Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Arthritis Rheumatol ; 75(6): 923-936, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36625730

RESUMEN

OBJECTIVE: The synovial lymphatic system (SLS) removes catabolic factors from the joint. Vascular endothelial growth factor C (VEGF-C) and its receptor, VEGFR-3, are crucial for lymphangiogenesis. However, their involvement in age-related osteoarthritis (OA) is unknown. This study was undertaken to determine whether the SLS and the VEGF-C/VEGFR-3 pathway contribute to the development and progression of age-related OA, using a murine model of naturally occurring joint disease. METHODS: SLS function was assessed in the knees of young (3-month-old) and aged (19-24-month-old) male and female C57BL/6J mice via a newly established in vivo IVIS-dextran imaging approach, which, in addition to histology, was used to assess the effects of VEGF-C treatment on SLS function and OA pathology in aged mice. RNA-sequencing of synovial tissue was performed to explore molecular mechanisms of the disease in the mouse knee joints. RESULTS: Results showed that aged mice had impaired SLS function, including decreases in joint clearance (mean T1/2 of signal intensity clearance, 2.8 hours in aged mice versus 0.5 hours in young mice; P < 0.0001), synovial influx (mean ± SD 1.7 ± 0.8% in aged mice versus 4.1 ± 1.9% in young mice; P = 0.0004), and lymph node draining capacity (mean ± SD epifluorescence total radiant intensity ([photons/second]/[µW/cm2 ]) 1.4 ± 0.8 in aged mice versus 3.7 ± 1.2 in young mice; P < 0.0001). RNA-sequencing of the synovial tissue showed that Vegf-c and Vegfr3 signaling genes were decreased in the synovium of aged mice. VEGF-C treatment resulted in improvements in SLS function in aged mice, including increased percentage of signal intensity joint clearance (mean ± SD 63 ± 9% in VEGF-C-treated aged mice versus 52 ± 15% in vehicle-treated aged mice; P = 0.012), increased total articular cartilage cross-sectional area (mean ± SD 0.38 ± 0.07 mm2 in VEGF-C-treated aged mice versus 0.26 ± 0.07 mm2 in vehicle-treated aged mice; P < 0.0001), and decreased percentage of matrix metallopeptidase 13-positive staining area within total synovial area in 22-month-old VEGF-C-treated mice versus 22-month-old vehicle-treated mice (mean ± SD decrease 7 ± 2% versus 4 ± 1%; P = 0.0004). CONCLUSION: SLS function is reduced in the knee joints of aged mice due to decreased VEGF-C/VEGFR-3 signaling. VEGF-C treatment attenuates OA joint damage and improves synovial lymphatic drainage in aged mice. The SLS and VEGF-C/VEGFR-3 signaling represent novel physiopathologic mechanisms that could potentially be used as therapeutic targets for age-related OA.


Asunto(s)
Osteoartritis , Factor C de Crecimiento Endotelial Vascular , Ratones , Masculino , Femenino , Animales , Receptor 3 de Factores de Crecimiento Endotelial Vascular , Ratones Endogámicos C57BL , Osteoartritis/metabolismo , Membrana Sinovial/metabolismo , ARN/metabolismo
2.
Sci Rep ; 12(1): 12751, 2022 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-35882971

RESUMEN

While rheumatoid arthritis patients and tumor necrosis factor transgenic (TNF-Tg) mice with inflammatory-erosive arthritis display lymphatic drainage deficits, the mechanisms responsible remain unknown. As ultrastructural studies of joint-draining popliteal lymphatic vessels (PLVs) in TNF-Tg mice revealed evidence of lymphatic muscle cell (LMC) damage, we aimed to evaluate PLV-LMC coverage in TNF-Tg mice. We tested the hypothesis that alpha smooth muscle actin (αSMA)+ PLV-LMC coverage decreases with severe inflammatory-erosive arthritis, and is recovered by anti-TNF therapy facilitated by increased PLV-LMC turnover during amelioration of joint disease. TNF-Tg mice with established disease received anti-TNF monoclonal antibody (mAb) or placebo IgG isotype control mAb therapy (n = 5) for 6-weeks, while wild-type (WT) littermates (n = 8) received vehicle (PBS). Bromodeoxyuridine (BrdU) was also administered daily during the treatment period to monitor PLV-LMC turnover. Effective anti-TNF therapy was confirmed by longitudinal assessment of popliteal lymph node (PLN) volume via ultrasound, PLV contraction frequency via near-infrared imaging of indocyanine green, and ankle bone volumes via micro-computed tomography (micro-CT). Terminal knee micro-CT, and ankle and knee histology were also performed. PLVs were immunostained for αSMA and BrdU to evaluate PLV-LMC coverage and turnover, respectively, via whole-mount fluorescent microscopy. Anti-TNF therapy reduced PLN volume, increased talus and patella bone volumes, and reduced tarsal and knee synovial areas compared to placebo treated TNF-Tg mice (p < 0.05), as expected. Anti-TNF therapy also increased PLV contraction frequency at 3-weeks (from 0.81 ± 1.0 to 3.2 ± 2.0 contractions per minute, p < 0.05). However, both anti-TNF and placebo treated TNF-Tg mice exhibited significantly reduced αSMA+ PLV-LMC coverage compared to WT (p < 0.05). There was no correlation of αSMA+ PLV-LMC coverage restoration with amelioration of inflammatory-erosive arthritis. Similarly, there was no difference in PLV-LMC turnover measured by BrdU labeling between WT, TNF-Tg placebo, and TNF-Tg anti-TNF groups with an average of < 1% BrdU+ PLV-LMCs incorporated per week. Taken together these results demonstrate that PLV-LMC turnover in adult mice is limited, and that recovery of PLV function during amelioration of inflammatory-erosive arthritis occurs without restoration of αSMA+ LMC coverage. Future studies are warranted to investigate the direct and indirect effects of chronic TNF exposure, and the role of proximal inflammatory cells on PLV contractility.


Asunto(s)
Artritis Reumatoide , Vasos Linfáticos , Animales , Anticuerpos Monoclonales/farmacología , Artritis Reumatoide/patología , Bromodesoxiuridina , Vasos Linfáticos/patología , Ratones , Ratones Transgénicos , Células Musculares , Inhibidores del Factor de Necrosis Tumoral/farmacología , Factor de Necrosis Tumoral alfa/uso terapéutico , Microtomografía por Rayos X
3.
Pharmaceutics ; 11(2)2019 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-30813284

RESUMEN

Osteomyelitis is a chronic bone infection that is often treated with adjuvant antibiotic-impregnated poly(methyl methacrylate) (PMMA) cement spacers in multi-staged revisions. However, failure rates remain substantial due to recurrence of infection, which is attributed to the poor performance of the PMMA cement as a drug release device. Hence, the objective of this study was to design and evaluate a bioresorbable calcium phosphate scaffold (CaPS) for sustained antimicrobial drug release and investigate its efficacy in a murine model of femoral implant-associated osteomyelitis. Incorporating rifampin and sitafloxacin, which are effective against bacterial phenotypes responsible for bacterial persistence, into 3D-printed CaPS coated with poly(lactic co-glycolic) acid, achieved controlled release for up to two weeks. Implantation into the murine infection model resulted in decreased bacterial colonization rates at 3- and 10-weeks post-revision for the 3D printed CaPS in comparison to gentamicin-laden PMMA. Furthermore, a significant increase in bone formation was observed for 3D printed CaPS incorporated with rifampin at 3 and 10 weeks. The results of this study demonstrate that osteoconductive 3D printed CaPS incorporated with antimicrobials demonstrate more efficacious bacterial colonization outcomes and bone growth in a single-stage revision in comparison to gentamicin-laden PMMA requiring a two-stage revision.

4.
Arthritis Rheumatol ; 71(2): 244-257, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30144298

RESUMEN

OBJECTIVE: To investigate the roles of the synovial lymphatic system in the severity and progression of joint tissue damage and functional responses of synovial lymphatic endothelial cells (LECs) to macrophage subsets, and to evaluate the therapeutic potential of the proteasome inhibitor bortezomib (BTZ) in a mouse model of experimental posttraumatic osteoarthritis (OA). METHODS: C57BL/6J wild-type mice received a meniscal ligamentous injury to induce posttraumatic knee OA. Lymphangiogenesis was blocked by a vascular endothelial growth factor receptor 3 (VEGFR-3) neutralizing antibody. Synovial lymphatic drainage was examined by near-infrared imaging. Joint damage was assessed by histology. RNA-sequencing and pathway analyses were applied to synovial LECs. Macrophage subsets in the mouse synovium were identified by flow cytometry and immunofluorescence staining. M1 and M2 macrophages were induced from mouse bone marrow cells, and their effects on LECs were examined in cocultures in the presence or absence of BTZ. The effects of BTZ on joint damage, LEC inflammation, and synovial lymphatic drainage were examined. RESULTS: Injection of a VEGFR-3 neutralizing antibody into the joints of mice with posttraumatic knee OA reduced synovial lymphatic drainage and accelerated joint tissue damage. Synovial LECs from the mouse OA joints had dysregulated inflammatory pathways and expressed high levels of inflammatory genes. The number of M1 macrophages was increased in the knee joints of mice with posttraumatic OA, thereby promoting the expression of inflammatory genes by LECs; this effect was blocked by BTZ. Treatment with BTZ decreased cartilage loss, reduced the expression of inflammatory genes by LECs, and improved lymphatic drainage in the knee joints of mice with posttraumatic OA. CONCLUSION: Experimental posttraumatic knee OA is associated with decreased synovial lymphatic drainage, increased numbers of M1 macrophages, and enhanced inflammatory gene expression by LECs, all of which was improved by treatment with BTZ. Intraarticular administration of BTZ may represent a new therapy for the restoration of synovial lymphatic function in subjects with posttraumatic knee OA.


Asunto(s)
Bortezomib/farmacología , Células Endoteliales/efectos de los fármacos , Vasos Linfáticos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Osteoartritis de la Rodilla/inmunología , Inhibidores de Proteasoma/farmacología , Membrana Sinovial/efectos de los fármacos , Animales , Anticuerpos Neutralizantes/farmacología , Bortezomib/uso terapéutico , Técnicas de Cocultivo , Progresión de la Enfermedad , Inflamación , Traumatismos de la Rodilla/complicaciones , Linfangiogénesis/efectos de los fármacos , Vasos Linfáticos/inmunología , Macrófagos/inmunología , Ratones , Osteoartritis de la Rodilla/tratamiento farmacológico , Osteoartritis de la Rodilla/etiología , Inhibidores de Proteasoma/uso terapéutico , Espectroscopía Infrarroja Corta , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptor 3 de Factores de Crecimiento Endotelial Vascular/inmunología
5.
mSphere ; 3(5)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30381352

RESUMEN

Drug repurposing offers an expedited and economical route to develop new clinical therapeutics in comparison to traditional drug development. Growth-based high-throughput screening is concomitant with drug repurposing and enables rapid identification of new therapeutic uses for investigated drugs; however, this traditional method is not compatible with microorganisms with abnormal growth patterns such as Staphylococcus aureus small-colony variants (SCV). SCV subpopulations are auxotrophic for key compounds in biosynthetic pathways, which result in low growth rate. SCV formation is also associated with reduced antibiotic susceptibility, and the SCV's ability to revert to the normal cell growth state is thought to contribute to recurrence of S. aureus infections. Thus, there is a critical need to identify antimicrobial agents that are potent against SCV in order to effectively treat chronic infections. Accordingly, here we describe adapting an adenylate kinase (AK)-based cell death reporter assay to identify members of a Food and Drug Administration (FDA)-approved drug library that display bactericidal activity against S. aureus SCV. Four library members, daunorubicin, ketoconazole, rifapentine, and sitafloxacin, exhibited potent SCV bactericidal activity against a stable S. aureus SCV. Further investigation showed that sitafloxacin was potent against methicillin-susceptible and -resistant S. aureus, as well as S. aureus within an established biofilm. Taken together, these results demonstrate the ability to use the AK assay to screen small-molecule libraries for SCV bactericidal agents and highlight the therapeutic potential of sitafloxacin to be repurposed to treat chronic S. aureus infections associated with SCV and/or biofilm growth states.IMPORTANCE Conventional antibiotics fail to successfully treat chronic osteomyelitis, endocarditis, and device-related and airway infections. These recurring infections are associated with the emergence of SCV, which are recalcitrant to conventional antibiotics. Studies have investigated antibiotic therapies to treat SCV-related infections but have had little success, emphasizing the need to identify novel antimicrobial drugs. However, drug discovery is a costly and time-consuming process. An alternative strategy is drug repurposing, which could identify FDA-approved and well-characterized drugs that could have off-label utility in treating SCV. In this study, we adapted a high-throughput AK-based assay to identify 4 FDA-approved drugs, daunorubicin, ketoconazole, rifapentine, and sitafloxacin, which display antimicrobial activity against S. aureus SCV, suggesting an avenue for drug repurposing in order to effectively treat SCV-related infections. Additionally, this screening paradigm can easily be adapted for other drug/chemical libraries to identify compounds bactericidal against SCV.


Asunto(s)
Antibacterianos/aislamiento & purificación , Evaluación Preclínica de Medicamentos/métodos , Reposicionamiento de Medicamentos/métodos , Infecciones Estafilocócicas/microbiología , Staphylococcus aureus/efectos de los fármacos , Adenilato Quinasa/análisis , Genes Reporteros , Viabilidad Microbiana/efectos de los fármacos , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/crecimiento & desarrollo
6.
Nat Rev Rheumatol ; 14(2): 94-106, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29323343

RESUMEN

Although clinical outcomes for patients with rheumatoid arthritis (RA) have greatly improved with the use of biologic and conventional DMARDs, approximately 40% of patients do not achieve primary clinical outcomes in randomized trials, and only a small proportion achieve lasting remission. Over the past decade, studies in murine models point to the critical role of the lymphatic system in the pathogenesis and therapy of inflammatory-erosive arthritis, presumably by the removal of catabolic factors, cytokines and inflammatory cells from the inflamed synovium. Murine studies demonstrate that lymphatic drainage increases at the onset of inflammatory-erosive arthritis but, as inflammation progresses to a more chronic phase, lymphatic clearance declines and both structural and cellular changes are observed in the draining lymph node. Specifically, chronic damage to the lymphatic vessel from persistent inflammation results in loss of lymphatic vessel contraction followed by lymph node collapse, reduced lymphatic drainage, and ultimately severe synovitis and joint erosion. Notably, clinical pilot studies in patients with RA report lymph node changes following treatment, and thus draining lymphatic vessels and nodes could represent a potential biomarker of arthritis activity and response to therapy. Most importantly, targeting lymphatics represents an innovative strategy for therapeutic intervention for RA.


Asunto(s)
Artritis Reumatoide/terapia , Sistema Linfático/patología , Factor C de Crecimiento Endotelial Vascular/genética , Animales , Artritis Reumatoide/genética , Artritis Reumatoide/patología , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Terapia Genética , Vectores Genéticos/administración & dosificación , Humanos , Sistema Linfático/efectos de los fármacos , Ratones
7.
PLoS One ; 12(10): e0185446, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29020057

RESUMEN

Small animal studies have demonstrated significant high-dose recombinant parathyroid hormone1-34 (rPTH1-34) effects on intercalary allograft healing. Towards a human adjuvant therapy to decrease non-unions, we evaluated rPTH1-34 safety and efficacy in a clinically relevant canine femoral allograft model. Adult female mongrel hounds (n = 20) received a 5cm mid-diaphyseal osteotomy reconstructed with a plated allograft, and were randomized to: 1) Placebo (n = 5; daily saline), 2) Continuous rPTH1-34 (n = 7; 5 µg/kg/day s.c. from day 1-55 post-op), or 3) Delayed rPTH1-34 (n = 8; 5 µg/kg/day s.c. from day 14-28 post-op). Safety was assessed by physical behavior and blood calcium monitoring. Cone beam CT (CB-CT) was performed on days 14, 28 and 56 post-op to assess 2D cortical healing, 3D bone volume, and Union Ratio. Biomechanical testing and dynamic histomorphometry were also performed. The high drug dose was poorly tolerated, as most dogs receiving rPTH1-34 had to be given intravenous saline, and one dog died from hypercalcemia. Continuous rPTH1-34 significantly increased 2D healing and callus volumes at 4-weeks versus Placebo, and sustained the significant increase in cortical union at 8-week (p<0.05). These rPTH1-34 effects were confirmed by histomorphometry, revealing significant increases in mineral apposition rates (MAR) on host bone and graft-host junctions (p<0.05). Delayed rPTH1-34 significantly increased callus volume and MAR at 8 weeks (p<0.05). Although no biomechanical differences were observed, as expected for early healing, the results demonstrated that 2D RUST scoring significantly correlated with torsional biomechanics (p<0.01). In conclusion, 8-weeks of intermittent high-dose rPTH1-34 treatment significantly increases callus formation and accelerates bony union of intercalary massive allografts in a clinically relevant canine model, but with serious side-effects from hypercalcemia.


Asunto(s)
Callo Óseo/diagnóstico por imagen , Callo Óseo/patología , Fémur/trasplante , Teriparatido/administración & dosificación , Teriparatido/uso terapéutico , Cicatrización de Heridas , Aloinjertos/efectos de los fármacos , Animales , Fenómenos Biomecánicos/efectos de los fármacos , Callo Óseo/efectos de los fármacos , Tomografía Computarizada de Haz Cónico , Modelos Animales de Enfermedad , Perros , Relación Dosis-Respuesta a Droga , Fémur/fisiopatología , Fémur/cirugía , Fluorescencia , Minerales/metabolismo , Cuidados Posoperatorios , Teriparatido/farmacología , Cicatrización de Heridas/efectos de los fármacos
8.
Biomaterials ; 35(13): 4026-34, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24529628

RESUMEN

Low temperature 3D printing of calcium phosphate scaffolds holds great promise for fabricating synthetic bone graft substitutes with enhanced performance over traditional techniques. Many design parameters, such as the binder solution properties, have yet to be optimized to ensure maximal biocompatibility and osteoconductivity with sufficient mechanical properties. This study tailored the phosphoric acid-based binder solution concentration to 8.75 wt% to maximize cytocompatibility and mechanical strength, with a supplementation of Tween 80 to improve printing. To further enhance the formulation, collagen was dissolved into the binder solution to fabricate collagen-calcium phosphate composites. Reducing the viscosity and surface tension through a physiologic heat treatment and Tween 80, respectively, enabled reliable thermal inkjet printing of the collagen solutions. Supplementing the binder solution with 1-2 wt% collagen significantly improved maximum flexural strength and cell viability. To assess the bone healing performance, we implanted 3D printed scaffolds into a critically sized murine femoral defect for 9 weeks. The implants were confirmed to be osteoconductive, with new bone growth incorporating the degrading scaffold materials. In conclusion, this study demonstrates optimization of material parameters for 3D printed calcium phosphate scaffolds and enhancement of material properties by volumetric collagen incorporation via inkjet printing.


Asunto(s)
Fosfatos de Calcio/química , Colágeno/química , Impresión Tridimensional , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Animales , Regeneración Ósea , Supervivencia Celular/efectos de los fármacos , Femenino , Ratones , Ratones Endogámicos BALB C , Andamios del Tejido/efectos adversos , Viscosidad
9.
Arthritis Rheum ; 65(1): 130-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23002006

RESUMEN

OBJECTIVE: B cell depletion therapy ameliorates rheumatoid arthritis by mechanisms that are incompletely understood. Arthritis flare in tumor necrosis factor (TNF)-transgenic mice is associated with efferent lymph node (LN) "collapse," triggered by B cell translocation into lymphatic spaces and decreased lymphatic drainage. The aim of this study was to examine whether the efficacy of B cell depletion therapy is associated with restoration of lymphatic drainage due to removal of obstructing nodal B cells. METHODS: We used contrast-enhanced magnetic resonance imaging, indocyanine green near-infrared imaging, and intravital immunofluorescence imaging to longitudinally assess synovitis, lymphatic flow, and cell migration in lymphatic vessels in TNF-transgenic mice. We conducted tests to determine whether the efficacy of B cell depletion therapy is associated with restoration of lymphatic draining and cell egress from arthritic joints. RESULTS: Unlike active lymphatics to normal and prearthritic knees, afferent lymphatic vessels to collapsed LNs in inflamed knees do not pulse. Intravital immunofluorescence imaging demonstrated that CD11b+ monocyte/macrophages in lymphatic vessels afferent to expanding LNs travel at high velocity (mean±SD 186±37 µm/second), while these cells are stationary in lymphatic vessels afferent to collapsed popliteal LNs. B cell depletion therapy for arthritis flares in TNF-transgenic mice significantly decreased knee synovium volume (by 50% from the baseline level) and significantly increased lymphatic clearance compared with placebo (P<0.05). This increased lymphatic drainage restored macrophage egress from inflamed joints without recovery of the lymphatic pulse. CONCLUSION: These results support a novel mechanism in which B cell depletion therapy for joint arthritis flares lessens inflammation by increasing lymphatic drainage and subsequent migration of cells and cytokines from the synovial space.


Asunto(s)
Artritis Reumatoide/terapia , Articulación de la Rodilla/patología , Vasos Linfáticos/patología , Depleción Linfocítica/métodos , Sinovitis/patología , Animales , Artritis Reumatoide/patología , Linfocitos B , Antígeno CD11b , Citometría de Flujo , Inmunohistoquímica , Ganglios Linfáticos , Imagen por Resonancia Magnética , Ratones , Ratones Transgénicos , Microscopía Fluorescente , Resultado del Tratamiento
10.
Cell Tissue Res ; 347(3): 575-88, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21695398

RESUMEN

Biomaterial scaffolds functionalized to stimulate endogenous repair mechanisms via the incorporation of osteogenic cues offer a potential alternative to bone grafting for the treatment of large bone defects. We first quantified the ability of a self-complementary adeno-associated viral vector encoding bone morphogenetic protein 2 (scAAV2.5-BMP2) to enhance human stem cell osteogenic differentiation in vitro. In two-dimensional culture, scAAV2.5-BMP2-transduced human mesenchymal stem cells (hMSCs) displayed significant increases in BMP2 production and alkaline phosphatase activity compared with controls. hMSCs and human amniotic-fluid-derived stem cells (hAFS cells) seeded on scAAV2.5-BMP2-coated three-dimensional porous polymer Poly(ε-caprolactone) (PCL) scaffolds also displayed significant increases in BMP2 production compared with controls during 12 weeks of culture, although only hMSC-seeded scaffolds displayed significantly increased mineral formation. PCL scaffolds coated with scAAV2.5-BMP2 were implanted into critically sized immunocompromised rat femoral defects, both with or without pre-seeding of hMSCs, representing ex vivo and in vivo gene therapy treatments, respectively. After 12 weeks, defects treated with acellular scAAV2.5-BMP2-coated scaffolds displayed increased bony bridging and had significantly higher bone ingrowth and mechanical properties compared with controls, whereas defects treated with scAAV2.5-BMP2 scaffolds pre-seeded with hMSCs failed to display significant differences relative to controls. When pooled, defect treatment with scAAV2.5-BMP2-coated scaffolds, both with or without inclusion of pre-seeded hMSCs, led to significant increases in defect mineral formation at all time points and increased mechanical properties compared with controls. This study thus presents a novel acellular bone-graft-free endogenous repair therapy for orthotopic tissue-engineered bone regeneration.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Huesos/efectos de los fármacos , Huesos/patología , Materiales Biocompatibles Revestidos/farmacología , Dependovirus/genética , Andamios del Tejido/química , Cicatrización de Heridas/efectos de los fármacos , Líquido Amniótico/citología , Animales , Proteína Morfogenética Ósea 2/metabolismo , Calcificación Fisiológica/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , ADN/metabolismo , Dependovirus/efectos de los fármacos , Femenino , Fémur/efectos de los fármacos , Fémur/patología , Humanos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Poliésteres/farmacología , Ratas , Ratas Desnudas , Transducción Genética , beta-Galactosidasa/metabolismo
11.
Arthritis Rheum ; 63(8): 2318-28, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21538325

RESUMEN

OBJECTIVE: To investigate whether the enhancement of joint lymphangiogenesis by injection of vascular endothelial growth factor C (VEGF-C) adeno-associated virus (AAV) into the affected joints has therapeutic efficacy in chronic inflammatory arthritis in mice. METHODS: Tumor necrosis factor-transgenic (TNF-Tg) mice were used as a model of chronic inflammatory arthritis. Human VEGF-C was cloned into an AAV expression vector to generate AAV-VEGF-C. The joints of TNF-Tg mice were injected with AAV-VEGF-C or AAV-luciferase (AAV-Luc) as a control. During the 4 months following injection, magnetic resonance imaging of the joints and lymphatic imaging were performed to assess changes in synovial volume and lymph flow from the joint tissues to local draining lymph nodes. Joint inflammation, bone erosion, and cartilage loss were examined by histologic analyses. Lymphatic vessel formation was assessed using immunohistochemistry. RESULTS: Intraarticular administration of AAV-VEGF-C virus significantly attenuated the increase in synovial volume and increased lymphatic vessel number in the joint sections, as compared with that in control AAV-Luc-injected joints, during the 4-month period. This was accompanied by a reduction in the area of inflammation, bone erosion, cartilage loss, and osteoclast numbers. Lymph flow from the joints to local draining lymph nodes was slower in TNF-Tg mice than in wild-type littermates, and was significantly improved with AAV-VEGF-C treatment. CONCLUSION: Intraarticular injection of AAV-VEGF-C increased lymphangiogenesis and improved lymphatic drainage from the inflamed joints of mice, resulting in attenuation of joint tissue damage. Thus, improvement of joint lymphatic function by local administration of lymphatic growth factors represents a new therapeutic approach for chronic inflammatory arthritis.


Asunto(s)
Artritis/terapia , Articulaciones/patología , Factor C de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Artritis/patología , Inflamación/metabolismo , Inflamación/patología , Inflamación/terapia , Articulaciones/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Ratones , Ratones Transgénicos , Factor C de Crecimiento Endotelial Vascular/metabolismo
12.
Ann N Y Acad Sci ; 1192: 84-94, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20392222

RESUMEN

The effects of antiresorptive agents (e.g., alendronate [Aln], osteoprotegerin [OPG]) on bone infection are unknown. Thus, their effects on implant-associated osteomyelitis (OM) were investigated in mice using PBS (placebo), gentamycin, and etanercept (TNFR:Fc) controls. None of the drugs affected humoral immunity, angiogenesis, or chronic infection. However, the significant (P < 0.05 vs. PBS) inhibition of cortical osteolysis and decreased draining lymph node size in Aln- and OPG-treated mice was associated with a significant (P < 0.05) increase in the incidence of high-grade infections during the establishment of OM. In contrast, the high-grade infections in TNFR:Fc-treated mice were associated with immunosuppression, as evidenced by the absence of granulomas and presence of Gram(+) biofilm in the bone marrow. Collectively, these findings indicate that although antiresorptive agents do not exacerbate chronic OM, they can increase the bacterial load during early infection by decreasing lymphatic drainage and preventing the removal of necrotic bone that harbors the bacteria.


Asunto(s)
Conservadores de la Densidad Ósea/farmacología , Enfermedades Maxilomandibulares/inducido químicamente , Osteomielitis/inducido químicamente , Osteonecrosis/inducido químicamente , Animales , Biopelículas/efectos de los fármacos , Enfermedad Crónica , Citocinas/efectos de los fármacos , Evaluación Preclínica de Medicamentos , Humanos , Inmunidad/efectos de los fármacos , Incidencia , Enfermedades Maxilomandibulares/epidemiología , Enfermedades Maxilomandibulares/inmunología , Ratones , Neovascularización Fisiológica/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/fisiología , Osteomielitis/complicaciones , Osteomielitis/epidemiología , Osteomielitis/inmunología , Osteonecrosis/epidemiología , Osteonecrosis/inmunología , Infecciones Estafilocócicas/inducido químicamente , Infecciones Estafilocócicas/complicaciones , Infecciones Estafilocócicas/epidemiología
13.
Arthritis Rheum ; 60(9): 2666-76, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19714652

RESUMEN

OBJECTIVE: This study was undertaken to investigate the effect of lymphatic inhibition on joint and draining lymph node (LN) pathology during the course of arthritis progression in mice. METHODS: Tumor necrosis factor (TNF)-transgenic mice were used as a model of chronic inflammatory arthritis. Mice were subjected to contrast-enhanced magnetic resonance imaging to obtain ankle and knee joint synovial volumes and draining popliteal LN volumes before and after 8 weeks of treatment with vascular endothelial growth factor receptor 3 (VEGFR-3) neutralizing antibody, VEGFR-2 neutralizing antibody, or isotype IgG. Animals were subjected to near-infrared lymphatic imaging to determine the effect of VEGFR-3 neutralization on lymph transport from paws to draining popliteal LNs. Histologic, immunohistochemical, and reverse transcriptase-polymerase chain reaction analyses were used to examine lymphatic vessel formation and the morphology of joints and popliteal LNs. RESULTS: Compared with IgG treatment, VEGFR-3 neutralizing antibody treatment significantly decreased the size of popliteal LNs, the number of lymphatic vessels in joints and popliteal LNs, lymphatic drainage from paws to popliteal LNs, and the number of VEGF-C-expressing CD11b+ myeloid cells in popliteal LNs. However, it increased the synovial volume and area of inflammation in ankle and knee joints. VEGFR-2 neutralizing antibody, in contrast, inhibited both lymphangiogenesis and joint inflammation. CONCLUSION: These findings indicate that lymphangiogenesis and lymphatic drainage are reciprocally related to the severity of joint lesions during the development of chronic arthritis. Lymphatic drainage plays a beneficial role in controlling the progression of chronic inflammation.


Asunto(s)
Artritis/metabolismo , Inflamación/metabolismo , Ganglios Linfáticos/irrigación sanguínea , Ganglios Linfáticos/metabolismo , Neovascularización Fisiológica/fisiología , Índice de Severidad de la Enfermedad , Receptor 3 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Animales , Anticuerpos/farmacología , Artritis/patología , Artritis/fisiopatología , Antígeno CD11b/metabolismo , Enfermedad Crónica , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Inflamación/patología , Inflamación/fisiopatología , Ganglios Linfáticos/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neovascularización Fisiológica/efectos de los fármacos , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Factor C de Crecimiento Endotelial Vascular/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo
14.
J Orthop Res ; 27(9): 1162-8, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19242999

RESUMEN

Gene therapy is a promising clinical tool that is no longer limited as a method to supplement genetic deficits, but rather is considered reliable for delivering proteins to specific tissues or cells. Recombinant adeno-associated virus (rAAV) vector is one of the most potent gene transfer vehicles. Many biomaterials have been used in reconstructive surgery, but their biological inactivity has limited their use. To overcome shortcomings of available bone-related biomaterials, we investigated the combination of rAAV with biomaterials. Taking advantage of the method of lyophilizing rAAV onto biomaterials, we showed that an rAAV coating successfully induced beta-galactosidase protein expression by rat fibroblasts on hydroxyapatite, beta-tricalcium phosphate, and titanium alloy in vitro. beta-Galactosidase expression was detected for 8 weeks after implantation of rAAV-coated hydroxyapatite into rat back muscles in vivo. A coating of bone morphogenetic protein-2-expressing rAAV induced significant de novo bone formation on hydroxyapatite in rat back muscles. Our study demonstrates that the combination of lyophilized rAAV and biomaterials presents a promising strategy for bone regenerative medicine.


Asunto(s)
Adenoviridae/genética , Proteína Morfogenética Ósea 2/genética , Regeneración Ósea/fisiología , Técnicas de Transferencia de Gen , Terapia Genética/métodos , Animales , Fosfatos de Calcio , Células Cultivadas , Materiales Biocompatibles Revestidos , Durapatita , Fibroblastos/citología , Fibroblastos/fisiología , Liofilización , Plásmidos/genética , Ratas , Ratas Wistar , Titanio , Transducción Genética/métodos , beta-Galactosidasa/genética
15.
Arthritis Res Ther ; 9 Suppl 1: S7, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17634146

RESUMEN

The receptor activator of nuclear factor-kappaB ligand (RANKL), its cognate receptor RANK, and its natural decoy receptor osteoprotegerin have been identified as the final effector molecules of osteoclastic bone resorption. This has provided an ideal target for therapeutic interventions in metabolic bone disease. As described in previous reviews in this supplement, RANKL signaling is required for osteoclast differentiation, activation, and survival. Furthermore, in vivo inhibition of RANKL leads to immediate osteoclast apoptosis, and there are no in vivo models of bone resorption that are refractory to RANKL inhibition. Thus, the only step remaining in the development of a clinical intervention is the generation of a safe, effective, and specific drug that can inhibit RANKL in humans. Here we review the clinical development of denosumab (formerly known as AMG 162), which is a fully human mAb directed against RANKL. This discussion includes the breadth of 21 human studies that have led to the current phase 3 clinical trials seeking approval for use of this agent to treat postmenopausal women with low bone mineral density (osteoporosis) and patients with metastatic lytic bone lesions (multiple myeloma, and prostate and breast cancer).


Asunto(s)
Ligando RANK/antagonistas & inhibidores , Ligando RANK/metabolismo , Tecnología Farmacéutica/métodos , Animales , Humanos , Ensayos Clínicos Controlados Aleatorios como Asunto/métodos , Receptor Activador del Factor Nuclear kappa-B/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/metabolismo
16.
J Cell Biochem ; 97(2): 226-32, 2006 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-16240334

RESUMEN

Focal bone loss around inflamed joints in patients with autoimmune disease, such as rheumatoid arthritis, remains a serious clinical problem. The recent elucidation of the RANK/RANK-ligand/OPG pathway and its role as the final effector of osteoclastogenesis and bone resorption has brought a tremendous understanding of the pathophysiology of inflammatory bone loss, and has heightened expectation of a novel intervention. Here, we review the etiology of inflammatory bone loss, the RANK/RANK-ligand/OPG pathway, and the clinical development of anti-RANK-ligand therapy.


Asunto(s)
Artritis/terapia , Enfermedades Óseas/terapia , Resorción Ósea/etiología , Proteínas Portadoras/fisiología , Inflamación/terapia , Glicoproteínas de Membrana/fisiología , Artritis/etiología , Artritis/metabolismo , Enfermedades Óseas/etiología , Resorción Ósea/inmunología , Resorción Ósea/terapia , Proteínas Portadoras/antagonistas & inhibidores , Glicoproteínas/fisiología , Humanos , Glicoproteínas de Membrana/antagonistas & inhibidores , Modelos Biológicos , Osteoclastos/fisiología , Osteoprotegerina , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B , Receptores Citoplasmáticos y Nucleares/fisiología , Receptores del Factor de Necrosis Tumoral/fisiología
17.
J Orthop Res ; 21(4): 676-84, 2003 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-12798068

RESUMEN

As dominant regulators of osteoclastogenesis and bone resorption, receptor activator of NFkappaB (RANK), receptor activator of NFkappaB ligand, and OPG have been identified as ideal drug targets for the treatment of metabolic bone disease. One concern regarding the therapeutic use of RANK signaling inhibitors is their effect on fracture healing. Therefore we tested if uncoupling and osteoclast depletion via RANK blockade affects callus formation, maturation and matrix remodeling, as well as union rates in a mouse tibia fracture model. Low dose (1 mg/kg i.p.) RANK:Fc therapy had no effect on callus formation, matrix maturation and remodeling, and resulted in 100% bony union by day 28. High dose RANK:Fc treatment (10 mg/kg i.p.) effectively eliminated osteoclasts at the fracture site on day 14, with no significant effects on fracture healing. When therapy was discontinued, normal numbers of osteoclasts were observed at the fracture site by day 28. However, continuous therapy resulted in a large osteopetrotic callus consisting of both mineralized and unmineralized matrix that was void of osteoclasts, but bony union was unaffected at day 28. We also evaluated this process in the complete absence of RANK signaling using RANK -/- mice. These animals exhibited significant radiographic and histologic evidence of callus formation, indicating that RANK signaling is not required for fracture callus formation. However, only 33% of RANK -/- animals formed bony unions compared to 100% of the osteopetrotic control mice. This defect was most likely a result of decreased blood flow, as evidenced by fewer blood vessels in the RANK -/- animals. Together, these data imply that osteoclast depletion via inhibition of RANK signaling is a viable option for the treatment of pathological bone loss since no adverse effects on fracture healing are observed when therapy is discontinued.


