222 results on '"Julie Kerr-Conte"'
Search Results
2. β-cell specific E2f1 deficiency impairs glucose homeostasis, β-cell identity and insulin secretion
- Author
-
Jean-Sébastien Annicotte, Patrick Collombat, Amélie Bonnefond, Benoit Pourcet, Philippe Froguel, François Pattou, Julie Kerr-Conte, Lluis Fajas, Patrick Meffre, Zohra Benfodda, Pierre-Damien Denechaud, Sarah Anissa Hannou, Etienne Blanc, Mehdi Derhourhi, Lionel Berberian, Souhila Amanzougarene, Emmanuelle Durand, Nabil Rabhi, Charlène Carney, Maeva Moreno, Xavier Gromada, Laure Rolland, Emilie Courty, Marika Elsa Friano, Cyril Bourouh, and Frédérik Oger
- Abstract
The loss of pancreatic β-cell identity emerges as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cell-autonomous role of the cell cycle regulator and transcription factor E2F1 in the maintenance of β-cell identity, insulin secretion and glucose homeostasis. We show that the β-cell-specific loss of E2f1 function in mice triggers glucose intolerance associated with defective insulin secretion, an altered endocrine cell mass, a downregulation of many β-cell genes and a concomitant increase of non-β-cell markers. Mechanistically, the epigenomic profiling of promoters of these non-β-cell upregulated genes identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, promoters of downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that specific E2f1 transcriptional, cistromic and epigenomic signatures are associated with these β-cell dysfunctions, with E2F1 directly regulating several β-cell genes at the chromatin level. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of β-cell identity genes. Our data suggest that E2F1 is critical for maintaining β-cell identity and function through a sustained control of β-cell and non β-cell transcriptional programs.
- Published
- 2023
- Full Text
- View/download PDF
3. β-cell specific E2f1 deficiency impairs glucose homeostasis, β-cell identity and insulin secretion
- Author
-
Frédérik Oger, Cyril Bourouh, Marika Elsa Friano, Emilie Courty, Laure Rolland, Xavier Gromada, Maeva Moreno, Charlène Carney, Nabil Rabhi, Emmanuelle Durand, Souhila Amanzougarene, Lionel Berberian, Mehdi Derhourhi, Etienne Blanc, Sarah Anissa Hannou, Pierre-Damien Denechaud, Zohra Benfodda, Patrick Meffre, Lluis Fajas, Julie Kerr-Conte, François Pattou, Philippe Froguel, Benoit Pourcet, Amélie Bonnefond, Patrick Collombat, and Jean-Sébastien Annicotte
- Subjects
Endocrinology, Diabetes and Metabolism ,Internal Medicine - Abstract
The loss of pancreatic β-cell identity emerges as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cellautonomous role of the cell cycle regulator and transcription factor E2F1 in the maintenance of β-cell identity, insulin secretion and glucose homeostasis. We show that the β-cell-specific loss of E2f1 function in mice triggers glucose intolerance associated with defective insulin secretion, an altered endocrine cell mass, a downregulation of many β-cell genes and a concomitant increase of non-β-cell markers. Mechanistically, the epigenomic profiling of promoters of these non-β-cell upregulated genes identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, promoters of downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that specific E2f1 transcriptional, cistromic and epigenomic signatures are associated with these β-cell dysfunctions, with E2F1 directly regulating several β-cell genes at the chromatin level. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of β-cell identity genes. Our data suggest that E2F1 is critical for maintaining β-cell identity and function through a sustained control of β-cell and non β-cell transcriptional programs.
- Published
- 2023
- Full Text
- View/download PDF
4. Advancing multi-day ex vivo kidney perfusion using spatially resolved metabolomics
- Author
-
Marlon J.A. de Haan, Franca M.R. Witjas, Annemarie M.A. de Graaf, Marleen E. Jacobs, Elena Sánchez-López, Sarantos Kostidis, Martin Giera, Mehdi Maanaoui, Thomas Hubert, Julie Kerr-Conte, François Pattou, Dorottya K. de Vries, Jesper Kers, Ian P.J. Alwayn, Cees van Kooten, Bram P.A.M. Heijs, Gangqi Wang, Marten A. Engelse, and Ton J. Rabelink
- Abstract
The ability to preserve metabolically active kidneys ex vivo for multiple days may permit reconditioning, repair and regeneration of deceased donor kidneys. However, the kidneys high metabolic demand limits its functional preservation. Current approaches focus on normothermic machine perfusion (NMP) at 37°C or hypothermic machine perfusion (HMP) at 4-8°C. At normothermia, kidneys are metabolically active butex vivopreservation is limited to hours. During hypothermia kidneys can be preserved up to 24 hours but are metabolically inactive and suffer cold-induced injury. Therefore, we revisited sub normothermic perfusion (at 25°C) as an alternative approach to preserve human kidneys in a metabolically active state for extended periods of time.In a custom-made platform that includes a cell-free perfusate enriched with TCA cycle fuels, urine recirculation, and continuous hemofiltration we perfused discarded human kidneys up to 8 days. Using spatially resolved single cell resolution isotope tracing we demonstrate active metabolism in all the different renal cell types over this period. However, beyond 4 days cell composition of nephron segments assessed with spatial lipidomics changed substantially and injury markers such as NGAL and LDH increased in the perfusate. Up to 4 days, perfused human discarded donor kidneys maintained metabolic fluxes, functional parameters and allow for reperfusion using a porcine auto transplantation model. These data underpin that extended multi-day metabolic preservation of human kidneys is achievable using a sub normothermic perfusion platform.
- Published
- 2023
- Full Text
- View/download PDF
5. Effects of subtotal pancreatectomy and long‐term glucose and lipid overload on insulin secretion and glucose homeostasis in minipigs
- Author
-
Rébecca Goutchtat, Audrey Quenon, Manon Clarisse, Nathalie Delalleau, Anaïs Coddeville, Mathilde Gobert, Valéry Gmyr, Julie Kerr‐Conte, François Pattou, and Thomas Hubert
- Subjects
Endocrinology, Diabetes and Metabolism - Published
- 2023
- Full Text
- View/download PDF
6. High-Resolution Glucose Fate-Mapping Reveals LDHB-Dependent Lactate Production by Human Pancreatic Islets
- Author
-
Federica Cuozzo, Katrina Viloria, Daniela Nasteska, Zicong Jiao, Hannah R. Smith, Caroline Bonner, Julie Kerr-Conte, Francois Pattou, Rita Nano, Lorenzo Piemonti, Jennie Roberts, Gareth G. Lavery, Anne Clark, Carlo Ceresa, Leanne Hodson, Amy Davies, Guy Rutter, Ildem Akerman, Daniel A. Tennant, Christian Ludwig, and David Hodson
- Published
- 2023
- Full Text
- View/download PDF
7. Pharmacological HDAC Inhibition Impairs Pancreatic β-Cell Function Through an Epigenome-Wide Reprogramming
- Author
-
Frederik Oger, Maeva Moreno, Mehdi Derhourhi, Bryan Thiroux, Lionel Berberian, Cyril Bourouh, Emmanuelle Durand, Souhila Amanzougarene, Alaa Badreddine, Etienne Blanc, Olivier Molendi-Coste, Laurent Pineau, Gianni Pasquetti, Laure Rolland, Charlene Carney, Florine Bornaque, Emilie Courty, Céline Gheeraert, Jerome Eeckhoute, David Dombrowicz, Julie Kerr-Conte, Francois Pattou, Bart Staels, Phillipe Froguel, Amélie Bonnefond, and Jean-Sebastien Annicotte
- Published
- 2023
- Full Text
- View/download PDF
8. Oral metformin transiently lowers post-prandial glucose response by reducing the apical expression of sodium-glucose co-transporter 1 in enterocytes
- Author
-
Lorea Zubiaga, Olivier Briand, Florent Auger, Veronique Touche, Thomas Hubert, Julien Thevenet, Camille Marciniak, Audrey Quenon, Caroline Bonner, Simon Peschard, Violeta Raverdy, Mehdi Daoudi, Julie Kerr-Conte, Gianni Pasquetti, Hermann Koepsell, Daniela Zdzieblo, Markus Mühlemann, Bernard Thorens, Nathalie D. Delzenne, Laure B. Bindels, Benoit Deprez, Marie C. Vantyghem, Blandine Laferrère, Bart Staels, Damien Huglo, Sophie Lestavel, François Pattou, and UCL - SSS/LDRI - Louvain Drug Research Institute
- Subjects
Multidisciplinary ,Endocrinology ,Drugs ,Molecular physiology - Abstract
Metformin (MET) is the most prescribed antidiabetic drug, but its mechanisms of action remain elusive. Recent data point to the gut as MET's primary target. Here, we explored the effect of MET on the gut glucose transport machinery. Using human enterocytes (Caco-2/TC7 cells) , we showed that MET transiently reduced the apical density of sodium-glucose transporter 1 (SGLT1) and decreased the absorption of glucose, without changes in the mRNA levels of the transporter. Administered 1 h before a glucose challenge in rats (Wistar, GK), C57BL6 mice and mice pigs, oral MET reduced the post-prandial glucose response (PGR). This effect was abrogated in SGLT1-KO mice. MET also reduced the luminal clearance of 2-(F)-fluoro-2-deoxy-D-glucose after oral administration in rats. In conclusion, oral metformin transiently lowers post-prandial glucose response by reducing the apical expression of SGLT1 in enterocytes, which may contribute to the clinical effects of the drug.
- Published
- 2023
9. Epigenetics ofPNLIPRP1in human pancreas reveals a molecular path between type 2 diabetes and pancreatic cancer
- Author
-
Lucas Maurin, Lorella Marselli, Lijiao Ning, Mathilde Boissel, Raphael Boutry, Mara Suleiman, Audrey Leloire, Vincent Pascat, Jared Maina, Bénédicte Toussaint, Souhila Amanzougarene, Alaa Badreddine, Mehdi Derhourhi, Inga Prokopenko, Anne Jörns, Sigurd Lenzen, François Pattou, Julie Kerr-Conte, Mickaël Canouil, Amélie Bonnefond, Piero Marchetti, Philippe Froguel, and Amna Khamis
- Abstract
BackgroundType 2 diabetes (T2D) increases the risk of pancreatic ductal adenocarcinoma (PDAC), which could be due to an epigenetic mechanism.MethodsWe explored the association between T2D and whole pancreas methylation in 141 individuals, of which 28 had T2D, using Illumina MethylationEPIC 850K BeadChip arrays. We performed downstream functional assessment in the rat acinar pancreas cell line AR42J. To further understand the role of our candidate gene in humans, we tested whether null variants were associated with T2D and related traits using the UK biobank.ResultsMethylation analysis identified one significant CpG associated with T2D: hypermethylation in an enhancer inPNLIPRP1, an acinar-specific gene.PNLIPRP1expression was decreased in T2D individuals. Using a rat acinar cell line, we 1/ confirmed decreasedPnliprp1in response to a diabetogenic treatment, and 2/ inPnliprp1knockdown, an up-regulation of cholesterol biosynthesis, cell cycle down-regulation, decreased expression of acinar markers and increased expression of ductal markers pointing towards acinar-to-ductal metaplasia (ADM), a hallmark of PDAC initiation. Using exome data from UK Biobank, we show that rarePNLIPRP1null variants associated with increased glucose, BMI and LDL-cholesterol.Conclusions/interpretationWe present evidence that an epigenetically-regulated gene associates with T2D risk, and might promote ADM and PDAC progression, opening new insights into early prevention of PDAC.
- Published
- 2022
- Full Text
- View/download PDF
10. High-resolution glucose fate-mapping revealsLDHB-dependent lactate production by human pancreatic β cells
- Author
-
Federica Cuozzo, Daniela Nasteska, Zicong Jiao, Hannah R. Smith, Caroline Bonner, Julie Kerr-Conte, Francois Pattou, Rita Nano, Lorenzo Piemonti, Jennie Roberts, Gareth G. Lavery, Ildem Akerman, Daniel A. Tennant, Christian Ludwig, and David J. Hodson
- Abstract
Using13C6glucose labeling coupled to GC-MS and 2D1H-13C HSQC NMR spectroscopy, we have obtained a comparative high-resolution map of glucose fate underpinning steady state insulin release and β cell function. In both mouse and human islets, the contribution of glucose to the TCA cycle is similar. Pyruvate-fueling of the TCA cycle is found to be mediated primarily by the activity of pyruvate dehydrogenase (PDH), with only a limited contribution from pyruvate carboxylase (PC). While conversion of pyruvate to lactate by lactate dehydrogenase (LDH) can be detected in both species, lactate accumulation via this route is six-fold higher in human islets. Transcriptomic analysis reveals that human β cells specifically express lactate dehydrogenase B (LDHB) at high levels, in keeping with the phenotype of patients harboring gain-of-function mutations in MCT1/ SLC16A1 (HHF7). Thus, glycolytically-derived acetyl CoA preferentially feeds the TCA cycle in both mouse and human β cells. However, human β cells possess the machinery needed to generate extra-mitochondrial lactate, which might reflect a key mechanism to balance the reducing activity of NADH-producing pathways.
- Published
- 2022
- Full Text
- View/download PDF
11. The HDAC inhibitor trichostatin A impairs pancreatic β-cell function through an epigenome-wide reprogramming
- Author
-
Frédérik Oger, Maeva Moreno, Mehdi Derhourhi, Bryan Thiroux, Lionel Berberian, Cyril Bourouh, Emmanuelle Durand, Souhila Amanzougarene, Alaa Badreddine, Etienne Blanc, Olivier Molendi-Coste, Laurent Pineau, Gianni Pasquetti, Laure Rolland, Charlène Carney, Florine Bornaque, Emilie Courty, Céline Gheeraert, Jérôme Eeckhoute, David Dombrowicz, Julie Kerr-Conte, François Pattou, Bart Staels, Philippe Froguel, Amélie Bonnefond, and Jean-Sébastien Annicotte
- Abstract
ObjectiveThe pancreatic islets of Langerhans contain distinct cell subtypes including insulin-producing β cells. Although their cell-specific gene expression pattern defines their identity, the underlying molecular network driving this transcriptional specificity is not fully understood. Among the numerous transcriptional regulators, histone deacetylases (HDAC) enzymes are potent chromatin modifiers which directly regulate gene expression through deacetylation of lysine residues within specific histone proteins. The precise molecular mechanisms underlying HDAC effects on cellular plasticity and β-cell identity are currently unknown.MethodsThe pharmacological inhibition of HDAC activity by trichostatin A (TSA) was studied in the mouse Min6 and human EndocBH1 cell lines, as well as primary mouse sorted β cells and human pancreatic islets. The molecular and functional effects of treating these complementary β-cell models with TSA was explored at the epigenomic and transcriptomic level through next-generation sequencing of chromatin immunoprecipitation (ChIP) assays (ChIP-seq) and RNA sequencing (RNA-seq) experiments, respectively.ResultsWe showed that TSA alters insulin secretion associated with β-cell specific transcriptome programming in both mouse and human β-cell lines, as well as on human pancreatic islets. We also demonstrated that this alternative β-cell transcriptional program in response to HDAC inhibition is related to an epigenome-wide remodeling at both promoters and enhancers.ConclusionsTaken together, our data indicate that full HDAC activity is required to safeguard the epigenome, to protect against loss of β-cell identity with unsuitable expression of genes associated with alternative cell fates.
- Published
- 2022
- Full Text
- View/download PDF
12. The challenge of HLA donor specific antibodies in the management of pancreatic islet transplantation: an illustrative case-series
- Author
-
Mehdi Maanaoui, Mikael Chetboun, Isabelle Top, Vincent Elsermans, Julie Kerr-Conte, Kristell Le Mapihan, Frederique Defrance, Valéry Gmyr, Thomas Hubert, Myriam Labalette, Marc Hazzan, Marie-Christine Vantyghem, and François Pattou
- Subjects
Adult ,Graft Rejection ,Multidisciplinary ,HLA Antigens ,Isoantibodies ,Graft Survival ,Islets of Langerhans Transplantation ,Humans ,Tissue Donors ,Retrospective Studies - 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 β-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.
- Published
- 2022
- Full Text
- View/download PDF
13. Pancreatic islet transplantation under the kidney capsule of mice: model of refinement for molecular and ex-vivo graft analysis
- Author
-
Valery Gmyr, Julie Kerr-Conte, Nathalie Delalleau, François Pattou, and Julien Thevenet
- Subjects
0303 health sciences ,geography ,geography.geographical_feature_category ,General Veterinary ,business.industry ,030209 endocrinology & metabolism ,Islet ,Bioinformatics ,medicine.disease ,Cell therapy ,Transplantation ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,In vivo ,Diabetes mellitus ,Medicine ,Animal Science and Zoology ,Pancreatic islet transplantation ,business ,Pancreas ,Ex vivo ,030304 developmental biology - Abstract
Diabetes cell therapy by human islet transplantation can restore an endogenous insulin secretion and normal glycaemic control in type 1 diabetic patients for as long as 10 years post transplantation. Before transplantation, each clinical islet preparation undergoes extensive in-vitro and in-vivo quality controls. The in-vivo quality control assay consists of transplanting human islets under the kidney capsule of immunocompromised mice. Currently, it is considered the best predictive factor to qualify clinical transplant efficiency. This chimeric model offers a wide area of study since it combines the possibility of producing not only quantitative but also a maximum of qualitative data. Today’s technological advances allow us to obtain more accurate and stronger data from the animals used in research while ensuring their comfort and well-being throughout the protocol, including cage enrichment and pain treatment during and after surgery. As demonstrated in this valuable model, we are able to generate more usable results (Refine), while reducing the number of animals used (Reduce), by focusing on the development of ex-vivo analysis techniques (Replace), which clearly highlights the Burch and Russell 3Rs concept.
- Published
- 2021
- Full Text
- View/download PDF
14. 1921–2021: From insulin discovery to islet transplantation in type 1 diabetes
- Author
-
Mikael Chetboun, Marie-Christine Vantyghem, Julie Kerr-Conte, François Pattou, and Arnaud Jannin
- Subjects
endocrine system ,medicine.medical_specialty ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,medicine.medical_treatment ,Islets of Langerhans Transplantation ,030209 endocrinology & metabolism ,Glycemic Control ,History, 21st Century ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Internal medicine ,medicine ,Humans ,Insulin ,Secretion ,Type 1 diabetes ,Kidney ,geography ,geography.geographical_feature_category ,C-Peptide ,business.industry ,Immunosuppression ,General Medicine ,History, 20th Century ,medicine.disease ,Islet ,Transplantation ,Diabetes Mellitus, Type 1 ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,France ,business ,Allotransplantation - Abstract
One century after the discovery of insulin, the French Health regulations have just authorized the reimbursement for islet transplantation. Intraportal islet allotransplantation from a pancreatic donor is indicated in patients with type 1 diabetes (T1D) complicated with lability or hypoglycemia unawareness, or in case of a functioning kidney graft; islet auto-transplantation may be indicated after pancreatic surgery.Compared with insulin even administered in closed-loop pumps, the specificity of islet allotransplantation is the restoration of C-peptide secretion. Long-term insulin-independence is observed when the engrafted islet mass is sufficient, at the cost of immunosuppression. Fewer low-glucose events and less glucose variability, are observed even with minimal functional islet graft, after islet transplantation as at onset of T1D, when a residual C-peptide secretion is maintained, an objective currently approached with less aggressive immuno-modulating therapies than in the past. Therefore, restoration or preservation of endogen insulin secretion is an important goal, allowing to maintain a long-term glucose balance with more than 70% of time in range 3.9-10mmol/L and less than 3% of time3.9mmol/L, thus reducing the occurrence of diabetic complications. In the clinical setting, - the preservation of C-peptide at early stage of T1D, - the use of technological ressources (multi-injections, sensors, insulin pump, closed-loop systems) at later stages, - and islet transplantation when hypoglycemia awareness becomes impaired are complementary for a personalized care all along the life of T1D patients.
- Published
- 2021
- Full Text
- View/download PDF
15. Thymidylate Synthase O-GlcNAcylation: a molecular mechanism of 5-FU sensitization in colorectal cancer
- Author
-
Julie Kerr-Conte, Julien Thevenet, Adeline Page, Vanessa Dehennaut, Céline Schulz, Gérard Vergoten, Ninon Very, Tony Lefebvre, Ikram El Yazidi-Belkoura, Amélie Decourcelle, Madjid Djouina, Stéphan Hardivillé, Unité de Glycobiologie Structurale et Fonctionnelle - UMR 8576 (UGSF), Université de Lille-Centre National de la Recherche Scientifique (CNRS), Cancer Heterogeneity, Plasticity and Resistance to Therapies - UMR 9020 - U 1277 (CANTHER), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille)-Centre National de la Recherche Scientifique (CNRS), Recherche translationnelle sur le diabète - U 1190 (RTD), Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Institute for Translational Research in Inflammation - U 1286 (INFINITE (Ex-Liric)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), SFR Biosciences, École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Université de Lille, École normale supérieure - Lyon (ENS Lyon)-Université Claude Bernard Lyon 1 (UCBL), EL YAZIDI-BELKOURA, IKRAM, CNRS, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF) - UMR 8576, Institut de Recherche Translationnelle sur l'Inflammation (INFINITE) - U1286, Cancer Heterogeneity, Plasticity and Resistance to Therapies - UMR 9020 - U 1277 [CANTHER], Recherche translationnelle sur le diabète (RTD) - U1190, Cancer Heterogeneity, Plasticity and Resistance to Therapies (CANTHER) - UMR 9020 - UMR 1277, Unité de Glycobiologie Structurale et Fonctionnelle UMR 8576 (UGSF), Lille Inflammation Research International Center - U 995 (LIRIC), and Miniaturisation pour la Synthèse, l’Analyse et la Protéomique - UAR 3290 (MSAP)
- Subjects
Cancer Research ,Gene knockdown ,Colorectal cancer ,[SDV]Life Sciences [q-bio] ,Thymidylate Synthase ,Biology ,medicine.disease ,medicine.disease_cause ,Thymidylate synthase ,In vitro ,[SDV] Life Sciences [q-bio] ,medicine.anatomical_structure ,In vivo ,Tumor progression ,Genetics ,medicine ,biology.protein ,Cancer research ,Carcinogenesis ,Molecular Biology ,Sensitization - Abstract
International audience; Alteration of O-GlcNAcylation, a dynamic post-translational modification, is associated with tumorigenesis and tumor progression. Its role in chemotherapy response is poorly investigated. Standard treatment for colorectal cancer (CRC), 5-fluorouracil (5-FU), mainly targets Thymidylate Synthase (TS). TS O-GlcNAcylation was reported but not investigated yet. We hypothesize that O-GlcNAcylation interferes with 5-FU CRC sensitivity by regulating TS. In vivo, we observed that combined 5-FU with Thiamet-G (O-GlcNAcase (OGA) inhibitor) treatment had a synergistic inhibitory effect on grade and tumor progression. 5-FU decreased O-GlcNAcylation and, reciprocally, elevation of O-GlcNAcylation was associated with TS increase. In vitro in non-cancerous and cancerous colon cells, we showed that 5-FU impacts O-GlcNAcylation by decreasing O-GlcNAc Transferase (OGT) expression both at mRNA and protein levels. Reciprocally, OGT knockdown decreased 5-FU-induced cancer cell apoptosis by reducing TS protein level and activity. Mass spectrometry, mutagenesis and structural studies mapped O-GlcNAcylated sites on T251 and T306 residues and deciphered their role in TS proteasomal degradation. We reveal a crosstalk between O-GlcNAcylation and 5-FU metabolism in vitro and in vivo that converges to 5-FU CRC sensitization by stabilizing TS. Overall, our data propose that combining 5-FU-based chemotherapy with Thiamet-G could be a new way to enhance CRC response to 5-FU.
- Published
- 2021
- Full Text
- View/download PDF
16. Genetic regulation of RNA splicing in human pancreatic islets
- Author
-
François Pattou, Anthony Beucher, Julie Kerr-Conte, Ignasi Moran, Anna L. Gloyn, Cuenca-Ardura M, Jorge Ferrer, Javier García-Hurtado, Mara Suleiman, Koning EJPd, Thierry Berney, Irimia M, Lorenzo Piemonti, Goutham Atla, Rashmi B. Prasad, Bonas-Guarch S, and Lorella Marselli
- Subjects
Genetics ,endocrine system ,geography ,geography.geographical_feature_category ,endocrine system diseases ,Effector ,Pancreatic islets ,Biology ,Islet ,medicine.anatomical_structure ,Genetic variation ,RNA splicing ,medicine ,Transcriptional regulation ,Gene ,Proinsulin - Abstract
Genetic variants that influence transcriptional regulation in pancreatic islets play a major role in the susceptibility to type 2 diabetes (T2D). For many susceptibility loci, however, the mechanisms are unknown. We examined splicing QTLs (sQTLs) in islets from 399 donors and observed that genetic variation has a widespread influence on splicing of genes with important functions in islet biology. In parallel, we profiled expression QTLs, and used transcriptome-wide association and co-localization studies to assign islet sQTLs or eQTLs to T2D susceptibility signals that lacked candidate effector genes. We found novel T2D associations, including an sQTL that creates a nonsense isoform in ERO1B, a regulator of ER-stress and proinsulin biosynthesis. The expanded list of T2D risk effectors revealed overrepresented pathways, including regulators of G-protein-mediated cAMP production. This data exposes an underappreciated layer of genetic regulation in pancreatic islets, and nominates molecular mediators of T2D susceptibility.
- Published
- 2021
- Full Text
- View/download PDF
17. Pushing the boundaries of organs before it’s too late: pre‐emptive regeneration
- Author
-
Mehdi Maanaoui and Julie Kerr-Conte
- Subjects
Transplantation ,Decellularization ,business.industry ,Regeneration (biology) ,Cell ,Mesenchymal stem cell ,Mesenchymal Stem Cells ,Organ Transplantation ,medicine.disease ,Cell biology ,Cell therapy ,medicine.anatomical_structure ,Mitochondrial biogenesis ,Fibrosis ,Regeneration ,Medicine ,business - Abstract
Solid organ transplantation is marked by accelerated aging and inexorable fibrosis. It is crucial to promote strategies to attenuate, or to reverse, damage before organ failure. Hence, the objective of this article is to provide insight into strategies, which aim to regenerate or rejuvenate the transplanted organs. Cell therapy with mesenchymal stromal cells is currently under investigation because of their antifibrotic properties. Their ability to promote mitochondrial biogenesis, and to transfer mitochondria to wounded cells, is another approach to boost the organ regeneration. Other teams have investigated bioengineered organs, which consists of decellularization of the damaged organ followed by recellularization. Lastly, the development of CAR-T cell-based technologies may revolutionize the field of transplantation, as recent preclinical studies showed that CAR-T cells could efficiently clear senescent cells from an organ and reverse fibrosis. Ultimately, these cutting-edge strategies may bring the holy grail of a pre-emptive regenerated organ closer to reality.
- Published
- 2021
- Full Text
- View/download PDF
18. REST is a major negative regulator of endocrine differentiation during pancreas organogenesis
- Author
-
Jose Luis Mosquera, Meritxell Rovira, François Pattou, Julie Kerr-Conte, Goutham Atla, Vane Grau, Jorge Ferrer, Miguel A. Maestro, Yasuhiro Yamada, Javier García-Hurtado, Maria Maqueda, Wellcome Trust, Medical Research Council (MRC), and Imperial College Healthcare NHS Trust- BRC Funding
- Subjects
PROMOTES ,Molecular biology ,Organogenesis ,Cellular differentiation ,SILENCING TRANSCRIPTION FACTOR ,Regulator ,Enteroendocrine cell ,MOUSE ,Mice ,0302 clinical medicine ,Endocrinologia ,pancreas ,Zebrafish ,11 Medical and Health Sciences ,Genetics & Heredity ,0303 health sciences ,PROGENITORS ,REST ,Gene Expression Regulation, Developmental ,Cell Differentiation ,EXPANSION ,β cells ,3. Good health ,Cell biology ,17 Psychology and Cognitive Sciences ,endocrine differentiation ,medicine.anatomical_structure ,Knockout mouse ,Pancreas ,Life Sciences & Biomedicine ,STEM-CELLS ,transcriptional repressors ,Research Paper ,EXPRESSION ,Biology ,Pàncrees ,03 medical and health sciences ,Organogènesi ,BETA-CELLS ,Genetics ,medicine ,TARGET GENES ,Animals ,Endocrine system ,Transcription factor ,Rest (music) ,Biologia molecular ,030304 developmental biology ,Science & Technology ,Cell Biology ,IN-VITRO ,06 Biological Sciences ,biology.organism_classification ,Embryonic stem cell ,bipotent progenitors ,beta cells ,pancreas development ,Genètica ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Multiple transcription factors have been shown to promote pancreatic β-cell differentiation, yet much less is known about negative regulators. Earlier epigenomic studies suggested that the transcriptional repressor REST could be a suppressor of endocrinogenesis in the embryonic pancreas. However, pancreatic Rest knockout mice failed to show abnormal numbers of endocrine cells, suggesting that REST is not a major regulator of endocrine differentiation. Using a different conditional allele that enables profound REST inactivation, we observed a marked increase in pancreatic endocrine cell formation. REST inhibition also promoted endocrinogenesis in zebrafish and mouse early postnatal ducts and induced β-cell-specific genes in human adult duct-derived organoids. We also defined genomic sites that are bound and repressed by REST in the embryonic pancreas. Our findings show that REST-dependent inhibition ensures a balanced production of endocrine cells from embryonic pancreatic progenitors. This research was supported by Ministerio de Ciencia, Innovación y Universidades (SAF2015-73226-JIN [Agencia Estatal de Investigación {AEI}/European Regional Development Fund, European Union {UE}] and RYC-2017-21950 [AEI/European Social Fund, UE] to M.R., and BFU2014-54284-R and RTI2018-095666-B-I00 to J.F.); the Medical Research Council (MR/L02036X/1), Wellcome Trust (WT101033), and European Research Council Advanced Grant (789055) (to J.F.); the Instituto de Salud Carlos III (CA18/00045 to J.L.M.); and a Spanish Ministry of Science, Innovation, and Universities (MCIU) fellowship (PTA2018-016371-I to M.M.). J.K.-C.'s and F.P.'s research was supported by L'Agence Nationale de la Recherche (ANR) grants, L'Institut Européen de Génomique du Diabète (EGID), ANR-10-LABX-0046, a French state fund managed by ANR under the frame program Investissements d'Avenir (I-SITE ULNE/ANR-16-IDEX-0004 ULNE to F.P.), and the European Consortium for Islet Transplantation funded by the Juvenile Diabetes Research Foundation International. Work in the Centre for Genomic Regulation was supported by the Centres de Recerca de Catalunya (CERCA) Programme, Generalitat de Catalunya, and Centro de Excelencia Severo Ochoa (SEV-2015-0510). Work at Institut d'Investigació Biomèdica de Bellvitge was supported by the CERCA Programme and Generalitat de Catalunya
- Published
- 2021
19. Insulin-producing organoids engineered from islet and amniotic epithelial cells to treat diabetes
- Author
-
Charles H Wassmer, Thierry Berney, David Goodman, Kevin Bellofatto, Ekaterine Berishvili, Vakhtang Kalandadze, Vanessa Lavallard, Antonia Follenzi, Romain Bonnet, Fanny Lebreton, Domenico Bosco, Michel Boulvain, Lisa Perez, and Julie Kerr-Conte
- Subjects
0301 basic medicine ,endocrine system diseases ,Angiogenesis ,medicine.medical_treatment ,Cell ,Islets of Langerhans Transplantation ,Insulins ,General Physics and Astronomy ,Mice, SCID ,Rats, Sprague-Dawley ,Tissue Culture Techniques ,surgery ,Mice ,Endocrinology ,0302 clinical medicine ,organ replacement ,Insulin ,lcsh:Science ,Multidisciplinary ,geography.geographical_feature_category ,ddc:618 ,diabetes ,ddc:617 ,General Commentary ,Graft Survival ,Islet ,Organoids ,medicine.anatomical_structure ,surgical procedures, operative ,Amniotic epithelial cells ,Regenerative medicine ,Heterografts ,medicine.symptom ,Organoids / transplantation ,Epithelial Cells / metabolism ,endocrine system ,Cell Survival ,Science ,Transplantation, Heterologous ,Inflammation ,Tissue Engineering / methods ,Streptozocin ,General Biochemistry, Genetics and Molecular Biology ,Article ,Diabetes Mellitus, Experimental ,03 medical and health sciences ,Islets of Langerhans ,stem cells ,Spheroids, Cellular ,medicine ,Organoid ,Diabetes Mellitus ,Animals ,Humans ,Tissue engineering ,Amnion ,geography ,business.industry ,Epithelial Cells ,General Chemistry ,Rats ,Transplantation ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Diabetes Mellitus, Type 1 / therapy ,Cancer research ,lcsh:Q ,business ,030217 neurology & neurosurgery ,Islets of Langerhans Transplantation / methods - Abstract
Maintaining long-term euglycemia after intraportal islet transplantation is hampered by the considerable islet loss in the peri-transplant period attributed to inflammation, ischemia and poor angiogenesis. Here, we show that viable and functional islet organoids can be successfully generated from dissociated islet cells (ICs) and human amniotic epithelial cells (hAECs). Incorporation of hAECs into islet organoids markedly enhances engraftment, viability and graft function in a mouse type 1 diabetes model. Our results demonstrate that the integration of hAECs into islet cell organoids has great potential in the development of cell-based therapies for type 1 diabetes. Engineering of functional mini-organs using this strategy will allow the exploration of more favorable implantation sites, and can be expanded to unlimited (stem-cell-derived or xenogeneic) sources of insulin-producing cells., Islet transplantation is a feasible approach to treat type I diabetes, however inflammation and poor vascularisation impair long-term engraftment. Here the authors show that incorporating human amniotic epithelial cells into islet organoids improves engraftment and function of organoids, through enhanced revascularisation.
- Published
- 2019
- Full Text
- View/download PDF
20. The transcription factor E2F1 controls the GLP-1 receptor pathway in pancreatic β cells
- Author
-
Cyril Bourouh, Emilie Courty, Laure Rolland, Gianni Pasquetti, Xavier Gromada, Nabil Rabhi, Charlène Carney, Maeva Moreno, Raphaël Boutry, Emilie Caron, Zohra Benfodda, Patrick Meffre, Julie Kerr-Conte, François Pattou, Philippe Froguel, Amélie Bonnefond, Frédérik Oger, Jean-Sébastien Annicotte, Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP), Université de Lille, Détection, évaluation, gestion des risques CHROniques et éMErgents (CHROME) / Université de Nîmes (CHROME), Université de Nîmes (UNIMES), Institut des Biomolécules Max Mousseron [Pôle Chimie Balard] (IBMM), Institut de Chimie du CNRS (INC)-Centre National de la Recherche Scientifique (CNRS)-Université de Montpellier (UM)-Ecole Nationale Supérieure de Chimie de Montpellier (ENSCM), Université de Montpellier (UM), Institut de Recherche en Cancérologie de Montpellier (IRCM - U1194 Inserm - UM), CRLCC Val d'Aurelle - Paul Lamarque-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM), ANR-17-CE14-0034,BETAPLASTICITY,Identification du rôle du facteur de transcription E2F1 dans l'identité et la plasticité de la cellule bétâ pancréatique(2017), and ANR-16-IDEX-0004,ULNE,ULNE(2016)
- Subjects
Glucose ,Diabetes Mellitus, Type 2 ,[SDV]Life Sciences [q-bio] ,Insulin-Secreting Cells ,Exenatide ,Humans ,Insulin ,[SDV.BBM.BM]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Molecular biology ,General Biochemistry, Genetics and Molecular Biology ,E2F1 Transcription Factor ,Glucagon-Like Peptide-1 Receptor - Abstract
The glucagon-like peptide 1 (Glp-1) has emerged as a hormone with broad pharmacological potential in type 2 diabetes (T2D) treatment, notably by improving β cell functions. The cell-cycle regulator and transcription factor E2f1 is involved in glucose homeostasis by modulating β cell mass and function. Here, we report that β cell-specific genetic ablation of E2f1 (E2f1
- Published
- 2021
- Full Text
- View/download PDF
21. A crosstalk between E2F1 and GLP-1 signaling pathways modulates insulin secretion
- Author
-
François Pattou, Patrick Meffre, Zohra Benfodda, Frédérik Oger, Gianni Pasquetti, Xavier Gromada, Julie Kerr-Conte, Amélie Bonnefond, Emilie Courty, Charlène Carney, Laure Rolland, Cyril Bourouh, Philippe Froguel, Emilie Caron, Jean-Sébastien Annicotte, Maeva Moreno, Raphael Boutry, and Nabil Rabhi
- Subjects
endocrine system ,medicine.medical_specialty ,biology ,Chemistry ,Pancreatic islets ,Retinoblastoma protein ,Incretin ,Glucagon-like peptide-1 ,Crosstalk (biology) ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,medicine ,biology.protein ,Glucose homeostasis ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,E2F - Abstract
Compromised β-cell function contributes to type 2 diabetes (T2D) development. The glucagon like peptide 1 (Glp-1) has emerged as a hormone with broad pharmacological potential toward T2D treatment, notably by improving β-cell functions. Recent data have shown that the transcription factor E2f1, besides its role as a cell cycle regulator, is involved in glucose homeostasis by modulating β-cell mass, function and identity. Here, we demonstrate a crosstalk between the E2F1, phosphorylation of retinoblastoma protein (pRb) and Glp-1 signaling pathways. We found that β-cell specificE2f1deficient mice (E2f1β−/−) presented with impaired glucose homeostasis and decreased glucose stimulated-insulin secretion mediated by exendin 4 (i.e., GLP1R agonist), which were associated with decreased expression ofGlp1rencoding Glp-1 receptor (GLP1R) inE2f1β−/−pancreatic islets. Decreasing E2F1 transcriptional activity with an E2F inhibitor in islets from nondiabetic humans decreasedGLP1Rlevels and blunted the incretin effect of exendin 4 on insulin secretion. Conversely, overexpressingE2f1in pancreatic β cells increasedGlp1rexpression associated with enhanced insulin secretion mediated by GLP1R agonist. Interestingly, kinome analysis of mouse islets demonstrated that an acute treatment with exendin 4 increased pRb phosphorylation and subsequent E2f1 transcriptional activity. This study suggests a molecular crosstalk between the E2F1/pRb and GLP1R signaling pathways that modulates insulin secretion and glucose homeostasis.
- Published
- 2021
- Full Text
- View/download PDF
22. Pancreatic islet transplantation under the kidney capsule of mice: model of refinement for molecular and
- Author
-
Julien, Thévenet, Valery, Gmyr, Nathalie, Delalleau, François, Pattou, and Julie, Kerr-Conte
- Subjects
Islets of Langerhans ,Mice ,Islets of Langerhans Transplantation ,Animals ,Humans ,Insulin ,Kidney - Abstract
Diabetes cell therapy by human islet transplantation can restore an endogenous insulin secretion and normal glycaemic control in type 1 diabetic patients for as long as 10 years post transplantation. Before transplantation, each clinical islet preparation undergoes extensive
- Published
- 2021
23. A human antibody against pathologic IAPP aggregates protects beta cells in type 2 diabetes
- Author
-
Karin Breu, Heather Denroche, Melania Osto, C. B. Verchere, François Pattou, Christoph Hock, Jan Grimm, Fabrice Heitz, Julien Thevenet, Ioana Combaluzier, Fabian Wirth, Paolo Arosio, Thomas A. Lutz, Christine Seeger, Roger M. Nitsch, Julie Kerr-Conte, and Marc Y. Donath
- Subjects
endocrine system ,Text mining ,biology ,business.industry ,biology.protein ,Cancer research ,Medicine ,Type 2 diabetes ,Antibody ,business ,Beta (finance) ,medicine.disease - Abstract
In patients with type 2 diabetes, pancreatic beta cells progressively degenerate and gradually lose their ability to produce insulin and regulate blood glucose. Beta cell dysfunction and loss is associated with an accumulation of aggregated forms of islet amyloid polypeptide (IAPP) consisting of soluble prefibrillar IAPP oligomers as well as insoluble IAPP fibrils in pancreatic islets. Here, we describe a novel human monoclonal antibody selectively targeting IAPP oligomers and neutralizing IAPP aggregate toxicity by preventing membrane disruption and apoptosis in vitro. Antibody treatment in rats and mice transgenic for human IAPP, and human islet-engrafted mouse models of type 2 diabetes triggered clearance of IAPP oligomers resulting in beta cell protection and improved glucose control. These results provide new evidence for the pathological role of IAPP oligomers and suggest that antibody-mediated removal of IAPP oligomers could be a pharmaceutical strategy to support beta cell function in type 2 diabetes.
- Published
- 2021
- Full Text
- View/download PDF
24. A Crosstalk between E2F1 and GLP-1 Signaling Pathways Modulates Insulin Secretion
- Author
-
Cyril Bourouh, Emilie Courty, Gianni Pasquetti, Xavier Gromada, Nabil Rabhi, Charlene Carney, Maeva Moreno, Raphael Boutry, Laure Rolland, Emilie Caron, Zohra Benfodda, Patrick Meffre, Julie Kerr-Conte, Francois Pattou, Phillipe Froguel, Amélie Bonnefond, Frederik Oger, and Jean-Sebastien Annicotte
- Published
- 2021
- Full Text
- View/download PDF
25. The Map3k12 (Dlk)/JNK3 signaling pathway is required for pancreatic beta-cell proliferation during postnatal development
- Author
-
Mathie Tenenbaum, Amar Abderrahmani, Amélie Bonnefond, Stéphane Dalle, Julien Bourry, Valery Gmyr, Philippe Froguel, Valérie Pawlowski, Nicole Beeler, Valérie Plaisance, Raphael Boutry, Hélène Ezanno, Clara Sanchez-Parra, Cécile Jacovetti, Romano Regazzi, Gianni Pasquetti, François Pattou, Syu-Ichi Hirai, Julie Kerr-Conte, Metabolic functional (epi)genomics and molecular mechanisms involved in type 2 diabetes and related diseases - UMR 8199 - UMR 1283 (EGENODIA (GI3M)), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre National de la Recherche Scientifique (CNRS), Institut d’Électronique, de Microélectronique et de Nanotechnologie - UMR 8520 (IEMN), Centrale Lille-Université de Lille-Centre National de la Recherche Scientifique (CNRS)-Université Polytechnique Hauts-de-France (UPHF)-JUNIA (JUNIA), Université catholique de Lille (UCL)-Université catholique de Lille (UCL), NanoBioInterfaces - IEMN (NBI - IEMN), Université catholique de Lille (UCL)-Université catholique de Lille (UCL)-Centrale Lille-Université de Lille-Centre National de la Recherche Scientifique (CNRS)-Université Polytechnique Hauts-de-France (UPHF)-JUNIA (JUNIA), Université de Lausanne = University of Lausanne (UNIL), Recherche translationnelle sur le diabète - U 1190 (RTD), Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Institut de Génomique Fonctionnelle (IGF), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS), Wakayama University, Imperial College London, This work was supported by grants from 'Société Francophone du Diabète (SFD)', 'European Genomic Institute for Diabetes' (E.G.I.D., ANR-10-LABEX-46) and European Commission, European Research Council (GEPIDIAB 294785 to P.F.), and by a grant from the Swiss National Science Foundation (310030-169480 to RR). Human islets were provided by the European Consortium for Islet Transplantation, funded by the Juvenile Diabetes Research Foundation International., ANR-10-LABX-0046,EGID,EGID Diabetes Pole(2010), European Project: 294785,EC:FP7:ERC,ERC-2011-ADG_20110310,GEPIDIAB(2012), Metabolic functional (epi)genomics and molecular mechanisms involved in type 2 diabetes and related diseases - UMR 8199 - UMR 1283 (GI3M), Université de Lausanne (UNIL), and Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
MAPK/ERK pathway ,endocrine system ,Biology ,Mapk ,Beta-cell mass ,Transcriptome ,Rats, Sprague-Dawley ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Mice ,[SPI]Engineering Sciences [physics] ,Mitogen-Activated Protein Kinase 10 ,Pregnancy ,Insulin-Secreting Cells ,Animals ,Cell Proliferation/drug effects ,Cyclin D1/genetics ,Cyclin D1/metabolism ,Cyclin D2/genetics ,Cyclin D2/metabolism ,Female ,Glucose/pharmacology ,Humans ,Insulin/metabolism ,Insulin-Secreting Cells/cytology ,Insulin-Secreting Cells/metabolism ,MAP Kinase Kinase Kinases/antagonists & inhibitors ,MAP Kinase Kinase Kinases/genetics ,MAP Kinase Kinase Kinases/metabolism ,Mice, Inbred C57BL ,Mitogen-Activated Protein Kinase 10/antagonists & inhibitors ,Mitogen-Activated Protein Kinase 10/genetics ,Mitogen-Activated Protein Kinase 10/metabolism ,Obesity/metabolism ,Obesity/pathology ,Pancreas/growth & development ,Pancreas/metabolism ,RNA Interference ,RNA, Small Interfering/metabolism ,Rats ,Signal Transduction/drug effects ,Obesity ,Postnatal development ,Gene silencing ,Cyclin D2 ,Insulin ,Cyclin D1 ,RNA, Small Interfering ,Molecular Biology ,Pancreas ,Cyclin ,Cell Proliferation ,Pharmacology ,Serine/threonine-specific protein kinase ,0303 health sciences ,geography ,geography.geographical_feature_category ,Kinase ,030302 biochemistry & molecular biology ,Cell Biology ,Islet ,MAP Kinase Kinase Kinases ,Cell biology ,Glucose ,Molecular Medicine ,Signal transduction ,Signal Transduction - Abstract
International audience; Unveiling the key pathways underlying postnatal beta-cell proliferation can be instrumental to decipher the mechanisms of beta-cell mass plasticity to increased physiological demand of insulin during weight gain and pregnancy. Using transcriptome and global Serine Threonine Kinase activity (STK) analyses of islets from newborn (10 days old) and adult rats, we found that highly proliferative neonatal rat islet cells display a substantially elevated activity of the mitogen activated protein 3 kinase 12, also called dual leucine zipper-bearing kinase (Dlk). As a key upstream component of the c-Jun amino terminal kinase (Jnk) pathway, Dlk overexpression was associated with increased Jnk3 activity and was mainly localized in the beta-cell cytoplasm. We provide the evidence that Dlk associates with and activates Jnk3, and that this cascade stimu- lates the expression of Ccnd1 and Ccnd2, two essential cyclins controlling postnatal beta-cell replication. Silencing of Dlk or of Jnk3 in neonatal islet cells dramatically hampered primary beta-cell replication and the expression of the two cyclins. Moreover, the expression of Dlk, Jnk3, Ccnd1 and Ccnd2 was induced in high replicative islet beta cells from ob/ob mice during weight gain, and from pregnant female rats. In human islets from non-diabetic obese individuals, DLK expression was also cytoplasmic and the rise of the mRNA level was associated with an increase of JNK3, CCND1 and CCND2 mRNA levels, when compared to islets from lean and obese patients with diabetes. In conclusion, we fjnd that activation of Jnk3 signalling by Dlk could be a key mechanism for adapting islet beta-cell mass during postnatal development and weight gain.
- Published
- 2021
- Full Text
- View/download PDF
26. Proglucagon-Derived Peptides Expression and Secretion in Rat Insulinoma INS-1 Cells
- Author
-
Ana Acosta-Montalvo, Chiara Saponaro, Julie Kerr-Conte, Jochen H. M. Prehn, François Pattou, and Caroline Bonner
- Subjects
endocrine system ,insulin ,glucagon ,lcsh:Biology (General) ,glicentin ,proglucagon ,INS-1 cells ,bi-hormonal cells ,lcsh:QH301-705.5 ,hormones, hormone substitutes, and hormone antagonists - Abstract
Rat insulinoma INS-1 cells are widely used to study insulin secretory mechanisms. Studies have shown that a population of INS-1 cells are bi-hormonal, co-expressing insulin, and proglucagon proteins. They coined this population as immature cells since they co-secrete proglucagon-derived peptides from the same secretory vesicles similar to that of insulin. Since proglucagon encodes multiple peptides including glucagon, glucagon-like-peptide-1 (GLP-1), GLP-2, oxyntomodulin, and glicentin, their specific expression and secretion are technically challenging. In this study, we aimed to focus on glucagon expression which shares the same amino acid sequence with glicentin and proglucagon. Validation of the anti-glucagon antibody (Abcam) by Western blotting techniques revealed that the antibody detects proglucagon (≈ 20 kDa), glicentin (≈ 9 kDa), and glucagon (≈ 3 kDa) in INS-1 cells and primary islets, all of which were absent in the kidney cell line (HEK293). Using the validated anti-glucagon antibody, we showed by immunofluorescence imaging that a population of INS-1 cells co-express insulin and proglucagon-derived proteins. Furthermore, we found that chronic treatment of INS-1 cells with high-glucose decreases insulin and glucagon content, and also reduces the percentage of bi-hormonal cells. In line with insulin secretion, we found glucagon and glicentin secretion to be induced in a glucose-dependent manner. We conclude that INS-1 cells are a useful model to study glucose-stimulated insulin secretion, but not that of glucagon or glicentin. Our study suggests Western blotting technique as an important tool for researchers to study proglucagon-derived peptides expression and regulation in primary islets in response to various metabolic stimuli.
- Published
- 2020
- Full Text
- View/download PDF
27. Pancreatic β-cell specific loss ofE2f1impairs insulin secretion and β-cell identity through the epigenetic repression of non β-cell programs
- Author
-
Amélie Bonnefond, Pierre-Damien Denechaud, Jean-Sébastien Annicotte, Nabil Rabhi, Zohra Benfodda, François Pattou, Mehdi Derhourhi, Xavier Gromada, Souhila Amanzougarene, Lluis Fajas, Cyril Bourouh, Sarah Anissa Hannou, Emmanuelle Durand, Julie Kerr-Conte, Philippe Froguel, Patrick Meffre, Lionel Berberian, Emilie Courty, Maeva Moreno, Charlène Carney, Laure Rolland, and Frédérik Oger
- Subjects
Histone ,biology ,Downregulation and upregulation ,Epigenetic Repression ,biology.protein ,H3K4me3 ,Histone deacetylase ,E2F ,Epigenomics ,Chromatin ,Cell biology - Abstract
The loss of pancreatic β-cell identity emerges as an important feature of type 2 diabetes development, but the molecular mechanisms are still elusive. Here, we explore the cell-autonomous role of the cell cycle regulator and transcription factor E2F1 in the maintenance of β-cell identity and insulin secretion. We show that the β-cell-specific loss ofE2f1function in mice triggers glucose intolerance associated with defective insulin secretion, an altered α-to-β-cell ratio, a downregulation of many β-cell genes and a concomitant increase of non-β-cell markers. Mechanistically, the epigenomic profiling of non-beta cell upregulated gene promoters identified an enrichment of bivalent H3K4me3/H3K27me3 or H3K27me3 marks. Conversely, downregulated genes were enriched in active chromatin H3K4me3 and H3K27ac histone marks. We find that histone deacetylase inhibitors modulate E2F1 transcriptional and epigenomic signatures associated with these β-cell dysfunctions. Finally, the pharmacological inhibition of E2F transcriptional activity in human islets also impairs insulin secretion and the expression of β-cell identity genes. Our data suggest that E2F1 is critical for maintaining β-cell identity through a sustained repression of non β-cell transcriptional programs.
- Published
- 2020
- Full Text
- View/download PDF
28. Predicting the Key Regulators of Cell Identity in Human Adult Pancreas
- Author
-
Lotte Vanheer, Federica Fantuzzi, San Kit To, Andrea Alex Schiavo, Matthias Van Haele, Tine Haesen, Xiaoyan Yi, Adrian Janiszewski, Joel Chappell, Adrien Rihoux, Toshiaki Sawatani, Tania Roskams, Francois Pattou, Julie Kerr-Conte, Miriam Cnop, and Vincent Pasque
- Subjects
Cell type ,medicine.anatomical_structure ,Gene expression ,Gene regulatory network ,medicine ,RNA ,Computational biology ,Biology ,Induced pluripotent stem cell ,Pancreas ,Transcription factor ,Alpha cell - Abstract
SUMMARYCellular identity during development is under the control of transcription factors that form gene regulatory networks. However, the transcription factors and gene regulatory networks underlying cellular identity in the human adult pancreas remain largely unexplored. Here, we integrate multiple single-cell RNA-sequencing datasets of the human adult pancreas, totaling 7393 cells, and comprehensively reconstruct gene regulatory networks. We show that a network of 142 transcription factors forms distinct regulatory modules that characterize pancreatic cell types. We present evidence that our approach identifies regulators of cell identity in the human adult pancreas. We predict that HEYL, BHLHE41 and JUND are active in acinar, beta and alpha cells, respectively, and show that these proteins are present in the human adult pancreas as well as in human induced pluripotent stem cell (hiPSC)-derived islet cells. Using single-cell transcriptomics, we found that JUND represses beta cell genes in hiPSC-alpha cells. Both BHLHE41 and JUND depletion seemed to increase the number of sc-enterochromaffin cells in hiPSC-derived islets. The comprehensive gene regulatory network atlas can be explored interactively online. We anticipate our analysis to be the starting point for a more sophisticated dissection of how transcription factors regulate cell identity in the human adult pancreas. Furthermore, given that transcription factors are major regulators of embryo development and are often perturbed in diseases, a comprehensive understanding of how transcription factors work will be relevant in development and disease.HIGHLIGHTSReconstruction of gene regulatory networks for human adult pancreatic cell typesAn interactive resource to explore and visualize gene expression and regulatory statesPrediction of putative transcription factors that drive pancreatic cell identityBHLHE41 depletion in primary islets induces apoptosis
- Published
- 2020
- Full Text
- View/download PDF
29. Coxsackievirus-B4 Infection Can Induce the Expression of Human Endogenous Retrovirus W in Primary Cells
- Author
-
Arthur, Dechaumes, Antoine, Bertin, Famara, Sane, Sandrine, Levet, Jennifer, Varghese, Benjamin, Charvet, Valéry, Gmyr, Julie, Kerr-Conte, Justine, Pierquin, Govindakarnavar, Arunkumar, François, Pattou, Hervé, Perron, and Didier, Hober
- Subjects
endocrine system ,endocrine system diseases ,enterovirus ,viruses ,nutritional and metabolic diseases ,Article ,macrophages ,pancreatic cells ,lcsh:Biology (General) ,immune system diseases ,endogenous retrovirus ,embryonic structures ,PBMCs ,lcsh:QH301-705.5 - Abstract
Human Endogenous Retrovirus W Envelope (HERV-W ENV) mRNA or protein can be found in peripheral blood mononuclear cells (PBMCs) and exocrine pancreas of patients with type 1 diabetes (T1D). Further, previous observations have shown an association between enteroviral infection and development of T1D, specifically, coxsackievirus-B (CV-B) has been detected in the blood and pancreas of patients with T1D. Notably, viruses can activate HERV-W expression. Hence, we evaluated the effect of CV-B4 infection on HERV-W ENV mRNA expression. Primary human pancreatic ductal cells were obtained from five brain-dead donors. In the pancreatic cells of three donors, the HERV-W ENV mRNA level measured using RT-qPCR was upregulated upon CV-B4 infection. The HERV-W ENV protein was detected in the infected cells using the immunoblot assay. In human PBMCs inoculated with CV-B4 or when CV-B4 was incubated with an enhancing serum, the HERV-W ENV mRNA level was higher than the background RNA level. In monocyte-derived macrophages obtained from 5 of 13 donors, the HERV-W ENV mRNA level was higher in cultures inoculated with CV-B4 than in the control. Therefore, CV-B4 can upregulate or induce the transcription of a certain HERV-W ENV copy (or copies) in primary cell cultures, such as monocytes, macrophages, and pancreatic cells.
- Published
- 2020
30. 2056-P: In Vivo Human Islet Compensation and Dysfunction to Diet-Induced Obesity: A Phase 0 Preclinical Model
- Author
-
Julien Thevenet, Clara Clabaut, François Pattou, Caroline Bonner, Pauline Petit, Sofia Gargani, Julie Kerr-Conte, Valery Gmyr, and Gianni Pasquetti
- Subjects
0301 basic medicine ,medicine.medical_specialty ,geography ,geography.geographical_feature_category ,business.industry ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,medicine.disease ,Islet ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Endocrinology ,Downregulation and upregulation ,Fibrosis ,In vivo ,Diabetes mellitus ,Internal medicine ,Gene expression ,Internal Medicine ,medicine ,Hepatic stellate cell ,business ,Hormone - Abstract
A translational discrepancy exists between animal models used for diabetes research and diabetes in man, thus human relevant models must become a priority. We showed that collectively human islet grafts from 26 donors statistically significantly adapted their function and endocrine and β-cell mass to HFD (high fat diet) in vivo in 233 RAG2KO mice. Human islet compensation occurs at 6 weeks and metabolic dysfunction upon chronic ≥10 weeks HFD. Herein, gene expression studies (GeneChip® Human Gene 2.0 ST Arrays) identified molecular pathways involved. Indeed, human islets compensate to HFD by upregulating hormones, transcription factors, antioxidants, and unfolded protein response genes, and then become dysfunctional at 10 wks -by downregulating these genes. Herein we show Krebs cycle and Mitochondrial electron transport chain was targeted with a transient increase at 6wk and then a drop-in expression at 10 wks; whereas uncouplinggene expression UCP2 and 3 increased. Glycolysis remained upregulated. Explanations for expanded islet and beta mass in HFD (vs. CTL) may be found in cell cycle heatmap. Lastly, a marker of activated stellate cells TAGLN, and matrix genes rose during islet dysfunction (fibrosis?). Conclusion: This human-based model system progressively depicts defects identified in human pre and T2DM that could serve as a “phase 0” in vivo model to test new targets and develop effective therapies in man. Disclosure J.A. Kerr-Conte: None. J. Thevenet: None. G. Pasquetti: None. P. Petit: None. C. Clabaut: None. V. Gmyr: None. C. Bonner: None. S. Gargani: None. F. Pattou: None. Funding JDRF; Programme d’investissements d’avenir; LabEx; European Genomic Institute for Diabetes (ANR-10-LABX-46)
- Published
- 2020
- Full Text
- View/download PDF
31. P1672IMPACT OF ISLET TRANSPLANTATION VERSUS INSULIN ALONE AFTER KIDNEY TRANSPLANTATION IN TYPE 1 DIABETES PATIENTS
- Author
-
Valery Gmyr, Thomas Hubert, Robert Caiazzo, Julie Kerr-Conte, François Pattou, Mikael Chetboun, Marc Hazzan, Marie-Christine Vantyghem, Arnaud Jannin, Mehdi Maanaoui, and Kristell Le Mapihan
- Subjects
Transplantation ,Type 1 diabetes ,medicine.medical_specialty ,geography ,geography.geographical_feature_category ,business.industry ,Insulin ,medicine.medical_treatment ,medicine.disease ,Islet ,Gastroenterology ,Nephrology ,Internal medicine ,medicine ,business ,Kidney transplantation - Abstract
Background and Aims In patients with type 1 diabetes and end-stage renal disease, kidney transplantation improves both quality of life and survival. When simultaneous kidney-pancreas transplantation appears too invasive, or after failure of the pancreatic graft, an islet after kidney transplantation (IAK) may be considered to restore a stable endocrine function. The aim of our work was to assess the impact of islet transplantation on kidney transplantation outcomes versus insulin alone. Method In this retrospective parallel-arm cohort study in Lille, we included all type 1 diabetes patients who received a kidney graft from 2000 to 2017, followed by an islet transplantation after kidney (IAK) or not (kidney alone, KA). The primary study endpoint was the change of renal function (estimated glomerular filtration rate, eGFR). Secondary endpoints were glycemic control-related markers, such as HBA1c. Results During the period of study, 14 patients were included in the KA group versus 15 in the IAK group (including 5 after failure of a simultaneous pancreatic graft) were enrolled. At baseline, kidney donor sex, BMI, cause of death, cold ischemia time and recipient sex, waiting time on dialysis, type of dialysis, and number of previous kidney transplantation, were similar between the two groups. Yet, there were significant differences between KA and IAK, considering donor age (resp. 56.0±15.0 vs 35.2±13.7 years, p Conclusion In patients with type 1 diabetes and a functioning kidney graft, IAK was associated with a better glucose control and a slower decrease of eGFR than standard insulin therapy. Our results suggest that IAK should be proposed to type 1 diabetes patients with a functional kidney graft.
- Published
- 2020
- Full Text
- View/download PDF
32. 1900-P: HNF1A Deficiency Leads to Perturbed Glucagon Secretion in Humans
- Author
-
Magali Chiral, Caroline Bonner, Ioannis Spiliotis, Marco Pontoglio, C. Saponaro, Jochen H. M. Prehn, Julien Thevenet, Gianni Pasquetti, Lia Anguelova, Anthony Piron, Katharine R. Owen, Miriam Cnop, Valery Gmyr, Julie Kerr-Conte, Ana Acosta-Montalvo, and François Pattou
- Subjects
endocrine system ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,Glucagon secretion ,Hypoglycemia ,medicine.disease ,Glucagon ,Maturity onset diabetes of the young ,Endocrinology ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,Medicine ,Glucose homeostasis ,Gliclazide ,business ,medicine.drug - Abstract
Mutations in HNF1A cause Maturity Onset Diabetes of the Young (HNF1A-MODY). Carriers are normoglycemic in childhood, but with age, they develop hyperglycemia associated with insulin secretory defects and beta cell dysfunction. Although endogenous glucose production has been reported, glucagon in HNF1A-MODY has been less explored. Patients respond well to low dose sulfonylureas such as gliclazide, which increases insulin secretion independent of glucose but may predispose to hypoglycemia. We aimed to investigate whether low dose gliclazide had an effect on glucagon secretion in HNF1A-MODY. Additionally, we explored the potential of GLP-1 to normalize hormone secretion using in vivo and in vitro models of HNF1A deficiency. A 75g oral glucose tolerance test was performed in 7 MODY patients before and 72h after stopping gliclazide. Glucose rise was greater off gliclazide (p=0.003), while c-peptide and glucagon levels were similar between the groups. When adjusting for glucose levels during the OGTT, the change from baseline glucagon levels (Δ glucagon/glucose ratio) showed a significant decrease in those taking gliclazide (p=0.048), while there was no difference in Δ c-peptide/glucose ratio. Hnf1a-/- mice compared to WT mice are hyperglycemic, have decreased plasma insulin levels and hyperglucagonemia that was not suppressed after a glucose challenge (p=0.008). Furthermore, induction of the HNF1A variant Pro291fsInsC in rat INS1 cells reduced insulin and increased glucagon secretion, both of which were improved by GLP-1 treatment. Using the TIGER database we found HNF1A to be heterogeneously expressed in human islets, and siHNF1A reduced insulin secretion at high glucose. Collectively, these findings indicate that HNF1A is also essential for the regulation of glucagon secretion and glucose homeostasis. The mechanism by which GLP-1 normalizes insulin and glucagon secretion needs further investigation but could contribute to a better understanding of treatment response in HNF1A-MODY. Disclosure C. Saponaro: None. A. Acosta-Montalvo: None. L. Anguelova: None. J. Thevenet: None. M. Chiral: None. G. Pasquetti: None. A. Piron: None. M. Cnop: None. V. Gmyr: None. J. Prehn: None. J.A. Kerr-Conte: None. F. Pattou: None. M. Pontoglio: None. K.R. Owen: None. I. Spiliotis: None. C. Bonner: None. Funding European Foundation for the Study of Diabetes/Lilly European Diabetes Research Programme; Oxford Health Sciences Research Committee (1233)
- Published
- 2020
- Full Text
- View/download PDF
33. A nanobody-based nuclear imaging tracer targeting dipeptidyl peptidase 6 to determine the mass of human beta cell grafts in mice
- Author
-
Julien Thevenet, Rita Garcia Ribeiro, Stéphane Demine, Lorella Marselli, Julie Kerr-Conte, François Pattou, Piero Marchetti, Decio L. Eizirik, Nick Devoogdt, Supporting clinical sciences, Medical Imaging, Translational Imaging Research Alliance, and Pathologic Biochemistry and Physiology
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Islets of Langerhans Transplantation ,Cell Count ,Mice, SCID ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Insulin-Secreting Cells ,Human islet imaging ,Pancreatic beta cell imaging ,Pancreatic beta cells ,PET ,SPECT ,Type 1 diabetes ,Cells, Cultured ,geography.geographical_feature_category ,biology ,Organotechnetium Compounds ,Islet ,3. Good health ,Endocrinologie ,Molecular Imaging ,Médecine interne ,medicine.anatomical_structure ,Cell Tracking ,Heterografts ,Female ,Antibody ,Beta cell ,Single Photon Emission Computed Tomography Computed Tomography ,030209 endocrinology & metabolism ,Gallium Radioisotopes ,Mice, Transgenic ,Dipeptidyl peptidase ,03 medical and health sciences ,Islets of Langerhans ,Métabolisme ,In vivo ,Spect imaging ,Internal Medicine ,medicine ,Animals ,Humans ,Propidium iodide ,Radioactive Tracers ,Dipeptidyl-Peptidases and Tripeptidyl-Peptidases ,geography ,Diabétologie ,Pancreatic islets ,Single-Domain Antibodies ,Molecular biology ,Mice, Inbred C57BL ,030104 developmental biology ,chemistry ,biology.protein - Abstract
Aims/hypothesis: Type 1 diabetes is characterised by a progressive decline in beta cell mass. This is also observed following implantation of pancreatic islet allografts, but there is no reliable information regarding the time course of beta cell loss. This is due to the limited availability of non-invasive pancreatic islet imaging techniques. We have previously described that dipeptidyl peptidase 6 (DPP6) is an alpha and beta cell-specific biomarker, and developed a camelid antibody (nanobody ‘4hD29’) against it. We demonstrated the possibility to detect DPP6-expressing cells by single-photon emission computed tomography (SPECT)/ computed tomography (CT), but the correlation between the number of cells grafted and the SPECT signal was not assessed. Here, we investigate whether the 4hD29 nanobody allows us to detect different amounts of human pancreatic islets implanted into immune-deficient mice. In addition, we also describe the adaptation of the probe for use with positron emission tomography (PET). Methods: DPP6 expression was assessed in human samples using tissue arrays and immunohistochemistry. The effect of the 4hD29 nanobody on cell death and glucose-stimulated insulin secretion was measured in EndoC-βH1 cells and in human islets using Hoechst/propidium iodide staining and an anti-insulin ELISA, respectively. We performed in vivo SPECT imaging on severe combined immunodeficient (SCID) mice transplanted with different amounts of EndoC-βH1 cells (2 × 106, 5 × 106 and 10 × 106 cells), human islets (1000 and 3000) or pancreatic exocrine tissue using 99mTc-labelled 4hD29 nanobody. This DPP6 nanobody was also conjugated to N-chlorosuccinimide (NCS)-1,4,7-triazacyclononane-1,4,7-triacetic acid (NOTA), radiolabelled with either 67Ga (SPECT) or 68Ga (PET) and used in a proof-of-principle experiment to detect DPP6-expressing cells (Kelly neuroblastoma) grafted in SCID mice. Results: The DPP6 protein is mainly expressed in pancreatic islets. Importantly, the anti-DPP6 nanobody 4hD29 allows non-invasive detection of high amounts of EndoC-βH1 cells or human islets grafted in immunodeficient mice. This suggests that the probe must be further improved to detect lower numbers of islet cells. The 4hD29 nanobody neither affected beta cell viability nor altered insulin secretion in EndoC-βH1 cells and human islets. The conversion of 4hD29 nanobody into a PET probe was successful and did not alter its specificity. Conclusions/interpretation: These findings suggest that the anti-DPP6 4hD29 nanobody may become a useful tool for the quantification of human islet grafts in mice and, pending future development, islet mass in individuals with diabetes., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2020
- Full Text
- View/download PDF
34. microRNA-375 regulates glucose metabolism-related signaling for insulin secretion
- Author
-
Marc Prentki, Christophe Duranton, Virginie Casamento, N Gautier, François Pattou, Charlotte Hinault, Michel Tauc, Gaia Fabris, Emmanuel Van Obberghen, Didier F. Pisani, Patricia Lebrun, Julie Kerr-Conte, Stéphane Dalle, Olivier Dumortier, Institut de Recherche sur le Cancer et le Vieillissement (IRCAN), Université Nice Sophia Antipolis (... - 2019) (UNS), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), Laboratoire de PhysioMédecine Moléculaire (LP2M), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-COMUE Université Côte d'Azur (2015-2019) (COMUE UCA)-Centre National de la Recherche Scientifique (CNRS)-Université Côte d'Azur (UCA), COMUE Université Côte d'Azur (2015-2019) (COMUE UCA), Institut de Génomique Fonctionnelle (IGF), Université de Montpellier (UM)-Université Montpellier 1 (UM1)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université Montpellier 2 - Sciences et Techniques (UM2)-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Recherche translationnelle sur le diabète - U 1190 (RTD), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Université de Montréal (UdeM), Guerineau, Nathalie C., Université Nice Sophia Antipolis (1965 - 2019) (UNS), and Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Montpellier (UM)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Adult ,Male ,medicine.medical_specialty ,insulin secretion ,Endocrinology, Diabetes and Metabolism ,pancreatic islets diabetes ,030209 endocrinology & metabolism ,Carbohydrate metabolism ,03 medical and health sciences ,Islets of Langerhans ,0302 clinical medicine ,Endocrinology ,Internal medicine ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,medicine ,Animals ,Humans ,Glycolysis ,Rats, Wistar ,030304 developmental biology ,[SDV.MHEP.EM] Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,0303 health sciences ,Glucose metabolism ,pancreatic islets ,Developmental maturation ,diabetes ,Chemistry ,Pancreatic islets ,[SDV.MHEP.EM]Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,Pyruvate dehydrogenase complex ,Pyruvate carboxylase ,Rats ,microRNAs ,medicine.anatomical_structure ,Glucose ,[SDV.BBM.GTP] Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Female ,Beta cell ,Homeostasis ,Signal Transduction - Abstract
Enhanced beta cell glycolytic and oxidative metabolism are necessary for glucose-induced insulin secretion. While several microRNAs modulate beta cell homeostasis, miR-375 stands out as it is highly expressed in beta cells where it regulates beta cell function, proliferation and differentiation. As glucose metabolism is central in all aspects of beta cell functioning, we investigated the role of miR-375 in this process using human and rat islets; the latter being an appropriate model for in-depth investigation. We used forced expression and repression of mR-375 in rat and human primary islet cells followed by analysis of insulin secretion and metabolism. Additionally, miR-375 expression and glucose-induced insulin secretion were compared in islets from rats at different developmental ages. We found that overexpressing of miR-375 in rat and human islet cells blunted insulin secretion in response to glucose but not to α-ketoisocaproate or KCl. Further, miR-375 reduced O2 consumption related to glycolysis and pyruvate metabolism, but not in response to α-ketoisocaproate. Concomitantly, lactate production was augmented suggesting that glucose-derived pyruvate is shifted away from mitochondria. Forced miR-375 expression in rat or human islets increased mRNA levels of pyruvate dehydrogenase kinase-4, but decreased those of pyruvate carboxylase and malate dehydrogenase1. Finally, reduced miR-375 expression was associated with maturation of fetal rat beta cells and acquisition of glucose-induced insulin secretion function. Altogether our findings identify miR-375 as an efficacious regulator of beta cell glucose metabolism and of insulin secretion, and could be determinant to functional beta cell developmental maturation.
- Published
- 2020
- Full Text
- View/download PDF
35. Optimizing primary graft function in islet allotransplantation: The Lille experience
- Author
-
Mikael Chetboun, Julie Kerr-Conte, Robert Caiazzo, François Pattou, Christian Noel, Violeta Raverdy, Kristell Le Mapihan, Marie-Christine Vantyghem, Valery Gmyr, and Thomas Hubert
- Subjects
endocrine system ,Type 1 diabetes ,geography ,Kidney ,medicine.medical_specialty ,geography.geographical_feature_category ,endocrine system diseases ,business.industry ,medicine.medical_treatment ,Urology ,Immunosuppressive regimen ,medicine.disease ,Islet ,Graft function ,Transplantation ,medicine.anatomical_structure ,medicine ,Beta cell ,business ,Allotransplantation - Abstract
Islet allotransplantation has been shown to be effective in restoring endogenous insulin secretion. Many initial reports showed, however, a sharp decline of islet graft function with time. In the late 1990s we hypothesized that islet transplantation success could be significantly prolonged by increasing islet primary graft function, through the deliberate repetition of islet infusions. We report here the favorable long-term outcome of this strategy in patients with type 1 diabetes and severe hypoglycemia unawareness or after a kidney graft with the Edmonton immunosuppressive regimen. Our results suggest that optimizing initial graft function is essential for the long-term success of beta cell replacement.
- Published
- 2020
- Full Text
- View/download PDF
36. In vivo quality control of human islets in the immunodeficient mouse to predict islet function in man: A retrospective study in 87 clinical transplants
- Author
-
Julien Thevenet, Marie-Christine Vantyghem, Mikael Chetboun, Thomas Hubert, Alan Apete, Gianni Pasquetti, Julie Kerr-Conte, François Pattou, Caroline Bonner, Valery Gmyr, and Nathalie Delalleau
- Subjects
endocrine system ,geography ,medicine.medical_specialty ,geography.geographical_feature_category ,Sterility ,business.industry ,Urology ,Retrospective cohort study ,Islet ,Transplantation ,In vivo ,medicine ,Potency ,Beta (finance) ,business ,Immunodeficient Mouse - Abstract
Objective. This retrospective study in 87 clinical islet transplants sought to determine if the quantitative in vivo islet potency assay (QIVIPA) based on human c-peptide measurement in normoglycemic immunodeficient mice transplanted with 1% of clinical islet transplant (n = 165) could predict islet function in man after transplantation (tx). Since each recipient receives a mean of 2.7 grafts in our center, the ultimate goal of this work was to prospectively determine the functional islet mass required in 1–3 transplants in the QIVIPA mice to achieve optimal long-term islet function (beta score ≥ 7) in man. Methods. Islet preparations that met in vitro release criteria (200,000 IEQ, viability ≥ 80%, sterility) were transplanted in severe type 1 diabetic patients (n = 87). 2% of the preparation was used for in vitro quality controls, and 2% for in vivo controls: tx under the kidney capsule of two normoglycemic nude or RAG2 mice where fasting c-peptide and glycemia were measured over 30 days. Human islet function was defined by immediate graft function (hCP increase 7 days after tx), primary graft function (PGF = beta score 1 month after the last tx) and beta score at 1 year. Results. The QIVIPA model is robust and reproducible with two mice transplanted per human islet preparation (P Conclusion. Evidence from this retrospective study on 87 clinical islet transplants in 165 mice suggests that in vivo quantitative islet potency assays like QIVIPA in normoglycemic immunodeficient are a key indicator of the quality of manufactured clinical graft preparations as they predict islet function in man, both early function at 7 days post tx, PGF (beta score 1 month after the last tx), and 1-year beta score even when multiple grafts are transplanted per recipient.
- Published
- 2020
- Full Text
- View/download PDF
37. Contributors
- Author
-
Peter Abrams, Joel T. Adler, Rodolfo Alejandro, Mohamed Alibashe-Ahmed, Ana Alvarez, Takayuki Anazawa, Axel Andres, Barbara Antonioli, Alan Apete, David A. Axelrod, Lionel Badet, David Baidal, Kaylene Barrera, Pierre-Yves Benhamou, Thierry Berney, Alain Gerald Bertoni, Federico Bertuzzi, Ugo Boggi, Caroline Bonner, Adel Bozorgzadeh, Julien Branchereau, Jonathan Bromberg, George W. Burke, Fanny Buron, Robert Caiazzo, Rossana Caldara, Stephanie S. Camhi, Diego Cantarovich, Massimo Cardillo, D. Castanares-Zapatero, Pierre Cattan, Suresh Rama Chandran, Erin Chang, Linda Chen, Mikael Chetboun, Pratik Choudhary, Gaetano Ciancio, Maria Pia Cicalese, Antonio Citro, C. Collienne, Caterina Conte, Claire Counter, Khaled Z. Dajani, Carly M. Darden, Francesco De Cobelli, Eelco J.P. de Koning, Hector De Leon, Nathalie Delalleau, Laura DiChiacchio, Jason B. Doppenberg, Cinthia B. Drachenberg, Erica Dugnani, Ty B. Dunn, Marten A. Engelse, Ahmed Farag, Alan Farney, Anne Elizabeth Farrow, Ibrahim Fathi, Jose Figueiro, Anneliese Flatt, Georgia Fousteri, Jonathan A. Fridell, Peter J. Friend, Giacomo Gastaldi, Valery Gmyr, Javier Gonzalez, Jeevan Prakash Gopal, Frans K. Gorus, Masafumi Goto, Mitsukazu Gotoh, Michel Greget, Dominique Grenet, Paolo Antonio Grossi, Rainer W.G. Gruessner, Angelika C. Gruessner, David I. Harriman, Wayne J. Hawthorne, Jarl Hellman, Brenda Lee Holbert, Thomas Hubert, Sara Iacopi, Marco Infante, Peter Jacob, Paul Johnson, Raja Kandaswamy, Georges Karam, Dixon B. Kaufman, W.F. Kendall Jr, Clark D. Kensinger, Norma S. Kenyon, Julie Kerr-Conte, Delphine Kervella, Laurence Kessler, Romain Kessler, Bart Keymeulen, Olle Korsgren, Sandrine Lablanche, Muhaib Lakhani, Neeraj Lalwani, P.F. Laterre, Michael C. Lawrence, Frances Tangherlini Lee, Roger Lehmann, Elina Linetsky, Barbara Ludwig, Torbjörn Lundgren, Xunrong Luo, SriGita Madiraju, Paola Maffi, Paola Magistretti, Kristell Le Mapihan, James F. Markmann, Geert Martens, Paulo N. Martins, Francesco Antonio Mazzotta, Kavya Chitra Mekala, Raffaella Melzi, Alessia Mercalli, Paolo Monti, Mahmoud Morsi, Irene Mosca, M. Mourad, Anand S. Rathnasamy Muthusamy, Rita Nano, Bashoo Naziruddin, Christian Noel, John O’Callaghan, Jon S. Odorico, Anne Olland, E.C. Opara, Giuseppe Orlando, Nathalia Padilla, John C. Papadimitriou, Vassilios E. Papalois, Klearchos K. Papas, Gianni Pasquetti, François Pattou, Silvia Pellegrini, Nadine Pernin, Vittorio Grazio Perrone, Lorenzo Piemonti, Rutger Ploeg, John A. Powelson, Alberto Pugliese, Shanthini K. Rajan, Karthik V. Ramanathan, Violeta Raverdy, Robert R. Redfield, John Renz, Michael R. Rickels, Charles G. Rickert, Camillo Ricordi, Jeffrey Rogers, Joseph R. Scalea, Jesse D. Schold, Hanne Scholz, Antonio Secchi, Oscar K. Serrano, A.M. James Shapiro, Sidharth Sharma, Edward Sharples, James A.M. Shaw, Sanjay Sinha, Carlo Socci, Jean-Paul G. Squifflet, Peter G. Stock, Robert J. Stratta, David E.R. Sutherland, Manfredi Tesauro, Olivier Thaunat, Julien Thévenet, Christoph Troppmann, Marie-Christine Vantyghem, Francesco Vendrame, Massimo Venturini, Rodrigo Vianna, Fabio Vistoli, Bengt von Zur-Mühlen, X. Wittebole, Anne Wojtusciszyn, Arya Zarinsefat, and Asha Zimmerman
- Published
- 2020
- Full Text
- View/download PDF
38. Erratum. Ten-Year Outcome of Islet Alone or Islet After Kidney Transplantation in Type 1 Diabetes: A Prospective Parallel-Arm Cohort Study. Diabetes Care 2019;42:2042-2049
- Author
-
Eric Van Belle, François Machuron, Marc Hazzan, Marie-Christine Vantyghem, Christian Noel, Thomas Hubert, Robert Caiazzo, Stéphanie Espiard, Julie Kerr-Conte, Arnaud Jannin, Violeta Raverdy, Pascal Pigny, François Pattou, Nathalie Delalleau, Valery Gmyr, Kristell Le Mapihan, Marie Frimat, Mikael Chetboun, Université de Lille, LillOA, Recherche translationnelle sur le diabète - U 1190 (RTD), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Université de Lille, Miniaturisation pour la Synthèse, l’Analyse et la Protéomique - UAR 3290 (MSAP), Université de Lille-Centre National de la Recherche Scientifique (CNRS), Miniaturisation pour la Synthèse, l’Analyse et la Protéomique - USR 3290 (MSAP), Institut de Chimie du CNRS (INC)-Université de Lille-Centre National de la Recherche Scientifique (CNRS), Inserm, CHU Lille, Recherche translationnelle sur le diabète (RTD) - U1190, Cancer Heterogeneity, Plasticity and Resistance to Therapies (CANTHER) - UMR 9020 - UMR 1277, Centre Hospitalier Régional Universitaire [Lille] [CHRU Lille], Récepteurs nucléaires, Maladies Cardiovasculaires et Diabète (EGID) - U1011, Facteurs de Risque et Déterminants Moléculaires des Maladies liées au Vieillissement - U 1167 [RID-AGE], Récepteurs nucléaires, maladies cardiovasculaires et diabète - U 1011 [RNMCD], Miniaturisation pour la Synthèse, l’Analyse et la Protéomique - UAR 3290 [MSAP], Lille Inflammation Research International Center - U 995 [LIRIC], and Recherche translationnelle sur le diabète - U 1190 [RTD]
- Subjects
Advanced and Specialized Nursing ,Type 1 diabetes ,medicine.medical_specialty ,geography ,geography.geographical_feature_category ,business.industry ,Endocrinology, Diabetes and Metabolism ,[SDV]Life Sciences [q-bio] ,MEDLINE ,030209 endocrinology & metabolism ,medicine.disease ,Islet ,3. Good health ,Islet after kidney ,Transplantation ,[SDV] Life Sciences [q-bio] ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,Internal Medicine ,medicine ,030212 general & internal medicine ,business ,Cohort study - Abstract
The names of the working groups that collaborated in the study reported in this article were inadvertently omitted from the byline. The corrected byline should … 2019;42
- Published
- 2020
- Full Text
- View/download PDF
39. Glucose Regulates m6A Methylation of RNA in Pancreatic Islets
- Author
-
Florine Bornaque, Clément Philippe Delannoy, Emilie Courty, Nabil Rabhi, Charlène Carney, Laure Rolland, Maeva Moreno, Xavier Gromada, Cyril Bourouh, Pauline Petit, Emmanuelle Durand, François Pattou, Julie Kerr-Conte, Philippe Froguel, Amélie Bonnefond, Frédérik Oger, and Jean-Sébastien Annicotte
- Subjects
insulin secretion ,Adenosine ,QH301-705.5 ,Palmitates ,AlkB Homolog 5, RNA Demethylase ,Alpha-Ketoglutarate-Dependent Dioxygenase FTO ,Down-Regulation ,General Medicine ,Methylation ,Article ,epitranscriptome ,pancreatic beta cell ,type 2 diabetes ,Cell Line ,Mice, Inbred C57BL ,Islets of Langerhans ,Mice ,Glucose ,Insulin-Secreting Cells ,Animals ,RNA ,RNA, Messenger ,Biology (General) - Abstract
Type 2 diabetes is characterized by chronic hyperglycemia associated with impaired insulin action and secretion. Although the heritability of type 2 diabetes is high, the environment, including blood components, could play a major role in the development of the disease. Amongst environmental effects, epitranscriptomic modifications have been recently shown to affect gene expression and glucose homeostasis. The epitranscriptome is characterized by reversible chemical changes in RNA, with one of the most prevalent being the m6A methylation of RNA. Since pancreatic β cells fine tune glucose levels and play a major role in type 2 diabetes physiopathology, we hypothesized that the environment, through variations in blood glucose or blood free fatty acid concentrations, could induce changes in m6A methylation of RNAs in pancreatic β cells. Here we observe a significant decrease in m6A methylation upon high glucose concentration, both in mice and human islets, associated with altered expression levels of m6A demethylases. In addition, the use of siRNA and/or specific inhibitors against selected m6A enzymes demonstrate that these enzymes modulate the expression of genes involved in pancreatic β-cell identity and glucose-stimulated insulin secretion. Our data suggest that environmental variations, such as glucose, control m6A methylation in pancreatic β cells, playing a key role in the control of gene expression and pancreatic β-cell functions. Our results highlight novel causes and new mechanisms potentially involved in type 2 diabetes physiopathology and may contribute to a better understanding of the etiology of this disease.
- Published
- 2022
- Full Text
- View/download PDF
40. Modulation de l’absorption intestinale postprandiale du glucose après Roux-en-Y Gastric Bypass chez le miniporc
- Author
-
Grégory Baud, Camille Marciniak, Vincent Vangelder, Mehdi Daoudi, Thomas Hubert, Violeta Raverdy, Julie Kerr-Conte, Valery Gmyr, Robert Caiazzo, and François Pattou
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,General Medicine - Abstract
Le DT2 est caracterise par un defaut combine de la secretion et de l’action de l’insuline. Depuis pres d’un demi siecle la chirurgie bariatrique et notamment le Roux-en-Y Gastric Bypass (RYGB) ont montre des effets spectaculaires sur le controle glycemique remettant en question le paradigme de la prise en charge medicale du DT2. L’exclusion gastro duodenale induite par le RYGB ameliore le metabolisme glucidique independemment de la perte de poids. Ainsi les modifications du flux biliaire semblent jouer un role, cependant les mecanismes sous-jacents ne sont pas clairs. Nous avons realises des RYGB chez le miniporc et nous avons montre que l'absorption intestinale du glucose est diminuee dans l’anse alimentaire (AL) depourvue de bile. L'absorption du glucose dans l’AL etait restauree par l'ajout de la bile, et cet effet etait inhibe lorsque le co transport actif sodium glucose 1 (SGLT1) etait bloquee par la phlorizine. SGLT1 restait exprimee dans la AL, cependant la teneur dans la lumiere de l’intestin en sodium etait nettement diminuee. L’ajout de sodium dans l'AL provoquait le meme effet que la bile sur l'absorption du glucose et augmentait egalement l’excursion glycemique post prandiale chez le miniporc au cours d’un repas test vigil. La diminution de l'absorption intestinale du glucose apres RYGB a ensuite ete confirmee chez l'homme. Nos resultats demontrent que la l’exclusion biliaire affecte le metabolisme post prandiale du glucose par modulation des co transporteurs intestinaux sodium-glucose.
- Published
- 2018
- Full Text
- View/download PDF
41. Islets for Research: Nothing Is Perfect, but We Can Do Better
- Author
-
Olle Korsgren, Raffaella Melzi, Marie Karlsson, Valery Gmyr, Julie Kerr-Conte, Lorenzo Piemonti, Vanessa Lavallard, Rita Nano, Alessia Mercalli, François Pattou, Domenico Bosco, Thierry Berney, Nano, Rita, Kerr-Conte, Julie A, Bosco, Domenico, Karlsson, Marie, Lavallard, Vanessa, Melzi, Raffaella, Gmyr, Valery, Mercalli, Alessia, Berney, Thierry, Pattou, Françoi, Korsgren, Olle, and Piemonti, Lorenzo
- Subjects
0301 basic medicine ,endocrine system ,endocrine system diseases ,Endocrinology, Diabetes and Metabolism ,Islets of Langerhans Transplantation ,MEDLINE ,030209 endocrinology & metabolism ,Bioinformatics ,Islets of Langerhans ,03 medical and health sciences ,0302 clinical medicine ,Nothing ,Diabetes mellitus ,Diabetes Mellitus ,Internal Medicine ,Insulin ,Humans ,Medicine ,Diabetes Mellitus / metabolism ,geography ,geography.geographical_feature_category ,ddc:617 ,business.industry ,Islet ,medicine.disease ,3. Good health ,Transplantation ,Islets of Langerhans / metabolism ,030104 developmental biology ,Insulin / metabolism ,business ,Insulin metabolism - Abstract
In December 2018, Diabetes and Diabetologia began requiring authors of papers reporting data obtained from studies on human islets to report critical characteristics of the human islets used for research. The islet community was asked to provide feedback on it. Here is the contribution by the European Consortium for Islet Transplantation.
- Published
- 2019
- Full Text
- View/download PDF
42. 207.5: Impact of HLA Donor Specific Antibodies in the Era of Cell Therapy: Clinical Trajectories in Islet Allotransplantation Illustrate a Challenging Model
- Author
-
François Pattou, Marc Hazzan, Mehdi Maanaoui, Valery Gmyr, Julie Kerr-Conte, Kristell Le Mapihan, Isabelle Top, Thomas Hubert, Mikael Chetboun, Marie-Christine Vantyghem, and Myriam Labalette
- Subjects
Cell therapy ,Transplantation ,geography ,geography.geographical_feature_category ,business.industry ,Donor specific antibodies ,medicine.medical_treatment ,Immunology ,Medicine ,Human leukocyte antigen ,business ,Islet ,Allotransplantation - Published
- 2021
- Full Text
- View/download PDF
43. Expression and functional assessment of candidate type 2 diabetes susceptibility genes identify four new genes contributing to human insulin secretion
- Author
-
Ana Ortalli, Marlène Huyvaert, Amélie Bonnefond, Lorella Marselli, Piero Marchetti, Marie Verbanck, Fatou K. Ndiaye, Julie Kerr-Conte, Raphaël Scharfmann, Emmanuelle Durand, Philippe Froguel, Amar Abderrahmani, Mickaël Canouil, Olivier Sand, Vikash Chandra, François Pattou, Raphaël Boutry, Valérie Pawlowski, Odile Poulain-Godefroy, C. Lecoeur, Iandry Rabearivelo, and Clara Salazar-Cardozo
- Subjects
0301 basic medicine ,endocrine system diseases ,medicine.medical_treatment ,EndoC-βH1 ,Gene regulatory network ,Mice, Obese ,EndoC-beta H1 ,Genome-wide association study ,Cell Cycle Proteins ,Type 2 diabetes ,VARIANTS ,Bioinformatics ,Expression analysis ,PATHWAY ,MELLITUS ,Mice ,Insulin-Secreting Cells ,OF-FUNCTION MUTATIONS ,Insulin ,Gene Regulatory Networks ,Genetics ,Insulin secretion ,3. Good health ,DNA-Binding Proteins ,Original Article ,Female ,ACCURATE ,Life Sciences & Biomedicine ,lcsh:Internal medicine ,Racemases and Epimerases ,LINE ,Biology ,EndoC-βH1 ,RNAi screening ,Molecular Biology ,Cell Biology ,Cell Line ,Endocrinology & Metabolism ,03 medical and health sciences ,medicine ,Animals ,Humans ,Secretion ,Genetic Predisposition to Disease ,GENOME-WIDE ASSOCIATION ,lcsh:RC31-1245 ,Gene ,Transcription factor ,Genetic association ,Adaptor Proteins, Signal Transducing ,Science & Technology ,nutritional and metabolic diseases ,medicine.disease ,Mice, Inbred C57BL ,030104 developmental biology ,Diabetes Mellitus, Type 2 ,PANCREATIC BETA-CELLS ,GLUCOSE-HOMEOSTASIS ,Transcription Factors - Abstract
Objectives Genome-wide association studies (GWAS) have identified >100 loci independently contributing to type 2 diabetes (T2D) risk. However, translational implications for precision medicine and for the development of novel treatments have been disappointing, due to poor knowledge of how these loci impact T2D pathophysiology. Here, we aimed to measure the expression of genes located nearby T2D associated signals and to assess their effect on insulin secretion from pancreatic beta cells. Methods The expression of 104 candidate T2D susceptibility genes was measured in a human multi-tissue panel, through PCR-free expression assay. The effects of the knockdown of beta-cell enriched genes were next investigated on insulin secretion from the human EndoC-βH1 beta-cell line. Finally, we performed RNA-sequencing (RNA-seq) so as to assess the pathways affected by the knockdown of the new genes impacting insulin secretion from EndoC-βH1, and we analyzed the expression of the new genes in mouse models with altered pancreatic beta-cell function. Results We found that the candidate T2D susceptibility genes' expression is significantly enriched in pancreatic beta cells obtained by laser capture microdissection or sorted by flow cytometry and in EndoC-βH1 cells, but not in insulin sensitive tissues. Furthermore, the knockdown of seven T2D-susceptibility genes (CDKN2A, GCK, HNF4A, KCNK16, SLC30A8, TBC1D4, and TCF19) with already known expression and/or function in beta cells changed insulin secretion, supporting our functional approach. We showed first evidence for a role in insulin secretion of four candidate T2D-susceptibility genes (PRC1, SRR, ZFAND3, and ZFAND6) with no previous knowledge of presence and function in beta cells. RNA-seq in EndoC-βH1 cells with decreased expression of PRC1, SRR, ZFAND6, or ZFAND3 identified specific gene networks related to T2D pathophysiology. Finally, a positive correlation between the expression of Ins2 and the expression of Prc1, Srr, Zfand6, and Zfand3 was found in mouse pancreatic islets with altered beta-cell function. Conclusions This study showed the ability of post-GWAS functional studies to identify new genes and pathways involved in human pancreatic beta-cell function and in T2D pathophysiology., Highlights • Expression of genes located nearby T2D associated signals is enriched in β cells. • Knockdown of 7 T2D genes with known role in β cell changes insulin secretion. • Knockdown of 4 T2D genes with unknown role in β cell impairs insulin secretion. • RNA-seq in cells with knockdown of these 4 genes detected T2D-related networks.
- Published
- 2017
44. Pharmacological approach to understanding the control of insulin secretion in human islets
- Author
-
Jean-Claude Henquin, Denis Dufrane, Valery Gmyr, Julie Kerr-Conte, and Myriam Nenquin
- Subjects
0301 basic medicine ,medicine.medical_specialty ,BK channel ,Tetraethylammonium ,Glucokinase ,Endocrinology, Diabetes and Metabolism ,Stimulation ,Biology ,Inhibitory postsynaptic potential ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Endocrinology ,Tolbutamide ,chemistry ,Internal medicine ,Internal Medicine ,medicine ,Tetrodotoxin ,Diazoxide ,biology.protein ,medicine.drug - Abstract
Aims To understand better the control of insulin secretion by human β cells and to identify similarities to and differences from rodent models. Methods Dynamic insulin secretion was measured in perifused human islets treated with pharmacological agents of known modes of action. Results Glucokinase activation (Ro28-1675) lowered the glucose threshold for stimulation of insulin secretion to 1 mmol/L (G1), augmented the response to G3-G5 but not to G8-G15, whereas tolbutamide remained active in G20, which indicates that not all KATP channels were closed by high glucose concentrations. An almost 2-fold greater response to G15 than to supramaximal tolbutamide in G3 or to KCl+diazoxide in G15 vs G3 quantified the contribution of metabolic amplification to insulin secretion. Both disruption (latrunculin-B) and stabilization (jasplakinolide) of microfilaments augmented insulin secretion without affecting metabolic amplification. Tolbutamide-induced insulin secretion was consistently greater in G10 than G3, with a threshold at 1 and maximum at 10 µmol/L tolbutamide in G10, vs 10 and 25 µmol/L in G3. Sulphonylurea effects were thus clearly glucose-dependent. Insulin secretion was also increased by inhibiting K channels other than KATP channels: Kv or BK channels (tetraethylammonium), TASK-1 channels (ML-365) and SK4 channels (TRAM-34). Opening KATP channels with diazoxide inhibited glucose-induced insulin secretion with half maximum inhibitory concentrations of 9.6 and 24 µmol/L at G7 and G15. Blockade of L-type Ca channels (nimodipine) abolished insulin secretion, whereas a blocker of T-type Ca channels (NNC-55-0396) was ineffective at specific concentrations. Blockade of Na channels (tetrodotoxin) did not affect glucose-induced insulin secretion. Conclusions In addition to sharing a KATP channel-dependent triggering pathway and a metabolic amplifying pathway, human and rodent β cells were found to display more similarities than differences in the control of insulin secretion.
- Published
- 2017
- Full Text
- View/download PDF
45. Loss-of-function mutations in MRAP2 are pathogenic in hyperphagic obesity with hyperglycemia and hypertension
- Author
-
Cécile Aubert, Maithé Tauber, Ronan Roussel, Bénédicte Toussaint, François Pattou, Philippe Froguel, Jean-Michel Borys, Amjad Ghulam, Hélène Loiselle, Mathilde Boissel, Emmanuelle Durand, Sylvia Franc, Julie Kerr-Conte, Julie Maillet, Morgane Baron, Marlène Huyvaert, Amélie Bonnefond, Raphaël Boutry, J. Philippe, Jérémy Thomas, Guillaume Charpentier, Claire Lévy-Marchal, Michel Marre, Mehdi Derhourhi, Aurélie Dechaume, Emmanuel Vaillant, B. Balkau, Stefan Gaget, Raphael Scharfmann, Jacques Weill, Mickaël Canouil, Metabolic functional (epi)genomics and molecular mechanisms involved in type 2 diabetes and related diseases - UMR 8199 - UMR 1283 (EGENODIA (GI3M)), Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre National de la Recherche Scientifique (CNRS), Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Centre d'études et de recherches pour l'intensification du traitement du diabète (CERITD), Centre Hospitalier Sud Francilien, Association Fleurbaix Laventie Ville Santé (FLVS), Centre d'Investigation Clinique - Epidemiologie Clinique/essais Cliniques Hopital Robert Debre, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre de Physiopathologie Toulouse Purpan (CPTP), Université Toulouse III - Paul Sabatier (UT3), Université de Toulouse (UT)-Université de Toulouse (UT)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Service d'endocrinologie pédiatrique [CHU Lille], Hôpital Jeanne de Flandre [Lille]-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Fondation ILDYS (ILDYS), Recherche translationnelle sur le diabète - U 1190 (RTD), Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre Hospitalier Régional Universitaire [Lille] (CHRU Lille), Centre de Recherche des Cordeliers (CRC (UMR_S_1138 / U1138)), École Pratique des Hautes Études (EPHE), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Université Paris Diderot - Paris 7 (UPD7)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Sorbonne Université (SU), AP-HP - Hôpital Bichat - Claude Bernard [Paris], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Université Paris Diderot, Sorbonne Paris Cité, Paris, France, Université Paris Diderot - Paris 7 (UPD7), Centre de recherche en épidémiologie et santé des populations (CESP), Université de Versailles Saint-Quentin-en-Yvelines (UVSQ)-Université Paris-Sud - Paris 11 (UP11)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM), AP-HP. Université Paris Saclay, Clinique Ambroise Paré [Centres Médico-Chirurgicaux Ambroise Pré, Pierre Cherest, Hartmann], Imperial College London, This work was supported by grants from the French-speaking Society of Diabetes (Société Française du Diabète) to A.B., from the European Foundation for the Study of Diabetes/Lilly (to A.B.), from the French National Research Agency (ANR-10-LABX-46 (European Genomics Institute for Diabetes) and ANR-10-EQPX-07-01 (LIGAN-PM) to P.F.), from the European Research Council (ERC GEPIDIAB-294785 to P.F. and ERC Reg-Seq-715575 to A.B.), from FEDER (to P.F.) and from the ‘Région Nord Pas-de-Calais’ (to P.F.). A.B. was supported by Inserm., ANR-10-EQPX-0007,LIGAN PM,Plate forme Lilloise de séquençage du génome humain pour une médecine personnalisée(2010), European Project: 294785,EC:FP7:ERC,ERC-2011-ADG_20110310,GEPIDIAB(2012), Metabolic functional (epi)genomics and molecular mechanisms involved in type 2 diabetes and related diseases - UMR 8199 - UMR 1283 (GI3M), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille-Centre National de la Recherche Scientifique (CNRS)-Institut Pasteur de Lille, Réseau International des Instituts Pasteur (RIIP)-Réseau International des Instituts Pasteur (RIIP), Laboratoire de Biochimie et Hormonologie [CHRU LIlle] (Centre de Biologie Pathologie), Service de Diabétologie [Orsay, Corbeil-Essonnes], Université Paris-Sud - Paris 11 (UP11)-Hôpital Sud Francilien Corbeil Essonne, Centre d'Etudes et de Recherches pour l'Intensification du Traitement du Diabète (CERITD), Association 'Fleurbaix Laventie Ville Santé' (FLVS), Association Fleurbaix Laventie Ville Santé, Université Fédérale Toulouse Midi-Pyrénées-Université Fédérale Toulouse Midi-Pyrénées-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Centre National de la Recherche Scientifique (CNRS)-Université Paris Descartes - Paris 5 (UPD5)-Institut National de la Santé et de la Recherche Médicale (INSERM), Ildys Foundation [Roscoff, France], Départment de Diabétologie, Endocrinologie et Nutrition [AP-HP Hôpital Bichat], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-DHU FIRE Centre de compétence des maladies pulmonaires rares, École pratique des hautes études (EPHE), UFR de Médecine - Sorbonne Paris Cité (Université Paris Diderot - Paris 7 - UPD7), Université Paris Diderot - Paris 7 (UPD7)-Sorbonne Université (SU), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Université Paris-Sud - Paris 11 (UP11)-Hôpital Paul Brousse-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Versailles Saint-Quentin-en-Yvelines (UVSQ), CMC Ambroise Paré [Neuilly-sur-Seine, France], Department of Metabolism [London, UK] (Section of Genomics of Common Disease), Bonnefond, Amelie, Plate forme Lilloise de séquençage du génome humain pour une médecine personnalisée - - LIGAN PM2010 - ANR-10-EQPX-0007 - EQPX - VALID, and Genetics and epigenetics of Type 2 Diabetes physiology - GEPIDIAB - - EC:FP7:ERC2012-11-01 - 2017-10-31 - 294785 - VALID
- Subjects
0301 basic medicine ,Male ,MESH: Hypertension / complications ,MESH: Hypertension / pathology ,[SDV.GEN] Life Sciences [q-bio]/Genetics ,0302 clinical medicine ,Loss of Function Mutation ,Risk Factors ,MESH: Obesity / complications ,Receptor ,Child ,[SDV.MHEP.EM] Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,MESH: Obesity / metabolism ,MESH: Loss of Function Mutation / genetics ,[SDV.MHEP] Life Sciences [q-bio]/Human health and pathology ,MESH: Receptor, Melanocortin, Type 4 / genetics ,General Medicine ,Middle Aged ,[SDV.MHEP.EM]Life Sciences [q-bio]/Human health and pathology/Endocrinology and metabolism ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Metabolic effects ,Hypertension ,Receptor, Melanocortin, Type 4 ,Female ,Melanocortin ,MESH: Hyperglycemia / pathology ,Adult ,medicine.medical_specialty ,Adolescent ,MESH: Energy Metabolism / genetics ,Obesity risk ,Hyperphagia ,General Biochemistry, Genetics and Molecular Biology ,Article ,03 medical and health sciences ,Islets of Langerhans ,Young Adult ,MESH: Genetic Predisposition to Disease ,Internal medicine ,medicine ,Humans ,MESH: Adaptor Proteins, Signal Transducing / genetics ,Genetic Predisposition to Disease ,Obesity ,Loss function ,Adaptor Proteins, Signal Transducing ,MESH: Hyperphagia / pathology ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,MESH: Humans ,business.industry ,Pancreatic islets ,Energy control ,medicine.disease ,030104 developmental biology ,Blood pressure ,Endocrinology ,Hyperglycemia ,MESH: Obesity / pathology ,business ,Energy Metabolism ,[SDV.MHEP]Life Sciences [q-bio]/Human health and pathology ,MESH: Hyperphagia / complications - Abstract
International audience; The G-protein-coupled receptor accessory protein MRAP2 is implicated in energy control in rodents, notably via the melanocortin-4 receptor1. Although some MRAP2 mutations have been described in people with obesity1-3, their functional consequences on adiposity remain elusive. Using large-scale sequencing of MRAP2 in 9,418 people, we identified 23 rare heterozygous variants associated with increased obesity risk in both adults and children. Functional assessment of each variant shows that loss-of-function MRAP2 variants are pathogenic for monogenic hyperphagic obesity, hyperglycemia and hypertension. This contrasts with other monogenic forms of obesity characterized by excessive hunger, including melanocortin-4 receptor deficiency, that present with low blood pressure and normal glucose tolerance4. The pleiotropic metabolic effect of loss-of-function mutations in MRAP2 might be due to the failure of different MRAP2-regulated G-protein-coupled receptors in various tissues including pancreatic islets.
- Published
- 2019
- Full Text
- View/download PDF
46. Shielding islets with human amniotic epithelial cells enhances islet engraftment and revascularization in a murine diabetes model
- Author
-
François Pattou, Begoña Martinez de Tejada, Ekaterine Berishvili, David Cottet-Dumoulin, Vanessa Lavallard, Charles H Wassmer, Julie Kerr-Conte, Lisa Perez, Fanny Lebreton, Géraldine Parnaud, Domenico Bosco, Kevin Bellofatto, and Véronique Othenin-Girard
- Subjects
endocrine system ,endocrine system diseases ,Tissue/organ engineering ,Islets of Langerhans Transplantation ,Basic (laboratory) research/science ,Stem cells ,030230 surgery ,Regenerative medicine ,Diabetes Mellitus, Experimental ,Translational research/science ,Andrology ,03 medical and health sciences ,Islets of Langerhans ,Mice ,0302 clinical medicine ,In vivo ,Immunology and Allergy ,Medicine ,Animals ,Humans ,Insulin ,Pharmacology (medical) ,type 1 [Diabetes] ,Islet transplantation ,Transplantation ,geography ,ddc:618 ,geography.geographical_feature_category ,ddc:617 ,business.industry ,Graft Survival ,Epithelial Cells ,Hypoxia (medical) ,Islet ,In vitro ,Rats ,Amniotic epithelial cells ,Immunohistochemistry ,Stem cell ,medicine.symptom ,business - Abstract
Hypoxia is a major cause of considerable islet loss during the early post-transplantation period. Here, we investigate whether shielding islets with human amniotic epithelial cells (hAECs), which possess anti-inflammatory and regenerative properties, improves islet engraftment and survival. Shielded islets were generated on agarose microwells by mixing rat (RI) or human (HI) islets and hAECs (100 hAECs/IEQ). Islet secretory function and viability were assessed after culture in hypoxia (1% O2) or normoxia (21% O2) in vitro. In vivo function was evaluated after transplantation under the kidney capsule of diabetic immunodeficient mice. Graft morphology and vascularization were evaluated by immunohistochemistry. Both shielded RI and HI show higher viability and increased glucose-stimulated insulin secretion after exposure to hypoxia in vitro compared to control islets. Transplantation of shielded islets results in considerably earlier normoglycemia and vascularization, an enhanced glucose tolerance and a higher β-cell mass. Our results how that hAECs have a clear cytoprotective effect against hypoxic damages in vitro. This strategy improves β-cell mass engraftment and islet revascularization, leading to an improved capacity of islets to reverse hyperglycaemia, and could be rapidly applicable in the clinical situation seeing that the modification to human islets are minor.
- Published
- 2019
47. Human pancreatic islet three-dimensional chromatin architecture provides insights into the genetics of type 2 diabetes
- Author
-
Lorenzo Piemonti, Goutham Atla, Torben Hansen, Claire Morgan, Inês Cebola, Jorge Ferrer, Inga Prokopenko, Irene Farabella, Irene Miguel-Escalada, Marc A. Marti-Renom, Lorenzo Pasquali, Emil V. R. Appel, Niels Grarup, Javier García-Hurtado, Leif Groop, Josep M. Mercader, Eelco J.P. de Koning, Mireia Ramos-Rodríguez, Anette P. Gjesing, Delphine M.Y. Rolando, Allan Linneberg, Sílvia Bonàs-Guarch, François Pattou, Philippe Ravassard, Daniel R. Witte, Biola M. Javierre, Julie Kerr-Conte, Joan Ponsa-Cobas, David Torrents, Anthony Beucher, Peter Fraser, Thierry Berney, Iryna O. Fedko, Ole Birger Pedersen, Julen Mendieta-Esteban, Ignasi Moran, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg (UNISTRA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS), Research Centre For Prevention and Health, Department 84/85, Copenhagen University Hospital Glostrup, Steno Diabetes Center, University of Copenhagen = Københavns Universitet (KU), Novo Nordisk Foundation Center for Basic Metabolic Research (CBMR), Faculty of Health and Medical Sciences, University of Copenhagen = Københavns Universitet (KU)-University of Copenhagen = Københavns Universitet (KU), Steno Diabetes Center and Hagedorn Research Institute, Institut du Cerveau et de la Moëlle Epinière = Brain and Spine Institute (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM)-CHU Pitié-Salpêtrière [AP-HP], Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Sorbonne Université (SU)-Centre National de la Recherche Scientifique (CNRS), Barcelona Supercomputing Center - Centro Nacional de Supercomputacion (BSC - CNS), IRCCS Ospedale San Raffaele [Milan, Italy], Centre médical universitaire de Genève (CMU), Therapie Cellulaire du Diabete, Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Lille, Droit et Santé, Department of Clinical Sciences, Diabetes and Endocrinology Unit, Lund University [Lund], Gene Regulation, Stem Cells and Cancer Program, CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST), Barcelona, Spain, The Babraham Institute, The Babraham Institute, Cambridge, APH - Mental Health, APH - Health Behaviors & Chronic Diseases, Amsterdam Neuroscience - Mood, Anxiety, Psychosis, Stress & Sleep, Biological Psychology, Wellcome Trust, Imperial College Healthcare NHS Trust- BRC Funding, Medical Research Council (MRC), Miguel-Escalada, Irene, Bonàs-Guarch, Silvia, Cebola, Inê, Ponsa-Cobas, Joan, Mendieta-Esteban, Julen, Atla, Goutham, Javierre, Biola M, Rolando, Delphine M Y, Farabella, Irene, Morgan, Claire C, García-Hurtado, Javier, Beucher, Anthony, Morán, Ignasi, Pasquali, Lorenzo, Ramos-Rodríguez, Mireia, Appel, Emil V R, Linneberg, Allan, Gjesing, Anette P, Witte, Daniel R, Pedersen, Oluf, Grarup, Niel, Ravassard, Philippe, Torrents, David, Mercader, Josep M, Piemonti, Lorenzo, Berney, Thierry, de Koning, Eelco J P, Kerr-Conte, Julie, Pattou, Françoi, Fedko, Iryna O, Groop, Leif, Prokopenko, Inga, Hansen, Torben, Marti-Renom, Marc A, Fraser, Peter, Ferrer, Jorge, and Hubrecht Institute for Developmental Biology and Stem Cell Research
- Subjects
Epigenomics ,endocrine system diseases ,Metabolic disorders ,Gene regulatory network ,Molecular Conformation ,Genome-wide association study ,VARIANTS ,Genome-wide association studies ,Genome ,Cohort Studies ,0302 clinical medicine ,Genetics research ,Insulin Secretion ,Gene Regulatory Networks ,TRANSCRIPTION ,Promoter Regions, Genetic ,11 Medical and Health Sciences ,ComputingMilieux_MISCELLANEOUS ,Genetics ,Genetics & Heredity ,RISK ,0303 health sciences ,geography.geographical_feature_category ,ddc:617 ,CELL IDENTITY ,Islet ,Chromatin ,READ ALIGNMENT ,medicine.anatomical_structure ,Enhancer Elements, Genetic ,Life Sciences & Biomedicine ,EXPRESSION ,endocrine system ,Biology ,MECHANISMS ,03 medical and health sciences ,Islets of Langerhans ,SDG 3 - Good Health and Well-being ,medicine ,Humans ,Genetic Predisposition to Disease ,GENOME-WIDE ASSOCIATION ,Enhancer ,Gene ,030304 developmental biology ,geography ,Science & Technology ,Pancreatic islets ,06 Biological Sciences ,SUPER-ENHANCERS ,TCF7L2 ,Diabetes Mellitus, Type 2 ,Gene Expression Regulation ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,030217 neurology & neurosurgery ,Genome-Wide Association Study ,Developmental Biology - Abstract
Genetic studies promise to provide insight into the molecular mechanisms underlying type 2 diabetes (T2D). Variants associated with T2D are often located in tissue-specific enhancer clusters or super-enhancers. So far, such domains have been defined through clustering of enhancers in linear genome maps rather than in three-dimensional (3D) space. Furthermore, their target genes are often unknown. We have created promoter capture Hi-C maps in human pancreatic islets. This linked diabetes-associated enhancers to their target genes, often located hundreds of kilobases away. It also revealed >1,300 groups of islet enhancers, super-enhancers and active promoters that form 3D hubs, some of which show coordinated glucose-dependent activity. We demonstrate that genetic variation in hubs impacts insulin secretion heritability, and show that hub annotations can be used for polygenic scores that predict T2D risk driven by islet regulatory variants. Human islet 3D chromatin architecture, therefore, provides a framework for interpretation of T2D genome-wide association study (GWAS) signals. This research was supported by the National Institute for Health Research Imperial Biomedical Research Centre. Work was funded by grants from the Wellcome Trust (nos. WT101033 to J.F. and WT205915 to I.P.), Horizon 2020 (Research and Innovation Programme nos. 667191, to J.F., 633595, to I.P., and 676556, to M.A.M.-R.; Marie Sklodowska-Curie 658145, to I.M.-E., and 43062 ZENCODE, to G.A.), European Research Council (nos. 789055, to J.F., and 609989, to M.A.M.-R.). Marató TV3 (no. 201611, to J.F. and M.A.M.-R.), Ministerio de Ciencia Innovación y Universidades (nos. BFU2014-54284-R, RTI2018-095666, to J.F., BFU2017-85926-P, to M.A.M.-R., IJCI-2015-23352, to I.F.), AGAUR (to M.A.M.-R.). UK Medical Research Council (no. MR/L007150/1, to P.F., MR/L02036X/1 to J.F.), World Cancer Research Fund (WCRF UK, to I.P.) and World Cancer Research Fund International (no. 2017/1641 to I.P.), Biobanking and Biomolecular Resources Research Infrastructure (nos. BBMRI-NL, NWO 184.021.007, to I.O.F.). Work in IDIBAPS, CRG and CNAG was supported by the CERCA Programme, Generalitat de Catalunya and Centros de Excelencia Severo Ochoa (no. SEV-2012-0208). Human islets were provided through the European islet distribution program for basic research supported by JDRF (no. 3-RSC-2016-160-I-X). We thank N. Ruiz-Gomez for technical assistance; R. L. Fernandes, T. Thorne (University of Reading) and A. Perdones-Montero (Imperial College London) for helpful discussions regarding Machine Learning approaches; B. Lenhard and M. Merkenschlager (London Institute of Medical Sciences, Imperial College London), F. Müller (University of Birmingham) and J. L. Gómez-Skarmeta (Centro Andaluz de Biología del Desarrollo) for critical comments on the draft; the CRG Genomics Unit; and the Imperial College High Performance Computing Service.
- Published
- 2019
- Full Text
- View/download PDF
48. Coxsackievirus-B4 Infection of Human Primary Pancreatic Ductal Cell Cultures Results in Impairment of Differentiation into Insulin-Producing Cells
- Author
-
Antoine Bertin, Famara Sane, Valery Gmyr, Delphine Lobert, Arthur Dechaumes, Julie Kerr-Conte, François Pattou, and Didier Hober
- Subjects
enterovirus ,Brief Report ,lcsh:QR1-502 ,insulin mRNA ,RT-PCR ,Pancreatic Ducts ,Coxsackievirus Infections ,in vitro ,Cell Differentiation ,Epithelial Cells ,c-peptide ,lcsh:Microbiology ,Enterovirus B, Human ,Insulin-Secreting Cells ,Humans ,Insulin ,Cells, Cultured - Abstract
Coxsackievirus-B4 (CV-B4) E2 can persist in the pancreatic ductal-like cells (Panc-1 cell line), which results in an impaired differentiation of these cells into islet-like cell aggregates (ICA). In this study, primary pancreatic ductal cells obtained as a by-product of islet isolation from the pancreas of seven brain-dead adults were inoculated with CV-B4 E2, followed-up for 29 days, and the impact was investigated. Viral titers in culture supernatants were analyzed throughout the culture. Intracellular viral RNA was detected by RT-PCR. Levels of ductal cell marker CK19 mRNA and of insulin mRNA were evaluated by qRT-PCR. The concentration of c-peptide in supernatants was determined by ELISA. Ductal cells exposed to trypsin and serum-free medium formed ICA and resulted in an increased insulin secretion. Ductal cells from five brain-dead donors were severely damaged by CV-B4 E2, whereas the virus persisted in cultures of cells obtained from the other two. The ICAs whose formation was induced on day 14 post-inoculation were scarce and appeared tiny in infected cultures. Also, insulin mRNA expression and c-peptide levels were strongly reduced compared to the controls. In conclusion, CV-B4 E2 lysed human primary pancreatic ductal cells or persisted in these cells, which resulted in the impairment of differentiation into insulin-producing cells.
- Published
- 2019
49. O-GlcNacylation Links TxNIP to Inflammasome Activation in Pancreatic β Cells
- Author
-
Gaelle Filhoulaud, Fadila Benhamed, Patrick Pagesy, Caroline Bonner, Yann Fardini, Anissa Ilias, Jamileh Movassat, Anne-Françoise Burnol, Sandra Guilmeau, Julie Kerr-Conte, François Pattou, Tarik Issad, and Catherine Postic
- Subjects
O-GlcNAcylation ,lcsh:RC648-665 ,inflammasome ,TXNIP (thioredoxin-interacting protein) ,hyperglycemia ,pancreatic beta cells ,lcsh:Diseases of the endocrine glands. Clinical endocrinology - Abstract
Thioredoxin interacting protein (TxNIP), which strongly responds to glucose, has emerged as a central mediator of glucotoxicity in pancreatic β 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 β (IL1β) secretion by pancreatic β cells. To better understand the regulation of TxNIP by glucose in pancreatic β cells, we investigated the implication of O-linked β-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 β 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 IL1β. 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 β cells.
- Published
- 2019
- Full Text
- View/download PDF
50. Fat mass impact of sirolimus after clinical islet transplantation, a case control study
- Author
-
Helene Hoth-Guechot, Arnaud Jannin, François Pattou, Stéphanie Espiard, Julie Kerr-Conte, and Marie-Christine Vantyghem
- Subjects
Transplantation ,medicine.medical_specialty ,geography ,geography.geographical_feature_category ,business.industry ,Sirolimus ,medicine ,Urology ,Case-control study ,business ,Islet ,medicine.drug ,Fat mass - Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.