Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Eur J Orthod ; 45(2): 224-234, 2023 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-36576115

RESUMEN

OBJECTIVE: Teriparatide (TPTD) and abaloparatide (ABL) are two osteoanabolic drugs targeting parathyroid hormone (PTH)1R signalling. This study aimed to investigate the effects of TPTD and ABL on the adolescent mandibular growth. METHOD: In total, 70 4-week-old male Sprague-Dawley rats were randomly divided into 14 groups, treated with intermittent TPDT or ABL at various doses, accompanied by mandibular advancement (MA) or not. 3D printing was used to fabricate an innovative splint for MA. After a 4-week treatment, morphological measurement, histological and immunohistochemical analysis were performed. Mandibular condylar chondrocytes (MCCs) were treated with TPTD or ABL, followed by CCK-8 assay, alcian blue staining, real time-PCR and immunofluorescent staining. RESULT: In vivo, TPTD or ABL alone increased the condylar length and cartilage thickness, with up-regulated SOX9 and COL II, whilst down-regulated COL X; however, when combined with MA, the promotive effects were attenuated. TPTD or ABL alone increased the mandibular body height and mandibular angle width, whilst increased the mandibular body length and alveolar bone width when combined with MA. In vitro, TPTD or ABL enhanced the MCC proliferation, glycosaminoglycan synthesis, COL II and SOX9 expression, whilst down-regulated COL X, Ihh and PTH1R expression. CONCLUSION: Both ABL and TPTD enhance mandibular growth in adolescent rats with site-specific and mechano-related effects, including propelling chondrogenesis at the condylar cartilage and promoting bone apposition at other mechano-responsive sites. They behave as promising drugs for mandibular growth modification, and in general ABL seems more potent than TPTD in this context.


Asunto(s)
Conservadores de la Densidad Ósea , Teriparatido , Ratas , Masculino , Animales , Teriparatido/farmacología , Teriparatido/uso terapéutico , Conservadores de la Densidad Ósea/farmacología , Conservadores de la Densidad Ósea/uso terapéutico , Ratas Sprague-Dawley , Proteína Relacionada con la Hormona Paratiroidea/farmacología , Proteína Relacionada con la Hormona Paratiroidea/uso terapéutico
2.
JCI Insight ; 5(8)2020 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-32208385

RESUMEN

Increased subchondral bone angiogenesis with blood vessels breaching the tidemark into the avascular cartilage is a diagnostic feature of human osteoarthritis. However, the mechanisms that initiate subchondral bone angiogenesis remain unclear. We show that abnormally increased platelet-derived growth factor-BB (PDGF-BB) secretion by mononuclear preosteoclasts induces subchondral bone angiogenesis, contributing to osteoarthritis development. In mice after destabilization of the medial meniscus (DMM), aberrant joint subchondral bone angiogenesis developed during an early stage of osteoarthritis, before articular cartilage damage occurred. Mononuclear preosteoclasts in subchondral bone secrete excessive amounts of PDGF-BB, which activates platelet-derived growth factor receptor-ß (PDGFR-ß) signaling in pericytes for neo-vessel formation. Selective knockout of PDGF-BB in preosteoclasts attenuates subchondral bone angiogenesis and abrogates joint degeneration and subchondral innervation induced by DMM. Transgenic mice that express PDGF-BB in preosteoclasts recapitulate pathological subchondral bone angiogenesis and develop joint degeneration and subchondral innervation spontaneously. Our study provides the first evidence to our knowledge that PDGF-BB derived from preosteoclasts is a key driver of pathological subchondral bone angiogenesis during osteoarthritis development and offers a new avenue for developing early treatments for this disease.


Asunto(s)
Becaplermina/metabolismo , Neovascularización Patológica/patología , Osteoartritis/patología , Osteoclastos/metabolismo , Animales , Huesos/metabolismo , Huesos/patología , Ratones , Ratones Endogámicos C57BL , Neovascularización Patológica/metabolismo , Osteoartritis/metabolismo , Osteoclastos/patología
3.
J Clin Invest ; 130(7): 3483-3498, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32191640

RESUMEN

The sensory nerve was recently identified as being involved in regulation of bone mass accrual. We previously discovered that prostaglandin E2 (PGE2) secreted by osteoblasts could activate sensory nerve EP4 receptor to promote bone formation by inhibiting sympathetic activity. However, the fundamental units of bone formation are active osteoblasts, which originate from mesenchymal stromal/stem cells (MSCs). Here, we found that after sensory denervation, knockout of the EP4 receptor in sensory nerves, or knockout of COX-2 in osteoblasts, could significantly promote adipogenesis and inhibit osteogenesis in adult mice. Furthermore, injection of SW033291 (a small molecule that locally increases the PGE2 level) or propranolol (a beta blocker) significantly promoted osteogenesis and inhibited adipogenesis. This effect of SW033291, but not propranolol, was abolished in conditional EP4-KO mice under normal conditions or in the bone repair process. We conclude that the PGE2/EP4 sensory nerve axis could regulate MSC differentiation in bone marrow of adult mice.


Asunto(s)
Adipogénesis , Dinoprostona/metabolismo , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Subtipo EP4 de Receptores de Prostaglandina E/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Ciclooxigenasa 2/metabolismo , Dinoprostona/genética , Técnicas de Inactivación de Genes , Células Madre Mesenquimatosas/patología , Ratones , Ratones Noqueados , Osteoblastos/metabolismo , Osteoblastos/patología , Subtipo EP4 de Receptores de Prostaglandina E/genética , Células Receptoras Sensoriales/patología
4.
J Bone Miner Res ; 35(6): 1188-1202, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32078184

RESUMEN

In the growing skeleton, angiogenesis is intimately coupled with osteogenesis. Chronic, high doses of glucocorticoids (GCs) are associated with decreased bone vasculature and induce osteoporosis and growth failure. The mechanism of GC-suppression of angiogenesis and relationship to osteoporosis and growth retardation remains largely unknown. Type H vessels, which are regulated by preosteoclast (POC) platelet-derived growth factor-BB (PDGF-BB), are specifically coupled with bone formation and development. We determined the effect of GCs on POC synthesis of PDGF-BB in relation to type H vessel formation, bone mass, and bone growth in the distal femur of 2-week-old young mice receiving prednisolone or vehicle for 2, 4, or 6 weeks. After 2 weeks of prednisolone, the number of POCs were unchanged while POC synthesis of PDGF-BB was reduced. Longer treatment with prednisolone reduced POCs numbers and PDGF-BB. These changes were associated with a reduction in type H vessels, bone formation rate, bone mass, and bone length at each time point. In vitro, excessive concentrations of prednisolone (10-6 M) resulted in decreased PDGF-BB concentration and POC numbers. Conditioned medium from POC cultures treated with control concentration of prednisolone (10-7 M) or recombinant PDGF-BB stimulated endothelial tube formation, whereas conditioned medium from control concentration of prednisolone-treated POC cultures neutralized by PDGF-BB antibody or excessive prednisolone inhibited endothelial tube formation. Administration of excessive prednisolone attenuated the P65 subunit of nuclear factor kappa B (NF-κB) binding to the Pdgfb promoter, resulting in lower Pdgfb transcription. Co-treatment with excessive prednisolone and the glucocorticoid receptor (GR) antagonist (RU486), GR siRNA, or TNFα rescued NF-κB binding to the Pdgfb promoter and endothelial tube formation. These results indicate that PDGF-BB synthesis in POCs is suppressed by GCs through transrepression of GR/NF-κB, thus inhibiting type H vessel formation and associated osteoporosis and growth failure. © 2020 American Society for Bone and Mineral Research.


Asunto(s)
Glucocorticoides , Osteoporosis , Proteínas Proto-Oncogénicas c-sis , Animales , Células Cultivadas , Glucocorticoides/farmacología , Ratones , FN-kappa B , Osteogénesis , Proteínas Proto-Oncogénicas c-sis/genética
5.
Bone Res ; 7: 29, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31666999

RESUMEN

Osteoarthritis (OA) causes the destruction of joints. Its pathogenesis is still under investigation, and there is no effective disease-modifying therapy. Here, we report that elevated cyclooxygenase-2 (COX-2) expression in the osteocytes of subchondral bone causes both spontaneous OA and rheumatoid arthritis (RA). The knockout of COX-2 in osteocytes or treatment with a COX-2 inhibitor effectively rescues the structure of subchondral bone and attenuates cartilage degeneration in spontaneous OA (STR/Ort) mice and tumor necrosis factor-α transgenic RA mice. Thus, elevated COX-2 expression in subchondral bone induces both OA-associated and RA-associated joint cartilage degeneration. The inhibition of COX-2 expression can potentially modify joint destruction in patients with arthritis.

6.
J Clin Invest ; 129(3): 1076-1093, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30530994

RESUMEN

Joint pain is the defining symptom of osteoarthritis (OA) but its origin and mechanisms remain unclear. Here, we investigated an unprecedented role of osteoclast-initiated subchondral bone remodeling in sensory innervation for OA pain. We show that osteoclasts secrete netrin-1 to induce sensory nerve axonal growth in subchondral bone. Reduction of osteoclast formation by knockout of receptor activator of nuclear factor kappa-B ligand (Rankl) in osteocytes inhibited the growth of sensory nerves into subchondral bone, dorsal root ganglion neuron hyperexcitability, and behavioral measures of pain hypersensitivity in OA mice. Moreover, we demonstrated a possible role for netrin-1 secreted by osteoclasts during aberrant subchondral bone remodeling in inducing sensory innervation and OA pain through its receptor DCC (deleted in colorectal cancer). Importantly, knockout of Netrin1 in tartrate-resistant acid phosphatase-positive (TRAP-positive) osteoclasts or knockdown of Dcc reduces OA pain behavior. In particular, inhibition of osteoclast activity by alendronate modifies aberrant subchondral bone remodeling and reduces innervation and pain behavior at the early stage of OA. These results suggest that intervention of the axonal guidance molecules (e.g., netrin-1) derived from aberrant subchondral bone remodeling may have therapeutic potential for OA pain.


Asunto(s)
Ganglios Espinales/metabolismo , Netrina-1/metabolismo , Osteoartritis/metabolismo , Osteoclastos/metabolismo , Dolor/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Remodelación Ósea/genética , Receptor DCC/genética , Receptor DCC/metabolismo , Ganglios Espinales/patología , Masculino , Ratones , Netrina-1/genética , Osteoartritis/genética , Osteoartritis/patología , Osteoclastos/patología , Dolor/genética , Dolor/patología , Células Receptoras Sensoriales/patología
7.
Biomaterials ; 181: 347-359, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30098570

RESUMEN

Peripheral nerve injuries often lead to incomplete recovery and contribute to significant disability to approximately 360,000 people in the USA each year. Stem cell therapy holds significant promise for peripheral nerve regeneration, but maintenance of stem cell viability and differentiation potential in vivo are still major obstacles for translation. Using a made-in-house 96-well vertical electrical stimulation (ES) platform, we investigated the effects of different stimulating pulse frequency, duration and field direction on human neural crest stem cell (NCSC) differentiation. We observed dendritic morphology with enhanced neuronal differentiation for NCSCs cultured on cathodes subject to 20 Hz, 100µs pulse at a potential gradient of 200 mV/mm. We further evaluated the effect of a novel cell-based therapy featuring optimized pulsatile ES of NCSCs for in vivo transplantation following peripheral nerve regeneration. 15 mm critical-sized sciatic nerve injuries were generated with subsequent surgical repair in sixty athymic nude rats. Injured animals were randomly assigned into five groups (N = 12 per group): blank control, ES, NCSC, NCSC + ES, and autologous nerve graft. The optimized ES was applied immediately after surgical repair for 1 h in ES and NCSC + ES groups. Recovery was assessed by behavioral (CatWalk gait analysis), wet muscle-mass, histomorphometric, and immunohistochemical analyses at either 6 or 12 weeks after surgery (N = 6 per group). Gastrocnemius muscle wet mass measurements in ES + NCSC group were comparable to autologous nerve transplantation and significantly higher than other groups (p < 0.05). Quantitative histomorphometric analysis and catwalk gait analysis showed similar improvements by ES on NCSCs (p < 0.05). A higher number of viable NCSCs was shown via immunochemical analysis, with higher Schwann cell (SC) differentiation in the NCSC + ES group compared to the NCSC group (p < 0.05). Overall, ES on NCSC transplantation significantly enhanced nerve regeneration after injury and repair, and was comparable to autograft treatment. Thus, ES can be a potent alternative to biochemical and physical cues for modulating stem cell survival and differentiation. This novel cell-based intervention presents an effective and safe approach for improved outcomes after peripheral nerve repair.


Asunto(s)
Regeneración Nerviosa/fisiología , Cresta Neural/citología , Traumatismos de los Nervios Periféricos/terapia , Animales , Diferenciación Celular/fisiología , Células Cultivadas , Estimulación Eléctrica/métodos , Humanos , Inmunohistoquímica , Células-Madre Neurales/citología , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Ratas , Trasplante de Células Madre/métodos
8.
Ann N Y Acad Sci ; 1433(1): 29-40, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30091466

RESUMEN

Camurati-Engelmann disease (CED) is a genetic bone-modeling disorder mainly caused by mutations in the gene that encodes transforming growth factor-ß1 (TGF-ß1). Symptoms of CED include bone pain, fractures, and dysplasia. Currently, effective therapies for bone fracture and dysplasia in CED are urgently needed. We have demonstrated that TGF-ß1 is a coupling factor for bone remodeling and is aberrantly activated in CED. Daily injection of TGF-ß type 1 receptor inhibitor (TßR1I) attenuated CED symptoms, but this systemic administration caused serious side effects. In this study, we created a conjugate linking TßR1I and alendronate, which delivered TßR1I specifically to bone. After weekly injection of the conjugate for 8 weeks, normal bone morphology and remodeling in CED mice was maintained with a minimum effective dose 700 times lower than TßR1I injection. Additionally, we found that the conjugate restored normal bone turnover by reducing the number of osteoblasts and osteoclasts, maintained a regular osteogenic microenvironment by regulating the formation of CD31 and Endomucin double-positive vessels, and preserved ordinary bone formation via inhibition of the migration of leptin-receptor-positive cells. Thus, targeting delivery of TßR1I to bone is a promising therapy for CED and other uncoupled bone remodeling disorders.


Asunto(s)
Remodelación Ósea/efectos de los fármacos , Síndrome de Camurati-Engelmann/tratamiento farmacológico , Receptor Tipo I de Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Alendronato/administración & dosificación , Alendronato/química , Animales , Remodelación Ósea/genética , Síndrome de Camurati-Engelmann/genética , Síndrome de Camurati-Engelmann/patología , Células Cultivadas , Modelos Animales de Enfermedad , Sistemas de Liberación de Medicamentos , Diseño de Fármacos , Células HeLa , Humanos , Masculino , Ratones , Ratones Mutantes , Ratones Transgénicos , Osteogénesis/efectos de los fármacos , Factor de Crecimiento Transformador beta1/genética
9.
Bone Res ; 6: 2, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29423331

RESUMEN

TGF-ß 1-3 are unique multi-functional growth factors that are only expressed in mammals, and mainly secreted and stored as a latent complex in the extracellular matrix (ECM). The biological functions of TGF-ß in adults can only be delivered after ligand activation, mostly in response to environmental perturbations. Although involved in multiple biological and pathological processes of the human body, the exact roles of TGF-ß in maintaining stem cells and tissue homeostasis have not been well-documented until recent advances, which delineate their functions in a given context. Our recent findings, along with data reported by others, have clearly shown that temporal and spatial activation of TGF-ß is involved in the recruitment of stem/progenitor cell participation in tissue regeneration/remodeling process, whereas sustained abnormalities in TGF-ß ligand activation, regardless of genetic or environmental origin, will inevitably disrupt the normal physiology and lead to pathobiology of major diseases. Modulation of TGF-ß signaling with different approaches has proven effective pre-clinically in the treatment of multiple pathologies such as sclerosis/fibrosis, tumor metastasis, osteoarthritis, and immune disorders. Thus, further elucidation of the mechanisms by which TGF-ß is activated in different tissues/organs and how targeted cells respond in a context-dependent way can likely be translated with clinical benefits in the management of a broad range of diseases with the involvement of TGF-ß.

10.
Nat Commun ; 9(1): 551, 2018 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-29416028

RESUMEN

Acquired heterotopic ossification (HO) is a painful and debilitating disease characterized by extraskeletal bone formation after injury. The exact pathogenesis of HO remains unknown. Here we show that TGF-ß initiates and promotes HO in mice. We find that calcified cartilage and newly formed bone resorb osteoclasts after onset of HO, which leads to high levels of active TGF-ß that recruit mesenchymal stromal/progenitor cells (MSPCs) in the HO microenvironment. Transgenic expression of active TGF-ß in tendon induces spontaneous HO, whereas systemic injection of a TGF-ß neutralizing antibody attenuates ectopic bone formation in traumatic and BMP-induced mouse HO models, and in a fibrodysplasia ossificans progressive mouse model. Moreover, inducible knockout of the TGF-ß type II receptor in MSPCs inhibits HO progression in HO mouse models. Our study points toward elevated levels of active TGF-ß as inducers and promoters of ectopic bone formation, and suggest that TGF-ß might be a therapeutic target in HO.


Asunto(s)
Osificación Heterotópica/metabolismo , Osteoclastos , Factor de Crecimiento Transformador beta/metabolismo , Tendón Calcáneo/efectos de los fármacos , Tendón Calcáneo/lesiones , Adulto , Animales , Anticuerpos Neutralizantes/farmacología , Becaplermina/metabolismo , Remodelación Ósea , Lesiones Traumáticas del Encéfalo , Cartílago , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Articulación del Codo/cirugía , Femenino , Fijación Interna de Fracturas , Fracturas Óseas , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Ratones Transgénicos , Persona de Mediana Edad , Músculo Esquelético/patología , Miositis Osificante/metabolismo , Osteogénesis/efectos de los fármacos , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Traumatismos de la Médula Espinal , Traumatismos de los Tendones , Tendones , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/metabolismo , Adulto Joven , Lesiones de Codo
11.
J Clin Invest ; 128(2): 846-860, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29355842

RESUMEN

Enthesopathy is a disorder of bone, tendon, or ligament insertion. It represents one-fourth of all tendon-ligament diseases and is one of the most difficult tendon-ligament disorders to treat. Despite its high prevalence, the exact pathogenesis of this condition remains unknown. Here, we show that TGF-ß was activated in both a semi-Achilles tendon transection (SMTS) mouse model and in a dorsiflexion immobilization (DI) mouse model of enthesopathy. High concentrations of active TGF-ß recruited mesenchymal stromal stem cells (MSCs) and led to excessive vessel formation, bone deterioration, and fibrocartilage calcification. Transgenic expression of active TGF-ß1 in bone also induced enthesopathy with a phenotype similar to that observed in SMTS and DI mice. Systemic inhibition of TGF-ß activity by injection of 1D11, a TGF-ß-neutralizing antibody, but not a vehicle antibody, attenuated the excessive vessel formation and restored uncoupled bone remodeling in SMTS mice. 1D11-treated SMTS fibrocartilage had increased proteoglycan and decreased collagen X and matrix metalloproteinase 13 expression relative to control antibody treatment. Notably, inducible knockout of the TGF-ß type II receptor in mouse MSCs preserved the bone microarchitecture and fibrocartilage composition after SMTS relative to the WT littermate controls. Thus, elevated levels of active TGF-ß in the enthesis bone marrow induce the initial pathological changes of enthesopathy, indicating that TGF-ß inhibition could be a potential therapeutic strategy.


Asunto(s)
Entesopatía/patología , Tendones/patología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Anticuerpos Neutralizantes/farmacología , Remodelación Ósea , Huesos/patología , Cartílago/patología , Colágeno/metabolismo , Modelos Animales de Enfermedad , Fibrocartílago/patología , Masculino , Células Madre Mesenquimatosas , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Osteoclastos/metabolismo , Fenotipo , Receptor Tipo II de Factor de Crecimiento Transformador beta/genética , Transducción de Señal , Tendones/metabolismo , Microtomografía por Rayos X
12.
Nat Commun ; 7: 11455, 2016 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-27126736

RESUMEN

Mesenchymal stem cells (MSCs) participate in the repair/remodelling of many tissues, where MSCs commit to different lineages dependent on the cues in the local microenvironment. Here we show that TGFß-activated RhoA/ROCK signalling functions as a molecular switch regarding the fate of MSCs in arterial repair/remodelling after injury. MSCs differentiate into myofibroblasts when RhoA/ROCK is turned on, endothelial cells when turned off. The former is pathophysiologic resulting in intimal hyperplasia, whereas the latter is physiological leading to endothelial repair. Further analysis revealed that MSC RhoA activation promotes formation of an extracellular matrix (ECM) complex consisting of connective tissue growth factor (CTGF) and vascular endothelial growth factor (VEGF). Inactivation of RhoA/ROCK in MSCs induces matrix metalloproteinase-3-mediated CTGF cleavage, resulting in VEGF release and MSC endothelial differentiation. Our findings uncover a novel mechanism by which cell-ECM interactions determine stem cell lineage specificity and offer additional molecular targets to manipulate MSC-involved tissue repair/regeneration.


Asunto(s)
Factor de Crecimiento del Tejido Conjuntivo/metabolismo , Células Endoteliales/metabolismo , Matriz Extracelular/metabolismo , Células Madre Mesenquimatosas/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo , Cicatrización de Heridas/genética , Proteínas de Unión al GTP rho/metabolismo , Animales , Diferenciación Celular , Factor de Crecimiento del Tejido Conjuntivo/genética , Células Endoteliales/citología , Matriz Extracelular/química , Arteria Femoral/lesiones , Arteria Femoral/metabolismo , Regulación de la Expresión Génica , Masculino , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Células Madre Mesenquimatosas/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miofibroblastos/metabolismo , Miofibroblastos/patología , Ratas , Ratas Sprague-Dawley , Regeneración/genética , Transducción de Señal , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor A de Crecimiento Endotelial Vascular/genética , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA
13.
Ann Rheum Dis ; 75(9): 1714-21, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-26470720

RESUMEN

OBJECTIVES: Examine whether osteoarthritis (OA) progression can be delayed by halofuginone in anterior cruciate ligament transection (ACLT) rodent models. METHODS: 3-month-old male C57BL/6J (wild type; WT) mice and Lewis rats were randomised to sham-operated, ACLT-operated, treated with vehicle, or ACLT-operated, treated with halofuginone. Articular cartilage degeneration was graded using the Osteoarthritis Research Society International (OARSI)-modified Mankin criteria. Immunostaining, flow cytometry, RT-PCR and western blot analyses were conducted to detect relative protein and RNA expression. Bone micro CT (µCT) and CT-based microangiography were quantitated to detect alterations of microarchitecture and vasculature in tibial subchondral bone. RESULTS: Halofuginone attenuated articular cartilage degeneration and subchondral bone deterioration, resulting in substantially lower OARSI scores. Specifically, we found that proteoglycan loss and calcification of articular cartilage were significantly decreased in halofuginone-treated ACLT rodents compared with vehicle-treated ACLT controls. Halofuginone reduced collagen X (Col X), matrix metalloproteinase-13 and A disintegrin and metalloproteinase with thrombospondin motifs 5 (ADAMTS 5) and increased lubricin, collagen II and aggrecan. In parallel, halofuginone-attenuated uncoupled subchondral bone remodelling as defined by reduced subchondral bone tissue volume, lower trabecular pattern factor (Tb.pf) and increased thickness of subchondral bone plate compared with vehicle-treated ACLT controls. We found that halofuginone exerted protective effects in part by suppressing Th17-induced osteoclastic bone resorption, inhibiting Smad2/3-dependent TGF-ß signalling to restore coupled bone remodelling and attenuating excessive angiogenesis in subchondral bone. CONCLUSIONS: Halofuginone attenuates OA progression by inhibition of subchondral bone TGF-ß activity and aberrant angiogenesis as a potential preventive therapy for OA.


Asunto(s)
Remodelación Ósea/efectos de los fármacos , Huesos/efectos de los fármacos , Osteoartritis/tratamiento farmacológico , Piperidinas/farmacología , Quinazolinonas/farmacología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Ligamento Cruzado Anterior/cirugía , Resorción Ósea/prevención & control , Huesos/metabolismo , Cartílago Articular/efectos de los fármacos , Cartílago Articular/metabolismo , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/efectos de los fármacos , Distribución Aleatoria , Ratas , Ratas Endogámicas Lew
14.
Nat Med ; 21(11): 1262-1271, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26457758

RESUMEN

Cancer-associated muscle weakness is a poorly understood phenomenon, and there is no effective treatment. Here we find that seven different mouse models of human osteolytic bone metastases-representing breast, lung and prostate cancers, as well as multiple myeloma-exhibited impaired muscle function, implicating a role for the tumor-bone microenvironment in cancer-associated muscle weakness. We found that transforming growth factor (TGF)-ß, released from the bone surface as a result of metastasis-induced bone destruction, upregulated NADPH oxidase 4 (Nox4), resulting in elevated oxidization of skeletal muscle proteins, including the ryanodine receptor and calcium (Ca(2+)) release channel (RyR1). The oxidized RyR1 channels leaked Ca(2+), resulting in lower intracellular signaling, which is required for proper muscle contraction. We found that inhibiting RyR1 leakage, TGF-ß signaling, TGF-ß release from bone or Nox4 activity improved muscle function in mice with MDA-MB-231 bone metastases. Humans with breast- or lung cancer-associated bone metastases also had oxidized skeletal muscle RyR1 that is not seen in normal muscle. Similarly, skeletal muscle weakness, increased Nox4 binding to RyR1 and oxidation of RyR1 were present in a mouse model of Camurati-Engelmann disease, a nonmalignant metabolic bone disorder associated with increased TGF-ß activity. Thus, pathological TGF-ß release from bone contributes to muscle weakness by decreasing Ca(2+)-induced muscle force production.


Asunto(s)
Neoplasias Óseas/metabolismo , Calcio/metabolismo , Debilidad Muscular/metabolismo , Músculo Esquelético/metabolismo , Neoplasias/metabolismo , Osteólisis/metabolismo , Canal Liberador de Calcio Receptor de Rianodina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Absorciometría de Fotón , Animales , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/secundario , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Señalización del Calcio , Síndrome de Camurati-Engelmann/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Células MCF-7 , Masculino , Ratones , Ratones Desnudos , Ratones SCID , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Contracción Muscular , Proteínas Musculares/metabolismo , Fuerza Muscular , Debilidad Muscular/etiología , NADPH Oxidasa 4 , NADPH Oxidasas/genética , NADPH Oxidasas/metabolismo , Neoplasias/complicaciones , Neoplasias/patología , Osteólisis/diagnóstico por imagen , Osteólisis/etiología , Oxidación-Reducción , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Regulación hacia Arriba , Microtomografía por Rayos X
15.
J Bone Miner Res ; 30(11): 2033-43, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25967237

RESUMEN

Rheumatoid arthritis (RA) is an autoimmune disease that often leads to joint destruction. A myriad of drugs targeting the immune abnormalities and downstream inflammatory cascades have been developed, but the joint destruction is not effectively halted. Here we report that aberrant activation of TGF-ß in the subchondral bone marrow by immune response increases osteoprogenitors and uncoupled bone resorption and formation in RA mouse/rat models. Importantly, either systemic or local blockade of TGF-ß activity in the subchondral bone attenuated articular cartilage degeneration in RA. Moreover, conditional deletion of TGF-ß receptor II (Tgfbr2) in nestin-positive cells also effectively halted progression of RA joint destruction. Our data demonstrate that aberrant activation of TGF-ß in the subchondral bone is involved at the onset of RA joint cartilage degeneration. Thus, modulation of subchondral bone TGF-ß activity could be a potential therapy for RA joint destruction.


Asunto(s)
Artritis Reumatoide/patología , Huesos/patología , Articulaciones/patología , Factor de Crecimiento Transformador beta/metabolismo , Animales , Artritis Reumatoide/metabolismo , Médula Ósea/patología , Resorción Ósea/patología , Huesos/metabolismo , Cartílago Articular/metabolismo , Cartílago Articular/patología , Colágeno , Modelos Animales de Enfermedad , Articulaciones/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Nestina/metabolismo , Pruebas de Neutralización , Osteogénesis , Ratas Endogámicas Lew , Transducción de Señal , Tibia/patología
16.
Nat Med ; 20(11): 1270-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25282358

RESUMEN

Osteogenesis during bone modeling and remodeling is coupled with angiogenesis. A recent study showed that a specific vessel subtype, strongly positive for CD31 and endomucin (CD31(hi)Emcn(hi)), couples angiogenesis and osteogenesis. Here, we found that platelet-derived growth factor-BB (PDGF-BB) secreted by preosteoclasts induces CD31(hi)Emcn(hi) vessel formation during bone modeling and remodeling. Mice with depletion of PDGF-BB in the tartrate-resistant acid phosphatase-positive cell lineage show significantly lower trabecular and cortical bone mass, serum and bone marrow PDGF-BB concentrations, and fewer CD31(hi)Emcn(hi) vessels compared to wild-type mice. In the ovariectomy (OVX)-induced osteoporotic mouse model, serum and bone marrow levels of PDGF-BB and numbers of CD31(hi)Emcn(hi) vessels are significantly lower compared to sham-operated controls. Treatment with exogenous PDGF-BB or inhibition of cathepsin K to increase the number of preosteoclasts, and thus the endogenous levels of PDGF-BB, increases CD31(hi)Emcn(hi) vessel number and stimulates bone formation in OVX mice. Thus, pharmacotherapies that increase PDGF-BB secretion from preosteoclasts offer a new therapeutic target for treating osteoporosis by promoting angiogenesis and thus bone formation.


Asunto(s)
Neovascularización Fisiológica , Osteoclastos/metabolismo , Osteogénesis , Proteínas Proto-Oncogénicas c-sis/metabolismo , Fosfatasa Ácida/metabolismo , Animales , Becaplermina , Catepsina K/antagonistas & inhibidores , Catepsina K/metabolismo , Recuento de Células , Movimiento Celular/efectos de los fármacos , Medios de Cultivo Condicionados/farmacología , Células Endoteliales/citología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Fémur/diagnóstico por imagen , Fémur/efectos de los fármacos , Fémur/metabolismo , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Isoenzimas/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Ratones Endogámicos C57BL , Neovascularización Fisiológica/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoclastos/enzimología , Osteogénesis/efectos de los fármacos , Ovariectomía , Fosforilación/efectos de los fármacos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Inhibidores de Proteasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatasa Ácida Tartratorresistente , Microtomografía por Rayos X
17.
J Bone Miner Res ; 29(5): 1141-57, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24166835

RESUMEN

Altered bone turnover is a key pathologic feature of chronic kidney disease-mineral and bone disorder (CKD-MBD). Expression of TGF-ß1, a known regulator of bone turnover, is increased in bone biopsies from individuals with CKD. Similarly, TGF-ß1 mRNA and downstream signaling is increased in bones from jck mice, a model of high-turnover renal osteodystrophy. A neutralizing anti-TGF-ß antibody (1D11) was used to explore TGF-ß's role in renal osteodystrophy. 1D11 administration to jck significantly attenuated elevated serum osteocalcin and type I collagen C-telopeptides. Histomorphometric analysis indicated that 1D11 administration increased bone volume and suppressed the elevated bone turnover in a dose-dependent manner. These effects were associated with reductions in osteoblast and osteoclast surface areas. Micro-computed tomography (µCT) confirmed the observed increase in trabecular bone volume and demonstrated improvements in trabecular architecture and increased cortical thickness. 1D11 administration was associated with significant reductions in expression of osteoblast marker genes (Runx2, alkaline phosphatase, osteocalcin) and the osteoclast marker gene, Trap5. Importantly, in this model, 1D11 did not improve kidney function or reduce serum parathyroid hormone (PTH) levels, indicating that 1D11 effects on bone are independent of changes in renal or parathyroid function. 1D11 also significantly attenuated high-turnover bone disease in the adenine-induced uremic rat model. Antibody administration was associated with a reduction in pSMAD2/SMAD2 in bone but not bone marrow as assessed by quantitative immunoblot analysis. Immunostaining revealed pSMAD staining in osteoblasts and osteocytes but not osteoclasts, suggesting 1D11 effects on osteoclasts may be indirect. Immunoblot and whole genome mRNA expression analysis confirmed our previous observation that repression of Wnt/ß-catenin expression in bone is correlated with increased osteoclast activity in jck mice and bone biopsies from CKD patients. Furthermore, our data suggest that elevated TGF-ß may contribute to the pathogenesis of high-turnover disease partially through inhibition of ß-catenin signaling.


Asunto(s)
Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/metabolismo , Osteoclastos/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Vía de Señalización Wnt , Animales , Anticuerpos Neutralizantes/farmacología , Antígenos de Diferenciación/metabolismo , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/diagnóstico por imagen , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/genética , Trastorno Mineral y Óseo Asociado a la Enfermedad Renal Crónica/patología , Colágeno Tipo I , Modelos Animales de Enfermedad , Masculino , Ratones , Osteocalcina/metabolismo , Osteoclastos/patología , Péptidos , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta1/genética , Microtomografía por Rayos X
18.
J Exp Stroke Transl Med ; 6: 7-17, 2013 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-24285991

RESUMEN

Although acute ischemic stroke has high mortality and morbidity rate but yet still has very limited treatment. In this study we have tested the concept of neuron protection by acute bioenergetic intervention or by pharmacological preconditioning with natural antioxidants. Adenosine triphosphate (ATP), pentobarbital, and suramin were encapsulated in pH-sensitive liposomes and used as bioenergy stabilizer. We induced ATP depletion model by incubating cells with media added with ATP-depleting agents for 2 hours. Treatment with bioenergy stabilizer started 10-min post inducing of ATP-depletion. The acute treatment with bioenergy stabilizer significantly increased cell viability in neuro-2a cells. In searching for a pharmacological preconditioning candidate for reducing ischemic injury, we tested cocoa-derived flavanols using bilateral common carotid artery occlusion (BCCAO). We pretreated mice with cocoa-derived flavanols (75 mg/kg) or water orally for 7 days and subjected mice for 12 minutes BCCAO. At 7 days post-ischemia, the number of surviving hippocampal CA1 neurons was significantly higher in the treated mice than in the water-treated controls. The protection from cocoa-derived flavanols was found associated with increased total antioxidant capacity in the brain. Our results indicate that for reducing acute ischemic injury bioenergetic intervention using advanced drug delivery tools is conceptually feasible, and for reducing reperfusion related secondary injury pharmacological preconditioning may provide significant protection.

19.
Int J Mol Sci ; 14(6): 11626-42, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23727934

RESUMEN

In the last decade, great advances have been made in epidermal stem cell studies at the cellular and molecular level. These studies reported various subpopulations and differentiations existing in the epidermal stem cell. Although controversies and unknown issues remain, epidermal stem cells possess an immune-privileged property in transplantation together with easy accessibility, which is favorable for future clinical application. In this review, we will summarize the biological characteristics of epidermal stem cells, and their potential in orthopedic regenerative medicine. Epidermal stem cells play a critical role via cell replacement, and demonstrate significant translational potential in the treatment of orthopedic injuries and diseases, including treatment for wound healing, peripheral nerve and spinal cord injury, and even muscle and bone remodeling.


Asunto(s)
Células Epidérmicas , Ortopedia , Medicina Regenerativa/métodos , Células Madre/citología , Animales , Humanos , Traumatismos de la Médula Espinal/terapia , Cicatrización de Heridas
20.
Nat Med ; 19(6): 704-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23685840

RESUMEN

Osteoarthritis is a highly prevalent and debilitating joint disorder. There is no effective medical therapy for the condition because of limited understanding of its pathogenesis. We show that transforming growth factor ß1 (TGF-ß1) is activated in subchondral bone in response to altered mechanical loading in an anterior cruciate ligament transection (ACLT) mouse model of osteoarthritis. TGF-ß1 concentrations are also high in subchondral bone from humans with osteoarthritis. High concentrations of TGF-ß1 induced formation of nestin-positive mesenchymal stem cell (MSC) clusters, leading to formation of marrow osteoid islets accompanied by high levels of angiogenesis. We found that transgenic expression of active TGF-ß1 in osteoblastic cells induced osteoarthritis, whereas inhibition of TGF-ß activity in subchondral bone attenuated the degeneration of articular cartilage. In particular, knockout of the TGF-ß type II receptor (TßRII) in nestin-positive MSCs led to less development of osteoarthritis relative to wild-type mice after ACLT. Thus, high concentrations of active TGF-ß1 in subchondral bone seem to initiate the pathological changes of osteoarthritis, and inhibition of this process could be a potential therapeutic approach to treating this disease.


Asunto(s)
Huesos/metabolismo , Células Madre Mesenquimatosas/fisiología , Osteoartritis/terapia , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Animales , Cartílago/patología , Células Cultivadas , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Ratas , Ratas Endogámicas Lew , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA