Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 165
Filtrar
1.
J Gynecol Obstet Biol Reprod (Paris) ; 38(7): 595-8, 2009 Nov.
Artigo em Francês | MEDLINE | ID: mdl-19540080

RESUMO

Takayasu's arteritis is a non-specific chronic vasculitis mainly involving the aorta and its main branches. A 37 year-old patient was diagnosed a Takayasu's arteritis during her last pregnancy. Her new pregnancy was characterised by a preeclampsia and an intrauterine growth restriction complicated by an intrauterine fetal death during the second trimester. Takayasu's arteritis is at risk of life-threatening complications for both the mother and the fetus. A multidisciplinary survey is recommended. Currently, management of this disease is unclear and no consensus is available during pregnancy. Trials testing antiplatelet agents and corticosteroids are now needed.


Assuntos
Morte Fetal/etiologia , Complicações Cardiovasculares na Gravidez , Arterite de Takayasu/complicações , Adulto , Feminino , Retardo do Crescimento Fetal/diagnóstico , Humanos , Pré-Eclâmpsia/diagnóstico , Gravidez
2.
J Mal Vasc ; 33(1): 1-11, 2008 Feb.
Artigo em Francês | MEDLINE | ID: mdl-18187280

RESUMO

INTRODUCTION: Peripheral arterial disease (PAD) is a frequent and serious condition with a risk of mortality comparable to that of certain cancers. However, in France, the literature on this medical condition is scarce and data on management, incidence of complications and prognosis are lacking. PURPOSES: The COPART I registry, set up in June 2004, in the Vascular Medicine Department of the University Hospital of Toulouse, France, constitutes an observational database on hospitalized patients with PAD, in order to evaluate management, follow-up and prognosis of the patients. The aim of the present work is to compare medical prescriptions at hospital discharge, with the recent guidelines of the French High Authority of Health. METHODS: All consecutive patients with PAD, hospitalized in the Vascular Medicine Department of the University of Toulouse, between June 1, 2004 and July 31, 2006 were included. Only surviving patients were analysed. RESULTS: Four hundred patients were included in the study. As expected, the majority were male (70%). Common cardiovascular risk factors were: arterial hypertension (66.7%), dyslipidemia (58.9%), diabetes (42.9%), and smoking (27.4%). Three patients out of 10 had claudication intermittens, nearly two out of 10 patients complained of persistent pain, and four out of 10 patients had Leriche and Fontaine stage IV arteriopathy. At hospital discharge, 86.9% of the patients were taking at least one antiplatelet treatment, 71.2% a statin, 54% a renin-angiotensin-system inhibitor. Nearly 66% of the patients (65.8%) received at least one antiplatelet agent and a statin. Nearly 50% of the patients (49.4%) had the three drugs recommended by the French High Authority of Health. We observed a change in prescription practices for statins (+30%), as well as for prescription of evidence-based tri-therapy (+29%) between 2004 and 2006. CONCLUSION: Treatments prescribed at hospital discharge of patient with PAD included in the COPART I registry are in compliance with the French High Authority of Health guidelines concerning antiplatelet drugs and statins. Inhibitors of the renin-angiotensin system seem insufficiently used. However, favorable trends in medical practices between 2004 and 2006 have been observed.


Assuntos
Alta do Paciente , Doenças Vasculares Periféricas/tratamento farmacológico , Antagonistas Adrenérgicos beta/administração & dosagem , Idoso , Idoso de 80 Anos ou mais , Angiotensinas/antagonistas & inibidores , Aspirina/administração & dosagem , Estudos de Coortes , Prescrições de Medicamentos , Feminino , França , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/administração & dosagem , Masculino , Pessoa de Meia-Idade , Inibidores da Agregação Plaquetária/administração & dosagem , Guias de Prática Clínica como Assunto , Piridinas/administração & dosagem , Sistema de Registros , Renina/antagonistas & inibidores , Sistema Renina-Angiotensina , Estudos Retrospectivos
3.
Ann Cardiol Angeiol (Paris) ; 56(2): 74-81, 2007 Apr.
Artigo em Francês | MEDLINE | ID: mdl-17484091

RESUMO

INTRODUCTION: Almost all patients with the most severe peripheral arterial diseases (PAD) patients are hospitalised. This means that the hospital is a particularly good place to observe the characteristics and outcome of PAD patients. It is for this reason that the hospitalised patient registry (COPART I) was created. RESULTS: From June 1st 2004 to May 31st 2005, we included 187 patients surviving at hospital discharge. As expected the majority were men (68.4%). The median age was 72 (+/- 13 years). Almost one third of the PAD of patients suffered from intermittent claudication and two thir (63,6%) from permanent ischemia. A large majority of this latter group had critical limb ischemia. We found a mortality rate of 17.1% at the on year follow-up. These deaths were mainly of cardiovascular origin (9.1%). Almost 2/3 of the deaths had already occurred by six months. One patient in four undergone major or minor amputation during the follow up 2/3 of them involving major amputation. This figure rose to fou patients in ten for critical limb disease. A previous history of both major and minor amputation is strongly related with new amputations (RR = (CI: 1.2-7.5) P = 0.02). After one year of follow-up, almost four patients in ten (42.6%) with permanent ischemia had died, undergone major amputation, or suffered an MI or an IS. CONCLUSION: Peripheral arterial disease remains a severe chronic disease linked to excess mortality of cardiovascular origin. Therefore patients should be given optimal treatment.


Assuntos
Doenças Vasculares Periféricas/mortalidade , Idoso , Amputação Cirúrgica , Estudos de Coortes , Feminino , Seguimentos , França/epidemiologia , Humanos , Claudicação Intermitente/mortalidade , Claudicação Intermitente/terapia , Isquemia/mortalidade , Isquemia/terapia , Perna (Membro)/irrigação sanguínea , Masculino , Doenças Vasculares Periféricas/terapia , Sistema de Registros
4.
Cancer Gene Ther ; 14(1): 19-29, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16990845

RESUMO

Pancreatic cancer is one of the most aggressive and devastating human malignancies. There is an urgent need for more effective therapy for patients with advanced disease. In this context, genetic therapy potentially represents a rational new approach to treating pancreatic cancer, which could provide an adjunct to conventional options. Because of the promise of recombinant SV40 vectors, we tested their ability to deliver a transgene, and to target a transcript, so as to inhibit pancreatic tumors growth in vivo. BxPC3 and Capan-1 cells were efficiently transduced using SV40 vectors without selection, as compared to synthetic vectors PEI. SV40 vectors were as efficient as adenoviral vectors, and provided long-term transgene expression. Next, we devised a SV40-derived, targeted gene therapy approach of pancreatic cancer, by combining hTR tumor-specific promoter with sst2 somatostatin receptor tumor-suppressor gene. In vitro cell proliferation was strongly impaired following administration of SV(hTR-sst2). SV40-derived sst2-mediated antiproliferative effect was dependent on the local production of somatostatin. In vivo, intratumoral gene transfer of sst2 using rSV40 vectors resulted in a marked inhibition of Capan-1 tumor progression, and proliferation. These results represent the initial steps toward a novel approach to the gene therapy of pancreatic cancer using SV40 as a vector.


Assuntos
Vírus Defeituosos/fisiologia , Técnicas de Transferência de Genes , Neoplasias Pancreáticas/patologia , Vírus 40 dos Símios/fisiologia , Replicação Viral , Sequência de Bases , Linhagem Celular Tumoral , Primers do DNA , Humanos , Reação em Cadeia da Polimerase , Transdução Genética
5.
FASEB J ; 15(12): 2300-2, 2001 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-11511520

RESUMO

Somatostatin receptor sst2 is an inhibitory G protein-coupled receptor, which inhibits normal and tumor cell growth by a mechanism involving the tyrosine phosphatase SHP-1. We reported previously that SHP-1 associates transiently with and is activated by sst2 and is a critical component for sst2 growth inhibitory signaling. Here, we demonstrate that in Chinese hamster ovary cells expressing sst2, SHP-1 is associated at the basal level with the neuronal nitric oxide synthase (nNOS). Following sst2 activation by the somatostatin analog RC-160, SHP-1 rapidly recruits nNOS tyrosine dephosphorylates and activates it. The resulting NO activates guanylate cyclase and inhibits cell proliferation. Coexpression of a catalytically inactive SHP-1 mutant with sst2 blocks RC-160-induced nNOS dephosphorylation and activation, as well as guanylate cyclase activation. In mouse pancreatic acini, RC-160 treatment reduces nNOS tyrosine phosphorylation accompanied by an increase of its activity. By opposition, in acini from viable motheaten (mev/mev) mice, which express a markedly inactive SHP-1, RC-160 has no effect on nNOS activity. Finally, expression of a dominant-negative form of nNOS prevents both RC-160-induced p27 up-regulation and cell proliferation inhibition. We therefore identified nNOS as a novel SHP-1 substrate critical for sst2-induced cell-growth arrest.


Assuntos
Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase/fisiologia , Proteínas Tirosina Fosfatases/metabolismo , Receptores de Somatostatina/fisiologia , Transdução de Sinais , Animais , Células CHO , Divisão Celular , Cricetinae , GMP Cíclico/biossíntese , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Modelos Biológicos , Óxido Nítrico/biossíntese , Óxido Nítrico Sintase Tipo I , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/fisiologia , Somatostatina/farmacologia
6.
Chemotherapy ; 47 Suppl 2: 30-9, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11275700

RESUMO

Over the past decade, antiproliferative effects of somatostatin and analogs have been reported in many somatostatin receptor-positive normal and tumor cell types. Regarding the molecular mechanisms involved, somatostatin or analogs mediate their action through both indirect and direct effects. Somatostatin acts through five somatostatin receptors (SSTR1-5) which are variably expressed in normal and tumor cells. These receptors regulate a variety of signal transduction pathways including inhibition of adenylate cyclase, regulation of ion channels, regulation of serine/threonine and tyrosine kinases and phosphatases. This review focuses on recent advances in biological mechanisms involved in the antineoplastic activity of somatostatin and analogs.


Assuntos
Antineoplásicos Hormonais/farmacologia , Divisão Celular/efeitos dos fármacos , Somatostatina/farmacologia , Animais , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Receptores de Somatostatina/metabolismo , Transdução de Sinais , Somatostatina/análogos & derivados
7.
J Biol Chem ; 276(16): 13461-8, 2001 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-11278805

RESUMO

The sst2 somatostatin receptor is an inhibitory G protein-coupled receptor, which exhibits anti-tumor properties. Expression of sst2 is lost in most human pancreatic cancers. We have cloned 2090 base pairs corresponding to the genomic DNA region upstream of the mouse sst2 (msst2) translation initiation codon (ATG). Deletion reporter analyses in mouse pituitary AtT-20 and human pancreatic cancer PANC-1, BxPC-3, and Capan-1 cells identify a region from nucleotide -260 to the ATG codon (325 base pairs) showing maximal activity, and a region between nucleotides -2025 and -260 likely to comprise silencer or transcriptional suppressor elements. In PANC-1 and AtT-20 cells, transforming growth factor (TGF)-beta up-regulates msst2 transcription. Transactivation is mediated by Smad4 and Smad3. The cis-acting region responsible for such regulation is comprised between nucleotides -1115 and -972 and includes Sp1 and CAGA-box sequences. Expression of Smad4 in Smad4-deficient Capan-1 and BxPC-3 cells restores TGF-beta-dependent and -independent msst2 transactivation. Expression of Smad4 in BxPC-3 cells reestablishes both endogenous sst2 expression and somatostatin-mediated inhibition of cell growth. These findings demonstrate that msst2 is a new target gene for TGF-beta transcription regulation and underlie the possibility that loss of Smad4 contributes to the lack of sst2 expression in human pancreatic cancer, which in turn may contribute to a stimulation of tumor growth.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Regiões Promotoras Genéticas , Receptores de Somatostatina/genética , Transativadores/metabolismo , Ativação Transcricional , Fator de Crescimento Transformador beta/farmacologia , Regiões 5' não Traduzidas/genética , Animais , Sequência de Bases , Clonagem Molecular , Códon , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Biblioteca Genômica , Humanos , Luciferases/genética , Camundongos , Dados de Sequência Molecular , Neoplasias Pancreáticas , Neoplasias Hipofisárias , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Deleção de Sequência , Proteína Smad4 , Transcrição Gênica , Ativação Transcricional/efeitos dos fármacos , Células Tumorais Cultivadas
8.
Gastroenterology ; 120(1): 200-9, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11208729

RESUMO

BACKGROUND & AIMS: The somatostatin receptor SST2 mediates the antiproliferative effect of stable somatostatin analogues. SST2 gene expression is lost in most human pancreatic carcinomas. We investigated the mechanisms that could be involved in this defect. METHODS: SST2 gene structure was investigated by sequencing and restriction fragment length polymorphism. Characterization of the polymorphism was performed by electrophoretic mobility shift, cross-linking, and transcription assays. RESULTS: No major deletion of the SST2 coding sequence was found in pancreatic carcinoma specimens, but 2 point mutations were frequently detected in the promoter sequence at positions -83 (A-->G) and -57 (C-->G) from the major transcription initiation site. These mutations were present in pancreatic cancer but also in normal pancreatic tissues or leukocytes and thus correspond to a genetic polymorphism. In the 2 human pancreatic cancer cell lines MiaPaCa-2 and AsPC-1, the naturally occurring mutation -57G had no effect on transcription of SST2 gene, whereas -83G mutation reduced it by 60%-70%. We showed that the -83G mutation creates a specific binding site for the nuclear factor I. Cotransfection experiments showed that the nuclear factor I-A1.1 isoform was responsible for SST2 promoter repression. CONCLUSIONS: The -83G polymorphism identified on human SST2 gene promoter is responsible for the specific fixation of nuclear factor I and repression of SST2 transcription in human pancreatic cancer cells. However, its contribution to pancreatic tumorigenesis remains unknown.


Assuntos
Proteínas de Ligação a DNA , Neoplasias Pancreáticas , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas/genética , Receptores de Somatostatina/genética , Fatores de Transcrição , Transcrição Gênica/fisiologia , Regiões 5' não Traduzidas/genética , Proteínas Estimuladoras de Ligação a CCAAT/genética , Proteínas Estimuladoras de Ligação a CCAAT/metabolismo , Deleção de Genes , Regulação Neoplásica da Expressão Gênica , Humanos , Técnicas In Vitro , Mutação/genética , Fatores de Transcrição NFI , Proteínas Nucleares , Pâncreas/fisiologia , RNA Mensageiro/genética , Células Tumorais Cultivadas , Proteína 1 de Ligação a Y-Box
9.
Proc Natl Acad Sci U S A ; 97(16): 9180-5, 2000 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-10900262

RESUMO

The sst2 somatostatin receptor mediates the antiproliferative effects of somatostatin analogs. The present study demonstrates that stable expression of sst2 in the hamster pancreatic cancer cells PC-1 and PC-1.0 activates an autocrine negative loop leading to an in vitro inhibition of cell proliferation. In vivo studies conducted in Syrian golden hamsters after orthotopic implantation of PC-1.0 cells showed that both tumor growth and metastatic progression of allografts containing 100% of sst2-expressing cells were significantly inhibited for up to 20 days after implantation, as compared with control allografts that did not express sst2. A local antitumor bystander effect was observed after induction of mixed tumors containing a 1:3 ratio of sst2-expressing cells to control cells. Tumor volume and incidence of metastases of mixed tumors were significantly reduced at day 13 post implantation. This effect decreased with time as at day 20, growth of mixed tumors was similar to that of control tumors. After administration of the cytotoxic somatostatin conjugate AN-238 on day 13, antitumor bystander effect observed in mixed tumors was significantly extended to day 20. We also observed that in vitro invasiveness of sst2-expressing PC-1.0 cells was significantly reduced. Tyrosine dephosphorylation of E-cadherin may participate in restoring the E-cadherin function, reducing in turn pancreatic cancer cell motility and invasiveness. This dephosphorylation depends on the tyrosine phosphatase src homology 2-containing tyrosine phosphatase 1 (SHP-1) positively coupled to sst2 receptor. The inhibitory effect of sst2 gene expression on pancreatic cancer growth and invasion combined with chemotherapy with targeted cytotoxic somatostatin analog administration provides a rationale for a therapeutic approach to gene therapy based on in vivo sst2 gene transfer.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Citotoxinas/administração & dosagem , Proteínas Fúngicas/fisiologia , Proteínas Ativadoras de GTPase , Neoplasias Pancreáticas/patologia , Proteínas de Saccharomyces cerevisiae , Animais , Caderinas/metabolismo , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Divisão Celular/fisiologia , Cricetinae , Doxorrubicina/administração & dosagem , Doxorrubicina/análogos & derivados , Proteínas Fúngicas/genética , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Mesocricetus , Metástase Neoplásica/prevenção & controle , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fosforilação , Proteína Fosfatase 1 , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/metabolismo , Pirróis/administração & dosagem , Células Tumorais Cultivadas , Tirosina/metabolismo
10.
J Biol Chem ; 275(23): 17321-7, 2000 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-10748160

RESUMO

Among the most conserved regions in the G-protein-coupled receptors is the (N/D)PX(2-3)Y motif of the seventh transmembrane domain (X represents any amino acid). The mutation of the Asn/Asp residue of this motif in different G-protein-coupled receptors was shown to affect the activation of either adenylyl cyclase or phospholipase C. We have mutated the Asn residue (Asn-391) of the NPXXY motif in the CCKBR to Ala and determined the effects of the mutation on binding, signaling, and G-proteins coupling after expression of the mutated receptor in COS cells. The mutated receptor displayed similar expression levels and high affinity CCK binding compared with the wild type CCKBR. However, unlike the wild type CCKBR, the mutated receptor was completely unable to mediate activation of either phospholipase C and protein kinase C-dependent and -independent mitogen-activated protein kinase pathways, indicating an essential role of Asn-391 in CCKBR signaling. Coimmunoprecipitation experiments allowed us to show that the inactive mutant retains an intact capacity to form stable complexes with G(q)alpha subunits in response to CCK. These results indicate that the formation of high affinity CCK-receptor-G(q) protein complexes is not sufficient to activate G(q) and suggest that Asn-391 is specifically involved in G(q) proteins activation.


Assuntos
Asparagina , Proteínas de Ligação ao GTP/metabolismo , Receptores da Colecistocinina/química , Receptores da Colecistocinina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Ácido Aspártico , Sítios de Ligação , Células COS , Colecistocinina/farmacologia , Sequência Conservada , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP , Guanosina 5'-O-(3-Tiotrifosfato)/farmacologia , Fosfatos de Inositol/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mutagênese Sítio-Dirigida , Fragmentos de Peptídeos/farmacologia , Proteína Quinase C/metabolismo , Ratos , Receptor de Colecistocinina B , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Transfecção , Fosfolipases Tipo C/metabolismo
11.
Int J Cancer ; 85(4): 555-62, 2000 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-10699930

RESUMO

Pancreatic tumors overexpress FGF-2 and t-PA, but the implication of the growth factor in t-PA synthesis and t-PA-dependent tumor invasion remains unknown. FGF-2 is present in different isoforms: The 18 kDa FGF-2 is secreted, while the 22.5 kDa one is nuclearized and exerts intracrine regulations bypassing cell-surface FGF receptors. Rat pancreatic carcinoma AR4-2J cells producing either the 18 or the 22.5 kDa FGF-2 after transfection with FGF-2 cDNAs have been used to analyze the role of FGF-2 in t-PA expression and t-PA-related cell spreading. The 22.5 kDa FGF-2 reduced t-PA and PAI-1 synthesis 2-fold. Addition of recombinant 18 kDa FGF-2 (rFGF-2) to cell cultures resulted in increased t-PA and decreased PAI-1 expression. By contrast, rFGF-2 did not significantly modify t-PA synthesis in cells producing the 22.5 kDa FGF-2. Cell spreading was t-PA-dependent. Furthermore, cells producing the 22.5 kDa FGF-2 migrated less than control cells and cells producing the 18 kDa FGF-2. Overall, our data show that secretory FGF-2 is involved in t-PA synthesis by pancreatic cancer cells and facilitates cell spreading. The 22.5 kDa FGF-2 exerts opposite effects by decreasing t-PA expression in basal conditions and during rFGF-2 stimulation. Since the expression of the 22.5 kDa FGF-2 is under specific controls, its up-regulation might have the potential to reduce spreading of pancreatic cancer cells.


Assuntos
Fator 2 de Crescimento de Fibroblastos/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias Pancreáticas/patologia , Inibidor 1 de Ativador de Plasminogênio/genética , Ativador de Plasminogênio Tecidual/genética , Animais , Divisão Celular/efeitos dos fármacos , Colágeno , Doxiciclina/toxicidade , Combinação de Medicamentos , Matriz Extracelular , Fator 2 de Crescimento de Fibroblastos/genética , Fator 2 de Crescimento de Fibroblastos/farmacologia , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Cinética , Laminina , Camundongos , Camundongos Nus , Peso Molecular , Mutagênese Sítio-Dirigida , Invasividade Neoplásica , Metástase Neoplásica , Neoplasias Pancreáticas/genética , Mutação Puntual , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Proteoglicanas , Ratos , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/farmacologia , Transcrição Gênica , Transfecção , Transplante Heterólogo , Células Tumorais Cultivadas , Ativador de Plasminogênio Tipo Uroquinase/genética
12.
Protein Sci ; 8(11): 2347-54, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10595537

RESUMO

The knowledge of the binding sites of G protein-coupled cholecystokinin receptors represents important insights that may serve to understand their activation processes and to design or optimize ligands. Our aim was to identify the amino acid of the cholecystokinin-A receptor (CCK-AR) binding site in an interaction with the sulfate of CCK, which is crucial for CCK binding and activity. A three-dimensional model of the [CCK-AR-CCK] complex was built. In this model, Arg197 was the best candidate residue for a ionic interaction with the sulfate of CCK. Arg197 was exchanged for a methionine by site-directed mutagenesis. Wild-type and mutated CCK-AR were transiently expressed in COS-7 cells for pharmacological and functional analysis. The mutated receptor on Arg197 did not bind the agonist radioligand 125I-BH-[Thr, Nle]-CCK-9; however, it bound the nonpeptide antagonist [3H]-SR27,897 as the wild-type receptor. The mutant was approximately 1,470- and 3,200-fold less potent than the wild-type CCK-AR to activate G proteins and to induce inositol phosphate production, respectively. This is consistent with the 500-fold lower potency and 800-fold lower affinity of nonsulfated CCK relative to sulfated CCK on the wild-type receptor. These data, together with those showing that the mutated receptor failed to discriminate nonsulfated and sulfated CCK while it retained other pharmacological features of the CCK-AR, strongly support an interaction between Arg197 of the CCK-AR binding site and the sulfate of CCK. In addition, the mutated CCK-AR resembled the low affinity state of the wild-type CCK-AR, suggesting that Arg197-sulfate interaction regulates conformational changes of the CCK-AR that are required for its physiological activation.


Assuntos
Arginina , Colecistocinina/química , Colecistocinina/metabolismo , Receptores da Colecistocinina/química , Receptores da Colecistocinina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sítios de Ligação , Células COS , Simulação por Computador , Humanos , Ácidos Indolacéticos/farmacocinética , Fosfatos de Inositol/metabolismo , Radioisótopos do Iodo , Cinética , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação Proteica , Ensaio Radioligante , Receptor de Colecistocinina A , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Tiazóis/farmacocinética , Transfecção , Trítio
13.
FASEB J ; 13(14): 2037-50, 1999 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-10544187

RESUMO

In CHO cells we had found that CCK positively regulated cell proliferation via the activation of a soluble guanylate cyclase. Here we demonstrate that CCK stimulated a nitric oxide synthase (NOS) activity. The production of NO was involved in the proliferative response elicited by CCK regarding the inhibitory effect of NOS inhibitors L-NAME and alpha-guanidinoglutaric acid. We identified the NOS activated by the peptide as the neuronal isoform: the expression of the C415A neuronal NOS mutant inhibited both CCK-induced stimulation of NOS activity and cell proliferation. These two effects were also inhibited after expression of the C459S tyrosine phosphatase SHP-2 mutant and the betaARK1 (495-689) sequestrant peptide, indicating the requirement of activated SHP-2 and G-betagamma subunit. Kinetic analysis (Western blot after coimmunoprecipitation and specific SHP-2 activity) revealed that in response to CCK-treatment, SHP-2 associated to G-beta1 subunit, became activated, and then dephosphorylated the neuronal NOS through a direct association. These data demonstrate that the neuronal NOS is implicated in proliferative effect evoked by CCK. A novel growth signaling pathway is described, involving the activation of neuronal NOS by dephosphorylation of tyrosyl residues.


Assuntos
Proteínas de Ligação ao GTP/fisiologia , Óxido Nítrico Sintase/metabolismo , Proteínas Tirosina Fosfatases/fisiologia , Animais , Células CHO , Divisão Celular/efeitos dos fármacos , Cricetinae , Dimerização , Ativação Enzimática , Peptídeos e Proteínas de Sinalização Intracelular , Óxido Nítrico/fisiologia , Óxido Nítrico Sintase Tipo I , Nitroprussiato/farmacologia , Fosforilação , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Receptores da Colecistocinina/fisiologia , Sincalida/farmacologia , Tirosina/metabolismo
14.
Diabetes ; 48(10): 2015-21, 1999 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-10512367

RESUMO

Gastrin (G) and cholecystokinin (CCK) are gastrointestinal neuropeptides that are released into circulation during a meal. G is also transiently expressed during embryogenic and early ontogenic development of the pancreas and is believed to act on islet-cell development. Both peptides act on pancreatic endocrine function; however, the effects are dependent on the species and on cellular and molecular underlying mechanisms that remain poorly characterized. Since CCK-B/G subtype receptor is predominant over the CCK-A subtype in the human pancreas, we hypothesized that it could be expressed by islet cells. Here we present reverse transcription-polymerase chain reaction and immunohistochemistry data demonstrating that the CCK-B/G receptor is expressed in islet cells and that islet glucagon-producing cells are the major site of CCK-B/G receptor expression in adult and fetal pancreas. Moreover, G immunoreactivity was detected in the fetal human pancreas at embryogenic week 22. G- and CCK-stimulated glucagon are released from purified human islets. Concentration of CCK and G eliciting a half-maximal level of glucagon secretion were 13 +/- 6 and 8 +/- 5 pmol/l, respectively. Maximal glucagon secretion was achieved in the presence of 30 pmol/l peptides and was similar to that obtained in the presence of 10 mmol/l L-arginine (1.6 pmol x ml(-1) x 90 min(-1)). The nonpeptide antagonist of the CCK-B/G receptor, RPR-101048, fully inhibited CCK- and G-stimulated glucagon secretion at 100 nmol/l concentration. These data are consistent with the view that the CCK-B/G receptor is involved in glucose homeostasis in adult humans and mediates the autocrine effects of G on islet differentiation and growth in the fetal pancreas.


Assuntos
Pâncreas/fisiologia , Receptores da Colecistocinina/fisiologia , Adulto , Células Cultivadas , Colecistocinina/metabolismo , Clonagem Molecular , Gastrinas/metabolismo , Regulação da Expressão Gênica , Glucagon/metabolismo , Humanos , Pâncreas/embriologia , RNA Mensageiro/metabolismo , Receptor de Colecistocinina B , Receptores da Colecistocinina/genética
15.
J Biol Chem ; 274(33): 23191-7, 1999 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-10438490

RESUMO

Recently, we reported that the mutation of His(207) to Phe located in the second extracellular loop of the cholecystokinin B receptor strongly affected cholecystokinin (CCK) binding (Silvente-Poirot, S., Escrieut, C., and Wank, S. A. (1998) Mol. Pharmacol. 54, 364-371). To characterize the functional group in CCK that interacts with His(207), we first substituted His(207) to Ala. This mutation decreased the affinity and the potency of CCK to produce total inositol phosphates 302-fold and 456-fold without affecting the expression of the mutant receptor. The screening of L-alanine-modified CCK peptides to bind and activate the wild type and mutant receptors allowed the identification of the interaction of the C-terminal Asp(8) of CCK with His(207). The H207A-CCKBR mutant, unlike the wild type receptor, was insensitive to substitution of Asp(8) of CCK to other amino acid residues. This interaction was further confirmed by mutating His(207) to Asp. The affinity of CCK for the H207D-CCKBR mutant was 100-fold lower than for the H207A-CCKBR mutant, consistent with an electrostatic repulsion between the negative charges of the two interacting aspartic acids. Peptides with neutral amino acids in position eight of CCK reversed this effect and displayed a gain of affinity for the H207D mutant compared with CCK. To date, this is the first report concerning the identification of a direct contact point between the CCKB receptor and CCK.


Assuntos
Ácido Aspártico/metabolismo , Colecistocinina/metabolismo , Histidina/metabolismo , Receptores da Colecistocinina/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Colecistocinina/química , Mutagênese Sítio-Dirigida , Receptor de Colecistocinina B , Receptores da Colecistocinina/química , Receptores da Colecistocinina/genética , Relação Estrutura-Atividade
16.
J Biol Chem ; 274(29): 20457-64, 1999 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-10400673

RESUMO

The cholecystokinin-A receptor (CCK-AR) is a G protein-coupled receptor that mediates important central and peripheral cholecystokinin actions. Residues of the CCK-AR binding site that interact with the C-terminal part of CCK that is endowed with biological activity are still unknown. Here we report on the identification of Arg-336 and Asn-333 of CCK-AR, which interact with the Asp-8 carboxylate and the C-terminal amide of CCK-9, respectively. Identification of the two amino acids was achieved by dynamics-based docking of CCK in a refined three-dimensional model of CCK-AR using, as constraints, previous results that demonstrated that Trp-39/Gln-40 and Met-195/Arg-197 interact with the N terminus and the sulfated tyrosine of CCK, respectively. Arg-336-Asp-8 and Asn-333-amide interactions were pharmacologically assessed by mutational exchange of Arg-336 and Asn-333 in the receptor or reciprocal elimination of the partner chemical functions in CCK. This study also allowed us to demonstrate that (i) the identified interactions are crucial for stabilizing the high affinity phospholipase C-coupled state of the CCK-AR.CCK complex, (ii) Arg-336 and Asn-333 are directly involved in interactions with nonpeptide antagonists SR-27,897 and L-364,718, and (iii) Arg-336 but not Asn-333 is directly involved in the binding of the peptide antagonist JMV 179 and the peptide partial agonist JMV 180. These data will be used to obtain an integrated dynamic view of the molecular processes that link agonist binding to receptor activation.


Assuntos
Arginina/metabolismo , Asparagina/metabolismo , Ácido Aspártico/metabolismo , Colecistocinina/metabolismo , Receptores da Colecistocinina/metabolismo , Amidas/metabolismo , Animais , Sítios de Ligação , Células COS , Colecistocinina/química , Simulação por Computador , Humanos , Modelos Moleculares , Mutagênese Sítio-Dirigida , Conformação Proteica , Receptor de Colecistocinina A , Receptores da Colecistocinina/química , Receptores da Colecistocinina/genética , Fosfolipases Tipo C/metabolismo
17.
J Biol Chem ; 274(29): 20657-63, 1999 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-10400698

RESUMO

We have analyzed in Chinese hamster ovary cells the upstream mediators by which the G protein-coupled receptor, gastrin/CCKB, activates the extracellular-regulated kinases (ERKs) and p85/p110-phosphatidylinositol 3-kinase (PI 3-kinase) pathways. Overexpression of an inhibitory mutant of Shc completely blocked gastrin-stimulated Shc.Grb2 complex formation but partially inhibited ERK-1 activation by this peptide. Expression of Csk, which inactivates Src-family kinases, totally inhibited gastrin-induced Src-like activity detected in anti-Src and anti-Shc precipitates but diminished by 50% Shc phosphorylation and ERK-1 activation. We observed a rapid tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1) and an increase in Src-like kinase activity in anti-IRS-1 immunoprecipitates from gastrin-stimulated cells, suggesting that IRS-1 may be a direct substrate of Src. This hypothesis was supported by the inhibition of gastrin-induced Src. IRS-1 complex formation and IRS-1 phosphorylation in Csk-transfected cells. In addition, the increase in PI 3-kinase activity measured in anti-p85 or anti-IRS-1 precipitates following gastrin stimulation was abolished by Csk. Our results demonstrate the existence of two mechanisms in gastrin-mediated ERKs activation. One requires Shc phosphorylation by Src-family kinases, and the other one is independent of these two proteins. They also indicate that tyrosine phosphorylation of IRS-1 by Src-family kinases could lead to the recruitment and the activation of the p85/p110-PI 3-kinase in response to gastrin.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , Proteínas Adaptadoras de Transporte Vesicular , Proteínas Quinases Dependentes de Cálcio-Calmodulina/metabolismo , Proteínas Quinases Ativadas por Mitógeno , Fosfatidilinositol 3-Quinases/metabolismo , Receptores da Colecistocinina/metabolismo , Quinases da Família src/metabolismo , Animais , Células CHO , Cricetinae , Replicação do DNA , Ativação Enzimática , Proteína Adaptadora GRB2 , Gastrinas/metabolismo , Humanos , Proteínas Substratos do Receptor de Insulina , Proteína Quinase 3 Ativada por Mitógeno , Fosfoproteínas/metabolismo , Fosforilação , Proteínas/metabolismo , Receptor de Colecistocinina B , Proteínas Adaptadoras da Sinalização Shc , Transdução de Sinais , Proteína 1 de Transformação que Contém Domínio 2 de Homologia de Src , Tirosina/metabolismo
18.
Hum Gene Ther ; 10(6): 995-1008, 1999 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-10223733

RESUMO

Human pancreatic adenocarcinomas lose the ability to express sst2, the somatostatin receptor, which mediates the antiproliferative effect of somatostatin. Reintroducing sst2 into human pancreatic cancer cells by stable expression evokes an autocrine negative feedback loop leading to a constitutive activation of the sst2 gene and an inhibition of cell proliferation and tumorigenicity. In vivo studies have been conducted in athymic mice to investigate the antitumor bystander effects resulting from the transfer of the sst2 gene into human pancreatic cancer cell line BxPC-3. In mixing experiments, a local bystander effect was observed: mixed tumors containing a ratio of sst2-expressing cells to control cells of 25:75, 50:50, and 75:25 grew with a time delay of 31, 44, and 50 days, respectively, when compared with control tumors derived from control cells. Tumors containing 100% sst2-expressing cells remained quiescent for up to 80 days. A significant increase in apoptosis and a decrease in the Ki67 index were detected in mixed and sst2 tumor when compared with control tumors. In combined experiments, mice were separately xenografted with control cells on one flank and with sst2-expressing cells on the other flank. A distant antitumor effect was induced: growth of control tumors was delayed by 33 days, the Ki67 index decreased significantly, and apoptosis increased when compared with control tumors that grew alone. The distant bystander effect may be explained in part by a significant increase in serum somatostatin-like immunoreactivity levels resulting from the autocrine feedback loop produced by sst2-expressing cells and inducing an upregulation of the type 1 somatostatin receptor, sst1, which also mediates the antiproliferative effect of somatostatin. In conclusion, the local and distant antitumor bystander effects obtained in this experimental model suggest that sst2 gene transfer may represent a new therapy for pancreatic cancer.


Assuntos
Adenocarcinoma/genética , Técnicas de Transferência de Genes , Terapia Genética , Neoplasias Pancreáticas/terapia , Receptores de Somatostatina/genética , Adenocarcinoma/imunologia , Adenocarcinoma/patologia , Animais , Apoptose , Feminino , Humanos , Marcação In Situ das Extremidades Cortadas , Antígeno Ki-67/imunologia , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , RNA Mensageiro/genética , Receptores de Somatostatina/metabolismo , Regulação para Cima
19.
J Biol Chem ; 274(21): 15186-93, 1999 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-10329727

RESUMO

Activation of the somatostatin receptor sst2 inhibits cell proliferation by a mechanism involving the stimulation of the protein-tyrosine phosphatase SHP-1. The cell cycle regulatory events leading to sst2-mediated growth arrest are not known. Here, we report that treatment of Chinese hamster ovary cells expressing sst2 with the somatostatin analogue, RC-160, led to G1 cell cycle arrest and inhibition of insulin-induced S-phase entry through induction of the cyclin-dependent kinase inhibitor p27(Kip1). Consequently, a decrease of p27(Kip1)-cdk2 association, an inhibition of insulin-induced cyclin E-cdk2 kinase activity, and an accumulation of hypophosphorylated retinoblastoma gene product (Rb) were observed. However, RC-160 had no effect on the p21(Waf1/Cip1). When sst2 was coexpressed with a catalytically inactive mutant SHP-1 in Chinese hamster ovary cells, mutant SHP-1 induced entry into cell cycle and down-regulation of p27(Kip1) and prevented modulation by insulin and RC-160 of p27(Kip1) expression, p27(Kip1)-cdk2 association, cyclin E-cdk2 kinase activity, and the phosphorylation state of Rb. In mouse pancreatic acini, RC-160 reverted down-regulation of p27(Kip1) induced by a mitogen, and this effect did not occur in acini from viable motheaten (mev/mev) mice expressing a mutant SHP-1 with markedly deficient enzymes. These findings provide the first evidence that sst2 induces cell cycle arrest through the up-regulation of p27(Kip1) and demonstrate that SHP-1 is required for maintaining high inhibitory levels of p27(Kip1) and is a critical target of the insulin, and somatostatin signaling cascade, leading to the modulation of p27(Kip1).


Assuntos
Quinases relacionadas a CDC2 e CDC28 , Proteínas de Ciclo Celular , Inibidores Enzimáticos , Fase G1/fisiologia , Proteínas Associadas aos Microtúbulos/fisiologia , Proteínas Tirosina Fosfatases/fisiologia , Receptores de Somatostatina/fisiologia , Fase S/fisiologia , Proteínas Supressoras de Tumor , Domínios de Homologia de src , Animais , Células CHO , Células Cultivadas , Cricetinae , Quinase 2 Dependente de Ciclina , Inibidor de Quinase Dependente de Ciclina p27 , Quinases Ciclina-Dependentes/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Fosforilação/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteína do Retinoblastoma/metabolismo , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Somatostatina/análogos & derivados , Somatostatina/farmacologia
20.
FEBS Lett ; 445(2-3): 251-5, 1999 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-10094466

RESUMO

The molecular events whereby gastrin occupancy of G/CCK(B) receptors leads to phosphatidylinositol (PI) 3-kinase activation have been examined. We report here that this peptide promotes the association between two non-receptor tyrosine kinases, p60Src and p125FAK, and elicits a parallel increase in tyrosine phosphorylation and activity of both kinases. Gastrin-induced PI 3-kinase activity was coprecipitated with p60Src and p125FAK and was inhibited by herbimycin A, the selective Src inhibitor PP-2 or cytochalasin D, which disrupts the actin cytoskeleton and prevents p125FAK activity. These results indicate, for the first time, that a p60Src/p125FAK complex acts upstream of the gastrin-stimulated PI 3-kinase pathway.


Assuntos
Moléculas de Adesão Celular/metabolismo , Gastrinas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Transdução de Sinais , Animais , Ativação Enzimática , Quinase 1 de Adesão Focal , Proteína-Tirosina Quinases de Adesão Focal , Gastrinas/farmacologia , Fosforilação , Testes de Precipitina , Ratos , Células Tumorais Cultivadas , Tirosina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA