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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Blood ; 119(18): 4205-14, 2012 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-22427205

RESUMO

BM stromal cells (BMSCs) are key players in the microenvironmental support of multiple myeloma (MM) cell growth and bone destruction. A spliced form of the X-box-binding protein-1 (XBP1s), a major proximal effector of unfolded protein response signaling, is highly expressed in MM cells and plays an indispensable role in MM pathogenesis. In the present study, we found that XBP1s is induced in the BMSCs of the MM microenvironment. XBP1s overexpression in healthy human BMSCs enhanced gene and/or protein expression of VCAM-1, IL-6, and receptor activator of NF-κB ligand (RANKL), enhancing BMSC support of MM cell growth and osteoclast formation in vitro and in vivo. Conversely, deficiency of XBP1 in healthy donor BMSCs displayed a range of effects on BMSCs that were opposite to those cells with overexpression of XBP1s. Knock-down of XBP1 in MM patient BMSCs greatly compromised their increased VCAM-1 protein expression and IL-6 and RANKL secretion in response to TNFα and reversed their enhanced support of MM-cell growth and osteoclast formation. Our results demonstrate that XBP1s is a pathogenic factor underlying BMSC support of MM cell growth and osteoclast formation and therefore represents a therapeutic target for MM bone disease.


Assuntos
Células da Medula Óssea/metabolismo , Proteínas de Ligação a DNA/fisiologia , Mieloma Múltiplo/patologia , Proteínas de Neoplasias/fisiologia , Osteoclastos/patologia , Células Estromais/metabolismo , Fatores de Transcrição/fisiologia , Animais , Células da Medula Óssea/patologia , Diferenciação Celular , Divisão Celular , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/metabolismo , Técnicas de Cocultura , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Endorribonucleases/deficiência , Endorribonucleases/genética , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Inflamação , Interleucina-6/biossíntese , Interleucina-6/genética , Interleucina-6/metabolismo , Camundongos , Camundongos SCID , Mieloma Múltiplo/metabolismo , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Proteínas Serina-Treonina Quinases/deficiência , Proteínas Serina-Treonina Quinases/genética , Ligante RANK/biossíntese , Ligante RANK/genética , Ligante RANK/metabolismo , Interferência de RNA , Proteínas Recombinantes de Fusão/fisiologia , Fatores de Transcrição de Fator Regulador X , Células Estromais/patologia , Fatores de Transcrição/antagonistas & inibidores , Fatores de Transcrição/deficiência , Fatores de Transcrição/genética , Microambiente Tumoral , Fator de Necrose Tumoral alfa/farmacologia , Molécula 1 de Adesão de Célula Vascular/biossíntese , Molécula 1 de Adesão de Célula Vascular/genética , Proteína 1 de Ligação a X-Box
2.
Blood ; 118(26): 6871-80, 2011 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-22042697

RESUMO

Protracted inhibition of osteoblast (OB) differentiation characterizes multiple myeloma (MM) bone disease and persists even when patients are in long-term remission. However, the underlying pathophysiology for this prolonged OB suppression is unknown. Therefore, we developed a mouse MM model in which the bone marrow stromal cells (BMSCs) remained unresponsive to OB differentiation signals after removal of MM cells. We found that BMSCs from both MM-bearing mice and MM patients had increased levels of the transcriptional repressor Gfi1 compared with controls and that Gfi1 was a novel transcriptional repressor of the critical OB transcription factor Runx2. Trichostatin-A blocked the effects of Gfi1, suggesting that it induces epigenetic changes in the Runx2 promoter. MM-BMSC cell-cell contact was not required for MM cells to increase Gfi1 and repress Runx2 levels in MC-4 before OBs or naive primary BMSCs, and Gfi1 induction was blocked by anti-TNF-α and anti-IL-7 antibodies. Importantly, BMSCs isolated from Gfi1(-/-) mice were significantly resistant to MM-induced OB suppression. Strikingly, siRNA knockdown of Gfi1 in BMSCs from MM patients significantly restored expression of Runx2 and OB differentiation markers. Thus, Gfi1 may have an important role in prolonged MM-induced OB suppression and provide a new therapeutic target for MM bone disease.


Assuntos
Neoplasias Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Mieloma Múltiplo/metabolismo , Osteoblastos/metabolismo , Células Estromais/metabolismo , Fatores de Transcrição/metabolismo , Células 3T3 , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Células da Medula Óssea/patologia , Neoplasias Ósseas/genética , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Proteínas de Ligação a DNA/genética , Feminino , Expressão Gênica , Humanos , Interleucina-7/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Mieloma Múltiplo/genética , Mieloma Múltiplo/patologia , Osteoblastos/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Células Estromais/patologia , Fatores de Transcrição/genética , Fator de Necrose Tumoral alfa/metabolismo
3.
Blood ; 115(1): 140-9, 2010 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-19887675

RESUMO

Increased osteoclastogenesis and angiogenesis occur in physiologic and pathologic conditions. However, it is unclear if or how these processes are linked. To test the hypothesis that osteoclasts stimulate angiogenesis, we modulated osteoclast formation in fetal mouse metatarsal explants or in adult mice and determined the effect on angiogenesis. Suppression of osteoclast formation with osteoprotegerin dose-dependently inhibited angiogenesis and osteoclastogenesis in metatarsal explants. Conversely, treatment with parathyroid hormone related protein (PTHrP) increased explant angiogenesis, which was completely blocked by osteoprotegerin. Further, treatment of mice with receptor activator of nuclear factor-kappaB ligand (RANKL) or PTHrP in vivo increased calvarial vessel density and osteoclast number. We next determined whether matrix metalloproteinase-9 (MMP-9), an angiogenic factor predominantly produced by osteoclasts in bone, was important for osteoclast-stimulated angiogenesis. The pro-angiogenic effects of PTHrP or RANKL were absent in metatarsal explants or calvaria in vivo, respectively, from Mmp9(-/-) mice, demonstrating the importance of MMP-9 for osteoclast-stimulated angiogenesis. Lack of MMP-9 decreased osteoclast numbers and abrogated angiogenesis in response to PTHrP or RANKL in explants and in vivo but did not decrease osteoclast differentiation in vitro. Thus, MMP-9 modulates osteoclast-stimulated angiogenesis primarily by affecting osteoclasts, most probably by previously reported migratory effects on osteoclasts. These results clearly demonstrate that osteoclasts stimulate angiogenesis in vivo through MMP-9.


Assuntos
Ossos do Metatarso/irrigação sanguínea , Neovascularização Fisiológica , Osteoclastos/fisiologia , Indutores da Angiogênese/metabolismo , Animais , Feminino , Feto/irrigação sanguínea , Feto/efeitos dos fármacos , Humanos , Masculino , Metaloproteinase 9 da Matriz/deficiência , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Ossos do Metatarso/efeitos dos fármacos , Ossos do Metatarso/embriologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Neovascularização Fisiológica/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoclastos/enzimologia , Proteína Relacionada ao Hormônio Paratireóideo/farmacologia , Ligante RANK/farmacologia , Crânio/citologia , Crânio/efeitos dos fármacos , Crânio/enzimologia , Regulação para Cima/efeitos dos fármacos
4.
Am J Pathol ; 174(3): 727-35, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19218349

RESUMO

Autosomal recessive osteopetrosis (ARO) is a group of genetic disorders that involve defects that preclude the normal function of osteoclasts, which differentiate from hematopoietic precursors. In half of human cases, ARO is the result of mutations in the TCIRG1 gene, which codes for a subunit of the vacuolar proton pump that plays a fundamental role in the acidification of the cell-bone interface. Functional mutations of this pump severely impair the resorption of bone mineral. Although postnatal hematopoietic stem cell transplantation can partially rescue the hematological phenotype of ARO, other stigmata of the disease, such as secondary neurological and growth defects, are not reversed. For this reason, ARO is a paradigm for genetic diseases that would benefit from effective prenatal treatment. Using the oc/oc mutant mouse, a murine model whose osteopetrotic phenotype closely recapitulates human TCIRG1-dependent ARO, we report that in utero transplantation of adult bone marrow hematopoietic stem cells can correct the ARO phenotype in a limited number of mice. Here we report that in utero injection of allogeneic fetal liver cells, which include hematopoietic stem cells, into oc/oc mouse fetuses at 13.5 days post coitum produces a high level of engraftment, and the oc/oc phenotype is completely rescued in a high percentage of these mice. Therefore, oc/oc pathology appears to be particularly sensitive to this form of early treatment of the ARO genetic disorder.


Assuntos
Transplante de Tecido Fetal , Transplante de Células-Tronco Hematopoéticas , Transplante de Fígado , Mutação , Osteopetrose/genética , Osteopetrose/cirurgia , ATPases Vacuolares Próton-Translocadoras/genética , Animais , Cruzamentos Genéticos , Primers do DNA , Modelos Animais de Doenças , Feminino , Feto , Genótipo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Osteopetrose/embriologia , Osteopetrose/patologia , Fenótipo , Reação em Cadeia da Polimerase , Gravidez
5.
J Bone Miner Res ; 26(1): 169-81, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20683884

RESUMO

ADAM8 expression is increased in the interface tissue around a loosened hip prosthesis and in the pannus and synovium of patients with rheumatoid arthritis, but its potential role in these processes is unclear. ADAM8 stimulates osteoclast (OCL) formation, but the effects of overexpression or loss of expression of ADAM8 in vivo and the mechanisms responsible for the effects of ADAM8 on osteoclastogenesis are unknown. Therefore, to determine the effects of modulating ADAM expression, we generated tartrate-resistant acid phosphatase (TRAP)-ADAM8 transgenic mice that overexpress ADAM8 in the OCL lineage and ADAM8 knockout (ADAM8 KO) mice. TRAP-ADAM8 mice developed osteopenia and had increased numbers of OCL precursors that formed hypermultinucleated OCLs with an increased bone-resorbing capacity per OCL. They also had an enhanced differentiation capacity, increased TRAF6 expression, and increased NF-κB, Erk, and Akt signaling compared with wild-type (WT) littermates. This increased bone-resorbing capacity per OCL was associated with increased levels of p-Pyk2 and p-Src activation. In contrast, ADAM8 KO mice did not display a bone phenotype in vivo, but unlike WT littermates, they did not increase RANKL production, OCL formation, or calvarial fibrosis in response to tumor necrosis factor α (TNF-α) in vivo. Since loss of ADAM8 does not inhibit basal bone remodeling but only blocks the enhanced OCL formation in response to TNF-α, these results suggest that ADAM8 may be an attractive therapeutic target for preventing bone destruction associated with inflammatory disease.


Assuntos
Proteínas ADAM/metabolismo , Antígenos CD/metabolismo , Proteínas de Membrana/metabolismo , Osteoclastos/citologia , Osteoclastos/enzimologia , Células-Tronco/citologia , Células-Tronco/enzimologia , Fosfatase Ácida/metabolismo , Animais , Biomarcadores/metabolismo , Reabsorção Óssea/patologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Fusão Celular , Ativação Enzimática/efeitos dos fármacos , Isoenzimas/metabolismo , Camundongos , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Tamanho do Órgão/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante RANK/farmacologia , Transdução de Sinais/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Fosfatase Ácida Resistente a Tartarato , Fator de Necrose Tumoral alfa/farmacologia , Quinases da Família src/metabolismo
6.
J Clin Invest ; 120(8): 2755-66, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20628199

RESUMO

Activating transcription factor 4 (ATF4) is a critical transcription factor for osteoblast (OBL) function and bone formation; however, a direct role in osteoclasts (OCLs) has not been established. Here, we targeted expression of ATF4 to the OCL lineage using the Trap promoter or through deletion of Atf4 in mice. OCL differentiation was drastically decreased in Atf4-/- bone marrow monocyte (BMM) cultures and bones. Coculture of Atf4-/- BMMs with WT OBLs or a high concentration of RANKL failed to restore the OCL differentiation defect. Conversely, Trap-Atf4-tg mice displayed severe osteopenia with dramatically increased osteoclastogenesis and bone resorption. We further showed that ATF4 was an upstream activator of the critical transcription factor Nfatc1 and was critical for RANKL activation of multiple MAPK pathways in OCL progenitors. Furthermore, ATF4 was crucial for M-CSF induction of RANK expression on BMMs, and lack of ATF4 caused a shift in OCL precursors to macrophages. Finally, ATF4 was largely modulated by M-CSF signaling and the PI3K/AKT pathways in BMMs. These results demonstrate that ATF4 plays a direct role in regulating OCL differentiation and suggest that it may be a therapeutic target for treating bone diseases associated with increased OCL activity.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Diferenciação Celular , Osteoclastos/citologia , Fator 4 Ativador da Transcrição/genética , Animais , Doenças Ósseas Metabólicas/etiologia , Células da Medula Óssea/patologia , Reabsorção Óssea/etiologia , Fator Estimulador de Colônias de Macrófagos/fisiologia , Macrófagos/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/fisiologia , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Ligante RANK/metabolismo
7.
PLoS One ; 4(10): e7583, 2009 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-19851510

RESUMO

Parathyroid hormone (PTH) is a potent anabolic agent for the treatment of osteoporosis. However, its mechanism of action in osteoblast and bone is not well understood. In this study, we show that the anabolic actions of PTH in bone are severely impaired in both growing and adult ovariectomized mice lacking bone-related activating transcription factor 4 (ATF4). Our study demonstrates that ATF4 deficiency suppresses PTH-stimulated osteoblast proliferation and survival and abolishes PTH-induced osteoblast differentiation, which, together, compromise the anabolic response. We further demonstrate that the PTH-dependent increase in osteoblast differentiation is correlated with ATF4-dependent up-regulation of Osterix. This regulation involves interactions of ATF4 with a specific enhancer sequence in the Osterix promoter. Furthermore, actions of PTH on Osterix require this same element and are associated with increased binding of ATF4 to chromatin. Taken together these experiments establish a fundamental role for ATF4 in the anabolic actions of PTH on the skeleton.


Assuntos
Fator 4 Ativador da Transcrição/fisiologia , Osso e Ossos/metabolismo , Regulação da Expressão Gênica , Hormônio Paratireóideo/metabolismo , Fatores de Transcrição/metabolismo , Fator 4 Ativador da Transcrição/metabolismo , Animais , Diferenciação Celular , AMP Cíclico/metabolismo , Feminino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Osteoblastos/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição Sp7 , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA