51
|
Grafe I, Alexander S, Peterson JR, Snider TN, Levi B, Lee B, Mishina Y. TGF-β Family Signaling in Mesenchymal Differentiation. Cold Spring Harb Perspect Biol 2018; 10:a022202. [PMID: 28507020 PMCID: PMC5932590 DOI: 10.1101/cshperspect.a022202] [Citation(s) in RCA: 186] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) can differentiate into several lineages during development and also contribute to tissue homeostasis and regeneration, although the requirements for both may be distinct. MSC lineage commitment and progression in differentiation are regulated by members of the transforming growth factor-β (TGF-β) family. This review focuses on the roles of TGF-β family signaling in mesenchymal lineage commitment and differentiation into osteoblasts, chondrocytes, myoblasts, adipocytes, and tenocytes. We summarize the reported findings of cell culture studies, animal models, and interactions with other signaling pathways and highlight how aberrations in TGF-β family signaling can drive human disease by affecting mesenchymal differentiation.
Collapse
Affiliation(s)
- Ingo Grafe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Stefanie Alexander
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Jonathan R Peterson
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Taylor Nicholas Snider
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Benjamin Levi
- Department of Surgery, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
| | - Yuji Mishina
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| |
Collapse
|
52
|
van der Kraan PM. Differential Role of Transforming Growth Factor-beta in an Osteoarthritic or a Healthy Joint. J Bone Metab 2018; 25:65-72. [PMID: 29900155 PMCID: PMC5995759 DOI: 10.11005/jbm.2018.25.2.65] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 05/26/2018] [Indexed: 12/24/2022] Open
Abstract
Transforming growth factor-β (TGF-β) is a cytokine that plays an important role in both normal joints and joints affected by osteoarthritis (OA), the most common joint disease. However, the role of this pleiotropic cytokine in a normal healthy joint is very different from its role in an OA joint. In a normal synovial joint, active TGF-β is only present after joint loading and only for a short period. In contrast, permanent and high levels of active TGF-β are detected in OA joints. Due to this difference in levels and exposure period of joint cells to active TGF-β, the function of TGF-β is strikingly different in normal and OA joints. The consequences of this difference in TGF-β levels on joint homeostasis and pathological changes in OA joints are discussed in this review.
Collapse
Affiliation(s)
- Peter M. van der Kraan
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Nijmegen, Netherlands
| |
Collapse
|
53
|
Moore ER, Jacobs CR. The primary cilium as a signaling nexus for growth plate function and subsequent skeletal development. J Orthop Res 2018; 36:533-545. [PMID: 28901584 PMCID: PMC5839937 DOI: 10.1002/jor.23732] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 09/07/2017] [Indexed: 02/04/2023]
Abstract
The primary cilium is a solitary, antenna-like sensory organelle with many important roles in cartilage and bone development, maintenance, and function. The primary cilium's potential role as a signaling nexus in the growth plate makes it an attractive therapeutic target for diseases and disorders associated with bone development and maintenance. Many signaling pathways that are mediated by the cilium-such as Hh, Wnt, Ihh/PTHrP, TGFβ, BMP, FGF, and Notch-are also known to influence endochondral ossification, primarily by directing growth plate formation and chondrocyte behavior. Although a few studies have demonstrated that these signaling pathways can be directly tied to the primary cilium, many pathways have yet to be evaluated in context of the cilium. This review serves to bridge this knowledge gap in the literature, as well as discuss the cilium's importance in the growth plate's ability to sense and respond to chemical and mechanical stimuli. Furthermore, we explore the importance of using the appropriate mechanism to study the cilium in vivo and suggest IFT88 deletion is the best available technique. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 36:533-545, 2018.
Collapse
Affiliation(s)
- Emily R. Moore
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| | - Christopher R. Jacobs
- Department of Biomedical Engineering; Columbia University; 351 Engineering Terrace, Mail Code 8904, 1210 Amsterdam Avenue New York 10027 New York
| |
Collapse
|
54
|
Xu X, Zheng L, Yuan Q, Zhen G, Crane JL, Zhou X, Cao X. Transforming growth factor-β in stem cells and tissue homeostasis. Bone Res 2018; 6:2. [PMID: 29423331 PMCID: PMC5802812 DOI: 10.1038/s41413-017-0005-4] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 11/12/2017] [Accepted: 11/15/2017] [Indexed: 02/05/2023] Open
Abstract
TGF-β 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-β in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-β in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-β is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-β ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-β signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-β is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-β.
Collapse
Affiliation(s)
- Xin Xu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Liwei Zheng
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Gehua Zhen
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Janet L. Crane
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
- Department of Pediatrics, Johns Hopkins University, Baltimore, MD USA
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xu Cao
- Department of Orthopedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
55
|
Wang C, Silverman RM, Shen J, O'Keefe RJ. Distinct metabolic programs induced by TGF-β1 and BMP2 in human articular chondrocytes with osteoarthritis. J Orthop Translat 2018; 12:66-73. [PMID: 29662780 PMCID: PMC5866480 DOI: 10.1016/j.jot.2017.12.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/13/2022] Open
Abstract
Objectives Cellular energy metabolism is important for the function of all tissues, including cartilage. Recent studies indicate that superficial and deep subpopulations of articular chondrocytes (ACs) have distinct metabolic profiles. At the cellular and molecular level, osteoarthritis (OA) is characterised by alteration from a healthy homoeostatic state towards a catabolic state. Several molecular pathways, including transforming growth factor beta (TGF-β) and bone morphogenetic protein (BMP) signalling, have been identified as critical players in the pathogenesis and progression of OA. However, the manner in which these factors influence cellular energy metabolism in ACs is not well understood. This study investigates the effect of TGF-β or BMP signalling on energy metabolism in human articular chondrocytes (hACs). Methods ACs were isolated from residual macroscopically full thickness and intact cartilage from the femoral condyle of human samples obtained from patients with OA. ACs were treated with Vehicle (control), TGF-β1 or BMP2 for 48–72 hours. Metabolic assays were performed to determine glucose consumption, lactate production and adenosine triphosphate (ATP) production, whereas the mitochondrial stress test was performed to determine oxygen consumption rate. Protein was isolated to assess translational activity and was evaluated using Western blot. Results We showed that TGF-β1, known to maintain chondrocyte homoeostasis, stimulated glycolysis by upregulating key glycolytic factors, such as glucose transporter 1 (Glut1) and hexokinase II, while reducing oxidative phosphorylation in hACs. In contrast, BMP2 enhanced mitochondrial metabolism and oxidative phosphorylation and had a minimal effect on key glycolytic regulators. Conclusions Our data revealed distinct metabolic programs induced by TGF-β1 and BMP2 in hACs, suggesting that the regulation of cellular metabolism may represent a new mechanism underlying the pathogenesis of OA. The translational potential of this article The findings define the regulation of energy metabolism as a potential novel therapeutic approach for the treatment of OA.
Collapse
Affiliation(s)
- Cuicui Wang
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | | | - Jie Shen
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
| | - Regis J. O'Keefe
- Department of Orthopaedic Surgery, School of Medicine, Washington University, St. Louis, MO 63110, USA
- Corresponding author. Department of Orthopaedic Surgery, Washington University in St. Louis, 660 S. Euclid, CB 8233, St. Louis, MO 63110, USA.Department of Orthopaedic SurgeryWashington University in St. Louis660 S. EuclidCB 8233St. LouisMO63110USA
| |
Collapse
|
56
|
Armiento AR, Stoddart MJ, Alini M, Eglin D. Biomaterials for articular cartilage tissue engineering: Learning from biology. Acta Biomater 2018; 65:1-20. [PMID: 29128537 DOI: 10.1016/j.actbio.2017.11.021] [Citation(s) in RCA: 390] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 10/05/2017] [Accepted: 11/07/2017] [Indexed: 12/27/2022]
Abstract
Articular cartilage is commonly described as a tissue that is made of up to 80% water, is devoid of blood vessels, nerves, and lymphatics, and is populated by only one cell type, the chondrocyte. At first glance, an easy tissue for clinicians to repair and for scientists to reproduce in a laboratory. Yet, chondral and osteochondral defects currently remain an open challenge in orthopedics and tissue engineering of the musculoskeletal system, without considering osteoarthritis. Why do we fail in repairing and regenerating articular cartilage? Behind its simple and homogenous appearance, articular cartilage hides a heterogeneous composition, a high level of organisation and specific biomechanical properties that, taken together, make articular cartilage a unique material that we are not yet able to repair or reproduce with high fidelity. This review highlights the available therapies for cartilage repair and retraces the research on different biomaterials developed for tissue engineering strategies. Their potential to recreate the structure, including composition and organisation, as well as the function of articular cartilage, intended as cell microenvironment and mechanically competent replacement, is described. A perspective of the limitations of the current research is given in the light of the emerging technologies supporting tissue engineering of articular cartilage. STATEMENT OF SIGNIFICANCE The mechanical properties of articular tissue reflect its functionally organised composition and the recreation of its structure challenges the success of in vitro and in vivo reproduction of the native cartilage. Tissue engineering and biomaterials science have revolutionised the way scientists approach the challenge of articular cartilage repair and regeneration by introducing the concept of the interdisciplinary approach. The clinical translation of the current approaches are not yet fully successful, but promising results are expected from the emerging and developing new generation technologies.
Collapse
Affiliation(s)
- A R Armiento
- AO Research Institute Davos, Davos Platz, Switzerland.
| | - M J Stoddart
- AO Research Institute Davos, Davos Platz, Switzerland; University Medical Center, Albert-Ludwigs University, Freiburg, Germany.
| | - M Alini
- AO Research Institute Davos, Davos Platz, Switzerland.
| | - D Eglin
- AO Research Institute Davos, Davos Platz, Switzerland.
| |
Collapse
|
57
|
Tabe S, Hikiji H, Ariyoshi W, Hashidate-Yoshida T, Shindou H, Shimizu T, Okinaga T, Seta Y, Tominaga K, Nishihara T. Lysophosphatidylcholine acyltransferase 4 is involved in chondrogenic differentiation of ATDC5 cells. Sci Rep 2017; 7:16701. [PMID: 29196633 PMCID: PMC5711957 DOI: 10.1038/s41598-017-16902-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 11/20/2017] [Indexed: 12/14/2022] Open
Abstract
Glycerophospholipids have important structural and functional roles in cells and are the main components of cellular membranes. Glycerophospholipids are formed via the de novo pathway (Kennedy pathway) and are subsequently matured in the remodeling pathway (Lands’ cycle). Lands’ cycle consists of two steps: deacylation of phospholipids by phospholipases A2 and reacylation of lysophospholipids by lysophospholipid acyltransferases (LPLATs). LPLATs play key roles in the maturation and maintenance of the fatty acid composition of biomembranes, and cell differentiation. We examined whether LPLATs are involved in chondrogenic differentiation of ATDC5 cells, which can differentiate into chondrocytes. Lysophosphatidylcholine acyltransferase 4 (LPCAT4) mRNA expression and LPCAT enzymatic activity towards 18:1-, 18:2-, 20:4-, and 22:6-CoA increased in the late stage of chondrogenic differentiation, when mineralization occurred. LPCAT4 knockdown decreased mRNA and protein levels of chondrogenic markers as well as Alcian blue staining intensity and alkaline phosphatase activity in ATDC5 cells. These results suggest that LPCAT4 plays important roles during the transition of chondrocytes into hypertrophic chondrocytes and/or a mineralized phenotype.
Collapse
Affiliation(s)
- Shirou Tabe
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan.,Division of Oral and Maxillofacial Surgery, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Hisako Hikiji
- School of Oral Health Sciences, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan.
| | - Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Tomomi Hashidate-Yoshida
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, 162-8655, Japan
| | - Hideo Shindou
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, 162-8655, Japan.,Agency for Medical Research and Development-Core Research for Evolutional Medical Science and Technology (AMED-CREST), AMED, Chiyoda-ku, Tokyo, 100-0004, Japan
| | - Takao Shimizu
- Department of Lipid Signaling, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, 162-8655, Japan.,Department of Lipidomics, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, 113-0033, Japan
| | - Toshinori Okinaga
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Yuji Seta
- Division of Anatomy, Department of Health Improvement, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Kazuhiro Tominaga
- Division of Oral and Maxillofacial Surgery, Department of Science of Physical Functions, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| | - Tatsuji Nishihara
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Kitakyushu, Fukuoka, 803-8580, Japan
| |
Collapse
|
58
|
Li Y, Zou N, Wang J, Wang KW, Li FY, Chen FX, Sun BY, Sun DJ. TGF-β1/Smad3 Signaling Pathway Mediates T-2 Toxin-Induced Decrease of Type II Collagen in Cultured Rat Chondrocytes. Toxins (Basel) 2017; 9:toxins9110359. [PMID: 29113082 PMCID: PMC5705974 DOI: 10.3390/toxins9110359] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 08/08/2017] [Accepted: 11/02/2017] [Indexed: 11/16/2022] Open
Abstract
T-2 toxin can cause damage to the articular cartilage, but the molecular mechanism remains unclear. By employing the culture of rat chondrocytes, we investigated the effect of the TGF-β1/Smad3 signaling pathway on the damage to chondrocytes induced by T-2 toxin. It was found that T-2 toxin could reduce cell viability and increased the number of apoptotic cells when compared with the control group. After the addition of the T-2 toxin, the production of type II collagen was reduced at mRNA and protein levels, while the levels of TGF-β1, Smad3, ALK5, and MMP13 were upregulated. The production of the P-Smad3 protein was also increased. Inhibitors of TGF-β1 and Smad3 were able to reverse the effect of the T-2 toxin on the protein level of above-mentioned signaling molecules. The T-2 toxin could promote the level of MMP13 via the stimulation of TGF-β1 signaling in chondrocytes, resulting in the downregulation of type II collagen and chondrocyte damage. Smad3 may be involved in the degradation of type II collagen, but the Smad3 has no connection with the regulation of MMP13 level. This study provides a new clue to elucidate the mechanism of T-2 toxin-induced chondrocyte damage.
Collapse
Affiliation(s)
- Yang Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| | - Ning Zou
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| | - Jing Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| | - Ke-Wei Wang
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
- China and Russia Medical Research Center, National Health and Family Planning Commission of the People's Republic of China, Harbin Medical University, Harbin 150081, China.
| | - Fu-Yuan Li
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| | - Fu-Xun Chen
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| | - Bing-Yu Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| | - Dian-Jun Sun
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
59
|
Marcucio RS, Qin L, Alsberg E, Boerckel JD. Reverse engineering development: Crosstalk opportunities between developmental biology and tissue engineering. J Orthop Res 2017; 35:2356-2368. [PMID: 28660712 DOI: 10.1002/jor.23636] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
The fields of developmental biology and tissue engineering have been revolutionized in recent years by technological advancements, expanded understanding, and biomaterials design, leading to the emerging paradigm of "developmental" or "biomimetic" tissue engineering. While developmental biology and tissue engineering have long overlapping histories, the fields have largely diverged in recent years at the same time that crosstalk opportunities for mutual benefit are more salient than ever. In this perspective article, we will use musculoskeletal development and tissue engineering as a platform on which to discuss these emerging crosstalk opportunities and will present our opinions on the bright future of these overlapping spheres of influence. The multicellular programs that control musculoskeletal development are rapidly becoming clarified, represented by shifting paradigms in our understanding of cellular function, identity, and lineage specification during development. Simultaneously, advancements in bioartificial matrices that replicate the biochemical, microstructural, and mechanical properties of developing tissues present new tools and approaches for recapitulating development in tissue engineering. Here, we introduce concepts and experimental approaches in musculoskeletal developmental biology and biomaterials design and discuss applications in tissue engineering as well as opportunities for tissue engineering approaches to inform our understanding of fundamental biology. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2356-2368, 2017.
Collapse
Affiliation(s)
- Ralph S Marcucio
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, California
| | - Ling Qin
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania
| | - Eben Alsberg
- Departments of Biomedical Engineering and Orthopaedic Surgery, Case Western Reserve University, Cleveland, Ohio
| | - Joel D Boerckel
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, 36th Street and Hamilton Walk, Philadelphia 19104-6081, Pennsylvania.,Department of Bioengineering, University of Pennslyvania, Philadelphia, Pennsylvania.,Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana
| |
Collapse
|
60
|
Wang Q, Tan Q, Xu W, Qi H, Chen D, Zhou S, Ni Z, Kuang L, Guo J, Huang J, Wang X, Wang Z, Su N, Chen L, Chen B, Jiang W, Gao Y, Chen H, Du X, Xie Y, Chen L. Cartilage-specific deletion of Alk5 gene results in a progressive osteoarthritis-like phenotype in mice. Osteoarthritis Cartilage 2017; 25:1868-1879. [PMID: 28716756 PMCID: PMC5694025 DOI: 10.1016/j.joca.2017.07.010] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 06/23/2017] [Accepted: 07/10/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Previous studies have shown that Transforming growth factor-β (TGF-β)/TGFβRII-Smad3 signaling is involved in articular cartilage homeostasis. However, the role of TGF-β/ALK5 signaling in articular cartilage homeostasis has not been fully defined. In this study, a combination of in vitro and in vivo approaches was used to elucidate the role of ALK5 signaling in articular cartilage homeostasis and the development of osteoarthritis (OA). DESIGN Mice with inducible cartilage-specific deletion of Alk5 were generated to assess the role of ALK5 in OA development. Alterations in cartilage structure were evaluated histologically. The expressions of genes associated with articular cartilage homeostasis and TGF-β signaling were analyzed by qRT-PCR, western blotting and immunohistochemistry. The chondrocyte apoptosis was detected by TUNEL staining and immunohistochemistry. In addition, the molecular mechanism underlying the effects of TGF-β/ALK5 signaling on articular cartilage homeostasis was explored by analyzing the TGF-β/ALK5 signaling-induced expression of proteoglycan 4 (PRG4) using specific inhibitors. RESULTS Postnatal cartilage-specific deletion of Alk5 induced an OA-like phenotype with degradation of articular cartilage, synovial hyperplasia, osteophyte formation, subchondral sclerosis, as well as enhanced chondrocyte apoptosis, overproduction of catabolic factors, and decreased expressions of anabolic factors in chondrocytes. In addition, the expressions of PRG4 mRNA and protein were decreased in Alk5 conditional knockout mice. Furthermore, our results showed, for the first time, that TGF-β/ALK5 signaling regulated PRG4 expression partially through the protein kinase A (PKA)-CREB signaling pathway. CONCLUSIONS TGF-β/ALK5 signaling maintains articular cartilage homeostasis, in part, by upregulating PRG4 expression through the PKA-CREB signaling pathway in articular chondrocytes.
Collapse
Affiliation(s)
- Q. Wang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Q.Y. Tan
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - W. Xu
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - H.B. Qi
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - D. Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL 60612, USA
| | - S. Zhou
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Z.H. Ni
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - L. Kuang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - J.Y. Guo
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - J.L. Huang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - X.X. Wang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Z.Q. Wang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - N. Su
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - L. Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - B. Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - W.L. Jiang
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - Y. Gao
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - H.G. Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | - X.L. Du
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China,Address correspondence and reprint requests to: X.L. Du, Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China. Fax: 86-23-68702991.
| | - Y.L. Xie
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China,Address correspondence and reprint requests to: Y.L. Xie, Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China. Fax: 86-23-68702991.
| | - L. Chen
- Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China,Address correspondence and reprint requests to: L. Chen, Department of Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing 400042, China. Fax: 86-23-68702991.
| |
Collapse
|
61
|
MacFarlane EG, Haupt J, Dietz HC, Shore EM. TGF-β Family Signaling in Connective Tissue and Skeletal Diseases. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022269. [PMID: 28246187 DOI: 10.1101/cshperspect.a022269] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The transforming growth factor β (TGF-β) family of signaling molecules, which includes TGF-βs, activins, inhibins, and numerous bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs), has important functions in all cells and tissues, including soft connective tissues and the skeleton. Specific TGF-β family members play different roles in these tissues, and their activities are often balanced with those of other TGF-β family members and by interactions with other signaling pathways. Perturbations in TGF-β family pathways are associated with numerous human diseases with prominent involvement of the skeletal and cardiovascular systems. This review focuses on the role of this family of signaling molecules in the pathologies of connective tissues that manifest in rare genetic syndromes (e.g., syndromic presentations of thoracic aortic aneurysm), as well as in more common disorders (e.g., osteoarthritis and osteoporosis). Many of these diseases are caused by or result in pathological alterations of the complex relationship between the TGF-β family of signaling mediators and the extracellular matrix in connective tissues.
Collapse
Affiliation(s)
- Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Julia Haupt
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,Howard Hughes Medical Institute, Bethesda, Maryland 21205
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
62
|
Mu W, Xu B, Ma H, Ji B, Zhang Z, Li J, Amat A, Cao L. Halofuginone attenuates articular cartilage degeneration by inhibition of elevated TGF‑β1 signaling in articular cartilage in a rodent osteoarthritis model. Mol Med Rep 2017; 16:7679-7684. [PMID: 28944864 DOI: 10.3892/mmr.2017.7549] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 08/14/2017] [Indexed: 11/05/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative condition of the weight‑bearing joints worldwide without effective medical therapy. In order to investigate whether administration of halofuginone (HF) may attenuate OA, the present study allocated 3‑month‑old male mice into Sham group, vehicle‑treated anterior cruciate ligament transection (ACLT) group and HF‑treated ACLT group. The present study determined that HF treatment reduced the expression of matrix metallopeptidase‑13 and collagen X in articular cartilage. Additionally, it lowered the Osteoarthritis Research Society International‑Modified Mankin score and prevented the loss of articular cartilage from Safranin O and Fast Green staining. HF reduced the progression of osteoarthritis by downregulating abnormally elevated TGF‑β1 activity in articular cartilage. Administration of HF may be a potential preventive therapy for OA.
Collapse
Affiliation(s)
- Wenbo Mu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Boyong Xu
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Hairong Ma
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Baochao Ji
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Zhendong Zhang
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Jiao Li
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Abdusami Amat
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| | - Li Cao
- Department of Orthopaedics, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, P.R. China
| |
Collapse
|
63
|
Hulme CH, Wilson EL, Peffers MJ, Roberts S, Simpson DM, Richardson JB, Gallacher P, Wright KT. Autologous chondrocyte implantation-derived synovial fluids display distinct responder and non-responder proteomic profiles. Arthritis Res Ther 2017; 19:150. [PMID: 28666451 PMCID: PMC5493128 DOI: 10.1186/s13075-017-1336-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/15/2017] [Indexed: 02/07/2023] Open
Abstract
Background Autologous chondrocyte implantation (ACI) can be used in the treatment of focal cartilage injuries to prevent the onset of osteoarthritis (OA). However, we are yet to understand fully why some individuals do not respond well to this intervention. Identification of a reliable and accurate biomarker panel that can predict which patients are likely to respond well to ACI is needed in order to assign the patient to the most appropriate therapy. This study aimed to compare the baseline and mid-treatment proteomic profiles of synovial fluids (SFs) obtained from responders and non-responders to ACI. Methods SFs were derived from 14 ACI responders (mean Lysholm improvement of 33 (17–54)) and 13 non-responders (mean Lysholm decrease of 14 (4–46)) at the two stages of surgery (cartilage harvest and chondrocyte implantation). Label-free proteome profiling of dynamically compressed SFs was used to identify predictive markers of ACI success or failure and to investigate the biological pathways involved in the clinical response to ACI. Results Only 1 protein displayed a ≥2.0-fold differential abundance in the preclinical SF of ACI responders versus non-responders. However, there is a marked difference between these two groups with regard to their proteome shift in response to cartilage harvest, with 24 and 92 proteins showing ≥2.0-fold differential abundance between Stages I and II in responders and non-responders, respectively. Proteomic data has been uploaded to ProteomeXchange (identifier: PXD005220). We have validated two biologically relevant protein changes associated with this response, demonstrating that matrix metalloproteinase 1 was prominently elevated and S100 calcium binding protein A13 was reduced in response to cartilage harvest in non-responders. Conclusions The differential proteomic response to cartilage harvest noted in responders versus non-responders is completely novel. Our analyses suggest several pathways which appear to be altered in non-responders that are worthy of further investigation to elucidate the mechanisms of ACI failure. These protein changes highlight many putative biomarkers that may have potential for prediction of ACI treatment success.
Collapse
Affiliation(s)
- Charlotte H Hulme
- Institute of Science and Technology in Medicine, Keele University, Keele, Staffordshire, UK.,Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Emma L Wilson
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK.,Institute of Medicine, Chester University, Chester, UK
| | - Mandy J Peffers
- Institute of Ageing and Chronic Disease, University of Liverpool, Liverpool, UK
| | - Sally Roberts
- Institute of Science and Technology in Medicine, Keele University, Keele, Staffordshire, UK.,Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Deborah M Simpson
- Centre for Proteome Research, Institute of Integrative Biology, University of Liverpool, Liverpool, UK
| | - James B Richardson
- Institute of Science and Technology in Medicine, Keele University, Keele, Staffordshire, UK.,Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Pete Gallacher
- Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK
| | - Karina T Wright
- Institute of Science and Technology in Medicine, Keele University, Keele, Staffordshire, UK. .,Robert Jones and Agnes Hunt Orthopaedic Hospital, Oswestry, Shropshire, UK.
| |
Collapse
|
64
|
Lu N, Lu J, Zhou C, Zhong F. Association between transforming growth factor-beta 1 gene single nucleotide polymorphisms and knee osteoarthritis susceptibility in a Chinese Han population. J Int Med Res 2017. [PMID: 28627979 PMCID: PMC5718715 DOI: 10.1177/0300060517705719] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Objective To investigate associations between single polymorphisms (SNPs) rs1800469 and rs1982073 in the transforming growth factor-β1 gene (TGF-β1) and knee osteoarthritis (OA) susceptibility in a Chinese Han population. Methods TGF-β1 rs1800469 and rs1982073 were genotyped in patients with knee OA and age- and sex-matched OA-free controls from a Chinese Han population. The association was further analyzed according to gender and age. Results A total of 765 patients with knee OA and 780 controls were included. CT and CT + CC genotypes of rs1982073, and variant C, were associated with a significantly increased risk of knee OA. Stratification analysis showed that the association between the OA risk and rs1982073 CT heterozygotes compared with TT homozygotes was stronger in females and those aged >65 years. In contrast, CT, TT, and CT + TT genotypes of rs1800469 were not significantly associated with the risk of knee OA, even after further stratification analysis for gender and age. Conclusions The TGF-β1 rs1982073 T to C change and the variant C genotype may contribute to knee OA risk in the Chinese Han population.
Collapse
Affiliation(s)
- Ning Lu
- 1 Tongde Hospital of Zhejiang Province, Department of Orthopaedics, Zhejiang, CN, P.R. China
| | - Jianwei Lu
- 1 Tongde Hospital of Zhejiang Province, Department of Orthopaedics, Zhejiang, CN, P.R. China
| | - Chenhe Zhou
- 2 Second Affiliated Hospital's Campus in Binjiang District, School of Medicine, Zhejiang University, Department of Orthopedic Surgery, Hangzhou, Zhejiang, P.R. China
| | - FuHua Zhong
- 1 Tongde Hospital of Zhejiang Province, Department of Orthopaedics, Zhejiang, CN, P.R. China
| |
Collapse
|
65
|
Longobardi L, Temple JD, Tagliafierro L, Willcockson H, Esposito A, D'Onofrio N, Stein E, Li T, Myers TJ, Ozkan H, Balestrieri ML, Ulici V, Loeser RF, Spagnoli A. Role of the C-C chemokine receptor-2 in a murine model of injury-induced osteoarthritis. Osteoarthritis Cartilage 2017; 25:914-925. [PMID: 27856294 PMCID: PMC5430000 DOI: 10.1016/j.joca.2016.11.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 09/26/2016] [Accepted: 11/07/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We previously found in our embryonic studies that proper regulation of the chemokine CCL12 through its sole receptor CCR2, is critical for joint and growth plate development. In the present study, we examined the role of CCR2 in injury-induced-osteoarthritis (OA). METHOD We used a murine model of injury-induced-OA (destabilization of medial meniscus, DMM), and systemically blocked CCR2 using a specific antagonist (RS504393) at different times during disease progression. We examined joint degeneration by assessing cartilage (cartilage loss, chondrocyte hypertrophy, MMP-13 expression) and bone lesions (bone sclerosis, osteophytes formation) with or without the CCR2 antagonist. We also performed pain behavioral studies by assessing the weight distribution between the normal and arthritic hind paws using the IITS incapacitance meter. RESULTS Testing early vs delayed administration of the CCR2 antagonist demonstrated differential effects on joint damage. We found that OA changes in articular cartilage and bone were ameliorated by pharmacological CCR2 blockade, if given early in OA development: specifically, pharmacological targeting of CCR2 during the first 4 weeks (wks) following injury, reduced OA cartilage and bone damage, with less effectiveness with later treatments. Importantly, our pain-related behavioral studies showed that blockade of CCR2 signaling during early, 1-4 wks post-surgery or moderate, 4-8 wks post-surgery, OA was sufficient to decrease pain measures, with sustained improvement at later stages, after treatment was stopped. CONCLUSIONS Our data highlight the potential efficacy of antagonizing CCR2 at early stages to slow the progression of post-injury OA and, in addition, improve pain symptoms.
Collapse
Affiliation(s)
- L Longobardi
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - J D Temple
- Department of Biomedical Engineering, UNC-Chapel Hill, NC, USA; Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| | - L Tagliafierro
- Department of Neurology, Duke University, Durham, NC, USA.
| | - H Willcockson
- Department of Cell Biology and Physiology, UNC-Chapel Hill, NC, USA.
| | - A Esposito
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| | - N D'Onofrio
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.
| | - E Stein
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| | - T Li
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| | - T J Myers
- Department of Pediatrics, UNC-Chapel Hill, NC, USA.
| | - H Ozkan
- Department of Orthopaedics, Gulhane Military Medical Academy, Etlik, Ankara, Turkey.
| | - M L Balestrieri
- Department of Biochemistry, Biophysics and General Pathology, Second University of Naples, Naples, Italy.
| | - V Ulici
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - R F Loeser
- Division of Rheumatology, Allergy and Immunology and the Thurston Arthritis Research Center, University of North Carolina-Chapel Hill, NC, USA.
| | - A Spagnoli
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA.
| |
Collapse
|
66
|
Fang J, Xiao L, Chen R, Zhao Z. Conditional removal of the canonical TGF-β1 signaling delays condylar cartilage degeneration induced by a partial discectomy in mice. PLoS One 2017; 12:e0177826. [PMID: 28542404 PMCID: PMC5436809 DOI: 10.1371/journal.pone.0177826] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/03/2017] [Indexed: 02/05/2023] Open
Abstract
Recent emerging data indicate that the increase in the expression and activity of the transforming growth factor beta 1 (Tgf-β1) signaling may have detrimental effect to mature articular cartilage of knee joints. However, there is no information about whether or not this is the case in condylar cartilages. The objective of this study is to investigate the protein expression and activity of Tgf-β1 signaling in degenerative condylar cartilages. We also investigate biological effects of the conditional deletion of transforming growth factor receptor type II (Tgfbr2) in condylar cartilage of adult mice after a partial discectomy. Two mouse models of osteoarthritis (OA) were used to examine protein expressions of Tgf-β1 and p-Smad2/3 in condylar cartilages at early degenerative stages. In addition, cartilage specific Tgfbr2-deficient adult mice were subjected to a partial discectomy. The morphological condition of condylar cartilages was evaluated in mice at 4 and 12 weeks after the surgery. We found that protein levels of Tgf-β1 and p-Smad2/3 were increased in the degenerative condylar cartilage of the mouse models. The conditional removal of Tgfbr2 in mature condylar cartilage significantly delayed the progressive progression of the cartilage degeneration induced by a partial discectomy. We conclude that the increase in the expression and activity of Tgf-β1 signaling may have detrimental effect to mature condylar cartilages. Therefore, inhibition of Tgf-β1 signaling may be able to protect condylar cartilages from being degraded in mature temporomandibular joints.
Collapse
Affiliation(s)
- Jie Fang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Li Xiao
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States of America
- Department of Stomatology, Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| | - Rebecca Chen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, United States of America
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
- * E-mail:
| |
Collapse
|
67
|
Hasei J, Teramura T, Takehara T, Onodera Y, Horii T, Olmer M, Hatada I, Fukuda K, Ozaki T, Lotz MK, Asahara H. TWIST1 induces MMP3 expression through up-regulating DNA hydroxymethylation and promotes catabolic responses in human chondrocytes. Sci Rep 2017; 7:42990. [PMID: 28220902 PMCID: PMC5318945 DOI: 10.1038/srep42990] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 01/18/2017] [Indexed: 02/05/2023] Open
Abstract
The objective was to investigate the levels of TWIST1 in normal and OA cartilage and examine its role in regulating gene expression in chondrocytes. Human cartilage tissues and chondrocytes were obtained at autopsy from normal knee joints and from OA-affected joints at the time of total knee arthroplasty. TWIST1 expression was increased in human OA knee cartilage compared to normal knee cartilage. TWIST1 induced matrix metalloproteinase 3 (MMP3) expression without direct binding to MMP3 promoter and increased the 5-hydroxymethylcytosine (5hmC) level at the MMP3 promoter. The effect of TWIST1 on expression of TET family (TET1, 2 and 3) was measured in stable TWIST1 transfected TC28 cells, and TET1 expression was up-regulated. TWIST1 dependent upregulation of Mmp3 expression was suppressed in Tet triple KO fibroblast derived from mouse ES cells. Increased TWIST1 expression is a feature of OA-affected cartilage. We identified a novel mechanism of catabolic reaction where TWIST1 up-regulates MMP3 expression by enriching 5hmC levels at the MMP3 promoter via TET1 induction. These findings implicate TWIST1 as an important factor regulating OA related gene expression. Clarifying epigenetic mechanisms of 5hmC induced by TWIST1 is a critical molecule to understanding OA pathogenesis.
Collapse
Affiliation(s)
- Joe Hasei
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Takeshi Teramura
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Toshiyuki Takehara
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Yuta Onodera
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Takuro Horii
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Merissa Olmer
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Izuho Hatada
- Laboratory of Genome Science, Biosignal Genome Resource Center, Institute for Molecular and Cellular Regulation, Gunma University, Gunma, Japan
| | - Kanji Fukuda
- Division of Cell Biology for Regenerative Medicine, Institute of Advanced Clinical Medicine, Kindai University, Faculty of Medicine, Osaka, Japan
- Department of Rehabilitation Medicine, Kindai University, Faculty of Medicine, Osaka, Japan
| | - Toshifumi Ozaki
- Science of Functional Recovery and Reconstruction, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Martin K. Lotz
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Hiroshi Asahara
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
68
|
|
69
|
Chavez RD, Coricor G, Perez J, Seo HS, Serra R. SOX9 protein is stabilized by TGF-β and regulates PAPSS2 mRNA expression in chondrocytes. Osteoarthritis Cartilage 2017; 25:332-340. [PMID: 27746378 PMCID: PMC5258840 DOI: 10.1016/j.joca.2016.10.007] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/29/2016] [Accepted: 10/05/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE We previously identified 3'-phosphoadenosine 5'-phosphosulfate synthase 2 (PAPSS2) as a transcriptional target of transforming growth factor β (TGF-β) in chondrocytes. PAPSS2 is required for proper sulfation of proteoglycans in cartilage. Defective sulfation in the matrix results in alterations in mechanical properties of the cartilage that would be expected to result in degeneration. The objective of this study was to identify factors that regulate PAPSS2 expression and compare to a known TGF-β responsive gene, proteoglycan 4/lubricin (PRG4). In this study, TGF-β-mediated regulation of SOX9 was characterized, and the involvement of SOX9 in regulation of PAPSS2 mRNA was investigated. DESIGN Primary bovine articular chondrocytes grown in micromass culture and ATDC5 cells were used as the model system. Adenoviruses were used to express SOX9 and SMAD3. siRNA was used to knock-down Sox9 and Smad3. Western blot and real-time quantitative RT-PCR (qPCR) were used to measure changes in protein and mRNA levels in response to treatment. RESULTS Over-expression of SOX9 was sufficient to up-regulate PAPSS2 mRNA. TGF-β treatment of SOX9-expressing cells resulted in enhanced up-regulation of PAPSS2 mRNA, suggesting that SOX9 cooperates with TGF-β signaling. Furthermore, Sox9 was required for full TGF-β-mediated induction of Papss2. In contrast, PRG4 was regulated by SMAD3 but not SOX9. SOX9 protein levels were increased after treatment with TGF-β, although SOX9 mRNA was not. SOX9 protein was post-translationally stabilized after treatment with TGF-β. CONCLUSIONS TGF-β stabilizes SOX9 protein, and SOX9 is sufficient and necessary for TGF-β-mediated regulation of PAPSS2 mRNA, providing a novel mechanism for TGF-β-mediated gene regulation in chondrocytes.
Collapse
Affiliation(s)
| | | | | | | | - R Serra
- corresponding author. Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, 1918 University Blvd., 660 MCLM, Birmingham, AL, 35294-0005
| |
Collapse
|
70
|
Gronau T, Krüger K, Prein C, Aszodi A, Gronau I, Iozzo RV, Mooren FC, Clausen-Schaumann H, Bertrand J, Pap T, Bruckner P, Dreier R. Forced exercise-induced osteoarthritis is attenuated in mice lacking the small leucine-rich proteoglycan decorin. Ann Rheum Dis 2017; 76:442-449. [PMID: 27377816 DOI: 10.1136/annrheumdis-2016-209319] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 06/05/2016] [Accepted: 06/14/2016] [Indexed: 11/04/2022]
Abstract
OBJECTIVE Interterritorial regions of articular cartilage matrix are rich in decorin, a small leucine-rich proteoglycan and important structural protein, also involved in many signalling events. Decorin sequesters transforming growth factor β (TGFβ), thereby regulating its activity. Here, we analysed whether increased bioavailability of TGFβ in decorin-deficient (Dcn-/-) cartilage leads to changes in biomechanical properties and resistance to osteoarthritis (OA). METHODS Unchallenged knee cartilage was analysed by atomic force microscopy (AFM) and immunohistochemistry. Active transforming growth factor β-1 (TGFβ1) content within cultured chondrocyte supernatants was measured by ELISA. Quantitative real-time (RT)-PCR was used to analyse mRNA expression of glycosaminoglycan (GAG)-modifying enzymes in C28/I2 cells following TGFβ1 treatment. In addition, OA was induced in Dcn-/- and wild-type (WT) mice via forced exercise on a treadmill. RESULTS AFM analysis revealed a strikingly higher compressive stiffness in Dcn-/- than in WT cartilage. This was accompanied by increased negative charge and enhanced sulfation of GAG chains, but not by alterations in the levels of collagens or proteoglycan core proteins. In addition, decorin-deficient chondrocytes were shown to release more active TGFβ1. Increased TGFβ signalling led to enhanced Chst11 sulfotransferase expression inducing an increased negative charge density of cartilage matrix. These negative charges might attract more water resulting in augmented compressive stiffness of the tissue. Therefore, decorin-deficient mice developed significantly less OA after forced exercise than WT mice. CONCLUSIONS Our study demonstrates that the disruption of decorin-restricted TGFβ signalling leads to higher stiffness of articular cartilage matrix, rendering joints more resistant to OA. Therefore, the loss of an important structural component can improve cartilage homeostasis.
Collapse
MESH Headings
- Animals
- Arthritis, Experimental/etiology
- Arthritis, Experimental/genetics
- Arthritis, Experimental/metabolism
- Biomechanical Phenomena
- Cartilage, Articular/metabolism
- Decorin/genetics
- Decorin/metabolism
- Enzyme-Linked Immunosorbent Assay
- Glycosaminoglycans/metabolism
- Immunohistochemistry
- Mice
- Mice, Knockout
- Microscopy, Atomic Force
- Osteoarthritis/etiology
- Osteoarthritis/genetics
- Osteoarthritis/metabolism
- Physical Conditioning, Animal/adverse effects
- Physical Conditioning, Animal/methods
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Real-Time Polymerase Chain Reaction
- Transforming Growth Factor beta/metabolism
- Transforming Growth Factor beta/pharmacology
Collapse
Affiliation(s)
- Tobias Gronau
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Karsten Krüger
- Institute of Sports Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Carina Prein
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences and Center for Nanoscience (CeNS), Munich, Germany
| | - Attila Aszodi
- Laboratory of Experimental Surgery and Regenerative Medicine, Department of General, Trauma and Reconstruction Surgery, Ludwig-Maximilians-University, Munich, Germany
| | - Isabel Gronau
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Renato V Iozzo
- Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, USA
| | - Frank C Mooren
- Institute of Sports Medicine, Justus-Liebig University Giessen, Giessen, Germany
| | - Hauke Clausen-Schaumann
- Center for Applied Tissue Engineering and Regenerative Medicine (CANTER), Munich University of Applied Sciences and Center for Nanoscience (CeNS), Munich, Germany
| | - Jessica Bertrand
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
- Department of Orthopaedic Surgery, Otto-von-Guericke University, Magdeburg, Germany
| | - Thomas Pap
- Institute of Experimental Musculoskeletal Medicine, University Hospital Münster, Münster, Germany
| | - Peter Bruckner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Rita Dreier
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| |
Collapse
|
71
|
Chen D, Shen J, Zhao W, Wang T, Han L, Hamilton JL, Im HJ. Osteoarthritis: toward a comprehensive understanding of pathological mechanism. Bone Res 2017; 5:16044. [PMID: 28149655 PMCID: PMC5240031 DOI: 10.1038/boneres.2016.44] [Citation(s) in RCA: 768] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease and a major cause of pain and disability in adult individuals. The etiology of OA includes joint injury, obesity, aging, and heredity. However, the detailed molecular mechanisms of OA initiation and progression remain poorly understood and, currently, there are no interventions available to restore degraded cartilage or decelerate disease progression. The diathrodial joint is a complicated organ and its function is to bear weight, perform physical activity and exhibit a joint-specific range of motion during movement. During OA development, the entire joint organ is affected, including articular cartilage, subchondral bone, synovial tissue and meniscus. A full understanding of the pathological mechanism of OA development relies on the discovery of the interplaying mechanisms among different OA symptoms, including articular cartilage degradation, osteophyte formation, subchondral sclerosis and synovial hyperplasia, and the signaling pathway(s) controlling these pathological processes.
Collapse
Affiliation(s)
- Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Jie Shen
- Department of Orthopaedic Surgery, Washington University, St Louis, MO, USA
| | - Weiwei Zhao
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
- Department of Orthopaedics & Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Tingyu Wang
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China
| | - Lin Han
- School of Biomedical Engineering, Science, and Health Systems, Drexel University, Philadelphia, PA, USA
| | - John L Hamilton
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Hee-Jeong Im
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
72
|
Abstract
Osteoarthritis (OA) was once defined as a non-inflammatory arthropathy, but it is now well-recognized that there is a major inflammatory component to this disease. In addition to synovial cells, articular chondrocytes and other cells of diarthrodial joints are also known to express inflammatory mediators. It has been proposed that targeting inflammation pathways could be a promising strategy to treat OA. There have been many reports of cross-talk between inflammation and epigenetic factors in cartilage. Specifically, inflammatory mediators have been shown to regulate levels of enzymes that catalyze changes in DNA methylation and histone structure, as well as alter levels of non-coding RNAs. In addition, expression levels of a number of these epigenetic factors have been shown to be altered in OA, thereby suggesting potential interplay between inflammation and epigenetics in this disease. This review provides information on inflammatory pathways in arthritis and summarizes published research on how epigenetic regulators are affected by inflammation in chondrocytes. Furthermore, we discuss data showing how altered expression of some of these epigenetic factors can induce either catabolic or anti-catabolic effects in response to inflammatory signals. A better understanding of how inflammation affects epigenetic factors in OA may provide us with novel therapeutic strategies to treat this condition.
Collapse
Affiliation(s)
- Jie Shen
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Yousef Abu-Amer
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Regis J. O'Keefe
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Audrey McAlinden
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO, USA,Department of Cell Biology & Physiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
73
|
Coricor G, Serra R. TGF-β regulates phosphorylation and stabilization of Sox9 protein in chondrocytes through p38 and Smad dependent mechanisms. Sci Rep 2016; 6:38616. [PMID: 27929080 PMCID: PMC5144132 DOI: 10.1038/srep38616] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/11/2016] [Indexed: 12/25/2022] Open
Abstract
Members of the TGF-β superfamily are important regulators of chondrocyte function. Sox9, a key transcriptional regulator of chondrogenesis, is required for TGF-β-mediated regulation of specific cartilage genes. TGF-β can signal through a canonical, Smad-mediated pathway or non-conical pathways, including p38. Here we show that both pathways are activated in chondrocytes after treatment with TGF-β and that TGF-β stabilizes Sox9 protein and increases phosphorylation of Sox9. Mutagenesis of potential serine phosphorylation sites on Sox9 was used to demonstrate that serine 211 is required to maintain normal basal levels of Sox9 as well as mediate increased Sox9 levels in response to TGF-β. The serine 211 site is in a motif that is targeted by p38 kinase. We used siRNA and pharmacological agents to show that p38 and Smad3 independently regulate the phosphorylation and stability of Sox9. Previously, we demonstrated that Papss2 is a downstream transcriptional target of Sox9 and TGF-β. Here we show that p38 is required for TGF-β-mediated regulation of Papss2 mRNA. Together the results suggest a new mechanism for TGF-β-mediated gene regulation in chondrocytes via p38 and phosphorylation and stabilization of Sox9. Understanding how TGF-β regulates Sox9 may lead to identification of therapeutic targets for OA.
Collapse
Affiliation(s)
- George Coricor
- University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama, 35294-0005, USA
| | - Rosa Serra
- University of Alabama at Birmingham, Department of Cell, Developmental, and Integrative Biology, Birmingham, Alabama, 35294-0005, USA
| |
Collapse
|
74
|
Wang W, Song B, Anbarchian T, Shirazyan A, Sadik JE, Lyons KM. Smad2 and Smad3 Regulate Chondrocyte Proliferation and Differentiation in the Growth Plate. PLoS Genet 2016; 12:e1006352. [PMID: 27741240 PMCID: PMC5065210 DOI: 10.1371/journal.pgen.1006352] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 09/08/2016] [Indexed: 12/30/2022] Open
Abstract
TGFβs act through canonical and non-canonical pathways, and canonical signals are transduced via Smad2 and Smad3. However, the contribution of canonical vs. non-canonical pathways in cartilage is unknown because the role of Smad2 in chondrogenesis has not been investigated in vivo. Therefore, we analyzed mice in which Smad2 is deleted in cartilage (Smad2CKO), global Smad3-/- mutants, and crosses of these strains. Growth plates at birth from all mutant strains exhibited expanded columnar and hypertrophic zones, linked to increased proliferation in resting chondrocytes. Defects were more severe in Smad2CKO and Smad2CKO;Smad3-/-(Smad2/3) mutant mice than in Smad3-/- mice, demonstrating that Smad2 plays a role in chondrogenesis. Increased levels of Ihh RNA, a key regulator of chondrocyte proliferation and differentiation, were seen in prehypertrophic chondrocytes in the three mutant strains at birth. In accordance, TGFβ treatment decreased Ihh RNA levels in primary chondrocytes from control (Smad2fx/fx) mice, but inhibition was impaired in cells from mutants. Consistent with the skeletal phenotype, the impact on TGFβ-mediated inhibition of Ihh RNA expression was more severe in Smad2CKO than in Smad3-/- cells. Putative Smad2/3 binding elements (SBEs) were identified in the proximal Ihh promoter. Mutagenesis demonstrated a role for three of them. ChIP analysis suggested that Smad2 and Smad3 have different affinities for these SBEs, and that the repressors SnoN and Ski were differentially recruited by Smad2 and Smad3, respectively. Furthermore, nuclear localization of the repressor Hdac4 was decreased in growth plates of Smad2CKO and double mutant mice. TGFβ induced association of Hdac4 with Smad2, but not with Smad3, on the Ihh promoter. Overall, these studies revealed that Smad2 plays an essential role in the development of the growth plate, that both Smads 2 and 3 inhibit Ihh expression in the neonatal growth plate, and suggested they accomplish this by binding to distinct SBEs, mediating assembly of distinct repressive complexes. The cartilage growth plate regulates the size and shape of nearly every skeletal element in the body. TGFβs are potent inducers of cartilage formation, but the mechanisms by which they transduce their signals in cartilage during development are poorly understood. Similarly, there is strong evidence that dysregulation of the TGFβ pathway increases the risk for osteoarthritis (OA) in humans, but the underlying mechanisms are unknown. TGFβs transduce their signals through a canonical pathway involving Smad2 and Smad3, and through several non-canonical pathways. However, the roles of canonical vs. noncanonical signaling are unknown in cartilage because the combined roles of Smad2 and Smad3 have not been determined. We generated mice lacking both Smad2 and Smad3 in cartilage in order to determine the role of canonical TGFβ signaling during embryonic development. We determined that Smad2 has a more prominent role than Smad3 in non-hypertrophic chondrocytes in the growth plate, and identified elevated levels of Ihh RNA in neonatal cartilage in Smad2 and Smad3 mutants. These findings may be important because Ihh is a vital regulator of cartilage proliferation and differentiation during cartilage development. More generally, the studies identify how Smad2 and Smad3 can regulate a common target gene through distinct mechanisms.
Collapse
Affiliation(s)
- Weiguang Wang
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Buer Song
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Teni Anbarchian
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Anna Shirazyan
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Joshua E. Sadik
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Karen M. Lyons
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, California, United States of America
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
75
|
Madej W, van Caam A, Blaney Davidson E, Buma P, van der Kraan PM. Unloading results in rapid loss of TGFβ signaling in articular cartilage: role of loading-induced TGFβ signaling in maintenance of articular chondrocyte phenotype? Osteoarthritis Cartilage 2016; 24:1807-1815. [PMID: 27268943 DOI: 10.1016/j.joca.2016.05.018] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 04/28/2016] [Accepted: 05/23/2016] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Recently it was shown that loading of articular cartilage explants activates TGFβ signaling. Here we investigated if in vivo chondrocytes express permanently high TGFβ signaling, and the consequence of the loss of compressive loading-mediated TGFβ signaling on chondrocyte function and phenotype. METHOD Bovine articular cartilage explants were collected within 10 min post mortem and stained immediately and after 30, 60 and 360 min for phosphorylated-Smad2, indicating active TGFβ signaling. Explants were unloaded for 48 h and subsequently repeatedly loaded with a compressive load of 3 MPa. In addition, explants were cultured unloaded for 2 weeks and the effect of loading or exogenous TGFβ on proteoglycan level and chondrocyte phenotype (Col10a1 mRNA expression) was analyzed. RESULTS Unloading of articular cartilage results in rapid loss of TGFβ signaling while subsequent compressive loading swiftly restored this. Loading and exogenous TGFβ enhanced expression of TGFβ1 and ALK5. Unloading of explants for 2 weeks resulted in proteoglycan loss and increased Col10a1 expression. Both loading and exogenous TGFβ inhibited elevated Col10a1 expression but not proteoglycan loss. CONCLUSION Our data might imply that in vivo regular physiological loading of articular cartilage leads to enduring TGFβ signaling and TGFβ-induced gene expression. We propose a hypothetical model in which loading activates a self-perpetuating system that prevents hypertrophic differentiation of chondrocytes and is crucial for cartilage homeostasis.
Collapse
Affiliation(s)
- W Madej
- Orthopedic Research Laboratory, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - A van Caam
- Experimental Rheumatology, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - E Blaney Davidson
- Experimental Rheumatology, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - P Buma
- Orthopedic Research Laboratory, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands
| | - P M van der Kraan
- Experimental Rheumatology, Radboudumc, PO Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
76
|
van der Kraan P, Matta C, Mobasheri A. Age-Related Alterations in Signaling Pathways in Articular Chondrocytes: Implications for the Pathogenesis and Progression of Osteoarthritis - A Mini-Review. Gerontology 2016; 63:29-35. [PMID: 27595269 DOI: 10.1159/000448711] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/27/2016] [Indexed: 11/19/2022] Open
Abstract
Musculoskeletal conditions are a major burden on individuals, healthcare systems, and social care systems throughout the world, with indirect costs having a predominant economic impact. Aging is a major contributing factor to the development and progression of arthritic and musculoskeletal diseases. Indeed, aging and inflammation (often referred to as 'inflammaging') are critical risk factors for the development of osteoarthritis (OA), which is one of the most common forms of joint disease. The term 'chondrosenescence' has recently been introduced to define the age-dependent deterioration of chondrocyte function and how it undermines cartilage function in OA. An important component of chondrosenescence is the age-related deregulation of subcellular signaling pathways in chondrocytes. This mini-review discusses the role of age-related alterations in chondrocyte signaling pathways. We focus our attention on two major areas: age-dependent alterations in transforming growth factor-β signaling and changes in protein kinase and phosphoprotein phosphatase activities in aging chondrocytes. A better understanding of the basic signaling mechanisms underlying aging in chondrocytes is likely to facilitate the development of new therapeutic and preventive strategies for OA and a range of other age-related osteoarticular disorders.
Collapse
Affiliation(s)
- Peter van der Kraan
- Department of Rheumatology, Experimental Rheumatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | | |
Collapse
|
77
|
Verschoyle RD, Edwards R, Nolan B, Greaves P. Articular Chondromatosis and Chrondroid Metaplasia in Transgenic TAg Mice. Toxicol Pathol 2016; 32:22-5. [PMID: 14713544 DOI: 10.1080/01926230490260691] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The C3(1)/SV40 T antigen transgenic mouse model for which rapid mammary and prostate tumor development has been documented uses the FVB/N mouse as a background strain. In this study, where the background strain used was the C57BL/6J mouse, neither mammary nor prostate tumors developed over periods of up to 40 weeks. However, a disturbance of hyaline cartilage in joints was observed similar to that found in synovial chondromatosis in humans. In addition, cartilage thickening in the external ears and cartilaginous metaplasia of the ascending aorta also occurred. This suggests that rearrangement of the transgene occurred in breeding on the C57BL background, thus modifying its expression. It raises the possibility that the genetic changes induced by the SV40 T antigen transforming sequence are important in cartilage homeostasis.
Collapse
Affiliation(s)
- Richard D Verschoyle
- Department of Oncology, University of Leicester, Leicester LE2 7LX, United Kingdom
| | | | | | | |
Collapse
|
78
|
Abstract
Strategies that seek to enhance musculoskeletal tissue regeneration and repair by modulating the biologic microenvironment at the site of injury have considerable therapeutic potential. Current and emerging biologic approaches include the use of growth factors, platelet-rich plasma, stem cell therapy, and scaffolds. The American Academy of Orthopaedic Surgeons hosted a research symposium in November 2015 to review the current state-of-the-art biologic treatments of articular cartilage, muscle, tendon, and bone injuries and identify knowledge gaps related to these emerging treatments. This review outlines the findings of the symposium and summarizes the consensus reached on how best to advance research on biologic treatment of orthopaedic injuries.
Collapse
|
79
|
The role of TGF-ß1 in osteoarthritis of the temporomandibular joint in two genetic mouse models. Arch Oral Biol 2016; 67:68-73. [DOI: 10.1016/j.archoralbio.2016.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 02/16/2016] [Accepted: 03/13/2016] [Indexed: 11/20/2022]
|
80
|
Wen ZH, Tang CC, Chang YC, Huang SY, Lin YY, Hsieh SP, Lee HP, Lin SC, Chen WF, Jean YH. Calcitonin attenuates cartilage degeneration and nociception in an experimental rat model of osteoarthritis: role of TGF-β in chondrocytes. Sci Rep 2016; 6:28862. [PMID: 27345362 PMCID: PMC4921823 DOI: 10.1038/srep28862] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 06/06/2016] [Indexed: 12/19/2022] Open
Abstract
We investigated the role of the calcitonin (Miacalcin) in the progression of osteoarthritis (OA) and in nociceptive behavior in an experimental rat model of OA and osteoporosis. OA was induced by anterior cruciate ligament transection (ACLT) of the right knee and by bilateral ovariectomy (OVX) in Wistar rats. Nociceptive behaviors (secondary mechanical allodynia and weight-bearing distribution of the hind paws) were analyzed prior to surgery and every week, beginning at 12 weeks after surgery, up to 20 weeks. At 20 weeks, histopathological studies were performed on the cartilage of the knee joints. Immunohistochemical analysis was performed to examine the effect of calcitonin on transforming growth factor (TGF)-β1 expression in articular cartilage chondrocytes. Rats subjected to ACLT + OVX surgery showed obvious OA changes in the joints. Animals subjected to ACLT + OVX and treated with calcitonin showed significantly less cartilage degeneration and improved nociceptive tests compared with animals subjected to ACLT + OVX surgeries alone. Moreover, calcitonin increased TGF-β1 expression in chondrocytes in ACLT + OVX-affected cartilage. Subcutaneous injection of calcitonin (1) attenuated the development of OA, (2) concomitantly reduced nociception, and (3) modulated chondrocyte metabolism, possibly by increasing cellular TGF-β1 expression.
Collapse
Affiliation(s)
- Zhi-Hong Wen
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Chi-Chieh Tang
- Department of Early Childhood Education, National Pintung University, Taiwan
| | - Yi-Chen Chang
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shi-Ying Huang
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Yen-You Lin
- Department of Marine Biotechnology &Resources, and Center for Translational Biopharmaceuticals, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Shih-Peng Hsieh
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Hsin-Pai Lee
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan.,Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Sung-Chun Lin
- Section of Pathology, Pingtung Christian Hospital, Pingtung, Taiwan.,Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| | - Wu-Fu Chen
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan.,Department of Neurosurgery, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Chang Gung University College of Medicine, Taiwan
| | - Yen-Hsuan Jean
- Department of Orthopedic Surgery, Pingtung Christian Hospital, Pingtung, Taiwan
| |
Collapse
|
81
|
Frazier K, Thomas R, Scicchitano M, Mirabile R, Boyce R, Zimmerman D, Grygielko E, Nold J, DeGouville AC, Huet S, Laping N, Gellibert F. Inhibition of ALK5 Signaling Induces Physeal Dysplasia in Rats. Toxicol Pathol 2016; 35:284-95. [PMID: 17366323 DOI: 10.1080/01926230701198469] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
TGF-|β|, and its type 1 (ALK5) receptor, are critical to the pathogenesis of fibrosis. In toxicologic studies of 4 or more days in 10-week-old Sprague–Dawley rats, using an ALK5 inhibitor (GW788388), expansion of hypertrophic and proliferation zones of femoral physes were noted. Subphyseal hyperostosis, chondrocyte hypertrophy/hyperplasia, and increased matrix were present. Physeal zones were laser microdissected from ALK5 inhibitor-treated and control rats sacrificed after 3 days of treatment. Transcripts for TGF-|β|1, TGF-|β|2, ALK5, IHH, VEGF, BMP-7, IGF-1, bFGF, and PTHrP were amplified by real-time PCR. IGF and IHH increased in all physis zones with treatment, but were most prominent in prehypertrophic zones. TGF-|β|2, bFGF and BMP7 expression increased in proliferative, pre- and hypertrophic zones. PTHrP expression was elevated in proliferative zones but decreased in hypertrophic zones. VEGF expression was increased after treatment in pre- and hypertrophic zones. ALK5 expression was elevated in prehypertrophic zones. Zymography demonstrated gelatinolytic activity was reduced after treatment. Apoptotic markers (TUNEL and caspase-3) were decreased in hypertrophic zones. Proliferation assessed by Topoisomerase II and Ki67 was increased in multiple zones. Movat stains demonstrated that proteoglycan deposition was altered. Physeal changes occurred at doses well above those resulting in fibrosis. Interactions of factors is important in producing the physeal dysplasia phenotype.
Collapse
MESH Headings
- Activin Receptors, Type I/antagonists & inhibitors
- Activin Receptors, Type I/genetics
- Activin Receptors, Type I/metabolism
- Animals
- Benzamides/adverse effects
- Bone Diseases, Developmental/chemically induced
- Bone Diseases, Developmental/pathology
- Cell Proliferation
- Dose-Response Relationship, Drug
- Gene Expression Regulation
- Growth Plate/drug effects
- Growth Plate/pathology
- Protein Serine-Threonine Kinases
- Pyrazoles/adverse effects
- Rats
- Rats, Sprague-Dawley
- Receptor, Transforming Growth Factor-beta Type I
- Receptors, Transforming Growth Factor beta/antagonists & inhibitors
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Transforming Growth Factor beta/physiology
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/physiology
Collapse
Affiliation(s)
- Kendall Frazier
- GlaxoSmithKline-Safety Assessment, King of Prussia, PA 19406, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Madej W, Buma P, van der Kraan P. Inflammatory conditions partly impair the mechanically mediated activation of Smad2/3 signaling in articular cartilage. Arthritis Res Ther 2016; 18:146. [PMID: 27334538 PMCID: PMC4918093 DOI: 10.1186/s13075-016-1038-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 05/31/2016] [Indexed: 01/14/2023] Open
Abstract
Background Joint trauma, which is frequently related with mechanical overloading of articular cartilage, is a well-established risk for osteoarthritis (OA) development. Additionally, reports show that trauma leads to synovial joint inflammation. In consequence, after joint trauma, cartilage is influenced by deleterious excessive loading combined with the catabolic activity of proinflammatory mediators. Since the activation of TGF-β signaling by loading is considered to be a key regulatory pathway for maintaining cartilage homeostasis, we tested the effect of proinflammatory conditions on mechanically mediated activation of TGF-β/Smad2/3P signaling in cartilage. Methods Cartilage explants were subjected to dynamic mechanical compression in the presence of interleukin-1 beta (IL-1β) or osteoarthritic synovium-conditioned medium (OAS-CM). Subsequently, the activation of the Smad2/3P pathway was monitored with QPCR analysis of reporter genes and additionally the expression of receptors activating the Smad2/3P pathway was analyzed. Finally, the ability for mechanically mediated activation of Smad2/3P was tested in human OA cartilage. Results IL-1β presence during compression did not impair the upregulation of Smad2/3P reporter genes, however the results were affected by IL-1β-mediated upregulations in unloaded controls. OAS-CM significantly impaired the compression-mediated upregulation of bSmad7 and Tgbfb1. IL-1β suppressed the compression-mediated bAlk5 upregulation where 12 MPa compression applied in the presence of OAS-CM downregulated the bTgfbr2. Mechanically driven upregulation of Smad2/3P reporter genes was present in OA cartilage. Conclusions Proinflammatory conditions partly impair the mechanically mediated activation of the protective TGF-β/Smad2/3P pathway. Additionally, the excessive mechanical compression, applied in the presence of proinflammatory conditions diminishes the expression of the type II TGF-β receptor, a receptor critical for maintenance of articular cartilage.
Collapse
Affiliation(s)
- Wojciech Madej
- Orthopedic Research Laboratory, Radboud University Medical Center, Geert Grooteplein-Zuid 10, route 547, 6525, GA, Nijmegen, The Netherlands.,Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 26-28, route 272, 6525, GA, Nijmegen, The Netherlands
| | - Pieter Buma
- Orthopedic Research Laboratory, Radboud University Medical Center, Geert Grooteplein-Zuid 10, route 547, 6525, GA, Nijmegen, The Netherlands
| | - Peter van der Kraan
- Experimental Rheumatology, Radboud University Medical Center, Geert Grooteplein 26-28, route 272, 6525, GA, Nijmegen, The Netherlands.
| |
Collapse
|
83
|
Ikegawa N, Sasho T, Yamaguchi S, Saito M, Akagi R, Muramatsu Y, Akatsu Y, Fukawa T, Nakagawa K, Nakajima A, Suzuki T, Takahashi K. Identification of genes required for the spontaneous repair of partial-thickness cartilage defects in immature rats. Connect Tissue Res 2016; 57:190-9. [PMID: 26719950 DOI: 10.3109/03008207.2015.1121250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
PURPOSE Our previous study showed that partial-thickness articular cartilage defects (PTCDs) created in immature rats spontaneously healed to resemble normal hyaline cartilage, but that of mature rats did not. To identify molecules involved in the spontaneous cartilage repair observed in this model, gene expression was compared between PTCD and sham-operated cartilage of immature and mature rats. MATERIALS AND METHODS Six sets of gene comparisons were made at 12, 24, and 48 hours after the creation of PTCDs in immature and mature rats using microarrays. All the genes upregulated in immature cartilage at 12 hours were selected for further analysis if their expression pattern was not irregular such that diminished at 24 hours and re-upregulated at 48 hours. Relationships among genes selected through the above steps were analyzed using Ingenuity Pathway Analysis (IPA) software. After deriving networks, important molecules were further narrowed down by location within a network. Genes were regarded as central if they had relationships with more than 10 molecules in a network. Protein localization in tissues was confirmed by immunohistochemistry. RESULTS Five networks were identified. Their functional annotations were gene expression, cell cycle, growth and proliferation, and cell signaling. Transforming growth factor-beta (TGF-β) was centrally located in the network with the highest IPA score and mothers against decapentaplegic homolog-3 (Smad3) were centrally located in the second highest ranking network. Phosphorylated Smad3 was detected in the nuclei of chondrocytes in immature cartilage. CONCLUSIONS Our data suggest the possible importance of Smad3 in the TGF-β signaling in the spontaneous healing of PTCDs in immature rats.
Collapse
Affiliation(s)
- Naoshi Ikegawa
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Takahisa Sasho
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Satoshi Yamaguchi
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Masahiko Saito
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Ryuichiro Akagi
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Yuta Muramatsu
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Yorikazu Akatsu
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Taisuke Fukawa
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Koichi Nakagawa
- b Department of Orthopaedic Surgery , Sakura Medical Center, Toho University , Sakura , Japan
| | - Arata Nakajima
- b Department of Orthopaedic Surgery , Sakura Medical Center, Toho University , Sakura , Japan
| | - Takane Suzuki
- c Department of Bioenvironmental Medicine , Graduate School of Medicine, Chiba University , Chiba , Japan
| | - Kazuhisa Takahashi
- a Department of Orthopaedic Surgery , Graduate School of Medicine, Chiba University , Chiba , Japan
| |
Collapse
|
84
|
Li YP, Wei XC, Li PC, Chen CW, Wang XH, Jiao Q, Wang DM, Wei FY, Zhang JZ, Wei L. The Role of miRNAs in Cartilage Homeostasis. Curr Genomics 2016; 16:393-404. [PMID: 27019614 PMCID: PMC4765526 DOI: 10.2174/1389202916666150817203144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 06/18/2015] [Accepted: 06/26/2015] [Indexed: 12/14/2022] Open
Abstract
Osteoarthritis (OA) is an age-related disease with poorly understood pathogenesis. Recent studies have demonstrated that miRNA might play a key role in OA initiation and development. We reviewed recent publications and elucidated the connection between miRNA and OA cartilage anabolic and catabolic signals, including four signaling pathways: TGF-β/Smads and BMPs signaling, associated with cartilage anabolism; and MAPK and NF-KB signaling, associated with cartilage catabolism. We also explored the relationships with MMP, ADAMTS and NOS (NitricOxide Synthases) families, as well as with the catabolic cytokines IL-1 and TNF-α. The potential role of miRNAs in biological processes such as cartilage degeneration, chondrocyte proliferation, and differentiation is discussed. Collective evidence indicates that miRNAs play a critical role in cartilage degeneration. These findings will aid in understanding the molecular network that governs articular cartilage homeostasis and in to elucidate the role of miRNA in the pathogenesis of OA.
Collapse
Affiliation(s)
- Yong Ping Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Xiao Chun Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Peng Cu Li
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Chun Wei Chen
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Xiao Hu Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Qiang Jiao
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Dong Ming Wang
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China
| | - Fang Yuan Wei
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jian Zhong Zhang
- Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Lei Wei
- Department of Orthopaedics, The Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi, China;; Foot and Ankle Orthopaedic Surgery Center, Beijing Tongren Hospital, Capital Medical University, Beijing, China; ; Department of Orthopaedics, The Warren Alpert Medical School of Brown University/Rhode Island Hospital, 1 Hoppin Street, Providence, RI 02903, USA
| |
Collapse
|
85
|
Wu M, Chen G, Li YP. TGF-β and BMP signaling in osteoblast, skeletal development, and bone formation, homeostasis and disease. Bone Res 2016; 4:16009. [PMID: 27563484 PMCID: PMC4985055 DOI: 10.1038/boneres.2016.9] [Citation(s) in RCA: 1146] [Impact Index Per Article: 127.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 03/04/2016] [Accepted: 03/07/2016] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor-beta (TGF-β) and bone morphogenic protein (BMP) signaling has fundamental roles in both embryonic skeletal development and postnatal bone homeostasis. TGF-βs and BMPs, acting on a tetrameric receptor complex, transduce signals to both the canonical Smad-dependent signaling pathway (that is, TGF-β/BMP ligands, receptors, and Smads) and the non-canonical-Smad-independent signaling pathway (that is, p38 mitogen-activated protein kinase/p38 MAPK) to regulate mesenchymal stem cell differentiation during skeletal development, bone formation and bone homeostasis. Both the Smad and p38 MAPK signaling pathways converge at transcription factors, for example, Runx2 to promote osteoblast differentiation and chondrocyte differentiation from mesenchymal precursor cells. TGF-β and BMP signaling is controlled by multiple factors, including the ubiquitin–proteasome system, epigenetic factors, and microRNA. Dysregulated TGF-β and BMP signaling result in a number of bone disorders in humans. Knockout or mutation of TGF-β and BMP signaling-related genes in mice leads to bone abnormalities of varying severity, which enable a better understanding of TGF-β/BMP signaling in bone and the signaling networks underlying osteoblast differentiation and bone formation. There is also crosstalk between TGF-β/BMP signaling and several critical cytokines’ signaling pathways (for example, Wnt, Hedgehog, Notch, PTHrP, and FGF) to coordinate osteogenesis, skeletal development, and bone homeostasis. This review summarizes the recent advances in our understanding of TGF-β/BMP signaling in osteoblast differentiation, chondrocyte differentiation, skeletal development, cartilage formation, bone formation, bone homeostasis, and related human bone diseases caused by the disruption of TGF-β/BMP signaling.
Collapse
Affiliation(s)
- Mengrui Wu
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| | - Guiqian Chen
- Department of Pathology, University of Alabama at Birmingham, Birmingham, USA; Department of neurology, Bruke Medical Research Institute, Weil Cornell Medicine of Cornell University, White Plains, USA
| | - Yi-Ping Li
- Department of Pathology, University of Alabama at Birmingham , Birmingham, USA
| |
Collapse
|
86
|
Huang H, Veien ES, Zhang H, Ayers DC, Song J. Skeletal Characterization of Smurf2-Deficient Mice and In Vitro Analysis of Smurf2-Deficient Chondrocytes. PLoS One 2016; 11:e0148088. [PMID: 26815610 PMCID: PMC4729489 DOI: 10.1371/journal.pone.0148088] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/12/2016] [Indexed: 12/21/2022] Open
Abstract
Overexpression of Smad ubiquitin regulatory factor 2 (Smurf2) in chondrocytes was reported to cause spontaneous osteoarthritis (OA) in mice. However, it is unclear whether Smurf2 is involved in bone and cartilage homeostasis and if it is required for OA pathogenesis. Here we characterized age-related changes in the bone and articular cartilage of Smurf2-deficient (MT) mice by microCT and histology, and examined whether reduced Smurf2 expression affected the severity of OA upon surgical destabilization of the medial meniscus (DMM). Using immature articular chondrocytes (iMAC) from MT and wild-type (WT) mice, we also examined how Smurf2 deficiency affects chondrogenic and catabolic gene expressions and Smurf2 and Smurf1 proteins upon TGF-β3 or IL-1β treatment in culture. We found no differences in cortical, subchondral and trabecular bone between WT and MT in young (4 months) and old mice (16–24 months). The articular cartilage and age-related alterations between WT and MT were also similar. However, 2 months following DMM, young MT showed milder OA compared to WT (~70% vs ~30% normal or exhibiting only mild OA cartilage phenotype). The majority of the older WT and MT mice developed moderate/severe OA 2 months after DMM, but a higher subset of aged MT cartilage (27% vs. 9% WT) remained largely normal. Chondrogenic gene expression (Sox9, Col2, Acan) trended higher in MT iMACs than WT with/without TGF-β3 treatment. IL-1β treatment suppressed chondrgenic gene expression, but Sox9 expression in MT remained significantly higher than WT. Smurf2 protein in WT iMACs increased upon TGF-β3 treatment and decreased upon IL-1β treatment in a dose-dependent manner. Smurf1 protein elevated more in MT than WT upon TGF-β3 treatment, suggesting a potential, but very mild compensatory effect. Overall, our data support a role of Smurf2 in regulating OA development but suggest that inhibiting Smurf2 alone may not be sufficient to prevent or consistently mitigate post-traumatic OA across a broad age range.
Collapse
MESH Headings
- Aging
- Animals
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cells, Cultured
- Chondrocytes/metabolism
- Chondrocytes/pathology
- Disease Models, Animal
- Female
- Gene Deletion
- Interleukin-1beta/metabolism
- Knee Joint
- Male
- Menisci, Tibial/pathology
- Menisci, Tibial/surgery
- Mice
- Mice, Inbred C57BL
- Osteoarthritis, Knee/genetics
- Osteoarthritis, Knee/metabolism
- Osteoarthritis, Knee/pathology
- Transforming Growth Factor beta3/metabolism
- Ubiquitin-Protein Ligases/genetics
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
- Henry Huang
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, United States of America
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Eric S. Veien
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Hong Zhang
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - David C. Ayers
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, United States of America
| | - Jie Song
- Department of Orthopedics and Physical Rehabilitation, University of Massachusetts Medical School, Worcester, MA, United States of America
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA, United States of America
- * E-mail:
| |
Collapse
|
87
|
Aury-Landas J, Marcelli C, Leclercq S, Boumédiene K, Baugé C. Genetic Determinism of Primary Early-Onset Osteoarthritis. Trends Mol Med 2016; 22:38-52. [DOI: 10.1016/j.molmed.2015.11.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 11/13/2015] [Accepted: 11/16/2015] [Indexed: 12/22/2022]
|
88
|
Miyazaki M, Yonemitsu I, Takei M, Kure-Hattori I, Ono T. The imbalance of masticatory muscle activity affects the asymmetric growth of condylar cartilage and subchondral bone in rats. Arch Oral Biol 2015; 63:22-31. [PMID: 26669214 DOI: 10.1016/j.archoralbio.2015.11.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 11/13/2015] [Accepted: 11/23/2015] [Indexed: 10/22/2022]
Abstract
OBJECTIVE To examine the effects of imbalance of masticatory muscle activity of the rat mandible on the condylar cartilage and subchondral bone during the growth period. DESIGN Forty 5-week-old male Wistar rats were randomly divided into experimental (n=20) and control (n=20) groups. In the experimental group, the left masseter muscles were resected. The rats were sacrificed at 7 or 9 weeks of age in both groups. Microcomputed tomography was used to determine the three-dimensional morphology and cancellous bone structure. For histological and histochemical examination, 5-μm-thick serial frontal sections of the condyle were stained with toluidine blue and immunostained with asporin and TGF-β1 to evaluate the promotion and inhibition of chondrogenesis. RESULTS In the experimental group, microcomputed tomography analysis showed asymmetric growth; the resected side condyles showed degenerative changes. Histological analysis showed that the total cartilage in the central region of the resected side was significantly thinner than in the non-resected side in the experimental group, as well as in the control group. Compared with the control group, the expression of asporin was significantly higher in the resected side, and significantly lower in the non-resected side. In contrast, the expression of TGF-β1-immunopositive cells in the non-resected side was significantly higher than in the resected side and the control group. CONCLUSIONS These findings imply that lateral imbalance of masseter muscle activity lead to inhibition of chondrogenesis and induce asymmetric formation of the condyle during the growth period.
Collapse
Affiliation(s)
- Mutsumi Miyazaki
- Orthodontic Science, Department of Orofacial Development and Function, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Ikuo Yonemitsu
- Orthodontic Science, Department of Orofacial Development and Function, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549, Japan.
| | - Maki Takei
- Orthodontic Science, Department of Orofacial Development and Function, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Ikuko Kure-Hattori
- Orthodontic Science, Department of Orofacial Development and Function, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549, Japan
| | - Takashi Ono
- Orthodontic Science, Department of Orofacial Development and Function, Division of Oral Health Sciences, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549, Japan
| |
Collapse
|
89
|
Upregulation of long non-coding RNA HIF 1α-anti-sense 1 induced by transforming growth factor-β-mediated targeting of sirtuin 1 promotes osteoblastic differentiation of human bone marrow stromal cells. Mol Med Rep 2015; 12:7233-8. [PMID: 26460121 PMCID: PMC4626181 DOI: 10.3892/mmr.2015.4415] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Accepted: 07/21/2015] [Indexed: 12/02/2022] Open
Abstract
The present study aimed to investigate the regulatory mechanism of long non-coding RNA hypoxia-inducible factor 1α-anti-sense 1 (lncRNA HIF1α-AS1) in osteoblast differentiation as well as its targeting by sirtuin 1 (SIRT1), which may be inhibited by transforming growth factor (TGF)-β in bone marrow stromal cells (BMSCs). Real-time polymerase chain reaction (PCR), western blot analysis, lncRNA PCR arrays and chromatin immunoprecipitation were performed in order to examine the interference of SIRT1 expression by TGF-β, the effects of SIRT1 overexpression on lncRNA HIF1α-AS1 and the regulation of the expression of homeobox (HOX)D10, which promotes BMSC differentiation, by lncRNA HIF1α-AS1. The results showed that TGF-β interfered with SIRT1 expression. Furthermore, lncRNA HIF1α-AS1 was significantly downregulated following overexpression of SIRT1. In addition, low expression of HIF1α-AS1 was sufficient to block the expression of HOXD10. The present study further demonstrated that downregulation of HOXD10 by HIF1α-AS1 interfered with acetylation, and subsequently resulted in the inhibition of osteoblast differentiation. These results suggested that HIF1α-AS1 is an essential mediator of osteoblast differentiation, and may thus represent a gene-therapeutic agent for the treatment of human bone diseases.
Collapse
|
90
|
Chen R, Mian M, Fu M, Zhao JY, Yang L, Li Y, Xu L. Attenuation of the progression of articular cartilage degeneration by inhibition of TGF-β1 signaling in a mouse model of osteoarthritis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:2875-85. [PMID: 26355014 DOI: 10.1016/j.ajpath.2015.07.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 06/22/2015] [Accepted: 07/26/2015] [Indexed: 10/23/2022]
Abstract
Transforming growth factor beta 1 (TGF-β1) is implicated in osteoarthritis. We therefore studied the role of TGF-β1 signaling in the development of osteoarthritis in a developmental stage-dependent manner. Three different mouse models were investigated. First, the Tgf-β receptor II (Tgfbr2) was specifically removed from the mature cartilage of joints. Tgfbr2-deficient mice were grown to 12 months of age and were then euthanized for collection of knee and temporomandibular joints. Second, Tgfbr2-deficient mice were subjected to destabilization of the medial meniscus (DMM) surgery. Knee joints were then collected from the mice at 8 and 16 weeks after the surgery. Third, wild-type mice were subjected to DMM at the age of 8 weeks. Immediately after the surgery, these mice were treated with the Tgfbr2 inhibitor losartan for 8 weeks and then euthanized for collection of knee joints. All joints were characterized for evidences of articular cartilage degeneration. Initiation or acceleration of articular cartilage degeneration was not observed by the genetic inactivation of Tgfbr2 in the joints at the age of 12 months. In fact, the removal of Tgfbr2 and treatment with losartan both delayed the progression of articular cartilage degeneration induced by DMM compared with control littermates. Therefore, we conclude that inhibition of Tgf-β1 signaling protects adult knee joints in mice against the development of osteoarthritis.
Collapse
Affiliation(s)
- Rebecca Chen
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Michelle Mian
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Martin Fu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Jing Ying Zhao
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Liang Yang
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts
| | - Yefu Li
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Faculty of Medicine, Harvard Medical School, Boston, Massachusetts.
| | - Lin Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts; Faculty of Medicine, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
91
|
Abstract
Cartilage remodeling is currently among the most popular topics in osteoarthritis research. Remodeling includes removal of the existing cartilage and replacement by neo-cartilage. As a loss of balance between removal and replacement of articular cartilage develops (particularly, the rate of removal surpasses the rate of replacement), joints will begin to degrade. In the last few years, significant progress in molecular understanding of the cartilage remodeling process has been made. In this brief review, we focus on the discussion of some current "controversial" observations in articular cartilage degeneration: (1) the biological effect of transforming growth factor-beta 1 on developing and mature articular cartilages, (2) the question of whether aggrecanase 1 (ADAMTS4) and aggrecanase 2 (ADAMTS5) are key enzymes in articular cartilage destruction, and (3) chondrocytes versus chondron in the development of osteoarthritis. It is hoped that continued discussion and investigation will follow to better clarify these topics. Clarification will be critical for those in search of novel therapeutic targets for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Yefu Li
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA ; Faculty of Medicine, Harvard Medical School, Boston, MA, USA
| | - Lin Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, MA, USA ; Faculty of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
92
|
Orth P, Madry H. Advancement of the Subchondral Bone Plate in Translational Models of Osteochondral Repair: Implications for Tissue Engineering Approaches. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:504-20. [PMID: 26066580 DOI: 10.1089/ten.teb.2015.0122] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Subchondral bone plate advancement is of increasing relevance for translational models of osteochondral repair in tissue engineering (TE). Especially for therapeutic TE approaches, a basic scientific knowledge of its chronological sequence, possible etiopathogenesis, and clinical implications are indispensable. This review summarizes the knowledge on this topic gained from a total of 31 translational investigations, including 1009 small and large animals. Experimental data indicate that the advancement of the subchondral bone plate frequently occurs during the spontaneous repair of osteochondral defects and following established articular cartilage repair approaches for chondral lesions such as marrow stimulation and TE-based strategies such as autologous chondrocyte implantation. Importantly, this subchondral bone reaction proceeds in a defined chronological and spatial pattern, reflecting both endochondral ossification and intramembranous bone formation. Subchondral bone plate advancement arises earlier in small animals and defects, but is more pronounced at the long term in large animals. Possible etiopathologies comprise a disturbed subchondral bone/articular cartilage crosstalk and altered biomechanical conditions or neovascularization. Of note, no significant correlation was found so far between subchondral bone plate advancement and articular cartilage repair. This evidence from translational animal models adverts to an increasing awareness of this previously underestimated pathology. Future research will shed more light on the advancement of the subchondral bone plate in TE models of cartilage repair.
Collapse
Affiliation(s)
- Patrick Orth
- 1 Center of Experimental Orthopedics, Saarland University , Homburg, Germany .,2 Department of Orthopedic Surgery, Saarland University Medical Center , Homburg, Germany
| | - Henning Madry
- 1 Center of Experimental Orthopedics, Saarland University , Homburg, Germany .,2 Department of Orthopedic Surgery, Saarland University Medical Center , Homburg, Germany
| |
Collapse
|
93
|
Shea CA, Rolfe RA, Murphy P. The importance of foetal movement for co-ordinated cartilage and bone development in utero : clinical consequences and potential for therapy. Bone Joint Res 2015; 4:105-16. [PMID: 26142413 PMCID: PMC4602203 DOI: 10.1302/2046-3758.47.2000387] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Construction of a functional skeleton is accomplished
through co-ordination of the developmental processes of chondrogenesis,
osteogenesis, and synovial joint formation. Infants whose movement in
utero is reduced or restricted and who subsequently suffer
from joint dysplasia (including joint contractures) and thin hypo-mineralised
bones, demonstrate that embryonic movement is crucial for appropriate
skeletogenesis. This has been confirmed in mouse, chick, and zebrafish
animal models, where reduced or eliminated movement consistently yields
similar malformations and which provide the possibility of experimentation
to uncover the precise disturbances and the mechanisms by which
movement impacts molecular regulation. Molecular genetic studies have
shown the important roles played by cell communication signalling
pathways, namely Wnt, Hedgehog, and transforming growth factor-beta/bone
morphogenetic protein. These pathways regulate cell behaviours such
as proliferation and differentiation to control maturation of the
skeletal elements, and are affected when movement is altered. Cell
contacts to the extra-cellular matrix as well as the cytoskeleton
offer a means of mechanotransduction which could integrate mechanical
cues with genetic regulation. Indeed, expression of cytoskeletal
genes has been shown to be affected by immobilisation. In addition
to furthering our understanding of a fundamental aspect of cell control
and differentiation during development, research in this area is
applicable to the engineering of stable skeletal tissues from stem
cells, which relies on an understanding of developmental mechanisms
including genetic and physical criteria. A deeper understanding
of how movement affects skeletogenesis therefore has broader implications
for regenerative therapeutics for injury or disease, as well as
for optimisation of physical therapy regimes for individuals affected
by skeletal abnormalities. Cite this article: Bone Joint Res 2015;4:105–116
Collapse
Affiliation(s)
- C A Shea
- Trinity College Dublin, College Green, Dublin, D2, Ireland
| | | | - P Murphy
- Trinity College Dublin, College Green, Dublin, D2, Ireland
| |
Collapse
|
94
|
Neumann S. WITHDRAWN: New hypothesis on the pathogenesis of osteoarthritis. Med Hypotheses 2015:S0306-9877(15)00252-2. [PMID: 26163059 DOI: 10.1016/j.mehy.2015.06.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 06/23/2015] [Accepted: 06/27/2015] [Indexed: 11/25/2022]
Abstract
This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Stephan Neumann
- Institute of Veterinary Medicine, University of Goettingen, Burckhardtweg 2, D-37077 Goettingen, Germany.
| |
Collapse
|
95
|
Tsang KY, Tsang SW, Chan D, Cheah KSE. The chondrocytic journey in endochondral bone growth and skeletal dysplasia. ACTA ACUST UNITED AC 2015; 102:52-73. [PMID: 24677723 DOI: 10.1002/bdrc.21060] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2014] [Accepted: 02/23/2014] [Indexed: 12/29/2022]
Abstract
The endochondral bones of the skeleton develop from a cartilage template and grow via a process involving a cascade of chondrocyte differentiation steps culminating in formation of a growth plate and the replacement of cartilage by bone. This process of endochondral ossification, driven by the generation of chondrocytes and their subsequent proliferation, differentiation, and production of extracellular matrix constitute a journey, deviation from which inevitably disrupts bone growth and development, and is the basis of human skeletal dysplasias with a wide range of phenotypic severity, from perinatal lethality to progressively deforming. This highly coordinated journey of chondrocyte specification and fate determination is controlled by a myriad of intrinsic and extrinsic factors. SOX9 is the master transcription factor that, in concert with varying partners along the way, directs the different phases of the journey from mesenchymal condensation, chondrogenesis, differentiation, proliferation, and maturation. Extracellular signals, including bone morphogenetic proteins, wingless-related MMTV integration site (WNT), fibroblast growth factor, Indian hedgehog, and parathyroid hormone-related peptide, are all indispensable for growth plate chondrocytes to align and organize into the appropriate columnar architecture and controls their maturation and transition to hypertrophy. Chondrocyte hypertrophy, marked by dramatic volume increase in phases, is controlled by transcription factors SOX9, Runt-related transcription factor, and FOXA2. Hypertrophic chondrocytes mediate the cartilage to bone transition and concomitantly face a live-or-die situation, a subject of much debate. We review recent insights into the coordination of the phases of the chondrocyte journey, and highlight the need for a systems level understanding of the regulatory networks that will facilitate the development of therapeutic approaches for skeletal dysplasia.
Collapse
Affiliation(s)
- Kwok Yeung Tsang
- Department of Biochemistry, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong, China
| | | | | | | |
Collapse
|
96
|
Wang W, Rigueur D, Lyons KM. TGFβ signaling in cartilage development and maintenance. ACTA ACUST UNITED AC 2015; 102:37-51. [PMID: 24677722 DOI: 10.1002/bdrc.21058] [Citation(s) in RCA: 198] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2013] [Accepted: 01/16/2014] [Indexed: 12/18/2022]
Abstract
Members of the transforming growth factor beta (TGFβ) superfamily of secreted factors play essential roles in nearly every aspect of cartilage formation and maintenance. However, the mechanisms by which TGFβs transduce their effects in cartilage in vivo remain poorly understood. Mutations in several TGFβ family members, their receptors, extracellular modulators, and intracellular transducers have been described, and these usually impact the development of the cartilaginous skeleton. Furthermore, genome-wide association studies have linked components of the (TGFβ) superfamily to susceptibility to osteoarthritis. This review focuses on recent discoveries from genetic studies in the mouse regarding the regulation of TGFβ signaling in developing growth plate and articular cartilage, as well as the different modes of crosstalk between canonical and noncanonical TGFβ signaling. These new insights into TGFβ signaling in cartilage may open new prospects for therapies that maintain healthy articular cartilage.
Collapse
Affiliation(s)
- Weiguang Wang
- Department of Orthopaedic Surgery and Orthopaedic Institute for Children, David Geffen School of Medicine, University of California, Los Angeles, California, 90095
| | | | | |
Collapse
|
97
|
Generation of articular chondrocytes from human pluripotent stem cells. Nat Biotechnol 2015; 33:638-45. [PMID: 25961409 DOI: 10.1038/nbt.3210] [Citation(s) in RCA: 144] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 03/25/2015] [Indexed: 12/19/2022]
Abstract
The replacement of articular cartilage through transplantation of chondrogenic cells or preformed cartilage tissue represents a potential new avenue for the treatment of degenerative joint diseases. Although many studies have described differentiation of human pluripotent stem cells (hPSCs) to the chondrogenic lineage, the generation of chondrocytes able to produce stable articular cartilage in vivo has not been demonstrated. Here we show that activation of the TGFβ pathway in hPSC-derived chondrogenic progenitors promotes the efficient development of articular chondrocytes that can form stable cartilage tissue in vitro and in vivo. In contrast, chondrocytes specified by BMP4 signaling display characteristics of hypertrophy and give rise to cartilage tissues that initiate the endochondral ossification process in vivo. These findings provide a simple serum-free and efficient approach for the routine generation of hPSC-derived articular chondrocytes for modeling diseases of the joint and developing cell therapy approaches to treat them.
Collapse
|
98
|
Tekari A, Luginbuehl R, Hofstetter W, Egli RJ. Transforming growth factor beta signaling is essential for the autonomous formation of cartilage-like tissue by expanded chondrocytes. PLoS One 2015; 10:e0120857. [PMID: 25775021 PMCID: PMC4361600 DOI: 10.1371/journal.pone.0120857] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Accepted: 01/27/2015] [Indexed: 02/07/2023] Open
Abstract
Cartilage is a tissue with limited self-healing potential. Hence, cartilage defects require surgical attention to prevent or postpone the development of osteoarthritis. For cell-based cartilage repair strategies, in particular autologous chondrocyte implantation, articular chondrocytes are isolated from cartilage and expanded in vitro to increase the number of cells required for therapy. During expansion, the cells lose the competence to autonomously form a cartilage-like tissue, that is in the absence of exogenously added chondrogenic growth factors, such as TGF-βs. We hypothesized that signaling elicited by autocrine and/or paracrine TGF-β is essential for the formation of cartilage-like tissue and that alterations within the TGF-β signaling pathway during expansion interfere with this process. Primary bovine articular chondrocytes were harvested and expanded in monolayer culture up to passage six and the formation of cartilage tissue was investigated in high density pellet cultures grown for three weeks. Chondrocytes expanded for up to three passages maintained the potential for autonomous cartilage-like tissue formation. After three passages, however, exogenous TGF-β1 was required to induce the formation of cartilage-like tissue. When TGF-β signaling was blocked by inhibiting the TGF-β receptor 1 kinase, the autonomous formation of cartilage-like tissue was abrogated. At the initiation of pellet culture, chondrocytes from passage three and later showed levels of transcripts coding for TGF-β receptors 1 and 2 and TGF-β2 to be three-, five- and five-fold decreased, respectively, as compared to primary chondrocytes. In conclusion, the autonomous formation of cartilage-like tissue by expanded chondrocytes is dependent on signaling induced by autocrine and/or paracrine TGF-β. We propose that a decrease in the expression of the chondrogenic growth factor TGF-β2 and of the TGF-β receptors in expanded chondrocytes accounts for a decrease in the activity of the TGF-β signaling pathway and hence for the loss of the potential for autonomous cartilage-like tissue formation.
Collapse
Affiliation(s)
- Adel Tekari
- Group for Bone Biology & Orthopaedic Research, Department Clinical Research, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | | | - Willy Hofstetter
- Group for Bone Biology & Orthopaedic Research, Department Clinical Research, University of Bern, Bern, Switzerland
| | - Rainer J. Egli
- Group for Bone Biology & Orthopaedic Research, Department Clinical Research, University of Bern, Bern, Switzerland
- RMS Foundation, Bettlach, Switzerland
| |
Collapse
|
99
|
Longobardi L, Li T, Tagliafierro L, Temple JD, Willcockson HH, Ye P, Esposito A, Xu F, Spagnoli A. Synovial joints: from development to homeostasis. Curr Osteoporos Rep 2015; 13:41-51. [PMID: 25431159 PMCID: PMC4306636 DOI: 10.1007/s11914-014-0247-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Synovial joint morphogenesis occurs through the condensation of mesenchymal cells into a non-cartilaginous region known as the interzone and the specification of progenitor cells that commit to the articular fate. Although several signaling molecules are expressed by the interzone, the mechanism is poorly understood. For treatments of cartilage injuries, it is critical to discover the presence of joint progenitor cells in adult tissues and their expression gene pattern. Potential stem cell niches have been found in different joint regions, such as the surface zone of articular cartilage, synovium, and groove of Ranvier. Inherited joint malformations as well as joint-degenerating conditions are often associated with other skeletal defects and may be seen as the failure of morphogenic factors to establish the correct microenvironment in cartilage and bone. Therefore, exploring how joints form can help us understand how cartilage and bone are damaged and develop drugs to reactivate this developing mechanism.
Collapse
Affiliation(s)
- Lara Longobardi
- Department of Pediatrics, University of North Carolina at Chapel Hill, 109 Mason Farm Road, Chapel Hill, NC, 27599-7039, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
100
|
Abstract
Osteoarthritis (OA) is a common degenerative joint disease, the pathological mechanism of which is currently unknown. Genetic alteration is one of the key contributing factors for OA pathology. Recent evidence suggests that epigenetic and microRNA regulation of critical genes may contribute to OA development. In this article, we review the epigenetic and microRNA regulations of genes related to OA development. Potential therapeutic strategies may be developed on the basis of novel findings.
Collapse
Affiliation(s)
- Di Chen
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jie Shen
- Department of Orthopedic Surgery, Washington University, St. Louis, MO, 63110, USA
| | - Tianqian Hui
- Department of Biochemistry, Rush University Medical Center, Chicago, IL, 60612, USA
| |
Collapse
|