significantly augmented. However, the mRNA expression of the DNMT3A was not affected during the ODMSCs though its methylation rate was increased. In addition, ZA could enhance the expression of the ZBTB16 and decrease its promoter regions methylation and on the opposite side, it diminished mRNA expression of Sox9, Twist1, and PPARγ2 genes and increased their methylation rates. Intriguingly, ZA did not show a significant impact on gene expression and methylation levels the OCN and DNMT3A. We found that methylation of the promoter regions of Sox9, OCN, and PPARγ2 genes might be one of the main mechanisms adjusting the genes expression during the ODMSCs. Furthermore, we noticed that ZA can accelerate the MSCs differentiation to the osteoblast cells via two regulatory processes; suppression of osteoblastic differentiation inhibitor genes including Sox9, Twist1, and PPARγ2, and through promotion of the ZBTB16 expression.
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Hassanzadeh
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Solali
- Department of Immunology, Division of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ghasem Vahedi
- Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Reza Mousavi Ardehaie
- Department of Medical Genetic, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sadegh Salarinasab
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammad Reza Aliparasti
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahnaz Ghaebi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Farshdousti Hagh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
159
|
Catheline SE, Hoak D, Chang M, Ketz JP, Hilton MJ, Zuscik MJ, Jonason JH. Chondrocyte-Specific RUNX2 Overexpression Accelerates Post-traumatic Osteoarthritis Progression in Adult Mice. J Bone Miner Res 2019; 34:1676-1689. [PMID: 31189030 PMCID: PMC7047611 DOI: 10.1002/jbmr.3737] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/18/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
RUNX2 is a transcription factor critical for chondrocyte maturation and normal endochondral bone formation. It promotes the expression of factors catabolic to the cartilage extracellular matrix and is upregulated in human osteoarthritic cartilage and in murine articular cartilage following joint injury. To date, in vivo studies of RUNX2 overexpression in cartilage have been limited to forced expression in osteochondroprogenitor cells preventing investigation into the effects of chondrocyte-specific RUNX2 overexpression in postnatal articular cartilage. Here, we used the Rosa26Runx2 allele in combination with the inducible Col2a1CreERT2 transgene or the inducible AcanCreERT2 knock-in allele to achieve chondrocyte-specific RUNX2 overexpression (OE) during embryonic development or in the articular cartilage of adult mice, respectively. RUNX2 OE was induced at embryonic day 13.5 (E13.5) for all developmental studies. Histology and in situ hybridization analyses suggest an early onset of chondrocyte hypertrophy and accelerated terminal maturation in the limbs of the RUNX2 OE embryos compared to control embryos. For all postnatal studies, RUNX2 OE was induced at 2 months of age. Surprisingly, no histopathological signs of cartilage degeneration were observed even 6 months following induction of RUNX2 OE. Using the meniscal/ligamentous injury (MLI), a surgical model of knee joint destabilization and meniscal injury, however, we found that RUNX2 OE accelerates the progression of cartilage degeneration following joint trauma. One month following MLI, the numbers of MMP13-positive and TUNEL-positive chondrocytes were significantly greater in the articular cartilage of the RUNX2 OE joints compared to control joints and 2 months following MLI, histomorphometry and Osteoarthritis Research Society International (OARSI) scoring revealed decreased cartilage area in the RUNX2 OE joints. Collectively, these results suggest that although RUNX2 overexpression alone may not be sufficient to initiate the OA degenerative process, it may predetermine the rate of OA onset and/or progression following traumatic joint injury. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Sarah E Catheline
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Donna Hoak
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Martin Chang
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - John P Ketz
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| | - Matthew J Hilton
- Department of Orthopaedic Surgery, Duke University, Durham, NC, USA
| | - Michael J Zuscik
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.,Orthopedic Research Center, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jennifer H Jonason
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Orthopaedics and Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
160
|
Ramachandran J, Liu Z, Gray RS, Vokes SA. PRMT5 is necessary to form distinct cartilage identities in the knee and long bone. Dev Biol 2019; 456:154-163. [PMID: 31442442 DOI: 10.1016/j.ydbio.2019.08.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 01/08/2023]
Abstract
During skeletal development, limb progenitors become specified as chondrocytes and subsequently differentiate into specialized cartilage compartments. We previously showed that the arginine dimethyl transferase, PRMT5, is essential for regulating the specification of progenitor cells into chondrocytes within early limb buds. Here, we report that PRMT5 regulates the survival of a separate progenitor domain that gives rise to the patella. Independent of its role in knee development, PRMT5 regulates several distinct types of chondrocyte differentiation within the long bones. Chondrocytes lacking PRMT5 have a striking blockage in hypertrophic chondrocyte differentiation and are marked by abnormal gene expression. PRMT5 remains important for articular cartilage and hypertrophic cell identity during adult stages, indicating an ongoing role in homeostasis of these tissues. We conclude that PRMT5 is required for distinct steps of early and late chondrogenic specialization and is thus a critical component of multiple aspects of long bone development and maintenance.
Collapse
Affiliation(s)
- Janani Ramachandran
- Department of Molecular Biosciences, University of Texas at Austin, 100 E 24th Street, Stop A5000, Austin, TX, 78712, USA
| | - Zhaoyang Liu
- Department of Pediatrics, Dell Pediatrics Research Institute, University of Texas at Austin Dell Medical School, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Ryan S Gray
- Department of Nutritional Sciences, University of Texas at Austin, 103 W. 24th Street, A2703, Austin, TX, 78712, USA; Department of Pediatrics, Dell Pediatrics Research Institute, University of Texas at Austin Dell Medical School, 1400 Barbara Jordan Blvd, Austin, TX, 78723, USA
| | - Steven A Vokes
- Department of Molecular Biosciences, University of Texas at Austin, 100 E 24th Street, Stop A5000, Austin, TX, 78712, USA.
| |
Collapse
|
161
|
Ouyang Y, Wang W, Tu B, Zhu Y, Fan C, Li Y. Overexpression of SOX9 alleviates the progression of human osteoarthritis in vitro and in vivo. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:2833-2842. [PMID: 31496660 PMCID: PMC6698167 DOI: 10.2147/dddt.s203974] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 06/29/2019] [Indexed: 12/19/2022]
Abstract
Purpose Recent findings have identified that SOX9 served as a key role during the pathogenesis of osteoarthritis (OA). This study aimed to investigate the mechanisms by which SOX9 regulated the formation of OA in vitro and in vivo. Materials and methods The relative expressions of SOX9 in patients with OA and normal fracture of thighbone were analyzed by real-time-PCR. In vitro, IL-1β induced inflammatory response in human chondrocytes was used to evaluate the function of SOX9. The recombinant SOX9 lentivirus vector (Lenti-SOX9) was used to upregulate the expression of SOX9 in cells. ELISA was used to measure the concentration of tumor necrosis factor-α (TNF-α). The protein expressions of SOX9, matrix metalloproteinase-13 (MMP13), Collagen II, Aggrecan and Smad3 were analyzed by Western blot. Cell proliferation and cell apoptosis were detected by CCK-8 assay and flow cytometry, respectively. In vivo, the effect of SOX9 on surgically induced OA mice was evaluated. Results The gene level of SOX9 was remarkably downregulated in patients with OA compared with normal people, while the concentration of TNF-α was upregulated. In addition, IL-1β reduced the expressions of SOX9, Collagen II and Aggrecan and increased the level of MMP13 in chondrocytes. Moreover, Lenti-SOX9 notably inhibited IL-1β-induced growth inhibition and apoptosis in chondrocytes via increasing the expression of Smad3. Finally, Lenti-SOX9 markedly alleviated the symptoms of OA mice in vivo. Conclusion Upregulation of SOX9 inhibited IL-1β-induced inflammatory response via increasing the level Smad3 in human chondrocytes and exhibited therapeutic effect on surgically induced OA mice in vivo. Therefore, SOX9 may serve as a potential target in the treatment of OA in the future.
Collapse
Affiliation(s)
- Yuanming Ouyang
- Department of Orthopedics, Shanghai Sixth People's Hospital East Campus Shanghai University of Medicine and Health, Shanghai 201306, People's Republic of China.,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Wei Wang
- Department of Orthopedics, Shanghai Sixth People's Hospital East Campus Shanghai University of Medicine and Health, Shanghai 201306, People's Republic of China.,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Bing Tu
- Department of Orthopedics, Shanghai Sixth People's Hospital East Campus Shanghai University of Medicine and Health, Shanghai 201306, People's Republic of China.,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yi Zhu
- Department of Orthopedics, Shanghai Sixth People's Hospital East Campus Shanghai University of Medicine and Health, Shanghai 201306, People's Republic of China.,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Sixth People's Hospital East Campus Shanghai University of Medicine and Health, Shanghai 201306, People's Republic of China.,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| | - Yanfeng Li
- Department of Orthopedics, Shanghai Sixth People's Hospital East Campus Shanghai University of Medicine and Health, Shanghai 201306, People's Republic of China.,Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China
| |
Collapse
|
162
|
SOX9 in cartilage development and disease. Curr Opin Cell Biol 2019; 61:39-47. [PMID: 31382142 DOI: 10.1016/j.ceb.2019.07.008] [Citation(s) in RCA: 166] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 07/06/2019] [Indexed: 12/18/2022]
Abstract
SOX9 is a pivotal transcription factor in chondrocytes, a lineage essential in skeletogenesis. Its mandatory role in transactivating many cartilage-specific genes is well established, whereas its pioneer role in lineage specification, which along with transactivation defines master transcription factors, remains to be better defined. Abundant, but yet incomplete evidence exists that intricate molecular networks control SOX9 activity during the multi-step chondrogenesis pathway. They include a highly modular genetic regulation, post-transcriptional and post-translational modifications, and varying sets of functional partners. Fully uncovering SOX9 actions and regulation is fundamental to explain mechanisms underlying many diseases that directly or indirectly affect SOX9 activities and to design effective disease treatments. We here review current knowledge, highlight recent discoveries, and propose new research directions to answer remaining questions.
Collapse
|
163
|
Diaz-Rodriguez P, Erndt-Marino JD, Gharat T, Munoz Pinto DJ, Samavedi S, Bearden R, Grunlan MA, Saunders WB, Hahn MS. Toward zonally tailored scaffolds for osteochondral differentiation of synovial mesenchymal stem cells. J Biomed Mater Res B Appl Biomater 2019; 107:2019-2029. [PMID: 30549205 PMCID: PMC6934364 DOI: 10.1002/jbm.b.34293] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 10/22/2018] [Accepted: 11/10/2018] [Indexed: 12/15/2022]
Abstract
Synovium-derived mesenchymal stem cells (SMSCs) are an emerging cell source for regenerative medicine applications, including osteochondral defect (OCD) repair. However, in contrast to bone marrow MSCs, scaffold compositions which promote SMSC chondrogenesis/osteogenesis are still being identified. In the present manuscript, we examine poly(ethylene) glycol (PEG)-based scaffolds containing zonally-specific biochemical cues to guide SMSC osteochondral differentiation. Specifically, SMSCs were encapsulated in PEG-based scaffolds incorporating glycosaminoglycans (hyaluronan or chondroitin-6-sulfate [CSC]), low-dose of chondrogenic and osteogenic growth factors (TGFβ1 and BMP2, respectively), or osteoinductive poly(dimethylsiloxane) (PDMS). Initial studies suggested that PEG-CSC-TGFβ1 scaffolds promoted enhanced SMSC chondrogenic differentiation, as assessed by significant increases in Sox9 and aggrecan. Conversely, PEG-PDMS-BMP2 scaffolds stimulated increased levels of osteoblastic markers with significant mineral deposition. A "Transition" zone formulation was then developed containing a graded mixture of the chondrogenic and osteogenic signals present in the PEG-CSC-TGFβ1 and PEG-PDMS-BMP2 constructs. SMSCs within the "Transition" formulation displayed a phenotypic profile similar to hypertrophic chondrocytes, with the highest expression of collagen X, intermediate levels of osteopontin, and mineralization levels equivalent to "bone" formulations. Overall, these results suggest that a graded transition from PEG-CSC-TGFβ1 to PEG-PDMS-BMP2 scaffolds elicits a gradual SMSC phenotypic shift from chondrocyte to hypertrophic chondrocyte to osteoblast-like. As such, further development of these scaffold formulations for use in SMSC-based OCD repair is warranted. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 2019-2029, 2019.
Collapse
Affiliation(s)
| | - Josh D Erndt-Marino
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Tanmay Gharat
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Dany J Munoz Pinto
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Satyavrata Samavedi
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, New York
| | - Robert Bearden
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Melissa A Grunlan
- Department of Biomedical Engineering, Texas A&M University, College Station, Texas
| | - W Brian Saunders
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, Texas
| | - Mariah S Hahn
- Department of Chemical and Biological Engineering, Rensselaer Polytechnic Institute, Troy, New York
| |
Collapse
|
164
|
Lui JC, Yue S, Lee A, Kikani B, Temnycky A, Barnes KM, Baron J. Persistent Sox9 expression in hypertrophic chondrocytes suppresses transdifferentiation into osteoblasts. Bone 2019; 125:169-177. [PMID: 31121357 PMCID: PMC7558415 DOI: 10.1016/j.bone.2019.05.027] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 05/09/2019] [Accepted: 05/19/2019] [Indexed: 11/25/2022]
Abstract
Longitudinal bone growth is driven by endochondral ossification, a process in which cartilage tissue is generated by growth plate chondrocytes and then remodeled into bone by osteoblasts. In the postnatal growth plate, as hypertrophic chondrocytes approach the chondro-osseous junction, they may undergo apoptosis, or directly transdifferentiate into osteoblasts. The molecular mechanisms governing this switch in cell lineage are poorly understood. Here we show that the physiological downregulation of Sox9 in hypertrophic chondrocyte is associated with upregulation of osteoblast-associated genes (such as Mmp13, Cola1, Ibsp) in hypertrophic chondrocytes, before they enter the metaphyseal bone. In transgenic mice that continued to express Sox9 in all cells derived from the chondrocytic lineage, upregulation of these osteoblast-associated genes in the hypertrophic zone failed to occur. Furthermore, lineage tracing experiments showed that, in transgenic mice expressing Sox9, the number of chondrocytes transdifferentiating into osteoblasts was markedly reduced. Collectively, our findings suggest that Sox9 downregulation in hypertrophic chondrocytes promotes expression of osteoblast-associated genes in hypertrophic chondrocytes and promotes the subsequent transdifferentiation of these cells into osteoblasts.
Collapse
Affiliation(s)
- Julian C Lui
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America.
| | - Shanna Yue
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Audrey Lee
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Bijal Kikani
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Adrian Temnycky
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Kevin M Barnes
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America
| | - Jeffrey Baron
- Section on Growth and Development, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, United States of America
| |
Collapse
|
165
|
Abstract
PURPOSE OF REVIEW The goal of the review is to summarize the current knowledge on the process of chondrocyte-to-osteoblast transdifferentiation during endochondral bone formation and its potential implications in fracture healing and disease. RECENT FINDINGS Lineage tracing experiments confirmed the transdifferentiation of chondrocytes into osteoblasts. More recent studies lead to the discovery of molecules involved in this process, as well as to the hypothesis that these cells may re-enter a stem cell-like phase prior to their osteoblastic differentiation. This review recapitulates the current knowledge regarding chondrocyte transdifferentiating into osteoblasts, the developmental and postnatal events where transdifferentiation appears to be relevant, and the molecules implicated in this process.
Collapse
Affiliation(s)
- Lena Ingeborg Wolff
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University Münster, Munster, Germany
| | - Christine Hartmann
- Institute of Musculoskeletal Medicine, Department of Bone and Skeletal Research, Medical Faculty of the Westphalian Wilhelms University Münster, Munster, Germany.
| |
Collapse
|
166
|
Khanshour AM, Kou I, Fan Y, Einarsdottir E, Makki N, Kidane YH, Kere J, Grauers A, Johnson TA, Paria N, Patel C, Singhania R, Kamiya N, Takeda K, Otomo N, Watanabe K, Luk KDK, Cheung KMC, Herring JA, Rios JJ, Ahituv N, Gerdhem P, Gurnett CA, Song YQ, Ikegawa S, Wise CA. Genome-wide meta-analysis and replication studies in multiple ethnicities identify novel adolescent idiopathic scoliosis susceptibility loci. Hum Mol Genet 2019; 27:3986-3998. [PMID: 30395268 DOI: 10.1093/hmg/ddy306] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 08/20/2018] [Indexed: 12/13/2022] Open
Abstract
Adolescent idiopathic scoliosis (AIS) is the most common musculoskeletal disorder of childhood development. The genetic architecture of AIS is complex, and the great majority of risk factors are undiscovered. To identify new AIS susceptibility loci, we conducted the first genome-wide meta-analysis of AIS genome-wide association studies, including 7956 cases and 88 459 controls from 3 ancestral groups. Three novel loci that surpassed genome-wide significance were uncovered in intragenic regions of the CDH13 (P-value_rs4513093 = 1.7E-15), ABO (P-value_ rs687621 = 7.3E-10) and SOX6 (P-value_rs1455114 = 2.98E-08) genes. Restricting the analysis to females improved the associations at multiple loci, most notably with variants within CDH13 despite the reduction in sample size. Genome-wide gene-functional enrichment analysis identified significant perturbation of pathways involving cartilage and connective tissue development. Expression of both SOX6 and CDH13 was detected in cartilage chondrocytes and chromatin immunoprecipitation sequencing experiments in that tissue revealed multiple HeK27ac-positive peaks overlapping associated loci. Our results further define the genetic architecture of AIS and highlight the importance of vertebral cartilage development in its pathogenesis.
Collapse
Affiliation(s)
- Anas M Khanshour
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Ikuyo Kou
- Laboratory of Bone & Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Yanhui Fan
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Elisabet Einarsdottir
- Folkhälsan Institute of Genetics, University of Helsinki, 00014 University of Helsinki, Finland.,Molecular Neurology Research Program, University of Helsinki, 00014 University of Helsinki, Finland.,Department of Biosciences & Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Nadja Makki
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Yared H Kidane
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Juha Kere
- Folkhälsan Institute of Genetics, University of Helsinki, 00014 University of Helsinki, Finland.,Molecular Neurology Research Program, University of Helsinki, 00014 University of Helsinki, Finland.,Department of Medical & Molecular Genetics, King's College London, Guy's Hospital, London SE1 9RT, UK.,Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, K54 Huddinge, Stockholm, Sweden
| | - Anna Grauers
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, K54 Huddinge, Stockholm, Sweden.,Department of Orthopedics, Sundsvall and Härnösand County Hospital, Sundsvall, Sweden
| | - Todd A Johnson
- Laboratory of Bone & Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Nandina Paria
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Chandreshkumar Patel
- McDermott Center for Human Growth & Development, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Richa Singhania
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | | | - Kazuki Takeda
- Laboratory of Bone & Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Nao Otomo
- Laboratory of Bone & Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan.,Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kota Watanabe
- Department of Orthopaedic Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Keith D K Luk
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - Kenneth M C Cheung
- Department of Orthopaedics & Traumatology, The University of Hong Kong, Hong Kong, China
| | - John A Herring
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA.,Department of Orthopaedic Surgery, Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan J Rios
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA.,McDermott Center for Human Growth & Development, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Orthopaedic Surgery, Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nadav Ahituv
- Department of Bioengineering & Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
| | - Paul Gerdhem
- Department of Clinical Science, Intervention & Technology (CLINTEC), Karolinska Institutet, K54 Huddinge, Stockholm, Sweden.,Department of Orthopedics, Karolinska University Hospital, K54 Huddinge, Stockholm, Sweden
| | - Christina A Gurnett
- Department of Neurology, School of Medicine, Washington University, St. Louis, MO, USA
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong, China
| | - Shiro Ikegawa
- Laboratory of Bone & Joint Diseases, RIKEN Center for Integrative Medical Sciences, Tokyo, Japan
| | - Carol A Wise
- Sarah M. & Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA.,McDermott Center for Human Growth & Development, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Department of Orthopaedic Surgery, Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
167
|
Diederichs S, Tonnier V, März M, Dreher SI, Geisbüsch A, Richter W. Regulation of WNT5A and WNT11 during MSC in vitro chondrogenesis: WNT inhibition lowers BMP and hedgehog activity, and reduces hypertrophy. CELLULAR AND MOLECULAR LIFE SCIENCES : CMLS 2019. [PMID: 30980110 DOI: 10.1007/s00018‐019‐03099‐0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Re-directing mesenchymal stromal cell (MSC) chondrogenesis towards a non-hypertrophic articular chondrocyte-(AC)-like phenotype is important for improving articular cartilage neogenesis to enhance clinical cartilage repair strategies. This study is the first to demonstrate that high levels of non-canonical WNT5A followed by WNT11 and LEF1 discriminated MSC chondrogenesis from AC re-differentiation. Moreover, β-catenin seemed incompletely silenced in differentiating MSCs, which altogether suggested a role for WNT signaling in hypertrophic MSC differentiation. WNT inhibition with the small molecule IWP-2 supported MSC chondrogenesis according to elevated proteoglycan deposition and reduced the characteristic upregulation of BMP4, BMP7 and their target ID1, as well as IHH and its target GLI1 observed during endochondral differentiation. Along with the pro-hypertrophic transcription factor MEF2C, multiple hypertrophic downstream targets including IBSP and alkaline phosphatase activity were reduced by IWP-2, demonstrating that WNT activity drives BMP and hedgehog upregulation, and MSC hypertrophy. WNT inhibition almost matched the strong anti-hypertrophic capacity of pulsed parathyroid hormone-related protein application, and both outperformed suppression of BMP signaling with dorsomorphin, which also reduced cartilage matrix deposition. Yet, hypertrophic marker expression under IWP-2 remained above AC level, and in vivo mineralization and ectopic bone formation were reduced but not eliminated. Overall, the strong anti-hypertrophic effects of IWP-2 involved inhibition but not silencing of pro-hypertrophic BMP and IHH pathways, and more advanced silencing of WNT activity as well as combined application of IHH or BMP antagonists should next be considered to install articular cartilage neogenesis from human MSCs.
