Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells

Abstract Background Adult human pancreatic beta cells are the “gold standard” for studies on diabetes pathogenesis, but their use is limited by insufficient availability and variable quality. An important effort has recently taken place to differentiate beta cells from human induced pluripotent stem...

Full description

Bibliographic Details
Main Authors: Stéphane Demine, Andrea Alex Schiavo, Sandra Marín-Cañas, Piero Marchetti, Miriam Cnop, Decio L. Eizirik
Format: Article
Language:English
Published: BMC 2020-01-01
Series:Stem Cell Research & Therapy
Subjects:
Online Access:https://doi.org/10.1186/s13287-019-1523-3
id doaj-6d4d18458ce44a819c6737f90ba938f9
record_format Article
spelling doaj-6d4d18458ce44a819c6737f90ba938f92021-01-03T12:05:03ZengBMCStem Cell Research & Therapy1757-65122020-01-0111111510.1186/s13287-019-1523-3Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cellsStéphane Demine0Andrea Alex Schiavo1Sandra Marín-Cañas2Piero Marchetti3Miriam Cnop4Decio L. Eizirik5ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB)ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB)ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB)Department of Clinical and Experimental Medicine, University of PisaULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB)ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB)Abstract Background Adult human pancreatic beta cells are the “gold standard” for studies on diabetes pathogenesis, but their use is limited by insufficient availability and variable quality. An important effort has recently taken place to differentiate beta cells from human induced pluripotent stem cells (iPSCs) and validate their use for diabetes research. We presently used a 7-stage protocol to generate beta cells from human iPSC and evaluated whether these cells are responsive to the pro-inflammatory cytokines (IFNγ, IL-1β, or IFNα) that play a role in type 1 diabetes. Methods The iPSC-derived islet-like cell clusters contained 40–50% beta and 10–15% alpha cells and expressed the receptors for IFNγ, IL-1β, or IFNα. Cells were exposed to either IFNγ (1000 U/mL) + IL-1β (50 U/mL) or IFNα alone (2000 U/mL) for 24/48 h. Apoptosis was quantified using Hoechst/propidium iodide staining or the RealTime Glo Apoptosis Kit (Promega). After treatment, CXCL10 secretion was quantified by ELISA. The expression of multiples genes (Ins, Gcg, Nkx2.2, Nkx6.1, Pdx1, Mafa, BiP, Chop, Atf3, CXCL10, CXCL9, CCL5, and HLA-ABC) was quantified by RT-qPCR. Phosphorylation state and total expression of STAT1/STAT2, as well as expression of PDL1 and of the ER chaperone BiP, were quantified by Western blotting. The co-localization of HLA-ABC or cleaved caspase-3 and Ins/Gcg expression was assessed by immunohistochemistry. The presence of HLA-ABC at the plasma membrane was measured by flow cytometry. Results IFNγ + IL-1β and IFNα induced apoptosis of the cells after 48 h of exposure. Cleaved caspase-3 co-localized mostly but not exclusively with Ins+ cells. Exposure to IFNγ + IL-1β induced a pro-inflammatory phenotype, including increased CXCL10, CXCL9, and CCL5 expression; CXCL10 secretion; and HLA-ABC expression. HLA overexpression was confirmed at the protein level by Western blotting and flow cytometry. Exposure to IFNγ + IL-1β (but not IFNα) also induced beta cell dedifferentiation and endoplasmic reticulum stress (increase in BiP, Chop, and Atf3 mRNA expression). Phosphorylation of STAT1 was stimulated already after 1 h by IFNγ + IL-1β and IFNα, while phosphorylation of STAT2 was only activated by IFNα at 1–4 h. PDL1 expression was increased by both IFNγ + IL-1β and IFNα. Conclusions Our data show that human iPSC-derived beta cells respond to pro-inflammatory cytokines IL-1β + IFNγ and IFNα, by activating the same pathogenic processes as adult human primary beta cells. These cells thus represent a valuable tool for future research on the pathogenesis of type 1 diabetes.https://doi.org/10.1186/s13287-019-1523-3Pancreatic beta cellsType 1 diabetesInduced pluripotent stem cellsCytokinesApoptosisEndoplasmic reticulum stress
collection DOAJ
language English
format Article
sources DOAJ
author Stéphane Demine
Andrea Alex Schiavo
Sandra Marín-Cañas
Piero Marchetti
Miriam Cnop
Decio L. Eizirik
spellingShingle Stéphane Demine
Andrea Alex Schiavo
Sandra Marín-Cañas
Piero Marchetti
Miriam Cnop
Decio L. Eizirik
Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
Stem Cell Research & Therapy
Pancreatic beta cells
Type 1 diabetes
Induced pluripotent stem cells
Cytokines
Apoptosis
Endoplasmic reticulum stress
author_facet Stéphane Demine
Andrea Alex Schiavo
Sandra Marín-Cañas
Piero Marchetti
Miriam Cnop
Decio L. Eizirik
author_sort Stéphane Demine
title Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
title_short Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
title_full Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
title_fullStr Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
title_full_unstemmed Pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human iPSC-derived beta cells
title_sort pro-inflammatory cytokines induce cell death, inflammatory responses, and endoplasmic reticulum stress in human ipsc-derived beta cells
publisher BMC
series Stem Cell Research & Therapy
issn 1757-6512
publishDate 2020-01-01
description Abstract Background Adult human pancreatic beta cells are the “gold standard” for studies on diabetes pathogenesis, but their use is limited by insufficient availability and variable quality. An important effort has recently taken place to differentiate beta cells from human induced pluripotent stem cells (iPSCs) and validate their use for diabetes research. We presently used a 7-stage protocol to generate beta cells from human iPSC and evaluated whether these cells are responsive to the pro-inflammatory cytokines (IFNγ, IL-1β, or IFNα) that play a role in type 1 diabetes. Methods The iPSC-derived islet-like cell clusters contained 40–50% beta and 10–15% alpha cells and expressed the receptors for IFNγ, IL-1β, or IFNα. Cells were exposed to either IFNγ (1000 U/mL) + IL-1β (50 U/mL) or IFNα alone (2000 U/mL) for 24/48 h. Apoptosis was quantified using Hoechst/propidium iodide staining or the RealTime Glo Apoptosis Kit (Promega). After treatment, CXCL10 secretion was quantified by ELISA. The expression of multiples genes (Ins, Gcg, Nkx2.2, Nkx6.1, Pdx1, Mafa, BiP, Chop, Atf3, CXCL10, CXCL9, CCL5, and HLA-ABC) was quantified by RT-qPCR. Phosphorylation state and total expression of STAT1/STAT2, as well as expression of PDL1 and of the ER chaperone BiP, were quantified by Western blotting. The co-localization of HLA-ABC or cleaved caspase-3 and Ins/Gcg expression was assessed by immunohistochemistry. The presence of HLA-ABC at the plasma membrane was measured by flow cytometry. Results IFNγ + IL-1β and IFNα induced apoptosis of the cells after 48 h of exposure. Cleaved caspase-3 co-localized mostly but not exclusively with Ins+ cells. Exposure to IFNγ + IL-1β induced a pro-inflammatory phenotype, including increased CXCL10, CXCL9, and CCL5 expression; CXCL10 secretion; and HLA-ABC expression. HLA overexpression was confirmed at the protein level by Western blotting and flow cytometry. Exposure to IFNγ + IL-1β (but not IFNα) also induced beta cell dedifferentiation and endoplasmic reticulum stress (increase in BiP, Chop, and Atf3 mRNA expression). Phosphorylation of STAT1 was stimulated already after 1 h by IFNγ + IL-1β and IFNα, while phosphorylation of STAT2 was only activated by IFNα at 1–4 h. PDL1 expression was increased by both IFNγ + IL-1β and IFNα. Conclusions Our data show that human iPSC-derived beta cells respond to pro-inflammatory cytokines IL-1β + IFNγ and IFNα, by activating the same pathogenic processes as adult human primary beta cells. These cells thus represent a valuable tool for future research on the pathogenesis of type 1 diabetes.
topic Pancreatic beta cells
Type 1 diabetes
Induced pluripotent stem cells
Cytokines
Apoptosis
Endoplasmic reticulum stress
url https://doi.org/10.1186/s13287-019-1523-3
work_keys_str_mv AT stephanedemine proinflammatorycytokinesinducecelldeathinflammatoryresponsesandendoplasmicreticulumstressinhumanipscderivedbetacells
AT andreaalexschiavo proinflammatorycytokinesinducecelldeathinflammatoryresponsesandendoplasmicreticulumstressinhumanipscderivedbetacells
AT sandramarincanas proinflammatorycytokinesinducecelldeathinflammatoryresponsesandendoplasmicreticulumstressinhumanipscderivedbetacells
AT pieromarchetti proinflammatorycytokinesinducecelldeathinflammatoryresponsesandendoplasmicreticulumstressinhumanipscderivedbetacells
AT miriamcnop proinflammatorycytokinesinducecelldeathinflammatoryresponsesandendoplasmicreticulumstressinhumanipscderivedbetacells
AT decioleizirik proinflammatorycytokinesinducecelldeathinflammatoryresponsesandendoplasmicreticulumstressinhumanipscderivedbetacells
_version_ 1724350851419471872