17 research outputs found

    Reproduction in vitro chez l'homme de la néogenÚse des cellules insulino-sécrétantes (recherche d'une source abondante de cellules précurseurs par transdifférenciation acinocanalaire)

    No full text
    La differentiation in vitro des cellules epitheliales canalaires pancreatiques, considerees comme les cellules souches du pancreas, pourrait etre une nouvelle source importante de cellules pour le traitement du diabete. Afin d'etayer cette hypothese, nous avons isole du pancreas des cellules de phenotype canalaire. Nous avons compare la methode classique d'isolement des cellules canalaires humaines a partir du canal de wirsung, le canal pancreatique principal, soit par culture, soit apres digestion enzymatique avec 2 methodes indirectes : a partir d'ilots humain mis en culture tridimensionnelle en collagene et a partir de culture de tissu exocrine en monocouche. Un nombre limite de cellules canalaires viables a ete obtenu du canal principal. La dissociation des structures kystiques formees a partir des preparations d'ilots cultivees en cqr a permis d'obtenir des cultures de phenotype canalaire. La plus grande quantite de cellules a cependant ete obtenue a partir du tissu exocrine en culture. Considerant la viabilite, l'extrapolation a un pancreas humain entier, cette derniere voie d'obtention permet d'obtenir 1696 52610 6 cellules de phenotype canalaire. Nous avons ensuite montre dans les premiers jours de culture, durant la perte du phenotype exocrine et l'augmentation des marqueurs canalaires, une expression augmentee de la proteine et des arnm de ipf1 (insulin promoter factor 1), respectivement de 3,2 fois et 10,5 fois (n = 5, p<0,001 vs jour 1). Ce facteur de transcription est une proteine homeodomaine qui regule l'ontogenese pancreatique et l'expression de plusieurs genes specifiques des cellules chez l'adulte. Les etudes immunohistochimiques, de proliferation et d'apoptose ont confirme la capacite de transdifferenciation du tissu exocrine. En conclusion, nous pouvons obtenir in vitro, une source abondante de cellules ayant un phenotype epithelial canalaire, considerees comme les cellules souches du pancreas.LILLE1-BU (590092102) / SudocSudocFranceF

    PPARÎł-dependent and -independent effects of Rosiglitazone on lipotoxic human pancreatic islets

    No full text
    International audienceWe explored the in vitro effects of Rosiglitazone (RZG), a PPARgamma agonist, on human pancreatic islet dysfunctions induced by chronic free fatty acid exposure. We demonstrated that RZG beneficial effects on insulin secretion and apoptosis did not imply PDX-1 or insulin gene modulation. It rather involved, through a PPARgamma-dependent mechanism, a reduction of iNOS overexpressed in lipotoxic islets. This reduction likely led to the restoration of ATP level and insulin secretion as well as the decrease in apoptosis. More interestingly, we also demonstrated that RZG beneficial effects involved PPARgamma-independent mechanisms. RZG treatment led to a limitation of oxidative stress exemplified by an increase of GPx and SOD expression. It also increased UCP2 expression that seemed to display antioxidant action in this model. Thus, RZG did not appear to exert a direct action on insulin expression but rather an indirect action on insulin secretion and apoptosis, through PPARgamma-dependent and -independent mechanisms, via regulation of nitrogen and oxygen reactive species injury

    Evidence for Epithelial–Mesenchymal Transition in Adult Human Pancreatic Exocrine Cells

    No full text
    It has been shown that adult pancreatic ductal cells can dedifferentiate and act as pancreatic progenitors. Dedifferentiation of epithelial cells is often associated with the epithelial–mesenchymal transition (EMT). In this study, we investigated the occurrence of EMT in adult human exocrine pancreatic cells both in vitro and in vivo. Cells of exocrine fraction isolated from the pancreas of brain-dead donors were first cultured in suspension for eight days. This led to the formation of spheroids, composed of a principal population of cells with duct-like phenotype. When cultivated in tissue culture-treated flasks, spheroid cells exhibited a proliferative capacity and coexpressed epithelial (cytokeratin7 and cytokeratin19) and mesenchymal (vimentin and α-smooth muscle actin) markers as well as marker of progenitor pancreatic cells (pancreatic duodenal homeobox factor-1) and surface markers of mesenchymal stem cells. The switch from E-cadherin to N-cadherin associated with Snail1 expression suggested that these cells underwent EMT. In addition, we showed coexpression of epithelial and mesenchymal markers in ductal cells of one normal adult pancreas and three type 2 diabetic pancreases. Some of the vimentin-positive cells were found to coexpress glucagon or amylase. These results point to the occurrence of EMT, which may take place on dedifferentiation of ductal cells during the regeneration or renewal of human pancreatic tissues. (J Histochem Cytochem 58:807–823, 2010

    Effects of subtotal pancreatectomy and long‐term glucose and lipid overload on insulin secretion and glucose homeostasis in minipigs

    No full text
    Abstract Introduction Nowadays, there are no strong diabetic pig models, yet they are required for various types of diabetes research. Using cutting‐edge techniques, we attempted to develop a type 2 diabetic minipig model in this study by combining a partial pancreatectomy (Px) with an energetic overload administered either orally or parenterally. Methods Different groups of minipigs, including Göttingen‐like (GL, n = 17) and Ossabaw (O, n = 4), were developed. Prior to and following each intervention, metabolic assessments were conducted. First, the metabolic responses of the Göttingen‐like (n = 3) and Ossabaw (n = 4) strains to a 2‐month High‐Fat, High‐Sucrose diet (HFHSD) were compared. Then, other groups of GL minipigs were established: with a single Px (n = 10), a Px combined with a 2‐month HFHSD (n = 6), and long‐term intraportal glucose and lipid infusions that were either preceded by a Px (n = 4) or not (n = 4). Results After the 2‐month HFHSD, there was no discernible change between the GL and O minipigs. The pancreatectomized group in GL minipigs showed a significantly lower Acute Insulin Response (AIR) (18.3 ± 10.0 IU/mL after Px vs. 34.9 ± 13.7 IU/mL before, p < .0005). In both long‐term intraportal infusion groups, an increase in the Insulinogenic (IGI) and Hepatic Insulin Resistance Indexes (HIRI) was found with a decrease in the AIR, especially in the pancreatectomized group (IGI: 4.2 ± 1.9 after vs. 1.5 ± 0.8 before, p < .05; HIRI (×10−5): 12.6 ± 7.9 after vs. 3.8 ± 4.3 before, p < .05; AIR: 24.4 ± 13.7 ”IU/mL after vs. 43.9 ± 14.5 ”IU/mL before, p < .005). Regardless of the group, there was no fasting hyperglycemia. Conclusions In this study, we used pancreatectomy followed by long‐term intraportal glucose and lipid infusions to develop an original minipig model with metabolic syndrome and early signs of glucose intolerance. We reaffirm the pig's usefulness as a preclinical model for the metabolic syndrome but without the fasting hyperglycemia that characterizes diabetes mellitus

    Influence of preservation solution on human islet isolation outcome

    No full text
    The influence of the preservation solution used for in situ perfusion of the donor and pancreas storage on islet isolation has received little attention

    The challenge of HLA donor specific antibodies in the management of pancreatic islet transplantation: an illustrative case-series

    No full text
    Abstract Islet transplantation is a unique paradigm in organ transplantation, since multiple donors are required to achieve complete insulin-independence. Preformed or de novo Donor Specific Antibodies (DSA) may target one or several donor islets, which adds complexity to the analysis of their impact. Adult patients with type 1 diabetes transplanted with pancreatic islets between 2005 and 2018 were included in a single-center observational study. Thirty-two recipients with available sera tested by solid-phase assays for anti-HLA antibodies during their whole follow-up were analyzed. Twenty-five recipients were islet-transplantation-alone recipients, and 7 islet-after-kidney recipients. Seven recipients presented with DSA at any time during follow-up (two with preformed DSA only, one with preformed and de novo DSA, 4 with de novo DSA only). Only islet-transplantation-alone recipients presented with de novo DSA. Three clinical trajectories were identified according to: 1/the presence of preformed DSA, 2/early de novo DSA or 3/late de novo DSA. Only late de novo DSA were associated with unfavorable outcomes, depicted by a decrease of the ÎČ-score. Islet transplantation with preformed DSA, even with high MFI values, is associated with favorable outcomes in our experience. On the contrary, de novo DSA, and especially late de novo DSA, may be associated with allograft loss

    Interindividual Heterogeneity of SGLT2 Expression and Function in Human Pancreatic Islets.

    No full text
    Studies implicating sodium-glucose cotransporter 2 (SGLT2) inhibitors in glucagon secretion by pancreatic α-cells reported controversial results. We hypothesized that interindividual heterogeneity in SGLT2 expression and regulation may affect glucagon secretion by human α-cells in response to SGLT2 inhibitors. An unbiased RNA-sequencing analysis of 207 donors revealed an unprecedented level of heterogeneity of SLC5A2 expression. To determine heterogeneity of SGLT2 expression at the protein level, the anti-SGLT2 antibody was first rigorously evaluated for specificity, followed by Western blot and immunofluorescence analysis on islets from 10 and 12 donors, respectively. The results revealed a high interdonor variability of SGLT2 protein expression. Quantitative analysis of 665 human islets showed a significant SGLT2 protein colocalization with glucagon but not with insulin or somatostatin. Moreover, glucagon secretion by islets from 31 donors at low glucose (1 mmol/L) was also heterogeneous and correlated with dapagliflozin-induced glucagon secretion at 6 mmol/L glucose. Intriguingly, islets from three donors did not secrete glucagon in response to either 1 mmol/L glucose or dapagliflozin, indicating a functional impairment of the islets of these donors to glucose sensing and SGLT2 inhibition. Collectively, these data suggest that heterogeneous expression of SGLT2 protein and variability in glucagon secretory responses contribute to interindividual differences in response to SGLT2 inhibitors.info:eu-repo/semantics/publishe

    Inhibition of the glucose transporter SGLT2 with dapagliflozin in pancreatic alpha cells triggers glucagon secretion

    No full text
    Type 2 diabetes (T2D) is characterized by chronic hyperglycemia resulting from a deficiency in insulin signaling, because of insulin resistance and/or defects in insulin secretion; it is also associated with increases in glucagon and endogenous glucose production (EGP). Gliflozins, including dapagliflozin, are a new class of approved oral antidiabetic agents that specifically inhibit sodium-glucose co-transporter 2 (SGLT2) function in the kidney, thus preventing renal glucose reabsorption and increasing glycosuria in diabetic individuals while reducing hyperglycemia. However, gliflozin treatment in subjects with T2D increases both plasma glucagon and EGP by unknown mechanisms. In spite of the rise in EGP, T2D patients treated with gliflozin have lower blood glucose levels than those receiving placebo, possibly because of increased glycosuria; however, the resulting increase in plasma glucagon levels represents a possible concerning side effect, especially in a patient population already affected by hyperglucagonemia. Here we demonstrate that SGLT2 is expressed in glucagon-secreting alpha cells of the pancreatic islets. We further found that expression of SLC5A2 (which encodes SGLT2) was lower and glucagon (GCG) gene expression was higher in islets from T2D individuals and in normal islets exposed to chronic hyperglycemia than in islets from non-diabetics. Moreover, hepatocyte nuclear factor 4-α (HNF4A) is specifically expressed in human alpha cells, in which it controls SLC5A2 expression, and its expression is downregulated by hyperglycemia. In addition, inhibition of either SLC5A2 via siRNA-induced gene silencing or SGLT2 via dapagliflozin treatment in human islets triggered glucagon secretion through K ATP channel activation. Finally, we found that dapagliflozin treatment further promotes glucagon secretion and hepatic gluconeogenesis in healthy mice, thereby limiting the decrease of plasma glucose induced by fasting. Collectively, these results identify a heretofore unknown role of SGLT2 and designate dapagliflozin an alpha cell secretagogue.SCOPUS: ar.jinfo:eu-repo/semantics/publishe
    corecore