1
|
Walker SL, Leete P, Boldison J. Tissue Resident and Infiltrating Immune Cells: Their Influence on the Demise of Beta Cells in Type 1 Diabetes. Biomolecules 2025; 15:441. [PMID: 40149976 PMCID: PMC11939886 DOI: 10.3390/biom15030441] [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: 02/10/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease that results in the selective loss of pancreatic beta cells and an eventual deficit in insulin production to maintain glucose homeostasis. It is now increasingly accepted that this dynamic disease process is multifactorial; involves a variety of immune cells which contribute to an inflamed pancreatic microenvironment; and that the condition is heterogenous, resulting in variable rates of subsequent beta cell damage. In this review, we will explore the current understanding of the cellular interactions between both resident and infiltrating immune cells within the pancreatic environment, highlighting key mechanisms which may promote the beta cell destruction and islet damage associated with T1D.
Collapse
Affiliation(s)
| | | | - Joanne Boldison
- Department of Clinical and Biomedical Sciences, University of Exeter, RILD Building (Level 4), Barrack Road, Exeter EX2 5DW, UK; (S.L.W.); (P.L.)
| |
Collapse
|
2
|
Aizenshtadt A, Wang C, Abadpour S, Menezes PD, Wilhelmsen I, Dalmao‐Fernandez A, Stokowiec J, Golovin A, Johnsen M, Combriat TMD, Røberg‐Larsen H, Gadegaard N, Scholz H, Busek M, Krauss SJK. Pump-Less, Recirculating Organ-on-Chip (rOoC) Platform to Model the Metabolic Crosstalk between Islets and Liver. Adv Healthc Mater 2024; 13:e2303785. [PMID: 38221504 PMCID: PMC11468483 DOI: 10.1002/adhm.202303785] [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: 10/31/2023] [Revised: 12/05/2023] [Indexed: 01/16/2024]
Abstract
Type 2 diabetes mellitus (T2DM), obesity, and metabolic dysfunction-associated steatotic liver disease (MASLD) are epidemiologically correlated disorders with a worldwide growing prevalence. While the mechanisms leading to the onset and development of these conditions are not fully understood, predictive tissue representations for studying the coordinated interactions between central organs that regulate energy metabolism, particularly the liver and pancreatic islets, are needed. Here, a dual pump-less recirculating organ-on-chip platform that combines human pluripotent stem cell (sc)-derived sc-liver and sc-islet organoids is presented. The platform reproduces key aspects of the metabolic cross-talk between both organs, including glucose levels and selected hormones, and supports the viability and functionality of both sc-islet and sc-liver organoids while preserving a reduced release of pro-inflammatory cytokines. In a model of metabolic disruption in response to treatment with high lipids and fructose, sc-liver organoids exhibit hallmarks of steatosis and insulin resistance, while sc-islets produce pro-inflammatory cytokines on-chip. Finally, the platform reproduces known effects of anti-diabetic drugs on-chip. Taken together, the platform provides a basis for functional studies of obesity, T2DM, and MASLD on-chip, as well as for testing potential therapeutic interventions.
Collapse
Affiliation(s)
- Aleksandra Aizenshtadt
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Chencheng Wang
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Shadab Abadpour
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
- Institute for Surgical ResearchOslo University HospitalOsloNorway
| | - Pedro Duarte Menezes
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Ingrid Wilhelmsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Andrea Dalmao‐Fernandez
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Department of PharmacyFaculty of Mathematics and Natural SciencesUniversity of OsloP.O. Box 1083Oslo0316Norway
| | - Justyna Stokowiec
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Alexey Golovin
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mads Johnsen
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Thomas M. D. Combriat
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
| | - Hanne Røberg‐Larsen
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Section for Chemical Life SciencesDepartment of ChemistryUniversity of OsloP.O. Box 1033Oslo0315Norway
| | - Nikolaj Gadegaard
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- James Watt School of EngineeringUniversity of GlasgowRankine BuildingGlasgowG12 8LTUK
| | - Hanne Scholz
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Transplantation MedicineExperimental Cell Transplantation Research GroupOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Mathias Busek
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| | - Stefan J. K. Krauss
- Hybrid Technology Hub Centre of ExcellenceInstitute of Basic Medical ScienceUniversity of OsloP.O. Box 1110Oslo0317Norway
- Dep. of Immunology and Transfusion MedicineOslo University HospitalP.O. Box 4950Oslo0424Norway
| |
Collapse
|
3
|
Kostopoulou E, Kalavrizioti D, Davoulou P, Papachristou E, Sinopidis X, Fouzas S, Dassios T, Gkentzi D, Kyriakou SI, Karatza A, Dimitriou G, Goumenos D, Spiliotis BE, Plotas P, Papasotiriou M. Monocyte Chemoattractant Protein-1 (MCP-1), Activin-A and Clusterin in Children and Adolescents with Obesity or Type-1 Diabetes Mellitus. Diagnostics (Basel) 2024; 14:450. [PMID: 38396489 PMCID: PMC10887959 DOI: 10.3390/diagnostics14040450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/04/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Inflammation plays a crucial role in diabetes and obesity through macrophage activation. Macrophage chemoattractant protein-1 (MCP-1), activin-A, and clusterin are chemokines with known roles in diabetes and obesity. The aim of this study is to investigate their possible diagnostic and/or early prognostic values in children and adolescents with obesity and type-1 diabetes mellitus (T1DM). METHODS We obtained serum samples from children and adolescents with a history of T1DM or obesity, in order to measure and compare MCP-1, activin-A, and clusterin concentrations. RESULTS Forty-three subjects were included in each of the three groups (controls, T1DM, and obesity). MCP-1 values were positively correlated to BMI z-score. Activin-A was increased in children with obesity compared to the control group. A trend for higher values was detected in children with T1DM. MCP-1 and activin-A levels were positively correlated. Clusterin levels showed a trend towards lower values in children with T1DM or obesity compared to the control group and were negatively correlated to renal function. CONCLUSIONS The inflammation markers MCP-1, activin-A, and clusterin are not altered in children with T1DM. Conversely, obesity in children is positively correlated to serum MCP-1 values and characterized by higher activin-A levels, which may reflect an already established systematic inflammation with obesity since childhood.
Collapse
Affiliation(s)
- Eirini Kostopoulou
- Division of Pediatric Endocrinology, Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (E.K.); (B.E.S.)
| | - Dimitra Kalavrizioti
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Panagiota Davoulou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Evangelos Papachristou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Xenophon Sinopidis
- Department of Pediatric Surgery, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Sotirios Fouzas
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Theodore Dassios
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Despoina Gkentzi
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Stavroula Ioanna Kyriakou
- Department of Pediatric Surgery, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece;
| | - Ageliki Karatza
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Gabriel Dimitriou
- Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (S.F.); (T.D.); (D.G.); (A.K.); (G.D.)
| | - Dimitrios Goumenos
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| | - Bessie E. Spiliotis
- Division of Pediatric Endocrinology, Department of Pediatrics, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (E.K.); (B.E.S.)
| | - Panagiotis Plotas
- Department of Speech and Language Therapy, School of Health Rehabilitation Sciences, University of Patras, 26504 Patras, Greece;
| | - Marios Papasotiriou
- Department of Nephrology and Kidney Transplantation, University Hospital of Patras, School of Medicine, University of Patras, 26504 Patras, Greece; (D.K.); (P.D.); (E.P.); (D.G.); (M.P.)
| |
Collapse
|
4
|
Riaz F, Wei P, Pan F. PPARs at the crossroads of T cell differentiation and type 1 diabetes. Front Immunol 2023; 14:1292238. [PMID: 37928539 PMCID: PMC10623333 DOI: 10.3389/fimmu.2023.1292238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
T-cell-mediated autoimmune type 1 diabetes (T1D) is characterized by the immune-mediated destruction of pancreatic beta cells (β-cells). The increasing prevalence of T1D poses significant challenges to the healthcare system, particularly in countries with struggling economies. This review paper highlights the multifaceted roles of Peroxisome Proliferator-Activated Receptors (PPARs) in the context of T1D, shedding light on their potential as regulators of immune responses and β-cell biology. Recent research has elucidated the intricate interplay between CD4+ T cell subsets, such as Tregs and Th17, in developing autoimmune diseases like T1D. Th17 cells drive inflammation, while Tregs exert immunosuppressive functions, highlighting the delicate balance crucial for immune homeostasis. Immunotherapy has shown promise in reinstating self-tolerance and restricting the destruction of autoimmune responses, but further investigations are required to refine these therapeutic strategies. Intriguingly, PPARs, initially recognized for their role in lipid metabolism, have emerged as potent modulators of inflammation in autoimmune diseases, particularly in T1D. Although evidence suggests that PPARs affect the β-cell function, their influence on T-cell responses and their potential impact on T1D remains largely unexplored. It was noted that PPARα is involved in restricting the transcription of IL17A and enhancing the expression of Foxp3 by minimizing its proteasomal degradation. Thus, antagonizing PPARs may exert beneficial effects in regulating the differentiation of CD4+ T cells and preventing T1D. Therefore, this review advocates for comprehensive investigations to delineate the precise roles of PPARs in T1D pathogenesis, offering innovative therapeutic avenues that target both the immune system and pancreatic function. This review paper seeks to bridge the knowledge gap between PPARs, immune responses, and T1D, providing insights that may revolutionize the treatment landscape for this autoimmune disorder. Moreover, further studies involving PPAR agonists in non-obese diabetic (NOD) mice hold promise for developing novel T1D therapies.
Collapse
Affiliation(s)
- Farooq Riaz
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Ping Wei
- Department of Otolaryngology, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
5
|
Camaya I, O’Brien B, Donnelly S. How do parasitic worms prevent diabetes? An exploration of their influence on macrophage and β-cell crosstalk. Front Endocrinol (Lausanne) 2023; 14:1205219. [PMID: 37564976 PMCID: PMC10411736 DOI: 10.3389/fendo.2023.1205219] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Diabetes is the fastest growing chronic disease globally, with prevalence increasing at a faster rate than heart disease and cancer. While the disease presents clinically as chronic hyperglycaemia, two distinct subtypes have been recognised. Type 1 diabetes (T1D) is characterised as an autoimmune disease in which the insulin-producing pancreatic β-cells are destroyed, and type 2 diabetes (T2D) arises due to metabolic insufficiency, in which inadequate amounts of insulin are produced, and/or the actions of insulin are diminished. It is now apparent that pro-inflammatory responses cause a loss of functional β-cell mass, and this is the common underlying mechanism of both T1D and T2D. Macrophages are the central immune cells in the pathogenesis of both diseases and play a major role in the initiation and perpetuation of the proinflammatory responses that compromise β-cell function. Furthermore, it is the crosstalk between macrophages and β-cells that orchestrates the inflammatory response and ensuing β-cell dysfunction/destruction. Conversely, this crosstalk can induce immune tolerance and preservation of β-cell mass and function. Thus, specifically targeting the intercellular communication between macrophages and β-cells offers a unique strategy to prevent/halt the islet inflammatory events underpinning T1D and T2D. Due to their potent ability to regulate mammalian immune responses, parasitic worms (helminths), and their excretory/secretory products, have been examined for their potential as therapeutic agents for both T1D and T2D. This research has yielded positive results in disease prevention, both clinically and in animal models. However, the focus of research has been on the modulation of immune cells and their effectors. This approach has ignored the direct effects of helminths and their products on β-cells, and the modulation of signal exchange between macrophages and β-cells. This review explores how the alterations to macrophages induced by helminths, and their products, influence the crosstalk with β-cells to promote their function and survival. In addition, the evidence that parasite-derived products interact directly with endocrine cells to influence their communication with macrophages to prevent β-cell death and enhance function is discussed. This new paradigm of two-way metabolic conversations between endocrine cells and macrophages opens new avenues for the treatment of immune-mediated metabolic disease.
Collapse
Affiliation(s)
| | | | - Sheila Donnelly
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| |
Collapse
|
6
|
Gurgul-Convey E. To Be or Not to Be: The Divergent Action and Metabolism of Sphingosine-1 Phosphate in Pancreatic Beta-Cells in Response to Cytokines and Fatty Acids. Int J Mol Sci 2022; 23:ijms23031638. [PMID: 35163559 PMCID: PMC8835924 DOI: 10.3390/ijms23031638] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/24/2022] [Accepted: 01/27/2022] [Indexed: 01/02/2023] Open
Abstract
Sphingosine-1 phosphate (S1P) is a bioactive sphingolipid with multiple functions conveyed by the activation of cell surface receptors and/or intracellular mediators. A growing body of evidence indicates its important role in pancreatic insulin-secreting beta-cells that are necessary for maintenance of glucose homeostasis. The dysfunction and/or death of beta-cells lead to diabetes development. Diabetes is a serious public health burden with incidence growing rapidly in recent decades. The two major types of diabetes are the autoimmune-mediated type 1 diabetes (T1DM) and the metabolic stress-related type 2 diabetes (T2DM). Despite many differences in the development, both types of diabetes are characterized by chronic hyperglycemia and inflammation. The inflammatory component of diabetes remains under-characterized. Recent years have brought new insights into the possible mechanism involved in the increased inflammatory response, suggesting that environmental factors such as a westernized diet may participate in this process. Dietary lipids, particularly palmitate, are substrates for the biosynthesis of bioactive sphingolipids. Disturbed serum sphingolipid profiles were observed in both T1DM and T2DM patients. Many polymorphisms were identified in genes encoding enzymes of the sphingolipid pathway, including sphingosine kinase 2 (SK2), the S1P generating enzyme which is highly expressed in beta-cells. Proinflammatory cytokines and free fatty acids have been shown to modulate the expression and activity of S1P-generating and S1P-catabolizing enzymes. In this review, the similarities and differences in the action of extracellular and intracellular S1P in beta-cells exposed to cytokines or free fatty acids will be identified and the outlook for future research will be discussed.
Collapse
Affiliation(s)
- Ewa Gurgul-Convey
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| |
Collapse
|
7
|
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
|
8
|
Ooms M, Strom A, Strassburger K, Menart B, Leslie RD, Schloot NC. Increased spontaneous CCL2 (MCP-1) and CCL5 (RANTES) secretion in vitro in LADA compared to type 1 diabetes and type 2 diabetes: Action LADA 14. Diabetes Metab Res Rev 2021; 37:e3431. [PMID: 33369072 DOI: 10.1002/dmrr.3431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/22/2020] [Accepted: 12/14/2020] [Indexed: 11/11/2022]
Abstract
AIMS Immune-mediated type 1 diabetes (T1D) in adulthood and latent autoimmune diabetes in adults (LADA) share similar pathological mechanisms but differ clinically in disease progression. The aim of this study was to acquire insights into spontaneous and stimulated chemokine secretion of immune cells in different diabetes types. MATERIALS AND METHODS We investigated in vitro spontaneous, mitogen (PI) and antigen (HSP60, p277, pGAD, pIA2) stimulated chemokine secretion of leucocytes from patients with T1D (n = 32), LADA (n = 22), type 2 diabetes (T2D; n = 49), and glucose-tolerant individuals (n = 13). Chemokine concentration in supernatants was measured for CCL2 (MCP-1), CXCL10 (IP10) and CCL5 (RANTES) using a multiplex bead array assay. RESULTS Spontaneous secretion of CCL2 and CCL5 were higher in LADA compared to T1D and T2D (all p < 0.05) while CXCL10 was similar in the groups. Mitogen-stimulated secretion of CCL2 in LADA was lower compared to T1D and T2D (all p < 0.05) while CXCL10 and CCL5 were similar in all groups. Upon stimulation with pIA2 the secretion of CCL2 in LADA was lower compared to T2D (p < 0.05). Spontaneous CXCL10 secretion in LADA was positively associated with body mass index (r2 = 0.35; p = 0.0035) and C-peptide (r2 = 0.30; p = 0.009). CONCLUSIONS Chemokine secretion is altered between different diabetes types. Increased spontaneous secretion of CCL2 and CCL5 and decreased secretion of CCL2, upon stimulation with PI and pIA2, in LADA compared to T1D and T2D could reflect altered immune responsiveness in LADA patients in association with their slower clinical progression compared to insulin dependence.
Collapse
Affiliation(s)
- Mark Ooms
- Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Duesseldorf, Germany
- Clinic and Policlinic for Oral and Maxillofacial Surgery, University Hospital Aachen, Aachen, Germany
| | - Alexander Strom
- Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Duesseldorf, Germany
| | - Klaus Strassburger
- Institute for Biometrics and Epidemiology, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Duesseldorf, Germany
| | - Barbara Menart
- Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Duesseldorf, Germany
| | - Richard D Leslie
- Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Nanette C Schloot
- Institute for Clinical Diabetology, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Duesseldorf, Germany
| |
Collapse
|
9
|
Cosentino C, Regazzi R. Crosstalk between Macrophages and Pancreatic β-Cells in Islet Development, Homeostasis and Disease. Int J Mol Sci 2021; 22:ijms22041765. [PMID: 33578952 PMCID: PMC7916718 DOI: 10.3390/ijms22041765] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/04/2021] [Accepted: 02/06/2021] [Indexed: 12/29/2022] Open
Abstract
Macrophages are highly heterogeneous and plastic immune cells with peculiar characteristics dependent on their origin and microenvironment. Following pathogen infection or damage, circulating monocytes can be recruited in different tissues where they differentiate into macrophages. Stimuli present in the surrounding milieu induce the polarisation of macrophages towards a pro-inflammatory or anti-inflammatory profile, mediating inflammatory or homeostatic responses, respectively. However, macrophages can also derive from embryonic hematopoietic precursors and reside in specific tissues, actively participating in the development and the homeostasis in physiological conditions. Pancreatic islet resident macrophages are present from the prenatal stages onwards and show specific surface markers and functions. They localise in close proximity to β-cells, being exquisite sensors of their secretory ability and viability. Over the years, the crucial role of macrophages in β-cell differentiation and homeostasis has been highlighted. In addition, macrophages are emerging as central players in the initiation of autoimmune insulitis in type 1 diabetes and in the low-grade chronic inflammation characteristic of obesity and type 2 diabetes pathogenesis. The present work reviews the current knowledge in the field, with a particular focus on the mechanisms of communication between β-cells and macrophages that have been described so far.
Collapse
Affiliation(s)
- Cristina Cosentino
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland;
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, CH-1005 Lausanne, Switzerland;
- Department of Biomedical Sciences, University of Lausanne, Rue du Bugnon 7, CH-1005 Lausanne, Switzerland
- Correspondence: ; Tel.: +41-21-692-52-80; Fax: +41-21-692-52-55
| |
Collapse
|
10
|
Li S, Vaziri ND, Swentek L, Takasu C, Vo K, Stamos MJ, Ricordi C, Ichii H. Prevention of Autoimmune Diabetes in NOD Mice by Dimethyl Fumarate. Antioxidants (Basel) 2021; 10:antiox10020193. [PMID: 33572792 PMCID: PMC7912218 DOI: 10.3390/antiox10020193] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 01/25/2021] [Accepted: 01/26/2021] [Indexed: 12/20/2022] Open
Abstract
Oxidative stress plays critical roles in the pathogenesis of diabetes. This study tested the hypothesis that by protecting β-cells against oxidative stress and inflammation, an Nrf2 activator, dimethyl fumarate (DMF), may prevent or delay the onset of type 1 diabetes in non-obese diabetic (NOD) mice. Firstly, islet isolation was conducted to confirm the antioxidative effects of DMF oral administration on islet cells. Secondly, in a spontaneous diabetes model, DMF (25 mg/kg) was fed to mice once daily starting at the age of 8 weeks up to the age of 22 weeks. In a cyclophosphamide-induced accelerated diabetes model, DMF (25 mg/kg) was fed to mice twice daily for 2 weeks. In the islet isolation study, DMF administration improved the isolation yield, attenuated oxidative stress and enhanced GCLC and NQO1 expression in the islets. In the spontaneous model, DMF significantly reduced the onset of diabetes compared to the control group (25% vs. 54.2%). In the accelerated model, DMF reduced the onset of diabetes from 58.3% to 16.7%. The insulitis score in the islets of the DMF treatment group (1.6 ± 0.32) was significantly lower than in the control group (3.47 ± 0.21). The serum IL-1α, IL-1β, IL-2, IL-4, IL-5, IL-6, IL-9, IL-12p70, IFN-γ, TNF-α, MCP-1 and CXCL16 levels in the DMF-treated group were lower than in the control group. In conclusion, DMF may protect islet cells and reduce the incidence of autoimmune diabetes in NOD mice by attenuating insulitis and proinflammatory cytokine production.
Collapse
Affiliation(s)
- Shiri Li
- Department of Surgery, University of California, Irvine, CA 92868, USA; (L.S.); (C.T.); (K.V.); (M.J.S.)
- Correspondence: (S.L.); (H.I.); Tel.: +1-714-456-5160 (S.L.); +1-714-456-8698 (H.I.)
| | | | - Lourdes Swentek
- Department of Surgery, University of California, Irvine, CA 92868, USA; (L.S.); (C.T.); (K.V.); (M.J.S.)
| | - Chie Takasu
- Department of Surgery, University of California, Irvine, CA 92868, USA; (L.S.); (C.T.); (K.V.); (M.J.S.)
| | - Kelly Vo
- Department of Surgery, University of California, Irvine, CA 92868, USA; (L.S.); (C.T.); (K.V.); (M.J.S.)
| | - Michael J. Stamos
- Department of Surgery, University of California, Irvine, CA 92868, USA; (L.S.); (C.T.); (K.V.); (M.J.S.)
| | - Camillo Ricordi
- Cell Transplant Center, Diabetes Research Institute, University of Miami, Miami, FL 33136, USA;
| | - Hirohito Ichii
- Department of Surgery, University of California, Irvine, CA 92868, USA; (L.S.); (C.T.); (K.V.); (M.J.S.)
- Correspondence: (S.L.); (H.I.); Tel.: +1-714-456-5160 (S.L.); +1-714-456-8698 (H.I.)
| |
Collapse
|
11
|
Lin XC, Pan M, Zhu LP, Sun Q, Zhou ZS, Li CC, Zhang GG. NFAT5 promotes arteriogenesis via MCP-1-dependent monocyte recruitment. J Cell Mol Med 2019; 24:2052-2063. [PMID: 31883300 PMCID: PMC6991654 DOI: 10.1111/jcmm.14904] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 09/28/2019] [Accepted: 10/05/2019] [Indexed: 01/24/2023] Open
Abstract
Studies have demonstrated that nuclear factor of activated T cells 5 (NFAT5) is not only a tonicity‐responsive transcription factor but also activated by other stimuli, so we aim to investigate whether NFAT5 participates in collateral arteries formation in rats. We performed femoral artery ligature (FAL) in rats for hindlimb ischaemia model and found that NFAT5 was up‐regulated in rat adductors with FAL compared with sham group. Knockdown of NFAT5 with locally injection of adenovirus‐mediated NFAT5‐shRNA in rats significantly inhibited hindlimb blood perfusion recovery and arteriogenesis. Moreover, NFAT5 knockdown decreased macrophages infiltration and monocyte chemotactic protein‐1 (MCP‐1) expression in rats adductors. In vitro, with interleukin‐1β (IL‐1β) stimulation and loss‐of‐function studies, we demonstrated that NFAT5 knockdown inhibits MCP‐1 expression in endothelial cells and chemotaxis of THP‐1 cells regulated by ERK1/2 pathway. More importantly, exogenous MCP‐1 delivery could recover hindlimb blood perfusion, promote arteriogenesis and macrophages infiltration in rats after FAL, which were depressed by NFAT5 knockdown. Besides, NFAT5 knockdown also inhibited angiogenesis in gastrocnemius muscles in rats. Our results indicate that NFAT5 is a critical regulator of arteriogenesis and angiogenesis via MCP‐1‐dependent monocyte recruitment, suggesting that NFAT5 may represent an alternative therapeutic target for ischaemic diseases.
Collapse
Affiliation(s)
- Xing-Chi Lin
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Miao Pan
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Ling-Ping Zhu
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Quan Sun
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zheng-Shi Zhou
- Department of Laboratory Animal, Xiangya School of Medicine, Central South University, Changsha, China
| | - Chuan-Chang Li
- Department of Geriatric Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guo-Gang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China.,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.,Department of Cardiovascular Medicine, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
12
|
Bae GD, Park EY, Kim K, Jang SE, Jun HS, Oh YS. Upregulation of caveolin-1 and its colocalization with cytokine receptors contributes to beta cell apoptosis. Sci Rep 2019; 9:16785. [PMID: 31728004 PMCID: PMC6856349 DOI: 10.1038/s41598-019-53278-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 10/30/2019] [Indexed: 02/07/2023] Open
Abstract
Caveolin-1 (cav-1), the principal structural and signalling protein of caveolae, is implicated in various signalling events, including apoptotic cell death in type 2 diabetes. However, the precise role of beta cells in apoptosis has not been clearly defined. In this study, we investigated the involvement of cav-1 in cytokine-induced beta cell apoptosis and its underlying mechanisms in the rat beta cell line, INS-1 and isolated islets. Treatment of cytokine mixture (CM, TNFα + IL-1β) significantly increased the mRNA and protein expression of cav-1, and resulting in increased formation of caveolae. We found that IL-1 receptor 1 and TNF receptor localized to plasma membrane lipid rafts in the control cells and CM treatment recruited these receptors to the caveolae domain. After cav-1 siRNA transfection, CM-dependent NF-κB activation was reduced and consequently downregulated the mRNA expression of iNOS and IL-1β. Finally, decreased cell viability by CM treatment was ameliorated in both INS-1 cells and isolated islets treated with cav-1 siRNA. These results suggest that increased cav-1 expression and recruitment of cytokine receptors into caveolae contribute to CM-induced beta cell apoptosis.
Collapse
Affiliation(s)
- Gong Deuk Bae
- Lee Gil Ya Cancer and Diabetes Institute, Department of Molecular Medicine, Gachon University, Incheon, South Korea
| | - Eun-Young Park
- College of Pharmacy, Mokpo National University, Jeonnam, South Korea
| | - Kyong Kim
- Department of Food and Nutrition, Eulji University, Seongnam, South Korea
| | - Se-Eun Jang
- Department of Food and Nutrition, Eulji University, Seongnam, South Korea
| | - Hee-Sook Jun
- Lee Gil Ya Cancer and Diabetes Institute, Department of Molecular Medicine, Gachon University, Incheon, South Korea
- College of Pharmacy and Gachon Institute of Pharmaceutical Science, Gachon University, Incheon, South Korea
| | - Yoon Sin Oh
- Department of Food and Nutrition, Eulji University, Seongnam, South Korea.
| |
Collapse
|
13
|
Yamamoto WR, Bone RN, Sohn P, Syed F, Reissaus CA, Mosley AL, Wijeratne AB, True JD, Tong X, Kono T, Evans-Molina C. Endoplasmic reticulum stress alters ryanodine receptor function in the murine pancreatic β cell. J Biol Chem 2019; 294:168-181. [PMID: 30420428 PMCID: PMC6322901 DOI: 10.1074/jbc.ra118.005683] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2018] [Revised: 11/08/2018] [Indexed: 01/23/2023] Open
Abstract
Alterations in endoplasmic reticulum (ER) calcium (Ca2+) levels diminish insulin secretion and reduce β-cell survival in both major forms of diabetes. The mechanisms responsible for ER Ca2+ loss in β cells remain incompletely understood. Moreover, a specific role for either ryanodine receptor (RyR) or inositol 1,4,5-triphosphate receptor (IP3R) dysfunction in the pathophysiology of diabetes remains largely untested. To this end, here we applied intracellular and ER Ca2+ imaging techniques in INS-1 β cells and isolated islets to determine whether diabetogenic stressors alter RyR or IP3R function. Our results revealed that the RyR is sensitive mainly to ER stress-induced dysfunction, whereas cytokine stress specifically alters IP3R activity. Consistent with this observation, pharmacological inhibition of the RyR with ryanodine and inhibition of the IP3R with xestospongin C prevented ER Ca2+ loss under ER and cytokine stress conditions, respectively. However, RyR blockade distinctly prevented β-cell death, propagation of the unfolded protein response (UPR), and dysfunctional glucose-induced Ca2+ oscillations in tunicamycin-treated INS-1 β cells and mouse islets and Akita islets. Monitoring at the single-cell level revealed that ER stress acutely increases the frequency of intracellular Ca2+ transients that depend on both ER Ca2+ leakage from the RyR and plasma membrane depolarization. Collectively, these findings indicate that RyR dysfunction shapes ER Ca2+ dynamics in β cells and regulates both UPR activation and cell death, suggesting that RyR-mediated loss of ER Ca2+ may be an early pathogenic event in diabetes.
Collapse
Affiliation(s)
- Wataru R Yamamoto
- Departments of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Robert N Bone
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Paul Sohn
- Departments of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Farooq Syed
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Christopher A Reissaus
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Amber L Mosley
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Aruna B Wijeratne
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Jason D True
- Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Xin Tong
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee 37235
| | - Tatsuyoshi Kono
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Carmella Evans-Molina
- Departments of Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202; Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202; Departments of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana 46202; Roudebush Veterans Affairs Medical Center, Indianapolis, Indiana 46202.
| |
Collapse
|
14
|
Chen YC, Kuo CH, Tsai YM, Lin YC, Hsiao HP, Chen BH, Chen YT, Wang SL, Hung CH. Suppressive effects of metformin on T-helper 1-related chemokines expression in the human monocytic leukemia cell line THP-1. Endocr Res 2018; 43:228-234. [PMID: 29630425 DOI: 10.1080/07435800.2018.1460605] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF THE STUDY Type 1 and type 2 diabetes mellitus (DM) are chronic T-cell-mediated inflammatory diseases. Metformin is a widely used drug for type 2 DM that reduces the need for insulin in type 1 DM. However, whether metformin has an anti-inflammatory effect for treating DM is unknown. We investigated the anti-inflammatory mechanism of metformin in the human monocytic leukemia cell line THP-1. MATERIALS AND METHODS The human monocytic leukemia cell line THP-1 was pretreated with metformin and stimulated with lipopolysaccharide (LPS). The production of T-helper (Th)-1-related chemokines including interferon-γ-induced protein-10 (IP-10) and monocyte chemoattractant protein-1 (MCP-1), Th2-related chemokine macrophage-derived chemokine, and the proinflammatory chemokine tumor necrosis factor-α was measured using enzyme-linked immunosorbent assay. Intracellular signaling pathways were investigated using Western blot analysis and chromatin immunoprecipitation assay. RESULTS Metformin suppressed LPS-induced IP-10 and MCP-1 production as well as LPS-induced phosphorylation of c-Jun N-terminal kinase (JNK), p38, extracellular signal-regulated kinase (ERK), and nuclear factor-kappa B (NF-κB). Moreover, metformin suppressed LPS-induced acetylation of histones H3 and H4 at the IP-10 promoter. CONCLUSIONS Metformin suppressed the production of Th1-related chemokines IP-10 and MCP-1 in THP-1 cells. Suppressive effects of metformin on IP-10 production might be attributed at least partially to the JNK, p38, ERK, and NF-κB pathways as well as to epigenetic regulation through the acetylation of histones H3 and H4. These results indicated the therapeutic anti-inflammatory potential of metformin.
Collapse
Affiliation(s)
- Yen-Chun Chen
- a Department of Pediatrics , Kaohsiung Municipal Hsiao-Kang Hospital , Kaohsiung , Taiwan
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Chang-Hung Kuo
- c Ta-Kuo Clinic , Kaohsiung , Taiwan
- d Department of Pediatrics , Kaohsiung Municipal Ta-Tung Hospital , Kaohsiung , Taiwan
| | - Ying-Ming Tsai
- e Department of Internal Medicine , Kaohsiung Municipal Ta-Tung Hospital , Kaohsiung , Taiwan
- f Graduate Institute of Clinical Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Yi-Ching Lin
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- f Graduate Institute of Clinical Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
- g Department of Laboratory Medicine, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Hui-Pin Hsiao
- a Department of Pediatrics , Kaohsiung Municipal Hsiao-Kang Hospital , Kaohsiung , Taiwan
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- h Department of Pediatrics, Faculty of Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Bai-Hsiun Chen
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- g Department of Laboratory Medicine, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- h Department of Pediatrics, Faculty of Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Yi-Ting Chen
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Shih-Ling Wang
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
| | - Chih-Hsing Hung
- a Department of Pediatrics , Kaohsiung Municipal Hsiao-Kang Hospital , Kaohsiung , Taiwan
- b Department of Pediatrics, Kaohsiung Medical University Hospital , Kaohsiung Medical University , Kaohsiung , Taiwan
- h Department of Pediatrics, Faculty of Medicine, College of Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
- i Research Center for Environmental Medicine , Kaohsiung Medical University , Kaohsiung , Taiwan
| |
Collapse
|
15
|
Um J, Jung N, Kim D, Choi S, Lee SH, Son Y, Park KS. Substance P preserves pancreatic β-cells in type 1 and type 2 diabetic mice. Biochem Biophys Res Commun 2018; 499:960-966. [PMID: 29626466 DOI: 10.1016/j.bbrc.2018.04.028] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 04/03/2018] [Indexed: 01/09/2023]
Abstract
Preservation of pancreatic β-cells is required for the development of therapies for type 1 and type 2 diabetes (T1D and T2D, respectively). Our previous study demonstrated that substance P (SP) preserves β-cell populations in mice with streptozotocin-induced T1D. Here, we demonstrated that chronic systemic treatment with SP restored the mass of β-cells both in nonobese diabetic (NOD) mice with T1D or db/db mice with T2D. SP delayed the onset of T1D in NOD mice via immune modulation. SP inhibited immune infiltration into islets and the salivary glands of NOD mice. In db/db mice, SP treatment rescued glucose intolerance. Moreover, SP inhibited apoptosis, as well as the activation of pancreatic stellate cells in pancreatic islets of db/db mice. SP downregulated the number of α-smooth muscle actin (α-SMA) expressing cells in db/db pancreatic islets. Cleaved-caspase-3 expression was reduced in islets of SP-treated db/db mice compared to that in the control. Therefore, these results suggested that SP may preserve pancreatic β-cells through immune modulation and protection from the stimulated activation of pancreatic stellate cells and apoptosis in T1D and T2D, respectively.
Collapse
Affiliation(s)
- Jihyun Um
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Nunggum Jung
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Dongjin Kim
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Sanghyuk Choi
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Sang-Ho Lee
- Kyung Hee University Hospital at Gangdong, Seoul 05278, South Korea
| | - Youngsook Son
- Graduate School of Biotechnology, Kyung Hee University, Yongin 17104, South Korea
| | - Ki-Sook Park
- Department of Biomedical Science and Technology, Graduate School, Kyung Hee University, Seoul 02447, South Korea; East-West Medical Research Institute, Kyung Hee University, Seoul 02447, South Korea; College of Medicine, Kyung Hee University, Seoul 02447, South Korea; Kyung Hee University Medical Center, Seoul 02447, South Korea.
| |
Collapse
|
16
|
Xie Q, Fei M, Fa Z, Wang L, Wang J, Zhang Y, Wang J, Deng X. Methane-rich saline alleviates cerulein-induced acute pancreatitis by inhibiting inflammatory response, oxidative stress and pancreatic apoptosis in mice. Int Immunopharmacol 2017; 51:17-24. [PMID: 28759809 DOI: 10.1016/j.intimp.2017.07.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 06/26/2017] [Accepted: 07/21/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND Acute pancreatitis (AP) is a potentially life-threatening gastrointestinal disease involving intracellular activation of digestive enzymes and pancreatic acinar cell injury. The present study was performed to investigate whether methane-rich saline (MS) was involved in the regulation of AP. METHODS MS (16ml/kg) was administered at different dosing frequencies on mice with cerulein-induced AP. Serum amylase, lipase and histopathological changes in the pancreas tissue were measured. Serum cytokine TNFα, IL-6, IFNγ and IL-10 were detected by ELISA. The mRNA levels of these inflammatory cytokines in the pancreas were detected by real time-PCR. Myeloperoxidase (MPO) and superoxide dismutase (SOD) were determined using commercial kits. Apoptosis was assessed by immunohistochemistry and Western blot. RESULTS MS treatment reversed the increased serum level of amylase and lipase, alleviated the pathological damage in the pancreas, and decreased the expression of TNFα, IL-6, IFNγ and IL-10 in cerulean-induced AP mice. In addition, MPO was down-regulated and SOD was up-regulated in the MS treated pancreas, indicating that MS had an anti-oxidant effect against AP. Furthermore, MS protected pancreatic cells against cerulean-induced apoptosis and abolished cleaved caspase-3. CONCLUSION MS exerted anti-inflammatory, anti-oxidant and anti-apoptotic effects on cerulein-induced AP in mice and may proved to be a promising therapeutic agent for the clinical treatment of pancreatitis.
Collapse
Affiliation(s)
- Qun Xie
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Miaomiao Fei
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Zhenzong Fa
- Shanghai Key Laboratory of Molecular Mycology, Department of Dermatology, Changzheng Hospital, Second Military Medical University, Shanghai 200003, China
| | - Liping Wang
- Department of Anesthesiology, Fuzhou General Hospital of Nanjing Military Region, Fuzhou 350025, Fujian Province, China
| | - Jun Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Yan Zhang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Jiafeng Wang
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| | - Xiaoming Deng
- Department of Anesthesiology and Intensive Care, Changhai Hospital, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
17
|
Dos Santos RS, Daures M, Philippi A, Romero S, Marselli L, Marchetti P, Senée V, Bacq D, Besse C, Baz B, Marroquí L, Ivanoff S, Masliah-Planchon J, Nicolino M, Soulier J, Socié G, Eizirik DL, Gautier JF, Julier C. dUTPase ( DUT) Is Mutated in a Novel Monogenic Syndrome With Diabetes and Bone Marrow Failure. Diabetes 2017; 66:1086-1096. [PMID: 28073829 DOI: 10.2337/db16-0839] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Accepted: 01/05/2017] [Indexed: 11/13/2022]
Abstract
We describe a new syndrome characterized by early-onset diabetes associated with bone marrow failure, affecting mostly the erythrocytic lineage. Using whole-exome sequencing in a remotely consanguineous patient from a family with two affected siblings, we identified a single homozygous missense mutation (chr15.hg19:g.48,626,619A>G) located in the dUTPase (DUT) gene (National Center for Biotechnology Information Gene ID 1854), affecting both the mitochondrial (DUT-M p.Y142C) and the nuclear (DUT-N p.Y54C) isoforms. We found the same homozygous mutation in an unrelated consanguineous patient with diabetes and bone marrow aplasia from a family with two affected siblings, whereas none of the >60,000 subjects from the Exome Aggregation Consortium (ExAC) was homozygous for this mutation. This replicated observation probability was highly significant, thus confirming the role of this DUT mutation in this syndrome. DUT is a key enzyme for maintaining DNA integrity by preventing misincorporation of uracil into DNA, which results in DNA toxicity and cell death. We showed that DUT silencing in human and rat pancreatic β-cells results in apoptosis via the intrinsic cell death pathway. Our findings support the importance of tight control of DNA metabolism for β-cell integrity and warrant close metabolic monitoring of patients treated by drugs affecting dUTP balance.
Collapse
Affiliation(s)
| | - Mathilde Daures
- INSERM UMRS 958, Faculté de Médecine Paris Diderot, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Anne Philippi
- INSERM UMRS 958, Faculté de Médecine Paris Diderot, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Sophie Romero
- INSERM UMRS 958, Faculté de Médecine Paris Diderot, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Valérie Senée
- INSERM UMRS 958, Faculté de Médecine Paris Diderot, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Delphine Bacq
- Centre National de Génotypage, Institut de Génomique, Commissariat à l'Energie Atomique, Evry, France
| | - Céline Besse
- Centre National de Génotypage, Institut de Génomique, Commissariat à l'Energie Atomique, Evry, France
| | - Baz Baz
- Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Department of Diabetes and Endocrinology, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Laura Marroquí
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Sarah Ivanoff
- Aplastic Anemia Reference Centre, Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, INSERM U944, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Julien Masliah-Planchon
- Aplastic Anemia Reference Centre, Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, INSERM U944, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Marc Nicolino
- Hôpital Femme-Mère-Enfant, Division of Pediatric Endocrinology, Hospices Civils de Lyon, Université Lyon 1, Lyon, France
| | - Jean Soulier
- Aplastic Anemia Reference Centre, Hematology Laboratory, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, INSERM U944, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Gérard Socié
- Hematology Transplantation, Department of Hematology, Immunology and Oncology, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-François Gautier
- Hôpital Lariboisière, Assistance Publique-Hôpitaux de Paris, Department of Diabetes and Endocrinology, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| | - Cécile Julier
- INSERM UMRS 958, Faculté de Médecine Paris Diderot, Université Paris Diderot-Paris 7, Université Sorbonne Paris Cité, Paris, France
| |
Collapse
|
18
|
Grieco FA, Sebastiani G, Juan-Mateu J, Villate O, Marroqui L, Ladrière L, Tugay K, Regazzi R, Bugliani M, Marchetti P, Dotta F, Eizirik DL. MicroRNAs miR-23a-3p, miR-23b-3p, and miR-149-5p Regulate the Expression of Proapoptotic BH3-Only Proteins DP5 and PUMA in Human Pancreatic β-Cells. Diabetes 2017; 66:100-112. [PMID: 27737950 PMCID: PMC5204315 DOI: 10.2337/db16-0592] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 10/08/2016] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease leading to β-cell destruction. MicroRNAs (miRNAs) are small noncoding RNAs that control gene expression and organ formation. They participate in the pathogenesis of several autoimmune diseases, but the nature of miRNAs contributing to β-cell death in T1D and their target genes remain to be clarified. We performed an miRNA expression profile on human islet preparations exposed to the cytokines IL-1β plus IFN-γ. Confirmation of miRNA and target gene modification in human β-cells was performed by real-time quantitative PCR. Single-stranded miRNAs inhibitors were used to block selected endogenous miRNAs. Cell death was measured by Hoechst/propidium iodide staining and activation of caspase-3. Fifty-seven miRNAs were detected as modulated by cytokines. Three of them, namely miR-23a-3p, miR-23b-3p, and miR-149-5p, were downregulated by cytokines and selected for further studies. These miRNAs were found to regulate the expression of the proapoptotic Bcl-2 proteins DP5 and PUMA and consequent human β-cell apoptosis. These results identify a novel cross talk between a key family of miRNAs and proapoptotic Bcl-2 proteins in human pancreatic β-cells, broadening our understanding of cytokine-induced β-cell apoptosis in early T1D.
Collapse
Affiliation(s)
- Fabio Arturo Grieco
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Umberto Di Mario ONLUS Foundation-Toscana Life Sciences Foundation, Siena, Italy
| | - Jonas Juan-Mateu
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Olatz Villate
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Laura Marroqui
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Ladrière
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Ksenya Tugay
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Romano Regazzi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Marco Bugliani
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Islet Cell Laboratory, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- Umberto Di Mario ONLUS Foundation-Toscana Life Sciences Foundation, Siena, Italy
| | - Décio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Brozzi F, Gerlo S, Grieco FA, Juusola M, Balhuizen A, Lievens S, Gysemans C, Bugliani M, Mathieu C, Marchetti P, Tavernier J, Eizirik DL. Ubiquitin D Regulates IRE1α/c-Jun N-terminal Kinase (JNK) Protein-dependent Apoptosis in Pancreatic Beta Cells. J Biol Chem 2016; 291:12040-56. [PMID: 27044747 DOI: 10.1074/jbc.m115.704619] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Indexed: 12/11/2022] Open
Abstract
Pro-inflammatory cytokines contribute to pancreatic beta cell apoptosis in type 1 diabetes at least in part by inducing endoplasmic reticulum (ER) stress and the consequent unfolded protein response (UPR). It remains to be determined what causes the transition from "physiological" to "apoptotic" UPR, but accumulating evidence indicates that signaling by the ER transmembrane protein IRE1α is critical for this transition. IRE1α activation is regulated by both intra-ER and cytosolic cues. We evaluated the role for the presently discovered cytokine-induced and IRE1α-interacting protein ubiquitin D (UBD) on the regulation of IRE1α and its downstream targets. UBD was identified by use of a MAPPIT (mammalian protein-protein interaction trap)-based IRE1α interactome screen followed by comparison against functional genomic analysis of human and rodent beta cells exposed to pro-inflammatory cytokines. Knockdown of UBD in human and rodent beta cells and detailed signal transduction studies indicated that UBD modulates cytokine-induced UPR/IRE1α activation and apoptosis. UBD expression is induced by the pro-inflammatory cytokines interleukin (IL)-1β and interferon (IFN)-γ in rat and human pancreatic beta cells, and it is also up-regulated in beta cells of inflamed islets from non-obese diabetic mice. UBD interacts with IRE1α in human and rodent beta cells, modulating IRE1α-dependent activation of JNK and cytokine-induced apoptosis. Our data suggest that UBD provides a negative feedback on cytokine-induced activation of the IRE1α/JNK pro-apoptotic pathway in cytokine-exposed beta cells.
Collapse
Affiliation(s)
- Flora Brozzi
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sarah Gerlo
- the Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), 9000 Ghent, Belgium, the Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Fabio Arturo Grieco
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Matilda Juusola
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Alexander Balhuizen
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium
| | - Sam Lievens
- the Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), 9000 Ghent, Belgium, the Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Conny Gysemans
- the Laboratory of Clinical and Experimental Endocrinology, KULeuven, 3000 Leuven, Belgium, and
| | - Marco Bugliani
- the Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56126 Pisa, Italy
| | - Chantal Mathieu
- the Laboratory of Clinical and Experimental Endocrinology, KULeuven, 3000 Leuven, Belgium, and
| | - Piero Marchetti
- the Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, 56126 Pisa, Italy
| | - Jan Tavernier
- the Department of Medical Protein Research, Flanders Interuniversity Institute for Biotechnology (VIB), 9000 Ghent, Belgium, the Department of Biochemistry, Ghent University, 9000 Ghent, Belgium
| | - Décio L Eizirik
- From the ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), 1070 Brussels, Belgium,
| |
Collapse
|
20
|
Dong H, Zhang Y, Song L, Kim DS, Wu H, Yang L, Li S, Morgan KA, Adams DB, Wang H. Cell-Permeable Peptide Blocks TLR4 Signaling and Improves Islet Allograft Survival. Cell Transplant 2016; 25:1319-29. [PMID: 26771084 DOI: 10.3727/096368916x690449] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Toll-like receptor 4 (TLR4) activation in pancreatic β cells activates aberrant islet graft cellular pathways and contributes to immune rejection in allogeneic islet transplantation. As an approach to overcoming this problem, we determined the capacity of a 33-amino acid peptide consisting of a protein transduction domain (PTD) from the Hph-1 virus and a fragment of the intracellular domain of TLR4 from the C3H mice (PTD-dnTLR4) to block TLR4 signaling and improve allogeneic islet survival in vitro and after transplantation. The efficacy of PTD-dnTLR4 in blocking TLR4 signaling was assessed in the Raw264.7 macrophage line, in the islets, and the βTC3 cell line. In Raw264.7 cells, preculture with the peptide reduced LPS-induced NF-κB activation and production of proinflammatory cytokines (IL-1β, TNF-α, iNOS, and IL-6). In islets and β cells, preincubation with PTD-dnTLR4 suppressed LPS-induced TNF-α expression via inhibition of NF-κB activation and protected them from stress-induced cell death. In vivo, preincubation of BALB/c (H-2(d)) islets with PTD-dnTLR4 resulted in significantly longer survival than control islets in a streptozotocin-induced diabetes model (two of seven grafts survived long term >100 days). PTD-dnTLR4-treated grafts exhibited reduced expression of TNF-α and iNOS and reduced macrophage infiltration posttransplant. The data indicate that PTD-dnTLR4 blocked TLR4 signaling in both macrophages and β cells, and prolonged allograft survival at least in part by suppressing inflammation and macrophage infiltration. This strategy for blocking TLR4 activity has potential utilization in the treatment of diseases where excessive TLR4 activation contributes to the pathologic cellular pathways such as islet transplantation.
Collapse
Affiliation(s)
- Huansheng Dong
- Department of Surgery, Medical University of South Carolina, Charleston, SC, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Song S, Roy S. Progress and challenges in macroencapsulation approaches for type 1 diabetes (T1D) treatment: Cells, biomaterials, and devices. Biotechnol Bioeng 2016; 113:1381-402. [PMID: 26615050 DOI: 10.1002/bit.25895] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 10/30/2015] [Accepted: 11/24/2015] [Indexed: 12/15/2022]
Abstract
Macroencapsulation technology has been an attractive topic in the field of treatment for Type 1 diabetes due to mechanical stability, versatility, and retrievability of the macro-capsule design. Macro-capsules can be categorized into extravascular and intravascular devices, in which solute transport relies either on diffusion or convection, respectively. Failure of macroencapsulation strategies can be due to limited regenerative capacity of the encased insulin-producing cells, sub-optimal performance of encapsulation biomaterials, insufficient immunoisolation, excessive blood thrombosis for vascular perfusion devices, and inadequate modes of mass transfer to support cell viability and function. However, significant technical advancements have been achieved in macroencapsulation technology, namely reducing diffusion distance for oxygen and nutrients, using pro-angiogenic factors to increase vascularization for islet engraftment, and optimizing membrane permeability and selectivity to prevent immune attacks from host's body. This review presents an overview of existing macroencapsulation devices and discusses the advances based on tissue-engineering approaches that will stimulate future research and development of macroencapsulation technology. Biotechnol. Bioeng. 2016;113: 1381-1402. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Shang Song
- Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco, San Francisco, California 94158
| | - Shuvo Roy
- Department of Bioengineering and Therapeutic Sciences, University of California-San Francisco, San Francisco, California 94158.
| |
Collapse
|
22
|
Santin I, Dos Santos RS, Eizirik DL. Pancreatic Beta Cell Survival and Signaling Pathways: Effects of Type 1 Diabetes-Associated Genetic Variants. Methods Mol Biol 2016; 1433:21-54. [PMID: 26936771 DOI: 10.1007/7651_2015_291] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Type 1 diabetes (T1D) is a complex autoimmune disease in which pancreatic beta cells are specifically destroyed by the immune system. The disease has an important genetic component and more than 50 loci across the genome have been associated with risk of developing T1D. The molecular mechanisms by which these putative T1D candidate genes modulate disease risk, however, remain poorly characterized and little is known about their effects in pancreatic beta cells. Functional studies in in vitro models of pancreatic beta cells, based on techniques to inhibit or overexpress T1D candidate genes, allow the functional characterization of several T1D candidate genes. This requires a multistage procedure comprising two major steps, namely accurate selection of genes of potential interest and then in vitro and/or in vivo mechanistic approaches to characterize their role in pancreatic beta cell dysfunction and death in T1D. This chapter details the methods and settings used by our groups to characterize the role of T1D candidate genes on pancreatic beta cell survival and signaling pathways, with particular focus on potentially relevant pathways in the pathogenesis of T1D, i.e., inflammation and innate immune responses, apoptosis, beta cell metabolism and function.
Collapse
Affiliation(s)
- Izortze Santin
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium.
- Endocrinology and Diabetes Research Group, BioCruces Health Research Institute, CIBERDEM, Spain.
| | - Reinaldo S Dos Santos
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
23
|
Ju SM, Youn GS, Cho YS, Choi SY, Park J. Celastrol ameliorates cytokine toxicity and pro-inflammatory immune responses by suppressing NF-κB activation in RINm5F beta cells. BMB Rep 2015; 48:172-7. [PMID: 25059279 PMCID: PMC4453024 DOI: 10.5483/bmbrep.2015.48.3.147] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Indexed: 01/09/2023] Open
Abstract
Upregulation of pro-inflammatory mediators contributes to β-cell destruction and enhanced infiltration of immune cells into pancreatic islets during development of type 1 diabetes mellitus. In this study, we examined the regulatory effects and the mechanisms of action of celastrol against cytotoxicity and pro-inflammatory immune responses in the RINm5F rat pancreatic β-cell line stimulated with a combination of interleukin-1 beta, tumor necrosis factor-alpha, and interferon-γ. Celastrol significantly restored cytokine-induced cell death and significantly inhibited cytokine-induced nitric oxide production. In addition, the protective effect of celastrol was correlated with a reduction in pro-inflammatory mediators, such as inducible nitric oxide synthase, cyclooxygenase-2, and CC chemokine ligand 2. Furthermore, celastrol significantly suppressed cytokine-induced signaling cascades leading to nuclear factor kappa B (NF-κB) activation, including IκB-kinase (IKK) activation, IκB degradation, p65 phosphorylation, and p65 DNA binding activity. These results suggest that celastrol may exert its cytoprotective activity by suppressing cytokine-induced expression of pro-inflammatory mediators by inhibiting activation of NF-κB in RINm5F cells. [BMB Reports 2015; 48(3): 172-177]
Collapse
Affiliation(s)
- Sung Mi Ju
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Gi Soo Youn
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Yoon Shin Cho
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 200-702, Korea
| | - Jinseu Park
- Department of Biomedical Science and Research Institute for Bioscience & Biotechnology, Hallym University, Chunchon 200-702, Korea
| |
Collapse
|
24
|
Brozzi F, Nardelli TR, Lopes M, Millard I, Barthson J, Igoillo-Esteve M, Grieco FA, Villate O, Oliveira JM, Casimir M, Bugliani M, Engin F, Hotamisligil GS, Marchetti P, Eizirik DL. Cytokines induce endoplasmic reticulum stress in human, rat and mouse beta cells via different mechanisms. Diabetologia 2015; 58:2307-16. [PMID: 26099855 DOI: 10.1007/s00125-015-3669-6] [Citation(s) in RCA: 164] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 05/29/2015] [Indexed: 01/01/2023]
Abstract
AIMS/HYPOTHESIS Proinflammatory cytokines contribute to beta cell damage in type 1 diabetes in part through activation of endoplasmic reticulum (ER) stress. In rat beta cells, cytokine-induced ER stress involves NO production and consequent inhibition of the ER Ca(2+) transporting ATPase sarco/endoplasmic reticulum Ca(2+) pump 2 (SERCA2B). However, the mechanisms by which cytokines induce ER stress and apoptosis in mouse and human pancreatic beta cells remain unclear. The purpose of this study is to elucidate the role of ER stress on cytokine-induced beta cell apoptosis in these three species and thus solve ongoing controversies in the field. METHODS Rat and mouse insulin-producing cells, human pancreatic islets and human EndoC-βH1 cells were exposed to the cytokines IL-1β, TNF-α and IFN-γ, with or without NO inhibition. A global comparison of cytokine-modulated gene expression in human, mouse and rat beta cells was also performed. The chemical chaperone tauroursodeoxycholic acid (TUDCA) and suppression of C/EBP homologous protein (CHOP) were used to assess the role of ER stress in cytokine-induced apoptosis of human beta cells. RESULTS NO plays a key role in cytokine-induced ER stress in rat islets, but not in mouse or human islets. Bioinformatics analysis indicated greater similarity between human and mouse than between human and rat global gene expression after cytokine exposure. The chemical chaperone TUDCA and suppression of CHOP or c-Jun N-terminal kinase (JNK) protected human beta cells against cytokine-induced apoptosis. CONCLUSIONS/INTERPRETATION These observations clarify previous results that were discrepant owing to the use of islets from different species, and confirm that cytokine-induced ER stress contributes to human beta cell death, at least in part via JNK activation.
Collapse
Affiliation(s)
- Flora Brozzi
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Tarlliza R Nardelli
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Miguel Lopes
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Isabelle Millard
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Jenny Barthson
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Mariana Igoillo-Esteve
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Fabio A Grieco
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Olatz Villate
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Joana M Oliveira
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Marina Casimir
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases, Sabri Ülker Center, Harvard School of Public Health, Boston, MA, 02115, USA
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Decio L Eizirik
- ULB-Center for Diabetes Research, Universitè Libre de Bruxelles (ULB), Route de Lennik, 808-CP618, 1070, Brussels, Belgium.
| |
Collapse
|
25
|
Burtea C, Laurent S, Crombez D, Delcambre S, Sermeus C, Millard I, Rorive S, Flamez D, Beckers MC, Salmon I, Vander Elst L, Eizirik DL, Muller RN. Development of a peptide-functionalized imaging nanoprobe for the targeting of (FXYD2)γa as a highly specific biomarker of pancreatic beta cells. CONTRAST MEDIA & MOLECULAR IMAGING 2015; 10:398-412. [PMID: 25930968 DOI: 10.1002/cmmi.1641] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 02/06/2015] [Accepted: 02/17/2015] [Indexed: 01/15/2023]
Abstract
Diabetes is characterized by a progressive decline of the pancreatic beta cell mass (BCM), which is responsible for insufficient insulin secretion and hyperglycaemia. There are currently no reliable methods to measure non-invasively the BCM in diabetic patients. Our work describes a phage display-derived peptide (P88) that is highly specific to (FXYD2)γa expressed by human beta cells and is proposed as a molecular vector for the development of functionalized imaging probes. P88 does not bind to the exocrine pancreas and is able to detect down to ~156 human pancreatic islets/mm(3) in vitro after conjugation to ultra-small particles of iron oxide (USPIO), as proven by the R2 measured on MR images. For in vivo evaluation, MRI studies were carried out on nude mice bearing Capan-2 tumours that also express (FXYD2)γa. A strong negative contrast was obtained subsequent to the injection of USPIO-P88, but not in negative controls. On human histological sections, USPIO-P88 seems to be specific to pancreatic beta cells, but not to duodenum, stomach or kidney tissues. USPIO-P88 thus represents a novel and promising tool for monitoring pancreatic BCM in diabetic patients. The quantitative correlation between BCM and R2 remains to be demonstrated in vivo, but the T2 mapping and the black pixel estimation after USPIO-P88 injection could provide important information for the future pancreatic BCM evaluation by MRI.
Collapse
Affiliation(s)
- Carmen Burtea
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Sophie Laurent
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Deborah Crombez
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Sébastien Delcambre
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Corine Sermeus
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Isabelle Millard
- Center for Diabetes Research, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium.,DIAPath, Center for Microscopy and Molecular Imaging, 8 rue Adrienne Bolland, 6041, Gosselies, Belgium
| | - Daisy Flamez
- Center for Diabetes Research, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Marie-Claire Beckers
- Eurogentec S.A., Liège Science Park, Rue du Bois Saint-Jean 5, B-4102, Seraing, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium.,DIAPath, Center for Microscopy and Molecular Imaging, 8 rue Adrienne Bolland, 6041, Gosselies, Belgium
| | - Luce Vander Elst
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium
| | - Decio L Eizirik
- Center for Diabetes Research, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Robert N Muller
- Department of General, Organic and Biomedical Chemistry, NMR and Molecular Imaging Laboratory, University of Mons, Avenue Maistriau 19, Mendeleev Building, B-7000, Mons, Belgium.,Center for Microscopy and Molecular Imaging, 8 rue Adrienne Bolland, 6041, Gosselies, Belgium
| |
Collapse
|
26
|
Dietary polyherbal supplementation decreases CD3+ cell infiltration into pancreatic islets and prevents hyperglycemia in nonobese diabetic mice. Nutr Res 2015; 35:328-36. [DOI: 10.1016/j.nutres.2014.12.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 12/17/2014] [Accepted: 12/19/2014] [Indexed: 01/26/2023]
|
27
|
Diana J, Lehuen A. Macrophages and β-cells are responsible for CXCR2-mediated neutrophil infiltration of the pancreas during autoimmune diabetes. EMBO Mol Med 2015; 6:1090-104. [PMID: 24968718 PMCID: PMC4154135 DOI: 10.15252/emmm.201404144] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Autoimmune type 1 diabetes (T1D) development results from the interaction between pancreatic β-cells, and the innate and the adaptive immune systems culminating with the destruction of the insulin-secreting β-cells by autoreactive T cells. This diabetogenic course starts during the first postnatal weeks by the infiltration of the pancreatic islets by innate immune cells and particularly neutrophils. Here, we aim to determine the cellular and molecular mechanism leading to the recruitment of this neutrophils in the pancreatic islets of non-obese diabetic (NOD) mice. Here, we show that neutrophil recruitment in the pancreatic islets is controlled by inflammatory macrophages and β-cells themselves. Macrophages and β-cells produce the chemokines CXCL1 and CXCL2, recruiting CXCR2-expressing neutrophils from the blood to the pancreatic islets. We further show that pancreatic macrophages secrete IL-1β-inducing CXCR2 ligand production by the β-cells. Finally, the blockade of neutrophil recruitment at early ages using CXCR2 antagonist dampens the diabetogenic T-cell response and the later development of autoimmune diabetes, supporting the therapeutic potential of this approach. Subject Categories Immunology; Metabolism
Collapse
Affiliation(s)
- Julien Diana
- Institut National de la Santé et de la Recherche Médicale (INSERM), U1151, Necker-Enfants Malades Institute (INEM) Necker Hospital, Paris, France Sorbonne Paris Cité, Université Paris Descartes, Paris, France
| | - Agnès Lehuen
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France Institut National de la Santé et de la Recherche Médicale (INSERM), U1016, Cochin Institute Cochin Hospital, Paris, France Laboratoire d'Excellence INFLAMEX, Paris, France
| |
Collapse
|
28
|
Brozzi F, Gerlo S, Grieco FA, Nardelli TR, Lievens S, Gysemans C, Marselli L, Marchetti P, Mathieu C, Tavernier J, Eizirik DL. A combined "omics" approach identifies N-Myc interactor as a novel cytokine-induced regulator of IRE1 protein and c-Jun N-terminal kinase in pancreatic beta cells. J Biol Chem 2015; 289:20677-93. [PMID: 24936061 DOI: 10.1074/jbc.m114.568808] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Type 1 diabetes is an autoimmune disease with a strong inflammatory component. The cytokines interleukin-1β and interferon-γ contribute to beta cell apoptosis in type 1 diabetes. These cytokines induce endoplasmic reticulum stress and the unfolded protein response (UPR), contributing to the loss of beta cells. IRE1α, one of the UPR mediators, triggers insulin degradation and inflammation in beta cells and is critical for the transition from "physiological" to "pathological" UPR. The mechanisms regulating inositol-requiring protein 1α (IRE1α) activation and its signaling for beta cell "adaptation," "stress response," or "apoptosis" remain to be clarified. To address these questions, we combined mammalian protein-protein interaction trap-based IRE1α interactome and functional genomic analysis of human and rodent beta cells exposed to pro-inflammatory cytokines to identify novel cytokine-induced regulators of IRE1α. Based on this approach, we identified N-Myc interactor (NMI) as an IRE1α-interacting/modulator protein in rodent and human pancreatic beta cells. An increased expression of NMI was detected in islets from nonobese diabetic mice with insulitis and in rodent or human beta cells exposed in vitro to the pro-inflammatory cytokines interleukin-1β and interferon-γ. Detailed mechanistic studies demonstrated that NMI negatively modulates IRE1α-dependent activation of JNK and apoptosis in rodent and human pancreatic beta cells. In conclusion, by using a combined omics approach, we identified NMI induction as a novel negative feedback mechanism that decreases IRE1α-dependent activation of JNK and apoptosis in cytokine-exposed beta cells
Collapse
|
29
|
Campanha-Rodrigues AL, Grazioli G, Oliveira TC, Campos-Lisbôa ACV, Mares-Guia TR, Sogayar MC. Therapeutic Potential of Laminin–Biodritin Microcapsules for Type 1 Diabetes Mellitus. Cell Transplant 2015; 24:247-61. [DOI: 10.3727/096368913x675160] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pancreatic islet microencapsulation constitutes an attractive therapy for type 1 diabetes mellitus; however, long-term β-cell function remains a major problem. Loss of extracellular matrix interactions during islet isolation dramatically affects β-cell viability. We have previously shown beneficial effects of laminin (LN) in human islet cultures. Herein, we investigated whether LN could improve the outcome of transplantation after islet microencapsulation in Biodritin, an alginate-based material. To test LN-Biodritin stability, microcapsules were subjected to different types of in vitro stress. Focusing on biocompatibility, empty microcapsules were coincubated with the RAW 264.7 macrophage cell line for up to 24 h, and empty beads were implanted IP in mice and retrieved for analyses after 7 and 30 days. Upon culturing for 48 h, mRNA, protein levels, and caspase 3 activity were evaluated in islets microencapsulated with LN-Biodritin. Mice rendered diabetic by streptozotocin injection were transplanted with microencapsulated islets, followed by assessment of body weight, glycemia, and graft function (evaluated by OGTT). Graft efficiency was observed upon microencapsulated islet explantation. The results obtained showed that LN-Biodritin microcapsules were as stable and biocompatible as Biodritin. Modulation of mRNA and protein levels suggested protection against apoptosis and islet stress. Mice transplanted with LN-Biodritin microencapsulated islets presented a better outcome at 198 days postsurgery. Graft explantation led animals to hyperglycemia. In conclusion, LN-Biodritin constitutes a very promising biomaterial for islet transplantation.
Collapse
Affiliation(s)
- Ana Lucia Campanha-Rodrigues
- Chemistry Institute, Biochemistry Department, Cell and Molecular Therapy Center (NUCEL/NETCEM), School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Gisella Grazioli
- Chemistry Institute, Biochemistry Department, Cell and Molecular Therapy Center (NUCEL/NETCEM), School of Medicine, University of São Paulo, São Paulo, SP, Brazil
- Cell Protect Biotechnology Ltda., São Paulo, SP, Brazil
| | - Talita C. Oliveira
- Chemistry Institute, Biochemistry Department, Cell and Molecular Therapy Center (NUCEL/NETCEM), School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| | - Ana Carolina V. Campos-Lisbôa
- Chemistry Institute, Biochemistry Department, Cell and Molecular Therapy Center (NUCEL/NETCEM), School of Medicine, University of São Paulo, São Paulo, SP, Brazil
- Cell Protect Biotechnology Ltda., São Paulo, SP, Brazil
| | - Thiago R. Mares-Guia
- Chemistry Institute, Biochemistry Department, Cell and Molecular Therapy Center (NUCEL/NETCEM), School of Medicine, University of São Paulo, São Paulo, SP, Brazil
- Cell Protect Biotechnology Ltda., São Paulo, SP, Brazil
| | - Mari C. Sogayar
- Chemistry Institute, Biochemistry Department, Cell and Molecular Therapy Center (NUCEL/NETCEM), School of Medicine, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
30
|
Morgan NG, Leete P, Foulis AK, Richardson SJ. Islet inflammation in human type 1 diabetes mellitus. IUBMB Life 2014; 66:723-34. [PMID: 25504835 DOI: 10.1002/iub.1330] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 11/17/2014] [Indexed: 01/12/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is caused by the selective deletion of pancreatic β-cells in response to an assault mounted within the pancreas by infiltrating immune cells. However, this apparently clear and focussed annunciation conceals a stark reality in which the cellular and molecular events leading to β-cell loss remain poorly understood in humans. This reflects the difficulty of studying these processes in living individuals and the fact that, using pathological specimens, islet inflammation has been analysed in fewer than 200 recent-onset cases of T1DM worldwide, over the past century. Nevertheless, insights have been gained and the composition of the islet infiltrate is being disclosed. This is shown to be primarily lymphocytic in nature, with populations of both CD8+ and CD4+ T cells displaying an autoreactivity against specific islet antigenic peptides. The T cells are often accompanied by influent CD20+ B cells, although new data imply that the proportions of these individual cell types vary and that patients fall into at least two distinct categories having either a hyper-immune (CD20Hi) or a pauci-immune (CD20Lo) phenotype. The overall rate of β-cell decline appears to correlate with these two phenotypes such that hyper-immune patients lose β-cells more quickly and tend to develop disease at an earlier age than those with the pauci-immune profile. In this article, we review the evidence which underpins our current understanding of the aetiology of T1DM and highlight both the established features as well as areas of on-going ambiguity and debate.
Collapse
Affiliation(s)
- Noel G Morgan
- Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, UK
| | | | | | | |
Collapse
|
31
|
Zhang J, Li G, Gao S, Yao Y, Pang L, Li Y, Wang W, Zhao Q, Kong D, Li C. Monocyte chemoattractant protein-1 released from polycaprolactone/chitosan hybrid membrane to promote angiogenesis in vivo. J BIOACT COMPAT POL 2014. [DOI: 10.1177/0883911514554146] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We have fabricated a hybrid membrane composed of polycaprolactone and a natural polysaccharide, chitosan. The incorporation of chitosan enabled heparinization of the material via electrostatic interaction between heparin and chitosan. More importantly, since multiple cytokines have exhibited binding affinity towards heparin, heparinization of the polycaprolactone/chitosan compound also facilitated immobilization of monocyte chemoattractant protein-1, which is a well-reported pro-angiogenic chemokine. Results demonstrated that the heparinized polycaprolactone/chitosan membrane with monocyte chemoattractant protein-1 immobilization was able to release monocyte chemoattractant protein-1 in a controlled and sustained manner. Bioactivity of the released monocyte chemoattractant protein-1 was uncompromised as shown by a chemotaxis chamber assay using isolated rat peripheral blood mononuclear cells. Enhanced local angiogenesis was subsequently observed in vivo after subcutaneous implantation of the heparinized polycaprolactone/chitosan membrane with monocyte chemoattractant protein-1-releasing property and the mechanisms underlying the angiogenic role of monocyte chemoattractant protein-1 were also investigated. We propose that the monocyte chemoattractant protein-1-induced local capillary formation is attributable to increased recruitment of macrophages, particularly the alternatively activated M2 macrophages, which have been implicated in wound healing. Moreover, a direct effect of monocyte chemoattractant protein-1 on angiogenesis was also observed, mainly via monocyte chemoattractant protein-1-stimulated vascular endothelial growth factor expression and activity. In summary, we report here a feasible way to fabricate a polycaprolactone/chitosan hybrid material that could be functionalized with angiogenic signalling agents, such as monocyte chemoattractant protein-1. Implantation of this material promoted angiogenesis and may therefore be developed into scaffold or dressing materials in treating local ischemia injuries or cutaneous wound.
Collapse
Affiliation(s)
- Ju Zhang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Guoping Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Shan Gao
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Yao Yao
- The Key Laboratory of Bioactive Materials of Ministry of Education, Institute of Molecular Biology, College of Life Science, Nankai University, Tianjin, China
| | - Liyun Pang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Yuejie Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Weiwei Wang
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| | - Qiang Zhao
- The Key Laboratory of Bioactive Materials of Ministry of Education, Institute of Molecular Biology, College of Life Science, Nankai University, Tianjin, China
| | - Deling Kong
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
- The Key Laboratory of Bioactive Materials of Ministry of Education, Institute of Molecular Biology, College of Life Science, Nankai University, Tianjin, China
| | - Chen Li
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Science and Peking Union Medical College, Tianjin, China
| |
Collapse
|
32
|
Mathijs I, Da Cunha DA, Himpe E, Ladriere L, Chellan N, Roux CR, Joubert E, Muller C, Cnop M, Louw J, Bouwens L. Phenylpropenoic acid glucoside augments pancreatic beta cell mass in high-fat diet-fed mice and protects beta cells from ER stress-induced apoptosis. Mol Nutr Food Res 2014; 58:1980-90. [PMID: 25044754 DOI: 10.1002/mnfr.201400211] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 12/24/2022]
Abstract
SCOPE A major goal of diabetes therapy is to identify novel drugs that preserve or expand pancreatic beta cell mass. Here, we examined the effect of a phenylpropenoic acid glucoside (PPAG) on the beta cell mass, and via which mechanism this effect is established. METHODS AND RESULTS Mice were fed a high-fat and fructose-containing diet to induce obesity and hyperglycemia. PPAG treatment protected obese mice from diet-induced hyperglycemia and resulted in a tripling of beta cell mass. The effect of the phytochemical on beta cell mass was neither due to increased proliferation, as determined by Ki67 immunostaining, nor to neogenesis, which was assessed by genetic lineage tracing. TUNEL staining revealed suppressed apoptosis in PPAG-treated obese mice. In vitro, PPAG protected beta cells from palmitate-induced apoptosis. It protected beta cells against ER stress by increasing expression of antiapoptotic B-cell lymphoma 2 (BCL2) protein without affecting proapoptotic signals. CONCLUSIONS We identified an antidiabetic phytochemical that protects pancreatic beta cells from ER stress and apoptosis induced by high-fat diet/lipotoxicity. At the tissue level, this led to a tripling of beta cell mass. At the molecular level, the protective effect of the phytochemical was mediated by increasing BCL2 expression in beta cells.
Collapse
Affiliation(s)
- Iris Mathijs
- Cell Differentiation Lab, Vrije Universiteit Brussel, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Marroquí L, Santin I, Dos Santos RS, Marselli L, Marchetti P, Eizirik DL. BACH2, a candidate risk gene for type 1 diabetes, regulates apoptosis in pancreatic β-cells via JNK1 modulation and crosstalk with the candidate gene PTPN2. Diabetes 2014; 63:2516-27. [PMID: 24608439 DOI: 10.2337/db13-1443] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Type 1 diabetes is a chronic autoimmune disease characterized by specific destruction of pancreatic β-cells by the immune system. Linkage and genome-wide association studies have identified more than 50 loci across the human genome associated with risk of type 1 diabetes. Recently, basic leucine zipper transcription factor 2 (BACH2) has been associated with genetic risk to develop type 1 diabetes, in an effect ascribed to the immune system. We evaluated whether BACH2 may also play a role in immune-mediated pancreatic β-cell apoptosis. BACH2 inhibition exacerbated cytokine-induced β-cell apoptosis in human and rodent β-cells by the mitochondrial pathway of cell death, whereas BACH2 overexpression had protective effects. BACH2 silencing and exposure to proinflammatory cytokines increased phosphorylation of the proapoptotic protein JNK1 by upregulation of mitogen-activated protein kinase kinase 7 (MKK7) and downregulation of PTPN2. JNK1 increased phosphorylation of the proapoptotic protein BIM, and both JNK1 and BIM knockdown protected β-cells against cytokine-induced apoptosis in BACH2-silenced cells. The present findings suggest that the type 1 diabetes candidate gene BACH2 regulates proinflammatory cytokine-induced apoptotic pathways in pancreatic β-cells by crosstalk with another candidate gene, PTPN2, and activation of JNK1 and BIM. This clarifies an unexpected and relevant mechanism by which BACH2 may contribute to diabetes.
Collapse
Affiliation(s)
- Laura Marroquí
- Laboratory of Experimental Medicine, Université Libre de Bruxelles Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Izortze Santin
- Laboratory of Experimental Medicine, Université Libre de Bruxelles Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, BelgiumEndocrinology and Diabetes Research Group, BioCruces Health Research Institute and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Barakaldo, Spain
| | - Reinaldo Sousa Dos Santos
- Laboratory of Experimental Medicine, Université Libre de Bruxelles Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Endocrinology and Metabolism, Metabolic Unit, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, Metabolic Unit, University of Pisa, Pisa, Italy
| | - Decio L Eizirik
- Laboratory of Experimental Medicine, Université Libre de Bruxelles Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
34
|
Cunha DA, Gurzov EN, Naamane N, Ortis F, Cardozo AK, Bugliani M, Marchetti P, Eizirik DL, Cnop M. JunB protects β-cells from lipotoxicity via the XBP1-AKT pathway. Cell Death Differ 2014; 21:1313-24. [PMID: 24786832 DOI: 10.1038/cdd.2014.53] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 03/18/2014] [Accepted: 03/20/2014] [Indexed: 12/24/2022] Open
Abstract
Diets rich in saturated fats may contribute to the loss of pancreatic β-cells in type 2 diabetes. JunB, a member of the activating protein 1 (AP-1) transcription factor family, promotes β-cell survival and mediates part of the beneficial effects of GLP-1 agonists. In this study we interrogated the molecular mechanisms involved in JunB-mediated β-cell protection from lipotoxicity. The saturated fatty acid palmitate decreased JunB expression, and this loss may contribute to β-cell apoptosis, as overexpression of JunB protected cells from lipotoxicity. Array analysis of JunB-deficient β-cells identified a gene expression signature of a downregulated endoplasmic reticulum (ER) stress response and inhibited AKT signaling. JunB stimulates XBP1 expression via the transcription factor c/EBPδ during ER stress, and forced expression of XBP1s rescued the viability of JunB-deficient cells, constituting an important antiapoptotic mechanism. JunB silencing inhibited AKT activation and activated the proapoptotic Bcl-2 protein BAD via its dephosphorylation. BAD knockdown reversed lipotoxic β-cell death potentiated by JunB siRNA. Interestingly, XBP1s links JunB and AKT signaling as XBP1 knockdown also reduced AKT phosphorylation. GLP-1 agonists induced cAMP-dependent AKT phosphorylation leading to β-cell protection against palmitate-induced apoptosis. JunB and XBP1 knockdown or IRE1 inhibition decreased AKT activation by cAMP, leading to β-cell apoptosis. In conclusion, JunB modulates the β-cell ER stress response and AKT signaling via the induction of XBP1s. The activation of the JunB gene network and the crosstalk between the ER stress and AKT pathway constitute a crucial defense mechanism by which GLP-1 agonists protect against lipotoxic β-cell death. These findings elucidate novel β-cell-protective signal transduction in type 2 diabetes.
Collapse
Affiliation(s)
- D A Cunha
- Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - E N Gurzov
- Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - N Naamane
- Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - F Ortis
- Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - A K Cardozo
- Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - M Bugliani
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - P Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - D L Eizirik
- Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - M Cnop
- 1] Laboratory of Experimental Medicine and ULB Center of Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium [2] Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
35
|
Wolden-Kirk H, Rondas D, Bugliani M, Korf H, Van Lommel L, Brusgaard K, Christesen HT, Schuit F, Proost P, Masini M, Marchetti P, Eizirik DL, Overbergh L, Mathieu C. Discovery of molecular pathways mediating 1,25-dihydroxyvitamin D3 protection against cytokine-induced inflammation and damage of human and male mouse islets of Langerhans. Endocrinology 2014; 155:736-47. [PMID: 24424042 DOI: 10.1210/en.2013-1409] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protection against insulitis and diabetes by active vitamin D, 1,25-dihydroxyvitamin D3 (1,25(OH)2D3), in nonobese diabetic mice has until now mainly been attributed to its immunomodulatory effects, but also protective effects of this hormone on inflammation-induced β-cell death have been reported. The aim of this study was to clarify the molecular mechanisms by which 1,25(OH)2D3 contributes to β-cell protection against cytokine-induced β-cell dysfunction and death. Human and mouse islets were exposed to IL-1β and interferon-γ in the presence or absence of 1,25(OH)2D3. Effects on insulin secretion and β-cell survival were analyzed by glucose-stimulated insulin release and electron microscopy or Hoechst/propidium iodide staining, respectively. Gene expression profiles were assessed by Affymetrix microarrays. Nuclear factor-κB activity was tested, whereas effects on secreted chemokines/cytokines were confirmed by ELISA and migration studies. Cytokine exposure caused a significant increase in β-cell apoptosis, which was almost completely prevented by 1,25(OH)2D3. In addition, 1,25(OH)2D3 restored insulin secretion from cytokine-exposed islets. Microarray analysis of murine islets revealed that the expression of approximately 4000 genes was affected by cytokines after 6 and 24 hours (n = 4; >1.3-fold; P < .02), of which nearly 250 genes were modified by 1,25(OH)2D3. These genes belong to functional groups involved in immune response, chemotaxis, cell death, and pancreatic β-cell function/phenotype. In conclusion, these findings demonstrate a direct protective effect of 1,25(OH)2D3 against inflammation-induced β-cell dysfunction and death in human and murine islets, with, in particular, alterations in chemokine production by the islets. These effects may contribute to the beneficial effects of 1,25(OH)2D3 against the induction of autoimmune diabetes.
Collapse
Affiliation(s)
- H Wolden-Kirk
- Clinical and Experimental Endocrinology (H.W.-K., D.R., H.K., L.O., C.M.), University Hospital Gasthuisberg, Gene Expression Unit (L.V.L., F.S.), Department of Molecular and Cellular Medicine, Department of Microbiology and Immunology (P.P.), B-3000 Leuven, Belgium; Hans Christian Andersen Children's Hospital (H.W.-K., H.T.C.) and Department of Clinical Genetics (K.B., D.L.E.), Odense University Hospital, DK-5000, Odense, Denmark; Department of Endocrinology and Metabolism (M.B., P.M.), Metabolic Unit, and Department of General Pathology (M.M.), University of Pisa, Pisa, Italy; and Laboratory of Experimental Medicine (D.L.E.), Université Libre de Bruxelles, B-1070 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Grieco FA, Moore F, Vigneron F, Santin I, Villate O, Marselli L, Rondas D, Korf H, Overbergh L, Dotta F, Marchetti P, Mathieu C, Eizirik DL. IL-17A increases the expression of proinflammatory chemokines in human pancreatic islets. Diabetologia 2014; 57:502-11. [PMID: 24352375 DOI: 10.1007/s00125-013-3135-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 11/15/2013] [Indexed: 01/05/2023]
Abstract
AIMS/HYPOTHESIS Cytotoxic T cells and macrophages contribute to beta cell destruction in type 1 diabetes at least in part through the production of cytokines such as IL-1β, IFN-γ and TNF-α. We have recently shown the IL-17 pathway to be activated in circulating T cells and pancreatic islets of type 1 diabetes patients. Here, we studied whether IL-17A upregulates the production of chemokines by human pancreatic islets, thus contributing to the build-up of insulitis. METHODS Human islets (from 18 donors), INS-1E cells and islets from wild-type and Stat1 knockout mice were studied. Dispersed islet cells were left untreated, or were treated with IL-17A alone or together with IL-1β+IFN-γ or TNF-α+IFN-γ. RNA interference was used to knock down signal transducer and activator of transcription 1 (STAT1). Chemokine expression was assessed by quantitative RT-PCR, ELISA and histology. Cell viability was evaluated with nuclear dyes. RESULTS IL-17A augmented IL-1β+IFN-γ- and TNF-α+IFN-γ-induced chemokine mRNA and protein expression, and apoptosis in human islets. Beta cells were at least in part the source of chemokine production. Knockdown of STAT1 in human islets prevented cytokine- or IL-17A+cytokine-induced apoptosis and the expression of particular chemokines, e.g. chemokine (C-X-C motif) ligands 9 and 10. Similar observations were made in islets isolated from Stat1 knockout mice. CONCLUSIONS/INTERPRETATION Our findings indicate that IL-17A exacerbates proinflammatory chemokine expression and secretion by human islets exposed to cytokines. This suggests that IL-17A contributes to the pathogenesis of type 1 diabetes by two mechanisms, namely the exacerbation of beta cell apoptosis and increased local production of chemokines, thus potentially aggravating insulitis.
Collapse
Affiliation(s)
- Fabio A Grieco
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Route de Lennik, 808 - CP618, 1070, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
GLIS3, a susceptibility gene for type 1 and type 2 diabetes, modulates pancreatic beta cell apoptosis via regulation of a splice variant of the BH3-only protein Bim. PLoS Genet 2013; 9:e1003532. [PMID: 23737756 PMCID: PMC3667755 DOI: 10.1371/journal.pgen.1003532] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 04/12/2013] [Indexed: 12/19/2022] Open
Abstract
Mutations in human Gli-similar (GLIS) 3 protein cause neonatal diabetes. The GLIS3 gene region has also been identified as a susceptibility risk locus for both type 1 and type 2 diabetes. GLIS3 plays a role in the generation of pancreatic beta cells and in insulin gene expression, but there is no information on the role of this gene on beta cell viability and/or susceptibility to immune- and metabolic-induced stress. GLIS3 knockdown (KD) in INS-1E cells, primary FACS-purified rat beta cells, and human islet cells decreased expression of MafA, Ins2, and Glut2 and inhibited glucose oxidation and insulin secretion, confirming the role of this transcription factor for the beta cell differentiated phenotype. GLIS3 KD increased beta cell apoptosis basally and sensitized the cells to death induced by pro-inflammatory cytokines (interleukin 1β + interferon-γ) or palmitate, agents that may contribute to beta cell loss in respectively type 1 and 2 diabetes. The increased cell death was due to activation of the intrinsic (mitochondrial) pathway of apoptosis, as indicated by cytochrome c release to the cytosol, Bax translocation to the mitochondria and activation of caspases 9 and 3. Analysis of the pathways implicated in beta cell apoptosis following GLIS3 KD indicated modulation of alternative splicing of the pro-apoptotic BH3-only protein Bim, favouring expression of the pro-death variant BimS via inhibition of the splicing factor SRp55. KD of Bim abrogated the pro-apoptotic effect of GLIS3 loss of function alone or in combination with cytokines or palmitate. The present data suggest that altered expression of the candidate gene GLIS3 may contribute to both type 1 and 2 type diabetes by favouring beta cell apoptosis. This is mediated by alternative splicing of the pro-apoptotic protein Bim and exacerbated formation of the most pro-apoptotic variant BimS. Pancreatic beta cell dysfunction and death is a central event in the pathogenesis of diabetes. Genome-wide association studies have identified a large number of associations between specific loci and the two main forms of diabetes, namely type 1 and type 2 diabetes, but the mechanisms by which these candidate genes predispose to diabetes remain to be clarified. The GLIS3 gene region has been identified as a susceptibility risk locus for both type 1 and type 2 diabetes—it is actually the only locus showing association with both forms of diabetes and the regulation of blood glucose. We show that decreased expression of GLIS3 may contribute to diabetes by favouring beta cell apoptosis. This is mediated by the mitochondrial pathway of apoptosis, activated via alternative splicing (a process by which exons are joined in multiple ways, leading to the generation of several proteins by a single gene) of the pro-apoptotic protein Bim, which favours formation of the most pro-apoptotic variant. The present data provides the first evidence that a susceptibility gene for diabetes may contribute to disease via regulation of alternative splicing of a pro-apoptotic gene in pancreatic beta cells.
Collapse
|
38
|
Taylor-Fishwick DA, Weaver JR, Grzesik W, Chakrabarti S, Green-Mitchell S, Imai Y, Kuhn N, Nadler JL. Production and function of IL-12 in islets and beta cells. Diabetologia 2013; 56:126-35. [PMID: 23052055 PMCID: PMC3651896 DOI: 10.1007/s00125-012-2732-9] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 08/09/2012] [Indexed: 10/27/2022]
Abstract
AIMS/HYPOTHESIS IL-12 is an important cytokine in early inflammatory responses and is implicated in the immune-mediated pathogenesis of pancreatic islets in diabetes. However, little is known about the direct effects of IL-12 on islets and beta cells. METHODS In this study, beta cell function, gene expression and protein production were assessed in primary human donor islets and murine beta cell lines in response to stimulation with IL-12 or a pro-inflammatory cytokine cocktail (TNF-α, IL-1β and IFN-γ). RESULTS The pro-inflammatory cytokine cocktail induced islet dysfunction and potently increased the expression and production of IL-12 ligand and IL-12 receptor in human islets. In human islets, the receptor for IL-12 co-localised to the cell surface of insulin-producing cells. Both IL-12 ligand and IL-12 receptor are expressed in the homogeneous beta cell line INS-1. IL-12 induced changes in gene expression, including a dose-dependent upregulation of IFNγ (also known as IFNG), in INS-1 cells. A neutralising antibody to IL-12 directly inhibited IFNγ gene expression in human donor islets induced by either IL-12 or pro-inflammatory cytokine stimulation. Functionally, IL-12 impaired glucose-stimulated insulin secretion (GSIS) in INS-1 cells and human donor islets. A neutralising antibody to IL-12 reversed the beta cell dysfunction (uncoupling of GSIS or induction of caspase-3 activity) induced by pro-inflammatory cytokines. CONCLUSIONS/INTERPRETATION These data identify beta cells as a local source of IL-12 ligand and suggest a direct role of IL-12 in mediating beta cell pathology.
Collapse
Affiliation(s)
- D A Taylor-Fishwick
- Strelitz Diabetes Center, Department of Internal Medicine, Eastern Virginia Medical School, 700 W. Olney Road, Norfolk, VA 23501, USA.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Antibodies recognizing specific Mycobacterium avium subsp. paratuberculosis's MAP3738c protein in type 1 diabetes mellitus children are associated with serum Th1 (CXCL10) chemokine. Cytokine 2012; 61:337-9. [PMID: 23265968 DOI: 10.1016/j.cyto.2012.11.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 09/26/2012] [Accepted: 11/16/2012] [Indexed: 11/24/2022]
Abstract
Recently Mycobacterium avium subsp. paratuberculosis (Map) was associated to type 1 diabetes mellitus (T1DM). In this study we investigated for Map presence in children affected by T1DM compared to healthy children. A pool of 212 sera from T1DM children at onset was compared to sera from 57 healthy children for humoral immune response towards the Map specific protein MAP3738c by ELISA. Serum concentrations of CXCL10 (pro-Th1) and CCL2 (pro-Th2) chemokines were also measured in both sera pool. Results showed that T1DM children had a stronger seropositivity towards MAP3738c protein compared to healthy children. Data highlighted also the correlation between serum activity of T1DM patients towards the specific protein of Map and the increase of CXCL10 concentration if compared to non-diabetic subjects. In conclusion, an immune response to Map in T1DM patients at onset was observed and this may indicate a role of the bacterium in triggering or precipitating the disease.
Collapse
|
40
|
Burke SJ, Goff MR, Updegraff BL, Lu D, Brown PL, Minkin SC, Biggerstaff JP, Zhao L, Karlstad MD, Collier JJ. Regulation of the CCL2 gene in pancreatic β-cells by IL-1β and glucocorticoids: role of MKP-1. PLoS One 2012; 7:e46986. [PMID: 23056550 PMCID: PMC3467264 DOI: 10.1371/journal.pone.0046986] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 09/07/2012] [Indexed: 02/06/2023] Open
Abstract
Release of pro-inflammatory cytokines from both resident and invading leukocytes within the pancreatic islets impacts the development of Type 1 diabetes mellitus. Synthesis and secretion of the chemokine CCL2 from pancreatic β-cells in response to pro-inflammatory signaling pathways influences immune cell recruitment into the pancreatic islets. Therefore, we investigated the positive and negative regulatory components controlling expression of the CCL2 gene using isolated rat islets and INS-1-derived β-cell lines. We discovered that activation of the CCL2 gene by IL-1β required the p65 subunit of NF-κB and was dependent on genomic response elements located in the -3.6 kb region of the proximal gene promoter. CCL2 gene transcription in response to IL-1β was blocked by pharmacological inhibition of the IKKβ and p38 MAPK pathways. The IL-1β-mediated increase in CCL2 secretion was also impaired by p38 MAPK inhibition and by glucocorticoids. Moreover, multiple synthetic glucocorticoids inhibited the IL-1β-stimulated induction of the CCL2 gene. Induction of the MAP Kinase Phosphatase-1 (MKP-1) gene by glucocorticoids or by adenoviral-mediated overexpression decreased p38 MAPK phosphorylation, which diminished CCL2 gene expression, promoter activity, and release of CCL2 protein. We conclude that glucocorticoid-mediated repression of IL-1β-induced CCL2 gene transcription and protein secretion occurs in part through the upregulation of the MKP-1 gene and subsequent deactivation of the p38 MAPK. Furthermore, the anti-inflammatory actions observed with MKP-1 overexpression were obtained without suppressing glucose-stimulated insulin secretion. Thus, MKP-1 is a possible target for anti-inflammatory therapeutic intervention with preservation of β-cell function.
Collapse
Affiliation(s)
- Susan J. Burke
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Matthew R. Goff
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Barrett L. Updegraff
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Danhong Lu
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Patricia L. Brown
- Advanced Microscopy and Imaging Center, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Steven C. Minkin
- Advanced Microscopy and Imaging Center, University of Tennessee, Knoxville, Tennessee, United States of America
| | - John P. Biggerstaff
- Advanced Microscopy and Imaging Center, University of Tennessee, Knoxville, Tennessee, United States of America
| | - Ling Zhao
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee, United States of America
- University of Tennessee Obesity Research Center, Knoxville, Tennessee, United States of America
| | - Michael D. Karlstad
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee, United States of America
- Department of Surgery, Graduate School of Medicine, University of Tennessee Medical Center, Knoxville, Tennessee, United States of America
- University of Tennessee Obesity Research Center, Knoxville, Tennessee, United States of America
| | - J. Jason Collier
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee, United States of America
- University of Tennessee Obesity Research Center, Knoxville, Tennessee, United States of America
| |
Collapse
|
41
|
Panee J. Monocyte Chemoattractant Protein 1 (MCP-1) in obesity and diabetes. Cytokine 2012; 60:1-12. [PMID: 22766373 DOI: 10.1016/j.cyto.2012.06.018] [Citation(s) in RCA: 299] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Revised: 05/30/2012] [Accepted: 06/04/2012] [Indexed: 12/23/2022]
Abstract
Monocyte Chemoattractant Protein-1 (MCP-1) is the first discovered and most extensively studied CC chemokine, and the amount of studies on its role in the etiologies of obesity- and diabetes-related diseases have increased exponentially during the past two decades. This review attempted to provide a panoramic perspective of the history, regulatory mechanisms, functions, and therapeutic strategies of this chemokine. The highlights of this review include the roles of MCP-1 in the development of obesity, diabetes, cardiovascular diseases, insulitis, diabetic nephropathy, and diabetic retinopathy. Therapies that specifically or non-specifically inhibit MCP-1 overproduction have been summarized.
Collapse
Affiliation(s)
- Jun Panee
- Department of Cell and Molecular Biology, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street BSB 222, Honolulu, HI 96813, USA.
| |
Collapse
|
42
|
Miani M, Colli ML, Ladrière L, Cnop M, Eizirik DL. Mild endoplasmic reticulum stress augments the proinflammatory effect of IL-1β in pancreatic rat β-cells via the IRE1α/XBP1s pathway. Endocrinology 2012; 153:3017-28. [PMID: 22529213 DOI: 10.1210/en.2011-2090] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The prevalence of obesity and type 1 diabetes in children is increasing worldwide. Insulin resistance and augmented circulating free fatty acids associated with obesity may cause pancreatic β-cell endoplasmic reticulum (ER) stress. We tested the hypothesis that mild ER stress predisposes β-cells to an exacerbated inflammatory response when exposed to IL-1β or TNF-α, cytokines that contribute to the pathogenesis of type 1 diabetes. INS-1E cells or primary rat β-cells were exposed to a low dose of the ER stressor cyclopiazonic acid (CPA) or free fatty acids, followed by low-dose IL-1β or TNF-α. ER stress signaling was inhibited by small interfering RNA. Cells were evaluated for proinflammatory gene expression by RT-PCR and ELISA, gene reporter activity, p65 activation by immunofluorescence, and apoptosis. CPA pretreatment enhanced IL-1β- induced, but not TNF-α-induced, expression of chemokine (C-C motif) ligand 2, chemokine (C-X-C motif) ligand 1, inducible nitric oxide synthase, and Fas via augmented nuclear factor κB (NF-κB) activation. X-box binding protein 1 (XBP1) and inositol-requiring enzyme 1, but not CCAAT/enhancer binding protein homologous protein, knockdown prevented the CPA-induced exacerbation of NF-κB-dependent genes and decreased IL-1β-induced NF-κB promoter activity. XBP1 modulated NF-κB activity via forkhead box O1 inhibition. In conclusion, rat β-cells facing mild ER stress are sensitized to IL-1β, generating a more intense and protracted inflammatory response through inositol-requiring enzyme 1/XBP1 activation. These observations link β-cell ER stress to the triggering of exacerbated local inflammation.
Collapse
Affiliation(s)
- Michela Miani
- Laboratory of Experimental Medicine, Erasmus Hospital, Université Libre de Bruxelles, B-1070 Brussels, Belgium
| | | | | | | | | |
Collapse
|
43
|
Eizirik DL, Sammeth M, Bouckenooghe T, Bottu G, Sisino G, Igoillo-Esteve M, Ortis F, Santin I, Colli ML, Barthson J, Bouwens L, Hughes L, Gregory L, Lunter G, Marselli L, Marchetti P, McCarthy MI, Cnop M. The human pancreatic islet transcriptome: expression of candidate genes for type 1 diabetes and the impact of pro-inflammatory cytokines. PLoS Genet 2012; 8:e1002552. [PMID: 22412385 PMCID: PMC3297576 DOI: 10.1371/journal.pgen.1002552] [Citation(s) in RCA: 375] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Accepted: 01/10/2012] [Indexed: 01/06/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease in which pancreatic beta cells are killed by infiltrating immune cells and by cytokines released by these cells. Signaling events occurring in the pancreatic beta cells are decisive for their survival or death in diabetes. We have used RNA sequencing (RNA–seq) to identify transcripts, including splice variants, expressed in human islets of Langerhans under control conditions or following exposure to the pro-inflammatory cytokines interleukin-1β (IL-1β) and interferon-γ (IFN-γ). Based on this unique dataset, we examined whether putative candidate genes for T1D, previously identified by GWAS, are expressed in human islets. A total of 29,776 transcripts were identified as expressed in human islets. Expression of around 20% of these transcripts was modified by pro-inflammatory cytokines, including apoptosis- and inflammation-related genes. Chemokines were among the transcripts most modified by cytokines, a finding confirmed at the protein level by ELISA. Interestingly, 35% of the genes expressed in human islets undergo alternative splicing as annotated in RefSeq, and cytokines caused substantial changes in spliced transcripts. Nova1, previously considered a brain-specific regulator of mRNA splicing, is expressed in islets and its knockdown modified splicing. 25/41 of the candidate genes for T1D are expressed in islets, and cytokines modified expression of several of these transcripts. The present study doubles the number of known genes expressed in human islets and shows that cytokines modify alternative splicing in human islet cells. Importantly, it indicates that more than half of the known T1D candidate genes are expressed in human islets. This, and the production of a large number of chemokines and cytokines by cytokine-exposed islets, reinforces the concept of a dialog between pancreatic islets and the immune system in T1D. This dialog is modulated by candidate genes for the disease at both the immune system and beta cell level. Pancreatic beta cells are destroyed by the immune system in type 1 diabetes mellitus, causing insulin dependence for life. Candidate genes for diabetes contribute to this process by acting both at the immune system and, as we suggest here, at the pancreatic beta cell level. We have utilized a novel technology, RNA sequencing, to define all transcripts expressed in human pancreatic islets under basal conditions and following exposure to cytokines, pro-inflammatory mediators that contribute to trigger diabetes. Our observations double the number of known genes present in human islets and indicate that >60% of the candidate genes for type 1 diabetes are expressed in beta cells. The data also show that pro-inflammatory cytokines modify alternative splicing in human islets, a process that may generate novel RNAs and proteins recognizable by the immune system. This, taken together with the findings that pancreatic beta cells themselves express and release many cytokines and chemokines (proteins that attract immune cells), indicates that early type 1 diabetes is characterized by a dialog between beta cells and the immune system. We suggest that candidate genes for diabetes function at least in part as “writers” for the beta cell words in this dialog.
Collapse
Affiliation(s)
- Décio L. Eizirik
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- * E-mail: (DLE); (MC)
| | - Michael Sammeth
- Functional Bioinformatics (FBI), Centre Nacional d'Anàlisi Genòmica (CNAG), Barcelona, Spain
| | - Thomas Bouckenooghe
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Guy Bottu
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Giorgia Sisino
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Mariana Igoillo-Esteve
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Fernanda Ortis
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Izortze Santin
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Maikel L. Colli
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Jenny Barthson
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Luc Bouwens
- Cell Differentiation Unit, Diabetes Research Centre, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Linda Hughes
- Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), Churchill Hospital, Oxford, United Kingdom
| | - Lorna Gregory
- Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), Churchill Hospital, Oxford, United Kingdom
| | - Gerton Lunter
- Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), Churchill Hospital, Oxford, United Kingdom
| | - Lorella Marselli
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, University of Pisa, Pisa, Italy
| | - Mark I. McCarthy
- Oxford Centre for Diabetes, Endocrinology, and Metabolism (OCDEM), Churchill Hospital, Oxford, United Kingdom
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Oxford NIHR Biomedical Research Centre, Churchill Hospital, Oxford, United Kingdom
| | - Miriam Cnop
- Laboratory of Experimental Medicine, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles (ULB), Brussels, Belgium
- * E-mail: (DLE); (MC)
| |
Collapse
|
44
|
Pancreatic islet expression of chemokine CCL2 suppresses autoimmune diabetes via tolerogenic CD11c+ CD11b+ dendritic cells. Proc Natl Acad Sci U S A 2012; 109:3457-62. [PMID: 22328150 DOI: 10.1073/pnas.1115308109] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Development of type 1 diabetes in the nonobese diabetic (NOD) mouse is preceded by an immune cell infiltrate in the pancreatic islets. The exact role of the attracted cells is still poorly understood. Chemokine CCL2/MCP-1 is known to attract CCR2(+) monocytes and dendritic cells (DCs). We have previously shown that transgenic expression of CCL2 in pancreatic islets via the rat insulin promoter induces nondestructive insulitis on a nonautoimmune background. We report here an unexpected reduction of diabetes development on the NOD background despite an increased islet cell infiltrate with markedly increased numbers of CD11c(+) CD11b(+) DCs. These DCs exhibited a hypoactive phenotype with low CD40, MHC II, CD80/CD86 expression, and reduced TNF-α but elevated IL-10 secretions. They failed to induce proliferation of diabetogenic CD4(+) T cells in vitro. Pancreatic lymph node CD4(+) T cells were down-regulated ex vivo and expressed the anergy marker Grail. By using an in vivo transfer system, we show that CD11c(+) CD11b(+) DCs from rat insulin promoter-CCL2 transgenic NOD mice were the most potent cells suppressing diabetes development. These findings support an unexpected beneficial role for CCL2 in type 1 diabetes with implications for current strategies interfering with the CCL2/CCR2 axis in humans, and for dendritic cell biology in autoimmunity.
Collapse
|
45
|
Volkmar M, Dedeurwaerder S, Cunha DA, Ndlovu MN, Defrance M, Deplus R, Calonne E, Volkmar U, Igoillo-Esteve M, Naamane N, Del Guerra S, Masini M, Bugliani M, Marchetti P, Cnop M, Eizirik DL, Fuks F. DNA methylation profiling identifies epigenetic dysregulation in pancreatic islets from type 2 diabetic patients. EMBO J 2012; 31:1405-26. [PMID: 22293752 PMCID: PMC3321176 DOI: 10.1038/emboj.2011.503] [Citation(s) in RCA: 296] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Accepted: 12/12/2011] [Indexed: 12/17/2022] Open
Abstract
In addition to genetic predisposition, environmental and lifestyle factors contribute to the pathogenesis of type 2 diabetes (T2D). Epigenetic changes may provide the link for translating environmental exposures into pathological mechanisms. In this study, we performed the first comprehensive DNA methylation profiling in pancreatic islets from T2D and non-diabetic donors. We uncovered 276 CpG loci affiliated to promoters of 254 genes displaying significant differential DNA methylation in diabetic islets. These methylation changes were not present in blood cells from T2D individuals nor were they experimentally induced in non-diabetic islets by exposure to high glucose. For a subgroup of the differentially methylated genes, concordant transcriptional changes were present. Functional annotation of the aberrantly methylated genes and RNAi experiments highlighted pathways implicated in β-cell survival and function; some are implicated in cellular dysfunction while others facilitate adaptation to stressors. Together, our findings offer new insights into the intricate mechanisms of T2D pathogenesis, underscore the important involvement of epigenetic dysregulation in diabetic islets and may advance our understanding of T2D aetiology.
Collapse
Affiliation(s)
- Michael Volkmar
- Laboratory of Cancer Epigenetics, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Arif S, Moore F, Marks K, Bouckenooghe T, Dayan CM, Planas R, Vives-Pi M, Powrie J, Tree T, Marchetti P, Huang GC, Gurzov EN, Pujol-Borrell R, Eizirik DL, Peakman M. Peripheral and islet interleukin-17 pathway activation characterizes human autoimmune diabetes and promotes cytokine-mediated β-cell death. Diabetes 2011; 60:2112-9. [PMID: 21659501 PMCID: PMC3142078 DOI: 10.2337/db10-1643] [Citation(s) in RCA: 163] [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: 11/30/2010] [Accepted: 05/05/2011] [Indexed: 12/17/2022]
Abstract
OBJECTIVE CD4 T-cells secreting interleukin (IL)-17 are implicated in several human autoimmune diseases, but their role in type 1 diabetes has not been defined. To address the relevance of such cells, we examined IL-17 secretion in response to β-cell autoantigens, IL-17A gene expression in islets, and the potential functional consequences of IL-17 release for β-cells. RESEARCH DESIGN AND METHODS Peripheral blood CD4 T-cell responses to β-cell autoantigens (proinsulin, insulinoma-associated protein, and GAD65 peptides) were measured by IL-17 enzyme-linked immunospot assay in patients with new-onset type 1 diabetes (n = 50). mRNA expression of IL-17A and IFNG pathway genes was studied by qRT-PCR using islets obtained from subjects who died 5 days and 10 years after diagnosis of disease, respectively, and from matched control subjects. IL-17 effects on the function of human islets, rat β-cells, and the rat insulinoma cell line INS-1E were examined. RESULTS A total of 27 patients (54%) showed IL-17 reactivity to one or more β-cell peptides versus 3 of 30 (10%) control subjects (P = 0.0001). In a single case examined close to diagnosis, islet expression of IL17A, RORC, and IL22 was detected. It is noteworthy that we show that IL-17 mediates significant and reproducible enhancement of IL-1β/interferon (IFN)-γ-induced and tumor necrosis factor (TNF)-α/IFN-γ-induced apoptosis in human islets, rat β-cells, and INS-1E cells, in association with significant upregulation of β-cell IL17RA expression via activation of the transcription factors STAT1 and nuclear factor (NF)-κB. CONCLUSIONS Circulating IL-17(+) β-cell-specific autoreactive CD4 T-cells are a feature of type 1 diabetes diagnosis. We disclose a novel pathway to β-cell death involving IL-17 and STAT1 and NF-κB, rendering this cytokine a novel disease biomarker and potential therapeutic target.
Collapse
Affiliation(s)
- Sefina Arif
- Department of Immunobiology, School of Medicine, King’s College London, London, U.K
| | - Fabrice Moore
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Katherine Marks
- Department of Immunobiology, School of Medicine, King’s College London, London, U.K
| | - Thomas Bouckenooghe
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Colin M. Dayan
- Henry Wellcome Laboratories for Integrative Neuroscience and Endocrinology, University of Bristol, Bristol, U.K
| | - Raquel Planas
- Laboratory of Immunobiology, Blood and Tissue Bank, Institut d’Investigacio Germans Trias i Pujol, Barcelona, Spain
| | - Marta Vives-Pi
- Laboratory of Immunobiology, Blood and Tissue Bank, Institut d’Investigacio Germans Trias i Pujol, Barcelona, Spain
| | - Jake Powrie
- Department of Diabetes and Endocrinology, Guy’s & St Thomas’ Hospital National Health Service (NHS) Foundation Trust, London, U.K
- National Institute for Health Research Biomedical Research Centre at Guy’s & St Thomas’ NHS Foundation Trust and King’s College London, London, U.K
| | - Timothy Tree
- Department of Immunobiology, School of Medicine, King’s College London, London, U.K
| | - Piero Marchetti
- Department of Endocrinology and Metabolism, Metabolic Unit, University of Pisa, Pisa, Italy
| | - Guo Cai Huang
- Diabetes Research Group, School of Medicine, King’s College London, London, U.K
| | - Esteban N. Gurzov
- Department of Endocrinology and Metabolism, Metabolic Unit, University of Pisa, Pisa, Italy
| | - Ricardo Pujol-Borrell
- Department of Diabetes and Endocrinology, Guy’s & St Thomas’ Hospital National Health Service (NHS) Foundation Trust, London, U.K
| | - Decio L. Eizirik
- Laboratory of Experimental Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Mark Peakman
- Department of Immunobiology, School of Medicine, King’s College London, London, U.K
- National Institute for Health Research Biomedical Research Centre at Guy’s & St Thomas’ NHS Foundation Trust and King’s College London, London, U.K
| |
Collapse
|
47
|
Shahaf G, Moser H, Ozeri E, Mizrahi M, Abecassis A, Lewis EC. α-1-antitrypsin gene delivery reduces inflammation, increases T-regulatory cell population size and prevents islet allograft rejection. Mol Med 2011; 17:1000-11. [PMID: 21670848 DOI: 10.2119/molmed.2011.00145] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2011] [Accepted: 05/27/2011] [Indexed: 01/12/2023] Open
Abstract
Antiinflammatory clinical-grade, plasma-derived human α-1 antitrypsin (hAAT) protects islets from allorejection as well as from autoimmune destruction. hAAT also interferes with disease progression in experimental autoimmune encephalomyelitis (EAE) and in collagen-induced arthritis (CIA) mouse models. hAAT increases IL-1 receptor antagonist expression in human mononuclear cells and T-regulatory (Treg) cell population size in animal models. Clinical-grade hAAT contains plasma impurities, multiple hAAT isoforms and various states of inactive hAAT. We thus wished to establish islet-protective activities and effect on Treg cells of plasmid-derived circulating hAAT in whole animals. Islet function was assessed in mice that received allogeneic islet transplants after mice were given hydrodynamic tail-vein injection with pEF-hAAT, a previously described Epstein-Barr virus (EBV) plasmid construct containing the EBV nuclear antigen 1 (EBNA1) and the family of repeat EBNA1 binding site components (designated "EF") alongside the hAAT gene. Sera collected from hAAT-expressing mice were added to lipopolysaccharide (LPS)-stimulated macrophages to assess macrophage responsiveness. Also, maturation of peritoneal cells from hAAT-expressing mice was evaluated. hAAT-expressing mice accepted islet allografts (n = 11), whereas phosphate-buffered saline-injected animals (n = 11), as well as mice treated with truncated-hAAT-plasmid (n = 6) and untreated animals (n = 20) rapidly rejected islet allografts. In hAAT-expressing animals, local Treg cells were abundant at graft sites, and the IL-1 receptor antagonist was elevated in grafts and circulation. Sera from hAAT-expressing mice, but not control mice, inhibited macrophage responses. Finally, peritoneal cells from hAAT-expressing mice exhibited a semimature phenotype. We conclude that plasmid-derived circulating hAAT protects islet allografts from acute rejection, and human plasma impurities are unrelated to islet protection. Future studies may use this in vivo approach to examine the structure-function characteristics of the protective activities of AAT by manipulation of the hAAT plasmid.
Collapse
Affiliation(s)
- Galit Shahaf
- Department of Clinical Biochemistry, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | | | | | | | | |
Collapse
|
48
|
Igoillo-Esteve M, Gurzov EN, Eizirik DL, Cnop M. The transcription factor B-cell lymphoma (BCL)-6 modulates pancreatic {beta}-cell inflammatory responses. Endocrinology 2011; 152:447-56. [PMID: 21190961 DOI: 10.1210/en.2010-0790] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Type 1 diabetes is a chronic autoimmune disease with a strong inflammatory component. We have previously shown that expression of the transcriptional repressor B-cell lymphoma (BCL)-6 is very low in pancreatic β-cells, which may favor prolonged proinflammatory responses after exposure to the cytokines IL-1β and interferon γ. Here we investigated whether cytokine-induced inflammation and apoptosis can be prevented in β-cells by BCL-6 expression using plasmid, prolactin, and adenoviral approaches. The induction of mild or abundant BCL-6 expression in β-cells by prolactin or an adenoviral BCL-6 expression construct, respectively, reduced cytokine-induced inflammatory responses in a dose-dependent manner through inhibition of nuclear factor-κB activation. BCL-6 decreased Fas and inducible nitric oxide synthase expression and nitric oxide production, but it inhibited the expression of the antiapoptotic proteins Bcl-2 and JunB while increasing the expression of the proapoptotic death protein 5. The net result of these opposite effects was an augmentation of β-cell apoptosis. In conclusion, BCL-6 expression tones down the unrestrained cytokine-induced proinflammatory response of β-cells but it also favors gene networks leading to apoptosis. This suggests that cytokine-induced proinflammatory and proapoptotic signals can be dissociated in β-cells. Further understanding of these pathways may open new possibilities to improve β-cell survival in early type 1 diabetes or after transplantation.
Collapse
Affiliation(s)
- Mariana Igoillo-Esteve
- Laboratory of Experimental Medicine, Erasmus Hospital, Universite´ Libre de Bruxelles, 1070 Brussels, Belgium
| | | | | | | |
Collapse
|
49
|
Interference with islet-specific homing of autoreactive T cells: an emerging therapeutic strategy for type 1 diabetes. Drug Discov Today 2010; 15:531-9. [PMID: 20685342 DOI: 10.1016/j.drudis.2010.05.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2009] [Revised: 05/11/2010] [Accepted: 05/21/2010] [Indexed: 11/21/2022]
Abstract
Pathogenesis of type 1 diabetes involves the activation of autoimmune T cells, consequent homing of activated lymphocytes to the pancreatic islets and ensuing destruction of insulin-producing b cells. Interaction between activated lymphocytes and endothelial cells in the islets is the hallmark of the homing process. Initial adhesion, firm adhesion and diapedesis of lymphocytes are the three crucial steps involved in the homing process. Cell-surface receptors including integrins, selectins and hyaluronate receptor CD44 mediate the initial steps of homing. Diapedesis relies on a series of proteolytic events mediated by matrix metalloproteinases. Here, molecular mechanisms governing transendothelial migration of the diabetogenic effector cells are discussed and resulting pharmacological strategies are considered.
Collapse
|
50
|
Lawrence MC, Naziruddin B, Levy MF, Jackson A, McGlynn K. Calcineurin/nuclear factor of activated T cells and MAPK signaling induce TNF-{alpha} gene expression in pancreatic islet endocrine cells. J Biol Chem 2010; 286:1025-36. [PMID: 21059644 DOI: 10.1074/jbc.m110.158675] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Cytokines contribute to pancreatic islet inflammation, leading to impaired glucose homeostasis and diabetic diseases. A plethora of data shows that proinflammatory cytokines are produced in pancreatic islets by infiltrating mononuclear immune cells. Here, we show that pancreatic islet α cells and β cells express tumor necrosis factor-α (TNF-α) and other cytokines capable of promoting islet inflammation when exposed to interleukin-1β (IL-1β). Cytokine expression by β cells was dependent on calcineurin (CN)/nuclear factor of activated T cells (NFAT) and MAPK signaling. NFAT associated with the TNF-α promoter in response to stimuli and synergistically activated promoter activity with ATF2 and c-Jun. In contrast, the β-cell-specific transcriptional activator MafA could repress NFAT-mediated TNF-α gene expression whenever C/EBP-β was bound to the promoter. NFAT differentially regulated the TNF-α gene depending upon the expression and MAPK-dependent activation of interacting basic leucine zipper partners in β cells. Both p38 and JNK were required for induction of TNF-α mRNA and protein expression. Collectively, the data show that glucose and IL-1β can activate signaling pathways, which control induction and repression of cytokines in pancreatic endocrine cells. Thus, by these mechanisms, pancreatic β cells themselves may contribute to islet inflammation and their own immunological destruction in the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Michael C Lawrence
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas 75390, USA.
| | | | | | | | | |
Collapse
|