151
|
Abstract
Osteoarticular disorders are the major cause of disability in Europe and North America. It is estimated that rheumatoid arthritis affects 1 % of the population and that more than two third of people over age 55 develop osteoarthritis. Because there are no satisfactory treatments, gene therapy offers a new therapeutic approach. The delivery of cDNA encoding anti-arthritic proteins to articular cells has shown therapeutic efficacy in numerous animal models in vivo. Through the development and the experimental progresses that have been made for both rheumatoid arthritis and osteoarthritis, this review discusses the different gene therapy strategies available today and the safety issues with which they may be associated. Among the different vectors available today, adeno-associated virus seems the best candidate for a direct in vivo gene delivery approach for the treatment of joint disorders.
Collapse
MESH Headings
- Aged
- Animals
- Arthritis, Rheumatoid/drug therapy
- Arthritis, Rheumatoid/physiopathology
- Arthritis, Rheumatoid/therapy
- Cartilage, Articular/metabolism
- Cartilage, Articular/pathology
- Cytokines/antagonists & inhibitors
- Cytokines/genetics
- DNA, Complementary/administration & dosage
- DNA, Complementary/therapeutic use
- Dependovirus/genetics
- Dogs
- Doxycycline/pharmacology
- Etanercept
- Gene Expression/drug effects
- Genes, Synthetic
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Genetic Vectors/adverse effects
- Genetic Vectors/therapeutic use
- Haplorhini
- Horses
- Humans
- Immunoglobulin G/therapeutic use
- Injections, Intra-Articular
- Mice
- Middle Aged
- Osteoarthritis/physiopathology
- Osteoarthritis/therapy
- Receptors, Tumor Necrosis Factor/therapeutic use
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Sirolimus/pharmacology
Collapse
Affiliation(s)
- Jean-Noël Gouze
- Department of Orthopaedics and Rehabilitation, Gene therapy laboratory, University of Florida, 1600 SW Archer Rd, Rm M2-210, Gainesville, FL 32610, USA.
| | | | | | | |
Collapse
|
152
|
Kim HA, Won Kim D, Park J, Choi SY. Transduction of Cu, Zn-superoxide dismutase mediated by an HIV-1 Tat protein basic domain into human chondrocytes. Arthritis Res Ther 2007; 8:R96. [PMID: 16792821 PMCID: PMC1779403 DOI: 10.1186/ar1972] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2006] [Revised: 05/04/2006] [Accepted: 05/12/2006] [Indexed: 01/01/2023] Open
Abstract
This study was performed to investigate the transduction of a full-length superoxide dismutase (SOD) protein fused to transactivator of transcription (Tat) into human chondrocytes, and to determine the regulatory function of transduced Tat-SOD in the inflammatory cytokine induced catabolic pathway. The pTat-SOD expression vector was constructed to express the basic domain of HIV-1 Tat as a fusion protein with Cu, Zn-SOD. We also purified histidine-tagged SOD without an HIV-1 Tat and Tat-GFP as control proteins. Cartilage samples were obtained from patients with osteoarthritis (OA) and chondrocytes were cultured in both a monolayer and an explant. For the transduction of fusion proteins, cells/explants were treated with a variety of concentrations of fusion proteins. The transduced protein was detected by fluorescein labeling, western blotting and SOD activity assay. Effects of transduced Tat-SOD on the regulation of IL-1 induced nitric oxide (NO) production and inducible nitric oxide synthase (iNOS) mRNA expression was assessed by the Griess reaction and reverse transcriptase PCR, respectively. Tat-SOD was successfully delivered into both the monolayer and explant cultured chondrocytes, whereas the control SOD was not. The intracellular transduction of Tat-SOD into cultured chondrocytes was detected after 1 hours, and the amount of transduced protein did not change significantly after further incubation. SOD enzyme activity increased in a dose-dependent manner. NO production and iNOS mRNA expression, in response to IL-1 stimulation, was significantly down-regulated by pretreatment with Tat-SOD fusion proteins. This study shows that protein delivery employing the Tat-protein transduction domain is feasible as a therapeutic modality to regulate catabolic processes in cartilage. Construction of additional Tat-fusion proteins that can regulate cartilage metabolism favorably and application of this technology in in vivo models of arthritis are the subjects of future studies.
Collapse
Affiliation(s)
- Hyun Ah Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, 896, Pyongchondong, Dongan-gu, Anyang, Kyunggi-do, 431-070, Korea
| | - Dae Won Kim
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Jinseu Park
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Soo Young Choi
- Department of Biomedical Sciences and Research Institute for Bioscience and Biotechnology, Hallym University, Chunchon 200-702, Korea
| |
Collapse
|
153
|
Cucchiarini M, Thurn T, Weimer A, Kohn D, Terwilliger EF, Madry H. Restoration of the extracellular matrix in human osteoarthritic articular cartilage by overexpression of the transcription factor SOX9. ACTA ACUST UNITED AC 2007; 56:158-67. [PMID: 17195218 DOI: 10.1002/art.22299] [Citation(s) in RCA: 121] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Human osteoarthritis (OA) is characterized by a pathologic shift in articular cartilage homeostasis toward the progressive loss of extracellular matrix (ECM). The purpose of this study was to investigate the ability of rAAV-mediated SOX9 overexpression to restore major ECM components in human OA articular cartilage. METHODS We monitored the synthesis and content of proteoglycans and type II collagen in 3-dimensional cultures of human normal and OA articular chondrocytes and in explant cultures of human normal and OA articular cartilage following direct application of a recombinant adeno-associated virus (rAAV) SOX9 vector in vitro and in situ. We also analyzed the effects of this treatment on cell proliferation in these systems. RESULTS Following SOX9 gene transfer, expression levels of proteoglycans and type II collagen increased over time in normal and OA articular chondrocytes in vitro. In situ, overexpression of SOX9 in normal and OA articular cartilage stimulated proteoglycan and type II collagen synthesis in a dose-dependent manner. These effects were not associated with changes in chondrocyte proliferation. Notably, expression of the 2 principal matrix components could be restored in OA articular cartilage to levels similar to those in normal cartilage. CONCLUSION These data support the concept of using direct, rAAV-mediated transfer of chondrogenic genes to articular cartilage for the treatment of OA in humans.
Collapse
|
154
|
Namazi H, Majd Z. Botulinum toxin and osteoarthritis: greater expectations. Med Hypotheses 2007; 68:1188. [PMID: 17257777 DOI: 10.1016/j.mehy.2006.01.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2006] [Revised: 01/24/2006] [Accepted: 01/26/2006] [Indexed: 11/23/2022]
|
155
|
Namazi H. Fasudil: A potential addition to the anti-arthritis weaponry. Med Hypotheses 2007; 68:1425. [PMID: 17257773 DOI: 10.1016/j.mehy.2006.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Accepted: 02/08/2006] [Indexed: 11/19/2022]
|
156
|
Steinert AF, Ghivizzani SC, Rethwilm A, Tuan RS, Evans CH, Nöth U. Major biological obstacles for persistent cell-based regeneration of articular cartilage. Arthritis Res Ther 2007; 9:213. [PMID: 17561986 PMCID: PMC2206353 DOI: 10.1186/ar2195] [Citation(s) in RCA: 236] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Hyaline articular cartilage, the load-bearing tissue of the joint, has very limited repair and regeneration capacities. The lack of efficient treatment modalities for large chondral defects has motivated attempts to engineer cartilage constructs in vitro by combining cells, scaffold materials and environmental factors, including growth factors, signaling molecules, and physical influences. Despite promising experimental approaches, however, none of the current cartilage repair strategies has generated long lasting hyaline cartilage replacement tissue that meets the functional demands placed upon this tissue in vivo. The reasons for this are diverse and can ultimately result in matrix degradation, differentiation or integration insufficiencies, or loss of the transplanted cells and tissues. This article aims to systematically review the different causes that lead to these impairments, including the lack of appropriate differentiation factors, hypertrophy, senescence, apoptosis, necrosis, inflammation, and mechanical stress. The current conceptual basis of the major biological obstacles for persistent cell-based regeneration of articular cartilage is discussed, as well as future trends to overcome these limitations.
Collapse
Affiliation(s)
- Andre F Steinert
- Orthopaedic Center for Musculoskeletal Research, König-Ludwig-Haus, Julius-Maximilians-University, Würzburg, Germany
| | - Steven C Ghivizzani
- Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL, USA
| | - Axel Rethwilm
- Institut für Virologie und Immunbiologie, Julius-Maximilians-University, Würzburg, Germany
| | - Rocky S Tuan
- Cartilage Biology and Orthopaedics Branch, National Institute of Arthritis, and Musculoskeletal and Skin Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, MD, USA
| | | | - Ulrich Nöth
- Orthopaedic Center for Musculoskeletal Research, König-Ludwig-Haus, Julius-Maximilians-University, Würzburg, Germany
| |
Collapse
|
157
|
Namazi H. Botulinum toxin as a novel addition to anti-arthritis armamentarium: An experimental study in rabbits. Int Immunopharmacol 2006; 6:1743-7. [DOI: 10.1016/j.intimp.2006.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2006] [Revised: 07/10/2006] [Accepted: 07/12/2006] [Indexed: 11/26/2022]
|
158
|
Porter RM, Akers RM, Howard RD, Forsten-Williams K. Transcriptional and proteolytic regulation of the insulin-like growth factor-I system of equine articular chondrocytes by recombinant equine interleukin-1beta. J Cell Physiol 2006; 209:542-50. [PMID: 16897751 DOI: 10.1002/jcp.20762] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Interleukin-1 (IL-1) and insulin-like growth factor-I (IGF-I), which have opposing effects on matrix metabolism within articular cartilage, are thought to play prominent roles in the pathogenesis of osteoarthritis. To better understand the link between these anabolic (IGF-I) and catabolic (IL-1) stimuli, we examined exogenous IL-1 regulation of the IGF-I signaling system of articular chondrocytes (ACs). Equine ACs from non-arthritic stifle joints were expanded in monolayer culture, encapsulated for 10 days in alginate beads, and stimulated as high-density monolayers with recombinant equine IL-1beta (0, 1, 10 ng/ml) for 48 h. IL-1beta enhanced expression of IGF-IR levels, as determined by both [125I]-IGF-I binding studies and Western blotting, while reducing the concentration of endogenous IGF-I detected in conditioned media by radioimmunoassay. Western ligand blotting revealed that chondrocytes primarily secreted IGF binding proteins (IGFBPs) with molecular weights of 28-30 and 32-34 kDa, which were identified as IGFBPs 5 and 2, respectively, and that IL-1beta treatment diminished IGFBP-2, the prominent homolog in conditioned media. Northern blot analysis suggested IL-1beta regulation of IGF-I and, to some extent, IGF-IR was mediated by transcription; however, the cytokine did not affect IGFBP-2 expression. To test for evidence of proteolysis by matrix metalloproteinases (MMPs), additional cultures were co-incubated with inhibitors for MMPs 2/9, 3, and 8. IGFBP-2 suppression was partially reversed by gelatinase (MMP-2/9) inhibition. In summary, these findings further delineate the role of IL-1 as a key regulator of the IGF-I system within articular cartilage, demonstrating that regulation occurs through both direct (transcriptional) and indirect (proteolytic) mechanisms.
Collapse
Affiliation(s)
- Ryan M Porter
- Department of Chemical Engineering, Virginia Polytechnic Institute & State University, Blacksburg, Virginia 24061-0211, USA
| | | | | | | |
Collapse
|
159
|
Ishihara A, Zachos TA, Bartlett JS, Bertone AL. Evaluation of permissiveness and cytotoxic effects in equine chondrocytes, synovial cells, and stem cells in response to infection with adenovirus 5 vectors for gene delivery. Am J Vet Res 2006; 67:1145-55. [PMID: 16817735 DOI: 10.2460/ajvr.67.7.1145] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To evaluate host cell permissiveness and cytotoxic effects of recombinant and modified adenoviral vectors in equine chondrocytes, synovial cells, and bone marrow-derived mesenchymal stem cells (BMD-MSCs). SAMPLE POPULATION Articular cartilage, synovium, and bone marrow from 15 adult horses. PROCEDURES Equine chondrocytes, synovial cells, and BMD-MSCs and human carcinoma (HeLa) cells were cultured and infected with an E-1-deficient adenovirus vector encoding the beta-galactosidase gene or the green fluorescent protein gene (Ad-GFP) and with a modified E-1-deficient vector with the arg-gly-asp capsid peptide insertion and containing the GFP gene (Ad-RGD-GFP). Percentages of transduced cells, total and transduced cell counts, and cell viability were assessed 2 and 7 days after infection. RESULTS -Permissiveness to adenoviral vector infection was significantly different among cell types and was ranked in decreasing order as follows: HeLa cells > BMD-MSCs > chondrocytes > synovial cells. Morphologic signs of cytotoxicity were evident in HeLa cells but not in equine cells. Numbers of transduced cells decreased by day 7 in all cell types except equine BMD-MSCs. Transduction efficiency was not significantly different between the Ad-GFP and Ad-RGD-GFP vectors. CONCLUSION AND CLINICAL RELEVANCE Sufficient gene transfer may be achieved by use of an adenovirus vector in equine cells. High vector doses can be used in equine cells because of relative resistance to cytotoxic effects in those cells. Greater permissiveness and sustained expression of transgenes in BMD-MSCs make them a preferential cell target for gene therapy in horses.
Collapse
Affiliation(s)
- Akikazu Ishihara
- Comparative Orthopedic Molecular Medicine Laboratory, Department of Veterinary Clinical Sciences, College of Veterinary Medicine, The Ohio State University, Columbus, 43210, USA
| | | | | | | |
Collapse
|
160
|
|
161
|
Abstract
Focal defects of articular cartilage are an unsolved problem in clinical orthopaedics. These lesions do not heal spontaneously and no treatment leads to complete and durable cartilage regeneration. Although the concept of gene therapy for cartilage damage appears elegant and straightforward, current research indicates that an adaptation of gene transfer techniques to the problem of a circumscribed cartilage defect is required in order to successfully implement this approach. In particular, the localised delivery into the defect of therapeutic gene constructs is desirable. Current strategies aim at inducing chondrogenic pathways in the repair tissue that fills such defects. These include the stimulation of chondrocyte proliferation, maturation, and matrix synthesis via direct or cell transplantation-mediated approaches. Among the most studied candidates, polypeptide growth factors have shown promise to enhance the structural quality of the repair tissue. A better understanding of the basic scientific aspects of cartilage defect repair, together with the identification of additional molecular targets and the development of improved gene-delivery techniques, may allow a clinical translation of gene therapy for cartilage defects. The first experimental steps provide reason for cautious optimism.
Collapse
Affiliation(s)
- Magali Cucchiarini
- Laboratory for Experimental Orthopaedics, Department of Orthopaedic Surgery, Saarland University Medical Center, 66421 Homburg/Saar, Germany
| | | |
Collapse
|
162
|
|
163
|
Abstract
OA is the most common articular disease. Age is its most notable risk factor. With the aging of the population of the developed world, there will be a growing need for better understanding of OA and for superior therapies. There is increasing appreciation for the role of inflammation in OA. There also is the realization that OA is not only a disease of cartilage, but of the entire joint. This has led to increasing interest in structure/disease modification as the goal in OA therapy. However, there are no universally proven structure-/ disease-modifying interventions.
Collapse
Affiliation(s)
- Roy D Altman
- Division of Rheumatology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
164
|
Le Maitre CL, Freemont AJ, Hoyland JA. A preliminary in vitro study into the use of IL-1Ra gene therapy for the inhibition of intervertebral disc degeneration. Int J Exp Pathol 2006; 87:17-28. [PMID: 16436110 PMCID: PMC2517345 DOI: 10.1111/j.0959-9673.2006.00449.x] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Conventional therapies for low back pain (LBP) are purely symptomatic and do not target the cause of LBP, which in approximately 40% of cases is caused by degeneration of the intervertebral disc (DIVD). Targeting therapies to inhibit the process of degeneration would be a potentially valuable treatment for LBP. There is increasing evidence for a role for IL-1 in DIVD. A natural inhibitor of IL-1 exists, IL-1Ra, which would be an ideal molecular target for inhibiting IL-1-mediated effects involved in DIVD and LBP. In this study, the feasibility of ex vivo gene transfer of IL-1Ra to the IVD was investigated. Monolayer and alginate cultures of normal and degenerate human intervertebral disc (IVD) cells were infected with an adenoviral vector carrying the IL-1Ra gene (Ad-IL-1Ra) and protein production measured using an enzyme-linked immunosorbent assay. The ability of these infected cells to inhibit the effects of IL-1 was also investigated. In addition, normal and degenerate IVD cells infected with Ad-IL-1Ra were injected into degenerate disc tissue explants and IL-1Ra production in these discs was assessed. This demonstrated that both nucleus pulposus and annulus fibrosus cells infected with Ad-IL-1Ra produced elevated levels of IL-1Ra for prolonged time periods, and these infected cells were resistant to IL-1. When the infected cells were injected into disc explants, IL-1Ra protein expression was increased which was maintained for 2 weeks of investigation. This in vitro study has shown that the use of ex vivo gene transfer to degenerate disc tissue is a feasible therapy for the inhibition of IL-1-mediated events during disc degeneration.
Collapse
Affiliation(s)
- Christine L Le Maitre
- Division of Laboratory and Regenerative Medicine, School of Medicine, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
165
|
Pelletier JP, Martel-Pelletier J, Raynauld JP. Most recent developments in strategies to reduce the progression of structural changes in osteoarthritis: today and tomorrow. Arthritis Res Ther 2006; 8:206. [PMID: 16569256 PMCID: PMC1526599 DOI: 10.1186/ar1932] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA), the most common of all arthritic conditions, is a social and financial burden to all nations. The most recent research has significantly advanced our understanding of the cause of OA and risk factors associated with it. These findings have provided useful information that has helped in the daily management of patients with OA. Some preventative measures and a number of therapeutic agents and drugs are available, which may help to reduce the progression of OA in certain patients. Moreover, the most recent progress in research has significantly enhanced our knowledge of the factors involved in the development of the disease and of the mechanisms responsible for its progression. This has allowed identification of several new therapeutic targets in a number of pathophysiological pathways. Consequently, the field is opening up to a new era in which drugs and agents that can specifically block important mechanisms responsible for the structural changes that occur in OA can be brought into development and eventually into clinical trials.
Collapse
Affiliation(s)
- Jean-Pierre Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Centre, Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Johanne Martel-Pelletier
- Osteoarthritis Research Unit, University of Montreal Hospital Centre, Notre-Dame Hospital, Montreal, Quebec, Canada
| | - Jean-Pierre Raynauld
- Osteoarthritis Research Unit, University of Montreal Hospital Centre, Notre-Dame Hospital, Montreal, Quebec, Canada
| |
Collapse
|
166
|
Zhang X, Yu C, Zhang C, Tang T, Dai K. Direct chitosan-mediated gene delivery to the rabbit knee joints in vitro and in vivo. Biochem Biophys Res Commun 2006; 341:202-8. [PMID: 16413501 DOI: 10.1016/j.bbrc.2005.12.171] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Accepted: 12/26/2005] [Indexed: 11/24/2022]
Abstract
Chitosan vector system is expected to be useful for direct gene therapy for joint disease. This study first sought to confirm that foreign genes can be transferred to articular chondrocytes in primary culture. Next, chitosan-DNA nanoparticles containing IL-1Ra or IL-10 gene were injected directly into the knee joint cavities of osteoarthritis rabbits to clarify the in vivo transfer availability of the chitosan vectors. Clear expression of IL-1Ra was detected in the knee joint synovial fluid of the chitosan IL-1Ra-injected group. While no expression was detected in the chitosan IL-10-injected group, this demonstrates that the transfection efficiency of chitosan-DNA nanoparticles was closely related to the type of the gene product. A significant reduction was also noted in the severity of histologic cartilage lesions in the group that received the chitosan IL-1Ra injection. This avenue may therefore represent a promising future treatment for osteoarthritis.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Institute of Health Sciences, Shanghai Jiao Tong University, School of Medicine and Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, PR China.
| | | | | | | | | |
Collapse
|
167
|
Young AA, McLennan S, Smith MM, Smith SM, Cake MA, Read RA, Melrose J, Sonnabend DH, Flannery CR, Little CB. Proteoglycan 4 downregulation in a sheep meniscectomy model of early osteoarthritis. Arthritis Res Ther 2006; 8:R41. [PMID: 16469119 PMCID: PMC1526595 DOI: 10.1186/ar1898] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2005] [Revised: 01/10/2006] [Accepted: 01/12/2006] [Indexed: 12/02/2022] Open
Abstract
Osteoarthritis is a disease of multifactorial aetiology characterised by progressive breakdown of articular cartilage. In the early stages of the disease, changes become apparent in the superficial zone of articular cartilage, including fibrillation and fissuring. Normally, a monolayer of lubricating molecules is adsorbed on the surface of cartilage and contributes to the minimal friction and wear properties of synovial joints. Proteoglycan 4 is the lubricating glycoprotein believed to be primarily responsible for this boundary lubrication. Here we have used an established ovine meniscectomy model of osteoarthritis, in which typical degenerative changes are observed in the operated knee joints at three months after surgery, to evaluate alterations in proteoglycan 4 expression and localisation in the early phases of the disease. In normal control joints, proteoglycan 4 was immunolocalised in the superficial zone of cartilage, particularly in those regions of the knee joint covered by a meniscus. After the onset of early osteoarthritis, we demonstrated a loss of cellular proteoglycan 4 immunostaining in degenerative articular cartilage, accompanied by a significant (p < 0.01) decrease in corresponding mRNA levels. Early loss of proteoglycan 4 from the cartilage surface in association with a decrease in its expression by superficial-zone chondrocytes might have a role in the pathogenesis of osteoarthritis.
Collapse
Affiliation(s)
- Allan A Young
- Raymond Purves Research Laboratory, Institute of Bone and Joint Research, Royal North Shore Hospital, University of Sydney, Pacific Highway, St Leonards, NSW 2065, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
168
|
Varoga D, Paulsen FP, Kohrs S, Grohmann S, Lippross S, Mentlein R, Tillmann BN, Goldring MB, Besch L, Pufe T. Expression and regulation of human β-defensin-2 in osteoarthritic cartilage. J Pathol 2006; 209:166-73. [PMID: 16622898 DOI: 10.1002/path.1974] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Defensins are antibiotic peptides that are involved in host defence at epithelial and mesenchymal surfaces. Previous studies have shown the induction of human beta-defensin-3 (HBD-3) in osteoarthritic joints, suggesting that these molecules have functions in addition to their ability to kill microbes. The aim of this study was to investigate the production of a further human beta-defensin, named HBD-2, in osteoarthritis (OA) and to determine its regulation by inflammatory cytokines. Healthy and osteoarthritic cartilage was assessed for HBD-2 expression by RT-PCR, immunohistochemistry, and ELISA. C28/I2 chondrocytes, primary chondrocytes, and cartilage explants were cultured for in vitro studies. After 24 h of stimulation with tumour necrosis factor-alpha (TNF-alpha), interleukin-1 (IL-1) or IL-6, real-time RT-PCR and ELISA experiments were performed to evaluate the effect of these cytokines on the production of HBD-2. In contrast to healthy cartilage, HBD-2 expression was identified in most of the OA samples examined (eight of ten). Cytokines that are potentially involved in the pathogenesis of OA, namely TNF-alpha, IL-1, and IL-6, were transcriptional inducers of HBD-2 in cultured chondrocytes and cartilage explants in vitro, as measured by real-time RT-PCR and ELISA. These results illustrate the induction of HBD-2 in osteoarthritic cartilage and suggest that it is a further factor in the pathogenesis of OA. However, further studies are required to elucidate the role played by HBD-2 in osteoarthritic cartilage.
Collapse
Affiliation(s)
- D Varoga
- Department of Orthopaedic and Trauma Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
169
|
Jacques C, Gosset M, Berenbaum F, Gabay C. The role of IL-1 and IL-1Ra in joint inflammation and cartilage degradation. VITAMINS AND HORMONES 2006; 74:371-403. [PMID: 17027524 DOI: 10.1016/s0083-6729(06)74016-x] [Citation(s) in RCA: 131] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Interleukin (IL)-1 is a cytokine that plays a major role in inflammatory responses in the context of infections and immune-mediated diseases. IL-1 refers to two different cytokines, termed IL-1alpha and IL-1beta, produced from two genes. IL-1alpha and IL-1beta are produced by different cell types following stimulation by bacterial products, cytokines, and immune complexes. Monocytes/macrophages are the primary source of IL-1beta. Both cytokines do not possess leader peptide sequences and do not follow a classical secretory pathway. IL-1alpha is mainly cell associated, whereas IL-1beta can be released from activated cells after cleavage of its amino-terminal region by caspase-1. IL-1 is present in the synovial tissue and fluids of patients with rheumatoid arthritis. Several in vitro studies have shown that IL-1 stimulates the production of mediators such as prostaglandin E(2), nitric oxide, cytokines, chemokines, and adhesion molecules that are involved in articular inflammation. Furthermore, IL-1 stimulates the synthesis and activity of matrix metalloproteinases and other enzymes involved in cartilage destruction in rheumatoid arthritis and osteoarthritis. The effects of IL-1 are inhibited in vitro and in vivo by natural inhibitors such as IL-1 receptor antagonist and soluble receptors. IL-1 receptor antagonist belongs to the IL-1 family of cytokines and binds to IL-1 receptors but does not induce any intracellular response. IL-1 receptor antagonist inhibits the effect of IL-1 by blocking its interaction with cell surface receptors. The use of IL-1 inhibitors in experimental models of inflammatory arthritis and osteoarthritis has provided a strong support for the role of IL-1 in the pathogeny of these diseases. Most importantly, these findings have been confirmed in clinical trials in patients with rheumatic diseases. Additional strategies aimed to block the effect of IL-1 are tested in clinical trials.
Collapse
Affiliation(s)
- Claire Jacques
- UMR 7079 CNRS, Physiology and Physiopathology Laboratory, University Paris 6, Paris, 75252 Cedex 5, France
| | | | | | | |
Collapse
|
170
|
Chowdhury TT, Bader DL, Lee DA. Anti-inflammatory effects of IL-4 and dynamic compression in IL-1β stimulated chondrocytes. Biochem Biophys Res Commun 2006; 339:241-7. [PMID: 16297873 DOI: 10.1016/j.bbrc.2005.11.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 11/02/2005] [Indexed: 10/25/2022]
Abstract
Mechanical loading can counteract inflammatory pathways induced by IL-1beta by inhibiting *NO and PGE2, catabolic mediators known to be involved in cartilage degradation. The current study investigates the potential of dynamic compression, in combination with the anti-inflammatory cytokine, IL-4, to further abrogate the IL-1beta induced effects. The data presented demonstrate that IL-4 alone can inhibit nitrite release in the presence and absence of IL-1beta and partially reverse the IL-1beta induced PGE2 release. When provided in combination, IL-4 and dynamic compression could further abrogate the IL-1beta induced nitrite and PGE2 release. IL-1beta inhibited [3H]thymidine incorporation and this effect could be reversed by IL-4 or dynamic strain alone or both in combination. By contrast, 35SO4 incorporation was not influenced by IL-4 and/or dynamic strain in IL-1beta stimulated constructs. IL-4 and mechanical loading may therefore provide a potential protective mechanism for cartilage destruction as observed in OA.
Collapse
Affiliation(s)
- T T Chowdhury
- Department of Engineering, Queen Mary, University of London, Mile End Road, London E1 4NS, UK.
| | | | | |
Collapse
|
171
|
|
172
|
Moore EE, Bendele AM, Thompson DL, Littau A, Waggie KS, Reardon B, Ellsworth JL. Fibroblast growth factor-18 stimulates chondrogenesis and cartilage repair in a rat model of injury-induced osteoarthritis. Osteoarthritis Cartilage 2005; 13:623-31. [PMID: 15896984 DOI: 10.1016/j.joca.2005.03.003] [Citation(s) in RCA: 241] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2004] [Accepted: 03/02/2005] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is the most common form of arthritis and a primary cause of disability, however, there are no treatments that can slow disease progression or repair damaged joint cartilage. Fibroblast growth factor-18 (FGF18) has been reported to have significant anabolic effects on cartilage. We therefore examined its effects on repair of cartilage damage in a rat meniscal tear model of OA. DESIGN Surgical damage to the meniscus in rats leads to joint instability and significant damage to the articular cartilage at 3 weeks post-surgery. At this time, animals received bi-weekly intra-articular injections of FGF18 for 3 weeks, and the knee joints were then harvested for histologic examination. RESULTS FGF18-induced dose-dependent increases in cartilage thickness of the tibial plateau, due to new cartilage formation at the articular surface and the joint periphery. The generation of new cartilage resulted in significant reductions in cartilage degeneration scores. The highest dose of FGF18 also induced an increase in chondrophyte size and increased remodeling of the subchondral bone. CONCLUSIONS The results of this study demonstrate that FGF18 can stimulate repair of damaged cartilage in a setting of rapidly progressive OA in rats.
Collapse
Affiliation(s)
- E E Moore
- ZymoGenetics, Inc., 1201 Eastlake Avenue East, Seattle, WA 98102, USA
| | | | | | | | | | | | | |
Collapse
|
173
|
Henrotin Y, Sanchez C, Balligand M. Pharmaceutical and nutraceutical management of canine osteoarthritis: present and future perspectives. Vet J 2005; 170:113-23. [PMID: 15993795 DOI: 10.1016/j.tvjl.2004.08.014] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2004] [Indexed: 12/01/2022]
Abstract
Osteoarthritis (OA) is one of the most common chronic musculoskeletal diseases and causes of lameness in the dogs. The osteoarthritic disease process involves the entire synovial joint, encompassing the synovium, cartilage and underlying bone. Joint failure results from an abnormal mechanical strain causing damage to normal tissue or failure of pathologically impaired articular cartilage and bone under the influence of normal physiological strain or a combination of both. In both cases, the end point is cartilage loss and joint impairment. Osteoarthritic chondrocytes show an altered phenotype characterised by an excess production of catabolic factors, including metalloproteinases and reactive oxygen species. These factors constitute potential therapeutic targets and some new drugs and nutraceuticals have been proposed to promote the return to homeostasis. Until now, the therapeutic management of OA in dogs has been dominated by nonsteroidal anti-inflammatory drugs, but some new compounds, including diacerhein, with potential structure-modifying effects, are already used to treat OA in humans and could be helpful to manage OA in the dog. In addition, novel nutraceuticals, such as avocado/soybean unsaponifiable substances, have shown symptomatic effects in knee OA in humans, and could offer an alternative to prevent OA progression. This paper provides an overview of recent discoveries in the pathophysiology and in the therapeutic management of osteoarthritis in dogs.
Collapse
Affiliation(s)
- Yves Henrotin
- Bone and Cartilage Research Unit, Institute of Pathology, Level +5, CHU Sart-Tilman, 4000 Liège, Belgium.
| | | | | |
Collapse
|
174
|
Evans CH, Ghivizzani SC, Herndon JH, Robbins PD. Gene therapy for the treatment of musculoskeletal diseases. J Am Acad Orthop Surg 2005; 13:230-42. [PMID: 16112980 DOI: 10.5435/00124635-200507000-00003] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Research into the orthopaedic applications of gene therapy has resulted in progress toward managing chronic and acute genetic and nongenetic disorders. Gene therapy for arthritis, the original focus of research, has progressed to the initiation of several phase I clinical trials. Preliminary findings support the application of gene therapy in the treatment of additional chronic conditions, including osteoporosis and aseptic loosening, as well as musculoskeletal tumors. The most rapid progress is likely to be in tissue repair because it requires neither long-term transgene expression nor closely regulated levels of transgene expression. Moreover, healing probably can be achieved with existing technology. In preclinical studies, genetically modulated stimulation of bone healing has shown impressive results in repairing segmental defects in the long bones and cranium and in improving the success of spinal fusions. An increasing amount of evidence indicates that gene transfer can aid the repair of articular cartilage, menisci, intervertebral disks, ligaments, and tendons. These developments have the potential to transform many areas of musculoskeletal care, leading to treatments that are less invasive, more effective, and less expensive than existing modalities.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopaedics, Department of Orthopaedic Surgery, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
175
|
Riyazi N, Slagboom E, de Craen AJM, Meulenbelt I, Houwing-Duistermaat JJ, Kroon HM, van Schaardenburg D, Rosendaal FR, Breedveld FC, Huizinga TWJ, Kloppenburg M. Association of the risk of osteoarthritis with high innate production of interleukin-1beta and low innate production of interleukin-10 ex vivo, upon lipopolysaccharide stimulation. ACTA ACUST UNITED AC 2005; 52:1443-50. [PMID: 15880595 DOI: 10.1002/art.21014] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
OBJECTIVE In a sibpair study of osteoarthritis (OA) patients, we investigated whether, upon stimulation with lipopolysaccharide (LPS), variations in the innate ex vivo production of interleukin-1beta (IL-1beta), IL-1 receptor antagonist (IL-1Ra), IL-10, and tumor necrosis factor alpha (TNFalpha) in whole-blood assays contribute to the risk of OA. METHODS Data from 305 patients with OA at multiple sites (hand, knee, hip, and spine), whose median age was 60 years (range 43-79 years), were compared with those from 137 controls. OA was defined in accordance with the American College of Rheumatology criteria. Whole-blood samples were stimulated with LPS (10 ng/ml). In the supernatants, cytokines were measured by enzyme-linked immunosorbent assay. Odds ratios (ORs) were used as measures of the relative risk of OA in relation to quartiles of IL-1beta, IL-1Ra, TNFalpha, and IL-10 production. The ORs were adjusted for sex and age, and 95% confidence intervals (95% CIs) were computed using robust standard errors to take into account the intrafamily effect. RESULTS Subjects in the highest quartile of IL-1beta and IL-1Ra had an increased risk of OA (OR 3.3, 95% CI 1.4-7.9 and OR 8.0, 95% CI 3.7-17.4, respectively), while subjects in the lowest quartile of IL-10 had a 3-fold increased risk of OA (OR 3.1, 95% CI 1.5-6.5). High innate ex vivo production of TNFalpha was not associated with an increased risk of OA. CONCLUSION Subjects with a high innate ex vivo production of IL-1beta and IL-1Ra and low innate ex vivo production of IL-10 have an increased risk of OA. These results suggest that a proportion of the genetic susceptibility to OA may be encoded for by variations in innate cytokine activity.
Collapse
Affiliation(s)
- Naghmeh Riyazi
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
176
|
Frisbie DD, McIlwraith CW. Gene therapy: future therapies in osteoarthritis. Vet Clin North Am Equine Pract 2005; 17:233-43, vi. [PMID: 15658173 DOI: 10.1016/s0749-0739(17)30059-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
The field of equine veterinary practice is in an ever-evolving state, requiring current technologies to be constantly evaluated for new applications. The specific use of gene therapy in the horse is a novel application. The authors want to help familiarize the equine practitioner with the concept of gene therapy, and introduce its use and potential future benefits for the equine industry in the treatment of osteoarthritis.
Collapse
Affiliation(s)
- D D Frisbie
- Department of Clinical Sciences, Colorado State University College of Veterinary Medicine and Biomedical Sciences, Fort Collins, Colorado 80524, USA.
| | | |
Collapse
|
177
|
Abstract
Osteoarthritis is a most common health problem in population over age 40 and a leading cause of pain and disability in the United States. Treatments of osteoarthritis incorporate combination of nonpharmacologic modalities, pharmacologic agents and surgical procedures. Unfortunately nonpharmacological modalities are underutilized in the management algorithm. In addition to physical and occupational therapy, diet and exercise play an extremely important role, thus patient education in the above areas is of great importance. Pharmacological measures should be examined carefully by physician and the patient weighting its existing risks and benefits. Surgical procedures are generally reserved for patients with severe arthritis who have persistent aim and significantly reduced function. Presently, there are no proven structure/disease-modifying interventions and therefore current therapy is aimed to symptom relief and rehabilitation.
Collapse
Affiliation(s)
- Elena V Barnes
- Division of Rheumatology, Department of Medicine, University of Florida, Gainesville 32610, USA.
| | | |
Collapse
|
178
|
Abstract
Osteoarthritis is common, incurable and difficult to treat. Because osteoarthritis is symptomatic only in a limited number of weight-bearing joints and lacks obvious extra-articular manifestations, it is well suited to local therapy administered by intra-articular injection. Several biologically based, local therapies of this type are either in clinical use or in development. Intra-articular injections of hyaluronic acid are widely used, but are highly controversial because their mode of action is unclear and clinical trials have provided contradictory results. The conclusions of meta-analyses are also discordant. An alternative therapy, based on the intra-articular injection of autologous conditioned serum, is used in Europe. This product, known as Orthokine, is generated by incubating venous blood with etched glass beads. In this way, peripheral blood leukocytes produce elevated amounts of the interleukin-1 receptor antagonist and other anti-inflammatory mediators that are recovered in the serum. Considerable symptomatic relief has been reported in clinical trials of this product. Alternatively, instead of injecting a heterogeneous, incompletely characterized mixture of native molecules into the joint, it is possible to inject recombinant growth factors and cytokine antagonists. None of these are in routine clinical use, but promising preliminary human trials have been performed with insulin-like growth factor-1 and the interleukin-1 receptor antagonist. It is possible that sustained intra-articular production of such factors could be achieved by gene transfer. Although gene therapy for osteoarthritis is not yet a clinical reality, the first human trial should begin next year.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopaedics, Harvard Medical School, Boston, Massachusetts 02115, USA.
| |
Collapse
|
179
|
Venkatesan N, Barré L, Benani A, Netter P, Magdalou J, Fournel-Gigleux S, Ouzzine M. Stimulation of proteoglycan synthesis by glucuronosyltransferase-I gene delivery: a strategy to promote cartilage repair. Proc Natl Acad Sci U S A 2004; 101:18087-92. [PMID: 15601778 PMCID: PMC535800 DOI: 10.1073/pnas.0404504102] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Osteoarthritis is a degenerative joint disease characterized by a progressive loss of articular cartilage components, mainly proteoglycans (PGs), leading to destruction of the tissue. We investigate a therapeutic strategy based on stimulation of PG synthesis by gene transfer of the glycosaminoglycan (GAG)-synthesizing enzyme, beta1,3-glucuronosyltransferase-I (GlcAT-I) to promote cartilage repair. We previously reported that IL-1beta down-regulated the expression and activity of GlcAT-I in primary rat chondrocytes. Here, by using antisense oligonucleotides, we demonstrate that GlcAT-I inhibition impaired PG synthesis and deposition in articular cartilage explants, emphasizing the crucial role of this enzyme in PG anabolism. Thus, primary chondrocytes and cartilage explants were engineered by lipid-mediated gene delivery to efficiently overexpress a human GlcAT-I cDNA. Interestingly, GlcAT-I overexpression significantly enhanced GAG synthesis and deposition as evidenced by (35)S-sulfate incorporation, histology, estimation of GAG content, and fluorophore-assisted carbohydrate electrophoresis analysis. Metabolic labeling and Western blot analyses further suggested that GlcAT-I expression led to an increase in the abundance rather than in the length of GAG chains. Importantly, GlcAT-I delivery was able to overcome IL-1beta-induced PG depletion and maintain the anabolic activity of chondrocytes. Moreover, GlcAT-I also restored PG synthesis to a normal level in cartilage explants previously depleted from endogenous PGs by IL-1beta-treatment. In concert, our investigations strongly indicated that GlcAT-I was able to control and reverse articular cartilage defects in terms of PG anabolism and GAG content associated with IL-1beta. This study provides a basis for a gene therapy approach to promote cartilage repair in degenerative joint diseases.
Collapse
Affiliation(s)
- N Venkatesan
- Unité Mixte de Recherche 7561, Centre National de la Recherche Scientifique, Université Henri Poincaré-Nancy 1, Faculté de Médecine, B.P. 184, 54505 Vanoeuvre-lès-Nancy, France
| | | | | | | | | | | | | |
Collapse
|
180
|
Abstract
We review progress in the field of orthopaedic gene therapy since the concept of using gene transfer to address orthopaedic problems was initiated approximately 15 years ago. The original target, arthritis, has been the subject of two successful Phase I clinical trials, and additional human studies are pending in rheumatoid arthritis and osteoarthritis. The repair of damaged musculoskeletal tissues also has proved to be a fruitful area of research, and impressive enhancement of bone healing has been achieved in preclinical models. Rapid progress also is being made in the use of gene transfer to improve cartilage repair, ligament healing, and restoration of various additional tissues, including tendon and meniscus. Other applications include intervertebral disc degeneration, aseptic loosening, osteoporosis, genetic diseases, and orthopaedic tumors. Of these various orthopaedic targets of gene therapy, tissue repair is likely to make the earliest clinical impact because it can be achieved with existing technology. Tissue repair may become one of the earliest clinical successes for gene therapy as a whole. Orthopaedics promises to be a leading discipline for the use of human gene therapy.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopaedics, Harvard Medical School, 221 Longwood Avenue, BL1-152, Boston, MA 02025, USA.
| | | | | |
Collapse
|
181
|
Affiliation(s)
- Gururaj A Rao
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL 32610, USA
| | | |
Collapse
|
182
|
Abstract
Chondrocytes are the single cellular component of hyaline cartilage. Under physiologic conditions, they show steady-state equilibrium between anabolic and catabolic activities that maintains the structural and functional integrity of the cartilage extracellular matrix. Implicit in the loss of cartilage matrix that is associated with osteoarthritis is that there is a disturbance in the regulation of synthetic (anabolic) and resorptive (catabolic) activities of the resident chondrocytes that results in a net loss of cartilage matrix components and deterioration in the structural and functional properties of the cartilage. Multiple mechanisms likely are involved in the disturbance of chondrocyte remodeling activities in OA. They include the development of acquired or age-related alterations in chondrocyte function, the effects of excessive mechanical loading, and the presence of dysregulated cytokine activities. Cytokines are soluble or cell-surface molecules that play an essential role in mediating cell-cell interactions. It is possible to classify the cytokines that regulate cartilage remodeling as catabolic, acting on target cells to increase products that enhance matrix degradation; as anticatabolic, tending to inhibit or antagonize the activity of the catabolic cytokines; and as anabolic, acting on chondrocytes to increase synthetic activity. This review will focus on the role of proinflammatory cytokines and their roles in mediating the increased matrix degradation that characterizes the osteoarthritic cartilage lesion.
Collapse
Affiliation(s)
- Steven R Goldring
- Rheumatology Division, Beth Israel Deaconess Medical Center, Harvard Institute of Medicine, Boston, MA 02115, USA.
| | | |
Collapse
|
183
|
Affiliation(s)
- Jean-Pierre Pelletier
- Osteoarthritis Research Unit, Centre Hospitalier de l'Université de Montréal, Hôpital Notre-Dame, Montreal, Quebec, Canada.
| | | |
Collapse
|
184
|
|
185
|
Salini V, Orso C. The Present and the Future of Cartilaginous Repair. EUR J INFLAMM 2004. [DOI: 10.1177/1721727x0400200301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The treatment of cartilage lesions constitutes one of the most binding challenges in the field of orthopaedics due to the fact that articular cartilage possesses a very limited intrinsic reparative capacity because of its scarse vascular content. There have been many attempts to obtain reconstruction with longeval hyaline cartilage, but none have obtained satisfactory results. One of the most promising procedures is tissue engineering coupled with gene therapy. This method renders it possible to obtain, for a prolonged period, high gene expression in the area of the lesion. Recent studies have demonstrated how, not only chondrocytes, but muscle derived cells as well, possess the capacity to transport genes in the location of the lesion. It has also been noted recently that between muscle derived cells there exist staminal cells which are possible candidates for gene therapy for the treatment of chondral lesion.
Collapse
Affiliation(s)
- V. Salini
- Department of Clinical Orthopaedics and Traumatology University “G. D'Annunzio”, Chieti, Italy
| | - C.A. Orso
- Department of Clinical Orthopaedics and Traumatology University “G. D'Annunzio”, Chieti, Italy
| |
Collapse
|
186
|
Zhang X, Mao Z, Yu C. Suppression of early experimental osteoarthritis by gene transfer of interleukin-1 receptor antagonist and interleukin-10. J Orthop Res 2004; 22:742-50. [PMID: 15183429 DOI: 10.1016/j.orthres.2003.12.007] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2003] [Accepted: 12/04/2003] [Indexed: 02/04/2023]
Abstract
Gene therapy offers a radically different approach to the treatment of arthritis. We demonstrated that cDNA coding for human interleukin-1 receptor-antagonist protein (IL-1Ra) and cDNA coding for human interleukin-10 (IL-10) can be delivered, by ex vivo techniques, to the synovial lining of joints, intra-articular expression of gene significantly reduced cartilage matrix degradation and cartilage breakdown. To achieve this, lapine synoviocytes were first transduced in culture by retroviral infection. The genetically modified synoviocytes were then transplanted by intra-articular injection into the knee joints of OA rabbits, assay of joint lavages confirmed that the gene expression was not lost 14 days after transfer. Knees receiving the IL-1Ra gene had significantly reduced cartilage breakdown. Delivery of the IL-10 gene was less effective, having only a moderate effect on cartilage breakdown. When both genes were injected together, there was a greater inhibition of cartilage breakdown, suggesting that simultaneous gene delivery may be necessary to treat OA by targeting the activities of multiple inflammatory effectors.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Institute of Sports Medicine, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing 100083, China
| | | | | |
Collapse
|
187
|
Van den Berg WB. Les mécanismes d’action de la diacerhéine, premier inhibiteur de l’interleukine 1 dans l’arthrose. Presse Med 2004. [DOI: 10.1016/s0755-4982(04)98702-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
188
|
Dumond H, Presle N, Pottie P, Pacquelet S, Terlain B, Netter P, Gepstein A, Livne E, Jouzeau JY. Site specific changes in gene expression and cartilage metabolism during early experimental osteoarthritis. Osteoarthritis Cartilage 2004; 12:284-95. [PMID: 15023380 DOI: 10.1016/j.joca.2003.11.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2003] [Accepted: 11/24/2003] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To characterize the molecular events underlying cartilage injury in the early phase of mono-iodoacetate-induced osteoarthritis (OA) in rats. METHODS Experimental osteoarthritis was induced by intra-articular injection of 0.03mg mono-iodoacetate (MIA) in Wistar rats. Animals were killed 2, 5, 10, 15 and 20 days post-injection. Synovial tissue and standardized biopsies from different areas of knee cartilage were examined. Proteoglycan synthesis ((35)S incorporation) and gelatinase activities (zymography), semi-quantitative RT-PCR and immunohistochemistry for IL1beta, iNOS, COX2 and PPARgamma, were performed on these samples. RESULTS Changes in proteoglycan synthesis and gelatinase activities were time and site-dependent. Proteoglycan synthesis inhibition was maximal by day 2 while the highest gelatinase activities were observed at day 5. Central part of patella and posterior plateaus and condyles, i.e. the weight-bearing cartilage areas, were the most affected. IL1beta and iNOS transcripts were induced early in cartilage at time of maximal proteoglycan synthesis inhibition, especially in weight-bearing areas. COX-2 was slightly up-regulated whereas PPARgamma gene expression remained unchanged. Gene expression profile in synovium paralleled that of cartilage, except for PPARgamma which was up-regulated at day 15 and 20. Immunostaining for IL1beta and iNOS showed that proteins were located in diseased cartilage areas at early stage of the experimental OA (day 2). At later time-points (day 20), IL1beta and iNOS were expressed in perilesional areas whereas immunostaining became below control level for COX-2 and PPARgamma. CONCLUSIONS Time-dependent degradation of cartilage after injection of low dose of MIA (0.03mg) into rat knee joint can be related to early loss of proteoglycan anabolism, increased gelatinase activities and expression of IL1beta and downstream inflammatory genes. Increased susceptibility to MIA in weight-bearing areas of cartilage further indicate that MIA-induced experimental OA is a relevant model to study not only metabolical but also biomechanical aspects of human OA.
Collapse
Affiliation(s)
- H Dumond
- Laboratoire de Pharmacologie et Physiopathologie articulaires, UMR CNRS-UHP 7561, Faculté de Médecine, Avenue de la Forêt de Haye, BP 184, 54505 Vandoeuvre-lès-Nancy, France
| | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Moulharat N, Lesur C, Thomas M, Rolland-Valognes G, Pastoureau P, Anract P, De Ceuninck F, Sabatini M. Effects of transforming growth factor-beta on aggrecanase production and proteoglycan degradation by human chondrocytes in vitro. Osteoarthritis Cartilage 2004; 12:296-305. [PMID: 15023381 DOI: 10.1016/j.joca.2003.11.009] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2003] [Accepted: 11/25/2003] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Aggrecan is degraded by Aggrecanases (ADAMTS-4 and -5) and MMPs, which cleave its core protein at different sites. Transforming growth factor (TGF)beta is known to stimulate matrix formation in cartilage, and ADAMTS-4 production in synoviocytes. The aim of this in-vitro study was to examine the effects of TGFbeta on aggrecanase production in human cartilage. DESIGN Expression of ADAMTS-4 and -5 in chondrocyte cultures from normal or osteoarthritic cartilage was studied at mRNA level by RT-PCR. Aggrecanase activity was examined by western blot of aggrecanase-generated neoepitope NITEGE, and by measure of proteoglycan degradation in cartilage explants. RESULTS TGFbeta strongly increased mRNA levels of ADAMTS-4, while ADAMTS-5 was expressed in a constitutive way in chondrocytes from normal and osteoathritic cartilage. TGFbeta also increased NITEGE levels and proteoglycan degradation. Addition of an aggrecanase inhibitor blocked the increase of NITEGE, and partially inhibited proteoglycan degradation. CONCLUSIONS TGFbeta stimulates ADAMTS-4 expression and aggrecan degradation in cartilage. This catabolic action seems to be partially mediated by aggrecanases. It is, therefore, proposed that the role of TGFbeta in cartilage matrix turnover is not limited to anabolic and anti-catabolic actions, but also extends to selective degradation of matrix components such as aggrecan.
Collapse
Affiliation(s)
- N Moulharat
- Division of Rheumatology, Institut de Recherches Servier, Suresnes, France
| | | | | | | | | | | | | | | |
Collapse
|
190
|
Affiliation(s)
- Shital N Parikh
- Division of Pediatric Orthopedic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio 45229, USA
| |
Collapse
|
191
|
Abstract
Osteoarthritis is the most common form of arthritis. The condition is characterised by loss or failure of the functional and/or biochemical integrity of the joint. The clinical symptoms include joint stiffness, pain and dysfunction, but the principal problem for the majority of patients is the pain. Although there are no pain receptors in the cartilage, the origin of the pain is thought to be due to stimulation of the A delta mechanoreceptors and the C polymodal nerve endings in the synovium and surrounding tissues. However, some of the pain experienced in and around the joints is referred pain or sympathetic efferent pain. In addition, there is a poor correlation of clinical symptoms with radiological or imaging appearance. This lack of correlation of clinical evaluation and imaging makes attempts at treatment difficult and compromises attempts to design studies and to evaluate the outcome of osteoarthritis in clinical trials.
Collapse
Affiliation(s)
- W F Kean
- Hamilton Health Sciences, McMaster University, Ontario, Canada.
| | | | | |
Collapse
|
192
|
Abstract
Osteoarthritis (OA) is the Western world's leading cause of disability. It is incurable, costly and responds poorly to treatment. This review discusses strategies for treating OA by gene therapy. As OA affects a limited number of weight-bearing joints and has no major extra-articular manifestations, it is well suited to local, intra-articular gene therapy. Possible intra-articular sites of gene transfer include the synovium and the cartilage. Most experimental progress has been made with gene transfer to synovium, a tissue amenable to genetic modification by a variety of vectors, using both in vivo and ex vivo protocols. The focus so far has been upon the transfer of genes whose products enhance synthesis of the cartilaginous matrix, or inhibit its breakdown, although there is certainly room for alternative targets. It is possible to build a convincing case implicating interleukin-1 (IL-1) as a key mediator of cartilage loss in OA, and the therapeutic effects of IL-1 receptor anatagonist (IL-1Ra) gene transfer have been confirmed in three different experimental models of OA. As transfer of IL-1Ra cDNA to human arthritic joints has already been accomplished safely, we argue that clinical studies of intra-articular IL-1Ra gene transfer in OA are indicated and should be funded. Of the available vector systems, recombinant adeno-associated virus may provide the best combination of safety with in vivo delivery using current technology.
Collapse
Affiliation(s)
- C H Evans
- Center for Molecular Orthopaedics, Harvard Medical School, MA 02115, USA
| | | | | | | | | |
Collapse
|
193
|
Abstract
Osteoarthritis (OA) is a major health problem in urgent need of better treatment. Gene therapy offers to meet this need. Of the different strategies for using gene therapy in OA, local gene transfer to synovium is in the most advanced stage of development. Local gene transfer brings several advantages, including a focused, local therapy that promises greater efficacy with reduced side-effects, potentially at far lower cost. Moreover, its clinical feasibility has already been confirmed in two Phase I studies of gene therapy for rheumatoid arthritis. Although there are numerous candidate genes of potential use in treating OA genetically, considerable evidence identifies interleukin-1 (IL-1) as a key target. The existence of a natural antagonist, the IL-1 receptor antagonist (IL-1Ra), provides a means with which to inhibit its biologic actions. Clinical studies are suggested in which IL-1Ra complementary DNA is transferred to knee joints shortly before they are surgically replaced with prostheses. This will permit the ready assessment of the safety and efficiency of gene transfer and expression in the human OA knee, as well as permitting preliminary functional data to be obtained, as a prelude to phase II efficacy studies. At this point, the major barriers to progress are financial rather than intellectual or technical.
Collapse
Affiliation(s)
- Christopher H Evans
- Center for Molecular Orthopedics, Harvard Medical School, 221 Longwood Avenue, BLI-152, Boston, MA 02115, USA.
| |
Collapse
|
194
|
Pelletier JP. Rationale for the use of structure-modifying drugs and agents in the treatment of osteoarthritis. Osteoarthritis Cartilage 2004; 12 Suppl A:S63-8. [PMID: 14698646 DOI: 10.1016/j.joca.2003.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
This paper summarizes our current stand on potential pathophysiological targets for osteoarthritis (OA) therapies. Although OA is a complicated disease, involving the cartilage, synovial membrane, and subchondral bone, a number of interactive pathways have been found to explain the structural changes in the disease process. Study of these three tissues has yielded a list of targets to examine for their potential to affect disease progression. At the cartilage level, therapeutic agents, such as growth factors, could be targeted to increase chondrocyte anabolism. At the synovial level, cytokines and cytokine receptor antagonists are potential targets for therapy. In the subchondral bone, cytokines, growth factors and eicosanoids, and locally synthesized factors affecting bone metabolism are also potential targets of therapy. Recent progress in the understanding of the pathophysiology of OA has led to exploration of several interesting new approaches toward the treatment of this disease. New classes of molecules that inhibit one or more OA disease processes are under evaluation for their potential to alter the degenerative process. The prospect of finding a cure for OA is more promising than ever. Based on the discovery of major pathophysiological pathways leading to the structural changes observed in OA, novel ways to treat the progression of OA lesions are emerging.
Collapse
Affiliation(s)
- Jean-Pierre Pelletier
- Head, Arthritis Division and Director, Osteoarthritis Research Unit, Centre Hospitalier de l'Université de Montréal (CHUM)-Hôpital Notre-Dame, Montréal, Québec H2L 4M1 Canada.
| |
Collapse
|
195
|
Pelletier JP, Martel-Pelletier J. Therapeutic targets in osteoarthritis: from today to tomorrow with new imaging technology. Ann Rheum Dis 2003; 62 Suppl 2:ii79-82. [PMID: 14532156 PMCID: PMC1766757 DOI: 10.1136/ard.62.suppl_2.ii79] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- J-P Pelletier
- University of Montreal Hospital Center, Notre-Dame Hospital, Osteoarthritis Research Unit, Montreal, Quebec, Canada.
| | | |
Collapse
|
196
|
Ricci S, Macchia G, Ruggiero P, Maggi T, Bossù P, Xu L, Medaglini D, Tagliabue A, Hammarström L, Pozzi G, Boraschi D. In vivo mucosal delivery of bioactive human interleukin 1 receptor antagonist produced by Streptococcus gordonii. BMC Biotechnol 2003; 3:15. [PMID: 13129437 PMCID: PMC222906 DOI: 10.1186/1472-6750-3-15] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2003] [Accepted: 09/17/2003] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Interleukin-1 (IL-1) is a cytokine involved in the initiation and amplification of the defence response in infectious and inflammatory diseases. IL-1 receptor antagonist (IL-1ra) is an inactive member of the IL-1 family and represents one of the most potent mechanisms for controlling IL-1-dependent inflammation. IL-1ra has proven effective in the therapy of acute and chronic inflammatory diseases in experimental animal models and also in preliminary clinical trials. However, optimisation of therapeutic schedules is still needed. For instance, the use of drug delivery systems targeting specific mucosal sites may be useful to improve topical bioavailability and avoid side effects associated with systemic administration. RESULTS In order to develop systems for the delivery of IL-1ra to mucosal target sites, a Streptococcus gordonii strain secreting human IL-1ra was constructed. The recombinant IL-1ra produced by S. gordonii was composed of the four amino acid residues RVFP of the fusion partner at the N-terminus, followed by the mature human IL-1ra protein. RFVP/IL-1ra displayed full biological activity in vitro in assays of inhibition of IL-1beta-induced lymphocyte proliferation and was released by recombinant S. gordonii in vivo both at the vaginal and the gastrointestinal mucosa of mice. RFVP/IL-1ra appeared beneficial in the model of ulcerative colitis represented by IL-2-/- mice (knock-out for the interleukin-2 gene), as shown by the body weight increase of IL-2-/- mice locally treated with S. gordonii producing RFVP/IL-1ra. CONCLUSIONS These results indicate that recombinant S. gordonii can be successfully used as a delivery system for the selective targeting of mucosal surfaces with therapeutic proteins.
Collapse
Affiliation(s)
- Susanna Ricci
- Laboratory of Molecular Microbiology and Biotechnology, Department of Molecular Biology, University of Siena, Policlinico "Le Scotte", Viale Bracci, 53100 Siena, Italy
| | - Giovanni Macchia
- Research Center Dompé S.p.A., Via Campo di Pile, 67100 L'Aquila, Italy
- Current address: European Patent Office, Patentlaan 7, 2288 EE Rijswijk, The Netherlands
| | - Paolo Ruggiero
- Research Center Dompé S.p.A., Via Campo di Pile, 67100 L'Aquila, Italy
- Current address: IRIS Research Center, Chiron S.r.l., Via Fiorentina 1, 53100 Siena, Italy
| | - Tiziana Maggi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Molecular Biology, University of Siena, Policlinico "Le Scotte", Viale Bracci, 53100 Siena, Italy
- Current address: IRIS Research Center, Chiron S.r.l., Via Fiorentina 1, 53100 Siena, Italy
| | - Paola Bossù
- IRCCS Fondazione Santa Lucia, Laboratory of Clinical and Behavioural Neurology, Via Ardeatina 306, 00179 Roma, Italy
| | - Li Xu
- Center for Biotechnology, Novum, Department of Microbiology, Pathology and Immunology, Karolinska Institute, Huddinge Hospital, Huddinge, Sweden
| | - Donata Medaglini
- Laboratory of Molecular Microbiology and Biotechnology, Department of Molecular Biology, University of Siena, Policlinico "Le Scotte", Viale Bracci, 53100 Siena, Italy
| | - Aldo Tagliabue
- International Vaccine Institute, SNU Research Park, San 4–8 Bongcheon-7 dong, Kwanak-gu, Seoul, Korea 151–818
| | - Lennart Hammarström
- Center for Biotechnology, Novum, Department of Microbiology, Pathology and Immunology, Karolinska Institute, Huddinge Hospital, Huddinge, Sweden
| | - Gianni Pozzi
- Laboratory of Molecular Microbiology and Biotechnology, Department of Molecular Biology, University of Siena, Policlinico "Le Scotte", Viale Bracci, 53100 Siena, Italy
| | - Diana Boraschi
- Laboratory of Cytokines, Institute of Biomedical Technologies, CNR, Area della Ricerca di S. Cataldo, Via G. Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
197
|
Müller-Ladner U, Pap T, Gay RE, Gay S. Gene transfer as a future therapy for rheumatoid arthritis. Expert Opin Biol Ther 2003; 3:587-98. [PMID: 12831364 DOI: 10.1517/14712598.3.4.587] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Inhibiting key pathogenic processes within the rheumatoid synovium is a most attractive goal to achieve, and the number of potential intra- and extracellular pathways operative in rheumatoid arthritis (RA) that could be used for a gene therapy strategy is increasing continuously. Gene transfer or gene therapy might also be one of the approaches to solve the problem of long-term expression of therapeutic genes, in order to replace the frequent application of recombinant proteins, in the future. However, at present, gene therapy has not reached a realistic clinical stage, which is mainly due to severe side effects in humans, the complexity of RA pathophysiology and the current state of available gene transfer techniques. On the other hand, novel gene delivery systems are not restricted to vectors or certain types of cells, as mobile cells including macrophages, dendritic cells, lymphocytes and multipotent stem cells can also be used as smart gene transfer vehicles. Moreover, the observation in animal models that application of viral vectors into a joint can exert additional therapeutic effects in nearby joints might also facilitate the transfer from animal to human gene therapy. Future strategies will also examine the potential of novel long-term expression vectors such as lentiviruses and cytomegalovirus (CMV)-based viruses as a basis for future clinical trials in RA.
Collapse
Affiliation(s)
- Ulf Müller-Ladner
- Department of Internal Medicine I, University of Regensburg, FJS-Allee 11, D-93042 Regensburg, Germany.
| | | | | | | |
Collapse
|
198
|
Abstract
Rheumatoid arthritis is an autoimmune disease with intra-articular inflammation and synovial hyperplasia that results in progressive degradation of cartilage and bone, in severe cases it causes systemic complications. Recently, biological agents that suppress the activities of proinflammatory cytokines have shown efficacy as antiarthritic drugs, but require frequent administration. Thus, gene transfer approaches are being developed as an alternative approach for targeted, more efficient and sustained delivery of inhibitors of inflammatory cytokines as well as other therapeutic agents. Indeed, the efficacy of gene transfer for the treatment of arthritis has been demonstrated in mouse, rat, rabbit, and horse models of disease whereas the feasibility of the approach has been demonstrated in Phase I clinical trials. In this review, the current status of both preclinical and clinical arthritis gene therapy is presented. In addition, the advantages and disadvantages of different types of vectors, target cells and therapeutic genes being developed for the treatment of arthritis are summarized. Finally, the future directions of the rapidly developed field of arthritis gene therapy are outlined.
Collapse
Affiliation(s)
- P D Robbins
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | | |
Collapse
|
199
|
Vuolteenaho K, Moilanen T, Hämäläinen M, Moilanen E. Regulation of nitric oxide production in osteoarthritic and rheumatoid cartilage. Role of endogenous IL-1 inhibitors. Scand J Rheumatol 2003; 32:19-24. [PMID: 12635941 DOI: 10.1080/03009740310000355] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
OBJECTIVE To investigate the endogenous regulation of interleukin-1 (IL-1) cytokine network in osteoarthritic (OA) and rheumatoid (RA) cartilage in relation to nitric oxide (NO) production. METHODS Cartilage specimen obtained from OA and RA patients undergoing knee replacement surgery were studied for iNOS expression, NO and IL-1 antagonist production in tissue culture. RESULTS OA cartilage responded to IL-1beta-stimulation with higher NO production than RA cartilage, whereas there was no difference in NO synthesis between OA and RA samples when stimulated by TNFalpha or LPS. Interleukin-1 receptor antagonist (IL-1Ra) production was higher in RA cartilage than in OA cartilage, and its production was increased by NO synthase inhibitor 1400W. CONCLUSION IL-1beta is a potent stimulator of NO production by the iNOS pathway in RA and more pronouncedly in OA cartilage. This process is regulated by cartilage derived IL-1 antagonists, and is implicated in cartilage destruction and synovial inflammation in OA and RA joints.
Collapse
Affiliation(s)
- Katriina Vuolteenaho
- The Immunopharmacological Research Group, Medical School, University of Tampere, and Tampere University Hospital, Finland
| | | | | | | |
Collapse
|
200
|
Jones HP, Appleyard RC, Mahajan S, Murrell GAC. Meniscal and Chondral Loss in the Anterior Cruciate Ligament Injured Knee. Sports Med 2003; 33:1075-89. [PMID: 14599234 DOI: 10.2165/00007256-200333140-00004] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Rupture of the anterior cruciate ligament (ACL) of the knee is a commonly occurring injury in the athletic population. Associated meniscal and chondral injury is well recognised. This occurs both at the time of index injury and also secondarily over time in the ACL-deficient knee as a result of several related pathways culminating in osteoarthritis. ACL reconstruction is a well established surgical technique for treatment of symptomatic instability in ACL-deficient knees but the role of ACL reconstruction in the prevention of osteoarthritis remains unclear. This article reviews the contemporary literature on the pathophysiology of chondral and meniscal loss in ACL-injured knees and the role of current treatment techniques, including surgical reconstruction of ligamentous, meniscal and chondral pathology, in altering the natural history of the ACL-deficient knee.
Collapse
Affiliation(s)
- Hugh P Jones
- Department of Orthopaedic Surgery, St George Hospital Campus, Kogarah, New South Wales, Australia.
| | | | | | | |
Collapse
|