Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
J Biol Chem ; 298(7): 102096, 2022 07.
Article in English | MEDLINE | ID: mdl-35660019

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is involved in the degradation of the low-density lipoprotein receptor. PCSK9 also targets proteins involved in lipid metabolism (very low-density lipoprotein receptor), immunity (major histocompatibility complex I), and viral infection (cluster of differentiation 81). Recent studies have also indicated that PCSK9 loss-of-function mutations are associated with an increased incidence of diabetes; however, the expression and function of PCSK9 in insulin-producing pancreatic beta cells remain unclear. Here, we studied PCSK9 regulation and function by performing loss- and gain-of-function experiments in the human beta cell line EndoC-ßH1. We demonstrate that PCSK9 is expressed and secreted by EndoC-ßH1 cells. We also found that PCSK9 expression is regulated by cholesterol and sterol regulatory element-binding protein transcription factors, as previously demonstrated in other cell types such as hepatocytes. Importantly, we show that PCSK9 knockdown using siRNA results in deregulation of various elements of the transcriptome, proteome, and secretome, and increases insulin secretion. We also observed that PCSK9 decreases low-density lipoprotein receptor and very low-density lipoprotein receptor levels via an extracellular signaling mechanism involving exogenous PCSK9, as well as levels of cluster of differentiation 36, a fatty acid transporter, through an intracellular signaling mechanism. Finally, we found that PCSK9 regulates the cell surface expression of PDL1 and HLA-ABC, proteins involved in cell-lymphocyte interaction, also via an intracellular mechanism. Collectively, these results highlight PCSK9 as a regulator of multiple cell surface receptors in pancreatic beta cells.


Subject(s)
Insulin-Secreting Cells , Membrane Proteins , Proprotein Convertase 9 , CD36 Antigens/metabolism , Cell Line , Gain of Function Mutation , Humans , Insulin-Secreting Cells/metabolism , Lipoproteins, VLDL/metabolism , Loss of Function Mutation , Membrane Proteins/metabolism , Proprotein Convertase 9/metabolism , Receptors, LDL/metabolism
2.
FASEB J ; 30(2): 748-60, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26534832

ABSTRACT

The major feature of the human pancreatic islet architecture is the organization of endocrine cells into clusters comprising central ß cells and peripheral α cells surrounded by vasculature. To have an insight into the mechanisms that govern this unique islet architecture, islet cells were isolated, and reaggregation of α and ß cells into islet-like structures (pseudoislets) after culture or transplantation into mice was studied by immunohistology. The pseudoislets formed in culture displayed an unusual cell arrangement, contrasting with the transplanted pseudoislets, which exhibited a cell arrangement similar to that observed in native pancreatic islet subunits. The pattern of revascularization and the distribution of extracellular matrix around transplanted pseudoislets were alike to those observed in native pancreatic islets. This organization of transplanted pseudoislets occurred also when revascularization was abolished by treating mice with an anti-VEGF antibody, but not when contact with extracellular matrix was prevented by encapsulation of pseudoislets within alginate hydrogel. These results indicate that the maintenance of islet cell arrangement is dependent on in vivo features such as extracellular matrix but independent of vascularization.


Subject(s)
Extracellular Matrix/pathology , Glucagon-Secreting Cells/pathology , Insulin-Secreting Cells/pathology , Islets of Langerhans Transplantation , Animals , Extracellular Matrix/metabolism , Glucagon-Secreting Cells/metabolism , Heterografts , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, SCID
3.
EBioMedicine ; 95: 104740, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37536063

ABSTRACT

BACKGROUND: To resist the autoimmune attack characteristic of type 1 diabetes, insulin producing pancreatic ß cells need to evade T-cell recognition. Such escape mechanisms may be conferred by low HLA class I (HLA-I) expression and upregulation of immune inhibitory molecules such as Programmed cell Death Ligand 1 (PD-L1). METHODS: The expression of PD-L1, HLA-I and CXCL10 was evaluated in the human ß cell line, ECN90, and in primary human and mouse pancreatic islets. Most genes were determined by real-time RT-PCR, flow cytometry and Western blot. Activator and inhibitor of the AKT signaling were used to modulate PD-L1 induction. Key results were validated by monitoring activity of CD8+ Jurkat T cells presenting ß cell specific T-cell receptor and transduced with reporter genes in contact culture with the human ß cell line, ECN90. FINDINGS: In this study, we identify tryptophan (TRP) as an agonist of PD-L1 induction through the AKT signaling pathway. TRP also synergistically enhanced PD-L1 expression on ß cells exposed to interferon-γ. Conversely, interferon-γ-mediated induction of HLA-I and CXCL10 genes was down-regulated upon TRP treatment. Finally, TRP and its derivatives inhibited the activation of islet-reactive CD8+ T cells by ß cells. INTERPRETATION: Collectively, our findings indicate that TRP could induce immune tolerance to ß cells by promoting their immune evasion through HLA-I downregulation and PD-L1 upregulation. FUNDING: Dutch Diabetes Research Foundation, DON Foundation, the Laboratoire d'Excellence consortium Revive (ANR-10-LABX-0073), Agence Nationale de la Recherche (ANR-19-CE15-0014-01), Fondation pour la Recherche Médicale (EQ U201903007793-EQU20193007831), Innovative Medicines InitiativeINNODIA and INNODIA HARVEST, Aides aux Jeunes Diabetiques (AJD) and Juvenile Diabetes Research Foundation Ltd (JDRF).


Subject(s)
Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Animals , Mice , Humans , Tryptophan , Interferon-gamma/metabolism , Insulin-Secreting Cells/metabolism , Immune Evasion , B7-H1 Antigen , Proto-Oncogene Proteins c-akt
4.
Biochim Biophys Acta Mol Basis Dis ; 1868(11): 166509, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35914653

ABSTRACT

Type 2 diabetes is associated with an inflammatory phenotype in the pancreatic islets. We previously demonstrated that proinflammatory cytokines potently activate the tryptophan/kynurenine pathway (TKP) in INS-1 cells and in normal rat islets. Here we examined: (1) the TKP enzymes expression in the diabetic GK islets; (2) the TKP enzymes expression profiles in the GK islets before and after the onset of diabetes; (3) The glucose-stimulated insulin secretion (GSIS) in vitro in GK islets after KMO knockdown using specific morpholino-oligonucleotides against KMO or KMO blockade using the specific inhibitor Ro618048; (4) The glucose tolerance and GSIS after acute in vivo exposure to Ro618048 in GK rats. We report a remarkable induction of the kmo gene in GK islets and in human islets exposed to proinflammatory conditions. It occurred prominently in beta cells. The increased expression and activity of KMO reflected an acquired adaptation. Both KMO knockdown and specific inhibitor Ro618048 enhanced GSIS in vitro in GK islets. Moreover, acute administration of Ro618048 in vivo improved glucose tolerance, GSIS and basal blood glucose levels in GK rats. These results demonstrate that targeting islet TKP is able to correct defective GSIS. KMO inhibition could represent a potential therapeutic strategy for type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Blood Glucose/metabolism , Cytokines/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Glucose/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/metabolism , Kynurenine/metabolism , Kynurenine 3-Monooxygenase/metabolism , Morpholinos , Rats , Rats, Wistar , Tryptophan/metabolism
5.
Cell Death Dis ; 12(12): 1136, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876563

ABSTRACT

Glucocorticoids (GCs) are widely prescribed for their anti-inflammatory and immunosuppressive properties as a treatment for a variety of diseases. The use of GCs is associated with important side effects, including diabetogenic effects. However, the underlying mechanisms of GC-mediated diabetogenic effects in ß-cells are not well understood. In this study we investigated the role of glycogen synthase kinase 3 (GSK3) in the mediation of ß-cell death and dysfunction induced by GCs. Using genetic and pharmacological approaches we showed that GSK3 is involved in GC-induced ß-cell death and impaired insulin secretion. Further, we unraveled the underlying mechanisms of GC-GSK3 crosstalk. We showed that GSK3 is marginally implicated in the nuclear localization of GC receptor (GR) upon ligand binding. Furthermore, we showed that GSK3 regulates the expression of GR at mRNA and protein levels. Finally, we dissected the proper contribution of each GSK3 isoform and showed that GSK3ß isoform is sufficient to mediate the pro-apoptotic effects of GCs in ß-cells. Collectively, in this work we identified GSK3 as a viable target to mitigate GC deleterious effects in pancreatic ß-cells.


