Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Biol Rep ; 51(1): 587, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38683225

RESUMEN

BACKGROUND: Patients with multiple myeloma exhibit malignant osteolytic bone disease due to excessive osteoclast formation and function. We recently identified that osteoclastogenic stimulator selenoprotein W (SELENOW) is upregulated via ERK signaling and downregulated via p38 signaling during receptor activator of nuclear factor (NF)-κΒ ligand (RANKL)-induced osteoclast differentiation. In the intrinsic physiological process, RANKL-induced downregulation of SELENOW maintains proper osteoclast differentiation; in contrast, forced overexpression of SELENOW leads to overactive osteoclast formation and function. METHODS AND RESULTS: We observed that SELENOW is highly expressed in multiple myeloma-derived peripheral blood mononuclear cells (PBMCs) and mature osteoclasts when compared to healthy controls. Also, the level of tumor necrosis factor alpha (TNFα), a pathological osteoclastogenic factor, is increased in the PBMCs and serum of patients with multiple myeloma. ERK activation by TNFα was more marked and sustained than that by RANKL, allowing SELENOW upregulation. Excessive expression of SELENOW in osteoclast progenitors and mature osteoclasts derived from multiple myeloma facilitated efficient nuclear translocation of osteoclastogenic transcription factors NF-κB and NFATc1, which are favorable for osteoclast formation. CONCLUSION: Our findings suggest a possibility that feedforward signaling of osteoclastogenic SELENOW by TNFα derived from multiple myeloma induces overactive osteoclast differentiation, leading to bone loss during multiple myeloma.


Asunto(s)
Diferenciación Celular , Mieloma Múltiple , Osteoclastos , Selenoproteína W , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Diferenciación Celular/genética , Leucocitos Mononucleares/metabolismo , Sistema de Señalización de MAP Quinasas , Mieloma Múltiple/metabolismo , Mieloma Múltiple/patología , Mieloma Múltiple/genética , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Factores de Transcripción NFATC/genética , Osteoclastos/metabolismo , Ligando RANK/metabolismo , Selenoproteína W/metabolismo , Selenoproteína W/genética , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
2.
Int J Mol Sci ; 20(23)2019 Dec 02.
Artículo en Inglés | MEDLINE | ID: mdl-31810213

RESUMEN

Mollusks have served as important sources of human food and medicine for a long time. Raw Pisidium coreanum, a freshwater bivalve of the phylum Mollusca, is used in traditional therapies in parts of Asia. However, the therapeutic effects of Pisidium coreanum on bone diseases are not known. We investigated the functional roles of Pisidium coreanum in osteoporotic bone diseases. Pisidium coreanum inhibited the differentiation of bone marrow-derived monocytic cells into mature osteoclasts in vitro. The ovariectomized mice that received oral administration of Pisidium coreanum showed improvements in both trabecular and cortical bones. This preventive activity of Pisidium coreanum against bone loss was due to limited osteoclast maturation with reduced osteoclast surface extent in trabecular bone tissue. The formation of large multinucleated osteoclasts in vitro was significantly decreased in response to Pisidium coreanum, consistent with the reduced expression levels of osteoclast markers and fusion-related genes, such as NFATc1, p65, integrin αvß3, DC-STAMP, OC-STAMP, Atp6v0d2, FAK, CD44, and MFR. These data suggest that Pisidium coreanum inhibits osteoclast differentiation by negatively regulating the fusion of mononuclear osteoclast precursors. Thus, our data demonstrate the ability of Pisidium coreanum to effectively prevent estrogen-deficient osteoporosis through inhibition of multinucleated osteoclast formation.


Asunto(s)
Bivalvos , Enfermedades Óseas/dietoterapia , Estrógenos/deficiencia , Osteoporosis/dietoterapia , Animales , Enfermedades Óseas/metabolismo , Enfermedades Óseas/fisiopatología , Resorción Ósea/dietoterapia , Resorción Ósea/prevención & control , Diferenciación Celular/efectos de los fármacos , Humanos , Ratones , Osteoclastos/efectos de los fármacos , Osteoporosis/metabolismo , Alimentos Marinos/análisis
3.
Int J Mol Sci ; 19(10)2018 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-30275408

RESUMEN

Bone undergoes continuous remodeling, which is homeostatically regulated by concerted communication between bone-forming osteoblasts and bone-degrading osteoclasts. Multinucleated giant osteoclasts are the only specialized cells that degrade or resorb the organic and inorganic bone components. They secrete proteases (e.g., cathepsin K) that degrade the organic collagenous matrix and establish localized acidosis at the bone-resorbing site through proton-pumping to facilitate the dissolution of inorganic mineral. Osteoporosis, the most common bone disease, is caused by excessive bone resorption, highlighting the crucial role of osteoclasts in intact bone remodeling. Signaling mediated by mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38, has been recognized to be critical for normal osteoclast differentiation and activation. Various exogenous (e.g., toll-like receptor agonists) and endogenous (e.g., growth factors and inflammatory cytokines) stimuli contribute to determining whether MAPKs positively or negatively regulate osteoclast adhesion, migration, fusion and survival, and osteoclastic bone resorption. In this review, we delineate the unique roles of MAPKs in osteoclast metabolism and provide an overview of the upstream regulators that activate or inhibit MAPKs and their downstream targets. Furthermore, we discuss the current knowledge about the differential kinetics of ERK, JNK, and p38, and the crosstalk between MAPKs in osteoclast metabolism.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoclastos/enzimología , Animales , Humanos , Cinética , Sistema de Señalización de MAP Quinasas , Modelos Biológicos
4.
Int J Mol Sci ; 18(10)2017 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-28946669

RESUMEN

Anti-osteoporotic activity of a blocker of the ubiquitin-proteasome system, bortezomib, has known to be achieved by directly opposed action in increased bone formation by osteoblasts and in decreased bone destruction by osteoclasts. However, the mechanisms underlying the proteasome blocker inhibition of osteoclast differentiation and function are not fully understood. Here, we observed that proteasome inhibitors, such as MG132 and bortezomib, in osteoclasts accelerated the degradation of c-Fms, a cognate receptor of macrophage colony-stimulating factor (M-CSF), and did not affect the amount of receptor activator of nuclear factor kappa-B (RANK), a receptor of receptor activator of nuclear factor kappa-B ligand (RANKL). c-Fms degradation induced by proteasome inhibitors was controlled by the activation of p38/tumor necrosis factor-alpha converting enzyme (TACE)-mediated regulated intramembrane proteolysis (RIPping). This was validated through the restoration of c-Fms using specific inhibitors of p38 and TACE, and a stimulation of p38-dependent TACE. In addition, c-Fms degradation by proteasome inhibition completely blocked M-CSF-mediated intrinsic signalling and led to the suppression of osteoclast differentiation and bone resorption. In a mouse model with intraperitoneal administration of lipopolysaccharide (LPS) that stimulates osteoclast formation and leads to bone loss, proteasome blockers prevented LPS-induced inflammatory bone resorption due to a decrease in the number of c-Fms-positive osteoclasts. Our study showed that accelerating c-Fms proteolysis by proteasome inhibitors may be a therapeutic option for inflammation-induced bone loss.


Asunto(s)
Resorción Ósea/etiología , Resorción Ósea/metabolismo , Inflamación/complicaciones , Osteoclastos/citología , Osteoclastos/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Ubiquitina/metabolismo , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Resorción Ósea/patología , Resorción Ósea/prevención & control , Bortezomib/farmacología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Humanos , Factor Estimulante de Colonias de Macrófagos/metabolismo , Masculino , Ratones , Inhibidores de Proteasoma/farmacología , Proteolisis , Receptor de Factor Estimulante de Colonias de Macrófagos/genética
5.
EMBO J ; 31(5): 1147-59, 2012 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-22227519

RESUMEN

RASSF2 belongs to the Ras-association domain family (RASSF) of proteins, which may be involved in the Hippo signalling pathway. However, the role of RASSF2 in vivo is unknown. Here, we show that Rassf2 knockout mice manifest a multisystemic phenotype including haematopoietic anomalies and defects in bone remodelling. Bone marrow (BM) transplantation showed that Rassf2(-/-) BM cells had a normal haematopoietic reconstitution activity, indicating no intrinsic haematopoietic defects. Notably, in vitro differentiation studies revealed that ablation of Rassf2 suppressed osteoblastogenesis but promoted osteoclastogenesis. Co-culture experiments showed that an intrinsic defect in osteoblast differentiation from Rassf2(-/-) osteoblast precursors likely leads to both haematopoiesis and osteoclast defects in Rassf2(-/-) mice. Moreover, Rassf2 deficiency resulted in hyperactivation of nuclear factor (NF)-κB during both osteoclast and osteoblast differentiation. RASSF2 associated with IκB kinase (IKK) α and ß forms, and suppressed IKK activity. Introduction of either RASSF2 or a dominant-negative form of IKK into Rassf2(-/-) osteoclast or osteoblast precursors inhibited NF-κB hyperactivation and normalized osteoclast and osteoblast differentiation. These observations indicate that RASSF2 regulates osteoblast and osteoclast differentiation by inhibiting NF-κB signalling.


Asunto(s)
Hematopoyesis , Quinasa I-kappa B/metabolismo , Osteoblastos/fisiología , Osteoclastos/fisiología , Proteínas Supresoras de Tumor/metabolismo , Animales , Resorción Ósea , Diferenciación Celular , Proliferación Celular , Quinasa I-kappa B/antagonistas & inhibidores , Ratones , Ratones Noqueados , FN-kappa B/biosíntesis , Osteogénesis , Unión Proteica , Mapeo de Interacción de Proteínas , Proteínas Supresoras de Tumor/deficiencia
6.
Int J Mol Sci ; 17(8)2016 Aug 09.
Artículo en Inglés | MEDLINE | ID: mdl-27517906

RESUMEN

Osteoclast progenitors undergo cell cycle arrest before differentiation into osteoclasts, induced by exposure to macrophage colony-stimulating factor (M-CSF) and receptor activator of nuclear factor-κB ligand (RANKL). The role of such cell cycle arrest in osteoclast differentiation has remained unclear, however. We here examined the effect of synchronized cell cycle arrest on osteoclast formation. Osteoclast progenitors deprived of M-CSF in culture adopted a uniform morphology and exhibited cell cycle arrest at the G0-G1 phase in association with both down-regulation of cyclins A and D1 as well as up-regulation of the cyclin-dependent kinase inhibitor p27(Kip1). Such M-CSF deprivation also promoted the differentiation of osteoclast progenitors into multinucleated osteoclasts expressing high levels of osteoclast marker proteins such as NFATc1, c-Fos, Atp6v0d2, cathepsin K, and integrin ß3 on subsequent exposure to M-CSF and RANKL. Our results suggest that synchronized arrest and reprogramming of osteoclast progenitors renders them poised to respond to inducers of osteoclast formation. Further characterization of such effects may facilitate induction of the differentiation of heterogeneous and multipotent cells into desired cell lineages.


Asunto(s)
Osteoclastos/citología , Animales , Catepsina K/metabolismo , Puntos de Control del Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Células Cultivadas , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Immunoblotting , Integrina beta3/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Ratones , Factores de Transcripción NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/fisiología , Proteínas Proto-Oncogénicas c-fos/metabolismo , Ligando RANK/metabolismo , ATPasas de Translocación de Protón Vacuolares/metabolismo
7.
J Immunol ; 190(4): 1623-30, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23303667

RESUMEN

Bone mineralization is a normal physiological process, whereas ectopic calcification of soft tissues is a pathological process that leads to irreversible tissue damage. We have established a coxsackievirus B3 (CVB3)-infected mouse model that manifests both osteoporosis and ectopic calcification specifically in heart, pancreas, and lung. The CVB3-infected mice showed increased serum concentrations of both cytokines including IL-1ß, TNF-α, and the receptor activator of NF-κB ligand (RANKL) that stimulate osteoclast formation and of the osteoclast-derived protein tartrate-resistant acid phosphatase 5b. They exhibited more osteoclasts in bone, with no change in the number of osteoblasts, and a decrease in bone formation and the serum concentration of osteoblast-produced osteocalcin. These results indicate that CVB3-induced osteoporosis is likely due to upregulation of osteoclast formation and function, in addition to decreased osteoblast activity. In addition, the serum in the CVB3-infected mice contained a high inorganic phosphate content, which causes ectopic calcification. RANKL treatment induced an increase in the in vitro cardiac fibroblast calcification by inorganic phosphate via the upregulation of osteogenic BMP2, SPARC, Runx2, Fra-1, and NF-κB signaling. We finally observed that i.p. administration of RANK-Fc, a recombinant antagonist of RANKL, prevented bone loss as well as ectopic calcification in CVB3-infected mice. Thus, our results indicate that RANKL may contribute to both abnormal calcium deposition in soft tissues and calcium depletion in bone. In addition, our animal model should provide a tool for the development of new therapeutic agents for calcium disturbance in soft and hard tissues.


Asunto(s)
Calcinosis/prevención & control , Infecciones por Coxsackievirus/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporosis/metabolismo , Osteoporosis/prevención & control , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Animales , Calcinosis/patología , Calcinosis/virología , Infecciones por Coxsackievirus/inmunología , Infecciones por Coxsackievirus/patología , Modelos Animales de Enfermedad , Células HeLa , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Osificación Heterotópica/metabolismo , Osificación Heterotópica/patología , Osificación Heterotópica/virología , Osteoblastos/patología , Osteoblastos/virología , Osteoclastos/patología , Osteoclastos/virología , Osteoporosis/virología , Ligando RANK/antagonistas & inhibidores , Ratas , Ratas Sprague-Dawley , Receptor Activador del Factor Nuclear kappa-B/administración & dosificación , Proteínas Recombinantes de Fusión/administración & dosificación , Proteínas Recombinantes de Fusión/genética
8.
Int J Mol Sci ; 16(12): 29305-14, 2015 Dec 09.
Artículo en Inglés | MEDLINE | ID: mdl-26690137

RESUMEN

Small G-protein adenosine diphosphate (ADP)-ribosylation factors (ARFs) regulate a variety of cellular functions, including actin cytoskeleton remodeling, plasma membrane reorganization, and vesicular transport. Here, we propose the functional roles of ARF1 in multiple stages of osteoclast differentiation. ARF1 was upregulated during receptor activator of nuclear factor kappa-B ligand (RANKL)-induced osteoclast differentiation and transiently activated in an initial stage of their differentiation. Differentiation of ARF1-deficient osteoclast precursors into mature osteoclasts temporarily increased in pre-maturation stage of osteoclasts followed by reduced formation of mature osteoclasts, indicating that ARF1 regulates the osteoclastogenic process. ARF1 deficiency resulted in reduced osteoclast precursor proliferation and migration as well as increasing cell-cell fusion. In addition, ARF1 silencing downregulated c-Jun N-terminal kinase (JNK), Akt, osteopontin, and macrophage colony-stimulating factor (M-CSF)-receptor c-Fms as well as upregulating several fusion-related genes including CD44, CD47, E-cadherin, and meltrin-α. Collectively, we showed that ARF1 stimulated proliferation and migration of osteoclast precursors while suppressing their fusion, suggesting that ARF1 may be a plausible inter-player that mediates the transition to osteoclast fusion at multiple steps during osteoclast differentiation.


Asunto(s)
Factor 1 de Ribosilacion-ADP/fisiología , Diferenciación Celular , Movimiento Celular , Proliferación Celular , Osteoclastos/fisiología , Animales , Fusión Celular , Células Cultivadas , Masculino , Ratones Endogámicos C57BL , Transducción de Señal
9.
Biochem Biophys Res Commun ; 453(4): 793-7, 2014 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-25445591

RESUMEN

Vascular calcification is an active cell-mediated process that reduces elasticity of blood vessels and increases blood pressure. Until now, the molecular basis of vascular calcification has not been fully understood. We previously reported that microtubule disturbances mediate vascular calcification. Here, we found that protein kinase C (PKC) signaling acted as a novel coordinator between cytoskeletal changes and hyperphosphatemia-induced vascular calcification. Phosphorylation and expression of both PKCα and PKCδ decreased during inorganic phosphate (Pi)-induced vascular smooth muscle cell (VSMC) calcification. Knockdown of PKC isoforms by short interfering RNA as well as PKC inactivation by Go6976 or rottlerin treatment revealed that specific inhibition of PKCα and PKCδ accelerated Pi-induced calcification both in VSMCs and ex vivo aorta culture through upregulation of osteogenic signaling. Additionally, inhibition of PKCα and PKCδ induced disassembly of microtubule and actin, respectively. In summary, our results indicate that cytoskeleton perturbation via PKCα and PKCδ inactivation potentiates vascular calcification through osteogenic signal induction.


Asunto(s)
Aorta/enzimología , Calcinosis/enzimología , Citoesqueleto/metabolismo , Osteogénesis , Enfermedades Vasculares Periféricas/enzimología , Proteína Quinasa C/metabolismo , Animales , Aorta/patología , Calcinosis/patología , Células Cultivadas , Citoesqueleto/patología , Regulación de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/patología , Proteína Quinasa C-delta/metabolismo
10.
Biochem Biophys Res Commun ; 451(3): 436-41, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25111813

RESUMEN

Vascular calcification is a strong predictor of cardiovascular morbidity and mortality, especially in individuals with chronic kidney disease or diabetes. The mechanism of vascular calcification has remained unclear, however, and no effective therapy is currently available. Our study was aimed at identifying the role of dynamic remodeling of microtubule cytoskeletons in hyperphosphatemia-induced vascular calcification. Exposure of primary cultures of mouse vascular smooth muscle cells (VSMCs) to inorganic phosphate (Pi) elicited ectopic calcification that was associated with changes in tubulin dynamics, induction of osteogenic signaling, and increased release of matrix vesicles. A microtubule depolymerizing agent enhanced Pi-dependent calcification, whereas microtubule stabilization by paclitaxel suppressed calcification both in VSMC cultures and in an ex vivo culture system for the mouse aorta. The inhibition of Pi-stimulated calcification by paclitaxel was associated with down-regulation of osteogenic signal and attenuation of matrix vesicle release. Our results indicate that microtubule plays a central role in vascular calcification, and that microtubule stabilization represents a potential new approach to the treatment of this condition.


Asunto(s)
Microtúbulos/metabolismo , Osteogénesis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Calcificación Vascular/prevención & control , Animales , Calcinosis/prevención & control , Citoesqueleto/fisiología , Masculino , Ratones , Músculo Liso Vascular/metabolismo , Osteogénesis/genética , Paclitaxel/farmacología , Fosfatos/metabolismo , Regulación hacia Arriba
11.
Biochem Biophys Res Commun ; 432(2): 378-83, 2013 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-23357417

RESUMEN

Ectopic mineralization of soft tissues is known to be a typical response to systemic imbalance of various metabolic factors as well as tissue injury, leading to severe clinical consequences. In this study, coxsackievirus B3 (CVB3) infection in mice resulted in significant tissue injury, especially in the heart and pancreas. Inflammatory damage and apoptotic cell death were observed in CVB3-infected heart and pancreas tissues. Along with tissue damage, substantial ectopic calcification was detected in CVB3-infected heart, pancreas, and lung tissues, as determined by von Kossa staining and calcium content quantification. In addition, CVB3 infection induced upregulation of osteogenic signals, including six genes (BMP2, SPARC, Runx2, osteopontin, collagen type I, and osterix) in the heart, three genes (SPARC, osteopontin, and collagen type I) in the pancreas, and two genes (BMP2 and alkaline phosphatase) in the lung, as determined by quantitative real-time PCR analysis. Intriguingly, we showed that α-lipoic acid diminished CVB3-mediated inflammatory and apoptotic tissue damage, subsequently ameliorating ectopic calcification via the suppression of osteogenic signals. Collectively, our data provide evidence that ectopic calcification induced by CVB3 infection is implicated in the induction of osteogenic propensity, and α-lipoic acid may be a potential therapeutic agent to ameliorate pathologic calcification.


Asunto(s)
Calcinosis/prevención & control , Infecciones por Coxsackievirus/tratamiento farmacológico , Enterovirus Humano B , Ácido Tióctico/uso terapéutico , Animales , Calcinosis/patología , Calcinosis/virología , Infecciones por Coxsackievirus/complicaciones , Infecciones por Coxsackievirus/patología , Femenino , Corazón/virología , Humanos , Pulmón/patología , Pulmón/virología , Masculino , Ratones , Páncreas/patología , Páncreas/virología
12.
Nat Med ; 12(12): 1403-9, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17128270

RESUMEN

Matrix-producing osteoblasts and bone-resorbing osteoclasts maintain bone homeostasis. Osteoclasts are multinucleated, giant cells of hematopoietic origin formed by the fusion of mononuclear pre-osteoclasts derived from myeloid cells. Fusion-mediated giant cell formation is critical for osteoclast maturation; without it, bone resorption is inefficient. To understand how osteoclasts differ from other myeloid lineage cells, we previously compared global mRNA expression patterns in these cells and identified genes of unknown function predominantly expressed in osteoclasts, one of which is the d2 isoform of vacuolar (H(+)) ATPase (v-ATPase) V(0) domain (Atp6v0d2). Here we show that inactivation of Atp6v0d2 in mice results in markedly increased bone mass due to defective osteoclasts and enhanced bone formation. Atp6v0d2 deficiency did not affect differentiation or the v-ATPase activity of osteoclasts. Rather, Atp6v0d2 was required for efficient pre-osteoclast fusion. Increased bone formation was probably due to osteoblast-extrinsic factors, as Atp6v02 was not expressed in osteoblasts and their differentiation ex vivo was not altered in the absence of Atp6v02. Our results identify Atp6v0d2 as a regulator of osteoclast fusion and bone formation, and provide genetic data showing that it is possible to simultaneously inhibit osteoclast maturation and stimulate bone formation by therapeutically targeting the function of a single gene.


Asunto(s)
Osteoclastos/fisiología , Osteogénesis/fisiología , Bombas de Protones/genética , ATPasas de Translocación de Protón Vacuolares/fisiología , Animales , Diferenciación Celular/efectos de los fármacos , Fusión Celular , Células Cultivadas , Humanos , Factor Estimulante de Colonias de Macrófagos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteogénesis/genética , Isoformas de Proteínas , Estructura Terciaria de Proteína , Ligando RANK/farmacología , ATPasas de Translocación de Protón Vacuolares/química
13.
Lasers Med Sci ; 28(6): 1495-502, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23324954

RESUMEN

This study was aimed to evaluate the effects of low-level laser therapy (LLLT) in the treatment of trabecular bone loss induced by skeletal unloading. Twelve mice have taken denervation operation. At 2 weeks after denervation, LLLT (wavelength, 660 nm; energy, 3 J) was applied to the right tibiae of 6 mice (LASER) for 5 days/week over 2 weeks by using a minimally invasive laser needle system (MILNS) which consists of a 100 µm optical fiber in a fine needle (diameter, 130 µm) [corrected]. Structural parameters and histograms of bone mineralization density distribution (BMDD) were obtained before LLLT and at 2 weeks after LLLT. In addition, osteocyte, osteoblast, and osteoclast populations were counted. Two weeks after LLLT, bone volume fraction, trabeculae number, and trabeculae thickness were significantly increased and trabecular separations, trabecular bone pattern factor, and structure model index were significantly decreased in LASER than SHAM (p < 0.05). BMDD in LASER was maintained while that in SHAM was shifted to lower mineralization. Osteocyte and osteoblast populations were significantly increased but osteoclast population was significantly decreased in LASER when compared with those in SHAM (p < 0.05). The results indicate that LLLT with the MILNS may enhance bone quality and bone homeostasis associated with enhancement of bone formation and suppression of bone resorption.


Asunto(s)
Resorción Ósea/radioterapia , Terapia por Luz de Baja Intensidad/instrumentación , Animales , Densidad Ósea/efectos de la radiación , Resorción Ósea/etiología , Resorción Ósea/patología , Femenino , Suspensión Trasera/efectos adversos , Ratones , Ratones Endogámicos ICR , Agujas , Fibras Ópticas , Osteoblastos/patología , Osteoblastos/efectos de la radiación , Osteoclastos/patología , Osteoclastos/efectos de la radiación , Osteocitos/patología , Osteocitos/efectos de la radiación
14.
J Cell Mol Med ; 16(2): 273-86, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21362131

RESUMEN

Vascular calcification is prevalent in patients with chronic kidney disease and leads to increased cardiovascular morbidity and mortality. Although several reports have implicated mitochondrial dysfunction in cardiovascular disease and chronic kidney disease, little is known about the potential role of mitochondrial dysfunction in the process of vascular calcification. This study investigated the effect of α-lipoic acid (ALA), a naturally occurring antioxidant that improves mitochondrial function, on vascular calcification in vitro and in vivo. Calcifying vascular smooth muscle cells (VSMCs) treated with inorganic phosphate (Pi) exhibited mitochondrial dysfunction, as demonstrated by decreased mitochondrial membrane potential and ATP production, the disruption of mitochondrial structural integrity and concurrently increased production of reactive oxygen species. These Pi-induced functional and structural mitochondrial defects were accompanied by mitochondria-dependent apoptotic events, including release of cytochrome c from the mitochondria into the cytosol, subsequent activation of caspase-9 and -3, and chromosomal DNA fragmentation. Intriguingly, ALA blocked the Pi-induced VSMC apoptosis and calcification by recovery of mitochondrial function and intracellular redox status. Moreover, ALA inhibited Pi-induced down-regulation of cell survival signals through the binding of growth arrest-specific gene 6 (Gas6) to its cognate receptor Axl and subsequent Akt activation, resulting in increased survival and decreased apoptosis. Finally, ALA significantly ameliorated vitamin D(3) -induced aortic calcification and mitochondrial damage in mice. Collectively, the findings suggest ALA attenuates vascular calcification by inhibiting VSMC apoptosis through two distinct mechanisms; preservation of mitochondrial function via its antioxidant potential and restoration of the Gas6/Axl/Akt survival pathway.


Asunto(s)
Mitocondrias/metabolismo , Músculo Liso Vascular/metabolismo , Ácido Tióctico/metabolismo , Calcificación Vascular/metabolismo , Animales , Apoptosis/efectos de los fármacos , Calcio/metabolismo , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Células Cultivadas , Colecalciferol/farmacología , Citocromos c , Fragmentación del ADN , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Enfermedades Renales/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/enzimología , Mitocondrias/patología , Músculo Liso Vascular/patología , Fosfatos/farmacología , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Tirosina Quinasa del Receptor Axl
15.
J Biol Chem ; 286(45): 39043-50, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-21949120

RESUMEN

Glycogen synthase kinase-3ß (GSK-3ß) is a serine/threonine kinase originally identified as a regulator of glycogen deposition. Although the role of GSK-3ß in osteoblasts is well characterized as a negative regulator of ß-catenin, its effect on osteoclast formation remains largely unidentified. Here, we show that the GSK-3ß inactivation upon receptor activator of NF-κB ligand (RANKL) stimulation is crucial for osteoclast differentiation. Regulation of GSK-3ß activity in bone marrow macrophages by retroviral expression of the constitutively active GSK-3ß (GSK3ß-S9A) mutant inhibits RANKL-induced osteoclastogenesis, whereas expression of the catalytically inactive GSK-3ß (GSK3ß-K85R) or small interfering RNA (siRNA)-mediated GSK-3ß silencing enhances osteoclast formation. Pharmacological inhibition of GSK-3ß further confirmed the negative role of GSK-3ß in osteoclast formation. We also show that overexpression of the GSK3ß-S9A mutant in bone marrow macrophages inhibits RANKL-mediated NFATc1 induction and Ca(2+) oscillations. Remarkably, transgenic mice expressing the GSK3ß-S9A mutant show an osteopetrotic phenotype due to impaired osteoclast differentiation. Further, osteoclast precursor cells from the transgenic mice show defects in expression and nuclear localization of NFATc1. These findings demonstrate a novel role for GSK-3ß in the regulation of bone remodeling through modulation of NFATc1 in RANKL signaling.


Asunto(s)
Células de la Médula Ósea/enzimología , Remodelación Ósea/fisiología , Diferenciación Celular/fisiología , Núcleo Celular/enzimología , Glucógeno Sintasa Quinasa 3/metabolismo , Osteoclastos/enzimología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Transporte Activo de Núcleo Celular/fisiología , Sustitución de Aminoácidos , Animales , Relojes Biológicos/efectos de los fármacos , Relojes Biológicos/fisiología , Células de la Médula Ósea/citología , Remodelación Ósea/efectos de los fármacos , Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/genética , Células Cultivadas , Activación Enzimática/efectos de los fármacos , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Transgénicos , Mutación Missense , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/citología , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
16.
Biochem Biophys Res Commun ; 418(1): 144-8, 2012 Feb 03.
Artículo en Inglés | MEDLINE | ID: mdl-22244876

RESUMEN

Acidic extracellular pH promotes osteoporotic bone loss by osteoclast activation. However, the change of osteoclastic cell behavior in acidosis-stimulated bone resorption process is unknown. We found that lowering extracellular pH induced an increase in the survival, adhesion, and migration of mature osteoclasts with a full actin ring, leading to enhanced pit formation on dentine slices. Acidosis upregulated osteopontin, which is an Arg-Gly-Asp (RGD) motif-containing matrix protein secreted from osteoclasts and acts as a common modulator for their survival, adhesion, and migration. A synthetic RGD peptide treatment blocked acidosis-induced osteoclast adhesion and migration, likely by competing with the RGD motif-containing extracellular matrix proteins for cell surface integrin binding. We finally observed that acidosis was associated with activation of osteoclast survival/adhesion/migration-related Pyk2, Cbl-b, and Src signals. Collectively, the findings indicate that extracellular acidosis stimulates bone resorption by extending osteoclast survival and facilitating osteoclast adhesion and migration.


Asunto(s)
Acidosis/fisiopatología , Resorción Ósea/fisiopatología , Movimiento Celular , Osteoclastos/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Resorción Ósea/metabolismo , Adhesión Celular , Supervivencia Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/metabolismo , Quinasa 2 de Adhesión Focal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-cbl/metabolismo , Familia-src Quinasas/metabolismo
17.
Mol Biol Rep ; 39(3): 3211-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21688145

RESUMEN

To assess the recovery effect of water-soluble components of nacre on wound healing of burns, water-soluble nacre (WSN) was obtained from powdered nacre. Alterations to WSN-mediated wound healing characteristics were examined in porcine skin with deep second-degree burns; porcine skin was used as a proxy for human. When WSN was applied to a burned area, the burn-induced granulation sites were rapidly filled with collagen, and the damaged dermis and epidermis were restored to the appearance of normal skin. WSN enhanced wound healing recovery properties for burn-induced apoptotic and necrotic cellular damage and spurred angiogenesis. Additionally, WSN-treated murine fibroblast NIH3T3 cells showed increased proliferation and collagen synthesis. Collectively, the findings indicate that WSN improves the process of wound healing in burns by expeditiously restoring angiogenesis and fibroblast activity. WSN may be useful as a therapeutic agent, with superior biocompatibility to powdered nacre, and evoking less discomfort when applied to a wounded area.


Asunto(s)
Quemaduras/fisiopatología , Fibroblastos/efectos de los fármacos , Nácar/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Piel/lesiones , Cicatrización de Heridas/efectos de los fármacos , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Colágeno/biosíntesis , Cartilla de ADN/genética , Fibroblastos/fisiología , Ratones , Células 3T3 NIH , Nácar/química , Neovascularización Fisiológica/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Solubilidad , Sus scrofa , Agua/química , Cicatrización de Heridas/fisiología
18.
J Bone Metab ; 29(4): 265-269, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36529869

RESUMEN

BACKGROUND: Cells have heterogeneous cellular diversity in size, morphology, cell cycle, metabolism, differentiation degree, and spatial distribution. The shift of specific cells towards the desired cells is crucial for maintaining uniform cellular function and can be represented by homogeneity and heterogeneity. Here, we developed a simple and direct method for evaluating the homogeneous distribution of desired cells in a constant region. METHODS: We differentiated osteoclast progenitors into bone-resorbing multinucleated giant osteoclasts in a 2-dimensional culture plate under 2 conditions. Cells were stained with tartrate-resistant acid phosphatase to assess osteoclast differentiation, images were taken using a microscope and divided into sectors, and the number of osteoclasts (≥3 nuclei) in each sector was counted. To assess the homogeneity of the spatial distribution of osteoclasts, the standard deviation (SD) was calculated from the mean number of osteoclasts within each sector. RESULTS: From the 2 groups, a value with a SD close to 0 indicates high spatial homogeneity while a relatively high SD represents low spatial homogeneity. CONCLUSIONS: Our findings suggest that spatial homogeneity can be represented as SD.

19.
Nanomaterials (Basel) ; 12(24)2022 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-36558270

RESUMEN

The purpose of this study was to evaluate the surface properties of ZnO nanomaterials based on their ability to photodegrade methyl blue dye (MB) and to show their antibacterial properties against different types of Gram-positive bacteria (Bacillus manliponensis, Micrococcus luteus, Staphylococcus aureus) and Gram-negative bacteria (Escherichia coli). In this study, ZnO nanomaterials were synthesized rapidly and easily in the presence of 1-4 M NaOH at a low temperature of 40 °C within 4 h. It was found that the ZnO nanomaterials obtained from the 1.0 M (ZnO-1M) and 2.0 M (ZnO-2M) aqueous solutions of NaOH had spherical and needle-shaped forms, respectively. As the concentration of NaOH increased, needle thickness increased and the particles became rod-like. Although the ZnO nanomaterial shapes were different, the bandgap size remained almost unchanged. However, as the NaOH concentration increased, the energy position of the conduction band shifted upward. Photo current curves and photoluminescence intensities suggested that the recombination between photoexcited electrons and holes was low in the ZnO-4M materials prepared in 4.0 M NaOH solution; however, charge transfer was easy. ∙O2- radicals were generated more than ∙OH radicals in ZnO-4M particles, showing stronger antibacterial activity against both Gram-positive and Gram-negative bacteria and stronger decomposition ability on MB dye. The results of this study suggest that on the ZnO nanomaterial surface, ∙O2- radicals generated are more critical for antibacterial activity than particle shape.

20.
J Cell Mol Med ; 15(10): 2082-94, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21159123

RESUMEN

Systemic transplantation of adipose-derived stem cells (ASCs) is emerging as a novel therapeutic option for functional recovery of diverse damaged tissues. This study investigated the effects of systemic transplantation of human ASCs (hASCs) on bone repair. We found that hASCs secrete various bone cell-activating factors, including hepatocyte growth factor and extracellular matrix proteins. Systemic transplantation of hASCs into ovariectomized mice induced an increased number of both osteoblasts and osteoclasts in bone tissue and thereby prevented bone loss. We also observed that conditioned medium from hASCs is capable of stimulating proliferation and differentiation of osteoblasts via Smad/extracellular signal-regulated kinase (ERK)/JNK (c-jun NH(2) -terminal kinase) activation as well as survival and differentiation of osteoclasts via ERK/JNK/p38 activation in vitro. Overall, our findings suggest that paracrine factors secreted from hASCs improve bone repair and that hASCs can be a valuable tool for use in osteoporosis therapy.


Asunto(s)
Adipocitos/metabolismo , Regeneración Ósea/fisiología , Trasplante de Células Madre/métodos , Células Madre/metabolismo , Adipocitos/citología , Animales , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Medios de Cultivo Condicionados , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Expresión Génica , Humanos , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Ratones , Osteoblastos/citología , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/metabolismo , Transducción de Señal , Células Madre/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA