Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(9): 832-841, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28504211

ABSTRACT

Ecto-F1-ATPase is a complex related to mitochondrial ATP synthase which has been identified as a plasma membrane receptor for apolipoprotein A-I (apoA-I), the major protein of high-density lipoprotein (HDL), and has been shown to contribute to HDL endocytosis in several cell types. On hepatocytes, apoA-I binding to ecto-F1-ATPase stimulates extracellular ATP hydrolysis into ADP, which subsequently activates a P2Y13-mediated HDL endocytosis pathway. Interestingly, other mitochondrial proteins have been found to be expressed at the plasma membrane of several cell types. Among these, adenine nucleotide translocase (ANT) is an ADP/ATP carrier but its role in controlling extracellular ADP levels and F1-ATPase-mediated HDL endocytosis has never been investigated. Here we confirmed the presence of ANT at the plasma membrane of human hepatocytes. We then showed that ecto-ANT activity increases or reduces extracellular ADP level, depending on the extracellular ADP/ATP ratio. Interestingly, ecto-ANT co-localized with ecto-F1-ATPase at the hepatocyte plasma membrane and pharmacological inhibition of ecto-ANT activity increased extracellular ADP level when ecto-F1-ATPase was activated by apoA-I. This increase in the bioavailability of extracellular ADP accordingly translated into an increase of HDL endocytosis on human hepatocytes. This study thus uncovered a new location and function of ANT for which activity at the cell surface of hepatocytes modulates the concentration of extracellular ADP and regulates HDL endocytosis.


Subject(s)
Adenosine Diphosphate/metabolism , Adenosine Triphosphate/metabolism , Endocytosis/physiology , Hepatocytes/metabolism , Lipoproteins, HDL/metabolism , Mitochondrial ADP, ATP Translocases/metabolism , Proton-Translocating ATPases/metabolism , Apolipoprotein A-I/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Hep G2 Cells , Humans , Mitochondrial Proteins/metabolism , Signal Transduction/physiology
2.
Asian Pac J Cancer Prev ; 16(12): 5119-24, 2015.
Article in English | MEDLINE | ID: mdl-26163653

ABSTRACT

BACKGROUND: HPV vaccination in Singapore is voluntary and physician prescription-based. This study investigated the current status and intention for HPV vaccination among Singapore nurses. MATERIALS AND METHODS: All female nurses in a general hospital were given an anonymous questionnaire on HPV vaccination experience and intention of vaccinating their daughters. The influence of age, knowledge and perceived-risk of cervical cancer, and cultural background on mother's intention of vaccinating their daughters was analyzed. RESULTS: Of 2,000 nurses, 1,622 (81.1%) responded and analysis was performed on 1,611 with valid data. They showed good awareness on association of cervical cancer with multiple sexual partners (81.9%), history of sexually transmissible diseases (78.2%), and history of genital warts/HPV infection (73.5%), and on cervical cancer preventive effects of HPV vaccination (54.6%). The prevailing misconceptions of the vaccines were: investigational nature (38.9%), side effects (27.9%) and indicated for women at high risk for cervical cancer (20.5%). CONCLUSIONS: Misconceptions on the nature, role and safety of HPV vaccines low vaccine up-take rates and daughters. Dissemination of adequate and accurate HPV vaccine information and a review for school-based vaccination are needed for optimal delivery of HPV vaccines in Singapore.


Subject(s)
Attitude to Health , Health Knowledge, Attitudes, Practice , Papillomavirus Infections/prevention & control , Papillomavirus Vaccines/therapeutic use , Patient Acceptance of Health Care/statistics & numerical data , Uterine Cervical Neoplasms/prevention & control , Vaccination/psychology , Adult , Female , Follow-Up Studies , Humans , Intention , Middle Aged , Mothers/psychology , Nuclear Family/psychology , Papillomaviridae/isolation & purification , Papillomavirus Infections/psychology , Papillomavirus Infections/virology , Prognosis , Surveys and Questionnaires , Uterine Cervical Neoplasms/psychology , Uterine Cervical Neoplasms/virology
3.
Oncogene ; 34(24): 3120-30, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-25109333

ABSTRACT

Angiogenesis is essential in tumor progression and metastatic process, and increased angiogenesis has been associated with poor prognosis and relapse of colorectal cancer (CRC). VEGF has become the main target of anti-angiogenic therapy. However, most patients relapse after an initial response or present a resistance to the treatment. Identification of new pro-angiogenic factors may help to improve anti-angiogenic therapy. In this study, we demonstrated that the pro-hormone progastrin (PG), over-expressed in CRC, recognized as a growth factor, is a potent pro-angiogenic factor. In transgenic mice and human colorectal HPs producing high levels of PG, we correlated PG overexpression with an increased vascularization. In vitro, exogenous PG and conditioned media (CM) from CRC cells producing PG increased endothelial cell proliferation and migration. We also showed that treatment with exogenous PG can increase the ability of endothelial cells to form capillary-like structures. Moreover, we demonstrated that PG enhanced endothelial permeability. The finding that PG stimulated the phosphorylation of vascular endothelial (VE)-cadherin, p125-FAK, paxillin and induced actin remodelling was consistent with a role of these components in PG-stimulated endothelial cell migration and permeability. The pro-angiogenic effects observed with CM were significantly inhibited when CRC cells expressed a PG shRNA. In vivo, we found an important decrease in tumor growth and neovascularization when the CRC cells expressing the PG shRNA were xenografted in mice or in the chick chorioallantoic membrane model. We also observed an increase in the coverage of blood vessels by pericytes and a decrease in endothelial permeability when PG expression was blocked. Our results demonstrate that PG is a new pro-angiogenic factor in CRC and an attractive therapeutic target.


Subject(s)
Colorectal Neoplasms/blood supply , Gastrins/physiology , Neovascularization, Pathologic/genetics , Protein Precursors/physiology , Animals , Cells, Cultured , Chick Embryo , Colorectal Neoplasms/pathology , Gastrins/genetics , Gastrins/pharmacology , HCT116 Cells , Human Umbilical Vein Endothelial Cells , Humans , Mice , Mice, SCID , Mice, Transgenic , Protein Precursors/genetics , Protein Precursors/pharmacology , RNA, Small Interfering/pharmacology
4.
Cell Mol Life Sci ; 65(19): 3110-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18810319

ABSTRACT

The suppressor of translation initiation 4E-BP1 functions as a key regulator in cellular growth, differentiation, apoptosis and survival. While the control of 4E-BP1 activity via phosphorylation has been widely studied, the molecular mechanisms and the signaling pathways that govern 4E-BP1 gene expression are largely unknown. Here we show that inactivation of phosphatidylinositol 3-kinase (PI3K) consequent to stable expression of the antiproliferative somatostatin receptor 2 (sst2) in pancreatic cancer cells leads to transcriptional accumulation of the hypophosphorylated forms of 4E-BP1 protein. In cancer cells, while 4E-BP1 gene promoter is maintained repressed in a PI3K-dependent mechanism, sst2-dependent inactivation of the PI3K/Akt pathway releases 4E-BP1 gene transcription. Furthermore, the use of a pharmacological inhibitor and dominant-negative or -positive mutants of PI3K all affect 4E-BP1 protein expression and promoter activity in different cell lines. These data show that, in addition to inactivation of 4E-BP1 via hyperphosphorylation, signaling through the PI3K pathway silences 4E-BP1 gene transcription.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gene Silencing , Phosphatidylinositol 3-Kinases/metabolism , Phosphoproteins/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , Adaptor Proteins, Signal Transducing/genetics , Animals , Base Sequence , Cell Cycle Proteins , Cell Line , Enzyme Activation , Genes, Reporter , Humans , Molecular Sequence Data , Ornithine Decarboxylase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphoproteins/genetics , Protein Biosynthesis , Repressor Proteins/genetics , Signal Transduction/physiology
5.
Clin Exp Immunol ; 141(3): 518-25, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16045742

ABSTRACT

Human immunodeficiency virus (HIV) codes for a protein, Rev, that mediates the viral RNA export from the nucleus to the cytoplasm. Recently, it has been found that Sam68, the substrate of Src associated in mitosis, is a functional homologue of Rev, and a synergistic activator of Rev activity. Thus, it has been suggested that Sam68 may play an important role in the post-transcriptional regulation of HIV. Sam68 contains an RNA binding motif named KH [homology to the nuclear ribonucleoprotein (hnRNP) K]. Tyrosine phosphorylation of Sam68 and binding to SH3 domains have been found to negatively regulate its RNA binding capacity. Besides, tyrosine phosphorylation of Sam68 allows the formation of signalling complexes with other proteins containing SH2 and SH3 domains, suggesting a role in signal transduction of different systems in human lymphocytes, such as the T cell receptor, and leptin receptor, or the insulin receptor in other cell types. In the present work, we have found that Sam68 is tyrosine phosphorylated in peripheral blood mononuclear cells (PBMC) from HIV infected subjects, leading to the formation of signalling complexes with p85 the regulatory subunit of PI3K, GAP and STAT-3, and decreasing its RNA binding capacity. In contrast, PBMC from HIV infected subjects have lower expression levels of Sam68 compared with controls. These results suggest that Sam68 may play some role in the immune function of lymphocytes in HIV infection.


Subject(s)
HIV Infections/immunology , Leukocytes, Mononuclear/immunology , Phosphotyrosine/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Adult , Blotting, Western , DNA-Binding Proteins , HIV Infections/metabolism , Humans , Immunoblotting/methods , Immunoprecipitation , Protein Binding , RNA/metabolism , RNA, Messenger/analysis , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction
6.
J Mol Endocrinol ; 34(1): 119-26, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15691882

ABSTRACT

Hyperhomocysteinemia and insulin resistance are independent factors for cardiovascular disease. Most of the angiotoxic effects of homocysteine are related to the formation of homocysteine thiolactone and the consequent increase in oxidative stress. We have recently found that homocysteine thiolactone inhibits insulin receptor tyrosine kinase activity, which results in decreased phosphatidylinositol 3-kinase (PI3K) activity and inhibition of glycogen synthesis. Oxidative stress seemed to be the mechanism underlying these effects, since glutathione was able to restore the insulin signaling as well as the insulin-mediated glycogen synthesis. In the present work we have further investigated insulin receptor signaling studying mitogen-activated protein kinase (MAPK), glycogen synthase kinase-3 (GSK-3) and p70 S6K phosphorylation. Again, homocysteine thiolactone (50 microM) prevented insulin-mediated MAPK, GSK-3 and p70 S6K phosphorylation and these effects were blocked by glutathione (250 microM). Since MAPK and PI3K pathways, including GSK3 and S6K, seem to mediate insulin-mediated growth and proliferation, we measured DNA and protein synthesis. We have found that homocysteine thiolactone (50 microM) inhibits insulin-mediated growth and proliferation, as previously shown for glycogen synthesis. Again, these effects seem to be mediated by oxidative stress, since 250 microM glutathione completely abolished the effects of homocysteine thiolactone on insulin-stimulated DNA and protein synthesis. In conclusion, these data suggest that homocysteine thiolactone impairs insulin signaling by a mechanism involving oxidative stress, leading to a defect in the action of insulin on growth and proliferation.


Subject(s)
Glycogen Synthase Kinase 3/metabolism , Homocysteine/analogs & derivatives , Homocysteine/metabolism , Mitogen-Activated Protein Kinases/metabolism , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Animals , Cell Proliferation , DNA Replication/physiology , Insulin/metabolism , Protein Biosynthesis/physiology , Rats
7.
Cell Mol Life Sci ; 62(1): 36-43, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15619005

ABSTRACT

Sam68, the substrate of Src in mitosis, belongs to the family of RNA binding proteins. Sam68 contains consensus sequences to interact with other proteins via specific domains. Thus, Sam68 has various proline-rich sequences to interact with SH3 domain-containing proteins. Moreover, Sam68 also has a C-terminal domain rich in tyrosine residues that is a substrate for tyrosine kinases. Tyrosine phosphorylation of Sam68 promotes its interaction with SH2 containing proteins. The association of Sam68 with SH3 domain-containing proteins, and its tyrosine phosphorylation may negatively regulate its RNA binding activity. The presence of these consensus sequences to interact with different domains allows this protein to participate in signal transduction pathways triggered by tyrosine kinases. Thus, Sam68 participates in the signaling of T cell receptors, leptin and insulin receptors. In these systems Sam68 is tyrosine phosphorylated and recruited to specific signaling complexes. The participation of Sam68 in signaling suggests that it may function as an adaptor molecule, working as a dock to recruit other signaling molecules. Finally, the connection between this role of Sam68 in protein-protein interaction with RNA binding activity may connect signal transduction of tyrosine kinases with the regulation of RNA metabolism.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , RNA-Binding Proteins/physiology , Signal Transduction/physiology , Animals , DNA-Binding Proteins , Humans , Protein Conformation , RNA/metabolism , Receptor, Insulin/physiology , Receptors, Antigen, T-Cell/physiology , Receptors, Cell Surface/physiology , Receptors, Leptin
8.
Cell Mol Life Sci ; 60(4): 751-8, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12785721

ABSTRACT

The role of Sam68, an RNA binding protein and putative substrate of the insulin receptor (IR) in insulin signaling was studied using CHO wild type (WT) cells, CHO cells overexpressing IR, and rat white adipocytes as a physiological system. In CHO-IR cells and adipocytes, Sam68 was tyrosine phosphorylated in response to insulin, and then associated with p85 phosphatidylinositol-3 kinase along with IRS-1. Sam68 was localized mainly in the nucleus of CHO-WT, and both in the nucleus and cytoplasm of CHO-IR cells, but only in the cytoplasm of rat white adipocytes. Insulin stimulation for 16 h enhanced the expression of Sam68 in rat adipocytes and CHO-IR cells. Moreover, CHO-IR cells expressed more Sam68 than CHO-WT, suggesting that overexpression of the IR is enough to induce the expression of Sam68. In summary, these results demonstrate that Sam68 works as a cytoplasmic docking protein which is recruited by IR signaling and whose expression is induced by insulin stimulation, suggesting a putative role for Sam68 in insulin signal transduction.


Subject(s)
Insulin/metabolism , RNA-Binding Proteins/genetics , Signal Transduction/physiology , Adipocytes , Animals , CHO Cells , Cricetinae , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Insulin/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/drug effects , RNA-Binding Proteins/metabolism , Rats , Receptor, Insulin/drug effects , Receptor, Insulin/metabolism , Signal Transduction/drug effects
9.
Clin Exp Immunol ; 133(1): 11-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12823272

ABSTRACT

Leptin is a an adipocyte-secreted hormone that regulates weight centrally. However, the leptin receptor is expressed not only in the central nervous system, but also in peripheral tissues, such as haematopoietic and immune systems. Therefore, the physiological role of leptin should not be limited to the regulation of food intake and energy expenditure. Moreover, the leptin receptor bears homology to members of the class I cytokine family, and recent data have demonstrated that leptin is able to modulate the immune response. Thus, the leptin receptor is expressed in human peripheral blood mononuclear cells, mediating the leptin effect on proliferation and activation. In vitro activation and HIV infection in vivo induce the expression of the long isoform of the leptin receptor in mononuclear cells. Also, leptin stimulates the production of proinflammatory cytokines from cultured monocytes and enhances the production of Th1 type cytokines from stimulated lymphocytes. Moreover, leptin has a trophic effect on monocytes, preventing apoptosis induced by serum deprivation. Leptin stimulation activates JAK-STAT, IRS-1-PI3K and MAPK signalling pathways. Leptin also stimulates Tyr-phosphorylation of the RNA-binding protein Sam68 mediating the dissociation from RNA. In this way, leptin signalling could modulate RNA metabolism. These signal transduction pathways provide possible mechanisms whereby leptin may modulate activation of peripheral blood mononuclear cells. Therefore, these data support the hypothesis regarding leptin as a proinflammatory cytokine with a possible role as a link between the nutritional status and the immune response. Moreover, these immunoregulatory functions of leptin could have some relevance in the pathophysiology of obesity.


Subject(s)
Adipocytes/metabolism , Leptin/immunology , Leukocytes, Mononuclear/immunology , Receptors, Cell Surface/metabolism , Signal Transduction/physiology , Apoptosis , Autoimmune Diseases/immunology , Cells, Cultured , Cytokines/immunology , Humans , Janus Kinase 1 , Leptin/pharmacology , Leukocytes, Mononuclear/drug effects , Lymphocyte Activation , MAP Kinase Signaling System , Nutritional Status , Obesity/immunology , Phosphatidylinositol 3-Kinases/metabolism , Protein Isoforms/metabolism , Protein-Tyrosine Kinases/metabolism , Receptors, Leptin , Th1 Cells/metabolism
10.
Saudi Med J ; 23(6): 672-4, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12070545

