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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Biol Rep ; 51(1): 587, 2024 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-38683225

RESUMO

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.


Assuntos
Diferenciação Celular , Mieloma Múltiplo , Osteoclastos , Selenoproteína W , Animais , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Diferenciação Celular/genética , Leucócitos Mononucleares/metabolismo , Sistema de Sinalização das MAP Quinases , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Mieloma Múltiplo/genética , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/metabolismo , Fatores de Transcrição NFATC/genética , Osteoclastos/metabolismo , Ligante RANK/metabolismo , Selenoproteína W/metabolismo , Selenoproteína W/genética , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
2.
Int J Mol Sci ; 20(23)2019 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-31810213

RESUMO

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.


Assuntos
Bivalves , Doenças Ósseas/dietoterapia , Estrogênios/deficiência , Osteoporose/dietoterapia , Animais , Doenças Ósseas/metabolismo , Doenças Ósseas/fisiopatologia , Reabsorção Óssea/dietoterapia , Reabsorção Óssea/prevenção & controle , Diferenciação Celular/efeitos dos fármacos , Humanos , Camundongos , Osteoclastos/efeitos dos fármacos , Osteoporose/metabolismo , Alimentos Marinhos/análise
3.
Int J Mol Sci ; 19(10)2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30275408

RESUMO

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.


Assuntos
Proteínas Quinases Ativadas por Mitógeno/metabolismo , Osteoclastos/enzimologia , Animais , Humanos , Cinética , Sistema de Sinalização das MAP Quinases , Modelos Biológicos
4.
Int J Mol Sci ; 18(10)2017 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-28946669

RESUMO

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.


Assuntos
Reabsorção Óssea/etiologia , Reabsorção Óssea/metabolismo , Inflamação/complicações , Osteoclastos/citologia , Osteoclastos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Ubiquitina/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Bortezomib/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Fator Estimulador de Colônias de Macrófagos/metabolismo , Masculino , Camundongos , Inibidores de Proteassoma/farmacologia , Proteólise , Receptor de Fator Estimulador de Colônias de Macrófagos/genética
5.
EMBO J ; 31(5): 1147-59, 2012 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-22227519

RESUMO

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.


Assuntos
Hematopoese , Quinase I-kappa B/metabolismo , Osteoblastos/fisiologia , Osteoclastos/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Reabsorção Óssea , Diferenciação Celular , Proliferação de Células , Quinase I-kappa B/antagonistas & inibidores , Camundongos , Camundongos Knockout , NF-kappa B/biossíntese , Osteogênese , Ligação Proteica , Mapeamento de Interação de Proteínas , Proteínas Supressoras de Tumor/deficiência
6.
Int J Mol Sci ; 17(8)2016 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-27517906

RESUMO

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.


Assuntos
Osteoclastos/citologia , Animais , Catepsina K/metabolismo , Pontos de Checagem do Ciclo Celular/fisiologia , Diferenciação Celular/fisiologia , Proliferação de Células/fisiologia , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Immunoblotting , Integrina beta3/metabolismo , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/metabolismo , Osteoclastos/fisiologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ligante RANK/metabolismo , ATPases Vacuolares Próton-Translocadoras/metabolismo
7.
J Immunol ; 190(4): 1623-30, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23303667

RESUMO

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.


Assuntos
Calcinose/prevenção & controle , Infecções por Coxsackievirus/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoporose/metabolismo , Osteoporose/prevenção & controle , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Animais , Calcinose/patologia , Calcinose/virologia , Infecções por Coxsackievirus/imunologia , Infecções por Coxsackievirus/patologia , Modelos Animais de Doenças , Células HeLa , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ossificação Heterotópica/metabolismo , Ossificação Heterotópica/patologia , Ossificação Heterotópica/virologia , Osteoblastos/patologia , Osteoblastos/virologia , Osteoclastos/patologia , Osteoclastos/virologia , Osteoporose/virologia , Ligante RANK/antagonistas & inibidores , Ratos , Ratos Sprague-Dawley , Receptor Ativador de Fator Nuclear kappa-B/administração & dosagem , Proteínas Recombinantes de Fusão/administração & dosagem , Proteínas Recombinantes de Fusão/genética
8.
Int J Mol Sci ; 16(12): 29305-14, 2015 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-26690137

RESUMO

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.


Assuntos
Fator 1 de Ribosilação do ADP/fisiologia , Diferenciação Celular , Movimento Celular , Proliferação de Células , Osteoclastos/fisiologia , Animais , Fusão Celular , Células Cultivadas , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais
9.
Biochem Biophys Res Commun ; 453(4): 793-7, 2014 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-25445591

RESUMO

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.


Assuntos
Aorta/enzimologia , Calcinose/enzimologia , Citoesqueleto/metabolismo , Osteogênese , Doenças Vasculares Periféricas/enzimologia , Proteína Quinase C/metabolismo , Animais , Aorta/patologia , Calcinose/patologia , Células Cultivadas , Citoesqueleto/patologia , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Miócitos de Músculo Liso/enzimologia , Miócitos de Músculo Liso/patologia , Proteína Quinase C-delta/metabolismo
10.
Biochem Biophys Res Commun ; 451(3): 436-41, 2014 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-25111813

RESUMO

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.


Assuntos
Microtúbulos/metabolismo , Osteogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Calcificação Vascular/prevenção & controle , Animais , Calcinose/prevenção & controle , Citoesqueleto/fisiologia , Masculino , Camundongos , Músculo Liso Vascular/metabolismo , Osteogênese/genética , Paclitaxel/farmacologia , Fosfatos/metabolismo , Regulação para Cima
11.
Biochem Biophys Res Commun ; 432(2): 378-83, 2013 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-23357417

RESUMO

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.


Assuntos
Calcinose/prevenção & controle , Infecções por Coxsackievirus/tratamento farmacológico , Enterovirus Humano B , Ácido Tióctico/uso terapêutico , Animais , Calcinose/patologia , Calcinose/virologia , Infecções por Coxsackievirus/complicações , Infecções por Coxsackievirus/patologia , Feminino , Coração/virologia , Humanos , Pulmão/patologia , Pulmão/virologia , Masculino , Camundongos , Pâncreas/patologia , Pâncreas/virologia
12.
Nat Med ; 12(12): 1403-9, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17128270

RESUMO

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.


Assuntos
Osteoclastos/fisiologia , Osteogênese/fisiologia , Bombas de Próton/genética , ATPases Vacuolares Próton-Translocadoras/fisiologia , Animais , Diferenciação Celular/efeitos dos fármacos , Fusão Celular , Células Cultivadas , Humanos , Fator Estimulador de Colônias de Macrófagos/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoclastos/citologia , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteogênese/genética , Isoformas de Proteínas , Estrutura Terciária de Proteína , Ligante RANK/farmacologia , ATPases Vacuolares Próton-Translocadoras/química
13.
Lasers Med Sci ; 28(6): 1495-502, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23324954

RESUMO

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.


Assuntos
Reabsorção Óssea/radioterapia , Terapia com Luz de Baixa Intensidade/instrumentação , Animais , Densidade Óssea/efeitos da radiação , Reabsorção Óssea/etiologia , Reabsorção Óssea/patologia , Feminino , Elevação dos Membros Posteriores/efeitos adversos , Camundongos , Camundongos Endogâmicos ICR , Agulhas , Fibras Ópticas , Osteoblastos/patologia , Osteoblastos/efeitos da radiação , Osteoclastos/patologia , Osteoclastos/efeitos da radiação , Osteócitos/patologia , Osteócitos/efeitos da radiação
14.
J Cell Mol Med ; 16(2): 273-86, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21362131

RESUMO

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.


Assuntos
Mitocôndrias/metabolismo , Músculo Liso Vascular/metabolismo , Ácido Tióctico/metabolismo , Calcificação Vascular/metabolismo , Animais , Apoptose/efeitos dos fármacos , Cálcio/metabolismo , Caspase 3/metabolismo , Caspase 9/metabolismo , Células Cultivadas , Colecalciferol/farmacologia , Citocromos c , Fragmentação do DNA , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Músculo Liso Vascular/patologia , Fosfatos/farmacologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Doenças Vasculares/genética , Doenças Vasculares/metabolismo , Receptor Tirosina Quinase Axl
15.
J Biol Chem ; 286(45): 39043-50, 2011 Nov 11.
Artigo em Inglês | MEDLINE | ID: mdl-21949120

RESUMO

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.


Assuntos
Células da Medula Óssea/enzimologia , Remodelação Óssea/fisiologia , Diferenciação Celular/fisiologia , Núcleo Celular/enzimologia , Quinase 3 da Glicogênio Sintase/metabolismo , Osteoclastos/enzimologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Substituição de Aminoácidos , Animais , Relógios Biológicos/efeitos dos fármacos , Relógios Biológicos/fisiologia , Células da Medula Óssea/citologia , Remodelação Óssea/efeitos dos fármacos , Cálcio/metabolismo , Diferenciação Celular/efeitos dos fármacos , Núcleo Celular/genética , Células Cultivadas , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Inibidores Enzimáticos/farmacologia , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Quinase 3 da Glicogênio Sintase/genética , Glicogênio Sintase Quinase 3 beta , Camundongos , Camundongos Transgênicos , Mutação de Sentido Incorreto , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Ligante RANK/genética , Ligante RANK/metabolismo , RNA Interferente Pequeno/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
16.
Biochem Biophys Res Commun ; 418(1): 144-8, 2012 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-22244876

RESUMO

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.


Assuntos
Acidose/fisiopatologia , Reabsorção Óssea/fisiopatologia , Movimento Celular , Osteoclastos/fisiologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Reabsorção Óssea/metabolismo , Adesão Celular , Sobrevivência Celular , Células Cultivadas , Proteínas da Matriz Extracelular/metabolismo , Quinase 2 de Adesão Focal/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteoclastos/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Quinases da Família src/metabolismo
17.
Mol Biol Rep ; 39(3): 3211-8, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21688145

RESUMO

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.


Assuntos
Queimaduras/fisiopatologia , Fibroblastos/efeitos dos fármacos , Nácar/farmacologia , Neovascularização Fisiológica/efeitos dos fármacos , Pele/lesões , Cicatrização/efeitos dos fármacos , Animais , Western Blotting , Proliferação de Células/efeitos dos fármacos , Colágeno/biossíntese , Primers do DNA/genética , Fibroblastos/fisiologia , Camundongos , Células NIH 3T3 , Nácar/química , Neovascularização Fisiológica/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Solubilidade , Sus scrofa , Água/química , Cicatrização/fisiologia
18.
J Bone Metab ; 29(4): 265-269, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36529869

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-36558270

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-21159123

RESUMO

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.


Assuntos
Adipócitos/metabolismo , Regeneração Óssea/fisiologia , Transplante de Células-Tronco/métodos , Células-Tronco/metabolismo , Adipócitos/citologia , Animais , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Meios de Cultivo Condicionados , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Humanos , MAP Quinase Quinase 4/genética , MAP Quinase Quinase 4/metabolismo , Camundongos , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoclastos/citologia , Osteoclastos/metabolismo , Transdução de Sinais , Células-Tronco/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA