Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Stem Cells Transl Med ; 8(8): 810-821, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31038850

RESUMEN

Xenogeneic-free media are required for translating advanced therapeutic medicinal products to the clinics. In addition, process efficiency is crucial for ensuring cost efficiency, especially when considering large-scale production of mesenchymal stem cells (MSCs). Human platelet lysate (HPL) has been increasingly adopted as an alternative for fetal bovine serum (FBS) for MSCs. However, its therapeutic and regenerative potential in vivo is largely unexplored. Herein, we compare the effects of FBS and HPL supplementation for a scalable, microcarrier-based dynamic expansion of human periosteum-derived cells (hPDCs) while assessing their bone forming capacity by subcutaneous implantation in small animal model. We observed that HPL resulted in faster cell proliferation with a total fold increase of 5.2 ± 0.61 in comparison to 2.7 ± 02.22-fold in FBS. Cell viability and trilineage differentiation capability were maintained by HPL, although a suppression of adipogenic differentiation potential was observed. Differences in mRNA expression profiles were also observed between the two on several markers. When implanted, we observed a significant difference between the bone forming capacity of cells expanded in FBS and HPL, with HPL supplementation resulting in almost three times more mineralized tissue within calcium phosphate scaffolds. FBS-expanded cells resulted in a fibrous tissue structure, whereas HPL resulted in mineralized tissue formation, which can be classified as newly formed bone, verified by µCT and histological analysis. We also observed the presence of blood vessels in our explants. In conclusion, we suggest that replacing FBS with HPL in bioreactor-based expansion of hPDCs is an optimal solution that increases expansion efficiency along with promoting bone forming capacity of these cells. Stem Cells Translational Medicine 2019;8:810&821.


Asunto(s)
Técnicas de Cultivo Celular por Lotes/métodos , Regeneración Ósea , Medios de Cultivo/farmacología , Cultivo Primario de Células/métodos , Células Madre/efectos de los fármacos , Adipogénesis , Animales , Técnicas de Cultivo Celular por Lotes/instrumentación , Reactores Biológicos , Plaquetas/metabolismo , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Medios de Cultivo/química , Humanos , Ratones , Ratones Desnudos , Osteogénesis , Periostio/citología , Cultivo Primario de Células/instrumentación , Trasplante de Células Madre/métodos , Células Madre/fisiología
2.
PLoS One ; 13(6): e0198104, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29897942

RESUMEN

Tissue calcification is an important physiological process required for the normal structure and function of bone. However, ectopic or excessive calcification contributes to diseases such as chondrocalcinosis, to calcium deposits in the skin or to vascular calcification. SMOC2 is a member of the BM-40/osteonectin family of calcium-binding secreted matricellular proteins. Using osteoprogenitor MC3T3-E1 cells stably overexpressing SMOC2, we show that SMOC2 inhibits osteogenic differentiation and extracellular matrix mineralization. Stable Smoc2 knockdown in these cells had no effect on mineralization suggesting that endogenous SMOC2 is not essential for the mineralization process. Mineralization in MC3T3-E1 cells overexpressing mutant SMOC2 lacking the extracellular calcium-binding domain was significantly increased compared to cells overexpressing full length SMOC2. When SMOC2 overexpressing cells were cultured in the presence of extracellular calcium supplementation, SMOC2's inhibitory effect on calcification was rescued. Our observations were translationally validated in primary human periosteal-derived cells. Furthermore, SMOC2 was able to impair mineralization in transdifferentiated human umbilical vein endothelial cells. Taken together, our data indicate that SMOC2 can act as an inhibitor of mineralization. We propose a possible role for SMOC2 to prevent calcification disorders.


Asunto(s)
Calcificación Fisiológica/genética , Proteínas de Unión al Calcio/fisiología , Diferenciación Celular/genética , Células Endoteliales/fisiología , Osteoblastos/fisiología , Animales , Proteínas de Unión al Calcio/genética , Células Cultivadas , Células Endoteliales de la Vena Umbilical Humana/fisiología , Humanos , Ratones , Osteogénesis/genética
3.
Acta Biomater ; 72: 447-460, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29626696

RESUMEN

Rapid neovascularization of a tissue-engineered (TE) construct by the host vasculature is quintessential to warrant effective bone regeneration. This process can be promoted through active induction of angiogenic growth factor secretion or by implementation of in vitro pre-vascularization strategies. In this study, we aimed at optimizing the pro-angiogenic effect of Cobalt (Co2+) to enhance vascular endothelial growth factor (VEGF) expression by human periosteum-derived mesenchymal stem cells (hPDCs). Simultaneously we set out to promote microvascular network formation by co-culturing with human umbilical vein endothelial cells (HUVECs). The results showed that Co2+ treatments (at 50, 100 or 150 µM) significantly upregulated in vitro VEGF expression, but inhibited hPDCs growth and HUVECs network formation in co-cultures. These inhibitory effects were mitigated at lower Co2+ concentrations (at 5, 10 or 25 µM) while VEGF expression remained significantly upregulated and further augmented in the presence of Ascorbic Acid and Dexamethasone possibly through Runx2 upregulation. The supplements also facilitated HUVECs network formation, which was dependent on the quantity and spatial distribution of collagen type-1 matrix deposited by the hPDCs. When applied to hPDCs seeded onto calcium phosphate scaffolds, the supplements significantly induced VEGF secretion in vitro, and promoted higher vascularization upon ectopic implantation in nude mice shown by an increase of CD31 positive blood vessels within the scaffolds. Our findings provided novel insights into the pleotropic effects of Co2+ on angiogenesis (i.e. promoted VEGF secretion and inhibited endothelial network formation), and showed potential to pre-condition TE constructs under one culture regime for improved implant neovascularization in vivo. STATEMENT OF SIGNIFICANT: Cobalt (Co2+) is known to upregulate vascular endothelial growth factor (VEGF) secretion, however it also inhibits in vitro angiogenesis through unknown Co2+-induced events. This limits the potential of Co2+ for pro-angiogenesis of tissue engineered (TE) implants. We showed that Co2+ upregulated VEGF expression by human periosteum-derived cells (hPDCs) but reduced the cell growth, and endothelial network formation due to reduction of col-1 matrix deposition. Supplementation with Ascorbic acid and Dexamethasone concurrently improved hPDCs growth, endothelial network formation, and upregulated VEGF secretion. In vitro pre-conditioning of hPDC-seeded TE constructs with this fine-tuned medium enhanced VEGF secretion and implant neovascularization. Our study provided novel insights into the pleotropic effects of Co2+ on angiogenesis and formed the basis for improving implant neovascularization.


Asunto(s)
Cobalto , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Implantes Experimentales , Células Madre Mesenquimatosas/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Periostio/metabolismo , Factor A de Crecimiento Endotelial Vascular/biosíntesis , Adulto , Cobalto/química , Cobalto/farmacología , Técnicas de Cocultivo , Femenino , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Periostio/citología
4.
Sci Rep ; 8(1): 2942, 2018 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-29440666

RESUMEN

Neural tube defects (NTDs) are severe congenital abnormalities, caused by failed closure of neural tube during early embryonic development. Periconceptional folic acid (FA) supplementation greatly reduces the risk of NTDs. However, the molecular mechanisms behind NTDs and the preventive role of FA remain unclear. Here, we use human induced pluripotent stem cells (iPSCs) derived from fetuses with spina bifida aperta (SBA) to study the pathophysiology of NTDs and explore the effects of FA exposure. We report that FA exposure in SBA model is necessary for the proper formation and maturation of neural tube structures and robust differentiation of mesodermal derivatives. Additionally, we show that the folate antagonist methotrexate dramatically affects the formation of neural tube structures and FA partially reverts this aberrant phenotype. In conclusion, we present a novel model for human NTDs and provide evidence that it is a powerful tool to investigate the molecular mechanisms underlying NTDs, test drugs for therapeutic approaches.


Asunto(s)
Ácido Fólico/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Fenotipo , Espina Bífida Quística/patología , Diferenciación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Desarrollo de Músculos/efectos de los fármacos , Neuronas/citología , Neuronas/efectos de los fármacos , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX7/genética , Esferoides Celulares/citología , Esferoides Celulares/efectos de los fármacos , Regulación hacia Arriba/efectos de los fármacos
5.
Tissue Eng Part C Methods ; 22(5): 473-86, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-27018617

RESUMEN

Successful application of cell-based strategies in cartilage and bone tissue engineering has been hampered by the lack of robust protocols to efficiently differentiate mesenchymal stem cells into the chondrogenic lineage. The development of chemically defined culture media supplemented with growth factors (GFs) has been proposed as a way to overcome this limitation. In this work, we applied a fractional design of experiment (DoE) strategy to screen the effect of multiple GFs (BMP2, BMP6, GDF5, TGF-ß1, and FGF2) on chondrogenic differentiation of human periosteum-derived mesenchymal stem cells (hPDCs) in vitro. In a micromass culture (µMass) system, BMP2 had a positive effect on glycosaminoglycan deposition at day 7 (p < 0.001), which in combination with BMP6 synergistically enhanced cartilage-like tissue formation that displayed in vitro mineralization capacity at day 14 (p < 0.001). Gene expression of µMasses cultured for 7 days with a medium formulation supplemented with 100 ng/mL of BMP2 and BMP6 and a low concentration of GDF5, TGF-ß1, and FGF2 showed increased expression of Sox9 (1.7-fold) and the matrix molecules aggrecan (7-fold increase) and COL2A1 (40-fold increase) compared to nonstimulated control µMasses. The DoE analysis indicated that in GF combinations, BMP2 was the strongest effector for chondrogenic differentiation of hPDCs. When transplanted ectopically in nude mice, the in vitro-differentiated µMasses showed maintenance of the cartilaginous phenotype after 4 weeks in vivo. This study indicates the power of using the DoE approach for the creation of new medium formulations for skeletal tissue engineering approaches.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/efectos de los fármacos , Condrocitos/citología , Condrogénesis/fisiología , Péptidos y Proteínas de Señalización Intercelular/farmacología , Periostio/citología , Ingeniería de Tejidos/métodos , Adolescente , Animales , Células Cultivadas , Condrocitos/efectos de los fármacos , Condrocitos/metabolismo , Condrogénesis/efectos de los fármacos , Femenino , Humanos , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Desnudos , Periostio/efectos de los fármacos , Periostio/metabolismo , Donantes de Tejidos
6.
Cell Metab ; 23(2): 265-79, 2016 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-26863487

RESUMEN

Cell-based therapy is a promising strategy in regenerative medicine, but the poor survival rate of the implanted cells remains a major challenge and limits clinical translation. We preconditioned periosteal cells to the hypoxic and ischemic environment of the bone defect site by deleting prolyl hydroxylase domain-containing protein 2 (PHD2), resulting in hypoxia-inducible factor 1 alpha (HIF-1α) stabilization. This strategy increased postimplantation cell survival and improved bone regeneration. The enhanced cell viability was angiogenesis independent but relied on combined changes in glutamine and glycogen metabolism. HIF-1α stabilization stimulated glutaminase-mediated glutathione synthesis, maintaining redox homeostasis at baseline and during oxidative or nutrient stress. Simultaneously, HIF-1α signaling increased glycogen storage, preventing an energy deficit during nutrient or oxygen deprivation. Pharmacological inhibition of PHD2 recapitulated the adaptations in glutamine and glycogen metabolism and, consequently, the beneficial effects on cell survival. Thus, targeting cellular metabolism is an appealing strategy for bone regeneration and cell-based therapy in general.


