Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 14 de 14
1.
Cell Rep ; 43(4): 114047, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38607916

Using 13C6 glucose labeling coupled to gas chromatography-mass spectrometry and 2D 1H-13C heteronuclear single quantum coherence NMR spectroscopy, we have obtained a comparative high-resolution map of glucose fate underpinning ß cell function. In both mouse and human islets, the contribution of glucose to the tricarboxylic acid (TCA) cycle is similar. Pyruvate fueling of the TCA cycle is primarily mediated by the activity of pyruvate dehydrogenase, with lower flux through pyruvate carboxylase. While the conversion of pyruvate to lactate by lactate dehydrogenase (LDH) can be detected in islets of both species, lactate accumulation is 6-fold higher in human islets. Human islets express LDH, with low-moderate LDHA expression and ß cell-specific LDHB expression. LDHB inhibition amplifies LDHA-dependent lactate generation in mouse and human ß cells and increases basal insulin release. Lastly, cis-instrument Mendelian randomization shows that low LDHB expression levels correlate with elevated fasting insulin in humans. Thus, LDHB limits lactate generation in ß cells to maintain appropriate insulin release.


Insulin Secretion , Insulin-Secreting Cells , L-Lactate Dehydrogenase , Lactic Acid , Humans , Insulin-Secreting Cells/metabolism , Animals , L-Lactate Dehydrogenase/metabolism , Mice , Lactic Acid/metabolism , Glucose/metabolism , Insulin/metabolism , Isoenzymes/metabolism , Citric Acid Cycle , Mice, Inbred C57BL , Male
2.
bioRxiv ; 2023 Sep 17.
Article En | MEDLINE | ID: mdl-37745505

Interferon (IFN)-α is the earliest cytokine signature observed in individuals at risk for type 1 diabetes (T1D), but its effect on the repertoire of HLA Class I (HLA-I)-bound peptides presented by pancreatic ß-cells is unknown. Using immunopeptidomics, we characterized the peptide/HLA-I presentation in in-vitro resting and IFN-α-exposed ß-cells. IFN-α increased HLA-I expression and peptide presentation, including neo-sequences derived from alternative mRNA splicing, post-translational modifications - notably glutathionylation - and protein cis-splicing. This antigenic landscape relied on processing by both the constitutive and immune proteasome. The resting ß-cell immunopeptidome was dominated by HLA-A-restricted ligands. However, IFN-α only marginally upregulated HLA-A and largely favored HLA-B, translating into a major increase in HLA-B-restricted peptides and into an increased activation of HLA-B-restricted vs. HLA-A-restricted CD8+ T-cells. A preferential HLA-B hyper-expression was also observed in the islets of T1D vs. non-diabetic donors, and we identified islet-infiltrating CD8+ T-cells from T1D donors reactive to HLA-B-restricted granule peptides. Thus, the inflammatory milieu of insulitis may skew the autoimmune response toward epitopes presented by HLA-B, hence recruiting a distinct T-cell repertoire that may be relevant to T1D pathogenesis.

3.
Nat Metab ; 5(10): 1673-1684, 2023 Oct.
Article En | MEDLINE | ID: mdl-37709961

The glucagon-like peptide 1 receptor (GLP1R) is a major drug target with several agonists being prescribed in individuals with type 2 diabetes and obesity1,2. The impact of genetic variability of GLP1R on receptor function and its association with metabolic traits are unclear with conflicting reports. Here, we show an unexpected diversity of phenotypes ranging from defective cell surface expression to complete or pathway-specific gain of function (GoF) and loss of function (LoF), after performing a functional profiling of 60 GLP1R variants across four signalling pathways. The defective insulin secretion of GLP1R LoF variants is rescued by allosteric GLP1R ligands or high concentrations of exendin-4/semaglutide in INS-1 823/3 cells. Genetic association studies in 200,000 participants from the UK Biobank show that impaired GLP1R cell surface expression contributes to poor glucose control and increased adiposity with increased glycated haemoglobin A1c and body mass index. This study defines impaired GLP1R cell surface expression as a risk factor for traits associated with type 2 diabetes and obesity and provides potential treatment options for GLP1R LoF variant carriers.


Blood Glucose , Diabetes Mellitus, Type 2 , Humans , Insulin/metabolism , Diabetes Mellitus, Type 2/genetics , Adiposity/genetics , Obesity/genetics
4.
Cells ; 12(1)2022 12 28.
Article En | MEDLINE | ID: mdl-36611907

Type 1 diabetes (T1D) is an auto-immune disease characterized by the progressive destruction of insulin-producing pancreatic beta cells. While beta cells are the target of the immune attack, the other islet endocrine cells, namely the alpha and delta cells, can also be affected by the inflammatory milieu. Here, using a flow cytometry-based strategy, we compared the impact of IFNγ, one of the main cytokines involved in T1D, on the three endocrine cell subsets isolated from C57BL/6 mouse islets. RNA-seq analyses revealed that alpha and delta cells exposed in vitro to IFNγ display a transcriptomic profile very similar to that of beta cells, with an increased expression of inflammation key genes such as MHC class I molecules, the CXCL10 chemokine and the programmed death-ligand 1 (PD-L1), three hallmarks of IFNγ signaling. Interestingly, at low IFNγ concentration, we observed two beta cell populations (responders and non-responders) based on PD-L1 protein expression. Our data indicate that this differential sensitivity relies on the location of the cells within the islet rather than on the existence of two different beta cells subsets. The same findings were corroborated by the in vivo analysis of pancreatic islets from the non-obese diabetic mouse model of T1D, showing more intense PD-L1 staining on endocrine cells close to immune infiltrate. Collectively, our work demonstrates that alpha and delta cells are as sensitive as beta cells to IFNγ, and suggests a gradual diffusion of the cytokine into an islet. These observations provide novel insights into the in situ inflammatory processes occurring in T1D progression.


Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Islets of Langerhans , Mice , Animals , Diabetes Mellitus, Type 1/genetics , B7-H1 Antigen/metabolism , Mice, Inbred C57BL , Islets of Langerhans/metabolism , Insulin-Secreting Cells/metabolism , Interferon-gamma/metabolism , Cytokines/metabolism
5.
Biochem Soc Trans ; 49(2): 645-661, 2021 04 30.
Article En | MEDLINE | ID: mdl-33860789

The MET receptor tyrosine kinase (RTK) and its cognate ligand hepatocyte growth factor (HGF) comprise a signaling axis essential for development, wound healing and tissue homeostasis. Aberrant HGF/MET signaling is a driver of many cancers and contributes to drug resistance to several approved therapeutics targeting other RTKs, making MET itself an important drug target. In RTKs, homeostatic receptor signaling is dependent on autoinhibition in the absence of ligand binding and orchestrated set of conformational changes induced by ligand-mediated receptor dimerization that result in activation of the intracellular kinase domains. A fundamental understanding of these mechanisms in the MET receptor remains incomplete, despite decades of research. This is due in part to the complex structure of the HGF ligand, which remains unknown in its full-length form, and a lack of high-resolution structures of the complete MET extracellular portion in an apo or ligand-bound state. A current view of HGF-dependent MET activation has evolved from biochemical and structural studies of HGF and MET fragments and here we review what these findings have thus far revealed.


Hepatocyte Growth Factor/chemistry , Hepatocyte Growth Factor/metabolism , Protein Domains , Proto-Oncogene Proteins c-met/chemistry , Proto-Oncogene Proteins c-met/metabolism , Animals , Hepatocyte Growth Factor/genetics , Humans , Ligands , Models, Molecular , Mutation , Protein Binding , Proto-Oncogene Proteins c-met/genetics , Signal Transduction/genetics
6.
Diabetologia ; 63(2): 395-409, 2020 02.
Article En | MEDLINE | ID: mdl-31796987

AIMS/HYPOTHESIS: During the onset of type 2 diabetes, excessive dietary intake of saturated NEFA and fructose lead to impaired insulin production and secretion by insulin-producing pancreatic beta cells. The majority of data on the deleterious effects of lipids on functional beta cell mass were obtained either in vivo in rodent models or in vitro using rodent islets and beta cell lines. Translating data from rodent to human beta cells remains challenging. Here, we used the human beta cell line EndoC-ßH1 and analysed its sensitivity to a lipotoxic and glucolipotoxic (high palmitate with or without high glucose) insult, as a way to model human beta cells in a type 2 diabetes environment. METHODS: EndoC-ßH1 cells were exposed to palmitate after knockdown of genes related to saturated NEFA metabolism. We analysed whether and how palmitate induces apoptosis, stress and inflammation and modulates beta cell identity. RESULTS: EndoC-ßH1 cells were insensitive to the deleterious effects of saturated NEFA (palmitate and stearate) unless stearoyl CoA desaturase (SCD) was silenced. SCD was abundantly expressed in EndoC-ßH1 cells, as well as in human islets and human induced pluripotent stem cell-derived beta cells. SCD silencing induced markers of inflammation and endoplasmic reticulum stress and also IAPP mRNA. Treatment with the SCD products oleate or palmitoleate reversed inflammation and endoplasmic reticulum stress. Upon SCD knockdown, palmitate induced expression of dedifferentiation markers such as SOX9, MYC and HES1. Interestingly, SCD knockdown by itself disrupted beta cell identity with a decrease in mature beta cell markers INS, MAFA and SLC30A8 and decreased insulin content and glucose-stimulated insulin secretion. CONCLUSIONS/INTERPRETATION: The present study delineates an important role for SCD in the protection against lipotoxicity and in the maintenance of human beta cell identity. DATA AVAILABILITY: Microarray data and all experimental details that support the findings of this study have been deposited in in the GEO database with the GSE130208 accession code.


Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Palmitic Acid/pharmacology , Stearoyl-CoA Desaturase/metabolism , Apoptosis/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/metabolism , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Insulin Secretion/drug effects , Proto-Oncogene Proteins c-myc/metabolism , SOX9 Transcription Factor/metabolism , Transcription Factor HES-1/metabolism
7.
Nat Immunol ; 19(9): 1035, 2018 Sep.
Article En | MEDLINE | ID: mdl-29880894

In the version of this Article originally published, the asterisks indicating statistical significance were missing from Supplementary Figure 6; the file with the correct figure is now available.

8.
JCI Insight ; 3(3)2018 02 08.
Article En | MEDLINE | ID: mdl-29415896

Type 1 diabetes (T1D) is a chronic disease characterized by an autoimmune-mediated destruction of insulin-producing pancreatic ß cells. Environmental factors such as viruses play an important role in the onset of T1D and interact with predisposing genes. Recent data suggest that viral infection of human islets leads to a decrease in insulin production rather than ß cell death, suggesting loss of ß cell identity. We undertook this study to examine whether viral infection could induce human ß cell dedifferentiation. Using the functional human ß cell line EndoC-ßH1, we demonstrate that polyinosinic-polycytidylic acid (PolyI:C), a synthetic double-stranded RNA that mimics a byproduct of viral replication, induces a decrease in ß cell-specific gene expression. In parallel with this loss, the expression of progenitor-like genes such as SOX9 was activated following PolyI:C treatment or enteroviral infection. SOX9 was induced by the NF-κB pathway and also in a paracrine non-cell-autonomous fashion through the secretion of IFN-α. Lastly, we identified SOX9 targets in human ß cells as potentially new markers of dedifferentiation in T1D. These findings reveal that inflammatory signaling has clear implications in human ß cell dedifferentiation.


Cell Dedifferentiation/immunology , Diabetes Mellitus, Type 1/immunology , Enterovirus Infections/immunology , Insulin-Secreting Cells/physiology , Cell Dedifferentiation/drug effects , Cell Line , Diabetes Mellitus, Type 1/virology , Enterovirus/immunology , Enterovirus Infections/virology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Humans , Interferon Inducers/pharmacology , Interferon-alpha/immunology , Interferon-alpha/metabolism , NF-kappa B/metabolism , Poly I-C/pharmacology , Primary Cell Culture , SOX9 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/immunology
9.
Mol Metab ; 10: 74-86, 2018 04.
Article En | MEDLINE | ID: mdl-29472102

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.


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
10.
Med Eng Phys ; 54: 22-31, 2018 04.
Article En | MEDLINE | ID: mdl-29475589

Orthopedic oscillating saws (OOSs) are widely used for plane processing in orthopedic surgery such as knee and hip replacement. However, sawing has been associated with bone breakthrough and necrosis problems. In this paper, a novel elliptical vibration assisted OOS was designed to achieve a low cutting force under the condition of deepening cut depth and reducing cutting speed, based on the analysis of brittle fractures of the bone and elliptical vibration assisted cutting kinematics. The elliptical vibration was generated using two parallel stacked piezoelectric actuators assembled with the fixture. In order to reduce the large cutting forces due to the large cutting depth, a series of experiments was also conducted to investigate the influence of processing parameters on cutting forces. It was demonstrated that cutting forces are significantly reduced by increasing the vibration frequency and vibration amplitude, and decreasing the sawing speed in the current design. The new design could minimize the cutting forces during sawing and allow surgeons to have better control over the sawing process.


Materials Testing , Mechanical Phenomena , Orthopedic Equipment , Vibration , Animals , Cattle , Equipment Design
11.
Diabetes ; 67(3): 461-472, 2018 03.
Article En | MEDLINE | ID: mdl-29282201

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.


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
12.
Nat Immunol ; 18(12): 1321-1331, 2017 Dec.
Article En | MEDLINE | ID: mdl-28991267

Type 1 diabetes (T1D) is an autoimmune disease that results from the destruction of pancreatic ß-cells by the immune system that involves innate and adaptive immune cells. Mucosal-associated invariant T cells (MAIT cells) are innate-like T-cells that recognize derivatives of precursors of bacterial riboflavin presented by the major histocompatibility complex (MHC) class I-related molecule MR1. Since T1D is associated with modification of the gut microbiota, we investigated MAIT cells in this pathology. In patients with T1D and mice of the non-obese diabetic (NOD) strain, we detected alterations in MAIT cells, including increased production of granzyme B, which occurred before the onset of diabetes. Analysis of NOD mice that were deficient in MR1, and therefore lacked MAIT cells, revealed a loss of gut integrity and increased anti-islet responses associated with exacerbated diabetes. Together our data highlight the role of MAIT cells in the maintenance of gut integrity and the control of anti-islet autoimmune responses. Monitoring of MAIT cells might represent a new biomarker of T1D, while manipulation of these cells might open new therapeutic strategies.


Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class I/analysis , Intestinal Mucosa/immunology , Minor Histocompatibility Antigens/analysis , Mucosal-Associated Invariant T Cells/immunology , Pancreas/immunology , Animals , Cells, Cultured , Gastrointestinal Microbiome/immunology , Granzymes/biosynthesis , Humans , Insulin-Secreting Cells/immunology , Intestinal Mucosa/cytology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Pancreas/cytology
13.
Mol Metab ; 6(4): 340-351, 2017 04.
Article En | MEDLINE | ID: mdl-28377873

OBJECTIVE: In type 2 diabetes (T2D), pancreatic ß cells become progressively dysfunctional, leading to a decline in insulin secretion over time. In this study, we aimed to identify key genes involved in pancreatic beta cell dysfunction by analyzing multiple mouse strains in parallel under metabolic stress. METHODS: Male mice from six commonly used non-diabetic mouse strains were fed a high fat or regular chow diet for three months. Pancreatic islets were extracted and phenotypic measurements were recorded at 2 days, 10 days, 30 days, and 90 days to assess diabetes progression. RNA-Seq was performed on islet tissue at each time-point and integrated with the phenotypic data in a network-based analysis. RESULTS: A module of co-expressed genes was selected for further investigation as it showed the strongest correlation to insulin secretion and oral glucose tolerance phenotypes. One of the predicted network hub genes was Elovl2, encoding Elongase of very long chain fatty acids 2. Elovl2 silencing decreased glucose-stimulated insulin secretion in mouse and human ß cell lines. CONCLUSION: Our results suggest a role for Elovl2 in ensuring normal insulin secretory responses to glucose. Moreover, the large comprehensive dataset and integrative network-based approach provides a new resource to dissect the molecular etiology of ß cell failure under metabolic stress.


Acetyltransferases/genetics , Diabetes Mellitus, Type 2/genetics , Insulin/metabolism , Acetyltransferases/metabolism , Animals , Cell Line , Diabetes Mellitus, Type 2/metabolism , Fatty Acid Elongases , Gene Regulatory Networks , Glucose/metabolism , Humans , Insulin Secretion , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Phenotype
14.
Stem Cells Dev ; 24(15): 1766-78, 2015 Aug 01.
Article En | MEDLINE | ID: mdl-25925840

A comparative analysis of mouse and human pancreatic development may reveal common mechanisms that control key steps as organ morphogenesis and cell proliferation and differentiation. More specifically, understanding beta cell development remains an issue, despite recent progress related to their generation from human embryonic and induced pluripotent stem cells. In this study, we use an integrated approach, including prospective isolation, organ culture, and characterization of intermediate stages, and report that cells from human and mouse fetal pancreas can be expanded in the long term and give rise to hollow duct-like structures in 3D cultures. The expanded cells express a combination of markers (E-cadherin, PDX1, NKX6-1, SOX9, and HNF1ß) that reveals pancreatic progenitor identity. Proliferation of embryonic progenitors was stimulated by the Wnt agonist R-spondin1 (RSPO1), FGF10, and EGF. This combination of growth factors allowed maintaining human fetal pancreatic progenitors in culture for many passages, a finding not reported previously. Importantly, in the absence of EGF, proliferation was reduced, while endocrine differentiation was significantly enhanced. We conclude that modulation of EGF signaling affects in vitro expansion and differentiation of progenitors from embryonic pancreas of both mice and man.


Cell Differentiation/physiology , Cell Proliferation/physiology , Embryonic Stem Cells/metabolism , Epidermal Growth Factor/metabolism , Pancreas/embryology , Signal Transduction/physiology , Animals , Cells, Cultured , Embryonic Stem Cells/cytology , Fetus , Humans , Mice , Pancreas/cytology
...