1
|
Posey KL. Curcumin and Resveratrol: Nutraceuticals with so Much Potential for Pseudoachondroplasia and Other ER-Stress Conditions. Biomolecules 2024; 14:154. [PMID: 38397390 PMCID: PMC10886985 DOI: 10.3390/biom14020154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/25/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Natural products with health benefits, nutraceuticals, have shown considerable promise in many studies; however, this potential has yet to translate into widespread clinical use for any condition. Notably, many drugs currently on the market, including the first analgesic aspirin, are derived from plant extracts, emphasizing the historical significance of natural products in drug development. Curcumin and resveratrol, well-studied nutraceuticals, have excellent safety profiles with relatively mild side effects. Their long history of safe use and the natural origins of numerous drugs contrast with the unfavorable reputation associated with nutraceuticals. This review aims to explore the nutraceutical potential for treating pseudoachondroplasia, a rare dwarfing condition, by relating the mechanisms of action of curcumin and resveratrol to molecular pathology. Specifically, we will examine the curcumin and resveratrol mechanisms of action related to endoplasmic reticulum stress, inflammation, oxidative stress, cartilage health, and pain. Additionally, the barriers to the effective use of nutraceuticals will be discussed. These challenges include poor bioavailability, variations in content and purity that lead to inconsistent results in clinical trials, as well as prevailing perceptions among both the public and medical professionals. Addressing these hurdles is crucial to realizing the full therapeutic potential of nutraceuticals in the context of pseudoachondroplasia and other health conditions that might benefit.
Collapse
Affiliation(s)
- Karen L Posey
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| |
Collapse
|
2
|
Hecht JT, Veerisetty AC, Patra D, Hossain MG, Chiu F, Mobed C, Gannon FH, Posey KL. Early Resveratrol Treatment Mitigates Joint Degeneration and Dampens Pain in a Mouse Model of Pseudoachondroplasia (PSACH). Biomolecules 2023; 13:1553. [PMID: 37892235 PMCID: PMC10605626 DOI: 10.3390/biom13101553] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/11/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
Pseudoachondroplasia (PSACH), a severe dwarfing condition associated with early-onset joint degeneration and lifelong joint pain, is caused by mutations in cartilage oligomeric matrix protein (COMP). The mechanisms underlying the mutant-COMP pathology have been defined using the MT-COMP mouse model of PSACH that has the common D469del mutation. Mutant-COMP protein does not fold properly, and it is retained in the rough endoplasmic reticulum (rER) of chondrocytes rather than being exported to the extracellular matrix (ECM), driving ER stress that stimulates oxidative stress and inflammation, driving a self-perpetuating cycle. CHOP (ER stress signaling protein) and TNFα inflammation drive high levels of mTORC1 signaling, shutting down autophagy and blocking ER clearance, resulting in premature loss of chondrocytes that negatively impacts linear growth and causes early joint degeneration in MT-COMP mice and PSACH. Previously, we have shown that resveratrol treatment from birth to 20 weeks prevents joint degeneration and decreases the pathological processes in articular chondrocytes. Resveratrol's therapeutic mechanism of action in the mutant-COMP pathology was shown to act by primarily stimulating autophagy and reducing inflammation. Importantly, we demonstrated that MT-COMP mice experience pain consistent with PSACH joint pain. Here, we show, in the MT-COMP mouse, that resveratrol treatment must begin within 4 weeks to preserve joint health and reduce pain. Resveratrol treatment started at 6 or 8 weeks (to 20 weeks) was not effective in preventing joint degeneration. Collectively, our findings in MT-COMP mice show that there is a postnatal resveratrol treatment window wherein the inevitable mutant-COMP joint degeneration and pain can be prevented.
Collapse
Affiliation(s)
- Jacqueline T. Hecht
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Alka C. Veerisetty
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Debabrata Patra
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA;
| | - Mohammad G. Hossain
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Frankie Chiu
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| | - Claire Mobed
- Department of Biology, Rice University, Houston, TX 77005, USA;
| | - Francis H. Gannon
- Departments of Pathology and Immunology and Orthopedic Surgery, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Karen L. Posey
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA; (J.T.H.); (A.C.V.); (M.G.H.); (F.C.)
| |
Collapse
|
3
|
Sheng R, Chen J, Wang H, Luo Y, Liu J, Chen Z, Mo Q, Chi J, Ling C, Tan X, Yao Q, Zhang W. Nanosilicate-Reinforced Silk Fibroin Hydrogel for Endogenous Regeneration of Both Cartilage and Subchondral Bone. Adv Healthc Mater 2022; 11:e2200602. [PMID: 35749970 DOI: 10.1002/adhm.202200602] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 06/10/2022] [Indexed: 01/27/2023]
Abstract
Osteochondral defects are characterized by injuries to both cartilage and subchondral bone, which is a result of trauma, inflammation, or inappropriate loading. Due to the unique biological properties of subchondral bone and cartilage, developing a tissue engineering scaffold that can promote dual-lineage regeneration of cartilage and bone simultaneously remains a great challenge. In this study, a microporous nanosilicate-reinforced enzymatically crosslinked silk fibroin (SF) hydrogel is fabricated by introducing montmorillonite (MMT) nanoparticles via intercalation chemistry. In vitro studies show that SF-MMT nanocomposite hydrogel has improved mechanical properties and hydrophilicity, as well as the bioactivities to promote the osteogenic differentiation of bone marrow mesenchymal stem cells and maintain chondrocyte phenotype compared with SF hydrogel. Global proteomic analysis verifies the dual-lineage bioactivities of SF-MMT nanocomposite hydrogel, which are probably regulated by multiple signaling pathways. Furthermore, it is observed that the biophysical interaction of cells and SF-MMT nanocomposite hydrogel is partially mediated by clathrin-mediated endocytosis and its downstream processes. In vivo, the SF-MMT nanocomposite hydrogel effectively promotes osteochondral regeneration as evidenced by macroscopic, micro-CT, and histological evaluation. In conclusion, a functionalized SF-MMT nanocomposite hydrogel is developed with dual-lineage bioactivity for osteochondral regeneration, indicating its potential in osteochondral tissue engineering.
Collapse
Affiliation(s)
- Renwang Sheng
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jialin Chen
- School of Medicine, Southeast University, Nanjing, 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| | - Hongmei Wang
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Yifan Luo
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jia Liu
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Zhixuan Chen
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Qingyun Mo
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Jiayu Chi
- School of Medicine, Southeast University, Nanjing, 210009, China
| | - Chen Ling
- Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Xin Tan
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Qingqiang Yao
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China.,Department of Orthopaedic Surgery, Institute of Digital Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Wei Zhang
- School of Medicine, Southeast University, Nanjing, 210009, China.,Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210096, China.,China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310058, China
| |
Collapse
|
4
|
Application of Alginate Hydrogels for Next-Generation Articular Cartilage Regeneration. Int J Mol Sci 2022; 23:ijms23031147. [PMID: 35163071 PMCID: PMC8835677 DOI: 10.3390/ijms23031147] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/28/2022] Open
Abstract
The articular cartilage has insufficient intrinsic healing abilities, and articular cartilage injuries often progress to osteoarthritis. Alginate-based scaffolds are attractive biomaterials for cartilage repair and regeneration, allowing for the delivery of cells and therapeutic drugs and gene sequences. In light of the heterogeneity of findings reporting the benefits of using alginate for cartilage regeneration, a better understanding of alginate-based systems is needed in order to improve the approaches aiming to enhance cartilage regeneration with this compound. This review provides an in-depth evaluation of the literature, focusing on the manipulation of alginate as a tool to support the processes involved in cartilage healing in order to demonstrate how such a material, used as a direct compound or combined with cell and gene therapy and with scaffold-guided gene transfer procedures, may assist cartilage regeneration in an optimal manner for future applications in patients.
Collapse
|
5
|
Andrés Sastre E, Maly K, Zhu M, Witte-Bouma J, Trompet D, Böhm AM, Brachvogel B, van Nieuwenhoven CA, Maes C, van Osch GJVM, Zaucke F, Farrell E. Spatiotemporal distribution of thrombospondin-4 and -5 in cartilage during endochondral bone formation and repair. Bone 2021; 150:115999. [PMID: 33971315 DOI: 10.1016/j.bone.2021.115999] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 12/12/2022]
Abstract
During skeletal development most bones are first formed as cartilage templates, which are gradually replaced by bone. If later in life those bones break, temporary cartilage structures emerge to bridge the fractured ends, guiding the regenerative process. This bone formation process, known as endochondral ossification (EO), has been widely studied for its potential to reveal factors that might be used to treat patients with large bone defects. The extracellular matrix of cartilage consists of different types of collagens, proteoglycans and a variety of non-collagenous proteins that organise the collagen fibers in complex networks. Thrombospondin-5, also known as cartilage oligomeric matrix protein (TSP-5/COMP) is abundant in cartilage, where it has been described to enhance collagen fibrillogenesis and to interact with a variety of growth factors, matrix proteins and cellular receptors. However, very little is known about the skeletal distribution of its homologue thrombospondin-4 (TSP-4). In our study, we compared the spatiotemporal expression of TSP-5 and TSP-4 during postnatal bone formation and fracture healing. Our results indicate that in both these settings, TSP-5 distributes across all layers of the transient cartilage, while the localisation of TSP-4 is restricted to the population of hypertrophic chondrocytes. Furthermore, in fractured bones we observed TSP-4 sparsely distributed in the periosteum, while TSP-5 was absent. Last, we analysed the chemoattractant effects of the two proteins on endothelial cells and bone marrow stem cells and hypothesised that, of the two thrombospondins, only TSP-4 might promote blood vessel invasion during ossification. We conclude that TSP-4 is a novel factor involved in bone formation. These findings reveal TSP-4 as an attractive candidate to be evaluated for bone tissue engineering purposes.
Collapse
Affiliation(s)
- E Andrés Sastre
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - K Maly
- Dr. Rolf Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - M Zhu
- Center for Biochemistry, Faculty of Medicine, University of Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Germany
| | - J Witte-Bouma
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands
| | - D Trompet
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Belgium
| | - A M Böhm
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Belgium
| | - B Brachvogel
- Center for Biochemistry, Faculty of Medicine, University of Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Germany
| | - C A van Nieuwenhoven
- Department of Plastic, Reconstructive and Hand Surgery, Erasmus MC-Sophia, University Medical Center, Rotterdam, the Netherlands
| | - C Maes
- Laboratory of Skeletal Cell Biology and Physiology (SCEBP), Skeletal Biology and Engineering Research Center (SBE), Department of Development and Regeneration, KU Leuven, Belgium
| | - G J V M van Osch
- Department of Orthopaedics and Sports Medicine, Erasmus MC University Medical Center, Rotterdam, the Netherlands; Department of Otorhinolaryngology, Erasmus MC University Medical Center, Rotterdam, the Netherlands
| | - F Zaucke
- Dr. Rolf Schwiete Research Unit for Osteoarthritis, Orthopaedic University Hospital Friedrichsheim, Frankfurt, Germany
| | - E Farrell
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, the Netherlands.
| |
Collapse
|
6
|
Joint Degeneration in a Mouse Model of Pseudoachondroplasia: ER Stress, Inflammation, and Block of Autophagy. Int J Mol Sci 2021; 22:ijms22179239. [PMID: 34502142 PMCID: PMC8431545 DOI: 10.3390/ijms22179239] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/05/2021] [Accepted: 08/18/2021] [Indexed: 01/05/2023] Open
Abstract
Pseudoachondroplasia (PSACH), a short limb skeletal dysplasia associated with premature joint degeneration, is caused by misfolding mutations in cartilage oligomeric matrix protein (COMP). Here, we define mutant-COMP-induced stress mechanisms that occur in articular chondrocytes of MT-COMP mice, a murine model of PSACH. The accumulation of mutant-COMP in the ER occurred early in MT-COMP articular chondrocytes and stimulated inflammation (TNFα) at 4 weeks, and articular chondrocyte death increased at 8 weeks while ER stress through CHOP was elevated by 12 weeks. Importantly, blockage of autophagy (pS6), the major mechanism that clears the ER, sustained cellular stress in MT-COMP articular chondrocytes. Degeneration of MT-COMP articular cartilage was similar to that observed in PSACH and was associated with increased MMPs, a family of degradative enzymes. Moreover, chronic cellular stresses stimulated senescence. Senescence-associated secretory phenotype (SASP) may play a role in generating and propagating a pro-degradative environment in the MT-COMP murine joint. The loss of CHOP or resveratrol treatment from birth preserved joint health in MT-COMP mice. Taken together, these results indicate that ER stress/CHOP signaling and autophagy blockage are central to mutant-COMP joint degeneration, and MT-COMP mice joint health can be preserved by decreasing articular chondrocyte stress. Future joint sparing therapeutics for PSACH may include resveratrol.
Collapse
|
7
|
Expression and Localization of Thrombospondins, Plastin 3, and STIM1 in Different Cartilage Compartments of the Osteoarthritic Varus Knee. Int J Mol Sci 2021; 22:ijms22063073. [PMID: 33802838 PMCID: PMC8002632 DOI: 10.3390/ijms22063073] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 02/08/2023] Open
Abstract
Osteoarthritis (OA) is a multifactorial disease which is characterized by a change in the homeostasis of the extracellular matrix (ECM). The ECM is essential for the function of the articular cartilage and plays an important role in cartilage mechanotransduction. To provide a better understanding of the interaction between the ECM and the actin cytoskeleton, we investigated the localization and expression of the Ca2+-dependent proteins cartilage oligomeric matrix protein (COMP), thrombospondin-1 (TSP-1), plastin 3 (PLS3) and stromal interaction molecule 1 (STIM1). We investigated 16 patients who suffered from varus knee OA and performed a topographical analysis of the cartilage from the medial and lateral compartment of the proximal tibial plateau. In a varus knee, OA is more pronounced in the medial compared to the lateral compartment as a result of an overloading due to the malalignment. We detected a location-dependent staining of PLS3 and STIM1 in the articular cartilage tissue. The staining intensity for both proteins correlated with the degree of cartilage degeneration. The staining intensity of TSP-1 was clearly reduced in the cartilage of the more affected medial compartment, an observation that was confirmed in cartilage extracts by immunoblotting. The total amount of COMP was unchanged; however, slight changes were detected in the localization of the protein. Our results provide novel information on alterations in OA cartilage suggesting that Ca2+-dependent mechanotransduction between the ECM and the actin cytoskeleton might play an essential role in the pathomechanism of OA.
Collapse
|
8
|
Tran V, Karsai A, Fong MC, Cai W, Fraley JG, Yik JHN, Klineberg E, Haudenschild DR, Liu GY. Direct Visualization of the Binding of Transforming Growth Factor Beta 1 with Cartilage Oligomeric Matrix Protein via High-Resolution Atomic Force Microscopy. J Phys Chem B 2020; 124:9497-9504. [PMID: 33052673 DOI: 10.1021/acs.jpcb.0c07286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
This work reports the first direct observations of binding and complex formation between transforming growth factor beta 1 (TGF-β1) and cartilage oligomeric matrix protein (COMP) using high-resolution atomic force microscopy (AFM). Each COMP molecule consists of pentamers whose five identical monomeric units bundle at N-termini. From this central point, the five monomers' flexible arms extend outward with C-terminal domains at the distal ends, forming a bouquet-like structure. In commonly used buffer solutions, TGF-β1 molecules typically form homodimers (majority), double dimers (minority), and aggregates (trace amount). Mixing TGF-β1 and COMP leads to rapid binding and complex formation. The TGF-β1/COMP complexes contain one to three COMP and multiple TGF-β1 molecules. For complexes with one COMP, the structure is more compact and less flexible than that of COMP alone. For complexes with two or more COMP molecules, the conformation varies to a large degree from one complex to another. This is attributed to the presence of double dimers or aggregates of TGF-β1 molecules, whose size and multiple binding sites enable binding to more than one COMP. The number and location of individual TGF-β1 dimers are also clearly visible in all complexes. This molecular-level information provides a new insight into the mechanism of chondrogenesis enhancement by TGF-β1/COMP complexes, i.e., simultaneous and multivalent presentation of growth factors. These presentations help explain the high efficacy in sustained activation of the signaling pathway to augment chondrogenesis.
Collapse
Affiliation(s)
- Victoria Tran
- Department of Chemistry, University of California, Davis, California 95616, United States
| | - Arpad Karsai
- Department of Chemistry, University of California, Davis, California 95616, United States
| | - Michael C Fong
- Department of Biomedical Engineering, University of California, Davis, California 95616, United States
| | - Weiliang Cai
- Department of Orthopaedic Surgery, University of California-Davis Medical Center, Sacramento, California 95817, United States
| | - J Gabriel Fraley
- Department of Orthopaedic Surgery, University of California-Davis Medical Center, Sacramento, California 95817, United States
| | - Jasper H N Yik
- Department of Orthopaedic Surgery, University of California-Davis Medical Center, Sacramento, California 95817, United States
| | - Eric Klineberg
- Department of Orthopaedic Surgery, University of California-Davis Medical Center, Sacramento, California 95817, United States
| | - Dominik R Haudenschild
- Department of Orthopaedic Surgery, University of California-Davis Medical Center, Sacramento, California 95817, United States
| | - Gang-Yu Liu
- Department of Chemistry, University of California, Davis, California 95616, United States
| |
Collapse
|
9
|
Caron MMJ, Janssen MPF, Peeters L, Haudenschild DR, Cremers A, Surtel DAM, van Rhijn LW, Emans PJ, Welting TJM. Aggrecan and COMP Improve Periosteal Chondrogenesis by Delaying Chondrocyte Hypertrophic Maturation. Front Bioeng Biotechnol 2020; 8:1036. [PMID: 32984292 PMCID: PMC7483497 DOI: 10.3389/fbioe.2020.01036] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Accepted: 08/11/2020] [Indexed: 12/29/2022] Open
Abstract
The generation of cartilage from progenitor cells for the purpose of cartilage repair is often hampered by hypertrophic differentiation of the engineered cartilaginous tissue caused by endochondral ossification. Since a healthy cartilage matrix contains high amounts of Aggrecan and COMP, we hypothesized that their supplementation in the biogel used in the generation of subperiosteal cartilage mimics the composition of the cartilage extracellular matrix environment, with beneficial properties for the engineered cartilage. Supplementation of COMP or Aggrecan was studied in vitro during chondrogenic differentiation of rabbit periosteum cells and periosteum-derived chondrocytes. Low melting agarose was supplemented with bovine Aggrecan, human recombinant COMP or vehicle and was injected between the bone and periosteum at the upper medial side of the tibia of New Zealand white rabbits. Generated subperiosteal cartilage tissue was analyzed for weight, GAG and DNA content and ALP activity. Key markers of different phases of endochondral ossification were measured by RT-qPCR. For the in vitro experiments, no significant differences in chondrogenic marker expression were detected following COMP or Aggrecan supplementation, while in vivo favorable chondrogenic marker expression was detected. Gene expression levels of hypertrophic markers as well as ALP activity were significantly decreased in the Aggrecan and COMP supplemented conditions compared to controls. The wet weight and GAG content of the in vivo generated subperiosteal cartilage tissue was not significantly different between groups. Data demonstrate the potential of Aggrecan and COMP to favorably influence the subperiosteal microenvironment for the in vivo generation of cartilage for the optimization of cartilage regenerative approaches.
Collapse
Affiliation(s)
- Marjolein M J Caron
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Maarten P F Janssen
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Laura Peeters
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Dominik R Haudenschild
- Department of Orthopedic Surgery, University of California Davis School of Medicine, Sacramento, CA, United States
| | - Andy Cremers
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Don A M Surtel
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Lodewijk W van Rhijn
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Pieter J Emans
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| | - Tim J M Welting
- Laboratory for Experimental Orthopedics, Department of Orthopedic Surgery, CAPHRI Care and Public Health Research Institute, Maastricht University Medical Center, Maastricht, Netherlands
| |
Collapse
|
10
|
Mouser VHM, Levato R, Mensinga A, Dhert WJA, Gawlitta D, Malda J. Bio-ink development for three-dimensional bioprinting of hetero-cellular cartilage constructs. Connect Tissue Res 2020; 61:137-151. [PMID: 30526130 DOI: 10.1080/03008207.2018.1553960] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Bioprinting is a promising tool to fabricate organized cartilage. This study aimed to investigate the printability of gelatin-methacryloyl/gellan gum (gelMA/gellan) hydrogels with and without methacrylated hyaluronic acid (HAMA), and to explore (zone-specific) chondrogenesis of chondrocytes, articular cartilage progenitor cells (ACPCs), and multipotent mesenchymal stromal cells (MSCs) embedded in these bio-inks.The incorporating of HAMA in gelMA/gellan bio-ink increased filament stability, as measured using a filament collapse assay, but did not influence (zone-specific) chondrogenesis of any of the cell types. Highest chondrogenic potential was observed for MSCs, followed by ACPCs, which displayed relatively high proteoglycan IV mRNA levels. Therefore, two-zone constructs were printed with gelMA/gellan/HAMA containing ACPCs in the superficial region and MSCs in the middle/deep region. Chondrogenic differentiation was confirmed, however, printing influence cellular differentiation.ACPC- and MSC-laden gelMA/gellan/HAMA hydrogels are of interest for the fabrication of cartilage constructs. Nevertheless, this study underscores the need for careful evaluation of the effects of printing on cellular differentiation.
Collapse
Affiliation(s)
- Vivian H M Mouser
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Anneloes Mensinga
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Wouter J A Dhert
- Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands.,Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
11
|
Posey KL, Coustry F, Veerisetty AC, Hossain MG, Gambello MJ, Hecht JT. Novel mTORC1 Mechanism Suggests Therapeutic Targets for COMPopathies. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:132-146. [PMID: 30553437 DOI: 10.1016/j.ajpath.2018.09.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Revised: 08/20/2018] [Accepted: 09/17/2018] [Indexed: 02/06/2023]
Abstract
Cartilage oligomeric matrix protein (COMP) is a large, multifunctional extracellular protein that, when mutated, is retained in the rough endoplasmic reticulum (ER). This retention elicits ER stress, inflammation, and oxidative stress, resulting in dysfunction and death of growth plate chondrocytes. While identifying the cellular pathologic mechanisms underlying the murine mutant (MT)-COMP model of pseudoachondroplasia, increased midline-1 (MID1) expression and mammalian target of rapamycin complex 1 (mTORC1) signaling was found. This novel role for MID1/mTORC1 signaling was investigated since treatments shown to repress the pathology also reduced Mid1/mTORC1. Although ER stress-inducing drugs or tumor necrosis factor α (TNFα) in rat chondrosarcoma cells increased Mid1, oxidative stress did not, establishing that ER stress- or TNFα-driven inflammation alone is sufficient to elevate MID1 expression. Since MID1 ubiquitinates protein phosphatase 2A (PP2A), a negative regulator of mTORC1, PP2A was evaluated in MT-COMP growth plate chondrocytes. PP2A was decreased, indicating de-repression of mTORC1 signaling. Rapamycin treatment in MT-COMP mice reduced mTORC1 signaling and intracellular retention of COMP, and increased proliferation, but did not change inflammatory markers IL-16 and eosinophil peroxidase. Lastly, mRNA from tuberous sclerosis-1/2-null mice brain tissue exhibiting ER stress had increased Mid1 expression, confirming the relationship between ER stress and MID1/mTORC1 signaling. These findings suggest a mechanistic link between ER stress and MID1/mTORC1 signaling that has implications extending to other conditions involving ER stress.
Collapse
Affiliation(s)
- Karen L Posey
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas.
| | - Francoise Coustry
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| | - Alka C Veerisetty
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| | - Mohammad G Hossain
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas
| | - Michael J Gambello
- Human Genetics and Pediatrics, Emory University School of Medicine, Atlanta, Georgia
| | - Jacqueline T Hecht
- Department of Pediatrics, McGovern Medical School, University of Texas Health Science Center, Houston, Texas; School of Dentistry, University of Texas Health Science Center, Houston, Texas
| |
Collapse
|
12
|
Tran V, Karsai A, Fong MC, Cai W, Yik JHN, Klineberg E, Haudenschild DR, Liu GY. Label-Free and Direct Visualization of Multivalent Binding of Bone Morphogenetic Protein-2 with Cartilage Oligomeric Matrix Protein. J Phys Chem B 2019; 123:39-46. [PMID: 30554512 DOI: 10.1021/acs.jpcb.8b08564] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
This work presents the first direct evidence of multivalent binding between bone morphogenetic protein-2 (BMP-2) and cartilage oligomeric matrix protein (COMP) using high-resolution atomic force microscopy (AFM) imaging. AFM topographic images reveal the molecular morphology of COMP, a pentameric protein whose five identical monomer units bundle together at N-termini, extending out with flexible chains to C-termini. Upon addition of BMP-2, COMP molecules undergo conformational changes at the C-termini to enable binding with BMP-2 molecules. AFM enables local structural changes of COMP to be revealed upon binding various numbers, 1-5, of BMP-2 molecules. These BMP-2/COMP complexes exhibit very different morphologies from those of COMP: much more compact and thus less flexible. These molecular-level insights deepen current understanding of the mechanism of how the BMP-2/COMP complex enhances osteogenesis among osteoprogenitor cells, i.e., multivalent presentation of BMP-2 via the stable and relatively rigid BMP-2/COMP complex could form a lattice of interaction between multiple BMP-2 and BMP-2 receptors. These ligand-receptor clusters lead to fast initiation and sustained activation of the Smad signaling pathway, resulting in enhanced osteogenesis. This work is also of translational importance as the outcome may enable use of lower BMP-2 dosage for bone repair and regeneration.
Collapse
Affiliation(s)
- Victoria Tran
- Department of Chemistry , University of California , Davis , California 95616 , United States
| | - Arpad Karsai
- Department of Chemistry , University of California , Davis , California 95616 , United States
| | - Michael C Fong
- Department of Biomedical Engineering , University of California , Davis , California 95616 , United States
| | - Weiliang Cai
- Department of Orthopaedic Surgery , University of California-Davis Medical Center , Sacramento , California 95817 , United States
| | - Jasper H N Yik
- Department of Orthopaedic Surgery , University of California-Davis Medical Center , Sacramento , California 95817 , United States
| | - Eric Klineberg
- Department of Orthopaedic Surgery , University of California-Davis Medical Center , Sacramento , California 95817 , United States
| | - Dominik R Haudenschild
- Department of Orthopaedic Surgery , University of California-Davis Medical Center , Sacramento , California 95817 , United States
| | - Gang-Yu Liu
- Department of Chemistry , University of California , Davis , California 95616 , United States
| |
Collapse
|
13
|
Wiggenhauser PS, Schwarz S, Rotter N. The distribution patterns of COMP and matrilin-3 in septal, alar and triangular cartilages of the human nose. Histochem Cell Biol 2018; 150:291-300. [PMID: 29721643 DOI: 10.1007/s00418-018-1672-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2018] [Indexed: 01/07/2023]
Abstract
The biomechanical characteristics of septal cartilage depend strongly on the distinct extracellular matrix of cartilage tissue; therefore, it is essential that the components of this matrix are identified and understood. Cartilage oligomeric matrix protein (COMP) and matrilin-3 are localised in articular cartilage. This study was the first to examine all subtypes of mature human nasal cartilages (alar, triangular and septal) with specific attention to the distribution of COMP and matrilin-3. Three whole fresh-frozen noses from human donors were dissected, and exemplary biopsies were examined using histochemical staining (haematoxylin and eosin and Alcian blue) and immunohistochemistry (collagen II, COMP and matrilin-3). The following three zones within the nasal cartilage were identified: superficial, intermediate and central. COMP was detected as highest in the intermediate zones in all three subtypes of nasal cartilage, whereas matrilin-3 was detected with pericellular deposition mainly within septal cartilage predominantly in the superficial zones. The distinct staining patterns of COMP and matrilin-3 underscore the different functional roles of both proteins in nasal cartilage. According to the literature, COMP might be involved with collagen II in the formation of networks, whereas matrilin-3 is reported to prevent ossification or regulate mechanosensitivity. The predominant staining observed in septal cartilage suggests matrilin-3's modulatory role because of its presence in the osteochondral junctional zone and given that the biomechanical load in septal cartilage is different from that in alar or triangular cartilage. In conclusion, COMP and matrilin-3 were detected in mature human nasal cartilage but displayed different staining patterns that might be explained by the functional roles of the respective matrix protein; however, further research is necessary to identify and define the functional aspects of this morphological difference.
Collapse
Affiliation(s)
- Paul Severin Wiggenhauser
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany.
- Department of Hand, Plastic and Aesthetic Surgery, University Hospital, Ludwig-Maximilians University, Pettenkoferstr. 8a, 80336, Munich, Germany.
| | - Silke Schwarz
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
- Department of Anatomy, Paracelsus Medical University, Prof. Ernst Nathan Str. 1, Salzburg, 90419, Nuremberg, Germany
| | - Nicole Rotter
- Department of Oto-Rhino-Laryngology, Head and Neck Surgery, Ulm University Medical Center, Frauensteige 12, 89075, Ulm, Germany
- Department of Oto-Rhino-Laryngology, University Hospital Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| |
Collapse
|
14
|
Hsueh MF, Kraus VB, Önnerfjord P. Cartilage matrix remodelling differs by disease state and joint type. Eur Cell Mater 2017; 34:70-82. [PMID: 28836259 PMCID: PMC5599932 DOI: 10.22203/ecm.v034a05] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Dramatic alterations in mechanical properties have been documented for osteoarthritic (OA) cartilage. However, the matrix composition underlying these changes has not been mapped and their aetiology is not entirely understood. We hypothesised that an understanding of the cartilage matrix heterogeneity could provide insights into the origin of these OA-related alterations. We generated serial transverse cryo sections for 7 different cartilage conditions: 2 joint sites (knee and hip), 2 disease states (healthy and OA) and 3 tissue depths (superficial, middle and deep). By laser capture microscopy, we acquired ~200 cartilage matrix specimens from territorial (T) and interterritorial (IT) regions for all 7 conditions. A standardised matrix area was collected for each condition for a total of 0.02 ± 0.001 mm3 (corresponding to 20 µg of tissue) from a total of 4800 specimens. Extracted proteins were analysed for abundance by targeted proteomics. For most proteins, a lower IT/T ratio was observed for the OA disease state and knee joint type. A major cause of the altered IT/T ratios was the decreased protein abundance in IT regions. The collagenase-derived type III collagen neo-epitope, indicative of collagen proteolysis, was significantly more abundant in OA cartilage. In addition, it was enriched on average of 1.45-fold in IT relative to T matrix. These results were consistent with an elevated proteolysis in IT regions of OA cartilage, due to degenerative influences originating from synovial tissue and/or produced locally by chondrocytes. In addition, they offered direct evidence for dynamic remodelling of cartilage and provided a cogent biochemical template for understanding the alterations of matrix mechanical properties.
Collapse
Affiliation(s)
- Ming-Feng Hsueh
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC 27701
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC 27701,Department of Medicine, Duke University School of Medicine, Duke University, Durham, NC 27701
| | - Patrik Önnerfjord
- Department of Clinical Sciences Lund, Section of Rheumatology and Molecular Skeletal Biology Center of Excellence in Biological and Medical Mass Spectrometry, Lund University, Lund, Sweden
| |
Collapse
|
15
|
Posey KL, Coustry F, Veerisetty AC, Hossain M, Gattis D, Booten S, Alcorn JL, Seth PP, Hecht JT. Antisense Reduction of Mutant COMP Reduces Growth Plate Chondrocyte Pathology. Mol Ther 2017; 25:705-714. [PMID: 28162960 DOI: 10.1016/j.ymthe.2016.12.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 11/28/2016] [Accepted: 12/25/2016] [Indexed: 01/25/2023] Open
Abstract
Mutations in cartilage oligomeric matrix protein cause pseudoachondroplasia, a severe disproportionate short stature disorder. Mutant cartilage oligomeric matrix protein produces massive intracellular retention of cartilage oligomeric matrix protein, stimulating ER and oxidative stresses and inflammation, culminating in post-natal loss of growth plate chondrocytes, which compromises linear bone growth. Treatments for pseudoachondroplasia are limited because cartilage is relatively avascular and considered inaccessible. Here we report successful delivery and treatment using antisense oligonucleotide technology in our transgenic pseudoachondroplasia mouse model. We demonstrate delivery of human cartilage oligomeric matrix protein-specific antisense oligonucleotides to cartilage and reduction of cartilage oligomeric matrix protein expression, which largely alleviates pseudoachondroplasia growth plate chondrocyte pathology. One antisense oligonucleotide reduced steady-state levels of cartilage oligomeric matrix protein mRNA and dampened intracellular retention of mutant cartilage oligomeric matrix protein, leading to a reduction of inflammatory markers and cell death and partial restoration of proliferation. This novel and exciting work demonstrates that antisense-based therapy is a viable approach for treating pseudoachondroplasia and other human cartilage disorders.
Collapse
Affiliation(s)
- Karen L Posey
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA.
| | - Francoise Coustry
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Alka C Veerisetty
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Mohammad Hossain
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Danielle Gattis
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Sheri Booten
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Joseph L Alcorn
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| | - Punit P Seth
- Department of Antisense Drug Discovery, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Jacqueline T Hecht
- Department of Pediatrics, McGovern Medical School at The University of Texas Health Science Center at Houston (UTHealth), Houston, TX 77030, USA
| |
Collapse
|
16
|
Guo T, Lembong J, Zhang LG, Fisher JP. Three-Dimensional Printing Articular Cartilage: Recapitulating the Complexity of Native Tissue<sup/>. TISSUE ENGINEERING PART B-REVIEWS 2016; 23:225-236. [PMID: 27875945 DOI: 10.1089/ten.teb.2016.0316] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
In the past few decades, the field of tissue engineering combined with rapid prototyping (RP) techniques has been successful in creating biological substitutes that mimic tissues. Its applications in regenerative medicine have drawn efforts in research from various scientific fields, diagnostics, and clinical translation to therapies. While some areas of therapeutics are well developed, such as skin replacement, many others such as cartilage repair can still greatly benefit from tissue engineering and RP due to the low success and/or inefficiency of current existing, often surgical treatments. Through fabrication of complex scaffolds and development of advanced materials, RP provides a new avenue for cartilage repair. Computer-aided design and three-dimensional (3D) printing allow the fabrication of modeled cartilage scaffolds for repair and regeneration of damaged cartilage tissues. Specifically, the various processes of 3D printing will be discussed in details, both cellular and acellular techniques, covering the different materials, geometries, and operational printing conditions for the development of tissue-engineered articular cartilage. Finally, we conclude with some insights on future applications and challenges related to this technology, especially using 3D printing techniques to recapitulate the complexity of native structure for advanced cartilage regeneration.
Collapse
Affiliation(s)
- Ting Guo
- 1 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | - Josephine Lembong
- 1 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| | - Lijie Grace Zhang
- 2 Department of Mechanical and Aerospace Engineering, The George Washington University , Washington, District of Columbia
| | - John P Fisher
- 1 Fischell Department of Bioengineering, University of Maryland , College Park, Maryland
| |
Collapse
|
17
|
Hsueh MF, Khabut A, Kjellström S, Önnerfjord P, Kraus VB. Elucidating the Molecular Composition of Cartilage by Proteomics. J Proteome Res 2016; 15:374-88. [PMID: 26632656 DOI: 10.1021/acs.jproteome.5b00946] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Articular cartilage consists of chondrocytes and two major components, a collagen-rich framework and highly abundant proteoglycans. Most prior studies defining the zonal distribution of cartilage have extracted proteins with guanidine-HCl. However, an unextracted collagen-rich residual is left after extraction. In addition, the high abundance of anionic polysaccharide molecules extracted from cartilage adversely affects the chromatographic separation. In this study, we established a method for removing chondrocytes from cartilage sections with minimal extracellular matrix protein loss. The addition of surfactant to guanidine-HCl extraction buffer improved protein solubility. Ultrafiltration removed interference from polysaccharides and salts. Almost four-times more collagen peptides were extracted by the in situ trypsin digestion method. However, as expected, proteoglycans were more abundant within the guanidine-HCl extraction. These different methods were used to extract cartilage sections from different cartilage layers (superficial, intermediate, and deep), joint types (knee and hip), and disease states (healthy and osteoarthritic), and the extractions were evaluated by quantitative and qualitative proteomic analyses. The results of this study led to the identifications of the potential biomarkers of osteoarthritis (OA), OA progression, and the joint specific biomarkers.
Collapse
Affiliation(s)
- Ming-Feng Hsueh
- Duke Molecular Physiology Institute, ‡Departments of Medicine, and §Pathology, Duke University School of Medicine, Duke University , Durham, North Carolina 27701, United States.,Department of Clinical Sciences Lund, Section of Rheumatology and Molecular Skeletal Biology and ¶Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University , SE 22184 Lund, Sweden
| | - Areej Khabut
- Duke Molecular Physiology Institute, ‡Departments of Medicine, and §Pathology, Duke University School of Medicine, Duke University , Durham, North Carolina 27701, United States.,Department of Clinical Sciences Lund, Section of Rheumatology and Molecular Skeletal Biology and ¶Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University , SE 22184 Lund, Sweden
| | - Sven Kjellström
- Duke Molecular Physiology Institute, ‡Departments of Medicine, and §Pathology, Duke University School of Medicine, Duke University , Durham, North Carolina 27701, United States.,Department of Clinical Sciences Lund, Section of Rheumatology and Molecular Skeletal Biology and ¶Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University , SE 22184 Lund, Sweden
| | - Patrik Önnerfjord
- Duke Molecular Physiology Institute, ‡Departments of Medicine, and §Pathology, Duke University School of Medicine, Duke University , Durham, North Carolina 27701, United States.,Department of Clinical Sciences Lund, Section of Rheumatology and Molecular Skeletal Biology and ¶Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University , SE 22184 Lund, Sweden
| | - Virginia Byers Kraus
- Duke Molecular Physiology Institute, ‡Departments of Medicine, and §Pathology, Duke University School of Medicine, Duke University , Durham, North Carolina 27701, United States.,Department of Clinical Sciences Lund, Section of Rheumatology and Molecular Skeletal Biology and ¶Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University , SE 22184 Lund, Sweden
| |
Collapse
|
18
|
Di Bella C, Fosang A, Donati DM, Wallace GG, Choong PFM. 3D Bioprinting of Cartilage for Orthopedic Surgeons: Reading between the Lines. Front Surg 2015; 2:39. [PMID: 26322314 PMCID: PMC4534805 DOI: 10.3389/fsurg.2015.00039] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Accepted: 07/31/2015] [Indexed: 12/14/2022] Open
Abstract
Chondral and osteochondral lesions represent one of the most challenging and frustrating scenarios for the orthopedic surgeon and for the patient. The lack of therapeutic strategies capable to reconstitute the function and structure of hyaline cartilage and to halt the progression toward osteoarthritis has brought clinicians and scientists together, to investigate the potential role of tissue engineering as a viable alternative to current treatment modalities. In particular, the role of bioprinting is emerging as an innovative technology that allows for the creation of organized 3D tissue constructs via a "layer-by-layer" deposition process. This process also has the capability to combine cells and biomaterials in an ordered and predetermined way. Here, we review the recent advances in cartilage bioprinting and we identify the current challenges and the directions for future developments in cartilage regeneration.
Collapse
Affiliation(s)
- Claudia Di Bella
- Department of Orthopaedic, St Vincent’s Hospital, Melbourne, VIC, Australia
- Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| | - Amanda Fosang
- Murdoch Childrens Research Institute, University of Melbourne, Parkville, VIC, Australia
| | - Davide M. Donati
- Unit of Orthopaedic Pathology and Osteoarticular Tissue Regeneration, Rizzoli Orthopaedic Institute, Bologna, Italy
| | - Gordon G. Wallace
- ARC Centre of Excellence for Electromaterials Science, AIIM Facility, Intelligent Polymer Research Institute, University of Wollongong, Wollongong, NSW, Australia
| | - Peter F. M. Choong
- Department of Orthopaedic, St Vincent’s Hospital, Melbourne, VIC, Australia
- Department of Surgery, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
19
|
Mateer JL, Hoch JM, Mattacola CG, Butterfield TA, Lattermann C. Serum Cartilage Oligomeric Matrix Protein Levels in Collegiate Soccer Athletes over the Duration of an Athletic Season: A Pilot Study. Cartilage 2015; 6:6-11. [PMID: 26069705 PMCID: PMC4462244 DOI: 10.1177/1947603514557944] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE The primary objective of this study was to measure serum cartilage oligomeric matrix protein (sCOMP) levels weekly in a group of collegiate soccer athletes over the duration of a spring soccer season and 2 weeks following the conclusion of the season while documenting minutes of exercise participation as a measure of exercise intensity. DESIGN A repeated-measures study design was employed. A volunteer sample of 6 female soccer athletes participated in this study. Serum samples were collected on 10 separate occasions, 1 week prior to the start of the season (baseline), once a week during the 8-week season (PX1-PX8), and once a week for 2 weeks following the conclusion of the season (postseason; PS1 and PS2). Minutes of participation were documented following all spring soccer activities for each week. Once all samples were collected, sCOMP concentrations were determined using a commercially available enzyme-linked immunosorbent assay. RESULTS The results of Friedman test revealed a significant effect for time (P = 0.003). Post hoc analysis revealed no significant differences between baseline and practice or postseason levels. A qualitative analysis of the sCOMP levels and minutes indicated higher sCOMP levels occurred when the athletes' participation in soccer-related activities was higher. CONCLUSIONS Qualitatively, our findings suggest that as minutes of participation increased, sCOMP levels increased. However, no statistically significant differences were identified. We speculate these increases were an increase in cartilage turnover and an interesting observation related to increases in physical activity. However, the implications are unclear as there was a return to near baseline levels.
Collapse
Affiliation(s)
- Jessica L. Mateer
- Sports Health Department, University of Florida, Gainesville, FL, USA
| | - Johanna M. Hoch
- School of Physical Therapy and Athletic Training, Old Dominion University, Norfolk, VA, USA
| | - Carl G. Mattacola
- Department of Rehabilitation Sciences, University of Kentucky, Lexington, KY, USA
| | | | - Christian Lattermann
- Department of Orthopaedic Surgery and Sports Medicine, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
20
|
Lord MS, Farrugia BL, Rnjak-Kovacina J, Whitelock JM. Current serological possibilities for the diagnosis of arthritis with special focus on proteins and proteoglycans from the extracellular matrix. Expert Rev Mol Diagn 2014; 15:77-95. [DOI: 10.1586/14737159.2015.979158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
21
|
Quantitative proteomics at different depths in human articular cartilage reveals unique patterns of protein distribution. Matrix Biol 2014; 40:34-45. [PMID: 25193283 DOI: 10.1016/j.matbio.2014.08.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 08/20/2014] [Accepted: 08/22/2014] [Indexed: 11/23/2022]
Abstract
The articular cartilage of synovial joints ensures friction-free mobility and attenuates mechanical impact on the joint during movement. These functions are mediated by the complex network of extracellular molecules characteristic for articular cartilage. Zonal differences in the extracellular matrix (ECM) are well recognized. However, knowledge about the precise molecular composition in the different zones remains limited. In the present study, we investigated the distribution of ECM molecules along the surface-to-bone axis, using quantitative non-targeted as well as targeted proteomics.\ In a discovery approach, iTRAQ mass spectrometry was used to identify all extractable ECM proteins in the different layers of a human lateral tibial plateau full thickness cartilage sample. A targeted MRM mass spectrometry approach was then applied to verify these findings and to extend the analysis to four medial tibial plateau samples. In the lateral tibial plateau sample, the unique distribution patterns of 70 ECM proteins were identified, revealing groups of proteins with a preferential distribution to the superficial, intermediate or deep regions of articular cartilage. The detailed analysis of selected 29 proteins confirmed these findings and revealed similar distribution patterns in the four medial tibial plateau samples. The results of this study allow, for the first time, an overview of the zonal distribution of a broad range of cartilage ECM proteins and open up further investigations of the functional roles of matrix proteins in the different zones of articular cartilage in health and disease.
Collapse
|
22
|
Acharya C, Yik JHN, Kishore A, Van Dinh V, Di Cesare PE, Haudenschild DR. Cartilage oligomeric matrix protein and its binding partners in the cartilage extracellular matrix: interaction, regulation and role in chondrogenesis. Matrix Biol 2014; 37:102-11. [PMID: 24997222 DOI: 10.1016/j.matbio.2014.06.001] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 06/05/2014] [Accepted: 06/05/2014] [Indexed: 10/25/2022]
Abstract
Thrombospondins (TSPs) are widely known as a family of five calcium-binding matricellular proteins. While these proteins belong to the same family, they are encoded by different genes, regulate different cellular functions and are localized to specific regions of the body. TSP-5 or Cartilage Oligomeric Matrix Protein (COMP) is the only TSP that has been associated with skeletal disorders in humans, including pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED). The pentameric structure of COMP, the evidence that it interacts with multiple cellular proteins, and the recent reports of COMP acting as a 'lattice' to present growth factors to cells, inspired this review of COMP and its interacting partners. In our review, we have compiled the interactions of COMP with other proteins in the cartilage extracellular matrix and summarized their importance in maintaining the structural integrity of cartilage as well as in regulating cellular functions.
Collapse
Affiliation(s)
- Chitrangada Acharya
- Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Jasper H N Yik
- Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Ashleen Kishore
- Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Victoria Van Dinh
- Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| | - Paul E Di Cesare
- Department of Orthopaedics and Rehabilitation, New York Hospital Queens, New York, NY 11355, USA
| | - Dominik R Haudenschild
- Department of Orthopaedic Surgery, Lawrence J. Ellison Musculoskeletal Research Center, University of California at Davis Medical Center, Sacramento, CA 95817, USA
| |
Collapse
|
23
|
Vuga LJ, Milosevic J, Pandit K, Ben-Yehudah A, Chu Y, Richards T, Sciurba J, Myerburg M, Zhang Y, Parwani AV, Gibson KF, Kaminski N. Cartilage oligomeric matrix protein in idiopathic pulmonary fibrosis. PLoS One 2013; 8:e83120. [PMID: 24376648 PMCID: PMC3869779 DOI: 10.1371/journal.pone.0083120] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 10/30/2013] [Indexed: 01/13/2023] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive and life threatening disease with median survival of 2.5-3 years. The IPF lung is characterized by abnormal lung remodeling, epithelial cell hyperplasia, myofibroblast foci formation, and extracellular matrix deposition. Analysis of gene expression microarray data revealed that cartilage oligomeric matrix protein (COMP), a non-collagenous extracellular matrix protein is among the most significantly up-regulated genes (Fold change 13, p-value <0.05) in IPF lungs. This finding was confirmed at the mRNA level by nCounter® expression analysis in additional 115 IPF lungs and 154 control lungs as well as at the protein level by western blot analysis. Immunohistochemical analysis revealed that COMP was expressed in dense fibrotic regions of IPF lungs and co-localized with vimentin and around pSMAD3 expressing cells. Stimulation of normal human lung fibroblasts with TGF-β1 induced an increase in COMP mRNA and protein expression. Silencing COMP in normal human lung fibroblasts significantly inhibited cell proliferation and negatively impacted the effects of TGF-β1 on COL1A1 and PAI1. COMP protein concentration measured by ELISA assay was significantly increased in serum of IPF patients compared to controls. Analysis of serum COMP concentrations in 23 patients who had prospective blood draws revealed that COMP levels increased in a time dependent fashion and correlated with declines in force vital capacity (FVC). Taken together, our results should encourage more research into the potential use of COMP as a biomarker for disease activity and TGF-β1 activity in patients with IPF. Hence, studies that explore modalities that affect COMP expression, alleviate extracellular matrix rigidity and lung restriction in IPF and interfere with the amplification of TGF-β1 signaling should be persuaded.
Collapse
Affiliation(s)
- Louis J. Vuga
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Jadranka Milosevic
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kusum Pandit
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Ahmi Ben-Yehudah
- Pittsburgh Development Center, Magee-Women’s Research Institute and Foundation, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Yanxia Chu
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Thomas Richards
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Joshua Sciurba
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Michael Myerburg
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Yingze Zhang
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Anil V. Parwani
- Department of Pathology, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Kevin F. Gibson
- Dorothy P and Richard P Simmons Center for Interstitial Lung Diseases, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Naftali Kaminski
- Pulmonary, Critical Care and Sleep Medicine, Yale School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
24
|
Verma P, Dalal K. Serum cartilage oligomeric matrix protein (COMP) in knee osteoarthritis: a novel diagnostic and prognostic biomarker. J Orthop Res 2013; 31:999-1006. [PMID: 23423905 DOI: 10.1002/jor.22324] [Citation(s) in RCA: 103] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 01/23/2013] [Indexed: 02/04/2023]
Abstract
A case-control study was conducted to estimate the association of cartilage oligomeric matrix protein (COMP) with knee osteoarthritis (OA) and to examine the potential utility of COMP as a diagnostic and prognostic biomarker in early knee OA. The COMP levels were estimated in the blood sera of 150 subjects belonging to study group (n = 100) and control one (n = 50). Patients with confirmed clinical isolated knee OA diagnosed through American College of Rheumatology criteria were included and were without any other cause of knee pain. ELISA was used to determine the levels of COMP, interleukin-1β (IL-1β) and tumor necrosis factor-α (TNF-α). The median (range) serum COMP levels were observed to be 1117.21 ng/ml (125.03-4209.75 ng/ml) in OA patients and 338.62 ng/ml (118-589 ng/ml) in control subjects with p < 0.001. The COMP levels of study group were negatively correlated (correlation factor -0.88) with disease duration and positively correlated with age, BMI, pain score and IL-1β with correlation factors 0.86, 0.63, 0.76, and 0.79, respectively with p < 0.001. Gender differentiation was found in study group with 52% higher COMP level in males as compared to that of females. There was no significant correlation of COMP levels with radiological grading, erythrocyte sedimentation rate (ESR), hemoglobin (Hb), and TNF-α. The serum COMP levels may be used as a diagnostic OA marker along with prognostic value in determining the patients at risk of rapidly progressing this debilitating joint disease. The serum COMP level remains significantly high in first 3 years of disease duration.
Collapse
Affiliation(s)
- Priyanka Verma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, India.
| | | |
Collapse
|
25
|
Schuurman W, Harimulyo EB, Gawlitta D, Woodfield TBF, Dhert WJA, van Weeren PR, Malda J. Three-dimensional assembly of tissue-engineered cartilage constructs results in cartilaginous tissue formation without retainment of zonal characteristics. J Tissue Eng Regen Med 2013; 10:315-24. [DOI: 10.1002/term.1726] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2012] [Revised: 08/08/2012] [Accepted: 01/22/2013] [Indexed: 01/15/2023]
Affiliation(s)
- W. Schuurman
- Department of Orthopaedics; University Medical Centre Utrecht; The Netherlands
- Department of Equine Sciences, Faculty of Veterinary Sciences; Utrecht University; The Netherlands
| | - E. B. Harimulyo
- Department of Orthopaedics; University Medical Centre Utrecht; The Netherlands
| | - D. Gawlitta
- Department of Orthopaedics; University Medical Centre Utrecht; The Netherlands
| | - T. B. F. Woodfield
- Department of Orthopaedic Surgery; University of Otago; Christchurch New Zealand
| | - W. J. A. Dhert
- Department of Orthopaedics; University Medical Centre Utrecht; The Netherlands
- Faculty of Veterinary Sciences; University of Utrecht; The Netherlands
| | - P. R. van Weeren
- Department of Equine Sciences, Faculty of Veterinary Sciences; Utrecht University; The Netherlands
| | - J. Malda
- Department of Orthopaedics; University Medical Centre Utrecht; The Netherlands
| |
Collapse
|
26
|
Skory RM, Bernabé BP, Galdones E, Broadbelt LJ, Shea LD, Woodruff TK. Microarray analysis identifies COMP as the most differentially regulated transcript throughout in vitro follicle growth. Mol Reprod Dev 2013; 80:132-44. [PMID: 23242557 DOI: 10.1002/mrd.22144] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2012] [Accepted: 12/09/2012] [Indexed: 12/15/2022]
Abstract
In vitro follicle growth has emerged as a technology that can provide new information about folliculogenesis and serve as part of a suite of methods currently under development to assist women whose fertility is threatened by cancer treatments. Though it has been shown that in vitro-grown follicles secrete peptide and steroid hormones, much of the follicular transcriptome remains unknown. Thus, microarray analysis was performed to characterize the transcriptome and secretome of in vitro-grown follicles. One prominently regulated gene product was cartilage oligomeric matrix protein (Comp): its mRNA was upregulated during the final 4 days of culture (P < 0.05) and COMP protein could be detected in medium from individual follicles. COMP expression localized to mural granulosa cells of large antral follicles both in vitro and in vivo, with maximal expression immediately preceding ovulation in cycling and chorionic gonadotropin-primed female mice. COMP was co-expressed with two known markers of follicle maturation, inhibin β(A) and gremlin, and was expressed only in TUNEL-negative follicles. In addition to other gene products identified in the microarray, COMP has potential utility as a marker of follicle maturation.
Collapse
Affiliation(s)
- Robin M Skory
- Department of Obstetrics and Gynecology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | | | | | | | | | |
Collapse
|
27
|
Cartilage oligomeric matrix protein in serum and epiphyseal plate in Kashin-Beck disease. CURRENT ORTHOPAEDIC PRACTICE 2013. [DOI: 10.1097/bco.0b013e3182793dde] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
28
|
Schuurman W, Klein TJ, Dhert WJA, van Weeren PR, Hutmacher DW, Malda J. Cartilage regeneration using zonal chondrocyte subpopulations: a promising approach or an overcomplicated strategy? J Tissue Eng Regen Med 2012; 9:669-78. [PMID: 23135870 DOI: 10.1002/term.1638] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 08/30/2012] [Accepted: 09/27/2012] [Indexed: 01/01/2023]
Abstract
Cartilage defects heal imperfectly and osteoarthritic changes develop frequently as a result. Although the existence of specific behaviours of chondrocytes derived from various depth-related zones in vitro has been known for over 20 years, only a relatively small body of in vitro studies has been performed with zonal chondrocytes and current clinical treatment strategies do not reflect these native depth-dependent (zonal) differences. This is surprising since mimicking the zonal organization of articular cartilage in neo-tissue by the use of zonal chondrocyte subpopulations could enhance the functionality of the graft. Although some research groups including our own have made considerable progress in tailoring culture conditions using specific growth factors and biomechanical loading protocols, we conclude that an optimal regime has not yet been determined. Other unmet challenges include the lack of specific zonal cell sorting protocols and limited amounts of cells harvested per zone. As a result, the engineering of functional tissue has not yet been realized and no long-term in vivo studies using zonal chondrocytes have been described. This paper critically reviews the research performed to date and outlines our view of the potential future significance of zonal chondrocyte populations in regenerative approaches for the treatment of cartilage defects. Secondly, we briefly discuss the capabilities of additive manufacturing technologies that can not only create patient-specific grafts directly from medical imaging data sets but could also more accurately reproduce the complex 3D zonal extracellular matrix architecture using techniques such as hydrogel-based cell printing.
Collapse
Affiliation(s)
- W Schuurman
- Department of Orthopaedics, University Medical Center Utrecht, The Netherlands.,Department of Equine Sciences, Faculty of Veterinary Sciences, Utrecht University, The Netherlands
| | - T J Klein
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia
| | - W J A Dhert
- Department of Orthopaedics, University Medical Center Utrecht, The Netherlands.,Faculty of Veterinary Sciences, University of Utrecht, The Netherlands
| | - P R van Weeren
- Department of Equine Sciences, Faculty of Veterinary Sciences, Utrecht University, The Netherlands
| | - D W Hutmacher
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia
| | - J Malda
- Department of Orthopaedics, University Medical Center Utrecht, The Netherlands.,Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Australia
| |
Collapse
|
29
|
Yang JH, Kim JH, Lim DS, Oh KJ. Effect of combined sex hormone replacement on bone/cartilage turnover in a murine model of osteoarthritis. Clin Orthop Surg 2012; 4:234-41. [PMID: 22949956 PMCID: PMC3425655 DOI: 10.4055/cios.2012.4.3.234] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 12/21/2011] [Indexed: 12/05/2022] Open
Abstract
Background Estrogens act on estrogen receptors distributed in articular cartilages, synovial membrane, and ligaments, which are thought to be related with degenerative changes. Meanwhile, progesterone is known to have a weak anabolic action on bone formation This study evaluates the effects of estrogen and progesterone hormone on bone/cartilage turnover in ovariectomized (OVX) rats. Methods Thirty-five 7-month-old female Sprague-Dawley rats were randomly divided into 5 groups and then ovariectomized bilaterally except the sham control group. The first and the second group acting as controls did not receive hormonal therapy, the third group received estrogen, the fourth group received progesterone, and the fifth group received combination of both hormones 10 weeks after surgery. Evaluations were done using the serum levels of cartilage oligomeric matrix protein (COMP) for cartilage turnover, collagen type I C-telopeptide (CTX-1) and osteocalcin (OC) for bone turnover at 11, 15, 19 weeks after OVX and histology using the Osteoarthritis Research Society International (OARSI) osteoarthritis (OA) cartilage histopathology assessment system. Results Significantly less cartilage degradation (decreased levels of COMP) was found in the combined hormone treated group in comparison with OVX group. Similarly, both hormonal treatment resulted in increased bone formation and decreased bone resorption i.e., a low overall bone turnover status (decrease in the serum OC and CTX-1 levels). Conclusions Combined estrogen and progesterone therapy was found to be convincing in terms of reducing the severity of OA in this experimental model.
Collapse
Affiliation(s)
- Jae-Hyuk Yang
- Department of Orthopaedic Surgery, Seoul Veterans Hospital, Seoul, Korea
| | | | | | | |
Collapse
|
30
|
Lai Y, Yu XP, Zhang Y, Tian Q, Song H, Mucignat MT, Perris R, Samuels J, Krasnokutsky S, Attur M, Greenberg JD, Abramson SB, Di Cesare PE, Liu C. Enhanced COMP catabolism detected in serum of patients with arthritis and animal disease models through a novel capture ELISA. Osteoarthritis Cartilage 2012; 20:854-62. [PMID: 22595227 PMCID: PMC3389204 DOI: 10.1016/j.joca.2012.05.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 04/15/2012] [Accepted: 05/06/2012] [Indexed: 02/02/2023]
Abstract
OBJECTIVE The study aimed determining whether assessment of cartilage oligomeric matrix protein (COMP) degradation products could serve as a serological disease course and therapeutic response predictor in arthritis. METHODS We generated a panel of monoclonal antibodies against COMP fragments and developed a novel capture enzyme-linked immunosorbent assay (ELISA) for detecting COMP fragments in patients with osteoarthritis (OA) and rheumatoid arthritis (RA). This test was also used to monitor COMP fragments in surgically-induced OA, collagen-induced arthritis (CIA), and tumor necrosis factor (TNF) transgenic animal models. RESULTS Compared with a commercial COMP ELISA kit that detected no significant difference in COMP levels between OA and control groups, a significant increase of the COMP fragments were noted in the serum of OA patients assayed by this newly established ELISA. In addition, serum COMP fragment levels were well correlated with severity in OA patients and the progression of surgically-induced OA in murine models. Furthermore, the serum levels of COMP fragments in RA patients, mice with CIA, and TNF transgenic mice were significantly higher when compared with their controls. Interestingly, treatment with TNFα inhibitors and methotrexate led to a significant decrease of serum COMP fragments in RA patients. Additionally, administration of Atsttrin [Tang, et al., Science 2011;332(6028):478] also resulted in a significant reduction in COMP fragments in arthritis mice models. CONCLUSION A novel sandwich ELISA is capable of reproducibly measuring serum COMP fragments in both arthritic patients and rodent arthritis models. This test also provides a valuable means to utilize serum COMP fragments for monitoring the effects of interventions in arthritis.
Collapse
Affiliation(s)
- Yongjie Lai
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003,Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China
| | - Xiu-Ping Yu
- Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong 250012, China,To whom correspondence should be addressed: Xiu-Ping Yu, Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China, Tel: 86-531-88382579, ; or Chuan-Ju Liu, PhD, Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, 10003, 301 East 17th Street, New York, NY 10003. Tel: 212-598-6103; Fax: 212-598-6096;
| | - Yuying Zhang
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Qingyun Tian
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Haicheng Song
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003
| | - Maria Teresa Mucignat
- Department of Genetics, Microbiology and Anthropology, University of Parma, Parma 43100, Italy
| | - Roberto Perris
- Department of Genetics, Microbiology and Anthropology, University of Parma, Parma 43100, Italy
| | - Jonathan Samuels
- Division of Rheumatology, NYU Hospital for Joint Diseases, New York, NY 10003
| | | | - Mukundan Attur
- Division of Rheumatology, NYU Hospital for Joint Diseases, New York, NY 10003
| | | | - Steven B. Abramson
- Division of Rheumatology, NYU Hospital for Joint Diseases, New York, NY 10003
| | - Paul E. Di Cesare
- Department of Orthopaedic Surgery, UC Davis Medical Center, Sacramento, CA 95817
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003,Department of Cell Biology, New York University School of Medicine, New York, NY 10016,To whom correspondence should be addressed: Xiu-Ping Yu, Institute of Pathogenic Biology, Shandong University School of Medicine, Jinan, Shandong 250012, P.R. China, Tel: 86-531-88382579, ; or Chuan-Ju Liu, PhD, Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, 10003, 301 East 17th Street, New York, NY 10003. Tel: 212-598-6103; Fax: 212-598-6096;
| |
Collapse
|
31
|
Xu L, Polur I, Servais JM, Hsieh S, Lee PL, Goldring MB, Li Y. Intact pericellular matrix of articular cartilage is required for unactivated discoidin domain receptor 2 in the mouse model. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:1338-46. [PMID: 21855682 DOI: 10.1016/j.ajpath.2011.05.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 04/29/2011] [Accepted: 05/24/2011] [Indexed: 10/17/2022]
Abstract
Increased expression of the discoidin domain receptor 2 (DDR2) results from its interaction with collagen type II. This induces expression of matrix metalloproteinase (MMP)-13, leading to osteoarthritis (OA). To investigate the impact of the pericellular matrix of chondrocytes on DDR2, we generated a mouse model with inducible overexpression of DDR2 in cartilage. Conditional overexpression of DDR2 in mature mouse articular cartilage was controlled via the cartilage oligomeric matrix protein promoter using the Tet-Off-inducible system. Doxycycline was withdrawn at 1 month of age, and knee joints were examined at 2, 3, and 4 months of age. Microsurgery was performed on 3-month-old transgenic mice overexpressing DDR2 to destabilize the medial meniscus, and serial paraffin sections were examined at 2, 4, 8, and 12 weeks after surgery. DDR2 expression increased in the knee joints of transgenic mice. However, the increased DDR2 did not induce MMP-13 expression. No OA-like changes were observed in the transgenic mice at the age of 4 months. When transgenic mice were subjected to destabilizing of the medial meniscus, we observed accelerated progression to OA, which was associated with DDR2 activation. Therefore, conditionally overexpressing DDR2 in the mature articular cartilage of mouse knee joints requires activation to induce OA, and altered biomechanical stress can accelerate the onset of cartilage loss and progression to OA in transgenic mice.
Collapse
Affiliation(s)
- Lin Xu
- Department of Developmental Biology, Harvard School of Dental Medicine, Boston, Massachusetts, USA.
| | | | | | | | | | | | | |
Collapse
|
32
|
Chop (Ddit3) is essential for D469del-COMP retention and cell death in chondrocytes in an inducible transgenic mouse model of pseudoachondroplasia. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 180:727-37. [PMID: 22154935 DOI: 10.1016/j.ajpath.2011.10.035] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 10/19/2011] [Accepted: 10/23/2011] [Indexed: 11/21/2022]
Abstract
Cartilage oligomeric matrix protein (COMP), a secreted glycoprotein synthesized by chondrocytes, regulates proliferation and type II collagen assembly. Mutations in the COMP gene cause pseudoachondroplasia and multiple epiphyseal dysplasia. Previously, we have shown that expression of D469del-COMP in transgenic mice causes intracellular retention of D469del-COMP, thereby recapitulating pseudoachondroplasia chondrocyte pathology. This inducible transgenic D469del-COMP mouse is the only in vivo model to replicate the critical cellular and clinical features of pseudoachondroplasia. Here, we report developmental studies of D469del-COMP-induced chondrocyte pathology from the prenatal period to adolescence. D469del-COMP retention was limited prenatally and did not negatively affect the growth plate until 3 weeks after birth. Results of immunostaining, transcriptome analysis, and qRT-PCR suggest a molecular model in which D469del-COMP triggers apoptosis during the first postnatal week. By 3 weeks (when most chondrocytes are retaining D469del-COMP), inflammation, oxidative stress, and DNA damage contribute to chondrocyte cell death by necroptosis. Importantly, by crossing the D469del-COMP mouse onto a Chop null background (Ddit3 null), thereby eliminating Chop, the unfolded protein response was disrupted, thus alleviating both D469del-COMP intracellular retention and premature chondrocyte cell death. Chop therefore plays a significant role in processes that mediate D469del-COMP retention. Taken together, these results suggest that there may be an optimal window before the induction of significant D469del-COMP retention during which endoplasmic reticulum stress could be targeted.
Collapse
|
33
|
Du Y, Wang Y, Wang L, Liu B, Tian Q, Liu CJ, Zhang T, Xu Q, Zhu Y, Ake O, Qi Y, Tang C, Kong W, Wang X. Cartilage Oligomeric Matrix Protein Inhibits Vascular Smooth Muscle Calcification by Interacting With Bone Morphogenetic Protein-2. Circ Res 2011; 108:917-28. [DOI: 10.1161/circresaha.110.234328] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale:
Vascular calcification is a significant contributor to cardiovascular morbidity and mortality. We recently reported that cartilage oligomeric matrix protein (COMP) is pivotal for maintaining the homeostasis of vascular smooth muscle cells (VSMCs). Whether COMP affects the process of vascular calcification is unknown.
Objective:
We aimed to test whether COMP modulates vascular calcification.
Methods and Results:
VSMC calcification in vitro was induced by calcifying media containing high inorganic phosphate or calcium. In vivo medial vessel calcification was induced in rats by 5/6 nephrectomy with a high-phosphate diet or by periadventitial application of CaCl
2
to the abdominal aorta. COMP protein level was markedly reduced in both calcified VSMCs and arteries. COMP deficiency remarkably exacerbated VSMC calcification, whereas ectopic expression of COMP greatly reduced calcification. Furthermore, COMP knockdown facilitated osteogenic markers expression by VSMCs even in the absence of calcifying media. By contrast, COMP overexpression significantly inhibited high phosphate– or high calcium–induced VSMC osteochondrogenic transition. Induction of osteogenic marker expression by COMP silencing was reversed by a soluble form of bone morphogenetic protein (BMP)-2 receptor IA, which suggests a BMP-2–dependent mechanism. Our data revealed that COMP bound directly to BMP-2 through the C terminus, inhibited BMP-2 receptor binding, and blocked BMP-2 osteogenic signaling, indicating COMP inhibits osteochondrogenic transition of VSMCs at least partially through inhibiting BMP-2.
Conclusions:
Our data strongly suggest that COMP is a novel inhibitor of vascular calcification. The imbalance between the effects of COMP and BMP-2 may provide new insights into the pathophysiology of vascular calcification.
Collapse
Affiliation(s)
- Yaoyao Du
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Yue Wang
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Li Wang
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Bo Liu
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Qingyun Tian
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Chuan-ju Liu
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Tao Zhang
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Qingbo Xu
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Yi Zhu
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Oldberg Ake
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Yongfen Qi
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Chaochu Tang
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Wei Kong
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| | - Xian Wang
- From the Department of Physiology and Pathophysiology (Y.D., L.W., B.L., Y.Z., Y.Q., C.T., W.K., X.W.), School of Basic Medical Sciences, Peking University, Beijing, People's Republic of China; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, People's Republic of China; Department of Nephrology (Y.W.), Peking University Third Hospital, Beijing, People's Republic of China; Department of Orthopaedic Surgery and Department of Cell Biology (Q.T., C.-j.L.), New York
| |
Collapse
|
34
|
Motaung SCKM, Di Cesare PE, Hari Reddi A. Differential response of cartilage oligomeric matrix protein (COMP) to morphogens of bone morphogenetic protein/transforming growth factor-β family in the surface, middle and deep zones of articular cartilage. J Tissue Eng Regen Med 2011; 5:e87-96. [DOI: 10.1002/term.358] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 07/12/2010] [Indexed: 12/21/2022]
|
35
|
Posey KL, Liu P, Wang HR, Veerisetty AC, Alcorn JL, Hecht JT. RNAi reduces expression and intracellular retention of mutant cartilage oligomeric matrix protein. PLoS One 2010; 5:e10302. [PMID: 20421976 PMCID: PMC2858657 DOI: 10.1371/journal.pone.0010302] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2010] [Accepted: 03/12/2010] [Indexed: 01/06/2023] Open
Abstract
Mutations in cartilage oligomeric matrix protein (COMP), a large extracellular glycoprotein expressed in musculoskeletal tissues, cause two skeletal dysplasias, pseudoachondroplasia and multiple epiphyseal dysplasia. These mutations lead to massive intracellular retention of COMP, chondrocyte death and loss of growth plate chondrocytes that are necessary for linear growth. In contrast, COMP null mice have only minor growth plate abnormalities, normal growth and longevity. This suggests that reducing mutant and wild-type COMP expression in chondrocytes may prevent the toxic cellular phenotype causing the skeletal dysplasias. We tested this hypothesis using RNA interference to reduce steady state levels of COMP mRNA. A panel of shRNAs directed against COMP was tested. One shRNA (3B) reduced endogenous and recombinant COMP mRNA dramatically, regardless of expression levels. The activity of the shRNA against COMP mRNA was maintained for up to 10 weeks. We also demonstrate that this treatment reduced ER stress. Moreover, we show that reducing steady state levels of COMP mRNA alleviates intracellular retention of other extracellular matrix proteins associated with the pseudoachondroplasia cellular pathology. These findings are a proof of principle and the foundation for the development of a therapeutic intervention based on reduction of COMP expression.
Collapse
Affiliation(s)
- Karen L Posey
- Department of Pediatrics, University of Texas Medical School at Houston, Houston, Texas, United States of America.
| | | | | | | | | | | |
Collapse
|
36
|
Cartilage oligomeric matrix protein promotes cell attachment via two independent mechanisms involving CD47 and alphaVbeta3 integrin. Mol Cell Biochem 2009; 338:215-24. [PMID: 20033473 DOI: 10.1007/s11010-009-0355-3] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2009] [Accepted: 12/03/2009] [Indexed: 10/20/2022]
Abstract
Cartilage oligomeric matrix protein (COMP) is a pentameric approximately 524 kDa multidomain extracellular matrix protein and is the fifth member of the thrombospondin family. COMP is abundantly expressed in proliferating and hypertrophic chondrocytes of the growth plate, articular cartilage, synovium, tendon, and ligament. The spatial localization of COMP highlights its importance in the phenotypes of pseudoachondroplasia (PSACH) and multiple epiphyseal dysplasia (MED), COMP disorders that are characterized by disproportionate short stature, brachydactyly, scoliosis, early-onset osteoarthritis, and joint hypermobility. In this study, the role of COMP in ligament was investigated with a series of cell attachment assays using ligament cells binding to COMP. A dose-dependent cell attachment activity was found, which was inhibited by a peptide containing the SFYVVMWK amino acid sequence derived from the globular C-terminal domain of COMP. This activity was independent of the recently described RGD-dependent attachment activity. Function-blocking antibodies to CD47 and alphaVbeta3 integrin reduced cell attachment to COMP, implicating the participation of these cell surface molecules in COMP cell binding. Immunofluorescence studies showed that cell attachment to COMP induced the formation of lamellae containing F-actin microspikes associated with fascin. We propose that COMP promotes cell attachment via two independent mechanisms involving cell surface CD47 and alphaVbeta3 integrin and that a consequence of cell attachment to COMP is the specific induction of fascin-stabilized actin microspikes.
Collapse
|
37
|
Schuurman W, Gawlitta D, Klein TJ, ten Hoope W, van Rijen MHP, Dhert WJA, van Weeren PR, Malda J. Zonal chondrocyte subpopulations reacquire zone-specific characteristics during in vitro redifferentiation. Am J Sports Med 2009; 37 Suppl 1:97S-104S. [PMID: 19846691 DOI: 10.1177/0363546509350978] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
BACKGROUND If chondrocytes from the superficial, middle, and deep zones of articular cartilage could maintain or regain their characteristic properties during in vitro culture, it would be feasible to create constructs comprising these distinctive zones. HYPOTHESIS Zone-specific characteristics of zonal cell populations will disappear during 2-dimensional expansion but will reappear after 3-dimensional redifferentiation, independent of the culture technique used (alginate beads versus pellet culture). STUDY DESIGN Controlled laboratory study. METHODS Equine articular chondrocytes from the 3 zones were expanded in monolayer culture (8 donors) and subsequently redifferentiated in pellet and alginate bead cultures for up to 4 weeks. Glycosaminoglycans and DNA were quantified, along with immunohistochemical assessment of the expression of various zonal markers, including cartilage oligomeric protein (marking cells from the deeper zones) and clusterin (specifically expressed by superficial chondrocytes). RESULTS Cell yield varied between zones, but proliferation rates did not show significant differences. Expression of all evaluated zonal markers was lost during expansion. Compared to the alginate bead cultures, pellet cultures showed a higher amount of glycosaminoglycans produced per DNA after redifferentiation. In contrast to cells in pellet cultures, cells in alginate beads regained zonal differences, as evidenced by zone-specific reappearance of cartilage oligomeric protein and clusterin, as well as significantly higher glycosaminoglycans production by cells from the deep zone compared to the superficial zone. CONCLUSION Chondrocytes isolated from the 3 zones of equine cartilage can restore their zone-specific matrix expression when cultured in alginate after in vitro expansion. CLINICAL RELEVANCE Appreciation of the zonal differences can lead to important advances in cartilage tissue engineering. Findings support the use of hydrogels such as alginate for engineering zonal cartilage constructs.
Collapse
Affiliation(s)
- Wouter Schuurman
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Posey KL, Veerisetty AC, Liu P, Wang HR, Poindexter BJ, Bick R, Alcorn JL, Hecht JT. An inducible cartilage oligomeric matrix protein mouse model recapitulates human pseudoachondroplasia phenotype. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 175:1555-63. [PMID: 19762713 PMCID: PMC2751552 DOI: 10.2353/ajpath.2009.090184] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/01/2009] [Indexed: 11/20/2022]
Abstract
Cartilage oligomeric matrix protein (COMP) is a pentameric extracellular protein expressed in cartilage and other musculoskeletal tissues. Mutations in the COMP gene cause pseudoachondroplasia (PSACH), a severe dwarfing condition that has a growth plate chondrocyte pathology. PSACH is characterized by intracellular retention of COMP and other extracellular matrix (ECM) proteins, which form an ordered matrix within large rough endoplasmic reticulum cisternae. This accumulation is cytotoxic and causes premature chondrocyte cell death, thereby depleting chondrocytes needed for normal long bone growth. Research to define the underlying molecular mechanisms of PSACH has been hampered by the lack of a suitable model system. In this study, we achieved robust expression of human mutant (MT) or wild-type (WT) COMP in mice by using a tetracycline-inducible promoter. Normal growth plate distribution of ECM proteins was observed in 1-month-old WT-COMP and C57BL\6 control mice. In contrast, the structure of the MT-COMP growth plate recapitulated the findings of human PSACH growth plate morphology, including (1) retention of ECM proteins, (2) intracellular matrix formation in the rER cisternae, and (3) increased chondrocyte apoptosis. Therefore, we have generated the first mouse model to show extensive intracellular retention of ECM proteins recapitulating the human PSACH disease process at the cellular level.
Collapse
Affiliation(s)
- Karen L Posey
- Department of Pediatrics, University of Texas Medical School, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
A broad screen for targets of immune complexes decorating arthritic joints highlights deposition of nucleosomes in rheumatoid arthritis. Proc Natl Acad Sci U S A 2009; 106:15867-72. [PMID: 19720992 DOI: 10.1073/pnas.0908032106] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Deposits of Ig and complement are abundant in affected joints of patients with rheumatoid arthritis (RA) and in animal models of RA in which antibodies are demonstrably pathogenic. To identify molecular targets of the Igs deposited in arthritic joints, which may activate local inflammation, we used a combination of mass spectrometry (MS) and protein microarrays. Immune complexes were affinity-purified from surgically removed joint tissues of 26 RA and osteoarthritis (OA) patients. Proteins complexed with IgG were identified by proteomic analysis using tandem MS. A striking diversity of components of the extracellular matrix, and some intracellular components, copurified specifically with IgG from RA and OA tissues. A smaller set of autoantigens was observed only in RA eluates. In complementary experiments, IgG fractions purified from joint immune complexes were tested on protein microarrays against a range of candidate autoantigens. These Igs bound a diverse subset of proteins and peptides from synovium and cartilage, different from that bound by normal serum Ig. One type of intracellular protein detected specifically in RA joints (histones H2A/B) was validated by immunohistology and found to be deposited on the cartilage surface of RA but not OA joints. Thus, autoantibodies to many determinants (whether deposited as "neoantigens" or normal constituents of the extracellular matrix) have the potential to contribute to arthritic inflammation.
Collapse
|
40
|
Tseng S, Reddi AH, Di Cesare PE. Cartilage Oligomeric Matrix Protein (COMP): A Biomarker of Arthritis. Biomark Insights 2009; 4:33-44. [PMID: 19652761 PMCID: PMC2716683 DOI: 10.4137/bmi.s645] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Arthritis is a chronic disease with a significant impact on the population. It damages the cartilage, synovium, and bone of the joints causing pain, impairment, and disability in patients. Current methods for diagnosis of and monitoring the disease are only able to detect clinical manifestations of arthritis late in the process. However, with the recent onset of successful treatments for rheumatoid arthritis and osteoarthritis, it becomes important to identify prognostic factors that can predict the evolution of arthritis. This is especially critical in the early phases of disease so that these treatments can be started as soon as possible to slow down progression of the disease. A valuable approach to monitor arthritis would be by measuring biological markers of cartilage degradation and repair to reflect variations in joint remodeling. One such potential biological marker of arthritis is cartilage oligomeric matrix protein (COMP). In various studies, COMP has shown promise as a diagnostic and prognostic indicator and as a marker of the disease severity and the effect of treatment. This review highlights the progress in the utilization of COMP as a biomarker of arthritis.
Collapse
Affiliation(s)
- Susan Tseng
- Department of Orthopaedic Surgery, UC Davis Medical Center, Sacramento, California, 95817, U.S.A
| | | | | |
Collapse
|
41
|
Adams JC, Bentley AA, Kvansakul M, Hatherley D, Hohenester E. Extracellular matrix retention of thrombospondin 1 is controlled by its conserved C-terminal region. J Cell Sci 2008; 121:784-95. [PMID: 18285447 DOI: 10.1242/jcs.021006] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Thrombospondins (TSPs) are an evolutionarily ancient family of extracellular calcium-binding glycoproteins. The five mammalian TSPs collectively have important roles in angiogenesis and vascular biology, synaptogenesis, wound repair and connective tissue organisation. Their complex functions relate to the multiple postsecretion fates of TSPs that can involve endocytic uptake, proteolysis or retention within the extracellular matrix (ECM). Surprisingly, the molecular and cellular mechanisms by which TSPs become retained within the ECM are poorly understood. We hypothesised that the highly conserved TSP C-terminal domain mediates ECM retention. We report that ECM incorporation as insoluble punctate deposits is an evolutionarily conserved property of TSPs. ECM retention of TSP1 is mediated by the C-terminal region in trimeric form, and not by C-terminal monomer or trimers of the N-terminal domain or type 1 repeats. Using a novel mRFP-tagged TSP1 C-terminal trimer, we demonstrate that ECM retention involves the RGD site and a novel site in the L-lectin domain with structural similarity to the ligand-binding site of cargo transport proteins. CD47 and beta1 integrins are dispensable for ECM retention, but beta1 integrins enhance activity. These novel data advance concepts of the molecular processes that lead to ECM retention of TSP1.
Collapse
Affiliation(s)
- Josephine C Adams
- Department of Cell Biology, Lerner Research Institute, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | | | | | | | |
Collapse
|
42
|
Gagarina V, Carlberg AL, Pereira-Mouries L, Hall DJ. Cartilage Oligomeric Matrix Protein Protects Cells against Death by Elevating Members of the IAP Family of Survival Proteins. J Biol Chem 2008; 283:648-659. [DOI: 10.1074/jbc.m704035200] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
43
|
Fujiki M, Shineha J, Yamanokuchi K, Misumi K, Sakamoto H. Effects of treatment with polysulfated glycosaminoglycan on serum cartilage oligomeric matrix protein and C-reactive protein concentrations, serum matrix metalloproteinase-2 and -9 activities, and lameness in dogs with osteoarthritis. Am J Vet Res 2007; 68:827-33. [PMID: 17669022 DOI: 10.2460/ajvr.68.8.827] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To investigate the effects of polysulfated glycosaminoglycan (PSGAG) treatment on serum cartilage oligomeric matrix protein (COMP) concentration, matrix metal-loproteinase-2 (MMP-2) and -9 (MMP-9) activities, C-reactive protein (CRP) concentration, and lameness scores in dogs with osteoarthritis. ANIMALS 16 dogs with osteoarthritis and 5 clinically normal dogs. PROCEDURES Dogs with osteoarthritis had a history of chronic lameness, and osteophytes were observed on radiographic evaluation of the affected joint. Polysulfated glycosaminoglycan was administered IM twice a week for a total of 8 treatments to all dogs with osteoarthritis and to clinically normal control dogs. RESULTS Lameness scores after PSGAG treatment in osteoarthritic dogs improved in 12 of the 16 dogs. Serum COMP concentrations in osteoarthritic dogs were significantly higher than in control dogs before treatment. Lameness scores in osteoarthritic dogs decreased significantly after treatment, compared with before treatment. Lameness scores of 9 dogs with hind limb lameness improved significantly after treatment; these dogs had corresponding decreases in serum COMP concentrations. After treatment, serum COMP concentrations and lameness scores of 7 dogs with forelimb lameness remained high and were significantly higher than those of dogs with hind limb lameness. Serum MMP-9 activities of dogs with forelimb lameness were significantly higher than in dogs with hind limb lameness after treatment. CONCLUSIONS AND CLINICAL RELEVANCE IM administration of PSGAG inhibited COMP degradation in dogs with osteoarthritis. Results indicate that decreases in serum COMP concentrations might be related to improvement in lameness after PSGAG treatment.
Collapse
Affiliation(s)
- Makoto Fujiki
- Laboratory of Veterinary Surgery, Department of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | | | | | | | | |
Collapse
|
44
|
Chen FH, Herndon ME, Patel N, Hecht JT, Tuan RS, Lawler J. Interaction of cartilage oligomeric matrix protein/thrombospondin 5 with aggrecan. J Biol Chem 2007; 282:24591-8. [PMID: 17588949 PMCID: PMC2905148 DOI: 10.1074/jbc.m611390200] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Cartilage oligomeric matrix protein/thrombospondin 5 (COMP/TSP5) is a major component of the extracellular matrix (ECM) of the musculoskeletal system. Its importance is underscored by its association with several growth disorders. In this report, we investigated its interaction with aggrecan, a major component of cartilage ECM. We also tested a COMP/TSP5 mutant, designated MUT3 that accounts for 30% of human pseudoachondroplasia cases, to determine if the mutation affects function. Using a solid-phase binding assay, we have shown that COMP/TSP5 can bind aggrecan. This binding was decreased with MUT3, or when COMP/TSP5 was treated with EDTA, indicating the presence of a conformation-dependent aggrecan binding site. Soluble glycosaminoglycans (GAGs) partially inhibited binding, suggesting that the interaction was mediated in part through aggrecan GAG side chains. Using affinity co-electrophoresis, we showed that COMP/TSP5, in its calcium-replete conformation, bound to heparin, chondroitin sulfates, and heparan sulfate; this binding was reduced with EDTA treatment of COMP/TSP5. MUT3 showed weaker binding than calcium-repleted COMP/TSP5. Using recombinant COMP/TSP5 fragments, we found that the "signature domain" could bind to aggrecan, suggesting that this domain can mediate the interaction of COMP/TSP5 and aggrecan. In summary, our data indicate that COMP/TSP5 is an aggrecan-binding protein, and this interaction is regulated by the calcium-sensitive conformation of COMP/TSP5; interaction of COMP with aggrecan can be mediated through the GAG side chains on aggrecan and the "signature domain" of COMP/TSP5. Our results suggest that COMP/TSP5 may function to support matrix interactions in cartilage ECM.
Collapse
Affiliation(s)
- Faye Hui Chen
- Cartilage Biology and Orthopaedics Branch, NIAMS, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | |
Collapse
|
45
|
Morozzi G, Fabbroni M, Bellisai F, Pucci G, Galeazzi M. Cartilage Oligomeric Matrix Protein Level in Rheumatic Diseases: Potential Use as a Marker for Measuring Articular Cartilage Damage and/or the Therapeutic Efficacy of Treatments. Ann N Y Acad Sci 2007; 1108:398-407. [PMID: 17894003 DOI: 10.1196/annals.1422.041] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cartilage oligomeric matrix protein (COMP) is a tissue-specific noncollagenous protein that was first detected in the serum and the synovial fluid of patients suffering from rheumatic disorders, such as rheumatoid arthritis, reactive arthritis, juvenile chronic arthritis, and osteoarthritis. In this review, the authors consider serum COMP levels in different diseases and discuss their study of patients with rheumatoid arthritis treated with anti-TNF-alpha, to evaluate whether COMP is able to predict a rapid and sustained clinical response to these drugs. They observe that patients with high COMP levels have a lower ACR 70 response independently of the state of systemic inflammation, and conclude that COMP seems to have a pathogenetic role that is independent of the mechanisms regulating inflammatory processes.
Collapse
Affiliation(s)
- Gabriella Morozzi
- Department of Clinical Medicine and Immunology, Rheumatology Section, University of Siena, Siena, Italy.
| | | | | | | | | |
Collapse
|
46
|
Qi C, Changlin H. Effects of Moving Training on Histology and Biomarkers Levels of Articular Cartilage. J Surg Res 2006; 135:352-63. [PMID: 16904690 DOI: 10.1016/j.jss.2006.03.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2006] [Revised: 02/22/2006] [Accepted: 03/08/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE To study the adaptation process and extent of articular cartilage in the canine knee joint to different modes of movements and to investigate if levels of cartilage oligomeric matrix protein (COMP), matrix metalloproteinases-1 (MMP-1), matrix metalloproteinases-3 (MMP-3), and tissue inhibitor of matrix metalloproteinases-1 (TIMP-1) in serum and synovial fluid can be used to predict effectively early sports injury and remolding degree of articular cartilage in the canine knee. MATERIALS AND METHODS Twenty adult dogs divided randomly into three groups (eight in the common training group, Training Group; eight in the intensified training group, Intensified Group; and four in the Control Group) were trained daily at different intensities. Magnetic resonance imaging (MRI) examinations were performed regularly (0, 2, 4, 6, 8, 10 weeks) to investigate changes of articular cartilage in the canine knee, while concentrations of COMP, MMP-1, MMP-3, and TIMP-1 in serum and synovial fluid were measured by ELISA assays. All of the dogs were euthanized after training for 10 weeks, and all of the knee joints were taken out to be examined histologically. RESULTS We could find imaging changes of early sport injury of articular cartilage in the Training Group and Intensified Group by MRI examination after 2 weeks of training; the damage images were most severe in 4-6 weeks, and then lightened gradually. We could not find the difference of cartilage injury and repair degree in MRI images between these two groups at different time points. Elevations of levels of COMP, MMP-1, MMP-3, TIMP-1, and MMP-3/TIMP-1 in serum and synovial fluid were seen during the training period, and their levels changed remarkably at different times. Levels of MMP-1, MMP-3, and MMP-3/TIMP-1 in the Intensified Group were lower than that in the Training Group in general, and levels of COMP were higher, which hinted that the injury trend of articular cartilage in the Intensified Group was lower than that in the Training group, and the repair trend was higher. Furthermore, there were statistically significant associations between biomarker levels in serum and in synovial fluid. Histological examinations in 10 weeks demonstrated that the signs of cartilage damage and repair in canine knee joint in the Training Group and the Intensified Group were obvious, and the Intensified Group could do better than the Training Group in promoting remodeling reconstruction of articular cartilage. CONCLUSIONS High-intensity and repetitive movement may easily induce sports injury, and it is followed with a repair process; intensified training can do better than common training in promoting remodeling reconstruction of articular cartilage. The sensitivity of these biomarkers reflecting articular cartilage pathological changes is better than MRI, and the associated application of several biomarkers to predict the extent of damage and repair, as well as changes of metabolism in articular cartilage, and to monitor change of disease course has very good value for clinical application.
Collapse
Affiliation(s)
- Chang Qi
- Orthopedics Institute, Xijing Hospital of PLA, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | | |
Collapse
|
47
|
Clements DN, Carter SD, Innes JF, Ollier WER. Genetic basis of secondary osteoarthritis in dogs with joint dysplasia. Am J Vet Res 2006; 67:909-18. [PMID: 16649929 DOI: 10.2460/ajvr.67.5.909] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Affiliation(s)
- Dylan N Clements
- Connective Tissue Research Group, Faculty of Veterinary Science, University of Liverpool, Liverpool, L69 3BX, UK
| | | | | | | |
Collapse
|
48
|
Ishii Y, Thomas AO, Guo XE, Hung CT, Chen FH. Localization and distribution of cartilage oligomeric matrix protein in the rat intervertebral disc. Spine (Phila Pa 1976) 2006; 31:1539-46. [PMID: 16778685 DOI: 10.1097/01.brs.0000221994.61882.4a] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Whole rat intervertebral disc (IVD), as well as the anulus fibrosus (AF) and the nucleus pulposus (NP) were studied using immunoblot, immunohistochemistry, and reverse-transcription followed by polymerase chain reaction (RT-PCR) methods to investigate the expression and distribution of cartilage oligomeric matrix protein (COMP). OBJECTIVES To investigate the expression and distribution patterns of COMP in normal IVD. SUMMARY OF BACKGROUND DATA COMP is an extracellular matrix protein abundantly expressed in articular and growth plate cartilage, as well as bone, ligament, tendon, and synovium. The potential importance of COMP to the spine has been underscored by its mutations that lead to skeletal dysplasia with characteristic platyspondyly. However, the expression and distribution of COMP in spine and IVD has not been illustrated before. METHODS The presence of COMP protein was investigated by immunoblotting using a COMP antibody F8 on protein extractions from whole IVD and AF or NP. To compare the expression levels of COMP between lumbar and tail IVDs, and between AF and NP of the IVD, wet weight of the tissues were used for normalization. To show that COMP can be made by IVD cells in situ, RT-PCR was used to investigate the COMP mRNA message. The distribution patterns of COMP in IVD were investigated using immunohistochemistry studies with COMP antibody F8. RESULTS COMP is expressed at both the protein and mRNA levels in both the AF and NP of both the lumbar spine and tail IVD. Immunohistochemistry studies show that COMP is found in the extracellular matrix of the IVD, exhibiting lamellar distribution pattern in the AF region. When normalized to wet weight, COMP is found to be expressed at higher levels in the lumbar than the tail IVD, and within the IVD, greater in the AF than the NP region. CONCLUSIONS Our results demonstrate the expression of COMP in both the AF and NP of the IVD. COMP is a component of the extracellular matrix of AF and NP, with a lamellar distribution pattern in the AF. Our data suggest that COMP may play a role in the normal structure of IVD.
Collapse
Affiliation(s)
- Yoshimasa Ishii
- Department of Orthopaedic Surgery, Columbia University, New York, NY, USA
| | | | | | | | | |
Collapse
|
49
|
Chen FH, Thomas AO, Hecht JT, Goldring MB, Lawler J. Cartilage oligomeric matrix protein/thrombospondin 5 supports chondrocyte attachment through interaction with integrins. J Biol Chem 2005; 280:32655-61. [PMID: 16051604 PMCID: PMC2896261 DOI: 10.1074/jbc.m504778200] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Cartilage oligomeric matrix protein/thrombospondin 5 (COMP/TSP5) is a major component of the extracellular matrix of the musculoskeletal system. Although COMP/TSP5 abnormalities are associated with several pathological conditions, its normal function remains unclear. This study was undertaken to delineate the function(s) of COMP/TSP5 in cartilage, especially regarding its interaction with chondrocytes. We show that COMP/TSP5 can support chondrocyte attachment and that the RGD sequence in COMP/TSP5 and the integrin receptors alpha5beta1 and alphaVbeta3 on the chondrocytes are involved in mediating this attachment. The interactions of COMP/TSP5 with the integrins are dependent on COMP/TSP5 conformation. Chondrocyte attachment to COMP/TSP5 in the calcium-replete conformation was inhibited by function-blocking integrin alpha5 and beta1 antibodies, suggesting the involvement of the alpha5beta1 integrin. Under this condition, a function-blocking antibody against alphaVbeta3 did not have any effect on cell attachment. On the other hand, chondrocyte attachment to reduced COMP/TSP5 was instead sensitive to alphaVbeta3 function-blocking antibodies, suggesting that COMP/TSP5 mediates attachment through chondrocyte alphaVbeta3 integrin under this condition. Cell attachment to reduced COMP/TSP5 was not inhibited by beta1 antibodies. These data indicate that COMP/TSP5 in different conformations can utilize different integrin receptors. These results are the first to demonstrate that COMP/TSP5 can mediate chondrocyte attachment through interactions with integrins. Through these interactions, COMP/TSP5 may be able to regulate cellular activities and respond to environment in the surrounding cartilage matrix.
Collapse
Affiliation(s)
- Faye Hui Chen
- Department of Orthopaedic Surgery, Columbia University, New York, New York 10032, USA
| | | | | | | | | |
Collapse
|
50
|
Kelman A, Lui L, Yao W, Krumme A, Nevitt M, Lane NE. Association of higher levels of serum cartilage oligomeric matrix protein and N-telopeptide crosslinks with the development of radiographic hip osteoarthritis in elderly women. ACTA ACUST UNITED AC 2005; 54:236-43. [PMID: 16385523 DOI: 10.1002/art.21527] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To examine the association of baseline concentrations of serum cartilage oligomeric matrix protein (COMP) and serum N-telopeptide crosslinks (NTX) with the development and progression of radiographic hip osteoarthritis (RHOA) in elderly women. METHODS Pelvic radiographs were obtained a mean of 8.3 years apart from white women > or =65 years of age enrolled in the Study of Osteoporotic Fractures. Random sampling from a cohort of 5,928 subjects was performed, with subjects ( approximately 200 per group) assigned to nested case-control studies, one focusing on RHOA incidence and the other on RHOA progression. Baseline serum levels of COMP and NTX were measured by enzyme-linked immunosorbent assay in duplicate. Odds ratios (ORs) and 95% confidence intervals (95% CIs), indicating the likelihood of baseline serum COMP and NTX levels to be predictive of the development or progression of RHOA, were calculated using logistic regression analysis, with adjustment for potential covariates. RESULTS At baseline, incident cases of RHOA were associated with higher serum levels of COMP and NTX (P < 0.05 for each) compared with the no RHOA control group. Higher baseline serum COMP and NTX levels were associated with an increased risk of incident RHOA compared with the no RHOA group, with an adjusted OR of 1.31 per SD increase in COMP (95% CI 1.02-1.68) and adjusted OR of 1.38 per SD increase in NTX (95% CI 1.07-1.79). In this community-based cohort, progression of RHOA was modest. However, there was a trend toward increased risk of RHOA progression with higher baseline COMP and NTX levels. CONCLUSION These data suggest that serum levels of COMP and NTX are modest risk markers for the development of RHOA in a community-based cohort of elderly white women.
Collapse
Affiliation(s)
- A Kelman
- Stanford University Medical Center, Stanford, California, USA
| | | | | | | | | | | |
Collapse
|