Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 27(51): 6550-60, 2008 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-18679424

RESUMO

Alveolar rhabdomyosarcoma is an aggressive skeletal muscle cancer of childhood. Our initial studies of rhabdomyosarcoma gene expression for patients enrolled in a national clinical trial suggested that platelet-derived growth factor receptor A (PDGFR-A) may be a mediator of disease progression and metastasis. Using our conditional mouse tumor models that authentically recapitulate the primary mutations and metastatic progression of alveolar rhabdomyosarcomas in humans, we found by immunoblotting and immunokinase assays that PDGFR-A and its downstream effectors, mitogen-activated protein kinase and Akt, were highly activated in both primary and metastatic tumors. Inhibition of PDGFR-A by RNA interference, small molecule inhibitor or neutralizing antibody had a dramatic effect on tumor cell growth both in vitro and in vivo, although resistance evolved in one-third of tumors. These results establish proof-of-principal for PDGFR-A as a therapeutic target in alveolar rhabdomyosarcoma.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Musculares/tratamento farmacológico , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/fisiologia , Rabdomiossarcoma Alveolar/tratamento farmacológico , Animais , Benzamidas , Linhagem Celular Tumoral , Células Cultivadas , Genes p16 , Humanos , Mesilato de Imatinib , Camundongos , Camundongos Knockout , Neoplasias Musculares/etiologia , Piperazinas/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Pirimidinas/uso terapêutico , Rabdomiossarcoma Alveolar/etiologia , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Bone ; 42(6): 1122-30, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18378205

RESUMO

Diabetes is a chronic disease associated with hyperglycemia and altered bone metabolism that may lead to complications including osteopenia, increased risk of fracture and osteoporosis. Hyperglycemia has been implicated in the pathogenesis of diabetic bone disease; however, the biologic effect of glucose on osteoclastogenesis is unclear. In the present study, we examined the effect of high d(+)glucose (d-Glc) and l(-)glucose (l-Glc; osmotic control) on RANKL-induced osteoclastogenesis using RAW264.7 cells and Bone Marrow Macrophages (BMM) as models. Cells were exposed to sustained high glucose levels to mimic diabetic conditions. Osteoclast formation was analyzed using tartrate resistant acid phosphatase (TRACP) assay, expression of calcitonin receptor (CTR) and cathepsin K mRNAs, and cultures were examined for reactive oxygen species (ROS) using dichlorodihydrofluorescein diacetate (DCF-DA) fluorescence, caspase-3 and Nuclear Factor kappaB (NF-kappaB) activity. Cellular function was assessed using a migration assay. Results show, for the first time, that high d-Glc inhibits osteoclast formation, ROS production, caspase-3 activity and migration in response to RANKL through a metabolic pathway. Our findings also suggest that high d-Glc may alter RANKL-induced osteoclast formation by inhibiting redox-sensitive NF-kappaB activity through an anti-oxidative mechanism. This study increases our understanding of the role of glucose in diabetes-associated bone disease. Our data suggest that high glucose levels may alter bone turnover by decreasing osteoclast differentiation and function in diabetes and provide new insight into the biologic effects of glucose on osteoclastogenesis.


Assuntos
Diferenciação Celular/fisiologia , Glucose/metabolismo , Osteoclastos/fisiologia , Ligante RANK/metabolismo , Fosfatase Ácida/metabolismo , Animais , Caspase 3/metabolismo , Catepsina K , Catepsinas/genética , Catepsinas/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Células Cultivadas , Humanos , Isoenzimas/metabolismo , Metaloproteinase 14 da Matriz/genética , Metaloproteinase 14 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fosfatase Ácida Resistente a Tartarato
3.
FASEB J ; 15(11): 1909-20, 2001 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-11532971

RESUMO

Angiotensin II (Ang II) exerts contractile and trophic effects in glomerular mesangial cells (MCs). One potential downstream target of Ang II is the protein kinase Akt/protein kinase B (PKB). We investigated the effect of Ang II on Akt/PKB activity in MCs. Ang II causes rapid activation of Akt/PKB (5-10 min) but delayed activation of phosphoinositide 3-kinase (PI3-K) (30 min). Activation of Akt/PKB by Ang II was not abrogated by the PI3-K inhibitors or by the introduction of a dominant negative PI3-K, indicating that in MCs, PI3-K is not an upstream mediator of Akt/PKB activation by Ang II. Incubation of MCs with phospholipase A2 inhibitors also blocked Akt/PKB activation by Ang II. AA mimicked the effect of Ang II. Inhibitors of cyclooxygenase-, lipoxyogenase-, and cytochrome P450-dependent metabolism did not influence AA-induced Akt/PKB activation. However, the antioxidants N-acetylcysteine and diphenylene iodonium inhibited both AA- and Ang II-induced Akt/PKB activation. Dominant negative mutant of Akt/PKB or antioxidants, but not the dominant negative form of PI3-K, inhibited Ang II-induced protein synthesis and cell hypertrophy. These data provide the first evidence that Ang II induces protein synthesis and hypertrophy in MCs through AA/redox-dependent pathway and Akt/PKB activation independent of PI3-K.


Assuntos
Angiotensina II/metabolismo , Ácido Araquidônico/metabolismo , Mesângio Glomerular/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas/metabolismo , Transdução de Sinais , Angiotensina II/farmacologia , Animais , Células Cultivadas , Ativação Enzimática , Mesângio Glomerular/citologia , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Oxirredução , Inibidores de Fosfoinositídeo-3 Quinase , Fosfolipases A/metabolismo , Fosfolipases A2 , Proteínas Proto-Oncogênicas c-akt , Ratos , Espécies Reativas de Oxigênio/metabolismo
4.
J Biol Chem ; 276(45): 42492-500, 2001 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-11509557

RESUMO

Insulin-like growth factor-I (IGF-I) may play an important role in the development of renal hypertrophy. In this study we determined the effect of IGF-I on cultured mesangial cells (MCs) and examined activation of key signaling pathways. IGF-I induced hypertrophy as determined by an increase in cell size and an increase in protein to DNA ratio and increased accumulation of extracellular matrix (ECM) proteins. IGF-I also activated both Erk1/Erk2 MAPK and phosphatidylinositol 3-kinase (PI3K) in MCs. Inhibition of either MAPK or PI3K, however, had no effect on IGF-I-induced hypertrophy or ECM production. Next, we examined the effect of IGF-I on activation of the calcium-dependent phosphatase calcineurin. IGF-I treatment stimulated calcineurin activity and increased the protein levels of calcineurin and the calcineurin binding protein, calmodulin. Cyclosporin A, an inhibitor of calcineurin, blocked both IGF-I-mediated hypertrophy and up-regulation of ECM. In addition, calcineurin resulted in sustained Akt activation, indicating possible cross-talk with other signaling pathways. Finally, IGF-I treatment resulted in the calcineurindependent nuclear localization of NFATc1. Therefore, IGF-I induces hypertrophy and increases ECM accumulation in MCs. IGF-I-mediated hypertrophy is associated with activation of Erk1/Erk2 MAPK and PI3K but does not require either of these pathways. Instead, IGF-I mediates hypertrophy via a calcineurin-dependent pathway.


Assuntos
Calcineurina/fisiologia , Fator de Crescimento Insulin-Like I/toxicidade , Rim/efeitos dos fármacos , Proteínas Nucleares , Proteínas Serina-Treonina Quinases , Animais , Células Cultivadas , Proteínas de Ligação a DNA/análise , Proteínas de Ligação a DNA/fisiologia , Proteínas da Matriz Extracelular/biossíntese , Hipertrofia , Rim/patologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Transcrição NFATC , Fosfatidilinositol 3-Quinases/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Ratos , Fatores de Transcrição/análise , Fatores de Transcrição/fisiologia , Fator de Crescimento Transformador beta/toxicidade
5.
Kidney Int ; 60(2): 495-504, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11473632

RESUMO

BACKGROUND: Altered regulation of signaling pathways may contribute to the pathogenesis of renal disease. We examined renal cortical signaling pathways in type 2 diabetes. METHODS: The status of renal cortical signaling pathways was examined in control and db/db mice with type 2 diabetes in the early phase of diabetic nephropathy associated with renal matrix expansion and albuminuria. RESULTS: Tyrosine phosphorylation of renal cortical proteins was increased in diabetic mice. Renal cortical activities of phosphatidylinositol 3-kinase (PI 3-kinase) in antiphosphotyrosine immunoprecipitates, Akt (PKB), and ERK1/2-type mitogen-activated protein (MAP) kinase activities were significantly augmented sixfold (P < 0.01), twofold (P < 0.0003), and sevenfold (P < 0.001), respectively, in diabetic mice compared with controls. A part of the increased renal cortical PI 3-kinase activity was due to insulin receptor activation, as PI 3-kinase activity associated with beta chain of the insulin receptor was increased nearly fourfold (P < 0.0235). Additionally, the kinase activity of the immunoprecipitated insulin receptor beta chain was augmented in the diabetic renal cortex, and tyrosine phosphorylation of the insulin receptor was increased. In the liver, activities of PI 3-kinase in the antiphosphotyrosine immunoprecipitates and Akt also were increased threefold (P < 0.05) and twofold (P < 0.0002), respectively. However, there was no change in the hepatic insulin receptor-associated PI 3-kinase activity. Additionally, the hepatic ERK1/2-type MAP kinase activity was inhibited by nearly 50% (P < 0.01). CONCLUSIONS: These studies demonstrate that a variety of receptor signaling pathways are activated in the renal cortex of mice with type 2 diabetes, and suggest a role for augmented insulin receptor activity in nephropathy of type 2 diabetes.


Assuntos
Diabetes Mellitus Tipo 2/metabolismo , Nefropatias Diabéticas/metabolismo , Córtex Renal/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Proteínas Serina-Treonina Quinases , Animais , Hiperinsulinismo/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Mutantes , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptor de Insulina/metabolismo , Tirosina/metabolismo
6.
Kidney Int ; 59(3): 866-75, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11231341

RESUMO

BACKGROUND: Augmented protein translation by insulin involves activation of eukaryotic initiation factor 4E (eIF4E) that follows release of eIF4E from a heterodimeric complex by phosphorylation of its inhibitory binding protein, 4E-BP1. We examined insulin regulation of 4E-BP1 phosphorylation in murine proximal tubular epithelial cells. METHODS AND RESULTS: Insulin (1 nmol/L) increased de novo protein synthesis by 58 +/- 11% (P < 0.001). Insulin also augmented 4E-BP1 phosphorylation and phosphatidylinositol 3-kinase (PI 3-kinase) activity in antiphosphotyrosine immunoprecipitates. This could be prevented by PI 3-kinase inhibitors, Wortmannin, and LY294002. Insulin also activated Akt that lies downstream of PI 3-kinase. Rapamycin abrogated 4E-BP1 phosphorylation in response to insulin, suggesting involvement of mammalian target of rapamycin (mTOR), a kinase downstream of Akt. Insulin-stimulated phosphorylation of 4E-BP1 was also inhibited by PD098059, implying involvement of Erk-1/-2 mitogen-activated protein (MAP) kinase. An increase in Erk-1/-2 type MAP kinase activity by insulin was directly confirmed in an immunokinase assay and was found to be PI 3-kinase dependent. CONCLUSIONS: In proximal tubular epithelial cells, insulin augments 4E-BP1 phosphorylation, which is PI 3-kinase and mTOR dependent. The requirement for Erk-1/-2 MAP kinase activation for 4E-BP1 phosphorylation by insulin suggests a cross-talk between PI 3-kinase and Erk-1/-2-type MAP kinase pathways.


Assuntos
Insulina/farmacologia , Túbulos Renais Proximais/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas de Transporte/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Células Epiteliais/metabolismo , Fator de Iniciação 4E em Eucariotos , Fatores de Iniciação em Eucariotos , Túbulos Renais Proximais/citologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fatores de Iniciação de Peptídeos/metabolismo , Fosfatidilinositol 3-Quinases/fisiologia , Fosfoproteínas/fisiologia , Fosforilação , Biossíntese de Proteínas , Proteínas Quinases/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteínas Proto-Oncogênicas c-akt , Serina-Treonina Quinases TOR , Tirosina/metabolismo
7.
Hepatology ; 33(3): 713-21, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11230753

RESUMO

Activated hepatic stellate cells (HSC) regulate the liver "wound-healing" response through expression of chemokines, including monocyte chemoattractant protein-1 (MCP-1), which participate in the formation of the inflammatory infiltrate during liver injury. Cyclooxygenase (COX) catalyzes the conversion of arachidonic acid into prostaglandins, which may contribute to the inflammatory response. In this study, we investigated the effects of COX inhibitors on the expression of MCP-1 in cultured HSC. Pretreatment of HSC with nonspecific COX inhibitors such as indomethacin or ibuprofen markedly reduced the expression of MCP-1 caused by exposure to tumor necrosis factor alpha (TNF-alpha) or interleukin-1alpha (IL-1alpha). NS-398, a specific COX-2 inhibitor, also resulted in a dose-dependent inhibition of MCP-1 gene and protein expression. These effects were dependent on reduced MCP-1 transcription, as established using a reporter plasmid. In contrast, the up-regulation of MCP-1 expression caused by interferon gamma (IFN-gamma) was not sensitive to COX inhibitors. Quiescent HSC did not show detectable expression of COX-2, which became evident after activation in culture, and while TNF-alpha and IL-1alpha markedly increased the expression of COX-2, IFN-gamma did not have any effects. Pretreatment of HSC with the stable cyclic adenosine monophosphate (cAMP) analog, 8-bromo cAMP, reverted the effects of the COX-2 inhibitor, but not of a nuclear factor-kappaB (NF-kappaB) inhibitor, demonstrating that prostaglandins modulate MCP-1 expression via production of cAMP. On the other hand, the action of NF-kappaB inhibitors was negligible in IFN-gamma-stimulated cells. These findings indicate that cross-talk between cytokines and a prostaglandin-cAMP pathway differentially regulates the proinflammatory potential of HSC, contributing to the modulation of liver tissue inflammation.


Assuntos
Quimiocina CCL2/agonistas , Quimiocina CCL2/metabolismo , Fígado/metabolismo , Prostaglandina-Endoperóxido Sintases/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Células Cultivadas , Quimiocina CCL2/genética , AMP Cíclico/fisiologia , Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase 2 , Inibidores de Ciclo-Oxigenase/farmacologia , Expressão Gênica/efeitos dos fármacos , Humanos , Ibuprofeno/farmacologia , Indometacina/farmacologia , Interleucina-1/farmacologia , Isoenzimas/antagonistas & inibidores , Isoenzimas/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Proteínas de Membrana , NF-kappa B/fisiologia , Nitrobenzenos/farmacologia , Sulfonamidas/farmacologia , Transcrição Gênica/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia
8.
J Biol Chem ; 275(13): 9527-33, 2000 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-10734101

RESUMO

Platelet-derived growth factor (PDGF) B-chain and PDGF receptor beta (PDGFR beta) are essential for glomerulogenesis. Mice deficient in PDGF B-chain or PDGFR beta exhibit an abnormal glomerular phenotype characterized by total lack of mesangial cells. In this study, we localized PDGFR beta in the developing rat kidney and explored the biological effects of PDGF in metanephric mesenchymal cells in an attempt to determine the mechanism by which PDGF regulates mesangial cell development. Immunohistochemical and in situ hybridization studies of rat embryonic kidneys reveal that PDGFR beta localizes to undifferentiated metanephric mesenchyme and is later expressed in the cleft of the comma-shaped and S-shaped bodies and in more mature glomeruli in a mesangial distribution. We also isolated and characterized cells from rat metanephric mesenchyme. Metanephric mesenchymal cells express vimentin and alpha-smooth muscle actin but not cytokeratin. These cells also express functional PDGFR beta, as demonstrated by autophosphorylation of the receptor as well as activation of phosphatidylinositol 3 kinase in response to PDGF B-chain homodimer. PDGF B-chain also induces migration and proliferation of metanephric mesenchymal cells. Taken together with the fact that PDGF B-chain is expressed in the glomerular epithelium and mesangial area, as demonstrated in the human embryonic kidney, we suggest that PDGF B-chain acts in a paracrine fashion to stimulate the migration and proliferation of mesangial cell precursors from undifferentiated metanephric mesenchyme to the mesangial area. PDGF B-chain also likely stimulates proliferation of mesangial cell precursors in an autocrine fashion once these cells migrate to the glomerular tuft.


Assuntos
Movimento Celular/fisiologia , Replicação do DNA/fisiologia , Rim/citologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/fisiologia , Animais , Células Cultivadas , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Imuno-Histoquímica , Rim/embriologia , Rim/metabolismo , Camundongos , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo
9.
Biochem Biophys Res Commun ; 258(2): 490-6, 1999 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-10329414

RESUMO

Bone morphogenetic protein-2 (BMP-2) is a member of the TGFbeta superfamily of growth and differentiation factors. We investigated the effect of BMP-2 on epidermal growth factor (EGF)-induced mitogenic signaling in kidney glomerular mesangial cells. BMP-2 dose-dependently inhibits EGF-induced DNA synthesis. Maximum effect was obtained at a concentration of 100 ng/ml. BMP-2 had no inhibitory effect on the EGF receptor (EGFR)-associated tyrosine kinase activity indicating that inhibition of DNA synthesis is due to regulation of post-receptor signaling event(s). EGF stimulates MAPK activity in mesangial cells in a time-dependent manner. Inhibition of MAPK by the MEK inhibitor PD098059 blocks EGF-induced DNA synthesis indicating the requirement of this enzyme activity in EGF-mediated mitogenic signaling. Furthermore, we show that exposure of mesangial cells to BMP-2 blocks EGF-induced MAPK activity which leads to phosphorylattion of Elk-1 transcription factor. Using a GAL-4 DNA binding-domain-Elk-1 transactivation domain fusion protein-based reporter assay, we demonstrate that BMP-2 inhibits EGF-induced Elk-1-mediated transcription. These data provide the first evidence that BMP-2 signaling in mesangial cells initiates a negative regulatory cross-talk with MAPK-based transcription to inhibit EGF-induced DNA synthesis.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas Quinases Dependentes de Cálcio-Calmodulina/antagonistas & inibidores , Replicação do DNA/efeitos dos fármacos , Proteínas de Ligação a DNA , Fator de Crescimento Epidérmico/farmacologia , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição , Ativação Transcricional/efeitos dos fármacos , Fator de Crescimento Transformador beta , Animais , Proteína Morfogenética Óssea 2 , Receptores ErbB/metabolismo , Mesângio Glomerular/citologia , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/metabolismo , Ratos , Proteínas Elk-1 do Domínio ets
10.
J Biol Chem ; 274(16): 10897-902, 1999 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-10196167

RESUMO

Bone morphogenetic proteins (BMPs) play an important role in nephrogenesis. The biologic effect and mechanism of action of these proteins in the adult kidney has not yet been studied. We investigated the effect of BMP2, a member of these growth and differentiation factors, on mitogenic signal transduction pathways induced by platelet-derived growth factor (PDGF) in glomerular mesangial cells. PDGF is a growth and survival factor for these cells in vitro and in vivo. Incubation of mesangial cells with increasing concentrations of BMP2 inhibited PDGF-induced DNA synthesis in a dose-dependent manner with maximum inhibition at 250 ng/ml. Immune complex tyrosine kinase assay of PDGF receptor beta immunoprecipitates from lysates of mesangial cells treated with PDGF showed no inhibitory effect of BMP2 on PDGF receptor tyrosine phosphorylation. This indicates that the inhibition of DNA synthesis is likely due to postreceptor events. However, BMP2 significantly inhibited PDGF-stimulated mitogen-activated protein kinase (MAPK) activity that phosphorylates the Elk-1 transcription factor, a component of the ternary complex factor. Using a fusion protein-based reporter assay, we also show that BMP2 blocks PDGF-induced Elk-1-mediated transcription. Furthermore, we demonstrate that BMP2 inhibits PDGF-induced transcription of c-fos gene, a natural target of Elk-1 that normally forms a ternary complex that activates the serum response element of the c-fos gene. These data provide the first evidence that in mesangial cells, BMP2 signaling cross-talks with MAPK-based transcriptional events to inhibit PDGF-induced DNA synthesis. One target for this inhibition is the early response gene c-fos.


Assuntos
Proteínas Morfogenéticas Ósseas/fisiologia , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Replicação do DNA/fisiologia , Genes fos , Fator de Crescimento Derivado de Plaquetas/fisiologia , Transcrição Gênica/fisiologia , Animais , Proteína Morfogenética Óssea 2 , Células Cultivadas , Ativação Enzimática , Fibronectinas/farmacologia , Mesângio Glomerular/citologia , Mesângio Glomerular/enzimologia , Mesângio Glomerular/metabolismo , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo , Fator de Crescimento Transformador beta/farmacologia
11.
Am J Physiol ; 276(3): F382-9, 1999 03.
Artigo em Inglês | MEDLINE | ID: mdl-10070161

RESUMO

BMP-7, a member of the bone morphogenic protein subfamily (BMPs) of the transforming growth factor-beta superfamily of secreted growth factors, is abundantly expressed in the fetal kidney. The precise role of this protein in renal physiology or pathology is unknown. A cDNA that encodes rat BMP-7 was cloned and used as a probe to localize BMP-7 mRNA expression by in situ hybridization in the adult rat kidney. The highest expression of BMP-7 mRNA could be seen in tubules of the outer medulla. In glomeruli, a few cells, mainly located at the periphery of the glomerular tuft, showed specific and strong signals. Also, high BMP-7 mRNA expression could be localized to the adventitia of renal arteries, as well as to the epithelial cell layer of the renal pelvis and the ureter. Preliminary evidence suggests that BMP-7 enhances recovery when infused into rats with ischemia-induced acute renal failure. We examined BMP-7 mRNA expression in kidneys with acute renal failure induced by unilateral renal artery clamping. BMP-7 mRNA abundance as analyzed by solution hybridization was reduced in ischemic kidneys after 6 and 16 h of reperfusion compared with the contralateral kidney. In situ hybridization in ischemic kidneys showed a marked decrease of BMP-7 mRNA in the outer medulla and in glomeruli. Utilizing rat metanephric mesenchymal cells in culture, we also demonstrate that BMP-7 induces epithelial cell differentiation. Taken together, these data suggest that BMP-7 is important in both stimulating and maintaining a healthy differentiated epithelial cell phenotype.


Assuntos
Proteínas Morfogenéticas Ósseas/genética , Isquemia/metabolismo , Rim/metabolismo , RNA Mensageiro/metabolismo , Circulação Renal , Fator de Crescimento Transformador beta , Animais , Proteína Morfogenética Óssea 7 , Diferenciação Celular/fisiologia , Embrião de Mamíferos/citologia , Rim/embriologia , Glomérulos Renais/metabolismo , Medula Renal/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Valores de Referência , Circulação Renal/fisiologia , Distribuição Tecidual
12.
Kidney Int ; 54(4): 1117-27, 1998 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-9767527

RESUMO

BACKGROUND: Shiga toxin 1 (Stx1) is a causative agent in hemolytic uremic syndrome (HUS). Its receptor, the glycosphingolipid globotriaosylceramide (Gb3), is expressed on cultured human endothelial and mesangial cells. Mesangial cell injury in HUS ranges from mild cellular edema to severe mesangiolysis and eventual glomerulosclerosis. We hypothesized that, in addition to endothelial cells, mesangial cells are targets of Stx1. METHODS: Human mesangial cells were exposed to Stx1. Protein synthesis was measured using [35S]-methionine/cysteine. Cell viability was measured as the lysosomal uptake of Neutral Red. Monocyte chemotactic peptide (MCP-1) mRNA and protein were analyzed by Northern blotting and ELISA. RESULTS: Stx1 (0.25 to 2500 ng/ml) resulted in a dose-dependent inhibition of protein synthesis. This effect of Stx1 was potentiated by preincubation of the cells with interleukin-1alpha (IL-1alpha; 2 ng/ml) or tumor necrosis-alpha (TNF-alpha; 500 U/ml). Stx1 had little effect on mesangial cell viability during the first 24 hours of exposure to Stx1. However, prolonged incubation with Stx1 for 48 and 72 hours resulted in a 68% and 80% decrease in cell-viability, respectively. Stx1 elicited a dose and time dependent increase in the levels of MCP-1 mRNA, an effect that was potentiated by preincubation with IL-1alpha. CONCLUSION: These data indicate that mesangial cells are susceptible to the effects of Stx1 in vitro. Stx1 exerts a spectrum of biologic effects on mesangial cells ranging from activation of chemokine genes to a lethal toxic injury. Immunoinflammatory cytokines potentiate the effects of Stx1. Thus, glomerular pathology in HUS may also result from a direct effect of Stx1 on mesangial cells.


Assuntos
Toxinas Bacterianas/toxicidade , Mesângio Glomerular/efeitos dos fármacos , Mesângio Glomerular/metabolismo , Tamanho Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Quimiocina CCL2/biossíntese , Quimiocina CCL2/genética , DNA/biossíntese , Endotélio Vascular/efeitos dos fármacos , Mesângio Glomerular/citologia , Síndrome Hemolítico-Urêmica/etiologia , Humanos , Interleucina-1/farmacologia , Modelos Biológicos , Biossíntese de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Toxinas Shiga , Fator de Necrose Tumoral alfa/farmacologia
13.
J Immunol ; 161(7): 3719-28, 1998 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-9759897

RESUMO

Monocyte chemoattractant protein-1 (MCP-1) is induced in chronic osseous inflammation, and is temporally and spatially correlated with monocyte recruitment. We investigated the mechanism of MCP-1 regulation in a human osteoblastic cell line in response to IFN-gamma, a potent mediator of the immune inflammatory response. Nuclear run-on and stability studies demonstrated that IFN-gamma stimulated MCP-1 transcription and did not enhance mRNA stabilization. Using MCP-1 promoter/reporter gene constructs, we determined that IFN-gamma-enhanced MCP-1 transcription is regulated by a 29-bp element located at -227 relative to the ATG start codon. This element contains a 13-bp CT-rich sequence (GCTTCCCTTTCCT) adjacent to a IFN-gamma activation site (GAS). Since deletion of the CT sequence enhanced both the magnitude and duration of IFN-gamma-stimulated, GAS-mediated transcription, we have termed it the IFN response-inhibitory sequence (IRIS). The combined IRIS/GAS sequence is highly conserved in mouse, rat, and bovine MCP-1 genes. In gel-shift assays, nuclear extracts from IFN-gamma-stimulated osteoblastic cells formed two specific inducible bands with labeled IRIS/GAS DNA. Both bands were supershifted by anti-STAT1 Abs, but not by Abs to STAT2, p48(ISGF-3y), IFN-regulatory factor-1, or IFN-regulatory factor-2. Formation of one of the bands required the presence of the IRIS moiety. IRIS/GAS DNA also formed a number of specific complexes with constitutively expressed factors, none of which were affected by the above Abs. These studies establish a mechanism for IFN-gamma-stimulated MCP-1 expression and identify a complex element that regulates MCP-1 gene transcription.


Assuntos
Quimiocina CCL2/genética , Interferon gama/fisiologia , Transcrição Gênica/imunologia , Sequência de Bases , Sequência Conservada/imunologia , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Dados de Sequência Molecular , Proteínas Nucleares/metabolismo , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Regiões Promotoras Genéticas/imunologia , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Transcrição Gênica/efeitos dos fármacos , Células Tumorais Cultivadas
14.
Kidney Int ; 51(3): 770-6, 1997 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9067909

RESUMO

Chemokines are a family of chemotactic cytokines whose participation in inflammation in vivo remains to be established. To study the role of monocyte-chemoattractant-protein-1 (MCP-1) on the glomerular accumulation of leukocytes, rats received a neutralizing anti-MCP-1 antiserum following the induction of an glomerulonephritis by an anti-thymocyte antibody (ATS). The infiltration of monocytes/macrophages (M/M) and granulocytes was analyzed by immunohistology. When studied by Northern blotting, glomerular mRNA levels of MCP-1, and interleukin 1 beta (IL-1 beta) increased at three hours and 24 hours following the induction of the injury. The glomerular mRNA expression of intercellular adhesion molecule-1 (ICAM-1) only increased marginally, whereas the expression of the chemokine RANTES was not enhanced. In animals that received anti-MCP-1 antibody glomerular MCP-1 mRNA expression increased. However, the chemoattractant activity for monocytes released into supernatants of isolated glomeruli was reduced. The anti-MCP-1 antibody did not affect glomerular IL-1 beta, ICAM-1 or RANTES mRNA levels. The induction of glomerulonephritis was associated with an increased glomerular recruitment of polymorphonuclear granulocytes (PMNs) at three hours and M/M at 24 hours, when compared with controls. The anti-MCP-1 antiserum significantly reduced the glomerular M/M infiltration at 24 hours by 40%, but was without effect on glomerular PMN recruitment or growth of the resident glomerular cells. These studies demonstrate that MCP-1 is an important mediator for monocyte recruitment in this model of glomerulonephritis. The reduction of M/M infiltration might affect this glomerular injury.


Assuntos
Quimiocina CCL2/fisiologia , Glomerulonefrite/patologia , Glomerulonefrite/fisiopatologia , Macrófagos/patologia , Macrófagos/fisiologia , Monócitos/patologia , Monócitos/fisiologia , Animais , Anticorpos , Soro Antilinfocitário/farmacologia , Quimiocina CCL2/antagonistas & inibidores , Quimiocina CCL2/imunologia , Quimiotaxia de Leucócito/fisiologia , Glomerulonefrite/etiologia , Técnicas In Vitro , Glomérulos Renais/patologia , Glomérulos Renais/fisiopatologia , Masculino , Testes de Neutralização , Ratos , Ratos Wistar
15.
Am J Physiol ; 270(2 Pt 2): F295-300, 1996 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8779890

RESUMO

Thrombin stimulates mitogenesis and tyrosine phosphorylation of several proteins in glomerular mesangial cells [T. Force, J. M. Kyriakis, J. Avruch, and J. V. Bonventre, J. Biol. Chem. 266: 6650-6656, 1991; and G. Grandaliano, G. Ghosh Choudhury, P. Biswas, and H. E. Abboud, Am. J. Physiol. 267 (Renal Fluid Electrolyte Physiol. 36: F528-F536, 1994]. However, none of the tyrosine phosphorylated proteins have been identified. Here we show that thrombin stimulates phosphorylation of four major proteins of molecular masses 170, 125, 97, and 47 kDa in antiphosphotyrosine immunoprecipitates in vitro. Immunoblot analysis of antiphosphotyrosine immunoprecipitates from lysates of thrombin-treated cells with anti-Nck antibody revealed the presence of this src homology domain-containing adaptor molecule in the tyrosine-phosphorylated protein fraction. In addition, in thrombin-treated cells, direct immunoblotting of Nck immunoprecipitates with antiphosphotyrosine antibody showed no tyrosine phosphorylation of Nck. In these immunoprecipitates, we detected a 125-kDa tyrosine-phosphorylated protein. We identified this protein as pp125FAK (FAK, focal adhesion kinase) after analyzing Nck immunoprecipitates by anti-FAK immunoblotting. Treatment of mesangial cells with thrombin resulted in stimulation of the tyrosine kinase activity of pp125FAK in vitro. We conclude that activation of the cytoplasmic protein tyrosine kinase pp125FAK by thrombin stimulates its association with the src homology domain-containing adaptor protein Nck. This indicates that Nck is a direct target for FAK in the thrombin-induced signal transduction pathway.


Assuntos
Moléculas de Adesão Celular/metabolismo , Genes src , Proteínas Oncogênicas/genética , Proteínas Oncogênicas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Homologia de Sequência , Trombina/farmacologia , Proteínas Adaptadoras de Transdução de Sinal , Células Cultivadas , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Mesângio Glomerular/citologia , Mesângio Glomerular/metabolismo , Humanos , Immunoblotting , Fosfotirosina/imunologia , Testes de Precipitina
16.
Am J Physiol ; 267(4 Pt 2): F528-36, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7524356

RESUMO

Thrombin elicits multiple biological effects on a variety of cells. We have previously shown that thrombin is a potent mitogen for human glomerular mesangial cells. This mitogenic effect of thrombin is associated with activation of phospholipase C (PLC) and induction of platelet-derived growth factor (PDGF) gene expression. The thrombin receptor, which belongs to the guanine nucleotide binding protein (G protein)-coupled receptor family, has recently been shown to induce rapid tyrosine phosphorylation of cellular proteins. In the present study, we investigated the role of protein-tyrosine phosphorylation in mediating the cellular responses elicited by thrombin in human glomerular mesangial cells. Amino acid labeling followed by immunoprecipitation with phosphotyrosine antibodies demonstrate that thrombin stimulates tyrosine phosphorylation of a set of cellular proteins. Treatment of mesangial cells with thrombin followed by immunoblotting with phosphotyrosine antibodies showed three major bands of tyrosine-phosphorylated proteins approximately 130, 70, and 44-42 kDa. Phosphorylation of these proteins was inhibited by two tyrosine kinase inhibitors, herbimycin A and genistein. Both compounds inhibited DNA synthesis and PDGF B-chain gene expression but had no effect on inositol phosphates production or increases in cytosolic calcium in response to thrombin. These data demonstrate that protein-tyrosine phosphorylation is not required for thrombin-induced PLC activation with inositol phosphate formation and subsequent intracellular calcium release, but it is an absolute requirement for thrombin-induced DNA synthesis and PDGF B-chain gene expression.


Assuntos
Mesângio Glomerular/efeitos dos fármacos , Mitógenos/farmacologia , Fosfoproteínas/metabolismo , Fator de Crescimento Derivado de Plaquetas/biossíntese , Proteínas Tirosina Quinases/metabolismo , Trombina/farmacologia , Tirosina/análogos & derivados , Benzoquinonas , Cálcio/metabolismo , Células Cultivadas , Cisteína/metabolismo , DNA/biossíntese , Expressão Gênica/efeitos dos fármacos , Genisteína , Mesângio Glomerular/citologia , Mesângio Glomerular/metabolismo , Humanos , Immunoblotting , Fosfatos de Inositol/metabolismo , Isoflavonas/farmacologia , Lactamas Macrocíclicas , Peso Molecular , Fosfoproteínas/biossíntese , Fosfoproteínas/isolamento & purificação , Fosforilação , Fosfotirosina , Fator de Crescimento Derivado de Plaquetas/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Quinonas/farmacologia , RNA Mensageiro/análise , RNA Mensageiro/biossíntese , Rifabutina/análogos & derivados , Radioisótopos de Enxofre , Tirosina/análise , Tirosina/metabolismo
17.
Gastroenterology ; 107(4): 1110-7, 1994 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-7926460

RESUMO

BACKGROUND/AIMS: Liver fat-storing cells (FSCs) actively proliferate and secrete extracellular matrix during liver injury. Platelet-derived growth factor (PDGF) is a potent mitogen for cultured FSCs. In the present study, we investigated the regulation of PDGF gene expression and production in cultured human liver FSCs. METHODS: PDGF A-chain and B-chain expression was analyzed by Northern blotting and ribonuclease protection assay, respectively. Secretion of PDGF was evaluated by immunoprecipitation and immunoblotting of conditioned medium and metabolic labeling of FSC followed by immunoprecipitation. RESULTS: Three PDGF A-chain transcripts were detectable. Stimulation of FSC with phorbol myristate acetate (10(-7) mol/L) or PDGF BB (20 ng/mL) increased steady-state levels of PDGF A-chain and B-chain messenger RNA. PDGF AA had a small stimulatory effect on A-chain but not B-chain messenger RNA levels. FSCs secrete PDGF in the conditioned medium. The secreted protein is bioactive, because concentrated conditioned medium induced an increase in thymidine incorporation that was inhibited by anti-PDGF antibodies. CONCLUSIONS: This study shows that cultured FSCs express PDGF A- and B-chain genes and release bioactive PDGF in the culture medium. These data raise the possibility of an autocrine or short-loop paracrine effect of PDGF in FSCs as a mechanism contributing to the maintenance of the proliferative state during liver injury.


Assuntos
Expressão Gênica , Metabolismo dos Lipídeos , Fígado/metabolismo , Fígado/fisiologia , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , Células Cultivadas , Humanos , Fígado/citologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-sis
18.
J Lab Clin Med ; 123(2): 282-9, 1994 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-8301205

RESUMO

Cell-mediated immunity and monocyte infiltration is a prominent histologic feature of several different types of glomerulonephritis. Monocyte influx to the glomerulus correlates with glomerular hypercellularity and proteinuria. Glomerular mesangial cells, in addition to being targets for inflammatory stimuli, are also effector cells that actively participate in glomerular pathology. Mesangial cells release monocyte chemotactic protein (MCP-1). In the present article, we characterized and studied the regulation of MCP-1 released by cultured human mesangial cells. Serum-deprived mesangial cells constitutively release chemotactic activity that is neutralized by specific anti-MCP-1 antibody. An antibody to baboon MCP-1 recognized 16, 15, and 11 kd proteins from concentrated conditioned medium that were consistent with the presence of different forms of MCP-1. Gamma interferon (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha), and interleukin-1 (IL-1) markedly stimulate the release of MCP-1 as measured by a specific and sensitive radioimmunoassay. The release of MCP-1 in response to these cytokines is at least partially dependent on de novo synthesis of the protein because all three cytokines markedly stimulate the expression of MCP-1 mRNA. These data demonstrate that human mesangial cells synthesize and release at least three different forms of MCP-1 and that IFN-gamma and other cytokines regulate the secretion of MCP-1. IFN-gamma and MCP-1 may play a major role in the recruitment and activation of monocytes to the inflamed glomerulus.


Assuntos
Fatores Quimiotáticos/biossíntese , Mesângio Glomerular/metabolismo , Interferon gama/farmacologia , Northern Blotting , Células Cultivadas , Quimiocina CCL2 , Fatores Quimiotáticos/genética , Fatores Quimiotáticos/metabolismo , Meios de Cultivo Condicionados , Citocinas , Humanos , Interleucina-1/farmacologia , RNA Mensageiro/metabolismo , Proteínas Recombinantes , Fator de Necrose Tumoral alfa/farmacologia
19.
Biochem J ; 297 ( Pt 2): 385-8, 1994 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-8297346

RESUMO

Mesangial cells express platelet-derived growth factor (PDGF) A- and B-chain mRNA and release PDGF. Several polypeptide growth factors, including PDGF itself, induce PDGF A- and B- chain mRNA abundance. To understand the molecular mechanisms associated with the changes in mRNA abundance, we measured the effects of PDGF BB homodimer on PDGF A- and B-chain gene transcription in cultured mesangial cells. The data demonstrate 2- and 4-fold increases in PDGF A-chain gene transcription in response to PDGF BB homodimer at 5 and 24 h time points respectively. PDGF B-chain gene transcription was also induced approximately 3-fold at 2, 5 and 24 h time points in response to treatment with PDGF BB homodimer. The effect of PDGF BB on the half-life of PDGF A- as well as PDGF B-chain mRNA was measured directly by the pulse-chase method. There was no effect on PDGF A-chain mRNA half-life whereas PDGF B-chain mRNA half-life was increased 1.5-fold. These studies indicate that, in human mesangial cells, the increase in the levels of PDGF A- and B-chain mRNA in response to PDGF- receptor(s) activation is mediated at the level of gene transcription. In addition, the regulation of PDGF B- but not PDGF A-chain gene involves increased mRNA stability. Mesangial cells are a useful model for studying molecular mechanisms of PDGF- gene regulation in non-transformed human cells.


Assuntos
Regulação da Expressão Gênica , Mesângio Glomerular/fisiologia , Fator de Crescimento Derivado de Plaquetas/genética , Células Cultivadas , Humanos , Técnicas In Vitro , Fator de Crescimento Derivado de Plaquetas/química , RNA Mensageiro/genética , Transcrição Gênica
20.
J Cell Physiol ; 158(1): 140-50, 1994 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-8263021

RESUMO

Platelet-derived growth factor (PDGF) occurs as homodimers or heterodimers of related polypeptide chains PDGF-BB, -AA, and -AB. There are two receptors that bind PDGF, termed alpha and beta. The beta receptor recognizes PDGF B chain and is dimerized in response to PDGF BB. The alpha receptor recognizes PDGF B as well as A chains and can be dimerized by the three dimeric forms of PDGF AA, AB, and BB. To characterize PDGF receptor signaling mechanisms and biologic activities in human mesangial cells (MC), we explored the effects of the three PDGF isoforms on DNA synthesis, phospholipase C activation, and PDGF protooncogene induction. PDGF-BB homodimer and AB heterodimer induced a marked increase in DNA synthesis, activation of phospholipase C, and autoinduction of PDGF A and B chain mRNAs, whereas PDGF-AA homodimer was without effect. The lack of response to PDGF AA could be accounted for by down-regulation of the PDGF-alpha receptor since preincubation of MC with suramin restored PDGF AA-induced DNA synthesis. Ligand binding studies demonstrate specific binding of labeled PDGF BB and AB and to a lower extent PDGF AA isoforms to mesangial cells. These results are consistent with predominant expression of PDGF beta receptor in MC, which is linked to phospholipase-C activation. The potent biologic effects of PDGF-AB heterodimer in cells that express very few alpha receptors and do not respond to PDGF AA are somewhat inconsistent with the currently accepted model of PDGF receptor interaction and suggest the presence of additional mechanisms for PDGF isoform binding and activation.


Assuntos
Mesângio Glomerular/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Transporte Biológico , Cálcio/metabolismo , Células Cultivadas , DNA/biossíntese , Regulação para Baixo , Mesângio Glomerular/citologia , Mesângio Glomerular/metabolismo , Humanos , Fosfatos de Inositol/biossíntese , Membranas Intracelulares/metabolismo , Isomerismo , Concentração Osmolar , Fator de Crescimento Derivado de Plaquetas/genética , Fator de Crescimento Derivado de Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Receptores do Fator de Crescimento Derivado de Plaquetas/classificação , Receptores do Fator de Crescimento Derivado de Plaquetas/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA