Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
J Gynecol Obstet Biol Reprod (Paris) ; 38(7): 595-8, 2009 Nov.
Article in French | MEDLINE | ID: mdl-19540080

ABSTRACT

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.


Subject(s)
Fetal Death/etiology , Pregnancy Complications, Cardiovascular , Takayasu Arteritis/complications , Adult , Female , Fetal Growth Retardation/diagnosis , Humans , Pre-Eclampsia/diagnosis , Pregnancy
2.
J Mal Vasc ; 33(1): 1-11, 2008 Feb.
Article in French | MEDLINE | ID: mdl-18187280

ABSTRACT

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.


Subject(s)
Patient Discharge , Peripheral Vascular Diseases/drug therapy , Adrenergic beta-Antagonists/administration & dosage , Aged , Aged, 80 and over , Angiotensins/antagonists & inhibitors , Aspirin/administration & dosage , Cohort Studies , Drug Prescriptions , Female , France , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Male , Middle Aged , Platelet Aggregation Inhibitors/administration & dosage , Practice Guidelines as Topic , Pyridines/administration & dosage , Registries , Renin/antagonists & inhibitors , Renin-Angiotensin System , Retrospective Studies
3.
J Clin Invest ; 97(8): 1874-83, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8621771

ABSTRACT

The somatostatin receptor subtype sst2 mediates both activation of a tyrosine phosphatase activity and inhibition of cell proliferation induced by somatostatin analogues. In the absence of exogenous ligand, expression of sst2 in NIH 3T3 cells resulted in inhibition of cell growth. Polymerase chain reaction coupled to reverse transcription demonstrated that expression of sst2 in NIH 3T3 cells stimulated the expression of preprosomatostatin mRNA accompanied by a production of immunoreactive somatostatin-like peptide which corresponded predominantly to somatostatin 14. Moreover anti-somatostatin antibodies suppressed sst2-promoted inhibition of cell proliferation. Inhibition of cell proliferation associated with increased secretion of somatostatin-like immunoreactivity was also observed after expression of sst2 in human pancreatic tumor cells BxPC3 devoid of endogenous receptors. In addition, expression of sst2 in NIH 3T3 cells was associated with constitutive activation of tyrosine phosphatase PTP1C that resulted from enhanced expression of the protein. Blocking of PTP1C tyrosine phosphatase activity with orthovanadate or that of PTP1C protein with antisense PTP1C oligonucleotides decreased the sst2-induced inhibition of cell proliferation. These results, taken together, show that expression of sst2 in NIH 3T3 cells generated a negative autocrine loop by stimulating sst2 ligand production and amplifying PTP1C sst2-transducer. Sst2/ligand may function as a determinant factor involved in the negative growth control of cells.


Subject(s)
Gene Expression , Protein Precursors/biosynthesis , Receptors, Somatostatin/biosynthesis , Somatostatin/biosynthesis , 3T3 Cells , Animals , Base Sequence , Cell Line , Chromatography, Gel , DNA Primers , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Mice , Molecular Sequence Data , Oligonucleotides, Antisense/pharmacology , Pancreatic Neoplasms , Polymerase Chain Reaction , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/metabolism , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Receptors, Somatostatin/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/metabolism , Signal Transduction , Somatostatin/analysis , Time Factors , Tumor Cells, Cultured , Vanadates/pharmacology
4.
Cancer Gene Ther ; 14(1): 19-29, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16990845

ABSTRACT

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.


Subject(s)
Defective Viruses/physiology , Gene Transfer Techniques , Pancreatic Neoplasms/pathology , Simian virus 40/physiology , Virus Replication , Base Sequence , Cell Line, Tumor , DNA Primers , Humans , Polymerase Chain Reaction , Transduction, Genetic
5.
Ann Cardiol Angeiol (Paris) ; 56(2): 74-81, 2007 Apr.
Article in French | MEDLINE | ID: mdl-17484091

ABSTRACT

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.


Subject(s)
Peripheral Vascular Diseases/mortality , Aged , Amputation, Surgical , Cohort Studies , Female , Follow-Up Studies , France/epidemiology , Humans , Intermittent Claudication/mortality , Intermittent Claudication/therapy , Ischemia/mortality , Ischemia/therapy , Leg/blood supply , Male , Peripheral Vascular Diseases/therapy , Registries
6.
Cancer Res ; 51(14): 3633-8, 1991 Jul 15.
Article in English | MEDLINE | ID: mdl-2065320

ABSTRACT

Capan 1, a human pancreatic cancer cell line, is routinely grown in 10% fetal calf serum (FCS). In order to characterize the factors needed for its proliferation, FCS was replaced by a synthetic serum (Ultroser G). For Capan 1 proliferation we found that Ultroser G was as efficient as FCS. A subfraction of Ultroser G containing insulin, transferrin, and lipids was found to be responsible for that response since a combination of these compounds reproduced the growth observed with 10% FCS. Lipids stimulated cell proliferation even in the absence of other factors. Other human (MIA PaCa 2, AsPC1, Panc 1) or rat (AR4-2J) pancreatic cancer cell lines tested proliferated in the reconstituted medium containing insulin (100 ng/ml), transferrin (100 micrograms/ml), fatty acid-free albumin (1 mg/ml), and bovine serum lipids (0.7%), as in 10% FCS. Furthermore, the growth of nonpancreatic cell lines (HT29, A431, CREF) was not enhanced by lipids. Lipoproteins were found to be involved in the mitogenic response of pancreatic cells to lipids, whereas phosphatidylcholine and fatty acids were either inefficient or inhibitors (MIA PaCa2 and AR4-2J). Alkaline phosphatase and amylase content, differentiation markers for Capan 1 and AR4-2J cells, respectively, were not modified by the reconstituted medium. These data suggest that lipids, insulin, and transferrin are the essential factors for the proliferation of pancreatic cancer cell lines, reproducing the growth effect of 10% FCS. Moreover, in the absence of most of the seric growth factors, pancreatic cells remained differentiated.


Subject(s)
Lipids/pharmacology , Pancreatic Neoplasms/pathology , Blood Substitutes/pharmacology , Cell Division/drug effects , Culture Media , Fetal Blood/physiology , Humans , Organic Chemicals , Serum Albumin/pharmacology , Tumor Cells, Cultured
7.
Cancer Res ; 50(18): 5829-33, 1990 Sep 15.
Article in English | MEDLINE | ID: mdl-2393852

ABSTRACT

Many reports emphasized the role of gastrin as growth factor on normal gastrointestinal mucosa and pancreas. In the present study, we analyzed the proliferative effects of cholecystokinin (CCK) and gastrin peptides on a rat tumoral pancreatic cell line, AR42J, which possesses both CCKA and CCKB receptor subtypes. The results showed a good correlation between the binding of gastrin to CCKB receptor [Kd 1.125 +/- 0.3 (SD) nM] and its ability to either induce ornithine decarboxylase activity [50% effective concentration, 0.6 +/- 0.3 nM] and [3H]-thymidine incorporation [50% effective concentration, 2 +/- 0.4 nM]. Furthermore, the ability of different cholecystokinin and gastrin antagonists such as proglumide and asperlicin derivatives (respectively, CR1409, CR1505, and L364,718) were tested. We found that all antagonists displaced 125I-labeled gastrin binding, with the following order of potencies: L364,718 greater than CR1409 greater than CR1505 greater than proglumide. Furthermore, the 50% inhibitory concentration of CR1409 and CR1505 to inhibit gastrin stimulated ornithine decarboxylase activity (an early event involved in cell proliferation) and [3H]thymidine incorporation were in agreement with their constants of inhibition (Ki) on gastrin binding. The L364,718 compound, at a concentration which fully occupied the CCKA without affecting the CCKB, had no effect on gastrin stimulated ornithine decarboxylase activity and [3H]thymidine incorporation. In addition, this compound appeared to be a full agonist on CCKB receptor. These results confirm the implication of the CCKB receptor in the proliferative response of AR42J cells to gastrin.


Subject(s)
DNA, Neoplasm/biosynthesis , Gastrins/pharmacology , Glutamine/analogs & derivatives , Pancreatic Neoplasms/metabolism , Proglumide/analogs & derivatives , Animals , Ornithine Decarboxylase/analysis , Pancreatic Neoplasms/pathology , Proglumide/pharmacology , Rats , Receptors, Cholecystokinin/analysis , Receptors, Cholecystokinin/physiology , Thymidine/metabolism , Tumor Cells, Cultured
8.
Cancer Res ; 56(8): 1742-5, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620486

ABSTRACT

The ornithine decarboxylase enzyme (ODC) is the key regulator of polyamine synthesis and is a member of the cellular proto-oncogene family. Its expression becomes constitutively activated by carcinogens, viruses, and oncogenes. ODC mRNA has a long 5' untranslated region that could be important in the regulation of enzyme levels by affecting translation. To test this hypothesis, we have determined the role of this region on the constitutive ODC hyperexpression measured in AR4-2J cells, an azaserine-induced, tumor-derived pancreatic acinar cell line. Construction of expression vectors in which ODC 5' leader sequence was placed flanking the chloramphenicol acetyltransferase reporter gene allowed us to identify three AR4-2J specific, different alternatively spliced ODC 5' leaders. The 5' ends of exons 2 and 3 were lengthened by 17 and 13 bases, respectively. Translation performed in a cell-free system as well as in COS7 transient transfection experiments demonstrated that AR4-2J isoforms induce a strong increase in the rate of translation. These results provide evidence that alternative splicing observed in tumoral cells, coupled with translation regulation, relieves the translation repression mediated by the long and structured 5' untranslated region of the ODC proto-oncogene.


Subject(s)
Alternative Splicing , Ornithine Decarboxylase/biosynthesis , Protein Biosynthesis , RNA, Messenger/metabolism , Animals , Base Sequence , Blotting, Western , Cell Line , Chloramphenicol O-Acetyltransferase/analysis , Chloramphenicol O-Acetyltransferase/biosynthesis , Chlorocebus aethiops , DNA Primers , Enzyme Repression , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Molecular Sequence Data , Pancreas , Pancreatic Neoplasms , RNA, Messenger/biosynthesis , Rats , Recombinant Proteins/analysis , Recombinant Proteins/biosynthesis , Restriction Mapping , Transfection , Tumor Cells, Cultured
9.
Cancer Res ; 53(5): 1182-7, 1993 Mar 01.
Article in English | MEDLINE | ID: mdl-7679948

ABSTRACT

The role of the different basic fibroblast growth factor (bFGF) forms on the regulation of pancreatic acinar cancer cells was analyzed on the rat cell line AR4-2J. This cell line expresses bFGF receptors but does not produce bFGF. AR4-2J cells were retrovirally transfected with the wild type or with point-mutated bFGF complementary DNAs in order to obtain the expression of all the bFGF forms (clone A4), or of that of the M(r) 22,500 form (clone A3), or of that of the M(r) 18,000 form (clone A5). Each clone was less tumorigenic in nude mice than AR4-2J cells. In culture, only the coexpression of all the bFGF forms modified cell morphology (fibroblast-like) and secretory enzyme synthesis (about a 20-fold decrease of amylase and lipase). Cells expressing the high molecular weight bFGF (A3 and A4) were able to grow in serum-free medium. As for AR4-2J, exogenously added bFGF still exerted mitogenic effects on the bFGF-producing cells. These results suggest that pancreatic acinar cancer cells may respond to endogenous bFGF; furthermore, they seem very sensitive to the coexpression of the different bFGF forms which is often described in cancer cells.


Subject(s)
Fibroblast Growth Factor 2/physiology , Pancreatic Neoplasms/pathology , Amylases/analysis , Amylases/biosynthesis , Amylases/genetics , Animals , Dexamethasone/pharmacology , Fibroblast Growth Factor 2/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/metabolism , Rats , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
10.
Cancer Res ; 56(8): 1823-7, 1996 Apr 15.
Article in English | MEDLINE | ID: mdl-8620499

ABSTRACT

Five somatostatin receptor subtypes (sst1 to sst5) have been cloned. We demonstrated previously that sst2 and sst5 mediate the antiproliferative effect of the somatostatin analogues octreotide and vapreotide. Using reverse transcription-PCR, we investigated gene expression of the five receptors in 47 human normal and cancerous tissues or cell lines from pancreatic and colorectal origin. mRNAs of somatostatin receptor subtypes were detected in 98% of samples, with more than two mRNA subtypes being expressed in 55% of cases. sst1, sst4, and sst5 were heterogeneously expressed in both normal and cancerous tissues; sst3 was rarely or not expressed. sst2 was present in normal pancreatic tissues but was absent in exocrine pancreatic carcinomas and their metastases. sst2 mRNAs were detected in normal colon, sporadic polyadenomas, and 50% of Dukes' stage B and 20% of Dukes' stage C carcinomas but were undetectable in Dukes' stage D carcinomas, hepatic metastases, and adenomas from familial adenomatous polyposis. The loss of sst2 expression could represent a growth advantage in these tumors and provide an explanation for the lack of therapeutic effect of somatostatin analogues in such adenocarcinomas. A subtyping of somatostatin receptors should be carried out before considering a somatostatin analogue treatment in patients with colorectal or pancreatic cancer.


Subject(s)
Colorectal Neoplasms/metabolism , Gene Expression , Intestine, Large/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Receptors, Somatostatin/biosynthesis , Receptors, Somatostatin/genetics , Cell Line , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Humans , Neoplasm Staging , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/surgery , Polymerase Chain Reaction , RNA, Messenger/analysis , RNA, Messenger/biosynthesis , Reference Values , Tumor Cells, Cultured
11.
Cancer Res ; 57(5): 956-62, 1997 Mar 01.
Article in English | MEDLINE | ID: mdl-9041201

ABSTRACT

Among the five cloned somatostatin receptor subtypes (sst1 to sst5), sst2 mediates the antiproliferative effect of somatostatin analogues in vitro. Somatostatin analogues have been shown to inhibit cell growth in vitro and in vivo in pancreatic cancer models that expressed sst2. We recently demonstrated the loss of sst2 gene expression in human pancreatic adenocarcinomas and most of the derived pancreatic cancer cell lines. In the present study, we corrected the sst2 defect in human pancreatic cancer BxPC-3 and Capan-1 cells by stable transfection with human sst2 cDNA. In the absence of exogenous ligand, both BxPC-3 and Capan-1 cells expressing sst2 showed a significant reduction in cell growth. This inhibitory effect was blocked by treatment with antiserum to somatostatin. sst2-expressing cells produced somatostatin-like immunoreactivity that mainly corresponded to somatostatin 14, indicating the induction of a negative autocrine loop. In other respects, sst2 expression in Capan-1 cells induced a significant reduction of clonogenicity in soft agar. Moreover, a significantly reduced (Capan-1 cells) or suppressed (BxPC-3 cells) tumor growth in athymic nude mice was observed. The reversal of tumorigenicity induced by the restoration of sst2 expression suggests that the loss of sst2 contributes to the malignancy of human pancreatic cancers.


Subject(s)
Receptors, Somatostatin/physiology , Animals , Cell Adhesion , Cell Division , Cell Transformation, Neoplastic , Gene Expression Regulation, Neoplastic , Humans , Mice , Mice, Nude , Neoplasm Transplantation , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Somatostatin/physiology , Transfection , Transplantation, Heterologous , Tumor Cells, Cultured
12.
Oncogene ; 16(17): 2219-27, 1998 Apr 30.
Article in English | MEDLINE | ID: mdl-9619831

ABSTRACT

Gastrin via its G-protein coupled specific receptor induces transcription of c-fos and c-jun genes through a ras-MAPK pathway. Ornithine Decarboxylase (ODC), a growth regulated proto-oncogene, was chosen to investigate gastrin effects on translation initiation of mRNAs exhibiting a 5'UnTranslated Region (5'UTR) responsible for translation repression in quiescent cells. In AR4-2J tumoral cells, we first demonstrated that gastrin increases ODC mRNA translation. Transient transfections with various CAT chimeric constructs suggested a direct involvement of the 5'UTR in this observation. Translation of this group of mRNAs is enhanced by the availability of the cap-binding protein (eIF4E) that is increased after phosphorylation of its specific binding protein eIF4E-BP1. We found that AR4-2J cells over-expressed eIF4E protein which was not modulated by gastrin treatment. Rapamycin which inhibits 4E-BP1 phosphorylation, completely prevents gastrin-mediated increase of ODC translation indicating that 4E-BP1 could be involved in regulating ODC translation. Implication of 4E-BP1 in mediating gastrin effects is corroborated by the capacity of the ligand to affect 4E-BP1 phosphorylation. These results indicate that gastrin enhances ornithine decarboxylase mRNA translation through a rapamycin sensitive pathway and provide the first evidence in the control of 4E-BP1 phosphorylation after occupancy of a G protein-coupled receptor.


Subject(s)
Carrier Proteins , Gastrins/pharmacology , Ornithine Decarboxylase/genetics , Peptide Chain Initiation, Translational/drug effects , Phosphoproteins/drug effects , Phosphoproteins/metabolism , RNA, Messenger/genetics , Animals , COS Cells , Enzyme Induction/drug effects , Gene Expression Regulation, Enzymologic/drug effects , Intracellular Signaling Peptides and Proteins , Ornithine Decarboxylase/biosynthesis , Ornithine Decarboxylase Inhibitors , Phosphorylation/drug effects , Polyenes/pharmacology , Protein Processing, Post-Translational/drug effects , RNA, Messenger/drug effects , Rats , Repressor Proteins/pharmacology , Sirolimus , Tumor Cells, Cultured
13.
Biochim Biophys Acta ; 720(4): 378-83, 1982 Jul 22.
Article in English | MEDLINE | ID: mdl-6180776

ABSTRACT

Dispersed acini from dog pancreas were used to examine the ability of dopamine to increase cyclic AMP cellular content and the binding of [3H] dopamine. Cyclic AMP accumulation caused by dopamine was detected at 1.10(-8) M and was half-maximal at 7.9 +/- 3.4 10(-7) M. The increase at 1.10(-5) M, (7.5-fold) was equal to the half-maximal increase caused by secretin at 1.10(-9) M. Haloperidol, a dopaminergic receptor antagonist inhibited cyclic AMP accumulation caused by dopamine. The IC50 value for haloperidol, calculated from the inhibition of cyclic AMP increase caused by 1.10(-5) M dopamine was 2.3 +/- 0.9.10(-6) M. Haloperidol did not alter basal or secretin-stimulated cyclic AMP content. [3H] Dopamine binding was studied on the same batch of cells as cyclic AMP accumulation. At 37 degrees C, it was rapid, reversible, saturable and stereospecific. The Kd value for high affinity binding sites was 0.43 +/- 0.1.10(-7) M and 4.7 +/- 1.6.10(-7) M for low affinity binding sites. The concentration of drugs necessary to inhibit specific binding of dopamine by 50% was 1.2 +/- 0.4.10(-7) M epinine, 4.1 +/- 1.8.10(-6) M fluphenazine, 8.0 +/- 1.6.10(-6) M haloperidol, 4.2 +/- 1.2.10(-6) M cis-flupenthixol, 2.7 +/- 4.0.10(-5) M trans-flupenthixol, less than 1.10(-5) M apomorphine, sulpiride, naloxone and isoproterenol.


Subject(s)
Cyclic AMP/metabolism , Dopamine/pharmacology , Pancreas/metabolism , Receptors, Dopamine/metabolism , Amylases/metabolism , Animals , Binding, Competitive , Dogs , Dopamine/metabolism , Haloperidol/pharmacology , Kinetics , Pancreas/drug effects , Secretin/pharmacology , Vasoactive Intestinal Peptide/pharmacology
14.
Diabetes ; 48(10): 2015-21, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10512367

ABSTRACT

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.


Subject(s)
Pancreas/physiology , Receptors, Cholecystokinin/physiology , Adult , Cells, Cultured , Cholecystokinin/metabolism , Cloning, Molecular , Gastrins/metabolism , Gene Expression Regulation , Glucagon/metabolism , Humans , Pancreas/embryology , RNA, Messenger/metabolism , Receptor, Cholecystokinin B , Receptors, Cholecystokinin/genetics
15.
FASEB J ; 15(12): 2300-2, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11511520

ABSTRACT

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.


Subject(s)
Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase/physiology , Protein Tyrosine Phosphatases/metabolism , Receptors, Somatostatin/physiology , Signal Transduction , Animals , CHO Cells , Cell Division , Cricetinae , Cyclic GMP/biosynthesis , Intracellular Signaling Peptides and Proteins , Mice , Models, Biological , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type I , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/physiology , Somatostatin/pharmacology
16.
Cell Calcium ; 19(6): 495-500, 1996 Jun.
Article in English | MEDLINE | ID: mdl-8842516

ABSTRACT

For the first time, we have demonstrated in AR4-2J cells, an experimental model of azaserine-induced carcinoma in the rat exocrine pancreas, the co-expression of alpha 1 subunit of dihydropyridine-sensitive Ca2+ channel and the alpha 1 sub-unit of omega-conotoxin-sensitive Ca2+ channel RNA messengers which share homologous sequences with, respectively, rbC II and rbB I sub-types described in the rat brain. These two types of voltage-dependent Ca2+ channels which are functionally expressed, emphasize the acquisition during carcinogenesis of neuroendocrine features of AR4-2J cells. Additionally, using antisense phosphorothioate oligodeoxynucleotide, we demonstrated clearly the involvement of dihydropyridine-sensitive Ca2+ channels in the control of AR4-2J cell proliferation.


Subject(s)
Calcium Channels/genetics , Calcium Channels/metabolism , Calcium/metabolism , Gene Expression Regulation, Neoplastic/genetics , 3-Pyridinecarboxylic acid, 1,4-dihydro-2,6-dimethyl-5-nitro-4-(2-(trifluoromethyl)phenyl)-, Methyl ester/pharmacology , Animals , Antisense Elements (Genetics)/genetics , Base Sequence , Calcium Channel Agonists/pharmacology , Calcium Channel Blockers/pharmacology , Carcinoma/chemically induced , Carcinoma/genetics , Carcinoma/pathology , Cell Division/genetics , Cell Division/physiology , DNA Primers/chemistry , Dihydropyridines/pharmacology , Dose-Response Relationship, Drug , Mollusk Venoms/pharmacology , Pancreas/cytology , Pancreatic Neoplasms/chemically induced , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Polymerase Chain Reaction , Rats , Tumor Cells, Cultured
17.
Hum Gene Ther ; 10(6): 995-1008, 1999 Apr 10.
Article in English | MEDLINE | ID: mdl-10223733

ABSTRACT

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.


Subject(s)
Adenocarcinoma/genetics , Gene Transfer Techniques , Genetic Therapy , Pancreatic Neoplasms/therapy , Receptors, Somatostatin/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Animals , Apoptosis , Female , Humans , In Situ Nick-End Labeling , Ki-67 Antigen/immunology , Mice , Mice, Nude , Neoplasm Transplantation , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , RNA, Messenger/genetics , Receptors, Somatostatin/metabolism , Up-Regulation
18.
Protein Sci ; 8(11): 2347-54, 1999 Nov.
Article in English | MEDLINE | ID: mdl-10595537

ABSTRACT

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.


Subject(s)
Arginine , Cholecystokinin/chemistry , Cholecystokinin/metabolism , Receptors, Cholecystokinin/chemistry , Receptors, Cholecystokinin/metabolism , Amino Acid Sequence , Amino Acid Substitution , Animals , Binding Sites , COS Cells , Computer Simulation , Humans , Indoleacetic Acids/pharmacokinetics , Inositol Phosphates/metabolism , Iodine Radioisotopes , Kinetics , Models, Molecular , Mutagenesis, Site-Directed , Protein Conformation , Radioligand Assay , Receptor, Cholecystokinin A , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Thiazoles/pharmacokinetics , Transfection , Tritium
19.
Eur J Cell Biol ; 69(4): 316-26, 1996 Apr.
Article in English | MEDLINE | ID: mdl-8741213

ABSTRACT

Fibroblast growth factor (FGF-2) is a multifunctional growth factor. In cells producing this factor, FGF-2 is synthesized as different molecular weight isoforms lacking the signal peptide sequence for secretion. All forms are highly concentrated in cells. The presence of a nuclearization signal sequence in some isoforms suggests the involvement of these isoforms in cell functions bypassing the cell surface receptors. Our aims were to better define the intracellular localizations of the FGF-2 isoforms by immunocytochemistry and confocal microscopy and to analyze whether these isoforms were involved in the expression of extracellular matrix (ECM) components. We chose the pancreatic acinar cell line AR4-2J since it does not synthesize FGF-2. These cells were retrovirally transfected by point-mutated FGF-2 cDNAs. The cell lines obtained produced either the 18 kDa form (A5 cells) or the 22.5 kDa form (A3 cells). In A5 cells, the 18 kDa form was found in the cytoplasm, on the cell surface reflecting its secretion, and in the nucleoli. Parental AR4-2J cells treated with exogenous FGF-2 exhibited identical localizations, suggesting that in A5 cells the 18 kDa form followed the same translocation pathways than the exogenous FGF-2. By contrast, in A3 cells the 22.5 kDa form was predominantly localized in the nucleoplasm but was undetectable on the cell surface, suggesting its direct translocation to the nucleus. Northern and Western blot analysis showed that cells expressing the high molecular weight form exhibited a decrease of laminin B1 protein level and mRNA stability. In contrast, collagen IV and fibronectin expressions were unmodified either in FGF-2-transfected cells or in parental cells treated by exogenous FGF-2. Thus, these data indicate that: 1) 18 and 22.5 kDa FGF-2 are preferentially localized in different nuclear compartments and 2) the high molecular weight form plays a role on the expression of some ECM components.


Subject(s)
Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/metabolism , Pancreas/metabolism , Animals , Cell Line , Cells, Cultured , Fibroblast Growth Factor 2/genetics , Fibroblast Growth Factor 2/pharmacology , Gene Expression , Humans , Pancreas/cytology , Pancreas/ultrastructure , Rabbits , Transfection
20.
Endocrinology ; 133(3): 1182-91, 1993 Sep.
Article in English | MEDLINE | ID: mdl-8365360

ABSTRACT

Cholecystokinin (CCK)/gastrin receptors were characterized in calf pancreatic plasma membranes from newborns, 28- and 119-day-old milk-fed preruminants, and 119-day-old weaned ruminants. Scatchard analysis of [125I]Bolton-Hunter reagent-[Thr28,Nle31]CCK-(25-33) binding indicated two classes of binding sites: high affinity sites exhibited significant higher affinity and binding capacity (P < 0.05) in 119-day-old ruminants than in 119-day-old preruminants (Kd = 0.13 +/- 0.02 vs. 0.35 +/- 0.08 nM; binding capacity (Bmax) = 53 +/- 12 vs. 18 +/- 5 fmol/mg protein). Pharmacological analysis using selective agonists and antagonists indicated the expression of the CCK-A receptor at birth, whereas the CCK-B receptor predominated at postnatal stages. At all stages, the binding was inhibited by guanosine 5'-[gamma-thio]triphosphate. Binding site identification by photoaffinity labeling showed that at birth, the labeling occurred mainly on a 78- to 96-kilodalton (kDa) component. In milk-fed animals, aged 28 and 119 days, two membrane-binding components were labeled at 78-96 and 43-52 kDa. In 119-day-old ruminants, labeling occurred mainly on a 40- to 47-kDa protein. Deglycosylation by endo-beta-N-acetylglucosaminidase-F of the 40- to 47- and 43- to 52-kDa components resulted in the formation of a 37-kDa membrane protein. Consequently, this study demonstrated 1) the differential expression of CCK-A and -B receptors in developing calf pancreas, 2) the predominance of CCK-B receptors in normal pancreas, and 3) the maturation of CCK-B receptors during the weaning period, which includes the glycosylation level. These results suggest that CCK may play a predominant role during the early postnatal development, while gastrin and CCK-B receptors can function progressively to regulate proliferation and exocrine secretion in the calf pancreas, especially from the weaning period.


Subject(s)
Pancreas/growth & development , Receptors, Cholecystokinin/metabolism , Affinity Labels , Aging/metabolism , Animals , Animals, Newborn/metabolism , Binding Sites , Cattle , Cell Membrane/metabolism , Cholecystokinin/analogs & derivatives , Cholecystokinin/metabolism , GTP-Binding Proteins/physiology , Gastrins/metabolism , Glycosylation , Indicators and Reagents , Kinetics , Pancreas/metabolism , Peptide Fragments/metabolism , Photochemistry , Succinimides
SELECTION OF CITATIONS
SEARCH DETAIL