Collapse
Affiliation(s)
- Solvig Diederichs
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Veronika Tonnier
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Melanie März
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Simon I Dreher
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Geisbüsch
- Clinic for Orthopaedics and Trauma Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Wiltrud Richter
- Research Center for Experimental Orthopaedics, Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
168
|
Pfeifer CG, Karl A, Kerschbaum M, Berner A, Lang S, Schupfner R, Koch M, Angele P, Nerlich M, Mueller MB. TGF- β Signalling is Suppressed under Pro-Hypertrophic Conditions in MSC Chondrogenesis Due to TGF- β Receptor Downregulation. Int J Stem Cells 2019; 12:139-150. [PMID: 30836731 PMCID: PMC6457698 DOI: 10.15283/ijsc18088] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 12/21/2018] [Accepted: 12/28/2018] [Indexed: 02/06/2023] Open
Abstract
Background and Objectives Mesenchymal stem cells (MSCs) become hypertrophic in long term despite chondrogenic differentiation following the pathway of growth plate chondrocytes. This terminal differentiation leads to phenotypically unstable cartilage and was mirrored in vitro by addition of hypertrophy inducing medium. We investigated how intrinsic TGF-β signaling is altered in pro-hypertrophic conditions. Methods and Results Human bone marrow derived MSC were chondrogenically differentiated in 3D culture. At day 14 medium conditions were changed to 1. pro-hypertrophic by addition of T3 and withdrawal of TGF-β and dexamethasone 2. pro-hypertrophic by addition of BMP 4 and withdrawal of TGF-β and dexamethasone and 3. kept in prochondrogenic medium conditions. All groups were treated with and without TGFβ-type-1-receptor inhibitor SB431542 from day 14 on. Aggregates were harvested for histo- and immunohistological analysis at d14 and d28, for gene expression analysis (rt-PCR) on d1, d3, d7, d14, d17, d21 and d28 and for Western blot analysis on d21 and d28. Induction of hypertrophy was achieved in the pro-hypertrophic groups while expression of TGFβ-type-1- and 2-receptor and Sox 9 were significantly downregulated compared to pro-chondrogenic conditions. Western blotting showed reduced phosphorylation of Smad 2 and 3 in hypertrophic samples, reduced TGF-β-1 receptor proteins and reduced SOX 9. Addition of SB431542 did not initiate hypertrophy under pro-chondrogenic conditions, but was capable of enhancing hypertrophy when applied simultaneously with BMP-4. Conclusions Our results suggest that the enhancement of hypertrophy in this model is a result of both activation of pro-hypertrophic BMP signaling and reduction of anti-hypertrophic TGFβ signaling.
Collapse
Affiliation(s)
- Christian G Pfeifer
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Alexandra Karl
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Maximilian Kerschbaum
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Arne Berner
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Siegmund Lang
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Rupert Schupfner
- Department of Trauma and Reconstructive Surgery, Klinikum Bayreuth, Bayreuth, Germany
| | - Matthias Koch
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Peter Angele
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Michael Nerlich
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany
| | - Michael B Mueller
- Laboratory of Experimental Trauma Surgery, Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma Surgery, University Regensburg Medical Centre, Regensburg, Germany.,Department of Trauma and Reconstructive Surgery, Klinikum Bayreuth, Bayreuth, Germany
| |
Collapse
|
169
|
Abstract
SOX transcription factors participate in the specification, differentiation and activities of many cell types in development and beyond. The 20 mammalian family members are distributed into eight groups based on sequence identity, and while co-expressed same-group proteins often have redundant functions, different-group proteins typically have distinct functions. More than a handful of SOX proteins have pivotal roles in skeletogenesis. Heterozygous mutations in their genes cause human diseases, in which skeletal dysmorphism is a major feature, such as campomelic dysplasia (SOX9), or a minor feature, such as LAMSHF syndrome (SOX5) and Coffin-Siris-like syndromes (SOX4 and SOX11). Loss- and gain-of-function experiments in animal models have revealed that SOX4 and SOX11 (SOXC group) promote skeletal progenitor survival and control skeleton patterning and growth; SOX8 (SOXE group) delays the differentiation of osteoblast progenitors; SOX9 (SOXE group) is essential for chondrocyte fate maintenance and differentiation, and works in cooperation with SOX5 and SOX6 (SOXD group) and other types of transcription factors. These and other SOX proteins have also been proposed, mainly through in vitro experiments, to have key roles in other aspects of skeletogenesis, such as SOX2 in osteoblast stem cell self-renewal. We here review current knowledge of well-established and proposed skeletogenic roles of SOX proteins, their transcriptional and non-transcriptional actions, and their modes of regulation at the gene, RNA and protein levels. We also discuss gaps in knowledge and directions for future research to further decipher mechanisms underlying skeletogenesis in health and diseases and identify treatment options for skeletal malformation and degeneration diseases.
Collapse
Affiliation(s)
- Véronique Lefebvre
- The Children's Hospital of Philadelphia, Philadelphia, PA, United States.
| |
Collapse
|
170
|
Merceron C, Ranganathan K, Wang E, Tata Z, Makkapati S, Khan MP, Mangiavini L, Yao AQ, Castellini L, Levi B, Giaccia AJ, Schipani E. Hypoxia-inducible factor 2α is a negative regulator of osteoblastogenesis and bone mass accrual. Bone Res 2019; 7:7. [PMID: 30792937 PMCID: PMC6382776 DOI: 10.1038/s41413-019-0045-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 11/28/2018] [Accepted: 12/12/2018] [Indexed: 12/24/2022] Open
Abstract
Osteoblasts, which are the bone-forming cells, operate in a hypoxic environment. The transcription factors hypoxia-inducible factor-1α (HIF1) and HIF2 are key mediators of the cellular response to hypoxia. Both are expressed in osteoblasts. HIF1 is known to be a positive regulator of bone formation. Conversely, the role of HIF2 in the control osteoblast biology is still poorly understood. In this study, we used mouse genetics to demonstrate that HIF2 is an inhibitor of osteoblastogenesis and bone mass accrual. Moreover, we provided evidence that HIF2 impairs osteoblast differentiation at least in part, by upregulating the transcription factor Sox9. Our findings constitute a paradigm shift, as activation of the hypoxia-signaling pathway has traditionally been associated with increased bone formation through HIF1. Inhibiting HIF2 could thus represent a therapeutic approach for the treatment of the low bone mass observed in chronic diseases, osteoporosis, or aging.
Collapse
Affiliation(s)
- Christophe Merceron
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Kavitha Ranganathan
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI USA
| | - Elizabeth Wang
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Zachary Tata
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Shreya Makkapati
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Mohd Parvez Khan
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Laura Mangiavini
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Angela Qing Yao
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| | - Laura Castellini
- Department of Radiation Oncology, Stanford University Medical School, Stanford, CA USA
| | - Benjamin Levi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, University of Michigan, Ann Arbor, MI USA
| | - Amato J. Giaccia
- Department of Radiation Oncology, Stanford University Medical School, Stanford, CA USA
| | - Ernestina Schipani
- Department of Orthopaedic Surgery, School of Medicine, University of Michigan, Ann Arbor, MI USA
| |
Collapse
|
171
|
Abstract
Accumulating evidence supports the idea that stem and progenitor cells play important roles in skeletal development. Over the last decade, the definition of skeletal stem and progenitor cells has evolved from cells simply defined by their in vitro behaviors to cells fully defined by a combination of sophisticated approaches, including serial transplantation assays and in vivo lineage-tracing experiments. These approaches have led to better identification of the characteristics of skeletal stem cells residing in multiple sites, including the perichondrium of the fetal bone, the resting zone of the postnatal growth plate, the bone marrow space and the periosteum in adulthood. These diverse groups of skeletal stem cells appear to closely collaborate and achieve a number of important biological functions of bones, including not only bone development and growth, but also bone maintenance and repair. Although these are important findings, we are only beginning to understand the diversity and the nature of skeletal stem and progenitor cells, and how they actually behave in vivo.
Collapse
Affiliation(s)
- Noriaki Ono
- University of Michigan School of Dentistry, Ann Arbor, MI, United States.
| | - Deepak H Balani
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Henry M Kronenberg
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
172
|
Coculture of hWJMSCs and pACs in Oriented Scaffold Enhances Hyaline Cartilage Regeneration In Vitro. Stem Cells Int 2019; 2019:5130152. [PMID: 30881462 PMCID: PMC6383394 DOI: 10.1155/2019/5130152] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 11/27/2018] [Indexed: 12/12/2022] Open
Abstract
Seed cells of articular cartilage tissue engineering face many obstacles in their application because of the dedifferentiation of chondrocytes or unstable chondrogenic differentiation status of pluripotent stem cells. To overcome mentioned dilemmas, a simulation of the articular cartilage microenvironment was constructed by primary articular cartilage cells (pACs) and acellular cartilage extracellular matrix- (ACECM-) oriented scaffold cocultured with human umbilical cord Wharton's jelly-derived mesenchymal stem cells (hWJMSCs) in vitro. The coculture groups showed more affluent cartilage special matrix ingredients including collagen II and aggrecan based on the results of histological staining and western blotting and cut down as many pACs as possible. The RT-PCR and cell viability experiments also demonstrated that hWJMSCs were successfully induced to differentiate into chondrocytes when cultured in the simulated cartilage microenvironment, as confirmed by the significant upregulation of collagen II and aggrecan, while the cell proliferation activity of pACs was significantly improved by cell-cell interactions. Therefore, compared with monoculture and chondrogenic induction of inducers, coculture providing a simulated native articular microenvironment was a potential and temperate way to regulate the biological behaviors of pACs and hWJMSCs to regenerate the hyaline articular cartilage.
Collapse
|
173
|
Johanson Z, Martin K, Fraser G, James K. The Synarcual of the Little Skate, Leucoraja erinacea: Novel Development Among the Vertebrates. Front Ecol Evol 2019. [DOI: 10.3389/fevo.2019.00012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
174
|
Shen B, Vardy K, Hughes P, Tasdogan A, Zhao Z, Yue R, Crane GM, Morrison SJ. Integrin alpha11 is an Osteolectin receptor and is required for the maintenance of adult skeletal bone mass. eLife 2019; 8:42274. [PMID: 30632962 PMCID: PMC6349404 DOI: 10.7554/elife.42274] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/05/2019] [Indexed: 12/13/2022] Open
Abstract
We previously discovered a new osteogenic growth factor that is required to maintain adult skeletal bone mass, Osteolectin/Clec11a. Osteolectin acts on Leptin Receptor+ (LepR+) skeletal stem cells and other osteogenic progenitors in bone marrow to promote their differentiation into osteoblasts. Here we identify a receptor for Osteolectin, integrin α11, which is expressed by LepR+ cells and osteoblasts. α11β1 integrin binds Osteolectin with nanomolar affinity and is required for the osteogenic response to Osteolectin. Deletion of Itga11 (which encodes α11) from mouse and human bone marrow stromal cells impaired osteogenic differentiation and blocked their response to Osteolectin. Like Osteolectin deficient mice, Lepr-cre; Itga11fl/fl mice appeared grossly normal but exhibited reduced osteogenesis and accelerated bone loss during adulthood. Osteolectin binding to α11β1 promoted Wnt pathway activation, which was necessary for the osteogenic response to Osteolectin. This reveals a new mechanism for maintenance of adult bone mass: Wnt pathway activation by Osteolectin/α11β1 signaling. Throughout our lives, our bones undergo constant remodeling. Cells called osteoclasts break down old bone and cells called osteoblasts lay down new. Normally, the two cell types work in balance but if the rate of breakdown outpaces new bone formation the skeleton can become weak. This weakness leads to a condition called osteoporosis, in which people suffer from fragile bones. Osteoporosis is hard to reverse, in part because our ability to encourage new bone to form is limited. In 2016, researchers discovered a protein called osteolectin, which promotes new bone formation during adulthood by helping skeletal stem cells transform into bone cells. But so far, it has been unclear how osteolectin achieves this. To investigate this further, Shen et al. – including some researchers involved in the 2016 study – marked osteolectin with a molecular tag and tested what it bound on the surface of mouse and human bone marrow cells. The experiments revealed that osteolectin binds to a specific receptor protein called α11 integrin, which can only be found on skeletal stem cells and the osteoblasts they give rise to. Once osteolectin binds to the receptor, it activates a signaling pathway that induces the stem cells to develop into osteoblasts. Mice that lacked either osteolectin or α11 integrin produced less bone and lost bone tissue faster as adults. Osteolectin could potentially be useful in the treatment of osteoporosis or broken bones. Since only skeletal stem cells and osteoblasts cells produce α11 integrin, osteolectin would specifically target these cells without affecting cells that do not form bones. A next step will be to assess how well osteolectin compares to existing treatments for fragile bones.
