Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
J Transl Med ; 16(1): 56, 2018 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-29523155

RESUMEN

BACKGROUND: MIV-711 is a highly potent and selective cathepsin K inhibitor. The current article summarizes the therapeutic effects of MIV-711 on joint pathology in rabbits subjected to anterior cruciate ligament transection (ACLT), and the prophylactic effects on joint pathology in dogs subjected to partial medial meniscectomy, two surgical models of osteoarthritis (OA). METHODS: Starting 1 week after surgery, rabbits were dosed daily via oral gavage with either MIV-711 or vehicle (n = 7/group) for 7 weeks. The four treatment groups were: (1) sham + vehicle; (2) ACLT + vehicle; (3) ACLT + MIV-711, 30 µmol/kg and (4) ACLT + MIV-711, 100 µmol/kg. Subchondral bone and articular cartilage structures were assessed by µCT, histomorphometry, and scoring. Dogs subjected to partial medial meniscectomy received either MIV-711 (30 µmol/kg) or vehicle (n = 15/group) via oral gavage once daily, starting 1 day before meniscectomy, for 28 days. Cartilage degradation was assessed at the macroscopic and microscopic levels. The exposures of MIV-711 were assessed in both studies and biomarkers reflecting bone resorption (HP-1 in rabbits, CTX-I in dogs) and cartilage degradation (CTX-II) were measured. RESULTS: In ACLT rabbits, MIV-711 decreased HP-1 levels by up to 72% (p < 0.001) and CTX-II levels by up to 74% (p < 0.001) compared to ACLT vehicle controls. ACLT surgery significantly reduced the total thickness of the subchondral bone plate and reduced trabecular bone volume in the femur and tibia. These effects were reversed by MIV-711. ACLT resulted in cartilage thickening, which was attenuated by MIV-711. MIV-711 did not affect osteophyte formation or Mankin scores. In dogs, MIV-711 reduced CTX-I and CTX-II levels by 86% (p < 0.001) and 80% (p < 0.001), respectively. Synovial CTX-II levels were reduced by 55-57% (p < 0.001) compared to baseline. MIV-711-treated animals had 25-37% lower macroscopic scores in the femur condyles and 13-33% lower macroscopic scores in the tibial plateaus. CONCLUSIONS: MIV-711 prevents subchondral bone loss and partially attenuates cartilage pathology in two animal models of OA. These beneficial effects of MIV-711 on joint pathology are observed in conjunction with decreases in bone and cartilage biomarkers that have been shown to be clinically attainable in human. The data support the further development of MIV-711 for the treatment of OA.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior/tratamiento farmacológico , Catepsina K/antagonistas & inhibidores , Inhibidores de Cisteína Proteinasa/uso terapéutico , Articulaciones/patología , Osteoartritis/tratamiento farmacológico , Animales , Ligamento Cruzado Anterior , Biomarcadores/sangre , Biomarcadores/orina , Resorción Ósea/patología , Cartílago Articular/diagnóstico por imagen , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Inhibidores de Cisteína Proteinasa/sangre , Inhibidores de Cisteína Proteinasa/farmacocinética , Inhibidores de Cisteína Proteinasa/farmacología , Modelos Animales de Enfermedad , Perros , Femenino , Articulaciones/diagnóstico por imagen , Articulaciones/efectos de los fármacos , Masculino , Compuestos Orgánicos , Osteoartritis/sangre , Osteoartritis/diagnóstico por imagen , Osteoartritis/patología , Análisis de Componente Principal , Conejos
2.
Int J Toxicol ; 37(1): 4-27, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29264927

RESUMEN

As the need for nasal, ocular, spinal, and articular therapeutic compounds increases, toxicology assessments of drugs administered via these routes play an important role in human safety. This symposium outlined the local and systemic evaluation to support safety during the development of these drugs in nonclinical models with some case studies. Discussions included selection of appropriate species for the intended route; conducting nonclinical studies that closely mimic the intended use with adequate duration; functional assessment, if deemed necessary; evaluation of local tissues with special histological staining procedure; and evaluations of safety margins based on local and systemic toxicity.


Asunto(s)
Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Preparaciones Farmacéuticas/administración & dosificación , Administración Intranasal/efectos adversos , Humanos , Inyecciones Intraarticulares/efectos adversos , Inyecciones Intraoculares/efectos adversos , Inyecciones Espinales/efectos adversos
3.
Mol Pharmacol ; 92(6): 694-706, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28974538

RESUMEN

Bromodomain (BD) and extra-terminal domain containing proteins (BET) are chromatin adapters that bind acetylated histone marks via two tandem BDs, BD1 and BD2, to regulate gene transcription. BET proteins are involved in transcriptional reprogramming in response to inflammatory stimuli. BET BD inhibitors (BETis) that are nonselective for BD1 or BD2 have recognized anti-inflammatory properties in vitro and counter pathology in models of inflammation or autoimmune disease. Although both BD1 and BD2 bind acetylated histone residues, they may independently regulate the expression of BET-sensitive genes. Here we characterized the ability of RVX-297, a novel orally active BETi with selectivity for BD2, to modulate inflammatory processes in vitro, in vivo, and ex vivo. RVX-297 suppressed inflammatory gene expression in multiple immune cell types in culture. Mechanistically, RVX-297 displaced BET proteins from the promoters of sensitive genes and disrupted recruitment of active RNA polymerase II, a property shared with pan-BETis that nonselectively bind BET BDs. In the lipopolysaccharide model of inflammation, RVX-297 reduced proinflammatory mediators assessed in splenic gene expression and serum proteins. RVX-297 also countered pathology in three rodent models of polyarthritis: rat and mouse collagen-induced arthritis, and mouse collagen antibody-induced arthritis. Further, RVX-297 prevented murine experimental autoimmune encephalomyelitis (a model of human multiple sclerosis) disease development when administered prophylactically and reduced hallmarks of pathology when administered therapeutically. We show for the first time that a BD2-selective BETi maintains anti-inflammatory properties and is effective in preclinical models of acute inflammation and autoimmunity.


Asunto(s)
Antiinflamatorios/farmacología , Artritis/tratamiento farmacológico , Enfermedades Autoinmunes/tratamiento farmacológico , Proteínas/antagonistas & inhibidores , Quinazolinonas/uso terapéutico , Enfermedad Aguda , Animales , Antiinflamatorios/uso terapéutico , Anticuerpos/inmunología , Artritis/inducido químicamente , Artritis/inmunología , Artritis/patología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Artritis Reumatoide/patología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/patología , Linfocitos B/efectos de los fármacos , Linfocitos B/metabolismo , Células Cultivadas , Colágeno/inmunología , Citocinas/biosíntesis , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipopolisacáridos/farmacología , Masculino , Ratones Endogámicos C57BL , Ratas Endogámicas Lew , Bazo/efectos de los fármacos , Bazo/metabolismo , Bazo/patología , Células U937
4.
Ann Rheum Dis ; 76(1): 295-302, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27381034

RESUMEN

OBJECTIVE: To investigate whether the effects of nerve growth factor (NGF) inhibition with tanezumab on rats with medial meniscal tear (MMT) effectively model rapidly progressive osteoarthritis (RPOA) observed in clinical trials. METHODS: Male Lewis rats underwent MMT surgery and were treated weekly with tanezumab (0.1, 1 or 10 mg/kg), isotype control or vehicle for 7, 14 or 28 days. Gait deficiency was measured to assess weight-bearing on the operated limb. Joint damage was assessed via histopathology. A second arm, delayed onset of treatment (starting 3-8 weeks after MMT surgery) was used to control for analgesia early in the disease process. A third arm, mid-tibial amputation, evaluated the dependency of the model on weight-bearing. RESULTS: Gait deficiency in untreated rats was present 3-7 days after MMT surgery, with a return to normal weight-bearing by days 14-28. Prophylactic treatment with tanezumab prevented gait deficiency and resulted in more severe cartilage damage. When onset of treatment with tanezumab was delayed to 3-8 weeks after MMT surgery, there was no increase in cartilage damage. Mid-tibial amputation completely prevented cartilage damage in untreated MMT rats. CONCLUSIONS: These data suggest that analgesia due to NGF inhibition during the acute injury phase is responsible for increased voluntary weight-bearing and subsequent cartilage damage in the rat MMT model. This model failed to replicate the hypotrophic bone response observed in tanezumab-treated patients with RPOA.


Asunto(s)
Anticuerpos Monoclonales Humanizados/farmacología , Cartílago Articular/lesiones , Factor de Crecimiento Nervioso/antagonistas & inhibidores , Lesiones de Menisco Tibial/tratamiento farmacológico , Animales , Anticuerpos Monoclonales Humanizados/administración & dosificación , Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticuerpos Monoclonales Humanizados/toxicidad , Artritis Experimental/inducido químicamente , Cartílago Articular/efectos de los fármacos , Cartílago Articular/patología , Cartílago Articular/fisiopatología , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Esquema de Medicación , Evaluación Preclínica de Medicamentos/métodos , Marcha , Masculino , Meniscos Tibiales/diagnóstico por imagen , Meniscos Tibiales/patología , Radiografía , Ratas Endogámicas Lew , Lesiones de Menisco Tibial/diagnóstico por imagen , Lesiones de Menisco Tibial/patología , Lesiones de Menisco Tibial/fisiopatología , Soporte de Peso , Microtomografía por Rayos X
5.
Mod Rheumatol ; 27(5): 838-848, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27846748

RESUMEN

OBJECTIVES: This study was performed to evaluate the potential efficacy of natural eggshell membrane (NEM) in collagen-induced arthritic rats, a well-established rodent model of inflammation and rheumatoid arthritis. METHODS: Rats with developing type II collagen-induced arthritis (CIA) were treated once daily by oral gavage on study days -14 to 17 with vehicle or NEM (52 mg/kg body weight). Rats were euthanized on study day 17. Efficacy was assessed by daily ankle caliper measurements, ankle diameter expressed as area under the curve (AUCd0-17), and histopathologic evaluation of ankles and knees. Serum biomarkers of cartilage function and inflammation [collagen type II C-telopeptide (CTXII), cartilage oligomeric matrix protein (COMP), and alpha-2-macroglobulin (A2M)] were measured by ELISA. RESULTS: Treatment with NEM resulted in significant beneficial effects on the daily ankle diameter measurements and ankle diameter AUC. Ankle and knee histopathology scores were significantly reduced (36% and 43% reduction of summed individual histopathology scores for ankle and knee, respectively; p < 0.05) toward normal for rats given NEM compared to vehicle controls. The percent reduction of serum CTXII, COMP, and A2M in NEM-treated rats ranged from 30% to 72% (p < 0.05). CONCLUSIONS: NEM significantly improved multiple aspects of inflammatory arthritis including inflammation, pannus, cartilage damage, bone resorption, and periosteal bone formation. This study provides further support for the use of CTXII, COMP, and A2M as relevant biomarkers that were responsive to NEM.


Asunto(s)
Artritis Experimental , Cáscara de Huevo , Animales , Antirreumáticos/farmacología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Artritis Experimental/patología , Productos Biológicos/farmacología , Conservadores de la Densidad Ósea/farmacología , Proteína de la Matriz Oligomérica del Cartílago/análisis , Colágeno Tipo II/análisis , Inflamación/tratamiento farmacológico , Masculino , Fragmentos de Péptidos/análisis , Ratas , Resultado del Tratamiento
6.
J Toxicol Pathol ; 29(3 Suppl): 49S-103S, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27621538

RESUMEN

The INHAND (International Harmonization of Nomenclature and Diagnostic Criteria for Lesions in Rats and Mice) Project (www.toxpath.org/inhand.asp) is an initiative of the Societies of Toxicological Pathology from Europe (ESTP), Great Britain (BSTP), Japan (JSTP) and North America (STP) to develop an internationally accepted nomenclature for proliferative and nonproliferative lesions in laboratory animals. The purpose of this publication is to provide a standardized nomenclature for classifying microscopic lesions observed in the skeletal tissues and teeth of laboratory rats and mice, with color photomicrographs illustrating examples of many common lesions. The standardized nomenclature presented in this document is also available on the internet (http://www.goreni.org/). Sources of material were databases from government, academic and industrial laboratories throughout the world.

7.
Arthritis Rheum ; 62(10): 3006-15, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20533541

RESUMEN

OBJECTIVE: To demonstrate that the novel highly selective matrix metalloproteinase 13 (MMP-13) inhibitor PF152 reduces joint lesions in adult dogs with osteoarthritis (OA) and decreases biomarkers of cartilage degradation. METHODS: The potency and selectivity of PF152 were evaluated in vitro using 16 MMPs, TACE, and ADAMTS-4 and ADAMTS-5, as well as ex vivo in human cartilage explants. In vivo effects were evaluated at 3 concentrations in mature beagles with partial medial meniscectomy. Gross and histologic changes in the femorotibial joints were evaluated using various measures of cartilage degeneration. Biomarkers of cartilage turnover were examined in serum, urine, or synovial fluid. Results were analyzed individually and in combination using multivariate analysis. RESULTS: The potent and selective MMP-13 inhibitor PF152 decreased human cartilage degradation ex vivo in a dose-dependent manner. PF152 treatment of dogs with OA reduced cartilage lesions and decreased biomarkers of type II collagen (type II collagen neoepitope) and aggrecan (peptides ending in ARGN or AGEG) degradation. The dose required for significant inhibition varied with the measure used, but multivariate analysis of 6 gross and histologic measures indicated that all doses differed significantly from vehicle but not from each other. Combined analysis of cartilage degradation markers showed similar results. CONCLUSION: This highly selective MMP-13 inhibitor exhibits chondroprotective effects in mature animals. Biomarkers of cartilage degradation, when evaluated in combination, parallel the joint structural changes induced by the MMP-13 inhibitor. These data support the potential therapeutic value of selective MMP-13 inhibitors and the use of a set of appropriate biomarkers to predict efficacy in OA clinical trials.


Asunto(s)
Cartílago Articular/fisiopatología , Metaloproteinasa 13 de la Matriz/fisiología , Inhibidores de la Metaloproteinasa de la Matriz , Osteoartritis de la Rodilla/fisiopatología , Pirimidinas/farmacología , Tetrazoles/farmacología , Animales , Artritis Experimental , Biomarcadores/orina , Cartílago Articular/patología , Colágeno Tipo II/orina , Perros , Femenino , Osteoartritis de la Rodilla/patología , Líquido Sinovial/química
8.
J Immunol ; 182(8): 5024-31, 2009 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-19342682

RESUMEN

Multiple inflammatory mediators in osteoarthritis (OA) cartilage, including S100/calgranulin ligands of receptor for advanced glycation end products (RAGE), promote chondrocyte hypertrophy, a differentiation state associated with matrix catabolism. In this study, we observed that RAGE knockout was not chondroprotective in instability-induced knee OA in 8-wk-old mice. Hence, we tested the hypothesis that expression of the alternative S100/calgranulin and patterning receptor CD36, identified here as a marker of growth plate chondrocyte hypertrophy, mediates chondrocyte inflammatory and differentiation responses that promote OA. In rat knee joint destabilization-induced OA, RAGE expression was initially sparse throughout cartilage but increased diffusely by 4 wk after surgery. In contrast, CD36 expression focally increased at sites of cartilage injury and colocalized with developing chondrocyte hypertrophy and aggrecan cleavage NITEGE neoepitope formation. However, CD36 transfection in normal human knee-immortalized chondrocytes (CH-8 cells) was associated with decreased capacity of S100A11 and TNF-alpha to induce chondrocyte hypertrophy and ADAMTS-4 and matrix metalloproteinase 13 expression. S100A11 lost the capacity to inhibit proteoglycans synthesis and gained the capacity to induce proteoglycan synthesis in CD36-transfected CH-8 cells. Moreover, S100A11 required the p38 MAPK pathway kinase MKK3 to induce NITEGE development in mouse articular cartilage explants. However, CH-8 cells transfected with CD36 demonstrated decreased S100A11-induced MKK3 and p38 phosphorylation. Therefore, RAGE and CD36 patterning receptor expression were linked with opposing effects on inflammatory, procatabolic responses to S100A11 and TNF-alpha in chondrocytes.


Asunto(s)
Antígenos CD36/inmunología , Condrocitos/inmunología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Línea Celular , Condrocitos/metabolismo , Modelos Animales de Enfermedad , Humanos , Hipertrofia/inmunología , Hipertrofia/metabolismo , Hipertrofia/patología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , MAP Quinasa Quinasa 3/metabolismo , Masculino , Ratones , Ratones Noqueados , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/inmunología , Osteoartritis de la Rodilla/metabolismo , Osteoartritis de la Rodilla/patología , Ratas , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/deficiencia , Receptores Inmunológicos/genética , Receptores Inmunológicos/metabolismo , Proteínas S100/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
9.
Eur J Med Chem ; 224: 113666, 2021 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-34245949

RESUMEN

Osteoarthritis (OA) is a chronic disorder that causes damage to the cartilage and surrounding tissues and is characterized by pain, stiffness, and loss of function. Current treatments for OA primarily involve providing only relief of symptoms but does not affect the overall trajectory of the disease. A major goal for treating OA has been to slow down or reverse disease progression. Matrix metalloproteinase-13 (MMP-13) is expressed by chondrocytes and synovial cells in human OA and is thought to play a critical role in cartilage destruction. Herein we report a new, allosteric MMP-13 inhibitor, AQU-019, that has been optimized for potency, metabolic stability, and oral bioavailability through a combination of structure activity relationship (SAR) and deuterium substitution as a potential disease modifying OA drug (DMOAD). The inhibitor was demonstrated to be chondroprotective when injected intraarticular (IA) in the monoiodoacetic acid (MIA) rat model of OA.


Asunto(s)
Metaloproteinasa 13 de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/uso terapéutico , Osteoartritis/tratamiento farmacológico , Amidas/química , Amidas/metabolismo , Amidas/uso terapéutico , Animales , Modelos Animales de Enfermedad , Semivida , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Articulaciones/patología , Metaloproteinasa 13 de la Matriz/metabolismo , Inhibidores de la Metaloproteinasa de la Matriz/química , Inhibidores de la Metaloproteinasa de la Matriz/farmacocinética , Osteoartritis/inducido químicamente , Osteoartritis/patología , Pirimidinas/química , Ratas , Relación Estructura-Actividad
10.
J Exp Orthop ; 7(1): 39, 2020 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-32476071

RESUMEN

PURPOSE: Intraarticular (IA) hyaluronic acid (HA) injection is used to reduce pain and improve mobility in knee osteoarthritis (OA). Little is known about histopathological changes underlying HA efficacy. This study investigated dose-related effects of 1% sodium hyaluronate (BioHA) on knee joint histopathology and pain responses in a medial meniscal tear (MMT) rat model of OA. METHODS: Following MMT surgery, rats were randomized into treatment groups: single IA injection of vehicle, BioHA, or an avian-derived hyaluronic acid (hylan G-F 20) on Day 7; or 3 weekly injections of vehicle or BioHA on Days 7, 14, and 21. On Day 35, joints were evaluated by microscopic histopathology for cartilage degeneration, collagen degeneration, synovitis, and cytokine expression (tumor necrosis factor α, transforming growth factor ß). RESULTS: Joint pathology for control animals was consistent with that expected for the MMT model. Rats treated with 3 injections of IA-BioHA had significantly reduced collagen degeneration (21%) relative to control animals. No significant change in collagen degeneration was observed for rats given a single injection of hylan G-F 20 or IA-BioHA compared to control animals. HA treatment did not affect cytokine expression. CONCLUSIONS: IA-BioHA viscosupplementation in a rat MMT model of OA showed preservation of joint cartilage and collagen. This effect was most pronounced on tibial surfaces having less severe injury, suggesting that treatment should be initiated early in the disease process. A comparison of responses to IA-BioHA or hylan G-F 20 in the MMT rat OA model suggest IA-BioHA may be more effective in preserving joint connective tissue.

11.
ACS Biomater Sci Eng ; 5(6): 3060-3067, 2019 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-31608307

RESUMEN

A poly(7-oxanorbornene-2-carboxylate) polymer containing pendent triethyleneglycol (TEG) chains of 2.8 MDa ("2.8M TEG") was synthesized and evaluated for long-term lubrication and wear reduction of ex vivo bovine cartilage as well as for synovitis in rats and dogs after intra-articular administration. Bovine cartilage surfaces were tested under torsional friction for 10,080 rotations while immersed in either saline, bovine synovial fluid (BSF), or 2.8M TEG. For each solution, coefficient of friction (µ), changes in surface roughness, and lost cartilage glycosaminoglycan were compared. To directly compare 2.8M TEG and BSF, additional samples were tested sequentially in BSF, BSF, 2.8M TEG, and then BSF. Finally, another set of samples were tested twice in saline to induce surface roughness and then tested in BSF, Synvisc, or 2.8M TEG to determine each treatment's effect on worn cartilage. Next, male Lewis rats were injected in one knee with 2.8M TEG or saline and evaluated for effects on gait, and female beagles were injected with either 2.8M TEG or saline in one knee, and their synovial tissues analyzed for inflammation by H&E staining. Treatment with 2.8M TEG lowers µ, lessens surface roughness, and minimizes glycosaminoglycan loss compared to saline. The 2.8M TEG also reduces µ compared to BSF in pairwise testing and on worn cartilage surfaces. Injection of 2.8M TEG in rat or beagle knees gives comparable effects to treatment with saline, and does not cause significant synovitis.

12.
ACS Appl Bio Mater ; 2(7): 2822-2832, 2019 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-35030816

RESUMEN

The most common treatment for osteoarthritis is daily oral administration of a nonsteroidal anti-inflammatory drug such as diclofenac. This daily dosage regime is often associated with severe side effects. In this study, we explored the potential of utilizing a high molecular weight cross-linked polyurethane polymer covalently linked to diclofenac (C-DCF-PU) for intra-articular administration. We aim to exploit the advantages of local drug delivery by developing an implant with improved efficacy and reduced side effects. The polymer was synthesized from a diclofenac-functionalized monomer unit in a simple one-pot reaction, followed by cross-linking. In vitro drug release studies showed zero-order drug release for 4 days, followed by a gradual decline in drug release rate until diclofenac was depleted after 15 days. The cross-linked polymer was triturated to yield an injectable microgel formulation for administration. Whole animal fluorescence imaging of the rhodamine-labeled C-DCF-RH-PU showed good retention of the polymer in the knee joints of healthy rats, with approximately 30% of the injected dose still present 2 weeks post intra-articular administration. In a reactivation arthritis animal model, the C-DCF-RH-PU formulation reduced pain and significantly reduced inflammation after a short lag phase, showing that this drug delivery system warrants further development for long-term treatment of osteoarthritis with the benefit of reduced side effects.

13.
Biomaterials ; 182: 13-20, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30099277

RESUMEN

Intra-articular injection of hyaluronic acid (HA) is used to treat osteoarthritis (OA) as a viscosupplement, yet it only provides short-term benefit because HA is cleaved by hyaluronidase and cleared out of the joint after several days. Therefore, we developed a new polymer biolubricant based on poly-oxanorbornane carboxylate to enhance joint lubrication for a prolonged time. Rheological and biotribological studies of the biolubricant reveal viscoelastic properties and coefficient of friction equivalent and superior to that of healthy synovial fluid, respectively. Furthermore, in an ex vivo bovine cartilage plug model, the biolubricant exhibits superior long-term reduction of friction and wear prevention compared to saline and healthy synovial fluid. ISO 10993 biocompatibility tests demonstrate that the biolubricant polymer is non-toxic. In an in vivo rat medial meniscal tear OA model, where the performance of the leading HA viscosupplement (Synvisc-one®) is comparable to the saline control, treatment with the biolubricant affords significant chondroprotection compared to the saline control.


Asunto(s)
Condrocitos/efectos de los fármacos , Furanos/administración & dosificación , Articulación de la Rodilla/efectos de los fármacos , Menisco/efectos de los fármacos , Polienos/administración & dosificación , Líquido Sinovial/efectos de los fármacos , Viscosuplementos/administración & dosificación , Animales , Fenómenos Biomecánicos , Línea Celular , Condrocitos/citología , Condrocitos/metabolismo , Furanos/farmacología , Furanos/uso terapéutico , Humanos , Inyecciones Intraarticulares , Traumatismos de la Rodilla/tratamiento farmacológico , Traumatismos de la Rodilla/metabolismo , Articulación de la Rodilla/metabolismo , Masculino , Menisco/lesiones , Menisco/metabolismo , Ratones , Células 3T3 NIH , Osteoartritis/tratamiento farmacológico , Polienos/farmacología , Polienos/uso terapéutico , Ratas Endogámicas Lew , Líquido Sinovial/metabolismo , Viscosuplementos/farmacología , Viscosuplementos/uso terapéutico
14.
J Orthop Res ; 35(10): 2260-2268, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28139016

RESUMEN

Current osteoarthritis (OA) research searches for treatments that modify the course of disease progression. Autologous Protein Solution (APS) is derived from whole blood and is a unique autologous therapy that contains high concentrations of white blood cells, platelets, and concentrated plasma, providing cytokines that can target the underlying mechanisms of disease progression. The APS Kit is currently under investigation for clinical use in the USA (NCT02262364). The aim of this study was to determine if APS has disease-modifying properties in the well-characterized rat meniscal tear-induced model of OA. Thirty male athymic rats underwent surgery to induce OA by a complete tear of the medial meniscus of the right knee. Seven days later, rats were administered 50 µl intra-articular (IA) APS from a human donor or phosphate buffered saline (PBS) control. Rats were euthanized 3 weeks following treatment and knee joints were processed for histological analysis. Collagen and cartilage degeneration were decreased by APS treatment, resulting in a significantly improved total joint score in APS-treated rats compared to those treated with the PBS control. No significant variations in gait analysis, weight gain, osteophyte score, or synovitis score were observed between APS- and PBS-treated animals. There were no adverse effects of APS reported in the study. Treatment with a single IA injection of APS reduced the cartilage degeneration that characterizes the progression of OA. Further studies are necessary to determine if APS can modify OA disease progression in humans. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:2260-2268, 2017.


Asunto(s)
Antiinflamatorios/uso terapéutico , Transfusión de Componentes Sanguíneos , Proteínas Sanguíneas/uso terapéutico , Osteoartritis de la Rodilla/prevención & control , Lesiones de Menisco Tibial/complicaciones , Animales , Antiinflamatorios/química , Humanos , Inyecciones Intraarticulares , Masculino , Osteoartritis de la Rodilla/etiología , Ratas Desnudas
15.
Pharmacol Res Perspect ; 5(3): e00316, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28603634

RESUMEN

Prostaglandin (PG) E2 is the key driver of inflammation associated with arthritic conditions. Inhibitors of PGE 2 production (NSAIDs and Coxibs) are used to treat these conditions, but carry significant side effect risks due to the inhibition of all prostanoids that play important physiological function. The activities of PGE 2 are transduced through various receptor sub-types. Prostaglandin E2 type 4 receptor (EP4) is associated with the development of inflammation and autoimmunity. We therefore are interested in identifying novel EP4 antagonists to treat the signs and symptoms of arthritis without the potential side effects of PGE 2 modulators such as NSAIDs and Coxibs. Novel EP4 antagonists representing distinct chemical scaffolds were identified using a variety of in vitro functional assays and were shown to be selective and potent. The compounds were shown to be efficacious in animal models of analgesia, inflammation, and arthritis.

16.
Arthritis Res Ther ; 18: 24, 2016 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-26794830

RESUMEN

BACKGROUND: As an initial step in the development of a local therapeutic to treat osteoarthritis (OA), a number of agents were tested for their ability to block activation of inflammation through nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB), subchondral bone changes through receptor activator of nuclear factor κB ligand (RANKL)-mediated osteoclastogenesis, and proteolytic degradation through matrix metalloproteinase (MMP)-13 activity. Candidates with low toxicity and predicted efficacy were further examined using either of two widely accepted models of OA joint degeneration in the rat: the monoiodoacetic acid (MIA) model or the medial meniscal tear/medial collateral ligament tear (MMT/MCLT) model. METHODS: Potential therapeutics were assessed for their effects on the activation of nuclear factor (NF)-κB, RANKL-mediated osteoclastogenesis, and MMP-13 activity in vitro using previously established assays. Toxicity was measured using HeLa cells, a synovial cell line, or primary human chondrocytes. Drugs predicted to perform well in vivo were tested either systemically or via intraarticular injection in the MIA or the MMT/MCLT model of OA. Pain behavior was measured by mechanical hyperalgesia using the digital Randall-Selitto test (dRS) or by incapacitance with weight bearing (WB). Joint degeneration was evaluated using micro computed tomography and a comprehensive semiquantitative scoring of cartilage, subchondral bone, and synovial histopathology. RESULTS: Several agents were effective both in vitro and in vivo. With regard to pain behavior, systemically delivered clonidine was superior in treating MIA-induced changes in WB or dRS, while systemic clonidine, curcumin, tacrolimus, and fluocinolone were all somewhat effective in modifying MMT/MCLT-induced changes in WB. Systemic tacrolimus was the most effective in slowing disease progression as measured by histopathology in the MMT/MCLT model. CONCLUSIONS: All of the agents that demonstrated highest benefit in vivo, excepting clonidine, were found to inhibit MMP-13, NF-κB, and bone matrix remodeling in vitro. The MIA and MMT/MCLT models of OA, previously shown to possess inflammatory characteristics and to display associated pain behavior, were affected to different degrees by the same drugs. Although no therapeutic was remarkable across all measures, the several which showed the most promise in either model merit continued study with alternative dosing and therapeutic strategies.


Asunto(s)
Antirreumáticos/uso terapéutico , Cartílago Articular/efectos de los fármacos , Progresión de la Enfermedad , Osteoartritis/tratamiento farmacológico , Dolor/tratamiento farmacológico , Ensayos Clínicos Controlados Aleatorios como Asunto , Animales , Antirreumáticos/farmacología , Cartílago Articular/metabolismo , Cartílago Articular/patología , Línea Celular , Relación Dosis-Respuesta a Droga , Células HeLa , Humanos , Mediadores de Inflamación/antagonistas & inhibidores , Mediadores de Inflamación/metabolismo , Masculino , Ratones , Osteoartritis/metabolismo , Osteoartritis/patología , Dolor/metabolismo , Dolor/patología , Ratas , Ratas Endogámicas Lew , Ratas Sprague-Dawley , Resultado del Tratamiento
17.
Am J Vet Res ; 66(1): 156-63, 2005 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-15691052

RESUMEN

OBJECTIVE: To determine whether decreases in peak vertical force of the hind limb after transection of the cranial cruciate ligament (CrCL) would be indicative of medial meniscal damage in dogs. ANIMALS: 39 purpose-bred adult male Walker Hounds. PROCEDURE: The right CrCL was transected arthroscopically. Force plate measurements of the right hind limb were made prior to and 2, 4, 10, and 18 weeks after transection of the CrCL. Only dogs with > or =10% decreases in peak vertical force after week 2 were considered to have potential meniscal damage. Dogs that did not have > or =10% decreases in peak vertical force at any time point after week 2 were assigned to group 1. Group 2 dogs had > or =10% decreases in peak vertical force from weeks 2 to 4 only. Group 3 and 4 dogs had > or =10% decreases in peak vertical force from weeks 4 to 10 only or from weeks 10 to 18 only, respectively. Damage to menisci and articular cartilage was graded at week 18, and grades for groups 2 to 4 were compared with those of group 1. RESULTS: The percentage change in peak vertical force and impulse area was significantly different in groups 2 (n = 4), 3 (4), and 4 (4) at the end of each measurement period (weeks 4, 10, and 18, respectively) than in group 1 (27). The meniscal grade for groups 2 to 4 was significantly higher than for group 1. A > or =10% decrease in peak vertical force had sensitivity of 52% and accuracy of 72% for identifying dogs with moderate to severe medial meniscal damage. CONCLUSIONS AND CLINICAL RELEVANCE: In dogs with transected or ruptured CrCLs, force plate analysis can detect acute exacerbation of lameness, which may be the result of secondary meniscal damage, and provide an objective noninvasive technique that delineates the temporal pattern of medial meniscal injury.


Asunto(s)
Perros/lesiones , Ligamentos Articulares/lesiones , Lesiones de Menisco Tibial , Animales , Enfermedades de los Perros , Miembro Posterior , Cojera Animal/fisiopatología , Masculino , Rotura/veterinaria , Estrés Mecánico , Soporte de Peso
18.
Exp Hematol ; 32(5): 441-9, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15145212

RESUMEN

OBJECTIVE: The aim of this study was to determine whether fusion proteins comprising human granulocyte colony-stimulating factor (G-CSF) joined to human immunoglobulin G1 and G4 (IgG1 and IgG4) Fc and C(H) domains are biologically active and have improved pharmacokinetic and hematopoietic properties in vivo. MATERIAL AND METHODS: Chimeric genes encoding human G-CSF fused to the N-termini of the Fc and C(H) domains of human IgG1 and IgG4 were constructed and used to transfect monkey COS cells. The fusion proteins were purified from the conditioned media by protein A affinity chromatography. Bioactivities of the proteins were measured in a G-CSF-dependent in vitro bioassay. Pharmacokinetic and granulopoietic properties of the G-CSF/IgG1-Fc fusion protein were measured in normal rats. RESULTS: The G-CSF/IgG-Fc and G-CSF/IgG-C(H) fusion proteins were secreted from transfected COS cells primarily as disulfide-linked homodimers. On a molar basis, the purified G-CSF/IgG-Fc fusion proteins were as active as G-CSF in in vitro bioassays, whereas bioactivities of the purified G-CSF/IgG-C(H) fusion proteins were decreased 3- to 4-fold. The G-CSF/IgG1-Fc fusion protein displayed a slower plasma clearance rate and stimulated greater and longer lasting increases in circulating neutrophils and white blood cells than G-CSF following intravenous and subcutaneous administration to rats. CONCLUSION: Fusion of G-CSF to human IgG domains results in homodimeric fusion proteins possessing high in vitro bioactivities, long circulating half-lives, and enhanced hematopoietic properties in vivo.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/farmacología , Hematopoyesis/efectos de los fármacos , Inmunoglobulina G/farmacología , Proteínas Recombinantes de Fusión/farmacología , Animales , Recuento de Células Sanguíneas , Examen de la Médula Ósea , Células COS , Factor Estimulante de Colonias de Granulocitos/sangre , Factor Estimulante de Colonias de Granulocitos/genética , Semivida , Humanos , Inmunoglobulina G/genética , Leucocitos/citología , Leucocitos/efectos de los fármacos , Masculino , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Farmacocinética , Ratas , Ratas Sprague-Dawley , Proteínas Recombinantes de Fusión/sangre , Proteínas Recombinantes de Fusión/genética , Transfección
19.
Inflammation ; 38(1): 40-60, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25189465

RESUMEN

Current therapeutic treatment options for osteoarthritis entail significant safety concerns. A novel ropivacaine crystalline microsuspension for bolus intra-articular (IA) delivery was thus developed and studied in a peptidoglycan polysaccharide (PGPS)-induced ankle swelling rat model. Compared with celecoxib controls, both oral and IA, ropivacaine IA treatment resulted in a significant reduction of pain upon successive PGPS reactivation, as demonstrated in two different pain models, gait analysis and incapacitance testing. The reduction in pain was attended by a significant reduction in histological inflammation, which in turn was accompanied by significant reductions in the cytokines IL-18 and IL-1ß. This may have been due to inhibition of substance P, which was also significantly reduced. Pharmacokinetic analysis indicated that the analgesic effects outlasted measurable ropivacaine levels in either blood or tissue. The results are discussed in the context of pharmacologic mechanisms both of local anesthetics as well as inflammatory arthritis.


Asunto(s)
Amidas/administración & dosificación , Artritis Experimental/tratamiento farmacológico , Celecoxib/administración & dosificación , Citocinas/antagonistas & inhibidores , Dolor/tratamiento farmacológico , Sustancia P/antagonistas & inhibidores , Administración Oral , Animales , Artritis Experimental/metabolismo , Artritis Experimental/patología , Citocinas/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Inflamación/patología , Inyecciones Intraarticulares , Masculino , Microesferas , Dolor/metabolismo , Dolor/patología , Ratas , Ropivacaína , Sustancia P/metabolismo
20.
Arthritis Res Ther ; 17: 5, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25592743

RESUMEN

INTRODUCTION: Cathepsin K (catK) expression is increased in cartilage, bone and synovium during osteoarthritis (OA). To study the role of catK expression and elevated cathepsin activity in the synovium on cartilage destruction in established OA, we overexpressed cystatin C (cysC), a natural cysteine protease inhibitor, in the synovium of rabbit OA joints. METHODS: The ability of cysC to inhibit activity of cathepsins in rabbit OA synovium lysates was tested in vitro using protease activity assay. In vivo, the tissue localization of recombinant adeno-associated virus (rAAV) with LacZ gene after intra-articular injection was determined by ß-galactosidase staining of rabbit joints 4 weeks later. To inhibit cathepsin activity in the synovium, a rAAV2-encoding cysC was delivered intra-articularly into rabbit joints 4 weeks after OA was induced by anterior cruciate ligament transection (ACLT). Seven weeks postinjection, endogenous catK and cysC levels as well as the vector-derived cysC expression in the synovium of normal and OA joints were examined by RNA quantification. Synovial cathepsin activity and catK, catB and catL protein levels were determined by activity and Western blot analyses, respectively. Synovitis and cartilage degradation were evaluated by histopathological scoring. RESULTS: In vitro, the ability of cysC to efficiently inhibit activity of purified catK and OA-induced cathepsins in rabbit synovial lysates was demonstrated. In vivo, the intra-articular delivery of rAAV2/LacZ showed transduction of mostly synovium. Induction of OA in rabbit joints resulted in fourfold increase in catK mRNA compared to sham controls while no change was detected in endogenous cysC mRNA levels in the synovium. Protein levels for catK, catB and catL were also increased in the synovium with a concomitant fourfold increase in cathepsin activity. Joints treated with rAAV2/cysC showed both detection of vector genomes and vector-derived cysC transcripts in the synovium. Production of functional cysC by the vector was demonstrated by complete block of cathepsin activity in the synovium. However, this did not decrease synovitis, bone sclerosis or progression of cartilage degradation. CONCLUSIONS: Increased production of natural cathepsin inhibitor, cysC, in OA synovium does not alleviate synovitis or cartilage pathology during a preexisting OA.


Asunto(s)
Cartílago Articular/metabolismo , Cistatina C/biosíntesis , Osteoartritis/metabolismo , Membrana Sinovial/metabolismo , Sinovitis/metabolismo , Animales , Cartílago Articular/patología , Regulación de la Expresión Génica , Humanos , Masculino , Osteoartritis/patología , Conejos , Membrana Sinovial/patología , Sinovitis/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA