Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Invest ; 121(6): 2290-300, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21555855

RESUMO

GPCR inhibitors are highly prevalent in modern therapeutics. However, interference with complex GPCR regulatory mechanisms leads to both therapeutic efficacy and adverse effects. Recently, the sphingosine-1-phosphate (S1P) receptor inhibitor FTY720 (also known as Fingolimod), which induces lymphopenia and prevents neuroinflammation, was adopted as a disease-modifying therapeutic in multiple sclerosis. Although highly efficacious, dose-dependent increases in adverse events have tempered its utility. We show here that FTY720P induces phosphorylation of the C-terminal domain of S1P receptor 1 (S1P1) at multiple sites, resulting in GPCR internalization, polyubiquitinylation, and degradation. We also identified the ubiquitin E3 ligase WWP2 in the GPCR complex and demonstrated its requirement in FTY720-induced receptor degradation. GPCR degradation was not essential for the induction of lymphopenia, but was critical for pulmonary vascular leak in vivo. Prevention of receptor phosphorylation, internalization, and degradation inhibited vascular leak, which suggests that discrete mechanisms of S1P receptor regulation are responsible for the efficacy and adverse events associated with this class of therapeutics.


Assuntos
Síndrome de Vazamento Capilar/fisiopatologia , Propilenoglicóis/toxicidade , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Esfingosina/análogos & derivados , Animais , Relação Dose-Resposta a Droga , Endocitose , Cloridrato de Fingolimode , Técnicas de Introdução de Genes , Linfopenia/induzido quimicamente , Lisofosfolipídeos/fisiologia , Camundongos , Organofosfatos/farmacologia , Peptídeo Hidrolases/metabolismo , Fosforilação/efeitos dos fármacos , Propilenoglicóis/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Estrutura Terciária de Proteína , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/fisiopatologia , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes de Fusão/fisiologia , Esfingosina/farmacologia , Esfingosina/fisiologia , Esfingosina/toxicidade , Receptores de Esfingosina-1-Fosfato , Ubiquitina-Proteína Ligases/genética , Ubiquitina-Proteína Ligases/fisiologia , Ubiquitinação/efeitos dos fármacos
2.
J Clin Invest ; 119(12): 3530-43, 2009 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19884656

RESUMO

The RNA-binding protein HuR (also known as ELAV1) binds to the 3'-untranslated region of mRNAs and regulates transcript stability and translation. However, the in vivo functions of HuR are not well understood. Here, we report that murine HuR is essential for life; postnatal global deletion of Elavl1 induced atrophy of hematopoietic organs, extensive loss of intestinal villi, obstructive enterocolitis, and lethality within 10 days. Upon Elavl1 deletion, progenitor cells in the BM, thymus, and intestine underwent apoptosis, whereas quiescent stem cells and differentiated cells were unaffected. The survival defect of hematopoietic progenitor cells was cell intrinsic, as transplant of Elavl1-/- BM led to compromised hematopoietic reconstitution but did not cause lethality. Expression of p53 and its downstream effectors critical for cell death were induced in progenitor cells as HuR levels declined. In mouse embryonic fibroblasts, HuR bound to and stabilized the mRNA for Mdm2, a critical negative regulator of p53. Furthermore, cell survival was restored by expression of Mdm2 in Elavl1-/- cells, suggesting that HuR keeps p53 levels in check in progenitor cells and thereby promotes cell survival. This regulation of cell stress response by HuR in progenitor cells, which we believe to be novel, could potentially be exploited in cytotoxic anticancer therapies as well as stem cell transplant therapy.


Assuntos
Antígenos de Superfície/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas de Ligação a RNA/metabolismo , Animais , Antígenos de Superfície/genética , Apoptose , Diferenciação Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Feminino , Trato Gastrointestinal/citologia , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/metabolismo , Deleção de Genes , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/efeitos dos fármacos , Masculino , Camundongos , Camundongos Knockout , Modelos Biológicos , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Estabilidade de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Tolerância a Radiação , Tamoxifeno/toxicidade , Quimeras de Transplante , Proteína Supressora de Tumor p53/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 27(6): 1312-8, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17431187

RESUMO

OBJECTIVES: S1P acts via the S1PR family of G protein-coupled receptors to regulate a variety of physiological responses. Whereas S1P1R activates G(i)- and PI-3-kinase-dependent signals to inhibit vascular permeability, the related S1P2R inhibits the PI-3-kinase pathway by coupling to the Rho-dependent activation of the PTEN phosphatase. However, cellular consequences of S1P2R signaling in the vascular cells are not well understood. METHODS AND RESULTS: Selective signaling of the S1P2R was achieved by adenoviral-mediated expression in endothelial cells. Secondly, endogenously expressed S1P2R was blocked by the specific pharmacological antagonist JTE013. Activation of S1P2R in endothelial cells resulted in Rho-ROCK- and PTEN-dependent disruption of adherens junctions, stimulation of stress fibers, and increased paracellular permeability. JTE013 treatment of naive endothelial cells potentiated the S1P1R-dependent effects such as formation of cortical actin, blockade of stress fibers, stimulation of adherens junction assembly, and improved barrier integrity. This observation was extended to the in vivo model of vascular permeability in the rat lung: the S1P2R antagonist JTE013 significantly inhibited H2O2-induced permeability in the rat lung perfused model. CONCLUSIONS: S1P2R activation in endothelial cells increases vascular permeability. The balance of S1P1 and S1P2 receptors in the endothelium may determine the regulation of vascular permeability by S1P.


Assuntos
Permeabilidade Capilar , Células Endoteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lisofosfolipídeos/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais , Esfingosina/análogos & derivados , Junções Aderentes/metabolismo , Animais , Antígenos CD/metabolismo , Caderinas/metabolismo , Permeabilidade Capilar/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio , Fosforilação , Edema Pulmonar/induzido quimicamente , Edema Pulmonar/metabolismo , Edema Pulmonar/fisiopatologia , Pirazóis/farmacologia , Piridinas/farmacologia , Ratos , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética , Receptores de Lisoesfingolipídeo/antagonistas & inibidores , Receptores de Lisoesfingolipídeo/genética , Transdução de Sinais/efeitos dos fármacos , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Fibras de Estresse/metabolismo , Fatores de Tempo , Transfecção , Proteínas rac de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho
4.
J Biol Chem ; 282(12): 9082-9, 2007 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-17237497

RESUMO

Sphingosine 1-phosphate (S1P), a multifunctional lipid mediator, regulates lymphocyte trafficking, vascular permeability, and angiogenesis by activation of the S1P1 receptor. This receptor is activated by FTY720-P, a phosphorylated derivative of the immunosuppressant and vasoactive compound FTY720. However, in contrast to the natural ligand S1P, FTY720-P appears to act as a functional antagonist, even though the mechanisms involved are poorly understood. In this study, we investigated the fate of endogenously expressed S1P1 receptor in agonist-activated human umbilical vein endothelial cells and human embryonic kidney 293 cells expressing green fluorescent protein-tagged S1P1. We show that FTY720-P is more potent than S1P at inducing receptor degradation. Pretreatment with an antagonist of S1P1, VPC 44116, prevented receptor internalization and degradation. FTY720-P did not induce degradation of internalization-deficient S1P1 receptor mutants. Further, small interfering RNA-mediated down-regulation of G protein-coupled receptor kinase-2 and beta-arrestins abolished FTY720-P-induced S1P1 receptor degradation. These data suggest that agonist-induced phosphorylation of S1P1 and subsequent endocytosis are required for FTY720-P-induced degradation of the receptor. S1P1 degradation is blocked by MG132, a proteasomal inhibitor. Indeed, FTY720-P strongly induced polyubiquitinylation of S1P1 receptor, whereas S1P at concentrations that induced complete internalization was not as efficient, suggesting that receptor internalization is required but not sufficient for ubiquitinylation and degradation. We propose that the ability of FTY720-P to target the S1P1 receptor to the ubiquitinylation and proteasomal degradation pathway may at least in part underlie its immunosuppressive and anti-angiogenic properties.


Assuntos
Inibidores da Angiogênese/farmacologia , Imunossupressores/farmacologia , Propilenoglicóis/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Lisoesfingolipídeo/agonistas , Esfingosina/análogos & derivados , Ubiquitina/química , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Cloridrato de Fingolimode , Proteínas de Fluorescência Verde/metabolismo , Humanos , Leupeptinas/farmacologia , Ligantes , Microscopia Confocal , Ligação Proteica , Esfingosina/farmacologia
5.
Prostaglandins Other Lipid Mediat ; 76(1-4): 48-58, 2005 May.
Artigo em Inglês | MEDLINE | ID: mdl-15967161

RESUMO

Prostaglandin E(2) (PGE(2)), a major metabolite of the cyclooxygenase pathway in the mammary gland, induces angiogenesis during mammary tumor progression. To better define the molecular mechanisms involved, we examined the role of the G protein-coupled receptors (GPCR) for PGE(2) in mammary tumor cell lines isolated from MMTV-cyclooxygenase-2 (COX-2) transgenic mice. Expression of the EP2 subtype of the PGE(2) receptor was correlated with the tumorigenic phenotype and the ability to induce vascular endothelial growth factor (VEGF). Overexpression of EP2 by adenoviral transduction into EP2-null cells resulted in the induction of VEGF expression in response to PGE(2) and CAY10399, an EP2 receptor agonist. The induction of VEGF by the EP2 receptor did not require the hypoxia inducible factor (HIF)-1alpha pathway, MAP kinase pathway, or phosphoinositide-3-kinase/Akt pathway, but required the cAMP/protein kinase A pathway. These results suggest that EP2 receptor is a critical element for PGE(2) mediated VEGF induction in mouse mammary tumor cells.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Receptores de Prostaglandina E/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Sequência de Bases , Western Blotting , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Primers do DNA , Feminino , Imunoprecipitação , Neoplasias Mamárias Experimentais/enzimologia , Neoplasias Mamárias Experimentais/patologia , Camundongos , Receptores de Prostaglandina E Subtipo EP2 , Células Tumorais Cultivadas
6.
Proc Natl Acad Sci U S A ; 102(12): 4312-7, 2005 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-15764699

RESUMO

PTEN, a tumor suppressor phosphatase, is important in the regulation of cell migration and invasion. Physiological regulation of PTEN (phosphatase and tensin homolog deleted on chromosome 10) by cell surface receptors has not been described. Here, we show that the bioactive lipid sphingosine 1-phosphate (S1P), which acts through the S1P2 receptor (S1P2R) G protein-coupled receptor (GPCR) to inhibit cell migration, utilizes PTEN as a signaling intermediate. S1P2R inhibition of cell migration is abrogated by dominant-negative PTEN expression. S1P was unable to efficiently inhibit the migration of Pten(DeltaloxP/DeltaloxP) mouse embryonic fibroblasts; however, the antimigratory effect was restored upon the expression of PTEN. S1P2R activation of Rho GTPase is not affected in Pten(DeltaloxP/DeltaloxP) cells, and dominant-negative Rho GTPase reversed S1P inhibition of cell migration in WT cells but not in Pten(DeltaloxP/DeltaloxP) cells, suggesting that PTEN acts downstream of the Rho GTPase. Ligand activation of the S1P2R receptor stimulated the coimmunoprecipitation of S1P2R and PTEN. Interestingly, S1P2R signaling increased PTEN phosphatase activity in membrane fractions. Furthermore, tyrosine phosphorylation of PTEN was stimulated by S1P2R signaling. These data suggest that the S1P2R receptor actively regulates the PTEN phosphatase by a Rho GTPase-dependent pathway to inhibit cell migration. GPCR regulation of PTEN maybe a general mechanism in signaling events of cell migration and invasion.


Assuntos
Monoéster Fosfórico Hidrolases/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Movimento Celular/fisiologia , Células Cultivadas , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , PTEN Fosfo-Hidrolase , Monoéster Fosfórico Hidrolases/química , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/deficiência , Proteínas Tirosina Fosfatases/genética , Receptores de Lisoesfingolipídeo/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Transfecção , Proteínas Supressoras de Tumor/química , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Tirosina/química , Proteínas rho de Ligação ao GTP/metabolismo , Quinases Associadas a rho
7.
J Biol Chem ; 278(47): 47281-90, 2003 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-12954648

RESUMO

FTY720, a potent immunosuppressive agent, is phosphorylated in vivo into FTY720-P, a high affinity agonist for sphingosine 1-phosphate (S1P) receptors. The effects of FTY720 on vascular cells, a major target of S1P action, have not been addressed. We now report the metabolic activation of FTY720 by sphingosine kinase-2 and potent activation of vascular endothelial cell functions in vitro and in vivo by phosphorylated FTY720 (FTY720-P). Incubation of endothelial cells with FTY720 resulted in phosphorylation by sphingosine kinase activity and formation of FTY720-P. Sphingosine kinase-2 effectively phosphorylated FTY720 in the human embryonic kidney 293T heterologous expression system. FTY720-P treatment of endothelial cells stimulated extracellular signal-activated kinase and Akt phosphorylation and adherens junction assembly and promoted cell survival. The effects of FTY720-P were inhibited by pertussis toxin, suggesting the requirement for Gi-coupled S1P receptors. Indeed, transmonolayer permeability induced by vascular endothelial cell growth factor was potently reversed by FTY720-P. Furthermore, oral FTY720 administration in mice potently blocked VEGF-induced vascular permeability in vivo. These findings suggest that FTY720 or its analogs may find utility in the therapeutic regulation of vascular permeability, an important process in angiogenesis, inflammation, and pathological conditions such as sepsis, hypoxia, and solid tumor growth.


Assuntos
Permeabilidade Capilar/efeitos dos fármacos , Propilenoglicóis/metabolismo , Propilenoglicóis/farmacologia , Proteínas Serina-Treonina Quinases , Fator A de Crescimento do Endotélio Vascular/efeitos adversos , Junções Aderentes/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Antagonismo de Drogas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Cloridrato de Fingolimode , Humanos , Imunossupressores/administração & dosagem , Imunossupressores/metabolismo , Imunossupressores/farmacologia , Camundongos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosforilação , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Propilenoglicóis/administração & dosagem , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/fisiologia , Receptores de Lisofosfolipídeos , Esfingosina/análogos & derivados , Veias Umbilicais/citologia
8.
J Biol Chem ; 278(27): 25227-33, 2003 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-12704185

RESUMO

The cyclooxygenase-2 (COX-2) gene encodes the inducible prostaglandin synthase enzyme implicated in inflammation, cell growth, and tumorigenesis. Regulation of the COX-2 gene expression at the post-transcriptional level is poorly understood. For example, protein factors that regulate the post-transcriptional mRNA metabolism of COX-2 have not been fully characterized. In this study, we demonstrate that the RNA-binding protein HuR binds to COX-2 mRNA and regulates its expression. We show that there are three binding sites for HuR in the 3'-untranslated region of human COX-2. These sites are located at the following positions in the COX-2 3'-untranslated region: 39-84 nucleotides (nt), 1155-1187 nt, and 1244-1256 nt (hereinafter referred to as Sites I, II and III, respectively). Although all three sites are present in the 4.6-kb COX-2 mRNA, only site I is present in the shorter 2.8-kb isoform. HuR in MDA-MB-231 cell extracts associated with COX-2 mRNA at the identified sites. Further, HuR location in the cytoplasm was induced by serum withdrawal, a stimulus known to induce COX-2 mRNA. Down-regulation of HuR by two independent methods, namely RNA interference as well as antisense RNA expression, significantly attenuated serum withdrawal-induced increase in COX-2 mRNA (both the 4.6- and 2.8-kb isoforms) and protein levels. These data suggest that HuR binding to COX-2 is critical for its post-transcriptional mRNA stabilization.


Assuntos
Antígenos de Superfície , Isoenzimas/genética , Prostaglandina-Endoperóxido Sintases/genética , Proteínas de Ligação a RNA/genética , Sequência de Bases , Ciclo-Oxigenase 2 , Proteínas ELAV , Proteína Semelhante a ELAV 1 , Regulação Enzimológica da Expressão Gênica , Humanos , Isoenzimas/biossíntese , Proteínas de Membrana , Dados de Sequência Molecular , Prostaglandina-Endoperóxido Sintases/biossíntese , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas de Ligação a RNA/metabolismo , Alinhamento de Sequência , Células Tumorais Cultivadas
9.
FEBS Lett ; 533(1-3): 25-8, 2003 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-12505153

RESUMO

Sphingosine 1-phosphate (S1P), a bioactive lipid mediator, signals via G protein-coupled receptors (GPCR). The prototypical S1P receptor, S1P1 (also known as EDG-1), a Gi-linked receptor, is critical for vascular maturation during development. Recent work suggested that platelet-derived growth factor (PDGF)-induced cell migration required the S1P1 receptor, representing a novel mechanism for cross-talk between receptor tyrosine kinases and GPCRs. Since both S1P and PDGF are implicated in vascular smooth muscle cell (VSMC) pathobiology and development, we investigated this issue in rat VSMC and in embryonic fibroblasts derived from S1P1 null mice. Our data suggest that the S1P1 receptor is critical for S1P-induced, Gi-dependent migration but not for PDGF-BB-induced, receptor tyrosine kinase-dependent chemotaxis in VSMC. In addition, lack of S1P1 receptor in mouse embryonic fibroblasts did not significantly affect PDGF-induced cell migration. These data question the generality of the concept that S1P1 GPCR is a critical downstream component of PDGF-induced chemotaxis.


Assuntos
Quimiotaxia/efeitos dos fármacos , Fator de Crescimento Derivado de Plaquetas/farmacologia , Receptores de Superfície Celular/fisiologia , Receptores Acoplados a Proteínas G , Animais , Linhagem Celular , Quimiotaxia/fisiologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/fisiologia , Proteínas de Ligação ao GTP/metabolismo , Camundongos , Camundongos Knockout , Músculo Liso Vascular/citologia , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/fisiologia , Fator de Crescimento Derivado de Plaquetas/fisiologia , Ratos , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores de Lisofosfolipídeos , Esfingosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...