201
|
Gouze JN, Gouze E, Palmer GD, Liew VS, Pascher A, Betz OB, Thornhill TS, Evans CH, Grodzinsky AJ, Ghivizzani SC. A comparative study of the inhibitory effects of interleukin-1 receptor antagonist following administration as a recombinant protein or by gene transfer. Arthritis Res Ther 2003; 5:R301-9. [PMID: 12932294 PMCID: PMC193732 DOI: 10.1186/ar795] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2002] [Revised: 06/23/2003] [Accepted: 07/01/2003] [Indexed: 11/28/2022] Open
Abstract
Anakinra, the recombinant form of IL-1 receptor antagonist (IL-1Ra), has been approved for clinical use in the treatment of rheumatoid arthritis as the drug Kineret trade mark, but it must be administered daily by subcutaneous injection. Gene transfer may offer a more effective means of delivery. In this study, using prostaglandin E2 production as a measure of stimulation, we quantitatively compared the ability of anakinra, as well as that of IL-1Ra delivered by gene transfer, to inhibit the biologic actions of IL-1beta. Human synovial fibroblast cultures were incubated with a range of doses of anakinra or HIG-82 cells genetically modified to constitutively express IL-1Ra. The cultures were then challenged with recombinant human IL-1beta either simultaneously with addition of the source of IL-1Ra or 24 hours later. In a similar manner, the potencies of the two sources of IL-1Ra were compared when human synovial fibroblasts were challenged with IL-1beta produced constitutively by genetically modified cells. No significant difference in inhibitory activity was observed between recombinant protein and IL-1Ra provided by the genetically modified cells, under static culture conditions, even following incubation for 4 days. However, under culture conditions that provided progressive dilution of the culture media, striking differences between these methods of protein delivery became readily apparent. Constitutive synthesis of IL-1Ra by the genetically modified cells provided sustained or increased protection from IL-1 stimulation over time, whereas the recombinant protein became progressively less effective. This was particularly evident under conditions of continuous IL-1beta synthesis.
Collapse
Affiliation(s)
- Jean-Noel Gouze
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Elvire Gouze
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Glyn D Palmer
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Victor S Liew
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Arnulf Pascher
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Oliver B Betz
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Thomas S Thornhill
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Christopher H Evans
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan J Grodzinsky
- Center for Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Steven C Ghivizzani
- Center for Molecular Orthopaedics, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
202
|
Lavigne P, Shi Q, Jolicoeur FC, Pelletier JP, Martel-Pelletier J, Fernandes JC. Modulation of IL-1beta, IL-6, TNF-alpha and PGE(2) by pharmacological agents in explants of membranes from failed total hip replacement. Osteoarthritis Cartilage 2002; 10:898-904. [PMID: 12435335 DOI: 10.1053/joca.2002.0846] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Introduction and goal Proinflammatory cytokines and prostaglandin E(2) (PGE(2)) play an important role in the pathophysiology of osteolysis and implant loosening. The aim of this study was to evaluate the role of pharmacological agents in the inhibition of Interleukin-1 (IL-1), interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) and PGE(2) in explants of interface membranes from failed total hip replacements (fTHR). Material and methods Membranes from fTHR were retrieved (N=20) and explants were incubated for 72h in the absence or presence of tenidap at three different concentrations (5, 20 or 50 microg/ml) or diclofenac (125 microg/l). IL-1beta, IL-6, TNF-alpha, and PGE(2) levels were measured in the culture medium using ELISA Capture or EIA kits. Statistical analysis was done using the Mann-Whitney U-test. Results A statistically significant inhibition in IL-1beta synthesis was found at tenidap concentrations of 20 microg/ml (71.3%, P< 0.05) and 50 microg/ml (79.3%,P< 0.02). Tenidap reduced IL-6 levels by 90.4% at 20 microg/ml (P< 0.005) and 96.0% (P< 0.05) at 50 microg/ml. Tenidap also reduced the synthesis of TNF-alpha by 66.9% (P< 0.05) and 77.4% at concentrations of 20 microg/ml and 50 microg/ml. Tenidap had a marked suppressive effect of over 90% (P< 0.0001) on PGE(2) synthesis in all three concentrations. Diclofenac (125 microg/l) decreased PGE(2) production by 95% (P< 0.0001), but had no significant effect in IL-1beta, IL-6, and TNF-alpha levels in the culture medium. Conclusion The ability to simultaneously suppress the release of proinflammatory cytokines and PGE(2) may help control osteolysis and prevent aseptic loosening of THR. This effect could increase implant longevity and lead the way to the pharmacological treatment of this pathology.
Collapse
Affiliation(s)
- P Lavigne
- Osteoarthritis Research Unit, Centre hospitalier de l'Université de Montréal, Hôpital Notre-Dame, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
203
|
Sabatini M, Bardiot A, Lesur C, Moulharat N, Thomas M, Richard I, Fradin A. Effects of agonists of peroxisome proliferator-activated receptor gamma on proteoglycan degradation and matrix metalloproteinase production in rat cartilage in vitro. Osteoarthritis Cartilage 2002; 10:673-9. [PMID: 12202119 DOI: 10.1053/joca.2002.0827] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To examine the effects of agonists of peroxisome proliferator-activated receptor (PPAR) gamma on proteoglycan degradation induced by interleukin (IL)-1beta or tumor necrosis factor (TNF)alpha in cartilage in vitro. DESIGN Proteoglycan degradation was measured as release of radioactivity from rat cartilage explants previously labeled with (35)SO2-4. Western blots were used to examine tissue levels of aggrecan neoepitopes NITEGE and VDIPEN, generated by aggrecanases and matrix metalloproteinases (MMP), respectively. Production of MMP-2, -3 and -9 by cultured rat chondrocytes was measured by zymography and by fluorimetric assay. RESULTS IL-1beta-induced proteoglycan degradation was likely due to aggrecanase, since it was associated with a strong increase of NITEGE signal. MMP-dependent VDIPEN signal increased only after further incubation with pro-MMP activator APMA. PPAR agonists 15d-PGJ(2) and GI262570 (10 microM) inhibited IL-1beta- and TNFalpha-induced proteoglycan degradation measured both before and after addition of APMA. The agonists also inhibited cytokine-induced MMP production by isolated chondrocytes. CONCLUSION This study shows that PPARgamma agonists inhibit cytokine-induced proteoglycan degradation mediated by both aggrecanase and MMP. This effect is associated with inhibition of production of MMP-3 and -9. These results support the interest for PPARgamma agonists as candidate inhibitors of pathological cartilage degradation.
Collapse
Affiliation(s)
- M Sabatini
- Division of Rheumatology, Institut de Recherches Servier (IdRS), 11 rue des Moulineaux, 92150 Suresnes, France.
| | | | | | | | | | | | | |
Collapse
|
204
|
Abstract
IL-1 is an important mediator of inflammation and tissue damage in multiple organs, both in experimental animal models of disease and in human diseases. The IL-1 family consists of two agonists, IL-1alpha and IL-1beta, two receptors, biologically active IL-1RI and inert IL-1RII, and a specific receptor antagonist, IL-1Ra. The balance between IL-1 and IL-1Ra in local tissues plays an important role in the susceptibility to and severity of many diseases. An allelic polymorphism in the IL-1Ra gene has been associated with a variety of human diseases primarily of epithelial and endothelial cell origin. This association may be secondary to an imbalance in the IL-1 system with enhanced production of IL-1beta and reduced production of the major intracellular isoform of IL-1Ra. Treatment of RA with daily subcutaneous injections of recombinant IL-1Ra protein has been shown to be efficacious. Gene therapy approaches with IL-1Ra are being evaluated for the treatment of RA and other human diseases.
Collapse
Affiliation(s)
- William P Arend
- B115 Division of Rheumatology, Health Sciences Centre, University of Colorado, 4200 East Ninth Avenue, Denver, CO 80262, USA.
| |
Collapse
|
205
|
Hardy MM, Seibert K, Manning PT, Currie MG, Woerner BM, Edwards D, Koki A, Tripp CS. Cyclooxygenase 2-dependent prostaglandin E2 modulates cartilage proteoglycan degradation in human osteoarthritis explants. ARTHRITIS AND RHEUMATISM 2002; 46:1789-803. [PMID: 12124863 DOI: 10.1002/art.10356] [Citation(s) in RCA: 240] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To examine cyclooxygenase-2 (COX-2) enzyme expression, its regulation by interleukin-1 beta (IL-1 beta), and the role of prostaglandin E(2) (PGE(2)) in proteoglycan degradation in human osteoarthritic (OA) cartilage. METHODS Samples of human OA articular cartilage, meniscus, synovial membrane, and osteophytic fibrocartilage were obtained at knee arthroplasty and cultured ex vivo with or without IL-1 beta and COX inhibitors. COX expression was evaluated by immunohistochemistry and Western blot analysis. The enzymatic activity of COX was measured by conversion of arachidonic acid to PGE(2). Cartilage degradation was evaluated by measuring the accumulation of sulfated glycosaminoglycans in the medium. RESULTS IL-1 beta induced robust expression of COX-2 and PGE(2) in OA meniscus, synovial membrane, and osteophytic fibrocartilage explants, whereas low levels were produced in OA articular cartilage. IL-1 beta also induced cartilage proteoglycan degradation in OA synovial membrane-cartilage cocultures. Increased proteoglycan degradation corresponded to the induction of COX-2 protein expression in, and PGE(2) production from, the synovial membrane. Dexamethasone, neutralizing IL-1 beta antibody, or the selective COX-2 inhibitor, SC-236, attenuated both the IL-1 beta-induced PGE(2) production and cartilage proteoglycan degradation in these cocultures. The addition of PGE(2) reversed the inhibition of proteoglycan degradation caused by SC-236. CONCLUSION IL-1 beta-induced production of COX-2 protein and PGE(2) was low in OA articular cartilage compared with that in the other OA tissues examined. IL-1 beta-mediated degradation of cartilage proteoglycans in OA synovial membrane-cartilage cocultures was blocked by the selective COX-2 inhibitor, SC-236, and the effect of SC-236 was reversed by the addition of exogenous PGE(2). Our data suggest that induction of synovial COX-2-produced PGE(2) is one mechanism by which IL-1 beta modulates cartilage proteoglycan degradation in OA.
Collapse
Affiliation(s)
- Medora M Hardy
- Arthritis and Inflammation Pharmaclogy, Pharmacia Corporation, St. Louis, Missouri 63017, USA.
| | | | | | | | | | | | | | | |
Collapse
|
206
|
Marijnissen ACA, van Roermund PM, Verzijl N, Tekoppele JM, Bijlsma JWJ, Lafeber FPJG. Steady progression of osteoarthritic features in the canine groove model. Osteoarthritis Cartilage 2002; 10:282-9. [PMID: 11950251 DOI: 10.1053/joca.2001.0507] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Recently we described a canine model of osteoarthritis (OA), the groove model with features of OA at 10 weeks after induction, identical to those seen in the canine anterior cruciate ligament transection (ACLT) model. This new model depends on cartilage damage accompanied by transient intensified loading of the affected joint. The present study evaluates this groove model at 20 and 40 weeks after induction, to assess whether the osteoarthritic features progress in time. METHODS Grooves were made in the femoral condyles of one knee without damaging the subchondral bone. After surgery the dogs were forced to load the experimental joint 3 days per week (4 hours/day) for 20 weeks by fixing the contralateral control limb to the trunk. After 20 weeks and 40 weeks (the last 20 weeks normal loading) joints were analysed for biochemical and histological features of OA. RESULTS All biochemical cartilage parameters were indicative of OA and all these parameters suggested a slow progression of degeneration over time from 20 to 40 weeks after induction, statistically significant for synthesis and content of proteoglycans as well as Mankin grade. Synovial inflammation, which was mild, diminished slightly in time. CONCLUSION The degenerative joint damage in the canine groove model is slowly progressive over time in the first year. The cartilage degeneration is induced by a one-time trauma and is not primarily mediated by synovial inflammation, which gives this model unique characteristics compared to presently available models for studying early osteoarthritic features in vivo. In the groove model the effect of treatment of cartilage damage is not counteracted by permanent joint instability or hampered by inflammation. Therefore, the model might be more sensitive to detect effects of therapy, aimed at cartilage protection and repair.
Collapse
Affiliation(s)
- A C A Marijnissen
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, 3508 GA Utrecht, The Netherlands.
| | | | | | | | | | | |
Collapse
|
207
|
|
208
|
Frisbie DD, Ghivizzani SC, Robbins PD, Evans CH, McIlwraith CW. Treatment of experimental equine osteoarthritis by in vivo delivery of the equine interleukin-1 receptor antagonist gene. Gene Ther 2002; 9:12-20. [PMID: 11850718 DOI: 10.1038/sj.gt.3301608] [Citation(s) in RCA: 254] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2001] [Accepted: 10/16/2001] [Indexed: 11/08/2022]
Abstract
Osteoarthritis in horses and in humans is a significant social and economic problem and continued research and improvements in therapy are needed. Because horses have naturally occurring osteoarthritis, which is similar to that of humans, the horse was chosen as a species with which to investigate gene transfer as a potential therapeutic modality for the clinical treatment of osteoarthritis. Using an established model of equine osteoarthritis that mimics clinical osteoarthritis, the therapeutic effects resulting from intra-articular overexpression of the equine interleukin-1 receptor antagonist gene through adenoviral-mediated gene transfer were investigated. In vivo delivery of the equine IL-IRa gene led to elevated intra-articular expression of interleukin-1 receptor antagonist for approximately 28 days, resulting in significant improvement in clinical parameters of pain and disease activity, preservation of articular cartilage, and beneficial effects on the histologic parameters of synovial membrane and articular cartilage. Based on these findings, gene transfer of interleukin-1 receptor antagonist is an attractive treatment modality for the equine patient and also offers future promise for human patients with osteoarthritis.
Collapse
Affiliation(s)
- D D Frisbie
- Equine Orthopaedic Research Laboratory, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO 80523, USA
| | | | | | | | | |
Collapse
|
209
|
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease characterized by joint inflammation as well as progressive cartilage and bone destruction. Advances in the understanding of the pathophysiology of RA have led to the development of new therapeutic strategies, including gene therapy. Gene therapy offers a new approach to deliver therapeutic proteins to the joints of arthritis patients. Local as well as systemic gene therapy can be envisaged for the treatment of arthritis. Several viral and non-viral vectors have been used in animal models for rheumatoid arthritis for ex vivo and in vivo delivery of therapeutic genes. Promising pre-clinical data have resulted from the application of these strategies. Using ex vivo gene delivery, successful and safe gene transfer has been demonstrated in the joints of RA patients. Although new insights into the role of cytokines and other mediators of chronic inflammation have provided novel targets for therapeutic intervention, the development of vectors that induce long-term and regulated gene expression remains a challenge.
Collapse
Affiliation(s)
- M J Vervoordeldonk
- Division of Clinical Immunology and Rheumatology, Department of Medicine, Academic Medical Centre, University of Amsterdam, Amsterdam, 1100 DD, The Netherlands
| | | |
Collapse
|
210
|
Pelletier JP, Martel-Pelletier J. The Novartis-ILAR Rheumatology Prize 2001 Osteoarthritis: from molecule to man. ARTHRITIS RESEARCH 2001; 4:13-9. [PMID: 11879533 PMCID: PMC128913 DOI: 10.1186/ar378] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2001] [Accepted: 10/04/2001] [Indexed: 11/18/2022]
Abstract
During our careers, we have developed new and innovative concepts pertaining to the pathophysiology of osteoarthritis which have assisted in the development of new therapeutic approaches. Moreover, our laboratory has long sought to develop protective agents for osteoarthritic structural joint tissues. The most significant concepts that have originated from our lab are briefly outlined in this commentary.
Collapse
Affiliation(s)
- Jean-Pierre Pelletier
- Osteoarthritis Research Unit, Centre Hospitalier de l'Université de Montréal (CHUM)-Hôpital Notre-Dame, Montréal, Québec, Canada.
| | | |
Collapse
|
211
|
Abstract
Mature articular cartilage has a poor reparative response to injury and its irreparable breakdown is the common feature of degenerative joint diseases. If articular cartilage lesions become symptomatic, the orthopaedic surgeon must decide on a treatment option. The treatment options include conversion of chondral lesions to osteochondral lesions, which facilitates migration of cells from the marrow space to effect repair. In recent years, a greater emphasis has been placed on tissue engineering strategies and thus several new treatment options have been introduced, including the use of cell transplantation. Several tissue sources and cell types can potentially be used for this type of therapy. These include autologous or allograft chondrocytes and mesenchymal progenitor cells from various tissues. These cells may be delivered to articular cartilage lesions by a variety of methods including direct cell injection to the lesion or seeding in a biodegradable scaffold prior to implantation. In this review, the potential of cell transplantation for articular cartilage repair and regeneration will be discussed. The authors will focus on the available technologies and the present limitations of cell-based therapies.
Collapse
Affiliation(s)
- B Johnstone
- Department of Orthopaedics, Case Western Reserve University, Cleveland, OH 44106, USA.
| | | |
Collapse
|
212
|
Abstract
Osteoarthritis (OA) is a joint disease that involves degeneration of articular cartilage, weakening of the subchondral bone and limited intra-articular inflammation manifested by synovitis. Since the pathogenesis of OA involves multiple aetiologies, including mechanical, biochemical and genetic factors, it has been difficult to identify unique targets for therapy. Current pharmacological interventions focus primarily on improving symptoms. The rationale for the use of anticytokine therapy in OA is based on evidence from studies in vitro and in vivo that interleukin-1 (IL-1) and tumour necrosis factor (TNF)-alpha are the predominant pro-inflammatory and catabolic cytokines involved in the initiation and progression of articular cartilage destruction. Since the increased levels of catabolic enzymes, prostaglandins, nitric oxide (NO) and other markers in OA fluids and tissues appear to be related to elevated levels of IL-1 and TNF-alpha, therapies that interfere with the expression or actions of these cytokines are most promising. Other cytokines that are anti-inflammatory and are often detected, paradoxically, in OA tissues are also potential therapeutic agents for counteracting the cartilage destruction in OA. Identification of methods for early diagnosis is of key importance, since therapeutic interventions aimed at blocking or reversing structural damage will be more effective when there is the possibility of preserving normal homeostasis. At later stages, cartilage tissue engineering with or without gene therapy will also require anticytokine therapy to block damage to newly repaired cartilage. This review will focus on experimental approaches currently under study that may lead to elucidation of effective strategies for therapy in OA, with special emphasis on anticytokine therapy.
Collapse
Affiliation(s)
- M B Goldring
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA 02115-5713, USA.
| |
Collapse
|
213
|
Saxer RA, Bent SJ, Brower-Toland BD, Mi Z, Robbins PD, Evans CH, Nixon AJ. Gene mediated insulin-like growth factor-I delivery to the synovium. J Orthop Res 2001; 19:759-67. [PMID: 11562119 DOI: 10.1016/s0736-0266(00)00077-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The feasibility of articular gene therapy using insulin-like growth factor-I transgene expression in synovial tissues was assessed in vitro by transfection of synovial explant and monolayer cultures. Synovial membrane was harvested from horses and distributed for explant culture in multiwell plates or digested for monolayer culture in multiwell plates and chamber slides. Synovial monolayers were cultured for 48 h after infection with 0, 100, 200, or 500 moi adenovirus-IGF-I (AdeIGF-I) to establish an optimum dose. Explants were then either infected with AdeIGF-I or adenoviral LacZ and cultured for 8 days, treated with 100 ng/ml recombinant IGF-I as a positive control, or remained as uninfected untreated culture controls. Expression of IGF-I in explants and monolayers was assessed by in situ hybridization and quantitative polymerase chain reaction (PCR), and translation confirmed by IGF-I radioimmunoassay (RIA) and tissue immunoreaction. Effects of IGF-I on synovial function was assessed by proteoglycan and hyaluronan assay, and northern blot assessment of decorin and collagen type I expression. Significant transgene expression in synovial cells was present for all AdeIGF-I concentrations. Similarly, medium IGF-I concentrations were significantly elevated in AdeIGF-I infected synovial monolayer and explant cultures at all time points. Peak IGF-I concentration of 246 +/- 43 ng/ml developed in explant cultures on day 4; IGF-I levels in control explant groups were unchanged over baseline values. In situ hybridization and immunolocalization for IGF-I indicated focal IGF-I expression in intimal and subintimal layers of infected explants, with diffuse immunoreaction throughout infected subintimal and fibrous layers. For monolayer cultures, intracellular immunoreaction to IGF-I was markedly higher in infected cells, and was most prominent at 100 moi. Effects of IGF-I on synoviocyte cultures were evident on northern blots, which showed decreased decorin expression and elevated type I collagen production in AdeIGF-I infected monolayers. Proteoglycan concentration in the medium from explant cultures rose over the initial 4 days but was similar between treatment groups. The concentration of hyaluronan in medium from explant cultures did not differ significantly within or between treated and control groups during the 8-day study period. These data indicate that IGF-I can be successfully introduced to synovial structures by adenoviral vectors and results in effective IGF-I ligand synthesis without untoward synovial morphologic effects.
Collapse
Affiliation(s)
- R A Saxer
- Comparative Orthopaedics Laboratory, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | |
Collapse
|
214
|
Budsberg SC. Long-term temporal evaluation of ground reaction forces during development of experimentally induced osteoarthritis in dogs. Am J Vet Res 2001; 62:1207-11. [PMID: 11497439 DOI: 10.2460/ajvr.2001.62.1207] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVE To describe changes in vertical ground reaction forces (GRF) over 48 months in dogs with osteoarthritis (OA) of the stifle joint induced by transection of a cranial cruciate ligament (CCL). ANIMALS 12 clinically normal adult dogs. PROCEDURE Vertical GRF (eg, peak force and impulse) were determined prior to and 1, 2, 3, 6, 10, and 12 months after transection of the right CCL. In 7 dogs, data were also collected 24, 32, 38, 42, and 48 months after transection. RESULTS Vertical peak force and impulse were significantly decreased in the right hind limb at all times after transection, compared with baseline values. From 10 through 48 months after transection, vertical GRF remained essentially static. Ground reaction forces in the unoperated (left) hind limb also changed significantly during the study. Left vertical impulse significantly increased 3 months after transection, whereas at 24, 38, 42, and 48 months after transection, left vertical peak force was significantly decreased, compared with the baseline value. Mean intradog coefficients of variation (CV) for peak vertical force and impulse ranged from 738 and 9.32, respectively, 1 month after transection to 1.96 and 2.76, respectively, at 42 months. CONCLUSIONS AND CLINICAL RELEVANCE Vertical GRF in the affected hind limb equilibrated approximately 10 months after CCL transection. Prior to this, force transmission across the affected stifle joint changed significantly over time. Intradog CV were small, indicating that GRF may be an appropriate outcome measurement for evaluation of OA development induced by CCL transection in dogs.
Collapse
Affiliation(s)
- S C Budsberg
- Department of Small Animal Medicine, College of Veterinary Medicine, University of Georgia, Athens 30602, USA
| |
Collapse
|
215
|
Gelse K, Jiang QJ, Aigner T, Ritter T, Wagner K, Pöschl E, von der Mark K, Schneider H. Fibroblast-mediated delivery of growth factor complementary DNA into mouse joints induces chondrogenesis but avoids the disadvantages of direct viral gene transfer. ARTHRITIS AND RHEUMATISM 2001; 44:1943-53. [PMID: 11508447 DOI: 10.1002/1529-0131(200108)44:8<1943::aid-art332>3.0.co;2-z] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE To assess the advantages and disadvantages of a direct adenoviral and a cell-mediated approach to the induction of cartilage formation in joints by transfer of growth factor genes. METHODS Adenoviral vectors carrying insulin-like growth factor 1 (IGF-1) or bone morphogenetic protein 2 (BMP-2) complementary DNA were constructed and applied to primary human and murine chondrocytes or fibroblasts. Transgene expression was quantified by enzyme-linked immunosorbent assay. Direct injection of these vectors or AdLacZ, a reporter gene vector, into mouse knee joints was compared with the transplantation of syngeneic fibroblasts (infected ex vivo with the same vectors) with respect to virus spread, immune response, and cartilage formation by use of histologic, immunohistochemical, and molecular analyses. RESULTS AdIGF-1 and AdBMP-2 efficiently infected all cell types tested. Human cells secreted biologically relevant levels of protein over a period of at least 28 days. Direct transfer of AdLacZ into mouse knee joints resulted in positively stained synovial tissues, whereas AdLacZ-infected fibroblasts settled on the surface of the synovial membranes. Inadvertent spread of vector DNA into the liver, lung, and spleen was identified by nested polymerase chain reaction in all mice that had received the vector directly; this rarely occurred following fibroblast-mediated gene transfer. Direct injection of AdBMP-2 induced the synthesis of new cartilage in periarticular mesenchyme, accompanied by extensive osteophyte formation. When AdBMP-2 was administered by injecting ex vivo-infected fibroblasts, cartilage formation was observed only in regions near the injected cells. AdIGF-1 treatment did not lead to morphologic changes. Importantly, fibroblast-mediated gene transfer avoided the strong immune response to adenovirus that was elicited following direct application of the vector. CONCLUSION Our results indicate that cell-mediated gene transfer provides sufficient BMP-2 levels in the joint to induce cartilage formation while avoiding inadvertent vector spread and immune reactions.
Collapse
Affiliation(s)
- K Gelse
- Department of Experimental Medicine I, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
216
|
Boyault S, Simonin MA, Bianchi A, Compe E, Liagre B, Mainard D, Bécuwe P, Dauça M, Netter P, Terlain B, Bordji K. 15-Deoxy-delta12,14-PGJ2, but not troglitazone, modulates IL-1beta effects in human chondrocytes by inhibiting NF-kappaB and AP-1 activation pathways. FEBS Lett 2001; 501:24-30. [PMID: 11457450 DOI: 10.1016/s0014-5793(01)02614-x] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The activation of peroxisome proliferator-activated receptor gamma (PPARgamma) has been shown to inhibit the production and the effects of proinflammatory cytokines. Since interleukin-1beta (IL-1beta) directly mediates cartilage degradation in osteoarthritis, we investigated the capability of PPARgamma ligands to modulate IL-1beta effects on human chondrocytes. RT-PCR and Western blot analysis revealed that PPARgamma expression was decreased by IL-1beta. 15-deoxy-Delta12,14-prostaglandin J2 (15d-PGJ2), in contrast to troglitazone, was highly potent to counteract IL-1beta-induced cyclooxygenase-2 and inductible nitric oxide synthase expression, NO production and the decrease in proteoglycan synthesis. Western blot and gel-shift analyses demonstrated that 15d-PGJ2 inhibited NF-kappaB activation, while troglitazone was ineffective. Although 15d-PGJ2 attenuated activator protein-1 binding on the DNA, it potentiated c-jun migration in the nucleus. The absence or the low effect of troglitazone suggests that 15d-PGJ2 action in human chondrocytes is mainly PPARgamma-independent.
Collapse
Affiliation(s)
- S Boyault
- Laboratoire de Pharmacologie, UMR 7561 CNRS-Université Henri Poincaré Nancy I, Faculté de Médecine, Vandoeuvre-lès-Nancy, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
217
|
Sabatini M, Thomas M, Deschamps C, Lesur C, Rolland G, de Nanteuil G, Bonnet J. Effects of ceramide on aggrecanase activity in rabbit articular cartilage. Biochem Biophys Res Commun 2001; 283:1105-10. [PMID: 11355886 DOI: 10.1006/bbrc.2001.4920] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Ceramide participates in signal transduction of IL-1 and TNF, two cytokines likely involved in cartilage degradation in osteoarthritis. We previously showed that ceramide stimulates proteoglycan degradation, mRNA expression of matrix metalloproteinase (MMP)-1, -3, and -13, and pro-MMP-3 production in rabbit cartilage. Since aggrecan, the main cartilage proteoglycan, can be cleaved by metalloproteinases both of MMP and aggrecanase type, the aim of this study was to determine if ceramide stimulates aggrecanase action and, if that is the case, in which measure aggrecanase mediates the degradative effect of ceramide. To this end, antibodies were used against the C terminal aggrecan neoepitopes generated by aggrecanases (NITEGE(373)) and MMPs (DIPEN(341)). Ceramide C(2) at 10(-5) to 10(-4) M dose-dependently increased NITEGE signal, without changing that of DIPEN, in cultured explants of rabbit cartilage. The effects of 10(-4) M C(2) on NITEGE signal and proteoglycan degradation were similarly antagonized by the metalloproteinase inhibitor batimastat, with return to the basal level at 10(-6) M. These results show that, similarly to IL-1 and TNF, ceramide-induced aggrecan degradation is mainly due to aggrecanases. That no increase of MMP activity was detected, despite stimulation of MMP expression, was probably due to lack of proenzyme conversion to mature form, since addition of a MMP activator to C(2)-treated cartilage increased both DIPEN signal and proteoglycan degradation. These findings support the hypothesis that cytokine-induced ceramide could play a mediatory role in situations of increased degradation of cartilage matrix.
Collapse
Affiliation(s)
- M Sabatini
- Division of Rheumatology, Institut de Recherches Servier, Suresnes, France.
| | | | | | | | | | | | | |
Collapse
|
218
|
Pelletier JP, Martel-Pelletier J, Abramson SB. Osteoarthritis, an inflammatory disease: potential implication for the selection of new therapeutic targets. ARTHRITIS AND RHEUMATISM 2001; 44:1237-47. [PMID: 11407681 DOI: 10.1002/1529-0131(200106)44:6<1237::aid-art214>3.0.co;2-f] [Citation(s) in RCA: 780] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Affiliation(s)
- J P Pelletier
- Centre Hospitalier de l'Université de Montréal, Hĵpital Notre-Dame, Quebec, Canada
| | | | | |
Collapse
|
219
|
Prud'homme GJ, Lawson BR, Theofilopoulos AN. Anticytokine gene therapy of autoimmune diseases. Expert Opin Biol Ther 2001; 1:359-73. [PMID: 11727511 DOI: 10.1517/14712598.1.3.359] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Viral and nonviral gene therapy vectors have been successfully employed to deliver inflammatory cytokine inhibitors (anticytokines), or anti-inflammatory cytokines, such as transforming growth factor beta-1 (TGF-beta 1), which protect against experimental autoimmune diseases. These vectors carry the relevant genes into a variety of tissues, for either localised or systemic release of the encoded protein. Administration of cDNA encoding soluble IFN-gamma receptor (IFN-gamma R)/IgG-Fc fusion proteins, soluble TNF-alpha receptors, or IL-1 receptor antagonist (IL-1ra), protects against either lupus, various forms of arthritis, autoimmune diabetes, or other autoimmune diseases. These inhibitors, unlike many cytokines, have little or no toxic potential. Similarly, TGF-beta 1 gene therapy protects against numerous forms of autoimmunity, though its administration entails more risk than anticytokine therapy. We have relied on the injection of naked plasmid DNA into skeletal muscle, with or without enhancement of gene transfer by in vivo electroporation. Expression plasmids offer interesting advantages over viral vectors, since they are simple to produce, non-immunogenic and nonpathogenic. They can be repeatedly administered and after each treatment the encoded proteins are produced for relatively long periods, ranging from weeks to months. Moreover, soluble receptors which block cytokine action, encoded by gene therapy vectors, can be constructed from non-immunogenic self elements that are unlikely to be neutralised by the host immune response (unlike monoclonal antibodies [mAbs]), allowing long-term gene therapy of chronic inflammatory disorders.
Collapse
Affiliation(s)
- G J Prud'homme
- Department of Pathology, McGill University, Montreal, Qc, H3A2B4, Canada.
| | | | | |
Collapse
|
220
|
McCarthy GM, Westfall PR, Masuda I, Christopherson PA, Cheung HS, Mitchell PG. Basic calcium phosphate crystals activate human osteoarthritic synovial fibroblasts and induce matrix metalloproteinase-13 (collagenase-3) in adult porcine articular chondrocytes. Ann Rheum Dis 2001; 60:399-406. [PMID: 11247873 PMCID: PMC1753595 DOI: 10.1136/ard.60.4.399] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVE To determine the ability of basic calcium phosphate (BCP) crystals to induce (a) mitogenesis, matrix metalloproteinase (MMP)-1, and MMP-13 in human osteoarthritic synovial fibroblasts (HOAS) and (b) MMP-13 in cultured porcine articular chondrocytes. METHODS Mitogenesis of HOAS was measured by [3H]thymidine incorporation assay and counts of cells in monolayer culture. MMP messenger RNA (mRNA) accumulation was determined either by northern blot analysis or reverse transcriptase-polymerase chain reaction (RT-PCR) of RNA from chondrocytes or HOAS treated with BCP crystals. MMP-13 secretion was identified by immunoprecipitation and MMP-1 secretion by western blot of conditioned media. RESULTS BCP crystals caused a 4.5-fold increase in [3H]thymidine incorporation by HOAS within 20 hours compared with untreated control cultures (p< or =0.05). BCP crystals induced MMP-13 mRNA accumulation and MMP-13 protein secretion by articular chondrocytes. In contrast, in HOAS, MMP-13 mRNA induced by BCP crystals was detectable only by RT-PCR, and MMP-13 protein was undetectable. BCP crystals induced MMP-1 mRNA accumulation and MMP-1 protein secretion by HOAS. MMP-1 expression was further augmented when HOAS were co-incubated with either BCP and tumour necrosis factor alpha (TNFalpha; threefold) or BCP and interleukin 1alpha (IL1alpha; twofold). CONCLUSION These data confirm the ability of BCP crystals to activate HOAS, leading to the induction of mitogenesis and MMP-1 production. MMP-13 production in response to BCP crystals is substantially more detectable in porcine articular chondrocytes than in HOAS. These data support the active role of BCP crystals in osteoarthritis and suggest that BCP crystals act synergistically with IL1alpha and TNFalpha to promote MMP production and subsequent joint degeneration.
Collapse
Affiliation(s)
- G M McCarthy
- Department of Clinical Pharmacology, The Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland.
| | | | | | | | | | | |
Collapse
|
221
|
Adachi N, Pelinkovic D, Lee CW, Fu FH, Huard J. Gene therapy and the future of cartilage repair. ACTA ACUST UNITED AC 2001. [DOI: 10.1016/s1048-6666(01)80023-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
222
|
Kubo T, Takahashi K, Hirasawa Y. Gene therapies for joint diseases. Mod Rheumatol 2001; 11:10-6. [PMID: 24387014 DOI: 10.3109/s101650170037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- T Kubo
- Department of Orthopaedic Surgery, Kyoto Prefectural University of Medicine , Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566 , Japan
| | | | | |
Collapse
|
223
|
Brower-Toland BD, Saxer RA, Goodrich LR, Mi Z, Robbins PD, Evans CH, Nixon AJ. Direct adenovirus-mediated insulin-like growth factor I gene transfer enhances transplant chondrocyte function. Hum Gene Ther 2001; 12:117-29. [PMID: 11177549 DOI: 10.1089/104303401750061186] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell-based cartilage-resurfacing procedures may be enhanced by the addition of insulin-like growth factor I (IGF-I) to the transplant biomatrix. Given the relatively short half-life of IGF-I in biological systems, however, maintenance of effective concentrations of this peptide necessitates either high initial doses, or repeated treatment. This study investigated IGF-I delivery via adenoviral gene therapy, targeting graftable articular chondrocytes. Cultured articular chondrocytes were infected with an E1-deleted adenoviral vector containing IGF-I-coding sequence under CMV promoter control. Increased adenovirus-IGF-I concentrations resulted in coordinate increase in IGF-I mRNA and ligand expression; however, chondrocyte matrix synthesis was maximized by the lower adenovirus-IGF-I concentration (100 MOI) without additional increase at 200 or 500 MOI. Using 100 MOI, infected monolayers produced medium IGF-I content of at least 10 ng/ml in each 48-hr period for 28 days, reaching a day 4 peak concentration of 66 +/- 4.0 ng/ml. These concentrations were sufficient to produce significant stimulation of normal cartilage matrix gene expression. The concentration of secreted matrix products in medium from infected monolayers was increased up to 8-fold over uninfected control cultures. Moreover, compared with uninfected cultures, cells in infected cultures were more resistant to de-differentiation over time under serum-starved conditions, maintaining a normal chondrocyte molecular phenotype for at least 28 days. These data indicate that cultured chondrocytes are readily transduced by recombinant adenoviral vectors. The adenoviral-IGF transgene is abundantly expressed and its product secreted at therapeutic concentrations for at least 28 days, resulting in increased matrix biosynthesis and maintenance of the chondrocytic phenotype. Combined, this information suggests that there may be significant value in preimplantation adenoviral-IGF gene therapy for chondrocytes destined for cartilage resurfacing.
Collapse
Affiliation(s)
- B D Brower-Toland
- Comparative Orthopaedics Laboratory, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | | | | | | | | | | | | |
Collapse
|
224
|
Brandi ML, Gennari L, Cerinic MM, Becherini L, Falchetti A, Masi L, Gennari C, Reginster JY. Genetic markers of osteoarticular disorders: facts and hopes. ARTHRITIS RESEARCH 2001; 3:270-80. [PMID: 11549368 PMCID: PMC128904 DOI: 10.1186/ar316] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2001] [Revised: 05/08/2001] [Accepted: 05/11/2001] [Indexed: 11/10/2022]
Abstract
Osteoarthritis and osteoporosis are the two most common age-related chronic disorders of articular joints and skeleton, representing a major public health problem in most developed countries. Apart from being influenced by environmental factors, both disorders have a strong genetic component, and there is now considerable evidence from large population studies that these two disorders are inversely related. Thus, an accurate analysis of the genetic component of one of these two multifactorial diseases may provide data of interest for the other. However, the existence of confounding factors must always be borne in mind in interpreting the genetic analysis. In addition, each patient must be given an accurate clinical evaluation, including family history, history of drug treatments, lifestyle, and environment, in order to reduce the background bias. Here, we review the impact of recent work in molecular genetics suggesting that powerful molecular biology techniques will soon make possible both a rapid accumulation of data on the genetics of both disorders and the development of novel diagnostic, prognostic, and therapeutic approaches.
Collapse
Affiliation(s)
- M L Brandi
- Department of Medicine, University of Florence, Viale Morgagni 85, 50135 Florence, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
225
|
Abstract
The pathogenesis of osteoarthritis involves multiple etiologies, including mechanical, biochemical, and genetic factors that contribute to the imbalance in the synthesis and destruction of articular cartilage. It is now well documented that interleukin-1 and tumor necrosis factor-alpha are the predominant proinflammatory and catabolic cytokines involved in disease initiation and progression. Other proinflammatory cytokines may amplify or modulate this process, whereas anti-inflammatory cytokines, which are often detected, paradoxically, in osteoarthritis tissues, may counteract the tissue destruction and inflammation. This review focuses on the role of cytokines in the pathogenesis of osteoarthritis with special emphasis on how findings in culture and animal models may be reflected in the human disease process.
Collapse
Affiliation(s)
- M B Goldring
- Beth Israel Deaconess Medical Center, New England Baptist Bone & Joint Institute, Harvard Institutes of Medicine, Room 246, 4 Blackfan Circle, Boston, MA 02115, USA.
| |
Collapse
|
226
|
Kerschner JE, Beste DJ, Lynch JB, Fox MC, Kehl KS. Interleukin-1 receptor antagonist as an adjunct in the treatment of Haemophilus influenzae otitis media in the chinchilla. Laryngoscope 2000; 110:1457-61. [PMID: 10983942 DOI: 10.1097/00005537-200009000-00009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVES This purpose of this study was to investigate the effect of blockade of the inflammatory cytokine pathway on experimentally induced otitis media in the chinchilla model. STUDY DESIGN Pilot, randomized placebo-controlled trial. METHODS Ampicillin-sensitive Haemophilus influenzae otitis media was induced in 45 adult chinchillas. The animals were randomly assigned to the following treatment groups: 1) transbullar injections (TBI) of interleukin-1 receptor antagonist (IL-1ra) and intramuscular ampicillin, 2) TBI of saline and intramuscular ampicillin, 3) TBI of IL-1ra and intramuscular sa-1 line or 4) TBI of saline and intramuscular saline. Blinded investigators measured resolution of otitis media by otomicroscopy, tympanogram, and culture results. RESULTS Comparisons were made between the treatment groups to assess the ability of IL-1ra to assist with resolution of otitis media using exact two-group binomial tests with the StatXact statistical program. The group with TBI of IL-1ra and intramuscular ampicillin as a treatment demonstrated trends suggesting more rapid resolution of positive cultures and more rapid and complete return to normal results on tympanograms and otomicroscopic findings compared with the group treated with TBI of saline and intramuscular ampicillin. These trends did not achieve statistical significance with the relatively small sample sizes used in this pilot study. CONCLUSIONS This investigation provides further evidence that the inflammatory cytokine cascade plays a significant role in the pathophysiology of otitis media and that modulation of this inflammatory pathway may provide novel and efficacious treatments for otitis media Further studies with larger groups of animals are warranted to determine whether the trends identified in this pilot study are reproducible and achieve statistical significance.
Collapse
Affiliation(s)
- J E Kerschner
- Department of Otolaryngology and Communication Sciences, Medical College of Wisconsin, Milwaukee, USA
| | | | | | | | | |
Collapse
|
227
|
Prud'homme GJ. Gene therapy of autoimmune diseases with vectors encoding regulatory cytokines or inflammatory cytokine inhibitors. J Gene Med 2000; 2:222-32. [PMID: 10953913 DOI: 10.1002/1521-2254(200007/08)2:4<222::aid-jgm117>3.0.co;2-p] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Gene therapy offers advantages for the immunotherapeutic delivery of cytokines or their inhibitors. After gene transfer, these mediators are produced at relatively constant, non-toxic levels and sometimes in a tissue-specific manner, obviating limitations of protein administration. Therapy with viral or nonviral vectors is effective in several animal models of autoimmunity including Type 1 diabetes mellitus (DM), experimental allergic encephalomyelitis (EAE), systemic lupus erythematosus (SLE), colitis, thyroiditis and various forms of arthritis. Genes encoding transforming growth factor beta, interleukin-4 (IL-4) and IL-10 are most frequently protective. Autoimmune/ inflammatory diseases are associated with excessive production of inflammatory cytokines such as IL-1, IL-12, tumor necrosis factor alpha (TNFalpha) and interferon gamma (IFNgamma). Vectors encoding inhibitors of these cytokines, such as IL-1 receptor antagonist, soluble IL-1 receptors, IL-12p40, soluble TNFalpha receptors or IFNgamma-receptor/IgG-Fc fusion proteins are protective in models of either arthritis, Type 1 DM, SLE or EAE. We use intramuscular injection of naked plasmid DNA for cytokine or anticytokine therapy. Muscle tissue is accessible, expression is usually more persistent than elsewhere, transfection efficiency can be increased by low-voltage in vivo electroporation, vector administration is simple and the method is inexpensive. Plasmids do not induce neutralizing immunity allowing repeated administration, and are suitable for the treatment of chronic immunological diseases.
Collapse
Affiliation(s)
- G J Prud'homme
- Department of Pathology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
228
|
Abstract
Articular cartilage, which enables smooth gliding of joints during skeletal motion, is vulnerable to injuries and degenerative diseases over time. Bone growth factors have a role in the preservation of the cartilage matrix. This article reviews the potential to treat cartilage damage for bone morphogenetic proteins, insulin-like growth factors, hepatocyte growth factor, basic fibroblast growth factor, and transforming growth factor beta.
Collapse
Affiliation(s)
- W J O'Connor
- Department of Orthopedic Surgery, Hospital for Special Surgery, New York, New York 10021, USA
| | | | | | | |
Collapse
|
229
|
Kohsaka H. Gene therapy for arthritis. Mod Rheumatol 2000; 10:78-82. [PMID: 24383560 DOI: 10.3109/s101650050003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Abstract An accumulating body of evidence shows that gene therapy can be successfully used to treat animal models of arthritis. Based on this success, a clinical trial of gene therapy for rheumatoid arthritis has been initiated. We review the methods and genes used for the previous gene transfer experiments, including our own. Retroviral ex vivo gene transfer and adenoviral in vivo gene transfer were utilized most frequently. Most of the gene transfer strategies aimed at suppression of synovial inflammation, while our study attempted to convert a phenotype of synovial cells. Gene transfer could be used for part of the future therapy for RA. In basic research studies, gene transfer is of great help in defining new target molecules to treat arthritis.
Collapse
Affiliation(s)
- H Kohsaka
- First Department of Internal Medicine, School of Medicine, Tokyo Medical and Dental University 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8519 , Japan
| |
Collapse
|
230
|
Kim SH, Evans CH, Kim S, Oligino T, Ghivizzani SC, Robbins PD. Gene therapy for established murine collagen-induced arthritis by local and systemic adenovirus-mediated delivery of interleukin-4. ARTHRITIS RESEARCH 2000; 2:293-302. [PMID: 11056670 PMCID: PMC17812 DOI: 10.1186/ar104] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/1999] [Revised: 03/20/2000] [Accepted: 04/05/2000] [Indexed: 12/04/2022]
Abstract
To determine whether IL-4 is therapeutic in treating established experimental arthritis, a recombinant adenovirus carrying the gene that encodes murine IL-4 (Ad-mIL-4) was used for periarticular injection into the ankle joints into mice with established collagen-induced arthritis (CIA). Periarticular injection of Ad-mIL-4 resulted in a reduction in the severity of arthritis and joint swelling compared with saline- and adenoviral control groups. Local expression of IL-4 also reduced macroscopic signs of joint inflammation and bone erosion. Moreover, injection of Ad-mIL-4 into the hind ankle joints resulted in a decrease in disease severity in the untreated front paws. Systemic delivery of murine IL-4 by intravenous injection of Ad-mIL-4 resulted in a significant reduction in the severity of early-stage arthritis.
Collapse
Affiliation(s)
- Seon Hee Kim
- University of Pittsburgh School of Medicine, Pittsburgh,
Pennsylvania, USA
| | | | | | - Thomas Oligino
- University of Pittsburgh School of Medicine, Pittsburgh,
Pennsylvania, USA
| | | | - Paul D Robbins
- University of Pittsburgh School of Medicine, Pittsburgh,
Pennsylvania, USA
| |
Collapse
|
231
|
Abstract
Studies in animal models of osteoarthritis (OA) have been used extensively to gain insight into the pathogenesis of OA, but early studies largely ignored inflammation except as a secondary phenomenon. Synovitis has often been noted as a feature in experimental OA, and more recent work has established a central role for inflammatory cytokines as biochemical signals which stimulate chondrocytes to release cartilage-degrading proteinases. Thus, proteinase inhibitors, cytokine antagonists and receptor blocking antibodies, and growth/differentiation factors have been considered as potential therapeutic agents and targets for gene therapy. Although there is some disagreement, it is generally accepted that IL-1 is the pivotal cytokine at early and late stages, while TNF-alpha is involved primarily in the onset of arthritis. Other cytokines released during the inflammatory process in the OA joint may be regulatory (IL-6, IL-8) or inhibitory (IL-4, IL-10, IL-13, IFN-gamma). Furthermore, studies in animal models have illustrated the potentially beneficial effects of anticytokine therapy with monoclonal antibodies or receptor antagonists, although local rather than systemic delivery would be necessary for the largely localized OA in humans. Transgenic or knockout mice have also provided insights into general mechanisms of cytokine-induced cartilage degradation but have not directly addressed OA pathogenesis. Similarly, animals with spontaneous or transgenic modifications in cartilage matrix components, growth/differentiation factors, or developmentally regulated transcription factors have provided information about potential gene defects that predispose to OA without addressing the role of inflammatory mediators in cartilage destruction. Although the multiple etiologies of human OA indicate that it is more complex than any animal model, the use of appropriate, well-defined animal models will establish the feasibility of novel forms of therapy.
Collapse
Affiliation(s)
- M B Goldring
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
232
|
Moldovan F, Pelletier JP, Jolicoeur FC, Cloutier JM, Martel-Pelletier J. Diacerhein and rhein reduce the ICE-induced IL-1beta and IL-18 activation in human osteoarthritic cartilage. Osteoarthritis Cartilage 2000; 8:186-96. [PMID: 10806046 DOI: 10.1053/joca.1999.0289] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE IL-1beta plays a fundamental role in osteoarthritis (OA) pathophysiology and cartilage destruction. Targeting the activation mechanism of this cytokine appears to be important as a therapeutic approach. As the interleukin-1 converting enzyme (ICE) is the physiologic modulator of the production of active IL-1beta, we investigated the effect of diacerhein and its active metabolite rhein used in the treatment of OA patients, on the enzyme expression and synthesis on human OA cartilage. Further, we looked at the effect of both drugs on the production of the active form of IL-1beta and IL-18. METHODS The expression and synthesis of ICE were investigated on human OA cartilage explants using in-situ hybridization and immunohistochemical methods, respectively. The effect of the drugs on ICE OA chondrocytes was also determined by Northern blotting and a specific ELISA assay. Furthermore, the effect of both drugs on the level of active IL-1beta and IL-18 was examined by immunohistochemistry. RESULTS Data showed that diacerhein and rhein have no true effect on reducing total ICE mRNA by both Northern blotting analysis and in-situ hybridization. A marked and statistically significant decrease was, however, found for protein production. ELISA showed a reduction of 31% (P< 0.04) for diacerhein and 50% (P< 0.02) for rhein. The drugs' immunohistological cell score reduction was similar to data from the ELISA, and a statistical significant reduction of ICE production was found at both superficial and deep zones of the cartilage. IL-1beta and IL-18 were both preferentially produced in chondrocytes of the superficial zone. For each of these cytokines, both drugs demonstrated a statistically significant decrease in this zone. A marked decrease was also noted in the deep zone, but statistical significance was reached only for rhein. CONCLUSION These results provide a novel regulatory mechanism by which diacerhein and rhein could exert a down-regulation on IL-1's effect on OA cartilage.
Collapse
Affiliation(s)
- F Moldovan
- Osteoarthritis Research Unit, Centre hospitalier de l'Université de Montréal (CHUM), Hôpital Notre-Dame, Montréal, Québec, H2L 4M1, Canada
| | | | | | | | | |
Collapse
|
233
|
Croxford JL, Triantaphyllopoulos KA, Neve RM, Feldmann M, Chernajovsky Y, Baker D. Gene therapy for chronic relapsing experimental allergic encephalomyelitis using cells expressing a novel soluble p75 dimeric TNF receptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:2776-81. [PMID: 10679120 DOI: 10.4049/jimmunol.164.5.2776] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In a murine relapsing experimental allergic encephalomyelitis (EAE) model, gene therapy to block TNF was investigated with the use of a retroviral dimeric p75 TNF receptor (dTNFR) construct. To effectively produce these TNF inhibitors in vivo, a conditionally immortalized syngeneic fibroblast line was established, using a temperature-sensitive SV40 large T Ag-expressing retrovirus. These cells were subsequently infected with a retrovirus expressing soluble dTNFR. CNS-injected cells could be detected 3 mo after transplantation and were shown to produce the transgene product by immunocytochemistry and ELISA of tissue fluids. These levels of dTNFR protein were biologically active and could significantly ameliorate both acute and relapsing EAE. This cell-based gene-vector approach is ideal for delivering proteins to the CNS and has particular relevance to the control of inflammatory CNS disease.
Collapse
MESH Headings
- Acute Disease
- Animals
- Antigens, CD/administration & dosage
- Antigens, CD/biosynthesis
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Brain Tissue Transplantation/immunology
- Cell Line, Transformed
- Chronic Disease
- Dimerization
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/therapy
- Fibroblasts/immunology
- Fibroblasts/metabolism
- Genetic Therapy/methods
- Genetic Vectors/chemical synthesis
- Genetic Vectors/immunology
- Humans
- Injections, Intraventricular
- Kidney/cytology
- Mice
- Receptors, Tumor Necrosis Factor/administration & dosage
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type II
- Recurrence
- Solubility
Collapse
Affiliation(s)
- J L Croxford
- Neuroinflammation Group, Institute of Neurology, and Department of Clinical Science, Institute of Ophthalmology, University College London, United Kingdom
| | | | | | | | | | | |
Collapse
|
234
|
Sabatini M, Rolland G, Léonce S, Thomas M, Lesur C, Pérez V, de Nanteuil G, Bonnet J. Effects of ceramide on apoptosis, proteoglycan degradation, and matrix metalloproteinase expression in rabbit articular cartilage. Biochem Biophys Res Commun 2000; 267:438-44. [PMID: 10623638 DOI: 10.1006/bbrc.1999.1983] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Cartilage loss in osteoarthritis is characterized by matrix degradation and chondrocyte death. The lipid messenger ceramide is implicated in signal transduction of the catabolic cytokines tumor necrosis factor (TNF) and interleukin-1 (IL-1), as well as in apoptosis. The aim of this study was to examine the in vitro effects of ceramide on proteoglycan degradation, matrix-metalloproteinase (MMP) expression and activity, and chondrocyte apoptosis in rabbit articular cartilage. Cell-permeant ceramide C(2) stimulated proteoglycan degradation in cartilage explants starting from 3 x 10(-5) M, with 100% increase at the dose of 10(-4) M. This effect was probably due to MMPs since it was blocked by the MMP inhibitor batimastat. Furthermore, in isolated chondrocytes, C(2) stimulated the expression of MMP-1, 3, and 13 at the mRNA level, MMP activity, and MMP-3 production. Ceramide also caused chondrocyte apoptosis at doses ranging from 10(-5) to 10(-4) M. This study supports the hypothesis that ceramide might play a mediatory role in both matrix degradation and apoptosis in processes of cartilage loss such as those observed in osteoarthritis.
Collapse
Affiliation(s)
- M Sabatini
- Division of Rheumatology, Institut de Recherches Servier, Suresnes, France.
| | | | | | | | | | | | | | | |
Collapse
|
235
|
Abstract
IL-1 is a pleiotropic cytokine shown to play a major role in synovitis and in the mechanisms that lead to the progressive joint destruction of rheumatoid arthritis (RA). IL-1 receptor antagonist (IL-1Ra), a member of the IL-1 family, binds IL-1 receptors but does not induce a cellular response. IL-1Ra competitively inhibits the binding of IL-1 to its cell surface receptors and thus acts as an endogenous anti-inflammatory mediator. In different experimental animal models of arthritis systemic administration of IL-1Ra, or local delivery into the joints by gene therapy attenuated the severity of the inflammatory response and reduced articular destruction. In addition, treatment of RA patients with IL-1Ra led to an improvement in different clinical and biological parameters and to a reduction in the radiological signs of joint erosions. Recently, interesting results were obtained using IL-1Ra in combination with methotrexate, a well-known antirheumatic drug, or in combination with other strategies designed to block the effects of tumour necrosis factor (TNF)-alpha. Encouraging results also have been reported in both in vitro and in vivo experimental models of arthritis by using other strategies designed to block the effects of IL-1.
Collapse
Affiliation(s)
- C Gabay
- Division of Rheumatology, University Hospital of Geneva, 26 Avenue Beau-Sejour, 1211 Geneva 14, Switzerland.
| |
Collapse
|
236
|
Martin I, Vunjak-Novakovic G, Yang J, Langer R, Freed LE. Mammalian chondrocytes expanded in the presence of fibroblast growth factor 2 maintain the ability to differentiate and regenerate three-dimensional cartilaginous tissue. Exp Cell Res 1999; 253:681-8. [PMID: 10585291 DOI: 10.1006/excr.1999.4708] [Citation(s) in RCA: 204] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The differentiated phenotype of chondrocytes from hyaline cartilage is gradually lost during expansion in monolayers. Chondrocytes can reexpress their differentiated phenotype by transfer into an environment that prevents cell flattening, but serially passaged cells never completely recover their chondrogenic potential. We report that chondrocytes expanded (up to 2000-fold) in the presence of fibroblast growth factor 2 (FGF-2) dedifferentiated, but fully maintained their potential for redifferentiation in response to environmental changes. After seeding onto three-dimensional polymer scaffolds, chondrocytes expanded in the presence of FGF-2 formed cartilaginous tissue that was histologically and biochemically comparable to that obtained using primary chondrocytes, in contrast to chondrocytes expanded to the same degree but in the absence of FGF-2. The presence of FGF-2 inhibited the formation of thick F-actin structures, which otherwise formed during monolayer expansion, were maintained during tissue cultivation, and were associated with reduced ability of chondrocytes to reexpress their differentiated phenotype. This study provides evidence that FGF-2 maintains the chondrogenic potential during chondrocyte expansion in monolayers, possibly due to changes in the architecture of F-actin elements and allows more efficient utilization of harvested tissue for cartilage tissue engineering.
Collapse
Affiliation(s)
- I Martin
- Division of Health Sciences & Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | | | | | | | | |
Collapse
|
237
|
Nishida K, Kang JD, Gilbertson LG, Moon SH, Suh JK, Vogt MT, Robbins PD, Evans CH. Modulation of the biologic activity of the rabbit intervertebral disc by gene therapy: an in vivo study of adenovirus-mediated transfer of the human transforming growth factor beta 1 encoding gene. Spine (Phila Pa 1976) 1999; 24:2419-25. [PMID: 10626303 DOI: 10.1097/00007632-199912010-00002] [Citation(s) in RCA: 237] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN In vivo studies using a rabbit model to determine the biologic effects of direct, adenovirus-mediated transfer of a therapeutic gene to the intervertebral disc. OBJECTIVES 1) To deliver an exogenous therapeutic gene to rabbit lumbar intervertebral discs in vivo, 2) to quantify the resulting amount of gene expression, and 3) to determine the effect on the biologic activity of the discs. SUMMARY OF BACKGROUND DATA Although growth factors such as transforming growth factor beta 1 appear to have promising therapeutic properties, there currently is no practical method for sustained delivery of exogenous growth factors to the disc for the management of certain chronic types of disease (e.g., disc degeneration). A possible solution is to modify the disc cells genetically through gene transfer such that the cells manufacture the desired growth factors endogenously on a continuous basis. METHODS Saline, with or without virus, was injected directly into lumbar discs of 22 skeletally mature female New Zealand white rabbits. Group 1 (n = 11) received the adenovirus construct Ad/CMV-hTGF beta 1 containing the therapeutic human transforming growth factor beta 1-encoding gene. Group 2 (n = 6) received adenovirus containing the luciferase marker gene. Group 3 (n = 5) received saline only. The rabbits were killed 1 week after injection. Immunohistochemical staining for human transforming growth factor beta 1 was performed on the disc tissues of one rabbit from Group 1. Nucleus pulposus tissues from the remaining rabbits were cultured in serumless medium. Bioassays were performed to determine human transforming growth factor beta 1 production and proteoglycan synthesis. RESULTS Discs injected with Ad/CMV-hTGF beta 1 exhibited extensive and intense positive immunostaining for transforming growth factor beta 1. The nucleus pulposus tissues from the discs injected with Ad/CMV-hTGF beta 1 exhibited a 30-fold increase in active transforming growth factor beta 1 production, and a 5-fold increase in total (active + latent) transforming growth factor beta 1 production over that from intact control discs (P < 0.05). Furthermore, these tissues exhibited a 100% increase in proteoglycan synthesis compared with intact control tissue, which was statistically significant (P < 0.05). CONCLUSIONS The results of this study suggest that the intervertebral disc is an appropriate site for adenovirus-mediated transfer of exogenous genes and subsequent production of therapeutic growth factors. Gene therapy therefore may have useful applications for study of the basic science of the intervertebral disc and for clinical management of degenerative disc disease.
Collapse
Affiliation(s)
- K Nishida
- Department of Orthopaedic Surgery, University of Pittsburgh, Pennsylvania, USA
| | | | | | | | | | | | | | | |
Collapse
|
238
|
|
239
|
Presle N, Cipolletta C, Jouzeau JY, Abid A, Netter P, Terlain B. Cartilage protection by nitric oxide synthase inhibitors after intraarticular injection of interleukin-1beta in rats. ARTHRITIS AND RHEUMATISM 1999; 42:2094-102. [PMID: 10524680 DOI: 10.1002/1529-0131(199910)42:10<2094::aid-anr9>3.0.co;2-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To evaluate the effect of nitric oxide synthase (NOS) inhibitors on proteoglycan synthesis following intraarticular administration of interleukin-1beta (IL-1beta) in rats. METHODS Recombinant human IL-1beta and NOS inhibitors with different selectivity for inducible NOS (N-monomethyl-L-arginine [L-NMA], N-iminoethyl-L-ornithine [L-NIO], and S-methylisothiourea [SMT]) were simultaneously administered in rats by a single intraarticular injection in each knee. L-NMA was also infused for 72 hours using an Alzet mini osmotic pump implanted into the peritoneal cavity 24 hours before IL-1beta challenge. NO production was determined as nitrate and nitrite, either in synovial fluid or ex vivo in supernatants of synovium and patellae. Proteoglycan synthesis was measured by ex vivo incorporation of 35SO4(2-) into patellar cartilage. RESULTS IL-1beta induced a time-dependent increase in NO production in synovial fluid. Synovium and patellae released large amounts of nitrate and nitrite under ex vivo conditions, indicating that both tissues are effective sources of NO within the joint. This production of NO was accompanied by a delayed inhibition of proteoglycan synthesis. The intraarticular administration of L-NMA and L-NIO reduced NO release in synovial fluid and resulted in a partial recovery of proteoglycan synthesis. Under our experimental conditions, SMT failed to reduce NO synthesis and to restore proteoglycan synthesis. The protection of cartilage was improved by the systemic and sustained delivery of L-NMA. However, the complete inhibition of NO production in synovial fluid was not sufficient to fully restore cartilage anabolism. CONCLUSION Our findings show that in rats: 1) NO may be an early mediator of the effect of IL-1beta on cartilage, 2) NO inhibition may have therapeutic relevance, although it is not sufficient to fully reverse the deleterious effects of IL-1beta, 3) among NOS inhibitors tested, only amino acid derivatives are effective, 4) protection can be achieved by local administration of NOS inhibitors, and 5) systemic and sustained delivery of the NOS inhibitor with the highest efficacy after intraarticular injection provides the most benefit.
Collapse
Affiliation(s)
- N Presle
- CNRS, Vandoeuvre les Nancy, France
| | | | | | | | | | | |
Collapse
|
240
|
McCarty MF, Russell AL. Niacinamide therapy for osteoarthritis--does it inhibit nitric oxide synthase induction by interleukin 1 in chondrocytes? Med Hypotheses 1999; 53:350-60. [PMID: 10608273 DOI: 10.1054/mehy.1998.0792] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Fifty years ago, Kaufman reported that high-dose niacinamide was beneficial in osteoarthritis (OA) and rheumatoid arthritis. A recent double-blind study confirms the efficacy of niacinamide in OA. It may be feasible to interpret this finding in the context of evidence that synovium-generated interleukin-1 (IL-1), by inducing nitric oxide (NO) synthase and thereby inhibiting chondrocyte synthesis of aggrecan and type II collagen, is crucial to the pathogenesis of OA. Niacinamide and other inhibitors of ADP-ribosylation have been shown to suppress cytokine-mediated induction of NO synthase in a number of types of cells; it is therefore reasonable to speculate that niacinamide will have a comparable effect in IL-1-exposed chondrocytes, blunting the anti-anabolic impact of IL-1. The chondroprotective antibiotic doxycycline may have a similar mechanism of action. Other nutrients reported to be useful in OA may likewise intervene in the activity or synthesis of IL-1. Supplemental glucosamine can be expected to stimulate synovial synthesis of hyaluronic acid; hyaluronic acid suppresses the anti-catabolic effect of IL-1 in chondrocyte cell cultures, and has documented therapeutic efficacy when injected intra-articularly. S-adenosylmethionine (SAM), another proven therapy for OA, upregulates the proteoglycan synthesis of chondrocytes, perhaps because it functions physiologically as a signal of sulfur availability. IL-1 is likely to decrease SAM levels in chondrocytes; supplemental SAM may compensate for this deficit. Adequate selenium nutrition may down-regulate cytokine signaling, and ample intakes of fish oil can be expected to decrease synovial IL-1 production; these nutrients should receive further evaluation in OA. These considerations suggest that non-toxic nutritional regimens, by intervening at multiple points in the signal transduction pathways that promote the synthesis and mediate the activity of IL-1, may provide a substantially superior alternative to NSAIDs (merely palliative and often dangerously toxic) in the treatment and perhaps prevention of OA.
Collapse
|
241
|
Kuboki T, Nakanishi T, Kanyama M, Sonoyama W, Fujisawa T, Kobayashi K, Ikeda T, Kubo T, Yamashita A, Takigawa M. Direct adenovirus-mediated gene delivery to the temporomandibular joint in guinea-pigs. Arch Oral Biol 1999; 44:701-9. [PMID: 10471154 DOI: 10.1016/s0003-9969(99)00069-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Adenovirus 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, recombinant adenovirus vectors harbouring beta-galactosidase gene (LacZ) was injected directly into the temporomandibular joints of Hartley guinea-pigs to clarify the in vivo transfer availability of the adenovirus vectors. Specifically, recombinant adenovirus harbouring LacZ gene (AxlCALacZ) was injected into the upper joint cavities of both mandibular joints of four male 6-week-old Hartley guinea-pigs. Either the same amount of recombinant adenovirus without LacZ gene (Axlw) suspension (placebo) or the same amount of phosphate-buffered saline solution (control) were injected into the upper joint cavities of both joints of another four male guinea-pigs. At 1, 2, 3 and 4 weeks after injection, the joints were dissected and the expression of delivered LacZ was examined by 5-bromo-4-chloro-3-indolyl-beta-D-galactopyranoside (X-gal) staining and reverse transcriptase-polymerase chain reaction (RT-PCR). To investigate the expression of transferred gene in other organs, total RNA was extracted from liver, kidney, heart and brain and the expression of LacZ mRNA and 18 S ribosomal RNA were analysed by RT-PCR. Clear expression of LacZ was observed in the articular surfaces of the temporal tubercle, articular disc and synovium of the temporomandibular joints even 4 weeks after injection in the AxlCALacZ-injected group, while no expression was detected in placebo and control groups. Histological examination confirmed that LacZ activity was clearly detected in a few cell layers of the articular surface tissues, which is much more efficient than in a previously study of the knee joint. In the other organs, expression of the delivered transgene was not observed. Based on these findings, direct gene delivery into the articular surface of the temporomandibular joint using the adenovirus vector is feasible as an effective in vivo method.
Collapse
Affiliation(s)
- T Kuboki
- Department of Fixed Prosthodontics, Okayama University Dental School, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
242
|
Saha N, Moldovan F, Tardif G, Pelletier JP, Cloutier JM, Martel-Pelletier J. Interleukin-1beta-converting enzyme/caspase-1 in human osteoarthritic tissues: localization and role in the maturation of interleukin-1beta and interleukin-18. ARTHRITIS AND RHEUMATISM 1999; 42:1577-87. [PMID: 10446854 DOI: 10.1002/1529-0131(199908)42:8<1577::aid-anr3>3.0.co;2-z] [Citation(s) in RCA: 105] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To study the expression and production of interleukin-1beta-converting enzyme (ICE) in human normal and osteoarthritic (OA) cartilage and synovium, quantitate the level of ICE in OA chondrocytes, and examine the relationship between the topographic distribution of ICE, interleukin-1beta (IL-1beta), and IL-18, as well as apoptosis of chondrocytes. METHODS The expression and synthesis of ICE were investigated in human normal and OA cartilage and synovial membrane using in situ hybridization and immunohistochemical methods. The intracellular level of ICE in OA chondrocytes was also measured by enzyme-linked immunosorbent assay (ELISA). Furthermore, the topographic relationship between the presence of ICE and mature IL-1beta and IL-18 was examined by immunohistochemistry, and apoptotic chondrocytes by the TUNEL technique. RESULTS ICE was expressed and synthesized in both human synovial membrane and cartilage, with a significantly greater number of cells staining positive in OA tissue than in normal tissue. ICE production was preferentially located in the superficial and upper intermediate layers of articular cartilage. With a specific ELISA, a level of 230.2+/-22.5 pg/5 x 10(5) cells (mean +/- SEM) of ICE was found in OA chondrocytes. In cartilage, IL-1beta and IL-18 stained positive at a topographic location similar to that of ICE. The production of mature IL-1beta in OA cartilage explants and chondrocytes was completely blocked by treatment with a specific ICE inhibitor, which also markedly diminished the number of IL-18-positive cells. The data show that there was no close relationship between the presence of ICE and the presence of apoptotic chondrocytes in OA cartilage. CONCLUSION This study shows, for the first time, the presence of active ICE in human articular cartilage, with a markedly increased cellular level in OA tissue. The relationship between active IL-1beta and ICE suggests that ICE may promote OA progression by activating this proinflammatory cytokine. The role of IL-18 in pathologic cartilage is discussed.
Collapse
Affiliation(s)
- N Saha
- Centre Hospitalier de l'Université de Montréal, Notre-Dame Hospital, Quebec, Canada
| | | | | | | | | | | |
Collapse
|
243
|
Abstract
OA is common, debilitating, costly, incurable, and, in many cases, resistant to treatment. Novel approaches to therapy are clearly required. Progress in understanding the biology of cartilage and OA have led to our suggestion of a gene therapy approach to treatment. Genes whose products stimulate chondrogenesis or inhibit breakdown of the cartilaginous matrix are obviously candidates for therapeutic use. These genes may be transferred to the synovium or cartilage of affected joints by in vivo or ex vivo means using a variety of vectors. Transfer of such genes to chondroprogenitor cells is a particularly attractive approach.
Collapse
Affiliation(s)
- C H Evans
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pennsylvania, USA.
| | | |
Collapse
|
244
|
Abstract
Gene therapy in simple terms is the introduction of a gene into a cell, in vivo, in order to ameliorate a disease process. Human clinical trials have focused on the correction of monogenic deficiency diseases, cancer and AIDS. This paper summarises the technology of gene therapy, gives a brief synopsis of the current applications of gene therapy to veterinary medicine and discusses some of the problems which need to be overcome so that gene therapy can become accepted clinical practice.
Collapse
Affiliation(s)
- D J Argyle
- Department of Veterinary Clinical Studies, University of Glasgow Veterinary School
| |
Collapse
|
245
|
Fernandes J, Tardif G, Martel-Pelletier J, Lascau-Coman V, Dupuis M, Moldovan F, Sheppard M, Krishnan BR, Pelletier JP. In vivo transfer of interleukin-1 receptor antagonist gene in osteoarthritic rabbit knee joints: prevention of osteoarthritis progression. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:1159-69. [PMID: 10233854 PMCID: PMC1866546 DOI: 10.1016/s0002-9440(10)65368-0] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/06/1998] [Indexed: 11/28/2022]
Abstract
The goal of this study was to determine the efficacy of local IL-1Ra gene therapy by intra-articular plasmid injections on structural changes in the meniscectomy rabbit model of osteoarthritis. A partial meniscectomy of the right knee was performed on the rabbits through a medial parapatellar incision. The rabbits were then divided into four experimental groups. Group 1 received no treatment. Group 2 received three consecutive intra-articular injections at 24-hour intervals of 0.9% saline containing a lipid, gammaAP-DLRIE/DOPE, and a DNA plasmid, VR1012. Group 3 received three consecutive injections of saline containing 1000 microg of canine IL-1Ra plasmid and lipid. The injections were given starting 4 weeks post-surgery. Rabbits from Group 1 were killed 4 weeks post-surgery, and all other rabbits 8 weeks post-surgery. The severity of macroscopic and microscopic changes on cartilage on the medial and femoral condyles and tibial plateaus and synovium were graded separately. Specimens were also processed for immunohistochemical staining using a rabbit polyclonal antibody against canine IL-1Ra. The level of canine IL-1Ra in synovial fluid was determined using enzyme-linked immunosorbent assay. The presence of the DNA plasmid in the synovium was tested by polymerase chain reaction. A significant reduction in the width of osteophytes and size of macroscopic lesions (P < 0.04) was observed, and was dependent on the amount of IL-1Ra plasmid injected. A significant reduction was also noted in the severity of histologic cartilage lesions (P < 0.01) in the group that received the highest dosage (1000 microg) of IL-1Ra plasmid. IL-1Ra was detected in synovial fluid by enzyme-linked immunosorbent assay and by immunohistochemical staining in the synovium and cartilage of rabbits that received injections containing the IL-1Ra plasmid. Polymerase chain reaction analysis of synovial DNA revealed the presence of the cloned cDNA dog IL-1Ra up to 4 weeks after the first intra-articular injection. This study demonstrates that direct in vivo transfer of the IL-1Ra gene into osteoarthritis knee cells using intra-articular injections of a plasmid vector and lipids can significantly reduce the progression of experimental osteoarthritis. This avenue may therefore represent a promising future treatment for osteoarthritis.
Collapse
Affiliation(s)
- J Fernandes
- Osteoarthritis Research Unit, Centre Hospitalier de l'Université de Montréal, Campus Notre-Dame, Montréal, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Müller-Ladner U, Evans CH, Franklin BN, Roberts CR, Gay RE, Robbins PD, Gay S. Gene transfer of cytokine inhibitors into human synovial fibroblasts in the SCID mouse model. ARTHRITIS AND RHEUMATISM 1999; 42:490-7. [PMID: 10088772 DOI: 10.1002/1529-0131(199904)42:3<490::aid-anr14>3.0.co;2-l] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate the effects of retrovirus-based gene delivery of inhibitory cytokines and cytokine inhibitors into human synovial fibroblasts in the SCID mouse model of rheumatoid arthritis (RA). METHODS The MFG vector was used for gene delivery of tumor necrosis factor alpha receptor (TNFalphaR) p55, viral interleukin-10 (IL-10), and murine IL-10 into RA synovial fibroblasts. The effect on invasion of these cells into human articular cartilage and on perichondrocytic cartilage degradation was examined after 60 days of coimplantation into the SCID mouse. RESULTS TNFalphaR p55 gene transfer showed only a limited effect on inhibition of RA synovial fibroblast invasiveness and cartilage degradation. In contrast, invasion of the RA synovial fibroblasts into the coimplanted cartilage was strongly inhibited by both viral and murine IL-10. Perichondrocytic cartilage degradation was not affected by either form of IL-10. CONCLUSION The data show that cytokines can be successfully inserted into the genome of human RA synovial fibroblasts using a retroviral vector delivery system, and that the SCID mouse model of human RA is a valuable tool for examining the effects of gene transfer. In addition, inhibition of more than one cytokine pathway may be required to inhibit both synovial- and chondrocyte-mediated cartilage destruction in RA.
Collapse
Affiliation(s)
- U Müller-Ladner
- Department of Internal Medicine I, University of Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
247
|
Evans CH, Ghivizzani SC, Kang R, Muzzonigro T, Wasko MC, Herndon JH, Robbins PD. Gene therapy for rheumatic diseases. ARTHRITIS AND RHEUMATISM 1999; 42:1-16. [PMID: 9920008 DOI: 10.1002/1529-0131(199901)42:1<1::aid-anr1>3.0.co;2-4] [Citation(s) in RCA: 128] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- C H Evans
- University of Pittsburgh School of Medicine, Pennsylvania, USA
| | | | | | | | | | | | | |
Collapse
|
248
|
Melchiorri C, Meliconi R, Frizziero L, Silvestri T, Pulsatelli L, Mazzetti I, Borzì RM, Uguccioni M, Facchini A. Enhanced and coordinated in vivo expression of inflammatory cytokines and nitric oxide synthase by chondrocytes from patients with osteoarthritis. ARTHRITIS AND RHEUMATISM 1998; 41:2165-74. [PMID: 9870873 DOI: 10.1002/1529-0131(199812)41:12<2165::aid-art11>3.0.co;2-o] [Citation(s) in RCA: 196] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVE To evaluate the sites of expression of interleukin-1beta (IL-1beta), tumor necrosis factor alpha (TNFalpha), and inducible nitric oxide synthase (iNOS) in patients with inflammatory and degenerative joint diseases. METHODS Cytokines and iNOS were detected by immunohistochemistry analysis of synovial and cartilage biopsy specimens obtained at knee arthroscopy in patients with rheumatoid arthritis (RA), psoriatic arthritis (PsA), osteoarthritis (OA), and traumatic knee arthritis. Cytokine and iNOS expression was quantified using computerized image analysis. RESULTS IL-1beta, TNFalpha, and iNOS were highly expressed by synovial cells (lining layer cells, infiltrating leukocytes, endothelial cells) from patients with inflammatory arthritides and significantly less by synovial cells from patients with OA and traumatic arthritis. In contrast, the latter patients showed high chondrocyte expression of cytokines and iNOS while RA and PsA patients had only minor chondrocyte positivity. In both joint compartments, IL-1beta expression, TNFalpha expression, and iNOS expression were strongly correlated. CONCLUSION The enhanced and coordinated expression of IL-1beta, TNFalpha, and iNOS by chondrocytes strongly supports the hypothesis that chondrocytes are the major site of production of mediators of inflammation in human OA, thus playing a primary role in the pathogenesis of this disease.
Collapse
Affiliation(s)
- C Melchiorri
- Istituto Codivilla Putti-Istituti Ortopedici Rizzoli, Bologna, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Nishida K, Kang JD, Suh JK, Robbins PD, Evans CH, Gilbertson LG. Adenovirus-mediated gene transfer to nucleus pulposus cells. Implications for the treatment of intervertebral disc degeneration. Spine (Phila Pa 1976) 1998; 23:2437-42; discussion 2443. [PMID: 9836359 DOI: 10.1097/00007632-199811150-00016] [Citation(s) in RCA: 136] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN In vitro and in vivo studies using a rabbit model were performed to determine the feasibility of adenovirus-mediated gene transfer to the intervertebral disc. OBJECTIVES This study was conducted to determine whether it is possible to transfer genes to cells within the intervertebral disc by direct injection of an adenovirus and to determine the duration of gene expression obtained by this method. SUMMARY OF BACKGROUND DATA Although growth factors have the potential to stimulate the regeneration of nucleus pulposus, sustained delivery of growth factors to a degenerated disc is clinically unfeasible with present technology. Novel approaches such as gene transfer should be investigated as possible solutions to this problem. METHODS The lacZ marker gene was used to evaluate gene delivery to cells within intervertebral discs. For the in vitro study, cell cultures were established from the nucleus pulposus tissue of New Zealand white rabbits and infected with an adenovirus encoding the lacZ gene (Ad-lacZ). For the in vivo study, the anterior aspects of lumbar intervertebral discs were surgically exposed, and Ad-lacZ in saline solution was directly injected into the nucleus pulposus. An equal volume of saline only was injected into control discs. Expression of the transferred gene was detected by staining with 5-bromo-4-chloro-3-indolyl-beta-galactosidase (X-Gal). RESULTS The in vitro experiments confirmed that nucleus pulposus cells were efficiently transduced by an adenoviral vector carrying the lacZ gene. In vivo injection of Ad-lacZ into the nucleus pulposus resulted in the transduction of a considerable number of cells. Marker gene expression in vivo persisted at an apparently undiminished level for at least 12 weeks. No staining was noted in control discs. CONCLUSIONS The results show the feasibility of adenovirus-mediated gene transfer to the intervertebral disc. Expression of the marker gene persisted at least 12 weeks in vivo. This successful demonstration of exogenous gene transfer to the disc and sustained, long-term expression suggests that the adenoviral vector may be suitable for delivery of appropriate genes to the disc for the treatment of spinal disorders.
Collapse
Affiliation(s)
- K Nishida
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pennsylvania, USA
| | | | | | | | | | | |
Collapse
|
250
|
Wooley PH, Sud S, Whalen JD, Nasser S. Pristane-induced arthritis in mice. V. Susceptibility to pristane-induced arthritis is determined by the genetic regulation of the T cell repertoire. ARTHRITIS AND RHEUMATISM 1998; 41:2022-31. [PMID: 9811058 DOI: 10.1002/1529-0131(199811)41:11<2022::aid-art18>3.0.co;2-p] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE Pristane-induced arthritis (PIA) is an experimental seropositive arthritis that is characterized by serologic and cellular immune abnormalities and is dependent on the presence of a competent CD4+ T cell population. We examined the regulation of PIA by genes of the major histocompatibility complex (MHC) and the Mls-1 loci to determine whether the selection of the T cells that infiltrate arthritic joints is a critical factor in disease susceptibility. METHODS Genetic regulation of PIA was investigated using F1 hybrid and congenic strain analysis to determine the influence of MHC and Mls-1 genes. The T cell receptor Vbeta phenotypes of lymph node cells and T cells infiltrating arthritic joints were examined with 2-color flow cytometry and reverse transcription-polymerase chain reaction techniques. RESULTS F1 hybrid offspring from 2 major PIA-susceptible strains (DBA/1 x BALB/c) were resistant to the induction of arthritis because of the interaction between genes of the MHC and the Mls-1 loci, which modified the T cell repertoire. This conclusion was supported by the observed resistance to PIA in BALB/ c-Mls-1a mice, where T cells expressing the Vbeta8.1 and Vbeta6 phenotypes were absent. The receptor phenotype of T cells infiltrating arthritic joints in DBA/1 mice was markedly skewed toward Vbeta8.1 and Vbeta6 compared with the population observed in lymph nodes from either PIA or normal control DBA/1 mice. CONCLUSION The data support the hypothesis that PIA is a T cell-mediated disease. While pristane causes a polyclonal T cell expansion that gives rise to lymphadenopathy, the development of arthritis in susceptible strains of mice occurs due to the preservation of specific T cell subsets with the capacity to infiltrate synovial joints.
Collapse
Affiliation(s)
- P H Wooley
- Wayne State University Medical School, Detroit, Michigan, USA
| | | | | | | |
Collapse
|