Subject(s)
Glucocorticoids , Glycogen Synthase Kinase 3 , Apoptosis , Cell Death , Glucocorticoids/adverse effects , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta/genetics
6.
Atherosclerosis ; 326: 47-55, 2021 06.
Article in English | MEDLINE | ID: mdl-33933263

ABSTRACT

BACKGROUND AND AIMS: Proprotein Convertase Subtilisin Kexin Type 9 (PCSK9) is an endogenous inhibitor of the LDL receptor (LDLR). Mendelian randomization studies suggest that PCSK9 deficiency increases diabetes risk, but the underlying mechanisms remain unknown. The aim of our study was to investigate whether PCSK9 or its inhibition may modulate beta cell function. METHODS: We assessed PCSK9 and insulin colocalization in human pancreatic sections by epifluorescent and confocal microscopy. We also investigated the expression and the function of PCSK9 in the human EndoC-ßH1 beta cell line, by ELISA and flow cytometry, respectively. PCSK9 was inhibited with Alirocumab or siRNA. LDLR expression and LDL uptake were assessed by flow cytometry. RESULTS: PCSK9 was expressed and secreted from beta cells isolated from human pancreas as well as from EndoC-ßH1 cells. PCSK9 secretion was enhanced by statin treatment. Recombinant PCSK9 decreased LDLR abundance at the surface of these cells, an effect abrogated by Alirocumab. Alirocumab as well as PCSK9 silencing increased LDLR expression at the surface of EndoC-ßH1 cells. Neither exogenous PCSK9, nor Alirocumab, nor PCSK9 silencing significantly altered glucose-stimulated insulin secretion (GSIS) from these cells. High-low density lipoproteins (LDL) concentrations decreased GSIS, but the addition of PCSK9 or its inhibition did not modulate this phenomenon. CONCLUSIONS: While PCSK9 regulates LDLR abundance in beta cells, inhibition of exogenous or endogenous PCSK9 does not appear to significantly impact insulin secretion. This is reassuring for the safety of PCSK9 inhibitors in terms of beta cell function.


Subject(s)
Insulin-Secreting Cells , Proprotein Convertase 9 , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Receptors, LDL , Subtilisins
7.
FASEB J ; 23(12): 4046-55, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19667121

ABSTRACT

Laminin-332 (LN-332) is a basement membrane component known to exert a beneficial effect on rat pancreatic beta cells in vitro. In this work, we analyzed the expression of LN-332 in human islets, its expression after inflammatory insults by cytokines, and the molecular mechanisms responsible for this effect. By Western blotting and RT-PCR, we showed that LN-332 was expressed in isolated human islets. By immunofluorescence on pancreas sections, we observed that labeling was confined to endocrine cells in islets. Confocal microscopy analysis on isolated islet cells revealed that labeling was granular but did not colocalize with hormone secretory granules. LN-332 was most abundant in cultured islets compared to freshly isolated islets and was found in culture medium, which suggests that it was secreted by islets. When islets were exposed to interleukin (IL)-1beta, expression and secretion of LN-332 increased as compared to control. No effect was observed with tumor necrosis factor (TNF)-alpha and interferon (IFN)-gamma. LY294002, an inhibitor of phosphatidylinositol 3-kinase (PI3-K) activity, inhibited culture- and IL-1beta-induced LN-332 expression in islets. These results show that LN-332, known to have some beneficial effect on beta cells in vitro, is produced and secreted by endocrine islet cells and is up-regulated by stressing conditions such as culture and IL-1beta-exposure.


Subject(s)
Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Gene Expression Regulation/physiology , Islets of Langerhans/metabolism , Cells, Cultured , Cytokines/pharmacology , Glucagon/pharmacology , Humans , Insulin/pharmacology , Islets of Langerhans/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Protein Subunits , Signal Transduction , Kalinin
8.
Biochimie ; 170: 26-35, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31838129

ABSTRACT

Type 2 diabetes mellitus is a disease characterized by the formation of amyloid fibrillar deposits consisting mainly in human islet amyloid polypeptide (hIAPP), a peptide co-produced and co-secreted with insulin. hIAPP and insulin are synthesized by pancreatic ß cells initially as prehormones resulting after sequential cleavages in the mature peptides as well as the two flanking peptides (N- and C-terminal) and the C-peptide, respectively. It has been suggested that in the secretory granules, the kinetics of hIAPP fibril formation could be modulated by some internal factors. Indeed, insulin is known to be a potent inhibitor of hIAPP fibril formation and hIAPP-induced cell toxicity. Here we investigate whether the flanking peptides could regulate hIAPP fibril formation and toxicity by combining biophysical and biological approaches. Our data reveal that both flanking peptides are not amyloidogenic. In solution and in the presence of phospholipid membranes, they are not able to totally inhibit hIAPP-fibril formation neither hIAPP-membrane damage. In the presence of INS-1 cells, a rat pancreatic ß-cell line, the flanking peptides do not modulate hIAPP fibrillation neither hIAPP-induced cell death while in the presence of human islets, they have a slightly tendency to reduce hIAPP fibril formation but not its toxicity. These data demonstrate that the flanking peptides do not strongly contribute to reduce mature hIAPP amyloidogenesis in solution and in living cells, suggesting that other biochemical factors present in the cells must act on mature hIAPP fibril formation and hIAPP-induced cell death.


Subject(s)
Amyloid/chemistry , Cell Death , Insulin-Secreting Cells/metabolism , Insulinoma/metabolism , Islet Amyloid Polypeptide/pharmacology , Pancreatic Neoplasms/metabolism , Secretory Vesicles/metabolism , Amino Acid Sequence , Amylin Receptor Agonists/pharmacology , Amyloid/drug effects , Animals , Cells, Cultured , Humans , Insulin/metabolism , Insulin-Secreting Cells/drug effects , Insulinoma/drug therapy , Insulinoma/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Rats , Secretory Vesicles/drug effects , Secretory Vesicles/pathology
9.
Sci Rep ; 10(1): 13469, 2020 08 10.
Article in English | MEDLINE | ID: mdl-32778664

ABSTRACT

G protein-coupled receptors are seven transmembrane signaling molecules that are involved in a wide variety of physiological processes. They constitute a large protein family of receptors with almost 300 members detected in human pancreatic islet preparations. However, the functional role of these receptors in pancreatic islets is unknown in most cases. We generated a new stable human beta cell line from neonatal pancreas. This cell line, named ECN90 expresses both subunits (GABBR1 and GABBR2) of the metabotropic GABAB receptor compared to human islet. In ECN90 cells, baclofen, a specific GABAB receptor agonist, inhibits cAMP signaling causing decreased expression of beta cell-specific genes such as MAFA and PCSK1, and reduced insulin secretion. We next demonstrated that in primary human islets, GABBR2 mRNA expression is strongly induced under cAMP signaling, while GABBR1 mRNA is constitutively expressed. We also found that induction and activation of the GABAB receptor in human islets modulates insulin secretion.


Subject(s)
Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Receptors, GABA-B/genetics , Baclofen/pharmacology , Cell Line , GABA-B Receptor Agonists/pharmacology , Humans , Insulin/metabolism , Insulin Secretion , Insulin-Secreting Cells/physiology , Islets of Langerhans/physiology , Pancreas/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, GABA-B/metabolism , Signal Transduction , gamma-Aminobutyric Acid/metabolism
10.
Stem Cell Res Ther ; 11(1): 158, 2020 04 17.
Article in English | MEDLINE | ID: mdl-32303252

ABSTRACT

BACKGROUND: Mesenchymal stromal cells (MSCs) represent an interesting tool to improve pancreatic islet transplantation. They have immunomodulatory properties and secrete supportive proteins. However, the functional properties of MSCs vary according to many factors such as donor characteristics, tissue origin, or isolation methods. To counteract this heterogeneity, we aimed to immortalize and characterize adherent cells derived from human pancreatic islets (hISCs), using phenotypic, transcriptomic, and functional analysis. METHODS: Adherent cells derived from human islets in culture were infected with a hTERT retrovirus vector and then characterized by microarray hybridization, flow cytometry analysis, and immunofluorescence assays. Osteogenic, adipogenic, and chondrogenic differentiation as well as PBMC proliferation suppression assays were used to compare the functional abilities of hISCs and MSCs. Extracellular matrix (ECM) gene expression profile analysis was performed using the SAM (Significance Analysis of Microarrays) software, and protein expression was confirmed by western blotting. RESULTS: hISCs kept an unlimited proliferative potential. They exhibited several properties of MSCs such as CD73, CD90, and CD105 expression and differentiation capacity. From a functional point of view, hISCs inhibited the proliferation of activated peripheral blood mononuclear cells. The transcriptomic profile of hISCs highly clusterized with bone marrow (BM)-MSCs and revealed a differential enrichment of genes involved in the organization of the ECM. Indeed, the expression and secretion profiles of ECM proteins including collagens I, IV, and VI, fibronectin, and laminins, known to be expressed in abundance around and within the islets, were different between hISCs and BM-MSCs. CONCLUSION: We generated a new human cell line from pancreatic islets, with MSCs properties and retaining some pancreatic specificities related to the production of ECM proteins. hISCs appear as a very promising tool in islet transplantation by their availability (as a source of inexhaustible source of cells) and ability to secrete a supportive "pancreatic" microenvironment.


Subject(s)
Islets of Langerhans , Mesenchymal Stem Cells , Bone Marrow Cells , Cell Differentiation , Cell Proliferation , Cells, Cultured , Chondrogenesis , Humans , Leukocytes, Mononuclear
12.
J Endocrinol ; 244(1): 133-148, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31600727

ABSTRACT

Islet inflammation is associated with defective ß cell function and mass in type 2 diabetes (T2D). Glycogen synthase kinase 3 (GSK3) has been identified as an important regulator of inflammation in different diseased conditions. However, the role of GSK3 in islet inflammation in the context of diabetes remains unexplored. In this study, we investigated the direct implication of GSK3 in islet inflammation in vitro and tested the impact of GSK3 inhibition in vivo, on the reduction of islet inflammation, and the improvement of glucose metabolism in the Goto-Kakizaki (GK) rat, a spontaneous model of T2D. GK rats were chronically treated with infra-therapeutic doses of lithium, a widely used inhibitor of GSK3. We analyzed parameters of glucose homeostasis as well as islet inflammation and fibrosis in the endocrine pancreas. Ex vivo, we tested the impact of GSK3 inhibition on the autonomous inflammatory response of non-diabetic rat and human islets, exposed to a mix of pro-inflammatory cytokines to mimic an inflammatory environment. Treatment of young GK rats with lithium prevented the development of overt diabetes. Lithium treatment resulted in reduced expression of pro-inflammatory cytokines in the islets. It decreased islet fibrosis and partially restored the glucose-induced insulin secretion in GK rats. Studies in non-diabetic human and rat islets exposed to inflammatory environment revealed the direct implication of GSK3 in the islet autonomous inflammatory response. We show for the first time, the implication of GSK3 in islet inflammation and suggest this enzyme as a viable target to treat diabetes-associated inflammation.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glycogen Synthase Kinase 3/metabolism , Islets of Langerhans/metabolism , Animals , Disease Models, Animal , Fibrosis , Glucose/metabolism , Humans , Inflammation , Insulin Secretion , Male , Rats , Rats, Wistar
13.
Nat Commun ; 11(1): 5982, 2020 11 25.
Article in English | MEDLINE | ID: mdl-33239617

ABSTRACT

Expanding the mass of pancreatic insulin-producing beta cells through re-activation of beta cell replication has been proposed as a therapy to prevent or delay the appearance of diabetes. Pancreatic beta cells exhibit an age-dependent decrease in their proliferative activity, partly related to changes in the systemic environment. Here we report the identification of CCN4/Wisp1 as a circulating factor more abundant in pre-weaning than in adult mice. We show that Wisp1 promotes endogenous and transplanted adult beta cell proliferation in vivo. We validate these findings using isolated mouse and human islets and find that the beta cell trophic effect of Wisp1 is dependent on Akt signaling. In summary, our study reveals the role of Wisp1 as an inducer of beta cell replication, supporting the idea that the use of young blood factors may be a useful strategy to expand adult beta cell mass.


Subject(s)
Aging/physiology , CCN Intercellular Signaling Proteins/metabolism , Insulin-Secreting Cells/physiology , Islets of Langerhans Transplantation/methods , Proto-Oncogene Proteins/metabolism , Aging/blood , Animals , CCN Intercellular Signaling Proteins/blood , CCN Intercellular Signaling Proteins/genetics , Cell Proliferation , Cells, Cultured , Culture Media/metabolism , Diabetes Mellitus/therapy , Female , Humans , Insulin-Secreting Cells/transplantation , Male , Mice , Mice, Knockout , Primary Cell Culture/methods , Proto-Oncogene Proteins/blood , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/physiology , Weaning
14.
Transpl Int ; 22(9): 931-9, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19453995

ABSTRACT

We explore herein the effect of TNF-related activation-induced cytokine (TRANCE) co-stimulatory pathway blockade on islet survival after allograft transplantation. Expression of TRANCE on murine C57Bl/6 (B6) CD4+ T cells after allogeneic activation was analyzed by fluorescence-activated cell sorter (FACS). The effect of a TRANCE receptor fusion protein (TR-Fc) and anti-CD154 antibody (MR1) on B6 spleen cell proliferation after allogeneic activation was assessed by mixed lymphocyte reaction (MLR). Three groups of B6 mice were transplanted with allogeneic islets (DBA2): Control; short-term TR-Fc-treatment (days 0-4); and prolonged TR-Fc-treatment (days -1 to 13). Donor-specific transfusion (DST) was performed at the time of islet transplantation in one independent experiment. Transplantectomy samples were analyzed by immunohistochemistry. TRANCE expression was upregulated in stimulated CD4+ T cells in vitro. In MLR experiments, TR-Fc and MR1 both reduced spleen cell proliferation, but less than the combination of both molecules. Short-course TR-Fc treatment did not prolong islet graft survival when compared with controls (10.6 +/- 1.9 vs. 10.7 +/- 1.5 days) in contrast to prolonged treatment (20.7 +/- 3.2 days; P < 0.05). After DST, primary non function (PNF) was observed in half of control mice, but never in TR-Fc-treated mice. Immunofluorescence staining for Mac-1 showed a clear decrease in macrophage recruitment in the treated groups. TRANCE-targeting may be an effective strategy for the prolongation of allogeneic islet graft survival, thanks to its inhibitory effects on co-stimulatory signals and macrophage recruitment.


Subject(s)
Islets of Langerhans Transplantation/methods , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , Animals , CD4-Positive T-Lymphocytes/immunology , CD40 Ligand/biosynthesis , Cell Separation , Flow Cytometry , Graft Survival , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Biological , Spleen/cytology , Transplantation, Homologous
15.
Diabetes ; 67(3): 461-472, 2018 03.
Article in English | MEDLINE | ID: mdl-29282201

ABSTRACT

Although the mechanisms by which glucose regulates insulin secretion from pancreatic ß-cells are now well described, the way glucose modulates gene expression in such cells needs more understanding. Here, we demonstrate that MondoA, but not its paralog carbohydrate-responsive element-binding protein, is the predominant glucose-responsive transcription factor in human pancreatic ß-EndoC-ßH1 cells and in human islets. In high-glucose conditions, MondoA shuttles to the nucleus where it is required for the induction of the glucose-responsive genes arrestin domain-containing protein 4 (ARRDC4) and thioredoxin interacting protein (TXNIP), the latter being a protein strongly linked to ß-cell dysfunction and diabetes. Importantly, increasing cAMP signaling in human ß-cells, using forskolin or the glucagon-like peptide 1 mimetic Exendin-4, inhibits the shuttling of MondoA and potently inhibits TXNIP and ARRDC4 expression. Furthermore, we demonstrate that silencing MondoA expression improves glucose uptake in EndoC-ßH1 cells. These results highlight MondoA as a novel target in ß-cells that coordinates transcriptional response to elevated glucose levels.


Subject(s)
Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Gene Expression Regulation , Glucose/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Second Messenger Systems , Active Transport, Cell Nucleus/drug effects , Animals , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cyclic AMP/metabolism , Exenatide , Gene Expression Regulation/drug effects , Humans , Incretins/pharmacology , Insulin Secretion , Insulin-Secreting Cells/drug effects , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Peptides/pharmacology , RNA Interference , Second Messenger Systems/drug effects , Thioredoxins/genetics , Thioredoxins/metabolism , Tissue Culture Techniques , Venoms/pharmacology
16.
Mol Metab ; 10: 74-86, 2018 04.
Article in English | MEDLINE | ID: mdl-29472102

ABSTRACT

OBJECTIVE: Dedifferentiation could explain reduced functional pancreatic ß-cell mass in type 2 diabetes (T2D). METHODS: Here we model human ß-cell dedifferentiation using growth factor stimulation in the human ß-cell line, EndoC-ßH1, and human pancreatic islets. RESULTS: Fibroblast growth factor 2 (FGF2) treatment reduced expression of ß-cell markers, (INS, MAFB, SLC2A2, SLC30A8, and GCK) and activated ectopic expression of MYC, HES1, SOX9, and NEUROG3. FGF2-induced dedifferentiation was time- and dose-dependent and reversible upon wash-out. Furthermore, FGF2 treatment induced expression of TNFRSF11B, a decoy receptor for RANKL and protected ß-cells against RANKL signaling. Finally, analyses of transcriptomic data revealed increased FGF2 expression in ductal, endothelial, and stellate cells in pancreas from T2D patients, whereas FGFR1, SOX,9 and HES1 expression increased in islets from T2D patients. CONCLUSIONS: We thus developed an FGF2-induced model of human ß-cell dedifferentiation, identified new markers of dedifferentiation, and found evidence for increased pancreatic FGF2, FGFR1, and ß-cell dedifferentiation in T2D.


Subject(s)
Cell Dedifferentiation , Diabetes Mellitus, Type 2/metabolism , Insulin-Secreting Cells/cytology , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Fibroblast Growth Factor 2/pharmacology , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Osteoprotegerin/genetics , Osteoprotegerin/metabolism , RANK Ligand/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
17.
Cell Metab ; 28(6): 946-960.e6, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30078552

ABSTRACT

Although CD8+ T-cell-mediated autoimmune ß cell destruction occurs in type 1 diabetes (T1D), the target epitopes processed and presented by ß cells are unknown. To identify them, we combined peptidomics and transcriptomics strategies. Inflammatory cytokines increased peptide presentation in vitro, paralleling upregulation of human leukocyte antigen (HLA) class I expression. Peptide sources featured several insulin granule proteins and all known ß cell antigens, barring islet-specific glucose-6-phosphatase catalytic subunit-related protein. Preproinsulin yielded HLA-A2-restricted epitopes previously described. Secretogranin V and its mRNA splice isoform SCG5-009, proconvertase-2, urocortin-3, the insulin gene enhancer protein ISL-1, and an islet amyloid polypeptide transpeptidation product emerged as antigens processed into HLA-A2-restricted epitopes, which, as those already described, were recognized by circulating naive CD8+ T cells in T1D and healthy donors and by pancreas-infiltrating cells in T1D donors. This peptidome opens new avenues to understand antigen processing by ß cells and for the development of T cell biomarkers and tolerogenic vaccination strategies.


Subject(s)
Antigen Presentation , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Transcriptome/immunology , Animals , Biomarkers/metabolism , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cell Line , Corticotropin-Releasing Hormone/metabolism , Cytokines/metabolism , HLA Antigens/metabolism , Humans , Insulin/metabolism , Islet Amyloid Polypeptide/metabolism , Mice , Neuroendocrine Secretory Protein 7B2/metabolism , Proprotein Convertase 2/metabolism , Protein Precursors/metabolism , Proteomics/methods , Urocortins/metabolism
18.
Diabetes ; 55(5): 1413-20, 2006 May.
Article in English | MEDLINE | ID: mdl-16644699

ABSTRACT

When attached on a matrix produced by a rat bladder carcinoma cell line (804G matrix), rat pancreatic beta-cells spread in response to glucose and secrete more insulin compared with cells attached on poly-l-lysine. The aim of this study was to determine whether laminin-5 and its corresponding cell receptor beta1 integrin are implicated in these phenomena. By using specific blocking antibodies, we demonstrated that laminin-5 is the component present in 804G matrix responsible for the effect of 804G matrix on beta-cell function and spreading. When expression of two well-known laminin-5 ligands, beta1 and beta4 integrin, was assessed by Western blot and RT-PCR, only the beta1 integrin was detected in beta-cells. Anti-beta1 integrin antibody reduced the spreading of beta-cells on 804G matrix. Blockade of the interaction between beta1 integrins and laminin-5 resulted in a reduction in glucose-stimulated insulin secretion. Blocking anti-beta1 integrin antibody also inhibited focal adhesion kinase phosphorylation induced by 804G matrix. In conclusion, anti-beta1 integrin and -laminin-5 antibodies interfere with spreading of beta-cells, resulting in decreased insulin secretion in response to glucose. Our findings indicate that outside-in signaling via engagement of beta1 integrins by laminin-5 is an important component of normal beta-cell function.


Subject(s)
Cell Adhesion/physiology , Extracellular Matrix/physiology , Insulin/metabolism , Integrin beta1/physiology , Islets of Langerhans/physiology , Laminin/antagonists & inhibitors , Animals , Antibodies/pharmacology , Base Sequence , Cell Adhesion/drug effects , Cell Line , Cell Movement/physiology , Cells, Cultured , DNA Primers , Extracellular Matrix/drug effects , Hemolytic Plaque Technique , Insulin Secretion , Integrin beta1/drug effects , Integrin beta1/genetics , Integrin beta4/genetics , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Laminin/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction
19.
Sci Rep ; 6: 25765, 2016 05 11.
Article in English | MEDLINE | ID: mdl-27166427

ABSTRACT

Acute or chronic metabolic complications such as diabetic ketoacidosis are often associated with extracellular acidification and pancreatic ß-cell dysfunction. However, the mechanisms by which human ß-cells sense and respond to acidic pH remain elusive. In this study, using the recently developed human ß-cell line EndoC-ßH2, we demonstrate that ß-cells respond to extracellular acidification through GPR68, which is the predominant proton sensing receptor of human ß-cells. Using gain- and loss-of-function studies, we provide evidence that the ß-cell enriched transcription factor RFX6 is a major regulator of GPR68. Further, we show that acidic pH stimulates the production and secretion of the chemokine IL-8 by ß-cells through NF-кB activation. Blocking of GPR68 or NF-кB activity severely attenuated acidification induced IL-8 production. Thus, we provide mechanistic insights into GPR68 mediated ß-cell response to acidic microenvironment, which could be a new target to protect ß-cell against acidosis induced inflammation.


Subject(s)
Acids/metabolism , Extracellular Space/chemistry , Insulin-Secreting Cells/metabolism , Interleukin-8/biosynthesis , Receptors, G-Protein-Coupled/metabolism , Cell Line , Cyclic AMP/biosynthesis , Humans , Hydrogen-Ion Concentration , Inflammation Mediators/metabolism , Inositol Phosphates/metabolism , NF-kappa B/metabolism , Neutrophils/metabolism , Protons , Regulatory Factor X Transcription Factors/metabolism
20.
Transplantation ; 79(3): 353-5, 2005 Feb 15.
Article in English | MEDLINE | ID: mdl-15699768

ABSTRACT

The aim of our study was to assess cell trafficking and early events after intraportal islet transplantation. Sprague-Dawley rat islets were incubated for various times, with various concentrations of 2-[F]fluoro-2deoxy-D-glucose (FDG), and in presence of various glucose concentrations. FDG-labeled syngeneic islets or FDG alone were injected in rats. Radioactivity was measured in the liver and in various organs by positron-emission tomography for 6 hours. FDG uptake increased with incubation time or FDG concentration and decreased in presence of glucose. In vivo, all islets implanted in the liver, with an uptake 4.4 times higher than controls (44.2% vs. 10.1%, P=0.02). Radioactivity in the liver decreased at the same rate after injection of labeled-islets and FDG alone. Ex vivo labeling of islets and imaging of posttransplant early events were feasible. Islets engrafted exclusively in the liver. No islet loss could be demonstrated 6 hours after transplantation.


Subject(s)
Islets of Langerhans Transplantation/physiology , Islets of Langerhans/diagnostic imaging , Positron-Emission Tomography , Animals , Fluorodeoxyglucose F18/pharmacokinetics , Male , Radiopharmaceuticals/pharmacokinetics , Rats , Rats, Sprague-Dawley , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL