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.
J Clin Invest ; 122(10): 3579-92, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22996695

RESUMO

The adenosine diphosphate (ADP) receptor P2RY12 (purinergic receptor P2Y, G protein coupled, 12) plays a critical role in platelet aggregation, and P2RY12 inhibitors are used clinically to prevent cardiac and cerebral thrombotic events. Extracellular ADP has also been shown to increase osteoclast (OC) activity, but the role of P2RY12 in OC biology is unknown. Here, we examined the role of mouse P2RY12 in OC function. Mice lacking P2ry12 had decreased OC activity and were partially protected from age-associated bone loss. P2ry12-/- OCs exhibited intact differentiation markers, but diminished resorptive function. Extracellular ADP enhanced OC adhesion and resorptive activity of WT, but not P2ry12-/-, OCs. In platelets, ADP stimulation of P2RY12 resulted in GTPase Ras-related protein (RAP1) activation and subsequent αIIbß3 integrin activation. Likewise, we found that ADP stimulation induced RAP1 activation in WT and integrin ß3 gene knockout (Itgb3-/-) OCs, but its effects were substantially blunted in P2ry12-/- OCs. In vivo, P2ry12-/- mice were partially protected from pathologic bone loss associated with serum transfer arthritis, tumor growth in bone, and ovariectomy-induced osteoporosis: all conditions associated with increased extracellular ADP. Finally, mice treated with the clinical inhibitor of P2RY12, clopidogrel, were protected from pathologic osteolysis. These results demonstrate that P2RY12 is the primary ADP receptor in OCs and suggest that P2RY12 inhibition is a potential therapeutic target for pathologic bone loss.


Assuntos
Difosfato de Adenosina/fisiologia , Remodelação Óssea/fisiologia , Osteoclastos/fisiologia , Osteoporose/fisiopatologia , Receptores Purinérgicos P2Y12/fisiologia , Animais , Artrite Experimental/complicações , Neoplasias Ósseas/complicações , Neoplasias Ósseas/secundário , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/fisiopatologia , Carcinoma/complicações , Carcinoma/secundário , Adesão Celular/efeitos dos fármacos , Células Cultivadas/efeitos dos fármacos , Clopidogrel , Ativação Enzimática/efeitos dos fármacos , Feminino , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Osteoporose/etiologia , Osteoporose/prevenção & controle , Ovariectomia , Fosfatidilinositol 3-Quinases/fisiologia , Inibidores de Fosfoinositídeo-3 Quinase , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Receptores Purinérgicos P2Y12/deficiência , Receptores Purinérgicos P2Y12/efeitos dos fármacos , Receptores Purinérgicos P2Y12/genética , Organismos Livres de Patógenos Específicos , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia , Ticlopidina/uso terapêutico , Proteínas rap1 de Ligação ao GTP/efeitos dos fármacos
2.
PLoS One ; 5(12): e15755, 2010 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-21209895

RESUMO

The ARF tumor suppressor regulates p53 as well as basic developmental processes independent of p53, including osteoclast activation, by controlling ribosomal biogenesis. Here we provide evidence that ARF is a master regulator of bone remodeling and osteosarcoma (OS) development in mice. Arf(-/-) mice displayed increased osteoblast (OB) and osteoclast (OC) activity with a significant net increase in trabecular bone volume. The long bones of Arf(-/-) mice had increased expression of OB genes while Arf(-/-) OB showed enhanced differentiation in vitro. Mice transgenic for the Tax oncogene develop lymphocytic tumors with associated osteolytic lesions, while Tax+Arf(-/-) mice uniformly developed spontaneous OS by 7 months of age. Tax+Arf(-/-) tumors were well differentiated OS characterized by an abundance of new bone with OC recruitment, expressed OB markers and displayed intact levels of p53 mRNA and reduced Rb transcript levels. Cell lines established from OS recapitulated characteristics of the primary tumor, including the expression of mature OB markers and ability to form mineralized tumors when transplanted. Loss of heterozygosity in OS tumors arising in Tax+Arf(+/-) mice emphasized the necessity of ARF-loss in OS development. Hypothesizing that inhibition of ARF-regulated bone remodeling would repress development of OS, we demonstrated that treatment of Tax+Arf(-/-) mice with zoledronic acid, a bisphosphonate inhibitor of OC activity and repressor of bone turnover, prevented or delayed the onset of OS. These data describe a novel role for ARF as a regulator of bone remodeling through effects on both OB and OC. Finally, these data underscore the potential of targeting bone remodeling as adjuvant therapy or in patients with genetic predispositions to prevent the development of OS.


Assuntos
Carcinoma/genética , Produtos do Gene tax/genética , Osteoblastos/citologia , Osteoclastos/citologia , Proteína Supressora de Tumor p14ARF/genética , Animais , Remodelação Óssea , Difosfonatos/química , Difosfonatos/farmacologia , Genes p53 , Heterozigoto , Imidazóis/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Genéticos , Proteína Supressora de Tumor p53/metabolismo , Ácido Zoledrônico
3.
Bone ; 44(5): 908-16, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19442620

RESUMO

Bisphosphonates (BPs), bone targeted drugs that disrupt osteoclast function, are routinely used to treat complications of bone metastasis. Studies in preclinical models of cancer have shown that BPs reduce skeletal tumor burden and increase survival. Similarly, we observed in the present study that administration of the Nitrogen-containing BP (N-BP), zoledronic acid (ZA) to osteolytic tumor-bearing Tax+ mice beginning at 6 months of age led to resolution of radiographic skeletal lesions. N-BPs inhibit farnesyl diphosphate (FPP) synthase, thereby inhibiting protein prenylation and causing cellular toxicity. We found that ZA decreased Tax+ tumor and B16 melanoma viability and caused the accumulation of unprenylated Rap1a proteins in vitro. However, it is presently unclear whether N-BPs exert anti-tumor effects in bone independent of inhibition of osteoclast (OC) function in vivo. Therefore, we evaluated the impact of treatment with ZA on B16 melanoma bone tumor burden in irradiated mice transplanted with splenic cells from src(-/-) mice, which have non-functioning OCs. OC-defective mice treated with ZA demonstrated a significant 88% decrease in tumor growth in bone compared to vehicle-treated OC-defective mice. These data support an osteoclast-independent role for N-BP therapy in bone metastasis.


Assuntos
Neoplasias Ósseas/tratamento farmacológico , Difosfonatos/farmacologia , Imidazóis/farmacologia , Osteoclastos/patologia , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Difosfonatos/administração & dosagem , Ativação Enzimática/efeitos dos fármacos , Geraniltranstransferase/metabolismo , Imidazóis/administração & dosagem , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Metástase Neoplásica/prevenção & controle , Osteoclastos/efeitos dos fármacos , Prenilação de Proteína/efeitos dos fármacos , Ácido Zoledrônico
4.
J Biol Chem ; 284(7): 4658-66, 2009 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-19059914

RESUMO

Interferon-gamma (IFN-gamma) has been shown to enhance anti-tumor immunity and inhibit the formation of bone-resorbing osteoclasts. We evaluated the role of IFN-gamma in bone metastases, tumor-associated bone destruction, and hypercalcemia in human T cell lymphotrophic virus type 1-Tax transgenic mice. Compared with Tax(+)IFN-gamma(+/+) mice, Tax(+)IFN-gamma(-/-) mice developed increased osteolytic bone lesions and soft tissue tumors, as well as increased osteoclast formation and activity. In vivo administration of IFN-gamma to tumor-bearing Tax(+)IFN-gamma(-/-) mice prevented new tumor development and resulted in decreased bromodeoxyuridine uptake by established tumors. In vitro, IFN-gamma directly decreased the viability of Tax(+) tumor cells through inhibition of proliferation, suppression of ERK phosphorylation, and induction of apoptosis and caspase 3 cleavage. IFN-gamma also inhibited macrophage colonystimulating factor-mediated proliferation and survival of osteoclast progenitors in vitro. Administration of IFN-gamma to C57BL/6 mice decreased Tax(+) tumor growth and prevented tumor-associated bone loss and hypercalcemia. In contrast, IFN-gamma treatment failed to protect IFN-gammaR1(-/-) mice from Tax(+) tumor-induced skeletal complications, despite decreasing tumor growth. These data demonstrate that IFN-gamma suppressed tumor-induced bone loss and hypercalcemia in Tax(+) mice by inhibiting both Tax(+) tumor cell growth and host-induced osteolysis. These data suggest a protective role for IFN-gamma in patients with bone metastases and hypercalcemia of malignancy.


Assuntos
Neoplasias Ósseas/metabolismo , Reabsorção Óssea/metabolismo , Vírus Linfotrópico T Tipo 1 Humano , Interferon gama/farmacologia , Osteoclastos/metabolismo , Neoplasias de Tecidos Moles/metabolismo , Animais , Neoplasias Ósseas/complicações , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/genética , Neoplasias Ósseas/secundário , Reabsorção Óssea/etiologia , Reabsorção Óssea/genética , Reabsorção Óssea/patologia , Reabsorção Óssea/prevenção & controle , Produtos do Gene tax/genética , Produtos do Gene tax/metabolismo , Humanos , Hipercalcemia/tratamento farmacológico , Hipercalcemia/etiologia , Hipercalcemia/genética , Hipercalcemia/metabolismo , Hipercalcemia/patologia , Interferon gama/genética , Interferon gama/metabolismo , Camundongos , Camundongos Knockout , Metástase Neoplásica , Osteoclastos/patologia , Neoplasias de Tecidos Moles/complicações , Neoplasias de Tecidos Moles/tratamento farmacológico , Neoplasias de Tecidos Moles/patologia
5.
J Cell Biochem ; 104(4): 1311-23, 2008 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-18260128

RESUMO

Platelets contribute to the development of metastasis, the most common cause of mortality in cancer patients, but the precise role that anti-platelet drugs play in cancer treatment is not defined. Metastatic tumor cells can produce platelet alphaIIb beta3 activators, such as ADP and thromboxane A(2) (TXA(2)). Inhibitors of platelet beta3 integrins decrease bone metastases in mice but are associated with significant bleeding. We examined the role of a novel soluble apyrase/ADPase, APT102, and an inhibitor of TXA(2) synthesis, acetylsalicylic acid (aspirin or ASA), in mouse models of experimental bone metastases. We found that treatment with ASA and APT102 in combination (ASA + APT102), but not either drug alone, significantly decreased breast cancer and melanoma bone metastases in mice with fewer bleeding complications than observed with alphaIIb beta3 inhibition. ASA + APT102 diminished tumor cell induced platelet aggregation but did not directly alter tumor cell viability. Notably, APT102 + ASA treatment did not affect initial tumor cell distribution and similar results were observed in beta3-/- mice. These results show that treatment with ASA + APT102 decreases bone metastases without significant bleeding complications. Anti-platelet drugs such as ASA + APT102 could be valuable experimental tools for studying the role of platelet activation in metastasis as well as a therapeutic option for the prevention of bone metastases.


Assuntos
Apirase/uso terapêutico , Aspirina/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/secundário , Metástase Neoplásica/tratamento farmacológico , Inibidores da Agregação Plaquetária/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Apirase/farmacologia , Aspirina/farmacologia , Diagnóstico por Imagem , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/patologia , Camundongos , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Carga Tumoral/efeitos dos fármacos
6.
Proc Natl Acad Sci U S A ; 104(35): 14062-7, 2007 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-17715292

RESUMO

CXCR4 regulates hematopoietic and tumor cell homing to bone, but its role during osteoclast (OC) development is unknown. We investigated the role of CXCR4 in osteoclastogenesis and in a model of bone metastasis. Compared with controls, mice reconstituted with CXCR4 null hematopoietic cells exhibited elevated markers of bone resorption, increased OC perimeter along bone, and increased bone loss. CXCR4-/- OCs demonstrated accelerated differentiation and enhanced bone resorption in vitro. Furthermore, tumor growth specifically in bone was significantly increased in mice reconstituted with CXCR4-/- hematopoietic cells. Finally, enhancement of bone tumor growth in the absence of CXCR4 was abrogated with the OC inhibitor, zoledronic acid. These data demonstrate that disruption of CXCR4 enhances osteoclastogenesis and suggest that inhibition of CXCR4 may enhance established skeletal tumor burden by increasing OC activity.


Assuntos
Neoplasias Ósseas/patologia , Receptores CXCR4/genética , Animais , Conservadores da Densidade Óssea/uso terapêutico , Neoplasias Ósseas/genética , Reabsorção Óssea , Divisão Celular , Difosfonatos/uso terapêutico , Imidazóis/uso terapêutico , Cinética , Macrófagos/patologia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Metástase Neoplásica , Osteoclastos/patologia , Receptores CXCR4/deficiência , Ácido Zoledrônico
7.
Blood ; 109(8): 3424-31, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17192391

RESUMO

Inhibition of osteoclast (OC) activity has been associated with decreased tumor growth in bone in animal models. Increased recognition of factors that promote osteoclastic bone resorption in cancer patients led us to investigate whether increased OC activation could enhance tumor growth in bone. Granulocyte colony-stimulating factor (G-CSF) is used to treat chemotherapy-induced neutropenia, but is also associated with increased markers of OC activity and decreased bone mineral density (BMD). We used G-CSF as a tool to investigate the impact of increased OC activity on tumor growth in 2 murine osteolytic tumor models. An 8-day course of G-CSF alone (without chemotherapy) significantly decreased BMD and increased OC perimeter along bone in mice. Mice administered G-CSF alone demonstrated significantly increased tumor growth in bone as quantitated by in vivo bioluminescence imaging and histologic bone marrow tumor analysis. Short-term administration of AMD3100, a CXCR4 inhibitor that mobilizes neutrophils with little effect on bone resorption, did not lead to increased tumor burden. However, OC-defective osteoprotegerin transgenic (OPG(Tg)) mice and bisphosphonate-treated mice were resistant to the effects of G-CSF administration upon bone tumor growth. These data demonstrate a G-CSF-induced stimulation of tumor growth in bone that is OC dependent.


Assuntos
Densidade Óssea/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Reabsorção Óssea/metabolismo , Fator Estimulador de Colônias de Granulócitos/efeitos adversos , Osteoclastos/metabolismo , Carga Tumoral/efeitos dos fármacos , Animais , Fármacos Anti-HIV/farmacologia , Benzilaminas , Neoplasias Ósseas/patologia , Neoplasias Ósseas/fisiopatologia , Reabsorção Óssea/patologia , Reabsorção Óssea/fisiopatologia , Movimento Celular/efeitos dos fármacos , Ciclamos , Fator Estimulador de Colônias de Granulócitos/farmacologia , Compostos Heterocíclicos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias Experimentais , Neutropenia/tratamento farmacológico , Neutrófilos/metabolismo , Neutrófilos/patologia , Osteoclastos/patologia , Receptores CXCR4/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA