1
|
Chen S, Wu P, Zhang T, Zhang J, Gao H. Global scientific trends on the islet transplantation in the 21st century: A bibliometric and visualized analysis. Medicine (Baltimore) 2024; 103:e37945. [PMID: 38669398 PMCID: PMC11049693 DOI: 10.1097/md.0000000000037945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/29/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND Islet transplantation (IT) has emerged as a significant research area for the treatment of diabetes mellitus and has witnessed a surge in scholarly attention. Despite its growing importance, there is a lack of bibliometric analyses that encapsulate the evolution and scientific underpinnings of this field. This study aims to fill this gap by conducting a comprehensive bibliometric analysis to delineate current research hotspots and forecast future trajectories within the IT domain with a particular focus on evidence-based medicine practices. METHODS This analysis scrutinized literature from January 1, 2000, to October 1, 2023, using the Web of Science Core Collection (WoSCC). Employing bibliometric tools such as VOSviewer, CiteSpace, and the R package "bibliometrix," we systematically evaluated the literature to uncover scientific trends and collaboration networks in IT research. RESULTS The analysis revealed 8388 publications from 82 countries, predominantly the United States and China. However, global cross-institutional collaboration in IT research requires further strengthening. The number of IT-related publications has increased annually. Leading research institutions in this field include Harvard University, the University of Alberta, the University of Miami, and the University of Minnesota. "Transplantation" emerges as the most frequently cited journal in this area. Shapiro and Ricordi were the most prolific authors, with 126 and 121 publications, respectively. Shapiro also led to co-citations, totaling 4808. Key research focuses on IT sites and procedures as well as novel therapies in IT. Emerging research hotspots are identified by terms like "xenotransplantation," "apoptosis," "stem cells," "immunosuppression," and "microencapsulation." CONCLUSIONS The findings underscore a mounting anticipation for future IT research, which is expected to delve deeper into evidence-based methodologies for IT sites, procedures, and novel therapeutic interventions. This shift toward evidence-based medicine underscores the field's commitment to enhancing the efficacy and safety of IT for diabetes treatment, signaling a promising direction for future investigations aimed at optimizing patient outcomes.
Collapse
Affiliation(s)
- Sheng Chen
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - PeiZhong Wu
- Graduate School, Guangxi University of Chinese Medicine, Nanning, China
| | - Ting Zhang
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Jianqiang Zhang
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Hongjun Gao
- Ruikang Hospital, Guangxi University of Chinese Medicine, Nanning, China
| |
Collapse
|
2
|
Atla G, Bonàs-Guarch S, Cuenca-Ardura M, Beucher A, Crouch DJM, Garcia-Hurtado J, Moran I, Irimia M, Prasad RB, Gloyn AL, Marselli L, Suleiman M, Berney T, de Koning EJP, Kerr-Conte J, Pattou F, Todd JA, Piemonti L, Ferrer J. Genetic regulation of RNA splicing in human pancreatic islets. Genome Biol 2022; 23:196. [PMID: 36109769 PMCID: PMC9479353 DOI: 10.1186/s13059-022-02757-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 08/23/2022] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Non-coding genetic variants that influence gene transcription in pancreatic islets play a major role in the susceptibility to type 2 diabetes (T2D), and likely also contribute to type 1 diabetes (T1D) risk. For many loci, however, the mechanisms through which non-coding variants influence diabetes susceptibility are unknown. RESULTS We examine splicing QTLs (sQTLs) in pancreatic islets from 399 human donors and observe that common genetic variation has a widespread influence on the splicing of genes with established roles in islet biology and diabetes. In parallel, we profile expression QTLs (eQTLs) and use transcriptome-wide association as well as genetic co-localization studies to assign islet sQTLs or eQTLs to T2D and T1D susceptibility signals, many of which lack candidate effector genes. This analysis reveals biologically plausible mechanisms, including the association of T2D with an sQTL that creates a nonsense isoform in ERO1B, a regulator of ER-stress and proinsulin biosynthesis. The expanded list of T2D risk effector genes reveals overrepresented pathways, including regulators of G-protein-mediated cAMP production. The analysis of sQTLs also reveals candidate effector genes for T1D susceptibility such as DCLRE1B, a senescence regulator, and lncRNA MEG3. CONCLUSIONS These data expose widespread effects of common genetic variants on RNA splicing in pancreatic islets. The results support a role for splicing variation in diabetes susceptibility, and offer a new set of genetic targets with potential therapeutic benefit.
Collapse
Affiliation(s)
- Goutham Atla
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en red Diabetes y enfermedades metabólicas asociadas (CIBERDEM), Barcelona, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Silvia Bonàs-Guarch
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en red Diabetes y enfermedades metabólicas asociadas (CIBERDEM), Barcelona, Spain.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| | - Mirabai Cuenca-Ardura
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en red Diabetes y enfermedades metabólicas asociadas (CIBERDEM), Barcelona, Spain
| | - Anthony Beucher
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en red Diabetes y enfermedades metabólicas asociadas (CIBERDEM), Barcelona, Spain
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Daniel J M Crouch
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Javier Garcia-Hurtado
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro de Investigación Biomédica en red Diabetes y enfermedades metabólicas asociadas (CIBERDEM), Barcelona, Spain
| | - Ignasi Moran
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Present Address: Life Sciences Department, Barcelona Supercomputing Center (BSC), 08034, Barcelona, Spain
| | - Manuel Irimia
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Rashmi B Prasad
- Lund University Diabetes Centre, Clinical Research Center, Malmö, Sweden
- Department of Clinical Sciences in Malmö, Lund University, Malmö, Sweden
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
- Department of Pediatrics, Division of Endocrinology, Stanford School of Medicine, Stanford, CA, USA
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, AOUP Cisanello University Hospital, University of Pisa, Pisa, Italy
| | - Thierry Berney
- Cell Isolation and Transplantation Center, University of Geneva, Geneva, Switzerland
| | - Eelco J P de Koning
- Department of Medicine, Leiden University Medical Center, Leiden, the Netherlands
- Hubrecht Institute/KNAW, Utrecht, the Netherlands
| | - Julie Kerr-Conte
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), Institute Pasteur Lille, U1190 -European Genomic Institute for Diabetes (EGID), F59000, Lille, France
| | - Francois Pattou
- University of Lille, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospitalier Universitaire de Lille (CHU Lille), Institute Pasteur Lille, U1190 -European Genomic Institute for Diabetes (EGID), F59000, Lille, France
| | - John A Todd
- JDRF/Wellcome Diabetes and Inflammation Laboratory, Wellcome Centre for Human Genetics, Nuffield Department of Medicine, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Lorenzo Piemonti
- Diabetes Research Institute, IRCCS Ospedale San Raffaele and Università Vita-Salute San Raffaele, Milan, Italy
| | - Jorge Ferrer
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.
- Centro de Investigación Biomédica en red Diabetes y enfermedades metabólicas asociadas (CIBERDEM), Barcelona, Spain.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
3
|
Liu J, Bailbé D, Raynal S, Carbonne C, Zhen D, Dairou J, Gausseres B, Armanet M, Domet T, Pitasi CL, Movassat J, Lim CK, Guillemin GJ, Autier V, Kergoat M, Portha B. Kynurenine-3-monooxygenase expression is activated in the pancreatic endocrine cells by diabetes and its blockade improves glucose-stimulated insulin secretion. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166509. [PMID: 35914653 DOI: 10.1016/j.bbadis.2022.166509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 07/25/2022] [Accepted: 07/25/2022] [Indexed: 11/25/2022]
Abstract
Type 2 diabetes is associated with an inflammatory phenotype in the pancreatic islets. We previously demonstrated that proinflammatory cytokines potently activate the tryptophan/kynurenine pathway (TKP) in INS-1 cells and in normal rat islets. Here we examined: (1) the TKP enzymes expression in the diabetic GK islets; (2) the TKP enzymes expression profiles in the GK islets before and after the onset of diabetes; (3) The glucose-stimulated insulin secretion (GSIS) in vitro in GK islets after KMO knockdown using specific morpholino-oligonucleotides against KMO or KMO blockade using the specific inhibitor Ro618048; (4) The glucose tolerance and GSIS after acute in vivo exposure to Ro618048 in GK rats. We report a remarkable induction of the kmo gene in GK islets and in human islets exposed to proinflammatory conditions. It occurred prominently in beta cells. The increased expression and activity of KMO reflected an acquired adaptation. Both KMO knockdown and specific inhibitor Ro618048 enhanced GSIS in vitro in GK islets. Moreover, acute administration of Ro618048 in vivo improved glucose tolerance, GSIS and basal blood glucose levels in GK rats. These results demonstrate that targeting islet TKP is able to correct defective GSIS. KMO inhibition could represent a potential therapeutic strategy for type 2 diabetes.
Collapse
Affiliation(s)
- Junjun Liu
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France; Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong, China; MetaBrain Research, Maisons-Alfort, France.
| | - Danielle Bailbé
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France
| | | | | | - Delong Zhen
- Shandong Institute of Endocrine and Metabolic Diseases, Shandong First Medical University, Jinan, Shandong, China
| | - Julien Dairou
- Laboratoire de Chimie et Biochimie Pharmacologiques et Toxicologiques, CNRS UMR8601, Université Paris-Cité, Paris, France
| | - Blandine Gausseres
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France
| | - Mathieu Armanet
- Cell Therapy Unit, Hôpital Saint-Louis, AP-HP, Université Paris-Cité, Paris, France
| | - Thomas Domet
- Cell Therapy Unit, Hôpital Saint-Louis, AP-HP, Université Paris-Cité, Paris, France
| | - Caterina L Pitasi
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France
| | - Jamileh Movassat
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France
| | - Chai K Lim
- Neuroinflammation Group, Macquarie Medicine School, Macquarie University, Sydney, Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Macquarie Medicine School, Macquarie University, Sydney, Australia
| | | | | | - Bernard Portha
- Laboratoire B2PE (Biologie et Pathologie du Pancréas Endocrine), Unité BFA (Biologie Fonctionnelle et Adaptive), CNRS UMR 8251, Université Paris-Cité, Paris, France.
| |
Collapse
|
4
|
Rakotoarisoa L, Wagner C, Munch M, Renaud Picard B, Grenet D, Olland A, Greget M, Enescu I, Bouilloud F, Bonnette P, Guth A, Bosco D, Mercier C, Rabilloud M, Berney T, Yves Benhamou P, Massard G, Camilo C, Colin C, Arnold C, Kessler R, Kessler L. Feasibility and efficacy of combined pancreatic islet-lung transplantation in cystic fibrosis-related diabetes-PIM study: A multicenter phase 1-2 trial. Am J Transplant 2022; 22:1861-1872. [PMID: 35403818 PMCID: PMC9540675 DOI: 10.1111/ajt.17058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/21/2022] [Accepted: 04/04/2022] [Indexed: 01/25/2023]
Abstract
Cystic fibrosis-related diabetes (CFRD) is a common complication of cystic fibrosis (CF), and restoring metabolic control in these patients may improve their management after lung transplantation. In this multicenter, prospective, phase 1-2 trial, we evaluate the feasibility and metabolic efficacy of combined pancreatic islet-lung transplantation from a single donor in patients with CFRD, terminal respiratory failure, and poorly controlled diabetes. Islets were infused via the portal vein under local anesthesia, 1 week after lung transplantation. At 1 year, the primary outcome was transplant success as evaluated by a composite score including four parameters (weight, fasting glycemia, HbA1c, and insulin requirements). Ten participants (age: 24 years [17-31], diabetes duration: 8 years [4-12]) received a combined islet-lung transplant with 2892 IEQ/kg [2293-6185]. Transplant success was achieved in 7 out of 10 participants at 1-year post transplant. Fasting plasma C-peptide increased from 0.91 μg/L [0.56-1.29] to 1.15 μg/L [0.77-2.2], HbA1c decreased from 7.8% [6.5-8.3] (62 mmol/mol [48-67]) to 6.7% [5.5-8.0] (50 mmol/mol [37-64]), with 38% decrease in daily insulin doses. No complications related to the islet injection procedure were reported. In this pilot study, combined pancreatic islet-lung transplantation restored satisfactory metabolic control and pulmonary function in patients with CF, without increasing the morbidity of lung transplantation.
Collapse
Affiliation(s)
- Luc Rakotoarisoa
- Department of Endocrinology, Diabetes and NutritionStrasbourg University HospitalFrance,Inserm UMR 1260Regenerative NanomedicineStrasbourgFrance
| | - Clothilde Wagner
- Department of Endocrinology, Diabetes and NutritionStrasbourg University HospitalFrance
| | - Marion Munch
- Department of Endocrinology, Diabetes and NutritionStrasbourg University HospitalFrance
| | - Benjamin Renaud Picard
- Inserm UMR 1260Regenerative NanomedicineStrasbourgFrance,Department of PneumologyCystic Fibrosis CenterStrasbourg UniversityStrasbourgFrance
| | - Dominique Grenet
- Department of PneumologyCystic Fibrosis CenterHôpital FochSuresnesFrance
| | - Anne Olland
- Inserm UMR 1260Regenerative NanomedicineStrasbourgFrance,Department of Thoracic SurgeryStrasbourg University HospitalStrasbourgFrance
| | - Michel Greget
- Department of RadiologyStrasbourg University HospitalStrasbourgFrance
| | - Iulian Enescu
- Department of RadiologyStrasbourg University HospitalStrasbourgFrance
| | | | | | - Axel Guth
- Department of RadiologyHôpital FochSuresnesFrance
| | - Domenico Bosco
- Department of Surgery, Islet Isolation, and TransplantationGeneva University HospitalsGenevaSwitzerland
| | - Catherine Mercier
- Pôle Santé PubliqueService de Biostatistique et BioinformatiqueHospices Civils de Lyon, LyonFrance,Université de LyonLyonFrance,Université Lyon 1VilleurbanneFrance,Laboratoire de Biométrie et Biologie ÉvolutiveÉquipe Biostatistique‐SantéCNRSUMR 5558VilleurbanneFrance
| | - Muriel Rabilloud
- Pôle Santé PubliqueService de Biostatistique et BioinformatiqueHospices Civils de Lyon, LyonFrance,Université de LyonLyonFrance,Université Lyon 1VilleurbanneFrance,Laboratoire de Biométrie et Biologie ÉvolutiveÉquipe Biostatistique‐SantéCNRSUMR 5558VilleurbanneFrance
| | - Thierry Berney
- Department of Surgery, Islet Isolation, and TransplantationGeneva University HospitalsGenevaSwitzerland
| | - Pierre Yves Benhamou
- Department of Endocrinology, Diabetes and NutritionGrenoble University HospitalGrenobleFrance
| | - Gilbert Massard
- Inserm UMR 1260Regenerative NanomedicineStrasbourgFrance,Department of Thoracic SurgeryStrasbourg University HospitalStrasbourgFrance
| | - Coralie Camilo
- Pôle Santé PubliqueService de Biostatistique et BioinformatiqueHospices Civils de Lyon, LyonFrance,Université de LyonLyonFrance,Université Lyon 1VilleurbanneFrance,Laboratoire de Biométrie et Biologie ÉvolutiveÉquipe Biostatistique‐SantéCNRSUMR 5558VilleurbanneFrance
| | - Cyrille Colin
- Pôle Santé PubliqueService de Biostatistique et BioinformatiqueHospices Civils de Lyon, LyonFrance,Université de LyonLyonFrance,Université Lyon 1VilleurbanneFrance,Laboratoire de Biométrie et Biologie ÉvolutiveÉquipe Biostatistique‐SantéCNRSUMR 5558VilleurbanneFrance
| | - Cécile Arnold
- Department of Clinical ResearchStrasbourg University HospitalStrasbourgFrance
| | - Romain Kessler
- Inserm UMR 1260Regenerative NanomedicineStrasbourgFrance,Department of PneumologyCystic Fibrosis CenterStrasbourg UniversityStrasbourgFrance
| | - Laurence Kessler
- Department of Endocrinology, Diabetes and NutritionStrasbourg University HospitalFrance,Inserm UMR 1260Regenerative NanomedicineStrasbourgFrance
| | | |
Collapse
|
5
|
Brown MR, Laouteouet D, Delobel M, Villard O, Broca C, Bertrand G, Wojtusciszyn A, Dalle S, Ravier MA, Matveyenko AV, Costes S. The nuclear receptor REV-ERBα is implicated in the alteration of β-cell autophagy and survival under diabetogenic conditions. Cell Death Dis 2022; 13:353. [PMID: 35428762 PMCID: PMC9012816 DOI: 10.1038/s41419-022-04767-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 01/07/2023]
Abstract
Pancreatic β-cell failure in type 2 diabetes mellitus (T2DM) is associated with impaired regulation of autophagy which controls β-cell development, function, and survival through clearance of misfolded proteins and damaged organelles. However, the mechanisms responsible for defective autophagy in T2DM β-cells remain unknown. Since recent studies identified circadian clock transcriptional repressor REV-ERBα as a novel regulator of autophagy in cancer, in this study we set out to test whether REV-ERBα-mediated inhibition of autophagy contributes to the β-cell failure in T2DM. Our study provides evidence that common diabetogenic stressors (e.g., glucotoxicity and cytokine-mediated inflammation) augment β-cell REV-ERBα expression and impair β-cell autophagy and survival. Notably, pharmacological activation of REV-ERBα was shown to phenocopy effects of diabetogenic stressors on the β-cell through inhibition of autophagic flux, survival, and insulin secretion. In contrast, negative modulation of REV-ERBα was shown to provide partial protection from inflammation and glucotoxicity-induced β-cell failure. Finally, using bioinformatic approaches, we provide further supporting evidence for augmented REV-ERBα activity in T2DM human islets associated with impaired transcriptional regulation of autophagy and protein degradation pathways. In conclusion, our study reveals a previously unexplored causative relationship between REV-ERBα expression, inhibition of autophagy, and β-cell failure in T2DM.
Collapse
Affiliation(s)
- Matthew R. Brown
- grid.66875.3a0000 0004 0459 167XDepartment of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN USA
| | - Damien Laouteouet
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Morgane Delobel
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Orianne Villard
- grid.157868.50000 0000 9961 060XLaboratory of Cell Therapy for Diabetes (LTCD), PRIMS facility, Institute for Regenerative Medicine and Biotherapy (IRMB), University hospital of Montpellier, Montpellier, France ,grid.157868.50000 0000 9961 060XDepartment of Endocrinology, Diabetes, and Nutrition, University Hospital of Montpellier, Montpellier, France
| | - Christophe Broca
- grid.157868.50000 0000 9961 060XLaboratory of Cell Therapy for Diabetes (LTCD), PRIMS facility, Institute for Regenerative Medicine and Biotherapy (IRMB), University hospital of Montpellier, Montpellier, France
| | - Gyslaine Bertrand
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Anne Wojtusciszyn
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France ,grid.157868.50000 0000 9961 060XLaboratory of Cell Therapy for Diabetes (LTCD), PRIMS facility, Institute for Regenerative Medicine and Biotherapy (IRMB), University hospital of Montpellier, Montpellier, France ,grid.157868.50000 0000 9961 060XDepartment of Endocrinology, Diabetes, and Nutrition, University Hospital of Montpellier, Montpellier, France
| | - Stéphane Dalle
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Magalie A. Ravier
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Aleksey V. Matveyenko
- grid.66875.3a0000 0004 0459 167XDepartment of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN USA ,grid.66875.3a0000 0004 0459 167XDivision of Endocrinology, Metabolism, Diabetes, and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN USA
| | - Safia Costes
- grid.121334.60000 0001 2097 0141Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
6
|
Delangre E, Liu J, Tolu S, Maouche K, Armanet M, Cattan P, Pommier G, Bailbé D, Movassat J. Underlying mechanisms of glucocorticoid-induced β-cell death and dysfunction: a new role for glycogen synthase kinase 3. Cell Death Dis 2021; 12:1136. [PMID: 34876563 PMCID: PMC8651641 DOI: 10.1038/s41419-021-04419-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 11/08/2022]
Abstract
Glucocorticoids (GCs) are widely prescribed for their anti-inflammatory and immunosuppressive properties as a treatment for a variety of diseases. The use of GCs is associated with important side effects, including diabetogenic effects. However, the underlying mechanisms of GC-mediated diabetogenic effects in β-cells are not well understood. In this study we investigated the role of glycogen synthase kinase 3 (GSK3) in the mediation of β-cell death and dysfunction induced by GCs. Using genetic and pharmacological approaches we showed that GSK3 is involved in GC-induced β-cell death and impaired insulin secretion. Further, we unraveled the underlying mechanisms of GC-GSK3 crosstalk. We showed that GSK3 is marginally implicated in the nuclear localization of GC receptor (GR) upon ligand binding. Furthermore, we showed that GSK3 regulates the expression of GR at mRNA and protein levels. Finally, we dissected the proper contribution of each GSK3 isoform and showed that GSK3β isoform is sufficient to mediate the pro-apoptotic effects of GCs in β-cells. Collectively, in this work we identified GSK3 as a viable target to mitigate GC deleterious effects in pancreatic β-cells.
Collapse
Affiliation(s)
- Etienne Delangre
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Junjun Liu
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
- Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Stefania Tolu
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Kamel Maouche
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Mathieu Armanet
- Cell Therapy Unit, Saint-Louis hospital, AP-HP, and Université de Paris, Paris, France
| | - Pierre Cattan
- Cell Therapy Unit, Saint-Louis hospital, AP-HP, and Université de Paris, Paris, France
| | - Gaëlle Pommier
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Danielle Bailbé
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France
| | - Jamileh Movassat
- Université de Paris, BFA, UMR 8251, CNRS, Team « Biologie et Pathologie du Pancréas Endocrine », Paris, France.
| |
Collapse
|
7
|
Kassem M, El Habhab A, Kreutter G, Amoura L, Baltzinger P, Abbas M, Sbat N, Zobairi F, Schini-Kerth VB, Kessler L, Toti F. In Vitro Impact of Pro-Senescent Endothelial Microvesicles on Isolated Pancreatic Rat Islets Function. Transplant Proc 2021; 53:1736-1743. [PMID: 33934912 DOI: 10.1016/j.transproceed.2021.02.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 01/15/2021] [Accepted: 02/24/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Ischemia-driven islet isolation procedure is one of the limiting causes of pancreatic islet transplantation. Ischemia-reperfusion process is associated with endothelium dysfunction and the release of pro-senescent microvesicles. We investigated whether pro-senescent endothelial microvesicles prompt islet senescence and dysfunction in vitro. MATERIAL AND METHODS Pancreatic islets were isolated from male young rats. Replicative endothelial senescence was induced by serial passaging of primary porcine coronary artery endothelial cells, and microvesicles were isolated either from young passage 1 (P1) or senescent passage 3 (P3) endothelial cells. Islet viability was assessed by fluorescence microscopy, apoptosis by flow cytometry, and Western blot. Function was assessed by insulin secretion and islet senescence markers p53, p21, and p16 by Western blot. Microvesicles were stained by the PKH26 lipid fluorescent probe and their islet integration assessed by microscopy and flow cytometry. RESULTS Regardless of the passage, half microvesicles were integrated in target islets after 24 hours incubation. Insulin secretion significantly decreased after treatment by senescent microvesicles (P3: 1.7 ± 0.2 vs untreated islet: 2.7 ± 0.2, P < .05) without altering the islet viability (89.47% ± 1.69 vs 93.15% ± 0.97) and with no significant apoptosis. Senescent microvesicles significantly doubled the expression of p53, p21, and p16 (P < .05), whereas young microvesicles had no significant effect. CONCLUSION Pro-senescent endothelial microvesicles specifically accelerate the senescence of islets and alter their function. These data suggest that islet isolation contributes to endothelial driven islet senescence.
Collapse
Affiliation(s)
- Mohamad Kassem
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Ali El Habhab
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Guillaume Kreutter
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Lamia Amoura
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Philippe Baltzinger
- Department of Diabetes and Nutrition Endocrinology, University Hospital of Strasbourg, Strasbourg, France
| | - Malak Abbas
- UMR CNRS 7213, Laboratory of Biophotonics and Pharmacology, Faculty of Pharmacy, University of Strasbourg, Illkirch-Graffenstaden, France
| | - Noura Sbat
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Fatiha Zobairi
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Valérie B Schini-Kerth
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France
| | - Laurence Kessler
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France; Department of Diabetes and Nutrition Endocrinology, University Hospital of Strasbourg, Strasbourg, France
| | - Florence Toti
- INSERM (French National Institute of Health and Medical Research), UMR 1260, Regenerative Nanomedicine (RNM), Faculty of Medicine, University of Strasbourg, Strasbourg, France.
| |
Collapse
|
8
|
Wassmer CH, Perrier Q, Combescure C, Pernin N, Parnaud G, Cottet-Dumoulin D, Brioudes E, Bellofatto K, Lebreton F, Berishvili E, Lablanche S, Kessler L, Wojtusciszyn A, Buron F, Borot S, Bosco D, Berney T, Lavallard V. Impact of ischemia time on islet isolation success and posttransplantation outcomes: A retrospective study of 452 pancreas isolations. Am J Transplant 2021; 21:1493-1502. [PMID: 32986297 DOI: 10.1111/ajt.16320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 08/03/2020] [Accepted: 09/04/2020] [Indexed: 01/25/2023]
Abstract
Many variables impact islet isolation, including pancreas ischemia time. The ischemia time upper limit that should be respected to avoid a negative impact on the isolation outcome is not well defined. We have performed a retrospective analysis of all islet isolations in our center between 2008 and 2018. Total ischemia time, cold ischemia time, and organ removal time were analyzed. Isolation success was defined as an islet yield ≥200 000 IEQ. Of the 452 pancreases included, 288 (64%) were successfully isolated. Probability of isolation success showed a significant decrease after 8 hours of total ischemia time, 7 hours of cold ischemia time, and 80 minutes of organ removal time. Although we observed an impact of ischemia time on islet yield, a probability of isolation success of 50% was still present even when total ischemia time exceeds 12 hours. Posttransplantation clinical outcomes were assessed in 32 recipients and no significant difference was found regardless of ischemia time. These data indicate that although shorter ischemia times are associated with better islet isolation outcomes, total ischemia time >12 hours can provide excellent results in appropriately selected donors.
Collapse
Affiliation(s)
- Charles-Henri Wassmer
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Quentin Perrier
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Christophe Combescure
- Division of Clinical Epidemiology, Faculty of Medicine, University of Geneva, and Geneva University Hospitals, Geneva, Switzerland
| | - Nadine Pernin
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Géraldine Parnaud
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - David Cottet-Dumoulin
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Estelle Brioudes
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Kevin Bellofatto
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland.,Institute of Medical Research, Ilia State University, Tbilisi, Georgia
| | - Sandrine Lablanche
- Endocrinology Department, Grenoble Alpes University Hospital, Grenoble, France
| | - Laurence Kessler
- Department of Diabetology, University Hospital, Strasbourg, France.,Federation of Translational Medicine of Strasbourg, University of Strasbourg, Strasbourg, France
| | - Anne Wojtusciszyn
- Department of Endocrinology, Diabetes, and Nutrition, Montpellier University Hospital, Montpellier, France.,Laboratory of Cell Therapy of Diabetes, Institute of Functional Genomics, Mixed Research Unit, French National Center for Scientific Research 5203, Inserm U1191, University of Montpellier, Montpellier, France
| | - Fanny Buron
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital, Hospices Civils de Lyon, Lyon, France
| | - Sophie Borot
- Endocrinology Department, Besancon University Hospital, Besancon, France
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland.,Faculty Diabetes Center, University of Geneva Medical Center, Geneva, Switzerland
| |
Collapse
|
9
|
Cayabyab F, Nih LR, Yoshihara E. Advances in Pancreatic Islet Transplantation Sites for the Treatment of Diabetes. Front Endocrinol (Lausanne) 2021; 12:732431. [PMID: 34589059 PMCID: PMC8473744 DOI: 10.3389/fendo.2021.732431] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/13/2021] [Indexed: 01/08/2023] Open
Abstract
Diabetes is a complex disease that affects over 400 million people worldwide. The life-long insulin injections and continuous blood glucose monitoring required in type 1 diabetes (T1D) represent a tremendous clinical and economic burdens that urges the need for a medical solution. Pancreatic islet transplantation holds great promise in the treatment of T1D; however, the difficulty in regulating post-transplantation immune reactions to avoid both allogenic and autoimmune graft rejection represent a bottleneck in the field of islet transplantation. Cell replacement strategies have been performed in hepatic, intramuscular, omentum, and subcutaneous sites, and have been performed in both animal models and human patients. However more optimal transplantation sites and methods of improving islet graft survival are needed to successfully translate these studies to a clinical relevant therapy. In this review, we summarize the current progress in the field as well as methods and sites of islet transplantation, including stem cell-derived functional human islets. We also discuss the contribution of immune cells, vessel formation, extracellular matrix, and nutritional supply on islet graft survival. Developing new transplantation sites with emerging technologies to improve islet graft survival and simplify immune regulation will greatly benefit the future success of islet cell therapy in the treatment of diabetes.
