Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Biochim Biophys Acta ; 1788(9): 1695-705, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19426705

RESUMO

Juvenile hormone (JH) controls insect development, metamorphosis and reproduction. In insect hemolymph a significant proportion of JH is bound to juvenile hormone binding protein (JHBP), which serves as a carrier supplying the hormone to the target tissues. To shed some light on JHBP passage within insect tissues, the interaction of this carrier with other proteins from Galleria mellonella (Lepidoptera) was investigated. Our studies revealed the presence of JHBP within the tracheal epithelium and fat body cells in both the membrane and cytoplasmic sections. We found that the interaction between JHBP and membrane proteins occurs with saturation kinetics and is specific and reversible. ATP synthase was indicated as a JHBP membrane binding protein based upon SPR-BIA and MS analysis. It was found that in G. mellonella fat body, this enzyme is present in mitochondrial fraction, plasma membranes and cytosol as well. In the model system containing bovine F(1) ATP synthase and JHBP, the interaction between these two components occurs with K(d)=0.86 nM. In hemolymph we detected JHBP binding to apolipophorin, arylphorin and hexamerin. These results provide the first demonstration of the physical interaction of JHBP with membrane and hemolymph proteins which can be involved in JHBP molecule traffic.


Assuntos
Proteínas de Transporte/metabolismo , Proteínas de Insetos/metabolismo , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Transporte Proteico/fisiologia , Animais , Apolipoproteínas/metabolismo , Corpo Adiposo/química , Corpo Adiposo/ultraestrutura , Hemolinfa/metabolismo , Hormônios Juvenis/metabolismo , Proteínas de Membrana/metabolismo , Mariposas , Ressonância de Plasmônio de Superfície
2.
Mol Pharmacol ; 75(3): 502-13, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19064631

RESUMO

Signaling of G protein-coupled receptors (GPCRs) is regulated by different mechanisms. One of these involves regulators of G protein signaling (RGS), which are diverse and multifunctional proteins that bind to active Galpha subunits of G proteins and act as GTPase-activating proteins. Little is known about the molecular mechanisms that govern the selective use of RGS proteins in living cells. We first demonstrated that CCK2R-mediated inositol phosphate production, known to be G(q)-dependent, is more sensitive to RGS2 than to RGS4 and is insensitive to RGS8. Both basal and agonist-stimulated activities of the CCK2R are regulated by RGS2. By combining biochemical, functional, and in silico structural approaches, we demonstrate that a direct and functional interaction occurs between RGS2 and agonist-stimulated cholecystokinin receptor-2 (CCK2R) and identified the precise residues involved: phosphorylated Ser434 and Thr439 located in the C-terminal tail of CCK2R and Lys62, Lys63, and Gln67, located in the N-terminal domain of RGS2. These findings confirm previous reports that RGS proteins can interact with GPCRs to modulate their signaling and provide a molecular basis for RGS2 recognition by the CCK2R.


Assuntos
Fragmentos de Peptídeos/fisiologia , Proteínas RGS/fisiologia , Receptor de Colecistocinina B/fisiologia , Transdução de Sinais/fisiologia , Motivos de Aminoácidos/fisiologia , Sequência de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Fosforilação/fisiologia , Ligação Proteica/fisiologia , Proteínas RGS/química , Proteínas RGS/metabolismo , Receptor de Colecistocinina B/química , Receptor de Colecistocinina B/metabolismo
3.
Mol Endocrinol ; 21(1): 229-46, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17021051

RESUMO

The receptor-like phosphotyrosine phosphatase eta (PTPeta) is an important intracellular effector of the cytostatic action of SST. Here we characterize, in Chinese hamster ovary-k1 cells, the intracellular pathway that from somatostatin receptor 1 (SSTR1), leads to the activation of PTPeta and that involves, in a multimeric complex and sequential activation, the tyrosine kinases Janus kinase (JAK) 2 and Src, and the cytosolic phosphotyrosine phosphatase SHP-2. We show that inhibitors of JAK2 and Src and dominant-negative mutants of SHP-2 and Src abolished the SSTR1-mediated PTPeta activation, suggesting that all these effectors participate in the activation of PTPeta. In basal conditions, JAK2 forms a multimeric complex with SHP-2, Src and PTPeta. In response to SST, JAK2 is activated in a G protein-dependent manner, dissociates from and phosphorylates SHP-2, increasing its activity. Subsequently, SHP-2 dissociates from Src, dephosphorylates the Src inhibitory tyrosine-529, and causes an autocatalytical increase of the phosphorylation of Src tyrosine 418, located inside its kinase activation loop. Active Src, in turn, controls the activity of PTPeta, via a direct interaction and phosphorylation of the phosphatase. These data for the first time depict an intracellular pathway involving a precise sequence of interactions and cross-activation among tyrosine phosphatases and kinases acting upstream of PTPeta. In particular the sequential activation of JAK2, SHP-2, and Src conveys the molecular signaling from SSTR1 to the activation of this phosphatase that is responsible for the final biological effects of SST.


Assuntos
Proteínas Tirosina Fosfatases/química , Receptores de Somatostatina/metabolismo , Animais , Células CHO , Cricetinae , Cricetulus , Citosol/enzimologia , Ativação Enzimática , Genes Dominantes , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Janus Quinase 2/metabolismo , Toxina Pertussis/farmacologia , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/metabolismo , Ratos , Transdução de Sinais , Quinases da Família src/metabolismo
4.
Biochim Biophys Acta ; 1763(10): 1098-107, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16963136

RESUMO

SHP-2 is a tyrosine phosphatase which functions as a positive regulator downstream of RTKs, activating growth-stimulatory signalling pathways. To date, very few G protein-coupled receptors (GPCRs) have been shown to be connected to SHP-2 and very little is known about the positive role of SHP-2 in GPCR signalling. The CCK2 receptor (CCK2R), a GPCR, is now recognized to mediate mitogenic effects of gastrin on gastrointestinal cells. In the present study, we demonstrate the role of SHP-2 in the activation of the AKT pathway by the CCK2R in COS-7 cells transfected with the CCK2R and in a pancreatic cancer cell line expressing the endogenous receptor. Using surface plasmon resonance analysis, we identified a highly conserved ITIM motif, containing the tyrosine residue 438, located in the C-terminal intracellular tail of the CCK2R which directly interacts with the SHP-2 SH2 domains. The interaction was confirmed by pull down assays and co-immunoprecipitation of the receptor with SHP-2. This interaction was transiently increased following gastrin stimulation of the CCK2R and correlated with the tyrosine phosphorylation of SHP-2. Mutational analysis of the key ITIM residue 438 confirmed that the CCK2R ITIM sequence is required for interaction with SHP-2 and the activation of the AKT pathway.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias Pancreáticas/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Colecistocinina B/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Gastrinas/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Camundongos , Dados de Sequência Molecular , Células NIH 3T3 , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/fisiologia , Ratos , Receptor de Colecistocinina B/genética , Receptor de Colecistocinina B/fisiologia , Transdução de Sinais , Tirosina/metabolismo
5.
Mol Biol Cell ; 14(9): 3911-28, 2003 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12972574

RESUMO

The G protein-coupled sst2 somatostatin receptor acts as a negative cell growth regulator. Sst2 transmits antimitogenic signaling by recruiting and activating the tyrosine phosphatase SHP-1. We now identified Src and SHP-2 as sst2-associated molecules and demonstrated their role in sst2 signaling. Surface plasmon resonance and mutation analyses revealed that SHP-2 directly associated with phosphorylated tyrosine 228 and 312, which are located in sst2 ITIMs (immunoreceptor tyrosine-based inhibitory motifs). This interaction was required for somatostatin-induced SHP-1 recruitment and activation and consequent inhibition of cell proliferation. Src interacted with sst2 and somatostatin promoted a transient Gbetagamma-dependent Src activation concomitant with sst2 tyrosine hyperphosphorylation and SHP-2 activation. These steps were abrogated with catalytically inactive Src. Both catalytically inactive Src and SHP-2 mutants abolished somatostatin-induced SHP-1 activation and cell growth inhibition. Sst2-Src-SHP-2 complex formation was dynamic. Somatostatin further induced sst2 tyrosine dephosphorylation and complex dissociation accompanied by Src and SHP-2 inhibition. These steps were defective in cells expressing a catalytically inactive Src mutant. All these data suggest that Src acts upstream of SHP-2 in sst2 signaling and provide evidence for a functional role for Src and SHP-2 downstream of an inhibitory G protein-coupled receptor.


Assuntos
Proteínas Tirosina Fosfatases/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Quinases da Família src/metabolismo , Motivos de Aminoácidos/fisiologia , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Células COS , Divisão Celular/fisiologia , Chlorocebus aethiops , Cricetinae , Ciclofosfamida , Análise Mutacional de DNA , Doxorrubicina , Peptídeos e Proteínas de Sinalização Intracelular , Modelos Moleculares , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Ratos , Receptores de Somatostatina/análise , Somatostatina/farmacologia , Ressonância de Plasmônio de Superfície , Vincristina
6.
Biosens Bioelectron ; 21(8): 1566-73, 2006 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-16099641

RESUMO

In order to detect xenoestrogens which induce perturbations of mammalian cells, design of biosensor using a mammalian cell line enable to detect these compounds is necessary. MELN cell line is suitable to detect estrogen activity, since they are stably transfect with an estrogen regulated luciferase gene. To realize this biosensor, it appeared necessary to add a protection to the mamalian cell, which is devoided, of the wall protecting yeasts or plant cells. With this aim in view, MELN cells have been isolated with a polyelectrolyte shell using the layer-by-layer technique. Among several polyelectrolyte-couples, the best cell survival (>80%) was obtained by alternating the polycation poly-diallyldimethyl ammonium chloride layer and the negatively charged poly-styrene sulfonate. We observed that the composition of the buffer used for layer-deposition was crucial to preserving cell viability, e.g. potassium ions were preferred to sodium ions during the coating. Furthermore, viability was increased when cells were allowed to recover for 2 h between each bilayer deposition. The use of engineered mammalian cells that synthesize luciferase as a response to exposure to estradiol, demonstrated that coating not only permits cell survival, but also allows essential metabolic functions, such as RNA and protein synthesis to take place. Capsule formation allows free diffusion of small molecules, while it prevents internalization in the cells of proteins larger than 60 kDa.


Assuntos
Cloreto de Amônio/química , Bioensaio/métodos , Técnicas Biossensoriais/métodos , Neoplasias da Mama/patologia , Técnicas de Cultura de Células/métodos , Estrogênios/administração & dosagem , Poliestirenos/química , Bioensaio/instrumentação , Técnicas Biossensoriais/instrumentação , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Materiais Revestidos Biocompatíveis/química , Estrogênios/análise , Humanos
7.
J Invest Dermatol ; 122(3): 747-54, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15086562

RESUMO

Corneodesmosin (CDSN), a glycoprotein expressed during the late stages of epidermal differentiation, localizes in the extracellular core of upper desmosomes and of corneodesmosomes. Since it displays homophilic adhesive properties, CDSN is thought to reinforce cell-cell cohesion within the upper layers of the epidermis. CDSN presents two serine- and glycine-rich domains in its N- and C-terminus that may fold into highly flexible and adhesive secondary structures called glycine loops. We analyzed the importance of these domains in CDSN homophilic adhesion by producing full-length and truncated recombinant forms of the protein deleted of the N- and/or the C-terminal domain. The adhesive properties of the various proteins were then tested in vitro by overlay binding assays and surface plasmon resonance quantitative analysis. Experiments evidenced the homophilic adhesive properties of the N-terminal glycine loop domain, confirming its involvement in CDSN-CDSN interactions. They further indicated that most of the C-terminal domain is not necessary for the adhesive properties of the protein. The dissociation constant (K(D)) was calculated to be 1.3x10(-5) M. This interaction strength might allow dynamic regulation of the CDSN-CDSN association to occur in vivo. Moreover, molecular filtration analyses demonstrated for the first time that non-glycosylated CDSN is able to spontaneously form large homo-oligomers in vitro and that the N-terminal glycine loop domain is necessary for the formation of these macromolecular complexes.


Assuntos
Glicoproteínas/química , Sequência de Aminoácidos , Dimerização , Glicina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Dados de Sequência Molecular , Ressonância de Plasmônio de Superfície
8.
FEBS Lett ; 568(1-3): 89-93, 2004 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-15196926

RESUMO

In ElasCCK2 transgenic mice expressing cholecystokinin (CCK2) receptor in acinar cells, pancreatic phenotypic alterations and preneoplastic lesions are observed. We determined whether activation of phospholipase C gamma1 (PLCgamma1), known to contribute to the tumorigenesis pathophysiology, could take place as a new signaling pathway induced by the CCK2 receptor. Overexpression and activation of the PLCgamma1 in response to gastrin was observed in acinar cells. The possibility that the C-terminal tyrosine 438 of the CCK2 receptor associates with the SH2 domains of PLCgamma1 was examined. A specific interaction was demonstrated using surface plasmon resonance, confirmed in a cellular system and by molecular modeling.


Assuntos
Receptor de Colecistocinina B/metabolismo , Fosfolipases Tipo C/metabolismo , Sequência de Aminoácidos , Animais , Ativação Enzimática , Imuno-Histoquímica , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Fosfolipase C gama , Ligação Proteica , Receptor de Colecistocinina B/química , Ressonância de Plasmônio de Superfície , Tirosina/metabolismo
9.
Mol Cell Biol ; 32(5): 1004-16, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22203038

RESUMO

Frequent oncogenic alterations occur in the phosphoinositide 3-kinase (PI3K) pathway, urging identification of novel negative controls. We previously reported an original mechanism for restraining PI3K activity, controlled by the somatostatin G protein-coupled receptor (GPCR) sst2 and involving a ligand-regulated interaction between sst2 with the PI3K regulatory p85 subunit. We here identify the scaffolding protein filamin A (FLNA) as a critical player regulating the dynamic of this complex. A preexisting sst2-p85 complex, which was shown to account for a significant basal PI3K activity in the absence of ligand, is disrupted upon sst2 activation. FLNA was here identified as a competitor of p85 for direct binding to two juxtaposed sites on sst2. Switching of GPCR binding preference from p85 toward FLNA is determined by changes in the tyrosine phosphorylation of p85- and FLNA-binding sites on sst2 upon activation. It results in the disruption of the sst2-p85 complex and the subsequent inhibition of PI3K. Knocking down FLNA expression, or abrogating FLNA recruitment to sst2, reversed the inhibition of PI3K and of tumor growth induced by sst2. Importantly, we report that this FLNA inhibitory control on PI3K can be generalized to another GPCR, the mu opioid receptor, thereby providing an unprecedented mechanism underlying GPCR-negative control on PI3K.


Assuntos
Classe Ia de Fosfatidilinositol 3-Quinase/metabolismo , Proteínas Contráteis/metabolismo , Proteínas dos Microfilamentos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais , Animais , Sítios de Ligação , Ligação Competitiva , Linhagem Celular , Filaminas , Fosforilação , Ligação Proteica , Subunidades Proteicas/genética
10.
FEBS J ; 276(17): 4866-74, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19664057

RESUMO

Transferrin is the main iron transport protein found in the circulation, and the level of transferrin saturation in the blood is an important indicator of iron status. The peptides amidated gastrin(17) (Gamide) and glycine-extended gastrin(17) (Ggly) are well known for their roles in controlling acid secretion and as growth factors in the gastrointestinal tract. Several lines of evidence, including the facts that transferrin binds gastrin, that gastrins bind ferric ions, and that the level of expression of gastrins positively correlates with transferrin saturation, suggest the possible involvement of the transferrin-gastrin interaction in iron homeostasis. In the present work, the interaction between gastrins and transferrin has been characterized by surface plasmon resonance and covalent crosslinking. First, an interaction between iron-free apo-transferrin and Gamide or Ggly was observed. The fact that no interaction was observed in the presence of the chelator EDTA suggested that the gastrin-ferric ion complex was the interacting species. Moreover, removal of ferric ions with EDTA reduced the stability of the complex between apo-transferrin and gastrins, and no interaction was observed between Gamide or Ggly and diferric transferrin. Second, some or all of glutamates at positions 8-10 of the Ggly molecule, together with the C-terminal domain, were necessary for the interaction with apo-transferrin. Third, monoferric transferrin mutants incapable of binding iron in either the N-terminal or C-terminal lobe still bound Ggly. These findings are consistent with the hypothesis that gastrin peptides bind to nonligand residues within the open cleft in each lobe of transferrin and are involved in iron loading of transferrin in vivo.


Assuntos
Gastrinas/química , Glicina/química , Transferrina/química , Cátions , Reagentes de Ligações Cruzadas/química , Ácido Edético/química , Gastrinas/genética , Glicina/genética , Ferro/química , Quelantes de Ferro/química , Mutação , Ligação Proteica
11.
J Biol Chem ; 281(28): 19156-71, 2006 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-16690617

RESUMO

Somatostatin receptor SST5 is an inhibitory G protein-coupled receptor that exerts a strong cytostatic effect on various cell types. We reported previously that the SST5 anti-proliferative effect results in the inhibition of mitogen-induced increases in intracellular cGMP levels and MAPK activity. This study was conducted to define the early molecular events accountable for the SST5-mediated anti-proliferative effect. Here, we demonstrate that, in Chinese hamster ovary cells expressing SST5 (CHO/SST5 cells), somatostatin inhibited cell proliferation induced by nitric oxide donors and overexpression of the neuronal nitric-oxide synthase (nNOS) protein isoform. Accordingly, nNOS activity and dimerization were strongly inhibited following SST5 activation by the somatostatin analog RC-160. In CHO/SST5 cells, nNOS was dynamically recruited by the SST5 receptor and phosphorylated at tyrosyl residues following RC-160 treatment. RC-160 induced SST5-p60(src) kinase complex formation and subsequent p60(src) kinase activation. Coexpression of an inactive p60(src) kinase mutant with SST5 blocked RC-160-induced nNOS phosphorylation and inactivation and prevented the SST5-mediated anti-proliferative effect. In CHO/SST5 cells, p60(src) kinase associated with nNOS to induce its inactivation by phosphorylation at tyrosyl residues following RC-160 treatment. Using recombinant proteins, we demonstrated that such phosphorylation prevented nNOS homodimerization. Next, surface plasmon resonance and mutation analysis revealed that p60(src) directly associated with nNOS phosphorylated Tyr604. SST5-mediated inhibition of nNOS activity was demonstrated to be essential to the RC-160 anti-proliferative effect on pancreatic endocrine tumor-derived cells. We therefore identified nNOS as a new p60(src) kinase substrate essential for SST5-mediated anti-proliferative action.


Assuntos
Regulação Enzimológica da Expressão Gênica , Óxido Nítrico Sintase Tipo I/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Animais , Células CHO , Linhagem Celular Tumoral , Proliferação de Células , Cricetinae , Dimerização , Humanos , Fosforilação , Isoformas de Proteínas , Proteínas Proto-Oncogênicas pp60(c-src)/química , Ratos , Proteínas Recombinantes
12.
EMBO J ; 25(17): 3943-54, 2006 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-16917505

RESUMO

Phosphatidylinositol 3-kinase (PI3K) regulates many cellular functions including growth and survival, and its excessive activation is a hallmark of cancer. Somatostatin, acting through its G protein-coupled receptor (GPCR) sst2, has potent proapoptotic and anti-invasive activities on normal and cancer cells. Here, we report a novel mechanism for inhibiting PI3K activity. Somatostatin, acting through sst2, inhibits PI3K activity by disrupting a pre-existing complex comprising the sst2 receptor and the p85 PI3K regulatory subunit. Surface plasmon resonance and molecular modeling identified the phosphorylated-Y71 residue of a p85-binding pYXXM motif in the first sst2 intracellular loop, and p85 COOH-terminal SH2 as direct interacting domains. Somatostatin-mediated dissociation of this complex as well as p85 tyrosine dephosphorylation correlates with sst2 tyrosine dephosphorylation on the Y71 residue. Mutating sst2-Y71 disabled sst2 to interact with p85 and somatostatin to inhibit PI3K, consequently abrogating sst2's ability to suppress cell survival and tumor growth. These results provide the first demonstration of a physical interaction between a GPCR and p85, revealing a novel mechanism for negative regulation by ligand-activated GPCR of PI3K-dependent survival pathways, which may be an important molecular target for antineoplastic therapy.


Assuntos
Fosfatidilinositol 3-Quinases/fisiologia , Receptores de Somatostatina/fisiologia , Somatostatina/fisiologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Ativação Enzimática , Feminino , Humanos , Camundongos , Camundongos Nus , Mutação , Transplante de Neoplasias , Neoplasias Experimentais/patologia , Fosfatidilinositol 3-Quinases/genética , Fosforilação , Ligação Proteica , Receptores de Somatostatina/genética , Transdução de Sinais , Ressonância de Plasmônio de Superfície , Transplante Heterólogo , Tirosina/metabolismo , Domínios de Homologia de src
13.
Chembiochem ; 6(1): 123-32, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15551357

RESUMO

The capacity of G-quadruplex ligands to stabilize four-stranded DNA makes them able to inhibit telomerase, which is involved in tumour cell proliferation. A series of cationic metalloporphyrin derivatives was prepared by making variations on a meso-tetrakis(4-N-methyl-pyridiniumyl)porphyrin skeleton (TMPyP). The DNA binding properties of nickel(II) and manganese(III) porphyrins were studied by surface plasmon resonance, and the capacity of the nickel porphyrins to inhibit telomerase was tested in a TRAP assay. The nature of the metal influences the kinetics (the process is faster for Ni than for Mn) and the mode of interaction (stacking or external binding). The chemical alterations did not lead to increased telomerase inhibition. The best selectivity for G-quadruplex DNA was observed for Mn-TMPyP, which has a tenfold preference for quadruplex over duplex.


Assuntos
Inibidores Enzimáticos/química , Metaloporfirinas/química , Telomerase/antagonistas & inibidores , Telômero/metabolismo , DNA/metabolismo , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/metabolismo , Quadruplex G , Humanos , Manganês/química , Manganês/metabolismo , Metaloporfirinas/síntese química , Metaloporfirinas/metabolismo , Níquel/química , Níquel/metabolismo , Técnicas de Amplificação de Ácido Nucleico , Ressonância de Plasmônio de Superfície
14.
Biochem Biophys Res Commun ; 326(4): 894-900, 2005 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-15607753

RESUMO

Recently, we have described a novel protein-protein interaction between the G-protein coupled bradykinin B2 receptor and tyrosine phosphatase SHP-2 via an immunoreceptor tyrosine-based inhibition motif (ITIM) sequence located in the C-terminal part of the B2 receptor and the Src homology (SH2) domains of SHP-2. Here we show that phospholipase C (PLC)gamma1, another SH2 domain containing protein, can also interact with this ITIM sequence. Using surface plasmon resonance analysis, we observed that PLCgamma1 interacted with a peptide containing the phosphorylated form of the bradykinin B2 receptor ITIM sequence. In CHO cells expressing the wild-type B2 receptor, bradykinin-induced transient recruitment and activation of PLCgamma1. Interestingly, this interaction was only observed in quiescent and not in proliferating cells. Mutation of the key ITIM residue abolished this interaction with and activation of PLCgamma1. Finally we also identified bradykinin-induced PLCgamma1 recruitment and activation in primary culture renal mesangial cells.


Assuntos
Mapeamento de Interação de Proteínas/métodos , Receptor B2 da Bradicinina/química , Receptor B2 da Bradicinina/metabolismo , Fosfolipases Tipo C/química , Fosfolipases Tipo C/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proliferação de Células , Cricetinae , Cricetulus , Dados de Sequência Molecular , Fosfolipase C gama , Ligação Proteica , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Ressonância de Plasmônio de Superfície/métodos
15.
Immunity ; 22(1): 71-80, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15664160

RESUMO

Vgamma9Vdelta2 T lymphocytes, a major gammadelta T lymphocyte subset in humans, display cytolytic activity against various tumor cells upon recognition of yet uncharacterized structures. Here, we show that an entity related to the mitochondrial F1-ATPase is expressed on tumor cell surface and promotes tumor recognition by Vgamma9Vdelta2 T cells. When immobilized, purified F1-ATPase induces selective activation of this lymphocyte subset. The Vgamma9Vdelta2 T cell receptors (TCR) and the F1-ATPase also bind a delipidated form of apolipoprotein A-I (apo A-I), as demonstrated by surface plasmon resonance. Moreover, the presence of apo A-I in the culture medium is required for optimal activation of Vgamma9Vdelta2 T cells by tumors expressing F1-ATPase. This study thus describes an unanticipated tumor recognition mechanism by Vgamma9Vdelta2 lymphocytes and a possible link between gammadelta T cell immunity and lipid metabolism.


Assuntos
Apolipoproteína A-I/metabolismo , Neoplasias/imunologia , ATPases Translocadoras de Prótons/metabolismo , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Receptores de Antígenos de Linfócitos T/metabolismo , Humanos , Imobilização , Células Jurkat , Cinética , Ligantes , Ativação Linfocitária , Ligação Proteica , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Ressonância de Plasmônio de Superfície , Propriedades de Superfície , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Células Tumorais Cultivadas
16.
Proteomics ; 3(4): 402-12, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12687608

RESUMO

Biological functions of most macromolecules depend on their ability to interact with other molecules and a great challenge is the complete description of the protein interaction networks. Biomolecular interaction analysis (BIA) is an optical technology that uses the surface plasmon resonance phenomenon for characterizing macromolecular interactions between an analyte in solution and its ligand immobilized on a sensor chip. Further identification of interacting proteins can be achieved by combining this nondestructive method to mass spectrometry (MS). The BIA-MS approach represents a promising tool in proteomics for the characterization of protein/protein interactions. In this study, we report on the improved sensitivity in the identification of an unknown protein bound to a known ligand by a rapid and simple BIA-MS approach. We took advantage of a new automatic and very reproducible microelution procedure available on BIACORE 3000 instruments, called "microrecovery", to elute the bound protein from the sensor chip. Protein identification was then achieved after tryptic digestion by matrix-assisted laser desorption/ionization-time of flight mass mapping and database search. The strategy was succesfully applied to the model protein SHP2 tyrosine phosphatase interacting with an immunoreceptor tyrosine-based inhibitory motif sequence of the sst2 somatostatin receptor. Optimization of the BIA-MS approach allowed the unambiguous identification of 10-20 fmol of the protein specifically trapped from a complex mixture of cytosolic extracts.


Assuntos
Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz/métodos , Sequência de Aminoácidos , Animais , Células CHO , Cromatografia de Afinidade , Cricetinae , Citosol/química , Espectroscopia de Ressonância de Spin Eletrônica , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/análise , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Somatostatina/química , Receptores de Somatostatina/metabolismo , Proteínas Recombinantes de Fusão/análise , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
17.
J Biol Chem ; 278(41): 39356-71, 2003 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-12878607

RESUMO

The G protein-coupled sst2 somatostatin receptor is a critical negative regulator of cell proliferation. sstII prevents growth factor-induced cell proliferation through activation of the tyrosine phosphatase SHP-1 leading to induction of the cyclin-dependent kinase inhibitor p27Kip1. Here, we investigate the signaling molecules linking sst2 to p27Kip1. In Chinese hamster ovary-DG-44 cells stably expressing sst2 (CHO/sst2), the somatostatin analogue RC-160 transiently stimulates ERK2 activity and potentiates insulin-stimulated ERK2 activity. RC-160 also stimulates ERK2 activity in pancreatic acini isolated from normal mice, which endogenously express sst2, but has no effect in pancreatic acini derived from sst2 knock-out mice. RC-160-induced p27Kip1 up-regulation and inhibition of insulin-dependent cell proliferation are both prevented by pretreatment of CHO/sst2 cells with the MEK1/2 inhibitor PD98059. In addition, using dominant negative mutants, we show that sst2-mediated ERK2 stimulation is dependent on the pertussis toxin-sensitive Gi/o protein, the tyrosine kinase Src, both small G proteins Ras and Rap1, and the MEK kinase B-Raf but is independent of Raf-1. Phosphatidylinositol 3-kinase (PI3K) and both tyrosine phosphatases, SHP-1 and SHP-2, are required upstream of Ras and Rap1. Taken together, our results identify a novel mechanism whereby a Gi/o protein-coupled receptor inhibits cell proliferation by stimulating ERK signaling via a SHP-1-SHP-2-PI3K/Ras-Rap1/B-Raf/MEK pathway.


Assuntos
Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-raf/metabolismo , Receptores de Somatostatina/metabolismo , Somatostatina/análogos & derivados , Proteínas rap1 de Ligação ao GTP/metabolismo , Proteínas ras/metabolismo , Animais , Células CHO , Proteínas de Ciclo Celular/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/fisiologia , Cricetinae , Inibidor de Quinase Dependente de Ciclina p27 , Ativação Enzimática/efeitos dos fármacos , Insulina/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína Quinase 3 Ativada por Mitógeno , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Modelos Biológicos , Pâncreas/efeitos dos fármacos , Pâncreas/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Proteínas Proto-Oncogênicas B-raf , Receptores de Somatostatina/deficiência , Receptores de Somatostatina/genética , Somatostatina/farmacologia , Proteínas Supressoras de Tumor/metabolismo
18.
J Biol Chem ; 277(43): 40375-83, 2002 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-12177051

RESUMO

Mitogenic G protein-coupled receptor (GPCR) signaling has been extensively studied. In contrast, little is known about anti-mitogenic GPCR signaling. We show here that anti-mitogenic signaling of a GPCR, the bradykinin B2 receptor, involves a novel direct protein-protein interaction. The antiproliferative effect of bradykinin was accompanied by a transient increase in protein-tyrosine phosphatase activity. Using surface plasmon resonance analysis, we observed that an immunoreceptor tyrosine-based inhibitory motif (ITIM) located in the C-terminal part of the B2 receptor interacted specifically with the protein-tyrosine phosphatase SHP-2. The interaction was confirmed in primary culture renal mesangial cells by co-immunoprecipitation of a B2 receptor.SHP-2 complex. The extent of the interaction was transiently increased by stimulation with bradykinin, which was accompanied by an increase in specific SHP-2 phosphatase activity. Mutational analysis of the key ITIM residue confirmed that the B2 receptor ITIM sequence is required for interaction with SHP-2, SHP-2 activation, and the anti-mitogenic effect of bradykinin. Finally, in mesangial cells transfected with a dominant-negative form of SHP-2, bradykinin lost the ability to inhibit cell proliferation. These observations demonstrate that bradykinin inhibits cell proliferation by a novel mechanism involving a direct protein-protein interaction between a GPCR (the B2 receptor) and SHP-2.


Assuntos
Bradicinina/farmacologia , Divisão Celular/efeitos dos fármacos , Proteínas de Ligação ao GTP/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptores da Bradicinina/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Células Cultivadas , Primers do DNA , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Dados de Sequência Molecular , Fosforilação , Testes de Precipitina , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Receptor B2 da Bradicinina , Homologia de Sequência de Aminoácidos , Tirosina/metabolismo
19.
Nature ; 421(6918): 75-9, 2003 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-12511957

RESUMO

The effect of high-density lipoprotein (HDL) in protecting against atherosclerosis is usually attributed to its role in 'reverse cholesterol transport'. In this process, HDL particles mediate the efflux and the transport of cholesterol from peripheral cells to the liver for further metabolism and bile excretion. Thus, cell-surface receptors for HDL on hepatocytes are chief partners in the regulation of cholesterol homeostasis. A high-affinity HDL receptor for apolipoprotein A-I (apoA-I) was previously identified on the surface of hepatocytes. Here we show that this receptor is identical to the beta-chain of ATP synthase, a principal protein complex of the mitochondrial inner membrane. Different experimental approaches confirm this ectopic localization of components of the ATP synthase complex and the presence of ATP hydrolase activity at the hepatocyte cell surface. Receptor stimulation by apoA-I triggers the endocytosis of holo-HDL particles (protein plus lipid) by a mechanism that depends strictly on the generation of ADP. We confirm this effect on endocytosis in perfused rat liver ex vivo by using a specific inhibitor of ATP synthase. Thus, membrane-bound ATP synthase has a previously unsuspected role in modulating the concentrations of extracellular ADP and is regulated by a principal plasma apolipoprotein.


Assuntos
Endocitose , Hepatócitos/citologia , Hepatócitos/metabolismo , Lipoproteínas HDL/metabolismo , ATPases Mitocondriais Próton-Translocadoras/química , ATPases Mitocondriais Próton-Translocadoras/metabolismo , Receptores de Lipoproteínas/metabolismo , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Apolipoproteína A-I/metabolismo , Linhagem Celular , Cricetinae , Endocitose/efeitos dos fármacos , Citometria de Fluxo , Imunofluorescência , Hepatócitos/efeitos dos fármacos , Humanos , Estrutura Secundária de Proteína , Ratos , Ressonância de Plasmônio de Superfície , Suínos , Células Tumorais Cultivadas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA