6 research outputs found

    Epithelial insulin signalling: A gatekeeper of the gut barrier function?

    No full text
    Accepted abstracts will be published in the Abstract Volume, a supplement issue of Diabetologia, the journal of the EASD. Furthermore abstracts accepted for presentation and discussion will be published on the Association’s website www.easd.org from 1 July 2022.International audienc

    Insulin activates hepatic Wnt/ÎČ- catenin signaling through stearoyl- CoA desaturase 1 and Porcupine

    No full text
    International audienceThe Wnt/ÎČ-catenin pathway plays a pivotal role in liver structural and metabolic homeostasis. Wnt activity is tightly regulated by the acyltransferase Porcupine through the addition of palmitoleate. Interestingly palmitoleate can be endogenously produced by the stearoyl-CoA desaturase 1 (SCD1), a lipogenic enzyme transcriptionally regulated by insulin. This study aimed to determine whether nutritional conditions, and insulin, regulate Wnt pathway activity in liver. An adenoviral TRE-Luciferase reporter was used as a readout of Wnt/ÎČ-catenin pathway activity, in vivo in mouse liver and in vitro in primary hepatocytes. Refeeding enhanced TRE-Luciferase activity and expression of Wnt target genes in mice liver, revealing a nutritional regulation of the Wnt/ÎČ-catenin pathway. This effect was inhibited in liver specific insulin receptor KO (iLIRKO) mice and upon wortmannin or rapamycin treatment. Overexpression or inhibition of SCD1 expression regulated Wnt/ÎČ-catenin activity in primary hepatocytes. Similarly, palmitoleate added exogenously or produced by SCD1-mediated desaturation of palmitate, induced Wnt signaling activity. Interestingly, this effect was abolished in the absence of Porcupine, suggesting that both SCD1 and Porcupine are key mediators of insulin-induced Wnt/ÎČ-catenin activity in hepatocytes. Altogether, our findings suggest that insulin and lipogenesis act as potential novel physiological inducers of hepatic Wnt/ÎČ-catenin pathway

    Regulation of hepatokine gene expression in response to fasting and feeding: Influence of PPARa and insulin-dependent signaling in hepatocytes

    No full text
    International audienceAim.-In hepatocyte, PPARa and insulin receptor (IR) are critical for the transcriptional responses to fasting and feeding, respectively. Here we analyzed the effects of the nutritional status (fasting vs feeding) on the expression of a large panel of hepatokines in hepatocyte-specific PPARa (Ppara hep−/−) and IR (IR hep−/−) null mice. Methods.-Ppara hep−/− , and IR hep−/− mice and their wild-type littermate were subjected to fasting or feeding metabolic challenges, and then analyzed for hepatokine gene expression. Experiments were conducted in mice of both sexes

    O-GlcNacylation Links TxNIP to Inflammasome Activation in Pancreatic ÎČ Cells

    No full text
    International audienceThioredoxin interacting protein (TxNIP), which strongly responds to glucose, has emerged as a central mediator of glucotoxicity in pancreatic ÎČ cells. TxNIP is a scaffold protein interacting with target proteins to inhibit or stimulate their activity. Recent studies reported that high glucose stimulates the interaction of TxNIP with the inflammasome protein NLRP3 (NLR family, pyrin domain containing 3) to increase interleukin-1 ÎČ (IL1ÎČ) secretion by pancreatic ÎČ cells. To better understand the regulation of TxNIP by glucose in pancreatic ÎČ cells, we investigated the implication of O-linked ÎČ-N-acetylglucosamine (O-GlcNAcylation) in regulating TxNIP at the posttranslational level. O-GlcNAcylation of proteins is controlled by two enzymes: the O-GlcNAc transferase (OGT), which transfers a monosaccharide to serine/threonine residues on target proteins, and the O-GlcNAcase (OGA), which removes it. Our study shows that TxNIP is subjected to O-GlcNAcylation in response to high glucose concentrations in ÎČ cell lines. Modification of the O-GlcNAcylation pathway through manipulation of OGT or OGA expression or activity significantly modulates TxNIP O-GlcNAcylation in INS1 832/13 cells. Interestingly, expression and O-GlcNAcylation of TxNIP appeared to be increased in islets of diabetic rodents. At the mechanistic level, the induction of the O-GlcNAcylation pathway in human and rat islets promotes inflammasome activation as evidenced by enhanced cleaved IL1ÎČ. Overexpression of OGT in HEK293 or INS1 832/13 cells stimulates TxNIP and NLRP3 interaction, while reducing TxNIP O-GlcNAcylation through OGA overexpression destabilizes this interaction. Altogether, our study reveals that O-GlcNAcylation represents an important regulatory mechanism for TxNIP activity in ÎČ cells
    corecore