Asunto(s)
Curación de Fractura/fisiología , Glicoproteínas/antagonistas & inhibidores , Glicoproteínas/genética , FN-kappa B/metabolismo , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/genética , Transducción de Señal/fisiología , Fracturas de la Tibia/metabolismo , Animales , Callo Óseo/metabolismo , Células CHO , Cricetinae , Curación de Fractura/efectos de los fármacos , Glicoproteínas/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Mutantes , Osteoblastos/fisiología , Osteoclastos/fisiología , Osteoprotegerina , Receptores del Factor de Necrosis Tumoral , Proteínas Recombinantes/farmacología , Flujo Sanguíneo Regional , Transducción de Señal/efectos de los fármacos
18.
Endocrinology ; 144(6): 2514-23, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12746314

RESUMEN

Whereas bone morphogenetic protein (BMP)-signaling events induce maturational characteristics in vitro, recent evidence suggests that the effects of other regulators might be mediated through BMP-signaling events. The present study examines the mechanism through which retinoic acid (RA) stimulates differentiation in chicken embryonic caudal sternal chondrocyte cultures. Both RA and BMP-2 induced expression of the chondrocyte maturational marker, colX, in chondrocyte cultures by 8 d. Though the RA effect was small, it synergistically enhanced the effect of BMP-2 on colX and phosphatase activity. Inhibition of either RA or BMP signaling, with selective inhibitors, interfered with the inductive effects of these agents but also inhibited the complementary pathway, demonstrating a codependence of RA and BMP signaling during chondrocyte maturation. BMP-2 did not enhance the effects of RA on an RA-responsive reporter construct, but RA enhanced basal activity and synergistically enhanced BMP-2 stimulation of the BMP-responsive chicken type X collagen reporter. A similar synergistic interaction between RA and BMP-2 was observed on colX expression. RA did not increase the expression of the type IA BMP receptor but did markedly up-regulate the expression of Smad1 and Smad5 proteins, important participants in the BMP pathway. Inhibition of RA signaling, with the selective inhibitor AGN 193109, blocked RA-mediated induction of the Smad proteins and chondrocyte differentiation. These findings demonstrate that RA induces the expression of BMP-signaling molecules and enhances BMP effects in chondrocytes.


Asunto(s)
Antineoplásicos/farmacología , Proteínas Morfogenéticas Óseas/farmacología , Condrocitos/citología , Proteínas de Unión al ADN/genética , Transactivadores/genética , Factor de Crecimiento Transformador beta , Tretinoina/farmacología , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2 , Diferenciación Celular/efectos de los fármacos , Embrión de Pollo , Pollos , Condrocitos/fisiología , Colágeno Tipo X/genética , Sinergismo Farmacológico , Fosfoproteínas/genética , Transducción de Señal/efectos de los fármacos , Proteínas Smad , Proteína Smad5 , Esternón/citología
19.
Radiat Res ; 157(1): 62-8, 2002 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-11754643

RESUMEN

Radiation therapy plays an important role as part of multimodality treatment for a number of childhood malignancies. The damaging effects of radiation on bone formation in children have been well documented. Recent work suggests that the postirradiation increase in cytosolic calcium is probably responsible for the deleterious effects of radiation on growth plate chondrocytes because it causes a specific suppression of the mitogen PTHrP. Using an in vitro model of avian growth plate chondrocytes, this study demonstrates that pentoxifylline is effective in increasing basal PTHrP mRNA levels and partially preventing the radiation-induced decrease in PTHrP mRNA. This effect of pentoxifylline is probably due to its ability to lower basal levels of cytosolic calcium and the radiation-induced increase in cytosolic calcium in chondrocytes. Pentoxifylline also prevented the radiation-induced decreases in [3H]thymidine uptake and BCL2 and PTHrP receptor mRNA levels in chondrocytes. The effects of pentoxifylline appear to be specific for the PTHrP signaling pathway because it did not alter basal TGFB mRNA levels or TGFB mRNA expression in irradiated chondrocytes. The results of the current study suggest that by decreasing basal cytosolic calcium levels and curtailing the radiation-induced increase in cytosolic calcium levels in chondrocytes, pentoxifylline is able to sustain PTHrP signaling in chondrocytes and maintains the proliferative signal that is necessary to prevent chondrocytes from undergoing apoptosis.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Condrocitos/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Placa de Crecimiento/efectos de los fármacos , Pentoxifilina/farmacología , Protectores contra Radiación/farmacología , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , División Celular/efectos de los fármacos , División Celular/efectos de la radiación , Embrión de Pollo , Condrocitos/efectos de la radiación , Citosol/metabolismo , Evaluación Preclínica de Medicamentos , Genes bcl-2/efectos de la radiación , Placa de Crecimiento/efectos de la radiación , Proteína Relacionada con la Hormona Paratiroidea , Biosíntesis de Proteínas , Proteínas/genética , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , ARN Mensajero/biosíntesis , Tolerancia a Radiación/efectos de los fármacos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA