1
|
Lokesh MN, Kumar R, Jacob N, Sachdeva N, Rawat A, Yadav J, Dayal D. Supplementation of High-Strength Oral Probiotics Improves Immune Regulation and Preserves Beta Cells among Children with New-Onset Type 1 Diabetes Mellitus: A Randomised, Double-Blind Placebo Control Trial. Indian J Pediatr 2025; 92:277-283. [PMID: 38557820 DOI: 10.1007/s12098-024-05074-5] [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: 09/30/2023] [Accepted: 01/24/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVES To investigate the mechanism of glycemic control in children with type 1 diabetes (T1D) following high-strength probiotics supplementation by assessing immune-regulatory markers. METHODS In this single-centre randomised double-blinded placebo-controlled study, children with new-onset T1D on regular insulin therapy were randomised into probiotic or placebo groups with 30 children each. The probiotics group received oral powder of Vivomixx®, and the placebo group received corn starch for six months. The primary outcome parameters included induced T regulatory cells (i-Tregs) percentage, insulin autoantibodies (IAA), insulinoma associated 2 autoantibodies (IA2), glutamic acid decarboxylase autoantibodies (GAD 65) and plasma interleukin-10 (IL-10) levels. The secondary outcome variables were changes in plasma C-peptide levels and glycemic control parameters. RESULTS Twenty-three children in the placebo group and 27 in the probiotic group completed the study. There was a significant increase in the percentage of iTregs (3.40 in the probiotic vs. 2.46 in the placebo group; p = 0.034). Median glycated hemoglobin (HbA1c) levels significantly decreased from 68 mmol/mol (8.35%) in the placebo group to 60 mmol/mol (7.55%) in the probiotic group (p = 0.017). Median C-peptide levels were significantly higher in probiotics (0.72 ng/ml) vs. placebo group (0.11 ng/ml) (p = 0.036). The plasma IL-10 levels significantly increased in the probiotic group after six months of treatment (p = 0.002). CONCLUSIONS The high-strength probiotics improved the immunoregulatory milieu, thereby preserving the beta-cell function and better glycemic control.
Collapse
Affiliation(s)
- M N Lokesh
- Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Rakesh Kumar
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India.
| | - Neenu Jacob
- Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Naresh Sachdeva
- Department of Endocrinology, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Amit Rawat
- Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Jaivinder Yadav
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India
| | - Devi Dayal
- Endocrinology and Diabetes Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Sector 12, Chandigarh, 160012, India
| |
Collapse
|
2
|
Cobb J, Rawson J, Gonzalez N, Singer M, Kandeel F, Husseiny MI. Mechanism of Action of Oral Salmonella-Based Vaccine to Prevent and Reverse Type 1 Diabetes in NOD Mice. Vaccines (Basel) 2024; 12:276. [PMID: 38543910 PMCID: PMC10975319 DOI: 10.3390/vaccines12030276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 02/27/2024] [Accepted: 03/04/2024] [Indexed: 03/06/2025] Open
Abstract
A combination therapy of preproinsulin (PPI) and immunomodulators (TGFβ+IL10) orally delivered via genetically modified Salmonella and anti-CD3 promoted glucose balance in in NOD mice with recent onset diabetes. The Salmonella bacteria were modified to express the diabetes-associated antigen PPI controlled by a bacterial promoter in conjunction with over-expressed immunomodulating molecules. The possible mechanisms of action of this vaccine to limit autoimmune diabetes remained undefined. In mice, the vaccine prevented and reversed ongoing diabetes. The vaccine-mediated beneficial effects were associated with increased numbers of antigen-specific CD4+CD25+Foxp3+ Tregs, CD4+CD49b+LAG3+ Tr1-cells, and tolerogenic dendritic-cells (tol-DCs) in the spleens and lymphatic organs of treated mice. Despite this, the immune response to Salmonella infection was not altered. Furthermore, the vaccine effects were associated with a reduction in islet-infiltrating lymphocytes and an increase in the islet beta-cell mass. This was associated with increased serum levels of the tolerogenic cytokines (IL10, IL2, and IL13) and chemokine ligand 2 (CCL2) and decreased levels of inflammatory cytokines (IFNγ, GM-CSF, IL6, IL12, and TNFα) and chemokines (CXCL1, CXCL2, and CXCL5). Overall, the data suggest that the Salmonella-based vaccine modulates the immune response, reduces inflammation, and promotes tolerance specifically to an antigen involved in autoimmune diabetes.
Collapse
Affiliation(s)
- Jacob Cobb
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Duarte, CA 91010, USA (F.K.)
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Jeffrey Rawson
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Duarte, CA 91010, USA (F.K.)
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Nelson Gonzalez
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Duarte, CA 91010, USA (F.K.)
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mahmoud Singer
- School of Medicine, University of California Irvine, Irvine, CA 92697, USA;
| | - Fouad Kandeel
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Duarte, CA 91010, USA (F.K.)
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Mohamed I. Husseiny
- Department of Translational Research & Cellular Therapeutics, Arthur Riggs Diabetes & Metabolism Research Institute, Duarte, CA 91010, USA (F.K.)
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
3
|
Thakkar S, Chopra A, Nagendra L, Kalra S, Bhattacharya S. Teplizumab in Type 1 Diabetes Mellitus: An Updated Review. TOUCHREVIEWS IN ENDOCRINOLOGY 2023; 19:22-30. [PMID: 38187075 PMCID: PMC10769466 DOI: 10.17925/ee.2023.19.2.7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 07/01/2023] [Indexed: 01/09/2024]
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic autoimmune condition characterized by the irreversible destruction of the β cells of the pancreas, which leads to a lifelong dependency on exogenous insulin. Despite the advancements in insulin delivery methods, the suboptimal outcomes of these methods have triggered the search for therapies that may prevent or reverse the disease. Given the autoimmune aetiology of T1DM, therapies counteracting the immune-mediated destruction of the β-cells are the obvious target. Although several treatment strategies have been attempted to target cellular, humoral and innate immunity, very few have had a clinically meaningful impact. Of all the available immunomodulatory agents, cluster of differentiation (CD) 3 antibodies have exhibited the most promising preclinical and clinical results. Muromonab-CD3, which also happened to be a murine CD3 antibody, was the first monoclonal antibody approved for clinical use and was primarily indicated for graft rejection. The adverse effects associated with muromonab-CD3 led to its withdrawal. Teplizumab, a newer CD3 antibody, has a better side-effect profile because of its humanized nature and non-Fc-receptor-binding domain. In November 2022, teplizumab became the first immunomodulatory agent to be licensed by the US Food and Drug Administration for delaying the onset of T1DM in high-risk adults and children over 8 years old. The mechanism seems to be enhancing regulatory T-cell activity and promoting immune tolerance. This article reviews the mechanism of action and the clinical trials of teplizumab in individuals with T1DM or at risk of developing the disease.
Collapse
Affiliation(s)
- Simran Thakkar
- Department of Endocrinology, Indraprastha Apollo Hospitals, New Delhi, India
| | - Aditi Chopra
- Department of Endocrinology, Manipal Hospital, Bengaluru, India
| | | | - Sanjay Kalra
- Department of Endocrinology, Bharti Hospital, Karnal, Haryana, India
| | | |
Collapse
|
4
|
Chen M, Zhang Q, Wei Y, Wan Q, Xu M, Chen X. Anti-CD20 therapy ameliorates β cell function and rebalances Th17/Treg cells in NOD mice. Endocrine 2022; 76:44-52. [PMID: 35067899 DOI: 10.1007/s12020-021-02965-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/12/2021] [Indexed: 12/15/2022]
Abstract
PURPOSE Anti-CD20 therapy delays type 1 diabetes mellitus (T1DM) progression in both nonobese diabetic (NOD) mice and new-onset patients. The mechanism is not completely defined. This study aimed to investigate the effects of anti-CD20 therapy on T helper 17 (Th17) cells and regulatory T cells (Tregs) in NOD mice. The role of B cell depletion in T1DM development was also examined. METHODS NOD mice were randomly divided into two groups. The mice in the experimental group were treated with an anti-CD20 antibody, while the control mice were treated with an isotype-matched control antibody. After treatment, islet morphology and inflammation, Th17 and Treg cell frequencies in the pancreas and spleen, serum cytokine and anti-glutamic acid decarboxylase (GAD) antibody levels, interleukin (IL)-17A levels in the pancreas and spleen, insulin expression in islet cells and islet β cell function were measured. RESULTS Decreased blood glucose and increased insulin secretion were found in the exprimental group compared with the CON group. A lower islet inflammation score was also found in the experimental group. Decreased Th17 cell and IL-17A levels and augmented Treg cell levels were found in the spleen and pancreas after anti-CD20 treatment. The serum levels of B cell activating factor (BAFF), IL-17A, IL-17F, IL-23 and anti-GAD autoantibodies were decreased in the experimental group, while higher serum levels of IL-10 and transforming growth factor (TGF)-β were found. CONCLUSION Anti-CD20 therapy might have some beneficial effects that improve β cell function by relieving islet inflammation through regulation of Th17/Treg cells and the proinflammatory/anti-inflammatory balance.
Collapse
Affiliation(s)
- Min Chen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Qianhui Zhang
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Yanhong Wei
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Qianqian Wan
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Min Xu
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China
| | - Xiaoqi Chen
- Department of Rheumatology and Immunology, Zhongnan Hospital, Wuhan University, 169 Donghu Road, Wuhan, China.
| |
Collapse
|
5
|
Hu W, Song X, Yu H, Sun J, Wang H, Zhao Y. Clinical Translational Potentials of Stem Cell-Derived Extracellular Vesicles in Type 1 Diabetes. Front Endocrinol (Lausanne) 2021; 12:682145. [PMID: 35095751 PMCID: PMC8789747 DOI: 10.3389/fendo.2021.682145] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 12/06/2021] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific disease characterized by the deficiency of insulin caused by the autoimmune destruction of pancreatic islet β cells. Stem cell-based therapies play essential roles in immunomodulation and tissue regeneration, both of which hold great promise for treating many autoimmune dysfunctions. However, their clinical translational potential has been limited by ethical issues and cell transplant rejections. Exosomes are small extracellular vesicles (EVs) released by almost all types of cells, performing a variety of cell functions through the delivery of their molecular contents such as proteins, DNAs, and RNAs. Increasing evidence suggests that stem cell-derived EVs exhibit similar functions as their parent cells, which may represent novel therapeutic agents for the treatment of autoimmune diseases including T1D. In this review, we summarize the current research progresses of stem cell-derived EVs for the treatment of T1D.
Collapse
Affiliation(s)
- Wei Hu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Xiang Song
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Haibo Yu
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
| | - Jingyu Sun
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ, United States
| | - Hongjun Wang
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ, United States
| | - Yong Zhao
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ, United States
- Throne Biotechnologies Inc., Paramus, NJ, United States
- *Correspondence: Yong Zhao,
| |
Collapse
|
6
|
Abela AG, Fava S. Why is the Incidence of Type 1 Diabetes Increasing? Curr Diabetes Rev 2021; 17:e030521193110. [PMID: 33949935 DOI: 10.2174/1573399817666210503133747] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 02/14/2021] [Accepted: 03/11/2021] [Indexed: 11/22/2022]
Abstract
Type 1 diabetes is a condition that can lead to serious long-term complications and can have significant psychological and quality of life implications. Its incidence is increasing in all parts of the world, but the reasons for this are incompletely understood. Genetic factors alone cannot explain such a rapid increase in incidence; therefore, environmental factors must be implicated. Lifestyle factors have been classically associated with type 2 diabetes. However, there are data implicating obesity and insulin resistance to type 1 diabetes as well (accelerator hypothesis). Cholesterol has also been shown to be correlated with the incidence of type 1 diabetes; this may be mediated by immunomodulatory effects of cholesterol. There is considerable interest in early life factors, including maternal diet, mode of delivery, infant feeding, childhood diet, microbial exposure (hygiene hypothesis), and use of anti-microbials in early childhood. Distance from the sea has recently been shown to be negatively correlated with the incidence of type 1 diabetes. This may contribute to the increasing incidence of type 1 diabetes since people are increasingly living closer to the sea. Postulated mediating mechanisms include hours of sunshine (and possibly vitamin D levels), mean temperature, dietary habits, and pollution. Ozone, polychlorinated biphenyls, phthalates, trichloroethylene, dioxin, heavy metals, bisphenol, nitrates/nitrites, and mercury are amongst the chemicals which may increase the risk of type 1 diabetes. Another area of research concerns the role of the skin and gut microbiome. The microbiome is affected by many of the factors mentioned above, including the mode of delivery, infant feeding, exposure to microbes, antibiotic use, and dietary habits. Research on the reasons why the incidence of type 1 diabetes is increasing not only sheds light on its pathogenesis but also offers insights into ways we can prevent type 1 diabetes.
Collapse
Affiliation(s)
- Alexia G Abela
- Department of Medicine, University of Malta & Mater Dei Hospital, Tal-Qroqq, Msida, Malta
| | - Stephen Fava
- Department of Medicine, University of Malta & Mater Dei Hospital, Tal-Qroqq, Msida, Malta
| |
Collapse
|
7
|
Lavoz C, Rayego-Mateos S, Orejudo M, Opazo-Ríos L, Marchant V, Marquez-Exposito L, Tejera-Muñoz A, Navarro-González JF, Droguett A, Ortiz A, Egido J, Mezzano S, Rodrigues-Diez RR, Ruiz-Ortega M. Could IL-17A Be a Novel Therapeutic Target in Diabetic Nephropathy? J Clin Med 2020; 9:E272. [PMID: 31963845 PMCID: PMC7019373 DOI: 10.3390/jcm9010272] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic kidney disease has become a major medical issue in recent years due to its high prevalence worldwide, its association with premature mortality, and its social and economic implications. A number of patients gradually progress to end-stage renal disease (ESRD), requiring then dialysis and kidney transplantation. Currently, approximately 40% of patients with diabetes develop kidney disease, making it the most prevalent cause of ESRD. Thus, more effective therapies for diabetic nephropathy are needed. In preclinical studies of diabetes, anti-inflammatory therapeutic strategies have been used to protect the kidneys. Recent evidence supports that immune cells play an active role in the pathogenesis of diabetic nephropathy. Th17 immune cells and their effector cytokine IL-17A have recently emerged as promising targets in several clinical conditions, including renal diseases. Here, we review current knowledge regarding the involvement of Th17/IL-17A in the genesis of diabetic renal injury, as well as the rationale behind targeting IL-17A as an additional therapy in patients with diabetic nephropathy.
Collapse
Affiliation(s)
- Carolina Lavoz
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.L.); (V.M.); (A.D.); (S.M.)
| | - Sandra Rayego-Mateos
- Vascular and Renal Translational Research Group, Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain;
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
| | - Macarena Orejudo
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Lucas Opazo-Ríos
- Renal, Vascular and Diabetes Research Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.O.-R.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Vanessa Marchant
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.L.); (V.M.); (A.D.); (S.M.)
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Laura Marquez-Exposito
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Antonio Tejera-Muñoz
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Juan F. Navarro-González
- Unidad de Investigación y Servicio de Nefrología, Hospital Universitario Nuestra Señora de Candelaria, 38010 Santa Cruz de Tenerife, Spain;
| | - Alejandra Droguett
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.L.); (V.M.); (A.D.); (S.M.)
| | - Alberto Ortiz
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
- Nephrology and Hypertension, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Jesús Egido
- Renal, Vascular and Diabetes Research Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain; (L.O.-R.); (J.E.)
- Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Sergio Mezzano
- Laboratorio de Nefrología, Facultad de Medicina, Universidad Austral de Chile, Valdivia 5090000, Chile; (C.L.); (V.M.); (A.D.); (S.M.)
| | - Raúl R. Rodrigues-Diez
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| | - Marta Ruiz-Ortega
- Red de Investigación Renal (REDINREN), Instituto de Salud Carlos III, 28029 Madrid, Spain; (M.O.); (L.M.-E.); (A.T.-M.); (A.O.)
- Cellular and Molecular Biology in Renal and Vascular Pathology Laboratory, Fundación Instituto de Investigación Sanitaria-Fundación Jiménez Díaz-Universidad Autónoma Madrid, 28040 Madrid, Spain
| |
Collapse
|
8
|
Cen L, Xing F, Xu L, Cao Y. Potential Role of Gene Regulator NFAT5 in the Pathogenesis of Diabetes Mellitus. J Diabetes Res 2020; 2020:6927429. [PMID: 33015193 PMCID: PMC7512074 DOI: 10.1155/2020/6927429] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/15/2020] [Accepted: 08/31/2020] [Indexed: 02/05/2023] Open
Abstract
Nuclear factor of activated T cells 5 (NFAT5), a Rel/nuclear factor- (NF-) κB family member, is the only known gene regulator of the mammalian adaptive response to osmotic stress. Exposure to elevated glucose increases the expression and nuclear translocation of NFAT5, as well as NFAT5-driven transcriptional activity in vivo and in vitro. Increased expression of NFAT5 is closely correlated with the progression of diabetes in patients. The distinct structure of NFAT5 governs its physiological and pathogenic roles, indicating its opposing functions. The ability of NFAT5 to maintain cell homeostasis and proliferation is impaired in patients with diabetes. NFAT5 promotes the formation of aldose reductase, pathogenesis of diabetic vascular complications, and insulin resistance. Additionally, NFAT5 activates inflammation at a very early stage of diabetes and induces persistent inflammation. Recent studies revealed that NFAT5 is an effective therapeutic target for diabetes. Here, we describe the current knowledge about NFAT5 and its relationship with diabetes, focusing on its diverse regulatory functions, and highlight the importance of this protein as a potential therapeutic target in patients with diabetes.
Collapse
Affiliation(s)
- Lusha Cen
- Department of Ophthalmology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Fengling Xing
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Liying Xu
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Cao
- Department of Dermatology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Youdian Rd. 54th, Hangzhou 310006, China
| |
Collapse
|
9
|
Mattner J, Mohammed JP, Fusakio ME, Giessler C, Hackstein CP, Opoka R, Wrage M, Schey R, Clark J, Fraser HI, Rainbow DB, Wicker LS. Genetic and functional data identifying Cd101 as a type 1 diabetes (T1D) susceptibility gene in nonobese diabetic (NOD) mice. PLoS Genet 2019; 15:e1008178. [PMID: 31199784 PMCID: PMC6568395 DOI: 10.1371/journal.pgen.1008178] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/07/2019] [Indexed: 12/16/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic multi-factorial disorder characterized by the immune-mediated destruction of insulin-producing pancreatic beta cells. Variations at a large number of genes influence susceptibility to spontaneous autoimmune T1D in non-obese diabetic (NOD) mice, one of the most frequently studied animal models for human disease. The genetic analysis of these mice allowed the identification of many insulin-dependent diabetes (Idd) loci and candidate genes, one of them being Cd101. CD101 is a heavily glycosylated transmembrane molecule which exhibits negative-costimulatory functions and promotes regulatory T (Treg) function. It is abundantly expressed on subsets of lymphoid and myeloid cells, particularly within the gastrointestinal tract. We have recently reported that the genotype-dependent expression of CD101 correlates with a decreased susceptibility to T1D in NOD.B6 Idd10 congenic mice compared to parental NOD controls. Here we show that the knockout of CD101 within the introgressed B6-derived Idd10 region increased T1D frequency to that of the NOD strain. This loss of protection from T1D was paralleled by decreased Gr1-expressing myeloid cells and FoxP3+ Tregs and an enhanced accumulation of CD4-positive over CD8-positive T lymphocytes in pancreatic tissues. As compared to CD101+/+ NOD.B6 Idd10 donors, adoptive T cell transfers from CD101-/- NOD.B6 Idd10 mice increased T1D frequency in lymphopenic NOD scid and NOD.B6 Idd10 scid recipients. Increased T1D frequency correlated with a more rapid expansion of the transferred CD101-/- T cells and a lower proportion of recipient Gr1-expressing myeloid cells in the pancreatic lymph nodes. Fewer of the Gr1+ cells in the recipients receiving CD101-/- T cells expressed CD101 and the cells had lower levels of IL-10 and TGF-β mRNA. Thus, our results connect the Cd101 haplotype-dependent protection from T1D to an anti-diabetogenic function of CD101-expressing Tregs and Gr1-positive myeloid cells and confirm the identity of Cd101 as Idd10.
Collapse
Affiliation(s)
- Jochen Mattner
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States of America
| | - Javid P. Mohammed
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States of America
| | - Michael E. Fusakio
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States of America
| | - Claudia Giessler
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Carl-Philipp Hackstein
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Robert Opoka
- Division of Immunobiology, Cincinnati Children's Hospital, Cincinnati, OH, United States of America
| | - Marius Wrage
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Regina Schey
- Mikrobiologisches Institut—Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen and Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Jan Clark
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust/MRC Building, Cambridge Institute for Medical Research, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Heather I. Fraser
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust/MRC Building, Cambridge Institute for Medical Research, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Daniel B. Rainbow
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust/MRC Building, Cambridge Institute for Medical Research, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Linda S. Wicker
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust/MRC Building, Cambridge Institute for Medical Research, NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Wellcome Trust Centre for Human Genetics, Nuffield Department of Medicine, National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Duan W, Ding Y, Yu X, Ma D, Yang B, Li Y, Huang L, Chen Z, Zheng J, Yang C. Metformin mitigates autoimmune insulitis by inhibiting Th1 and Th17 responses while promoting Treg production. Am J Transl Res 2019; 11:2393-2402. [PMID: 31105845 PMCID: PMC6511786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Accepted: 03/18/2019] [Indexed: 06/09/2023]
Abstract
Type 1 diabetes mellitus (T1DM) is still one of the major threats on global public health. This autoimmune condition is mainly caused by the imbalance of auto-reactive inflammatory effector T cells (Teffs) and protective regulatory T cells (Tregs). Therefore, inhibiting the development of Teffs and/or promoting Tregs provides a therapeutic strategy for preventing the development of T1DM. Pathways of energy metabolism have been shown to play a pivotal role in dictating the activation, differentiation and immune function of T cells. Studies have shown that inhibition of glycolysis suppresses the development of Th1 and Th17 cells, but promotes Treg production. AMP-activated protein kinase (AMPK) is a master sensor and regulator of cellular energy metabolism in mammals, which has also been demonstrated to interfere with T cell differentiation and effector function through inhibiting mammalian target of rapamycin (mTOR) and subsequent inhibition of glycolysis, and enhancement of lipid oxidation. In this study, we found that AMPK activator metformin suppresses T cell proliferation and inhibits the differentiation of Th1 and Th17 cells while promoting the development of Tregs in vitro in a dose-dependent manner. Treatment of NOD mice with metformin significantly mitigated autoimmune insulitis and substantially decreased the number of pro-inflammatory IFN-γ+ as well as IL17+ CD4 T cells in the spleens of NOD mice. However, a significantly increased percentage of regulatory IL-10+ and Foxp3+ CD4 T cells were seen. We provided a novel potential therapeutic method--by regulating T cell metabolism through targeting AMPK, to reduce the severity of autoimmune insulitis.
Collapse
Affiliation(s)
- Wu Duan
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yunchuan Ding
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Xuefeng Yu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Dongxia Ma
- Department of Allergy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Yi Li
- Department of Hematology and Oncology, Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Li Huang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Zhonghua Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
| | - Junmeng Zheng
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510120, China
| | - Chao Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhan, China
- Department of Cardiovascular Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen UniversityGuangzhou 510120, China
| |
Collapse
|
11
|
Singh K, Martinell M, Luo Z, Espes D, Stålhammar J, Sandler S, Carlsson PO. Cellular immunological changes in patients with LADA are a mixture of those seen in patients with type 1 and type 2 diabetes. Clin Exp Immunol 2019; 197:64-73. [PMID: 30843600 PMCID: PMC6591143 DOI: 10.1111/cei.13289] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/01/2019] [Indexed: 12/19/2022] Open
Abstract
There is currently scarce knowledge of the immunological profile of patients with latent autoimmune diabetes mellitus in the adult (LADA) when compared with healthy controls (HC) and patients with classical type 1 diabetes (T1D) and type 2 diabetes (T2D). The objective of this study was to investigate the cellular immunological profile of LADA patients and compare to HC and patients with T1D and T2D. All patients and age‐matched HC were recruited from Uppsala County. Peripheral blood mononuclear cells were isolated from freshly collected blood to determine the proportions of immune cells by flow cytometry. Plasma concentrations of the cytokine interleukin (IL)‐35 were measured by enzyme‐linked immunosorbent assay (ELISA). The proportion of CD11c+CD123– antigen‐presenting cells (APCs) was lower, while the proportions of CD11c+CD123+ APCs and IL‐35+ tolerogenic APCs were higher in LADA patients than in T1D patients. The proportion of CD3–CD56highCD16+ natural killer (NK) cells was higher in LADA patients than in both HC and T2D patients. The frequency of IL‐35+ regulatory T cells and plasma IL‐35 concentrations in LADA patients were similar to those in T1D and T2D patients, but lower than in HC. The proportion of regulatory B cells in LADA patients was higher than in healthy controls, T1D and T2D patients, and the frequency of IL‐35+ regulatory B cells was higher than in T1D patients. LADA presents a mixed cellular immunological pattern with features overlapping with both T1D and T2D.
Collapse
Affiliation(s)
- K Singh
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - M Martinell
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Z Luo
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - D Espes
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - J Stålhammar
- Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - S Sandler
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden
| | - P-O Carlsson
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.,Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
12
|
Xin GLL, Khee YP, Ying TY, Chellian J, Gupta G, Kunnath AP, Nammi S, Collet T, Hansbro PM, Dua K, Chellappan DK. Current Status on Immunological Therapies for Type 1 Diabetes Mellitus. Curr Diab Rep 2019; 19:22. [PMID: 30905013 DOI: 10.1007/s11892-019-1144-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) occurs when there is destruction of beta cells within the islets of Langerhans in the pancreas due to autoimmunity. It is considered a complex disease, and different complications can surface and worsen the condition if T1D is not managed well. Since it is an incurable disease, numerous treatments and therapies have been postulated in order to control T1D by balancing hyperglycemia control while minimizing hypoglycemic episodes. The purpose of this review is to primarily look into the current state of the available immunological therapies and their advantages for the treatment of T1D. RECENT FINDINGS Over the years, immunological therapy has become the center of attraction to treat T1D. Immunomodulatory approaches on non-antigens involving agents such as cyclosporine A, mycophenolate mofetil, anti-CD20, cytotoxic T cells, anti-TNF, anti-CD3, and anti-thymocyte globulin as well as immunomodulative approaches on antigens such as insulin, glutamic acid decarboxylase, and heat shock protein 60 have been studied. Aside from these two approaches, studies and trials have also been conducted on regulatory T cells, dendritic cells, interleukin 2, interleukin 4, M2 macrophages, and rapamycin/interleukin 2 combination therapy to test their effects on patients with T1D. Many of these agents have successfully suppressed T1D in non-obese diabetic (NOD) mice and in human trials. However, some have shown negative results. To date, the insights into the management of the immune system have been increasing rapidly to search for potential therapies and treatments for T1D. Nevertheless, some of the challenges are still inevitable. A lot of work and effort need to be put into the investigation on T1D through immunological therapy, particularly to reduce complications to improve and enhance clinical outcomes.
Collapse
Affiliation(s)
- Griselda Lim Loo Xin
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Yap Pui Khee
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Tan Yoke Ying
- School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Jestin Chellian
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Gaurav Gupta
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, 302017, India
| | - Anil Philip Kunnath
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Srinivas Nammi
- School of Science and Health, Western Sydney University, Sydney, NSW, 2751, Australia
- NICM Health Research Institute, Western Sydney University, Sydney, NSW, 2751, Australia
| | - Trudi Collet
- Innovative Medicines Group, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, 4059, Australia
| | - Philip Michael Hansbro
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, Newcastle, NSW, 2308, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney (UTS), Ultimo, NSW, 2007, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy, The University of Newcastle (UoN), Callaghan, Newcastle, NSW, 2308, Australia
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
13
|
Lee N, Kim D, Kim WU. Role of NFAT5 in the Immune System and Pathogenesis of Autoimmune Diseases. Front Immunol 2019; 10:270. [PMID: 30873159 PMCID: PMC6401628 DOI: 10.3389/fimmu.2019.00270] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 01/31/2019] [Indexed: 12/29/2022] Open
Abstract
The nuclear factor of activated T cells (NFAT5), also known as a tonicity-responsive enhancer-binding protein, was originally identified as a key transcription factor involved in maintaining cellular homeostasis against hypertonic and hyperosmotic environments. Although NFAT5 has been expressed and studied in various types of hyperosmolar tissues, evidence has emerged that NFAT5 plays a role in the development and activation of immune cells, especially T cells and macrophages. The immune-regulatory function of NFAT5 is achieved by inducing different target genes and different signaling pathways in both tonicity-dependent and -independent manners. Particularly in response to hyperosmotic stress, NFAT5 induces the generation of pathogenic TH17 cells and pro-inflammatory macrophages, contributing to autoimmune and inflammatory diseases. Meanwhile, with tonicity-independent stimuli, including activation of the Toll-like receptors and inflammatory cytokines, NFAT5 also can be activated and promotes immune cell survival, proliferation, migration, and angiogenesis. Moreover, under isotonic conditions, NFAT5 has been implicated in the pathogenesis of a variety of inflammatory and autoimmune diseases including rheumatoid arthritis. This review describes the current knowledge of NFAT5, focusing on its immune-regulatory functions, and it highlights the importance of NFAT5 as a novel therapeutic target for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Naeun Lee
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, South Korea
| | - Donghyun Kim
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, South Korea.,Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, South Korea.,Institute of Infectious Diseases, Seoul National University College of Medicine, Seoul, South Korea
| | - Wan-Uk Kim
- Center for Integrative Rheumatoid Transcriptomics and Dynamics, The Catholic University of Korea, Seoul, South Korea.,Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
14
|
Taherian M, Mahin Samadi P, Rastegar H, Faramarzi MA, Rostami-Nejad M, Yazdi MH, Rezaei-Tavirani M, Yazdi Z. An Overview on Probiotics as an Alternative Strategy for Prevention and Treatment of Human Diseases. IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH : IJPR 2019; 18:31-50. [PMID: 32802088 PMCID: PMC7393061 DOI: 10.22037/ijpr.2020.112232.13620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Probiotics are viable and useful microorganisms, which are beneficial factors for human and animal health by altering their microbial flora. Most of the probiotics belong to a large group of bacteria in the human gastrointestinal tract. There are several clinical shreds of evidence that show anti-carcinogenic effects of probiotics through altering digestive enzymes, inhibition of carcinogenic agents, and modulating the immune responses in experimental animals. Many studies have been performed to evaluate the potential effectiveness of probiotics in treating or preventing neurological diseases such as MS and novel treatment modality for T1D. The purpose of this study is to have an overview on probiotic microorganisms and to review the previous researches on the effects of probiotics on health through currently available literatures. The study was performed using following keywords; Probiotics, Cancer, Immune system, Multiple Sclerosis (MS) and Diabetes mellitus. PubMed/Medline, Clinicaltrials.gov, Ovid, Google Scholar, and Reaxcys databases used to find the full text of related articles. According to the current available data on probiotics and related health-promoting benefits, it seems that, consumption of probiotics can lead to the prevention and reduction the risk of cancer, diabetes, and multiple sclerosis. Although for the better and more decisive conclusion, there is a need to larger sample size clinical studies with more focus on the safety of these biological agents and their possible beneficial effects on different population.
Collapse
Affiliation(s)
- Mahdi Taherian
- Food and Drug Laboratory Research Center, Iran Food and Drug Organization (FDO), Ministry of Health and Medical Education (MOH), Tehran, Iran.
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Pariya Mahin Samadi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hossein Rastegar
- Food and Drug Cosmetic Research Center, Iran Food and Drug Organization (FDO), Ministry of Health and Medical Education (MOH), Tehran, Iran.
| | - Mohammad Ali Faramarzi
- Department of Pharmaceutical Biotechnology and Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Rostami-Nejad
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohammad Hossein Yazdi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran.
- Evidence-based Evaluation of Cost-Effectiveness and Clinical Outcomes, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, Tehran, Iran.
| | - Mostafa Rezaei-Tavirani
- Proteomics Research Center, Faculty of Paramedical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zeinab Yazdi
- Department of Medicine and epidemiology, School of veterinary Medicine, University of California, Davis, USA.
| |
Collapse
|
15
|
Kumar P, Saini S, Khan S, Surendra Lele S, Prabhakar BS. Restoring self-tolerance in autoimmune diseases by enhancing regulatory T-cells. Cell Immunol 2018; 339:41-49. [PMID: 30482489 DOI: 10.1016/j.cellimm.2018.09.008] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 09/14/2018] [Accepted: 09/28/2018] [Indexed: 12/16/2022]
Abstract
Self-tolerance, the state of unresponsiveness to self-tissues/antigens, is maintained through central and peripheral tolerance mechanisms, and a breach of these mechanisms leads to autoimmune diseases. Foxp3 + T-regulatory cells (Tregs) play an essential role in suppressing autoimmune response directed against self-antigens and thereby regulate self-tolerance. Natural Tregs are differentiated in the thymus on the basis of their higher TCR-affinity to self-antigens and migrate to the periphery where they maintain peripheral tolerance. In addition, extra-thymic differentiation of induced Tregs can occur in the periphery which can control abrupt immune responses under inflammatory conditions. A defect in Treg cell numbers and/or function is found to be associated with the development of autoimmune disease in several experimental models and human autoimmune diseases. Moreover, augmentation of Tregs has been shown to be beneficial in treating autoimmunity in preclinical models, and Treg based cellular therapy has shown initial promise in clinical trials. However, emerging studies have identified an unstable subpopulation of Tregs which expresses pro-inflammatory cytokines under both homeostatic and autoimmune conditions, as well as in ex vivo cultures. In addition, clinical translation of Treg cellular therapy is impeded by limitations such as lack of easier methods for selective expansion of Tregs and higher cost associated with GMP-facilities required for cell sorting, ex vivo expansion and infusion of ex vivo expanded Tregs. Here, we discuss the recent advances in molecular mechanisms regulating Treg differentiation, Foxp3 expression and lineage stability, the role of Tregs in the prevention of various autoimmune diseases, and critically review their clinical utility for treating human autoimmune diseases.
Collapse
Affiliation(s)
- Prabhakaran Kumar
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Shikha Saini
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Saad Khan
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Swarali Surendra Lele
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois - College of Medicine, Chicago, IL, USA.
| |
Collapse
|
16
|
Zhang Y, Lu S, Alahdal M, Gao H, Shen Y, Pan Y, Wu J, Xing Y, Jin L. Novel mutant P277 peptide VP to ameliorate atherogenic side-effects and to preserve anti-diabetic effects in NOD mice. Exp Cell Res 2018; 371:399-408. [PMID: 30179603 DOI: 10.1016/j.yexcr.2018.08.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/31/2018] [Indexed: 01/22/2023]
Abstract
P277 is a 24 amino-acids peptide, residues 437-460 of human heat shock protein 60 (HSP60). P277 or sequence repeated 6 × P277 was previously found showing potency preventive and therapeutic anti-diabetes functions in NOD mice, but aroused atherosclerosis due to the induction of anti-HSP65 autoantibodies as reported. To determine the intrinsic B epitope sequence, we screened P277 with pepscan method and then proved by detection of sera IgG from peptide fragments vaccinated mouse and rabbits. Results indicated HSP60 443-448 (ALLRCI) is potential intrinsic B epitope sequence of P277. We modified P277 by deleting the former three amino acids of ALLRCI (VP) or replacing these six with alanine (AP). The detection of serum lipid parameter in NOD mice and aorta endothelial damage levels in high-cholesterol diets fed rabbits demonstrated that VP induced higher anti-diabetes efficacy and caused less arteriosclerosis-liked diseases separately. With less TLR2/4 activation of dendritic cells and macrophages, VP treatment reduced Th1 related P277 specific pro-inflammatory cytokines production and increased regulatory immune responses both in vivo and in vitro. These results indicated that optimized VP peptide might serve as a promising candidate for mouse type 1 diabetes therapy.
Collapse
Affiliation(s)
- Yanfeng Zhang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Shiping Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Murad Alahdal
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Huashan Gao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Yumeng Shen
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Yi Pan
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Jie Wu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China; Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Yun Xing
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China
| | - Liang Jin
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Screening, Minigene Pharmacy Laboratory, School of Life Science and Technology, China Pharmaceutical University, China.
| |
Collapse
|
17
|
Wang X, Zheng P, Huang G, Yang L, Zhou Z. Dipeptidyl peptidase-4(DPP-4) inhibitors: promising new agents for autoimmune diabetes. Clin Exp Med 2018; 18:473-480. [PMID: 30022375 DOI: 10.1007/s10238-018-0519-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/08/2018] [Indexed: 12/16/2022]
Abstract
Dipeptidyl peptidase-4 (DPP-4) inhibitors constitute a novel class of anti-diabetic agents confirmed to improve glycemic control and preserve β-cell function in type 2 diabetes. Three major large-scale studies, EXAMINE, SAVOR-TIMI 53, and TECOS, have confirmed the cardiovascular safety profile of DPP-4 inhibitors. Based on these results, DPP-4 inhibitors have gained widespread use in type 2 diabetes treatment. It is currently unknown, however, whether DPP-4 inhibitors have similar therapeutic efficacy against autoimmune diabetes. Several in vitro and in vivo studies have addressed this issue, but the results remain controversial. In this review, we summarize experimental findings and preliminary clinical trial results, and identify potentially effective immune modulation targets of DPP-4 inhibitors for autoimmune diabetes.
Collapse
Affiliation(s)
- Xia Wang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China.,Department of Metabolism and Endocrinology, Hunan Provincial People's Hospital, Changsha, Hunan, China
| | - Peilin Zheng
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| | - Gan Huang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| | - Lin Yang
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China
| | - Zhiguang Zhou
- Department of Metabolism and Endocrinology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China. .,Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, Hunan, China.
| |
Collapse
|
18
|
Gamble A, Pepper AR, Bruni A, Shapiro AMJ. The journey of islet cell transplantation and future development. Islets 2018; 10:80-94. [PMID: 29394145 PMCID: PMC5895174 DOI: 10.1080/19382014.2018.1428511] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 02/06/2023] Open
Abstract
Intraportal islet transplantation has proven to be efficacious in preventing severe hypoglycemia and restoring insulin independence in selected patients with type 1 diabetes. Multiple islet infusions are often required to achieve and maintain insulin independence. Many challenges remain in clinical islet transplantation, including substantial islet cell loss early and late after islet infusion. Contributions to graft loss include the instant blood-mediated inflammatory reaction, potent host auto- and alloimmune responses, and beta cell toxicity from immunosuppressive agents. Protective strategies are being tested to circumvent several of these events including exploration of alternative transplantation sites, stem cell-derived insulin producing cell therapies, co-transplantation with mesenchymal stem cells or exploration of novel immune protective agents. Herein, we provide a brief introduction and history of islet cell transplantation, limitations associated with this procedure and methods to alleviate islet cell loss as a means to improve engraftment outcomes.
Collapse
Affiliation(s)
- Anissa Gamble
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Canada
| | - Andrew R. Pepper
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Canada
| | - Antonio Bruni
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Canada
| | - A. M. James Shapiro
- Alberta Diabetes Institute, University of Alberta, Edmonton, AB, Canada
- Clinical Islet Transplant Program, University of Alberta, Edmonton, AB, Canada
- Members of the Canadian National Transplant Research Project (CNTRP), Canada
| |
Collapse
|
19
|
Seyfarth J, Ahlert H, Rosenbauer J, Baechle C, Roden M, Holl RW, Mayatepek E, Meissner T, Jacobsen M. CISH promoter polymorphism effects on T cell cytokine receptor signaling and type 1 diabetes susceptibility. Mol Cell Pediatr 2018; 5:2. [PMID: 29411179 PMCID: PMC5801137 DOI: 10.1186/s40348-018-0080-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 01/22/2018] [Indexed: 11/25/2022] Open
Abstract
Background Impaired regulatory T cell immunity plays a central role in the development of type 1 diabetes (T1D). Interleukin-2 receptor (IL-2R) signaling is essential for regulatory T cells (TREG), and cytokine-inducible SH2-containing protein (CIS) regulates IL-2R signaling as a feedback inhibitor. Previous studies identified association of CISH promoter region single nucleotide polymorphisms (SNPs) with susceptibility to infectious diseases. Methods Here we analyzed allele frequencies of three CISH SNPs (i.e., rs809451, rs414171, rs2239751) in a study of T1D patients (n = 260, onset age < 5 years, duration > 10 years). Minor allele frequencies were compared to a control cohort of the 1000 Genomes Project. Assigned haplotypes were determined for effects on T1D manifestation and severity. Finally, the CISH haplotype influence on cytokine signaling and function was explored in T cells from healthy donors. Results We detected similar minor allele frequencies between T1D patients and the control cohort. T1D onset age, residual serum C-peptide level, and insulin requirement were comparable between different haplotypes. Only minor differences between the haplotypes were found for in vitro cytokine (i.e., IL-2, IL-7)-induced CIS mRNA expression. STAT5 phosphorylation was induced by IL-2 or IL-7, but no differences were found between the haplotypes. TREG purified from healthy donors with the two most common haplotypes showed similar capacity to inhibit heterologous effector T cells. Conclusions This study provides no evidence for an association of CISH promoter SNPs with susceptibility to T1D or severity of disease. In contrast to previous studies, no influence of different haplotypes on CIS mRNA expression or T cell-mediated functions was found. Electronic supplementary material The online version of this article (10.1186/s40348-018-0080-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Seyfarth
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), 85764, Munich, Neuherberg, Germany
| | - Heinz Ahlert
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Joachim Rosenbauer
- German Center for Diabetes Research (DZD), 85764, Munich, Neuherberg, Germany.,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225, Duesseldorf, Germany
| | - Christina Baechle
- German Center for Diabetes Research (DZD), 85764, Munich, Neuherberg, Germany.,Institute for Biometrics and Epidemiology, German Diabetes Center, Leibniz Center for Diabetes Research at Heinrich Heine University Düsseldorf, 40225, Duesseldorf, Germany
| | - Michael Roden
- German Center for Diabetes Research (DZD), 85764, Munich, Neuherberg, Germany.,Division of Endocrinology and Diabetology, Medical Faculty, Heinrich Heine University Düsseldorf, 40225, Düsseldorf, Germany.,Institute for Clinical Diabetology, German Diabetes Center, Leibniz Center at Heinrich-Heine University Düsseldorf, 40225, Düsseldorf, Germany
| | - Reinhard W Holl
- German Center for Diabetes Research (DZD), 85764, Munich, Neuherberg, Germany.,ZIBMT, Institute of Epidemiology and Medical Biometry, University of Ulm, 89081, Ulm, Germany
| | - Ertan Mayatepek
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany
| | - Thomas Meissner
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.,German Center for Diabetes Research (DZD), 85764, Munich, Neuherberg, Germany
| | - Marc Jacobsen
- Department of General Pediatrics, Neonatology, and Pediatric Cardiology, University Children's Hospital, Moorenstr. 5, 40225, Duesseldorf, Germany.
| |
Collapse
|
20
|
Abstract
Autoimmune diseases are characterized by the abnormal immune response against self-tissue, which are caused by the failure of nature immune homeostasis. Nature immune homeostasis represents the normal state of appropriate immune response to nonself-antigen and unresponsiveness to self-antigens. In normal situation, immune homeostasis is regulated by immunosuppressive signal and immunostimulating signal together. Accumulating data have demonstrated that the adenosinergic pathway played key roles in immune suppression and shield body from an excessive inflammatory response. The deficiency of adenosinergic pathway results in the imbalance between the pro- and anti-inflammatory activities. Thus, researchers pay much attention to the role of adenosinergic pathway in autoimmune diseases development. To date, accumulating data have suggested an important role of adenosinergic pathway-related molecules (i.e., CD39, CD73, ADA, adenosine receptors, etc.) in many types of human autoimmune diseases. More importantly, these findings have presented potential value of adenosinergic pathway analysis to be used for autoimmune diseases diagnosis, monitoring and treatment. In this review, we will provide a comprehensive description of the role of adenosinergic pathway in human autoimmune diseases.
Collapse
Affiliation(s)
- Ke Dong
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China.
| | - Zhao-Wei Gao
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui-Zhong Zhang
- Department of Clinical Diagnosis, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
21
|
Zhang W, Zhou L, Dang J, Zhang X, Wang J, Chen Y, Liang J, Li D, Ma J, Yuan J, Chen W, Zadeh HH, Olsen N, Zheng SG. Human Gingiva-Derived Mesenchymal Stem Cells Ameliorate Streptozoticin-induced T1DM in mice via Suppression of T effector cells and Up-regulating Treg Subsets. Sci Rep 2017; 7:15249. [PMID: 29127315 PMCID: PMC5681565 DOI: 10.1038/s41598-017-14979-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Accepted: 10/19/2017] [Indexed: 12/16/2022] Open
Abstract
There is yet no cure for type 1 diabetes (T1DM) so far. A significant body of evidence has demonstrated that bone marrow-derived mesenchymal stem cells (BMSCs) showed great potential in controlling T1DM. But there exists much difficulty in using BMSCs as a clinical therapy. We here test whether a new population of mesenchymal stem cells from human gingiva (GMSCs), which has many advantages over BMSCs, can delay or prevent progress of T1DM. GMSCs were adoptively transferred to multiple low-dose streptozotocin (STZ)-induced T1DM. Blood glucose levels and disease severities were analyzed. T cells subsets in blood, spleen and lymph nodes were detected dynamically by flow cytometry. GMSC distribution was dynamically analyzed. We found that infusion of GMSCs but not fibroblast cells significantly controlled blood glucose levels, delayed diabetes onset, ameliorated pathology scores in pancreas, and down-regulated production of IL-17 and IFN-γ in CD4+ and CD8+ T cells in spleens, pancreatic lymph nodes (pLN) and other lymph nodes. GMSCs also up-regulated the levels of CD4+ Treg induced in the periphery. Mechanismly, GMSCs could migrate to pancreas and local lymph node and function through CD39/CD73 pathway to regulate effector T cells. Thus, GMSCs show a potential promise in treating T1DM in the clinic.
Collapse
Affiliation(s)
- Wei Zhang
- Expert Workstation and Division of Endocrinology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan Province, China
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA
| | - Li Zhou
- Department of Clinical Immunology and Division of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA
| | - Junlong Dang
- Department of Clinical Immunology and Division of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA
| | - Ximei Zhang
- Department of Clinical Immunology and Division of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Julie Wang
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA
| | - Yanming Chen
- Department of Clinical Immunology and Division of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
| | - Jichao Liang
- Expert Workstation and Division of Endocrinology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan Province, China
| | - Dongqing Li
- Expert Workstation and Division of Endocrinology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan Province, China
| | - Jilin Ma
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA
- Division of Nephrology, Zhejiang Traditional Chinese Medicine and Western Medicine Hospital, Hangzhou, Zhejiang Province, China
| | - Jia Yuan
- Department of Clinical Immunology and Division of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Weiwen Chen
- Expert Workstation and Division of Endocrinology, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan Province, China.
| | - Homayoun H Zadeh
- Division of Periodontology, Diagnostic Sciences & Dental Hygiene, University of Southern California Ostrow School of Dentistry, Los Angeles, CA, 90089, USA
| | - Nancy Olsen
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA
| | - Song Guo Zheng
- Department of Clinical Immunology and Division of Endocrinology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong Province, China.
- Division of Rheumatology, The Pennsylvania State University, College of Medicine, 500 University Drive, Hershey, 17033, PA, USA.
| |
Collapse
|
22
|
Kowalewicz-Kulbat M, Locht C. BCG and protection against inflammatory and auto-immune diseases. Expert Rev Vaccines 2017; 16:1-10. [PMID: 28532186 DOI: 10.1080/14760584.2017.1333906] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Bacillus Calmette-Guérin (BCG) is the only available vaccine against tuberculosis. Although its protective efficacy against pulmonary tuberculosis is still under debate, it provides protection against other mycobacterial diseases. BCG is also an effective therapy against superficial bladder cancer and potentially decreases overall childhood mortality. Areas covered: The purpose of this paper is to provide a state-of-the-art summary of the beneficial effects of BCG in inflammatory and auto-immune diseases. As a strong inducer of Th1 type immunity, BCG has been reported to protect against atopic conditions, such as allergic asthma, a Th2-driven disorder. Its protective effect has been well documented in mice, but still awaits definitive evidence in humans. Similarly, murine studies have shown a protective effect of BCG against auto-immune diseases, such as multiple sclerosis and insulin-dependent diabetes, but studies in humans have come to conflicting conclusions. Expert commentary: Studies in mice have shown a beneficial effect of the BCG vaccine against allergic asthma, multiple sclerosis and diabetes. However, the understanding of its mechanism is still fragmentary and requires further in depth research. Some observational or intervention studies in humans have also suggested a beneficial effect, but definitive evidence for this requires confirmation in carefully conducted prospective studies.
Collapse
Affiliation(s)
- Magdalena Kowalewicz-Kulbat
- a Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology , University of Lodz , Lodz , Poland
| | - Camille Locht
- a Department of Immunology and Infectious Biology, Institute of Microbiology, Biotechnology and Immunology , University of Lodz , Lodz , Poland.,b Center for Infection and Immunity of Lille , Institut Pasteur de Lille , Lille , France.,c Center for Infection and Immunity of Lille , Inserm U1019 , Lille , France.,d Center for Infection and Immunity of Lille , CNRS UMR 8204 , Lille , France.,e Center for Infection and Immunity of Lille , Université Lille Nord de France , Lille , France
| |
Collapse
|
23
|
Daneshmandi S, Karimi MH, Pourfathollah AA. TGF-β engineered mesenchymal stem cells (TGF-β/MSCs) for treatment of Type 1 diabetes (T1D) mice model. Int Immunopharmacol 2017; 44:191-196. [PMID: 28110219 DOI: 10.1016/j.intimp.2017.01.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/07/2017] [Accepted: 01/10/2017] [Indexed: 12/28/2022]
Abstract
OBJECTIVE Mesenchymal stem cells (MSCs) are advantageous candidates for cell therapy of Type 1 diabetes (T1D). Considering immunomodulatory effect of MSC, in this study, we engineered MSCs with TGF-β gene to increase MSC potency for T1D therapy in mouse model. MATERIALS AND METHODS Two plans were designed for prevention and treatment of diabetes, respectively. In both of them, MSCs were injected i.v. and then, the diabetes features including serum insulin, blood glucose, glucose tolerance, splenocytes proliferation, and IL-4/IFN-γ production were evaluated. RESULTS TGF-β/MSCs treatment program resulted in the restoration of serum glucose after 3weeks, while prevention program could delay diabetes progression for two weeks. TGF-β/MSCs treatment elevated the levels of serum insulin and Th2 cytokine shift on 5th week after start of treatment. TGF-β/MSCs (and MSCs alone) could also diminish body weight and enhance mice survival comparing to untreated diabetic mice. CONCLUSION Engineered TGF-β/MSCs could restore some T1D features, including the regulation of adverse immune responses and could be potent tools for cell therapy of T1D comparing MSCs alone.
Collapse
Affiliation(s)
- Saeed Daneshmandi
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | | - Ali Akbar Pourfathollah
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
24
|
Impact of T-cell-specific Smad4 deficiency on the development of autoimmune diabetes in NOD mice. Immunol Cell Biol 2016; 95:287-296. [PMID: 27686408 PMCID: PMC5364321 DOI: 10.1038/icb.2016.98] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 09/12/2016] [Accepted: 09/22/2016] [Indexed: 12/27/2022]
Abstract
Type 1 diabetes results from autoimmune-mediated pancreatic beta-cell destruction and transforming growth factor-beta (TGF-β) is known to play a preventive role in type 1 diabetes in non-obese diabetic (NOD) mice. In this study, we investigated the role of Smad4, a key molecule for Smad-dependent TGF-β signaling, in T cells of NOD mice in the pathogenesis of autoimmune diabetes. We generated T-cell-specific Smad4 knockout (Smad4 tKO) NOD mice and assessed the pathological and immunological changes. Smad4 tKO showed earlier onset and increased incidence of diabetes than wild type (WT) NOD mice. Pathological features such as insulitis, anti-glutamic acid decarboxylase auto-antibody levels and serum IFN-γ levels were significantly increased in Smad4 tKO compared with WT NOD mice. Proportion and number of activated/memory CD4+ T cell were significantly increased in pancreatic lymph nodes of Smad4 tKO compared with WT NOD mice. However, the proportion and function of regulatory T cells was not different. Effector CD4+ T cells from Smad4 tKO were more resistant to suppression by regulatory T cells than effector cells from WT NOD mice. The proliferative potential of effector T cells from Smad4 tKO was significantly elevated compared with WT NOD mice, and activation of sterol regulatory element binding protein-1c (SREBP-1c) in T cells of Smad4 tKO NOD mice was correlated with this proliferative activity. We conclude that Smad4 deletion in T cells of NOD mice accelerated the development of autoimmune diabetes and increased the incidence of the disease by dysregulation of T cell activation at least in part via SREBP-1c activation.
Collapse
|
25
|
Qiao YC, Shen J, Hong XZ, Liang L, Bo CS, Sui Y, Zhao HL. Changes of regulatory T cells, transforming growth factor-beta and interleukin-10 in patients with type 1 diabetes mellitus: A systematic review and meta-analysis. Clin Immunol 2016; 170:61-9. [DOI: 10.1016/j.clim.2016.08.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 07/28/2016] [Accepted: 08/02/2016] [Indexed: 12/27/2022]
|
26
|
Sromova L, Busek P, Posova H, Potockova J, Skrha P, Andel M, Sedo A. The effect of dipeptidyl peptidase-IV inhibition on circulating T cell subpopulations in patients with type 2 diabetes mellitus. Diabetes Res Clin Pract 2016; 118:183-92. [PMID: 27388675 DOI: 10.1016/j.diabres.2016.06.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/23/2016] [Accepted: 06/05/2016] [Indexed: 02/06/2023]
Abstract
AIM To assess intraindividually the effects of DPP-IV inhibition on the subpopulations of immune cells in type 2 diabetes mellitus (DM2) patients during the course of treatment with sitagliptin. METHODS In this open label non-randomized observational study with a control group DM2 patients were examined before the initiation of the DPP-IV inhibitor administration (sitagliptin 100mg once daily) and then after 4weeks and 12months. Inhibition of the blood plasma DPP-IV enzymatic activity was determined by a chromogenic assay, the immunophenotyping of the blood cell subpopulations was performed using flow cytometry and blood plasma cytokine concentrations were quantified using an array-based multiplex ELISA. All parameters were evaluated in relation to the entry values in individual patients. RESULTS The blood plasma DPP-IV enzymatic activity was effectively inhibited during the sitagliptin treatment. A significant decrease of the proportion of Treg cells (to 86±31% (median±SD) of entry values, p=0.001) and an increase of Th1 cells (to 120±103% (median±SD) of entry values, p=0.004) were observed after 4weeks but not after one year of the sitagliptin treatment. No changes were observed in the ratio of CD4(+)/CD8(+) cells, in the quantity of NK and Th2 cells and blood plasma cytokine levels. CONCLUSIONS Sitagliptin treatment may cause temporary changes of the proportion of lymphocyte subpopulations in patients with DM2. The consequent deregulation of the immune system should be considered as a possible cause of the eventual side effects of long term DPP-IV inhibition.
Collapse
Affiliation(s)
- Lucie Sromova
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, U nemocnice 5, 12853 Prague 2, Czech Republic.
| | - Petr Busek
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, U nemocnice 5, 12853 Prague 2, Czech Republic.
| | - Helena Posova
- Institute of Immunology and Microbiology, First Faculty of Medicine, Charles University in Prague, Studnickova 7, 12000 Prague 2, Czech Republic.
| | - Jana Potockova
- 2nd Department of Internal Medicine, 3rd Faculty of Medicine and Faculty Hospital Královské Vinohrady, Charles University in Prague, Srobarova 1150, 10034 Prague 10, Czech Republic.
| | - Pavel Skrha
- 2nd Department of Internal Medicine, 3rd Faculty of Medicine and Faculty Hospital Královské Vinohrady, Charles University in Prague, Srobarova 1150, 10034 Prague 10, Czech Republic.
| | - Michal Andel
- 2nd Department of Internal Medicine, 3rd Faculty of Medicine and Faculty Hospital Královské Vinohrady, Charles University in Prague, Srobarova 1150, 10034 Prague 10, Czech Republic.
| | - Aleksi Sedo
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of Medicine, Charles University in Prague, U nemocnice 5, 12853 Prague 2, Czech Republic.
| |
Collapse
|
27
|
Veluthakal R, Sidarala V, Kowluru A. NSC23766, a Known Inhibitor of Tiam1-Rac1 Signaling Module, Prevents the Onset of Type 1 Diabetes in the NOD Mouse Model. Cell Physiol Biochem 2016; 39:760-7. [PMID: 27467102 DOI: 10.1159/000445666] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND/AIMS Type 1 diabetes (T1D) is characterized by absolute insulin deficiency due to destruction of pancreatic β-cells by cytokines (e.g., interleukin-1β; IL-1β) released by invading immune cells. The mechanisms by which these cytokines induce β-cell dysfunction remain poorly understood. Recent evidence suggests that excessive generation of reactive oxygen species (ROS) by the phagocyte-like NADPH oxidase2 (Nox2), along with significantly low levels of antioxidants in β-cells, drive them toward oxidative damage. Rac1, a small G-protein, is one of the members of Nox2 holoenzyme. We recently reported that NSC23766, a known inhibitor of Rac1, significantly attenuated cytokine-induced Nox2 activation and ROS generation in pancreatic islet β-cells in vitro. Herein, we determined the effects of NSC23766 (2.5 mg/kg/day, i.p/daily) on the development of diabetes in the NOD mouse, a model for T1D. METHODS Two groups of experimental animals (Balb/c and NOD mice) received NSC23766, while the two control groups received equal volume of saline. Body weights and blood glucose were measured every week for 34 weeks. Rac1 activation in pancreatic islets was measured by GLISA activation assay. Rac1 and CHOP expression was determined by Western Blotting. RESULTS Our findings indicate that administration of NSC23766 significantly prevented the development of spontaneous diabetes in the NOD mice. Furthermore, NSC23766 markedly suppressed Rac1 expression and activity and the endoplasmic reticulum stress (CHOP expression) in NOD islets. CONCLUSIONS Our findings provide the first evidence implicating the role of Tiam1-Rac1-Nox2 signaling pathway in the onset of spontaneous diabetes in the NOD mouse model.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Department of Molecular and Cellular Endocrinology, Diabetes and Metabolic Research Institute, City of Hope/BRI, Duarte, USA
| | | | | |
Collapse
|
28
|
Davis IC, Randell J, Davis SN. Immunotherapies currently in development for the treatment of type 1 diabetes. Expert Opin Investig Drugs 2016; 24:1331-41. [PMID: 26364507 DOI: 10.1517/13543784.2015.1075973] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Type I diabetes (T1DM) is an autoimmune disorder that affects the pancreas' ability to produce insulin. While T1DM can be managed using insulin therapy, patients face financial burden, serious complications and premature mortality, from the disease. Efforts have sought to define and ultimately suppress the underlying autoimmune attack that results in T1DM. AREAS COVERED The authors lay out promising immunosuppressive and immunomodulating drugs currently in development for T1DM and outline options for future immune treatment for the disorder. There have been several pharmacological strategies to combat the immune attack which will serve as the organization for this review: antigen-specific therapies; monoclonal antibodies; fusion proteins; alternate Treg affectors. EXPERT OPINION Immunosuppression and immunomodulation studies in T1DM demonstrated differing levels of slowing the progression of the immune attack; however, no single therapeutic approach provides a lasting halt of the immune attack and remission of the disease. The immunosuppressants (teplizumab, rituximab and abatacept) show promise in slowing the T1DM progressions for a specific subpopulation of T1DM patients, but this approach appears temporary and has the potential for unwanted side affects. Combination therapies may have the greatest chance of achieving durable cessation of the T1DM autoimmune attack.
Collapse
Affiliation(s)
- Ian C Davis
- a 1 University of Maryland School of Medicine , 3805 Greenway, Baltimore, MD 21218, USA
| | | | - Stephen N Davis
- c 3 University of Maryland School of Medicine , 22 South Greene St. N3W42, Baltimore, MD 21201, USA +1 41 0328 2488 ; +1 41 0328 8688 ;
| |
Collapse
|
29
|
Ferretti C, La Cava A. Adaptive immune regulation in autoimmune diabetes. Autoimmun Rev 2016; 15:236-41. [DOI: 10.1016/j.autrev.2015.11.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 11/19/2015] [Indexed: 12/16/2022]
|
30
|
Delic-Sarac M, Mutevelic S, Karamehic J, Subasic D, Jukic T, Coric J, Ridjic O, Panjeta M, Zunic L. ELISA Test for Analyzing of Incidence of Type 1 Diabetes Autoantibodies (GAD and IA2) in Children and Adolescents. Acta Inform Med 2016; 24:61-5. [PMID: 27041813 PMCID: PMC4789625 DOI: 10.5455/aim.2016.24.61-65] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 01/07/2016] [Indexed: 12/30/2022] Open
Abstract
Introduction: Anti GAD (antibodies on glutamic acid decarboxylase) and anti-IA2 antibodies (against tyrosine phosphatase), today, have their place and importance in diagnosis and prognosis of Type 1 diabetes. Huge number of patients with diabetes mellitus type 1 have these antibodies. Insulin antibodies are of critical importance in diagnosis of diabetes mellitus type 1 for pediatric population. Materials and methods: During 2014, the samples of 80 patients from Clinical Center University Sarajevo (CCUS) Pediatrics clinic’s, Endocrinology department were analyzed on anti-GAD and IA2 antibodies. The samples of serums of all patients were analyzed with ELISA tests using Anti GAD ELISA (IgG) kites from EUROIMMUN company. These are quantitative in vitro tests for human antibodies against decarboxylase of glutamine acid (GAD) and IA2, in serum or EDTA plasm. Results: During the period of one year, in CCUS’s Organizational unit, Institute for Clinical Immunology, 80 samples of patients with anti GAD and IA2 antibodies were analyzed. Out of total number of samples, 41 were male patients, or 51% and 39 female, or 49%. The youngest patient was born in 2012, and the oldest in 1993. Age average was represented by the patients born in 2001. Share of positive results for IA2 antibodies and GAD antibodies was 37% for IA2 antibodies, and 63% for GAD antibodies. Discussion: During an autoimmune – mediated Diabetes mellitus type 1 leads to T-cell mediated destruction of beta cells of pancreatic islets, reduced production of insulin and glucose metabolism. Studies have shown that these bodies are the most intense single marker for identifying persons with increased risk for diabetes development.
Collapse
Affiliation(s)
- Marina Delic-Sarac
- Department of Clinical Immunology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Selma Mutevelic
- Department of Biomedicine and Health, Medical Faculty, Josip Juraj Štrosmajer University, Osijek, Croatia
| | - Jasenko Karamehic
- Department of Clinical Immunology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Djemo Subasic
- Department of Clinical Immunology, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Tomislav Jukic
- Department of Biomedicine and Health, Medical Faculty, Josip Juraj Štrosmajer University, Osijek, Croatia
| | - Jozo Coric
- Department of Clinical Chemistry and Biochemistry, Clinical Center University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | - Ognjen Ridjic
- Sarajevo School of Science and Technology (SSST), Economics Department, International University of Sarajevo (IUS), Sarajevo, Bosnia and Herzegovina
| | - Mirsad Panjeta
- Faculty for Health Sciences, University of Zenica, Bosnia and Herzegovina
| | - Lejla Zunic
- Faculty for Health Sciences, University of Zenica, Bosnia and Herzegovina
| |
Collapse
|
31
|
LIU HUI, LIU HONGYANG, JIANG YINONG, LI NAN. Protective effect of thymoquinone improves cardiovascular function, and attenuates oxidative stress, inflammation and apoptosis by mediating the PI3K/Akt pathway in diabetic rats. Mol Med Rep 2016; 13:2836-42. [DOI: 10.3892/mmr.2016.4823] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 12/02/2015] [Indexed: 11/06/2022] Open
|
32
|
Staphylococcus enterotoxin B-induced T cells can efficaciously protect against type 1 diabetes in non-obese diabetic mice. Cent Eur J Immunol 2015; 40:292-9. [PMID: 26648772 PMCID: PMC4655378 DOI: 10.5114/ceji.2015.54589] [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: 03/19/2015] [Accepted: 05/26/2015] [Indexed: 11/17/2022] Open
Abstract
Type 1 diabetes (T1D), an autoimmune disease, can be protected against by natural killer T (NKT) cells. Several attempts demonstrate that NKT cells also can be produced by inducing with Staphylococcus enterotoxin B (SEB) in addition to its classical activated antigen α-galactosylceramide. Here, we examined a potential usage of SEB-induced T (SEB-T) cells for the treatment of T1D. We established the immunophenotypes of SEB-T cells via flow cytometry, and in consequence, enriched in CD8+NKT cells after SEB stimulated. A high level of transforming growth factor β (TGF-β), detected by RT-PCR and ELISA, was first observed to be expressed and secreted by these SEB-T cells. Mixed lymphocyte reactions indicated that SEB-T cells could not produce a response to mitogens and allogeneic lymphocyte, and can inhibit lymphocytes response to mitogens. In an animal model, our data indicated that infusion of SEB-T cells in non-obese diabetic mice was well tolerated and could ameliorate hyperglycemia and maintain the blood glucose nearly on normal level until sacrifice. Strikingly, infusion of SEB-T cells resulted in an increase in the serum TGF-β level. These data raise the possibility that SEB-T cells can protect against T1D, which is associated with NKT cells generated in these SEB-induced cells.
Collapse
|
33
|
He B, Li X, Yu H, Zhou Z. Therapeutic potential of umbilical cord blood cells for type 1 diabetes mellitus. J Diabetes 2015; 7:762-73. [PMID: 25799887 DOI: 10.1111/1753-0407.12286] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 02/25/2015] [Accepted: 03/09/2015] [Indexed: 12/18/2022] Open
Abstract
Type 1 diabetes mellitus (T1DM) is a chronic disorder that results from autoimmune-mediated destruction of pancreatic islet β-cells. However, to date, no conventional intervention has successfully treated the disease. The optimal therapeutic method for T1DM should effectively control the autoimmunity, restore immune homeostasis, preserve residual β-cells, reverse β-cell destruction, and protect the regenerated insulin-producing cells against re-attack. Umbilical cord blood is rich in regulatory T (T(reg)) cells and multiple types of stem cells that exhibit immunomodulating potential and hold promise in their ability to restore peripheral tolerance towards pancreatic islet β-cells through remodeling of immune responses and suppression of autoreactive T cells. Recently, reinfusion of autologous umbilical cord blood or immune cells from cord blood has been proposed as a novel therapy for T1DM, with the advantages of no risk to the donors, minimal ethical concerns, a low incidence of graft-versus-host disease and easy accessibility. In this review, we revisit the role of autologous umbilical cord blood or immune cells from cord blood-based applications for the treatment of T1DM.
Collapse
Affiliation(s)
- Binbin He
- Institute of Metabolism and Endocrinology, 2nd Xiangya Hospital, Central South University, Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Xia Li
- Institute of Metabolism and Endocrinology, 2nd Xiangya Hospital, Central South University, Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Haibo Yu
- Institute of Metabolism and Endocrinology, 2nd Xiangya Hospital, Central South University, Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| | - Zhiguang Zhou
- Institute of Metabolism and Endocrinology, 2nd Xiangya Hospital, Central South University, Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Changsha, China
| |
Collapse
|
34
|
TAK1 inhibition prevents the development of autoimmune diabetes in NOD mice. Sci Rep 2015; 5:14593. [PMID: 26459028 PMCID: PMC4602205 DOI: 10.1038/srep14593] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 09/02/2015] [Indexed: 01/29/2023] Open
Abstract
Transforming growth factor-β activated kinase-1 (TAK1, Map3k7), a member of the mitogen-activated protein kinase kinase kinase (MAP3K) family, is essential in innate and adaptive immune responses. We postulated that blockade of TAK1 would affect autoimmune diabetes in non-obese diabetic (NOD) mice. Administration of 5Z-7-oxozeaenol (OZ), a TAK1 inhibitor, decreased the incidence and delayed the onset of autoimmune diabetes in both spontaneous and accelerated (cyclophosphamide-induced) experimental NOD mice. OZ also reduced insulitis, preserved islet function, increased the expression of α1- antitrypsin (AAT), and severely inhibited NF-κB and JNK/AP-1 signaling pathways in immune organs and pancreatic tissues. Importantly, TAK1 inhibition by OZ elicited a Th1 to Th2 cytokine shift, and increased TGF-β1 production in cultured T lymphocytes supernatants. Systemic TAK1 inhibition induced immature DCs with lower expressions of MHC-II and CD86, attenuated DC-mediated T cell proliferation in allogeneic MLR, and production of cytokine IL-12p70 in DCs suspensions. The results indicate that TAK1 inhibition with OZ was associated with a lower frequency of autoimmune diabetes in NOD mice. The net effect of TAK1 inhibition in NOD mice therefore appears to be protective rather than disease-enhancing. Strategies targeting TAK1 specifically in NOD mice might prove useful for the treatment of autoimmune diabetes in general.
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW Although insulin is lifesaving and sustaining for those with type 1 diabetes (T1D), curing the disease will be much more complex than simple replacement of this hormone. T1D is an autoimmune disease orchestrated by T cells, and includes many arms of the immune response. Tremendous effort has gone into understanding its underlying immune, genetic, and environmental causes, and this progress has led to immunologically based clinical trials in T1D. This review will focus primarily on the clinical trials of the past decade that have attempted to translate these fundamental findings. RECENT FINDINGS It is known that powerful, nonspecific immune suppressants can temporarily slow the course of newly diagnosed T1D, yet are too toxic for long-term use, especially in children. Recent clinical trials to reverse T1D have used newly developed therapies that target specific components of the immune process believed to be involved with T1D. Although well justified and designed, no recent approach has resulted in clinical remission and few have had any effect on disease course. SUMMARY Advances in our fundamental understanding of how the human diabetes immune response is activated and regulated coupled with lessons that have been learnt from the most recent era of completed trials are guiding us toward the development of more effective, multipronged therapies to ablate diabetes autoimmunity, restore immune tolerance, preserve β cells, and, ultimately, improve the lives of patients with T1D.
Collapse
Affiliation(s)
- Mark R Rigby
- aIndiana University School of Medicine and Riley Hospital for Children at IU Health, Indianapolis, Indiana bImmune Tolerance Network, San Francisco, California, USA
| | | |
Collapse
|
36
|
Tersey SA, Colvin SC, Maier B, Mirmira RG. Protective effects of polyamine depletion in mouse models of type 1 diabetes: implications for therapy. Amino Acids 2014; 46:633-42. [PMID: 23846959 PMCID: PMC3888834 DOI: 10.1007/s00726-013-1560-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2013] [Accepted: 07/03/2013] [Indexed: 01/08/2023]
Abstract
The underlying pathophysiology of type 1 diabetes involves autoimmune-mediated islet inflammation, leading to dysfunction and death of insulin-secreting islet β cells. Recent studies have shown that polyamines, which are essential for mRNA translation, cellular replication, and the formation of the hypusine modification of eIF5A may play an important role in the progression of cellular inflammation. To test a role for polyamines in type 1 diabetes pathogenesis, we administered the ornithine decarboxylase inhibitor difluoromethylornithine to two mouse models--the low-dose streptozotocin model and the NOD model--to deplete intracellular polyamines, and administered streptozotocin to a third model, which was haploinsufficient for the gene encoding the hypusination enzyme deoxyhypusine synthase. Subsequent development of diabetes and/or glucose intolerance was monitored. In the low-dose streptozotocin mouse model, continuous difluoromethylornithine administration dose-dependently reduced the incidence of hyperglycemia and led to the preservation of β cell area, whereas in the NOD mouse model of autoimmune diabetes difluoromethylornithine reduced diabetes incidence by 50%, preserved β cell area and insulin secretion, led to reductions in both islet inflammation and potentially diabetogenic Th17 cells in pancreatic lymph nodes. Difluoromethylornithine treatment reduced hypusinated eIF5A levels in both immune cells and islets. Animals haploinsufficient for the gene encoding deoxyhypusine synthase were partially protected from hyperglycemia induced by streptozotocin. Collectively, these studies suggest that interventions that interfere with polyamine biosynthesis and/or eIF5A hypusination may represent viable approaches in the treatment of diabetes.
Collapse
MESH Headings
- Animals
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/drug therapy
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/drug therapy
- Diabetes Mellitus, Type 1/metabolism
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Eflornithine/administration & dosage
- Female
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Oxidoreductases Acting on CH-NH Group Donors/deficiency
- Oxidoreductases Acting on CH-NH Group Donors/metabolism
- Peptide Initiation Factors/metabolism
- Polyamines/metabolism
- RNA-Binding Proteins/metabolism
- Streptozocin/administration & dosage
- Eukaryotic Translation Initiation Factor 5A
Collapse
Affiliation(s)
- Sarah A. Tersey
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Stephanie C. Colvin
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Bernhard Maier
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Raghavendra G. Mirmira
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
- Departments of Medicine, Cellular and Integrative Physiology, and Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
37
|
Zhu H, Yu L, He Y, Wang B. Nonhuman primate models of type 1 diabetes mellitus for islet transplantation. J Diabetes Res 2014; 2014:785948. [PMID: 25389531 PMCID: PMC4217338 DOI: 10.1155/2014/785948] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Revised: 10/02/2014] [Accepted: 10/07/2014] [Indexed: 12/19/2022] Open
Abstract
Islet transplantation is an attractive treatment of type 1 diabetes mellitus (T1DM). Animal models of diabetes mellitus (DM) contribute a lot to the experimental studies of islet transplantation and to evaluations of isolated islet grafts for future clinical applications. Diabetic nonhuman primates (NHPs) represent the suitable models of DMs to better evaluate the effectiveness of islet transplantation, to assess new strategies for controlling blood glucose (BG), relieving immune rejection, or prolonging islet survival, and eventually to translate the preclinical data into tangible clinical practice. This review introduces some NHP models of DM, clarifies why and how the models should be used, and elucidates the usefulness and limitations of the models in islet transplantation.
Collapse
Affiliation(s)
- Haitao Zhu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Liang Yu
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Yayi He
- Department of Endocrinology, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an 710061, China
| | - Bo Wang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Medical College, Xi'an Jiaotong University, Xi'an 710061, China
- *Bo Wang:
| |
Collapse
|
38
|
Colvin SC, Maier B, Morris DL, Tersey SA, Mirmira RG. Deoxyhypusine synthase promotes differentiation and proliferation of T helper type 1 (Th1) cells in autoimmune diabetes. J Biol Chem 2013; 288:36226-35. [PMID: 24196968 DOI: 10.1074/jbc.m113.473942] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In type 1 diabetes, cytokines arising from immune cells cause islet β cell dysfunction even before overt hyperglycemia. Deoxyhypusine synthase catalyzes the crucial hypusine modification of the factor eIF5A, which promotes the translation of a subset of mRNAs involved in cytokine responses. Here, we tested the hypothesis that deoxyhypusine synthase and, secondarily, hypusinated eIF5A contribute to the pathogenesis of type 1 diabetes using the non-obese diabetic (NOD) mouse model. Pre-diabetic NOD mice that received injections of the deoxyhypusine inhibitor N1-guanyl-1,7-diaminoheptane (GC7) demonstrated significantly improved glucose tolerance, more robust insulin secretion, and reduced insulitis compared with control animals. Analysis of tissues from treated mice revealed selective reductions in diabetogenic T helper type 1 (Th1) cells in the pancreatic lymph nodes, a primary site of antigen presentation. Isolated mouse CD90.2(+) splenocytes stimulated in vitro with anti-CD3/anti-CD28 and IL-2 to mimic autoimmune T cell activation exhibited proliferation and differentiation of CD4(+) T cell subsets (Th1, Th17, and Treg), but those treated with the deoxyhypusine synthase inhibitor GC7 showed a dose-dependent block in T cell proliferation with selective reduction in Th1 cells, similar to that observed in NOD mice. Inhibition of deoxyhypusine synthase blocked post-transcriptional expression of CD25, the high affinity IL-2 receptor α chain. Our results suggest a previously unrecognized role for deoxyhypusine synthase in promoting T cell proliferation and differentiation via regulation of CD25. Inhibition of deoxyhypusine synthase may provide a strategy for reducing diabetogenic Th1 cells and preserving β cell function in type 1 diabetes.
Collapse
Affiliation(s)
- Stephanie C Colvin
- From the Department of Pediatrics and the Herman B. Wells Center for Pediatric Research and
| | | | | | | | | |
Collapse
|
39
|
da Rosa LC, Chiuso-Minicucci F, Zorzella-Pezavento SFG, França TGD, Ishikawa LLW, Colavite PM, Balbino B, Tavares LCB, Silva CL, Marques C, Ikoma MRV, Sartori A. Bacille Calmette-Guérin/DNAhsp65 prime-boost is protective against diabetes in non-obese diabetic mice but not in the streptozotocin model of type 1 diabetes. Clin Exp Immunol 2013; 173:430-7. [PMID: 23692306 DOI: 10.1111/cei.12140] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2013] [Indexed: 12/14/2022] Open
Abstract
Type I diabetes is a disease caused by autoimmune destruction of the beta cells in the pancreas that leads to a deficiency in insulin production. The aim of this study was to evaluate the prophylactic potential of a prime-boost strategy involving bacille Calmette-Guérin (BCG) and the pVAXhsp65 vaccine (BCG/DNAhsp65) in diabetes induced by streptozotocin (STZ) in C57BL/6 mice and also in spontaneous type 1 diabetes in non-obese diabetic (NOD) mice. BCG/DNAhsp65 vaccination in NOD mice determined weight gain, protection against hyperglycaemia, decreased islet inflammation, higher levels of cytokine production by the spleen and a reduced number of regulatory T cells in the spleen compared with non-immunized NOD mice. In the STZ model, however, there was no significant difference in the clinical parameters. Although this vaccination strategy did not protect mice in the STZ model, it was very effective in NOD mice. This is the first report demonstrating that a prime-boost strategy could be explored as an immunomodulatory procedure in autoimmune diseases.
Collapse
Affiliation(s)
- L C da Rosa
- Department of Microbiology and Immunology, Biosciences Institute, Universidade Estadual Paulista (UNESP), Botucatu, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Eric Gershwin M, Shoenfeld Y. Abul Abbas: An epitome of scholarship. J Autoimmun 2013; 45:1-6. [DOI: 10.1016/j.jaut.2013.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 07/14/2013] [Indexed: 11/29/2022]
|
41
|
Liu L, Perry AM, Cao W, Smith LM, Hsi ED, Liu X, Mo JQ, Dotlic S, Mosunjac M, Talmon G, Weisenburger DD, Fu K. Relationship between Rosai-Dorfman disease and IgG4-related disease: study of 32 cases. Am J Clin Pathol 2013; 140:395-402. [PMID: 23955459 DOI: 10.1309/ajcpfh0sj6yilxju] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES To assess the association between Rosai-Dorfman disease (RDD) and IgG4-related disease (IgG4-RD). METHODS We studied the number of IgG4-positive plasma cells and the IgG4/IgG ratio in 32 biopsy specimens (13 nodal, 19 extranodal) from 29 patients with RDD and compared the findings with those in IgG4-RD of the pancreas and reactive lymph nodes. We also assessed the number of FOXP3-positive regulatory T cells (Tregs) since they were reported to be increased in IgG4-RD. RESULTS We found that RDD cases had much lower numbers of IgG4-positive plasma cells and lower IgG4/IgG ratios compared with IgG4-RD but were similar to reactive lymph nodes. Furthermore, RDD had lower numbers of FOXP3-positive Tregs than did IgG4-RD. There were no significant differences in the number of IgG4-positive plasma cells and the IgG4/IgG ratio between the nodal and extranodal RDD cases. CONCLUSIONS Our study suggests that RDD does not belong in the spectrum of IgG4-RD.
Collapse
Affiliation(s)
- Liping Liu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha
| | | | - Wenfeng Cao
- Department of Pathology, Tianjin Medical University Cancer Hospital, Tianjin, China
| | - Lynette M. Smith
- Department of Biostatistics, University of Nebraska Medical Center, Omaha
| | - Eric D. Hsi
- Department of Clinical Pathology, Cleveland Clinic, Cleveland, OH
| | - Xiuli Liu
- Department of Clinical Pathology, Cleveland Clinic, Cleveland, OH
| | - Jun Q. Mo
- Division of Pediatric Pathology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH
| | - Snjezana Dotlic
- Department of Pathology, University Hospital Center Rebro, Zagreb, Croatia
| | - Marina Mosunjac
- Department of Pathology, Grady Memorial Hospital, Emory University School of Medicine, Atlanta, GA
| | - Geoffrey Talmon
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha
| | | | - Kai Fu
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha
| |
Collapse
|
42
|
Cabrera SM, Colvin SC, Tersey SA, Maier B, Nadler JL, Mirmira RG. Effects of combination therapy with dipeptidyl peptidase-IV and histone deacetylase inhibitors in the non-obese diabetic mouse model of type 1 diabetes. Clin Exp Immunol 2013; 172:375-82. [PMID: 23600825 DOI: 10.1111/cei.12068] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/10/2013] [Indexed: 12/24/2022] Open
Abstract
Type 1 diabetes (T1D) results from T helper type 1 (Th1)-mediated autoimmune destruction of insulin-producing β cells. Novel experimental therapies for T1D target immunomodulation, β cell survival and inflammation. We examined combination therapy with the dipeptidyl peptidase-IV inhibitor MK-626 and the histone deacetylase inhibitor vorinostat in the non-obese diabetic (NOD) mouse model of T1D. We hypothesized that combination therapy would ameliorate T1D by providing protection from β cell inflammatory destruction while simultaneously shifting the immune response towards immune-tolerizing regulatory T cells (T(regs)). Although neither mono- nor combination therapies with MK-626 and vorinostat caused disease remission in diabetic NOD mice, the combination of MK-626 and vorinostat increased β cell area and reduced the mean insulitis score compared to diabetic control mice. In prediabetic NOD mice, MK-626 monotherapy resulted in improved glucose tolerance, a reduction in mean insulitis score and an increase in pancreatic lymph node T(reg) percentage, and combination therapy with MK-626 and vorinostat increased pancreatic lymph node T(reg) percentage. We conclude that neither single nor combination therapies using MK-626 and vorinostat induce diabetes remission in NOD mice, but combination therapy appears to have beneficial effects on β cell area, insulitis and T(reg) populations. Combinations of vorinostat and MK-626 may serve as beneficial adjunctive therapy in clinical trials for T1D prevention or remission.
Collapse
Affiliation(s)
- S M Cabrera
- Department of Pediatrics and the Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
43
|
Chhabra P, Brayman KL. Stem cell therapy to cure type 1 diabetes: from hype to hope. Stem Cells Transl Med 2013; 2:328-336. [PMID: 23572052 PMCID: PMC3667565 DOI: 10.5966/sctm.2012-0116] [Citation(s) in RCA: 112] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 02/01/2013] [Indexed: 02/05/2023] Open
Abstract
Type 1 diabetes mellitus (T1D) is a chronic, multifactorial autoimmune disease that involves the progressive destruction of pancreatic β-cells, ultimately resulting in the loss of insulin production and secretion. The goal of clinical intervention is to prevent or arrest the onset and progression of autoimmunity, reverse β-cell destruction, and restore glycometabolic and immune homeostasis. Despite promising outcomes observed with islet transplantation and advancements in immunomodulatory therapies, the need for an effective cell replacement strategy for curing T1D still persists. Stem cell therapy offers a solution to the cited challenges of islet transplantation. While the regenerative potential of stem cells can be harnessed to make available a self-replenishing supply of glucose-responsive insulin-producing cells, their immunomodulatory properties may potentially be used to prevent, arrest, or reverse autoimmunity, ameliorate innate/alloimmune graft rejection, and prevent recurrence of the disease. Herein, we discuss the therapeutic potential of stem cells derived from a variety of sources for the cure of T1D, for example, embryonic stem cells, induced pluripotent stem cells, bone marrow-derived hematopoietic stem cells, and multipotent mesenchymal stromal cells derived from bone marrow, umbilical cord blood, and adipose tissue. The benefits of combinatorial approaches designed to ensure the successful clinical translation of stem cell therapeutic strategies, such as approaches combining effective stem cell strategies with islet transplantation, immunomodulatory drug regimens, and/or novel bioengineering techniques, are also discussed. To conclude, the application of stem cell therapy in the cure for T1D appears extremely promising.
Collapse
Affiliation(s)
- Preeti Chhabra
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| | - Kenneth L. Brayman
- Department of Surgery, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|