Asunto(s)
Metabolismo Energético , Glutamina/metabolismo , Glucógeno/metabolismo , Homeostasis , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Osteocitos/trasplante , Animales , Regeneración Ósea , Respiración de la Célula , Supervivencia Celular , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Silenciador del Gen , Glutaminasa/metabolismo , Ratones , Neovascularización Fisiológica , Osteocitos/metabolismo , Oxidación-Reducción , Estrés Oxidativo , Periostio/patología , Especies Reactivas de Oxígeno/metabolismo
7.
Biores Open Access ; 3(6): 265-77, 2014 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-25469312

RESUMEN

Functionalization of tissue engineering scaffolds with in vitro-generated bone-like extracellular matrix (ECM) represents an effective biomimetic approach to promote osteogenic differentiation of stem cells in vitro. However, the bone-forming capacity of these constructs (seeded with or without cells) is so far not apparent. In this study, we aimed at developing a mineralizing culture condition to biofunctionalize three-dimensional (3D) porous scaffolds with highly mineralized ECM in order to produce devitalized, osteoinductive mineralized carriers for human periosteal-derived progenitors (hPDCs). For this, three medium formulations [i.e., growth medium only (BM1), with ascorbic acid (BM2), and with ascorbic acid and dexamethasone (BM3)] supplemented with calcium (Ca(2+)) and phosphate (PO4 (3-)) ions simultaneously as mineralizing source were investigated. The results showed that, besides the significant impacts on enhancing cell proliferation (the highest in BM3 condition), the formulated mineralizing media differentially regulated the osteochondro-related gene markers in a medium-dependent manner (e.g., significant upregulation of BMP2, bone sialoprotein, osteocalcin, and Wnt5a in BM2 condition). This has resulted in distinguished cell populations that were identifiable by specific gene signatures as demonstrated by the principle component analysis. Through devitalization, mineralized carriers with apatite crystal structures unique to each medium condition (by X-ray diffraction and SEM analysis) were obtained. Quantitatively, BM3 condition produced carriers with the highest mineral and collagen contents as well as human-specific VEGF proteins, followed by BM2 and BM1 conditions. Encouragingly, all mineralized carriers (after reseeded with hPDCs) induced bone formation after 8 weeks of subcutaneous implantation in nude mice models, with BM2-carriers inducing the highest bone volume, and the lowest in the BM3 condition (as quantitated by nano-computed tomography [nano-CT]). Histological analysis revealed different bone formation patterns, either bone ossicles containing bone marrow surrounding the scaffold struts (in BM2) or bone apposition directly on the struts' surface (in BM1 and BM3). In conclusion, we have presented experimental data on the feasibility to produce devitalized osteoinductive mineralized carriers by functionalizing 3D porous scaffolds with an in vitro cell-made mineralized matrix under the mineralizing culture conditions.

8.
Arthritis Res Ther ; 14(2): R59, 2012 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-22410100

RESUMEN

INTRODUCTION: Studies in the spontaneous ankylosis model in aging male DBA/1 mice and in patients with ankylosing spondylitis provide evidence that inflammation and new tissue formation leading to joint or spine ankylosis are likely linked but largely uncoupled processes. We previously proposed the 'entheseal stress' hypothesis that defines microdamage or cell stress in the enthesis as a trigger for these disease processes. Here, we further investigated the relationship between inflammation and ankylosis by focusing on the early phase of the spontaneous arthritis model. METHODS: Aging male DBA/1 mice from different litters were caged together at the age of ten weeks and studied for signs of arthritis. A group of DBA/1 mice were treated daily with dexamethasone (0.5 µg/g body weight). Severity of disease was assessed by histomorphology and by positron emission tomography (PET) using 2-[18F]fluoro-2-deoxy-D-glucose (18F-FDG) as a tracer. Bone loss in dexamethasone-treated or control mice was determined by in vivo dual-energy X-ray absorptiometry. Chemokine gene expression was studied ex vivo in dissected paws and in vitro in mesenchymal cells (periosteal and bone marrow stromal cells) by quantitative real-time PCR in the presence or absence of bone morphogenetic protein 2 (BMP2) and dexamethasone. RESULTS: Dexamethasone treatment did not affect incidence or severity of ankylosis, but led to an expected reduction in inflammation in the paws at week 15 as measured by PET tracer uptake. Treatment with dexamethasone negatively affected bone mineral density. Chemokines attracting neutrophils and lymphocytes were expressed in affected paws. In vitro, BMP2 stimulation upregulated chemokines in different mesenchymal joint-associated cell types, an effect that was inhibited by dexamethasone. CONCLUSIONS: BMP signaling may be a trigger for both inflammation and ankylosis in the spontaneous model of ankylosing enthesitis. The lack of inhibition by glucocorticoids on new bone formation while causing systemic bone loss highlights the paradoxical simultaneous loss and gain of bone in patients with ankylosing spondylitis.


Asunto(s)
Envejecimiento/patología , Anquilosis/metabolismo , Anquilosis/patología , Artritis Experimental/tratamiento farmacológico , Artritis Experimental/metabolismo , Glucocorticoides/uso terapéutico , Estrés Fisiológico/inmunología , Envejecimiento/metabolismo , Animales , Anquilosis/tratamiento farmacológico , Artritis Experimental/patología , Densidad Ósea/efectos de los fármacos , Densidad Ósea/fisiología , Proteína Morfogenética Ósea 2/metabolismo , Células Cultivadas , Dexametasona/uso terapéutico , Humanos , Inflamación/metabolismo , Inflamación/patología , Inflamación/prevención & control , Masculino , Ratones , Ratones Endogámicos DBA
9.
Biomaterials ; 33(11): 3127-42, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22269651

RESUMEN

Stem cell-based strategies for bone regeneration, which use calcium phosphate (CaP)-based biomaterials in combination with developmentally relevant progenitor populations, have significant potential for clinical repair of skeletal defects. However, the exact mechanism of action and the stem cell-host-material interactions are still poorly understood. We studied if pre-conditioning of human periosteum-derived cells (hPDCs) in vitro could enhance, in combination with a CaP-based biomaterial carrier, ectopic bone formation in vivo. By culturing hPDCs in a biomimetic calcium (Ca(2+)) and phosphate (P(i)) enriched culture conditions, we observed an enhanced cell proliferation, decreased expression of mesenchymal stem cell (MSC) markers and upregulation of osteogenic genes including osterix, Runx2, osteocalcin, osteopontin, and BMP-2. However, the in vitro pre-conditioning protocols were non-predictive for in vivo ectopic bone formation. Surprisingly, culturing in the presence of Ca(2+) and P(i) supplements resulted in partial or complete abrogation of in vivo ectopic bone formation. Through histological, immunohistochemical and microfocus X-ray computed tomography (µCT) analysis of the explants, we found that in situ proliferation, collagen matrix deposition and the mediation of osteoclastic activity by hPDCs are associated to their ectopic bone forming capacity. These data were validated by the multivariate analysis and partial least square regression modelling confirming the non-predictability of in vitro parameters on in vivo ectopic bone formation. Our series of experiments provided further insights on the stem cell-host-material interactions that govern in vivo ectopic bone induction driven by hPDCs on CaP-based biomaterials.


Asunto(s)
Desarrollo Óseo/fisiología , Fosfatos de Calcio/química , Osteoblastos/citología , Osteogénesis/fisiología , Células Madre/citología , Ingeniería de Tejidos/métodos , Adolescente , Materiales Biocompatibles/síntesis química , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Femenino , Humanos , Masculino , Ensayo de Materiales , Osteoblastos/fisiología , Células Madre/fisiología
10.
Tissue Eng Part C Methods ; 18(2): 90-103, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21933019

RESUMEN

We report on the in vitro use of Ca(2+)/P(i) supplementation as a bio-instructive medium to drive human periosteum-derived cells (hPDCs) toward osteogenic differentiation on three-dimensional (3D) porous Ti6Al4V scaffolds. Through a multilevel factorial analysis, we have systematically investigated the biological effect and interactions of Ca(2+) or P(i) supplementation in three selected media preparations (i.e., basic growth medium, osteogenic medium [OM], and osteogenic medium without ß-glycerophosphate [OM(-)]) and have identified specific conditions which induce proliferation and significant osteogenic differentiation of two-dimensional (2D) hPDC cultures. These findings were translated from 2D to 3D cultures conditions to instruct hPDCs to populate porous Ti6Al4V scaffolds and to differentiate into the osteoblast lineage with collagenous matrix production and subsequent matrix mineralization on the 3D structures. These osteogenic hybrids may potentially serve as a clinically relevant customizable bone reparative unit, providing a biomimetic template to more effectively mediate in vivo bone regeneration.


Asunto(s)
Materiales Biocompatibles/farmacología , Biomimética/métodos , Calcio/farmacología , Análisis Multinivel , Osteogénesis/efectos de los fármacos , Fosfatos/farmacología , Ingeniería de Tejidos/métodos , Adolescente , Fosfatasa Alcalina/metabolismo , Aleaciones , Calcificación Fisiológica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Forma de la Célula/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/metabolismo , Análisis Factorial , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Osteogénesis/genética , Periostio/citología , Periostio/efectos de los fármacos , Periostio/enzimología , Porosidad/efectos de los fármacos , Andamios del Tejido/química , Titanio/farmacología
11.
Ann Rheum Dis ; 70(1): 208-13, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20805298

RESUMEN

OBJECTIVES: A functional polymorphism leading to reduced levels of growth and differentiation factor 5 (GDF5) was recently identified as a susceptibility factor for osteoarthritis. The authors studied the potential mechanisms of GDF5 involvement in osteoarthritis using haploinsufficient Gdf5(Bp-J/+) mice. METHODS: Gdf5(Bp-J/+) mice were challenged in the collagenase-induced arthritis model, the medial meniscus destabilisation model, the papain-induced arthritis model and a treadmill running model. Bone density and subchondral bone parameters were determined using dual energy x-ray absorptiometry and peripheral quantitative CT. Additional in-vitro and ex-vivo analyses studied cartilage metabolism, gait and collagen characteristics. RESULTS: Gdf5(Bp-J/+) mice appeared phenotypically normal. No difference in osteoarthritis severity was found in the different models, with the exception of increased synovial hyperplasia in the joints of Gdf5(Bp-J/+) mice in the treadmill model. However, in the collagenase-induced model severe joint damage was found in the contralateral joints of Gdf5(Bp-J/+) mice. Gait analysis demonstrated an aberrant walking pattern in Gdf5(Bp-J/+) mice. In addition, Gdf5(Bp-J/+) mice have a decreased subchondral bone density and a distorted arrangement of collagen fibres in bone. CONCLUSIONS: These data suggest that decreased GDF5 levels in mice can contribute to osteoarthritis development by different mechanisms including altered loading and subchondral bone changes. This highlights the importance of the joint as an organ with different tissues involved in joint disease.


Asunto(s)
Artritis Experimental/etiología , Factor 5 de Diferenciación de Crecimiento/deficiencia , Inestabilidad de la Articulación/etiología , Cojera Animal/etiología , Osteoartritis/etiología , Animales , Artritis Experimental/genética , Densidad Ósea/fisiología , Cartílago Articular/metabolismo , Colagenasas , Factor 5 de Diferenciación de Crecimiento/genética , Haploinsuficiencia , Inestabilidad de la Articulación/genética , Cojera Animal/genética , Ratones , Ratones Endogámicos A , Ratones Mutantes , Actividad Motora , Osteoartritis/genética , Papaína
12.
Arthritis Res Ther ; 9(2): 207, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17381828

RESUMEN

Joint destruction and tissue responses determine the outcome of chronic arthritis. Joint inflammation and damage are often the dominant clinical presentation. However, in some arthritic diseases, in particular the spondyloarthritides, joint remodeling is a prominent feature, with new cartilage and bone formation leading to ankylosis and contributing to loss of function. A role for bone morphogenetic proteins in joint remodeling has been demonstrated in the formation of both enthesophytes and osteophytes. Data from genetic models support a role for bone morphogenetic protein signaling in cartilage homeostasis. Finally, this signaling pathway is likely to play a steering role in the synovium.


Asunto(s)
Artritis/metabolismo , Artritis/patología , Proteínas Morfogenéticas Óseas/metabolismo , Articulaciones/metabolismo , Articulaciones/patología , Animales , Remodelación Ósea/fisiología , Cartílago/metabolismo , Cartílago/patología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA