Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
Cell Death Differ ; 13(11): 1845-56, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16470222

RESUMO

Osteosarcoma is the most common primary bone tumour in young adults. Despite improved prognosis, resistance to chemotherapy remains responsible for failure of osteosarcoma treatment. The identification of signals that promote apoptosis may provide clues to develop new therapeutic strategies for chemoresistant osteosarcoma. Here, we show that lipophilic statins (atorvastatin, simvastatin, cerivastatin) markedly induce caspases-dependent apoptosis in various human osteosarcoma cells, independently of bone morphogenetic protein (BMP)-2 signaling and cell differentiation. Although statins increased BMP-2 expression, the proapoptotic effect of statins was not prevented by the BMP antagonist noggin, and was abolished by mevalonate and geranylgeranylpyrophosphate, suggesting the involvement of defective protein geranylgeranylation. Consistently, lipophilic statins induced membrane RhoA relocalization to the cytosol and inhibited RhoA activity, which resulted in decreased phospho-p42/p44- mitogen-activated protein kinases (MAPKs) and Bcl-2 levels. Constitutively active RhoA rescued phospho-p42/p44-MAPKs and Bcl-2 and abolished statin-induced apoptosis. Thus, lipophilic statins induce caspase-dependent osteosarcoma cell apoptosis by a RhoA-p42/p44 MAPKs-Bcl-2-mediated mechanism, independently of BMP-2 signaling and cell differentiation.


Assuntos
Anticolesterolemiantes/farmacologia , Apoptose/efeitos dos fármacos , Proteínas Morfogenéticas Ósseas/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Osteossarcoma/patologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Atorvastatina , Proteína Morfogenética Óssea 2 , Proteínas Morfogenéticas Ósseas/genética , Caspases/metabolismo , Diferenciação Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Ácidos Heptanoicos/farmacologia , Humanos , Osteossarcoma/genética , Fenótipo , Fosforilação/efeitos dos fármacos , Fosfatos de Poli-Isoprenil/metabolismo , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Piridinas/farmacologia , Pirróis/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Fator de Crescimento Transformador beta/genética , Proteína rhoA de Ligação ao GTP/antagonistas & inibidores
2.
Oncogene ; 34(24): 3207-13, 2015 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-25065593

RESUMO

Osteosarcoma is the most prevalent primary pediatric cancer-related bone disease. These tumors frequently develop resistance to chemotherapy and are highly metastatic, leading to poor outcome. Thus, there is a need for new therapeutic strategies that can prevent cell dissemination. We previously showed that CYR61/CCN1 expression in osteosarcoma cells is correlated to aggressiveness both in vitro and in vivo in mouse models, as well as in patients. In this study, we found that CYR61 is a critical contributor to the vascularization of primary tumor. We demonstrate that silencing CYR61, using lentiviral transduction, leads to a significant reduction in expression level of pro-angiogenic markers such as VEGF, FGF2, PECAM and angiopoietins concomitantly to an increased expression of major anti-angiogenic markers such as thrombospondin-1 and SPARC. Matrix metalloproteinase-2 family member expression, a key pathway in osteosarcoma metastatic capacity was also downregulated when CYR61 was downregulated in osteosarcoma cells. Using a metastatic murine model, we show that CYR61 silencing in osteosarcoma cells results in reduced tumor vasculature and slows tumor growth compared with control. We also find that microvessel density correlates with lung metastasis occurrence and that CYR61 silencing in osteosarcoma cells limits the number of metastases. Taken together, our data indicate that CYR61 silencing can blunt the malignant behavior of osteosarcoma tumor cells by limiting primary tumor growth and dissemination process.


Assuntos
Neoplasias Ósseas/irrigação sanguínea , Proteína Rica em Cisteína 61/genética , Neovascularização Patológica/genética , Osteossarcoma/irrigação sanguínea , Animais , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Proliferação de Células/genética , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Camundongos Transgênicos , Invasividade Neoplásica , Metástase Neoplásica , Osteossarcoma/genética , Osteossarcoma/patologia , Interferência de RNA , Células Tumorais Cultivadas
3.
J Bone Miner Res ; 15(11): 2211-21, 2000 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-11092402

RESUMO

Breast cancer frequently spreads to bone and is almost always associated with osteolysis. This tumor-induced osteolysis is caused by increased osteoclastic bone resorption. Bisphosphonates are used successfully to inhibit bone resorption in tumor bone disease and may prevent development of new osteolytic lesions. The classical view is that bisphosphonates only act on bone cells. We investigated their effects on breast cancer cells using three human cell lines, namely, MCF-7, T47D, and MDA.MB.231, and we tested four structurally different bisphosphonates: clodronate, pamidronate, ibandronate, and zoledronate. We performed time course studies for each bisphosphonate at various concentrations and found that all four compounds induced a nonreversible growth inhibition in both MCF-7 and T47D cell lines in a time- and dose-dependent manner. The MDA.MB.231 cell line was less responsive. Bisphosphonates induced apoptosis in MCF-7 and cell necrosis in T47D cells. The inhibition of MCF-7 cell proliferation could be reverted almost completely by the benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethyl ketone (z-VAD-fmk) inhibitor of caspases, suggesting that the apoptotic process observed in the MCF-7 cell line is mediated, at least partly, by the caspase system. Caspase activity was little changed by bisphosphonates in T47D cells and the inhibitor of caspase did not modify bisphosphonates effects. In summary, we found that bisphosphonates inhibit breast cancer cell growth by inducing cell death in vitro. Such effects could contribute to the beneficial role of bisphosphonates in the treatment and the prevention of tumor-induced osteolysis.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Difosfonatos/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Anexinas/metabolismo , Apoptose/efeitos dos fármacos , Neoplasias da Mama/metabolismo , Inibidores de Caspase , Caspases/metabolismo , Morte Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Ácido Clodrônico/farmacologia , Inibidores de Cisteína Proteinase/farmacologia , Relação Dose-Resposta a Droga , Feminino , Humanos , Ácido Ibandrônico , Imidazóis/farmacologia , Cinética , Pamidronato , Células Tumorais Cultivadas , Ácido Zoledrônico
4.
J Bone Miner Res ; 16(9): 1600-10, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11547830

RESUMO

Breast cancer is associated frequently with skeletal metastases, which cause significant morbidity. The main mechanism is an increase in osteoclast-mediated bone resorption. We postulated that osteoblasts could be other essential target cells and previously showed that conditioned medium (CM) of breast cancer cells (BCCs) inhibits the proliferation of osteoblast-like cells. In this study, we investigated the effects of BCC-secreted products on osteoprogenitor cells using a clonal fetal human bone marrow stromal preosteoblastic cell line (FHSO-6) that expresses alkaline phosphatase (ALP) activity, type I collagen (COLI), and increased osteocalcin (OC) and osteopontin under treatment with dexamethasone (Dex), 1,25-dihydroxyvitamin D [1,25(OH)2D], or recombinant human bone morphogenetic protein 2 (rhBMP-2). Treatment with MCF-7 CM inhibited FHSO-6 cell survival in a dose-dependent and irreversible manner. Morphological investigation indicated that MCF-7 CM increased both apoptotic and necrotic cell number. MCF-7 CM increased caspases activity and a broad inhibitor of caspase activity (benzyloxycarbonyl-Val-Ala-Asp(OMe)-fluoromethyl ketone [z-VAD-fmk]) partly reversed the CM-induced inhibition of FHSO-6 cell survival. Western blot analyses revealed an increased bax/bcl-2 ratio in MCF-7 CM-treated FHSO-6 cells. MCF-7 cells exhibit FasLigand as membrane-bound protein and as a soluble cytokine in the CM. Deprivation of MCF-7 CM from active FasLigand by saturation with a soluble Fas molecule suppressed the induction of FHSO-6 apoptosis, whereas fibroblast CM, which did not contain FasLigand, only weakly modified FHSO-6 cell survival because of increased cell necrosis. These data indicate that FasLigand secreted by BCCs induces apoptosis and necrosis of human preosteoblastic stromal cells through caspase cascade modulated by the bax and bcl-2 protein level. The induction of apoptosis in human bone marrow stromal cells by BCCs may contribute to the inappropriately low osteoblast reaction and bone formation during tumor-induced osteolysis in bone metastases.


Assuntos
Apoptose , Células da Medula Óssea/citologia , Neoplasias da Mama/metabolismo , Osteoclastos/citologia , Células-Tronco/citologia , Células Estromais/citologia , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Sobrevivência Celular , Células Cultivadas , Meios de Cultivo Condicionados , Proteína Ligante Fas , Feminino , Humanos , Glicoproteínas de Membrana/metabolismo , Necrose , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas , Proteína X Associada a bcl-2
5.
Histol Histopathol ; 19(1): 151-7, 2004 01.
Artigo em Inglês | MEDLINE | ID: mdl-14702183

RESUMO

Human bone marrow stroma contains pluripotent mesenchymal progenitor cells that can give rise to many mesenchymal lineages, including chondroblasts, adipocytes or osteoblasts. The differentiation of these cells towards a specific lineage is dependent on hormonal and local factors activating specific transcription factors. Attempts have been recently made to identify osteoprogenitor cells in the human bone marrow and to identify the molecular mechanisms responsible for lineage-specific differentiation of human bone marrow stromal cells. Using a clonal pluripotent human bone marrow stromal cell line with tri-potential characteristics, we have provided evidence for a controlled reciprocal regulation of osteoblast/chondroblast and osteoblast/adipocyte differentiation of human bone marrow stromal cells. We have also shown that administration of TGFss that regulates the expression of specific osteoblast and adipocyte transcription factors can promote osteoblast differentiation and inhibit adipocyte conversion of rat marrow stromal cells in vivo. This indicates that the reciprocal relationship between osteoblastogenesis and adipogenesis can be manipulated in vivo in order to improve bone formation. Future studies will have to identify key signals for lineage-specific differentiation of human marrow stromal cells. This may result in the development of therapeutic strategies to promote the differentiation of these cells towards the osteoblast lineage and to inhibit excessive bone marrow adipogenesis associated with aging.


Assuntos
Envelhecimento , Doenças Ósseas Metabólicas/terapia , Células da Medula Óssea/citologia , Células Estromais/citologia , Adipócitos/efeitos dos fármacos , Adipócitos/metabolismo , Animais , Doenças Ósseas Metabólicas/patologia , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Linhagem da Célula , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Modelos Biológicos , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo , Fatores de Transcrição/efeitos dos fármacos , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/toxicidade
6.
Cell Death Dis ; 4: e874, 2013 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-24157868

RESUMO

Osteosarcoma is the most common primary tumor of bone occurring in children and adolescents. The histological response to chemotherapy represents a key clinical factor related to survival. We previously showed that statins exhibit antitumor effects in vitro, inducing apoptotic cell death, reducing cell migration and invasion capacities and strengthening cytotoxic effects in combination with standard drugs. Comparative transcriptomic analysis between control and statin-treated cells revealed strong expression of several genes, including metallothionein (MT) 2A. MT2A overexpression by lentiviral transduction reduced bioavailable zinc levels, an effect associated with reduced osteosarcoma cell viability and enhanced cell differentiation. In contrast, MT2A silencing did not modify cell viability but strongly inhibited expression of osteoblastic markers and differentiation process. MT2A overexpression induced chemoresistance to cytotoxic drugs through direct chelation of platinum-containing drugs and indirect action on p53 zinc-dependent activity. In contrast, abrogation of MT2A enhanced cytotoxic action of chemotherapeutic drugs on osteosarcoma cells. Finally, clinical samples derived from chemonaive biopsies revealed that tumor cells expressing low MT2A levels correspond to good prognostic (good responder patients with longer survival rate), whereas high MT2A levels were associated with adverse prognosis (poor responder patients). Taken together, these data show that MT2A contributes to chemotherapy resistance in osteosarcoma, an effect partially mediated by zinc chelation. The data also suggest that MT2A may be a potential new prognostic marker for osteosarcoma sensitivity to chemotherapy.


Assuntos
Quelantes/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Metalotioneína/metabolismo , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Zinco/metabolismo , Adolescente , Atorvastatina , Morte Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Criança , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Ácidos Heptanoicos/farmacologia , Humanos , Metalotioneína/genética , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteossarcoma/genética , Prognóstico , Conformação Proteica , Pirróis/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteína Supressora de Tumor p53/química , Proteína Supressora de Tumor p53/metabolismo
7.
Exp Cell Res ; 302(1): 129-42, 2005 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-15541732

RESUMO

To determine the molecular mechanisms by which N-cadherin transcription is regulated, we cloned and sequenced a 3681-bp of the 5'-flanking region of the human N-cadherin gene. Deletion analysis of the proximal region identified a minimal 318-bp region with strong promoter activity in human osteoblasts. The cryptic promoter is characterized by high GC content and a GA-rich binding core that may bind zing finger transcription factors. Electrophoretic mobility shift assays (EMSA), competition and supershift EMSA revealed that an Sp1/Sp3 binding site acts as a basal regulatory element of the promoter in osteoblasts. Incubation of osteoblast nuclear extracts with -163/-131 wild-type probe containing the GA-rich binding core revealed another specific complex, which was not formed with a -163/-131 probe mutated in the GA repeat. EMSA identified the nuclear factor involved as myeloid zinc finger-1 (MZF1). Mutation analysis showed that Sp1/Sp3 and MZF1 binding sites contribute to basal promoter activity. Cotransfection analyses showed that Sp1 and MZF1 overexpression increases whereas Sp3 antagonizes Sp1-induced N-cadherin promoter activity in osteoblasts. RT-PCR analysis showed that human osteoblastic cells express MZF1 and that Sp1/MZF1 overexpression increased N-cadherin expression. These results indicate that Sp1/Sp3 and MZF1 are important transcription factors regulating N-cadherin promoter activity and expression in osteoblasts.


Assuntos
Caderinas/genética , Proteínas de Ligação a DNA/metabolismo , Osteoblastos/metabolismo , Regiões Promotoras Genéticas/genética , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Região 5'-Flanqueadora/genética , Sequência de Aminoácidos/genética , Sequência de Bases/genética , Sítios de Ligação/genética , Diferenciação Celular/genética , Linhagem Celular Transformada , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Éxons/genética , Regulação da Expressão Gênica/genética , Genes Reguladores/genética , Humanos , Fatores de Transcrição Kruppel-Like , Sondas Moleculares , Dados de Sequência Molecular , Estrutura Terciária de Proteína/genética , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp3 , Fatores de Transcrição/genética , Regulação para Cima/genética
8.
Exp Cell Res ; 303(1): 138-47, 2005 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-15572034

RESUMO

The molecular mechanisms regulating the adipogenic differentiation of bone marrow stromal cells in vivo remain largely unknown. In this study, we investigated the regulatory effects of transforming growth factor beta-2 (TGF-beta2) on transcription factors involved in adipogenic differentiation induced by hind limb suspension in rat bone marrow stromal cells in vivo. Time course real-time quantitative reverse-transcription polymerase chain reaction (RT-PCR) analysis of gene expression showed that skeletal unloading progressively increases the expression of CCAAT/enhancer-binding protein (C/EBP)alpha and C/EBPbeta alpha at 5 days in bone marrow stromal cells resulting in increased peroxisome proliferator-activated receptor gamma (PPARgamma2) transcripts at 7 days. TGF-beta2 administration in unloaded rats corrected the rise in C/EBPalpha and C/EBPbeta transcripts induced by unloading in bone marrow stromal cells. This resulted in inhibition of PPARgamma2 expression that was associated with increased Runx2 expression. Additionally, the inhibition of C/EBPalpha and C/EBPbeta expression by TGF-beta2 was associated with increased PPARgamma serine phosphorylation in bone marrow stromal cells, a mechanism that inhibits PPARgamma transactivating activity. The sequential inhibitory effect of TGF-beta2 on C/EBPalpha, C/EBPbeta, and PPARgamma2 resulted in reduced LPL expression and abolition of bone marrow stromal cell adipogenic differentiation, which contributed to prevent bone loss induced by skeletal unloading. We conclude that TGF-beta2 inhibits the excessive adipogenic differentiation of bone marrow stromal cells induced by skeletal unloading by inhibiting C/EBPalpha, C/EBPbeta, and PPARgamma expression and activity, which provides a sequential mechanism by which TGF-beta2 regulates adipogenic differentiation of bone marrow stromal cells in vivo.


Assuntos
Células da Medula Óssea/citologia , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , PPAR gama/metabolismo , Células Estromais/citologia , Fator de Crescimento Transformador beta/farmacologia , Animais , Células da Medula Óssea/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/fisiologia , Masculino , Ratos , Ratos Wistar , Células Estromais/metabolismo , Fator de Crescimento Transformador beta2
9.
J Endocrinol Invest ; 25(6): 539-46, 2002 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-12109626

RESUMO

The key pharmacological action for the clinical use of bisphosphonates lies in the inhibition of osteoclast-mediated bone resorption. Osteoblasts could be other target cells for bisphosphonates. We studied the effects of bisphosphonates on the proliferation and the differentiation of normal human bone trabecular osteoblastic cells (hOB). We tested 4 different compounds: clodronate, pamidronate and 2 newer compounds: ibandronate, a nitrogen-containing bisphosphonate and zoledronate, which is a heterocyclic imidazole compound. Ibandronate and zoledronate stimulated hOB cell proliferation by up to 30% (p<0.05) after 72 h for concentrations ranging from 10(-8) M to 10(-5) M. Clodronate transiently enhanced hOB cell survival after only 24 h (+60%, p<0.001) whereas pamidronate had no effect. Longer time course studies, in presence of fetal calf serum, revealed that cell growth was finally reduced by all 4 bisphosphonates (40% after 7 days). Type I collagen synthesis was transiently increased by all 4 bisphosphonates after only 48 h incubation (+17% to +67%, p<0.05). Clodronate increased ALP activity by up to 1.7-fold after 4 days of culture (p<0.05) whereas ibandronate or zoledronate exhibited lesser stimulatory effects (+17 to +30%), and pamidronate had no significant effect. In conclusion, we found that different bisphosphonates, currently used or tested in various clinical conditions, transiently stimulated the growth of preosteoblastic cells and thereafter increased their differentiation according to sequential events (type I collagen synthesis first, then ALP activity to a lesser extent). Our data suggest that the beneficial effects of bisphosphonate treatment on bone mass and integrity could be partly mediated through a direct action on osteoblasts.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Difosfonatos/farmacologia , Osteoblastos/citologia , Adulto , Idoso , Fosfatase Alcalina/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Ácido Clodrônico/farmacologia , Colágeno Tipo I/biossíntese , Meios de Cultura , Meios de Cultura Livres de Soro , Sangue Fetal , Humanos , Ácido Ibandrônico , Imidazóis/farmacologia , Pessoa de Meia-Idade , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Pamidronato , Ácido Zoledrônico
10.
Cytokine ; 9(8): 613-23, 1997 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-9245490

RESUMO

We have studied the effects of rhTGF-beta2, dexamethanosome (dex) and 1,25(OH)2D3 on human bone marrow stromal (HBMS) cells in long-term culture. A fraction on HBMS expressed type I collagen (Col I) and osteopontin, and transient treatment (48 h) with dex increased the number of alkaline phosphatase (ALP) positive cells. Treatment with rhTGF-beta2 inhibited DNA synthesis and attenuated the stimulatory effect of dex on cell growth at 3-4 weeks of culture. 1,25(OH)2D3 inhibited DNA synthesis at 1-4 weeks, and dex partially blocked the inhibitory effect of 1,25(OH)2D3 on cell growth. rhTGF-beta2 and dex increased Col I synthesis at 1 week of culture. Both dex and 1,25(OH)2D3 increased ALP activity and mRNA levels independently, and when combined they had an additive or synergistic effect, whereas rhTGF-beta2 antagonized the stimulatory effect of dex on ALP activity. In addition, dex attenuated the increased osteocalcin expression induced by 1,25(OH)2D3. These results show that rhTGF-beta2, dex and 1,25(OH)2D3 have distinct effects and modulate the action of each other on human marrow stromal cell proliferation and differentiation at different time points during the culture.


Assuntos
Medula Óssea/efeitos dos fármacos , Calcitriol/farmacologia , Dexametasona/farmacologia , Fator de Crescimento Transformador beta/farmacologia , Idoso , Fosfatase Alcalina/genética , Fosfatase Alcalina/metabolismo , Células da Medula Óssea , Calcificação Fisiológica , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/biossíntese , Colágeno/genética , DNA/biossíntese , Expressão Gênica , Humanos , Osteocalcina/biossíntese , Osteocalcina/genética , Reação em Cadeia da Polimerase/métodos , Proteínas Recombinantes/farmacologia , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
11.
Br J Cancer ; 89(1): 178-84, 2003 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-12838321

RESUMO

Bone tissue constitutes a fertile 'soil' for metastatic tumours, notably breast cancer. High concentrations of growth factors in bone matrix favour cancer cell proliferation and survival, and a vicious cycle settles between bone matrix, osteoclasts and cancer cells. Classically, bisphosphonates interrupt this vicious cycle by inhibiting osteoclast-mediated bone resorption. We and others recently reported that bisphosphonates can also induce human breast cancer cell death in vitro, which could contribute to their beneficial clinical effects. We hypothesised that bisphosphonates could inhibit the favourable effects of 'bone-derived' growth factors, and indeed found that bisphosphonates reduced or abolished the stimulatory effects of growth factors (IGFs, FGF-2) on MCF-7 and T47D cell proliferation and inhibited their protective effects on apoptotic cell death in vitro under serum-free conditions. This could happen through an interaction with growth factors' intracellular phosphorylation transduction pathways, such as ERK1/2-MAPK. In conclusion, we report that bisphosphonates antagonised the stimulatory effects of growth factors on human breast cancer cell survival and reduced their protective effects against apoptotic cell death. Bisphosphonates and growth factors thus appear to be concurrent compounds for tumour cell growth and survival in bone tissue. This could represent a new mechanism of action of bisphosphonates in their protective effects against breast cancer-induced osteolysis.


Assuntos
Apoptose , Neoplasias Ósseas/secundário , Neoplasias da Mama/patologia , Sobrevivência Celular , Difosfonatos/farmacologia , Substâncias de Crescimento/farmacologia , Osso e Ossos/fisiologia , Feminino , Humanos , Osteólise/prevenção & controle , Fosforilação , Células Tumorais Cultivadas
12.
J Biol Chem ; 276(31): 29028-36, 2001 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-11395480

RESUMO

Bone morphogenetic protein-2 (BMP-2), a member of the transforming growth factor-beta (TGF-beta) family, regulates osteoblast differentiation and bone formation. Here we show a novel function of BMP-2 in human osteoblasts and identify a signaling pathway involved in this function. BMP-2 promotes apoptosis in primary human calvaria osteoblasts and in immortalized human neonatal calvaria osteoblasts, as shown by terminal deoxynucleotidyl transferase-mediated nick end labeling analysis. In contrast, TGF-beta 2 inhibits apoptosis in human osteoblasts. Studies of the mechanisms of action showed that BMP-2 increases the Bax/Bcl-2 ratio, whereas TG beta-2 has a negative effect. Moreover, BMP-2 increases the release of mitochondrial cytochrome c to the cytosol. Consistent with these results, BMP-2 increases caspase-9 and caspase-3, -6, and -7 activity, and an anti-caspase-9 agent suppresses BMP-2-induced apoptosis. Overexpression of dominant-negative Smad1 effectively blocks BMP-2-induced expression of the osteoblast transcription factor Runx2 but not the activation of caspases or apoptosis induced by BMP-2, indicating that the Smad1 signaling pathway is not involved in the BMP-2-induced apoptosis. The proapoptotic effect of BMP-2 is PKC-dependent, because BMP-2 increases PKC activity, and the selective PKC inhibitor calphostin C blocks the BMP-2-induced increased Bax/Bcl-2, caspase activity, and apoptosis. In contrast, the cAMP-dependent protein kinase A inhibitor H89, the p38 MAPK inhibitor SB203580, and the MEK inhibitor PD-98059 have no effect. The results show that BMP-2 uses a Smad-independent, PKC-dependent pathway to promote apoptosis via a Bax/Bcl-2 and cytochrome c-caspase-9-caspase-3, -6, -7 cascade in human osteoblasts.


Assuntos
Apoptose/fisiologia , Proteínas Morfogenéticas Ósseas/farmacologia , Proteínas de Neoplasias , Osteoblastos/citologia , Osteoblastos/fisiologia , Proteína Quinase C/metabolismo , Transdução de Sinais/fisiologia , Sulfonamidas , Apoptose/efeitos dos fármacos , Proteína Morfogenética Óssea 2 , Caspases/metabolismo , Células Cultivadas , Subunidade alfa 1 de Fator de Ligação ao Core , Grupo dos Citocromos c/metabolismo , Citosol/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Etoposídeo/farmacologia , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Marcação In Situ das Extremidades Cortadas , Recém-Nascido , Isoquinolinas/farmacologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Naftalenos/farmacologia , Osteoblastos/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Recombinantes/farmacologia , Transdução de Sinais/efeitos dos fármacos , Crânio/citologia , Proteínas Smad , Proteína Smad1 , Transativadores/genética , Transativadores/metabolismo , Fatores de Transcrição/genética , Fator de Crescimento Transformador beta/farmacologia , Proteína X Associada a bcl-2
13.
J Cell Biochem ; 68(4): 411-26, 1998 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-9493905

RESUMO

Osteoprogenitor cells in the human bone marrow stroma can be induced to differentiate into osteoblasts under stimulation with hormonal and local factors. We previously showed that human bone marrow stromal (HBMS) cells respond to dexamethasone and vitamin D by expressing several osteoblastic markers. In this study, we investigated the effects and interactions of local factors (BMP-2 and TGF-beta2) on HBMS cell proliferation and differentiation in short-term and long-term cultures. We found that rhTGF-beta2 increased DNA content and stimulated type I collagen synthesis, but inhibited ALP activity and mRNA levels, osteocalcin production, and mineralization of the matrix formed by HBMS cells. In contrast, rhBMP-2 increased ALP activity and mRNA levels, osteocalcin levels and calcium deposition in the extracellular matrix without affecting type I collagen synthesis and mRNA levels, showing that rhBMP-2 and rhTGF-beta2 regulate differentially HBMS cells. Co-treatment with rhBMP-2 and rhTGF-beta2 led to intermediate effects on HBMS cell proliferation and differentiation markers. rhTGF-beta2 attenuated the stimulatory effect of rhBMP-2 on osteocalcin levels, and ALP activity and mRNA levels, whereas rhBMP-2 reduced the rhTGF-beta2-enhanced DNA synthesis and type I collagen synthesis. We also investigated the effects of sequential treatments with rhBMP-2 and rhTGF-beta2 on HBMS cell differentiation in long-term culture. A transient (9 days) treatment with rhBMP-2 abolished the rhTGF-beta2 response of HBMS cells on ALP activity. In contrast, a transient (10 days) treatment with rhTGF-beta2 did not influence the subsequent rhBMP-2 action on HBMS cell differentiation. The data show that TGF-beta2 acts by increasing HBMS cell proliferation and type I collagen synthesis whereas BMP-2 acts by promoting HBMS cell differentiation. These observations suggest that TGF-beta2 and BMP-2 may act in a sequential manner at different stages to promote human bone marrow stromal cell differentiation towards the osteoblast phenotype.


Assuntos
Células da Medula Óssea/citologia , Proteínas Morfogenéticas Ósseas/farmacologia , Osteoblastos/citologia , Fator de Crescimento Transformador beta/farmacologia , Fosfatase Alcalina/metabolismo , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Proteína Morfogenética Óssea 2 , Calcificação Fisiológica/efeitos dos fármacos , Cálcio/análise , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/biossíntese , DNA/análise , Relação Dose-Resposta a Droga , Interações Medicamentosas , Expressão Gênica , Humanos , Osteocalcina/biossíntese , Proteínas Recombinantes/farmacologia , Células Estromais/citologia , Células Estromais/efeitos dos fármacos , Células Estromais/metabolismo
14.
Calcif Tissue Int ; 64(5): 394-401, 1999 May.
Artigo em Inglês | MEDLINE | ID: mdl-10203416

RESUMO

In the present study, we established a new adult human trabecular osteoblastic (AHTO) cell line, immortalized by SV-40 Large T (LT) oncogene. From seven proliferative colonies identified, we selected clone 7 with high alkaline phosphatase (ALP) activity for further analysis. AHTO-7 cells were able to grow for at least 8 months and 25 passages, with a doubling time of about 22 hours. Immunocytochemistry staining and RT-PCR analysis indicated that the extended life-span of AHTO-7 cells results in genomic insertion of SV-40 LT oncogene. The cells responded to PTH and PGE2 in terms of cAMP accumulation. The time course study, in the presence of 10(-8) M vitamin D3 (vit D3) showed a marked increase (fourfold) in ALP activity with a peak at day 3. Furthermore, in the presence of ascorbic acid (50 microg/ml) and inorganic phosphate (3 mM), AHTO-7 cells produced abundant calcified extracellular matrix, as examined by the von Kossa staining after 2 weeks of culture. Molecular analysis of mRNAs for phenotypic osteoblast markers at day 15 showed the expression of ALP, osteocalcin (OC), and collagen type I (Col I) mRNAs constitutively. Col I expression was inhibited by vit D3 and dexamethasone treatment. In contrast, treatment with vit D3 induced a marked increase of ALP and OC transcripts. Therefore, the immortalized AHTO-7 cells express osteoblast markers that are induced by calciotropic hormones, and constitute a suitable model for identifying specific osteoblastic genes and their regulation during human osteoblast differentiation.


Assuntos
Antígenos Transformantes de Poliomavirus/genética , Osso e Ossos/citologia , Transformação Celular Viral/genética , Técnicas de Transferência de Genes , Osteoblastos/citologia , Vírus 40 dos Símios/imunologia , Idoso , Fosfatase Alcalina/metabolismo , Ácido Ascórbico/farmacologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/virologia , Calcificação Fisiológica , Diferenciação Celular , Divisão Celular , Colecalciferol/farmacologia , Colágeno/metabolismo , Primers do DNA/química , DNA Viral/análise , Dexametasona/farmacologia , Matriz Extracelular/efeitos dos fármacos , Matriz Extracelular/metabolismo , Humanos , Masculino , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/virologia , Osteocalcina/metabolismo , Fenótipo , Fosfatos/farmacologia , RNA/análise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Am J Pathol ; 158(5): 1833-42, 2001 May.
Artigo em Inglês | MEDLINE | ID: mdl-11337381

RESUMO

Apert syndrome is an autosomal dominant disorder characterized by premature cranial ossification resulting from fibroblast growth factor receptor-2 (FGFR-2)-activating mutations. We have studied the effects of the prominent S252W FGFR-2 Apert mutation on apoptosis and the underlying mechanisms in human mutant osteoblasts. In vivo analysis of terminal deoxynucleotidyl transferase-mediated nick-end labeling revealed premature apoptosis of mature osteoblasts and osteocytes in the Apert suture compared to normal coronal suture. In vitro, mutant osteoblasts showed increased apoptosis, as demonstrated by terminal deoxynucleotidyl transferase-mediated nick-end labeling analysis, trypan blue staining, and DNA fragmentation. Mutant osteoblasts also showed increased activity of caspase-8 and effector caspases (-3, -6, -7) constitutively. This was related to protein kinase C activation because the selective protein kinase C inhibitor calphostin C inhibited caspase-8, effector caspases, and apoptosis in mutant osteoblasts. Apert osteoblasts also showed increased expression of interleukin (IL)-1alpha, IL-1beta, Fas, and Bax, and decreased Bcl-2 levels. Specific neutralizing anti-IL-1 antibody reduced Fas levels, Bax expression, effector caspases activity, and apoptosis in mutant cells. Thus, the Apert S252W FGFR-2 mutation promotes apoptosis in human osteoblasts through activation of protein kinase C, overexpression of IL-1 and Fas, activation of caspase-8, and increased Bax/Bcl-2 levels, leading to increased effector caspases and DNA fragmentation. This identifies a complex FGFR-2 signaling pathway involved in the premature apoptosis induced by the Apert S252W FGFR-2 mutation in human calvaria osteoblasts.


Assuntos
Acrocefalossindactilia/patologia , Apoptose , Craniossinostoses/patologia , Osteoblastos/patologia , Acrocefalossindactilia/genética , Substituição de Aminoácidos , Caspase 3 , Caspase 8 , Caspase 9 , Caspases/metabolismo , Células Cultivadas , Craniossinostoses/genética , Humanos , Interleucina-1/metabolismo , Interleucina-1/fisiologia , Mutação , Osteoblastos/metabolismo , Proteína Quinase C/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/genética , Proteína X Associada a bcl-2
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA