Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Marion Cornu is active.

Publication


Featured researches published by Marion Cornu.


Diabetes | 2008

Exendin-4 Protects β-Cells From Interleukin-1β–Induced Apoptosis by Interfering With the c-Jun NH2-Terminal Kinase Pathway

Mourad Ferdaoussi; Saida Abdelli; Jiang-Yan Yang; Marion Cornu; Guy Niederhauser; Dimitri Favre; Christian Widmann; Romano Regazzi; Bernard Thorens; Gérard Waeber; Amar Abderrahmani

OBJECTIVE— The pro-inflammatory cytokine interleukin-1β (IL-1β) generates pancreatic β-cells apoptosis mainly through activation of the c-Jun NH2-terminal kinase (JNK) pathway. This study was designed to investigate whether the long-acting agonist of the hormone glucagon-like peptide 1 (GLP-1) receptor exendin-4 (ex-4), which mediates protective effects against cytokine-induced β-cell apoptosis, could interfere with the JNK pathway. RESEARCH DESIGN AND METHODS— Isolated human, rat, and mouse islets and the rat insulin-secreting INS-1E cells were incubated with ex-4 in the presence or absence of IL-1β. JNK activity was assessed by solid-phase JNK kinase assay and quantification of c-Jun expression. Cell apoptosis was determined by scoring cells displaying pycnotic nuclei. RESULTS— Ex-4 inhibited induction of the JNK pathway elicited by IL-1β. This effect was mimicked with the use of cAMP-raising agents isobutylmethylxanthine and forskolin and required activation of the protein kinase A. Inhibition of the JNK pathway by ex-4 or IBMX and forskolin was concomitant with a rise in the levels of islet-brain 1 (IB1), a potent blocker of the stress-induced JNK pathway. In fact, ex-4 as well as IBMX and forskolin induced expression of IB1 at the promoter level through cAMP response element binding transcription factor 1. Suppression of IB1 levels with the use of RNA interference strategy impaired the protective effects of ex-4 against apoptosis induced by IL-1β. CONCLUSIONS— The data establish the requirement of IB1 in the protective action of ex-4 against apoptosis elicited by IL-1β and highlight the GLP-1 mimetics as new potent inhibitors of the JNK signaling induced by cytokines.


Journal of Clinical Investigation | 2012

MicroRNAs contribute to compensatory β cell expansion during pregnancy and obesity

Cécile Jacovetti; Amar Abderrahmani; Géraldine Parnaud; Jean-Christophe Jonas; Marie-Line Peyot; Marion Cornu; Ross Laybutt; Sophie Rome; Bernard Thorens; Marc Prentki; Domenico Bosco; Romano Regazzi

Pregnancy and obesity are frequently associated with diminished insulin sensitivity, which is normally compensated for by an expansion of the functional β cell mass that prevents chronic hyperglycemia and development of diabetes mellitus. The molecular basis underlying compensatory β cell mass expansion is largely unknown. We found in rodents that β cell mass expansion during pregnancy and obesity is associated with changes in the expression of several islet microRNAs, including miR-338-3p. In isolated pancreatic islets, we recapitulated the decreased miR-338-3p level observed in gestation and obesity by activating the G protein-coupled estrogen receptor GPR30 and the glucagon-like peptide 1 (GLP1) receptor. Blockade of miR-338-3p in β cells using specific anti-miR molecules mimicked gene expression changes occurring during β cell mass expansion and resulted in increased proliferation and improved survival both in vitro and in vivo. These findings point to a major role for miR-338-3p in compensatory β cell mass expansion occurring under different insulin resistance states.


Diabetes | 2009

Glucagon-Like Peptide-1 Protects β-Cells Against Apoptosis by Increasing the Activity of an Igf-2/Igf-1 Receptor Autocrine Loop

Marion Cornu; Jiang-Yan Yang; Evrim Jaccard; Carine Poussin; Christian Widmann; Bernard Thorens

OBJECTIVE The gluco-incretin hormones glucagon-like peptide (GLP)-1 and gastric inhibitory peptide (GIP) protect β-cells against cytokine-induced apoptosis. Their action is initiated by binding to specific receptors that activate the cAMP signaling pathway, but the downstream events are not fully elucidated. Here we searched for mechanisms that may underlie this protective effect. RESEARCH DESIGN AND METHODS We performed comparative transcriptomic analysis of islets from control and GipR−/−;Glp-1-R−/− mice, which have increased sensitivity to cytokine-induced apoptosis. We found that IGF-1 receptor expression was markedly reduced in the mutant islets. Because the IGF-1 receptor signaling pathway is known for its antiapoptotic effect, we explored the relationship between gluco-incretin action, IGF-1 receptor expression and signaling, and apoptosis. RESULTS We found that GLP-1 robustly stimulated IGF-1 receptor expression and Akt phosphorylation and that increased Akt phosphorylation was dependent on IGF-1 but not insulin receptor expression. We demonstrated that GLP-1–induced Akt phosphorylation required active secretion, indicating the presence of an autocrine activation mechanism; we showed that activation of IGF-1 receptor signaling was dependent on the secretion of IGF-2. We demonstrated, both in MIN6 cell line and primary β-cells, that reducing IGF-1 receptor or IGF-2 expression or neutralizing secreted IGF-2 suppressed GLP-1–induced protection against apoptosis. CONCLUSIONS An IGF-2/IGF-1 receptor autocrine loop operates in β-cells. GLP-1 increases its activity by augmenting IGF-1 receptor expression and by stimulating secretion; this mechanism is required for GLP-1–induced protection against apoptosis. These findings may lead to novel ways of preventing β-cell loss in the pathogenesis of diabetes.


Journal of Biological Chemistry | 2010

Glucagon-like Peptide-1 Increases β-Cell Glucose Competence and Proliferation by Translational Induction of Insulin-like Growth Factor-1 Receptor Expression

Marion Cornu; Honey Modi; Dan Kawamori; Rohit N. Kulkarni; Magali Joffraud; Bernard Thorens

Glucagon-like peptide-1 (GLP-1) protects β-cells against apoptosis, increases their glucose competence, and induces their proliferation. We previously demonstrated that the anti-apoptotic effect was mediated by an increase in insulin-like growth factor-1 receptor (IGF-1R) expression and signaling, which was dependent on autocrine secretion of insulin-like growth factor 2 (IGF-2). Here, we further investigated how GLP-1 induces IGF-1R expression and whether the IGF-2/IGF-1R autocrine loop is also involved in mediating GLP-1-increase in glucose competence and proliferation. We show that GLP-1 up-regulated IGF-1R expression by a protein kinase A-dependent translational control mechanism, whereas isobutylmethylxanthine, which led to higher intracellular accumulation of cAMP than GLP-1, increased both IGF-1R transcription and translation. We then demonstrated, using MIN6 cells and primary islets, that the glucose competence of these cells was dependent on the level of IGF-1R expression and on IGF-2 secretion. We showed that GLP-1-induced primary β-cell proliferation was suppressed by Igf-1r gene inactivation and by IGF-2 immunoneutralization or knockdown. Together our data show that regulation of β-cell number and function by GLP-1 depends on the cAMP/protein kinase A mediated-induction of IGF-1R expression and the increased activity of an IGF-2/IGF-1R autocrine loop.


Diabetes | 2011

In Vivo Conditional Pax4 Overexpression in Mature Islet β-Cells Prevents Stress-Induced Hyperglycemia in Mice

Kai Hui Hu He; Petra Isabel Lorenzo; Thierry Brun; Carmen M. Jimenez Moreno; Deborah Aeberhard; Jorge Vallejo Ortega; Marion Cornu; Fabrizio Thorel; Asllan Gjinovci; Bernard Thorens; Pedro Luis Herrera; Paolo Meda; Claes B. Wollheim; Benoit R. Gauthier

OBJECTIVE To establish the role of the transcription factor Pax4 in pancreatic islet expansion and survival in response to physiological stress and its impact on glucose metabolism, we generated transgenic mice conditionally and selectively overexpressing Pax4 or a diabetes-linked mutant variant (Pax4R129W) in β-cells. RESEARCH DESIGN AND METHODS Glucose homeostasis and β-cell death and proliferation were assessed in Pax4- or Pax4R129W-overexpressing transgenic animals challenged with or without streptozotocin. Isolated transgenic islets were also exposed to cytokines, and apoptosis was evaluated by DNA fragmentation or cytochrome C release. The expression profiles of proliferation and apoptotic genes and β-cell markers were studied by immunohistochemistry and quantitative RT-PCR. RESULTS Pax4 but not Pax4R129W protected animals against streptozotocin-induced hyperglycemia and isolated islets from cytokine-mediated β-cell apoptosis. Cytochrome C release was abrogated in Pax4 islets treated with cytokines. Interleukin-1β transcript levels were suppressed in Pax4 islets, whereas they were increased along with NOS2 in Pax4R129W islets. Bcl-2, Cdk4, and c-myc expression levels were increased in Pax4 islets while MafA, insulin, and GLUT2 transcript levels were suppressed in both animal models. Long-term Pax4 expression promoted proliferation of a Pdx1-positive cell subpopulation while impeding insulin secretion. Suppression of Pax4 rescued this defect with a concomitant increase in pancreatic insulin content. CONCLUSIONS Pax4 protects adult islets from stress-induced apoptosis by suppressing selective nuclear factor-κB target genes while increasing Bcl-2 levels. Furthermore, it promotes dedifferentiation and proliferation of β-cells through MafA repression, with a concomitant increase in Cdk4 and c-myc expression.


Journal of Biological Chemistry | 2005

Expression of an Uncleavable N-terminal RasGAP Fragment in Insulin-secreting Cells Increases Their Resistance toward Apoptotic Stimuli without Affecting Their Glucose-induced Insulin Secretion

Jiang-Yan Yang; Joël Walicki; Amar Abderrahmani; Marion Cornu; Gérard Waeber; Bernard Thorens; Christian Widmann

Apoptosis of pancreatic β cells is implicated in the onset of type 1 and type 2 diabetes. Consequently, strategies aimed at increasing the resistance of β cells toward apoptosis could be beneficial in the treatment of diabetes. RasGAP, a regulator of Ras and Rho GTPases, is an atypical caspase substrate, since it inhibits, rather than favors, apoptosis when it is partially cleaved by caspase-3 at position 455. The antiapoptotic signal generated by the partial processing of RasGAP is mediated by the N-terminal fragment (fragment N) in a Ras-phosphatidylinositol 3-kinase-Akt-dependent, but NF-κB-independent, manner. Further cleavage of fragment N at position 157 abrogates its antiapoptotic properties. Here we demonstrate that an uncleavable form of fragment N activates Akt, represses NF-κB activity, and protects the conditionally immortalized pancreatic insulinoma βTC-tet cell line against various insults, including exposure to genotoxins, trophic support withdrawal, and incubation with inflammatory cytokines. Fragment N also induced Akt activity and protection against cytokine-induced apoptosis in primary pancreatic islet cells. Fragment N did not alter insulin cell content and insulin secretion in response to glucose. These data indicate that fragment N protects β cells without affecting their function. The pathways regulated by fragment N are therefore promising targets for antidiabetogenic therapy.


Islets | 2009

GLP-1 protects β-cells against apoptosis by enhancing the activity of an IGF-2/IGF1-receptor autocrine loop

Marion Cornu; Bernard Thorens

GLP-1 protects β-cells against apoptosis by still incompletely understood mechanisms. In a recent study, we searched for novel anti-apoptotic pathways by performing comparative transcriptomic analysis of islets from Gipr-/-;Glp-1r-/- mice, which show increased susceptibility to cytokine-induced apoptosis. We observed a strong reduction in IGF-1R expression in the knockout islets suggesting a link between the gluco-incretin and IGF-1R signaling pathways. Using MIN6 and primary islet cells, we demonstrated that GLP-1 strongly stimulates IGF-1R expression and that activation of the IGF-1R/Akt signaling pathway required active secretion of IGF-2 by the β-cells. We showed that inactivation of the IGF-1 receptor gene in β-cells or preventing its up-regulation by GLP-1, as well as suppressing IGF-2 expression or action, blocked the protective effect of GLP-1 against cytokine-induced apoptosis. Thus, an IGF-2/IGF-1 receptor autocrine loop operates in β-cells and GLP-1 increases its activity by enhancing IGF-1R expression and by stimulating IGF-2 secretion. This mechanism is required for GLP-1 to protect β-cells against apoptosis.


Journal of Biological Chemistry | 2014

Glutamine stimulates biosynthesis and secretion of insulin-like growth factor 2 (IGF2), an autocrine regulator of beta cell mass and function.

Honey Modi; Marion Cornu; Bernard Thorens

Background: An IGF2/IGF1R autocrine loop regulates beta cell glucose competence, proliferation, and apoptosis, and GLP-1 increases IGF1R expression. Results: Glutamine enhances IGF2 biosynthesis, secretion, and IGF2-dependent Akt phosphorylation. Conclusion: Glutamine controls the activity of the IGF2/IGF1R autocrine loop through IGF2 up-regulation. Significance: The IGF2/IGF1R autocrine loop integrates changes in feeding and metabolic state to adapt beta cell mass and function. IGF2 is an autocrine ligand for the beta cell IGF1R receptor and GLP-1 increases the activity of this autocrine loop by enhancing IGF1R expression, a mechanism that mediates the trophic effects of GLP-1 on beta cell mass and function. Here, we investigated the regulation of IGF2 biosynthesis and secretion. We showed that glutamine rapidly and strongly induced IGF2 mRNA translation using reporter constructs transduced in MIN6 cells and primary islet cells. This was followed by rapid secretion of IGF2 via the regulated pathway, as revealed by the presence of mature IGF2 in insulin granule fractions and by inhibition of secretion by nimodipine and diazoxide. When maximally stimulated by glutamine, the amount of secreted IGF2 rapidly exceeded its initial intracellular pool and tolbutamide, and high K+ increased IGF2 secretion only marginally. This indicates that the intracellular pool of IGF2 is small and that sustained secretion requires de novo synthesis. The stimulatory effect of glutamine necessitates its metabolism but not mTOR activation. Finally, exposure of insulinomas or beta cells to glutamine induced Akt phosphorylation, an effect that was dependent on IGF2 secretion, and reduced cytokine-induced apoptosis. Thus, glutamine controls the activity of the beta cell IGF2/IGF1R autocrine loop by increasing the biosynthesis and secretion of IGF2. This autocrine loop can thus integrate changes in feeding and metabolic state to adapt beta cell mass and function.


Diabetes & Metabolism | 2008

O2 Glucagon-like peptide-1 protège les cellules bêta contre l’apoptose et augmente la sécrétion d’insuline induite par le glucose via l’induction d’une boucle autocrine dépendante du récepteur à l’Insulin-like growth factor-1

Marion Cornu; Carine Poussin; Jiang-Yan Yang; Christian Widmann; Bernard Thorens

Introduction Des experiences precedentes ont revele chez les ilots de souris deficientes pour les recepteurs glucose-dependent insulinotropic polypeptide (GIP) et glucagon-like peptide-1 (GLP-1) un defaut de secretion d’insuline tout en maintenant une structure et un contenu en insuline normal. Materiels et methodes Dans le but d’identifier les possibles causes de cette deregulation fonctionnelle, nous avons effectue une analyse transcriptomique des ARNm exprimes dans les ilots des souris deficientes pour les recepteurs GIP et GLP-1. L’expression des genes ainsi que leurs fonctions modulees par ces gluco-incretines, ont ete etudiees dans des ilots pancreatiques et une lignee de cellules beta (MIN6) de souris. Resultats Nous avons remarque que l’expression de l’Insulin-like growth factor 1-receptor (IGF1-R) etait fortement reduite dans les ilots des souris deficientes pour les recepteurs des gluco-incretines a la fois au niveau ARNm et proteique et que le GLP-1 induit l’expression d’IGF1-R au niveau proteique dans les ilots de souris controles. Nous avons montre qu’apres traitement des MIN6, le GLP-1 augmente l’expression d’IGF1-R seulement au niveau proteique (8 fois) tandis que l’inhibition de la phosphodiesterase, par l’IBMX, augmente l’expression d’IGF1-R a la fois au niveau ARNm (20 fois) et proteique (30 fois). L’expression d’IGF1-R induite par le GLP-1 ou l’IBMX, presente une cinetique similaire a celle de la phosphorylation d’Akt mais differente de celle de Erk. De plus, la phosphorylation d’Akt est fortement diminuee apres inhibition de l’expression d’IGF1-R par siRNA ou par l’inhibition de la secretion induite par le glucose. Enfin, la diminution de l’expression d’IGF1-R supprime l’effet protecteur du GLP-1 contre l’apoptose induite par les cytokines et reduit la secretion d’insuline stimulee par le glucose. Discussion Bien que le produit de secretion qui active l’IGF1-R reste encore a determiner, nous avons montre que la secretion d’IGF-II etait regulee par le glucose. Conclusion Nos donnees indiquent que le traitement des cellules beta de souris par le GLP-1 entraine l’activation d’une boucle autocrine dependante d’IGF1-R, qui est necessaire pour la protection contre l’apoptose et l’augmentation de la secretion d’insuline induite par le glucose.


Diabetes | 2009

Abstracts 外因性のglucose-dependent insulinotropic polypeptide (GIP)は2型糖尿病における食後高血糖を悪化させる

Marion Cornu; Jiang-Yan Yang; Evrim Jaccard

Collaboration


Dive into the Marion Cornu's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Honey Modi

University of Lausanne

View shared research outputs
Researchain Logo
Decentralizing Knowledge