Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur Cell Mater ; 36: 156-170, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30311630

RESUMO

Successful clinical translation of mesenchymal stem cell (MSC)-based therapies for cartilage repair will likely require the implementation of standardised protocols and broadly applicable tools to facilitate the comparisons among cell types and chondroinduction methods. The present study investigated the utility of recombinant lentiviral reporter vectors as reliable tools for comparing chondrogenic potential among primary cell populations and distinguishing cellular-level variations of chondrogenic activity in widely used three-dimensional (3D) culture systems. Primary equine MSCs and chondrocytes were transduced with vectors containing combinations of fluorescent and luciferase reporter genes under constitutive cytomeglavirus (CMV) or chondrocyte-lineage (Col2) promoters. Reporter activity was measured by fluorescence imaging and luciferase assay. In 3D cultures of MSC aggregates and polyethylene glycol-hyaluronic acid (PEG-HA) hydrogels, transforming growth factor beta 3 (TGF-ß3)-mediated chondroinduction increased Col2 reporter activity, demonstrating close correlation with histology and mRNA expression levels of COL2A1 and SOX9. Comparison of chondrogenic activities among MSC populations using a secretable luciferase reporter revealed enhanced chondrogenesis in bone-marrow-derived MSCs relative to MSC populations from synovium and adipose tissues. A dual fluorescence reporter - enabling discrimination of highly chondrogenic (Col2-GFP) cells within an MSC population (CMV-tdTomato) - revealed marked heterogeneity in differentiating aggregate cultures and identified chondrogenic cells in chondrocyte-seeded PEG-HA hydrogels after 6 weeks in a subcutaneous implant model - indicating stable, long-term reporter expression in vivo. These results suggested that lentiviral reporter vectors may be used to address fundamental questions regarding chondrogenic activity in chondroprogenitor cell populations and accelerate clinical translation of cell-based cartilage repair strategies.


Assuntos
Condrócitos/metabolismo , Condrogênese , Genes Reporter , Lentivirus/genética , Animais , Agregação Celular , Diferenciação Celular , Células Cultivadas , Condrócitos/citologia , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Fluorescência , Cavalos , Ácido Hialurônico/farmacologia , Hidrogênio/farmacologia , Implantes Experimentais , Luciferases/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Polietilenoglicóis/farmacologia , Regiões Promotoras Genéticas/genética
2.
Gene Ther ; 20(6): 670-7, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23151520

RESUMO

With the long-term goal of developing a gene-based treatment for osteoarthritis (OA), we performed studies to evaluate the equine joint as a model for adeno-associated virus (AAV)-mediated gene transfer to large, weight-bearing human joints. A self-complementary AAV2 vector containing the coding regions for human interleukin-1-receptor antagonist (hIL-1Ra) or green fluorescent protein was packaged in AAV capsid serotypes 1, 2, 5, 8 and 9. Following infection of human and equine synovial fibroblasts in culture, we found that both were only receptive to transduction with AAV1, 2 and 5. For these serotypes, however, transgene expression from the equine cells was consistently at least 10-fold higher. Analyses of AAV surface receptor molecules and intracellular trafficking of vector genomes implicate enhanced viral uptake by the equine cells. Following delivery of 1 × 10(11) vector genomes of serotypes 2, 5 and 8 into the forelimb joints of the horse, all three enabled hIL-1Ra expression at biologically relevant levels and effectively transduced the same cell types, primarily synovial fibroblasts and, to a lesser degree, chondrocytes in articular cartilage. These results provide optimism that AAV vectors can be effectively adapted for gene delivery to large human joints affected by OA.


Assuntos
Dependovirus/genética , Técnicas de Transferência de Genes , Proteína Antagonista do Receptor de Interleucina 1/genética , Osteoartrite/genética , Animais , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Cartilagem Articular/virologia , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Cavalos , Humanos , Interleucina-1/genética , Articulações/metabolismo , Articulações/patologia , Articulações/virologia , Osteoartrite/terapia , Membrana Sinovial/metabolismo , Membrana Sinovial/patologia , Membrana Sinovial/virologia
3.
J Cell Physiol ; 227(2): 416-20, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21948071

RESUMO

Orthopedic gene therapy has been the topic of considerable research for two decades. The preclinical data are impressive and many orthopedic conditions are well suited to genetic therapies. But there have been few clinical trials and no FDA-approved product exists. This paper examines why this is so. The reasons are multifactorial. Clinical translation is expensive and difficult to fund by traditional academic routes. Because gene therapy is viewed as unsafe and risky, it does not attract major funding from the pharmaceutical industry. Start-up companies are burdened by the complex intellectual property environment and difficulties in dealing with the technology transfer offices of major universities. Successful translation requires close interactions between scientists, clinicians and experts in regulatory and compliance issues. It is difficult to create such a favorable translational environment. Other promising fields of biological therapy have contemplated similar frustrations approximately 20 years after their founding, so there seem to be more general constraints on translation that are difficult to define. Gene therapy has noted some major clinical successes in recent years, and a sense of optimism is returning to the field. We hope that orthopedic applications will benefit collaterally from this upswing and move expeditiously into advanced clinical trials.


Assuntos
Terapia Genética/métodos , Doenças Musculoesqueléticas/genética , Doenças Musculoesqueléticas/terapia , Ortopedia/métodos , Ensaios Clínicos como Assunto , Humanos , Doenças Musculoesqueléticas/patologia , Medicina Regenerativa , Engenharia Tecidual
4.
Gene Ther ; 16(8): 944-52, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19675584

RESUMO

Gene therapies directed toward the treatment of arthritis and tissue repair continue to be the most active areas of research for bone and joint diseases. In the past 2 years, two trials in rheumatoid arthritis have been completed. a Phase I study reporting safety and a Phase I/II study that has yet to be published. An additional, small study has reported the first evidence of clinical efficacy. Two Phase I trials of gene therapy for osteoarthritis have also been initiated. There is much preclinical activity in developing AAV vectors for future trials in the gene therapy of arthritis. Research into tissue repair and regeneration remains at the preclinical stage, but a considerable volume of research attests to the promise of gene transfer in this arena, especially in the context of bone healing. For tissue repair, the major research questions are still which genes to use and how best to deliver them.


Assuntos
Doenças Ósseas/terapia , Artropatias/terapia , Artrite/terapia , Ensaios Clínicos como Assunto , Técnicas de Transferência de Genes , Terapia Genética
5.
Gene Ther ; 15(5): 357-63, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18283289

RESUMO

Based upon the powerful bridging and charge-masking properties of lanthanide cations (Ln3+), we have investigated their use to improve the transduction efficiency of adenovirus vectors. Using a luciferase marker gene, it was possible to increase transgene expression by the murine mesenchymal stem cell line C3H10T(1/2) by up to four log orders when using very low multiplicities of infection in conjunction with Ln3+; La3+ was superior to Gd3+, Y3+ and Lu3+ in this regard. All Ln3+ were more effective than Ca2+. Flow cytometry, using a green fluorescent protein marker gene, confirmed that La3+ increased both the percentage of transduced cells and the level of transgene expression per cell. Transduction of primary cultures of a variety of different mesenchymal cells from human, rabbit, bovine and rat sources, as well as gene transfer to synovium and muscle in vivo, was also greatly enhanced. Our findings suggest that this lanthanide-based method holds much promise for expediting both experimental and clinical applications of gene transfer with adenoviral vectors.


Assuntos
Adenoviridae/genética , Terapia Genética/métodos , Vetores Genéticos/genética , Elementos da Série dos Lantanídeos/farmacologia , Transdução Genética/métodos , Animais , Cátions , Bovinos , Linhagem Celular , Expressão Gênica , Engenharia Genética , Proteínas de Fluorescência Verde/genética , Humanos , Proteína Antagonista do Receptor de Interleucina 1/genética , Luciferases/genética , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Coelhos , Ratos , Ratos Wistar , Transgenes , Ítrio/farmacologia
6.
Adv Drug Deliv Rev ; 58(2): 243-58, 2006 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-16563557

RESUMO

Osteoarthritis (OA) is common, debilitating, expensive, incurable and very difficult to treat. Gene transfer to the synovial linings of affected joints is a promising strategy for achieving sustained, therapeutic, intraarticular concentrations of anti-arthritic gene products. This is not reasonably possible with existing, alternative technologies. The present review summarizes progress in achieving direct, in vivo intraarticular gene delivery and expression. Numerous non-viral vectors have been evaluated for their ability to transfect the synovia of experimental animals following intraarticular injection. None have given more than low levels of temporary transgene expression and many are inflammatory. Several viral vectors, however, are very effective in this regard and successfully treat experimental models of OA. Adeno-associated virus has been used in a phase I study for the gene therapy of rheumatoid arthritis. Its use in a clinical trial for treating OA is pending.


Assuntos
Terapia Genética , Osteoartrite/terapia , Animais , Artrite Reumatoide/genética , Artrite Reumatoide/terapia , Ensaios Clínicos como Assunto , Vetores Genéticos , Humanos , Vírus/genética
7.
Gene Ther ; 12(2): 177-86, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15578043

RESUMO

Combination of growth factor gene-enhanced cartilage matrix synthesis with interleukin-1 receptor antagonist protein (IL-1Ra) abrogation of cartilage matrix degradation may reduce and possibly reverse cartilage loss in synovitis and osteoarthritis. The feasibility of cotransduction of synovial membrane with two such genes that may act on cartilage homeostasis was investigated in an in vitro coculture system. Cultured synoviocytes in monolayer were cotransduced with E1-deleted adenoviral vectors, one containing IGF-I coding sequence under cytomegalovirus (CMV) promoter control (200 multiplicities of infection (moi)), and the second containing IL-1Ra sequence under CMV promoter control (100 moi). Adenovirus-IGF-I (AdIGF-I) transduction and AdIGF-I/AdIL-1Ra cotransduction of synovial monolayer cultures resulted in increased IGF-I mRNA and ligand expression, and similarly AdIL-1Ra and AdIGF-I/AdIL-1Ra-transduced cultures expressed high levels of IL-1Ra. Northern analysis confirmed a single mRNA transcript of the appropriate size for both IGF-I and IL-1Ra transgene expression. Synovial cell monolayer and cartilage explant coculture experiments were used to examine the effects of IGF-I and IL-1Ra protein expressed by transduced synoviocytes on normal and IL-1-depleted cartilage. Transduced monolayer cultures produced peak medium IGF-I content of 114+/-20.2 ng/ml and IL-1Ra levels of 241.8+/-10.5 ng/ml at 48 h after transduction. These IGF-I concentrations were sufficient to produce significantly increased proteoglycan (PG) content of normal cartilage cultured in medium conditioned by AdIGF-I and AdIGF-I/AdIL-1Ra-transduced synoviocytes. Interleukin-1-exposed cartilage was markedly depleted of PG, and this catabolic state was partially reversed in AdIGF-I-transduced cultures and fully reversed by AdIGF-I/AdIL-1Ra-transduced synovial cocultures. These data indicate that cultured synoviocytes are readily cotransduced by two recombinant adenoviral vectors containing transgenes active in restoring joint health. The AdIL-1Ra and AdIGF-I transgenes were abundantly expressed and the secreted products achieved therapeutic concentrations by day 2. The resulting increase in matrix biosynthesis returned cartilage PG content to normal levels. These data suggest that there may be significant value in cotransduction of synovial membrane to attenuate cartilage malacia associated with synovitis, injury, or early arthritis.


Assuntos
Cartilagem/patologia , Terapia Genética/métodos , Fator de Crescimento Insulin-Like I/genética , Osteoartrite/terapia , Sialoglicoproteínas/genética , Sinovite/terapia , Adenoviridae/genética , Animais , Cartilagem/imunologia , Expressão Gênica , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Cavalos , Proteína Antagonista do Receptor de Interleucina 1 , Modelos Animais , Osteoartrite/imunologia , Osteoartrite/patologia , Sinovite/imunologia , Sinovite/patologia , Técnicas de Cultura de Tecidos , Transdução Genética/métodos
8.
Osteoarthritis Cartilage ; 12(3): 217-24, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14972338

RESUMO

OBJECTIVE: To determine whether overexpression of glutamine: fructose-6-phosphate amidotransferase (GFAT) in synoviocytes will antagonize the response to interleukin-1beta (IL-1beta) of chondrocytes and synovial fibroblasts in co-culture. METHODS: Synovial fibroblasts from the rat were transduced by an adenovirus carrying the cDNA for GFAT and then co-cultured with rat chondrocytes encapsulated in alginate beads. Following challenge with 1, 5, or 10 ng/ml of IL-1beta for 24 h, proteoglycan synthesis by the chondrocytes was determined by incorporation of Na2(35)SO4. Production of nitric oxide (NO) and prostaglandin E2 (PGE2) were monitored by assay of conditioned medium from the co-culture. RESULTS: IL-1beta treatment of untransduced-synoviocyte/chondrocyte co-cultures resulted in markedly decreased proteoglycan synthesis by the chondrocytes, and increased NO and PGE2 levels in the culture medium. In contrast, adenovirus-mediated transfer of GFAT in synoviocytes prevented both the decrease in chondrocyte proteoglycan synthesis and increases in NO and PGE2 provoked by IL-1beta. CONCLUSIONS: Our study suggests that in a synoviocyte/chondrocyte co-culture system, overexpression of GFAT by synoviocytes significantly inhibits subsequent stimulation by IL-1beta in vitro. Since GFAT is the rate limiting enzyme in the synthesis of intracellular glucosamine and its derivatives, these results may open new possibilities for osteoarthritis treatment.


Assuntos
Condrócitos/efeitos dos fármacos , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante)/genética , Interleucina-1/farmacologia , Adenoviridae/genética , Animais , Condrócitos/metabolismo , Técnicas de Cocultura , Dinoprostona/biossíntese , Expressão Gênica , Vetores Genéticos , Glutamina-Frutose-6-Fosfato Transaminase (Isomerizante)/metabolismo , Masculino , Óxido Nítrico/biossíntese , Proteoglicanas/biossíntese , Ratos , Membrana Sinovial/citologia , Membrana Sinovial/metabolismo , Transdução Genética , Transgenes
9.
Gene Ther ; 11(2): 133-41, 2004 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-14712297

RESUMO

The long-term goal of the present study is to develop a clinically applicable approach to enhance natural repair mechanisms within cartilage lesions by targeting bone marrow-derived cells for genetic modification. To determine if bone marrow-derived cells infiltrating osteochondral defects could be transduced in situ, we implanted collagen-glycosaminoglycan (CG) matrices preloaded with adenoviral vectors containing various marker genes into lesions surgically generated in rabbit femoral condyles. Analysis of the recovered implants showed transgenic expression up to 21 days; however, a considerable portion was found in the synovial lining, indicating leakage of the vector and/or transduced cells from the matrix. As an alternative medium for gene delivery, we investigated the feasibility of using coagulated bone marrow aspirates. Mixture of an adenoviral suspension with the fluid phase of freshly aspirated bone marrow resulted in uniform dispersion of the vector throughout, and levels of transgenic expression in direct proportion to the density of nucleated cells in the ensuing clot. Furthermore, cultures of mesenchymal progenitor cells, previously transduced ex vivo with recombinant adenovirus, were readily incorporated into the coagulate when mixed with fresh aspirate. These vector-seeded and cell-seeded bone marrow clots were found to maintain their structural integrity following extensive culture and maintained transgenic expression in this manner for several weeks. When used in place of the CG matrix as a gene delivery vehicle in vivo, genetically modified bone marrow clots were able to generate similarly high levels of transgenic expression in osteochondral defects with better containment of the vector within the defect. Our results suggest that coagulates formed from aspirated bone marrow may be useful as a means of gene delivery to cartilage and perhaps other musculoskeletal tissues. Cells within the fluid can be readily modified with an adenoviral vector, and the matrix formed from the clot is completely natural, native to the host and is the fundamental platform on which healing and repair of mesenchymal tissues is based.


Assuntos
Adenoviridae/genética , Transplante de Medula Óssea , Doenças das Cartilagens/terapia , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Animais , Doenças das Cartilagens/patologia , Expressão Gênica , Modelos Animais , Coelhos , Transplante de Células-Tronco , Transdução Genética/métodos , Transgenes , Transplante Autólogo
10.
Gene Ther ; 11(4): 351-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14724680

RESUMO

Gene transfer technology has opened novel treatment avenues toward the treatment of damaged musculoskeletal tissues, and may be particularly beneficial to articular cartilage. There is no natural repair mechanism to heal damaged or diseased cartilage. Existing pharmacologic, surgical and cell based treatments may offer temporary relief but are incapable of restoring damaged cartilage to its normal phenotype. Gene transfer provides the capability to achieve sustained, localized presentation of bioactive proteins or gene products to sites of tissue damage. A variety of cDNAs have been cloned which may be used to stimulate biological processes that could improve cartilage healing by (1) inducing mitosis and the synthesis and deposition of cartilage extracellular matrix components by chondrocytes, (2) induction of chondrogenesis by mesenchymal progenitor cells, or (3) inhibiting cellular responses to inflammatory stimuli. The challenge is to adapt this technology into a useful clinical treatment modality. Using different marker genes, the principle of gene delivery to synovium, chondrocytes and mesenchymal progenitor cells has been convincingly demonstrated. Following this, research efforts have begun to move to functional studies. This involves the identification of appropriate gene or gene combinations, incorporation of these cDNAs into appropriate vectors and delivery to specific target cells within the proper biological context to achieve a meaningful therapeutic response. Methods currently being explored range from those as simple as direct delivery of a vector to a cartilage defect, to synthesis of cartilaginous implants through gene-enhanced tissue engineering. Data from recent efficacy studies provide optimism that gene delivery can be harnessed to guide biological processes toward both accelerated and improved articular cartilage repair.


Assuntos
Cartilagem Articular/lesões , Terapia Genética/métodos , Animais , Condrócitos/metabolismo , Técnicas de Transferência de Genes , Substâncias de Crescimento/genética , Humanos , Membrana Sinovial/metabolismo , Cicatrização
11.
Gene Ther ; 11(4): 379-89, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14724685

RESUMO

Osteoarthritis (OA) is the Western world's leading cause of disability. It is incurable, costly and responds poorly to treatment. This review discusses strategies for treating OA by gene therapy. As OA affects a limited number of weight-bearing joints and has no major extra-articular manifestations, it is well suited to local, intra-articular gene therapy. Possible intra-articular sites of gene transfer include the synovium and the cartilage. Most experimental progress has been made with gene transfer to synovium, a tissue amenable to genetic modification by a variety of vectors, using both in vivo and ex vivo protocols. The focus so far has been upon the transfer of genes whose products enhance synthesis of the cartilaginous matrix, or inhibit its breakdown, although there is certainly room for alternative targets. It is possible to build a convincing case implicating interleukin-1 (IL-1) as a key mediator of cartilage loss in OA, and the therapeutic effects of IL-1 receptor anatagonist (IL-1Ra) gene transfer have been confirmed in three different experimental models of OA. As transfer of IL-1Ra cDNA to human arthritic joints has already been accomplished safely, we argue that clinical studies of intra-articular IL-1Ra gene transfer in OA are indicated and should be funded. Of the available vector systems, recombinant adeno-associated virus may provide the best combination of safety with in vivo delivery using current technology.


Assuntos
Terapia Genética/métodos , Osteoartrite/terapia , Animais , Modelos Animais de Doenças , Técnicas de Transferência de Genes , Humanos , Proteína Antagonista do Receptor de Interleucina 1 , Receptores de Interleucina-1/antagonistas & inibidores , Sialoglicoproteínas/genética
12.
Gene Ther ; 10(12): 1055-60, 2003 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12776164

RESUMO

Rheumatoid arthritis (RA) is an inflammatory autoimmune disease that primarily affects joints. In rheumatoid joints there is extensive synovial proliferation with diseased synovium becoming highly aggressive, attaching to the articular cartilage and bone to form what is termed a pannus. The formation of active pannus is central to erosive disease and resulting joint destruction. In this study, we examined the ability to eliminate the hyperplastic synovium by adenoviral-mediated gene transfer of human TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF family that is able to induce apoptosis through interaction with receptors containing death domains, DR4 and DR5. Infection of synovial cells derived from RA patients with Ad.TRAIL resulted in significant apoptosis in three out of five lines. Moreover, primary rabbit synovial fibroblasts were also sensitive to Ad.TRAIL-mediated gene transfer. In a rabbit model of arthritis, intra-articular gene transfer of TRAIL induced apoptosis in cells within the synovial lining, reduced leukocytic infiltration and stimulated new matrix synthesis by cartilage. These results demonstrate that TRAIL can affect the viability of the cells populating the activated synovium in arthritic joints and suggest that the delivery of TRAIL to arthritic joints may represent a non-invasive mechanism for inducing pannus regression.


Assuntos
Artrite Experimental/terapia , Técnicas de Transferência de Genes , Terapia Genética/métodos , Glicoproteínas de Membrana/genética , Membrana Sinovial/patologia , Fator de Necrose Tumoral alfa/genética , Adenoviridae/genética , Animais , Apoptose , Proteínas Reguladoras de Apoptose , Artrite Experimental/patologia , Artrite Reumatoide/patologia , Linhagem Celular , Vetores Genéticos , Humanos , Hiperplasia , Injeções Intra-Articulares , Glicoproteínas de Membrana/fisiologia , Coelhos , Ligante Indutor de Apoptose Relacionado a TNF , Fator de Necrose Tumoral alfa/fisiologia
13.
Hum Gene Ther ; 13(7): 791-802, 2002 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-11975846

RESUMO

Our primary objective was to fabricate a porous gene-supplemented collagen-glycosaminoglycan (GSCG) matrix for sustained delivery (over a period of several weeks) of plasmid DNA to articular chondrocytes when implanted into cartilage lesions. The specific aims of this in vitro study were to determine the release kinetics profiles of plasmid DNA from the GSCG matrices, and to determine the ability of the released plasmid DNA to transfect adult canine articular chondrocytes. In particular, we evaluated the effects of two variables, cross-linking treatment and the pH at which the DNA was incorporated into the matrices, on the amount of the plasmid DNA that remained bound to the GSCG matrices after passive (nonenzymatic) leaching and on the expression of a reporter gene in articular chondrocytes grown in the GSCG matrices. Collagen-glycosaminoglycan matrices were synthesized without cross-linking, and by three cross-linking treatments: dehydrothermal (DHT) treatment, 1-ethyl-3-(3-dimethyl aminopropyl) carbodiimide (EDC) treatment, and exposure to ultraviolet (UV) radiation. The plasmid DNA was incorporated into the collagen-glycosaminoglycan matrices in solutions at pH 2.5 or 7.5. Transmission electron microscopy studies revealed plasmid DNA bound to the walls of the porous GSCG matrices. In general, the GSCG matrices fabricated at pH 2.5 retained a larger fraction of the initial DNA load after 28 days of incubation in Tris-EDTA buffer. The passive, solvent-mediated release of the plasmid DNA from the GSCG matrices showed a biphasic pattern consisting of a faster, early release rate over the initial 8 hr of leaching followed by a slower, late release rate that was relatively constant over the subsequent 28 days of leaching. Electrophoretic analyses revealed that the plasmid DNA released from the GSCG matrices fabricated at pH 2.5 had been linearized and/or degraded; whereas the plasmid DNA leached from the GSCG matrices prepared with a DNA solution at pH 7.5 was primarily supercoiled and linear. Plasmid DNA released from all GSCG matrix formulations was able to generate luciferase reporter gene expression in monolayer-cultured chondrocytes transfected with the aid of a commercial lipid reagent, and in chondrocytes cultured in the GSCG matrices without the aid of a supplemental transfection reagent. Luciferase expression in chondrocyte-seeded GSCG constructs was evident throughout the culture period (28 days), with the EDC and UV cross-linked matrices prepared at pH 7.5 providing the highest transgene expression levels. We conclude that released plasmid DNA continually transfected canine articular chondrocytes seeded into GSCG matrices in vitro for a 4-week period as evidenced by luciferase reporter gene expression. Thus, GSCG matrices can be fabricated to provide sustained release of plasmid DNA carrying a potential therapeutic gene.


Assuntos
Colágeno/metabolismo , Técnicas de Transferência de Genes , Glicosaminoglicanos/metabolismo , Plasmídeos/metabolismo , Animais , Células Cultivadas , Condrócitos/metabolismo , DNA/metabolismo , Cães , Eletroforese em Gel de Ágar , Terapia Genética/métodos , Concentração de Íons de Hidrogênio , Cinética , Luciferases/metabolismo , Microscopia Eletrônica , Microscopia Eletrônica de Varredura , Ligação Proteica , Fatores de Tempo , Transfecção , Raios Ultravioleta
14.
Gene Ther ; 9(1): 12-20, 2002 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11850718

RESUMO

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.


Assuntos
Terapia Genética/métodos , Doenças dos Cavalos/terapia , Osteoartrite/terapia , Osteoartrite/veterinária , Receptores de Interleucina-1/antagonistas & inibidores , Adenoviridae/genética , Análise de Variância , Animais , Expressão Gênica , Vetores Genéticos/administração & dosagem , Cavalos , Injeções Intra-Articulares , Modelos Animais , Transdução Genética/métodos
15.
Expert Opin Biol Ther ; 1(6): 971-8, 2001 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11728228

RESUMO

Rheumatoid arthritis (RA) is a disabling, painful disorder affecting 1% of the world's population. Although the aetiology of RA remains unknown, recent advances in understanding its pathophysiology have led to the characterisation of several proteins whose activities may be anti-arthritic. Clinical application of such proteins has greatly improved the treatment of RA, but the disease remains incurable and difficult to manage in a substantial number of patients. Thus, there are continued efforts to develop new therapeutic strategies. Because RA is a chronic condition, effective treatment will probably require the presence of therapeutic agents for extended periods of time. In the case of proteins, this is problematic. Gene therapy may offer a solution to this problem. Experimental studies have confirmed the feasibility, efficacy and safety of gene therapy for the treatment of animal models of arthritis. Several different approaches have shown promise in this regard, including gene transfer to the synovial lining cells of individual joints and the systemic delivery of genes to extra-articular locations. One unexpected finding has been the 'contralateral effect' in which gene delivery to one joint of an animal with polyarticular disease leads to improvement of multiple joints. Investigation of this phenomenon has led to interest in cell trafficking and the genetic modification of antigen-presenting cells (APC). The first Phase I clinical trial tested the feasibility and safety of ex vivo gene transfer to the synovial lining of human joints. This clinical trial has been successfully completed and two other Phase I trials are in progress. A Phase II study is now being planned to investigate the efficacy of gene transfer to the joints of patients with early stage RA.


Assuntos
Artrite Reumatoide/terapia , Terapia Genética , Ensaios Clínicos como Assunto , Vetores Genéticos , Humanos
16.
Mol Ther ; 3(6): 901-10, 2001 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-11407904

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease that primarily affects joints. During the pathogenesis of rheumatoid arthritis, the synovial lining becomes dramatically thickened and hyperplastic. This highly aggressive tissue invades and destroys articular cartilage and bone. Several lines of evidence suggest that the proliferation of the synovial tissue may be due to disruption in the control of the cell cycle or apoptotic pathways. In particular, mutations in the tumor suppressor protein p53 have been found in synovial tissue from RA joints. We have examined the effects of overexpression of p53 by adenoviral infection in synovial cells in culture and in synovial tissue in vivo in a rabbit model of arthritis. Here we demonstrate that p53 overexpression resulted in significant apoptosis in human and rabbit synovial cells in culture. Furthermore, intraarticular injection of Ad-p53 resulted in extensive and rapid induction of synovial apoptosis in the rabbit knee without affecting cartilage metabolism. Interestingly, a significant reduction in the leukocytic infiltrate was observed within 24 h postinfection of Ad.p53. These results suggest that intraarticular gene transfer of p53 is able to induce synovial apoptosis as well as reduce inflammation and thus may be useful clinically for the treatment of RA.


Assuntos
Adenoviridae/genética , Apoptose/genética , Artrite Reumatoide/terapia , Cartilagem Articular/patologia , Membrana Sinovial/patologia , Proteína Supressora de Tumor p53/genética , Animais , Artrite Reumatoide/induzido quimicamente , Artrite Reumatoide/patologia , Células Cultivadas , Fibroblastos , Vetores Genéticos , Humanos , Marcação In Situ das Extremidades Cortadas , Técnicas In Vitro , Interleucina-1/farmacologia , Articulação do Joelho/patologia , Óperon Lac/fisiologia , Proteoglicanas/biossíntese , Coelhos , Sais de Tetrazólio , Tiazóis , Proteína Supressora de Tumor p53/metabolismo
17.
Bone ; 28(5): 499-506, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11344049

RESUMO

The identification of bone morphogenetic proteins (BMPs) has stimulated intense interest in BMP delivery approaches. Ex vivo BMP-2 gene delivery has recently been described using skeletal muscle-derived cells. Skeletal muscle-derived cells, because of proven efficient transgene delivery and osteocompetence, represent an attractive cell population on which to base ex vivo BMP-2 gene delivery. However, the early in vivo fate of BMP-2-expressing muscle-derived cells is unknown. This study investigates the in vivo effects of BMP-2 secretion on skeletal muscle-derived cells in terms of cell survival and cell differentiation. The first experiment compared survival of BMP-2-expressing cells with control cells during the first 48 h after in vivo implantation. The results demonstrate that BMP-2 secretion did not adversely affect cell survival 8, 24, or 48 h after intramuscular implantation. The second experiment histologically compared the fate of BMP-2-expressing muscle-derived cells to the same cells not expressing BMP-2. The results show that BMP-2 expression prevented in vivo myogenic differentiation and promoted osteogenic differentiation of the transduced cells. This study further supports the existence of osteoprogenitor cells residing within skeletal muscle. Moreover, it is demonstrated that BMP-2 secretion does not adversely affect early cell survival of muscle-derived cells. These data are important for future investigations into BMP-2 gene delivery approaches to the musculoskeletal system.


Assuntos
Doenças Ósseas/terapia , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular/fisiologia , Células Cultivadas/transplante , Sobrevivência de Enxerto/fisiologia , Músculo Esquelético/transplante , Transplante de Células-Tronco , Fator de Crescimento Transformador beta , Animais , Desenvolvimento Ósseo/genética , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/metabolismo , Osso e Ossos/citologia , Osso e Ossos/metabolismo , Técnicas de Cultura de Células , Genes Reporter/fisiologia , Vetores Genéticos , Membro Posterior/diagnóstico por imagem , Membro Posterior/crescimento & desenvolvimento , Membro Posterior/metabolismo , Camundongos , Desenvolvimento Muscular , Músculo Esquelético/citologia , Músculo Esquelético/crescimento & desenvolvimento , Radiografia , Fatores de Tempo , Transdução Genética , beta-Galactosidase/análise , beta-Galactosidase/genética
18.
Arthritis Res ; 3(3): 142-6, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11299054

RESUMO

Recombinant adenoviruses are straightforward to produce at high titres, have a promiscuous host-range, and, because of their ability to infect nondividing cells, lend themselves to in vivo gene delivery. Such advantages have led to their widespread and successful use in preclinical studies of arthritis gene therapy. While adenoviral vectors are well suited to 'proof of principle' experiments in laboratory animals, there are several barriers to their use in human studies at this time. Transient transgene expression limits their application to strategies, such as synovial ablation, which do not require extended periods of gene expression. Moreover, there are strong immunological barriers to repeat dosing. In addition, safety concerns predicate local, rather than systemic, delivery of the virus. Continued engineering of the adenoviral genome is producing vectors with improved properties, which may eventually overcome these issues. Promising avenues include the development of 'gutted' vectors encoding no endogenous viral genes and of adenovirus-AAV chimeras. Whether these will offer advantages over existing vectors, which may already provide safe, long-term gene expression following in vivo delivery, remains to be seen.


Assuntos
Adenovírus Humanos/genética , Artrite Reumatoide/terapia , Terapia Genética , Osteoartrite/terapia , Expressão Gênica , Vetores Genéticos , Humanos , Recombinação Genética
19.
Curr Rheumatol Rep ; 3(1): 79-85, 2001 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11177774

RESUMO

Rheumatoid arthritis (RA) is a painful chronic disorder. Conventional therapies are palliative, not curative. Advances in the understanding of the pathophysiology of RA have led to the development of new therapeutic strategies, including gene therapy. Multiple studies in several different animal models provide proof supporting the use of gene therapy in arthritis. A phase I clinical trial has already been performed successfully on nine women with end-stage RA in the United States, and two other trials are in progress. Limited duration of gene expression impedes the development of a clinically useful genetic treatment for arthritis.


Assuntos
Artrite Reumatoide/genética , Terapia Genética , Animais , Artrite Reumatoide/imunologia , Ensaios Clínicos Fase I como Assunto , Expressão Gênica/genética , Expressão Gênica/imunologia , Vetores Genéticos/genética , Vetores Genéticos/imunologia , Vetores Genéticos/uso terapêutico , HIV/genética , HIV/imunologia , Humanos , Vírus da Leucemia Murina de Moloney/genética , Vírus da Leucemia Murina de Moloney/imunologia , Estados Unidos
20.
Drug Discov Today ; 6(5): 259-267, 2001 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11182599

RESUMO

Gene therapy offers a novel and innovative approach to the delivery of therapeutic proteins to the joints of patients with arthritis. Several viral vectors, including adenovirus, adeno-associated virus, retrovirus and herpes simplex virus, are capable of delivering exogenous cDNAs to the synovial lining, enabling effective levels of intra-articular transgene expression following direct injection to the joint. The expression of certain gene products has proven to be sufficient to inhibit the progression of disease in animals with experimental arthritis. Non-viral methods of gene transfer, however, are less satisfactory, and are limited by toxicity and transience of expression. Although the principle of direct gene delivery to the joint has been demonstrated, maintaining persistent intra-articular transgene expression remains a challenge.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...