425 results on '"Decio L. Eizirik"'
Search Results
2. Untangling the genetics of beta cell dysfunction and death in type 1 diabetes
- Author
-
Catherine C. Robertson, Ruth M. Elgamal, Belle A. Henry-Kanarek, Peter Arvan, Shuibing Chen, Sangeeta Dhawan, Decio L. Eizirik, John S. Kaddis, Golnaz Vahedi, Stephen C.J. Parker, Kyle J. Gaulton, and Scott A. Soleimanpour
- Subjects
GWAS ,QTL ,SNP ,Autoimmunity ,Apoptosis ,Islet ,Internal medicine ,RC31-1245 - Abstract
Background: Type 1 diabetes (T1D) is a complex multi-system disease which arises from both environmental and genetic factors, resulting in the destruction of insulin-producing pancreatic beta cells. Over the past two decades, human genetic studies have provided new insight into the etiology of T1D, including an appreciation for the role of beta cells in their own demise. Scope of Review: Here, we outline models supported by human genetic data for the role of beta cell dysfunction and death in T1D. We highlight the importance of strong evidence linking T1D genetic associations to bona fide candidate genes for mechanistic and therapeutic consideration. To guide rigorous interpretation of genetic associations, we describe molecular profiling approaches, genomic resources, and disease models that may be used to construct variant-to-gene links and to investigate candidate genes and their role in T1D. Major Conclusions: We profile advances in understanding the genetic causes of beta cell dysfunction and death at individual T1D risk loci. We discuss how genetic risk prediction models can be used to address disease heterogeneity. Further, we present areas where investment will be critical for the future use of genetics to address open questions in the development of new treatment and prevention strategies for T1D.
- Published
- 2024
- Full Text
- View/download PDF
3. Regulation of β-cell death by ADP-ribosylhydrolase ARH3 via lipid signaling in insulitis
- Author
-
Soumyadeep Sarkar, Cailin Deiter, Jennifer E. Kyle, Michelle A. Guney, Dylan Sarbaugh, Ruichuan Yin, Xiangtang Li, Yi Cui, Mireia Ramos-Rodriguez, Carrie D. Nicora, Farooq Syed, Jonas Juan-Mateu, Charanya Muralidharan, Lorenzo Pasquali, Carmella Evans-Molina, Decio L. Eizirik, Bobbie-Jo M. Webb-Robertson, Kristin Burnum-Johnson, Galya Orr, Julia Laskin, Thomas O. Metz, Raghavendra G. Mirmira, Lori Sussel, Charles Ansong, and Ernesto S. Nakayasu
- Subjects
Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Lipids are regulators of insulitis and β-cell death in type 1 diabetes development, but the underlying mechanisms are poorly understood. Here, we investigated how the islet lipid composition and downstream signaling regulate β-cell death. Methods We performed lipidomics using three models of insulitis: human islets and EndoC-βH1 β cells treated with the pro-inflammatory cytokines interlukine-1β and interferon-γ, and islets from pre-diabetic non-obese mice. We also performed mass spectrometry and fluorescence imaging to determine the localization of lipids and enzyme in islets. RNAi, apoptotic assay, and qPCR were performed to determine the role of a specific factor in lipid-mediated cytokine signaling. Results Across all three models, lipidomic analyses showed a consistent increase of lysophosphatidylcholine species and phosphatidylcholines with polyunsaturated fatty acids and a reduction of triacylglycerol species. Imaging assays showed that phosphatidylcholines with polyunsaturated fatty acids and their hydrolyzing enzyme phospholipase PLA2G6 are enriched in islets. In downstream signaling, omega-3 fatty acids reduce cytokine-induced β-cell death by improving the expression of ADP-ribosylhydrolase ARH3. The mechanism involves omega-3 fatty acid-mediated reduction of the histone methylation polycomb complex PRC2 component Suz12, upregulating the expression of Arh3, which in turn decreases cell apoptosis. Conclusions Our data provide insights into the change of lipidomics landscape in β cells during insulitis and identify a protective mechanism by omega-3 fatty acids. Video Abstract
- Published
- 2024
- Full Text
- View/download PDF
4. EndoC-βH5 cells are storable and ready-to-use human pancreatic beta cells with physiological insulin secretion
- Author
-
Bruno Blanchi, Marion Taurand, Claire Colace, Sofia Thomaidou, Charlotte Audeoud, Federica Fantuzzi, Toshiaki Sawatani, Sevda Gheibi, Joan Sabadell-Basallote, Fransje W.J. Boot, Thibault Chantier, Aline Piet, Charlotte Cavanihac, Marion Pilette, Adélie Balguerie, Hamza Olleik, Françoise Carlotti, Miriam Ejarque, Malin Fex, Hindrik Mulder, Miriam Cnop, Decio L. Eizirik, Ouardane Jouannot, Anne-Lise Gaffuri, Paul Czernichow, Arnaud Zaldumbide, Raphaël Scharfmann, and Philippe Ravassard
- Subjects
Human pancreatic beta cell line ,Human beta cell function ,Glucose and incretin stimulated insulin secretion ,Type-I diabetes disease model ,Internal medicine ,RC31-1245 - Abstract
Objectives: Readily accessible human pancreatic beta cells that are functionally close to primary adult beta cells are a crucial model to better understand human beta cell physiology and develop new treatments for diabetes. We here report the characterization of EndoC-βH5 cells, the latest in the EndoC-βH cell family. Methods: EndoC-βH5 cells were generated by integrative gene transfer of immortalizing transgenes hTERT and SV40 large T along with Herpes Simplex Virus-1 thymidine kinase into human fetal pancreas. Immortalizing transgenes were removed after amplification using CRE activation and remaining non-excized cells eliminated using ganciclovir. Resulting cells were distributed as ready to use EndoC-βH5 cells. We performed transcriptome, immunological and extensive functional assays. Results: Ready to use EndoC-βH5 cells display highly efficient glucose dependent insulin secretion. A robust 10-fold insulin secretion index was observed and reproduced in four independent laboratories across Europe. EndoC-βH5 cells secrete insulin in a dynamic manner in response to glucose and secretion is further potentiated by GIP and GLP-1 analogs. RNA-seq confirmed abundant expression of beta cell transcription factors and functional markers, including incretin receptors. Cytokines induce a gene expression signature of inflammatory pathways and antigen processing and presentation. Finally, modified HLA-A2 expressing EndoC-βH5 cells elicit specific A2-alloreactive CD8 T cell activation. Conclusions: EndoC-βH5 cells represent a unique storable and ready to use human pancreatic beta cell model with highly robust and reproducible features. Such cells are thus relevant for the study of beta cell function, screening and validation of new drugs, and development of disease models.
- Published
- 2023
- Full Text
- View/download PDF
5. The type 1 diabetes gene TYK2 regulates β-cell development and its responses to interferon-α
- Author
-
Vikash Chandra, Hazem Ibrahim, Clémentine Halliez, Rashmi B. Prasad, Federica Vecchio, Om Prakash Dwivedi, Jouni Kvist, Diego Balboa, Jonna Saarimäki-Vire, Hossam Montaser, Tom Barsby, Väinö Lithovius, Isabella Artner, Swetha Gopalakrishnan, Leif Groop, Roberto Mallone, Decio L. Eizirik, and Timo Otonkoski
- Subjects
Science - Abstract
The TYK2 gene is associated with development of type 1 diabetes. Here the authors show that TYK2 regulates β-cell development, but at the same time TYK2 inhibition in the islets prevents IFNα responses and enhances their survival against CD8+ T-cell cytotoxicity; representing a potent therapeutic target to halt T1D progression.
- Published
- 2022
- Full Text
- View/download PDF
6. Gene expression signature predicts rate of type 1 diabetes progressionResearch in context
- Author
-
Tomi Suomi, Inna Starskaia, Ubaid Ullah Kalim, Omid Rasool, Maria K. Jaakkola, Toni Grönroos, Tommi Välikangas, Caroline Brorsson, Gianluca Mazzoni, Sylvaine Bruggraber, Lut Overbergh, David Dunger, Mark Peakman, Piotr Chmura, Søren Brunak, Anke M. Schulte, Chantal Mathieu, Mikael Knip, Riitta Lahesmaa, Laura L. Elo, Pieter Gillard, Kristina Casteels, Lutgart Overbergh, Chris Wallace, Mark Evans, Ajay Thankamony, Emile Hendriks, Loredana Marcoveccchio, Timothy Tree, Noel G. Morgan, Sarah Richardson, John A. Todd, Linda Wicker, Adrian Mander, Colin Dayan, Mohammad Alhadj Ali, Thomas Pieber, Decio L. Eizirik, Myriam Cnop, Flemming Pociot, Jesper Johannesen, Peter Rossing, Cristina Legido Quigley, Roberto Mallone, Raphael Scharfmann, Christian Boitard, Timo Otonkoski, Riitta Veijola, Matej Oresic, Jorma Toppari, Thomas Danne, Anette G. Ziegler, Peter Achenbach, Teresa Rodriguez-Calvo, Michele Solimena, Ezio E. Bonifacio, Stephan Speier, Reinhard Holl, Francesco Dotta, Francesco Chiarelli, Piero Marchetti, Emanuele Bosi, Stefano Cianfarani, Paolo Ciampalini, Carine De Beaufort, Knut Dahl-Jørgensen, Torild Skrivarhaug, Geir Joner, Lars Krogvold, Przemka Jarosz-Chobot, Tadej Battelino, Bernard Thorens, Martin Gotthardt, Bart O. Roep, Tanja Nikolic, Arnaud Zaldumbide, Ake Lernmark, Marcus Lundgren, Guillaume Costacalde, Thorsten Strube, Almut Nitsche, Jose Vela, Matthias Von Herrath, Johnna Wesley, Antonella Napolitano-Rosen, Melissa Thomas, Nanette Schloot, Allison Goldfine, Frank Waldron-Lynch, Jill Kompa, Aruna Vedala, Nicole Hartmann, Gwenaelle Nicolas, Jean van Rampelbergh, Nicolas Bovy, Sanjoy Dutta, Jeannette Soderberg, Simi Ahmed, Frank Martin, Esther Latres, Gina Agiostratidou, Anne Koralova, Ruben Willemsen, Anne Smith, Binu Anand, Vipan Datta, Vijith Puthi, Sagen Zac-Varghese, Renuka Dias, Premkumar Sundaram, Bijay Vaidya, Catherine Patterson, Katharine Owen, Barbara Piel, Simon Heller, Tabitha Randell, Tasso Gazis, Elise Bismuth Reismen, Jean-Claude Carel, Jean-Pierre Riveline, Jean-Francoise Gautier, Fabrizion Andreelli, Florence Travert, Emmanuel Cosson, Alfred Penfornis, Catherine Petit, Bruno Feve, Nadine Lucidarme, Jean-Paul Beressi, Catherina Ajzenman, Alina Radu, Stephanie Greteau-Hamoumou, Cecile Bibal, Thomas Meissner, Bettina Heidtmann, Sonia Toni, Birgit Rami-Merhar, Bart Eeckhout, Bernard Peene, N. Vantongerloo, Toon Maes, and Leen Gommers
- Subjects
Type 1 diabetes ,Autoantibodies ,RNA-seq ,Gene expression signature ,Predictive model ,Medicine ,Medicine (General) ,R5-920 - Abstract
Summary: Background: Type 1 diabetes is a complex heterogenous autoimmune disease without therapeutic interventions available to prevent or reverse the disease. This study aimed to identify transcriptional changes associated with the disease progression in patients with recent-onset type 1 diabetes. Methods: Whole-blood samples were collected as part of the INNODIA study at baseline and 12 months after diagnosis of type 1 diabetes. We used linear mixed-effects modelling on RNA-seq data to identify genes associated with age, sex, or disease progression. Cell-type proportions were estimated from the RNA-seq data using computational deconvolution. Associations to clinical variables were estimated using Pearson's or point-biserial correlation for continuous and dichotomous variables, respectively, using only complete pairs of observations. Findings: We found that genes and pathways related to innate immunity were downregulated during the first year after diagnosis. Significant associations of the gene expression changes were found with ZnT8A autoantibody positivity. Rate of change in the expression of 16 genes between baseline and 12 months was found to predict the decline in C-peptide at 24 months. Interestingly and consistent with earlier reports, increased B cell levels and decreased neutrophil levels were associated with the rapid progression. Interpretation: There is considerable individual variation in the rate of progression from appearance of type 1 diabetes-specific autoantibodies to clinical disease. Patient stratification and prediction of disease progression can help in developing more personalised therapeutic strategies for different disease endotypes. Funding: A full list of funding bodies can be found under Acknowledgments.
- Published
- 2023
- Full Text
- View/download PDF
7. Mining the transcriptome of target tissues of autoimmune and degenerative pancreatic β-cell and brain diseases to discover therapies
- Author
-
Xiaoyan Yi, Bianca Marmontel de Souza, Toshiaki Sawatani, Florian Szymczak, Lorella Marselli, Piero Marchetti, Miriam Cnop, and Decio L. Eizirik
- Subjects
Biological sciences ,Immunology ,Transcriptomics ,Science - Abstract
Summary: Target tissues of autoimmune and degenerative diseases show signals of inflammation. We used publicly available RNA-seq data to study whether pancreatic β-cells in type 1 and type 2 diabetes and neuronal tissue in multiple sclerosis and Alzheimer’s disease share inflammatory gene signatures. We observed concordantly upregulated genes in pairwise diseases, many of them related to signaling by interleukins and interferons. We next mined these signatures to identify therapies that could be re-purposed/shared among the diseases and identified the bromodomain inhibitors as potential perturbagens to revert the transcriptional signatures. We experimentally confirmed in human β-cells that bromodomain inhibitors I-BET151 and GSK046 prevent the deleterious effects of the pro-inflammatory cytokines interleukin-1β and interferon-γ and at least some of the effects of the metabolic stressor palmitate. These results demonstrate that key inflammation-induced molecular mechanisms are shared between β-cells and brain in autoimmune and degenerative diseases and that these signatures can be mined for drug discovery.
- Published
- 2022
- Full Text
- View/download PDF
8. Combined transcriptome and proteome profiling of the pancreatic β-cell response to palmitate unveils key pathways of β-cell lipotoxicity
- Author
-
Maria Lytrivi, Kassem Ghaddar, Miguel Lopes, Victoria Rosengren, Anthony Piron, Xiaoyan Yi, Henrik Johansson, Janne Lehtiö, Mariana Igoillo-Esteve, Daniel A. Cunha, Lorella Marselli, Piero Marchetti, Henrik Ortsäter, Decio L. Eizirik, and Miriam Cnop
- Subjects
Pancreatic islets ,Beta-cells ,Free fatty acids ,Lipid metabolism ,Endoplasmic reticulum stress ,RNA-sequencing ,Biotechnology ,TP248.13-248.65 ,Genetics ,QH426-470 - Abstract
Abstract Background Prolonged exposure to elevated free fatty acids induces β-cell failure (lipotoxicity) and contributes to the pathogenesis of type 2 diabetes. In vitro exposure of β-cells to the saturated free fatty acid palmitate is a valuable model of lipotoxicity, reproducing features of β-cell failure observed in type 2 diabetes. In order to map the β-cell response to lipotoxicity, we combined RNA-sequencing of palmitate-treated human islets with iTRAQ proteomics of insulin-secreting INS-1E cells following a time course exposure to palmitate. Results Crossing transcriptome and proteome of palmitate-treated β-cells revealed 85 upregulated and 122 downregulated genes at both transcript and protein level. Pathway analysis identified lipid metabolism, oxidative stress, amino-acid metabolism and cell cycle pathways among the most enriched palmitate-modified pathways. Palmitate induced gene expression changes compatible with increased free fatty acid mitochondrial import and β-oxidation, decreased lipogenesis and modified cholesterol transport. Palmitate modified genes regulating endoplasmic reticulum (ER) function, ER-to-Golgi transport and ER stress pathways. Furthermore, palmitate modulated cAMP/protein kinase A (PKA) signaling, inhibiting expression of PKA anchoring proteins and downregulating the GLP-1 receptor. SLC7 family amino-acid transporters were upregulated in response to palmitate but this induction did not contribute to β-cell demise. To unravel critical mediators of lipotoxicity upstream of the palmitate-modified genes, we identified overrepresented transcription factor binding sites and performed network inference analysis. These identified LXR, PPARα, FOXO1 and BACH1 as key transcription factors orchestrating the metabolic and oxidative stress responses to palmitate. Conclusions This is the first study to combine transcriptomic and sensitive time course proteomic profiling of palmitate-exposed β-cells. Our results provide comprehensive insight into gene and protein expression changes, corroborating and expanding beyond previous findings. The identification of critical drivers and pathways of the β-cell lipotoxic response points to novel therapeutic targets for type 2 diabetes.
- Published
- 2020
- Full Text
- View/download PDF
9. Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
- Author
-
Stéphane Demine, Andrea Alex Schiavo, Sandra Marín-Cañas, Piero Marchetti, Miriam Cnop, and Decio L. Eizirik
- Subjects
Pancreatic beta cells ,Type 1 diabetes ,Induced pluripotent stem cells ,Cytokines ,Apoptosis ,Endoplasmic reticulum stress ,Medicine (General) ,R5-920 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Adult human pancreatic beta cells are the “gold standard” for studies on diabetes pathogenesis, but their use is limited by insufficient availability and variable quality. An important effort has recently taken place to differentiate beta cells from human induced pluripotent stem cells (iPSCs) and validate their use for diabetes research. We presently used a 7-stage protocol to generate beta cells from human iPSC and evaluated whether these cells are responsive to the pro-inflammatory cytokines (IFNγ, IL-1β, or IFNα) that play a role in type 1 diabetes. Methods The iPSC-derived islet-like cell clusters contained 40–50% beta and 10–15% alpha cells and expressed the receptors for IFNγ, IL-1β, or IFNα. Cells were exposed to either IFNγ (1000 U/mL) + IL-1β (50 U/mL) or IFNα alone (2000 U/mL) for 24/48 h. Apoptosis was quantified using Hoechst/propidium iodide staining or the RealTime Glo Apoptosis Kit (Promega). After treatment, CXCL10 secretion was quantified by ELISA. The expression of multiples genes (Ins, Gcg, Nkx2.2, Nkx6.1, Pdx1, Mafa, BiP, Chop, Atf3, CXCL10, CXCL9, CCL5, and HLA-ABC) was quantified by RT-qPCR. Phosphorylation state and total expression of STAT1/STAT2, as well as expression of PDL1 and of the ER chaperone BiP, were quantified by Western blotting. The co-localization of HLA-ABC or cleaved caspase-3 and Ins/Gcg expression was assessed by immunohistochemistry. The presence of HLA-ABC at the plasma membrane was measured by flow cytometry. Results IFNγ + IL-1β and IFNα induced apoptosis of the cells after 48 h of exposure. Cleaved caspase-3 co-localized mostly but not exclusively with Ins+ cells. Exposure to IFNγ + IL-1β induced a pro-inflammatory phenotype, including increased CXCL10, CXCL9, and CCL5 expression; CXCL10 secretion; and HLA-ABC expression. HLA overexpression was confirmed at the protein level by Western blotting and flow cytometry. Exposure to IFNγ + IL-1β (but not IFNα) also induced beta cell dedifferentiation and endoplasmic reticulum stress (increase in BiP, Chop, and Atf3 mRNA expression). Phosphorylation of STAT1 was stimulated already after 1 h by IFNγ + IL-1β and IFNα, while phosphorylation of STAT2 was only activated by IFNα at 1–4 h. PDL1 expression was increased by both IFNγ + IL-1β and IFNα. Conclusions Our data show that human iPSC-derived beta cells respond to pro-inflammatory cytokines IL-1β + IFNγ and IFNα, by activating the same pathogenic processes as adult human primary beta cells. These cells thus represent a valuable tool for future research on the pathogenesis of type 1 diabetes.
- Published
- 2020
- Full Text
- View/download PDF
10. Neuropeptide Y1 receptor antagonism protects β-cells and improves glycemic control in type 2 diabetes
- Author
-
Chieh-Hsin Yang, Danise Ann-Onda, Xuzhu Lin, Stacey Fynch, Shaktypreya Nadarajah, Evan G. Pappas, Xin Liu, John W. Scott, Jonathan S. Oakhill, Sandra Galic, Yanchuan Shi, Alba Moreno-Asso, Cassandra Smith, Thomas Loudovaris, Itamar Levinger, Decio L. Eizirik, D. Ross Laybutt, Herbert Herzog, Helen E. Thomas, and Kim Loh
- Subjects
NPY ,β-Cell ,Insulin secretion ,Y1 receptor ,Type 2 diabetes ,Internal medicine ,RC31-1245 - Abstract
Objectives: Loss of functional β-cell mass is a key factor contributing to poor glycemic control in advanced type 2 diabetes (T2D). We have previously reported that the inhibition of the neuropeptide Y1 receptor improves the islet transplantation outcome in type 1 diabetes (T1D). The aim of this study was to identify the pathophysiological role of the neuropeptide Y (NPY) system in human T2D and further evaluate the therapeutic potential of using the Y1 receptor antagonist BIBO3304 to improve β-cell function and survival in T2D. Methods: The gene expression of the NPY system in human islets from nondiabetic subjects and subjects with T2D was determined and correlated with the stimulation index. The glucose-lowering and β-cell-protective effects of BIBO3304, a selective orally bioavailable Y1 receptor antagonist, in high-fat diet (HFD)/multiple low-dose streptozotocin (STZ)-induced and genetically obese (db/db) T2D mouse models were assessed. Results: In this study, we identified a more than 2-fold increase in NPY1R and its ligand, NPY mRNA expression in human islets from subjects with T2D, which was significantly associated with reduced insulin secretion. Consistently, the pharmacological inhibition of Y1 receptors by BIBO3304 significantly protected β cells from dysfunction and death under multiple diabetogenic conditions in islets. In a preclinical study, we demonstrated that the inhibition of Y1 receptors by BIBO3304 led to reduced adiposity and enhanced insulin action in the skeletal muscle. Importantly, the Y1 receptor antagonist BIBO3304 treatment also improved β-cell function and preserved functional β-cell mass, thereby resulting in better glycemic control in both HFD/multiple low-dose STZ-induced and db/db T2D mice. Conclusions: Our results revealed a novel causal link between increased islet NPY-Y1 receptor gene expression and β-cell dysfunction and failure in human T2D, contributing to the understanding of the pathophysiology of T2D. Furthermore, our results demonstrate that the inhibition of the Y1 receptor by BIBO3304 represents a potential β-cell-protective therapy for improving functional β-cell mass and glycemic control in T2D.
- Published
- 2022
- Full Text
- View/download PDF
11. A Humanized Mouse Strain That Develops Spontaneously Immune-Mediated Diabetes
- Author
-
Sandrine Luce, Sophie Guinoiseau, Alexis Gadault, Franck Letourneur, Patrick Nitschke, Marc Bras, Michel Vidaud, Pierre Charneau, Etienne Larger, Maikel L. Colli, Decio L. Eizirik, François Lemonnier, and Christian Boitard
- Subjects
autoimmunity ,type 1 diabetes (T1D) ,humanized mouse ,HLA-DQ8 ,epitopes ,preproinsulin ,Immunologic diseases. Allergy ,RC581-607 - Abstract
To circumvent the limitations of available preclinical models for the study of type 1 diabetes (T1D), we developed a new humanized model, the YES-RIP-hB7.1 mouse. This mouse is deficient of murine major histocompatibility complex class I and class II, the murine insulin genes, and expresses as transgenes the HLA-A*02:01 allele, the diabetes high-susceptibility HLA-DQ8A and B alleles, the human insulin gene, and the human co-stimulatory molecule B7.1 in insulin-secreting cells. It develops spontaneous T1D along with CD4+ and CD8+ T-cell responses to human preproinsulin epitopes. Most of the responses identified in these mice were validated in T1D patients. This model is amenable to characterization of hPPI-specific epitopes involved in T1D and to the identification of factors that may trigger autoimmune response to insulin-secreting cells in human T1D. It will allow evaluating peptide-based immunotherapy that may directly apply to T1D in human and complete preclinical model availability to address the issue of clinical heterogeneity of human disease.
- Published
- 2021
- Full Text
- View/download PDF
12. Pro-Inflammatory Cytokines Promote the Transcription of Circular RNAs in Human Pancreatic β Cells
- Author
-
Simranjeet Kaur, Caroline Frørup, Aashiq H. Mirza, Tina Fløyel, Reza Yarani, Maikel L. Colli, Jesper Johannesen, Joachim Størling, Decio L. Eizirik, and Flemming Pociot
- Subjects
non-coding RNA ,type 1 diabetes ,circRNA ,miRNA ,human islets ,microarray ,Genetics ,QH426-470 - Abstract
Circular RNAs (circRNAs) have recently been implicated in impaired β-cell function in diabetes. Using microarray-based profiling of circRNAs in human EndoC-βH1 cells treated with pro-inflammatory cytokines, this study aimed to investigate the expression and possible regulatory roles of circRNAs in human β cells. We identified ~5000 β-cell-expressed circRNAs, of which 84 were differentially expressed (DE) after cytokine exposure. Pathway analysis of the host genes of the DE circRNAs revealed the enrichment of cytokine signaling pathways, indicative of circRNA transcription from inflammatory genes in response to cytokines. Multiple binding sites for β-cell-enriched microRNAs and RNA-binding proteins were observed for the highly upregulated circRNAs, supporting their function as ‘sponges’ or ‘decoys’. We also present evidence for circRNA sequence conservation in multiple species, the presence of cytokine-induced regulatory elements, and putative protein-coding potential for the DE circRNAs. This study highlights the complex regulatory potential of circRNAs, which may play a crucial role during immune-mediated β-cell destruction in type 1 diabetes.
- Published
- 2022
- Full Text
- View/download PDF
13. The Protective Action of Metformin against Pro-Inflammatory Cytokine-Induced Human Islet Cell Damage and the Mechanisms Involved
- Author
-
Laura Giusti, Marta Tesi, Federica Ciregia, Lorella Marselli, Lorenzo Zallocco, Mara Suleiman, Carmela De Luca, Silvia Del Guerra, Mariachiara Zuccarini, Marco Trerotola, Decio L. Eizirik, Miriam Cnop, Maria R. Mazzoni, Piero Marchetti, Antonio Lucacchini, and Maurizio Ronci
- Subjects
β-cell ,cytokines ,metformin ,proteomics ,label-free shotgun analysis ,Cytology ,QH573-671 - Abstract
Metformin, a drug widely used in type 2 diabetes (T2D), has been shown to protect human β-cells exposed to gluco- and/or lipotoxic conditions and those in islets from T2D donors. We assessed whether metformin could relieve the human β-cell stress induced by pro-inflammatory cytokines (which mediate β-cells damage in type 1 diabetes, T1D) and investigated the underlying mechanisms using shotgun proteomics. Human islets were exposed to 50 U/mL interleukin-1β plus 1000 U/mL interferon-γ for 48 h, with or without 2.4 µg/mL metformin. Glucose-stimulated insulin secretion (GSIS) and caspase 3/7 activity were studied, and a shotgun label free proteomics analysis was performed. Metformin prevented the reduction of GSIS and the activation of caspase 3/7 induced by cytokines. Proteomics analysis identified more than 3000 proteins in human islets. Cytokines alone altered the expression of 244 proteins (145 up- and 99 down-regulated), while, in the presence of metformin, cytokine-exposure modified the expression of 231 proteins (128 up- and 103 downregulated). Among the proteins inversely regulated in the two conditions, we found proteins involved in vesicle motility, defense against oxidative stress (including peroxiredoxins), metabolism, protein synthesis, glycolysis and its regulation, and cytoskeletal proteins. Metformin inhibited pathways linked to inflammation, immune reactions, mammalian target of rapamycin (mTOR) signaling, and cell senescence. Some of the changes were confirmed by Western blot. Therefore, metformin prevented part of the deleterious actions of pro-inflammatory cytokines in human β-cells, which was accompanied by islet proteome modifications. This suggests that metformin, besides use in T2D, might be considered for β-cell protection in other types of diabetes, possibly including early T1D.
- Published
- 2022
- Full Text
- View/download PDF
14. Persistent or Transient Human β Cell Dysfunction Induced by Metabolic Stress: Specific Signatures and Shared Gene Expression with Type 2 Diabetes
- Author
-
Lorella Marselli, Anthony Piron, Mara Suleiman, Maikel L. Colli, Xiaoyan Yi, Amna Khamis, Gaelle R. Carrat, Guy A. Rutter, Marco Bugliani, Laura Giusti, Maurizio Ronci, Mark Ibberson, Jean-Valery Turatsinze, Ugo Boggi, Paolo De Simone, Vincenzo De Tata, Miguel Lopes, Daniela Nasteska, Carmela De Luca, Marta Tesi, Emanuele Bosi, Pratibha Singh, Daniela Campani, Anke M. Schulte, Michele Solimena, Peter Hecht, Brian Rady, Ivona Bakaj, Alessandro Pocai, Lisa Norquay, Bernard Thorens, Mickaël Canouil, Philippe Froguel, Decio L. Eizirik, Miriam Cnop, and Piero Marchetti
- Subjects
type 2 diabetes ,lipoglucotoxicity ,glucolipotoxicity ,human pancreatic islets ,beta cells ,damage ,Biology (General) ,QH301-705.5 - Abstract
Summary: Pancreatic β cell failure is key to type 2 diabetes (T2D) onset and progression. Here, we assess whether human β cell dysfunction induced by metabolic stress is reversible, evaluate the molecular pathways underlying persistent or transient damage, and explore the relationships with T2D islet traits. Twenty-six islet preparations are exposed to several lipotoxic/glucotoxic conditions, some of which impair insulin release, depending on stressor type, concentration, and combination. The reversal of dysfunction occurs after washout for some, although not all, of the lipoglucotoxic insults. Islet transcriptomes assessed by RNA sequencing and expression quantitative trait loci (eQTL) analysis identify specific pathways underlying β cell failure and recovery. Comparison of a large number of human T2D islet transcriptomes with those of persistent or reversible β cell lipoglucotoxicity show shared gene expression signatures. The identification of mechanisms associated with human β cell dysfunction and recovery and their overlap with T2D islet traits provide insights into T2D pathogenesis, fostering the development of improved β cell-targeted therapeutic strategies.
- Published
- 2020
- Full Text
- View/download PDF
15. SARS-CoV-2 Receptor Angiotensin I-Converting Enzyme Type 2 (ACE2) Is Expressed in Human Pancreatic β-Cells and in the Human Pancreas Microvasculature
- Author
-
Daniela Fignani, Giada Licata, Noemi Brusco, Laura Nigi, Giuseppina E. Grieco, Lorella Marselli, Lut Overbergh, Conny Gysemans, Maikel L. Colli, Piero Marchetti, Chantal Mathieu, Decio L. Eizirik, Guido Sebastiani, and Francesco Dotta
- Subjects
diabetes ,COVID-19 ,angiotensin I-converting enzyme type 2 (ACE2) ,beta-cell ,human pancreatic islets ,SARS-CoV-2 ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Increasing evidence demonstrated that the expression of Angiotensin I-Converting Enzyme type 2 (ACE2) is a necessary step for SARS-CoV-2 infection permissiveness. In light of the recent data highlighting an association between COVID-19 and diabetes, a detailed analysis aimed at evaluating ACE2 expression pattern distribution in human pancreas is still lacking. Here, we took advantage of INNODIA network EUnPOD biobank collection to thoroughly analyze ACE2, both at mRNA and protein level, in multiple human pancreatic tissues and using several methodologies. Using multiple reagents and antibodies, we showed that ACE2 is expressed in human pancreatic islets, where it is preferentially expressed in subsets of insulin producing β-cells. ACE2 is also highly expressed in pancreas microvasculature pericytes and moderately expressed in rare scattered ductal cells. By using different ACE2 antibodies we showed that a recently described short-ACE2 isoform is also prevalently expressed in human β-cells. Finally, using RT-qPCR, RNA-seq and High-Content imaging screening analysis, we demonstrated that pro-inflammatory cytokines, but not palmitate, increase ACE2 expression in the β-cell line EndoC-βH1 and in primary human pancreatic islets. Taken together, our data indicate a potential link between SARS-CoV-2 and diabetes through putative infection of pancreatic microvasculature and/or ductal cells and/or through direct β-cell virus tropism.
- Published
- 2020
- Full Text
- View/download PDF
16. Molecular Footprints of the Immune Assault on Pancreatic Beta Cells in Type 1 Diabetes
- Author
-
Maikel L. Colli, Florian Szymczak, and Decio L. Eizirik
- Subjects
type 1 diabetes ,beta cells ,pancreatic islets ,insulitis ,inflammation ,therapeutics ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Type 1 diabetes (T1D) is a chronic disease caused by the selective destruction of the insulin-producing pancreatic beta cells by infiltrating immune cells. We presently evaluated the transcriptomic signature observed in beta cells in early T1D and compared it with the signatures observed following in vitro exposure of human islets to inflammatory or metabolic stresses, with the aim of identifying “footprints” of the immune assault in the target beta cells. We detected similarities between the beta cell signatures induced by cytokines present at different moments of the disease, i.e., interferon-α (early disease) and interleukin-1β plus interferon-γ (later stages) and the beta cells from T1D patients, identifying biological process and signaling pathways activated during early and late stages of the disease. Among the first responses triggered on beta cells was an enrichment in antiviral responses, pattern recognition receptors activation, protein modification and MHC class I antigen presentation. During putative later stages of insulitis the processes were dominated by T-cell recruitment and activation and attempts of beta cells to defend themselves through the activation of anti-inflammatory pathways (i.e., IL10, IL4/13) and immune check-point proteins (i.e., PDL1 and HLA-E). Finally, we mined the beta cell signature in islets from T1D patients using the Connectivity Map, a large database of chemical compounds/drugs, and identified interesting candidates to potentially revert the effects of insulitis on beta cells.
- Published
- 2020
- Full Text
- View/download PDF
17. Pro-Inflammatory Cytokines Induce Insulin and Glucagon Double Positive Human Islet Cells That Are Resistant to Apoptosis
- Author
-
Marta Tesi, Marco Bugliani, Gianmarco Ferri, Mara Suleiman, Carmela De Luca, Emanuele Bosi, Matilde Masini, Vincenzo De Tata, Conny Gysemans, Francesco Cardarelli, Miriam Cnop, Decio L. Eizirik, Piero Marchetti, and Lorella Marselli
- Subjects
human islets ,α-cells ,β-cells ,insulin ,glucagon ,diabetes ,Microbiology ,QR1-502 - Abstract
The presence of islet cells double positive for insulin and glucagon (Ins+/Glu+) has been described in the pancreas from both type 2 (T2D) and type 1 (T1D) diabetic subjects. We studied the role of pro-inflammatory cytokines on the occurrence, trajectory, and characteristics of Ins+/Glu+ cells in human pancreatic islets. Pancreas samples, isolated islets, and dispersed islet cells from 3 T1D and 11 non-diabetic (ND) multi-organ donors were studied by immunofluorescence, confocal microscopy, and/or electron microscopy. ND islet cells were exposed to interleukin-1β and interferon-γ for up to 120 h. In T1D islets, we confirmed an increased prevalence of Ins+/Glu+ cells. Cytokine-exposed islets showed a progressive increase of Ins+/Glu+ cells that represented around 50% of endocrine cells after 120h. Concomitantly, cells expressing insulin granules only decreased significantly over time, whereas those containing only glucagon granules remained stable. Interestingly, Ins+/Glu+ cells were less prone to cytokine-induced apoptosis than cells containing only insulin. Cytokine-exposed islets showed down-regulation of β-cell identity genes. In conclusion, pro-inflammatory cytokines induce Ins+/Glu+ cells in human islets, possibly due to a switch from a β- to a β-/α-cell phenotype. These Ins+/Glu+ cells appear to be resistant to cytokine-induced apoptosis.
- Published
- 2021
- Full Text
- View/download PDF
18. The lipid sensor GPR120 promotes brown fat activation and FGF21 release from adipocytes
- Author
-
Tania Quesada-López, Rubén Cereijo, Jean-Valery Turatsinze, Anna Planavila, Montserrat Cairó, Aleix Gavaldà-Navarro, Marion Peyrou, Ricardo Moure, Roser Iglesias, Marta Giralt, Decio L. Eizirik, and Francesc Villarroya
- Subjects
Science - Abstract
GPR120 is a G-protein-coupled receptor that binds polyunsaturated fatty acids. Here, the authors show that GPR120 is upregulated in brown fat in cold-exposed mice, and mediates thermogenic activation of brown fat via a mechanism that, at least in part, depends on the release of the adipokine FGF21.
- Published
- 2016
- Full Text
- View/download PDF
19. Pancreatic α Cells are Resistant to Metabolic Stress-induced Apoptosis in Type 2 Diabetes
- Author
-
Laura Marroqui, Matilde Masini, Beatriz Merino, Fabio A. Grieco, Isabelle Millard, Christine Dubois, Ivan Quesada, Piero Marchetti, Miriam Cnop, and Decio L. Eizirik
- Subjects
Pancreatic α cells ,Type 2 diabetes ,Apoptosis ,Metabolic stress ,Palmitate ,Bcl2l1 ,ER stress ,Medicine ,Medicine (General) ,R5-920 - Abstract
Pancreatic α cells are exposed to metabolic stress during the evolution of type 2 diabetes (T2D), but it remains unclear whether this affects their survival. We used electron microscopy to search for markers of apoptosis and endoplasmic reticulum (ER) stress in α and β cells in islets from T2D or non-diabetic individuals. There was a significant increase in apoptotic β cells (from 0.4% in control to 6.0% in T2D), but no α cell apoptosis. We observed, however, similar ER stress in α and β cells from T2D patients. Human islets or fluorescence-activated cell sorting (FACS)-purified rat β and α cells exposed in vitro to the saturated free fatty acid palmitate showed a similar response as the T2D islets, i.e. both cell types showed signs of ER stress but only β cells progressed to apoptosis. Mechanistic experiments indicate that this α cell resistance to palmitate-induced apoptosis is explained, at least in part, by abundant expression of the anti-apoptotic protein Bcl2l1 (also known as Bcl-xL).
- Published
- 2015
- Full Text
- View/download PDF
20. Clic4, a novel protein that sensitizes β-cells to apoptosis
- Author
-
Dhaval Patel, Damien Ythier, Flora Brozzi, Decio L. Eizirik, and Bernard Thorens
- Subjects
Clic4 ,β-cells ,Apoptosis ,Cytokines ,Bcl-2 ,Diabetes ,Internal medicine ,RC31-1245 - Abstract
Objectives: Chloride intracellular channel protein 4 (Clic4) is a ubiquitously expressed protein involved in multiple cellular processes including cell-cycle control, cell differentiation, and apoptosis. Here, we investigated the role of Clic4 in pancreatic β-cell apoptosis. Methods: We used βTC-tet cells and islets from β-cell specific Clic4 knockout mice (βClic4KO) and assessed cytokine-induced apoptosis, Bcl2 family protein expression and stability, and identified Clic4-interacting proteins by co-immunoprecipitation and mass spectrometry analysis. Results: We show that cytokines increased Clic4 expression in βTC-tet cells and in mouse islets and siRNA-mediated silencing of Clic4 expression in βTC-tet cells or its genetic inactivation in islets β-cells, reduced cytokine-induced apoptosis. This was associated with increased expression of Bcl-2 and increased expression and phosphorylation of Bad. Measurement of Bcl-2 and Bad half-lives in βTC-tet cells showed that Clic4 silencing increased the stability of these proteins. In primary islets β-cells, absence of Clic4 expression increased Bcl-2 and Bcl-xL expression as well as expression and phosphorylation of Bad. Mass-spectrometry analysis of proteins co-immunoprecipitated with Clic4 from βTC-tet cells showed no association of Clic4 with Bcl-2 family proteins. However, Clic4 co-purified with proteins from the proteasome suggesting a possible role for Clic4 in regulating protein degradation. Conclusions: Collectively, our data show that Clic4 is a cytokine-induced gene that sensitizes β-cells to apoptosis by reducing the steady state levels of Bcl-2, Bad and phosphorylated Bad.
- Published
- 2015
- Full Text
- View/download PDF
21. Imaging of Human Insulin Secreting Cells with Gd-DOTA-P88, a Paramagnetic Contrast Agent Targeting the Beta Cell Biomarker FXYD2γa
- Author
-
Stéphane Demine, Alexander Balhuizen, Vinciane Debaille, Lieke Joosten, Maïté Fereau, Satya Narayana Murthy Chilla, Isabelle Millard, Raphaël Scharfmann, Dominique Egrise, Serge Goldman, Piero Marchetti, Martin Gotthardt, Sophie Laurent, Carmen Burtea, and Decio L. Eizirik
- Subjects
peptide-based imaging ,beta cell imaging ,paramagnetic contrast agent ,non-invasive imaging ,MRI ,pancreatic beta cell ,Type 1 diabetes ,Type 2 diabetes ,Organic chemistry ,QD241-441 - Abstract
Non-invasive imaging and quantification of human beta cell mass remains a major challenge. We performed pre-clinical in vivo validation of a peptide previously discovered by our group, namely, P88 that targets a beta cell specific biomarker, FXYD2γa. We conjugated P88 with DOTA and then complexed it with GdCl3 to obtain the MRI (magnetic resonance imaging) contrast agent (CA) Gd-DOTA-P88. A scrambled peptide was used as a negative control CA, namely Gd-DOTA-Scramble. The CAs were injected in immunodeficient mice implanted with EndoC-βH1 cells, a human beta cell line that expresses FXYD2γa similarly to primary human beta cells. The xenograft-bearing mice were analyzed by MRI. At the end, the mice were euthanized and the CA biodistribution was evaluated on the excised tissues by measuring the Gd concentration with inductively coupled plasma mass spectrometry (ICP-MS). The MRI and biodistribution studies indicated that Gd-DOTA-P88 accumulates in EndoC-βH1 xenografts above the level observed in the background tissue, and that its uptake is significantly higher than that observed for Gd-DOTA-Scramble. In addition, the Gd-DOTA-P88 showed good xenograft-to-muscle and xenograft-to-liver uptake ratios, two potential sites of human islets transplantation. The CA shows good potential for future use to non-invasively image implanted human beta cells.
- Published
- 2018
- Full Text
- View/download PDF
22. Why does the immune system destroy pancreatic β-cells but not α-cells in type 1 diabetes?
- Author
-
Decio L. Eizirik, Florian Szymczak, and Roberto Mallone
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism - Published
- 2023
- Full Text
- View/download PDF
23. Inhibition of the type 1 diabetes candidate gene PTPN2 aggravates TNF-α-induced human beta cell dysfunction and death
- Author
-
Arturo Roca-Rivada, Sandra Marín-Cañas, Maikel L. Colli, Chiara Vinci, Toshiaki Sawatani, Lorella Marselli, Miriam Cnop, Piero Marchetti, and Decio L. Eizirik
- Subjects
Endocrinology, Diabetes and Metabolism ,Internal Medicine - Published
- 2023
- Full Text
- View/download PDF
24. BCL-XL Overexpression Protects Pancreatic β-Cells against Cytokine- and Palmitate-Induced Apoptosis
- Author
-
Atenea A. Perez-Serna, Reinaldo S. Dos Santos, Cristina Ripoll, Angel Nadal, Decio L. Eizirik, and Laura Marroqui
- Subjects
Inorganic Chemistry ,apoptosis ,BCL-XL ,Ca2+ signaling ,cytokines ,insulin secretion ,palmitate ,pancreatic β-cells ,Organic Chemistry ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
Diabetes is a chronic disease that affects glucose metabolism, either by autoimmune-driven β-cell loss or by the progressive loss of β-cell function, due to continued metabolic stresses. Although both α- and β-cells are exposed to the same stressors, such as proinflammatory cytokines and saturated free fatty acids (e.g., palmitate), only α-cells survive. We previously reported that the abundant expression of BCL-XL, an anti-apoptotic member of the BCL-2 family of proteins, is part of the α-cell defense mechanism against palmitate-induced cell death. Here, we investigated whether BCL-XL overexpression could protect β-cells against the apoptosis induced by proinflammatory and metabolic insults. For this purpose, BCL-XL was overexpressed in two β-cell lines—namely, rat insulinoma-derived INS-1E and human insulin-producing EndoC-βH1 cells—using adenoviral vectors. We observed that the BCL-XL overexpression in INS-1E cells was slightly reduced in intracellular Ca2+ responses and glucose-stimulated insulin secretion, whereas these effects were not observed in the human EndoC-βH1 cells. In INS-1E cells, BCL-XL overexpression partially decreased cytokine- and palmitate-induced β-cell apoptosis (around 40% protection). On the other hand, the overexpression of BCL-XL markedly protected EndoC-βH1 cells against the apoptosis triggered by these insults (>80% protection). Analysis of the expression of endoplasmic reticulum (ER) stress markers suggests that resistance to the cytokine and palmitate conferred by BCL-XL overexpression might be, at least in part, due to the alleviation of ER stress. Altogether, our data indicate that BCL-XL plays a dual role in β-cells, participating both in cellular processes related to β-cell physiology and in fostering survival against pro-apoptotic insults.
- Published
- 2023
- Full Text
- View/download PDF
25. GLP-1R agonists demonstrate potential to treat Wolfram syndrome in human preclinical models
- Author
-
Vyron Gorgogietas, Bahareh Rajaei, Chae Heeyoung, Bruno J. Santacreu, Sandra Marín-Cañas, Paraskevi Salpea, Toshiaki Sawatani, Anyishai Musuaya, María N. Arroyo, Cristina Moreno-Castro, Khadija Benabdallah, Celine Demarez, Sanna Toivonen, Cristina Cosentino, Nathalie Pachera, Maria Lytrivi, Ying Cai, Lode Carnel, Cris Brown, Fumihiko Urano, Piero Marchetti, Patrick Gilon, Decio L. Eizirik, Miriam Cnop, Mariana Igoillo-Esteve, and UCL - SSS/IREC/EDIN - Pôle d'endocrinologie, diabète et nutrition
- Subjects
Mice, Knockout ,Wolfram syndrome ,GLP-1R agonists ,Endocrinology, Diabetes and Metabolism ,Induced Pluripotent Stem Cells ,iPSC-derived neurons ,Mice ,Optic Atrophy ,Insulin-Secreting Cells ,Internal Medicine ,Humans ,Animals ,Exenatide ,Human pancreatic beta cells ,iPSC-derived beta cells - Abstract
Aims/hypothesis Wolfram syndrome is a rare autosomal recessive disorder caused by pathogenic variants in the WFS1 gene. It is characterised by insulin-dependent diabetes mellitus, optic nerve atrophy, diabetes insipidus, hearing loss and neurodegeneration. Considering the unmet treatment need for this orphan disease, this study aimed to evaluate the therapeutic potential of glucagon-like peptide 1 receptor (GLP-1R) agonists under wolframin (WFS1) deficiency with a particular focus on human beta cells and neurons. Methods The effect of the GLP-1R agonists dulaglutide and exenatide was examined in Wfs1 knockout mice and in an array of human preclinical models of Wolfram syndrome, including WFS1-deficient human beta cells, human induced pluripotent stem cell (iPSC)-derived beta-like cells and neurons from control individuals and individuals affected by Wolfram syndrome, and humanised mice. Results Our study shows that the long-lasting GLP-1R agonist dulaglutide reverses impaired glucose tolerance in WFS1-deficient mice, and that exenatide and dulaglutide improve beta cell function and prevent apoptosis in different human WFS1-deficient models including iPSC-derived beta cells from people with Wolfram syndrome. Exenatide improved mitochondrial function, reduced oxidative stress and prevented apoptosis in Wolfram syndrome iPSC-derived neural precursors and cerebellar neurons. Conclusions/interpretation Our study provides novel evidence for the beneficial effect of GLP-1R agonists on WFS1-deficient human pancreatic beta cells and neurons, suggesting that these drugs may be considered as a treatment for individuals with Wolfram syndrome. Graphical abstract
- Published
- 2023
26. Impact of Proinflammatory Cytokines on Alternative Splicing Patterns in Human Islets
- Author
-
Raghavendra G. Mirmira, Edward Simpson, Farooq Syed, Yunlong Liu, Chih-Chun Lee, Wenting Wu, Chuanpeng Dong, Garrick Chang, Jing Liu, Carmella Evans-Molina, Clayton Seitz, and Decio L. Eizirik
- Subjects
biology ,Endocrinology, Diabetes and Metabolism ,Alternative splicing ,RNA-binding protein ,Epitope ,MHC Class II Gene ,Cell biology ,Proinflammatory cytokine ,Exon ,Islet Studies ,MHC class I ,RNA splicing ,Internal Medicine ,biology.protein - Abstract
Alternative splicing (AS) within the β-cell has been proposed as one potential pathway that may exacerbate autoimmunity and unveil novel immunogenic epitopes in type 1 diabetes (T1D). We used a computational strategy to prioritize pathogenic splicing events in human islets treated with interleukin-1β plus interferon-γ as an ex vivo model of T1D and coupled this analysis with a k-mer–based approach to predict RNA-binding proteins involved in AS. In total, 969 AS events were identified in cytokine-treated islets, with a majority (44.8%) involving a skipped exon. ExonImpact identified 129 events predicted to affect protein structure. AS occurred with high frequency in MHC class II–related mRNAs, and targeted quantitative PCR validated reduced inclusion of exon 5 in the MHC class II gene HLA-DMB. Single-molecule RNA fluorescence in situ hybridization confirmed increased HLA-DMB splicing in β-cells from human donors with established T1D and autoantibody positivity. Serine/arginine-rich splicing factor 2 was implicated in 37.2% of potentially pathogenic events, including exon 5 exclusion in HLA-DMB. Together, these data suggest that dynamic control of AS plays a role in the β-cell response to inflammatory signals during T1D evolution.
- Published
- 2021
- Full Text
- View/download PDF
27. The power of TOPMed imputation for the discovery of Latino enriched rare variants associated with type 2 diabetes
- Author
-
Alicia Huerta-Chagoya, Philip Schroeder, Ravi Mandla, Aaron J. Deutsch, Wanying Zhu, Lauren Petty, Xiaoyan Yi, Joanne B. Cole, Miriam S. Udler, Peter Dornbos, Bianca Porneala, Daniel DiCorpo, Ching-Ti Liu, Josephine H. Li, Lukasz Szczerbiński, Varinderpal Kaur, Joohyun Kim, Yingchang Lu, Alicia Martin, Decio L. Eizirik, Piero Marchetti, Lorella Marselli, Ling Chen, Shylaja Srinivasan, Jennifer Todd, Jason Flannick, Rose Gubitosi-Klug, Lynne Levitsky, Rachana Shah, Megan Kelsey, Brian Burke, Dana M. Dabelea, Jasmin Divers, Santica Marcovina, Lauren Stalbow, Ruth J.F. Loos, Burcu F. Darst, Charles Kooperberg, Laura M. Raffield, Christopher Haiman, Quan Sun, Joseph B. McCormick, Susan P. Fisher-Hoch, Maria L. Ordoñez, James Meigs, Leslie J. Baier, Clicerio González-Villalpando, Maria Elena González-Villalpando, Lorena Orozco, Andrés Moreno, Carlos A. Aguilar-Salinas, Teresa Tusié, Josée Dupuis, Maggie C.Y. Ng, Alisa Manning, Heather M. Highland, Miriam Cnop, Robert Hanson, Jennifer Below, Jose C. Florez, Aaron Leong, and Josep M. Mercader
- Abstract
HypothesisThe prevalence of type 2 diabetes is higher in Latino populations compared with other major ancestry groups. Not only has the Latino population been systematically underrepresented in large-scale genetic analyses, but previous studies relied on the imputation of ungenotyped variants based on the 1000 Genomes (1000G) imputation reference panel, which results in suboptimal capture of low-frequency or Latino-enriched variants. The NHLBI Trans-Omics for Precision Medicine (TOPMed) reference panel represents a unique opportunity to analyze rare genetic variations in the Latino population.MethodsWe evaluate the TOPMed imputation performance using genotyping array and whole-exome sequence data in 6 Latino cohorts. To evaluate the ability of TOPMed imputation of increasing the identified loci, we performed a Latino type 2 diabetes GWAS meta-analysis in 8,150 type 2 diabetes cases and 10,735 controls and replicated the results in 6 additional cohorts including whole-genome sequence data from the All of Us cohort.ResultsWe show that, compared to imputation with 1000G, the TOPMed panel improves the identification of rare and low-frequency variants. We identified 26 distinct signals including a novel genome-wide significant variant (minor allele frequency 1.6%, OR=2.0, P=3.4×10−9) near ORC5. A Latino-tailored polygenic score constructed from our data and GWAS data from East Asian and European populations improves the prediction accuracy in a Latino target dataset, explaining up to 7.6% of the type 2 diabetes risk variance.ConclusionsOur results demonstrate the utility of TOPMed imputation for identifying low-frequency variation in understudied populations, leading to the discovery of novel disease associations and the improvement of polygenic scores.
- Published
- 2022
- Full Text
- View/download PDF
28. The impact of interferon-[alpha] on global gene expression in iPSC-derived [beta]- and [alpha]-like cells
- Author
-
Sandra, Marin-Canas, primary, Florian, Szymczak, additional, Maria, Ines Alvelos, additional, Stephane, Demine, additional, Maikel, L Colli, additional, de, Beeck Anne Op, additional, Sofia, Thomaidou, additional, Lorella, Marselli, additional, Arnaud, Zaldumbide, additional, Piero, Marchetti, additional, and Decio, L Eizirik, additional
- Published
- 2022
- Full Text
- View/download PDF
29. Exercise as a non-pharmacological intervention to protect pancreatic beta cells in patients with type 1 and type 2 diabetes
- Author
-
de, Brachene Alexandra Coomans, primary, Corentin, Scoubeau, additional, Anyishai, E Musuaya, additional, Angela, Castela, additional, Julie, Carpentier, additional, Vitalie, Faoro, additional, Malgorzata, Klass, additional, Miriam, Cnop, additional, and Decio, L Eizirik, additional
- Published
- 2022
- Full Text
- View/download PDF
30. 100 YEARS OF INSULIN: 100 years of insulin: progress, current perspectives and future challenges
- Author
-
James Cantley and Decio L. Eizirik
- Subjects
medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Insulin ,medicine.medical_treatment ,History, 20th Century ,History, 21st Century ,Endocrinology ,Internal medicine ,Insulin Secretion ,Diabetes Mellitus ,medicine ,Humans ,Current (fluid) ,Intensive care medicine ,business - Published
- 2022
- Full Text
- View/download PDF
31. 9-OR: ADA Presidents' Select Abstract: m6A mRNA Methylation Regulates the Innate Immune Response in Human Beta Cells
- Author
-
DARIO F. DE JESUS, ZIJIE ZHANG, NATALIE K. BROWN, GIORGIO BASILE, LING XIAO, SEVIM KAHRAMAN, CLAYTON E. MATHEWS, ALVIN C. POWERS, MARK A. ATKINSON, DECIO L. EIZIRIK, and ROHIT KULKARNI
- Subjects
Endocrinology, Diabetes and Metabolism ,Internal Medicine - Abstract
Innate immunity is directly associated with Type 1 diabetes (T1D) . Recently, N6-methyladenosine (m6A) has been shown to modulate human β-cell biology and innate immunity. Here, we focused on the contribution of m6A in modulating T1D development. RNA-seq. in β-cells revealed downregulation of the m6A writers (Mettl3, Mettl14, and Wtap) in pre-diabetic NOD mice. Consistently, pancreases from female NOD mice and human T1D showed downregulation of METTL3 protein levels in insulin-positive cells as the disease progressed. m6A-sequencing in human T1D versus control islets and intersection with differentially expressed genes (DEGs) in T1D β-cells from a single-cell RNA seq. dataset, revealed enriched pathways associated with ER function, T1D, and apoptosis. Pro-inflammatory cytokines are directly linked to β-cell autoimmunity and are considered a useful tool to study T1D onset. We applied m6A-sequencing in cytokine-treated human islet preparations, and in parallel studies, treated EndoC-βH1 cells with cytokines followed by RNA-sequencing. Intersection of m6A-decorated and common DEGs in human islets and EndoC-βH1 cells revealed enrichment in pathways associated with the innate immune response. METTL3 silencing in EndoC-βH1 impacted several innate immune mediators upon cytokine challenge. Incubation of human islets from multiple donors with ER-stress inducers, ROS donors, NO donors, and ROS or NO scavengers revealed that pre-existing ER stress and ROS decrease METTL3, while physiological levels of NO upregulate METTL3. We conclude that ROS drives METTL3 downregulation associated with T1D. Exploring therapeutic relevance by AAV-8 mediated gene delivery showed that β-cell specific upregulation of Mettl3 leads to a decrease in hyperglycemia during early stages of T1D in female NOD mice. Together, these results demonstrate a novel layer of regulation in human β-cells in the context of T1D and point to targeting METTL3 as a therapeutic approach to promote β-cell survival and improve glycemia. Disclosure D.F.De jesus: n/a. D.L.Eizirik: None. R.Kulkarni: Advisory Panel; Biomea, Novo Nordisk, REDD Pharma, Consultant; Relay Therapeutics, Research Support; Inversago, SerPlus. Z.Zhang: Research Support; SinoVac. N.K.Brown: None. G.Basile: None. L.Xiao: None. S.Kahraman: Employee; Boehringer Ingelheim International GmbH. C.E.Mathews: None. A.C.Powers: None. M.A.Atkinson: None. Funding American Diabetes Association (7-21-PDF-140) ; R01 DK067536R01 DK103215R01 DK117639
- Published
- 2022
- Full Text
- View/download PDF
32. SUMOylation of Pdia3 exacerbates proinsulin misfolding and ER stress in pancreatic beta cells
- Author
-
Na Li, Ping Yang, Xinqiang Wang, Qilin Yu, Cong-Yi Wang, Shu Zhang, Jipin Jiang, Fei Xiong, Zhiguang Zhou, Zhishui Chen, Quan Gong, Decio L. Eizirik, Jiajun Zhao, Jing Xu, and Xi Luo
- Subjects
Proteomics ,Protein Folding ,Génétique clinique ,Protein Disulfide-Isomerases ,SUMO protein ,PDIA3 ,Pharmacologie ,Endoplasmic Reticulum ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Insulin-Secreting Cells ,Drug Discovery ,Animals ,Humans ,Pdia3 ,Protein disulfide-isomerase ,Genetics (clinical) ,Proinsulin ,Proinsulin misfolding ,Chemistry ,Endoplasmic reticulum ,Sumoylation ,Biologie moléculaire ,Endoplasmic Reticulum Stress ,Subcellular localization ,SUMOylation ,Cell biology ,Beta cell ,Unfolded protein response ,Molecular Medicine ,ER stress ,030215 immunology - Abstract
SUMOylation has long been recognized to regulate multiple biological processes in pancreatic beta cells, but its impact on proinsulin disulfide maturation and endoplasmic reticulum (ER) stress remains elusive. Herein, we conducted comparative proteomic analyses of SUMOylated proteins in primary mouse/human islets following proinflammatory cytokine stimulation. Cytokine challenge rendered beta cells to undergo a SUMOylation turnover manifested by the changes of SUMOylation substrates and SUMOylation levels for multiple substrates. Our data support that SUMOylation may play a crucial role to regulate proinsulin misfolding and ER stress at least by targeting Protein Disulfide Isomerase a3 (Pdia3). SUMOylation regulates Pdia3 enzymatic activity, subcellular localization, and protein binding ability. Furthermore, SUMOylation of Pdia3 exacerbated proinsulin misfolding and ER stress, and repressed Stat3 activation. In contrast, disruption of Pdia3 SUMOylation markedly rescued the outcomes. Collectively, our study expands the understanding how SUMOylation regulates ER stress in beta cells, which shed light on developing potential strategies against beta cell dysfunction., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2020
- Full Text
- View/download PDF
33. Presumption of innocence for beta cells: why are they vulnerable autoimmune targets in type 1 diabetes?
- Author
-
Decio L. Eizirik and Roberto Mallone
- Subjects
Islet ,0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,T cell ,Autoimmunity ,030209 endocrinology & metabolism ,Inflammation ,Review ,Disease ,CD8-Positive T-Lymphocytes ,Biology ,medicine.disease_cause ,Endotype ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Métabolisme ,Insulin-Secreting Cells ,Insulin Secretion ,Internal Medicine ,medicine ,Humans ,Insulin ,Coxsackievirus ,Genetic Predisposition to Disease ,Diabétologie ,Type 1 diabetes ,Secretory Vesicles ,16. Peace & justice ,medicine.disease ,Endocrinologie ,3. Good health ,Médecine interne ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,medicine.anatomical_structure ,Benign ,Antigen ,Immunology ,Unfolded Protein Response ,Epitope ,Beta cell ,medicine.symptom ,CD8 ,Proinsulin - Abstract
It is increasingly appreciated that the pathogenic mechanisms of type 1 diabetes involve both the autoimmune aggressors and their beta cell targets, which engage in a conflicting dialogue within and possibly outside the pancreas. Indeed, autoimmune CD8+ T cells, which are the final mediators of beta cell destruction, circulate at similar frequencies in type 1 diabetic and healthy individuals. Hence a universal state of ‘benign’ islet autoimmunity exists, and we hypothesise that its progression to type 1 diabetes may at least partially rely on a higher vulnerability of beta cells, which play a key, active role in disease development and/or amplification. We posit that this autoimmune vulnerability is rooted in some features of beta cell biology: the stress imposed by the high rate of production of insulin and other granule proteins, their dense vascularisation and the secretion of their products directly into the bloodstream. Gene variants that may predispose individuals to this vulnerability have been identified, e.g. MDA5, TYK2, PTPN2. They interact with environmental cues, such as viral infections, that may drive this genetic potential towards exacerbated local inflammation and progressive beta cell loss. On top of this, beta cells set up compensatory responses, such as the unfolded protein response, that become deleterious in the long term. The relative contribution of immune and beta cell drivers may vary and phenotypic subtypes (endotypes) are likely to exist. This dual view argues for the use of circulating biomarkers of both autoimmunity and beta cell stress for disease staging, and for the implementation of both immunomodulatory and beta cell-protective therapeutic strategies. [Figure not available: see fulltext.], SCOPUS: re.j, info:eu-repo/semantics/published
- Published
- 2020
- Full Text
- View/download PDF
34. An integrated multi-omics approach identifies the landscape of interferon-α-mediated responses of human pancreatic beta cells
- Author
-
Mireia Ramos-Rodríguez, Lorenzo Pasquali, Decio L. Eizirik, Knut Dahl-Jørgensen, Lars Krogvold, Helena Raurell-Vila, Jean-Valery Turatsinze, Sarah J. Richardson, Jonàs Juan-Mateu, Jessica L. E. Hill, Alexandra Coomans de Brachène, Piero Marchetti, Maikel Luis Colli, Mark Russell, Noel G. Morgan, Angela Castela, Bobbie-Jo M. Webb-Robertson, Thomas O. Metz, Lorella Marselli, Maria Inês Alvelos, Miguel Lopes, and Ernesto S. Nakayasu
- Subjects
0301 basic medicine ,Cell death ,Proteomics ,Science ,General Physics and Astronomy ,General Biochemistry, Genetics and Molecular Biology ,Chromatin remodeling ,Article ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,Antigen ,Interferon ,Insulin-Secreting Cells ,MHC class I ,medicine ,Data Mining ,Humans ,Gene Regulatory Networks ,Protein Interaction Maps ,Beta (finance) ,lcsh:Science ,health care economics and organizations ,Cells, Cultured ,Multidisciplinary ,biology ,Alternative splicing ,Interferon-alpha ,General Chemistry ,Chromatin ,3. Good health ,Cell biology ,Alternative Splicing ,030104 developmental biology ,Type 1 diabetes ,Diabetes Mellitus, Type 1 ,Gene Expression Regulation ,biology.protein ,lcsh:Q ,Beta cell ,Transcription Initiation Site ,Systems biology ,030217 neurology & neurosurgery ,medicine.drug ,Cell signalling ,Transcription Factors - Abstract
Interferon-α (IFNα), a type I interferon, is expressed in the islets of type 1 diabetic individuals, and its expression and signaling are regulated by T1D genetic risk variants and viral infections associated with T1D. We presently characterize human beta cell responses to IFNα by combining ATAC-seq, RNA-seq and proteomics assays. The initial response to IFNα is characterized by chromatin remodeling, followed by changes in transcriptional and translational regulation. IFNα induces changes in alternative splicing (AS) and first exon usage, increasing the diversity of transcripts expressed by the beta cells. This, combined with changes observed on protein modification/degradation, ER stress and MHC class I, may expand antigens presented by beta cells to the immune system. Beta cells also up-regulate the checkpoint proteins PDL1 and HLA-E that may exert a protective role against the autoimmune assault. Data mining of the present multi-omics analysis identifies two compound classes that antagonize IFNα effects on human beta cells., The cytokine IFNα is expressed in the islets of individuals with type 1 diabetes and contributes to local inflammation and destruction of beta cells. Here, the authors provide a global multiomics view of IFNα-induced changes in human beta cells at the level of chromatin, mRNA and protein expression.
- Published
- 2020
- Full Text
- View/download PDF
35. Extracellular HMGB1 exacerbates autoimmune progression and recurrence of type 1 diabetes by impairing regulatory T cell stability
- Author
-
Bao Ling Adam, Kun Huang, Jiahui Luo, Decio L. Eizirik, Yang Li, Cong-Yi Wang, Fei Xiong, Zhiguang Zhou, Fei Sun, Yuan Zou, Jing Zhang, Zhishui Chen, Qilin Yu, Faheem Ahmed Khan, Faxi Wang, Ping Yang, Longmin Chen, Jingyi Li, Jing Liu, Jinxiu Li, and Shu Zhang
- Subjects
0301 basic medicine ,Endocrinology, Diabetes and Metabolism ,Islets of Langerhans Transplantation ,medicine.disease_cause ,T-Lymphocytes, Regulatory ,Autoimmunity ,Mice ,Phosphatidylinositol 3-Kinases ,0302 clinical medicine ,Mice, Inbred NOD ,Islet transplantation ,HMGB1 Protein ,Cells, Cultured ,NOD mice ,HMGB1 ,Mice, Inbred BALB C ,FOXP3 ,Regulatory T cells ,Colitis ,Endocrinologie ,Médecine interne ,Type 1 diabetes ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Female ,Regulatory T cell ,Blotting, Western ,chemical and pharmacologic phenomena ,Article ,03 medical and health sciences ,High-mobility group box 1 ,Immune system ,Métabolisme ,Diabetes mellitus ,Internal Medicine ,medicine ,Animals ,Humans ,Diabetes reversal ,PI3K/AKT/mTOR pathway ,Diabétologie ,business.industry ,Beta cell mass turnover ,medicine.disease ,Antibodies, Neutralizing ,Diabetes Mellitus, Type 1 ,030104 developmental biology ,Immunology ,business - Abstract
Aims/hypothesis: High-mobility group box 1 (HMGB1), an evolutionarily conserved chromosomal protein, was rediscovered to be a ‘danger signal’ (alarmin) that alerts the immune system once released extracellularly. Therefore, it has been recognised contributing to the pathogenesis of autoimmune diabetes, but its exact impact on the initiation and progression of type 1 diabetes, as well as the related molecular mechanisms, are yet to be fully characterised. Methods: In the current report, we employed NOD mice as a model to dissect the impact of blocking HMGB1 on the prevention, treatment and reversal of type 1 diabetes. To study the mechanism involved, we extensively examined the characteristics of regulatory T cells (Tregs) and their related signalling pathways upon HMGB1 stimulation. Furthermore, we investigated the relevance of our data to human autoimmune diabetes. Results: Neutralising HMGB1 both delayed diabetes onset and, of particular relevance, reversed diabetes in 13 out of 20 new-onset diabetic NOD mice. Consistently, blockade of HMGB1 prevented islet isografts from autoimmune attack in diabetic NOD mice. Using transgenic reporter mice that carry a Foxp3 lineage reporter construct, we found that administration of HMGB1 impairs Treg stability and function. Mechanistic studies revealed that HMGB1 activates receptor for AGE (RAGE) and toll-like receptor (TLR)4 to enhance phosphatidylinositol 3-kinase (PI3K)–Akt–mechanistic target of rapamycin (mTOR) signalling, thereby impairing Treg stability and functionality. Indeed, high circulating levels of HMGB1 in human participants with type 1 diabetes contribute to Treg instability, suggesting that blockade of HMGB1 could be an effective therapy against type 1 diabetes in clinical settings. Conclusions/interpretation: The present data support the possibility that HMGB1 could be a viable therapeutic target to prevent the initiation, progression and recurrence of autoimmunity in the setting of type 1 diabetes., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2020
- Full Text
- View/download PDF
36. The gene signatures of human alpha cells in types 1 and 2 diabetes indicate disease-specific pathways of alpha cell dysfunction
- Author
-
Emanuele Bosi, Piero Marchetti, Guy A. Rutter, and Decio L. Eizirik
- Subjects
History ,endocrine system ,Polymers and Plastics ,endocrine system diseases ,nutritional and metabolic diseases ,Business and International Management ,Industrial and Manufacturing Engineering - Abstract
Glucagon secretion is perturbed in both type 1 and type 2 diabetes (T1D, T2D) the pathophysiological changes at the level of individual pancreatic alpha cells are still largely obscure. Using recently-curated single-cell RNA data from human donors with either T1D or T2D and appropriate controls, we leveraged alpha cell transcriptomic alterations consistent with both common and discrete pathways. Firstly, altered expression of genes associated with alpha cell identity (ARX, MAFB) was common to both diseases. In contrast, increased expression of cytokine-regulated genes and genes involved in glucagon biosynthesis and processing were apparent in T1D, whereas mitochondrial genes associated with reactive oxygen species generation (COX7B, NQO2) were dysregulated in alpha cells from T2D patients. Conversely, T1D alpha cells displayed alterations in genes associated with autoimmune-induced ER stress (ERLEC1, HSP90) whilst those from T2D patients showed changes in glycolytic and citrate cycle genes (LDH, PDHB, PDK4) which were unaffected in T1D. These findings suggest that despite some similarities related to loss-of-function, the alterations of alpha cells present important disease-specific signatures, suggesting that they are secondary to the main pathogenic events characteristic to each disease, namely immune-mediated-or metabolic-mediated-stress in respectively T1D and T2D.
- Published
- 2022
- Full Text
- View/download PDF
37. Recognition of mRNA Splice Variant and Secretory Granule Epitopes by CD4+ T Cells in Type 1 Diabetes
- Author
-
Perrin Guyer, David Arribas-Layton, Anthony Manganaro, Cate Speake, Sandra Lord, Decio L. Eizirik, Sally C. Kent, Roberto Mallone, Eddie A. James, University of Massachusetts Medical School [Worcester] (UMASS), University of Massachusetts System (UMASS), Benaroya Research Institute [Seattle] (BRI), Université libre de Bruxelles (ULB), Institut Cochin (IC UM3 (UMR 8104 / U1016)), Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Université Paris Cité (UPCité), Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), and Mallone, Roberto
- Subjects
CD4-Positive T-Lymphocytes ,epitope ,splice variant ,Endocrinology, Diabetes and Metabolism ,Secretory Vesicles ,Epitopes, T-Lymphocyte ,CD4+ T cells ,Type 1 diabetes ,Diabetes Mellitus, Type 1 ,Cyclin I ,[SDV.IMM.IA]Life Sciences [q-bio]/Immunology/Adaptive immunology ,[SDV.IMM.IA] Life Sciences [q-bio]/Immunology/Adaptive immunology ,Internal Medicine ,beta cell stress ,Humans ,secretory granule ,Peptides - Abstract
A recent discovery effort resulted in identification of novel splice variant and secretory granule antigens within the HLA class I peptidome of human islets and documentation of their recognition by CD8+ T cells from peripheral blood and human islets. In the current study, we applied a systematic discovery process to identify novel CD4+ T cell epitopes derived from these candidate antigens. We predicted 145 potential epitopes spanning unique splice junctions and within conventional secretory granule antigens and measured their in vitro binding to DRB1*04:01. We generated HLA class II tetramers for the 35 peptides with detectable binding and used these to assess immunogenicity and isolate T cell clones. Tetramers corresponding to peptides with verified immunogenicity were then used to label T cells specific for these putative epitopes in peripheral blood. T cells that recognize distinct epitopes derived from a cyclin I splice variant, neuroendocrine convertase 2, and urocortin-3 were detected at frequencies that were similar to those of an immunodominant proinsulin epitope. Cells specific for these novel epitopes predominantly exhibited a Th1-like surface phenotype. Among the three epitopes, responses to the cyclin I peptide exhibited a distinct memory profile. Responses to neuroendocrine convertase 2 were detected among pancreatic infiltrating T cells. These results further establish the contribution of unconventional antigens to the loss of tolerance in autoimmune diabetes.
- Published
- 2022
- Full Text
- View/download PDF
38. A single-cell human islet interactome atlas identifies disrupted autocrine and paracrine communications in type 2 diabetes
- Author
-
Emanuele Bosi, Lorella Marselli, Mara Suleiman, Marta Tesi, Carmela De Luca, Silvia Del Guerra, Miriam Cnop, Decio L Eizirik, and Piero Marchetti
- Subjects
Structural Biology ,Applied Mathematics ,Genetics ,Molecular Biology ,Computer Science Applications - Abstract
A sensible control of hormone secretion from pancreatic islets requires concerted inter-cellular communications, but a comprehensive picture of the whole islet interactome is presently missing. Single-cell transcriptomics allows to overcome this and we used here a single-cell dataset from type 2 diabetic (T2D) and non-diabetic (ND) donors to leverage islet interaction networks. The single-cell dataset contains 3046 cells classified in 7 cell types. The interactions across cell types in T2D and ND were obtained and resulting networks analysed to identify high-centrality genes and altered interactions in T2D. The T2D interactome displayed a higher number of interactions (10 787) than ND (9707); 1289 interactions involved beta cells (1147 in ND). High-centrality genes included EGFR, FGFR1 and FGFR2, important for cell survival and proliferation. In conclusion, this analysis represents the first in silico model of the human islet interactome, enabling the identification of signatures potentially relevant for T2D pathophysiology.
- Published
- 2022
- Full Text
- View/download PDF
39. The Role of Beta Cell Recovery in Type 2 Diabetes Remission
- Author
-
Mara Suleiman, Lorella Marselli, Miriam Cnop, Decio L. Eizirik, Carmela De Luca, Francesca R. Femia, Marta Tesi, Silvia Del Guerra, and Piero Marchetti
- Subjects
Inorganic Chemistry ,Diabetes Mellitus, Type 2 ,Insulin-Secreting Cells ,Organic Chemistry ,Insulin Secretion ,Humans ,Insulin ,General Medicine ,Physical and Theoretical Chemistry ,Molecular Biology ,Spectroscopy ,Catalysis ,Computer Science Applications - Abstract
Type 2 diabetes (T2D) has been considered a relentlessly worsening disease, due to the progressive deterioration of the pancreatic beta cell functional mass. Recent evidence indicates, however, that remission of T2D may occur in variable proportions of patients after specific treatments that are associated with recovery of beta cell function. Here we review the available information on the recovery of beta cells in (a) non-diabetic individuals previously exposed to metabolic stress; (b) T2D patients following low-calorie diets, pharmacological therapies or bariatric surgery; (c) human islets isolated from non-diabetic organ donors that recover from “lipo-glucotoxic” conditions; and (d) human islets isolated from T2D organ donors and exposed to specific treatments. The improvement of insulin secretion reported by these studies and the associated molecular traits unveil the possibility to promote T2D remission by directly targeting pancreatic beta cells.
- Published
- 2022
- Full Text
- View/download PDF
40. T1DBase: integration and presentation of complex data for type 1 diabetes research.
- Author
-
Erin M. Hulbert, Luc J. Smink, Ellen C. Adlem, James E. Allen, David B. Burdick, Oliver S. Burren, Christopher C. Cavnor, Geoffrey E. Dolman, Daisy Flamez, Karen F. Friery, Barry C. Healy, Sarah A. Killcoyne, Burak Kutlu, Helen Schuilenburg, Neil M. Walker, Josyf Mychaleckyj, Decio L. Eizirik, Linda S. Wicker, John A. Todd, and Nathan Goodman
- Published
- 2007
- Full Text
- View/download PDF
41. T1DBase, a community web-based resource for type 1 diabetes research.
- Author
-
Luc J. Smink, Erin M. Helton, Barry C. Healy, Christopher C. Cavnor, Alex C. Lam, Daisy Flamez, Oliver S. Burren, Yang Wang, Geoffrey E. Dolman, David B. Burdick, Vincent H. Everett, Gustavo Glusman, Davide Laneri, Lee Rowen, Helen Schuilenburg, Neil M. Walker, Josyf Mychaleckyj, Linda S. Wicker, Decio L. Eizirik, John A. Todd, and Nathan Goodman
- Published
- 2005
- Full Text
- View/download PDF
42. CD8+ T Cells Variably Recognize Native versus Citrullinated GRP78 Epitopes in Type 1 Diabetes
- Author
-
Maki Nakayama, Maikel Luis Colli, Yann Verdier, Noemi Brusco, Lut Overbergh, Mark J. Mamula, Aïsha Callebaut, Joëlle Vinh, Guido Sebastiani, Georgia Afonso, Laura Nigi, Matthieu Giraud, Barbara Brandao, Zhicheng Zhou, Sylvaine You, Alexia Carré, Mijke Buitinga, Sheena Pinto, Marie Eliane Azoury, Decio L. Eizirik, Francesco Dotta, Chantal Mathieu, Søren Buus, Roberto Mallone, Fatoumata Samassa, Ana Ines Lalanne, Magali Irla, Spectrométrie de Masse Biologique et Protéomique (USR3149 / FRE2032) (SMBP), Ecole Superieure de Physique et de Chimie Industrielles de la Ville de Paris (ESPCI Paris), Université Paris sciences et lettres (PSL)-Université Paris sciences et lettres (PSL)-Centre National de la Recherche Scientifique (CNRS), and Université Paris sciences et lettres (PSL)
- Subjects
chemistry.chemical_classification ,0303 health sciences ,Endocrinology, Diabetes and Metabolism ,Citrullination ,030209 endocrinology & metabolism ,Peptide ,Biology ,Molecular biology ,Epitope ,03 medical and health sciences ,0302 clinical medicine ,chemistry ,PADI2 ,PADI4 ,Internal Medicine ,Cytotoxic T cell ,[SDV.IMM]Life Sciences [q-bio]/Immunology ,CD8 ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,NOD mice - Abstract
In type 1 diabetes, autoimmune β-cell destruction may be favored by neo-antigens harboring post-translational modifications such as citrullination. We studied the recognition of native and citrullinated glucose-regulated protein (GRP)78 peptides by CD8+ T cells. Citrullination modulated T-cell recognition and, to a lesser extent, HLA-A2 binding. GRP78-reactive CD8+ T cells circulated at similar frequencies in type 1 diabetic and healthy donors and preferentially recognized either native or citrullinated versions, without cross-reactivity. Rather, the preference for native GRP78 epitopes was associated with CD8+ T cells cross-reactive with bacterial mimotopes. In the pancreas, a dominant GRP78 peptide was instead preferentially recognized when citrullinated. To further clarify these recognition patterns, we considered the possibility of citrullination in the thymus. Citrullinating peptidyl-arginine deiminase (Padi) enzymes were expressed in murine and human medullary epithelial cells (mTECs), with citrullinated proteins detected in murine mTECs. However, Padi2 and Padi4 expression was diminished in mature mTECs from NOD mice versus C57BL/6 mice. We conclude that, on one hand, the CD8+ T-cell preference for native GRP78 peptides may be shaped by cross-reactivity with bacterial mimotopes. On the other hand, post-translational modifications may not invariably favor loss of tolerance because thymic citrullination, although impaired in NOD mice, may drive deletion of citrulline-reactive T cells.
- Published
- 2021
- Full Text
- View/download PDF
43. Recovery of human type 2 diabetes beta cell function associates with transcriptomic signatures that point to novel therapeutic targets
- Author
-
Mark Ibberson, Ugo Boggi, Carmela De Luca, Miriam Cnop, Emanuele Bosi, Mara Suleiman, Frédéric Burdet, Piero Marchetti, L Marselli, Silvia Del Guerra, Xiaoyan Yi, Paolo De Simone, Decio L. Eizirik, Marta Tesi, Alessandro Pocai, and Camille Kessler
- Subjects
Transcriptome ,endocrine system ,endocrine system diseases ,business.industry ,Diabetes mellitus ,Medicine ,Beta-cell Function ,Computational biology ,Human type ,business ,medicine.disease - Abstract
Remission of type 2 diabetes (T2D) may occur after very low-calorie diets or bariatric surgery, and is associated with improved pancreatic beta cell function. Here, we evaluated if T2D beta cell dysfunction can be rescued ex-vivo and which are the molecular mechanisms involved. Islets from 19 T2D donors were studied after isolation (“basal”) and following culture at 5.5 or 11.1 mmol/l glucose (“cultured”). We evaluated glucose-stimulated insulin secretion (GSIS) and transcriptomes by RNA sequencing, correlated insulin secretion changes (“cultured” vs “basal”) to global gene expression, and searched for potential therapeutic gene targets and compounds that mimic gene signatures of recovered beta cell function in T2D islets. GSIS improved in 12 out of 19 islet preparations from T2D donors after culture at 5.5 mmol/l glucose (insulin stimulation index increased from 1.4±0.1 to 2.3±0.2, p
- Published
- 2021
- Full Text
- View/download PDF
44. 287-OR: M6A mRNA Methylation Regulates the Innate Immune Response in Human ß-Cells
- Author
-
Dario F. De Jesus, Sevim Kahraman, Clayton E. Mathews, Mark A. Atkinson, Natalie K. Brown, Decio L. Eizirik, Alvin C. Powers, Rohit N. Kulkarni, Zijie Zhang, Chuan He, and Jiang Hu
- Subjects
Autoimmune disease ,Candidate gene ,Innate immune system ,Endocrinology, Diabetes and Metabolism ,Extrinsic apoptotic signaling pathway ,Biology ,medicine.disease ,Immune system ,Downregulation and upregulation ,Apoptosis ,Immunology ,Internal Medicine ,medicine ,MRNA methylation - Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by a progressive destruction of β-cells and several candidate genes associated with T1D have been linked to innate immunity. Recently, N6-methyladenosine (m6A) has been shown to modulate human β-cell biology and to target type I interferons. Here, we focused on the contribution of m6A mRNA methylation in modulating innate immunity and β-cell destruction. Immune cells contribute to β-cell apoptosis by the release of pro-inflammatory cytokines during the onset of T1D. We assessed whether m6A abundance or expression of m6A modulators was altered in human islets and EndoC-βH1 cells treated with cytokines to mimick T1D onset. LC-MS/MS analyses of human islets showed that treatment with IL-1β plus IFN-α for 48h increased m6A levels. Consistently, IL-1β plus IFN-α treatment of human islets and EndoC-βH1 induced upregulation of the m6A writer METTL3. Next, we applied m6A-sequencing in cytokine-treated human islet preparations, and in parallel studies, treated EndoC-βH1 cells followed by RNA-sequencing. Intersection of m6A-decorated genes in cytokine-treated human islets with common differentially expressed genes in human islets and EndoC-βH1 cells revealed enrichment in pathways associated with the innate immune response, negative regulation of immune response, and extrinsic apoptotic signaling pathway. Differentially expressed and m6A decorated genes included genes associated with T1D that are involved in innate immunity. Re-analyses of RNA-seq. data in sorted-β-cells from T1D patients revealed downregulation of METTL3. These data suggest that while healthy β-cells are able to upregulate m6A levels and METTL3 early in T1D (e.g., when they are exposed to cytokines), this response is lost in established T1D that allows an exacerbated immune response and β-cell death. Together, these results point to a possible therapeutic approach in targeting METTL3 to modulate innate immunity and promote β-cell survival. Disclosure D. F. De jesus: None. C. He: None. R. Kulkarni: None. Z. Zhang: None. N. K. Brown: None. J. Hu: None. S. Kahraman: None. C. E. Mathews: None. A. C. Powers: None. M. A. Atkinson: None. D. L. Eizirik: None. Funding National Institutes of Health (1UC4DK116278)
- Published
- 2021
- Full Text
- View/download PDF
45. TIGER: The gene expression regulatory variation landscape of human pancreatic islets
- Author
-
Ji Chen, David Torrents, Goutham Atla, Lorena Alonso, Lorella Marselli, Montserrat Puiggròs, Jason M. Torres, Jorge Ferrer, Hindrik Mulder, Matthieu Defrance, Vibe Nylander, Marta Guindo-Martínez, Miriam Cnop, Decio L. Eizirik, Sílvia Bonàs-Guarch, Romina Royo, Mara Suleiman, Piero Marchetti, Jonathan L.S. Esguerra, Irene Miguel-Escalada, Josep M. Mercader, Ignasi Moran, Xavier Garcia-Hurtado, Anthony Piron, Leif Groop, Jean-Valery Turatsinze, Anna L. Gloyn, Ramon Amela, and Lena Eliasson
- Subjects
endocrine system ,0303 health sciences ,geography ,geography.geographical_feature_category ,endocrine system diseases ,Pancreatic islets ,Genome-wide association study ,Computational biology ,Biology ,Islet ,3. Good health ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Genetic variation ,Genotype ,Expression quantitative trait loci ,medicine ,Allele ,Genotyping ,030217 neurology & neurosurgery ,030304 developmental biology - Abstract
GWAS have identified more than 700 genetic signals associated with type 2 diabetes (T2D). To gain insight into the underlying molecular mechanisms, we created the Translational human pancreatic Islet Genotype tissue-Expression Resource (TIGER), aggregating >500 human islet RNA-seq and genotyping datasets. We imputed genotypes using 4 reference panels and meta-analyzed cohorts to improve coverage of expression quantitative trait loci (eQTL) and developed a method to combine allele-specific expression across samples (cASE). We identified >1 million islet eQTLs (56% novel), of which 53 colocalize with T2D signals (60% novel). Among them, a low-frequency allele that reduces T2D risk by half increases CCND2 expression. We identified 8 novel cASE colocalizations, among which an SLC30A8 T2D associated variant. We make all the data available through the open-access TIGER portal (http://tiger.bsc.es), which represents a comprehensive human islet genomic data resource to elucidate how genetic variation affects islet function and translate this into therapeutic insight and precision medicine for T2D.
- Published
- 2021
- Full Text
- View/download PDF
46. Elevated Neuropeptide Y1 Receptor Signaling Contributes to β-cell Dysfunction and Failure in Type 2 Diabetes
- Author
-
Xuzhu Lin, Kim Loh, Alba Moreno-Asso, Chieh Hsin Yang, Ross Laybutt, Herbert Herzog, John W. Scott, Cassandra Smith, Stacey Fynch, Decio L. Eizirik, Liu X, Helen E. Thomas, Jonathan S. Oakhill, Evan Pappas, Nadarajah S, Yan-Chuan Shi, Sandra Galic, Loudovaris T, Itamar Levinger, and Ann-Onda D
- Subjects
medicine.medical_specialty ,geography ,geography.geographical_feature_category ,Chemistry ,Neuropeptide ,Type 2 diabetes ,Neuropeptide Y receptor ,medicine.disease ,Streptozotocin ,Islet ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,Diabetes mellitus ,medicine ,Beta (finance) ,medicine.drug - Abstract
Loss of functional β-cell mass is a key factor contributing to the poor glycaemic control in type 2 diabetes. However, therapies that directly target these underlying processes remains lacking. Here we demonstrate that gene expression of neuropeptide Y1 receptor and its ligand, neuropeptide Y, was significantly upregulated in human islets from subjects with type 2 diabetes. Importantly, the reduced insulin secretion in type 2 diabetes was associated with increased neuropeptide Y and Y1 receptor expression in human islets. Consistently, pharmacological inhibition of Y1 receptors by BIBO3304 significantly protected β-cells from dysfunction and death under multiple diabetogenic conditions in islets. In a preclinical study, Y1 receptor antagonist BIBO3304 treatment improved β-cell function and preserved functional β-cell mass, thereby resulting in better glycaemic control in both high-fat-diet/multiple low-dose streptozotocin- and db/db type 2 diabetic mice. Collectively, our results uncovered a novel causal link of increased islet NPY-Y1 receptor signaling to β-cell dysfunction and failure in human type 2 diabetes. These results further demonstrate that inhibition of Y1 receptor by BIBO3304 represents a novel and effective β-cell protective therapy for improving functional β-cell mass and glycaemic control in type 2 diabetes.
- Published
- 2021
- Full Text
- View/download PDF
47. Non-canonical mitochondrial STAT3 signaling mediates exercise-induced insulin secretion down-regulation
- Author
-
Jane Cristina de Souza, N. C. Leite, P. F. Paula, Antonio C. Boschero, Patricia Cristine Borck, Jonàs Juan-Mateu, Cláudio Cesar Zoppi, Everardo M. Carneiro, Jacques Jonas, C. Lubackzeuski, and Decio L. Eizirik
- Subjects
geography ,medicine.medical_specialty ,Gene knockdown ,geography.geographical_feature_category ,biology ,Chemistry ,Mitochondrion ,Islet ,In vitro ,Endocrinology ,Downregulation and upregulation ,Internal medicine ,biology.protein ,medicine ,Phosphorylation ,STAT3 ,Receptor - Abstract
Chronic exercise protects pancreatic beta cells from diabetogenic stress, reducing insulin secretion through unknown mechanisms. We tested the hypothesis that the IL-6/mitochondrial STAT3 (pS-STAT3) axis plays a role in this protective effect. C57BL/6N mice were subjected to endurance training ahead of pancreatic islet isolation and functional analysis. Similar in vitro experiments were performed using insulin-producing INS-1E cells and islets from untrained mice, cultured with serum from trained animals and treated with or without an IL-6 receptor (IL-6R) inhibitor. Then, IL-6R/pS-STAT3 pathway activation and its effects on mitochondrial function and insulin secretion were assessed. Exercise-induced down-regulation of insulin secretion was prevented by inhibition of IL-6R signaling and following STAT3 knockdown. IL-6R activation promoted STAT3 phosphorylation and translocation to the mitochondria, increasing oxygen consumption. Accordingly, lower H2O2 content was reported in islets from trained mice and beta cells exposed to exercise-conditioned serum, while exposure to exogenous H2O2 blocked the down-regulatory effect of training on insulin secretion. Similar findings were observed in islets from obese-trained mice. Together, these findings suggest that the IL-6R/pS-STAT3 axis mediates exercise-induced down-regulation of insulin secretion through modulation of the mitochondrial redox state.
- Published
- 2021
- Full Text
- View/download PDF
48. Correction to ‘Integration of single-cell datasets reveals novel transcriptomic signatures of β-cells in human type 2 diabetes’
- Author
-
Lorella Marselli, Decio L. Eizirik, Mara Suleiman, Carmela De Luca, Piero Marchetti, Miriam Cnop, Emanuele Bosi, Mark Ibberson, and Marta Tesi
- Subjects
AcademicSubjects/SCI01140 ,AcademicSubjects/SCI01060 ,business.industry ,AcademicSubjects/SCI00030 ,Cell ,Computational biology ,Human type ,Biology ,AcademicSubjects/SCI01180 ,medicine.disease ,Transcriptome ,medicine.anatomical_structure ,Text mining ,Diabetes mellitus ,medicine ,AcademicSubjects/SCI00980 ,Corrigendum ,business - Abstract
Pancreatic islet β-cell failure is key to the onset and progression of type 2 diabetes (T2D). The advent of single-cell RNA sequencing (scRNA-seq) has opened the possibility to determine transcriptional signatures specifically relevant for T2D at the β-cell level. Yet, applications of this technique have been underwhelming, as three independent studies failed to show shared differentially expressed genes in T2D β-cells. We performed an integrative analysis of the available datasets from these studies to overcome confounding sources of variability and better highlight common T2D β-cell transcriptomic signatures. After removing low-quality transcriptomes, we retained 3046 single cells expressing 27 931 genes. Cells were integrated to attenuate dataset-specific biases, and clustered into cell type groups. In T2D β-cells (
- Published
- 2021
- Full Text
- View/download PDF
49. CD8
- Author
-
Marie Eliane, Azoury, Fatoumata, Samassa, Mijke, Buitinga, Laura, Nigi, Noemi, Brusco, Aïsha, Callebaut, Matthieu, Giraud, Magali, Irla, Ana Ines, Lalanne, Alexia, Carré, Georgia, Afonso, Zhicheng, Zhou, Barbara, Brandao, Maikel L, Colli, Guido, Sebastiani, Francesco, Dotta, Maki, Nakayama, Decio L, Eizirik, Sylvaine, You, Sheena, Pinto, Mark J, Mamula, Yann, Verdier, Joelle, Vinh, Soren, Buus, Chantal, Mathieu, Lut, Overbergh, and Roberto, Mallone
- Subjects
Adult ,Male ,Adolescent ,Epitopes, T-Lymphocyte ,CD8-Positive T-Lymphocytes ,Middle Aged ,Lymphocyte Activation ,Mice, Inbred C57BL ,Mice ,Young Adult ,Diabetes Mellitus, Type 1 ,Mice, Inbred NOD ,Animals ,Humans ,Citrullination ,Female ,Immunology and Transplantation ,Child ,Endoplasmic Reticulum Chaperone BiP ,Protein Processing, Post-Translational - Abstract
In type 1 diabetes, autoimmune β-cell destruction may be favored by neoantigens harboring posttranslational modifications (PTMs) such as citrullination. We studied the recognition of native and citrullinated glucose-regulated protein (GRP)78 peptides by CD8(+) T cells. Citrullination modulated T-cell recognition and, to a lesser extent, HLA-A2 binding. GRP78-reactive CD8(+) T cells circulated at similar frequencies in healthy donors and donors with type 1 diabetes and preferentially recognized either native or citrullinated versions, without cross-reactivity. Rather, the preference for native GRP78 epitopes was associated with CD8(+) T cells cross-reactive with bacterial mimotopes. In the pancreas, a dominant GRP78 peptide was instead preferentially recognized when citrullinated. To further clarify these recognition patterns, we considered the possibility of citrullination in the thymus. Citrullinating peptidylarginine deiminase (Padi) enzymes were expressed in murine and human medullary epithelial cells (mTECs), with citrullinated proteins detected in murine mTECs. However, Padi2 and Padi4 expression was diminished in mature mTECs from NOD mice versus C57BL/6 mice. We conclude that, on one hand, the CD8(+) T cell preference for native GRP78 peptides may be shaped by cross-reactivity with bacterial mimotopes. On the other hand, PTMs may not invariably favor loss of tolerance because thymic citrullination, although impaired in NOD mice, may drive deletion of citrulline-reactive T cells.
- Published
- 2021
50. A functional genomic approach to identify reference genes for human pancreatic beta cell real-time quantitative RT-PCR analysis
- Author
-
Miriam Cnop, Ioannis Gkantounas, Piero Marchetti, Ângela Castela, Sandra Marín-Cañas, Florian Szymczak, Mariana Igoillo-Esteve, Decio L. Eizirik, Maikel Luis Colli, Maria Inês Alvelos, Federica Fantuzzi, Lorella Marselli, Cristina Cosentino, and Bianca Marmontel de Souza
- Subjects
Endocrinology, Diabetes and Metabolism ,Coefficient of variation ,medicine.medical_treatment ,Type 2 diabetes ,Biology ,Real-Time Polymerase Chain Reaction ,03 medical and health sciences ,Endocrinology ,Text mining ,0302 clinical medicine ,Rt pcr analysis ,Insulin-Secreting Cells ,Pancreatic beta Cells ,Reference genes ,Gene expression ,medicine ,Humans ,Beta (finance) ,Gene ,030304 developmental biology ,0303 health sciences ,Reverse Transcriptase Polymerase Chain Reaction ,business.industry ,Genomics ,medicine.disease ,Molecular biology ,Cytokine ,RAB7A ,030220 oncology & carcinogenesis ,Reference genes/ beta cells/ diabetes/ RNA-sequencing/ quantitative real-time pcr ,Beta cell ,business ,030217 neurology & neurosurgery ,Research Article ,Research Paper - Abstract
Exposure of human pancreatic beta cells to pro-inflammatory cytokines or metabolic stressors is used to model events related to type 1 and type 2 diabetes, respectively. Quantitative real-time PCR is commonly used to quantify changes in gene expression. The selection of the most adequate reference gene(s) for gene expression normalization is an important pre-requisite to obtain accurate and reliable results. There are no universally applicable reference genes, and the human beta cell expression of commonly used reference genes can be altered by different stressors. Here we aimed to identify the most stably expressed genes in human beta cells to normalize quantitative real-time PCR gene expression.We used comprehensive RNA-sequencing data from the human pancreatic beta cell line EndoC-βH1, human islets exposed to cytokines or the free fatty acid palmitate in order to identify the most stably expressed genes. Genes were filtered based on their level of significance (adjusted P-value >0.05), fold-change (|fold-change| We identified a total of 264 genes stably expressed in EndoC-βH1 cells and human islets following cytokine- or palmitate-induced stress, displaying a low coefficient of variation. Validation by quantitative real-time PCR of the top five genes ARF1, CWC15, RAB7A, SIAH1 and VAPA corroborated their expression stability under most of the tested conditions. Further validation in independent samples indicated that the geometric mean of ACTB and VAPA expression can be used as a reliable normalizing factor in human beta cells.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.