Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cells Dev ; 26(6): 419-430, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-27917698

RESUMO

Glucocorticoid-induced osteoporosis (GIOP) is a widespread clinical complication due to the common use of glucocorticoids. Excess glucocorticoids induce apoptosis of bone marrow-derived mesenchymal stem cells (BMSCs), which have been shown to play an increasingly important role in the pathogenesis and therapy of osteoporosis. Tetramethylpyrazine (TMP), an extract from one of the most recognized herbs in traditional Chinese medicine (Chuanxiong), has been reported to have antiapoptotic properties. In this study, we tested whether TMP protects rat BMSCs following exposure to glucocorticoids in vitro and in vivo. We treated BMSCs with different concentrations of TMP (50, 100, or 200 µM) and exposed them to 10-6 M dexamethasone (Dex) for 48 h in vitro. Our data showed that TMP inhibited Dex-induced cytotoxicity and protected BMSCs from apoptosis. Interestingly, further results demonstrated that TMP prevented apoptosis in BMSCs by promoting autophagy in an AMP-activated protein kinase (AMPK) and mammalian target of rapamycin (mTOR) pathway-dependent manner. In addition, calcein fluorescence double labeling and microcomputed tomography scanning indicated that 12 weeks of TMP administration augmented bone formation and protected trabecular bone mass in GIOP rats. We also discovered that first-passage BMSCs isolated from the TMP treatment group had a lower rate of apoptosis and a higher light chain 3 (LC3)-II/LC3-I ratio than the GIOP group. Our findings demonstrate for the first time that TMP can protect BMSCs from exposure to excess glucocorticoids by promoting autophagy through AMPK/mTOR pathway and might be an effective agent for the prevention and treatment of GIOP.


Assuntos
Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Osso e Ossos/patologia , Glucocorticoides/efeitos adversos , Células-Tronco Mesenquimais/citologia , Osteoporose/induzido quimicamente , Osteoporose/tratamento farmacológico , Pirazinas/uso terapêutico , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Osso e Ossos/efeitos dos fármacos , Dexametasona , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Osteoporose/patologia , Substâncias Protetoras/farmacologia , Substâncias Protetoras/uso terapêutico , Pirazinas/farmacologia , Ratos Sprague-Dawley , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo
2.
Int J Clin Exp Pathol ; 8(7): 8446-52, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26339416

RESUMO

BACKGROUND: Giant cell tumor of bone (GCT) is a potentially malignant tumor. CD147 is a multifunctional protein, which expresses itself in many tumors. In this study, we have investigated the correlation between CD147 and PCNA, VEGF, MMPs expression in giant cell tumor of bones. We have also explored the relationship between its clinical pathology and prognosis. RESULTS: A significant difference of the expression level of CD147, MMPs was found in cases of GCT with Jaffe grading and prognosis (P<0.05). But, it was not in accordance with the patient's age and sex. An expression of CD147 was positively correlated with MMP-9, VEGF, MVD, PCNA (r=0.271, P=0.025; r=0.411, P=0.000; r=0.872, P=0.000; r=0.394, P=0.001). CONCLUSION: The expression level of CD147 in giant cell tumors of bones is correlated with the development of cancers and relapse. There was a positive correlation between expressions of CD147 and MMP-9, VEGF, MVD, PCNA, and CD147. This is an important indicator in evaluating the malignant degree and prognosis of giant cell tumors of bone. It may be the new target for ensuring chemopreventive strategies.


Assuntos
Basigina/análise , Neoplasias Ósseas/química , Tumor de Células Gigantes do Osso/química , Metaloproteinase 9 da Matriz/análise , Antígeno Nuclear de Célula em Proliferação/análise , Fator A de Crescimento do Endotélio Vascular/análise , Adolescente , Adulto , Neoplasias Ósseas/patologia , Criança , Feminino , Tumor de Células Gigantes do Osso/patologia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Valor Preditivo dos Testes , Adulto Jovem
3.
Bone ; 73: 132-44, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25554600

RESUMO

Increased levels of reactive oxygen species (ROS) are a crucial pathogenic factor of osteoporosis. Gastrodin, isolated from the traditional Chinese herbal agent Gastrodia elata, is a potent antioxidant. We hypothesized that gastrodin demonstrates protective effects against osteoporosis by partially reducing reactive oxygen species in human bone marrow mesenchymal stem cells (hBMMSCs) and a macrophage cell line (RAW264.7 cells). We investigated gastrodin on osteogenic and adipogenic differentiation under oxidative stress in hBMMSCs. We also tested gastrodin on osteoclastic differentiation in RAW264.7 cells. Hydrogen peroxide (H2O2) was used to establish an oxidative cell injury model. Our results showed that gastrodin significantly promoted the proliferation of hBMMSCs, improved some osteogenic markers, reduced lipid generation and inhibited the mRNA expression of several adipogenic genes in hBMMSCs. Moreover, gastrodin reduced the number of osteoclasts, TRAP activity and the expression of osteoclast-specific genes in RAW264.7 cells. Gastrodin suppressed the production of reactive oxygen species in both hBMMSCs and RAW264.7 cells. In vivo, we established a murine ovariectomized (OVX) osteoporosis model. Our data revealed that gastrodin treatment reduced the activity of serum bone degradation markers, such as CTX-1 and TRAP. Importantly, it ameliorated the micro-architecture of trabecular bones. Gastrodin decreased osteoclast numbers in vivo by TRAP staining. To conclude, these results indicated that gastrodin shows protective effects against osteoporosis linking to a reduction in reactive oxygen species, suggesting that gastrodin may be useful in the prevention and treatment of osteoporosis.


Assuntos
Álcoois Benzílicos/farmacologia , Medicamentos de Ervas Chinesas/farmacologia , Glucosídeos/farmacologia , Osteoporose/prevenção & controle , Espécies Reativas de Oxigênio/metabolismo , Animais , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Feminino , Humanos , Interleucina-6/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos BALB C , Osteoporose/metabolismo , Ovariectomia , Ligante RANK/antagonistas & inibidores
4.
Stem Cells Dev ; 24(6): 781-90, 2015 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-25314926

RESUMO

Apoptosis of bone marrow-derived mesenchymal stem cells (BMMSCs) is an essential pathogenic factor of osteoporosis. Ginsenoside-Rb2 (Rb2), a 20(S)-protopanaxadiol glycoside extracted from ginseng, is a potent treatment for bone loss, which raises interest regarding the bone metabolism area. In the present study, we found that dose-response Rb2 inhibited high dosage of dexamethasone (Dex)-induced apoptosis in primary murine BMMSCs. Interestingly, Rb2 promoted GPR120 induction, which is the unsaturated long-chain fatty acid receptor. We further confirmed that GPR120-specific ShRNA reversed the inhibition of Rb2 on Dex-induced apoptosis by activating caspase-3 and reducing cell viability. In addition, Rb2 notably increased phosphorylated ERK1/2 levels and Ras kinase activity dependently through the GPR120. The ERK1/2 activity-specific inhibitor U0126 remarkably blocked the Rb2-induced antiapoptotic effect in response to Dex-induced apoptosis. Together, dose-response Rb2 protected BMMSCs against Dex-induced apoptosis dependently by inducing GPR120 promoted Ras-ERK1/2 signaling pathway. Therefore, in the prevalence of the abuse of Dex in the clinic, our findings suggest for the first time that Rb2 is not only a key to understand the link between Chinese medicine and the pathology of osteoporosis but also an underlying target for the treatment of bone complications in the foreseeable future.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Ginsenosídeos/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Células Cultivadas , Dexametasona/toxicidade , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/genética
5.
PLoS One ; 9(6): e99137, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24918446

RESUMO

Fat infiltration within marrow cavity is one of multitudinous features of estrogen deficiency, which leads to a decline in bone formation functionality. The origin of this fat is unclear, but one possibility is that it is derived from osteoblasts, which transdifferentiate into adipocytes that produce bone marrow fat. We examined the dose-dependent effect of 17ß-estradiol on the ability of MC3T3-E1 cells and murine bone marrow-derived mesenchymal stem cell (BMMSC)-derived osteoblasts to undergo osteo-adipogenic transdifferentiation. We found that 17ß-estradiol significantly increased alkaline phosphatase activity (P<0.05); calcium deposition; and Alp, Col1a1, Runx2, and Ocn expression levels dose-dependently. By contrast, 17ß-estradiol significantly decreased the number and size of lipid droplets, and Fabp4 and PPARγ expression levels during osteo-adipogenic transdifferentiation (P<0.05). Moreover, the expression levels of brown adipocyte markers (Myf5, Elovl3, and Cidea) and undifferentiated adipocyte markers (Dlk1, Gata2, and Wnt10b) were also affected by 17ß-estradiol during osteo-adipogenic transdifferentiation. Western blotting and immunostaining further showed that canonical Wnt signaling can be activated by estrogen to exert its inhibitory effect of osteo-adipogenesis. This is the first study to demonstrate the dose-dependent effect of 17ß-estradiol on the osteo-adipogenic transdifferentiation of MC3T3-E1 cells and BMMSCs likely via canonical Wnt signaling. In summary, our results indicate that osteo-adipogenic transdifferentiation modulated by canonical Wnt signaling pathway in bone metabolism may be a new explanation for the gradually increased bone marrow fat in estrogen-inefficient condition.


Assuntos
Tecido Adiposo/citologia , Diferenciação Celular/efeitos dos fármacos , Estrogênios/farmacologia , Osteoblastos/citologia , Via de Sinalização Wnt , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Relação Dose-Resposta a Droga , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Ovariectomia
6.
Lab Invest ; 94(8): 906-16, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24933421

RESUMO

Although many studies have suggested that estrogen prevents postmenopausal bone loss partially due to its anti-apoptosis effects in osteoblasts, the underlying mechanism has not been fully elucidated. In the present study, we found that 17ß-estradiol (17ß-E2), one of the primary estrogens, inhibited endoplasmic reticulum (ER) stress-induced apoptosis in MC3T3-E1 cells and primary osteoblasts. Interestingly, 17ß-E2-promoted Grp78 induction, but not CHOP induction in response to ER stress. We further confirmed that Grp78-specific siRNA reversed the inhibition of 17ß-E2 on ER stress-induced apoptosis by activating caspase-12 and caspase-3. Moreover, we found that 17ß-E2 markedly increased the phosphorylated TFII-I levels and nuclear localization of TFII-I in ER stress conditions. 17ß-E2 stimulated Grp78 promoter activity in a dose-dependent manner in the presence of TFII-I and enhanced the binding of TFII-I to the Grp78 promoter. In addition, 17ß-E2 notably increased phosphorylated ERK1/2 levels and Ras kinase activity in MC3T3-E1 cells. The ERK1/2 activity-specific inhibitor U0126 remarkably blocked 17ß-E2-induced TFII-I phosphorylation and Grp78 expression in response to ER stress. Together, 17ß-E2 protected MC3T3-E1 cells against ER stress-induced apoptosis by promoting Ras-ERK1/2-TFII-I signaling pathway-dependent Grp78 induction.


Assuntos
Apoptose/efeitos dos fármacos , Conservadores da Densidade Óssea/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Estradiol/farmacologia , Proteínas de Choque Térmico/agonistas , Osteoblastos/efeitos dos fármacos , Fator de Transcrição TFIIA/agonistas , Animais , Animais Recém-Nascidos , Conservadores da Densidade Óssea/química , Conservadores da Densidade Óssea/uso terapêutico , Linhagem Celular , Células Cultivadas , Chaperona BiP do Retículo Endoplasmático , Estradiol/química , Estradiol/uso terapêutico , Estrogênios/química , Estrogênios/farmacologia , Estrogênios/uso terapêutico , Proteínas de Choque Térmico/antagonistas & inibidores , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteoporose/induzido quimicamente , Osteoporose/metabolismo , Osteoporose/prevenção & controle , Fosforilação/efeitos dos fármacos , Regiões Promotoras Genéticas/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Interferência de RNA , Distribuição Aleatória , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Fator de Transcrição TFIIA/genética , Fator de Transcrição TFIIA/metabolismo
7.
PLoS One ; 9(3): e90220, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24595049

RESUMO

Osteosarcoma, the most common primary bone tumor in children and young adolescents, is characterized by local invasion and distant metastasis. But the detailed mechanisms of osteosarcoma metastasis are not well known. In the present study, we found that ßig-h3 promotes metastatic potential of human osteosarcoma cells in vitro and in vivo. Furthermore, ßig-h3 co-localized with integrin α2ß1 in osteosarcoma cells. But ßig-h3 did not change integrin α2ß1 expression in Saos-2 cells. Interaction of ßig-h3 with integrin α2ß1 mediates metastasis of human osteosarcoma cells. The second FAS1 domain of ßig-h3 but not the first FAS1 domain, the third FAS1 domain or the fourth FAS1 domain mediates human osteosarcoma cells metastasis, which is the α2ß1 integrin-interacting domain. We further demonstrated that PI3K/AKT signaling pathway is involved in ßig-h3-induced human osteosarcoma cells metastasis process. Together, these results reveal ßig-h3 enhances the metastasis potentials of human osteosarcoma cells via integrin α2ß1-mediated PI3K/AKT signal pathways. The discovery of ßig-h3-mediated pathway helps us to understand the mechanism of human osteosarcoma metastasis and provides evidence for the possibility that ßig-h3 can be a potential therapeutic target for osteosarcoma treatment.


Assuntos
Proteínas da Matriz Extracelular/metabolismo , Integrina alfa2beta1/metabolismo , Osteossarcoma/enzimologia , Osteossarcoma/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Regulação para Baixo , Ativação Enzimática , Proteínas da Matriz Extracelular/química , Humanos , Imunoprecipitação , Invasividade Neoplásica , Metástase Neoplásica , Ligação Proteica , Estrutura Terciária de Proteína , Fator de Crescimento Transformador beta/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...