ABSTRACT

OBJECTIVE: This study is aimed at determining the incidence of surgical site infections, with antibiotic use. Furthermore, to correlate infection with type of operations, length of intervention, number of stitches, pre-operative hospitalization, age and sex of the patient. METHODS: All Yemeni patients (N=601) who underwent surgical intervention, with preoperative antibiotic prophylaxis, at Al-Kuwait University Hospital, Sana'a, Republic of Yemen, during 1st August 2000 through to 30th November 2000 were followed up until stitch removal or discharge. Data was registered in a spreadsheet and processed statistically by statistical package for social sciences 10.0. RESULTS: Overall incidence of surgical site infections was 2.2%, 0.5% in clean operation, 2.8% in clean contaminated, 9.1% in contaminated and 2.3% in dirty operations. Surgical site infections were found positively correlated with duration of operation (P=0.015) and number of stitches (P=0.017), but insignificantly associated with sex, age, type of operation and pre-operative hospitalization. CONCLUSION: In conclusion, surgical site infections, with antibiotic use, were found low. Higher number of stitches and longer duration of operation were the risk factors.


Subject(s)
Antibiotic Prophylaxis , Surgical Procedures, Operative/methods , Surgical Wound Infection/prevention & control , Adolescent , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Hospitals, University , Humans , Infant , Male , Middle Aged , Risk Factors , Yemen
11.
Mol Cell Endocrinol ; 183(1-2): 113-21, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11604231

ABSTRACT

The 68 kDa Src substrate associated during mitosis (Sam68) is an RNA binding protein with Src homology (SH) 2 and 3 domain binding sites. We have recently found that Sam68 is a substrate of the insulin receptor (IR) and that Tyr-phosphorylated Sam68 associates with the SH2 domains of p85 PI3K. In the present work, using HTC-IR cells, we have found that insulin stimulation promotes the relocalization of Sam68 from the nucleus to the cytoplasm, and we have further studied the role of Sam68 in insulin receptor signaling complexes, by co-precipitating experiments. Thus, Sam68 is co-precipitated with p85 PI3K, IRS-1 and IR. The association of Sam68 with these complexes is mediated by the SH2 domains of PI3K. Moreover, we have found that Sam68 is a p120GAP associated protein after Tyr-phosphorylation by the IR. This association is mediated by the SH2 domains of GAP (preferentially the C-terminal SH2). Thus, Sam68 is linking p120GAP to PI3K signaling pathway. In fact, PI3K activity was increased in both anti-Sam68 and anti-GAP immmunoprecipitates upon insulin stimulation. We propose that the recruitment of the docking protein Sam68 to the PI3K pathway may serve to allow the association of other signaling molecules, i.e. p120GAP. In this way, these signaling complexes may modulate other signaling cascades of IR, such as p21Ras pathway.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , RNA-Binding Proteins/metabolism , Receptor, Insulin/metabolism , Signal Transduction , p120 GTPase Activating Protein/metabolism , Animals , Culture Media, Serum-Free , Insulin/pharmacology , Macromolecular Substances , Protein Structure, Tertiary , Protein Transport , Rats , Recombinant Fusion Proteins/metabolism , Tumor Cells, Cultured , src-Family Kinases/metabolism
12.
J Hepatol ; 35(1): 80-5, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11495046

ABSTRACT

BACKGROUND/AIMS: Pancreastatin, a chromogranin A-derived peptide, has a counter-regulatory effect on insulin action. We have previously characterized pancreastatin receptor and signalling in rat liver and HTC hepatoma cells. A G alpha(q/11)-PLC-beta pathway leads to an increase in [Ca2+]i, PKC and mitogen activated protein kinase (MAPK) activation. These data suggested that pancreastatin might have a role in growth and proliferation, similar to other calcium-mobilizing hormones. METHODS: DNA and protein synthesis were measured as [3H]-thymidine and [3H]-leucine incorporation. Nitric oxide (NO) was determined by the Griess method and cGMP production was quantified by enzyme-linked immunoassay. RESULTS: Contrary to the expected results, we have found that pancreastatin inhibits protein and DNA synthesis in HTC hepatoma cells. On the other hand, when the activity of NO synthase was inhibited by N-monomethyl-L-arginine (NMLA), the inhibitory effect of pancreastatin on DNA and protein synthesis was not only reverted, but a dose-dependent stimulatory effect was observed, probably due to MAPK activation, since it was prevented by PD98059. These data strongly suggested the role of NO in the inhibitory effect of pancreastatin on protein and DNA synthesis, which is overcoming the effect on MAPK activation. Moreover, pancreastatin dose-dependently increased NO production in parallel to cyclic guanosine monophosphate (cGMP). Both effects were prevented by NMLA. Finally, an indirect effect of pancreastatin through the induction of apoptosis was ruled out. CONCLUSIONS: Therefore, the NO and the cGMP produced by the NO-activated guanylate cyclase may mediate the dose-dependent inhibitory effect of pancreastatin on growth and proliferation in HTC hepatoma cells.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , DNA/antagonists & inhibitors , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Nitric Oxide/biosynthesis , Pancreatic Hormones/pharmacology , Protein Synthesis Inhibitors/pharmacology , Animals , Carcinoma, Hepatocellular/pathology , Cell Division/drug effects , Chromogranin A , Chromogranins , Cyclic GMP/biosynthesis , DNA/biosynthesis , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Liver Neoplasms/pathology , Nitric Oxide/antagonists & inhibitors , Pancreatic Hormones/chemistry , Rats , Tumor Cells, Cultured , omega-N-Methylarginine/pharmacology
13.
J Mol Endocrinol ; 27(1): 85-91, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11463579

ABSTRACT

Hyperhomocysteinemia and insulin resistance are independent factors for cardiovascular disease. Most of the angiotoxic effects of homocysteine are related to the formation of homocysteine thiolactone and the consequent increase in oxidative stress. The oxidative stress has also been shown to impair insulin action, therefore leading to insulin resistance. In order to study a putative direct effect of homocysteine on insulin signaling, we have characterized the molecular counter-regulation of the early events in the signal transduction of the insulin receptor, and the metabolic end-point of glycogen synthesis. We employed HTC rat hepatoma cells transfected with the human insulin receptor. A 10 min exposure to homocysteine thiolactone (50 microM) resulted in a significant inhibition of insulin-stimulated tyrosine phosphorylation of the insulin receptor beta-subunit and its substrates IRS-1 and p60-70, as well as their association with the p85 regulatory subunit of phosphatidylinositol 3-kinase. These effects led to impairment of the insulin-stimulated phosphatidylinositol 3-kinase activity, which plays a central role in regulating insulin action. Thus, insulin-stimulated glycogen synthesis was also inhibited by homocysteine thiolactone. To investigate whether oxidative stress was mediating the counter-regulatory effect of homocysteine thiolactone on insulin signaling, we preincubated the cells (5 min) with 250 microM glutathione prior to the incubation with homocysteine (10 min) and subsequent insulin challenge. Glutathione completely abolished the effects of homocysteine thiolactone on insulin-receptor signaling and restored the insulin-stimulated glycogen synthesis. In conclusion, these data suggest that homocysteine thiolactone impairs insulin signaling by a mechanism involving oxidative stress, leading to a defect in insulin action.


Subject(s)
Glutathione/physiology , Homocysteine/pharmacology , Insulin/metabolism , Signal Transduction/drug effects , Animals , Homocysteine/analogs & derivatives , Insulin Receptor Substrate Proteins , Oxidative Stress , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/metabolism , Phosphorylation , Rats , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism , Signal Transduction/physiology , Tumor Cells, Cultured , Tyrosine/metabolism
14.
Eur J Pharmacol ; 397(2-3): 229-35, 2000 Jun 02.
Article in English | MEDLINE | ID: mdl-10844119

ABSTRACT

Pancreastatin, a chromogranin A-derived peptide widely distributed throughout the neuroendocrine system, has a general inhibitory effect on endocrine secretion and a counterregulatory effect on insulin action. We have recently described the cross-talk of pancreastatin with insulin signaling in rat hepatoma cells (HTC), where it inhibits insulin action and signaling through the serine phosphorylation of the insulin receptor, thereby impairing tyrosine kinase activity. Here, we have characterized pancreastatin receptors and signaling in HTC cells. The pancreastatin effector systems were studied by determining phospholipase C activity in HTC membranes and mitogen-activated protein kinase (MAPK) phosphorylation activity in HTC cells. Binding studies with radiolabeled pancreastatin showed a population of high affinity binding sites, with a B(max) of 8 fmol/mg protein and a K(d) of 0.6 nM. Moreover, we assessed the coupling of the receptor with a G protein system by inhibiting the binding with guanine nucleotide and by measuring the GTP binding to HTC membranes. We found that pancreastatin receptor was coupled with a G alpha(q/11) protein which activates phospholipase C-beta(1) and phospholipase C-beta(3), in addition to MAPK via both beta gamma and alpha(q/11).


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Receptors, Gastrointestinal Hormone/physiology , Signal Transduction , Animals , Binding, Competitive , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Membrane/metabolism , Chromogranin A , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , GTP-Binding Proteins/drug effects , GTP-Binding Proteins/metabolism , MAP Kinase Signaling System/drug effects , Pancreatic Hormones/metabolism , Pancreatic Hormones/pharmacology , Protein Binding , Rats , Receptors, Gastrointestinal Hormone/metabolism , Tumor Cells, Cultured , Type C Phospholipases/drug effects , Type C Phospholipases/metabolism
16.
FEBS Lett ; 455(3): 307-10, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10437794

ABSTRACT

The 68 kDa Src substrate associated during mitosis is an RNA binding protein with Src homology 2 and 3 domain binding sites. A role for Src associated in mitosis 68 as an adaptor protein in signaling transduction has been proposed in different systems such as T-cell receptors. In the present work, we have sought to assess the possible role of Src associated in mitosis 68 in insulin receptor signaling. We performed in vivo studies in HTC-IR cells and in vitro studies using recombinant Src associated in mitosis 68, purified insulin receptor and fusion proteins containing either the N-terminal or the C-terminal Src homology 2 domain of p85 phosphatidylinositol-3-kinase. We have found that Src associated in mitosis 68 is a substrate of the insulin receptor both in vivo and in vitro. Moreover, tyrosine-phosphorylated Src associated in mitosis 68 was found to associate with p85 phosphatidylinositol-3-kinase in response to insulin, as assessed by co-immunoprecipitation studies. Therefore, Src associated in mitosis 68 may be part of the signaling complexes of insulin receptor along with p85. In vitro studies demonstrate that Src associated in mitosis 68 associates with the Src homology 2 domains of p85 after tyrosine phosphorylation by the activated insulin receptor. Moreover, tyr-phosphorylated Src associated in mitosis 68 binds with a higher affinity to the N-terminal Src homology 2 domain of p85 compared to the C-terminal Src homology 2 domain of p85, suggesting a preferential association of Src associated in mitosis 68 with the N-terminal Src homology 2 domain of p85. This association may be important for the link of the signaling with RNA metabolism.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , RNA-Binding Proteins/metabolism , Receptor, Insulin/metabolism , Adaptor Proteins, Signal Transducing , Animals , DNA-Binding Proteins , Humans , Insulin/pharmacology , Mitosis , Phosphatidylinositol 3-Kinases/chemistry , Phosphorylation , RNA-Binding Proteins/chemistry , Rats , Receptor, Insulin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Tumor Cells, Cultured , Tyrosine/metabolism , src Homology Domains
17.
Cell Mol Life Sci ; 55(1): 142-7, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10065161

ABSTRACT

Insulin action is initiated by binding to its cognate receptor, which then triggers multiple cellular responses by activating different signaling pathways. There is evidence that insulin receptor signaling may involve G protein activation in different target cells. We have studied the activation of G proteins in rat hepatoma (HTC) cells. We found that insulin stimulated binding of guanosine 5'-O-(3-thiotriphosphate) (GTP-gamma-35S) to plasma membrane proteins of HTC cells, in a dose-dependent manner. This effect was completely blocked by pertussis toxin treatment of the membranes, suggesting the involvement of G proteins of the G alpha i/G alpha o family. The expression of these G alpha proteins was checked by Western blotting. Next, we used blocking antibodies to sort out the specific G alpha protein activated by insulin stimulation. Anti-G alpha il,2 antibodies completely prevented insulin-stimulated GTP binding, whereas anti-G alpha o,i3 did not modify this effect of insulin on GTP binding. Moreover, we found physical association of the insulin receptor with G alpha il,2 by copurification studies. These results further support the involvement of a pertussis toxin-sensitive G protein in insulin receptor signaling and provides some evidence of specific association and activation of G alpha il,2 protein by insulin. These findings suggest that G alpha il,2 proteins might be involved in insulin action.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Cell Membrane/metabolism , GTP-Binding Proteins/metabolism , Insulin/pharmacology , Animals , Antibodies/pharmacology , Cells, Cultured , GTP-Binding Proteins/immunology , Guanosine Triphosphate/metabolism , Pertussis Toxin , Protein Binding/drug effects , Rats , Receptor, Insulin/metabolism , Virulence Factors, Bordetella/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...