Collapse
Affiliation(s)
- Fritz Cayabyab
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
| | - Lina R. Nih
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
| | - Eiji Yoshihara
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, United States
- David Geffen School of Medicine at University of California, Los Angeles, CA, United States
- *Correspondence: Eiji Yoshihara,
| |
Collapse
|
10
|
Heterogeneity of Human Pancreatic Islet Isolation Around Europe: Results of a Survey Study. Transplantation 2020; 104:190-196. [PMID: 31365472 DOI: 10.1097/tp.0000000000002777] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Europe is currently the most active region in the field of pancreatic islet transplantation, and many of the leading groups are actually achieving similar good outcomes. Further collaborative advances in the field require the standardization of islet cell product isolation processes, and this work aimed to identify differences in the human pancreatic islet isolation processes within European countries. METHODS A web-based questionnaire about critical steps, including donor selection, pancreas processing, pancreas perfusion and digestion, islet counting and culture, islet quality evaluation, microbiological evaluation, and release criteria of the product, was completed by isolation facilities participating at the Ninth International European Pancreas and Islet Transplant Association (EPITA) Workshop on Islet-Beta Cell Replacement in Milan. RESULTS Eleven islet isolation facilities completed the questionnaire. The facilities reported 445 and 53 islet isolations per year over the last 3 years from deceased organ donors and pancreatectomized patients, respectively. This activity resulted in 120 and 40 infusions per year in allograft and autograft recipients, respectively. Differences among facilities emerged in donor selection (age, cold ischemia time, intensive care unit length, amylase concentration), pancreas procurement, isolation procedures (brand and concentration of collagenase, additive, maximum acceptable digestion time), quality evaluation, and release criteria for transplantation (glucose-stimulated insulin secretion tests, islet numbers, and purity). Moreover, even when a high concordance about the relevance of one parameter was evident, thresholds for the acceptance were different among facilities. CONCLUSIONS The result highlighted the presence of a heterogeneity in the islet cell product process and product release criteria.
Collapse
|
11
|
Meier RPH, Meyer J, Muller YD, Szot GL, Bédat B, Andres A, Massé N, Lablanche S, Puppa G, Bosco D, Berney T. Pancreas collagen digestion during islet of Langerhans isolation-a prospective study. Transpl Int 2020; 33:1516-1528. [PMID: 32852857 DOI: 10.1111/tri.13725] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/03/2020] [Accepted: 08/20/2020] [Indexed: 12/12/2022]
Abstract
The success of pancreas islet isolation largely depends on donor characteristics, including extracellular matrix composition of which collagen is the main element. We hypothesized that isolation yields are proportional to collagen digestion percentage, and aimed to determine a threshold that predicts isolation success. The amount of pancreas collagen (I-V) was determined using colorimetry prior to and after the digestion process in 52 human islet isolations. Collagen I-V and VI were also assessed histologically. We identified a collagen digestion threshold of ≥ 60% as an independent factor beyond which an islet preparation has a ninefold increased odds of yielding ≥ 250 000 islet equivalents (IEQ) (P = 0.009) and a sixfold increased odds of being transplanted (P = 0.015). Preparations with ≥ 60% collagen digestion (n = 35) yielded 283 017 ± 164 214 IEQ versus 180 142 ± 85 397 in the < 60% collagen digestion group (n = 17) (P = 0.016); respectively 62.9% versus 29.4% of those were transplanted (P = 0.024). Common donor characteristics, initial collagen content, enzyme blend, and digestion times were not associated with collagen digestion percentage variations. Donor age positively correlated with the amount of collagen VI (P = 0.013). There was no difference in islet graft survival between high and low digestion groups. We determined that a 60% pancreas collagen digestion is the threshold beyond which an islet isolation is likely to be successful and transplanted.
Collapse
Affiliation(s)
- Raphael P H Meier
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland.,Department of Surgery, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jeremy Meyer
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| | - Yannick D Muller
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland.,Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Gregory L Szot
- Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Benoît Bédat
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| | - Axel Andres
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland.,Division of Transplantation, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| | - Nathalie Massé
- Division of Transplantation, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| | - Sandrine Lablanche
- Grenoble Alpes University, Grenoble, France.,Laboratory of Fundamental and Applied Bioenergetics Grenoble, Inserm U1055, Grenoble, France
| | - Giacomo Puppa
- Diagnostic Department, Geneva University Hospital, Geneva, Switzerland.,Department of Pathology and Immunology, University of Geneva Medical School, Geneva, Switzerland
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland.,Division of Transplantation, Department of Surgery, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| |
Collapse
|
12
|
Castex F, Leroy J, Broca C, Mezghenna K, Duranton F, Lavallard V, Lebreton F, Gross R, Wojtusciszyn A, Lajoix AD. Differential sensitivity of human islets from obese versus lean donors to chronic high glucose or palmitate. J Diabetes 2020; 12:532-541. [PMID: 32090456 DOI: 10.1111/1753-0407.13026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 01/15/2020] [Accepted: 02/19/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND Due to the shortage of multi-organ donors, human pancreatic islet transplantation has now been extended to islets originating from obese subjects. In this study, our aim is to compare the respective sensitivity of human islets from lean vs obese donors to chronic high glucose or high palmitate. METHODS Human islets were isolated from pancreases harvested from brain-dead multi-organ donors. Islets were cultured during 72 hours in the presence of moderate (16.7 mmol/L) or high (28 mmoL/L) glucose concentrations, or glucose (5.6 mmoL/L) and palmitate (0.4 mmoL/L), before measurement of their response to glucose. RESULTS We first observed a greater insulin response in islets from obese donors under both basal and high-glucose conditions, confirming their hyperresponsiveness to glucose. When islets from obese donors were cultured in the presence of moderate or high glucose concentrations, insulin response to glucose remained unchanged or was slightly reduced, as opposed to that observed in lean subjects. Moreover, culturing islets from obese donors with high palmitate also induced less reduction in insulin response to glucose than in lean subjects. This partial protection of obese islets is associated with less induction of inducible nitric oxide synthase in islets, together with a greater expression of the transcription factor forkhead box O1 (FOXO1). CONCLUSIONS Our data suggest that in addition to an increased sensitivity to glucose, islets from obese subjects can be considered as more resistant to glucose and fatty acid excursions and are thus valuable candidates for transplantation.
Collapse
Affiliation(s)
- Françoise Castex
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Jeremy Leroy
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Christophe Broca
- Laboratory of Cell Therapy for Diabetes, Institute for Regenerative Medicine & Biotherapy (IRMB), University Montpellier, INSERM, University Hospital Montpellier, Montpellier, France
| | - Karima Mezghenna
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Flore Duranton
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
- RD Néphrologie, Montpellier, France
| | - Vanessa Lavallard
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Genève, Switzerland
| | | | - René Gross
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| | - Anne Wojtusciszyn
- Laboratory of Cell Therapy for Diabetes, Institute for Regenerative Medicine & Biotherapy (IRMB), University Montpellier, INSERM, University Hospital Montpellier, Montpellier, France
| | - Anne-Dominique Lajoix
- Biocommunication in Cardio-Metabolism (BC2M), University Montpellier, Montpellier, France
| |
Collapse
|
13
|
Brioudes E, Alibashe-Ahmed M, Lavallard V, Berney T, Bosco D. Syndecan-4 is regulated by IL-1β in β-cells and human islets. Mol Cell Endocrinol 2020; 510:110815. [PMID: 32315719 DOI: 10.1016/j.mce.2020.110815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/30/2020] [Accepted: 04/01/2020] [Indexed: 12/13/2022]
Abstract
Syndecans (SDC) are important multifunctional components of the extracellular matrix mainly described in endothelial cells. We studied the expression and regulation of SDC in cultured MIN6B1 cells and pancreatic islets. qRT-PCR revealed that syndecan-4 (SDC4) was the predominant isoform expressed in MIN6B1 cells and islets compared to other forms of SDC. Immunofluorescence in mouse and human pancreas sections revealed that SDC4 is mainly expressed in β-cells compared to other pancreatic cells. Exposure of MIN6B1 and human islets to IL-1β dose-dependently induced a rapid and transient expression of SDC4 while SRC and STAT3 inhibitors decreased this effect. Exposure of human islets to Il-1β caused an increase of SDC4 shedding, however treatment with STAT3 and SRC inhibitors inhibited this effect. These results indicate that SDC4 is upregulated by IL-1β through the SRC-STAT3 pathway and this pathway is also involved in SDC4 shedding in islets.
Collapse
Affiliation(s)
- Estelle Brioudes
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211, Geneva 4, Geneva, Switzerland; Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva 4, Geneva, Switzerland.
| | - Mohamed Alibashe-Ahmed
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211, Geneva 4, Geneva, Switzerland; Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211, Geneva 4, Geneva, Switzerland; Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211, Geneva 4, Geneva, Switzerland; Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, 1211, Geneva 4, Geneva, Switzerland; Diabetes Center of the Faculty of Medicine, University of Geneva, 1211, Geneva 4, Geneva, Switzerland
| |
Collapse
|
14
|
The impact of proinflammatory cytokines on the β-cell regulatory landscape provides insights into the genetics of type 1 diabetes. Nat Genet 2019; 51:1588-1595. [PMID: 31676868 PMCID: PMC7040466 DOI: 10.1038/s41588-019-0524-6] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 09/27/2019] [Indexed: 01/31/2023]
Abstract
Early stages of type 1 diabetes (T1D) are characterized by local autoimmune inflammation and progressive loss of insulin-producing pancreatic β cells. We show here that exposure to pro-inflammatory cytokines unmasks a marked plasticity of the β-cell regulatory landscape. We expand the repertoire of human islet regulatory elements by mapping stimulus-responsive enhancers linked to changes in the β-cell transcriptome, proteome and 3D chromatin structure. Our data indicate that the β cell response to cytokines is mediated by the induction of new regulatory regions as well as the activation of primed regulatory elements prebound by islet-specific transcription factors. We find that T1D-associated loci are enriched of the newly mapped cis-regulatory regions and identify T1D-associated variants disrupting cytokine-responsive enhancer activity in human β cells. Our study illustrates how β cells respond to a pro-inflammatory environment and implicate a role for stimulus-response islet enhancers in T1D.
Collapse
|
15
|
Human pancreatic islet three-dimensional chromatin architecture provides insights into the genetics of type 2 diabetes. Nat Genet 2019; 51:1137-1148. [PMID: 31253982 DOI: 10.1038/s41588-019-0457-0] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Accepted: 05/29/2019] [Indexed: 01/07/2023]
Abstract
Genetic studies promise to provide insight into the molecular mechanisms underlying type 2 diabetes (T2D). Variants associated with T2D are often located in tissue-specific enhancer clusters or super-enhancers. So far, such domains have been defined through clustering of enhancers in linear genome maps rather than in three-dimensional (3D) space. Furthermore, their target genes are often unknown. We have created promoter capture Hi-C maps in human pancreatic islets. This linked diabetes-associated enhancers to their target genes, often located hundreds of kilobases away. It also revealed >1,300 groups of islet enhancers, super-enhancers and active promoters that form 3D hubs, some of which show coordinated glucose-dependent activity. We demonstrate that genetic variation in hubs impacts insulin secretion heritability, and show that hub annotations can be used for polygenic scores that predict T2D risk driven by islet regulatory variants. Human islet 3D chromatin architecture, therefore, provides a framework for interpretation of T2D genome-wide association study (GWAS) signals.
Collapse
|
16
|
Rickels MR, Robertson RP. Pancreatic Islet Transplantation in Humans: Recent Progress and Future Directions. Endocr Rev 2019; 40:631-668. [PMID: 30541144 PMCID: PMC6424003 DOI: 10.1210/er.2018-00154] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 10/26/2018] [Indexed: 12/11/2022]
Abstract
Pancreatic islet transplantation has become an established approach to β-cell replacement therapy for the treatment of insulin-deficient diabetes. Recent progress in techniques for islet isolation, islet culture, and peritransplant management of the islet transplant recipient has resulted in substantial improvements in metabolic and safety outcomes for patients. For patients requiring total or subtotal pancreatectomy for benign disease of the pancreas, isolation of islets from the diseased pancreas with intrahepatic transplantation of autologous islets can prevent or ameliorate postsurgical diabetes, and for patients previously experiencing painful recurrent acute or chronic pancreatitis, quality of life is substantially improved. For patients with type 1 diabetes or insulin-deficient forms of pancreatogenic (type 3c) diabetes, isolation of islets from a deceased donor pancreas with intrahepatic transplantation of allogeneic islets can ameliorate problematic hypoglycemia, stabilize glycemic lability, and maintain on-target glycemic control, consequently with improved quality of life, and often without the requirement for insulin therapy. Because the metabolic benefits are dependent on the numbers of islets transplanted that survive engraftment, recipients of autoislets are limited to receive the number of islets isolated from their own pancreas, whereas recipients of alloislets may receive islets isolated from more than one donor pancreas. The development of alternative sources of islet cells for transplantation, whether from autologous, allogeneic, or xenogeneic tissues, is an active area of investigation that promises to expand access and indications for islet transplantation in the future treatment of diabetes.
Collapse
Affiliation(s)
- Michael R Rickels
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - R Paul Robertson
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, Washington
- Division of Endocrinology, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
- Pacific Northwest Diabetes Research Institute, Seattle, Washington
| |
Collapse
|
17
|
Brandhorst H, Johnson PR, Mönch J, Kurfürst M, Korsgren O, Brandhorst D. Comparison of Clostripain and Neutral Protease as Supplementary Enzymes for Human Islet Isolation. Cell Transplant 2018; 28:176-184. [PMID: 30419762 PMCID: PMC6362525 DOI: 10.1177/0963689718811614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Although human islet transplantation has been established as valid and safe treatment for patients with type 1 diabetes, the utilization rates of human pancreases for clinical islet transplantation are still limited and substantially determined by the quality and composition of collagenase blends. While function and integrity of collagenase has been extensively investigated, information is still lacking about the most suitable supplementary neutral proteases. The present study compared islet isolation outcome after pancreas digestion by means of collagenase used alone or supplemented with either neutral protease (NP), clostripain (CP), or both proteases. Decent amounts of islet equivalents (IEQ) were isolated using collagenase alone (3090 ± 550 IEQ/g), or in combination with NP (2340 ± 450 IEQ/g) or CP (2740 ± 280 IEQ/g). Nevertheless, the proportion of undigested tissue was higher after using collagenase alone (21.1 ± 1.1%, P < 0.05) compared with addition of NP (13.3 ± 2.2%) or CP plus NP (13.7 ± 2.6%). Likewise, the percentage of embedded islets was highest using collagenase only (13 ± 2%) and lowest adding NP plus CP (4 ± 1%, P < 0.01). The latter combination resulted in lowest post-culture overall survival (42.7 ± 3.9%), while highest survival was observed after supplementation with CP (74.5 ± 4.8%, P < 0.01). An insulin response toward glucose challenge was present in all experimental groups, but the stimulation index was significantly decreased using collagenase plus NP (2.0 ± 0.12) compared with supplementation with CP (3.16 ± 0.4, P < 0.001). This study demonstrates for the first time that it is possible to isolate significant numbers of human islets combining collagenase only with CP. The supplementation with CP is an effective means to substantially reduce NP activity, which significantly decreases survival and viability after culture. This will facilitate the manufacturing of enzyme blends with less harmful characteristics.
Collapse
Affiliation(s)
- Heide Brandhorst
- 1 Nuffield Department of Surgical Sciences, University of Oxford, UK.,2 Oxford Centre for Diabetes, Endocrinology and Metabolism, UK.,3 Department of Immunology, Genetics and Pathology, University of Uppsala, Sweden
| | - Paul R Johnson
- 1 Nuffield Department of Surgical Sciences, University of Oxford, UK.,2 Oxford Centre for Diabetes, Endocrinology and Metabolism, UK.,4 Oxford NIHR Biomedical Research Centre, UK
| | | | | | - Olle Korsgren
- 3 Department of Immunology, Genetics and Pathology, University of Uppsala, Sweden
| | - Daniel Brandhorst
- 1 Nuffield Department of Surgical Sciences, University of Oxford, UK.,2 Oxford Centre for Diabetes, Endocrinology and Metabolism, UK.,3 Department of Immunology, Genetics and Pathology, University of Uppsala, Sweden
| |
Collapse
|
18
|
Lebreton F, Berishvili E, Parnaud G, Rouget C, Bosco D, Berney T, Lavallard V. NLRP3 inflammasome is expressed and regulated in human islets. Cell Death Dis 2018; 9:726. [PMID: 29941940 PMCID: PMC6018156 DOI: 10.1038/s41419-018-0764-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 06/05/2018] [Accepted: 06/08/2018] [Indexed: 02/02/2023]
Abstract
NRLP3 inflammasome is a protein complex involved in the maturation of IL1β. In the onset of type 1 diabetes as well as in islet transplantation, IL-1β is one of the cytokines involved in the recruitment of immune cells in islets and eventually in islet destruction. Whether IL-1β is produced by islet cells is still under debate and NLRP3 inflammasome-dependent IL-1β production has not been yet determined in human islets. The aim of this study was to determine the expression and the regulation of the NRLP3 inflammasome in human islets. Human islets were stimulated with LPS and successively with ATP (LPS + ATP) in the presence or absence of the inflammasome inhibitor glyburide. Islets were also incubated in hypoxic or normoxic conditions for 24 h in the presence or absence of glyburide. Then, IL1B and NLRP3 expression was studied by real time PCR, protein expression by western blot, protein localization by immunofluorescence and protein secretion by ELISA. LPS + ATP increased gene expression of NRLP3 and IL1B. Glyburide partially prevented this effect. IL-1β protein was localized in β and non-β cells. Moreover, LPS + ATP increased IL-1β protein expression and production, which were prevented by glyburide. Hypoxia increased gene expression of NRLP3 and IL1B and induced IL-1β and caspase-1 production. Finally, hypoxia-induced cell death which was not prevented by inhibition of NLRP3 inflammasome. NRLP3 inflammasome is expressed and plays a role in IL-1β production by human islets. By contrast, NRLP3 inflammasome activation is not involved in islet cell death induced by hypoxia.
Collapse
Affiliation(s)
- Fanny Lebreton
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Ekaterine Berishvili
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Géraldine Parnaud
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Caroline Rouget
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland
| | - Vanessa Lavallard
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University, Hospitals and University of Geneva, Geneva, Switzerland.
| |
Collapse
|
19
|
Ruiz L, Gurlo T, Ravier MA, Wojtusciszyn A, Mathieu J, Brown MR, Broca C, Bertrand G, Butler PC, Matveyenko AV, Dalle S, Costes S. Proteasomal degradation of the histone acetyl transferase p300 contributes to beta-cell injury in a diabetes environment. Cell Death Dis 2018; 9:600. [PMID: 29789539 PMCID: PMC5964068 DOI: 10.1038/s41419-018-0603-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 03/09/2018] [Accepted: 04/17/2018] [Indexed: 12/25/2022]
Abstract
In type 2 diabetes, amyloid oligomers, chronic hyperglycemia, lipotoxicity, and pro-inflammatory cytokines are detrimental to beta-cells, causing apoptosis and impaired insulin secretion. The histone acetyl transferase p300, involved in remodeling of chromatin structure by epigenetic mechanisms, is a key ubiquitous activator of the transcriptional machinery. In this study, we report that loss of p300 acetyl transferase activity and expression leads to beta-cell apoptosis, and most importantly, that stress situations known to be associated with diabetes alter p300 levels and functional integrity. We found that proteasomal degradation is the mechanism subserving p300 loss in beta-cells exposed to hyperglycemia or pro-inflammatory cytokines. We also report that melatonin, a hormone produced in the pineal gland and known to play key roles in beta-cell health, preserves p300 levels altered by these toxic conditions. Collectively, these data imply an important role for p300 in the pathophysiology of diabetes.
Collapse
Affiliation(s)
- Lucie Ruiz
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Tatyana Gurlo
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Magalie A Ravier
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Anne Wojtusciszyn
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.,Laboratory of Cell Therapy for Diabetes (LTCD), Institute for Regenerative Medicine and Biotherapy (IRMB), University Hospital of Montpellier, Montpellier, France.,Department of Endocrinology, Diabetes, and Nutrition, University Hospital of Montpellier, Montpellier, France
| | - Julia Mathieu
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Matthew R Brown
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Christophe Broca
- Laboratory of Cell Therapy for Diabetes (LTCD), Institute for Regenerative Medicine and Biotherapy (IRMB), University Hospital of Montpellier, Montpellier, France
| | | | - Peter C Butler
- Larry L. Hillblom Islet Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Aleksey V Matveyenko
- Department of Physiology and Biomedical Engineering, Mayo Clinic School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Stéphane Dalle
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France
| | - Safia Costes
- IGF, CNRS, INSERM, University of Montpellier, Montpellier, France.
| |
Collapse
|
20
|
Meier RPH, Andrey DO, Sun P, Niclauss N, Bédat B, Demuylder-Mischler S, Borot S, Benhamou PY, Wojtusciszyn A, Buron F, Pernin N, Muller YD, Bosco D, van Delden C, Berney T. Pancreas preservation fluid microbial contamination is associated with poor islet isolation outcomes - a multi-centre cohort study. Transpl Int 2018; 31:917-929. [PMID: 29603452 DOI: 10.1111/tri.13159] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/09/2018] [Accepted: 03/21/2018] [Indexed: 12/15/2022]
Abstract
The microbiological safety of islet preparations is paramount. Preservation medium contamination is frequent, and its impact on islet yield and function remains unclear. Microbiological samples collected during islet isolations from 2006 to 2016 were analyzed and correlated to isolation and allo- and autotransplantation outcomes. Microbial contamination of preservation medium was found in 64.4% of processed donor pancreases (291/452). We identified 464 microorganisms including Staphylococcus (253/464, 54.5%), Streptococcus (31/464, 6.7%), and Candida species (25/464, 5.4%). Microbial contamination was associated with longer warm and cold ischemia times and lower numbers of postpurification islet equivalents, purity, transplant rate, and stimulation index (all P < 0.05). Six percent of the preparations accepted for transplantation showed microbial contamination after isolation (12/200); 9 of 12 were Candida species. Six patients were transplanted with a sample with late microbial growth discovered after the infusion. Insulin independence rate was not affected. This risk of transplanting a contaminated islets preparation was reduced by half following the implementation of an additional sampling after 24 h of islet culture. Pancreas preservation fluid microbial contamination is associated with lower transplant rate and poorer in vitro function, but not with changes in graft survival. Culture medium testing 1 day after isolation reduces the risk of incidental transplantation with contaminated islets.
Collapse
Affiliation(s)
- Raphael P H Meier
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland.,Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland.,Transplant Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Diego O Andrey
- Transplant Infectious Diseases Unit, Department of Medical Specialties, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Pamela Sun
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Nadja Niclauss
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland.,Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Benoît Bédat
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Sandrine Demuylder-Mischler
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Sophie Borot
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland.,Service d'Endocrinologie-Métabolisme et Diabétologie-Nutrition, Centre Hospitalier Universitaire Jean Minjoz, Besançon, France
| | - Pierre-Yves Benhamou
- Department of Endocrinology, Pôle DigiDune, Grenoble University Hospital, University Grenoble Alpes, Grenoble, France
| | - Anne Wojtusciszyn
- Laboratory of Cell Therapy for Diabetes, IRMB, Saint Eloi Hospital, Montpellier University Hospital, Montpellier, France.,Department of Endocrinology, Diabetes and Nutrition, Lapeyronie Hospital, Montpellier University Hospital, Montpellier, France
| | - Fanny Buron
- Nephrology Unit, Department of Transplantation, Edouard Herriot Hospital, Lyon, France
| | - Nadine Pernin
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Yannick D Muller
- Transplant Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Domenico Bosco
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Christian van Delden
- Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland.,Transplant Infectious Diseases Unit, Department of Medical Specialties, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| | - Thierry Berney
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland.,Visceral and Transplant Surgery, Department of Surgery, Geneva University Hospital, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
21
|
Brandhorst D, Brandhorst H, Johnson PRV. Enzyme Development for Human Islet Isolation: Five Decades of Progress or Stagnation? Rev Diabet Stud 2017. [PMID: 28632819 DOI: 10.1900/rds.2017.14.22] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
In comparison to procedures used for the separation of individual cell types from other organs, the process of human pancreatic islet isolation aims to digest the pancreatic exocrine matrix completely without dispersing the individual cells within the endocrine cell cluster. This objective is unique within the field of tissue separation, and outlines the challenge of islet isolation to balance two opposing priorities. Although significant progress has been made in the characterization and production of enzyme blends for islet isolation, there are still numerous areas which require improvement. The ultimate goal of enzyme production, namely the routine production of a consistent and standardized enzyme blend, has still not been realized. This seems to be mainly the result of a lack of detailed knowledge regarding the structure of the pancreatic extracellular matrix and the synergistic interplay between collagenase and different supplementary proteases during the degradation of the extracellular matrix. Furthermore, the activation of intrinsic proteolytic enzymes produced by the pancreatic acinar cells, also impacts on the chance of a successful outcome of human islet isolation. This overview discusses the challenges of pancreatic enzymatic digestion during human islet isolation, and outlines the developments in this field over the past 5 decades.
Collapse
Affiliation(s)
- Daniel Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Heide Brandhorst
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| | - Paul R V Johnson
- Nuffield Department of Surgical Sciences, University of Oxford, United Kingdom
| |
Collapse
|
22
|
Kaviani M, Azarpira N. Insight into microenvironment remodeling in pancreatic endocrine tissue engineering: Biological and biomaterial approaches. Tissue Eng Regen Med 2016; 13:475-484. [PMID: 30603429 PMCID: PMC6170842 DOI: 10.1007/s13770-016-0014-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 02/03/2016] [Accepted: 02/05/2016] [Indexed: 01/04/2023] Open
Abstract
The treatment of diabetes mellitus, as a chronic and complicated disease, is a valuable purpose. Islet transplantation can provide metabolic stability and insulin independence in type 1 diabetes patients. Diet and insulin therapy are only diabetes controllers and cannot remove all of the diabetes complications. Moreover, islet transplantation is more promising treatment than whole pancreas transplantation because of lesser invasive surgical procedure and morbidity and mortality. According to the importance of extracellular matrix for islet viability and function, microenvironment remodeling of pancreatic endocrine tissue can lead to more success in diabetes treatment by pancreatic islets. Production of bioengineered pancreas and remodeling of pancreas extracellular matrix provide essential microenvironment for re-vascularization, re-innervation and signaling cascades triggering. Therefore, islets show better viability and function in these conditions. Researchers conduct various scaffolds with different biomaterials for the improvement of islet viability, function and transplantation outcome. The attention to normal pancreas anatomy, embryology and histology is critical to understand the pancreatic Langerhans islets niche and finally to achieve efficient engineered structure. Therefore, in the present study, the status and components of the islets niche is mentioned and fundamental issues related to the tissue engineering of this structure is considered. The purpose of this review article is summarization of recent progress in the endocrine pancreas tissue engineering and biomaterials and biological aspects of it.
Collapse
Affiliation(s)
- Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Mohamad Rasulallah Research Tower, Khalili street, Shiraz, 7193635899 Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Mohamad Rasulallah Research Tower, Khalili street, Shiraz, 7193635899 Iran
| |
Collapse
|
23
|
Lavallard V, Armanet M, Parnaud G, Meyer J, Barbieux C, Montanari E, Meier R, Morel P, Berney T, Bosco D. Cell rearrangement in transplanted human islets. FASEB J 2016; 30:748-760. [PMID: 26534832 DOI: 10.1096/fj.15-273805] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/13/2015] [Indexed: 11/03/2023]
Abstract
The major feature of the human pancreatic islet architecture is the organization of endocrine cells into clusters comprising central β cells and peripheral α cells surrounded by vasculature. To have an insight into the mechanisms that govern this unique islet architecture, islet cells were isolated, and reaggregation of α and β cells into islet-like structures (pseudoislets) after culture or transplantation into mice was studied by immunohistology. The pseudoislets formed in culture displayed an unusual cell arrangement, contrasting with the transplanted pseudoislets, which exhibited a cell arrangement similar to that observed in native pancreatic islet subunits. The pattern of revascularization and the distribution of extracellular matrix around transplanted pseudoislets were alike to those observed in native pancreatic islets. This organization of transplanted pseudoislets occurred also when revascularization was abolished by treating mice with an anti-VEGF antibody, but not when contact with extracellular matrix was prevented by encapsulation of pseudoislets within alginate hydrogel. These results indicate that the maintenance of islet cell arrangement is dependent on in vivo features such as extracellular matrix but independent of vascularization.
Collapse
Affiliation(s)
- Vanessa Lavallard
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Mathieu Armanet
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Géraldine Parnaud
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Jérémy Meyer
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Charlotte Barbieux
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Elisa Montanari
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Raphaël Meier
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Philippe Morel
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Thierry Berney
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| | - Domenico Bosco
- *Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Cell Therapy Unit, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, and University Paris 7, Paris, France; and Institut National de la Santé et de la Recherche Médicale, Unité Mixte de Recherche Scientifique 1138, Centre de Recherches des Cordeliers, Paris, France
| |
Collapse
|
24
|
Inhibition of the MAP3 kinase Tpl2 protects rodent and human β-cells from apoptosis and dysfunction induced by cytokines and enhances anti-inflammatory actions of exendin-4. Cell Death Dis 2016; 7:e2065. [PMID: 26794660 PMCID: PMC4816180 DOI: 10.1038/cddis.2015.399] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Revised: 11/27/2015] [Accepted: 12/01/2015] [Indexed: 02/06/2023]
Abstract
Proinflammatory cytokines exert cytotoxic effects on β-cells, and are involved in the pathogenesis of type I and type II diabetes and in the drastic loss of β-cells following islet transplantation. Cytokines induce apoptosis and alter the function of differentiated β-cells. Although the MAP3 kinase tumor progression locus 2 (Tpl2) is known to integrate signals from inflammatory stimuli in macrophages, fibroblasts and adipocytes, its role in β-cells is unknown. We demonstrate that Tpl2 is expressed in INS-1E β-cells, mouse and human islets, is activated and upregulated by cytokines and mediates ERK1/2, JNK and p38 activation. Tpl2 inhibition protects β-cells, mouse and human islets from cytokine-induced apoptosis and preserves glucose-induced insulin secretion in mouse and human islets exposed to cytokines. Moreover, Tpl2 inhibition does not affect survival or positive effects of glucose (i.e., ERK1/2 phosphorylation and basal insulin secretion). The protection against cytokine-induced β-cell apoptosis is strengthened when Tpl2 inhibition is combined with the glucagon-like peptide-1 (GLP-1) analog exendin-4 in INS-1E cells. Furthermore, when combined with exendin-4, Tpl2 inhibition prevents cytokine-induced death and dysfunction of human islets. This study proposes that Tpl2 inhibitors, used either alone or combined with a GLP-1 analog, represent potential novel and effective therapeutic strategies to protect diabetic β-cells.
Collapse
|
25
|
Calcineurin inhibitor-free immunosuppressive regimen in type 1 diabetes patients receiving islet transplantation: single-group phase 1/2 trial. Transplantation 2015; 98:1301-9. [PMID: 25286053 DOI: 10.1097/tp.0000000000000396] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Our final objective is to develop an adoptive therapy with tolerogenic donor-specific type 1 T regulatory cells for patients with type 1 diabetes undergoing islet transplantation. The achievement of this objective depends on the availability of an immunosuppressive treatment compatible with the survival, function, and expansion of type 1 T regulatory cells. METHODS For this purpose, we designed a single-group, phase 1 to 2 trial with an immunosuppression protocol including: (i) rapamycin treatment before the first islet infusion (starting ≥ 30 days before transplantation); (ii) induction therapy with anti-thymocyte globulin (ATG) instead of anti-interleukin-2Ra monoclonal antibody (after the first islet infusion only); (iii) short-term treatment with steroids and interleukin-1Ra (right before and for 2 weeks after each infusion); rapamycin+mycophenolate mofetil treatment as maintenance therapy. The target enrollment was 10 patients. RESULTS Ten of 15 patients who started the pretransplant rapamycin treatment completed it. Nine of 10 patients did not complete the induction therapy with ATG, and three of 10 required adaptation of maintenance immunosuppression caused by side effects. Four of 10 patients acquired insulin independence which can be maintained up to year 3 after last infusion. All six other patients have lost their graft, and the early graft loss was associated with lower dose of ATG during induction. CONCLUSION This protocol resulted feasible, safe but less efficient in maintaining graft survival during the time than other T-cell depletion-based protocols. An adequate induction at the first infusion should be considered to improve the overall clinical outcome.
Collapse
|
26
|
Ståhle M, Foss A, Gustafsson B, Lempinen M, Lundgren T, Rafael E, Tufveson G, Korsgren O, Friberg A. Clostripain, the Missing Link in the Enzyme Blend for Efficient Human Islet Isolation. Transplant Direct 2015; 1:e19. [PMID: 27500221 PMCID: PMC4946465 DOI: 10.1097/txd.0000000000000528] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 05/26/2015] [Indexed: 11/23/2022] Open
Abstract
UNLABELLED Effective digestive enzymes are crucial for successful islet isolation. Supplemental proteases are essential as they synergize with collagenase for effective pancreas digestion. The presence of tryptic-like activity has been implicated in efficient enzyme blends and the present study aimed to evaluate if addition of clostripain, an enzyme with tryptic-like activity, could improve efficacy of the islet isolation procedure. METHODS Clostripain was added to the enzyme blend just before pancreas perfusion. Islets were isolated per standard method and numerous isolation parameters, islet quality control, and the number of isolations fulfilling standard transplantation criteria were evaluated. Two control organs per clostripain organ were chosen by blindly matching against body mass index, cold ischemia time, hemoglobin A1c, donor sex, and donor age. RESULTS There were no differences in pancreas weight, dissection time, digestion time, harvest time, percent digested pancreas, or total pellet volume before islet purification between control or clostripain pancreases. Glucose-stimulated insulin release results were similar between groups. Total isolation islet equivalents, purified tissue volume and islet equivalents/g pancreas as well as fulfillment of transplantation criteria favored clostripain processed pancreases. CONCLUSIONS The addition of clostripain to the enzyme blend soundly improved islet yields and transplantation rates. It gently aided pancreas digestion and maintained proper islet functionality. The addition of clostripain to the enzyme blend has now been implemented into standard isolation protocols at the isolation centers in Uppsala and in Oslo.
Collapse
Affiliation(s)
- Magnus Ståhle
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Aksel Foss
- Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Bengt Gustafsson
- Department of Transplantation, University Hospital, Gothenburg, Sweden
| | - Marko Lempinen
- Department of Transplantation and Liver Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Torbjörn Lundgren
- Division of Transplantation Surgery, CLINTEC, Karolinska Institute, Stockholm, Sweden
| | - Ehab Rafael
- Transplantation Unit, Department of Surgery, Skåne University Hospital, Malmö, Sweden
| | - Gunnar Tufveson
- Division of Transplantation Surgery, Department of Surgical Sciences, Uppsala University Hospital, Uppsala, Sweden
| | - Olle Korsgren
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Andrew Friberg
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
27
|
Lebreton F, Pirog A, Belouah I, Bosco D, Berney T, Meda P, Bornat Y, Catargi B, Renaud S, Raoux M, Lang J. Slow potentials encode intercellular coupling and insulin demand in pancreatic beta cells. Diabetologia 2015; 58:1291-9. [PMID: 25788295 DOI: 10.1007/s00125-015-3558-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Accepted: 02/23/2015] [Indexed: 11/24/2022]
Abstract
AIMS/HYPOTHESIS Ion fluxes constitute a major integrative signal in beta cells that leads to insulin secretion and regulation of gene expression. Understanding these electrical signals is important for deciphering the endogenous algorithms used by islets to attain homeostasis and for the design of new sensors for monitoring beta cell function. METHODS Mouse and human islets were cultured on multielectrode arrays (MEAs) for 3-13 days. Extracellular electrical activities received on each electrode were continuously amplified and recorded for offline characterisation. RESULTS Differential band-pass filtering of MEA recordings of mouse islets showed two extracellular voltage waveforms: action potentials (lasting 40-60 ms) and very robust slow potentials (SPs, lasting 800-1,500 ms), the latter of which have not been described previously. The frequency of SPs directly correlated with glucose concentration, peaked at 10 mmol/l glucose and was further augmented by picomolar concentrations of glucagon-like peptide-1. SPs required the closure of ATP-dependent potassium channels as they were induced by glucose or glibenclamide but were not elicited by KCl-induced depolarisation. Pharmacological tools and the use of beta cell specific knockout mice showed that SPs reflected cell coupling via connexin 36. Moreover, increasing and decreasing glucose ramps showed hysteresis with reduced glucose sensitivity during the decreasing phase. SPs were also observed in human islets and could be continuously recorded over 24 h. CONCLUSIONS/INTERPRETATION This novel electrical signature reflects the syncytial function of the islets and is specific to beta cells. Moreover, the observed hysteresis provides evidence for an endogenous algorithm naturally present in islets to protect against hypoglycaemia.
Collapse
Affiliation(s)
- Fanny Lebreton
- CNRS UMR 5248, Chimie et Biologie des Membranes et Nano-objets, Université de Bordeaux, Batiment B14, Allée Geoffroy St Hilaire, CS90063, 33615, Pessac, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
The Choice of Enzyme for Human Pancreas Digestion is a Critical Factor for Increasing the Success of Islet Isolation. Transplant Direct 2015; 1. [PMID: 26146662 DOI: 10.1097/txd.0000000000000522] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND We evaluated three commercially available enzymes for pancreatic digestion by comparing key parameters during the islet isolation process, as well as islet quality post-isolation. METHODS Retrospectively compared and analyzed islet isolations from pancreata using three different enzyme groups: Liberase HI (n=63), Collagenase NB1/Neutral Protease (NP) (n=43), and Liberase Mammalian Tissue Free Collagenase/Thermolysin (MTF C/T) (n=115). A standardized islet isolation and purification method was used. Islet quality assessment was carried out using islet count, viability, in vitro glucose-stimulated insulin secretion (GSIS), glucose-stimulated oxygen consumption rate (ΔOCR), and in vivo transplantation model in mice. RESULTS Donor characteristics were not significantly different among the three enzyme groups used in terms of age, sex, hospital stay duration, cause of death, body mass index (BMI), hemoglobin A1c (HbA1c), cold ischemia time (CIT), and pancreas weight. Digestion efficacy (percentage of digested tissue by weight) was significantly higher in the Liberase MTF C/T group (73.5 ± 1.5 %) when compared to the Liberase HI group (63.6 ± 2.3 %) (p<0.001) and the Collagenase NB1/NP group (61.7 ± 2.9%) (p<0.001). The stimulation index for GSIS was significantly higher in the Liberase MTF C/T group (5.3 ± 0.5) as compared to the Liberase HI (2.9 ± 0.2) (p<0.0001) and the Collagenase NB1/NP (3.6 ± 2.9) (p=0.012) groups. Furthermore, the Liberase MTF C/T enzymes showed the highest success rate of transplantation in diabetic NOD Scid mice (65%), which was significantly higher than the Liberase HI (42%, p=0.001) and the Collagenase NB1/NP enzymes (41%, p<0.001). CONCLUSIONS Liberase MTF C/T is superior to Liberase HI and Collagenase NB1/NP in terms of digestion efficacy and glucose-stimulated insulin secretion in vitro. Moreover, Liberase MTF C/T had a significantly higher success rate of transplantation in diabetic NOD Scid mice compared to Liberase HI and Collagenase NB1/NP enzymes.
Collapse
|
29
|
Lablanche S, Cottet-Rousselle C, Argaud L, Laporte C, Lamarche F, Richard MJ, Berney T, Benhamou PY, Fontaine E. Respective effects of oxygen and energy substrate deprivation on beta cell viability. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1847:629-39. [PMID: 25868875 DOI: 10.1016/j.bbabio.2015.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/30/2015] [Accepted: 04/05/2015] [Indexed: 12/25/2022]
Abstract
Deficit in oxygen and energetic substrates delivery is a key factor in islet loss during islet transplantation. Permeability transition pore (PTP) is a mitochondrial channel involved in cell death. We have studied the respective effects of oxygen and energy substrate deprivation on beta cell viability as well as the involvement of oxidative stress and PTP opening. Energy substrate deprivation for 1h followed by incubation in normal conditions led to a cyclosporin A (CsA)-sensitive-PTP-opening in INS-1 cells and human islets. Such a procedure dramatically decreased INS-1 cells viability except when transient removal of energy substrates was performed in anoxia, in the presence of antioxidant N-acetylcysteine (NAC) or when CsA or metformin inhibited PTP opening. Superoxide production increased during removal of energy substrates and increased again when normal energy substrates were restored. NAC, anoxia or metformin prevented the two phases of oxidative stress while CsA prevented the second one only. Hypoxia or anoxia alone did not induce oxidative stress, PTP opening or cell death. In conclusion, energy substrate deprivation leads to an oxidative stress followed by PTP opening, triggering beta cell death. Pharmacological prevention of PTP opening during islet transplantation may be a suitable option to improve islet survival and graft success.
Collapse
Affiliation(s)
- Sandrine Lablanche
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France.
| | - Cécile Cottet-Rousselle
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | | | - Camille Laporte
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | - Frédéric Lamarche
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France
| | - Marie-Jeanne Richard
- Cellular Therapy Unit, EFS Rhône-Alpes, Grenoble University Hospital, Grenoble, France
| | - Thierry Berney
- Cell Isolation and Transplant Center, University of Geneva, Level R, 1 rue Michel Servet, Geneva 4, CH-1211, Switzerland
| | - Pierre-Yves Benhamou
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France
| | - Eric Fontaine
- University of Grenoble Alpes, LBFA, Grenoble F-38000, France; U1055, INSERM, Grenoble F-38000, France; Department of Endocrinology, Grenoble University Hospital, Grenoble F-38043, France
| |
Collapse
|
30
|
Parnaud G, Lavallard V, Bedat B, Matthey-Doret D, Morel P, Berney T, Bosco D. Cadherin engagement improves insulin secretion of single human β-cells. Diabetes 2015; 64:887-96. [PMID: 25277393 DOI: 10.2337/db14-0257] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The aim of this study was to assess whether cadherin-mediated adhesion of human islet cells was affected by insulin secretagogues and explore the role of cadherins in the secretory activity of β-cells. Experiments were carried out with single islet cells adherent to chimeric proteins made of functional E-, N-, or P-cadherin ectodomains fused to the Fc fragment of immunoglobulin (E-cad/Fc, N-cad/Fc, and P-cad/Fc) and immobilized on an inert substrate. We observed that cadherin expression in islet cells was not affected by insulin secretagogues. Adhesion tests showed that islet cells attached to N-cad/Fc and E-cad/Fc acquired, in a time- and secretagogue-dependent manner, a spreading form that was inhibited by blocking cadherin antibodies. By reverse hemolytic plaque assay, we showed that glucose-stimulated insulin secretion of single β-cells was increased by N-cad/Fc and E-cad/Fc adhesion compared with control. In the presence of E-cad/Fc and after glucose stimulation, we showed that total insulin secretion was six times higher in spreading β-cells compared with round β-cells. Furthermore, cadherin-mediated adhesion induced an asymmetric distribution of cortical actin in β-cells. Our results demonstrate that adhesion of β-cells to E- and N-cadherins is regulated by insulin secretagogues and that E- and N-cadherin engagement promotes stimulated insulin secretion.
Collapse
Affiliation(s)
- Geraldine Parnaud
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Vanessa Lavallard
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Benoît Bedat
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - David Matthey-Doret
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Philippe Morel
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Thierry Berney
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Domenico Bosco
- Department of Surgery, Cell Isolation and Transplantation Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| |
Collapse
|
31
|
Giovannoni L, Muller YD, Lacotte S, Parnaud G, Borot S, Meier RP, Lavallard V, Bédat B, Toso C, Daubeuf B, Elson G, Shang L, Morel P, Kosco-Vilbois M, Bosco D, Berney T. Enhancement of Islet Engraftment and Achievement of Long-Term Islet Allograft Survival by Toll-Like Receptor 4 Blockade. Transplantation 2015; 99:29-35. [DOI: 10.1097/tp.0000000000000468] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
32
|
Impact of anti-insulin antibodies on islet transplantation outcome: data from the GRAGIL Network. Transplantation 2014; 98:475-82. [PMID: 24837539 DOI: 10.1097/tp.0000000000000081] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND In patients with type 1 diabetes, insulin antibodies (IA), altering the pharmacokinetics of circulating insulin, might be associated with high glucose concentration, prolonged hypoglycemia, and higher insulin requirement. The impact of IA on islet transplantation has never been explored. Our aim was to evaluate islet transplantation results at 1 year according to the presence of IA. METHODS Our work is a retrospective, case-control study, comparing IA-negative and IA-positive patients among the cohort of patients with type 1 diabetes transplanted within the Swiss-French GRAGIL network between 2003 and 2010. RESULTS Data about IA were available for 17 patients. Before islet transplantation, 10 patients (59%) were screened positive for IA. At 12 months after transplantation, IA-positive patients reached insulin independence less frequently than IA-negative patients (cumulative incidence of insulin independence, 22.2% vs. 71.4%; P=0.02); β score was ≥7 in 43% of IA-negative patients versus 0% in IA-positive patients (P=0.022). When comparing IA-positive patients with IA-negative patients, insulin dose was 0.15 U/kg (0.10-0.18 U/kg) versus 0.01 U/kg (0-0.09 U/kg) (P=0.2); HbA1c was 6.1% (5.8%-6.3%) versus 6.1% (5.9%-6.8%) (P=0.16); basal C-peptide level was 460 ρmol/L (350-510 ρmol/L) versus 265 ρmol/L (177-405 ρmol/L) (P=0.28); occurrence of hypoglycemia was 12.5% versus 16.5% (P=0.9); and homeostatic model assessment insulin resistance was 1.25 (1-2.4) versus 0.7 (0.52-0.92) (P=0.01). CONCLUSION After islet transplantation, IA-positive patients achieved insulin independence less frequently, exhibiting lower β score and higher homeostatic model assessment insulin resistance compared with IA-negative patients. However, in both groups, islet transplantation restored good glycemic control and drastically reduced hypoglycemia and insulin requirements.
Collapse
|
33
|
Acute nutrient regulation of the mitochondrial glutathione redox state in pancreatic β-cells. Biochem J 2014; 460:411-23. [PMID: 24678915 DOI: 10.1042/bj20131361] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The glucose stimulation of insulin secretion by pancreatic β-cells depends on increased production of metabolic coupling factors, among which changes in NADPH and ROS (reactive oxygen species) may alter the glutathione redox state (EGSH) and signal through changes in thiol oxidation. However, whether nutrients affect EGSH in β-cell subcellular compartments is unknown. Using redox-sensitive GFP2 fused to glutaredoxin 1 and its mitochondria-targeted form, we studied the acute nutrient regulation of EGSH in the cytosol/nucleus or the mitochondrial matrix of rat islet cells. These probes were mainly expressed in β-cells and reacted to low concentrations of exogenous H2O2 and menadione. Under control conditions, cytosolic/nuclear EGSH was close to -300 mV and unaffected by glucose (from 0 to 30 mM). In comparison, mitochondrial EGSH was less negative and rapidly regulated by glucose and other nutrients, ranging from -280 mV in the absence of glucose to -299 mV in 30 mM glucose. These changes were largely independent from changes in intracellular Ca(2+) concentration and in mitochondrial pH. They were unaffected by overexpression of SOD2 (superoxide dismutase 2) and mitochondria-targeted catalase, but were inversely correlated with changes in NAD(P)H autofluorescence, suggesting that they indirectly resulted from increased NADPH availability rather than from changes in ROS concentration. Interestingly, the opposite regulation of mitochondrial EGSH and NAD(P)H autofluorescence by glucose was also observed in human islets isolated from two donors. In conclusion, the present study demonstrates that glucose and other nutrients acutely reduce mitochondrial, but not cytosolic/nuclear, EGSH in pancreatic β-cells under control conditions.
Collapse
|
34
|
Hilling DE, Bouwman E, Terpstra OT, Marang-Van De Mheen PJ. Effects of Donor-, Pancreas-, and Isolation-Related Variables on Human Islet Isolation Outcome: A Systematic Review. Cell Transplant 2014; 23:921-8. [DOI: 10.3727/096368913x666412] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Different factors have been reported to influence islet isolation outcome, but their importance varies between studies and are hampered by the small sample sizes in most studies. The purpose of this study was to perform a systematic review to assess the impact of donor-, pancreas-, and isolation-related variables on successful human islet isolation outcome. PubMed, Embase, and Web of Science were searched electronically in April 2009. All studies reporting on donor-, pancreas-, and isolation-related factors relating to prepurification and postpurification islet isolation yield and proportion of successful islet isolations were selected. Seventy-four retrospective studies had sufficient data and were included in the analyses. Higher pre- and postpurification islet yields and a higher proportion of successful islet isolations were obtained when pancreata were preserved with the two-layer method rather than University of Wisconsin solution in donors with shorter cold ischemia times (CITs) [1 h longer CIT resulted in an average decline of prepurification and postpurification yields and proportion of successful isolations of 59 islet equivalents (IEQs)/g, 54 IEQs/g, and 21%, respectively]. Higher prepurification yields and higher percentage of successful islet isolations were found in younger donors with higher body mass index. Lower yields were found in donation after brain death donors compared to donation after cardiac death donors. Higher postpurification yields were found for isolation with Serva collagenase. This review identified donor-, pancreas-, and isolation-related factors that influence islet isolation yield. Standardized reports of these factors in all future studies may improve the power and identify additional factors and thereby contribute to improving islet isolation yield.
Collapse
Affiliation(s)
- Denise E. Hilling
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Eelco Bouwman
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Onno T. Terpstra
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | | |
Collapse
|
35
|
Meier RPH, Sert I, Morel P, Muller YD, Borot S, Badet L, Toso C, Bosco D, Berney T. Islet of Langerhans isolation from pediatric and juvenile donor pancreases. Transpl Int 2014; 27:949-55. [PMID: 24890668 DOI: 10.1111/tri.12367] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 03/28/2014] [Accepted: 05/26/2014] [Indexed: 11/30/2022]
Abstract
Islet grafts isolated from young donors allow superior functional outcomes but are often associated with poor islet isolation yields. The objective of this study was to comparatively analyze the outcomes of islet isolation between young and older donors. We retrospectively analyzed 564 pancreas isolations performed at our institution. Isolation outcomes were compared between donors aged ≤20 years (n = 42, YD) and >20 years (n = 522, OD). Isolation procedure was identical in both groups. Prepurification percentage of embedded islets was higher in YD (44.3 ± 22.7% vs. 24.9 ± 20.9%, P < 0.001). This led to a lower recovery rate in YD (48% vs. 76%, P = 0.002) and hence lower postpurification IEQ/g pancreas in YD (2 412 ± 1 789 IEQ/g vs. 3 194 ± 1 892 IEQ/g, P = 0.01). Final yield was 180 982 ± 128 073 IEQ in YD and 244 167 ± 134 137 IEQ in OD, (P = 0.006). In vitro function was markedly, albeit nonsignificantly, higher in YD (SI: 4.5 ± 5.1 vs. 3.0 ± 5.7, P = 0.350). Proportion of transplanted preparations was similar in both groups, 38% (16/42) in YD vs. 43% (224/522) in OD, P = 0.628. In spite of isolation and purification difficulties, pancreases from young donors allowed similar islet transplantation rates as older donors. Efforts should be directed at improving islet extraction in these donors to realize their full potential for islet transplantation.
Collapse
Affiliation(s)
- Raphael P H Meier
- Department of Surgery, Cell Isolation and Transplantation Center, University of Geneva Hospitals and School of Medicine, Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Islet transplantation has become a promising treatment for selected patients with type 1 diabetes. Here we provide an overview of the procedure including its history, the process of donor selection, and the techniques and procedures involved in a successful transplant. A brief overview of the current immunosuppressive regimens, the long-term follow-up and the reported outcomes will also be discussed. While islet transplantation is currently generally reserved for adults with type 1 diabetes with severe hypoglycemia or glycemic lability, we herein consider the possibility of its application to the pediatric population.
Collapse
Affiliation(s)
- Michael McCall
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada
| | - A M James Shapiro
- Department of Surgery, University of Alberta, Edmonton, Alberta, Canada; Clinical Islet Transplant Program, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
37
|
Broca C, Varin E, Armanet M, Tourrel-Cuzin C, Bosco D, Dalle S, Wojtusciszyn A. Proteasome dysfunction mediates high glucose-induced apoptosis in rodent beta cells and human islets. PLoS One 2014; 9:e92066. [PMID: 24642635 PMCID: PMC3958412 DOI: 10.1371/journal.pone.0092066] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/17/2014] [Indexed: 01/23/2023] Open
Abstract
The ubiquitin/proteasome system (UPS), a major cellular protein degradation machinery, plays key roles in the regulation of many cell functions. Glucotoxicity mediated by chronic hyperglycaemia is detrimental to the function and survival of pancreatic beta cells. The aim of our study was to determine whether proteasome dysfunction could be involved in beta cell apoptosis in glucotoxic conditions, and to evaluate whether such a dysfunction might be pharmacologically corrected. Therefore, UPS activity was measured in GK rats islets, INS-1E beta cells or human islets after high glucose and/or UPS inhibitor exposure. Immunoblotting was used to quantify polyubiquitinated proteins, endoplasmic reticulum (ER) stress through CHOP expression, and apoptosis through the cleavage of PARP and caspase-3, whereas total cell death was detected through histone-associated DNA fragments measurement. In vitro, we found that chronic exposure of INS-1E cells to high glucose concentrations significantly decreases the three proteasome activities by 20% and leads to caspase-3-dependent apoptosis. We showed that pharmacological blockade of UPS activity by 20% leads to apoptosis in a same way. Indeed, ER stress was involved in both conditions. These results were confirmed in human islets, and proteasome activities were also decreased in hyperglycemic GK rats islets. Moreover, we observed that a high glucose treatment hypersensitized beta cells to the apoptotic effect of proteasome inhibitors. Noteworthily, the decreased proteasome activity can be corrected with Exendin-4, which also protected against glucotoxicity-induced apoptosis. Taken together, our findings reveal an important role of proteasome activity in high glucose-induced beta cell apoptosis, potentially linking ER stress and glucotoxicity. These proteasome dysfunctions can be reversed by a GLP-1 analog. Thus, UPS may be a potent target to treat deleterious metabolic conditions leading to type 2 diabetes.
Collapse
Affiliation(s)
- Christophe Broca
- CNRS UMR 5203, INSERM U661, and Montpellier 1 & 2 University, Institute of Functional Genomics, Montpellier, France
- Laboratory for Diabetes Cell Therapy, Institute for Research in Biotherapy, University Hospital St-Eloi, Montpellier, France
| | - Elodie Varin
- CNRS UMR 5203, INSERM U661, and Montpellier 1 & 2 University, Institute of Functional Genomics, Montpellier, France
- Laboratory for Diabetes Cell Therapy, Institute for Research in Biotherapy, University Hospital St-Eloi, Montpellier, France
| | - Mathieu Armanet
- Laboratory for Diabetes Cell Therapy, Institute for Research in Biotherapy, University Hospital St-Eloi, Montpellier, France
| | - Cécile Tourrel-Cuzin
- B2PE Laboratory (Biology & Pathology of Endocrine Pancreas), BFA Unit, Univ. Paris-Diderot, CNRS EAC4413, Paris, France
| | - Domenico Bosco
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Stéphane Dalle
- CNRS UMR 5203, INSERM U661, and Montpellier 1 & 2 University, Institute of Functional Genomics, Montpellier, France
- Laboratory for Diabetes Cell Therapy, Institute for Research in Biotherapy, University Hospital St-Eloi, Montpellier, France
| | - Anne Wojtusciszyn
- CNRS UMR 5203, INSERM U661, and Montpellier 1 & 2 University, Institute of Functional Genomics, Montpellier, France
- Laboratory for Diabetes Cell Therapy, Institute for Research in Biotherapy, University Hospital St-Eloi, Montpellier, France
- Department of Endocrinology-Diabetes-Nutrition, University Hospital Lapeyronie, Montpellier, France
| |
Collapse
|
38
|
Pasquali L, Gaulton KJ, Rodríguez-Seguí SA, Mularoni L, Miguel-Escalada I, Akerman İ, Tena JJ, Morán I, Gómez-Marín C, van de Bunt M, Ponsa-Cobas J, Castro N, Nammo T, Cebola I, García-Hurtado J, Maestro MA, Pattou F, Piemonti L, Berney T, Gloyn AL, Ravassard P, Skarmeta JLG, Müller F, McCarthy MI, Ferrer J. Pancreatic islet enhancer clusters enriched in type 2 diabetes risk-associated variants. Nat Genet 2014; 46:136-143. [PMID: 24413736 PMCID: PMC3935450 DOI: 10.1038/ng.2870] [Citation(s) in RCA: 396] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 12/12/2013] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes affects over 300 million people, causing severe complications and premature death, yet the underlying molecular mechanisms are largely unknown. Pancreatic islet dysfunction is central for type 2 diabetes pathogenesis, and therefore understanding islet genome regulation could provide valuable mechanistic insights. We have now mapped and examined the function of human islet cis-regulatory networks. We identify genomic sequences that are targeted by islet transcription factors to drive islet-specific gene activity, and show that most such sequences reside in clusters of enhancers that form physical 3D chromatin domains. We find that sequence variants associated with type 2 diabetes and fasting glycemia are enriched in these clustered islet enhancers, and identify trait-associated variants that disrupt DNA-binding and islet enhancer activity. Our studies illustrate how islet transcription factors interact functionally with the epigenome, and provide systematic evidence that dysregulation of islet enhancers is relevant to the mechanisms underlying type 2 diabetes.
Collapse
Affiliation(s)
- Lorenzo Pasquali
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Kyle J Gaulton
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Santiago A Rodríguez-Seguí
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Loris Mularoni
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Irene Miguel-Escalada
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, United Kingdom
| | - İldem Akerman
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Juan J Tena
- Centro Andaluz de Biología del Desarrollo (CABD) CSIC-UPO-Junta de Andalucía, Sevilla, Spain
| | - Ignasi Morán
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Carlos Gómez-Marín
- Centro Andaluz de Biología del Desarrollo (CABD) CSIC-UPO-Junta de Andalucía, Sevilla, Spain
| | - Martijn van de Bunt
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Joan Ponsa-Cobas
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Natalia Castro
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Takao Nammo
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Inês Cebola
- Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| | - Javier García-Hurtado
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Miguel Angel Maestro
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - François Pattou
- University of Lille 2, INSERM U859 Biotherapies Diabete, Lille, France
| | - Lorenzo Piemonti
- Clinical Transplant Unit, San Raffaele Scientific Institute, Milano, Italy
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Department of Surgery, Geneva University Hospitals and University of Geneva, Switzerland
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Philippe Ravassard
- Centre de Recherche de l'Institut du Cerveau et de la Moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris
| | | | - Ferenc Müller
- School of Clinical and Experimental Medicine, College of Medical and Dental Sciences, University of Birmingham, United Kingdom
| | - Mark I McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom.,Oxford Centre for Diabetes, Endocrinology, and Metabolism, Churchill Hospital, Oxford, United Kingdom.,Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Jorge Ferrer
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.,Department of Medicine, Imperial College London, London W12 0NN, United Kingdom
| |
Collapse
|
39
|
Azzi J, Geara AS, El-Sayegh S, Abdi R. Immunological aspects of pancreatic islet cell transplantation. Expert Rev Clin Immunol 2014; 6:111-24. [DOI: 10.1586/eci.09.67] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
40
|
Rheinheimer J, Ziegelmann PK, Carlessi R, Reck LR, Bauer AC, Leitão CB, Crispim D. Different digestion enzymes used for human pancreatic islet isolation: a mixed treatment comparison (MTC) meta-analysis. Islets 2014; 6:e977118. [PMID: 25437379 PMCID: PMC4588164 DOI: 10.4161/19382014.2014.977118] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Collagenases are critical reagents determining yield and quality of isolated human pancreatic islets and may affect islet transplantation outcome. Some islet transplantation centers have compared 2 or more collagenase blends; however, the results regarding differences in quantity and quality of islets are conflicting. Thus, for the first time, a mixed treatment comparison (MTC) meta-analysis was carried out to compile data about the effect of different collagenases used for human pancreas digestion on islet yield, purity, viability and stimulation index (SI). Pubmed, Embase and Cochrane libraries were searched. Of 755 articles retrieved, a total of 15 articles fulfilled the eligibility criteria and were included in the MTC meta-analysis. Our results revealed that Vitacyte and Liberase MTF were associated with a small increase in islet yield (islet equivalent number/g pancreas) when compared with Sevac enzyme [standardized mean difference (95% credible interval - CrI) = -2.19 (-4.25 to -0.21) and -2.28 (-4.49 to -0.23), respectively]. However, all other enzyme comparisons did not show any significant difference regarding islet yield. Purity and viability percentages were not significantly different among any of the analyzed digestion enzymes. Interestingly, Vitacyte and Serva NB1 were associated with increased SI when compared with Liberase MTF enzyme [unstandardized weighted mean difference (95% CrI) = -1.69 (-2.87 to -0.51) and -1.07 (-1.79 to -0.39), respectively]. In conclusion, our MTC meta-analysis suggests that the digestion enzymes currently being used for islet isolation works with similar efficiency regarding islet yield, purity and viability; however, Vitacyte and Serva NB1 enzymes seem to be associated with an improved SI as compared with Liberase MTF.
Collapse
Key Words
- BMI, body mass index
- CIT, cold ischemia time
- CrIs, credible intervals
- FE, fixed effect
- GRADE, grading of recommendations assessment, development and evaluation
- IEQ, islet equivalent number
- MTC, mixed treatment comparison
- NB, neutral protease
- RE, random effect
- SI, stimulation index
- SMD, standardized mean difference
- T1DM, type 1 diabetes mellitus
- WMD, weighted mean difference
- cGMP, current good manufacturing practice
- digestion collagenase
- enzyme
- human islet isolation
- meta-analysis
- mixed treatment comparison
Collapse
Affiliation(s)
- Jakeline Rheinheimer
- Laboratory of Biology of Human Pancreatic Islets; Endocrine Division; Hospital de Clinicas de Porto Alegre; Porto Alegre, Brazil
- Post-Graduation Program in Medical Sciences; Endocrinology; Federal University of Rio Grande do Sul. Porto Alegre; Porto Alegre, Brazil
| | - Patrícia Klarmann Ziegelmann
- Statistics Department and Post-Graduation Program in Cardiology; Federal University of Rio Grande do Sul; Porto Alegre, Brazil
| | - Rodrigo Carlessi
- Laboratory of Biology of Human Pancreatic Islets; Endocrine Division; Hospital de Clinicas de Porto Alegre; Porto Alegre, Brazil
- Post-Graduation Program in Medical Sciences; Endocrinology; Federal University of Rio Grande do Sul. Porto Alegre; Porto Alegre, Brazil
| | - Luciana Ross Reck
- Post-Graduation Program in Medical Sciences; Endocrinology; Federal University of Rio Grande do Sul. Porto Alegre; Porto Alegre, Brazil
| | - Andrea Carla Bauer
- Laboratory of Biology of Human Pancreatic Islets; Endocrine Division; Hospital de Clinicas de Porto Alegre; Porto Alegre, Brazil
| | - Cristiane Bauermann Leitão
- Laboratory of Biology of Human Pancreatic Islets; Endocrine Division; Hospital de Clinicas de Porto Alegre; Porto Alegre, Brazil
- Post-Graduation Program in Medical Sciences; Endocrinology; Federal University of Rio Grande do Sul. Porto Alegre; Porto Alegre, Brazil
| | - Daisy Crispim
- Laboratory of Biology of Human Pancreatic Islets; Endocrine Division; Hospital de Clinicas de Porto Alegre; Porto Alegre, Brazil
- Post-Graduation Program in Medical Sciences; Endocrinology; Federal University of Rio Grande do Sul. Porto Alegre; Porto Alegre, Brazil
- Correspondence to: Daisy Crispim;
| |
Collapse
|
41
|
Quantification of Islet Loss and Graft Functionality During Immune Rejection by 3-Tesla MRI in a Rat Model. Transplantation 2013; 96:438-44. [DOI: 10.1097/tp.0b013e31829b080f] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Rickels MR, Liu C, Shlansky-Goldberg RD, Soleimanpour SA, Vivek K, Kamoun M, Min Z, Markmann E, Palangian M, Dalton-Bakes C, Fuller C, Chiou AJ, Barker CF, Luning Prak ET, Naji A. Improvement in β-cell secretory capacity after human islet transplantation according to the CIT07 protocol. Diabetes 2013; 62:2890-7. [PMID: 23630300 PMCID: PMC3717864 DOI: 10.2337/db12-1802] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 04/24/2013] [Indexed: 12/15/2022]
Abstract
The Clinical Islet Transplantation 07 (CIT07) protocol uses antithymocyte globulin and etanercept induction, islet culture, heparinization, and intensive insulin therapy with the same low-dose tacrolimus and sirolimus maintenance immunosuppression as in the Edmonton protocol. To determine whether CIT07 improves engrafted islet β-cell mass, our center measured β-cell secretory capacity from glucose-potentiated arginine tests at days 75 and 365 after transplantation and compared those results with the results previously achieved by our group using the Edmonton protocol and normal subjects. All subjects were insulin free, with CIT07 subjects receiving fewer islet equivalents from a median of one donor compared with two donors for Edmonton protocol subjects. The acute insulin response to glucose-potentiated arginine (AIRpot) was greater in the CIT07 protocol than in the Edmonton protocol and was less in both cohorts than in normal subjects, with similar findings for C-peptide. The CIT07 subjects who completed reassessment at day 365 exhibited increasing AIRpot by trend relative to that of day 75. These data indicate that engrafted islet β-cell mass is markedly improved with the CIT07 protocol, especially given more frequent use of single islet donors. Although several peritransplant differences may have each contributed to this improvement, the lack of deterioration in β-cell secretory capacity over time in the CIT07 protocol suggests that low-dose tacrolimus and sirolimus are not toxic to islets.
Collapse
Affiliation(s)
- Michael R Rickels
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pulimeno P, Mannic T, Sage D, Giovannoni L, Salmon P, Lemeille S, Giry-Laterriere M, Unser M, Bosco D, Bauer C, Morf J, Halban P, Philippe J, Dibner C. Autonomous and self-sustained circadian oscillators displayed in human islet cells. Diabetologia 2013; 56:497-507. [PMID: 23242133 PMCID: PMC3563957 DOI: 10.1007/s00125-012-2779-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/18/2012] [Indexed: 12/11/2022]
Abstract
AIMS/HYPOTHESIS Following on from the emerging importance of the pancreas circadian clock on islet function and the development of type 2 diabetes in rodent models, we aimed to examine circadian gene expression in human islets. The oscillator properties were assessed in intact islets as well as in beta cells. METHODS We established a system for long-term bioluminescence recording in cultured human islets, employing lentivector gene delivery of the core clock gene Bmal1 (also known as Arntl)-luciferase reporter. Beta cells were stably labelled using a rat insulin2 promoter fluorescent construct. Single-islet/cell oscillation profiles were measured by combined bioluminescence-fluorescence time-lapse microscopy. RESULTS Human islets synchronised in vitro exhibited self-sustained circadian oscillations of Bmal1-luciferase expression at both the population and single-islet levels, with period lengths of 23.6 and 23.9 h, respectively. Endogenous BMAL1 and CRY1 transcript expression was circadian in synchronised islets over 48 h, and antiphasic to REV-ERBα (also known as NR1D1), PER1, PER2, PER3 and DBP transcript circadian profiles. HNF1A and PDX1 exhibited weak circadian oscillations, in phase with the REV-ERBα transcript. Dispersed islet cells were strongly oscillating as well, at population and single-cell levels. Importantly, beta and non-beta cells revealed oscillatory profiles that were well synchronised with each other. CONCLUSIONS/INTERPRETATION We provide for the first time compelling evidence for high-amplitude cell-autonomous circadian oscillators displayed in human pancreatic islets and in dispersed human islet cells. Moreover, these clocks are synchronised between beta and non-beta cells in primary human islet cell cultures.
Collapse
Affiliation(s)
- P. Pulimeno
- Division of Endocrinology, Diabetes and Nutrition, University Hospital of Geneva (HUG), Aile Jura 4-771.2, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland
| | - T. Mannic
- Division of Endocrinology, Diabetes and Nutrition, University Hospital of Geneva (HUG), Aile Jura 4-771.2, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland
| | - D. Sage
- Biomedical Imaging Group, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - L. Giovannoni
- Division of Endocrinology, Diabetes and Nutrition, University Hospital of Geneva (HUG), Aile Jura 4-771.2, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland
| | - P. Salmon
- Department of Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - S. Lemeille
- Division of Endocrinology, Diabetes and Nutrition, University Hospital of Geneva (HUG), Aile Jura 4-771.2, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland
| | - M. Giry-Laterriere
- Department of Neurosciences, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - M. Unser
- Biomedical Imaging Group, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - D. Bosco
- Cell Isolation and Transplantation Centre, University Hospital, Geneva, Switzerland
| | - C. Bauer
- NCCR Frontiers in Genetics, Bioimaging Platform, Sciences II, University of Geneva, Geneva, Switzerland
| | - J. Morf
- Department of Molecular Biology, Sciences II, University of Geneva, Geneva, Switzerland
| | - P. Halban
- Department of Genetic Medicine and Development, University Medical Centre, University of Geneva, Geneva, Switzerland
| | - J. Philippe
- Division of Endocrinology, Diabetes and Nutrition, University Hospital of Geneva (HUG), Aile Jura 4-771.2, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland
| | - C. Dibner
- Division of Endocrinology, Diabetes and Nutrition, University Hospital of Geneva (HUG), Aile Jura 4-771.2, Rue Gabrielle-Perret-Gentil, 4, CH-1211 Geneva, Switzerland
| |
Collapse
|
44
|
Shapiro AMJ. Islet transplantation in type 1 diabetes: ongoing challenges, refined procedures, and long-term outcome. Rev Diabet Stud 2012; 9:385-406. [PMID: 23804275 DOI: 10.1900/rds.2012.9.385] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Remarkable progress has been made in islet transplantation over a span of 40 years. Once just an experimental curiosity in mice, this therapy has moved forward, and can now provide robust therapy for highly selected patients with type 1 diabetes (T1D), refractory to stabilization by other means. This progress could not have occurred without extensive dynamic international collaboration. Currently, 1,085 patients have undergone islet transplantation at 40 international sites since the Edmonton Protocol was reported in 2000 (752 allografts, 333 autografts), according to the Collaborative Islet Transplant Registry. The long-term results of islet transplantation in selected centers now match registry data of pancreas-alone transplantation, with 6 sites reporting five-year insulin independence rates ≥50%. Islet transplantation has been criticized for the use of multiple donor pancreas organs, but progress has also occurred in single-donor success, with 10 sites reporting increased single-donor engraftment. The next wave of innovative clinical trial interventions will address instant blood-mediated inflammatory reaction (IBMIR), apoptosis, and inflammation, and will translate into further marked improvements in single-donor success. Effective control of auto- and alloimmunity is the key to long-term islet function, and high-resolution cellular and antibody-based assays will add considerable precision to this process. Advances in immunosuppression, with new antibody-based targeting of costimulatory blockade and other T-B cellular signaling, will have further profound impact on the safety record of immunotherapy. Clinical trials will move forward shortly to test out new human stem cell derived islets, and in parallel trials will move forward, testing pig islets for compatibility in patients. Induction of immunological tolerance to self-islet antigens and to allografts is a difficult challenge, but potentially within our grasp.
Collapse
Affiliation(s)
- A M James Shapiro
- Clinical Islet Transplant Program, University of Alberta, 2000 College Plaza, 8215 112th Street, Edmonton AB Canada T6G 2C8.
| |
Collapse
|
45
|
Morán I, Akerman İ, van de Bunt M, Xie R, Benazra M, Nammo T, Arnes L, Nakić N, García-Hurtado J, Rodríguez-Seguí S, Pasquali L, Sauty-Colace C, Beucher A, Scharfmann R, van Arensbergen J, Johnson PR, Berry A, Lee C, Harkins T, Gmyr V, Pattou F, Kerr-Conte J, Piemonti L, Berney T, Hanley NA, Gloyn AL, Sussel L, Langman L, Brayman KL, Sander M, McCarthy MI, Ravassard P, Ferrer J. Human β cell transcriptome analysis uncovers lncRNAs that are tissue-specific, dynamically regulated, and abnormally expressed in type 2 diabetes. Cell Metab 2012; 16:435-48. [PMID: 23040067 PMCID: PMC3475176 DOI: 10.1016/j.cmet.2012.08.010] [Citation(s) in RCA: 339] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2012] [Revised: 07/30/2012] [Accepted: 08/31/2012] [Indexed: 02/08/2023]
Abstract
A significant portion of the genome is transcribed as long noncoding RNAs (lncRNAs), several of which are known to control gene expression. The repertoire and regulation of lncRNAs in disease-relevant tissues, however, has not been systematically explored. We report a comprehensive strand-specific transcriptome map of human pancreatic islets and β cells, and uncover >1100 intergenic and antisense islet-cell lncRNA genes. We find islet lncRNAs that are dynamically regulated and show that they are an integral component of the β cell differentiation and maturation program. We sequenced the mouse islet transcriptome and identify lncRNA orthologs that are regulated like their human counterparts. Depletion of HI-LNC25, a β cell-specific lncRNA, downregulated GLIS3 mRNA, thus exemplifying a gene regulatory function of islet lncRNAs. Finally, selected islet lncRNAs were dysregulated in type 2 diabetes or mapped to genetic loci underlying diabetes susceptibility. These findings reveal a new class of islet-cell genes relevant to β cell programming and diabetes pathophysiology.
Collapse
Affiliation(s)
- Ignasi Morán
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - İldem Akerman
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Martijn van de Bunt
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
| | - Ruiyu Xie
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California, USA
| | - Marion Benazra
- Centre de recherche de l’institut du cerveau et de la moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris, France
| | - Takao Nammo
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Metabolic Disorders, Diabetes Research Center, Research Institute, National Center for Global Health and Medicine, Shinjuku-ku, Tokyo, Japan
| | - Luis Arnes
- Department of Genetics and Development, Russ Berrie Medical Pavilion, Columbia University, New York, USA
| | - Nikolina Nakić
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Javier García-Hurtado
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Santiago Rodríguez-Seguí
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Lorenzo Pasquali
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Claire Sauty-Colace
- Centre de recherche de l’institut du cerveau et de la moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris, France
| | - Anthony Beucher
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Raphael Scharfmann
- Institut National de la Santé et de la Recherche Médicale (INSERM) U845, Research Center Growth and Signalling, Paris Descartes University, Sorbonne Paris Cité, Necker Hospital, Paris, France
| | - Joris van Arensbergen
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Paul R Johnson
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford Islet Transplant Programme, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Andrew Berry
- Developmental Biomedicine Research Group, School of Biomedicine, Manchester Academic Health Sciences Centre, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Clarence Lee
- Genome Sequencing Collaborations Group, Life Technologies, Beverly, Massachusetts USA
| | - Timothy Harkins
- Genome Sequencing Collaborations Group, Life Technologies, Beverly, Massachusetts USA
| | - Valery Gmyr
- University of Lille Nord de France, INSERM U859 Biotherapies of Diabete, Lille, France
| | - François Pattou
- University of Lille Nord de France, INSERM U859 Biotherapies of Diabete, Lille, France
| | - Julie Kerr-Conte
- University of Lille Nord de France, INSERM U859 Biotherapies of Diabete, Lille, France
| | - Lorenzo Piemonti
- Diabetes research institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Thierry Berney
- Cell Isolation and Transplantation Center, Geneva, Switzerland
| | - Neil A Hanley
- Developmental Biomedicine Research Group, School of Biomedicine, Manchester Academic Health Sciences Centre, University of Manchester and Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Anna L Gloyn
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, UK
| | - Lori Sussel
- Department of Genetics and Development, Russ Berrie Medical Pavilion, Columbia University, New York, USA
| | - Linda Langman
- Division of Transplantation, Department of Surgery, Center for Cellular Therapy and Biotherapeutics, University of Virginia, USA
| | - Kenneth L Brayman
- Division of Transplantation, Department of Surgery, Center for Cellular Therapy and Biotherapeutics, University of Virginia, USA
| | - Maike Sander
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California, USA
| | - Mark I. McCarthy
- The Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, Oxford, UK
- Diabetes research institute (HSR-DRI), San Raffaele Scientific Institute, Milano, Italy
| | - Philippe Ravassard
- Centre de recherche de l’institut du cerveau et de la moelle, Biotechnology & Biotherapy team, CNRS UMR7225; INSERM U975; University Pierre et Marie Curie, Paris, France
| | - Jorge Ferrer
- Genomic Programming of Beta-cells Laboratory, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Department of Endocrinology and Nutrition, Hospital Clínic de Barcelona, Barcelona, Spain
| |
Collapse
|
46
|
Protective effect of rat pancreatic progenitors cells expressing Pdx1 and nestin on islets survival and function in vitro and in vivo. J Physiol Biochem 2012; 68:603-10. [PMID: 22644623 DOI: 10.1007/s13105-012-0180-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2011] [Accepted: 05/16/2012] [Indexed: 01/08/2023]
Abstract
To maintain islets survival and function is critical in successful pancreatic transplantation. Pancreatic progenitors cells (PPCs) with lineage potentials, giving rise to exocrine, endocrine, and duct cells, reside in developing and adult pancreas. As tissue-specific stem cells, they can produce pancreatic tissue-specific matrix factors to promote islets survival and function. The aim of our research was to investigate the protective effect of rat pancreatic-duodenal homeobox 1 (Pdx1)(+)/nestin(+) PPCs on islets. In vitro, co-culturing islets with Pdx1(+)/nestin(+) PPCs prolonged the former survival from 7 to 14 days. Furthermore, with high glucose (300.8 mg/dl) stimuli, the yield of insulin in co-cultures was significantly higher than that in control group (single islets group). In vivo, co-transplanting islets and Pdx1(+)/nestin(+) PPCs for 3 days, the blood glucose of diabetic rat was significantly decreased to normal level and sustained for 2 weeks. Without Pdx1(+)/nestin(+) PPCs in islets transplantation, hyperglycemia was reversed at day 7 and recovered at day 15. Pathology analysis showed that islets had remnants in co-transplantation at day 21, as complete graft rejection in alone islets transplantation. Our study showed that Pdx1(+)/nestin(+) PPCs displayed the ability of preserving islets viability and function in vitro and prolonging their survival in vivo.
Collapse
|
47
|
Comparative impact on islet isolation and transplant outcome of the preservation solutions Institut Georges Lopez-1, University of Wisconsin, and Celsior. Transplantation 2012; 93:703-8. [PMID: 22343333 DOI: 10.1097/tp.0b013e3182476cc8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Institut Georges Lopez-1 (IGL-1) is a preservation solution similar to University of Wisconsin (UW) with reversed Na/K contents. In this study, we assessed the impact of IGL-1, UW, and Celsior (CS) solutions on islet isolation and transplant outcome. METHODS We retrospectively analyzed 376 islet isolations from pancreases flushed and transported with IGL-1 (n=95), UW (n=204), or CS (n=77). We determined isolation outcome and β-cell function in vitro. Transplanted patients were divided into three groups depending on preservation solution of pancreas, and islet graft function was assessed by decrease in daily insulin needs, C-peptide/glucose ratios, β-scores, and transplant estimated function at 1- and 6-month follow-up. RESULTS IGL-1, UW, and CS groups were similar according to donor age, body mass index, and pancreas weight. There was no difference in islet yields between the three groups. Success rates, transplant rates, β-cell secretory function, and viability were similar for all three groups. We observed no difference in decreased insulin needs, C-peptide glucose ratios, β-scores, and transplant estimated function at 1- and 6-month follow-up between IGL-1, UW, and CS groups. CONCLUSIONS Our study shows that IGL-1 is equivalent to UW or CS solutions for pancreas perfusion and cold storage before islet isolation and transplantation.
Collapse
|
48
|
Danzer C, Eckhardt K, Schmidt A, Fankhauser N, Ribrioux S, Wollscheid B, Müller L, Schiess R, Züllig R, Lehmann R, Spinas G, Aebersold R, Krek W. Comprehensive description of the N-glycoproteome of mouse pancreatic β-cells and human islets. J Proteome Res 2012; 11:1598-608. [PMID: 22148984 DOI: 10.1021/pr2007895] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Cell surface N-glycoproteins provide a key interface of cells to their environment and therapeutic entry points for drug and biomarker discovery. Their comprehensive description denotes therefore a formidable challenge. The β-cells of the pancreas play a crucial role in blood glucose homeostasis, and disruption of their function contributes to diabetes. By combining cell surface and whole cell capturing technologies with high-throughput quantitative proteomic analysis, we report on the identification of a total of 956 unique N-glycoproteins from mouse MIN6 β-cells and human islets. Three-hundred-forty-nine of these proteins encompass potential surface N-glycoproteins and include orphan G-protein-coupled receptors, novel proteases, receptor protein kinases, and phosphatases. Interestingly, stimulation of MIN6 β-cells with glucose and the hormone GLP1, known stimulators of insulin secretion, causes significant changes in surface N-glycoproteome expression. Taken together, this β-cell N-glycoproteome resource provides a comprehensive view on the composition of β-cell surface proteins and expands the scope of signaling systems potentially involved in mediating responses of β-cells to various forms of (patho)physiologic stress and the extent of dynamic remodeling of surface N-glycoprotein expression associated with metabolic and hormonal stimulation. Moreover, it provides a foundation for the development of diabetes medicines that target or are derived from the β-cell surface N-glycoproteome.
Collapse
Affiliation(s)
- Carsten Danzer
- Institute of Cell Biology, ETH Zurich , CH-8093 Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
OBJECTIVES The canine model has been used extensively to improve the human pancreatic islet isolation technique. At the functional level, dog islets show high similarity to human islets and thus can be a helpful tool for islet research. We describe and compare 2 manual isolation methods, M1 (initial) and M2 (modified), and analyze the variables associated with the outcomes, including islet yield, purity, and glucose-stimulated insulin secretion (GSIS). METHODS Male mongrel dogs were used in the study. M2 (n = 7) included higher collagenase concentration, shorter digestion time, faster shaking speed, colder purification temperature, and higher differential density gradient than M1 (n = 7). RESULTS Islet yield was similar between methods (3111.0 ± 309.1 and 3155.8 ± 644.5 islets/g, M1 and M2, respectively; P = 0.951). Pancreas weight and purity together were directly associated with the yield (adjusted R(2) = 0.61; P = 0.002). Purity was considerably improved with M2 (96.7% ± 1.2% vs 75.0% ± 6.3%; P = 0.006). M2 improved GSIS (P = 0.021). Independently, digestion time was inversely associated with GSIS. CONCLUSIONS We describe an isolation method (M2) to obtain a highly pure yield of dog islets with adequate β-cell glucose responsiveness. The isolation variables associated with the outcomes in our canine model confirm previous reports in other species, including humans.
Collapse
|
50
|
Impact of the number of infusions on 2-year results of islet-after-kidney transplantation in the GRAGIL network. Transplantation 2011; 92:1031-8. [PMID: 21926944 DOI: 10.1097/tp.0b013e318230c236] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Insulin independence after islet transplantation is generally achieved after multiple infusions. However, single infusion would increase the number of recipients. Our aim was to evaluate the results of islet-after-kidney transplantation according to the number of infusions. METHODS Islets were isolated at the Geneva University, shipped, and transplanted into French patients from the Swiss-French GRAGIL network, on the "Edmonton" immunosuppression protocol between 2004 and 2010. RESULTS Nineteen patients were transplanted with 33 preparations. Fifteen patients reached 24 months follow-up; eight subjects were single-graft recipients and seven were double-graft recipients. Finally, single-graft recipients received a median of 5312 islet equivalents/kg (5186-6388) vs. 10,564 (10,054-11,375) for double-graft recipients (P=0.0003) with similar islet mass at first infusion. Insulin independence was achieved in five of eight single-graft subjects (62.5%) versus five of seven in double-graft subjects (71.4%), not significant. Median insulin independence duration was 4.7 (3.1-15.2) months after one infusion vs. 19 (9.6-20.8) months after two infusions (not significant). At 24 months posttransplant, comparing single- with double-graft patients, insulin doses were 0.23 (0.11-0.34) U/kg vs. 0.02 (0.0-0.23) U/kg, P=0.11; HbA1c was 6.5% (5.9%-6.8%) vs. 6.2% (5.9%-6.3%), P=0.16; and basal C-peptide was 302 (143-480) pmol/L vs. 599 (393-806) pmol/L, P=0.05. Only 37.5% of single-graft patients had a β-score ≥4 compared with 100% of double-graft patients (P=0.03). Two recipients experienced postinfusion bleeding, and two patients (13%) showed renal dysfunction in the absence of biopsy-proven rejection. CONCLUSIONS One infusion achieves good glycemic control and sometimes insulin independence. However, double-graft patients remain insulin-free longer, tend to have lower HbA1c, and show better graft function 24 months after transplant.
Collapse
|