Collapse
Affiliation(s)
- Bo Shen
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Kristy Vardy
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Payton Hughes
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Alpaslan Tasdogan
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Zhiyu Zhao
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Rui Yue
- Institute of Regenerative Medicine, Shanghai East Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Genevieve M Crane
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States
| | - Sean J Morrison
- Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, United States.,Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, United States.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, United States
| |
Collapse
|
175
|
Bahney CS, Zondervan RL, Allison P, Theologis A, Ashley JW, Ahn J, Miclau T, Marcucio RS, Hankenson KD. Cellular biology of fracture healing. J Orthop Res 2019; 37:35-50. [PMID: 30370699 PMCID: PMC6542569 DOI: 10.1002/jor.24170] [Citation(s) in RCA: 351] [Impact Index Per Article: 58.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 08/27/2018] [Indexed: 02/04/2023]
Abstract
The biology of bone healing is a rapidly developing science. Advances in transgenic and gene-targeted mice have enabled tissue and cell-specific investigations of skeletal regeneration. As an example, only recently has it been recognized that chondrocytes convert to osteoblasts during healing bone, and only several years prior, seminal publications reported definitively that the primary tissues contributing bone forming cells during regeneration were the periosteum and endosteum. While genetically modified animals offer incredible insights into the temporal and spatial importance of various gene products, the complexity and rapidity of healing-coupled with the heterogeneity of animal models-renders studies of regenerative biology challenging. Herein, cells that play a key role in bone healing will be reviewed and extracellular mediators regulating their behavior discussed. We will focus on recent studies that explore novel roles of inflammation in bone healing, and the origins and fates of various cells in the fracture environment. © 2018 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res.
Collapse
Affiliation(s)
- Chelsea S. Bahney
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Robert L. Zondervan
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| | - Patrick Allison
- Department of Physiology, College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
| | - Alekos Theologis
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Jason W. Ashley
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Jaimo Ahn
- Department of Biology, Eastern Washington University, Cheney, Washington
| | - Theodore Miclau
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Ralph S. Marcucio
- Department of Orthopaedic Surgery, University of California at San Francisco, San Francisco, California
| | - Kurt D. Hankenson
- Department of Orthopaedic Surgery, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
176
|
Ji Q, Zheng Y, Zhang G, Hu Y, Fan X, Hou Y, Wen L, Li L, Xu Y, Wang Y, Tang F. Single-cell RNA-seq analysis reveals the progression of human osteoarthritis. Ann Rheum Dis 2019; 78:100-110. [PMID: 30026257 PMCID: PMC6317448 DOI: 10.1136/annrheumdis-2017-212863] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 05/21/2018] [Accepted: 05/28/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES Understanding the molecular mechanisms underlying human cartilage degeneration and regeneration is helpful for improving therapeutic strategies for treating osteoarthritis (OA). Here, we report the molecular programmes and lineage progression patterns controlling human OA pathogenesis using single-cell RNA sequencing (scRNA-seq). METHODS We performed unbiased transcriptome-wide scRNA-seq analysis, computational analysis and histological assays on 1464 chondrocytes from 10 patients with OA undergoing knee arthroplasty surgery. We investigated the relationship between transcriptional programmes of the OA landscape and clinical outcome using severity index and correspondence analysis. RESULTS We identified seven molecularly defined populations of chondrocytes in the human OA cartilage, including three novel phenotypes with distinct functions. We presented gene expression profiles at different OA stages at single-cell resolution. We found a potential transition among proliferative chondrocytes, prehypertrophic chondrocytes and hypertrophic chondrocytes (HTCs) and defined a new subdivision within HTCs. We revealed novel markers for cartilage progenitor cells (CPCs) and demonstrated a relationship between CPCs and fibrocartilage chondrocytes using computational analysis. Notably, we derived predictive targets with respect to clinical outcomes and clarified the role of different cell types for the early diagnosis and treatment of OA. CONCLUSIONS Our results provide new insights into chondrocyte taxonomy and present potential clues for effective and functional manipulation of human OA cartilage regeneration that could lead to improved health.
Collapse
Affiliation(s)
- Quanbo Ji
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing, China
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
| | - Yuxuan Zheng
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Guoqiang Zhang
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Yuqiong Hu
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Xiaoying Fan
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
| | - Yu Hou
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
| | - Lu Wen
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
| | - Li Li
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
| | - Yameng Xu
- Department of Traditional Chinese Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan Wang
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing, China
| | - Fuchou Tang
- Biomedical Institute for Pioneering Investigation via Convergence and Ministry of Education Key Laboratory of Cell Proliferation and Differentiation, Beijing, China
- Beijing Advanced Innovation Center for Genomics (ICG), College of Life Science, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| |
Collapse
|
177
|
Baker CE, Moore-Lotridge SN, Hysong AA, Posey SL, Robinette JP, Blum DM, Benvenuti MA, Cole HA, Egawa S, Okawa A, Saito M, McCarthy JR, Nyman JS, Yuasa M, Schoenecker JG. Bone Fracture Acute Phase Response-A Unifying Theory of Fracture Repair: Clinical and Scientific Implications. Clin Rev Bone Miner Metab 2018; 16:142-158. [PMID: 30930699 PMCID: PMC6404386 DOI: 10.1007/s12018-018-9256-x] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Bone fractures create five problems that must be resolved: bleeding, risk of infection, hypoxia, disproportionate strain, and inability to bear weight. There have been enormous advancements in our understanding of the molecular mechanisms that resolve these problems after fractures, and in best clinical practices of repairing fractures. We put forth a modern, comprehensive model of fracture repair that synthesizes the literature on the biology and biomechanics of fracture repair to address the primary problems of fractures. This updated model is a framework for both fracture management and future studies aimed at understanding and treating this complex process. This model is based upon the fracture acute phase response (APR), which encompasses the molecular mechanisms that respond to injury. The APR is divided into sequential stages of "survival" and "repair." Early in convalescence, during "survival," bleeding and infection are resolved by collaborative efforts of the hemostatic and inflammatory pathways. Later, in "repair," avascular and biomechanically insufficient bone is replaced by a variable combination of intramembranous and endochondral ossification. Progression to repair cannot occur until survival has been ensured. A disproportionate APR-either insufficient or exuberant-leads to complications of survival (hemorrhage, thrombosis, systemic inflammatory response syndrome, infection, death) and/or repair (delayed- or non-union). The type of ossification utilized for fracture repair is dependent on the relative amounts of strain and vascularity in the fracture microenvironment, but any failure along this process can disrupt or delay fracture healing and result in a similar non-union. Therefore, incomplete understanding of the principles herein can result in mismanagement of fracture care or application of hardware that interferes with fracture repair. This unifying model of fracture repair not only informs clinicians how their interventions fit within the framework of normal biological healing but also instructs investigators about the critical variables and outputs to assess during a study of fracture repair.
Collapse
Affiliation(s)
- Courtney E Baker
- 1Department of Orthopaedics, Mayo Clinic, 200 1st Ave SW, Rochester, MN 55903 USA
| | - Stephanie N Moore-Lotridge
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,3Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN 37232 USA
| | - Alexander A Hysong
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - Samuel L Posey
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - J Patton Robinette
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - Deke M Blum
- 4Vanderbilt University School of Medicine, 1161 21st Ave S, #D3300, Nashville, TN 37232 USA
| | - Michael A Benvenuti
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA
| | - Heather A Cole
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA
| | - Satoru Egawa
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Atsushi Okawa
- 5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Masanori Saito
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Jason R McCarthy
- Masonic Research Institute, 2150 Bleecker St, Utica, NY 13501 USA
| | - Jeffry S Nyman
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,7Department of Biomedical Engineering, Vanderbilt University, PMB 351631, 2301 Vanderbilt Place, Nashville, TN 37235 USA.,Department of Veterans Affairs, Tennessee Valley Health Care System, F-519 VA Acre Building, 1210 24th Ave. South, Nashville, TN 37232 USA
| | - Masato Yuasa
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,5Department of Orthopaedic Surgery, Tokyo Medical and Dental University, Yushima Bunkyo Ward, Tokyo, 113-8519 Japan
| | - Jonathan G Schoenecker
- 2Department of Orthopaedics and Rehabilitation, Vanderbilt University Medical Center, 1215 21st Ave. South, Suite 4200 MCE, South Tower, Nashville, TN 37232 USA.,3Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, 1161 21st Ave. South, Nashville, TN 37232 USA.,9Department of Pharmacology, Vanderbilt University, 2200 Pierce Ave, Robinson Research Building, Nashville, TN 37232 USA.,10Department of Pediatrics, Vanderbilt University Medical Center, 4202 Doctor's Office Tower, 2200 Children's Way, Nashville, TN 37232 USA
| |
Collapse
|
178
|
Graceffa V, Vinatier C, Guicheux J, Evans CH, Stoddart M, Alini M, Zeugolis DI. State of art and limitations in genetic engineering to induce stable chondrogenic phenotype. Biotechnol Adv 2018; 36:1855-1869. [PMID: 30012541 DOI: 10.1016/j.biotechadv.2018.07.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 05/16/2018] [Accepted: 07/12/2018] [Indexed: 12/18/2022]
|
179
|
Schliermann A, Nickel J. Unraveling the Connection between Fibroblast Growth Factor and Bone Morphogenetic Protein Signaling. Int J Mol Sci 2018; 19:ijms19103220. [PMID: 30340367 PMCID: PMC6214098 DOI: 10.3390/ijms19103220] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 10/07/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022] Open
Abstract
Ontogeny of higher organisms as well the regulation of tissue homeostasis in adult individuals requires a fine-balanced interplay of regulating factors that individually trigger the fate of particular cells to either stay undifferentiated or to differentiate towards distinct tissue specific lineages. In some cases, these factors act synergistically to promote certain cellular responses, whereas in other tissues the same factors antagonize each other. However, the molecular basis of this obvious dual signaling activity is still only poorly understood. Bone morphogenetic proteins (BMPs) and fibroblast growth factors (FGFs) are two major signal protein families that have a lot in common: They are both highly preserved between different species, involved in essential cellular functions, and their ligands vastly outnumber their receptors, making extensive signal regulation necessary. In this review we discuss where and how BMP and FGF signaling cross paths. The compiled data reflect that both factors synchronously act in many tissues, and that antagonism and synergism both exist in a context-dependent manner. Therefore, by challenging a generalization of the connection between these two pathways a new chapter in BMP FGF signaling research will be introduced.
Collapse
Affiliation(s)
- Anna Schliermann
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
| | - Joachim Nickel
- Lehrstuhl für Tissue Engineering und Regenerative Medizin, Universitätsklinikum Würzburg, Röntgenring 11, 97222 Würzburg, Germany.
- Fraunhofer Institut für Silicatforschung, Translationszentrum TLZ-RT, Röntgenring 11, 97222 Würzburg, Germany.
| |
Collapse
|
180
|
Gao J, Li X, Zhang Y, Wang H. Endochondral ossification in hindlimbs during bufo gargarizans
metamorphosis: A model of studying skeletal development in vertebrates. Dev Dyn 2018; 247:1121-1134. [DOI: 10.1002/dvdy.24669] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/22/2018] [Accepted: 08/22/2018] [Indexed: 12/17/2022] Open
Affiliation(s)
- Jinshu Gao
- College of Life Science; Shaanxi Normal University; Xi'an, 710119 China
| | - Xinyi Li
- College of Life Science; Shaanxi Normal University; Xi'an, 710119 China
| | - Yuhui Zhang
- College of Life Science; Shaanxi Normal University; Xi'an, 710119 China
| | - Hongyuan Wang
- College of Life Science; Shaanxi Normal University; Xi'an, 710119 China
| |
Collapse
|
181
|
Guzhi Zengsheng Zhitongwan, a Traditional Chinese Medicinal Formulation, Stimulates Chondrocyte Proliferation through Control of Multiple Genes Involved in Chondrocyte Proliferation and Differentiation. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 2018:7265939. [PMID: 30275866 PMCID: PMC6157105 DOI: 10.1155/2018/7265939] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 08/07/2018] [Accepted: 08/19/2018] [Indexed: 11/30/2022]
Abstract
Chinese materia medica (CMM) are essential components of traditional Chinese medicine, and Chinese medicinal formulas consisting of 2 or more types of CMM are widely used. These formulations have played a pivotal role in health protection and disease control for thousands of years. Guzhi Zengsheng Zhitongwan (GZZSZTW), which represents one of the Chinese medicinal formulations, has been used for several decades to treat joint diseases. However, the exact molecular mechanism underlying its efficacy in treating osteoarthritis remains to be elucidated. In the present study, we investigated the effects of GZZSZTW on primary chondrocytes. We demonstrated that GZZSZTW significantly promoted chondrocyte viability, maintained chondrocytes in a continuous proliferative state, and prevented their further differentiation. These effects were achieved by the synergistic interactions of various herbs and their active components in GZZSZTW through an increase in the expression levels of functional genes participating in chondrocyte commitment and proliferation and a decrease in the expression levels of genes involved in chondrocyte differentiation. GZZSZTW treatment also decreased the expression levels of genes that inhibited chondrocyte proliferation. Thus, this study has greatly deepened the current knowledge about the molecular effects of GZZSZTW on chondrocytes. It has also shed new light on possible strategies to further prevent and treat cartilage-related diseases by using traditional Chinese medicinal formulations.
Collapse
|
182
|
Cheng X, Li PZ, Wang G, Yan Y, Li K, Brand-Saberi B, Yang X. Microbiota-derived lipopolysaccharide retards chondrocyte hypertrophy in the growth plate through elevating Sox9 expression. J Cell Physiol 2018; 234:2593-2605. [PMID: 30264889 DOI: 10.1002/jcp.27025] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Accepted: 06/25/2018] [Indexed: 12/22/2022]
Abstract
Accumulating data show that the cytotoxicity of bacterial lipopolysaccharides (LPS) from microbiota or infection is associated with many disorders observed in the clinics. However, it is still obscure whether or not embryonic osteogenesis is affected by the LPS exposure during gestation. Using the early chicken embryo model, we could demonstrate that LPS exposure inhibits chondrogenesis of the 8-day chicken embryos by Alcian Blue-staining and osteogenesis of 17-day by Alcian Blue and Alizarin Red staining. Further analysis of the growth plates showed that the length of the proliferating zone (PZ) increases whereas that of the hypertrophic zone (HZ) decreased following LPS exposure. However there is no significant change on cell proliferation in the growth plates. Immunofluorescent staining, western blot analysis, and quantitive polymerase chain reaction revealed that Sox9 and Col2a1 are highly expressed at the messenger RNA level and their protein products are also abundant. LPS exposure causes a downregulation of Runx2 and Col10a1 expression in 8-day hindlimbs, and a suppression of Runx2, Col10a1, and Vegfa expression in 17-day phalanges. Knocking down Sox9 in ATDC5 cells by small interfering RNA transfection lead to the expression reduction of Col2a1, Runx2, and Col10a1, implying the vital role of Sox9 in the process of LPS-induced delay in the transition from proliferating chondrocytes to hypertrophic chondrocytes in the growth plate. In the presence of LPS, the antioxidant defense regulator nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is highly expressed, and the activities of superoxide dismutase 1 (SOD1), SOD2, and glutaredoxin rise in 17-day phalanges and ADTC5 cells. Simultaneously, an increase of intracellular ROS is observed. When Nrf2 expression was knocked down in ATDC5 cells, the expressions of Sox9, Col2a1, Runx2, Col10a1, and Vegfa were also going down as well. Taken together, our current data suggest that LPS exposure during gestation could restrict the chondrocytes conversion from proliferating to hypertrophic in the growth plate, in which LPS-induced Sox9 plays a crucial role to trigger the cascade of downstream genes by excessive ROS production and Nrf2 elevation.
Collapse
Affiliation(s)
- Xin Cheng
- Department of Histology and Embryology, International Joint Laboratory for Embryonic, Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Pei-Zhi Li
- Department of Histology and Embryology, International Joint Laboratory for Embryonic, Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Guang Wang
- Department of Histology and Embryology, International Joint Laboratory for Embryonic, Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Yu Yan
- Department of Histology and Embryology, International Joint Laboratory for Embryonic, Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Ke Li
- Department of Histology and Embryology, International Joint Laboratory for Embryonic, Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Beate Brand-Saberi
- Department of Anatomy and Molecular Embryology, Ruhr-University Bochum, Bochum, Germany
| | - Xuesong Yang
- Department of Histology and Embryology, International Joint Laboratory for Embryonic, Development & Prenatal Medicine, Medical College, Jinan University, Guangzhou, China.,Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| |
Collapse
|
183
|
Jin Y, Cong Q, Gvozdenovic-Jeremic J, Hu J, Zhang Y, Terkeltaub R, Yang Y. Enpp1 inhibits ectopic joint calcification and maintains articular chondrocytes by repressing hedgehog signaling. Development 2018; 145:dev.164830. [PMID: 30111653 DOI: 10.1242/dev.164830] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 07/12/2018] [Indexed: 01/15/2023]
Abstract
The differentiated phenotype of articular chondrocytes of synovial joints needs to be maintained throughout life. Disruption of the articular cartilage, frequently associated with chondrocyte hypertrophy and calcification, is a central feature in osteoarthritis (OA). However, the molecular mechanisms whereby phenotypes of articular chondrocytes are maintained and pathological calcification is inhibited remain poorly understood. Recently, the ecto-enzyme Enpp1, a suppressor of pathological calcification, was reported to be decreased in joint cartilage with OA in both human and mouse, and Enpp1 deficiency causes joint calcification. Here, we found that hedgehog (Hh) signaling activation contributes to ectopic joint calcification in the Enpp1-/- mice. In the Enpp1-/- joints, Hh signaling was upregulated. Further activation of Hh signaling by removing the patched 1 gene in the Enpp1-/- mice enhanced ectopic joint calcification, whereas removing Gli2 partially rescued the ectopic calcification phenotype. In addition, reduction of Gαs in the Enpp1-/- mice enhanced joint calcification, suggesting that Enpp1 inhibits Hh signaling and chondrocyte hypertrophy by activating Gαs-PKA signaling. Our findings provide new insights into the mechanisms underlying Enpp1 regulation of joint integrity.
Collapse
Affiliation(s)
- Yunyun Jin
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA.,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai 200241, China
| | - Qian Cong
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| | | | - Jiajie Hu
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Yiqun Zhang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| | - Robert Terkeltaub
- Department of Medicine, Veterans Affairs Healthcare System, University of California San Diego, 111K, 3350 La Jolla Village Dr., San Diego, CA 92161, USA
| | - Yingzi Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Harvard Stem Cell Institute, Boston, MA 02115, USA
| |
Collapse
|
184
|
Cartilage Differentiation of Bone Marrow-Derived Mesenchymal Stem Cells in Three-Dimensional Silica Nonwoven Fabrics. APPLIED SCIENCES-BASEL 2018. [DOI: 10.3390/app8081398] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
In cartilage tissue engineering, three-dimensional (3D) scaffolds provide native extracellular matrix (ECM) environments that induce tissue ingrowth and ECM deposition for in vitro and in vivo tissue regeneration. In this report, we investigated 3D silica nonwoven fabrics (Cellbed®) as a scaffold for mesenchymal stem cells (MSCs) in cartilage tissue engineering applications. The unique, highly porous microstructure of 3D silica fabrics allows for immediate cell infiltration for tissue repair and orientation of cell–cell interaction. It is expected that the morphological similarity of silica fibers to that of fibrillar ECM contributes to the functionalization of cells. Human bone marrow-derived MSCs were cultured in 3D silica fabrics, and chondrogenic differentiation was induced by culture in chondrogenic differentiation medium. The characteristics of chondrogenic differentiation including cellular growth, ECM deposition of glycosaminoglycan and collagen, and gene expression were evaluated. Because of the highly interconnected network structure, stiffness, and permeability of the 3D silica fabrics, the level of chondrogenesis observed in MSCs seeded within was comparable to that observed in MSCs maintained on atelocollagen gels, which are widely used to study the chondrogenesis of MSCs in vitro and in vivo. These results indicated that 3D silica nonwoven fabrics are a promising scaffold for the regeneration of articular cartilage defects using MSCs, showing the particular importance of high elasticity.
Collapse
|
185
|
Chao W, Zhang Y, Chai L, Wang H. Transcriptomics provides mechanistic indicators of fluoride toxicology on endochondral ossification in the hind limb of Bufo gargarizans. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2018; 201:138-150. [PMID: 29908452 DOI: 10.1016/j.aquatox.2018.06.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 06/02/2018] [Accepted: 06/06/2018] [Indexed: 06/08/2023]
Abstract
Endochondral ossification, the process by which most of the bone is formed, is regulated by many specific groups of molecules and extracellular matrix components. Hind limb of Bufo gargarizans is a model to study endochondral ossification during metamorphosis. Chinese toad (Bufo gargarizans) were exposed to different fluoride concentrations (0, 1, 5, 10 and 20 mg L-1) from G3 to G42. The development of hind limb of B. gargarizans was observed using the double staining methodology. The transcriptome of hind limb of B. gargarizans was conducted using RNA-seq approach, and differentially expressed gene was also validated. In addition, the location of Sox9 and Ihh in the growth cartilage was determined using in situ hybridization. Our results showed that 5 mg L-1 stimulated bone mineralization, while 10 and 20 mg L-1 exposure could inhibit the tibio-fibula, tarsus and metacarpals ossification. Besides, 10 mg F/L treatment could down-regulate Ihh, Sox9, D2, D3, TRα, TRβ, Wnt10, FGF3 and BMP6 expression, while up-regulate ObRb and HHAT mRNA expression in the hind limb of B. gargarizans. Transcript level changes of Ihh, Sox9, D2, D3, TRα, TRβ, Wnt10, FGF3 and BMP6 were consistent with the results of RT-qPCR. In situ hybridization revealed that Ihh was expressed in prehypertrophic chondrocytes, while Sox9 was abundantly expressed in proliferous, prehypertrophic and hypertrophic chondrocytes. However, 10 mg F-/L did not cause any affect in the location of the Ihh and Sox9 mRNA. Therefore, high concentration of fluoride could affect the ossification-related genes mRNA expression and then inhibit the endochondral ossification. The present study thus will greatly contribute to our understanding of the effect of environmental contaminant on ossification in amphibian.
Collapse
Affiliation(s)
- Wu Chao
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China; State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, Jiangsu Province, 210023, China
| | - Yuhui Zhang
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China
| | - Lihong Chai
- School of Environmental Science and Engineering, Chang'an University, Xi'an, 710054, China; Key Laboratory of Subsurface Hydrology and Ecological Effect in Arid Region of Ministry of Education, Xi'an, 710062, China
| | - Hongyuan Wang
- College of Life Science, Shaanxi Normal University, Xi'an, 710119, China.
| |
Collapse
|
186
|
Wang C, Tan Z, Niu B, Tsang KY, Tai A, Chan WCW, Lo RLK, Leung KKH, Dung NWF, Itoh N, Zhang MQ, Chan D, Cheah KSE. Inhibiting the integrated stress response pathway prevents aberrant chondrocyte differentiation thereby alleviating chondrodysplasia. eLife 2018; 7:37673. [PMID: 30024379 PMCID: PMC6053305 DOI: 10.7554/elife.37673] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/05/2018] [Indexed: 12/16/2022] Open
Abstract
The integrated stress response (ISR) is activated by diverse forms of cellular stress, including endoplasmic reticulum (ER) stress, and is associated with diseases. However, the molecular mechanism(s) whereby the ISR impacts on differentiation is incompletely understood. Here, we exploited a mouse model of Metaphyseal Chondrodysplasia type Schmid (MCDS) to provide insight into the impact of the ISR on cell fate. We show the protein kinase RNA-like ER kinase (PERK) pathway that mediates preferential synthesis of ATF4 and CHOP, dominates in causing dysplasia by reverting chondrocyte differentiation via ATF4-directed transactivation of Sox9. Chondrocyte survival is enabled, cell autonomously, by CHOP and dual CHOP-ATF4 transactivation of Fgf21. Treatment of mutant mice with a chemical inhibitor of PERK signaling prevents the differentiation defects and ameliorates chondrodysplasia. By preventing aberrant differentiation, titrated inhibition of the ISR emerges as a rationale therapeutic strategy for stress-induced skeletal disorders.
Collapse
Affiliation(s)
- Cheng Wang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Zhijia Tan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Ben Niu
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Kwok Yeung Tsang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Andrew Tai
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Wilson C W Chan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Rebecca L K Lo
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Keith K H Leung
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Nelson W F Dung
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, University of Kyoto, Kyoto, Japan
| | - Michael Q Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Richardson, United States.,MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, Tsinghua University, Beijing, China
| | - Danny Chan
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, China
| | | |
Collapse
|
187
|
Singh P, Marcu KB, Goldring MB, Otero M. Phenotypic instability of chondrocytes in osteoarthritis: on a path to hypertrophy. Ann N Y Acad Sci 2018; 1442:17-34. [PMID: 30008181 DOI: 10.1111/nyas.13930] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/11/2018] [Accepted: 06/21/2018] [Indexed: 12/24/2022]
Abstract
Articular chondrocytes are quiescent, fully differentiated cells responsible for the homeostasis of adult articular cartilage by maintaining cellular survival functions and the fine-tuned balance between anabolic and catabolic functions. This balance requires phenotypic stability that is lost in osteoarthritis (OA), a disease that affects and involves all joint tissues and especially impacts articular cartilage structural integrity. In OA, articular chondrocytes respond to the accumulation of injurious biochemical and biomechanical insults by shifting toward a degradative and hypertrophy-like state, involving abnormal matrix production and increased aggrecanase and collagenase activities. Hypertrophy is a necessary, transient developmental stage in growth plate chondrocytes that culminates in bone formation; in OA, however, chondrocyte hypertrophy is catastrophic and it is believed to initiate and perpetuate a cascade of events that ultimately result in permanent cartilage damage. Emphasizing changes in DNA methylation status and alterations in NF-κB signaling in OA, this review summarizes the data from the literature highlighting the loss of phenotypic stability and the hypertrophic differentiation of OA chondrocytes as central contributing factors to OA pathogenesis.
Collapse
Affiliation(s)
- Purva Singh
- HSS Research Institute, Hospital for Special Surgery, New York, New York
| | - Kenneth B Marcu
- Biochemistry and Cell Biology Department, Stony Brook University, Stony Brook, New York
| | - Mary B Goldring
- HSS Research Institute, Hospital for Special Surgery, New York, New York.,Department of Cell and Developmental Biology, Weill Cornell Medical College and Weill Cornell Graduate School of Medical Sciences, New York, New York
| | - Miguel Otero
- HSS Research Institute, Hospital for Special Surgery, New York, New York
| |
Collapse
|
188
|
Tan Z, Niu B, Tsang KY, Melhado IG, Ohba S, He X, Huang Y, Wang C, McMahon AP, Jauch R, Chan D, Zhang MQ, Cheah KSE. Synergistic co-regulation and competition by a SOX9-GLI-FOXA phasic transcriptional network coordinate chondrocyte differentiation transitions. PLoS Genet 2018; 14:e1007346. [PMID: 29659575 PMCID: PMC5919691 DOI: 10.1371/journal.pgen.1007346] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 04/26/2018] [Accepted: 03/29/2018] [Indexed: 11/18/2022] Open
Abstract
The growth plate mediates bone growth where SOX9 and GLI factors control chondrocyte proliferation, differentiation and entry into hypertrophy. FOXA factors regulate hypertrophic chondrocyte maturation. How these factors integrate into a Gene Regulatory Network (GRN) controlling these differentiation transitions is incompletely understood. We adopted a genome-wide whole tissue approach to establish a Growth Plate Differential Gene Expression Library (GP-DGEL) for fractionated proliferating, pre-hypertrophic, early and late hypertrophic chondrocytes, as an overarching resource for discovery of pathways and disease candidates. De novo motif discovery revealed the enrichment of SOX9 and GLI binding sites in the genes preferentially expressed in proliferating and prehypertrophic chondrocytes, suggesting the potential cooperation between SOX9 and GLI proteins. We integrated the analyses of the transcriptome, SOX9, GLI1 and GLI3 ChIP-seq datasets, with functional validation by transactivation assays and mouse mutants. We identified new SOX9 targets and showed SOX9-GLI directly and cooperatively regulate many genes such as Trps1, Sox9, Sox5, Sox6, Col2a1, Ptch1, Gli1 and Gli2. Further, FOXA2 competes with SOX9 for the transactivation of target genes. The data support a model of SOX9-GLI-FOXA phasic GRN in chondrocyte development. Together, SOX9-GLI auto-regulate and cooperate to activate and repress genes in proliferating chondrocytes. Upon hypertrophy, FOXA competes with SOX9, and control toward terminal differentiation passes to FOXA, RUNX, AP1 and MEF2 factors. In the development of the mammalian growth plate, while several transcription factors are individually well known for their key roles in regulating phases of chondrocyte differentiation, there is little information on how they interact and cooperate with each other. We took an unbiased genome wide approach to identify the transcription factors and signaling pathways that play dominant roles in the chondrocyte differentiation cascade. We developed a searchable library of differentially expressed genes, GP-DGEL, which has fine spatial resolution and global transcriptomic coverage for discovery of processes, pathways and disease candidates. Our work identifies a novel regulatory mechanism that integrates the action of three transcription factors, SOX9, GLI and FOXA. SOX9-GLI auto-regulate and cooperate to activate and repress genes in proliferating chondrocytes. Upon entry into prehypertrophy, FOXA competes with SOX9, and control of hypertrophy passes to FOXA, RUNX, AP1 and MEF2 factors.
Collapse
Affiliation(s)
- Zhijia Tan
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Ben Niu
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Kwok Yeung Tsang
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Ian G. Melhado
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Shinsuke Ohba
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Xinjun He
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Yongheng Huang
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Cheng Wang
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Andrew P. McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad-CIRM Center for Regenerative Medicine and Stem Cell Research, W.M. Keck School of Medicine of the University of Southern California, Los Angeles, California, United States of America
| | - Ralf Jauch
- Genome Regulation Laboratory, Guangzhou Institutes of Biomedicine and Health, Guangzhou, China
| | - Danny Chan
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
| | - Michael Q. Zhang
- Department of Biological Sciences, Center for Systems Biology, The University of Texas at Dallas, Dallas, Texas, United States of America
- MOE Key Laboratory of Bioinformatics, Center for Synthetic and Systems Biology, TNLIST, Tsinghua University, Beijing, China
| | - Kathryn S. E. Cheah
- School of Biomedical Sciences, LKS Faculty of Medicine, the University of Hong Kong, Pokfulam, Hong Kong
- * E-mail:
| |
Collapse
|
189
|
Zuo C, Wang L, Kamalesh RM, Bowen ME, Moore DC, Dooner MS, Reginato AM, Wu Q, Schorl C, Song Y, Warman ML, Neel BG, Ehrlich MG, Yang W. SHP2 regulates skeletal cell fate by modifying SOX9 expression and transcriptional activity. Bone Res 2018; 6:12. [PMID: 29644115 PMCID: PMC5886981 DOI: 10.1038/s41413-018-0013-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2017] [Revised: 01/15/2018] [Accepted: 02/28/2018] [Indexed: 02/05/2023] Open
Abstract
Chondrocytes and osteoblasts differentiate from a common mesenchymal precursor, the osteochondroprogenitor (OCP), and help build the vertebrate skeleton. The signaling pathways that control lineage commitment for OCPs are incompletely understood. We asked whether the ubiquitously expressed protein-tyrosine phosphatase SHP2 (encoded by Ptpn11) affects skeletal lineage commitment by conditionally deleting Ptpn11 in mouse limb and head mesenchyme using "Cre-loxP"-mediated gene excision. SHP2-deficient mice have increased cartilage mass and deficient ossification, suggesting that SHP2-deficient OCPs become chondrocytes and not osteoblasts. Consistent with these observations, the expression of the master chondrogenic transcription factor SOX9 and its target genes Acan, Col2a1, and Col10a1 were increased in SHP2-deficient chondrocytes, as revealed by gene expression arrays, qRT-PCR, in situ hybridization, and immunostaining. Mechanistic studies demonstrate that SHP2 regulates OCP fate determination via the phosphorylation and SUMOylation of SOX9, mediated at least in part via the PKA signaling pathway. Our data indicate that SHP2 is critical for skeletal cell lineage differentiation and could thus be a pharmacologic target for bone and cartilage regeneration.
Collapse
Affiliation(s)
- Chunlin Zuo
- 1Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA.,9Present Address: Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022 China
| | - Lijun Wang
- 1Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| | - Raghavendra M Kamalesh
- 1Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| | - Margot E Bowen
- 2Orthopaedic Research Laboratories and Howard Hughes Medical Institute, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA 02115 USA
| | - Douglas C Moore
- 1Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| | - Mark S Dooner
- 3Division of Hematology and Oncology, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| | - Anthony M Reginato
- 4Division of Rheumatology, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| | - Qian Wu
- 5Department of Pathology and Laboratory Medicine, University of Connecticut Health Center, Farmington, CT 06030 USA
| | - Christoph Schorl
- 6Department of Molecular and Cell Biology and Biochemistry, Brown University, 70 Ship Street, Providence, RI 02912 USA
| | - Yueming Song
- 7Department of Orthopedic Surgery, West China Hospital of Sichuan University, Chengdu, 610041 China
| | - Matthew L Warman
- 2Orthopaedic Research Laboratories and Howard Hughes Medical Institute, Boston Children's Hospital and Department of Genetics, Harvard Medical School, Boston, MA 02115 USA
| | - Benjamin G Neel
- 8Laura and Issac Perlmutter Cancer Center, NYU Langone Medical Center, New York, NY 10016 USA
| | - Michael G Ehrlich
- 1Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| | - Wentian Yang
- 1Department of Orthopaedics, Brown University Alpert Medical School and Rhode Island Hospital, Providence, RI 02903 USA
| |
Collapse
|
190
|
Zayed M, Adair S, Ursini T, Schumacher J, Misk N, Dhar M. Concepts and challenges in the use of mesenchymal stem cells as a treatment for cartilage damage in the horse. Res Vet Sci 2018; 118:317-323. [PMID: 29601969 DOI: 10.1016/j.rvsc.2018.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/13/2018] [Accepted: 03/18/2018] [Indexed: 12/26/2022]
Abstract
Osteoarthritis (OA), the most common form of joint disease affecting humans and horses, is characterized by the advance and decline of cartilage and loss of function of the affected joint. The progression of OA is steadily accompanied with biochemical events, which interfere with the cytokines and proteolytic enzymes responsible for progress of the disease. Recently, regenerative therapies have been used with an assumption that mesenchymal stem cells (MSCs) possess the potential to prevent the advancement of cartilage damage and potentially regenerate the injured tissue with an ultimate goal of preventing OA. We believe that despite various challenges, the use of allogenic versus autologous MSCs in cartilage regeneration, is a major issue which can directly or indirectly affect the other factors including, the timing of implantation, dose or cell numbers for implantation, and the source of MSCs. Current knowledge reporting some of these challenges that the clinicians might face in the treatment of cartilage damage in horses are presented. In this regard we conducted two independent studies. In the first study we compared donor matched bone marrow and synovial fluid - derived equine MSCs in vitro, and showed that the SFMSCs were similar to the BMMSCs in their proliferation, expression of CD29, CD44 and CD90, but, exhibited a significantly different chondrogenesis. Additionally, 3.2-21% of all SFMSCs were positive for MHC II, whereas, BMMSCs were negative. In the second study we observed that injection of both the autologous and allogenic SFMSCs into the tarsocrural joint resulted in elevated levels of total protein and total nucleated cell counts. Further experiments to evaluate the in vivo acute or chronic response to allogenic or autologous MSCs are imperative.
Collapse
Affiliation(s)
- Mohammed Zayed
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - Steve Adair
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - Tena Ursini
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - James Schumacher
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA
| | - Nabil Misk
- Department of Animal Surgery, College of Veterinary Medicine, Assuit University, 71526 Assuit, Egypt
| | - Madhu Dhar
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, University of Tennessee, Knoxville, TN 37996, USA.
| |
Collapse
|
191
|
Roberto VP, Gavaia P, Nunes MJ, Rodrigues E, Cancela ML, Tiago DM. Evidences for a New Role of miR-214 in Chondrogenesis. Sci Rep 2018; 8:3704. [PMID: 29487295 PMCID: PMC5829070 DOI: 10.1038/s41598-018-21735-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 02/08/2018] [Indexed: 12/27/2022] Open
Abstract
miR-214 is known to play a role in mammalian skeletal development through inhibition of osteogenesis and stimulation of osteoclastogenesis, but data regarding other vertebrates, as well as a possible role in chondrogenesis, remain unknown. Here, we show that miR-214 expression is detected in bone and cartilage of zebrafish skeleton, and is downregulated during murine ATDC5 chondrocyte differentiation. Additionally, we observed a conservation of the transcriptional regulation of miR-214 primary transcript Dnm3os in vertebrates, being regulated by Ets1 in ATDC5 chondrogenic cells. Moreover, overexpression of miR-214 in vitro and in vivo mitigated chondrocyte differentiation probably by targeting activating transcription factor 4 (Atf4). Indeed, miR-214 overexpression in vivo hampered cranial cartilage formation of zebrafish and coincided with downregulation of atf4 and of the key chondrogenic players sox9 and col2a1. We show that miR-214 overexpression exerts a negative role in chondrogenesis by impacting on chondrocyte differentiation possibly through conserved mechanisms.
Collapse
Affiliation(s)
- Vânia Palma Roberto
- Centre of Marine Sciences (CCMAR/CIMAR-LA), University of Algarve, 8005-139, Faro, Portugal.,PhD Program in Biomedical Sciences, DCBM, University of Algarve, 8005-139, Faro, Portugal.,Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal.,Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
| | - Paulo Gavaia
- Centre of Marine Sciences (CCMAR/CIMAR-LA), University of Algarve, 8005-139, Faro, Portugal.,Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal
| | - Maria João Nunes
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Elsa Rodrigues
- Instituto de Investigação do Medicamento (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal.,Department of Biochemistry and Human Biology, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisbon, Portugal
| | - Maria Leonor Cancela
- Centre of Marine Sciences (CCMAR/CIMAR-LA), University of Algarve, 8005-139, Faro, Portugal. .,Algarve Biomedical Center, University of Algarve, Campus de Gambelas, 8005-139, Faro, Portugal. .,Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139, Faro, Portugal.
| | - Daniel Martins Tiago
- Centre of Marine Sciences (CCMAR/CIMAR-LA), University of Algarve, 8005-139, Faro, Portugal.
| |
Collapse
|
192
|
Li IMH, Liu K, Neal A, Clegg PD, De Val S, Bou-Gharios G. Differential tissue specific, temporal and spatial expression patterns of the Aggrecan gene is modulated by independent enhancer elements. Sci Rep 2018; 8:950. [PMID: 29343853 PMCID: PMC5772622 DOI: 10.1038/s41598-018-19186-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023] Open
Abstract
The transcriptional mechanism through which chondrocytes control the spatial and temporal composition of the cartilage tissue has remained largely elusive. The central aim of this study was to identify whether transcriptional enhancers played a role in the organisation of the chondrocytes in cartilaginous tissue. We focused on the Aggrecan gene (Acan) as it is essential for the normal structure and function of cartilage and it is expressed developmentally in different stages of chondrocyte maturation. Using transgenic reporter studies in mice we identified four elements, two of which showed individual chondrocyte developmental stage specificity. In particular, one enhancer (-80) distinguishes itself from the others by being predominantly active in adult cartilage. Furthermore, the -62 element uniquely drove reporter activity in early chondrocytes. The remaining chondrocyte specific enhancers, +28 and -30, showed no preference to chondrocyte type. The transcription factor SOX9 interacted with all the enhancers in vitro and mutation of SOX9 binding sites in one of the enhancers (-30) resulted in a loss of its chondrocyte specificity and ectopic enhancer reporter activity. Thus, the Acan enhancers orchestrate the precise spatiotemporal expression of this gene in cartilage types at different stages of development and adulthood.
Collapse
Affiliation(s)
- Ian M H Li
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Ke Liu
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Alice Neal
- Ludwig Cancer Research Ltd, University of Oxford, Oxford, UK
| | - Peter D Clegg
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK
| | - Sarah De Val
- Ludwig Cancer Research Ltd, University of Oxford, Oxford, UK
| | - George Bou-Gharios
- Institute of Ageing and Chronic Disease, University of Liverpool, William Henry Duncan Building, Liverpool, L7 8TX, UK.
| |
Collapse
|
193
|
Nishimura R, Hata K, Nakamura E, Murakami T, Takahata Y. Transcriptional network systems in cartilage development and disease. Histochem Cell Biol 2018; 149:353-363. [PMID: 29308531 DOI: 10.1007/s00418-017-1628-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/22/2017] [Indexed: 12/13/2022]
Abstract
Transcription factors play important roles in the regulation of cartilage development by controlling the expression of chondrogenic genes. Genetic studies have revealed that Sox9/Sox5/Sox6, Runx2/Runx3 and Osterix in particular are essential for the sequential steps of cartilage development. Importantly, these transcription factors form network systems that are also required for appropriate cartilage development. Molecular cloning approaches have largely contributed to the identification of several transcriptional partners for Sox9 and Runx2 during cartilage development. Although the importance of a negative-feedback loop between Indian hedgehog (Ihh) and parathyroid hormone-related protein (PTHrP) in chondrocyte hypertrophy has been well established, recent studies indicate that several transcription factors interact with the Ihh-PTHrP loop and demonstrated that Ihh has multiple functions in the regulation of cartilage development. The most common cartilage disorder, osteoarthritis, has been reported to result from the pathological action of several transcription factors, including Runx2, C/EBPβ and HIF-2α. On the other hand, NFAT family members appear to play roles in the protection of cartilage from osteoarthritis. It is also becoming important to understand the homeostasis and regulation of articular chondrocytes, because they have different cellular and molecular features from chondrocytes of the growth plate. This review summarizes the regulation and roles of transcriptional network systems in cartilage development and their pathological roles in osteoarthritis.
Collapse
Affiliation(s)
- Riko Nishimura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Kenji Hata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Eriko Nakamura
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomohiko Murakami
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yoshifumi Takahata
- Department of Molecular and Cellular Biochemistry, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
194
|
Yao B, Zhang M, Liu M, Wang Q, Liu M, Zhao Y. Sox9 Functions as a Master Regulator of Antler Growth by Controlling Multiple Cell Lineages. DNA Cell Biol 2018; 37:15-22. [DOI: 10.1089/dna.2017.3885] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Baojin Yao
- Chinese Medicine and Bioengineering Research and Development Center, Changchun University of Chinese Medicine, Changchun, China
| | - Mei Zhang
- Innovation Practice Center, Changchun University of Chinese Medicine, Changchun, China
| | - Meichen Liu
- Chinese Medicine and Bioengineering Research and Development Center, Changchun University of Chinese Medicine, Changchun, China
| | - Qun Wang
- Chinese Medicine and Bioengineering Research and Development Center, Changchun University of Chinese Medicine, Changchun, China
| | - Meixin Liu
- Chinese Medicine and Bioengineering Research and Development Center, Changchun University of Chinese Medicine, Changchun, China
| | - Yu Zhao
- Chinese Medicine and Bioengineering Research and Development Center, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
195
|
Mendes LF, Tam WL, Chai YC, Geris L, Luyten FP, Roberts SJ. Combinatorial Analysis of Growth Factors Reveals the Contribution of Bone Morphogenetic Proteins to Chondrogenic Differentiation of Human Periosteal Cells. Tissue Eng Part C Methods 2017; 22:473-86. [PMID: 27018617 DOI: 10.1089/ten.tec.2015.0436] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Successful application of cell-based strategies in cartilage and bone tissue engineering has been hampered by the lack of robust protocols to efficiently differentiate mesenchymal stem cells into the chondrogenic lineage. The development of chemically defined culture media supplemented with growth factors (GFs) has been proposed as a way to overcome this limitation. In this work, we applied a fractional design of experiment (DoE) strategy to screen the effect of multiple GFs (BMP2, BMP6, GDF5, TGF-β1, and FGF2) on chondrogenic differentiation of human periosteum-derived mesenchymal stem cells (hPDCs) in vitro. In a micromass culture (μMass) system, BMP2 had a positive effect on glycosaminoglycan deposition at day 7 (p < 0.001), which in combination with BMP6 synergistically enhanced cartilage-like tissue formation that displayed in vitro mineralization capacity at day 14 (p < 0.001). Gene expression of μMasses cultured for 7 days with a medium formulation supplemented with 100 ng/mL of BMP2 and BMP6 and a low concentration of GDF5, TGF-β1, and FGF2 showed increased expression of Sox9 (1.7-fold) and the matrix molecules aggrecan (7-fold increase) and COL2A1 (40-fold increase) compared to nonstimulated control μMasses. The DoE analysis indicated that in GF combinations, BMP2 was the strongest effector for chondrogenic differentiation of hPDCs. When transplanted ectopically in nude mice, the in vitro-differentiated μMasses showed maintenance of the cartilaginous phenotype after 4 weeks in vivo. This study indicates the power of using the DoE approach for the creation of new medium formulations for skeletal tissue engineering approaches.
Collapse
Affiliation(s)
- Luis Filipe Mendes
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Wai Long Tam
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Yoke Chin Chai
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Liesbet Geris
- 2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium .,3 Biomechanics Research Unit, University of Liege , Liege, Belgium .,4 Department of Mechanical Engineering, Biomechanics Section, Katholieke Universiteit Leuven, Heverlee, Belgium
| | - Frank P Luyten
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium
| | - Scott J Roberts
- 1 Tissue Engineering Laboratory, Skeletal Biology and Engineering Research Center , Katholieke Universiteit Leuven, Leuven, Belgium .,2 Prometheus, Division of Skeletal Tissue Engineering, Katholieke Universiteit Leuven , Leuven, Belgium .,5 Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London , The Royal National Orthopaedic Hospital, London, United Kingdom
| |
Collapse
|
196
|
Li Y, Wang J, Song X, Bai H, Ma T, Zhang Z, Li X, Jiang R, Wang G, Fan X, Liu X, Gao L. Effects of baicalein on IL-1β-induced inflammation and apoptosis in rat articular chondrocytes. Oncotarget 2017; 8:90781-90795. [PMID: 29207603 PMCID: PMC5710884 DOI: 10.18632/oncotarget.21796] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 09/21/2017] [Indexed: 01/15/2023] Open
Abstract
In osteoarthritis (OA), activated synoviocytes and articular chondrocytes produce pro-inflammatory cytokines, such as IL-1β, that promote chondrocyte apoptosis and activate the NF-κB signaling pathway to induce catabolic factors. In this study, we examined the anti-inflammatory and anti-apoptotic effect of baicalein on IL-1β signaling and NF-κB-regulated gene products in rat chondrocytes. Rat chondrocytes were pretreated with 10 ng/ml IL-1β for 24 h and then co-treated with 10 ng/ml IL-1β and 50 μM baicalein for 0, 12, 24, 36 and 48h. The expression levels of poly(ADP-ribose) polymerase (PARP), Bcl-2, caspase-3, matrix metalloproteinase (MMP)-9, MMP-3, cyclooxygenase (COX)-2 and SOX-9 were detected by Western blot and quantitative reverse transcription-PCR (qPCR). The effects of baicalein on the translocation and phosphorylation of the NF-κB system were studied by Western blotting and immunofluorescence. Baicalein stimulated the expression of anti-apoptotic genes and reduced the pro-apoptotic and pro-inflammatory gene products in chondrocytes. Baicalein promoted SOX-9 expression in a time-dependent manner in chondrocytes. Baicalein inhibited the NF-κB activation that was induced by IL-1β in a time-dependent manner in chondrocytes. Our results suggest that the anti-inflammatory and anti-apoptotic effects of baicalein are mediated through the inhibition of the translocation of phosphorylated p65 to the nucleus.
Collapse
Affiliation(s)
- Yue Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Jinglu Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaopeng Song
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Hui Bai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Tianwen Ma
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Zhiheng Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Xinran Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Renli Jiang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Guanying Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Xiaojing Fan
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Xu Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| | - Li Gao
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, People's Republic of China
| |
Collapse
|
197
|
Wang L, Huang J, Moore DC, Zuo C, Wu Q, Xie L, von der Mark K, Yuan X, Chen D, Warman ML, Ehrlich MG, Yang W. SHP2 Regulates the Osteogenic Fate of Growth Plate Hypertrophic Chondrocytes. Sci Rep 2017; 7:12699. [PMID: 28983104 PMCID: PMC5629218 DOI: 10.1038/s41598-017-12767-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 09/14/2017] [Indexed: 02/07/2023] Open
Abstract
Transdifferentiation of hypertrophic chondrocytes into bone-forming osteoblasts has been reported, yet the underlying molecular mechanism remains incompletely understood. SHP2 is an ubiquitously expressed cytoplasmic protein tyrosine phosphatase. SHP2 loss-of-function mutations in chondroid cells are linked to metachondromatosis in humans and mice, suggesting a crucial role for SHP2 in the skeleton. However, the specific role of SHP2 in skeletal cells has not been elucidated. To approach this question, we ablated SHP2 in collagen 2α1(Col2α1)-Cre- and collagen 10α1(Col10α1)-Cre-expressing cells, predominantly proliferating and hypertrophic chondrocytes, using "Cre-loxP"-mediated gene excision. Mice lacking SHP2 in Col2α1-Cre-expressing cells die at mid-gestation. Postnatal SHP2 ablation in the same cell population caused dwarfism, chondrodysplasia and exostoses. In contrast, mice in which SHP2 was ablated in the Col10α1-Cre-expressing cells appeared normal but were osteopenic. Further mechanistic studies revealed that SHP2 exerted its influence partly by regulating the abundance of SOX9 in chondrocytes. Elevated and sustained SOX9 in SHP2-deficient hypertrophic chondrocytes impaired their differentiation to osteoblasts and impaired endochondral ossification. Our study uncovered an important role of SHP2 in bone development and cartilage homeostasis by influencing the osteogenic differentiation of hypertrophic chondrocytes and provided insight into the pathogenesis and potential treatment of skeletal diseases, such as osteopenia and osteoporosis.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Jiahui Huang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Douglas C Moore
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Chunlin Zuo
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
- Department of Endocrinology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, P.R. China
| | - Qian Wu
- Department of Pathology and Laboratory Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Liqin Xie
- Regeneron Pharmaceuticals, Tarrytown, NY, 10591, USA
| | - Klaus von der Mark
- Department of Experimental Medicine, University of Erlangen-Nürnberg, Gluckstrasse 6, 91054, Erlangen, Germany
| | - Xin Yuan
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, 02115, USA
| | - Di Chen
- Department of Biochemistry, Rush University, 600 S. Paulina St., Chicago, IL, 60612, USA
| | - Matthew L Warman
- Orthopaedic Research Laboratories and Howard Hughes Medical Institute, Boston Children's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Michael G Ehrlich
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA
| | - Wentian Yang
- Department of Orthopaedic Surgery, Brown University Alpert Medical School, Providence, RI, 02903, USA.
| |
Collapse
|
198
|
COUP-TFII is required for morphogenesis of the neural crest-derived tympanic ring. Sci Rep 2017; 7:12386. [PMID: 28959031 PMCID: PMC5620064 DOI: 10.1038/s41598-017-12665-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 09/18/2017] [Indexed: 12/27/2022] Open
Abstract
Chicken Ovalbumin Upstream Promoter-Transcription Factor II (COUP-TFII) plays pivotal roles in cell growth, cell differentiation, and cell fate determination. Although genome-wide studies have identified COUP-TFII binding on gene sets mainly involved in neural crest cell (NCC) development and craniofacial morphogenesis, the direct functional connection between COUP-TFII and NCCs in vivo has not been well characterized. In this study, we show that COUP-TFII is expressed in the subpopulation of NCCs and its derivatives, and targeted ablation of COUP-TFII in mouse NCCs results in markedly shortened and bifurcated tympanic rings, which in turn disturb the caudal direction of external acoustic meatus invagination. However, formation of the manubrium of the malleus (MM) in Wnt1-Cre/+;COUP-TFIIflox/flox mice is not perturbed, suggesting that the rostral half of the tympanic ring is sufficient to support proper MM development. Interestingly, we found that loss of COUP-TFII up-regulates Sox9 in the tympanic ring primordium and affects the distribution of preosteoblasts before mesenchymal condensation. Together, our results demonstrate that COUP-TFII plays an essential role in regulating the patterning of the NCC-derived tympanic ring.
Collapse
|
199
|
Hu DP, Ferro F, Yang F, Taylor AJ, Chang W, Miclau T, Marcucio RS, Bahney CS. Cartilage to bone transformation during fracture healing is coordinated by the invading vasculature and induction of the core pluripotency genes. Development 2017; 144:221-234. [PMID: 28096214 DOI: 10.1242/dev.130807] [Citation(s) in RCA: 163] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 11/29/2016] [Indexed: 01/01/2023]
Abstract
Fractures heal predominantly through the process of endochondral ossification. The classic model of endochondral ossification holds that chondrocytes mature to hypertrophy, undergo apoptosis and new bone forms by invading osteoprogenitors. However, recent data demonstrate that chondrocytes transdifferentiate to osteoblasts in the growth plate and during regeneration, yet the mechanism(s) regulating this process remain unknown. Here, we show a spatially-dependent phenotypic overlap between hypertrophic chondrocytes and osteoblasts at the chondro-osseous border in the fracture callus, in a region we define as the transition zone (TZ). Hypertrophic chondrocytes in the TZ activate expression of the pluripotency factors [Sox2, Oct4 (Pou5f1), Nanog], and conditional knock-out of Sox2 during fracture healing results in reduction of the fracture callus and a delay in conversion of cartilage to bone. The signal(s) triggering expression of the pluripotency genes are unknown, but we demonstrate that endothelial cell conditioned medium upregulates these genes in ex vivo fracture cultures, supporting histological evidence that transdifferentiation occurs adjacent to the vasculature. Elucidating the cellular and molecular mechanisms underlying fracture repair is important for understanding why some fractures fail to heal and for developing novel therapeutic interventions.
Collapse
Affiliation(s)
- Diane P Hu
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| | - Federico Ferro
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| | - Frank Yang
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| | - Aaron J Taylor
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| | - Wenhan Chang
- University of California, San Francisco (UCSF) & San Francisco Veterans Affairs Medical Center (VAMC), Department of Medicine, 1700 Owens Street, 4th Floor, San Francisco, CA 94158, USA
| | - Theodore Miclau
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| | - Ralph S Marcucio
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| | - Chelsea S Bahney
- University of California, San Francisco (UCSF) & San Francisco General Hospital (SFGH), Department of Orthopaedic Surgery, Orthopaedic Trauma Institute, 2550 23rd Street, Building 9, 3rd Floor, San Francisco, CA 94110, USA
| |
Collapse
|
200
|
Ontogenic Identification and Analysis of Mesenchymal Stromal Cell Populations during Mouse Limb and Long Bone Development. Stem Cell Reports 2017; 9:1124-1138. [PMID: 28919259 PMCID: PMC5639212 DOI: 10.1016/j.stemcr.2017.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 08/14/2017] [Accepted: 08/14/2017] [Indexed: 12/21/2022] Open
Abstract
Bone-derived mesenchymal stromal cells (MSCs) differentiate into multiple lineages including chondro- and osteogenic fates and function in establishing the hematopoietic compartment of the bone marrow. Here, we analyze the emergence of different MSC types during mouse limb and long bone development. In particular, PDGFRαposSCA-1pos (PαS) cells and mouse skeletal stem cells (mSSCs) are detected within the PDGFRαposCD51pos (PαCD51) mesenchymal progenitors, which are the most abundant progenitors in early limb buds and developing long bones until birth. Long-bone-derived PαS cells and mSSCs are most prevalent in newborn mice, and molecular analysis shows that they constitute distinct progenitor populations from the earliest stages onward. Differential expression of CD90 and CD73 identifies four PαS subpopulations that display distinct chondro- and osteogenic differentiation potentials. Finally, we show that cartilage constructs generated from CD90pos PαS cells are remodeled into bone organoids encompassing functional endothelial and hematopoietic compartments, which makes these cells suited for bone tissue engineering. Ontogenic profiling of MSC populations during mouse limb and long bone development PαCD51-positive cells are the most prevalent mesenchymal population PαS cells and mSSCs arise as distinct populations within the PαCD51 progenitor pool Cartilage constructs from one PαS subpopulation are remodeled into bone organoids
Collapse
|