1
|
Vasilev G, Kokudeva M, Siliogka E, Padilla N, Shumnalieva R, Della-Morte D, Ricordi C, Mihova A, Infante M, Velikova T. T helper 17 cells and interleukin-17 immunity in type 1 diabetes: From pathophysiology to targeted immunotherapies. World J Diabetes 2025; 16:99936. [PMID: 40236846 PMCID: PMC11947927 DOI: 10.4239/wjd.v16.i4.99936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/06/2024] [Accepted: 02/07/2025] [Indexed: 02/28/2025] Open
Abstract
Type 1 diabetes (T1D) is a chronic organ-specific autoimmune disorder characterized by a progressive loss of the insulin-secreting pancreatic beta cells, which ultimately results in insulinopenia, hyperglycemia and lifelong need for exogenous insulin therapy. In the pathophysiological landscape of T1D, T helper 17 cells (Th17 cells) and their hallmark cytokine, interleukin (IL)-17, play pivotal roles from disease onset to disease progression. In this narrative mini-review, we discuss the dynamic interplay between Th17 cells and IL-17 in the context of T1D, providing insights into the underlying immunologic mechanisms contributing to the IL-17-immunity-mediated pancreatic beta-cell destruction. Furthermore, we summarized the main animal and clinical studies that investigated Th17- and IL-17-targeted interventions as promising immunotherapies able to alter the natural history of T1D.
Collapse
Affiliation(s)
- Georgi Vasilev
- Clinic of Neurology and Department of Emergency Medicine, UMHAT "Sv. Georgi", Plovdiv 4000, Bulgaria
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| | - Maria Kokudeva
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Medical University of Sofia, Sofia 1000, Bulgaria
| | - Elina Siliogka
- Faculty of Medicine, National and Kapodistrian University of Athens, Athens 11527, Attikí, Greece
| | - Nathalia Padilla
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Russka Shumnalieva
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
- Department of Rheumatology, Clinic of Rheumatology, University Hospital "St. Anna", Medical University-Sofia, Sofia 1612, Bulgaria
| | - David Della-Morte
- Department of Biomedicine and Prevention, Section of Clinical Nutrition and Nutrigenomics, University of Rome Tor Vergata, Rome 00133, Italy
| | - Camillo Ricordi
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | | | - Marco Infante
- Section of Diabetes & Metabolic Disorders, UniCamillus, Saint Camillus International University of Health Sciences, Rome 00131, Italy
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University St. Kliment Ohridski, Sofia 1407, Bulgaria
| |
Collapse
|
2
|
Nigi L, Pedace E, Dotta F, Sebastiani G. Neutrophils in Type 1 Diabetes: Untangling the Intricate Web of Pathways and Hypothesis. Biomolecules 2025; 15:505. [PMID: 40305198 PMCID: PMC12025241 DOI: 10.3390/biom15040505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Neutrophils are increasingly recognized as key contributors to the pathogenesis of Type 1 Diabetes (T1D), yet their precise mechanistic role in disease onset and progression remains incompletely understood. While these innate immune cells reside in pancreatic tissue and support tissue homeostasis under physiological conditions, they can also drive tissue damage by triggering innate immune responses and modulating inflammation. Within the inflammatory milieu, neutrophils establish complex, bidirectional interactions with various immune cells, including macrophages, dendritic cells, natural killer cells, and lymphocytes. Once activated, they may enhance the innate immune response through direct or indirect crosstalk with immune cells, antigen presentation, and β-cell destruction or dysfunction. These mechanisms underscore the multifaceted and dynamic role of neutrophils in T1D, shaped by their intricate immunological interactions. Further research into the diverse functional capabilities of neutrophils is crucial for uncovering novel aspects of their involvement in T1D, potentially revealing new therapeutic targets to modulate disease progression.
Collapse
Affiliation(s)
- Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| | - Erika Pedace
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
- Tuscany Centre for Precision Medicine, 53100 Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (L.N.); (E.P.); (G.S.)
- Fondazione Umberto Di Mario ONLUS, Toscana Life Sciences, 53100 Siena, Italy
| |
Collapse
|
3
|
Li R, Yang T, Zhang M, Ren K, Li J, Sato I, Yi SQ. A new histopathological phenomenon: Pancreatic islet cell loss in the elderly population. Dig Liver Dis 2024; 56:1039-1045. [PMID: 38065700 DOI: 10.1016/j.dld.2023.11.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 05/28/2024]
Abstract
BACKGROUND We observed the phenomenon of pancreatic islet cell loss (ICL) in our previous histopathological study. Multiple studies have reported that a decrease in β-cells is correlated with diabetes or chronic pancreatitis. Few studies have reported ICL in a healthy population. METHODS Thirty-three pancreatic tissue samples were obtained from cadavers (age: 65-104 years) who had never been diagnosed with any pancreatic diseases before death. The pancreatic body sections were used for an immunohistochemical study of pancreatic islet cells, and area calculations were performed using ImageJ to determine the degree of ICL and islet cell proportions. RESULTS The proportion of β-cells showed a downward trend as the degree of ICL increased (r=-0.414, P = 0.011), and the proportion of women with severe ICL was significantly higher than that of men with severe ICL (P = 0.016). The probability of severe ICL decreased with age in the population over 70 years of age (P = 0.069, linear correlation). Severe ICL may be associated with higher pancreatic intraepithelial neoplasia lesions (P = 0.059). CONCLUSION The phenomenon of ICL in the elderly population was mainly due to pancreatic β-cell reduction. It may be one of the direct causes of age-related diabetes.
Collapse
Affiliation(s)
- Rujia Li
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan
| | - Ting Yang
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan
| | - Mingshou Zhang
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan
| | - Ke Ren
- Faculty of Physical Education, Qu Jing Normal University, Yun Nan, China
| | - Jun Li
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Iwao Sato
- Department of Anatomy, Tokyo Medical University, Tokyo, Japan
| | - Shuang-Qin Yi
- Department of Frontier Health Sciences, Graduate School of Human Health Sciences, Tokyo Metropolitan University, 116-8551 Tokyo, Japan.
| |
Collapse
|
4
|
Liu YC, Liao YT, Chen VCH, Chen YL. Association Between Maternal Mood Disorders and Schizophrenia and the Risk of Type 1 Diabetes in Offspring: A Nationwide Cohort Study. Neuropsychiatr Dis Treat 2023; 19:2511-2518. [PMID: 38029045 PMCID: PMC10674753 DOI: 10.2147/ndt.s437430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023] Open
Abstract
Objective Our study aimed to determine whether mothers with bipolar disorder, major depressive disorder, schizophrenia, or schizoaffective disorder affected the risk of type 1 diabetes (T1D) in their offspring. Methods We conducted a nationwide cohort study by using data from Taiwan's National Health Insurance Research Database and the Maternal and Child Health Database from 2004 to 2018. A total of 2,556,640 mother-child pairs were identified. Cox proportional hazards models were used to compare the risk of T1D between children born to mothers with mood disorders and schizophrenia and those without. Results No significant difference in risk of T1D was observed between the offspring of mothers with major psychiatric disorders and those without (adjusted hazard ratio (aHR) of 0.86 with a 95% confidence interval (CI) of 0.58-1.24). In subgroup analysis, we found an aHR of 1.81 with a 95% CI of 0.83-3.82 in the maternal bipolar disorder on the risk of T1D in offspring and an aHR of 0.87 (95% CI: 0.59-1.25) in maternal major depressive disorder. In the schizophrenia/schizoaffective disorder group, aHR cannot be obtained due to lesser than three events in the analysis. Conclusion The risk of T1D in offspring of mothers with mood disorders and schizophrenia was not significant. However, children born to mothers with bipolar disorder may have a tendency to develop T1D. The relationship between maternal psychiatric disorders and the risk of T1D in offspring warrants further investigation in studies with longer follow-up periods.
Collapse
Affiliation(s)
- Yi-Chun Liu
- Department of Psychiatry, Changhua Christian Children’s Hospital, Changhua, 500, Taiwan
- Department of Psychiatry, Changhua Christian Hospital, Changhua, 500, Taiwan
- Department of Healthcare Administration, Asia University, Taichung, 413, Taiwan
- Department of Eldercare, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Yin-To Liao
- Department of Psychiatry, China Medical University Hospital, Taichung, 404, Taiwan
- China Medical University, Taichung, 406, Taiwan
| | - Vincent Chin-Hung Chen
- School of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
- Department of Psychiatry, Chiayi Chang Gung Memorial Hospital, Chiayi, 613, Taiwan
| | - Yi-Lung Chen
- Department of Healthcare Administration, Asia University, Taichung, 413, Taiwan
- Department of Psychology, Asia University, Taichung, 413, Taiwan
| |
Collapse
|
5
|
Harsini S, Rezaei N. Autoimmune diseases. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
6
|
Xu Q, Zhang X, Li T, Shao S. Exenatide regulates Th17/Treg balance via PI3K/Akt/FoxO1 pathway in db/db mice. Mol Med 2022; 28:144. [PMID: 36463128 PMCID: PMC9719171 DOI: 10.1186/s10020-022-00574-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 11/11/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND The T helper 17 (Th17)/T regulatory (Treg) cell imbalance is involved in the course of obesity and type 2 diabetes mellitus (T2DM). In the current study, the exact role of glucagon-like peptide-1 receptor agonist (GLP-1RA) exenatide on regulating the Th17/Treg balance and the underlying molecular mechanisms are investigated in obese diabetic mice model. METHODS Metabolic parameters were monitored in db/db mice treated with/without exenatide during 8-week study period. The frequencies of Th17 and Treg cells from peripheral blood and pancreas in db/db mice were assessed. The phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/Forkhead box O1 (FoxO1) pathway in Th17 and Treg cells from the spleens of male C57BL/6J mice was detected by western blotting. In addition, the expression of glucagon-like peptide-1 receptor (GLP-1R) in peripheral blood mononuclear cells (PBMCs) of male C57BL/6J mice was analyzed. RESULTS Exenatide treatment improved β-cell function and insulitis in addition to glucose, insulin sensitivity and weight. Increased Th17 and decreased Treg cells in peripheral blood were present as diabetes progressed while exenatide corrected this imbalance. Progressive IL-17 + T cell infiltration of pancreatic islets was alleviated by exenatide intervention. In vitro study showed no significant difference in the level of GLP-1R expression in PBMCs between control and palmitate (PA) groups. In addition, PA could promote Th17 but suppress Treg differentiation along with down-regulating the phosphorylation of PI3K/Akt/FoxO1, which was reversed by exenatide intervention. FoxO1 inhibitor AS1842856 could abrogate all these effects of exenatide against lipid stress. CONCLUSIONS Exenatide could restore systemic Th17/Treg balance via regulating FoxO1 pathway with the progression of diabetes in db/db mice. The protection of pancreatic β-cell function may be partially mediated by inhibiting Th17 cell infiltration into pancreatic islets, and the resultant alleviation of islet inflammation.
Collapse
Affiliation(s)
- Qinqin Xu
- grid.33199.310000 0004 0368 7223Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 430030 Hubei Province People’s Republic of China ,Branch of National Clinical Research Center for Metabolic Diseases, Hubei, People’s Republic of China
| | - Xiaoling Zhang
- grid.33199.310000 0004 0368 7223Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 430030 Hubei Province People’s Republic of China ,Branch of National Clinical Research Center for Metabolic Diseases, Hubei, People’s Republic of China
| | - Tao Li
- grid.33199.310000 0004 0368 7223Division of Ophthalmology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 430030 Hubei Province People’s Republic of China
| | - Shiying Shao
- grid.33199.310000 0004 0368 7223Division of Endocrinology, Tongji Hospital, Huazhong University of Science and Technology, Jiefang Road 1095, Wuhan, 430030 Hubei Province People’s Republic of China ,Branch of National Clinical Research Center for Metabolic Diseases, Hubei, People’s Republic of China
| |
Collapse
|
7
|
Fantuzzi F, Toivonen S, Schiavo AA, Chae H, Tariq M, Sawatani T, Pachera N, Cai Y, Vinci C, Virgilio E, Ladriere L, Suleiman M, Marchetti P, Jonas JC, Gilon P, Eizirik DL, Igoillo-Esteve M, Cnop M. In depth functional characterization of human induced pluripotent stem cell-derived beta cells in vitro and in vivo. Front Cell Dev Biol 2022; 10:967765. [PMID: 36060810 PMCID: PMC9428245 DOI: 10.3389/fcell.2022.967765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/06/2022] [Indexed: 01/05/2023] Open
Abstract
In vitro differentiation of human induced pluripotent stem cells (iPSCs) into beta cells represents an important cell source for diabetes research. Here, we fully characterized iPSC-derived beta cell function in vitro and in vivo in humanized mice. Using a 7-stage protocol, human iPSCs were differentiated into islet-like aggregates with a yield of insulin-positive beta cells comparable to that of human islets. The last three stages of differentiation were conducted with two different 3D culture systems, rotating suspension or static microwells. In the latter, homogeneously small-sized islet-like aggregates were obtained, while in rotating suspension size was heterogeneous and aggregates often clumped. In vitro function was assessed by glucose-stimulated insulin secretion, NAD(P)H and calcium fluctuations. Stage 7 aggregates slightly increased insulin release in response to glucose in vitro. Aggregates were transplanted under the kidney capsule of NOD-SCID mice to allow for further in vivo beta cell maturation. In transplanted mice, grafts showed glucose-responsiveness and maintained normoglycemia after streptozotocin injection. In situ kidney perfusion assays showed modulation of human insulin secretion in response to different secretagogues. In conclusion, iPSCs differentiated with equal efficiency into beta cells in microwells compared to rotating suspension, but the former had a higher experimental success rate. In vitro differentiation generated aggregates lacking fully mature beta cell function. In vivo, beta cells acquired the functional characteristics typical of human islets. With this technology an unlimited supply of islet-like organoids can be generated from human iPSCs that will be instrumental to study beta cell biology and dysfunction in diabetes.
Collapse
Affiliation(s)
- Federica Fantuzzi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium,Endocrinology and Metabolism, Department of Medicine and Surgery, University of Parma, Parma, Italy,*Correspondence: Miriam Cnop, ; Federica Fantuzzi,
| | - Sanna Toivonen
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrea Alex Schiavo
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Heeyoung Chae
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Mohammad Tariq
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Toshiaki Sawatani
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Pachera
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Ying Cai
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Chiara Vinci
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Enrico Virgilio
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Laurence Ladriere
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Jean-Christophe Jonas
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Patrick Gilon
- Institut de Recherche Expérimentale et Clinique, Pôle d’Endocrinologie, Diabète et Nutrition, Université Catholique de Louvain, Brussels, Belgium
| | - Décio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium,Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium,*Correspondence: Miriam Cnop, ; Federica Fantuzzi,
| |
Collapse
|
8
|
Niinistö S, Miettinen ME, Cuthbertson D, Honkanen J, Hakola L, Autio R, Erlund I, Arohonka P, Vuorela A, Härkönen T, Hyöty H, Krischer JP, Vaarala O, Knip M, Virtanen SM. Associations Between Serum Fatty Acids and Immunological Markers in Children Developing Islet Autoimmunity-The TRIGR Nested Case-Control Study. Front Immunol 2022; 13:858875. [PMID: 35693790 PMCID: PMC9175567 DOI: 10.3389/fimmu.2022.858875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 04/27/2022] [Indexed: 12/03/2022] Open
Abstract
Aims Altered immune functions as well as fatty acid intake and status have been associated with the development of type 1 diabetes. We aimed to study the relationship between fatty acids and immunological markers in young children with increased genetic risk for type 1 diabetes in order to define putative mechanisms related to development of islet autoimmunity. Methods Serum samples for fatty acid and immunological marker measurements were obtained in the Trial to Reduce IDDM in the Genetically at Risk (TRIGR) ancillary study (Divia) from children born between 2002 and 2007 in 15 countries. Case children (n = 95) were defined as having repeated positivity for at least two out of four diabetes-associated autoantibodies. For each case child, control children were selected matched for country and date of birth (n = 173). Serum fatty acids and immunological markers were measured from cord serum and at the age of 6 and 12 months. Spearman correlation coefficients were calculated between fatty acids and immunological markers. Results Correlations between circulating fatty acids and immunological markers were different in case children who developed islet autoimmunity than in control children already at birth continuing across the first year of life. In case children, saturated fatty acids (SFAs) showed stronger correlations with immunological markers, while in controls, polyunsaturated fatty acids (PUFAs) showed stronger correlations. Conclusions In cases, SFAs were associated with several immunological markers (CXCL10, IL-6, IL-9, IL-17, and CM-CSF) previously linked to the type 1 diabetes disease process. Findings indicate that fatty acids could have immunomodulatory potential in the early phase of the disease development, although causality between fatty acids and the immunological pathways remains to be explored. Trial registry number NCT00179777.
Collapse
Affiliation(s)
- Sari Niinistö
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Maija E. Miettinen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - David Cuthbertson
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Jarno Honkanen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Leena Hakola
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
- Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland
| | - Reija Autio
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
| | - Iris Erlund
- Department of Government Services, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Petra Arohonka
- Department of Government Services, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Arja Vuorela
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Taina Härkönen
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Heikki Hyöty
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Fimlab Laboratories, Pirkanmaa Hospital District, Tampere, Finland
| | - Jeffrey P. Krischer
- Health Informatics Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States
| | - Outi Vaarala
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Knip
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, University of Helsinki, Helsinki, Finland
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | - Suvi M. Virtanen
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Unit of Health Sciences, Faculty of Social Sciences, Tampere University, Tampere, Finland
- Tampere University Hospital, Research, Development and Innovation Center, Tampere, Finland
- Center for Child Health Research, Tampere University and Tampere University Hospital, Tampere, Finland
| | | |
Collapse
|
9
|
Xiao P, Takiishi T, Violato NM, Licata G, Dotta F, Sebastiani G, Marselli L, Singh SP, Sze M, Van Loo G, Dejardin E, Gurzov EN, Cardozo AK. NF-κB-inducing kinase (NIK) is activated in pancreatic β-cells but does not contribute to the development of diabetes. Cell Death Dis 2022; 13:476. [PMID: 35589698 PMCID: PMC9120028 DOI: 10.1038/s41419-022-04931-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/04/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
The transcription factor nuclear factor-κB (NF-κB) has a key role in the pathogenesis of diabetes and its complications. Although activation of the canonical NF-κB pathway in β-cells is generally deleterious, little is known about the role of the non-canonical NF-κB signalling and its main regulator, the NF-κB-inducing kinase (NIK), on pancreatic β-cell survival and function. Previous studies based on models of NIK overexpression in pancreatic islet cells showed that NIK induced either spontaneous β-cell death due to islet inflammation or glucose intolerance during diet-induced obesity (DIO) in mice. Therefore, NIK has been proposed as a potential target for diabetes therapy. However, no clear studies showed whether inhibition of NIK improves diabetes development. Here we show that genetic silencing of NIK in pancreatic β-cells neither modifies diabetes incidence nor inflammatory responses in a mouse model of immune-mediated diabetes. Moreover, NIK silencing in DIO mice did not influence body weight gain, nor glucose metabolism. In vitro studies corroborated the in vivo findings in terms of β-cell survival, function, and downstream gene regulation. Taken together, our data suggest that NIK activation is dispensable for the development of diabetes.
Collapse
Affiliation(s)
- Peng Xiao
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Tatiana Takiishi
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Natalia Moretti Violato
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Giada Licata
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Francesco Dotta
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
- Tuscany Centre for Precision Medicine (CReMeP), Siena, Italy
| | - Guido Sebastiani
- Department of Medical Sciences, Surgery and Neurosciences, University of Siena, Siena, Italy
- Fondazione Umberto Di Mario, c/o Toscana Life Sciences, Siena, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Sumeet Pal Singh
- Institute for Interdisciplinary Research in Human and Molecular Biology, Medical Faculty, Université libre de Bruxelles, Brussels, Belgium
| | - Mozes Sze
- Center for Inflammation Research, VIB, B-9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052, Ghent, Belgium
| | - Geert Van Loo
- Center for Inflammation Research, VIB, B-9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, B-9052, Ghent, Belgium
| | - Emmanuel Dejardin
- Laboratory of Molecular Immunology and Signal Transduction, GIGA-Insitute, ULiege, Liège, Belgium
| | - Esteban Nicolas Gurzov
- Signal Transduction and Metabolism Laboratory, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium
| | - Alessandra Kupper Cardozo
- Inflammation and Cell Death Signalling group, Laboratoire de Gastroentérologie Expérimental et Endotools, Université libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|
10
|
Rajendran S, Quesada-Masachs E, Zilberman S, Graef M, Kiosses WB, Chu T, Benkahla MA, Lee JHM, von Herrath M. IL-17 is expressed on beta and alpha cells of donors with type 1 and type 2 diabetes. J Autoimmun 2021; 123:102708. [PMID: 34358764 DOI: 10.1016/j.jaut.2021.102708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 12/30/2022]
Abstract
PURPOSE IL-17 is an important effector cytokine driving immune-mediated destruction in autoimmune diseases such as psoriasis. Blockade of the IL-17 pathway after the initiation of insulitis was effective in delaying or preventing the onset of type 1 diabetes (T1D) in rodent models. Expression of IL-17 transcripts in islets from a donor with recent-onset T1D has been reported, however, studies regarding IL-17 protein expression are lacking. We aimed to study whether IL-17 is being expressed in the islets of diabetic donors. METHODS We stained human pancreatic tissues from non-diabetic (n = 5), auto-antibody positive (aab+) (n = 5), T1D (n = 6) and T2D (n = 5) donors for IL-17, Insulin, and Glucagon, and for CD45 in selected cases. High resolution images were acquired with Zeiss laser scanning confocal microscope LSM780 and analyzed with Zen blue 2.3 software. Cases stained for CD45 were also acquired with widefield slide scanner and analyzed with QuPath software. RESULTS We observed a clear cytoplasmic staining for IL-17 in insulin-containing islets of donors with T1D and T2D, accounting for an average of 7.8 ± 8.4% and 14.9 ± 16.8% of total islet area, respectively. Both beta and alpha cells were sources of IL-17, but CD45+ cells were not a major source of IL-17 in those donors. Expression of IL-17 was reduced in islets of non-diabetic donors, aab+ donors and in insulin-deficient islets of donors with T1D. CONCLUSION Our finding that IL-17 is expressed in islets of donors with T1D or T2D is quite intriguing and warrants further mechanistic studies in human islets to understand the role of IL-17 in the context of metabolic and immune stress in beta cells.
Collapse
Affiliation(s)
| | | | | | | | | | - Tiffany Chu
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | | |
Collapse
|
11
|
Preventive and therapeutic significance of octreotide combined with lansoprazole on post-ERCP pancreatitis and its effect on serum amylase, inflammatory factors and immune function. Exp Ther Med 2021; 21:251. [PMID: 33603859 PMCID: PMC7851608 DOI: 10.3892/etm.2021.9682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/30/2020] [Indexed: 12/31/2022] Open
Abstract
The study aimed to investigate the preventive and therapeutic significance of octreotide combined with lansoprazole on post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis (PEP) and its effects on serum amylase (AMS), inflammatory factors and immune function. A total of 132 patients who underwent ERCP in Shaoxing People's Hospital (Shaoxing, China) were enrolled in the study and allocated into two groups: The study group (octreotide plus lansoprazole, 68 cases) and the control group (octreotide alone, 64 cases). The incidence of PEP and post-ERCP hyperamylasemia (PEH), the concentrations of serum AMS, interleukin-17 (IL-17) and tumor necrosis factor-α (TNF-α), as well as the T-lymphocyte population in peripheral blood were detected. The AMS levels in the study group were significantly lower than those in the control group at 6 and 24 h after treatment (P<0.001). The incidence of PEP and PEH, symptom disappearance time and hospital stay in the study group were significantly lower than those in the control group after treatment (P<0.05). The levels of IL-17 and TNF-α in the study group were significantly lower than those in the control group after treatment (all P<0.05). The percentage of CD3+, CD4+, CD8+ cells and the ratio of CD4+/CD8+ in the study group were significantly higher than those in the control group after treatment (all P<0.05). The results indicated that octreotide combined with lansoprazole reduces AMS levels and the incidence of PEP, alleviates inflammation and improves the immune function.
Collapse
|
12
|
Qiu AW, Cao X, Zhang WW, Liu QH. IL-17A is involved in diabetic inflammatory pathogenesis by its receptor IL-17RA. Exp Biol Med (Maywood) 2021; 246:57-65. [PMID: 32903039 PMCID: PMC7798001 DOI: 10.1177/1535370220956943] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 08/17/2020] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT The participation of interleukin (IL)-17A in diabetic pathogenesis is suggested in animal models of autoimmune diabetes and in patients with type 1 diabetes (T1D), but with some contradictory results. Particularly, evidence for a direct injury of IL-17A to pancreatic β cells is lacking. We showed that IL-17A deficiency alleviated diabetic signs including hyperglycemia, hypoinsulinemia, and inflammatory response in Ins2Akita (Akita) mice, a T1D model with spontaneous mutation in insulin 2 gene leading to β-cell apoptosis. IL-17A enhanced inflammatory reaction, oxidative stress, and cell apoptosis but attenuated insulin level in mouse insulin-producing MIN6 cells. IL-17A had also a synergistic destruction to MIN6 cells with streptozotocin (STZ), a pancreatic β-cell-specific cytotoxin. Blocking IL-17 receptor A (IL-17RA) reduced all these deleterious effects of IL-17A on MIN6 cells. The results demonstrate the role and the importance of IL-17A in T1D pathogenesis and suggest a potential therapeutic strategy for T1D targeting IL-17A and/or IL-17RA.
Collapse
Affiliation(s)
- Ao-Wang Qiu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Xin Cao
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Wei-Wei Zhang
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Qing-Huai Liu
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
13
|
Good Cop, Bad Cop: The Opposing Effects of Macrophage Activation State on Maintaining or Damaging Functional β-Cell Mass. Metabolites 2020; 10:metabo10120485. [PMID: 33256225 PMCID: PMC7761161 DOI: 10.3390/metabo10120485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 12/16/2022] Open
Abstract
Loss of functional β-cell mass is a hallmark of Type 1 and Type 2 Diabetes. Macrophages play an integral role in the maintenance or destruction of pancreatic β-cells. The effect of the macrophage β-cell interaction is dependent on the activation state of the macrophage. Macrophages can be activated across a spectrum, from pro-inflammatory to anti-inflammatory and tissue remodeling. The factors secreted by these differentially activated macrophages and their effect on β-cells define the effect on functional β-cell mass. In this review, the spectrum of macrophage activation is discussed, as are the positive and negative effects on β-cell survival, expansion, and function as well as the defined factors released from macrophages that impinge on functional β-cell mass.
Collapse
|
14
|
Sphingolipids in Type 1 Diabetes: Focus on Beta-Cells. Cells 2020; 9:cells9081835. [PMID: 32759843 PMCID: PMC7465050 DOI: 10.3390/cells9081835] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/28/2022] Open
Abstract
Type 1 diabetes (T1DM) is a chronic autoimmune disease, with a strong genetic background, leading to a gradual loss of pancreatic beta-cells, which secrete insulin and control glucose homeostasis. Patients with T1DM require life-long substitution with insulin and are at high risk for development of severe secondary complications. The incidence of T1DM has been continuously growing in the last decades, indicating an important contribution of environmental factors. Accumulating data indicates that sphingolipids may be crucially involved in T1DM development. The serum lipidome of T1DM patients is characterized by significantly altered sphingolipid composition compared to nondiabetic, healthy probands. Recently, several polymorphisms in the genes encoding the enzymatic machinery for sphingolipid production have been identified in T1DM individuals. Evidence gained from studies in rodent islets and beta-cells exposed to cytokines indicates dysregulation of the sphingolipid biosynthetic pathway and impaired function of several sphingolipids. Moreover, a number of glycosphingolipids have been suggested to act as beta-cell autoantigens. Studies in animal models of autoimmune diabetes, such as the Non Obese Diabetic (NOD) mouse and the LEW.1AR1-iddm (IDDM) rat, indicate a crucial role of sphingolipids in immune cell trafficking, islet infiltration and diabetes development. In this review, the up-to-date status on the findings about sphingolipids in T1DM will be provided, the under-investigated research areas will be identified and perspectives for future studies will be given.
Collapse
|
15
|
Catterall T, Fynch S, Kay TWH, Thomas HE, Sutherland APR. IL-17F induces inflammation, dysfunction and cell death in mouse islets. Sci Rep 2020; 10:13077. [PMID: 32753746 PMCID: PMC7403586 DOI: 10.1038/s41598-020-69805-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/16/2020] [Indexed: 01/10/2023] Open
Abstract
Type 17 immune responses, typified by the production of the cytokines IL-17A and IL-17F, have been implicated in the development of type 1 diabetes in animal models and human patients, however the underlying pathogenic mechanisms have not been clearly elucidated. While previous studies show that IL-17A enhances inflammatory gene expression and cell death in mouse β-cells and human islets, the function of IL-17F in pancreatic β-cells is completely untested to date. Here we show that IL-17F exhibits potent pathogenic effects in mouse β-cell lines and islets. IL-17F signals via the IL-17RA and -RC subunits in β-cells and in combination with other inflammatory cytokines induces expression of chemokine transcripts, suppresses the expression of β-cell identity genes and impairs glucose stimulated insulin secretion. Further IL-17F induces cell death in primary mouse islets. This occurs via Jnk, p38 and NF-κB dependent induction of Nos2 and is completely ablated in the presence of an inducible nitric oxide synthase (iNOS) inhibitor. Together these data indicate that IL-17F possesses similar pathogenic activities to IL-17A in mouse β-cell lines and islets and is likely to be a type 17 associated pathogenic factor in type 1 diabetes.
Collapse
Affiliation(s)
- Tara Catterall
- St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Melbourne, VIC, 3065, Australia
| | - Stacey Fynch
- St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Melbourne, VIC, 3065, Australia
| | - Thomas W H Kay
- St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Helen E Thomas
- St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Melbourne, VIC, 3065, Australia.,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| | - Andrew P R Sutherland
- St. Vincent's Institute of Medical Research, 9 Princes Street, Fitzroy, Melbourne, VIC, 3065, Australia. .,Department of Medicine, St. Vincent's Hospital, The University of Melbourne, Fitzroy, VIC, Australia.
| |
Collapse
|
16
|
Maddaloni E, Moretti C, Mignogna C, Buzzetti R. Adult-onset autoimmune diabetes in 2020: An update. Maturitas 2020; 137:37-44. [DOI: 10.1016/j.maturitas.2020.04.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/18/2020] [Accepted: 04/21/2020] [Indexed: 12/11/2022]
|
17
|
Jörns A, Ishikawa D, Teraoku H, Yoshimoto T, Wedekind D, Lenzen S. Remission of autoimmune diabetes by anti-TCR combination therapies with anti-IL-17A or/and anti-IL-6 in the IDDM rat model of type 1 diabetes. BMC Med 2020; 18:33. [PMID: 32106855 PMCID: PMC7047363 DOI: 10.1186/s12916-020-1503-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The cytokine IL-17 is a key player in autoimmune processes, while the cytokine IL-6 is responsible for the chronification of inflammation. However, their roles in type 1 diabetes development are still unknown. METHODS Therefore, therapies for 5 days with anti-IL-17A or anti-IL-6 in combination with a T cell-specific antibody, anti-TCR, or in a triple combination were initiated immediately after disease manifestation to reverse the diabetic metabolic state in the LEW.1AR1-iddm (IDDM) rat, a model of human type 1 diabetes. RESULTS Monotherapies with anti-IL-6 or anti-IL-17 showed no sustained anti-diabetic effects. Only the combination therapy of anti-TCR with anti-IL-6 or anti-IL-17 at starting blood glucose concentrations up to 12 mmol/l restored normoglycaemia. The triple antibody combination therapy was effective even up to very high initial blood glucose concentrations (17 mmol/l). The β cell mass was raised to values of around 6 mg corresponding to those of normoglycaemic controls. In parallel, the apoptosis rate of β cells was reduced and the proliferation rate increased as well as the islet immune cell infiltrate was strongly reduced in double and abolished in triple combination therapies. CONCLUSIONS The anti-TCR combination therapy with anti-IL-17 preferentially raised the β cell mass as a result of β cell proliferation while anti-IL-6 strongly reduced β cell apoptosis and the islet immune cell infiltrate with a modest increase of the β cell mass only. The triple combination therapy achieved both goals in a complimentary anti-autoimmune and anti-inflammatory action resulting in sustained normoglycaemia with normalized serum C-peptide concentrations.
Collapse
Affiliation(s)
- Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
| | - Daichi Ishikawa
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Institute of Experimental Diabetes Research, Hannover Medical School, 30623, Hannover, Germany
| | - Hiroki Teraoku
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Institute of Experimental Diabetes Research, Hannover Medical School, 30623, Hannover, Germany
| | - Toshiaki Yoshimoto
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany
- Institute of Experimental Diabetes Research, Hannover Medical School, 30623, Hannover, Germany
| | - Dirk Wedekind
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Hannover, Germany.
- Institute of Experimental Diabetes Research, Hannover Medical School, 30623, Hannover, Germany.
| |
Collapse
|
18
|
In Vivo and In Vitro Models of Diabetes: A Focus on Pregnancy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1307:553-576. [PMID: 32504388 DOI: 10.1007/5584_2020_536] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Diabetes in pregnancy is associated with an increased risk of poor outcomes, both for the mother and her offspring. Although clinical and epidemiological studies are invaluable to assess these outcomes and the effectiveness of potential treatments, there are certain ethical and practical limitations to what can be assessed in human studies.Thus, both in vivo and in vitro models can aid us in the understanding of the mechanisms behind these complications and, in the long run, towards their prevention and treatment. This review summarizes the existing animal and cell models used to mimic diabetes, with a specific focus on the intrauterine environment. Summary of this review.
Collapse
|
19
|
Grieco FA, Schiavo AA, Brozzi F, Juan-Mateu J, Bugliani M, Marchetti P, Eizirik DL. The miRNAs miR-211-5p and miR-204-5p modulate ER stress in human beta cells. J Mol Endocrinol 2019; 63:139-149. [PMID: 31277072 PMCID: PMC6938585 DOI: 10.1530/jme-19-0066] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Accepted: 07/05/2019] [Indexed: 12/16/2022]
Abstract
miRNAs are a class of small non-coding RNAs that regulate gene expression. Type 1 diabetes is an autoimmune disease characterized by insulitis (islets inflammation) and pancreatic beta cell destruction. The pro-inflammatory cytokines interleukin 1 beta (IL1B) and interferon gamma (IFNG) are released during insulitis and trigger endoplasmic reticulum (ER) stress and expression of pro-apoptotic members of the BCL2 protein family in beta cells, thus contributing to their death. The nature of miRNAs that regulate ER stress and beta cell apoptosis remains to be elucidated. We have performed a global miRNA expression profile on cytokine-treated human islets and observed a marked downregulation of miR-211-5p. By real-time PCR and Western blot analysis, we confirmed cytokine-induced changes in the expression of miR-211-5p and the closely related miR-204-5p and downstream ER stress related genes in human beta cells. Blocking of endogenous miRNA-211-5p and miR-204-5p by the same inhibitor (it is not possible to block separately these two miRs) increased human beta cell apoptosis, as measured by Hoechst/propidium Iodide staining and by determination of cleaved caspase-3 activation. Interestingly, miRs-211-5p and 204-5p regulate the expression of several ER stress markers downstream of PERK, particularly the pro-apoptotic protein DDIT3 (also known as CHOP). Blocking CHOP expression by a specific siRNA partially prevented the increased apoptosis observed following miR-211-5p/miR-204-5p inhibition. These observations identify a novel crosstalk between miRNAs, ER stress and beta cell apoptosis in early type 1 diabetes.
Collapse
Affiliation(s)
- Fabio Arturo Grieco
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Andrea Alex Schiavo
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Flora Brozzi
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Jonas Juan-Mateu
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Marco Bugliani
- 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
| | - Décio L. Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
- Corresponding author: Dr. Décio L. Eizirik, ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium, 808 Route de Lennik, 1070 Brussels, Belgium, Phone: +32 2 555 6242, Fax: +32 2 555 6239,
| |
Collapse
|
20
|
Ikumi K, Odanaka M, Shime H, Imai M, Osaga S, Taguchi O, Nishida E, Hemmi H, Kaisho T, Morita A, Yamazaki S. Hyperglycemia Is Associated with Psoriatic Inflammation in Both Humans and Mice. J Invest Dermatol 2019; 139:1329-1338.e7. [PMID: 30776434 DOI: 10.1016/j.jid.2019.01.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 01/16/2019] [Accepted: 01/25/2019] [Indexed: 12/29/2022]
Abstract
Chronic low-grade inflammation can cause several metabolic syndromes. Patients with psoriasis, a chronic immunological skin inflammation, often develop diabetes. However, it is not clear to date how psoriasis leads to, or is correlated with, glucose intolerance. Here, we investigate whether psoriasis itself is correlated with hyperglycemia in humans and mice. In patients, the severity of psoriasis was correlated with high blood glucose levels, and treatment of psoriasis by phototherapy improved insulin secretion. Imiquimod-induced systemic and cutaneous inflammation in mice, with features of human psoriasis, also resulted in hyperglycemia. Although it should be determined if psoriasis-like cutaneous inflammation alone can induce hyperglycemia, imiquimod-treated mice showed impairment of insulin secretion without significant islet inflammation. Administration of anti-IL-17A monoclonal antibody improved hyperglycemia in patients with psoriasis and imiquimod-treated mice with psoriasiform features. These results suggest that hyperglycemia is highly associated with psoriasis, mainly through IL-17.
Collapse
Affiliation(s)
- Kyoko Ikumi
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan; Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Mizuyu Odanaka
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroaki Shime
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Masaki Imai
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoshi Osaga
- Clinical Research Management Center, Nagoya City University Hospital, Nagoya, Japan
| | - Osamu Taguchi
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Emi Nishida
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroaki Hemmi
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan; Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Tsuneyasu Kaisho
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan; Laboratory for Immune Regulation, World Premier International Research Center Initiative, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Sayuri Yamazaki
- Department of Immunology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| |
Collapse
|
21
|
Bugliani M, Syed F, Paula FMM, Omar BA, Suleiman M, Mossuto S, Grano F, Cardarelli F, Boggi U, Vistoli F, Filipponi F, De Simone P, Marselli L, De Tata V, Ahren B, Eizirik DL, Marchetti P. DPP-4 is expressed in human pancreatic beta cells and its direct inhibition improves beta cell function and survival in type 2 diabetes. Mol Cell Endocrinol 2018; 473:186-193. [PMID: 29409957 DOI: 10.1016/j.mce.2018.01.019] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 12/20/2017] [Accepted: 01/29/2018] [Indexed: 11/26/2022]
Abstract
It has been reported that the incretin system, including regulated GLP-1 secretion and locally expressed DPP-4, is present in pancreatic islets. In this study we comprehensively evaluated the expression and role of DPP-4 in islet alpha and beta cells from non-diabetic (ND) and type 2 diabetic (T2D) individuals, including the effects of its inhibition on beta cell function and survival. Isolated islets were prepared from 25 ND and 18 T2D organ donors; studies were also performed with the human insulin-producing EndoC-βH1 cells. Morphological (including confocal microscopy), ultrastructural (electron microscopy, EM), functional (glucose-stimulated insulin secretion), survival (EM and nuclear dyes) and molecular (RNAseq, qPCR and western blot) studies were performed under several different experimental conditions. DPP-4 co-localized with glucagon and was also expressed in human islet insulin-containing cells. Furthermore, DPP-4 was expressed in EndoC-βH1 cells. The proportions of DPP-4 positive alpha and beta cells and DPP-4 gene expression were significantly lower in T2D islets. A DPP-4 inhibitor protected ND human beta cells and EndoC-βH1 cells against cytokine-induced toxicity, which was at least in part independent from GLP1 and associated with reduced NFKB1 expression. Finally, DPP-4 inhibition augmented glucose-stimulated insulin secretion, reduced apoptosis and improved ultrastructure in T2D beta cells. These results demonstrate the presence of DPP-4 in human islet alpha and beta cells, with reduced expression in T2D islets, and show that DPP-4 inhibition has beneficial effects on human ND and T2D beta cells. This suggests that DPP-4, besides playing a role in incretin effects, directly affects beta cell function and survival.
Collapse
Affiliation(s)
- Marco Bugliani
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Farooq Syed
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Flavia M M Paula
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Bilal A Omar
- Lund University, Department of Clinical Sciences, Lund Sweden
| | - Mara Suleiman
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Sandra Mossuto
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Francesca Grano
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Francesco Cardarelli
- National Enterprise for NanoScience and NanoTechnology (NEST), CNR and Scuola Normale Superiore, Pisa, Italy
| | - Ugo Boggi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Fabio Vistoli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Franco Filipponi
- Department of Surgical Pathology, Medicine, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - Paolo De Simone
- Department of Surgical Pathology, Medicine, Molecular and Critical Area, University of Pisa, Pisa, Italy
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy
| | - Vincenzo De Tata
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Bo Ahren
- Lund University, Department of Clinical Sciences, Lund Sweden
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Cell Laboratory, University of Pisa, Pisa, Italy.
| |
Collapse
|
22
|
Intestinal Immunomodulatory Cells (T Lymphocytes): A Bridge between Gut Microbiota and Diabetes. Mediators Inflamm 2018; 2018:9830939. [PMID: 29713241 PMCID: PMC5866888 DOI: 10.1155/2018/9830939] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/10/2018] [Accepted: 01/28/2018] [Indexed: 12/20/2022] Open
Abstract
Diabetes mellitus (DM) is one of the most familiar chronic diseases threatening human health. Recent studies have shown that the development of diabetes is closely related to an imbalance of the gut microbiota. Accordingly, there is increasing interest in how changes in the gut microbiota affect diabetes and its underlying mechanisms. Immunomodulatory cells play important roles in maintaining the normal functioning of the human immune system and in maintaining homeostasis. Intestinal immunomodulatory cells (IICs) are located in the intestinal mucosa and are regarded as an intermediary by which the gut microbiota affects physiological and pathological properties. Diabetes can be regulated by IICs, which act as a bridge linking the gut microbiota and DM. Understanding this bridge role of IICs may clarify the mechanisms by which the gut microbiota contributes to DM. Based on recent research, we summarize this process, thereby providing a basis for further studies of diabetes and other similar immune-related diseases.
Collapse
|
23
|
Pinto LC, Cerqueira-Lima AT, Suzarth SDS, de Souza R, Tosta BR, da Silva HB, Pires ADO, Queiroz GDA, Teixeira TO, Dourado KMC, Costa VO, Baqueiro V, Oliveira DP, Brandão HR, de Souza CO, Druzian JI, Medeiros KCDP, Dantas Alves CDA, Lopes MV, Figueiredo CAV. Anonna muricata L. (soursop) seed oil improves type 1 diabetes parameters in vivo and in vitro. PHARMANUTRITION 2018. [DOI: 10.1016/j.phanu.2017.11.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
24
|
Tsonkova VG, Sand FW, Wolf XA, Grunnet LG, Kirstine Ringgaard A, Ingvorsen C, Winkel L, Kalisz M, Dalgaard K, Bruun C, Fels JJ, Helgstrand C, Hastrup S, Öberg FK, Vernet E, Sandrini MPB, Shaw AC, Jessen C, Grønborg M, Hald J, Willenbrock H, Madsen D, Wernersson R, Hansson L, Jensen JN, Plesner A, Alanentalo T, Petersen MBK, Grapin-Botton A, Honoré C, Ahnfelt-Rønne J, Hecksher-Sørensen J, Ravassard P, Madsen OD, Rescan C, Frogne T. The EndoC-βH1 cell line is a valid model of human beta cells and applicable for screenings to identify novel drug target candidates. Mol Metab 2018; 8:144-157. [PMID: 29307512 PMCID: PMC5985049 DOI: 10.1016/j.molmet.2017.12.007] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 12/12/2017] [Accepted: 12/13/2017] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE To characterize the EndoC-βH1 cell line as a model for human beta cells and evaluate its beta cell functionality, focusing on insulin secretion, proliferation, apoptosis and ER stress, with the objective to assess its potential as a screening platform for identification of novel anti-diabetic drug candidates. METHODS EndoC-βH1 was transplanted into mice for validation of in vivo functionality. Insulin secretion was evaluated in cells cultured as monolayer and as pseudoislets, as well as in diabetic mice. Cytokine induced apoptosis, glucolipotoxicity, and ER stress responses were assessed. Beta cell relevant mRNA and protein expression were investigated by qPCR and antibody staining. Hundreds of proteins or peptides were tested for their effect on insulin secretion and proliferation. RESULTS Transplantation of EndoC-βH1 cells restored normoglycemia in streptozotocin induced diabetic mice. Both in vitro and in vivo, we observed a clear insulin response to glucose, and, in vitro, we found a significant increase in insulin secretion from EndoC-βH1 pseudoislets compared to monolayer cultures for both glucose and incretins. Apoptosis and ER stress were inducible in the cells and caspase 3/7 activity was elevated in response to cytokines, but not affected by the saturated fatty acid palmitate. By screening of various proteins and peptides, we found Bombesin (BB) receptor agonists and Pituitary Adenylate Cyclase-Activating Polypeptides (PACAP) to significantly induce insulin secretion and the proteins SerpinA6, STC1, and APOH to significantly stimulate proliferation. ER stress was readily induced by Tunicamycin and resulted in a reduction of insulin mRNA. Somatostatin (SST) was found to be expressed by 1% of the cells and manipulation of the SST receptors was found to significantly affect insulin secretion. CONCLUSIONS Overall, the EndoC-βH1 cells strongly resemble human islet beta cells in terms of glucose and incretin stimulated insulin secretion capabilities. The cell line has an active cytokine induced caspase 3/7 apoptotic pathway and is responsive to ER stress initiation factors. The cells' ability to proliferate can be further increased by already known compounds as well as by novel peptides and proteins. Based on its robust performance during the functionality assessment assays, the EndoC-βH1 cell line was successfully used as a screening platform for identification of novel anti-diabetic drug candidates.
Collapse
Affiliation(s)
- Violeta Georgieva Tsonkova
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark; University of Copenhagen, Department of Biomedical Sciences, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Fredrik Wolfhagen Sand
- Novo Nordisk A/S, Diabetes Research, GLP-1 & T2D Pharmacology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Xenia Asbæk Wolf
- Novo Nordisk A/S, Diabetes Research, GLP-1 & T2D Pharmacology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Lars Groth Grunnet
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Anna Kirstine Ringgaard
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark; University of Copenhagen, Department of Biomedical Sciences, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Camilla Ingvorsen
- Novo Nordisk A/S, Diabetes Research, Histology & Imaging, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Louise Winkel
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Mark Kalisz
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Kevin Dalgaard
- Novo Nordisk A/S, Diabetes Research, GLP-1 & T2D Pharmacology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Christine Bruun
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Johannes Josef Fels
- Novo Nordisk A/S, Discovery Biology & Technology, Research Bioanalysis, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Charlotte Helgstrand
- Novo Nordisk A/S, Protein Engineering, Expression Technologies 1, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Sven Hastrup
- Novo Nordisk A/S, Protein Engineering, Expression Technologies 1, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Fredrik Kryh Öberg
- Novo Nordisk A/S, Protein Engineering, Expression Technologies 1, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Erik Vernet
- Novo Nordisk Research Center Seattle Inc., Protein Engineering, NNRC Seattle, Inc., 530 Fairview Avenue North, 98109, Seattle, WA, USA
| | | | - Allan Christian Shaw
- Novo Nordisk A/S, Protein Engineering, Characterisation & Modelling Technology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Carsten Jessen
- Novo Nordisk A/S, Protein Engineering, Protein & Peptide Chemistry 2, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Mads Grønborg
- Novo Nordisk A/S, Discovery Biology & Technology, Discovery ADME, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Jacob Hald
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Hanni Willenbrock
- Novo Nordisk A/S, Discovery Biology & Technology, Bioinformatics, Maaloev, Denmark
| | - Dennis Madsen
- Novo Nordisk A/S, Discovery Biology & Technology, Bioinformatics, Maaloev, Denmark
| | | | - Lena Hansson
- Intomics A/S, Lottenborgvej 26, DK-2800, Lyngby, Denmark; Novo Nordisk Pharma Ltd., Research Centre Oxford, Bioinformatics, Novo Nordisk Ltd., 3 City Place Beehive Ring Road, Gatwick, RH6 0PA, West Sussex, United Kingdom
| | - Jan Nygaard Jensen
- Novo Nordisk Pharma Ltd., Research Centre Oxford, Bioinformatics, Novo Nordisk Ltd., 3 City Place Beehive Ring Road, Gatwick, RH6 0PA, West Sussex, United Kingdom
| | - Annette Plesner
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Tomas Alanentalo
- Novo Nordisk A/S, Diabetes Research, Histology & Imaging, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Maja Borup Kjær Petersen
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark; University of Copenhagen, DanStem, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Anne Grapin-Botton
- University of Copenhagen, DanStem, Blegdamsvej 3B, DK-2200, Copenhagen, Denmark
| | - Christian Honoré
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Jonas Ahnfelt-Rønne
- Novo Nordisk A/S, Diabetes Research, Histology & Imaging, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Jacob Hecksher-Sørensen
- Novo Nordisk A/S, Diabetes Research, Histology & Imaging, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Philippe Ravassard
- Institut du cerveau et de la moelle (ICM) - Hôpital Pitié-Salpêtrière, Boulevard de l'Hôpital, Sorbonne Universités, Inserm, CNRS, UPMC Univ, Paris 06, Paris, France
| | - Ole D Madsen
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Claude Rescan
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark
| | - Thomas Frogne
- Novo Nordisk A/S, Diabetes Research, Department of Islet & Stem Cell Biology, Novo Nordisk Park, 2760, Maaloev, Denmark.
| |
Collapse
|
25
|
Andreone L, Gimeno ML, Perone MJ. Interactions Between the Neuroendocrine System and T Lymphocytes in Diabetes. Front Endocrinol (Lausanne) 2018; 9:229. [PMID: 29867762 PMCID: PMC5966545 DOI: 10.3389/fendo.2018.00229] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Accepted: 04/20/2018] [Indexed: 12/16/2022] Open
Abstract
It is well established that there is a fine-tuned bidirectional communication between the immune and neuroendocrine tissues in maintaining homeostasis. Several types of immune cells, hormones, and neurotransmitters of different chemical nature are involved as communicators between organs. Apart of being key players of the adaptive arm of the immune system, it has been recently described that T lymphocytes are involved in the modulation of metabolism of several tissues in health and disease. Diabetes may result mainly from lack of insulin production (type 1 diabetes) or insufficient insulin and insulin resistance (type 2 diabetes), both influenced by genetic and environmental components. Herein, we discuss accumulating data regarding the role of the adaptive arm of the immune system in the pathogenesis of diabetes; including the action of several hormones and neurotransmitters influencing on central and peripheral T lymphocytes development and maturation, particularly under the metabolic burden triggered by diabetes. In addition, we comment on the role of T-effector lymphocytes in adipose and liver tissues during diabetes, which together enhances pancreatic β-cell stress aggravating the disease.
Collapse
|
26
|
Chou FC, Chen HY, Chen HH, Lin GJ, Lin SH, Sytwu HK. Differential modulation of IL-12 family cytokines in autoimmune islet graft failure in mice. Diabetologia 2017; 60:2409-2417. [PMID: 28929188 DOI: 10.1007/s00125-017-4418-9] [Citation(s) in RCA: 4] [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: 04/06/2017] [Accepted: 07/06/2017] [Indexed: 01/05/2023]
Abstract
AIMS/HYPOTHESIS The relative contribution of T helper (Th)1 and Th17 cells in graft rejection is inconclusive, on the basis of evidence provided by different T cell-related cytokine-deficient animal models and graft types. METHODS We used novel antigen-presenting-cell-specific Il-12p35 (also known as Il12a)-knockout (KO), IL-23p19-knockdown (KD) and IL-27p28-KD strategies to investigate T cell differentiation in islet graft rejection. RESULTS In vitro dendritic cell-T cell coculture experiments revealed that dendritic cells from Il-12p35-KO and IL-23p19-KD mice showed reduced ability to stimulate IFN-γ and IL-17 production in T cells, respectively. To further explore the T cell responses in islet graft rejection, we transplanted islets into streptozotocin-induced diabetic NOD/severe combined immunodeficiency (SCID) recipient mice with IL-12-, IL-23-, or IL-27-deficient backgrounds and then challenged them with NOD.BDC2.5 T cells. The survival of islet grafts was significantly prolonged in Il-12p35-KO and IL-23p19-KD recipients compared with the control recipients. T cell infiltrations and Th1 cell populations were also decreased in the grafts, correlating with prolonged graft survival. CONCLUSIONS/INTERPRETATION Our results suggest that IL-12 and IL-23 promote and/or maintain Th1 cell-mediated islet graft rejection. Thus, blockade of IL-12 and IL-23 might act as therapeutic strategies for reducing rejection responses.
Collapse
Affiliation(s)
- Feng-Cheng Chou
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, 161, Section 6, MinChuan East Road, Neihu, Taipei 114, Taiwan
| | - Heng-Yi Chen
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Hsin-Hui Chen
- Department of Medicine, National Defense Medical Center, Taipei, Taiwan
| | - Gu-Jiun Lin
- Department of Biology and Anatomy, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Hua Lin
- Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Huey-Kang Sytwu
- Department and Graduate Institute of Microbiology and Immunology, National Defense Medical Center, 161, Section 6, MinChuan East Road, Neihu, Taipei 114, Taiwan.
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
27
|
Meyerovich K, Violato NM, Fukaya M, Dirix V, Pachera N, Marselli L, Marchetti P, Strasser A, Eizirik DL, Cardozo AK. MCL-1 Is a Key Antiapoptotic Protein in Human and Rodent Pancreatic β-Cells. Diabetes 2017; 66:2446-2458. [PMID: 28667119 DOI: 10.2337/db16-1252] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 06/20/2017] [Indexed: 11/13/2022]
Abstract
Induction of endoplasmic reticulum stress and activation of the intrinsic apoptotic pathway is widely believed to contribute to β-cell death in type 1 diabetes (T1D). MCL-1 is an antiapoptotic member of the BCL-2 protein family, whose depletion causes apoptosis in rodent β-cells in vitro. Importantly, decreased MCL-1 expression was observed in islets from patients with T1D. We report here that MCL-1 downregulation is associated with cytokine-mediated killing of human β-cells, a process partially prevented by MCL-1 overexpression. By generating a β-cell-specific Mcl-1 knockout mouse strain (βMcl-1KO), we observed that, surprisingly, MCL-1 ablation does not affect islet development and function. β-Cells from βMcl-1KO mice were, however, more susceptible to cytokine-induced apoptosis. Moreover, βMcl-1KO mice displayed higher hyperglycemia and lower pancreatic insulin content after multiple low-dose streptozotocin treatment. We found that the kinase GSK3β, the E3 ligases MULE and βTrCP, and the deubiquitinase USP9x regulate cytokine-mediated MCL-1 protein turnover in rodent β-cells. Our results identify MCL-1 as a critical prosurvival protein for preventing β-cell death and clarify the mechanisms behind its downregulation by proinflammatory cytokines. Development of strategies to prevent MCL-1 loss in the early stages of T1D may enhance β-cell survival and thereby delay or prevent disease progression.
Collapse
Affiliation(s)
- Kira Meyerovich
- Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Natalia M Violato
- Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Makiko Fukaya
- Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Violette Dirix
- Laboratory of Vaccinology and Mucosal Immunity, Université Libre de Bruxelles, Brussels, Belgium
| | - Nathalie Pachera
- Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Islet Laboratory, University of Pisa, Pisa, Italy
| | - Andreas Strasser
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Decio L Eizirik
- Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Alessandra K Cardozo
- Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
28
|
Insuela DBR, Carvalho VF. Glucagon and glucagon-like peptide-1 as novel anti-inflammatory and immunomodulatory compounds. Eur J Pharmacol 2017; 812:64-72. [PMID: 28688914 DOI: 10.1016/j.ejphar.2017.07.015] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 07/04/2017] [Accepted: 07/05/2017] [Indexed: 12/25/2022]
Abstract
Glucagon and glucagon-like peptide-1 (GLP-1) are polypeptide hormones that are produced by pancreatic α-cells and the intestine, respectively, whose main function is to control glucose homeostasis. The glucagon and GLP-1 levels are imbalanced in diabetes. Furthermore, type 1 diabetic patients and animals present with a diminished inflammatory response, which is related to some morbidities of diabetes, such as a higher incidence of infectious diseases, including sepsis. The focus of this review is to briefly summarize the state of the art concerning the effects of glucagon and GLP-1 on the inflammatory response. Here, we propose that glucagon and GLP-1 have anti-inflammatory properties, making them possible prototypes for the design and synthesis of new compounds to treat inflammatory diseases. In addition, glucagon, GLP-1 or their analogues or new derivatives may not only be important for managing inflammatory diseases but may also have the therapeutic potential to prevent, cure or ameliorate diabetes in patients by counteracting the deleterious effects of pro-inflammatory cytokines on the function and viability of pancreatic β-cells. In addition, GLP-1, its analogues or drugs that inhibit GLP-1 metabolism may have a doubly beneficial effect in diabetic patients by inhibiting the inflammatory response and reducing glycaemia.
Collapse
Affiliation(s)
- Daniella B R Insuela
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n°4365, Manguinhos, CEP 21040-360 Rio de Janeiro, Brazil
| | - Vinicius F Carvalho
- Laboratório de Inflamação, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Av. Brasil, n°4365, Manguinhos, CEP 21040-360 Rio de Janeiro, Brazil; National Institute of Science and Technology on Neuroimmunomodulation (INCT-NIM), Brazil.
| |
Collapse
|
29
|
Badal D, Kumar R, Paul M, Dayal D, Bhansali A, Bhadada SK, Kumar R, Sachdeva N. Peripheral blood mononuclear cells of patients with latent autoimmune diabetes secrete higher levels of pro- & anti-inflammatory cytokines compared to those with type-1 diabetes mellitus following in vitro stimulation with β-cell autoantigens. Indian J Med Res 2017; 145:767-776. [PMID: 29067979 PMCID: PMC5674547 DOI: 10.4103/ijmr.ijmr_1563_15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND & OBJECTIVES Type-1 diabetes mellitus (T1DM) and latent autoimmune diabetes in adults (LADA) share similar pathological features but differ in age of onset and progression. There is a scarcity of information on differences in CD4+ T-cell responses, particularly, cytokine secretion, between the two forms of autoimmune diabetes. Here proliferative potential and concentration of pro- and anti-inflammatory cytokines secreted by peripheral blood mononuclear cells (PBMCs) of T1DM and LADA patients were compared, after in vitro stimulation with β-cell autoantigens. METHODS A total of 19 patients with LADA, 37 with T1DM and 20 healthy controls were compared on the basis of lymphocyte proliferation and secretion of pro- and anti-inflammatory cytokines belonging to different T-helper types after in vitro stimulation of PBMCs with insulin and glutamic acid decarboxylase 65 (GAD65). RESULTS Following insulin stimulation, LADA group secreted higher concentration of interleukin-17 (IL-17) (P=0.02) and had higher proportion of interferon gamma (IFN-γ) secretors (P<0.001) than T1DM group. Post-GAD65 stimulation, higher proportion of LADA patients secreted IL-23 than T1DM group (P=0.02). Proportion of responders , as well as levels of secreted IL-10, were significantly higher in LADA than T1DM group, following stimulation with both insulin (P=0.01) and GAD65 (P=0.03). A significant positive correlation was observed between body mass index and IL-17 levels (r=0.41, P=0.04) and fasting plasma C-peptide with IL-10 levels (r=0.37, P=0.04). INTERPRETATION & CONCLUSIONS There are differences in the portfolio of cytokine secretion in diabetic subjects with varying rates of β-cell destruction as LADA subjects secrete higher levels of both pro- and anti-inflammatory cytokines on exposure to β-cell autoantigens, thus highlighting another distinguishing feature in the pathophysiology of the two forms of autoimmune diabetes.
Collapse
Affiliation(s)
- Darshan Badal
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Rajendra Kumar
- University Institute of Pharmaceutical Sciences (UIPS), Chandigarh, India
| | - Mahinder Paul
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Devi Dayal
- Department of Pediatrics, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Anil Bhansali
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Sanjay Kumar Bhadada
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Rajesh Kumar
- Department of Statistics, Panjab University, Chandigarh, India
| | - Naresh Sachdeva
- Department of Endocrinology, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| |
Collapse
|
30
|
Chemistry and biology of reactive species with special reference to the antioxidative defence status in pancreatic β-cells. Biochim Biophys Acta Gen Subj 2017; 1861:1929-1942. [PMID: 28527893 DOI: 10.1016/j.bbagen.2017.05.013] [Citation(s) in RCA: 95] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 05/12/2017] [Accepted: 05/16/2017] [Indexed: 01/08/2023]
Abstract
BACKGROUND Diabetes mellitus is a serious metabolic disease. Dysfunction and subsequent loss of the β-cells in the islets of Langerhans through apoptosis ultimately cause a life-threatening insulin deficiency. The underlying reason for the particular vulnerability of the β-cells is an extraordinary sensitivity to the toxicity of reactive oxygen and nitrogen species (ROS and RNS) due to its low antioxidative defense status. SCOPE REVIEW This review considers the different aspects of the chemistry and biology of the biologically most important reactive species and their chemico-biological interactions in the β-cell toxicity of proinflammatory cytokines in type 1 diabetes and of lipotoxicity in type 2 diabetes development. MAJOR CONCLUSION The weak antioxidative defense equipment in the different subcellular organelles makes the β-cells particularly vulnerable and prone to mitochondrial, peroxisomal and ER stress. Looking upon the enzyme deficiencies which are responsible for the low antioxidative defense status of the pancreatic β-cells it is the lack of enzymatic capacity for H2O2 inactivation at all major subcellular sites. GENERAL SIGNIFICANCE Diabetes is the most prevalent metabolic disorder with a steadily increasing incidence of both type 1 and type 2 diabetes worldwide. The weak protection of the pancreatic β-cells against oxidative stress is a major reason for their particular vulnerability. Thus, careful protection of the β-cells is required for prevention of the disease.
Collapse
|
31
|
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
|
32
|
Gurgul-Convey E, Mehmeti I, Plötz T, Jörns A, Lenzen S. Sensitivity profile of the human EndoC-βH1 beta cell line to proinflammatory cytokines. Diabetologia 2016; 59:2125-33. [PMID: 27460666 DOI: 10.1007/s00125-016-4060-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 07/05/2016] [Indexed: 01/27/2023]
Abstract
AIMS/HYPOTHESIS The aim of this study was to perform a detailed analysis of cytokine toxicity in the new human EndoC-βH1 beta cell line. METHODS The expression profile of the antioxidative enzymes in the new human EndoC-βH1 beta cells was characterised and compared with that of primary beta cells in the human pancreas. The effects of proinflammatory cytokines on reactive oxygen species formation, insulin secretory responsiveness and apoptosis of EndoC-βH1 beta cells were determined. RESULTS EndoC-βH1 beta cells were sensitive to the toxic action of proinflammatory cytokines. Glucose-dependent stimulation of insulin secretion and an increase in the ATP/ADP ratio was abolished by proinflammatory cytokines without induction of IL-1β expression. Cytokine-mediated caspase-3 activation was accompanied by reactive oxygen species formation and developed more slowly than in rodent beta cells. Cytokines transiently increased the expression of unfolded protein response genes, without inducing endoplasmic reticulum stress-marker genes. Cytokine-mediated NFκB activation was too weak to induce inducible nitric oxide synthase expression. The resultant lack of nitric oxide generation in EndoC-βH1 cells, in contrast to rodent beta cells, makes these cells dependent on exogenously generated nitric oxide, which is released from infiltrating immune cells in human type 1 diabetes, for full expression of proinflammatory cytokine toxicity. CONCLUSIONS/INTERPRETATION EndoC-βH1 beta cells are characterised by an imbalance between H2O2-generating and -inactivating enzymes, and react to cytokine exposure in a similar manner to primary human beta cells. They are a suitable beta cell surrogate for cytokine-toxicity studies.
Collapse
Affiliation(s)
- Ewa Gurgul-Convey
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Ilir Mehmeti
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Thomas Plötz
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Anne Jörns
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany
| | - Sigurd Lenzen
- Institute of Clinical Biochemistry, Hannover Medical School, Carl-Neuberg-Str. 1, 30625, Hannover, Germany.
- Institute of Experimental Diabetes Research, Hannover Medical School, Hannover, Germany.
| |
Collapse
|
33
|
Baharlou R, Ahmadi-Vasmehjani A, Davami MH, Faraji F, Atashzar MR, Karimipour F, Sadeghi A, Asadi MA, Khoubyari M. Elevated Levels of T-helper 17-associated Cytokines in Diabetes Type I Patients: Indicators for Following the Course of Disease. Immunol Invest 2016; 45:641-51. [DOI: 10.1080/08820139.2016.1197243] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
34
|
Scharfmann R, Didiesheim M, Richards P, Chandra V, Oshima M, Albagli O. Mass production of functional human pancreatic β-cells: why and how? Diabetes Obes Metab 2016; 18 Suppl 1:128-36. [PMID: 27615142 DOI: 10.1111/dom.12728] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 05/17/2016] [Indexed: 12/17/2022]
Abstract
Diabetes (either type 1 or type 2) is due to insufficient functional β-cell mass. Research has, therefore, aimed to discover new ways to maintain or increase either β-cell mass or function. For this purpose, rodents have mainly been used as model systems and a large number of discoveries have been made. Meanwhile, although we have learned that rodent models represent powerful systems to model β-cell development, function and destruction, we realize that there are limitations when attempting to transfer the data to what is occurring in humans. Indeed, while human β-cells share many similarities with rodent β-cells, they also differ on a number of important parameters. In this context, developing ways to study human β-cell development, function and death represents an important challenge. This review will describe recent data on the development and use of convenient sources of human β-cells that should be useful tools to discover new ways to modulate functional β-cell mass in humans.
Collapse
Affiliation(s)
- R Scharfmann
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France.
| | - M Didiesheim
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - P Richards
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - V Chandra
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - M Oshima
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| | - O Albagli
- INSERM U1016, Université Paris-Descartes, Institut Cochin, Paris, France
| |
Collapse
|
35
|
Berchtold LA, Prause M, Størling J, Mandrup-Poulsen T. Cytokines and Pancreatic β-Cell Apoptosis. Adv Clin Chem 2016; 75:99-158. [PMID: 27346618 DOI: 10.1016/bs.acc.2016.02.001] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The discovery 30 years ago that inflammatory cytokines cause a concentration, activity, and time-dependent bimodal response in pancreatic β-cell function and viability has been a game-changer in the fields of research directed at understanding inflammatory regulation of β-cell function and survival and the causes of β-cell failure and destruction in diabetes. Having until then been confined to the use of pathophysiologically irrelevant β-cell toxic chemicals as a model of β-cell death, researchers could now mimic endocrine and paracrine effects of the cytokine response in vitro by titrating concentrations in the low to the high picomolar-femtomolar range and vary exposure time for up to 14-16h to reproduce the acute regulatory effects of systemic inflammation on β-cell secretory responses, with a shift to inhibition at high picomolar concentrations or more than 16h of exposure to illustrate adverse effects of local, chronic islet inflammation. Since then, numerous studies have clarified how these bimodal responses depend on discrete signaling pathways. Most interest has been devoted to the proapoptotic response dependent upon mainly nuclear factor κ B and mitogen-activated protein kinase activation, leading to gene expressional changes, endoplasmic reticulum stress, and triggering of mitochondrial dysfunction. Preclinical studies have shown preventive effects of cytokine antagonism in animal models of diabetes, and clinical trials demonstrating proof of concept are emerging. The full clinical potential of anticytokine therapies has yet to be shown by testing the incremental effects of appropriate dosing, timing, and combinations of treatments. Due to the considerable translational importance of enhancing the precision, specificity, and safety of antiinflammatory treatments of diabetes, we review here the cellular, preclinical, and clinical evidence of which of the death pathways recently proposed in the Nomenclature Committee on Cell Death 2012 Recommendations are activated by inflammatory cytokines in the pancreatic β-cell to guide the identification of antidiabetic targets. Although there are still scarce human data, the cellular and preclinical studies point to the caspase-dependent intrinsic apoptosis pathway as the prime effector of inflammatory β-cell apoptosis.
Collapse
Affiliation(s)
| | - M Prause
- University of Copenhagen, Copenhagen, Denmark
| | - J Størling
- Copenhagen Diabetes Research Center, Beta Cell Biology Group, Copenhagen University Hospital Herlev, Herlev, Denmark
| | | |
Collapse
|
36
|
Marroqui L, Dos Santos RS, Fløyel T, Grieco FA, Santin I, Op de Beeck A, Marselli L, Marchetti P, Pociot F, Eizirik DL. TYK2, a Candidate Gene for Type 1 Diabetes, Modulates Apoptosis and the Innate Immune Response in Human Pancreatic β-Cells. Diabetes 2015; 64:3808-17. [PMID: 26239055 DOI: 10.2337/db15-0362] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 06/12/2015] [Indexed: 11/13/2022]
Abstract
Pancreatic β-cells are destroyed by an autoimmune attack in type 1 diabetes. Linkage and genome-wide association studies point to >50 loci that are associated with the disease in the human genome. Pathway analysis of candidate genes expressed in human islets identified a central role for interferon (IFN)-regulated pathways and tyrosine kinase 2 (TYK2). Polymorphisms in the TYK2 gene predicted to decrease function are associated with a decreased risk of developing type 1 diabetes. We presently evaluated whether TYK2 plays a role in human pancreatic β-cell apoptosis and production of proinflammatory mediators. TYK2-silenced human β-cells exposed to polyinosinic-polycitidilic acid (PIC) (a mimick of double-stranded RNA produced during viral infection) showed less type I IFN pathway activation and lower production of IFNα and CXCL10. These cells also had decreased expression of major histocompatibility complex (MHC) class I proteins, a hallmark of early β-cell inflammation in type 1 diabetes. Importantly, TYK2 inhibition prevented PIC-induced β-cell apoptosis via the mitochondrial pathway of cell death. The present findings suggest that TYK2 regulates apoptotic and proinflammatory pathways in pancreatic β-cells via modulation of IFNα signaling, subsequent increase in MHC class I protein, and modulation of chemokines such as CXCL10 that are important for recruitment of T cells to the islets.
Collapse
Affiliation(s)
- Laura Marroqui
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | | | - Tina Fløyel
- Department of Pediatrics, Herlev University Hospital, Herlev, Denmark
| | - Fabio A Grieco
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Izortze Santin
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium Endocrinology and Diabetes Research Group, BioCruces Health Research Institute and Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders, Barakaldo, Spain
| | - Anne Op de Beeck
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Lorella Marselli
- Department of Clinical and Experimental Medicine, Pancreatic Islet Laboratory, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, Pancreatic Islet Laboratory, University of Pisa, Pisa, Italy
| | - Flemming Pociot
- Department of Pediatrics, Herlev University Hospital, Herlev, Denmark
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
37
|
Olson NC, Doyle MF, de Boer IH, Huber SA, Jenny NS, Kronmal RA, Psaty BM, Tracy RP. Associations of Circulating Lymphocyte Subpopulations with Type 2 Diabetes: Cross-Sectional Results from the Multi-Ethnic Study of Atherosclerosis (MESA). PLoS One 2015; 10:e0139962. [PMID: 26458065 PMCID: PMC4601795 DOI: 10.1371/journal.pone.0139962] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 09/18/2015] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Distinct lymphocyte subpopulations have been implicated in the regulation of glucose homeostasis and obesity-associated inflammation in mouse models of insulin resistance. Information on the relationships of lymphocyte subpopulations with type 2 diabetes remain limited in human population-based cohort studies. METHODS Circulating levels of innate (γδ T, natural killer (NK)) and adaptive immune (CD4+ naive, CD4+ memory, Th1, and Th2) lymphocyte subpopulations were measured by flow cytometry in the peripheral blood of 929 free-living participants of the Multi-Ethnic Study of Atherosclerosis (MESA). Cross-sectional relationships of lymphocyte subpopulations with type 2 diabetes (n = 154) and fasting glucose and insulin concentrations were evaluated by generalized linear models. RESULTS Each standard deviation (SD) higher CD4+ memory cells was associated with a 21% higher odds of type 2 diabetes (95% CI: 1-47%) and each SD higher naive cells was associated with a 22% lower odds (95% CI: 4-36%) (adjusted for age, gender, race/ethnicity, and BMI). Among participants not using diabetes medication, higher memory and lower naive CD4+ cells were associated with higher fasting glucose concentrations (p<0.05, adjusted for age, sex, and race/ethnicity). There were no associations of γδ T, NK, Th1, or Th2 cells with type 2 diabetes, glucose, or insulin. CONCLUSIONS A higher degree of chronic adaptive immune activation, reflected by higher memory and lower naive CD4+ cells, was positively associated with type 2 diabetes. These results are consistent with a role of chronic immune activation and exhaustion augmenting chronic inflammatory diseases, and support the importance of prospective studies evaluating adaptive immune activation and type 2 diabetes.
Collapse
Affiliation(s)
- Nels C. Olson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Margaret F. Doyle
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Ian H. de Boer
- Division of Nephrology, Department of Medicine, University of Washington, Seattle, Washington, United States of America
- Kidney Research Institute, University of Washington, Seattle, Washington, United States of America
| | - Sally A. Huber
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Nancy Swords Jenny
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
| | - Richard A. Kronmal
- Collaborative Health Studies Coordinating Center, Department of Biostatistics, University of Washington, Seattle, Washington, United States of America
| | - Bruce M. Psaty
- Departments of Medicine, Epidemiology, Health Services, and Cardiovascular Health Research Unit, University of Washington, Seattle, Washington, United States of America
- Group Health Research Institute, Group Health Cooperative, Seattle, Washington, United States of America
| | - Russell P. Tracy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, Vermont, United States of America
- Department of Biochemistry, University of Vermont College of Medicine, Burlington, Vermont, United States of America
- * E-mail:
| |
Collapse
|
38
|
Eljaafari A, Robert M, Chehimi M, Chanon S, Durand C, Vial G, Bendridi N, Madec AM, Disse E, Laville M, Rieusset J, Lefai E, Vidal H, Pirola L. Adipose Tissue-Derived Stem Cells From Obese Subjects Contribute to Inflammation and Reduced Insulin Response in Adipocytes Through Differential Regulation of the Th1/Th17 Balance and Monocyte Activation. Diabetes 2015; 64:2477-88. [PMID: 25765019 DOI: 10.2337/db15-0162] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 03/04/2015] [Indexed: 12/19/2022]
Abstract
Obesity, through low-grade inflammation, can drive insulin resistance and type 2 diabetes. While infiltration of adipose tissue (AT) with mononuclear cells (MNCs) is well established in obesity, the functional consequences of these interactions are less understood. Herein, we cocultured human adipose-derived stem cells (ASCs) from obese individuals with MNCs and analyzed their reciprocal behavior. Presence of ASCs 1) enhanced interleukin (IL)-17A secretion by Th17 cells, 2) inhibited γ-interferon and tumor necrosis factor α secretion by Th1 cells, and 3) increased monocyte-mediated IL-1β secretion. IL-17A secretion also occurred in stromal vascular fractions issued from obese but not lean individuals. Th17 polarization mostly depended on physical contacts between ASCs and MNCs-with a contribution of intracellular adhesion molecule-1-and occurred through activation of the inflammasome and phosphatidylinositol 3-kinase pathways. ASCs favored STAT3 over STAT5 transcription factor binding on STAT binding sites within the IL-17A/F gene locus. Finally, conditioned media from activated ASC-MNC cocultures inhibited adipocyte differentiation mRNA markers and impaired insulin-mediated Akt phosphorylation and lipolysis inhibition. In conclusion, we report that obese- but not lean-derived ASCs induce Th17 promotion and monocyte activation. This proinflammatory environment, in turn, inhibits adipogenesis and adipocyte insulin response. The demonstration of an ASC-Th17-monocyte cell axis reveals a novel proinflammatory process taking place in AT during obesity and defines novel putative therapeutic targets.
Collapse
Affiliation(s)
- Assia Eljaafari
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France Clinical Research Department, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Maud Robert
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France Gastroenterology and Surgery Department, Edouard Herriot Hospital, Lyon, France
| | - Marwa Chehimi
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Stephanie Chanon
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Christine Durand
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Guillaume Vial
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Nadia Bendridi
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Anne-Marie Madec
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Emmanuel Disse
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France Clinical Research Department, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Martine Laville
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France Clinical Research Department, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Jennifer Rieusset
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Etienne Lefai
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Hubert Vidal
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| | - Luciano Pirola
- CarMeN Laboratory, INSERM U1060, Lyon-1 University, INRA U1397, INSA-Lyon, Lyon, France
| |
Collapse
|
39
|
Astry B, Venkatesha SH, Moudgil KD. Involvement of the IL-23/IL-17 axis and the Th17/Treg balance in the pathogenesis and control of autoimmune arthritis. Cytokine 2015; 74:54-61. [PMID: 25595306 PMCID: PMC4457562 DOI: 10.1016/j.cyto.2014.11.020] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 11/15/2014] [Accepted: 11/18/2014] [Indexed: 02/07/2023]
Abstract
The T helper (Th) cell subsets are characterized by the type of cytokines produced and the master transcription factor expressed. Th1 cells participate in cell-mediated immunity, whereas Th2 cells promote humoral immunity. Furthermore, the two subsets can control each other. Thereby, Th1-Th2 balance offered a key paradigm in understanding the induction and regulation of immune pathology in autoimmune and other diseases. However, over the past decade, Th17 cells producing interleukin-17 (IL-17) have emerged as the major pathogenic T cell subset in many pathological conditions that were previously attributed to Th1 cells. In addition, the role of CD4+CD25+T regulatory cells (Treg) in controlling the activity of Th17 and other T cell subsets has increasingly been realized. Thereby, examination of the Th17/Treg balance in the course of autoimmune diseases has significantly advanced our understanding of the pathogenesis of these disorders. The differentiation of Th17 and Treg cells from naïve T cells is inter-related and controlled in part by the cytokine milieu. For example, transforming growth factor β (TGFβ) is required for Treg induction, whereas the same cytokine in the presence of IL-6 (or IL-1) promotes the differentiation of Th17. Furthermore, IL-23 plays a role in the maintenance of Th17. Accordingly, novel therapeutic approaches are being developed to target IL-23/IL-17 as well as to modulate the Th17/Treg balance in favor of immune regulation to control autoimmunity.
Collapse
Affiliation(s)
- Brian Astry
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Shivaprasad H Venkatesha
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States
| | - Kamal D Moudgil
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, United States.
| |
Collapse
|
40
|
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
|
41
|
Reinert-Hartwall L, Honkanen J, Salo HM, Nieminen JK, Luopajärvi K, Härkönen T, Veijola R, Simell O, Ilonen J, Peet A, Tillmann V, Knip M, Vaarala O. Th1/Th17 plasticity is a marker of advanced β cell autoimmunity and impaired glucose tolerance in humans. THE JOURNAL OF IMMUNOLOGY 2014; 194:68-75. [PMID: 25480564 DOI: 10.4049/jimmunol.1401653] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Upregulation of IL-17 immunity and detrimental effects of IL-17 on human islets have been implicated in human type 1 diabetes. In animal models, the plasticity of Th1/Th17 cells contributes to the development of autoimmune diabetes. In this study, we demonstrate that the upregulation of the IL-17 pathway and Th1/Th17 plasticity in peripheral blood are markers of advanced β cell autoimmunity and impaired β cell function in human type 1 diabetes. Activated Th17 immunity was observed in the late stage of preclinical diabetes in children with β cell autoimmunity and impaired glucose tolerance, but not in children with early β cell autoimmunity. We found an increased ratio of IFN-γ/IL-17 expression in Th17 cells in children with advanced β cell autoimmunity, which correlated with HbA1c and plasma glucose concentrations in an oral glucose tolerance test, and thus impaired β cell function. Low expression of Helios was seen in Th17 cells, suggesting that Th1/Th17 cells are not converted thymus-derived regulatory T cells. Our results suggest that the development of Th1/Th17 plasticity may serve as a biomarker of disease progression from β cell autoantibody positivity to type 1 diabetes. These data in human type 1 diabetes emphasize the role of Th1/Th17 plasticity as a potential contributor to tissue destruction in autoimmune conditions.
Collapse
Affiliation(s)
- Linnea Reinert-Hartwall
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Jarno Honkanen
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Harri M Salo
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Janne K Nieminen
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland
| | - Kristiina Luopajärvi
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, 00271 Helsinki, Finland; Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland
| | - Taina Härkönen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland
| | - Riitta Veijola
- Department of Pediatrics, Medical Research Center, Oulu University Hospital and University of Oulu, 90014 Oulu, Finland
| | - Olli Simell
- Department of Pediatrics, University of Turku, 20520 Turku, Finland
| | - Jorma Ilonen
- Immunogenetics Laboratory, University of Turku, 20520 Turku, Finland; Department of Clinical Microbiology, University of Eastern Finland, 70211 Kuopio, Finland
| | - Aleksandr Peet
- Department of Pediatrics, University of Tartu and Children's Clinic of Tartu University Hospital, Tartu 51014, Estonia
| | - Vallo Tillmann
- Department of Pediatrics, University of Tartu and Children's Clinic of Tartu University Hospital, Tartu 51014, Estonia
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland; Folkhälsan Research Center, 00290 Helsinki, Finland; Diabetes and Obesity Research Program, University of Helsinki, 00290 Helsinki, Finland; Department of Pediatrics, Tampere University Hospital, 33521 Tampere, Finland; and
| | - Outi Vaarala
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, 00281 Helsinki, Finland; Respiratory, Inflammatory and Autoimmune Diseases, Innovative Medicine, AstraZeneca, 43183 Mölndal, Sweden
| | | | | |
Collapse
|
42
|
Mathieu C, Overbergh L. The Story of NAIMIT - A Framework 7 Project on Type 1 Diabetes. EUROPEAN ENDOCRINOLOGY 2014; 10:100-105. [PMID: 29872472 DOI: 10.17925/ee.2014.10.02.100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/06/2014] [Indexed: 11/24/2022]
Abstract
NAIMIT (acronym for Natural Immune Modulation for Intervention in Type 1 Diabetes) is a large-scale collaborative programme of the 7th framework from the European Commission. The aim of the consortium is to bring together a group of leading European researchers spanning the field from genetics, through pancreatic beta-cell, dendritic cells and T-cell biology, towards clinical interventions. The ultimate goal is to develop novel and personalised interventional therapies in recent-onset type 1 diabetic patients, with minimal immune system interference, leading to beta-cell protection and restoration, based on a solid understanding of the disease pathogenesis, enabling experimental findings to be adopted for clinical applications.
Collapse
Affiliation(s)
| | - Lut Overbergh
- Senior Researcher, NAIMIT Consortium, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| |
Collapse
|