1
|
Zhang S, Ma J, Ma Y, Yi J, Wang B, Wang H, Yang Q, Zhang K, Yan X, Sun D, You J. Engineering Probiotics for Diabetes Management: Advances, Challenges, and Future Directions in Translational Microbiology. Int J Nanomedicine 2024; 19:10917-10940. [PMID: 39493275 PMCID: PMC11530765 DOI: 10.2147/ijn.s492651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Background Diabetes Mellitus (DM) is a substantial health concern worldwide, and its incidence is progressively escalating. Conventional pharmacological interventions frequently entail undesirable side effects, and while probiotics offer benefits, they are hindered by constraints such as diminished stability and effectiveness within the gastrointestinal milieu. Given these complications, the advent of bioengineered probiotics is a promising alternative for DM management. Aim of Review The objective of this review is to provide an exhaustive synthesis of the most recent studies on the use of engineered probiotics in the management of DM. This study aimed to clarify the mechanisms through which these probiotics function, evaluate their clinical effectiveness, and enhance public awareness of their prospective advantages in the treatment of DM. Key Scientific Concepts of Review Scholarly critiques have explored diverse methodologies of probiotic engineering, including physical alteration, bioenrichment, and genetic manipulation. These techniques augment the therapeutic potency of probiotics by ameliorating gut microbiota, fortifying the intestinal barrier, modulating metabolic pathways, and regulating immune responses. Such advancements have established engineered probiotics as a credible therapeutic strategy for DM, potentially providing enhanced results compared to conventional treatments.
Collapse
Affiliation(s)
- Shenghao Zhang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Beier Wang
- Department of Hepatobiliary-Pancreatic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077, People’s Republic of China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, People’s Republic of China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, People’s Republic of China
| | - Xiaoqing Yan
- The Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu hospital of Wenzhou Medical University, Yiwu, 322000, People’s Republic of China
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, 324000, People’s Republic of China
| |
Collapse
|
2
|
Zaongo SD, Zongo AW, Chen Y. Mechanisms underlying the development of type 1 diabetes in ART-treated people living with HIV: an enigmatic puzzle. Front Immunol 2024; 15:1470308. [PMID: 39257582 PMCID: PMC11383789 DOI: 10.3389/fimmu.2024.1470308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 08/12/2024] [Indexed: 09/12/2024] Open
Abstract
The immunopathogenesis of HIV infection remains poorly understood. Despite the widespread use of effective modern antiretroviral therapy (ART), people living with HIV (PLWH) are known to develop several comorbidities, including type 1 diabetes (T1DM). However, the etiology and critical mechanisms accounting for the onset of T1DM in the preceding context remain unknown. This article proposes to address this topic in order to provide further understanding and future research directions.
Collapse
Affiliation(s)
- Silvere D Zaongo
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Abel W Zongo
- College of Food Science and Technology, Zhejiang University of Technology, Hangzhou, China
| | - Yaokai Chen
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
3
|
Pearson JA, Hu Y, Peng J, Wong FS, Wen L. TLR5-deficiency controls dendritic cell subset development in an autoimmune diabetes-susceptible model. Front Immunol 2024; 15:1333967. [PMID: 38482010 PMCID: PMC10935730 DOI: 10.3389/fimmu.2024.1333967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 02/06/2024] [Indexed: 03/17/2024] Open
Abstract
Introduction The incidence of the autoimmune disease, type 1 diabetes (T1D), has been increasing worldwide and recent studies have shown that the gut microbiota are associated with modulating susceptibility to T1D. Toll-like receptor 5 (TLR5) recognizes bacterial flagellin and is widely expressed on many cells, including dendritic cells (DCs), which are potent antigen-presenting cells (APCs). TLR5 modulates susceptibility to obesity and alters metabolism through gut microbiota; however, little is known about the role TLR5 plays in autoimmunity, especially in T1D. Methods To fill this knowledge gap, we generated a TLR5-deficient non-obese diabetic (NOD) mouse, an animal model of human T1D, for study. Results We found that TLR5-deficiency led to a reduction in CD11c+ DC development in utero, prior to microbial colonization, which was maintained into adulthood. This was associated with a bias in the DC populations expressing CD103, with or without CD8α co-expression, and hyper-secretion of different cytokines, both in vitro (after stimulation) and directly ex vivo. We also found that TLR5-deficient DCs were able to promote polyclonal and islet antigen-specific CD4+ T cell proliferation and proinflammatory cytokine secretion. Interestingly, only older TLR5-deficient NOD mice had a greater risk of developing spontaneous T1D compared to wild-type mice. Discussion In summary, our data show that TLR5 modulates DC development and enhances cytokine secretion and diabetogenic CD4+ T cell responses. Further investigation into the role of TLR5 in DC development and autoimmune diabetes may give additional insights into the pathogenesis of Type 1 diabetes.
Collapse
Affiliation(s)
- James Alexander Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Youjia Hu
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
| | - Jian Peng
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
| | - F. Susan Wong
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
4
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM, Kuhn KA. Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022; 14. [PMID: 36288282 PMCID: PMC9804515 DOI: 10.1126/scitranslmed.abn5166 10.1126/scitranslmed.abn5166] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The mucosal origins hypothesis of rheumatoid arthritis (RA) proposes a central role for mucosal immune responses in the initiation or perpetuation of the systemic autoimmunity that occurs with disease. However, the connection between the mucosa and systemic autoimmunity in RA remains unclear. Using dual immunoglobulin A (IgA) and IgG family plasmablast-derived monoclonal autoantibodies obtained from peripheral blood of individuals at risk for RA, we identified cross-reactivity between RA-relevant autoantigens and bacterial taxa in the closely related families Lachnospiraceae and Ruminococcaceae. After generating bacterial isolates within the Lachnospiraceae/Ruminococcaceae genus Subdoligranulum from the feces of an individual, we confirmed monoclonal antibody binding and CD4+ T cell activation in individuals with RA compared to control individuals. In addition, when Subdoligranulum isolate 7 but not isolate 1 colonized germ-free mice, it stimulated TH17 cell expansion, serum RA-relevant IgG autoantibodies, and joint swelling reminiscent of early RA, with histopathology characterized by antibody deposition and complement activation. Systemic immune responses were likely due to mucosal invasion along with the generation of colon-isolated lymphoid follicles driving increased fecal and serum IgA by isolate 7, because B and CD4+ T cell depletion not only halted intestinal immune responses but also eliminated detectable clinical disease. In aggregate, these findings demonstrate a mechanism of RA pathogenesis through which a specific intestinal strain of bacteria can drive systemic autoantibody generation and joint-centered antibody deposition and immune activation.
Collapse
Affiliation(s)
- Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael R. Clay
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alex Ren Hsu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jennifer Seifert
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marie L. Feser
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA 98101
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth A. Bemis
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | - Daniel N. Frank
- Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - M. Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | | | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,Corresponding Author:
| |
Collapse
|
5
|
Chriswell ME, Lefferts AR, Clay MR, Hsu AR, Seifert J, Feser ML, Rims C, Bloom MS, Bemis EA, Liu S, Maerz MD, Frank DN, Demoruelle MK, Deane KD, James EA, Buckner JH, Robinson WH, Holers VM, Kuhn KA. Clonal IgA and IgG autoantibodies from individuals at risk for rheumatoid arthritis identify an arthritogenic strain of Subdoligranulum. Sci Transl Med 2022; 14:eabn5166. [PMID: 36288282 PMCID: PMC9804515 DOI: 10.1126/scitranslmed.abn5166] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The mucosal origins hypothesis of rheumatoid arthritis (RA) proposes a central role for mucosal immune responses in the initiation or perpetuation of the systemic autoimmunity that occurs with disease. However, the connection between the mucosa and systemic autoimmunity in RA remains unclear. Using dual immunoglobulin A (IgA) and IgG family plasmablast-derived monoclonal autoantibodies obtained from peripheral blood of individuals at risk for RA, we identified cross-reactivity between RA-relevant autoantigens and bacterial taxa in the closely related families Lachnospiraceae and Ruminococcaceae. After generating bacterial isolates within the Lachnospiraceae/Ruminococcaceae genus Subdoligranulum from the feces of an individual, we confirmed monoclonal antibody binding and CD4+ T cell activation in individuals with RA compared to control individuals. In addition, when Subdoligranulum isolate 7 but not isolate 1 colonized germ-free mice, it stimulated TH17 cell expansion, serum RA-relevant IgG autoantibodies, and joint swelling reminiscent of early RA, with histopathology characterized by antibody deposition and complement activation. Systemic immune responses were likely due to mucosal invasion along with the generation of colon-isolated lymphoid follicles driving increased fecal and serum IgA by isolate 7, because B and CD4+ T cell depletion not only halted intestinal immune responses but also eliminated detectable clinical disease. In aggregate, these findings demonstrate a mechanism of RA pathogenesis through which a specific intestinal strain of bacteria can drive systemic autoantibody generation and joint-centered antibody deposition and immune activation.
Collapse
Affiliation(s)
- Meagan E. Chriswell
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Adam R. Lefferts
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Michael R. Clay
- Department of Pathology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Alex Ren Hsu
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Jennifer Seifert
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Marie L. Feser
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Cliff Rims
- Benaroya Research Institute, Seattle, WA 98101
| | - Michelle S. Bloom
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - Elizabeth A. Bemis
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Sucai Liu
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | - Daniel N. Frank
- Division of Infectious Disease, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - M. Kristen Demoruelle
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kevin D. Deane
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | | | | | - William H. Robinson
- Division of Immunology and Rheumatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305
| | - V. Michael Holers
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045
| | - Kristine A. Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045,Corresponding Author:
| |
Collapse
|
6
|
Hu J, Zhang R, Zou H, Xie L, Zhou Z, Xiao Y. Latent Autoimmune Diabetes in Adults (LADA): From Immunopathogenesis to Immunotherapy. Front Endocrinol (Lausanne) 2022; 13:917169. [PMID: 35937817 PMCID: PMC9350734 DOI: 10.3389/fendo.2022.917169] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/26/2022] [Indexed: 11/26/2022] Open
Abstract
Latent autoimmune diabetes in adults (LADA) is a type of diabetes characterized by slow autoimmune damage of pancreatic β cells without insulin treatment in the early clinical stage. There are differences between LADA and classical type 1 diabetes (T1D) and type 2 diabetes (T2D) in genetic background, autoimmune response, rate of islet function decline, clinical metabolic characteristics, and so on. The disease progression and drug response of patients with LADA are closely related to the level of islet autoimmunity, thus exploring the pathogenesis of LADA is of great significance for its prevention and treatment. Previous studies reported that adaptive immunity and innate immunity play a critical role in the etiology of LADA. Recent studies have shown that the intestinal microbiota which impacts host immunity hugely, participates in the pathogenesis of LADA. In addition, the progression of autoimmune pancreatic β cell destruction in LADA is slower than in classical T1D, providing a wider window of opportunities for intervention. Therefore, therapies including antidiabetic drugs with immune-regulation effects and immunomodulators could contribute to promising interventions for LADA. We also shed light on potential interventions targeting the gut microbiota and gut-associated immunity, which may be envisaged to halt or delay the process of autoimmunity in LADA.
Collapse
|
7
|
Houeiss P, Luce S, Boitard C. Environmental Triggering of Type 1 Diabetes Autoimmunity. Front Endocrinol (Lausanne) 2022; 13:933965. [PMID: 35937815 PMCID: PMC9353023 DOI: 10.3389/fendo.2022.933965] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 06/20/2022] [Indexed: 12/15/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease in which pancreatic islet β cells are destroyed by immune cells, ultimately leading to overt diabetes. The progressive increase in T1D incidence over the years points to the role of environmental factors in triggering or accelerating the disease process which develops on a highly multigenic susceptibility background. Evidence that environmental factors induce T1D has mostly been obtained in animal models. In the human, associations between viruses, dietary habits or changes in the microbiota and the development of islet cell autoantibodies or overt diabetes have been reported. So far, prediction of T1D development is mostly based on autoantibody detection. Future work should focus on identifying a causality between the different environmental risk factors and T1D development to improve prediction scores. This should allow developing preventive strategies to limit the T1D burden in the future.
Collapse
Affiliation(s)
- Pamela Houeiss
- Laboratory Immunology of Diabetes, Department EMD, Cochin Institute, INSERMU1016, Paris, France
- Medical Faculty, Paris University, Paris, France
| | - Sandrine Luce
- Laboratory Immunology of Diabetes, Department EMD, Cochin Institute, INSERMU1016, Paris, France
- Medical Faculty, Paris University, Paris, France
| | - Christian Boitard
- Laboratory Immunology of Diabetes, Department EMD, Cochin Institute, INSERMU1016, Paris, France
- Medical Faculty, Paris University, Paris, France
- *Correspondence: Christian Boitard,
| |
Collapse
|
8
|
Chen Y, Wang M. New Insights of Anti-Hyperglycemic Agents and Traditional Chinese Medicine on Gut Microbiota in Type 2 Diabetes. Drug Des Devel Ther 2021; 15:4849-4863. [PMID: 34876807 PMCID: PMC8643148 DOI: 10.2147/dddt.s334325] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 11/19/2021] [Indexed: 12/11/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a widespread metabolic disease characterized by chronic hyperglycemia. Human microbiota, which is regarded as a “hidden organ”, plays an important role in the initiation and development of T2DM. In addition, anti-hyperglycemic agents and traditional Chinese medicine may affect the composition of gut microbiota and consequently improve glucose metabolism. However, the relationship between gut microbiota, T2DM and anti-hyperglycemic agents or traditional Chinese medicine is poorly understood. In this review, we summarized pre-clinical and clinical studies to elucidate the possible underlying mechanism. Some anti-hyperglycemic agents and traditional Chinese medicine may partly exert hypoglycemic effects by altering the gut microbiota composition in ways that reduce metabolic endotoxemia, maintain the integrity of intestinal mucosal barrier, promote the production of short-chain fatty acids (SCFAs), decrease trimethylamine-N-oxide (TMAO) and regulate bile acid metabolism. In conclusion, gut microbiota may provide some new therapeutic targets for treatment of patients with diabetes mellitus.
Collapse
Affiliation(s)
- Yanxia Chen
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Mian Wang
- Department of Endocrinology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050051, People's Republic of China
| |
Collapse
|
9
|
Toll-Like Receptors (TLRs) and their potential therapeutic applications in diabetic neuropathy. Int Immunopharmacol 2021; 102:108398. [PMID: 34863652 DOI: 10.1016/j.intimp.2021.108398] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/03/2021] [Accepted: 11/19/2021] [Indexed: 02/08/2023]
Abstract
One of the most common diabetic microvascular complications is diabetic neuropathy (DN). Immune cell infiltration in the peripheral nerve system (PNS), myelin loss, Schwann cell death, and axonal damage are all hallmarks of DN, which is currently believed to be a chronic inflammatory disease. Toll-like receptors (TLRs) are found in various types of nervous system cells, including Schwann cells, microglia, oligodendrocytes, astrocytes, and neurons. Proinflammatory mediators released at the end of TLR signal transduction can trigger an inflammatory response involving the nervous system. Studies on the association between TLRs and DN began as early as 2004. Since then, several studies have been conducted to assess the involvement of TLRs in the pathogenesis of DN. The focus of this review is to give a complete summary of the researches that have been done in this context, as well as an overview of the role of TLRs and their therapeutic applications in DN.
Collapse
|
10
|
Luce S, Guinoiseau S, Gadault A, Letourneur F, Nitschke P, Bras M, Vidaud M, Charneau P, Larger E, Colli ML, Eizirik DL, Lemonnier F, Boitard C. A Humanized Mouse Strain That Develops Spontaneously Immune-Mediated Diabetes. Front Immunol 2021; 12:748679. [PMID: 34721418 PMCID: PMC8551915 DOI: 10.3389/fimmu.2021.748679] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Accepted: 09/22/2021] [Indexed: 12/03/2022] Open
Abstract
To circumvent the limitations of available preclinical models for the study of type 1 diabetes (T1D), we developed a new humanized model, the YES-RIP-hB7.1 mouse. This mouse is deficient of murine major histocompatibility complex class I and class II, the murine insulin genes, and expresses as transgenes the HLA-A*02:01 allele, the diabetes high-susceptibility HLA-DQ8A and B alleles, the human insulin gene, and the human co-stimulatory molecule B7.1 in insulin-secreting cells. It develops spontaneous T1D along with CD4+ and CD8+ T-cell responses to human preproinsulin epitopes. Most of the responses identified in these mice were validated in T1D patients. This model is amenable to characterization of hPPI-specific epitopes involved in T1D and to the identification of factors that may trigger autoimmune response to insulin-secreting cells in human T1D. It will allow evaluating peptide-based immunotherapy that may directly apply to T1D in human and complete preclinical model availability to address the issue of clinical heterogeneity of human disease.
Collapse
Affiliation(s)
- Sandrine Luce
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France.,Medical Faculty, Paris University, Paris, France
| | - Sophie Guinoiseau
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France.,Medical Faculty, Paris University, Paris, France
| | - Alexis Gadault
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France.,Medical Faculty, Paris University, Paris, France
| | - Franck Letourneur
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France
| | | | - Marc Bras
- Medical Faculty, Paris University, Paris, France
| | - Michel Vidaud
- Biochemistry and Molecular Genetics Department, Cochin Hospital, Paris, France
| | - Pierre Charneau
- Molecular Virology and Vaccinology, Pasteur Institute, Paris, France
| | - Etienne Larger
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France.,Diabetology Department, Cochin Hospital, Paris, France
| | - Maikel L Colli
- Université Libre de Bruxelles (ULB) Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Decio L Eizirik
- Université Libre de Bruxelles (ULB) Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium.,Diabetes Center, Indiana Biosciences Research Institute (IBRI), Indianapolis, IN, United States
| | - François Lemonnier
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France.,Medical Faculty, Paris University, Paris, France
| | - Christian Boitard
- Laboratory Immunology of Diabetes, INSERMU1016, Department EMD, Cochin Institute, Paris, France.,Medical Faculty, Paris University, Paris, France.,Diabetology Department, Cochin Hospital, Paris, France
| |
Collapse
|
11
|
Zipris D. Visceral Adipose Tissue: A New Target Organ in Virus-Induced Type 1 Diabetes. Front Immunol 2021; 12:702506. [PMID: 34421908 PMCID: PMC8371384 DOI: 10.3389/fimmu.2021.702506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 07/19/2021] [Indexed: 12/14/2022] Open
Abstract
Type 1 diabetes (T1D) is a proinflammatory pathology that leads to the specific destruction of insulin producing β-cells and hyperglycaemia. Much of the knowledge about type 1 diabetes (T1D) has focused on mechanisms of disease progression such as adaptive immune cells and the cytokines that control their function, whereas mechanisms linked with the initiation of the disease remain unknown. It has been hypothesized that in addition to genetics, environmental factors play a pivotal role in triggering β-cell autoimmunity. The BioBreeding Diabetes Resistant (BBDR) and LEW1.WR1 rats have been used to decipher the mechanisms that lead to virus-induced T1D. Both animals develop β-cell inflammation and hyperglycemia upon infection with the parvovirus Kilham Rat Virus (KRV). Our earlier in vitro and in vivo studies indicated that KRV-induced innate immune upregulation early in the disease course plays a causal role in triggering β-cell inflammation and destruction. Furthermore, we recently found for the first time that infection with KRV induces inflammation in visceral adipose tissue (VAT) detectable as early as day 1 post-infection prior to insulitis and hyperglycemia. The proinflammatory response in VAT is associated with macrophage recruitment, proinflammatory cytokine and chemokine upregulation, endoplasmic reticulum (ER) and oxidative stress responses, apoptosis, and downregulation of adipokines and molecules that mediate insulin signaling. Downregulation of inflammation suppresses VAT inflammation and T1D development. These observations are strikingly reminiscent of data from obesity and type 2 diabetes (T2D) in which VAT inflammation is believed to play a causal role in disease mechanisms. We propose that VAT inflammation and dysfunction may be linked with the mechanism of T1D progression.
Collapse
Affiliation(s)
- Danny Zipris
- Innate Biotechnologies LLC, Denver, CO, United States
| |
Collapse
|
12
|
Gradisteanu Pircalabioru G, Corcionivoschi N, Gundogdu O, Chifiriuc MC, Marutescu LG, Ispas B, Savu O. Dysbiosis in the Development of Type I Diabetes and Associated Complications: From Mechanisms to Targeted Gut Microbes Manipulation Therapies. Int J Mol Sci 2021; 22:2763. [PMID: 33803255 PMCID: PMC7967220 DOI: 10.3390/ijms22052763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Globally, we are facing a worrying increase in type 1 diabetes mellitus (T1DM) incidence, with onset at younger age shedding light on the need to better understand the mechanisms of disease and step-up prevention. Given its implication in immune system development and regulation of metabolism, there is no surprise that the gut microbiota is a possible culprit behind T1DM pathogenesis. Additionally, microbiota manipulation by probiotics, prebiotics, dietary factors and microbiota transplantation can all modulate early host-microbiota interactions by enabling beneficial microbes with protective potential for individuals with T1DM or at high risk of developing T1DM. In this review, we discuss the challenges and perspectives of translating microbiome data into clinical practice. Nevertheless, this progress will only be possible if we focus our interest on developing numerous longitudinal, multicenter, interventional and double-blind randomized clinical trials to confirm their efficacy and safety of these therapeutic approaches.
Collapse
Affiliation(s)
| | - Nicolae Corcionivoschi
- Bacteriology Branch, Veterinary Sciences Division, Agri-Food and Biosciences Institute, Belfast BT9 5PX, UK;
| | - Ozan Gundogdu
- Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK;
| | - Mariana-Carmen Chifiriuc
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania; (G.G.P.); (L.G.M.); (B.I.)
- Academy of Romanian Scientists, 050045 Bucharest, Romania
| | | | - Bogdan Ispas
- Research Institute of University of Bucharest (ICUB), 300645 Bucharest, Romania; (G.G.P.); (L.G.M.); (B.I.)
| | - Octavian Savu
- “N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 2nd District, 020042 Bucharest, Romania;
- Department of Doctoral School, “Carol Davila” University of Medicine and Pharmacy, 5th District, 050474 Bucharest, Romania
| |
Collapse
|
13
|
Gut-Lung Dysbiosis Accompanied by Diabetes Mellitus Leads to Pulmonary Fibrotic Change through the NF-κB Signaling Pathway. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:838-856. [PMID: 33705752 DOI: 10.1016/j.ajpath.2021.02.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 01/25/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Growing evidence shows that the lungs are an unavoidable target organ of diabetic complications. However, the pathologic mechanisms of diabetic lung injury are still controversial. This study demonstrated the dysbiosis of the gut and lung microbiome, pulmonary alveolar wall thickening, and fibrotic change in streptozotocin-induced diabetic mice and antibiotic-induced gut dysbiosis mice compared with controls. In both animal models, the NF-κB signaling pathway was activated in the lungs. Enhanced pulmonary alveolar well thickening and fibrotic change appeared in the lungs of transgenic mice expressing a constitutively active NF-κB mutant compared with wild type. When lincomycin hydrochloride-induced gut dysbiosis was ameliorated by fecal microbiota transplant, enhanced inflammatory response in the intestine and pulmonary fibrotic change in the lungs were significantly decreased compared with lincomycin hydrochloride-treated mice. Furthermore, the application of fecal microbiota transplant and baicalin could also redress the microbial dysbiosis of the gut and lungs in streptozotocin-induced diabetic mice. Taken together, these data suggest that multiple as yet undefined factors related to microbial dysbiosis of gut and lungs cause pulmonary fibrogenesis associated with diabetes mellitus through an NF-κB signaling pathway.
Collapse
|
14
|
TLR3-Dependent Activation of TLR2 Endogenous Ligands via the MyD88 Signaling Pathway Augments the Innate Immune Response. Cells 2020; 9:cells9081910. [PMID: 32824595 PMCID: PMC7464415 DOI: 10.3390/cells9081910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/12/2020] [Accepted: 08/12/2020] [Indexed: 12/26/2022] Open
Abstract
The role of the adaptor molecule MyD88 is thought to be independent of Toll-like receptor 3 (TLR3) signaling. In this report, we demonstrate a previously unknown role of MyD88 in TLR3 signaling in inducing endogenous ligands of TLR2 to elicit innate immune responses. Of the various TLR ligands examined, the TLR3-specific ligand polyinosinic:polycytidylic acid (poly I:C), significantly induced TNF production and the upregulation of other TLR transcripts, in particular, TLR2. Accordingly, TLR3 stimulation also led to a significant upregulation of endogenous TLR2 ligands mainly, HMGB1 and Hsp60. By contrast, the silencing of TLR3 significantly downregulated MyD88 and TLR2 gene expression and pro-inflammatory IL1β, TNF, and IL8 secretion. The silencing of MyD88 similarly led to the downregulation of TLR2, IL1β, TNF and IL8, thus suggesting MyD88 to somehow act downstream of TLR3. Corroborating in vitro data, Myd88−/− knockout mice downregulated TNF, CXCL1; and phospho-p65 and phospho-IRF3 nuclear localization, upon poly I:C treatment in a mouse model of skin infection. Taken together, we identified a previously unknown role for MyD88 in the TLR3 signaling pathway, underlying the importance of TLRs and adapter protein interplay in modulating endogenous TLR ligands culminating in pro-inflammatory cytokine regulation.
Collapse
|
15
|
Zhou H, Sun L, Zhang S, Zhao X, Gang X, Wang G. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front Endocrinol (Lausanne) 2020; 11:125. [PMID: 32265832 PMCID: PMC7105744 DOI: 10.3389/fendo.2020.00125] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is a multifactorial autoimmune disease mediated by genetic, epigenetic, and environmental factors. In recent years, the emergence of high-throughput sequencing has allowed us to investigate the role of gut microbiota in the development of T1D. Significant changes in the composition of gut microbiome, also termed dysbiosis, have been found in subjects with clinical or preclinical T1D. However, whether the dysbiosis is a cause or an effect of the disease remains unclear. Currently, increasing evidence has supported a causal link between intestine microflora and T1D development. The current review will focus on recent research regarding the associations between intestine microbiome and T1D progression with an intention to evaluate the causality. We will also discuss the possible mechanisms by which imbalanced gut microbiota leads to the development of T1D.
Collapse
|
16
|
Yang J, Fukuchi KI. TIR-Domain-Containing Adaptor-Inducing Interferon- β (TRIF) Is Involved in Glucose Metabolism in Adipose Tissue through the Insulin/AKT Signaling Pathway. Int J Endocrinol 2020; 2020:6942307. [PMID: 33376487 PMCID: PMC7744180 DOI: 10.1155/2020/6942307] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/09/2020] [Accepted: 11/25/2020] [Indexed: 01/06/2023] Open
Abstract
Obesity significantly increases the risk of developing type 2 diabetes mellitus and other metabolic diseases. Obesity is associated with chronic low-grade inflammation in white adipose tissues, which is thought to play an essential role in developing insulin resistance. Many lines of evidence indicate that toll-like receptors (TLRs) and their downstream signaling pathways are involved in development of chronic low-grade inflammation and insulin resistance, which are associated with obesity. Mice lacking molecules positively involved in the TLR signaling pathways are generally protected from high-fat diet-induced inflammation and insulin resistance. In this study, we have determined the effects of genetic deficiency of toll/interleukin-1 receptor-domain-containing adaptor-inducing interferon-β (TRIF) on food intake, bodyweight, glucose metabolism, adipose tissue macrophage polarization, and insulin signaling in normal chow diet-fed mice to investigate the role of the TRIF-dependent TLR signaling in adipose tissue metabolism and inflammation. TRIF deficiency (TRIF-/-) increased food intake and bodyweight. The significant increase in bodyweight in TRIF-/- mice was discernible as early as 24 weeks of age and sustained thereafter. TRIF-/- mice showed impaired glucose tolerance in glucose tolerance tests, but their insulin tolerance tests were similar to those in TRIF+/+ mice. Although no difference was found in the epididymal adipose mass between the two groups, the percentage of CD206+ M2 macrophages in epididymal adipose tissue decreased in TRIF-/- mice compared with those in TRIF+/+ mice. Furthermore, activation of epididymal adipose AKT in response to insulin stimulation was remarkably diminished in TRIF-/- mice compared with TRIF+/+ mice. Our results indicate that the TRIF-dependent TLR signaling contributes to maintaining insulin/AKT signaling and M2 macrophages in epididymal adipose tissue under a normal chow diet and provide new evidence that TLR4-targeted therapies for type 2 diabetes require caution.
Collapse
Affiliation(s)
- Junling Yang
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, 1 Illini Drive, Peoria, IL 61605, USA
| | - Ken-Ichiro Fukuchi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine Peoria, 1 Illini Drive, Peoria, IL 61605, USA
| |
Collapse
|
17
|
Meng X, Zhou HY, Shen HH, Lufumpa E, Li XM, Guo B, Li BZ. Microbe-metabolite-host axis, two-way action in the pathogenesis and treatment of human autoimmunity. Autoimmun Rev 2019; 18:455-475. [PMID: 30844549 DOI: 10.1016/j.autrev.2019.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 12/14/2022]
Abstract
The role of microorganism in human diseases cannot be ignored. These microorganisms have evolved together with humans and worked together with body's mechanism to maintain immune and metabolic function. Emerging evidence shows that gut microbe and their metabolites open up new doors for the study of human response mechanism. The complexity and interdependence of these microbe-metabolite-host interactions are rapidly being elucidated. There are various changes of microbial levels in models or in patients of various autoimmune diseases (AIDs). In addition, the relevant metabolites involved in mechanism mainly include short-chain fatty acids (SCFAs), bile acids (BAs), and polysaccharide A (PSA). Meanwhile, the interaction between microbes and host genes is also a factor that must be considered. It has been demonstrated that human microbes are involved in the development of a variety of AIDs, including organ-specific AIDs and systemic AIDs. At the same time, microbes or related products can be used to remodel body's response to alleviate or cure diseases. This review summarizes the latest research of microbes and their related metabolites in AIDs. More importantly, it highlights novel and potential therapeutics, including fecal microbial transplantation, probiotics, prebiotics, and synbiotics. Nonetheless, exact mechanisms still remain elusive, and future research will focus on finding a specific strain that can act as a biomarker of an autoimmune disease.
Collapse
Affiliation(s)
- Xiang Meng
- School of Stomatology, Anhui Medical University, Hefei, Anhui, China
| | - Hao-Yue Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, 81 Meishan Road, Hefei, Anhui, China
| | - Hui-Hui Shen
- Department of Clinical Medicine, The second School of Clinical Medicine, Anhui Medical University, Anhui, Hefei, China
| | - Eniya Lufumpa
- Institute of Applied Health Research, University of Birmingham, Birmingham, UK
| | - Xiao-Mei Li
- Department of Rheumatology & Immunology, Anhui Provincial Hospital, Anhui, Hefei, China
| | - Biao Guo
- The Second Affiliated Hospital of Anhui Medical University, Anhui, Hefei, China
| | - Bao-Zhu Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China; Anhui Province Key Laboratory of Major Autoimmune Diseases, 81 Meishan Road, Hefei, Anhui, China.
| |
Collapse
|
18
|
Liu Y, Zheng W, Pan Y, Hu J. Low expression of miR-186-5p regulates cell apoptosis by targeting toll-like receptor 3 in high glucose-induced cardiomyocytes. J Cell Biochem 2018; 120:9532-9538. [PMID: 30506923 DOI: 10.1002/jcb.28229] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/15/2018] [Indexed: 12/20/2022]
Abstract
To investigate the effect and mechanism of microRNA-186-5p (miR-186-5p) on the apoptosis in high glucose (HG)-treated cardiomyocytes. Diabetic cardiomyopathy model was established in cardiomyocytes by stimulating with HG. The expressions of miR-186-5p and toll-like receptor 3 (TLR3) were detected by quantitative polymerase chain reaction or Western blot analysis, respectively. Apoptosis was detected in HG-treated cardiomyocytes by flow cytometry and Western blot analysis. The interaction between miR-186-5p and TLR3 was explored by bioinformatics analysis and luciferase activity assay. Results showed that miR-186-5p expression was downregulated in HG-treated cardiomyocytes and its overexpression reversed HG-induced apoptosis and cleaved caspase-3 protein expression. Moreover, TLR3 was indicated as a target of miR-186-5p and regulated by miR-186-5p. Knockdown of TLR3 suppressed HG-induced apoptosis and cleaved caspase-3 protein expression. Besides, restoration of TLR3 ablated the effect of miR-186-5p on cell apoptosis. Collectively, miR-186-5p attenuated HG-induced apoptosis by regulating TLR3 in cardiomyocytes, providing novel biomarker for treatment of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Ye Liu
- Electrocardial Center of the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Wei Zheng
- Electrocardial Center of the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yue Pan
- Electrocardial Center of the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Jing Hu
- Electrocardial Center of the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
19
|
Pearson JA, Agriantonis A, Wong FS, Wen L. Modulation of the immune system by the gut microbiota in the development of type 1 diabetes. Hum Vaccin Immunother 2018; 14:2580-2596. [PMID: 30156993 PMCID: PMC6314421 DOI: 10.1080/21645515.2018.1514354] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 07/29/2018] [Accepted: 08/17/2018] [Indexed: 02/08/2023] Open
Abstract
T1D is an autoimmune disease characterized by T cell-mediated destruction of insulin-producing β-cells in the pancreatic islets of Langerhans, resulting in hyperglycemia, with patients requiring lifelong insulin treatment. Many studies have shown that genetics alone are not sufficient for the increase in T1D incidence and thus other factors have been suggested to modify the disease risk. T1D incidence has sharply increased in the developed world, especially amongst youth. In Europe, T1D incidence is increasing at an annual rate of 3-4%. Increasing evidence shows that gut microbiota, as one of the environmental factors influencing diabetes development, play an important role in development of T1D. Here, we summarize the current knowledge about the relationship between the microbiota and T1D. We also discuss the possibility of T1D prevention by changing the composition of gut microbiota.
Collapse
Affiliation(s)
- James A. Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - Andrew Agriantonis
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - F. Susan Wong
- Diabetes Research Group, Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| |
Collapse
|
20
|
Luce S, Guinoiseau S, Gadault A, Letourneur F, Blondeau B, Nitschke P, Pasmant E, Vidaud M, Lemonnier F, Boitard C. Humanized Mouse Model to Study Type 1 Diabetes. Diabetes 2018; 67:1816-1829. [PMID: 29967002 DOI: 10.2337/db18-0202] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 06/15/2018] [Indexed: 12/15/2022]
Abstract
Key requirements in type 1 diabetes (T1D) are in setting up new assays as diagnostic biomarkers that will apply to prediabetes, likely T-cell assays, and in designing antigen-specific therapies to prevent T1D development. New preclinical models of T1D will be required to help with advancing both aims. By crossing mouse strains that lack either murine MHC class I and class II genes and insulin genes, we developed YES mice that instead express human HLA-A*02:01, HLA-DQ8, and insulin genes as transgenes. The metabolic and immune phenotype of YES mice is basically identical to that of the parental strains. YES mice remain insulitis and diabetes free up to 1 year of follow-up, maintain normoglycemia to an intraperitoneal glucose challenge in the long-term range, have a normal β-cell mass, and show normal immune responses to conventional antigens. This new model has been designed to evaluate adaptive immune responses to human insulin on a genetic background that recapitulates a human high-susceptibility HLA-DQ8 genetic background. Although insulitis free, YES mice develop T1D when challenged with polyinosinic-polycytidylic acid. They allow the characterization of preproinsulin epitopes recognized by CD8+ and CD4+ T cells upon immunization against human preproinsulin or during diabetes development.
Collapse
MESH Headings
- Adaptive Immunity/drug effects
- Aging
- Animals
- Autoimmune Diseases/immunology
- Autoimmune Diseases/metabolism
- Autoimmune Diseases/pathology
- Autoimmune Diseases/physiopathology
- Biomarkers/blood
- Biomarkers/metabolism
- Crosses, Genetic
- Diabetes Mellitus, Type 1/immunology
- Diabetes Mellitus, Type 1/metabolism
- Diabetes Mellitus, Type 1/pathology
- Diabetes Mellitus, Type 1/physiopathology
- Disease Models, Animal
- Disease Progression
- Female
- HLA-A2 Antigen/genetics
- HLA-A2 Antigen/metabolism
- HLA-DQ Antigens/blood
- HLA-DQ Antigens/genetics
- HLA-DQ Antigens/metabolism
- Humans
- Insulin/blood
- Insulin/genetics
- Insulin/metabolism
- Islets of Langerhans/immunology
- Islets of Langerhans/metabolism
- Islets of Langerhans/pathology
- Islets of Langerhans/physiopathology
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Mice, Knockout
- Mice, Transgenic
- Poly I-C/toxicity
- Prediabetic State/immunology
- Prediabetic State/metabolism
- Prediabetic State/pathology
- Prediabetic State/physiopathology
- Protein Precursors/blood
- Protein Precursors/genetics
- Protein Precursors/metabolism
- Specific Pathogen-Free Organisms
Collapse
Affiliation(s)
- Sandrine Luce
- INSERM U1016, Institut Cochin, Paris, France
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
| | - Sophie Guinoiseau
- INSERM U1016, Institut Cochin, Paris, France
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
| | - Alexis Gadault
- INSERM U1016, Institut Cochin, Paris, France
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
| | | | | | - Patrick Nitschke
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
| | - Eric Pasmant
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
- Service de Biochimie et Génétique Moléculaire, Hôpital COCHIN, Paris, France
| | - Michel Vidaud
- Service de Biochimie et Génétique Moléculaire, Hôpital COCHIN, Paris, France
| | - François Lemonnier
- INSERM U1016, Institut Cochin, Paris, France
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
| | - Christian Boitard
- INSERM U1016, Institut Cochin, Paris, France
- Faculté de Médecine René Descartes, Université Paris Descartes, Paris, France
| |
Collapse
|
21
|
Jubair WK, Hendrickson JD, Severs EL, Schulz HM, Adhikari S, Ir D, Pagan J, Anthony R, Robertson CE, Frank DN, Banda NK, Kuhn KA. Modulation of Inflammatory Arthritis in Mice by Gut Microbiota Through Mucosal Inflammation and Autoantibody Generation. Arthritis Rheumatol 2018; 70:1220-1233. [PMID: 29534332 PMCID: PMC6105374 DOI: 10.1002/art.40490] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 03/06/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Observations of microbial dysbiosis in patients with rheumatoid arthritis (RA) have raised interest in studying microbial-mucosal interactions as a potential trigger of RA. Using the murine collagen-induced arthritis (CIA) model, we undertook this study to test our hypothesis that microbiota modulate immune responses leading to autoimmune arthritis. METHODS CIA was induced by immunization of mice with type II collagen (CII) in adjuvant on days 0 and 21, with arthritis appearing on days 23 and 24. Intestinal microbiota were profiled by 16S ribosomal RNA sequencing every 7 days during the course of CIA, and intestinal mucosal changes were evaluated on days 14 and 35. Then, microbiota were depleted either early (7 days before immunization) or late (day 21 after immunization) by administration of broad-spectrum antibiotics. Disease severity, autoantibody and systemic cytokine production, and intestinal mucosal responses were monitored in the setting of microbial reduction. RESULTS Significant dysbiosis and mucosal inflammation occurred early in CIA, prior to visible arthritis, and continued to evolve during the course of disease. Depletion of the microbiota prior to the induction of CIA resulted in an ~40% reduction in disease severity and in significantly reduced levels of serum inflammatory cytokines and anti-CII antibodies. In intestinal tissue, production of interleukin-17A (IL-17A) and IL-22 was delayed. Unexpectedly, microbial depletion during the late phase of CIA resulted in a >50% decrease in disease severity. Anti-CII antibodies were mildly reduced but were significantly impaired in their ability to activate complement, likely due to altered glycosylation profiles. CONCLUSION These data support a model in which intestinal dysbiosis triggers mucosal immune responses that stimulate T and B cells that are key for the development of inflammatory arthritis.
Collapse
Affiliation(s)
- Widian K. Jubair
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Jason D. Hendrickson
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Erin L. Severs
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Hanna M. Schulz
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Sumitra Adhikari
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
| | - Diana Ir
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Jose Pagan
- Harvard University, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA
| | - Robert Anthony
- Harvard University, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA
| | - Charles E. Robertson
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Daniel N. Frank
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Nirmal K. Banda
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
| | - Kristine A. Kuhn
- Department of Medicine, Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| |
Collapse
|
22
|
Kuhn KA, Schulz HM, Regner EH, Severs EL, Hendrickson JD, Mehta G, Whitney AK, Ir D, Ohri N, Robertson CE, Frank DN, Campbell EL, Colgan SP. Bacteroidales recruit IL-6-producing intraepithelial lymphocytes in the colon to promote barrier integrity. Mucosal Immunol 2018; 11:357-368. [PMID: 28812548 PMCID: PMC5815964 DOI: 10.1038/mi.2017.55] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/12/2017] [Indexed: 02/04/2023]
Abstract
Interactions between the microbiota and distal gut are important for the maintenance of a healthy intestinal barrier; dysbiosis of intestinal microbial communities has emerged as a likely contributor to diseases that arise at the level of the mucosa. Intraepithelial lymphocytes (IELs) are positioned within the epithelial barrier, and in the small intestine they function to maintain epithelial homeostasis. We hypothesized that colon IELs promote epithelial barrier function through the expression of cytokines in response to interactions with commensal bacteria. Profiling of bacterial 16S ribosomal RNA revealed that candidate bacteria in the order Bacteroidales are sufficient to promote IEL presence in the colon that in turn produce interleukin-6 (IL-6) in a MyD88 (myeloid differentiation primary response 88)-dependent manner. IEL-derived IL-6 is functionally important in the maintenance of the epithelial barrier as IL-6-/- mice were noted to have increased paracellular permeability, decreased claudin-1 expression, and a thinner mucus gel layer, all of which were reversed by transfer of IL-6+/+ IELs, leading to protection of mice in response to Citrobacter rodentium infection. Therefore, we conclude that microbiota provide a homeostatic role for epithelial barrier function through regulation of IEL-derived IL-6.
Collapse
Affiliation(s)
- Kristine A. Kuhn
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Hanna M. Schulz
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Emilie H. Regner
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
| | - Erin L. Severs
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Jason D. Hendrickson
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Gaurav Mehta
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Alyssa K. Whitney
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Neha Ohri
- Division of Rheumatology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Charles E. Robertson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Daniel N. Frank
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora CO
| | - Eric L. Campbell
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| | - Sean P. Colgan
- Division of Gastroenterology, University of Colorado School of Medicine, Aurora CO
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora CO
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW Despite immense research efforts, type 1 diabetes (T1D) remains an autoimmune disease without a known trigger or approved intervention. Over the last three decades, studies have primarily focused on delineating the role of the adaptive immune system in the mechanism of T1D. The discovery of Toll-like receptors in the 1990s has advanced the knowledge on the role of the innate immune system in host defense as well as mechanisms that regulate adaptive immunity including the function of autoreactive T cells. RECENT FINDINGS Recent investigations suggest that inflammation plays a key role in promoting a large number of autoimmune disorders including T1D. Data from the LEW1.WR1 rat model of virus-induced disease and the RIP-B7.1 mouse model of diabetes suggest that innate immune signaling plays a key role in triggering disease progression. There is also evidence that innate immunity may be involved in the course of T1D in humans; however, a small number of clinical trials have shown that interfering with the function of the innate immune system following disease onset exerts only a modest effect on β-cell function. The data implying that innate immune pathways are linked with mechanisms of islet autoimmunity hold great promise for the identification of novel disease pathways that may be harnessed for clinical intervention. Nevertheless, more work needs to be done to better understand mechanisms by which innate immunity triggers β-cell destruction and assess the therapeutic value in blocking innate immunity for diabetes prevention.
Collapse
Affiliation(s)
- James C Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, 80045, USA
| | - Danny Zipris
- Innate Biotechnologies LLC, Denver, CO, 80231, USA.
| |
Collapse
|
24
|
Dai D, Prussin AJ, Marr LC, Vikesland PJ, Edwards MA, Pruden A. Factors Shaping the Human Exposome in the Built Environment: Opportunities for Engineering Control. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2017; 51:7759-7774. [PMID: 28677960 DOI: 10.1021/acs.est.7b01097] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The "exposome" is a term describing the summation of one's lifetime exposure to microbes and chemicals. Such exposures are now recognized as major drivers of human health and disease. Because humans spend ∼90% of their time indoors, the built environment exposome merits particular attention. Herein we utilize an engineering perspective to advance understanding of the factors that shape the built environment exposome and its influence on human wellness and disease, while simultaneously informing development of a framework for intentionally controlling the exposome to protect public health. Historically, engineers have been focused on controlling chemical and physical contaminants and on eradicating microbes; however, there is a growing awareness of the role of "beneficial" microbes. Here we consider the potential to selectively control the materials and chemistry of the built environment to positively influence the microbial and chemical components of the indoor exposome. Finally, we discuss research gaps that must be addressed to enable intentional engineering design, including the need to define a "healthy" built environment exposome and how to control it.
Collapse
Affiliation(s)
- Dongjuan Dai
- Via Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University , Blacksburg Virginia 24061, United States
| | - Aaron J Prussin
- Via Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University , Blacksburg Virginia 24061, United States
| | - Linsey C Marr
- Via Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University , Blacksburg Virginia 24061, United States
| | - Peter J Vikesland
- Via Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University , Blacksburg Virginia 24061, United States
| | - Marc A Edwards
- Via Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University , Blacksburg Virginia 24061, United States
| | - Amy Pruden
- Via Department of Civil and Environmental Engineering, Virginia Polytechnic Institute and State University , Blacksburg Virginia 24061, United States
| |
Collapse
|
25
|
Chen K, Chen H, Faas MM, de Haan BJ, Li J, Xiao P, Zhang H, Diana J, de Vos P, Sun J. Specific inulin-type fructan fibers protect against autoimmune diabetes by modulating gut immunity, barrier function, and microbiota homeostasis. Mol Nutr Food Res 2017; 61. [PMID: 28218451 DOI: 10.1002/mnfr.201601006] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 01/07/2017] [Accepted: 02/02/2017] [Indexed: 12/30/2022]
Abstract
SCOPE Dietary fibers capable of modifying gut barrier and microbiota homeostasis affect the progression of type 1 diabetes (T1D). Here, we aim to compare modulatory effects of inulin-type fructans (ITFs), natural soluble dietary fibers with different degrees of fermentability from chicory root, on T1D development in nonobese diabetic mice. METHODS AND RESULTS Female nonobese diabetic mice were weaned to long- and short-chain ITFs [ITF(l) and ITF(s), 5%] supplemented diet up to 24 weeks. T1D incidence, pancreatic-gut immune responses, gut barrier function, and microbiota composition were analyzed. ITF(l) but not ITF(s) supplementation dampened the incidence of T1D. ITF(l) promoted modulatory T-cell responses, as evidenced by increased CD25+ Foxp3+ CD4+ regulatory T cells, decreased IL17A+ CD4+ Th17 cells, and modulated cytokine production profile in the pancreas, spleen, and colon. Furthermore, ITF(l) suppressed NOD like receptor protein 3 caspase-1-p20-IL-1β inflammasome in the colon. Expression of barrier reinforcing tight junction proteins occludin and claudin-2, antimicrobial peptides β-defensin-1, and cathelicidin-related antimicrobial peptide as well as short-chain fatty acid production were enhanced by ITF(l). Next-generation sequencing analysis revealed that ITF(l) enhanced Firmicutes/Bacteroidetes ratio to an antidiabetogenic balance and enriched modulatory Ruminococcaceae and Lactobacilli. CONCLUSION Our data demonstrate that ITF(l) but not ITF(s) delays the development of T1D via modulation of gut-pancreatic immunity, barrier function, and microbiota homeostasis.
Collapse
Affiliation(s)
- Kang Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
| | - Hao Chen
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
| | - Marijke M Faas
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart J de Haan
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jiahong Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
| | - Ping Xiao
- Institute of Clinical Medical Research, Affiliated Foshan Hospital of Sun Yat-sen University, Foshan, P.R. China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China
| | - Julien Diana
- Sorbonne Paris Cité, Université Paris Descartes, Paris, France.,Institut National de la Santé et de la Recherche Médicale (INSERM), Unité 1151, Institut Necker-Enfants Malades (INEM), Centre National de la Recherche ITF(l)enctifique, Paris, France
| | - Paul de Vos
- Immunoendocrinology, Division of Medical Biology, Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jia Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, P. R. China.,School of Medicine, Jiangnan University, Wuxi, P.R. China
| |
Collapse
|
26
|
Abstract
The gastrointestinal system represents one of the largest interfaces between the human internal microenvironment and the external world. This system harbors trillions of commensal bacteria that reside in symbiosis with the host. Intestinal bacteria play a crucial role in maintaining systemic and intestinal immune and metabolic homeostasis because of their effect on nutrient absorption and immune development and function. Recently, altered gut bacterial composition (dysbiosis) was hypothesized to be involved in mechanisms through which islet autoimmunity is triggered. Evidence from animal models indicates that alterations in the gut bacterial composition precede disease onset, thus implicating a causal role for the gut microbiome in islet destruction. However, it remains unclear whether dysbiosis is directly linked to the mechanisms of human type 1 diabetes (T1D). In this review, we discuss data implicating the gut microbiota in disease progression with an emphasis on our recent studies performed in humans and in rodent models of T1D.
Collapse
Affiliation(s)
- James C Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO, 80045, USA
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO, 80045, USA.
| |
Collapse
|
27
|
Bugging inflammation: role of the gut microbiota. Clin Transl Immunology 2016; 5:e72. [PMID: 27195115 PMCID: PMC4855262 DOI: 10.1038/cti.2016.12] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 03/10/2016] [Accepted: 03/10/2016] [Indexed: 12/12/2022] Open
Abstract
The advent of vaccination and improved hygiene have eliminated many of the deadly infectious pathogens in developed nations. However, the incidences of inflammatory diseases, such as inflammatory bowel disease, asthma, obesity and diabetes are increasing dramatically. Research in the recent decades revealed that it is indeed the lack of early childhood microbial exposure, increase use of antibiotics, as well as increase consumption of processed foods high in carbohydrates and fats, and lacking fibre, which wreak havoc on the proper development of immunity and predispose the host to elevated inflammatory conditions. Although largely unexplored and under-appreciated until recent years, these factors impact significantly on the composition of the gut microbiota (a collection of microorganisms that live within the host mucosal tissue) and inadvertently play intricate and pivotal roles in modulating an appropriate host immune response. The suggestion that shifts in the composition of host microbiota is a risk factor for inflammatory disease raises an exciting opportunity whereby the microbiota may also present as a potential modifiable component or therapeutic target for inflammatory diseases. This review provides insights into the interactions between the microbiota and the immune system, how these affect disease phenotypes, and explore current and emerging therapies that target the gut microbiota as potential treatment for inflammatory diseases.
Collapse
|
28
|
Mellado-Gil JM, Jiménez-Moreno CM, Martin-Montalvo A, Alvarez-Mercado AI, Fuente-Martin E, Cobo-Vuilleumier N, Lorenzo PI, Bru-Tari E, Herrera-Gómez IDG, López-Noriega L, Pérez-Florido J, Santoyo-López J, Spyrantis A, Meda P, Boehm BO, Quesada I, Gauthier BR. PAX4 preserves endoplasmic reticulum integrity preventing beta cell degeneration in a mouse model of type 1 diabetes mellitus. Diabetologia 2016; 59:755-65. [PMID: 26813254 PMCID: PMC4779135 DOI: 10.1007/s00125-016-3864-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/17/2015] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS A strategy to enhance pancreatic islet functional beta cell mass (BCM) while restraining inflammation, through the manipulation of molecular and cellular targets, would provide a means to counteract the deteriorating glycaemic control associated with diabetes mellitus. The aims of the current study were to investigate the therapeutic potential of such a target, the islet-enriched and diabetes-linked transcription factor paired box 4 (PAX4), to restrain experimental autoimmune diabetes (EAD) in the RIP-B7.1 mouse model background and to characterise putative cellular mechanisms associated with preserved BCM. METHODS Two groups of RIP-B7.1 mice were genetically engineered to: (1) conditionally express either PAX4 (BPTL) or its diabetes-linked mutant variant R129W (mutBPTL) using doxycycline (DOX); and (2) constitutively express luciferase in beta cells through the use of RIP. Mice were treated or not with DOX, and EAD was induced by immunisation with a murine preproinsulin II cDNA expression plasmid. The development of hyperglycaemia was monitored for up to 4 weeks following immunisation and alterations in the BCM were assessed weekly by non-invasive in vivo bioluminescence intensity (BLI). In parallel, BCM, islet cell proliferation and apoptosis were evaluated by immunocytochemistry. Alterations in PAX4- and PAX4R129W-mediated islet gene expression were investigated by microarray profiling. PAX4 preservation of endoplasmic reticulum (ER) homeostasis was assessed using thapsigargin, electron microscopy and intracellular calcium measurements. RESULTS PAX4 overexpression blunted EAD, whereas the diabetes-linked mutant variant PAX4R129W did not convey protection. PAX4-expressing islets exhibited reduced insulitis and decreased beta cell apoptosis, correlating with diminished DNA damage and increased islet cell proliferation. Microarray profiling revealed that PAX4 but not PAX4R129W targeted expression of genes implicated in cell cycle and ER homeostasis. Consistent with the latter, islets overexpressing PAX4 were protected against thapsigargin-mediated ER-stress-related apoptosis. Luminal swelling associated with ER stress induced by thapsigargin was rescued in PAX4-overexpressing beta cells, correlating with preserved cytosolic calcium oscillations in response to glucose. In contrast, RNA interference mediated repression of PAX4-sensitised MIN6 cells to thapsigargin cell death. CONCLUSIONS/INTERPRETATION The coordinated regulation of distinct cellular pathways particularly related to ER homeostasis by PAX4 not achieved by the mutant variant PAX4R129W alleviates beta cell degeneration and protects against diabetes mellitus. The raw data for the RNA microarray described herein are accessible in the Gene Expression Omnibus database under accession number GSE62846.
Collapse
Affiliation(s)
- José Manuel Mellado-Gil
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Carmen María Jiménez-Moreno
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Alejandro Martin-Montalvo
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Ana Isabel Alvarez-Mercado
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Esther Fuente-Martin
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Nadia Cobo-Vuilleumier
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Petra Isabel Lorenzo
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Eva Bru-Tari
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
- Instituto de Bioingeniería, Universidad Miguel Hernandez, Elche, Spain
| | - Irene de Gracia Herrera-Gómez
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Livia López-Noriega
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain
| | - Javier Pérez-Florido
- Medical Genome Project, Genomics & Bioinformatics Platform of Andalusia, Seville, Spain
| | - Javier Santoyo-López
- Medical Genome Project, Genomics & Bioinformatics Platform of Andalusia, Seville, Spain
- Edinburgh Genomics, University of Edinburgh, Edinburgh, UK
| | - Andreas Spyrantis
- Department of Internal Medicine, Ulm University Medical Centre, Ulm, Germany
| | - Paolo Meda
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Bernhard O Boehm
- Department of Internal Medicine, Ulm University Medical Centre, Ulm, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Republic of Singapore
- Imperial College, London, UK
| | - Ivan Quesada
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Spain
- Instituto de Bioingeniería, Universidad Miguel Hernandez, Elche, Spain
| | - Benoit R Gauthier
- Pancreatic Islet Development and Regeneration Unit, Department of Stem Cells, Andalusian Center for Molecular Biology and Regenerative Medicine (CABIMER), Avda Américo Vespucio, Parque Científico y Tecnológico Cartuja 93, 41092, Seville, Spain.
| |
Collapse
|
29
|
Hauser LJ, Ir D, Kingdom TT, Robertson CE, Frank DN, Ramakrishnan VR. Evaluation of bacterial transmission to the paranasal sinuses through sinus irrigation. Int Forum Allergy Rhinol 2016; 6:800-6. [PMID: 26990369 DOI: 10.1002/alr.21755] [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] [Received: 03/27/2015] [Revised: 01/19/2016] [Accepted: 01/22/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND Saline nasal irrigation is effective in the treatment of sinonasal disorders, including chronic rhinosinusitis (CRS). Despite bacterial contamination in rinse bottles and reports of infections from contaminated irrigation water, tap water is still used by ∼50% of irrigation users, raising a potential public health concern. This study aimed to determine whether bacteria from the water supply used in sinus irrigations colonizes the paranasal sinuses. METHODS Samples were taken from the: (1) water used for irrigation, (2) faucet or container the water originated from, (3) rinse bottle, and (4) postoperative ethmoid cavity from 13 subjects with CRS. Microbiota were characterized using quantitative polymerase chain reaction (qPCR) and 16S ribosomal RNA (rRNA) gene sequencing. The Morisita-Horn beta-diversity index (M-H) was used to assess similarity in microbiota between samples, and genomic analysis was performed to assess clonality of cultured bacteria. RESULTS Of 13 subjects, 6 used distilled water, 6 used tap water, and 1 used well water in this institutional review board (IRB)-approved observational study. Well-water had markedly more bacteria than tap or distilled water. There was a trend toward tap having more bacteria than distilled water. The sinus samples were notably dissimilar to the bottle, faucet, and irrigant (M-H 0.15, 0.09, and 0.18, respectively). There was no difference in postoperative microbiotas between distilled and tap water users. CONCLUSION The current study suggests that irrigation plays little role in establishing the sinus microbiome. Although rinsing with tap water may never be formally recommended, these data are useful to counsel patients who prefer to do so in non-endemic areas if the municipal water supply is appropriately treated.
Collapse
Affiliation(s)
- Leah J Hauser
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado, Aurora, CO
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado, Aurora, CO
| | - Todd T Kingdom
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado, Aurora, CO
| | - Charles E Robertson
- The Microbiome Research Consortium, University of Colorado, Aurora, CO.,Department of Biostatistics and Informatics, University of Colorado, Aurora, CO.,Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Aurora, CO
| | - Daniel N Frank
- Division of Infectious Diseases, University of Colorado, Aurora, CO.,The Microbiome Research Consortium, University of Colorado, Aurora, CO
| | - Vijay R Ramakrishnan
- Department of Otolaryngology-Head and Neck Surgery, University of Colorado, Aurora, CO
| |
Collapse
|
30
|
Bendtsen KM, Fisker L, Hansen AK, Hansen CHF, Nielsen DS. The influence of the young microbiome on inflammatory diseases--Lessons from animal studies. ACTA ACUST UNITED AC 2015; 105:278-95. [PMID: 26663871 DOI: 10.1002/bdrc.21116] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Chronic inflammatory diseases are on the rise in the Westernized world. This rise has been correlated to a range of environmental factors, such as birth mode, rural versus urban living conditions, and use of antibiotics. Such environmental factors also influence early life gut microbiota (GM) colonization and maturation--and there is growing evidence that the negative effects of these factors on human health are mediated via GM alterations. Colonization of the gut initiates priming of the immune system from birth, driving tolerance towards non-harmful microorganisms and dietary antigens and proper reactions towards invading pathogens. This early colonization is crucial for the establishment of a healthy GM, and throughout life the balanced interaction of GM and immune system is a key element in maintaining health. An immune system out of balance increases the risk for later life inflammatory diseases. Animal models are indispensable in the studies of GM influence on disease mechanisms and progression, and focus points include studies of GM modification during pregnancy and perinatal life. Here, we present an overview of animal studies which have contributed to our understanding of GM functions in early life and how alterations affect risk and expression of certain inflammatory diseases with juvenile onset, including interventions, such as birth mode, antibiotics, and probiotics.
Collapse
Affiliation(s)
- Katja M Bendtsen
- Department of Veterinary Disease Biology, Faculty of Health, University of Copenhagen, Denmark
| | - Line Fisker
- Department of Veterinary Disease Biology, Faculty of Health, University of Copenhagen, Denmark
| | - Axel K Hansen
- Department of Veterinary Disease Biology, Faculty of Health, University of Copenhagen, Denmark
| | - Camilla H F Hansen
- Department of Veterinary Disease Biology, Faculty of Health, University of Copenhagen, Denmark
| | - Dennis S Nielsen
- Department of Food Science, Food Microbiology, Faculty of Science, University of Copenhagen, Denmark
| |
Collapse
|
31
|
Abstract
The innate immune system includes several classes of pattern recognition receptors (PRRs), including membrane-bound Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). These receptors detect pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) in the extracellular and intracellular space. Intracellular NLRs constitute inflammasomes, which activate and release caspase-1, IL-1β, and IL-18 thereby initiating an inflammatory response. Systemic and local low-grade inflammation and release of proinflammatory cytokines are implicated in the development and progression of diabetes mellitus and diabetic nephropathy. TLR2, TLR4, and the NLRP3 inflammasome can induce the production of various proinflammatory cytokines and are critically involved in inflammatory responses in pancreatic islets, and in adipose, liver and kidney tissues. This Review describes how innate immune system-driven inflammatory processes can lead to apoptosis, tissue fibrosis, and organ dysfunction resulting in insulin resistance, impaired insulin secretion, and renal failure. We propose that careful targeting of TLR2, TLR4, and NLRP3 signalling pathways could be beneficial for the treatment of diabetes mellitus and diabetic nephropathy.
Collapse
|
32
|
Alkanani AK, Hara N, Gottlieb PA, Ir D, Robertson CE, Wagner BD, Frank DN, Zipris D. Alterations in Intestinal Microbiota Correlate With Susceptibility to Type 1 Diabetes. Diabetes 2015; 64:3510-20. [PMID: 26068542 PMCID: PMC4587635 DOI: 10.2337/db14-1847] [Citation(s) in RCA: 209] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 05/29/2015] [Indexed: 12/26/2022]
Abstract
We tested the hypothesis that alterations in the intestinal microbiota are linked with the progression of type 1 diabetes (T1D). Herein, we present results from a study performed in subjects with islet autoimmunity living in the U.S. High-throughput sequencing of bacterial 16S rRNA genes and adjustment for sex, age, autoantibody presence, and HLA indicated that the gut microbiomes of seropositive subjects differed from those of autoantibody-free first-degree relatives (FDRs) in the abundance of four taxa. Furthermore, subjects with autoantibodies, seronegative FDRs, and new-onset patients had different levels of the Firmicutes genera Lactobacillus and Staphylococcus compared with healthy control subjects with no family history of autoimmunity. Further analysis revealed trends toward increased and reduced abundances of the Bacteroidetes genera Bacteroides and Prevotella, respectively, in seropositive subjects with multiple versus one autoantibody. Canonical discriminant analysis suggested that the gut microbiomes of autoantibody-positive individuals and seronegative FDRs clustered together but separate from those of new-onset patients and unrelated healthy control subjects. Finally, no differences in biodiversity were evident in seropositive versus seronegative FDRs. These observations suggest that altered intestinal microbiota may be associated with disease susceptibility.
Collapse
Affiliation(s)
- Aimon K Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| | - Diana Ir
- Division of Infectious Diseases, School of Medicine, University of Colorado Denver, Aurora, CO
| | - Charles E Robertson
- Division of Infectious Diseases, School of Medicine, University of Colorado Denver, Aurora, CO University of Colorado Denver Microbiome Research Consortium, School of Medicine, University of Colorado Denver, Aurora, CO
| | - Brandie D Wagner
- University of Colorado Denver Microbiome Research Consortium, School of Medicine, University of Colorado Denver, Aurora, CO Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO
| | - Daniel N Frank
- Division of Infectious Diseases, School of Medicine, University of Colorado Denver, Aurora, CO University of Colorado Denver Microbiome Research Consortium, School of Medicine, University of Colorado Denver, Aurora, CO
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO
| |
Collapse
|
33
|
Pearson JA, Wong FS, Wen L. The importance of the Non Obese Diabetic (NOD) mouse model in autoimmune diabetes. J Autoimmun 2015; 66:76-88. [PMID: 26403950 DOI: 10.1016/j.jaut.2015.08.019] [Citation(s) in RCA: 197] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 08/26/2015] [Indexed: 02/06/2023]
Abstract
Type 1 Diabetes (T1D) is an autoimmune disease characterized by the pancreatic infiltration of immune cells resulting in T cell-mediated destruction of the insulin-producing beta cells. The successes of the Non-Obese Diabetic (NOD) mouse model have come in multiple forms including identifying key genetic and environmental risk factors e.g. Idd loci and effects of microorganisms including the gut microbiota, respectively, and how they may contribute to disease susceptibility and pathogenesis. Furthermore, the NOD model also provides insights into the roles of the innate immune cells as well as the B cells in contributing to the T cell-mediated disease. Unlike many autoimmune disease models, the NOD mouse develops spontaneous disease and has many similarities to human T1D. Through exploiting these similarities many targets have been identified for immune-intervention strategies. Although many of these immunotherapies did not have a significant impact on human T1D, they have been shown to be effective in the NOD mouse in early stage disease, which is not equivalent to trials in newly-diagnosed patients with diabetes. However, the continued development of humanized NOD mice would enable further clinical developments, bringing T1D research to a new translational level. Therefore, it is the aim of this review to discuss the importance of the NOD model in identifying the roles of the innate immune system and the interaction with the gut microbiota in modifying diabetes susceptibility. In addition, the role of the B cells will also be discussed with new insights gained through B cell depletion experiments and the impact on translational developments. Finally, this review will also discuss the future of the NOD mouse and the development of humanized NOD mice, providing novel insights into human T1D.
Collapse
Affiliation(s)
- James A Pearson
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA
| | - F Susan Wong
- Diabetes Research Group, Institute of Molecular & Experimental Medicine, School of Medicine, Cardiff University, Wales, UK
| | - Li Wen
- Section of Endocrinology, School of Medicine, Yale University, New Haven, CT, USA.
| |
Collapse
|
34
|
Bessesen MT, Kotter CV, Wagner BD, Adams JC, Kingery S, Benoit JB, Robertson CE, Janoff EN, Frank DN. MRSA colonization and the nasal microbiome in adults at high risk of colonization and infection. J Infect 2015; 71:649-57. [PMID: 26335708 DOI: 10.1016/j.jinf.2015.08.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Revised: 08/11/2015] [Accepted: 08/24/2015] [Indexed: 12/15/2022]
Abstract
OBJECTIVE The objective of this study was to define the nasal microbiome of hospital inpatients who are persistently colonized with methicillin-resistant Staphylococcus aureus (MRSA) compared with matched, non-colonized controls. METHODS Twenty-six persistently MRSA-colonized subjects and 26 matched non-colonized controls were selected from the screening records of the infection control program at the Department of the Veteran Affairs Eastern Colorado Health Care System (VA-ECHCS). The nasal microbiotas were analyzed with PCR amplification and sequencing of the 16S ribosomal RNA (rRNA) gene. Comparison of all variables across the groups was performed using stratified logistic regression to account for the one-to-one matching. Canonical discriminant analysis was performed to assess differences in bacterial community across the two groups. Competing organisms were cocultured with MRSA in vitro. RESULTS There was a negative association between MRSA colonization and colonization with Streptococcus spp. At the species level, multivariate analysis demonstrated a statistically significant negative association between colonization with Streptococcus mitis or Lactobacillus gasseri and MRSA. Coculture experiments revealed in vitro competition between S. mitis and all of the 22 MRSA strains isolated from subjects. Competition was blocked by addition of catalase to the media. Persistently colonized subjects had lesser microbial diversity than the non-colonized controls. CONCLUSION In a high-risk inpatient setting, bacterial competition in the nasal niche protects some patients from MRSA colonization.
Collapse
Affiliation(s)
- Mary T Bessesen
- Division of Adult Infectious Diseases, University of Colorado Denver, Aurora, CO, USA; Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA.
| | - Cassandra Vogel Kotter
- Division of Adult Infectious Diseases, University of Colorado Denver, Aurora, CO, USA; Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA
| | - Brandie D Wagner
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, USA; Microbiome Research Consortium (MiRC), University of Colorado Denver, Aurora, CO, USA
| | - Jill C Adams
- Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA
| | - Shannon Kingery
- Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA
| | - Jeanne B Benoit
- Division of Adult Infectious Diseases, University of Colorado Denver, Aurora, CO, USA; Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA; Microbiome Research Consortium (MiRC), University of Colorado Denver, Aurora, CO, USA
| | - Charles E Robertson
- Division of Adult Infectious Diseases, University of Colorado Denver, Aurora, CO, USA; Microbiome Research Consortium (MiRC), University of Colorado Denver, Aurora, CO, USA
| | - Edward N Janoff
- Division of Adult Infectious Diseases, University of Colorado Denver, Aurora, CO, USA; Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA; Microbiome Research Consortium (MiRC), University of Colorado Denver, Aurora, CO, USA; Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, CO, USA
| | - Daniel N Frank
- Division of Adult Infectious Diseases, University of Colorado Denver, Aurora, CO, USA; Department of Veterans Affairs Eastern Colorado Healthcare System, Denver, CO, USA; Microbiome Research Consortium (MiRC), University of Colorado Denver, Aurora, CO, USA; Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
35
|
Fichna M, Żurawek M, Fichna P, Januszkiewicz-Lewandowska D, Ruchała M, Nowak J. Polymorphisms of the Toll-Like Receptor-3 Gene in Autoimmune Adrenal Failure and Type 1 Diabetes in Polish Patients. Arch Immunol Ther Exp (Warsz) 2015; 64:83-7. [PMID: 26318769 PMCID: PMC4713709 DOI: 10.1007/s00005-015-0360-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Accepted: 06/08/2015] [Indexed: 12/16/2022]
Abstract
Infectious agents are plausible environmental triggers for autoimmunity in genetically susceptible individuals. Polymorphic variants of genes implicated in innate immunity may affect immune responses and hence promote auto-aggressive reactions. Genes such as Toll-like receptor-3 (TLR3), which participate in recognizing conserved foreign molecules and mounting the first line of defence against viral infections, are promising functional candidates in autoimmune conditions. We investigated the association of the TLR3 variants, rs13126816 and rs3775291, with the autoimmune endocrine disorders, Addison's disease (AD) and type 1 diabetes (T1D) in the Polish population. The study comprised 168 AD patients, 524 individuals with T1D and 592 healthy controls. Genotyping was performed by real-time PCR. Distribution of the TLR3 genotypes and alleles did not reveal significant differences between patients and controls (p > 0.05). No effect on age at disease onset was found in affected cohorts. This analysis does not support an association between TLR3 variants and the risk for autoimmune destruction of the adrenal cortex and beta cells. However, innate immunity merits further studies in autoimmune endocrine conditions.
Collapse
Affiliation(s)
- Marta Fichna
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland. .,Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland. .,Department of Clinical Immunology, Poznan University of Medical Sciences, Poznan, Poland.
| | - Magdalena Żurawek
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| | - Piotr Fichna
- Department of Paediatric Diabetes and Obesity, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Marek Ruchała
- Department of Endocrinology, Metabolism and Internal Medicine, Poznan University of Medical Sciences, Poznan, Poland
| | - Jerzy Nowak
- Institute of Human Genetics, Polish Academy of Sciences, Strzeszynska 32, 60-479, Poznan, Poland
| |
Collapse
|
36
|
Frank DN, Bales ES, Monks J, Jackman MJ, MacLean PS, Ir D, Robertson CE, Orlicky DJ, McManaman JL. Perilipin-2 Modulates Lipid Absorption and Microbiome Responses in the Mouse Intestine. PLoS One 2015; 10:e0131944. [PMID: 26147095 PMCID: PMC4493139 DOI: 10.1371/journal.pone.0131944] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Accepted: 06/08/2015] [Indexed: 02/06/2023] Open
Abstract
Obesity and its co-morbidities, such as fatty liver disease, are increasingly prevalent worldwide health problems. Intestinal microorganisms have emerged as critical factors linking diet to host physiology and metabolic function, particularly in the context of lipid homeostasis. We previously demonstrated that deletion of the cytoplasmic lipid drop (CLD) protein Perilipin-2 (Plin2) in mice largely abrogates long-term deleterious effects of a high fat (HF) diet. Here we test the hypotheses that Plin2 function impacts the earliest steps of HF diet-mediated pathogenesis as well as the dynamics of diet-associated changes in gut microbiome diversity and function. WT and perilipin-2 null mice raised on a standard chow diet were randomized to either low fat (LF) or HF diets. After four days, animals were assessed for changes in physiological (body weight, energy balance, and fecal triglyceride levels), histochemical (enterocyte CLD content), and fecal microbiome parameters. Plin2-null mice had significantly lower respiratory exchange ratios, diminished frequencies of enterocyte CLDs, and increased fecal triglyceride levels compared with WT mice. Microbiome analyses, employing both 16S rRNA profiling and metagenomic deep sequencing, indicated that dietary fat content and Plin2 genotype were significantly and independently associated with gut microbiome composition, diversity, and functional differences. These data demonstrate that Plin2 modulates rapid effects of diet on fecal lipid levels, enterocyte CLD contents, and fuel utilization properties of mice that correlate with structural and functional differences in their gut microbial communities. Collectively, the data provide evidence of Plin2 regulated intestinal lipid uptake, which contributes to rapid changes in the gut microbial communities implicated in diet-induced obesity.
Collapse
Affiliation(s)
- Daniel N. Frank
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Microbiome Research Consortium, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Elise S. Bales
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Jenifer Monks
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Matthew J. Jackman
- Division of Endocrinology and Metabolism, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Paul S. MacLean
- Division of Endocrinology and Metabolism, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Diana Ir
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Division of Infectious Disease, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- Microbiome Research Consortium, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - David J. Orlicky
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - James L. McManaman
- Division of Basic Reproductive Sciences, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- The Center for Human Nutrition, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| |
Collapse
|
37
|
Tai N, Wong FS, Wen L. The role of gut microbiota in the development of type 1, type 2 diabetes mellitus and obesity. Rev Endocr Metab Disord 2015; 16:55-65. [PMID: 25619480 PMCID: PMC4348024 DOI: 10.1007/s11154-015-9309-0] [Citation(s) in RCA: 186] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Diabetes is a group of metabolic disorders characterized by persistent hyperglycemia and has become a major public health concern. Autoimmune type 1 diabetes (T1D) and insulin resistant type 2 diabetes (T2D) are the two main types. A combination of genetic and environmental factors contributes to the development of these diseases. Gut microbiota have emerged recently as an essential player in the development of T1D, T2D and obesity. Altered gut microbiota have been strongly linked to disease in both rodent models and humans. Both classic 16S rRNA sequencing and shot-gun metagenomic pyrosequencing analysis have been successfully applied to explore the gut microbiota composition and functionality. This review focuses on the association between gut microbiota and diabetes and discusses the potential mechanisms by which gut microbiota regulate disease development in T1D, T2D and obesity.
Collapse
Affiliation(s)
- Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, Yale School of Medicine, New Haven, USA
| | | | | |
Collapse
|
38
|
Flass T, Tong S, Frank DN, Wagner BD, Robertson CE, Kotter CV, Sokol RJ, Zemanick E, Accurso F, Hoffenberg EJ, Narkewicz MR. Intestinal lesions are associated with altered intestinal microbiome and are more frequent in children and young adults with cystic fibrosis and cirrhosis. PLoS One 2015; 10:e0116967. [PMID: 25658710 PMCID: PMC4319904 DOI: 10.1371/journal.pone.0116967] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 12/17/2014] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIMS Cirrhosis (CIR) occurs in 5-7% of cystic fibrosis (CF) patients. We hypothesized that alterations in intestinal function in CF contribute to the development of CIR. AIMS Determine the frequency of macroscopic intestinal lesions, intestinal inflammation, intestinal permeability and characterize fecal microbiome in CF CIR subjects and CF subjects with no liver disease (CFnoLIV). METHODS 11 subjects with CFCIR (6 M, 12.8 yrs ± 3.8) and 19 matched with CFnoLIV (10 M, 12.6 yrs ± 3.4) underwent small bowel capsule endoscopy, intestinal permeability testing by urinary lactulose: mannitol excretion ratio, fecal calprotectin determination and fecal microbiome characterization. RESULTS CFCIR and CFnoLIV did not differ in key demographics or CF complications. CFCIR had higher GGT (59±51 U/L vs 17±4 p = 0.02) and lower platelet count (187±126 vs 283±60 p = 0.04) and weight (-0.86 ± 1.0 vs 0.30 ± 0.9 p = 0.002) z scores. CFCIR had more severe intestinal mucosal lesions on capsule endoscopy (score ≥4, 4/11 vs 0/19 p = 0.01). Fecal calprotectin was similar between CFCIR and CFnoLIV (166 μg/g ±175 vs 136 ± 193 p = 0.58, nl <120). Lactulose:mannitol ratio was elevated in 27/28 subjects and was slightly lower in CFCIR vs CFnoLIV (0.08±0.02 vs 0.11±0.05, p = 0.04, nl ≤0.03). Small bowel transit time was longer in CFCIR vs CFnoLIV (195±42 min vs 167±68 p<0.001, nl 274 ± 41). Bacteroides were decreased in relative abundance in CFCIR and were associated with lower capsule endoscopy score whereas Clostridium were more abundant in CFCIR and associated with higher capsule endoscopy score. CONCLUSIONS CFCIR is associated with increased intestinal mucosal lesions, slower small bowel transit time and alterations in fecal microbiome. Abnormal intestinal permeability and elevated fecal calprotectin are common in all CF subjects. Disturbances in intestinal function in CF combined with changes in the microbiome may contribute to the development of hepatic fibrosis and intestinal lesions.
Collapse
Affiliation(s)
- Thomas Flass
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Suhong Tong
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital, Aurora, Colorado, United States of America
| | - Daniel N. Frank
- Department of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Brandie D. Wagner
- Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital, Aurora, Colorado, United States of America
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado Denver, Aurora, CO, United States of America
| | - Charles E. Robertson
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado Boulder, Boulder, Colorado, United States of America
| | - Cassandra Vogel Kotter
- Department of Infectious Diseases, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Ronald J. Sokol
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
- Colorado Clinical and Translational Sciences Institute, University of Colorado School of Medicine, Aurora, CO, United States of America
| | - Edith Zemanick
- Section of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Frank Accurso
- Section of Pediatric Pulmonary Medicine, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Edward J. Hoffenberg
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| | - Michael R. Narkewicz
- Section of Pediatric Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, University of Colorado School of Medicine and Children’s Hospital Colorado, Aurora, CO, United States of America
| |
Collapse
|
39
|
Biedermann L, Rogler G. The intestinal microbiota: its role in health and disease. Eur J Pediatr 2015; 174:151-67. [PMID: 25563215 DOI: 10.1007/s00431-014-2476-2] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/08/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022]
Abstract
UNLABELLED The intestinal microbiota (previously referred to as "intestinal flora") has entered the focus of research interest not only in microbiology but also in medicine. Huge progress has been made with respect to the analysis of composition and functions of the human microbiota. An "imbalance" of the microbiota, frequently also called a "dysbiosis," has been associated with different diseases in recent years. Crohn's disease and ulcerative colitis as two major forms of inflammatory bowel disease, irritable bowel syndrome (IBS) and some infectious intestinal diseases such as Clostridium difficile colitis feature a dysbiosis of the intestinal flora. Whereas this is somehow expected or less surprising, an imbalance of the microbiota or an enrichment of specific bacterial strains in the flora has been associated with an increasing number of other diseases such as diabetes, metabolic syndrome, non-alcoholic fatty liver disease or steatohepatitis and even psychiatric disorders such as depression or multiple sclerosis. It is important to understand the different aspects of potential contributions of the microbiota to pathophysiology of the mentioned diseases. CONCLUSION With the present manuscript, we aim to summarize the current knowledge and provide an overview of the different concepts on how bacteria contribute to health and disease in animal models and-more importantly-humans. In addition, it has to be borne in mind that we are only at the very beginning to understand the complex mechanisms of host-microbial interactions.
Collapse
Affiliation(s)
- Luc Biedermann
- Division of Gastroenterology and Hepatology, University Hospital Zürich, Rämistrasse 100, 8091, Zürich, Switzerland,
| | | |
Collapse
|
40
|
Abstract
The microbiota of the human metaorganism is not a mere bystander. These microbes have coevolved with us and are pivotal to normal development and homoeostasis. Dysbiosis of the GI microbiota is associated with many disease susceptibilities, including obesity, malignancy, liver disease and GI pathology such as IBD. It is clear that there is direct and indirect crosstalk between this microbial community and host immune response. However, the precise mechanism of this microbial influence in disease pathogenesis remains elusive and is now a major research focus. There is emerging literature on the role of the microbiota in the pathogenesis of autoimmune disease, with clear and increasing evidence that changes in the microbiota are associated with some of these diseases. Examples include type 1 diabetes, coeliac disease and rheumatoid arthritis, and these contribute significantly to global morbidity and mortality. Understanding the role of the microbiota in autoimmune diseases may offer novel insight into factors that initiate and drive disease progression, stratify patient risk for complications and ultimately deliver new therapeutic strategies. This review summarises the current status on the role of the microbiota in autoimmune diseases.
Collapse
Affiliation(s)
- Mairi H McLean
- Laboratory of Molecular Immunoregulation, Cancer & Inflammation Program, National Cancer Institute, Frederick, Maryland, USA
| | - Dario Dieguez
- Society for Women’s Health Research, Scientific Affairs, Washington, DC, USA
| | - Lindsey M Miller
- Society for Women’s Health Research, Scientific Affairs, Washington, DC, USA
| | - Howard A Young
- Laboratory of Molecular Immunoregulation, Cancer & Inflammation Program, National Cancer Institute, Frederick, Maryland, USA
| |
Collapse
|
41
|
Rosenbaum JT, Lin P, Asquith M, Costello ME, Kenna TJ, Brown MA. Does the microbiome play a causal role in spondyloarthritis? Clin Rheumatol 2014; 33:763-7. [PMID: 24810703 DOI: 10.1007/s10067-014-2664-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 03/24/2014] [Indexed: 01/20/2023]
Abstract
The purpose of this study is to review the potential causal role of the microbiome in the pathogenesis of spondyloarthritis. The method used for the study is literature review. The microbiome plays a major role in educating the immune response. The microbiome is strongly implicated in inflammatory bowel disease which has clinical and genetic overlap with spondyloarthritis. The microbiome also plays a causal role in bowel and joint disease in HLA B27/human beta 2 microglobulin transgenic rats. The mechanism(s) by which HLA B27 could influence the microbiome is unknown but theories include an immune response gene selectivity, an effect on dendritic cell function, or a mucosal immunodeficiency. Bacteria are strongly implicated in the pathogenesis of spondyloarthritis. Studies to understand how HLA B27 affects bacterial ecosystems should be encouraged.
Collapse
Affiliation(s)
- James T Rosenbaum
- Division of Arthritis and Rheumatic Diseases and Department of Ophthalmology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd. L467Ad, Portland, OR, 97239, USA,
| | | | | | | | | | | |
Collapse
|
42
|
Nielsen DS, Krych Ł, Buschard K, Hansen CHF, Hansen AK. Beyond genetics. Influence of dietary factors and gut microbiota on type 1 diabetes. FEBS Lett 2014; 588:4234-43. [PMID: 24746688 DOI: 10.1016/j.febslet.2014.04.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Revised: 04/04/2014] [Accepted: 04/07/2014] [Indexed: 12/31/2022]
Abstract
Type 1 diabetes (T1D) is an autoimmune disease ultimately leading to destruction of insulin secreting β-cells in the pancreas. Genetic susceptibility plays an important role in T1D etiology, but even mono-zygotic twins only have a concordance rate of around 50%, underlining that other factors than purely genetic are involved in disease development. Here we review the influence of dietary and environmental factors on T1D development in humans as well as animal models. Even though data are still inconclusive, there are strong indications that gut microbiota dysbiosis plays an important role in T1D development and evidence from animal models suggests that gut microbiota manipulation might prove valuable in future prevention of T1D in genetically susceptible individuals.
Collapse
Affiliation(s)
- Dennis S Nielsen
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg C, Denmark.
| | - Łukasz Krych
- Department of Food Science, Faculty of Science, University of Copenhagen, 1958 Frederiksberg C, Denmark
| | | | - Camilla H F Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1871 Frederiksberg C, Denmark
| | - Axel K Hansen
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen, 1871 Frederiksberg C, Denmark
| |
Collapse
|