1
|
Makio T, Chen J, Simmen T. ER stress as a sentinel mechanism for ER Ca 2+ homeostasis. Cell Calcium 2024; 124:102961. [PMID: 39471738 DOI: 10.1016/j.ceca.2024.102961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/01/2024]
Abstract
Endoplasmic reticulum (ER) stress is triggered upon the interference with oxidative protein folding that aims to produce fully folded, disulfide-bonded and glycosylated proteins, which are then competent to exit the ER. Many of the enzymes catalyzing this process require the binding of Ca2+ ions, including the chaperones BiP/GRP78, calnexin and calreticulin. The induction of ER stress with a variety of drugs interferes with chaperone Ca2+ binding, increases cytosolic Ca2+through the opening of ER Ca2+ channels, and activates store-operated Ca2+ entry (SOCE). Posttranslational modifications (PTMs) of the ER Ca2+ handling proteins through ER stress-dependent phosphorylation or oxidation control these mechanisms, as demonstrated in the case of the sarco/endoplasmic reticulum ATPase (SERCA), inositol 1,4,5 trisphosphate receptors (IP3Rs) or stromal interaction molecule 1 (STIM1). Their aim is to restore ER Ca2+ homeostasis but also to increase Ca2+ transfer from the ER to mitochondria during ER stress. This latter function boosts ER bioenergetics, but also triggers apoptosis if ER Ca2+ signaling persists. ER Ca2+ toolkit oxidative modifications upon ER stress can occur within the ER lumen or in the adjacent cytosol. Enzymes involved in this redox control include ER oxidoreductin 1 (ERO1) or the thioredoxin-family protein disulfide isomerases (PDI) and ERp57. A tight, but adaptive connection between ER Ca2+ content, ER stress and mitochondrial readouts allows for the proper functioning of many tissues, including skeletal muscle, the liver, and the pancreas, where ER stress either maintains or compromises their function, depending on its extent and context. Upon mutation of key regulators of ER Ca2+ signaling, diseases such as muscular defects (e.g., from mutated selenoprotein N, SEPN1/SELENON), or diabetes (e.g., from mutated PERK) are the result.
Collapse
Affiliation(s)
- Tadashi Makio
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Junsheng Chen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada
| | - Thomas Simmen
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton T6G2H7, Alberta, Canada.
| |
Collapse
|
2
|
Srivastava N, Vomund AN, Peterson OJ, Abousaway O, Li T, Kain L, Stone P, Clement CC, Sharma S, Zhang B, Liu C, Joglekar AV, Campisi L, Hsieh CS, Santambrogio L, Teyton L, Arbelaez AM, Lichti CF, Wan X. A post-translational cysteine-to-serine conversion in human and mouse insulin generates a diabetogenic neoepitope. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.07.622538. [PMID: 39605669 PMCID: PMC11601459 DOI: 10.1101/2024.11.07.622538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Type 1 diabetes (T1D) affects a genetically susceptible population that develops autoreactive T cells attacking insulin-producing pancreatic β cells. Increasingly, neoantigens are recognized as critical drivers of this autoimmune response. Here, we report a novel insulin neoepitope generated via post-translational cysteine-to-serine conversion (C>S) in human patients, which is also seen in the autoimmune-prone non-obese diabetic (NOD) mice. This modification is driven by oxidative stress within the microenvironment of pancreatic β cells and is further amplified by T1D-relevant inflammatory cytokines, enhancing neoantigen formation in both pancreatic β cells and dendritic cells. We discover that C>S-modified insulin is specifically recognized by CD4 + T cells in human T1D patients and NOD mice. In humans with established T1D, HLA-DQ8-restricted, C>S-specific CD4 + T cells exhibit an activated memory phenotype and lack regulatory signatures. In NOD mice, these neoepitope-specific T cells can orchestrate islet infiltration and promote diabetes progression. Collectively, these data advance a concept that microenvironment-driven and context-dependent post-translational modifications (PTMs) can generate neoantigens that contribute to organ-specific autoimmunity.
Collapse
|
3
|
Roy S, Pokharel P, Piganelli JD. Decoding the immune dance: Unraveling the interplay between beta cells and type 1 diabetes. Mol Metab 2024; 88:101998. [PMID: 39069156 PMCID: PMC11342121 DOI: 10.1016/j.molmet.2024.101998] [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: 05/11/2024] [Revised: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024] Open
Abstract
BACKGROUND Type 1 diabetes (T1D) is an autoimmune disease characterized by the specific destruction of insulin-producing beta cells in the pancreas by the immune system, including CD4 cells which orchestrate the attack and CD8 cells which directly destroy the beta cells, resulting in the loss of glucose homeostasis. SCOPE OF REVIEW This comprehensive document delves into the complex interplay between the immune system and beta cells, aiming to shed light on the mechanisms driving their destruction in T1D. Insights into the genetic predisposition, environmental triggers, and autoimmune responses provide a foundation for understanding the autoimmune attack on beta cells. From the role of viral infections as potential triggers to the inflammatory response of beta cells, an intricate puzzle starts to unfold. This exploration highlights the importance of beta cells in breaking immune tolerance and the factors contributing to their targeted destruction. Furthermore, it examines the potential role of autophagy and the impact of cytokine signaling on beta cell function and survival. MAJOR CONCLUSIONS This review collectively represents current research findings on T1D which offers valuable perspectives on novel therapeutic approaches for preserving beta cell mass, restoring immune tolerance, and ultimately preventing or halting the progression of T1D. By unraveling the complex dynamics between the immune system and beta cells, we inch closer to a comprehensive understanding of T1D pathogenesis, paving the way for more effective treatments and ultimately a cure.
Collapse
Affiliation(s)
- Saptarshi Roy
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Pravil Pokharel
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States
| | - Jon D Piganelli
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, IN, 46202, United States.
| |
Collapse
|
4
|
Fagundes RR, Zaldumbide A, Taylor CT. Role of hypoxia-inducible factor 1 in type 1 diabetes. Trends Pharmacol Sci 2024; 45:798-810. [PMID: 39127527 DOI: 10.1016/j.tips.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/08/2024] [Accepted: 07/11/2024] [Indexed: 08/12/2024]
Abstract
Type 1 diabetes (T1D) is a common autoimmune disease in which dysregulated glucose metabolism is a key feature. T1D is both poorly understood and in need of improved therapeutics. Hypoxia is frequently encountered in multiple tissues in T1D patients including the pancreas and sites of diabetic complications. Hypoxia-inducible factor (HIF)-1, a ubiquitous master regulator of the adaptive response to hypoxia, promotes glucose metabolism through transcriptional and non-transcriptional mechanisms and alters disease progression in multiple preclinical T1D models. However, how HIF-1 activation in β-cells of the pancreas and immune cells (two key cell types in T1D) ultimately affects disease progression remains controversial. We discuss recent advances in our understanding of the role of hypoxia/HIF-1-induced glycolysis in T1D and explore the possible use of drugs targeting this pathway as potential new therapeutics.
Collapse
Affiliation(s)
- Raphael R Fagundes
- Department of Cell and Chemical Biology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical Biology, Leiden University Medical Center, Albinusdreef 2, Leiden, The Netherlands
| | - Cormac T Taylor
- School of Medicine and Conway Institute of Biomolecular and Biomedical Research and Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
5
|
Wang L, Yang S, Zhu G, Li J, Meng G, Chen X, Zhang M, Wang S, Li X, Pan Y, Huang Y, Wang L, Wu Y. Immunopeptidome mining reveals a novel ERS-induced target in T1D. Cell Mol Immunol 2024; 21:604-619. [PMID: 38689020 PMCID: PMC11143349 DOI: 10.1038/s41423-024-01150-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 03/03/2024] [Indexed: 05/02/2024] Open
Abstract
Autoreactive CD8+ T cells play a key role in type 1 diabetes (T1D), but the antigen spectrum that activates autoreactive CD8+ T cells remains unclear. Endoplasmic reticulum stress (ERS) has been implicated in β-cell autoantigen generation. Here, we analyzed the major histocompatibility complex class I (MHC-I)-associated immunopeptidome (MIP) of islet β-cells under steady and ERS conditions and found that ERS reshaped the MIP of β-cells and promoted the MHC-I presentation of a panel of conventional self-peptides. Among them, OTUB258-66 showed immunodominance, and the corresponding autoreactive CD8+ T cells were diabetogenic in nonobese diabetic (NOD) mice. High glucose intake upregulated pancreatic OTUB2 expression and amplified the OTUB258-66-specific CD8+ T-cell response in NOD mice. Repeated OTUB258-66 administration significantly reduced the incidence of T1D in NOD mice. Interestingly, peripheral blood mononuclear cells (PBMCs) from patients with T1D, but not from healthy controls, showed a positive IFN-γ response to human OTUB2 peptides. This study provides not only a new explanation for the role of ERS in promoting β-cell-targeted autoimmunity but also a potential target for the prevention and treatment of T1D. The data are available via ProteomeXchange with the identifier PXD041227.
Collapse
Affiliation(s)
- Lina Wang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, 261053, China
- Department of Immunology, Medical College of Qingdao University, Qingdao, 266071, China
| | - Shushu Yang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Gaohui Zhu
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Jie Li
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Gang Meng
- Department of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiaoling Chen
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Shufeng Wang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Xiangqian Li
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Yu Pan
- Department of Endocrinology, Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Yi Huang
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Infection and Immunity, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China
| | - Li Wang
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Yuzhang Wu
- Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
6
|
James EA, Joglekar AV, Linnemann AK, Russ HA, Kent SC. The beta cell-immune cell interface in type 1 diabetes (T1D). Mol Metab 2023; 78:101809. [PMID: 37734713 PMCID: PMC10622886 DOI: 10.1016/j.molmet.2023.101809] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Revised: 09/01/2023] [Accepted: 09/15/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND T1D is an autoimmune disease in which pancreatic islets of Langerhans are infiltrated by immune cells resulting in the specific destruction of insulin-producing islet beta cells. Our understanding of the factors leading to islet infiltration and the interplay of the immune cells with target beta cells is incomplete, especially in human disease. While murine models of T1D have provided crucial information for both beta cell and autoimmune cell function, the translation of successful therapies in the murine model to human disease has been a challenge. SCOPE OF REVIEW Here, we discuss current state of the art and consider knowledge gaps concerning the interface of the islet beta cell with immune infiltrates, with a focus on T cells. We discuss pancreatic and immune cell phenotypes and their impact on cell function in health and disease, which we deem important to investigate further to attain a more comprehensive understanding of human T1D disease etiology. MAJOR CONCLUSIONS The last years have seen accelerated development of approaches that allow comprehensive study of human T1D. Critically, recent studies have contributed to our revised understanding that the pancreatic beta cell assumes an active role, rather than a passive position, during autoimmune disease progression. The T cell-beta cell interface is a critical axis that dictates beta cell fate and shapes autoimmune responses. This includes the state of the beta cell after processing internal and external cues (e.g., stress, inflammation, genetic risk) that that contributes to the breaking of tolerance by hyperexpression of human leukocyte antigen (HLA) class I with presentation of native and neoepitopes and secretion of chemotactic factors to attract immune cells. We anticipate that emerging insights about the molecular and cellular aspects of disease initiation and progression processes will catalyze the development of novel and innovative intervention points to provide additional therapies to individuals affected by T1D.
Collapse
Affiliation(s)
- Eddie A James
- Center for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Alok V Joglekar
- Center for Systems Immunology and Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amelia K Linnemann
- Center for Diabetes and Metabolic Diseases, and Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Holger A Russ
- Diabetes Institute, University of Florida, Gainesville, FL, USA; Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL, USA
| | - Sally C Kent
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
7
|
Weldemariam MM, Sudhir PR, Woo J, Zhang Q. Effects of multiple stressors on pancreatic human islets proteome reveal new insights into the pathways involved. Proteomics 2023; 23:e2300022. [PMID: 37489002 PMCID: PMC10591809 DOI: 10.1002/pmic.202300022] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 07/26/2023]
Abstract
Pancreatic β-cell dysfunction is an early hallmark of type 1 diabetes mellitus. Among the potentially critical factors that cause β-cell dysfunction are cytokine attack, glucotoxicity, induction of endoplasmic reticulum (ER) or mitochondria stress. However, the exact molecular mechanism underlying β-cell's inability to maintain glucose homeostasis under severe stresses is unknown. This study used proinflammatory cytokines, thapsigargin, and rotenone in the presence of high concentration glucose to mimicking the conditions experienced by dysfunctional β-cells in human pancreatic islets, and profiled the alterations to the islet proteome with TMT-based proteomics. The results were further verified with label-free quantitative proteomics. The differentially expressed proteins under stress conditions reveal that immune related pathways are mostly perturbed by cytokines, while the respiratory electron transport chains and protein processing in ER pathways by rotenone. Thapsigargin together with high glucose induces dramatic increases of proteins in lipid synthesis and peroxisomal protein import pathways, with energy metabolism and vesicle secretion related pathways downregulated. High concentration glucose, on the other hand, alleviated complex I inhibition induced by rotenone. Our results contribute to a more comprehensive understanding of the molecular events involved in β-cell dysfunction.
Collapse
Affiliation(s)
- Mehari Muuz Weldemariam
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
| | - Putty-Reddy Sudhir
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
| | - Jongmin Woo
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
| | - Qibin Zhang
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, North Carolina 28081, United States
- Department of Chemistry and Biochemistry, University of North Carolina at Greensboro, Greensboro, North Carolina 27402, United States
| |
Collapse
|
8
|
The Role of ER Stress in Diabetes: Exploring Pathological Mechanisms Using Wolfram Syndrome. Int J Mol Sci 2022; 24:ijms24010230. [PMID: 36613674 PMCID: PMC9820298 DOI: 10.3390/ijms24010230] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/25/2022] Open
Abstract
The endoplasmic reticulum (ER) is a cytosolic organelle that plays an essential role in the folding and processing of new secretory proteins, including insulin. The pathogenesis of diabetes, a group of metabolic disorders caused by dysfunctional insulin secretion (Type 1 diabetes, T1DM) or insulin sensitivity (Type 2 diabetes, T2DM), is known to involve the excess accumulation of "poorly folded proteins", namely, the induction of pathogenic ER stress in pancreatic β-cells. ER stress is known to contribute to the dysfunction of the insulin-producing pancreatic β-cells. T1DM and T2DM are multifactorial diseases, especially T2DM; both environmental and genetic factors are involved in their pathogenesis, making it difficult to create experimental disease models. In recent years, however, the development of induced pluripotent stem cells (iPSCs) and other regenerative technologies has greatly expanded research capabilities, leading to the development of new candidate therapies. In this review, we will discuss the mechanism by which dysregulated ER stress responses contribute to T2DM pathogenesis. Moreover, we describe new treatment methods targeting protein folding and ER stress pathways with a particular focus on pivotal studies of Wolfram syndrome, a monogenic form of syndromic diabetes caused by pathogenic variants in the WFS1 gene, which also leads to ER dysfunction.
Collapse
|
9
|
Butterfield JSS, Yamada K, Bertolini TB, Syed F, Kumar SRP, Li X, Arisa S, Piñeros AR, Tapia A, Rogers CA, Li N, Rana J, Biswas M, Terhorst C, Kaufman RJ, de Jong YP, Herzog RW. IL-15 blockade and rapamycin rescue multifactorial loss of factor VIII from AAV-transduced hepatocytes in hemophilia A mice. Mol Ther 2022; 30:3552-3569. [PMID: 35821634 PMCID: PMC9734025 DOI: 10.1016/j.ymthe.2022.07.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/09/2022] [Accepted: 07/09/2022] [Indexed: 12/14/2022] Open
Abstract
Hepatic adeno-associated viral (AAV) gene transfer has the potential to cure the X-linked bleeding disorder hemophilia A. However, declining therapeutic coagulation factor VIII (FVIII) expression has plagued clinical trials. To assess the mechanistic underpinnings of this loss of FVIII expression, we developed a hemophilia A mouse model that shares key features observed in clinical trials. Following liver-directed AAV8 gene transfer in the presence of rapamycin, initial FVIII protein expression declines over time in the absence of antibody formation. Surprisingly, loss of FVIII protein production occurs despite persistence of transgene and mRNA, suggesting a translational shutdown rather than a loss of transduced hepatocytes. Some of the animals develop ER stress, which may be linked to hepatic inflammatory cytokine expression. FVIII protein expression is preserved by interleukin-15/interleukin-15 receptor blockade, which suppresses CD8+ T and natural killer cell responses. Interestingly, mice with initial FVIII levels >100% of normal had diminishing expression while still under immune suppression. Taken together, our findings of interanimal variability of the response, and the ability of the immune system to shut down transgene expression without utilizing cytolytic or antibody-mediated mechanisms, illustrate the challenges associated with FVIII gene transfer. Our protocols based upon cytokine blockade should help to maintain efficient FVIII expression.
Collapse
Affiliation(s)
- John S S Butterfield
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32607, USA
| | - Kentaro Yamada
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Thais B Bertolini
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Farooq Syed
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Sandeep R P Kumar
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Xin Li
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Sreevani Arisa
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Annie R Piñeros
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Alejandro Tapia
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Christopher A Rogers
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Ning Li
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Jyoti Rana
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Moanaro Biswas
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA 02215, USA
| | - Randal J Kaufman
- Center for Genetic Disorders and Aging Research, Samford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Roland W Herzog
- Herman B. Wells Center for Pediatric Research, Indiana University, Indianapolis, IN 46202, USA.
| |
Collapse
|
10
|
Yang ML, Kibbey RG, Mamula MJ. Biomarkers of autoimmunity and beta cell metabolism in type 1 diabetes. Front Immunol 2022; 13:1028130. [PMID: 36389721 PMCID: PMC9647083 DOI: 10.3389/fimmu.2022.1028130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/13/2022] [Indexed: 09/10/2023] Open
Abstract
Posttranslational protein modifications (PTMs) are an inherent response to physiological changes causing altered protein structure and potentially modulating important biological functions of the modified protein. Besides cellular metabolic pathways that may be dictated by PTMs, the subtle change of proteins also may provoke immune attack in numerous autoimmune diseases. Type 1 diabetes (T1D) is a chronic autoimmune disease destroying insulin-producing beta cells within the pancreatic islets, a result of tissue inflammation to specific autoantigens. This review summarizes how PTMs arise and the potential pathological consequence of PTMs, with particular focus on specific autoimmunity to pancreatic beta cells and cellular metabolic dysfunction in T1D. Moreover, we review PTM-associated biomarkers in the prediction, diagnosis and in monitoring disease activity in T1D. Finally, we will discuss potential preventive and therapeutic approaches of targeting PTMs in repairing or restoring normal metabolic pathways in pancreatic islets.
Collapse
Affiliation(s)
- Mei-Ling Yang
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Richard G. Kibbey
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Mark J. Mamula
- Section of Rheumatology, Allergy and Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
11
|
Makam AA, Biswas A, Kothegala L, Gandasi NR. Setting the Stage for Insulin Granule Dysfunction during Type-1-Diabetes: Is ER Stress the Culprit? Biomedicines 2022; 10:2695. [PMID: 36359215 PMCID: PMC9687317 DOI: 10.3390/biomedicines10112695] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/07/2022] [Accepted: 10/19/2022] [Indexed: 02/06/2025] Open
Abstract
Type-1-diabetes (T1D) is a multifactorial disorder with a global incidence of about 8.4 million individuals in 2021. It is primarily classified as an autoimmune disorder, where the pancreatic β-cells are unable to secrete sufficient insulin. This leads to elevated blood glucose levels (hyperglycemia). The development of T1D is an intricate interplay between various risk factors, such as genetic, environmental, and cellular elements. In this review, we focus on the cellular elements, such as ER (endoplasmic reticulum) stress and its consequences for T1D pathogenesis. One of the major repercussions of ER stress is defective protein processing. A well-studied example is that of islet amyloid polypeptide (IAPP), which is known to form cytotoxic amyloid plaques when misfolded. This review discusses the possible association between ER stress, IAPP, and amyloid formation in β-cells and its consequences in T1D. Additionally, ER stress also leads to autoantigen generation. This is driven by the loss of Ca++ ion homeostasis. Imbalanced Ca++ levels lead to abnormal activation of enzymes, causing post-translational modification of β-cell proteins. These modified proteins act as autoantigens and trigger the autoimmune response seen in T1D islets. Several of these autoantigens are also crucial for insulin granule biogenesis, processing, and release. Here, we explore the possible associations between ER stress leading to defects in insulin secretion and ultimately β-cell destruction.
Collapse
Affiliation(s)
- Aishwarya A. Makam
- Cell metabolism Lab (GA-08), Department of Molecular Reproduction, Development and Genetics (MRDG), Indian Institute of Science (IISc), Bengaluru 560012, India
| | - Anusmita Biswas
- Cell metabolism Lab (GA-08), Department of Molecular Reproduction, Development and Genetics (MRDG), Indian Institute of Science (IISc), Bengaluru 560012, India
| | - Lakshmi Kothegala
- Cell metabolism Lab (GA-08), Department of Molecular Reproduction, Development and Genetics (MRDG), Indian Institute of Science (IISc), Bengaluru 560012, India
- Unit of Metabolic Physiology, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Nikhil R. Gandasi
- Cell metabolism Lab (GA-08), Department of Molecular Reproduction, Development and Genetics (MRDG), Indian Institute of Science (IISc), Bengaluru 560012, India
- Unit of Metabolic Physiology, University of Gothenburg, 405 30 Gothenburg, Sweden
- Department of Medical Cell Biology, Uppsala University, BMC 571, 751 23 Uppsala, Sweden
| |
Collapse
|
12
|
Hu A, Zou H, Chen B, Zhong J. Posttranslational modifications in diabetes: Mechanisms and functions. Rev Endocr Metab Disord 2022; 23:1011-1033. [PMID: 35697961 DOI: 10.1007/s11154-022-09740-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2022] [Indexed: 12/15/2022]
Abstract
As one of the most widespread chronic diseases, diabetes and its accompanying complications affect approximately one tenth of individuals worldwide and represent a growing cause of morbidity and mortality. Accumulating evidence has proven that the process of diabetes is complex and interactive, involving various cellular responses and signaling cascades by posttranslational modifications (PTMs). Therefore, understanding the mechanisms and functions of PTMs in regulatory networks has fundamental importance for understanding the prediction, onset, diagnosis, progression, and treatment of diabetes. In this review, we offer a holistic summary and illustration of the crosstalk between PTMs and diabetes, including both types 1 and 2. Meanwhile, we discuss the potential use of PTMs in diabetes treatment and provide a prospective direction for deeply understanding the metabolic diseases.
Collapse
Affiliation(s)
- Ang Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China
| | - Haohong Zou
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China
| | - Bin Chen
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China
- The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jianing Zhong
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, 323 National Road, Ganzhou, 341000, Jiangxi, China.
| |
Collapse
|
13
|
Piñeros AR, Kulkarni A, Gao H, Orr KS, Glenn L, Huang F, Liu Y, Gannon M, Syed F, Wu W, Anderson CM, Evans-Molina C, McDuffie M, Nadler JL, Morris MA, Mirmira RG, Tersey SA. Proinflammatory signaling in islet β cells propagates invasion of pathogenic immune cells in autoimmune diabetes. Cell Rep 2022; 39:111011. [PMID: 35767947 PMCID: PMC9297711 DOI: 10.1016/j.celrep.2022.111011] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 04/10/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
Type 1 diabetes is a disorder of immune tolerance that leads to death of insulin-producing islet β cells. We hypothesize that inflammatory signaling within β cells promotes progression of autoimmunity within the islet microenvironment. To test this hypothesis, we deleted the proinflammatory gene encoding 12/15-lipoxygenase (Alox15) in β cells of non-obese diabetic mice at a pre-diabetic time point when islet inflammation is a feature. Deletion of Alox15 leads to preservation of β cell mass, reduces populations of infiltrating T cells, and protects against spontaneous autoimmune diabetes in both sexes. Mice lacking Alox15 in β cells exhibit an increase in a population of β cells expressing the gene encoding the protein programmed death ligand 1 (PD-L1), which engages receptors on immune cells to suppress autoimmunity. Delivery of a monoclonal antibody against PD-L1 recovers the diabetes phenotype in knockout animals. Our results support the contention that inflammatory signaling in β cells promotes autoimmunity during type 1 diabetes progression.
Collapse
Affiliation(s)
- Annie R Piñeros
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Abhishek Kulkarni
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kara S Orr
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Lindsey Glenn
- Department of Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Fei Huang
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maureen Gannon
- Department of Medicine, Vanderbilt University and Department of Veterans Affairs, Tennessee Valley Authority, Nashville, TN, USA
| | - Farooq Syed
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Wenting Wu
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Cara M Anderson
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA
| | - Carmella Evans-Molina
- Department of Pediatrics and the Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA; Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Marcia McDuffie
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, USA
| | - Jerry L Nadler
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Margaret A Morris
- Department of Medicine, Eastern Virginia Medical School, Norfolk, VA, USA
| | - Raghavendra G Mirmira
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA.
| | - Sarah A Tersey
- Department of Medicine and the Kovler Diabetes Center, The University of Chicago, Chicago, IL 60637, USA.
| |
Collapse
|
14
|
Khilji MS, Faridi P, Pinheiro-Machado E, Hoefner C, Dahlby T, Aranha R, Buus S, Nielsen M, Klusek J, Mandrup-Poulsen T, Pandey K, Purcell AW, Marzec MT. Defective Proinsulin Handling Modulates the MHC I Bound Peptidome and Activates the Inflammasome in β-Cells. Biomedicines 2022; 10:biomedicines10040814. [PMID: 35453564 PMCID: PMC9024965 DOI: 10.3390/biomedicines10040814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/23/2022] [Accepted: 03/28/2022] [Indexed: 12/04/2022] Open
Abstract
How immune tolerance is lost to pancreatic β-cell peptides triggering autoimmune type 1 diabetes is enigmatic. We have shown that loss of the proinsulin chaperone glucose-regulated protein (GRP) 94 from the endoplasmic reticulum (ER) leads to mishandling of proinsulin, ER stress, and activation of the immunoproteasome. We hypothesize that inadequate ER proinsulin folding capacity relative to biosynthetic need may lead to an altered β-cell major histocompatibility complex (MHC) class-I bound peptidome and inflammasome activation, sensitizing β-cells to immune attack. We used INS-1E cells with or without GRP94 knockout (KO), or in the presence or absence of GRP94 inhibitor PU-WS13 (GRP94i, 20 µM), or exposed to proinflammatory cytokines interleukin (IL)-1β or interferon gamma (IFNγ) (15 pg/mL and 10 ng/mL, respectively) for 24 h. RT1.A (rat MHC I) expression was evaluated using flow cytometry. The total RT1.A-bound peptidome analysis was performed on cell lysates fractionated by reverse-phase high-performance liquid chromatography (RP-HPLC), followed by liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS). The nucleotide-binding oligomerization domain, leucine rich repeat and pyrin domain containing protein (NLRP1), nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha (IκBα), and (pro) IL-1β expression and secretion were investigated by Western blotting. GRP94 KO increased RT1.A expression in β-cells, as did cytokine exposure compared to relevant controls. Immunopeptidome analysis showed increased RT1.A-bound peptide repertoire in GRP94 KO/i cells as well as in the cells exposed to cytokines. The GRP94 KO/cytokine exposure groups showed partial overlap in their peptide repertoire. Notably, proinsulin-derived peptide diversity increased among the total RT1.A peptidome in GRP94 KO/i along with cytokines exposure. NLRP1 expression was upregulated in GRP94 deficient cells along with decreased IκBα content while proIL-1β cellular levels declined, coupled with increased secretion of mature IL-1β. Our results suggest that limiting β-cell proinsulin chaperoning enhances RT1.A expression alters the MHC-I peptidome including proinsulin peptides and activates inflammatory pathways, suggesting that stress associated with impeding proinsulin handling may sensitize β-cells to immune-attack.
Collapse
Affiliation(s)
- Muhammad Saad Khilji
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
- Department of Physiology, University of Veterinary and Animal Sciences, Lahore 54000, Pakistan
| | - Pouya Faridi
- Department of Medicine, School of Clinical Sciences, Monash Univesity, Clayton, VIC 3168, Australia;
| | - Erika Pinheiro-Machado
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, 9713 Groningen, The Netherlands;
| | - Carolin Hoefner
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
| | - Tina Dahlby
- Laboratory of Translational Nutrition Biology, Department of Health Sciences and Technology, Institute of Food, Nutrition and Health, ETH Zürich, 8603 Zürich, Switzerland;
| | - Ritchlynn Aranha
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
| | - Søren Buus
- Department of Immunology and Microbiology, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Morten Nielsen
- Department of Health Technology, Section for Bioinformatics, Technical University of Denmark, 2800 Lyngby, Denmark;
- Instituto de Investigaciones Biotecnológicas, Universidad Nacional de San Martín, San Martín CP1650, Argentina
| | - Justyna Klusek
- Laboratory of Medical Genetics, Department of Surgical Medicine, Collegium Medicum, Jan Kochanowski University, 25-369 Kielce, Poland;
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
| | - Kirti Pandey
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia; (R.A.); (K.P.)
- Correspondence: (A.W.P.); (M.T.M.); Tel.: +61-39-902-9265 (A.W.P.); +45-25-520-256 (M.T.M.)
| | - Michal T. Marzec
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (M.S.K.); (C.H.); (T.M.-P.)
- Institute of Health Sciences, Collegium Medicum, Jan Kochanowski University, 25-002 Kielce, Poland
- Correspondence: (A.W.P.); (M.T.M.); Tel.: +61-39-902-9265 (A.W.P.); +45-25-520-256 (M.T.M.)
| |
Collapse
|
15
|
Houeiss P, Luce S, Boitard C. Environmental Triggering of Type 1 Diabetes Autoimmunity. Front Endocrinol (Lausanne) 2022; 13:933965. [PMID: 35937815 PMCID: PMC9353023 DOI: 10.3389/fendo.2022.933965] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/20/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease in which pancreatic islet β cells are destroyed by immune cells, ultimately leading to overt diabetes. The progressive increase in T1D incidence over the years points to the role of environmental factors in triggering or accelerating the disease process which develops on a highly multigenic susceptibility background. Evidence that environmental factors induce T1D has mostly been obtained in animal models. In the human, associations between viruses, dietary habits or changes in the microbiota and the development of islet cell autoantibodies or overt diabetes have been reported. So far, prediction of T1D development is mostly based on autoantibody detection. Future work should focus on identifying a causality between the different environmental risk factors and T1D development to improve prediction scores. This should allow developing preventive strategies to limit the T1D burden in the future.
Collapse
Affiliation(s)
- Pamela Houeiss
- Laboratory Immunology of Diabetes, Department EMD, Cochin Institute, INSERMU1016, Paris, France
- Medical Faculty, Paris University, Paris, France
| | - Sandrine Luce
- Laboratory Immunology of Diabetes, Department EMD, Cochin Institute, INSERMU1016, Paris, France
- Medical Faculty, Paris University, Paris, France
| | - Christian Boitard
- Laboratory Immunology of Diabetes, Department EMD, Cochin Institute, INSERMU1016, Paris, France
- Medical Faculty, Paris University, Paris, France
- *Correspondence: Christian Boitard,
| |
Collapse
|
16
|
Li X, Wang L, Meng G, Chen X, Yang S, Zhang M, Zheng Z, Zhou J, Lan Z, Wu Y, Wang L. Sustained high glucose intake accelerates type 1 diabetes in NOD mice. Front Endocrinol (Lausanne) 2022; 13:1037822. [PMID: 36545340 PMCID: PMC9760976 DOI: 10.3389/fendo.2022.1037822] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 11/18/2022] [Indexed: 12/11/2022] Open
Abstract
INTRODUCTION Epidemiological studies have suggested that dietary factors, especially high consumption of high glycaemic index carbohydrates and sugars, may trigger or exacerbate the progression of type 1 diabetes. We aimed to provide experimental evidence to confirm this relevance and to explore the underlying mechanisms. METHODS NOD mice were given sustained high-glucose drinking or glucose-free water and observed for the incidence of type 1 diabetes and islet inflammation. RNAseq was performed to detect the transcriptome changes of the NOD islet beta cell line NIT-1 after high glucose treatment, and mass spectrometry was performed to detect the proteome changes of NIT-1-cells-derived sEVs. RESULTS Sustained high glucose drinking significantly aggravates islet inflammation and accelerates the onset of type 1 diabetes in NOD mice. Mechanistically, high glucose treatment induces aberrant ER stress and up-regulates the expression of autoantigens in islet beta cell. Moreover, high glucose treatment alters the proteome of beta-cells-derived sEVs, and significantly enhances the ability of sEVs to promote DC maturation and stimulate immune inflammatory response. DISCUSSION This study provides evidence for negative effect of high glucose intake as a dietary factor on the pathogenesis of type 1 diabetes in genetically predisposed individuals. Therefore, avoiding high sugar intake may be an effective disease prevention strategy for children or adults susceptible to type 1 diabetes.
Collapse
Affiliation(s)
- Xiangqian Li
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Lina Wang
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- Department of Immunology, College of Basic Medicine, Qingdao University. Qingdao, Shandong, China
- Department of Immunology, College of Basic Medicine, Weifang Medical University, Weifang, China
| | - Gang Meng
- Department of Pathology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaoling Chen
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Shushu Yang
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Mengjun Zhang
- Department of Pharmaceutical Analysis, College of Pharmacy, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhengni Zheng
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Jie Zhou
- Department of Dermatology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhu Lan
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Wang
- Institute of Immunology People's Liberation Army (PLA) & Department of Immunology, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Li Wang,
| |
Collapse
|
17
|
Callebaut A, Bruggeman Y, Zamit C, Sodré FMC, Irla M, Mathieu C, Buitinga M, Overbergh L. Aberrant expression of transglutaminase 2 in pancreas and thymus of NOD mice underscores the importance of deamidation in neoantigen generation. Front Endocrinol (Lausanne) 2022; 13:908248. [PMID: 35966081 PMCID: PMC9367685 DOI: 10.3389/fendo.2022.908248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022] Open
Abstract
Post-translational modifications can lead to a break in immune tolerance in autoimmune diseases such as type 1 diabetes (T1D). Deamidation, the conversion of glutamine to glutamic acid by transglutaminase (TGM) enzymes, is a post-translational modification of interest, with deamidated peptides being reported as autoantigens in T1D. However, little is known about how Tgm2, the most ubiquitously expressed Tgm isoform, is regulated and how tolerance against deamidated peptides is lost. Here, we report on the aberrant expression and regulation of Tgm2 in the pancreas and thymus of NOD mice. We demonstrate that Tgm2 expression is induced by the inflammatory cytokines IL1β and IFNγ in a synergistic manner and that murine pancreatic islets of NOD mice have higher Tgm2 levels, while Tgm2 levels in medullary thymic epithelial cells are reduced. We thus provide the first direct evidence to our knowledge that central tolerance establishment against deamidated peptides might be impaired due to lower Tgm2 expression in NOD medullary thymic epithelial cells, which together with the aberrantly high levels of deamidated peptides in NOD β-cells underscores the role of deamidation in amplifying T-cell reactivity.
Collapse
Affiliation(s)
- Aїsha Callebaut
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Ylke Bruggeman
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Cloé Zamit
- CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
| | - Fernanda Marques Câmara Sodré
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Microbiology, University of Sao Paulo, Sao Paulo, Brazil
| | - Magali Irla
- CNRS, INSERM, Centre d’Immunologie de Marseille-Luminy, Aix-Marseille University, Marseille, France
| | - Chantal Mathieu
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
| | - Mijke Buitinga
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- Department of Nutrition and Movement Sciences, Maastricht University, Maastricht, Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Lut Overbergh
- Laboratory of Clinical and Experimental Endocrinology, Department of Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
- *Correspondence: Lut Overbergh,
| |
Collapse
|
18
|
Sahin GS, Lee H, Engin F. An accomplice more than a mere victim: The impact of β-cell ER stress on type 1 diabetes pathogenesis. Mol Metab 2021; 54:101365. [PMID: 34728341 PMCID: PMC8606542 DOI: 10.1016/j.molmet.2021.101365] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pancreatic β-cells are the insulin factory of an organism with a mission to regulate glucose homeostasis in the body. Due to their high secretory activity, β-cells rely on a functional and intact endoplasmic reticulum (ER). Perturbations to ER homeostasis and unmitigated stress lead to β-cell dysfunction and death. Type 1 diabetes (T1D) is a chronic inflammatory disease caused by the autoimmune-mediated destruction of β-cells. Although autoimmunity is an essential component of T1D pathogenesis, accumulating evidence suggests an important role of β-cell ER stress and aberrant unfolded protein response (UPR) in disease initiation and progression. SCOPE OF REVIEW In this article, we introduce ER stress and the UPR, review β-cell ER stress in various mouse models, evaluate its involvement in inflammation, and discuss the effects of ER stress on β-cell plasticity and demise, and islet autoimmunity in T1D. We also highlight the relationship of ER stress with other stress response pathways and provide insight into ongoing clinical studies targeting ER stress and the UPR for the prevention or treatment of T1D. MAJOR CONCLUSIONS Evidence from ex vivo studies, in vivo mouse models, and tissue samples from patients suggest that β-cell ER stress and a defective UPR contribute to T1D pathogenesis. Thus, restoration of β-cell ER homeostasis at various stages of disease presents a plausible therapeutic strategy for T1D. Identifying the specific functions and regulation of each UPR sensor in β-cells and uncovering the crosstalk between stressed β-cells and immune cells during T1D progression would provide a better understanding of the molecular mechanisms of disease process, and may reveal novel targets for development of effective therapies for T1D.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA; Department of Cell & Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
19
|
Baraja A, Sunarto FR, Adji AS, Handajani F, Rahman FS. Deletion of the RNLS Gene using CRISPR/Cas9 as Pancreatic Cell β Protection against Autoimmune and ER Stress for Type 1 Diabetes Mellitus. Open Access Maced J Med Sci 2021. [DOI: 10.3889/oamjms.2021.7658] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND: Type 1 diabetes mellitus (T1DM) is a chronic disease in children which is usually caused by autoimmunity that damages pancreatic a and b cells which have functions as blood glucose regulators. Some studies stated that Renalase (RNLS) gene deletion will protect these b cells from autoimmune reactions and Endoplasmic Reticulum (ER) stress. RNLS deletion by genome editing Clustered Regular interspersed Short Palindromic Repeats-CRISPR-related (CRISPR/Cas9) is believed to have the potential to be a therapy for T1DM Patients.
AIM: This research was conducted to know the potential of RNLS deletion using the CRISPR/Cas9 as an effective therapy and whether it has a permanent effect on T1DM patients.
METHODS: The method applied in this research summarized articles by analyzing the titles and abstracts of various predetermined keywords. In this case, the author chose a full-text article published within the past 10 years by prioritizing searches in the last 5 years through PubMed, Google Scholar, Science Direct, Cochrane, American Diabetes Association, and official guidelines from IDAI.
RESULTS: RNLS deletion using CRISPR/Cas9 in mice weakened the response of polyclonal -cell-reactive CD8+ T cells and disrupted the immune recognition to cells so that autoimmune killing did occur. In addition, such deletion prevents RNLS ER stress by increasing the threshold, triggering the unfolded protein response so that ER stress is difficult to occur. RNLS mutations in b cells also increase b cell survivability to oxidative stress.
CONCLUSION: b cells RNLS deletion by genome editing CRISPR/Cas9 is effective in protecting b cells from autoimmune reactions and RE stress. However, further research is needed to determine the side effects and safety of its use.
Collapse
|
20
|
Toren E, Burnette KS, Banerjee RR, Hunter CS, Tse HM. Partners in Crime: Beta-Cells and Autoimmune Responses Complicit in Type 1 Diabetes Pathogenesis. Front Immunol 2021; 12:756548. [PMID: 34691077 PMCID: PMC8529969 DOI: 10.3389/fimmu.2021.756548] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease characterized by autoreactive T cell-mediated destruction of insulin-producing pancreatic beta-cells. Loss of beta-cells leads to insulin insufficiency and hyperglycemia, with patients eventually requiring lifelong insulin therapy to maintain normal glycemic control. Since T1D has been historically defined as a disease of immune system dysregulation, there has been little focus on the state and response of beta-cells and how they may also contribute to their own demise. Major hurdles to identifying a cure for T1D include a limited understanding of disease etiology and how functional and transcriptional beta-cell heterogeneity may be involved in disease progression. Recent studies indicate that the beta-cell response is not simply a passive aspect of T1D pathogenesis, but rather an interplay between the beta-cell and the immune system actively contributing to disease. Here, we comprehensively review the current literature describing beta-cell vulnerability, heterogeneity, and contributions to pathophysiology of T1D, how these responses are influenced by autoimmunity, and describe pathways that can potentially be exploited to delay T1D.
Collapse
Affiliation(s)
- Eliana Toren
- Department of Medicine, Division of Endocrinology Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - KaLia S. Burnette
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Ronadip R. Banerjee
- Division of Endocrinology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Chad S. Hunter
- Department of Medicine, Division of Endocrinology Diabetes and Metabolism, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hubert M. Tse
- Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
21
|
Martins CP, New LA, O’Connor EC, Previte DM, Cargill KR, Tse IL, Sims- Lucas S, Piganelli JD. Glycolysis Inhibition Induces Functional and Metabolic Exhaustion of CD4 + T Cells in Type 1 Diabetes. Front Immunol 2021; 12:669456. [PMID: 34163475 PMCID: PMC8216385 DOI: 10.3389/fimmu.2021.669456] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 05/17/2021] [Indexed: 01/10/2023] Open
Abstract
In Type 1 Diabetes (T1D), CD4+ T cells initiate autoimmune attack of pancreatic islet β cells. Importantly, bioenergetic programs dictate T cell function, with specific pathways required for progression through the T cell lifecycle. During activation, CD4+ T cells undergo metabolic reprogramming to the less efficient aerobic glycolysis, similarly to highly proliferative cancer cells. In an effort to limit tumor growth in cancer, use of glycolytic inhibitors have been successfully employed in preclinical and clinical studies. This strategy has also been utilized to suppress T cell responses in autoimmune diseases like Systemic Lupus Erythematosus (SLE), Multiple Sclerosis (MS), and Rheumatoid Arthritis (RA). However, modulating T cell metabolism in the context of T1D has remained an understudied therapeutic opportunity. In this study, we utilized the small molecule PFK15, a competitive inhibitor of the rate limiting glycolysis enzyme 6-phosphofructo-2-kinase/fructose-2,6- biphosphatase 3 (PFKFB3). Our results confirmed PFK15 inhibited glycolysis utilization by diabetogenic CD4+ T cells and reduced T cell responses to β cell antigen in vitro. In an adoptive transfer model of T1D, PFK15 treatment delayed diabetes onset, with 57% of animals remaining euglycemic at the end of the study period. Protection was due to induction of a hyporesponsive T cell phenotype, characterized by increased and sustained expression of the checkpoint molecules PD-1 and LAG-3 and downstream functional and metabolic exhaustion. Glycolysis inhibition terminally exhausted diabetogenic CD4+ T cells, which was irreversible through restimulation or checkpoint blockade in vitro and in vivo. In sum, our results demonstrate a novel therapeutic strategy to control aberrant T cell responses by exploiting the metabolic reprogramming of these cells during T1D. Moreover, the data presented here highlight a key role for nutrient availability in fueling T cell function and has implications in our understanding of T cell biology in chronic infection, cancer, and autoimmunity.
Collapse
Affiliation(s)
- Christina P. Martins
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Lee A. New
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Erin C. O’Connor
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Dana M. Previte
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Kasey R. Cargill
- Department of Pediatrics, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Isabelle L. Tse
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Sunder Sims- Lucas
- Department of Pediatrics, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| | - Jon D. Piganelli
- Department of Pediatric Surgery, Rangos Research Center, UPMC Children’s Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, United States
| |
Collapse
|
22
|
Yang ML, Sodré FMC, Mamula MJ, Overbergh L. Citrullination and PAD Enzyme Biology in Type 1 Diabetes - Regulators of Inflammation, Autoimmunity, and Pathology. Front Immunol 2021; 12:678953. [PMID: 34140951 PMCID: PMC8204103 DOI: 10.3389/fimmu.2021.678953] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/11/2021] [Indexed: 02/06/2023] Open
Abstract
The generation of post-translational modifications (PTMs) in human proteins is a physiological process leading to structural and immunologic variety in proteins, with potentially altered biological functions. PTMs often arise through normal responses to cellular stress, including general oxidative changes in the tissue microenvironment and intracellular stress to the endoplasmic reticulum or immune-mediated inflammatory stresses. Many studies have now illustrated the presence of 'neoepitopes' consisting of PTM self-proteins that induce robust autoimmune responses. These pathways of inflammatory neoepitope generation are commonly observed in many autoimmune diseases including systemic lupus erythematosus, rheumatoid arthritis, multiple sclerosis, and type 1 diabetes (T1D), among others. This review will focus on one specific PTM to self-proteins known as citrullination. Citrullination is mediated by calcium-dependent peptidylarginine deiminase (PAD) enzymes, which catalyze deimination, the conversion of arginine into the non-classical amino acid citrulline. PADs and citrullinated peptides have been associated with different autoimmune diseases, notably with a prominent role in the diagnosis and pathology of rheumatoid arthritis. More recently, an important role for PADs and citrullinated self-proteins has emerged in T1D. In this review we will provide a comprehensive overview on the pathogenic role for PADs and citrullination in inflammation and autoimmunity, with specific focus on evidence for their role in T1D. The general role of PADs in epigenetic and transcriptional processes, as well as their crucial role in histone citrullination, neutrophil biology and neutrophil extracellular trap (NET) formation will be discussed. The latter is important in view of increasing evidence for a role of neutrophils and NETosis in the pathogenesis of T1D. Further, we will discuss the underlying processes leading to citrullination, the genetic susceptibility factors for increased recognition of citrullinated epitopes by T1D HLA-susceptibility types and provide an overview of reported autoreactive responses against citrullinated epitopes, both of T cells and autoantibodies in T1D patients. Finally, we will discuss recent observations obtained in NOD mice, pointing to prevention of diabetes development through PAD inhibition, and the potential role of PAD inhibitors as novel therapeutic strategy in autoimmunity and in T1D in particular.
Collapse
Affiliation(s)
- Mei-Ling Yang
- Section of Rheumatology, Allergy and Clinical Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Fernanda M C Sodré
- Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| | - Mark J Mamula
- Section of Rheumatology, Allergy and Clinical Immunology, Department of Internal Medicine, Yale University, New Haven, CT, United States
| | - Lut Overbergh
- Department of Chronic Diseases, Metabolism and Ageing, Laboratory of Clinical and Experimental Endocrinology (CEE), KU Leuven, Leuven, Belgium
| |
Collapse
|
23
|
Resistance exercise training improves glucose homeostasis by enhancing insulin secretion in C57BL/6 mice. Sci Rep 2021; 11:8574. [PMID: 33883630 PMCID: PMC8060292 DOI: 10.1038/s41598-021-88105-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 04/06/2021] [Indexed: 01/14/2023] Open
Abstract
Resistance exercise exerts beneficial effects on glycemic control, which could be mediated by exercise-induced humoral factors released in the bloodstream. Here, we used C57Bl/6 healthy mice, submitted to resistance exercise training for 10 weeks. Trained mice presented higher muscle weight and maximum voluntary carrying capacity, combined with reduced body weight gain and fat deposition. Resistance training improved glucose tolerance and reduced glycemia, with no alterations in insulin sensitivity. In addition, trained mice displayed higher insulinemia in fed state, associated with increased glucose-stimulated insulin secretion. Islets from trained mice showed reduced expression of genes related to endoplasmic reticulum (ER) stress, associated with increased expression of Ins2. INS-1E beta-cells incubated with serum from trained mice displayed similar pattern of insulin secretion and gene expression than isolated islets from trained mice. When exposed to CPA (an ER stress inducer), the serum from trained mice partially preserved the secretory function of INS-1E cells, and prevented CPA-induced apoptosis. These data suggest that resistance training, in healthy mice, improves glucose homeostasis by enhancing insulin secretion, which could be driven, at least in part, by humoral factors.
Collapse
|
24
|
Rodriguez-Calvo T, Johnson JD, Overbergh L, Dunne JL. Neoepitopes in Type 1 Diabetes: Etiological Insights, Biomarkers and Therapeutic Targets. Front Immunol 2021; 12:667989. [PMID: 33953728 PMCID: PMC8089389 DOI: 10.3389/fimmu.2021.667989] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 03/29/2021] [Indexed: 12/12/2022] Open
Abstract
The mechanisms underlying type 1 diabetes (T1D) pathogenesis remain largely unknown. While autoantibodies to pancreatic beta-cell antigens are often the first biological response and thereby a useful biomarker for identifying individuals in early stages of T1D, their role in T1D pathogenesis is not well understood. Recognition of these antigenic targets by autoreactive T-cells plays a pathological role in T1D development. Recently, several beta-cell neoantigens have been described, indicating that both neoantigens and known T1D antigens escape central or peripheral tolerance. Several questions regarding the mechanisms by which tolerance is broken in T1D remain unanswered. Further delineating the timing and nature of antigenic responses could allow their use as biomarkers to improve staging, as targets for therapeutic intervention, and lead to a better understanding of the mechanisms leading to loss of tolerance. Multiple factors that contribute to cellular stress may result in the generation of beta-cell derived neoepitopes and contribute to autoimmunity. Understanding the cellular mechanisms that induce beta-cells to produce neoantigens has direct implications on development of therapies to intercept T1D disease progression. In this perspective, we will discuss evidence for the role of neoantigens in the pathogenesis of T1D, including antigenic responses and cellular mechanisms. We will additionally discuss the pathways leading to neoepitope formation and the cross talk between the immune system and the beta-cells in this regard. Ultimately, delineating the timing of neoepitope generation in T1D pathogenesis will determine their role as biomarkers as well as therapeutic targets.
Collapse
Affiliation(s)
- Teresa Rodriguez-Calvo
- Institute of Diabetes Research, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health, Munich, Germany
| | - James D. Johnson
- Diabetes Research Group, Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Lut Overbergh
- Laboratory Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Jessica L. Dunne
- Janssen Research and Development, LLC, Raritan, NJ, United States
| |
Collapse
|
25
|
Joshi K, Cameron F, Tiwari S, Mannering SI, Elefanty AG, Stanley EG. Modeling Type 1 Diabetes Using Pluripotent Stem Cell Technology. Front Endocrinol (Lausanne) 2021; 12:635662. [PMID: 33868170 PMCID: PMC8047192 DOI: 10.3389/fendo.2021.635662] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/03/2021] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) technology is increasingly being used to create in vitro models of monogenic human disorders. This is possible because, by and large, the phenotypic consequences of such genetic variants are often confined to a specific and known cell type, and the genetic variants themselves can be clearly identified and controlled for using a standardized genetic background. In contrast, complex conditions such as autoimmune Type 1 diabetes (T1D) have a polygenic inheritance and are subject to diverse environmental influences. Moreover, the potential cell types thought to contribute to disease progression are many and varied. Furthermore, as HLA matching is critical for cell-cell interactions in disease pathogenesis, any model that seeks to test the involvement of particular cell types must take this restriction into account. As such, creation of an in vitro model of T1D will require a system that is cognizant of genetic background and enables the interaction of cells representing multiple lineages to be examined in the context of the relevant environmental disease triggers. In addition, as many of the lineages critical to the development of T1D cannot be easily generated from iPSCs, such models will likely require combinations of cell types derived from in vitro and in vivo sources. In this review we imagine what an ideal in vitro model of T1D might look like and discuss how the required elements could be feasibly assembled using existing technologies. We also examine recent advances towards this goal and discuss potential uses of this technology in contributing to our understanding of the mechanisms underlying this autoimmune condition.
Collapse
Affiliation(s)
- Kriti Joshi
- Department of Endocrinology and Metabolism, All India Institute of Medical Sciences Rishikesh, Uttarakhand, India
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
| | - Fergus Cameron
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Endocrinology and Diabetes, The Royal Children’s Hospital, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
| | - Swasti Tiwari
- Department of Molecular Medicine & Biotechnology, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Lucknow, India
| | - Stuart I. Mannering
- Immunology and Diabetes Unit, St. Vincent’s Institute of Medical Research, Fitzroy, Vic, Australia
| | - Andrew G. Elefanty
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| | - Edouard G. Stanley
- Department of Cell Biology, Murdoch Children’s Research Institute, Parkville, Vic, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Vic, Australia
| |
Collapse
|
26
|
Non-Genetically Encoded Epitopes Are Relevant Targets in Autoimmune Diabetes. Biomedicines 2021; 9:biomedicines9020202. [PMID: 33671312 PMCID: PMC7922826 DOI: 10.3390/biomedicines9020202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 12/16/2022] Open
Abstract
Islet antigen reactive T cells play a key role in promoting beta cell destruction in type 1 diabetes (T1D). Self-reactive T cells are typically deleted through negative selection in the thymus or deviated to a regulatory phenotype. Nevertheless, those processes are imperfect such that even healthy individuals have a reservoir of potentially autoreactive T cells. What remains less clear is how tolerance is lost to insulin and other beta cell specific antigens. Islet autoantibodies, the best predictor of disease risk, are known to recognize classical antigens such as proinsulin, GAD65, IA-2, and ZnT8. These antibodies are thought to be supported by the expansion of autoreactive CD4+ T cells that recognize these same antigenic targets. However, recent studies have identified new classes of non-genetically encoded epitopes that may reflect crucial gaps in central and peripheral tolerance. Notably, some of these specificities, including epitopes from enzymatically post-translationally modified antigens and hybrid insulin peptides, are present at relatively high frequencies in the peripheral blood of patients with T1D. We conclude that CD4+ T cells that recognize non-genetically encoded epitopes are likely to make an important contribution to the progression of islet autoimmunity in T1D. We further propose that these classes of neo-epitopes should be considered as possible targets for strategies to induce antigen specific tolerance.
Collapse
|
27
|
Callebaut A, Derua R, Vig S, Delong T, Mathieu C, Overbergh L. Identification of Deamidated Peptides in Cytokine-Exposed MIN6 Cells through LC-MS/MS Using a Shortened Digestion Time and Inspection of MS2 Spectra. J Proteome Res 2020; 20:1405-1414. [PMID: 33372785 DOI: 10.1021/acs.jproteome.0c00801] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Enzymatic deamidation, the conversion of glutamine (Gln) into glutamic acid (Glu) residues, mediated by tissue transglutaminase enzymes, can provoke autoimmunity by generating altered self-epitopes, a process well-known in celiac disease and more recently also described in type 1 diabetes (T1D). To identify deamidated proteins, liquid chromatography-tandem mass spectrometry is the method of choice. However, as nonenzymatic deamidations on asparagine (Asn) and to a minor extent on Gln are frequently induced in vitro during proteomics sample preparation, the accurate detection of in vivo deamidation can be hampered. Here we report on the optimization of a method to reduce in vitro generated deamidation by 70% using improved trypsin digestion conditions (90 min/pH 8). We also point to the critical importance of manual inspection of MS2 spectra, considering that only 55% of the high quality peptides with Gln deamidation were assigned correctly using an automated search algorithm. As proof of principal, using these criteria, we showed a significant increase in levels of both Asn and Gln deamidation in cytokine-exposed murine MIN6 β-cells, paralleled by an increase in tissue transglutaminase activity. These findings add evidence to the hypothesis that deamidation is occurring in stressed β-cell proteins and can be involved in the autoimmune process in T1D.
Collapse
Affiliation(s)
- Aïsha Callebaut
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, 3000 Leuven, Belgium.,SyBioMa, KU Leuven, 3000 Leuven, Belgium
| | - Saurabh Vig
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Thomas Delong
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz, Aurora, Colorado 80045, United States
| | - Chantal Mathieu
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
28
|
Madrid FF, Grossman LI, Aras S. Mitochondria Autoimmunity and MNRR1 in Breast Carcinogenesis: A Review. JOURNAL OF CANCER IMMUNOLOGY 2020; 2:138-158. [PMID: 33615312 PMCID: PMC7894625 DOI: 10.33696/cancerimmunol.2.027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
We review here the evidence for participation of mitochondrial autoimmunity in BC inception and progression and propose a new paradigm that may challenge the prevailing thinking in oncogenesis by suggesting that mitochondrial autoimmunity is a major contributor to breast carcinogenesis and probably to the inception and progression of other solid tumors. It has been shown that MNRR1 mediated mitochondrial-nuclear function promotes BC cell growth and migration and the development of metastasis and constitutes a proof of concept supporting the participation of mitochondrial autoimmunity in breast carcinogenesis. The resemblance of the autoantibody profile in BC detected by IFA with that in the rheumatic autoimmune diseases suggested that studies on the autoantibody response to tumor associated antigens and the characterization of the mtDNA- and nDNA-encoded antigens may provide functional data on breast carcinogenesis. We also review the studies supporting the view that a panel of autoreactive nDNA-encoded mitochondrial antigens in addition to MNRR1 may be involved in breast carcinogenesis. These include GAPDH, PKM2, GSTP1, SPATA5, MFF, ncRNA PINK1-AS/DDOST as probably contributing to BC progression and metastases and the evidence suggesting that DDX21 orchestrates a complex signaling network with participation of JUND and ATF3 driving chronic inflammation and breast tumorigenesis. We suggest that the widespread autoreactivity of mtDNA- and nDNA-encoded mitochondrial proteins found in BC sera may be the reflection of autoimmunity triggered by mitochondrial and non-mitochondrial tumor associated antigens involved in multiple tumorigenic pathways. Furthermore, we suggest that mitochondrial proteins may contribute to mitochondrial dysfunction in BC even if mitochondrial respiration is found to be within normal limits. However, although the studies show that mitochondrial autoimmunity is a major factor in breast cancer inception and progression, it is not the only factor since there is a multiplex autoantibody profile targeting centrosome and stem cell antigens as well as anti-idiotypic antibodies, revealing the complex signaling network involved in breast carcinogenesis. In summary, the studies reviewed here open new, unexpected therapeutic avenues for cancer prevention and treatment of patients with cancer derived from an entirely new perspective of breast carcinogenesis.
Collapse
Affiliation(s)
- Félix Fernández Madrid
- Department of Medicine, Division of Rheumatology, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201 USA
- Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Lawrence I. Grossman
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201 USA
| | - Siddhesh Aras
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI 48201 USA
| |
Collapse
|
29
|
Cai EP, Ishikawa Y, Zhang W, Leite NC, Li J, Hou S, Kiaf B, Hollister-Lock J, Yilmaz NK, Schiffer CA, Melton DA, Kissler S, Yi P. Genome-scale in vivo CRISPR screen identifies RNLS as a target for beta cell protection in type 1 diabetes. Nat Metab 2020; 2:934-945. [PMID: 32719542 PMCID: PMC7502486 DOI: 10.1038/s42255-020-0254-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 06/26/2020] [Indexed: 02/06/2023]
Abstract
Type 1 diabetes (T1D) is caused by the autoimmune destruction of pancreatic beta cells. Pluripotent stem cells can now be differentiated into beta cells, thus raising the prospect of a cell replacement therapy for T1D. However, autoimmunity would rapidly destroy newly transplanted beta cells. Using a genome-scale CRISPR screen in a mouse model for T1D, we show that deleting RNLS, a genome-wide association study candidate gene for T1D, made beta cells resistant to autoimmune killing. Structure-based modelling identified the U.S. Food and Drug Administration-approved drug pargyline as a potential RNLS inhibitor. Oral pargyline treatment protected transplanted beta cells in diabetic mice, thus leading to disease reversal. Furthermore, pargyline prevented or delayed diabetes onset in several mouse models for T1D. Our results identify RNLS as a modifier of beta cell vulnerability and as a potential therapeutic target to avert beta cell loss in T1D.
Collapse
Affiliation(s)
- Erica P Cai
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Yuki Ishikawa
- Section for Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Wei Zhang
- Section for Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nayara C Leite
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Jian Li
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Shurong Hou
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Badr Kiaf
- Section for Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Jennifer Hollister-Lock
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Nese Kurt Yilmaz
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Celia A Schiffer
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Douglas A Melton
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| | - Peng Yi
- Islet Cell and Regenerative Biology, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
30
|
Mudali D, Jeevanandam J, Danquah MK. Probing the characteristics and biofunctional effects of disease-affected cells and drug response via machine learning applications. Crit Rev Biotechnol 2020; 40:951-977. [PMID: 32633615 DOI: 10.1080/07388551.2020.1789062] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Drug-induced transformations in disease characteristics at the cellular and molecular level offers the opportunity to predict and evaluate the efficacy of pharmaceutical ingredients whilst enabling the optimal design of new and improved drugs with enhanced pharmacokinetics and pharmacodynamics. Machine learning is a promising in-silico tool used to simulate cells with specific disease properties and to determine their response toward drug uptake. Differences in the properties of normal and infected cells, including biophysical, biochemical and physiological characteristics, plays a key role in developing fundamental cellular probing platforms for machine learning applications. Cellular features can be extracted periodically from both the drug treated, infected, and normal cells via image segmentations in order to probe dynamic differences in cell behavior. Cellular segmentation can be evaluated to reflect the levels of drug effect on a distinct cell or group of cells via probability scoring. This article provides an account for the use of machine learning methods to probe differences in the biophysical, biochemical and physiological characteristics of infected cells in response to pharmacokinetics uptake of drug ingredients for application in cancer, diabetes and neurodegenerative disease therapies.
Collapse
Affiliation(s)
- Deborah Mudali
- Department of Computer Science, University of Tennessee, Chattanooga, TN, USA
| | - Jaison Jeevanandam
- Department of Chemical Engineering, Faculty of Engineering and Science, Curtin University, Miri, Malaysia
| | - Michael K Danquah
- Chemical Engineering Department, University of Tennessee, Chattanooga, TN, USA
| |
Collapse
|
31
|
Tong Y, Yang L, Shao F, Yan X, Li X, Huang G, Xiao Y, Zhou Z. Distinct secretion pattern of serum proinsulin in different types of diabetes. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:452. [PMID: 32395496 PMCID: PMC7210169 DOI: 10.21037/atm.2020.03.189] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background Latent autoimmune diabetes in adults (LADA) is characterized by autoimmunity, late-onset and intermediate beta-cell deprivation rate between type 2 diabetes mellitus (T2DM) and type 1 diabetes mellitus (T1DM). Herein, we investigated proinsulin (PI) secretion patterns and the endoplasmic reticulum (ER) dysfunction biomarker, PI-to-C-peptide (PI:CP) ratio, to elucidate beta-cell intrinsic pathogenesis mechanisms in different types of diabetes. Methods Total serum fasting PI (FPI) were measured in adult-onset and newly-diagnosed diabetes patients, including 60 T1DM, 60 LADA and 60 T2DM. Thirty of each type underwent mixed meal tolerance tests (MMTTs), and hence 120 min postprandial PI (PPI) were detected. PI:CP ratio = PI (pmol/L) ÷ CP (pmol/L) × 100%. PI-related measurements among types of diabetes were compared. Correlation between PI-related measurements and beta-cell autoimmunity were analyzed. The possibility of discriminating LADA from T1DM and T2DM with PI-related measurements were tested. Results FPI and PPI were significantly higher in LADA than T1DM (P<0.001 for both comparisons), but lower than those in T2DM (P<0.001 and P=0.026, respectively). Fasting PI:CP ratio was significantly higher in T1DM than both LADA and T2DM (median 3.25% vs. 2.13% and 2.32%, P=0.011 and P=0.017, respectively). In LADA, positive autoantibody numbers increased by both fasting and postprandial PI:CP ratio (P=0.007 and P=0.034, respectively). Areas under receiver operation characteristic curves (AUCROC) of FPI and PPI for discriminating LADA from adult-onset T1DM were 0.751 (P<0.001) and 0.838 (P<0.001), respectively. Between LADA and T2DM, AUCROC of FPI and PPI were 0.685 (P<0.001) and 0.741 (P=0.001), respectively. Conclusions In the development of autoimmune diabetes, interplays between ER stress and beta-cell autoimmunity are potentially responsible for severer beta-cell destruction. PI-related measurements could help in differentiating LADA from adult-onset T1DM and T2DM.
Collapse
Affiliation(s)
- Yue Tong
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Lin Yang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Feng Shao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Xiang Yan
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Xia Li
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Yang Xiao
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha 410011, China.,National Clinical Research Center for Metabolic Diseases, Changsha 410011, China.,Key Laboratory of Diabetes Immunology, Central South University, Ministry of Education, Changsha 410011, China
| |
Collapse
|
32
|
Kim JY, Kim MH, Lee HJ, Huh JW, Lee SR, Lee HS, Lee DS. Peroxiredoxin 4 inhibits insulin-induced adipogenesis through regulation of ER stress in 3T3-L1 cells. Mol Cell Biochem 2020; 468:97-109. [PMID: 32185676 DOI: 10.1007/s11010-020-03714-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/06/2020] [Indexed: 02/06/2023]
Abstract
Obesity was originally considered a disease endemic to developed countries but has since emerged as a global health problem. Obesity is characterized by abnormal or excessive lipid accumulation (World Health Organization, WHO) resulting from pre-adipocyte differentiation (adipogenesis). The endoplasmic reticulum (ER) produces proteins and cholesterol and shuttles these compounds to their target sites. Many studies have implicated ER stress, indicative of ER dysfunction, in adipogenesis. Reactive oxygen species (ROS) are also known to be involved in pre-adipocyte differentiation. Prx4 specific to the ER lumen exhibits ROS scavenging activity, and we thereby focused on ER-specific Prx4 in tracking changes in adipocyte differentiation and lipid accumulation. Overexpression of Prx4 reduced ER stress and suppressed lipid accumulation by regulating adipogenic gene expression during adipogenesis. Our results demonstrate that Prx4 inhibits ER stress, lowers ROS levels, and attenuates pre-adipocyte differentiation. These findings suggested enhancing the activity of Prx4 may be helpful in the treatment of obesity; the data also support the development of new therapeutic approaches to obesity and obesity-related metabolic disorders.
Collapse
Affiliation(s)
- Jae Yeop Kim
- School of Life Sciences, BK21 Plus KNU Creative Bioresearch Group, Kyungpook National University, Daegu, Republic of Korea.,School of Life Sciences & Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Mi Hye Kim
- School of Life Sciences, BK21 Plus KNU Creative Bioresearch Group, Kyungpook National University, Daegu, Republic of Korea.,School of Life Sciences & Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Hong Jun Lee
- College of Medicine, Chungbuk National University, Chungbuk, Republic of Korea.,Research Institute, E-Biogen Inc, Seoul, Korea
| | - Jae-Won Huh
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
| | - Sang-Rae Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju, Republic of Korea
| | - Hyun-Shik Lee
- School of Life Sciences, BK21 Plus KNU Creative Bioresearch Group, Kyungpook National University, Daegu, Republic of Korea.,School of Life Sciences & Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Dong-Seok Lee
- School of Life Sciences, BK21 Plus KNU Creative Bioresearch Group, Kyungpook National University, Daegu, Republic of Korea. .,School of Life Sciences & Biotechnology, College of Natural Sciences, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
33
|
Chen J, Chen J, Cheng Y, Fu Y, Zhao H, Tang M, Zhao H, Lin N, Shi X, Lei Y, Wang S, Huang L, Wu W, Tan J. Mesenchymal stem cell-derived exosomes protect beta cells against hypoxia-induced apoptosis via miR-21 by alleviating ER stress and inhibiting p38 MAPK phosphorylation. Stem Cell Res Ther 2020; 11:97. [PMID: 32127037 PMCID: PMC7055095 DOI: 10.1186/s13287-020-01610-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 02/01/2020] [Accepted: 02/14/2020] [Indexed: 02/08/2023] Open
Abstract
Background Hypoxia is a major cause of beta cell death and dysfunction after transplantation. The aim of this study was to investigate the effect of exosomes derived from mesenchymal stem cells (MSCs) on beta cells under hypoxic conditions and the potential underlying mechanisms. Methods Exosomes were isolated from the conditioned medium of human umbilical cord MSCs and identified by WB, NTA, and transmission electron microscopy. Beta cells (βTC-6) were cultured in serum-free medium in the presence or absence of exosomes under 2% oxygen conditions. Cell viability and apoptosis were analysed with a CCK-8 assay and a flow cytometry-based annexin V-FITC/PI apoptosis detection kit, respectively. Endoplasmic reticulum stress (ER stress) proteins and apoptosis-related proteins were detected by the WB method. MiRNAs contained in MSC exosomes were determined by Illumina HiSeq, and treatment with specific miRNA mimics or inhibitors of the most abundant miRNAs was used to reveal the underlying mechanism of exosomes. Results Exosomes derived from MSC-conditioned culture medium were 40–100 nm in diameter and expressed the exosome markers CD9, CD63, CD81, HSP70, and Flotillin 1, as well as the MSC markers CD73, CD90, and CD105. Hypoxia significantly induced beta cell apoptosis, while MSC exosomes remarkably improved beta cell survival. The WB results showed that ER stress-related proteins, including GRP78, GRP94, p-eIF2α and CHOP, and the apoptosis-related proteins cleaved caspase 3 and PARP, were upregulated under hypoxic conditions but were inhibited by MSC exosomes. Moreover, the p38 MAPK signalling pathway was activated by hypoxia and was inhibited by MSC exosomes. The Illumina HiSeq results show that MSC exosomes were rich in miR-21, let-7 g, miR-1246, miR-381, and miR-100. After transfection with miRNA mimics, the viability of beta cells under hypoxia was increased significantly by miR-21 mimic, and the p38 MAPK and ER stress-related proteins in beta cells were downregulated. These changes were reversed after exosomes were pretreated with miR-21 inhibitor. Conclusions Exosomes derived from MSCs could protect beta cells against apoptosis induced by hypoxia, largely by carrying miR-21, alleviating ER stress and inhibiting p38 MAPK signalling. This result indicated that MSC exosomes might improve encapsulated islet survival and benefit diabetes patients.
Collapse
Affiliation(s)
- Jin Chen
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China. .,Organ Transplant Institute, 900th Hospital, Clinical Medical Institute of Fujian Medical University, 156th XiErHuan Road, Fuzhou, 350025, China.
| | - Junqiu Chen
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China.,Xiangyang Central Hospital, Hubei University of Arts and Science, Xiangyang, 441000, China
| | - Yuanhang Cheng
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Yunfeng Fu
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Hongzhou Zhao
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Minying Tang
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Hu Zhao
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Na Lin
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Xiaohua Shi
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Yan Lei
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Shuiliang Wang
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China. .,Organ Transplant Institute, 900th Hospital, Clinical Medical Institute of Fujian Medical University, 156th XiErHuan Road, Fuzhou, 350025, China.
| | - Lianghu Huang
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China
| | - Weizhen Wu
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China. .,Organ Transplant Institute, 900th Hospital, Clinical Medical Institute of Fujian Medical University, 156th XiErHuan Road, Fuzhou, 350025, China.
| | - Jianming Tan
- Fujian Provincial Key Laboratory of Transplant Biology, 900th Hospital, Xiamen University, 156th XiErHuan Road, Fuzhou, 350025, China.,Organ Transplant Institute, 900th Hospital, Clinical Medical Institute of Fujian Medical University, 156th XiErHuan Road, Fuzhou, 350025, China
| |
Collapse
|
34
|
Johar D, Ahmed SM, El Hayek S, Al-Dewik N, Bahbah EI, Omar NH, Mustafa M, Salman DO, Fahmey A, Mottawea M, Azouz RAM, Bernstein L. Diabetes-induced Proteome Changes Throughout Development. Endocr Metab Immune Disord Drug Targets 2020; 19:732-743. [PMID: 31038056 DOI: 10.2174/1871530319666190305153810] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/31/2018] [Accepted: 11/25/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND Diabetes Mellitus (DM) is a multisystemic disease involving the homeostasis of insulin secretion by the pancreatic islet beta cells (β-cells). It is associated with hypertension, renal disease, and arterial and arteriolar vascular diseases. DISCUSSION The classification of diabetes is identified as type 1 (gene linked β-cell destruction in childhood) and type 2 (late onset associated with β-cell overload and insulin resistance in peripheral tissues. Type 1 diabetes is characterized by insulin deficiency, type 2 diabetes by both insulin deficiency and insulin resistance. The former is a genetically programmed loss of insulin secretion whereas the latter constitutes a disruption of the homeostatic relationship between the opposing activity of β- cell insulin and alpha cell (α-cell) glucagon of the Islets of Langerhans. The condition could also occur in pregnancy, as a prenatal occurring event, possibly triggered by the hormonal changes of pregnancy combined with β-cell overload. This review discusses the molecular basis of the biomolecular changes that occur with respect to glucose homeostasis and related diseases in DM. The underlying link between pancreatic, renal, and microvascular diseases in DM is based on oxidative stress and the Unfolded Protein Response (UPR). CONCLUSION Studying proteome changes in diabetes can deepen our understanding of the biomolecular basis of disease and help us acquire more efficient therapies.
Collapse
Affiliation(s)
- Dina Johar
- Biomedical Science Program, University of Science and Technology, Zewail City of Science and Technology, Giza, Egypt and Biochemistry and Nutrition Department, Ain Shams University Faculty of Women for Arts, Sciences and Education, Heliopolis, Cairo, Egypt
| | - Sara M Ahmed
- Clinical Pathology Department, Faculty of Medicine (Girls), Al-Azhar University, Nasr City, Cairo, Egypt
| | - Samer El Hayek
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nader Al-Dewik
- Qatar Medical Genetic Center, Pediatrics Department, Hamad General Hospital (HGH), Hamad Medical Corporation (HMC), Doha, Qatar
| | - Eshak I Bahbah
- Faculty of Medicine, Al-Azhar University, Damietta, P.C. 34511, Egypt
| | - Nabil H Omar
- Pharmacy Department, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | | | - Doaa O Salman
- Genetics Unit, Histology and Cell biology department, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Asmaa Fahmey
- Faculty of Pharmacy, Al-Mansoura University, Al-Mansoura, Egypt
| | - Mohamed Mottawea
- Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Rasha A M Azouz
- Molecular Biology Department, Genetic Engineering and Biotechnology Research Division, National Research Centre, Dokki, 12622 Giza, Egypt
| | - Larry Bernstein
- Triplex Consulting, 54 Firethorn Lane, Northampton, MA 01060, United States
| |
Collapse
|
35
|
Piganelli JD, Mamula MJ, James EA. The Role of β Cell Stress and Neo-Epitopes in the Immunopathology of Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:624590. [PMID: 33679609 PMCID: PMC7930070 DOI: 10.3389/fendo.2020.624590] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/23/2020] [Indexed: 12/15/2022] Open
Abstract
Due to their secretory function, β cells are predisposed to higher levels of endoplasmic reticulum (ER) stress and greater sensitivity to inflammation than other cell types. These stresses elicit changes in β cells that alter their function and immunogenicity, including defective ribosomal initiation, post-translational modifications (PTMs) of endogenous β cell proteins, and alternative splicing. Multiple published reports confirm the presence of not only CD8+ T cells, but also autoreactive CD4+ T cells within pancreatic islets. Although the specificities of T cells that infiltrate human islets are incompletely characterized, they have been confirmed to include neo-epitopes that are formed through stress-related enzymatic modifications of β cell proteins. This article summarizes emerging knowledge about stress-induced changes in β cells and data supporting a role for neo-antigen formation and cross-talk between immune cells and β cells that provokes autoimmune attack - leading to a breakdown in tissue-specific tolerance in subjects who develop type 1 diabetes.
Collapse
Affiliation(s)
- Jon D. Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, United States
| | - Mark J. Mamula
- Section of Rheumatology, Department of Medicine, Yale School of Medicine, New Haven, CT, United States
| | - Eddie A. James
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
- *Correspondence: Eddie A. James,
| |
Collapse
|
36
|
Zheng Z, Shang Y, Tao J, Zhang J, Sha B. Endoplasmic Reticulum Stress Signaling Pathways: Activation and Diseases. Curr Protein Pept Sci 2019; 20:935-943. [PMID: 31223084 DOI: 10.2174/1389203720666190621103145] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 02/06/2023]
Abstract
Secretory and membrane proteins are folded in the endoplasmic reticulum (ER) prior to their exit. When ER function is disturbed by exogenous and endogenous factors, such as heat shock, ultraviolet radiation, hypoxia, or hypoglycemia, the misfolded proteins may accumulate, promoting ER stress. To rescue this unfavorable situation, the unfolded protein response is activated to reduce misfolded proteins within the ER. Upon ER stress, the ER transmembrane sensor molecules inositol-requiring enzyme 1 (IRE1), RNA-dependent protein kinase (PKR)-like ER kinase (PERK), and activating transcription factor 6, are activated. Here, we discuss the mechanisms of PERK and IRE1 activation and describe two working models for ER stress initiation: the BiP-dependent model and the ligand-driven model. ER stress activation has been linked to multiple diseases, including cancers, Alzheimer's disease, and diabetes. Thus, the regulation of ER stress may provide potential therapeutic targets for these diseases.
Collapse
Affiliation(s)
- Zhi Zheng
- Department of Cell, Developmental and Integrative Biology (CDIB), University of Alabama at Birmingham, Birmingham, AL 35294, United States.,Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xi-Cheng District, Beijing 100050, China
| | - Yuxi Shang
- Department of Hematology, Fuxing Hospital, Eighth Clinical Medical College, Capital Medical University, Beijing 100038, China
| | - Jiahui Tao
- Department of Cell, Developmental and Integrative Biology (CDIB), University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Jun Zhang
- Department of General Surgery, Beijing Friendship Hospital, Capital Medical University, 95 Yong-an Road, Xi-Cheng District, Beijing 100050, China
| | - Bingdong Sha
- Department of Cell, Developmental and Integrative Biology (CDIB), University of Alabama at Birmingham, Birmingham, AL 35294, United States
| |
Collapse
|
37
|
Sankrityayan H, Oza MJ, Kulkarni YA, Mulay SR, Gaikwad AB. ER stress response mediates diabetic microvascular complications. Drug Discov Today 2019; 24:2247-2257. [PMID: 31430543 DOI: 10.1016/j.drudis.2019.08.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 07/19/2019] [Accepted: 08/01/2019] [Indexed: 12/16/2022]
Abstract
Endoplasmic reticulum (ER) homeostasis orchestrates the folding, modification, and trafficking of secretory and membrane proteins to the Golgi compartment, thus governing cellular functions. Alterations in ER homeostasis result in the activation of signaling pathways, such as the unfolded protein response (UPR), to regain ER homeostasis. Nevertheless, failure of UPR leads to activation of autophagy-mediated cell death. Several recent studies emphasized the association of the ER stress (ERS) response with the initiation and progression of diabetes. In this review, we highlight the contribution of the ERS response, such as UPR and autophagy, in the initiation and progression of diabetes and associated microvascular complications, including diabetic nephropathy (DN), retinopathy, and neuropathy, in various experimental models, as well as in humans. We highlight the ERS as a putative therapeutic target for the treatment of diabetic microvascular complications and, thus, the urgent need for the development of improved synthetic and natural inhibitors of ERS.
Collapse
Affiliation(s)
- Himanshu Sankrityayan
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India
| | - Manisha J Oza
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India; SVKM's Dr Bhanuben Nanavati College of Pharmacy, Vile Parle (W), Mumbai 400056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400056, India
| | - Shrikant R Mulay
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow 226031, India
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
38
|
Coudriet GM, Stoops J, Orr AV, Bhushan B, Koral K, Lee S, Previte DM, Dong HH, Michalopoulos GK, Mars WM, Piganelli JD. A Noncanonical Role for Plasminogen Activator Inhibitor Type 1 in Obesity-Induced Diabetes. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:1413-1422. [PMID: 31054988 DOI: 10.1016/j.ajpath.2019.04.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 04/01/2019] [Accepted: 04/02/2019] [Indexed: 01/07/2023]
Abstract
Obesity is a major risk factor for type 2 diabetes because of chronic hepatic inflammation and resultant insulin resistance. Hepatocyte growth factor (HGF) is responsible for resetting hepatic homeostasis after injury following activation by urokinase-type plasminogen activator (u-PA; encoded by the PLAU gene). Plasminogen activator inhibitor type-1 (PAI-1; encoded by the SERPINE1 gene), a u-PA inhibitor and antifibrinolytic agent, is often elevated in obesity and is linked to cardiovascular events. We hypothesized that, in addition to its role in preventing fibrinolysis, elevated PAI-1 inhibits HGF's activation by u-PA and the resultant anti-inflammatory and hepatoprotective properties. Wild-type and PAI-1 knockout (KO) mice on a high-fat diet both became significantly heavier than lean controls; however, the obese KO mice demonstrated improved glucose metabolism compared with wild-type mice. Obese KO mice also exhibited an increase in conversion of latent single-chain HGF to active two-chain HGF, coinciding with an increase in the phosphorylation of the HGF receptor (HGFR or MET, encoded by the MET gene), as well as dampened inflammation. These results strongly suggest that, in addition to its other functions, PAI-mediated inhibition of HGF activation prohibits the resolution of inflammation in the context of obesity-induced type 2 diabetes.
Collapse
Affiliation(s)
- Gina M Coudriet
- Department of Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - John Stoops
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Anne V Orr
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Bharat Bhushan
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Kelly Koral
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Sojin Lee
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Dana M Previte
- Department of Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - H Henry Dong
- Department of Pediatrics, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Wendy M Mars
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| | - Jon D Piganelli
- Department of Surgery, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
39
|
Vig S, Buitinga M, Rondas D, Crèvecoeur I, van Zandvoort M, Waelkens E, Eizirik DL, Gysemans C, Baatsen P, Mathieu C, Overbergh L. Cytokine-induced translocation of GRP78 to the plasma membrane triggers a pro-apoptotic feedback loop in pancreatic beta cells. Cell Death Dis 2019; 10:309. [PMID: 30952835 PMCID: PMC6450900 DOI: 10.1038/s41419-019-1518-0] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 03/04/2019] [Accepted: 03/05/2019] [Indexed: 11/16/2022]
Abstract
The 78-kDa glucose-regulated protein (GRP78) is an ubiquitously expressed endoplasmic reticulum chaperone, with a central role in maintaining protein homeostasis. Recently, an alternative role for GRP78 under stress conditions has been proposed, with stress-induced extracellular secretion and translocation of GRP78 to the cell surface where it acts as a multifunctional signaling receptor. Here we demonstrate translocation of GRP78 to the surface of human EndoC-βH1 cells and primary human islets upon cytokine exposure, in analogy to observations in rodent INS-1E and MIN6 beta cell lines. We show that GRP78 is shuttled via the anterograde secretory pathway, through the Golgi complex and secretory granules, and identify the DNAJ homolog subfamily C member 3 (DNAJC3) as a GRP78-interacting protein that facilitates its membrane translocation. Evaluation of downstream signaling pathways, using N- and C-terminal anti-GRP78 blocking antibodies, demonstrates that both GRP78 signaling domains initiate pro-apoptotic signaling cascades in beta cells. Extracellular GRP78 itself is identified as a ligand for cell surface GRP78 (sGRP78), increasing caspase 3/7 activity and cell death upon binding, which is accompanied by enhanced Chop and Bax mRNA expression. These results suggest that inflammatory cytokines induce a self-destructive pro-apoptotic feedback loop through the secretion and membrane translocation of GRP78. This proapoptotic function distinguishes the role of sGRP78 in beta cells from its reported anti-apoptotic and proliferative role in cancer cells, opening the road for the use of compounds that block sGRP78 as potential beta cell-preserving therapies in type 1 diabetes.
Collapse
Affiliation(s)
- Saurabh Vig
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Mijke Buitinga
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Dieter Rondas
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Inne Crèvecoeur
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Marc van Zandvoort
- Department of Molecular Cell Biology and School for Nutrition and Translational Research in Metabolism NUTRIM, Maastricht University, Maastricht, The Netherlands
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven, Belgium.,SyBioMa, KU Leuven, Leuven, Belgium
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Universite Libre de Bruxelles, Brussels, Belgium
| | - Conny Gysemans
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Pieter Baatsen
- Electron Microscopy Platform of VIB Bio Imaging Core at KU Leuven and VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium
| | - Chantal Mathieu
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Lut Overbergh
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
40
|
Becker MW, Simonovich JA, Phelps EA. Engineered microenvironments and microdevices for modeling the pathophysiology of type 1 diabetes. Biomaterials 2019; 198:49-62. [DOI: 10.1016/j.biomaterials.2018.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/21/2018] [Accepted: 07/01/2018] [Indexed: 01/09/2023]
|
41
|
Aivazidis S, Anderson CC, Roede JR. Toxicant-mediated redox control of proteostasis in neurodegeneration. CURRENT OPINION IN TOXICOLOGY 2019; 13:22-34. [PMID: 31602419 PMCID: PMC6785977 DOI: 10.1016/j.cotox.2018.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Disruption in redox signaling and control of cellular processes has emerged as a key player in many pathologies including neurodegeneration. As protein aggregations are a common hallmark of several neuronal pathologies, a firm understanding of the interplay between redox signaling, oxidative and free radical stress, and proteinopathies is required to sort out the complex mechanisms in these diseases. Fortunately, models of toxicant-induced neurodegeneration can be utilized to evaluate and report mechanistic alterations in the proteostasis network (PN). The epidemiological links between environmental toxicants and neurological disease gives further credence into characterizing the toxicant-mediated PN disruptions observed in these conditions. Reviewed here are examples of mechanistic interaction between oxidative or free radical stress and PN alterations. Additionally, investigations into toxicant-mediated PN disruptions, specifically focusing on environmental metals and pesticides, are discussed. Finally, we emphasize the need to distinguish whether the presence of protein aggregations are contributory to phenotypes related to neurodegeneration, or if they are a byproduct of PN deficiencies.
Collapse
Affiliation(s)
- Stefanos Aivazidis
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Colin C Anderson
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| |
Collapse
|
42
|
Harnessing CXCL12 signaling to protect and preserve functional β-cell mass and for cell replacement in type 1 diabetes. Pharmacol Ther 2019; 193:63-74. [DOI: 10.1016/j.pharmthera.2018.08.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
43
|
Burg AR, Tse HM. Redox-Sensitive Innate Immune Pathways During Macrophage Activation in Type 1 Diabetes. Antioxid Redox Signal 2018; 29:1373-1398. [PMID: 29037052 PMCID: PMC6166692 DOI: 10.1089/ars.2017.7243] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
SIGNIFICANCE Type 1 diabetes (T1D) is an autoimmune disease resulting in β-cell destruction mediated by islet-infiltrating leukocytes. The role of oxidative stress in human and murine models of T1D is highly significant as these noxious molecules contribute to diabetic complications and β-cell lysis, but their direct impact on dysregulated autoimmune responses is highly understudied. Pro-inflammatory macrophages play a vital role in the initiation and effector phases of T1D by producing free radicals and pro-inflammatory cytokines to facilitate β-cell destruction and to present antigen to autoreactive T cells. Recent Advances: Redox modulation of macrophage functions may play critical roles in autoimmunity. These include enhancing pro-inflammatory innate immune signaling pathways in response to environmental triggers, enforcing an M1 macrophage differentiation program, controlling antigen processing, and altering peptide recognition by oxidative post-translational modification. Therefore, an oxidative environment may act on multiple macrophage functions to orchestrate T1D pathogenesis. CRITICAL ISSUES Mechanisms involved in the initiation of T1D remain unclear, making preventive and early therapeutics difficult to develop. Although many of these advances in the redox regulation of macrophages are in their infancy, they provide insight into how oxidative stress aids in the precipitating event of autoimmune activation. FUTURE DIRECTIONS Future studies should be aimed at mechanistically determining which redox-regulated macrophage functions are pertinent in T1D pathogenesis, as well as at investigating potential targetable therapeutics to halt and/or dampen innate immune activation in T1D.
Collapse
Affiliation(s)
- Ashley R Burg
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham , Birmingham, Alabama
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
44
|
Previte DM, Piganelli JD. Reactive Oxygen Species and Their Implications on CD4 + T Cells in Type 1 Diabetes. Antioxid Redox Signal 2018; 29:1399-1414. [PMID: 28990401 DOI: 10.1089/ars.2017.7357] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Previous work has indicated that type 1 diabetes (T1D) pathology is highly driven by reactive oxygen species (ROS). One way in which ROS shape the autoimmune response demonstrated in T1D is by promoting CD4+ T cell activation and differentiation. As CD4+ T cells are a significant contributor to pancreatic β cell destruction in T1D, understanding how ROS impact their development, activation, and differentiation is critical. Recent Advances: CD4+ T cells themselves generate ROS via nicotinamide adenine dinucleotide phosphate (NADPH) oxidase expression and electron transport chain activity. Moreover, T cells can also be exposed to exogenous ROS generated by other immune cells (e.g., macrophages and dendritic cells) and β cells. Genetically modified animals and ROS inhibitors have demonstrated that ROS blockade during activation results in CD4+ T cell hyporesponsiveness and reduced diabetes incidence. Critical Issues and Future Directions: Although the majority of studies with regard to T1D and CD4+ T cells have been done to examine the influence of redox on CD4+ T cell activation, this is not the only circumstance in which a T cell can be impacted by redox. ROS and redox have also been shown to play roles in CD4+ T cell-related tolerogenic mechanisms, including thymic selection and regulatory T cell-mediated suppression. However, the effect of these mechanisms with respect to T1D pathogenesis remains elusive. Therefore, pursuing these avenues may provide valuable insight into the global role of ROS and redox in autoreactive CD4+ T cell formation and function.
Collapse
Affiliation(s)
- Dana M Previte
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| | - Jon D Piganelli
- Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center , Pittsburgh, Pennsylvania
| |
Collapse
|
45
|
Yang ML, Doyle HA, Clarke SG, Herold KC, Mamula MJ. Oxidative Modifications in Tissue Pathology and Autoimmune Disease. Antioxid Redox Signal 2018; 29:1415-1431. [PMID: 29088923 PMCID: PMC6166690 DOI: 10.1089/ars.2017.7382] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
SIGNIFICANCE Various autoimmune syndromes are characterized by abnormalities found at the level of tissues and cells, as well as by microenvironmental influences, such as reactive oxygen species (ROS), that alter intracellular metabolism and protein expression. Moreover, the convergence of genetic, epigenetic, and even environmental influences can result in B and T lymphocyte autoimmunity and tissue pathology. Recent Advances: This review describes how oxidative stress to cells and tissues may alter post-translational protein modifications, both directly and indirectly, as well as potentially lead to aberrant gene expression. For example, it has been clearly observed in many systems how oxidative stress directly amplifies carbonyl protein modifications. However, ROS also lead to a number of nonenzymatic spontaneous modifications including deamidation and isoaspartate modification as well as to enzyme-mediated citrullination of self-proteins. ROS have direct effects on DNA methylation, leading to influences in gene expression, chromosome inactivation, and the silencing of genetic elements. Finally, ROS can alter many other cellular pathways, including the initiation of apoptosis and NETosis, triggering the release of modified intracellular autoantigens. CRITICAL ISSUES This review will detail specific post-translational protein modifications, the pathways that control autoimmunity to modified self-proteins, and how products of ROS may be important biomarkers of tissue pathogenesis. FUTURE DIRECTIONS A clear understanding of the many pathways affected by ROS will lead to potential therapeutic manipulations to alter the onset and/or progression of autoimmune disease.
Collapse
Affiliation(s)
- Mei-Ling Yang
- 1 Section of Rheumatology, Yale University School of Medicine , New Haven, Connecticut.,2 Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Hester A Doyle
- 1 Section of Rheumatology, Yale University School of Medicine , New Haven, Connecticut.,2 Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| | - Steven G Clarke
- 3 Department of Chemistry and Biochemistry, University of California , Los Angeles, Los Angeles, California
| | - Kevan C Herold
- 2 Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut.,4 Department of Immunobiology, Yale University School of Medicine , New Haven, Connecticut
| | - Mark J Mamula
- 1 Section of Rheumatology, Yale University School of Medicine , New Haven, Connecticut.,2 Department of Internal Medicine, Yale University School of Medicine , New Haven, Connecticut
| |
Collapse
|
46
|
Buitinga M, Callebaut A, Marques Câmara Sodré F, Crèvecoeur I, Blahnik-Fagan G, Yang ML, Bugliani M, Arribas-Layton D, Marré M, Cook DP, Waelkens E, Mallone R, Piganelli JD, Marchetti P, Mamula MJ, Derua R, James EA, Mathieu C, Overbergh L. Inflammation-Induced Citrullinated Glucose-Regulated Protein 78 Elicits Immune Responses in Human Type 1 Diabetes. Diabetes 2018; 67:2337-2348. [PMID: 30348823 PMCID: PMC6973547 DOI: 10.2337/db18-0295] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/31/2018] [Indexed: 12/16/2022]
Abstract
The β-cell has become recognized as a central player in the pathogenesis of type 1 diabetes with the generation of neoantigens as potential triggers for breaking immune tolerance. We report that posttranslationally modified glucose-regulated protein 78 (GRP78) is a novel autoantigen in human type 1 diabetes. When human islets were exposed to inflammatory stress induced by interleukin-1β, tumor necrosis factor-α, and interferon-γ, arginine residue R510 within GRP78 was converted into citrulline, as evidenced by liquid chromatography-tandem mass spectrometry. This conversion, known as citrullination, led to the generation of neoepitopes, which effectively could be presented by HLA-DRB1*04:01 molecules. With the use of HLA-DRB1*04:01 tetramers and ELISA techniques, we demonstrate enhanced antigenicity of citrullinated GRP78 with significantly increased CD4+ T-cell responses and autoantibody titers in patients with type 1 diabetes compared with healthy control subjects. Of note, patients with type 1 diabetes had a predominantly higher percentage of central memory cells and a lower percentage of effector memory cells directed against citrullinated GRP78 compared with the native epitope. These results strongly suggest that citrullination of β-cell proteins, exemplified here by the citrullination of GRP78, contributes to loss of self-tolerance toward β-cells in human type 1 diabetes, indicating that β-cells actively participate in their own demise.
Collapse
Affiliation(s)
- Mijke Buitinga
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Aïsha Callebaut
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | | | - Inne Crèvecoeur
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | | | | | | | | | - Meghan Marré
- Division of Pediatric Surgery, University of Pittsburgh, Pittsburgh, PA
| | - Dana P Cook
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Etienne Waelkens
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven, Belgium
- SyBioMa, KU Leuven, Leuven, Belgium
| | - Roberto Mallone
- INSERM, U1016, CNRS, UMR8104, Paris Descartes University, Sorbonne Paris Cité, Cochin Institute, Paris, France
| | - Jon D Piganelli
- Division of Pediatric Surgery, University of Pittsburgh, Pittsburgh, PA
| | | | | | - Rita Derua
- Laboratory of Protein Phosphorylation and Proteomics, KU Leuven, Leuven, Belgium
- SyBioMa, KU Leuven, Leuven, Belgium
| | | | - Chantal Mathieu
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Lut Overbergh
- Laboratory for Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|
47
|
Marré ML, Piganelli JD, James EA. Protecting functional β cells with a therapeutic peptide. ANNALS OF TRANSLATIONAL MEDICINE 2018; 6:372. [PMID: 30370299 DOI: 10.21037/atm.2018.07.26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Meghan L Marré
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jon D Piganelli
- Division of Pediatric Surgery, Department of Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eddie A James
- Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| |
Collapse
|
48
|
Hu L, Dai SC, Luan X, Chen J, Cannavicci A. Dysfunction and Therapeutic Potential of Endothelial Progenitor Cells in Diabetes Mellitus. J Clin Med Res 2018; 10:752-757. [PMID: 30214646 PMCID: PMC6134996 DOI: 10.14740/jocmr3581w] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 08/16/2018] [Indexed: 12/25/2022] Open
Abstract
Diabetes mellitus (DM) is a chronic, multifactorial metabolic disease whereby insulin deficiency or resistance results in hyperglycemia. Endothelial cells (ECs) form the innermost layer of the blood vessel and produce and release a variety of vasoactive substances and growth factors to regulate vascular homeostasis and angiogenesis. Hyperglycemia and insulin resistance can cause endothelial dysfunction, leading to vascular complications such as coronary artery disease, peripheral arterial disease, diabetic nephropathy, neuropathy and retinopathy. The detrimental effect exerted on ECs by hyperglycemia and insulin resistance underlines the importance of reparatory mechanisms in DM. Endothelial progenitor cells (EPCs), derived from bone marrow, have been recognized as endogenous cells involved in endothelial repair and new blood vessel formation. Initially isolated from a subset of circulating CD34+ mononuclear cells, EPCs were found to possess the ability to differentiate into ECs when cultured in vitro and incorporate into newly formed vessels upon transplantation in animal models of ischemia. Due to the low frequency of CD34+ cells in circulation, the vast majority of studies investigating EPC actions have used cells that are generated through the culture of peripheral blood mononuclear cells (PBMNCs) for 4 - 7 days in endothelial selective medium. These cells, mainly of myeloid hematopoietic cell origin, were termed “Early EPCs,” of which, few expressed stem/progenitor-cell markers. Therefore, early EPCs were also termed “myeloid angiogenic cells” (MACs). When PBMNCs are cultured for over 2 weeks, early EPCs gradually diminish while so-called late EPCs appear. Late EPCs share phenotypic features with mature ECs and are therefore also termed blood-derived ECs; they will not be addressed in this review. MAC dysfunction has been observed in a variety of disease conditions including DM. In this article we review the activities and therapeutic potential of MACs in DM. We will interchangeably use “EPCs” and “MACs” to refer to the cells procured by culture of PBMNCs in EC selective medium for approximately 7 days.
Collapse
Affiliation(s)
- Lidong Hu
- Department of Endocrinology, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Si-Cheng Dai
- Medical Sciences Program, Western University, London, ON, Canada.,Keenan Research Center for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| | - Xiaojun Luan
- Department of Endocrinology, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Jingsong Chen
- Department of Endocrinology, The First People's Hospital of Foshan, Foshan 528000, Guangdong, China
| | - Anthony Cannavicci
- Keenan Research Center for Biomedical Science, St. Michael's Hospital, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
49
|
Thomaidou S, Zaldumbide A, Roep BO. Islet stress, degradation and autoimmunity. Diabetes Obes Metab 2018; 20 Suppl 2:88-94. [PMID: 30230178 PMCID: PMC6174957 DOI: 10.1111/dom.13387] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 05/18/2018] [Accepted: 05/30/2018] [Indexed: 12/15/2022]
Abstract
β-cell destruction in type 1 diabetes (T1D) results from the effect of inflammation and autoimmunity. In response to inflammatory signals, islet cells engage adaptive mechanisms to restore and maintain cellular homeostasis. Among these mechanisms, the unfolded protein response (UPR) leads to a reduction of the general protein translation rate, increased production of endoplasmic reticulum chaperones and the initiation of degradation by activation of the ER associated degradation pathway (ERAD) in which newly synthetized proteins are ubiquitinylated and processed through the proteasome. This adaptive phase is also believed to play a critical role in the development of autoimmunity by the generation of neoantigens. While we have previously investigated the effect of stress on transcription, translation and post-translational events as possible source for neoantigens, the participation of the degradation machinery, yet crucial in the generation of antigenic peptides, remains to be investigated in the context of T1D pathology. In this review, we will describe the relation between the unfolded protein response and the Ubiquitin Proteasome System (UPS) and address the role of the cellular degradation machinery in the generation of antigens. Learning from tumour immunology, we propose how these processes may unmask β-cells by triggering the generation of aberrant peptides recognized by the immune cells.
Collapse
Affiliation(s)
- Sofia Thomaidou
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Arnaud Zaldumbide
- Department of Cell and Chemical BiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Bart O. Roep
- Department of Immunohematology and Blood bank Leiden University Medical CenterLeiden University Medical CenterLeidenThe Netherlands
- Department of Diabetes ImmunologyDiabetes & Metabolism Research Institute at the Beckman Research InstituteDuarteCalifornia
| |
Collapse
|
50
|
Creusot RJ, Postigo-Fernandez J, Teteloshvili N. Altered Function of Antigen-Presenting Cells in Type 1 Diabetes: A Challenge for Antigen-Specific Immunotherapy? Diabetes 2018; 67:1481-1494. [PMID: 30030289 PMCID: PMC6054431 DOI: 10.2337/db17-1564] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 05/03/2018] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) arises from a failure to maintain tolerance to specific β-cell antigens. Antigen-specific immunotherapy (ASIT) aims to reestablish immune tolerance through the supply of pertinent antigens to specific cell types or environments that are suitable for eliciting tolerogenic responses. However, antigen-presenting cells (APCs) in T1D patients and in animal models of T1D are affected by a number of alterations, some due to genetic polymorphism. Combination of these alterations, impacting the number, phenotype, and function of APC subsets, may account for both the underlying tolerance deficiency and for the limited efficacy of ASITs so far. In this comprehensive review, we examine different aspects of APC function that are pertinent to tolerance induction and summarize how they are altered in the context of T1D. We attempt to reconcile 25 years of studies on this topic, highlighting genetic, phenotypic, and functional features that are common or distinct between humans and animal models. Finally, we discuss the implications of these defects and the challenges they might pose for the use of ASITs to treat T1D. Better understanding of these APC alterations will help us design more efficient ways to induce tolerance.
Collapse
Affiliation(s)
- Rémi J Creusot
- Columbia Center for Translational Immunology, Naomi Berrie Diabetes Center and Department of Medicine, Columbia University Medical Center, New York, NY
| | - Jorge Postigo-Fernandez
- Columbia Center for Translational Immunology, Naomi Berrie Diabetes Center and Department of Medicine, Columbia University Medical Center, New York, NY
| | - Nato Teteloshvili
- Columbia Center for Translational Immunology, Naomi Berrie Diabetes Center and Department of Medicine, Columbia University Medical Center, New York, NY
| |
Collapse
|