1
|
Sun F, Yang CL, Wang FX, Rong SJ, Luo JH, Lu WY, Yue TT, Wang CY, Liu SW. Pancreatic draining lymph nodes (PLNs) serve as a pathogenic hub contributing to the development of type 1 diabetes. Cell Biosci 2023; 13:156. [PMID: 37641145 PMCID: PMC10464122 DOI: 10.1186/s13578-023-01110-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 08/21/2023] [Indexed: 08/31/2023] Open
Abstract
Type 1 diabetes (T1D) is a chronic, progressive autoinflammatory disorder resulting from the breakdown of self-tolerance and unrestrained β cell-reactive immune response. Activation of immune cells is initiated in islet and amplified in lymphoid tissues, especially those pancreatic draining lymph nodes (PLNs). The knowledge of PLNs as the hub of aberrant immune response is continuously being replenished and renewed. Here we provide a PLN-centered view of T1D pathogenesis and emphasize that PLNs integrate signal inputs from the pancreas, gut, viral infection or peripheral circulation, undergo immune remodeling within the local microenvironment and export effector cell components into pancreas to affect T1D progression. In accordance, we suggest that T1D intervention can be implemented by three major ways: cutting off the signal inputs into PLNs (reduce inflammatory β cell damage, enhance gut integrity and control pathogenic viral infections), modulating the immune activation status of PLNs and blocking the outputs of PLNs towards pancreatic islets. Given the dynamic and complex nature of T1D etiology, the corresponding intervention strategy is thus required to be comprehensive to ensure optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Fei Sun
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chun-Liang Yang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fa-Xi Wang
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan-Jie Rong
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Hui Luo
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wan-Ying Lu
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tian-Tian Yue
- Devision of Nutrition, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cong-Yi Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China.
- NHC Key Laboratory of Respiratory Diseases, Department of Respiratory and Critical Care Medicine, The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shi-Wei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China.
| |
Collapse
|
2
|
Wang K, Li L, Jin J, An Y, Wang Z, Zhou S, Zhang J, Abuduaini B, Cheng C, Li N. Fatty acid synthase (Fasn) inhibits the expression levels of immune response genes via alteration of alternative splicing in islet cells. J Diabetes Complications 2022; 36:108159. [PMID: 35210136 DOI: 10.1016/j.jdiacomp.2022.108159] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Revised: 12/17/2021] [Accepted: 02/11/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND Increasing evidence has shown that fatty acid synthase (Fasn) is associated with diabetes mellitus (DM) and insulin resistance, however, it remains unclear how Fasn upregulation leads to dysregulation of energy homeostasis in islet cells. Consequently, uncovering the function of Fasn in islet cells. Consequently, uncovering the function of FASN in islet cells is immensely important for finding a treatment target. AIM In this study, we elucidated the biological function of Fasn on the target genes in a rat insulinoma INS-1 cell line. METHODS We created a Fasn overexpressing rat insulinoma cell line (Fasn-OE), and performed bulk RNA-sequencing (RNA-seq) experiments on Fasn-OE and INS-1 (control) cells. We first identified differentially expressed genes (DEGs) using Bioconductor package edgeR, and then discovered enriched gene ontology terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways using the KEGG Orthology Based Annotation System (KOBAS) 2.0 web server. Furthermore, we identified alternative splicing events (ASEs) and regulated alternative splicing events (RASEs) by applying the ABLas pipeline. The reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used for validation of selected differentially expressed genes (DEGs) and Fasn-regulated alternative splicing genes (RASGs). RESULTS In this study we found that Fasn overexpression led to significant changes of gene expression profiles, including downregulations of mRNA levels of immune related genes, including Bst2, Ddit3, Isg15, Mx2, Oas1a, Oasl, and RT1-S3 in INS-1 cell line. Furthermore, Fasn positively regulated the expression of transcription factors such as Fat1 and Ncl diabetes-related genes. Importantly, Fasn overexpression to result in alternative splicing events including in a metabolism-associated ATP binding protein mRNA Abcc5. In Gene Ontology analysis, the downregulated genes in Fasn-OE cells were mainly enriched in inflammatory response and innate immune response. In Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, the downregulated genes were mainly enriched in TNF signaling pathway and cytokine-mediated signaling pathways. CONCLUSIONS Our findings showed that upregulation of Fasn may play a critical role in islet cell immunmetabolism via modifications of immune/inflammatory related genes on transcription and alternative splicing level, which provide novel insights into characterizing the function of Fasn in islet cell immunity and for the development of chemo/immune therapies.
Collapse
Affiliation(s)
- Kunling Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, PR China
| | - Lin Li
- Department of Molecular Biology, Xinjiang Medical University, Urumqi, Xinjiang 830054, PR China
| | - Jing Jin
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, PR China
| | - Yanli An
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, PR China
| | - Zhongjuan Wang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, PR China
| | - Shiying Zhou
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830054, PR China
| | - Jiyuan Zhang
- The First Clinical Institute of Xinjiang Medical University
| | - Buzukela Abuduaini
- Department of Intensive Care Unit, The First Affiliated Hospital of Xinjiang Medical University.
| | - Chao Cheng
- ABLife BioBigData Institute, Wuhan, Hubei, 430075, China
| | - Ning Li
- ABLife BioBigData Institute, Wuhan, Hubei, 430075, China
| |
Collapse
|
3
|
Needell JC, Brown MN, Zipris D. Involvement of adipose tissue inflammation and dysfunction in virus-induced type 1 diabetes. J Endocrinol 2018; 238:61-75. [PMID: 29743341 DOI: 10.1530/joe-18-0131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/08/2018] [Indexed: 12/16/2022]
Abstract
The etiopathogenesis of type 1 diabetes (T1D) remains poorly understood. We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to better understand the role of the innate immune system in the mechanism of virus-induced disease. We observed that infection with KRV results in cell influx into visceral adipose tissue soon following infection prior to insulitis and hyperglycemia. In sharp contrast, subcutaneous adipose tissue is free of cellular infiltration, whereas β cell inflammation and diabetes are observed beginning on day 14 post infection. Immunofluorescence studies further demonstrate that KRV triggers CD68+ macrophage recruitment and the expression of KRV transcripts and proinflammatory cytokines and chemokines in visceral adipose tissue. Adipocytes from naive rats cultured in the presence of KRV express virus transcripts and upregulate cytokine and chemokine gene expression. KRV induces apoptosis in visceral adipose tissue in vivo, which is reflected by positive TUNEL staining and the expression of cleaved caspase-3. Moreover, KRV leads to an oxidative stress response and downregulates the expression of adipokines and genes associated with mediating insulin signaling. Activation of innate immunity with Poly I:C in the absence of KRV leads to CD68+ macrophage recruitment to visceral adipose tissue and a decrease in adipokine expression detected 5 days following Poly (I:C) treatment. Finally, proof-of-principle studies show that brief anti-inflammatory steroid therapy suppresses visceral adipose tissue inflammation and protects from virus-induced disease. Our studies provide evidence raising the hypothesis that visceral adipose tissue inflammation and dysfunction may be involved in early mechanisms triggering β cell autoimmunity.
Collapse
Affiliation(s)
- James C Needell
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Denver, Aurora, Colorado, USA
| | - Madalyn N Brown
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Denver, Aurora, Colorado, USA
| | - Danny Zipris
- Barbara Davis Center for Childhood DiabetesUniversity of Colorado Denver, Aurora, Colorado, USA
| |
Collapse
|
4
|
Arts RJW, Joosten LAB, Netea MG. The Potential Role of Trained Immunity in Autoimmune and Autoinflammatory Disorders. Front Immunol 2018. [PMID: 29515591 PMCID: PMC5826224 DOI: 10.3389/fimmu.2018.00298] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
During induction of trained immunity, monocytes and macrophages undergo a functional and transcriptional reprogramming toward increased activation. Important rewiring of cellular metabolism of the myeloid cells takes place during induction of trained immunity, including a shift toward glycolysis induced through the mTOR pathway, as well as glutaminolysis and cholesterol synthesis. Subsequently, this leads to modulation of the function of epigenetic enzymes, resulting in important changes in chromatin architecture that enables increased gene transcription. However, in addition to the beneficial effects of trained immunity as a host defense mechanism, we hypothesize that trained immunity also plays a deleterious role in the induction and/or maintenance of autoimmune and autoinflammatory diseases if inappropriately activated.
Collapse
Affiliation(s)
- Rob J W Arts
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands
| | - Leo A B Joosten
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Mihai G Netea
- Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, Netherlands.,Department for Genomics and Immunoregulation, Life and Medical Sciences Institute (LIMES), University of Bonn, Bonn, Germany
| |
Collapse
|
5
|
Mohamed WA, Schaalan MF. Antidiabetic efficacy of lactoferrin in type 2 diabetic pediatrics; controlling impact on PPAR-γ, SIRT-1, and TLR4 downstream signaling pathway. Diabetol Metab Syndr 2018; 10:89. [PMID: 30534206 PMCID: PMC6280363 DOI: 10.1186/s13098-018-0390-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 11/27/2018] [Indexed: 12/27/2022] Open
Abstract
The current study aims to investigate the antidiabetic efficacy of camel milk-derived lactoferrin and potential involvement of PPAR-γ and SIRT-1 via TLR-4/NFκB signaling pathway in obese diabetic pediatric population. Sixty young obese patients with type 2 diabetes were selected from the Pediatric Endocrine Metabolic Unit, Cairo University and were randomly divided among two age and sex-matched groups so as to receive either standard therapy without lactoferrin in one arm or to be treated with oral lactoferrin capsules (250 mg/day, p.o) for 3 months in the other arm. Both groups were compared to 50 control healthy volunteers. Measurements of HbA1c, lipid profile, antioxidant capacity (SOD, Nrf2), proinflammatory interleukins; (IL-1β, IL-6, IL-18), Cyclin D-1, lipocalin-2, and PPAR-γ expression levels were done at the beginning and 3 months after daily consumption of lactoferrin. The mechanistic involvement of TLR4-SIRT-1-NFκB signaling cascade was also investigated. The antidiabetic efficacy of lactoferrin was confirmed by significant improvement of the baseline levels of HbA1c, BMI and lipid profile of the obese pediatric cohort, which is evidenced by increased PPAR-γ and SIRT-1 expression. Moreover, the anti-inflammatory effect was evident by the significant decrease in serum levels of IL-1β, IL-6, IL-18, TNF-α, lipocalin 2 in type 2 diabetic post-treatment group, which corresponded by decreased NFκB downstream signaling indicators. The antioxidant efficacy was evident by stimulated SOD levels and NrF2 expression; compared with the pre-treatment group (all at P ≤ 0.001). The consumption of high concentrations of lactoferrin explains its hypoglycemic efficacy and counts for its insulin-sensitizing, anti-inflammatory and immunomodulatory effects via TLR4-NFκB-SIRT-1 signaling cascade. Recommendations on regular intake of lactoferrin could ensure better glycemic control, compared to conventional antidiabetics alone.
Collapse
Affiliation(s)
- Waleed A. Mohamed
- Department of Chemistry, Kasr El Aini Teaching Hospitals, Cairo University, Cairo, Egypt
| | - Mona F. Schaalan
- Department of Clinical Pharmacy and Pharmacy Practice, Translational Research Unit, Faculty of Pharmacy, Misr International University, Cairo, Egypt
| |
Collapse
|
6
|
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
|
7
|
Needell JC, Ir D, Robertson CE, Kroehl ME, Frank DN, Zipris D. Maternal treatment with short-chain fatty acids modulates the intestinal microbiota and immunity and ameliorates type 1 diabetes in the offspring. PLoS One 2017; 12:e0183786. [PMID: 28886045 PMCID: PMC5590848 DOI: 10.1371/journal.pone.0183786] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 08/13/2017] [Indexed: 02/06/2023] Open
Abstract
We recently hypothesized that the intestinal microbiota and the innate immune system play key roles in the mechanism of Kilham Rat Virus-induced type 1 diabetes in the LEW1.WR1 rat. We used this animal model to test the hypothesis that maternal therapy with short-chain fatty acids can modulate the intestinal microbiota and reverse virus-induced proinflammatory responses and type 1 diabetes in rat offspring. We observed that administration of short-chain fatty acids to rat breeders via drinking water prior to pregnancy and further treatment of the offspring with short-chain fatty acids after weaning led to disease amelioration. In contrast, rats that were administered short-chain fatty acids beginning at weaning were not protected from type 1 diabetes. Short-chain fatty acid therapy exerted a profound effect on the intestinal microbiome in the offspring reflected by a reduction and an increase in the abundances of Firmicutes and Bacteroidetes taxa, respectively, on day 5 post-infection, and reversed virus-induced alterations in certain bacterial taxa. Principal component analysis and permutation multivariate analysis of variance tests further revealed that short-chain fatty acids induce a distinct intestinal microbiota compared with uninfected animals or rats that receive the virus only. Short-chain fatty acids downregulated Kilham Rat Virus-induced proinflammatory responses in the intestine. Finally, short-chain fatty acids altered the B and T cell compartments in Peyer’s patches. These data demonstrate that short-chain fatty acids can reshape the intestinal microbiota and prevent virus-induced islet autoimmunity and may therefore represent a useful therapeutic strategy for disease prevention.
Collapse
Affiliation(s)
- James C. Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- University of Colorado Microbiome Research Consortium (MiRC), Aurora, Colorado, United States of America
| | - Miranda E. Kroehl
- Department of Biostatistics and Informatics, Colorado School of Public Health and University of Colorado Denver, Aurora, Colorado, United States of America
| | - Daniel N. Frank
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- University of Colorado Microbiome Research Consortium (MiRC), Aurora, Colorado, United States of America
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
- * E-mail:
| |
Collapse
|
8
|
Needell JC, Dinarello CA, Ir D, Robertson CE, Ryan SM, Kroehl ME, Frank DN, Zipris D. Implication of the intestinal microbiome as a potential surrogate marker of immune responsiveness to experimental therapies in autoimmune diabetes. PLoS One 2017; 12:e0173968. [PMID: 28301545 PMCID: PMC5354421 DOI: 10.1371/journal.pone.0173968] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 03/01/2017] [Indexed: 01/13/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune proinflammatory disease with no effective intervention. A major obstacle in developing new immunotherapies for T1D is the lack of means for monitoring immune responsiveness to experimental therapies. The LEW1.WR1 rat develops autoimmunity following infection with the parvovirus Kilham rat virus (KRV) via mechanisms linked with activation of proinflammatory pathways and alterations in the gut bacterial composition. We used this animal to test the hypothesis that intervention with agents that block innate immunity and diabetes is associated with a shift in the gut microbiota. We observed that infection with KRV results in the induction of proinflammatory gene activation in both the spleen and pancreatic lymph nodes. Furthermore, administering animals the histone deacetylase inhibitor ITF-2357 and IL-1 receptor antagonist (Anakinra) induced differential STAT-1 and the p40 unit of IL-12/IL-23 gene expression. Sequencing of bacterial 16S rRNA genes demonstrated that both ITF-2357 and Anakinra alter microbial diversity. ITF-2357 and Anakinra modulated the abundance of 23 and 8 bacterial taxa in KRV-infected animals, respectively, of which 5 overlapped between the two agents. Lastly, principal component analysis implied that ITF-2357 and Anakinra induce distinct gut microbiomes compared with those from untreated animals or rats provided KRV only. Together, the data suggest that ITF-2357 and Anakinra differentially influence the innate immune system and the intestinal microbiota and highlight the potential use of the gut microbiome as a surrogate means of assessing anti-inflammatory immune effects in type 1 diabetes.
Collapse
Affiliation(s)
- James C. Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| | - Charles A. Dinarello
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Diana Ir
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
| | - Charles E. Robertson
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- University of Colorado Microbiome Research Consortium (MiRC), Aurora, Colorado, United States of America
| | - Sarah M. Ryan
- Department of Biostatistics and Informatics, Colorado School of Public Health and University of Colorado Denver, Aurora, Colorado, United States of America
| | - Miranda E. Kroehl
- Department of Biostatistics and Informatics, Colorado School of Public Health and University of Colorado Denver, Aurora, Colorado, United States of America
| | - Daniel N. Frank
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, Colorado, United States of America
- University of Colorado Microbiome Research Consortium (MiRC), Aurora, Colorado, United States of America
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America
| |
Collapse
|
9
|
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
|
10
|
Schneider DA, von Herrath MG. Potential viral pathogenic mechanism in human type 1 diabetes. Diabetologia 2014; 57:2009-18. [PMID: 25073445 PMCID: PMC4153966 DOI: 10.1007/s00125-014-3340-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 06/09/2014] [Indexed: 12/15/2022]
Abstract
In type 1 diabetes, as a result of as yet unknown triggering events, auto-aggressive CD8(+) T cells, together with a significant number of other inflammatory cells, including CD8(+) T lymphocytes with unknown specificity, infiltrate the pancreas, leading to insulitis and destruction of the insulin-producing beta cells. Type 1 diabetes is a multifactorial disease caused by an interactive combination of genetic and environmental factors. Viruses are major environmental candidates with known potential effects on specific key points in the pathogenesis of type 1 diabetes and recent findings seem to confirm this presumption. However, we still lack well-grounded mechanistic explanations for how exactly viruses may influence type 1 diabetes aetiology. In this review we provide a summary of experimentally defined viral mechanisms potentially involved in the ontology of type 1 diabetes and discuss some novel hypotheses of how viruses may affect the initiation and natural history of the disease.
Collapse
Affiliation(s)
- Darius A. Schneider
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037 USA
- Department of Medicine, UC San Diego, La Jolla, CA USA
| | - Matthias G. von Herrath
- La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037 USA
- Novo Nordisk Type 1 Diabetes Research Center, Seattle, WA 98109 USA
| |
Collapse
|
11
|
Alkanani AK, Hara N, Gianani R, Zipris D. Kilham Rat Virus-induced type 1 diabetes involves beta cell infection and intra-islet JAK-STAT activation prior to insulitis. Virology 2014; 468-470:19-27. [PMID: 25129435 DOI: 10.1016/j.virol.2014.07.041] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 07/17/2014] [Accepted: 07/21/2014] [Indexed: 11/24/2022]
Abstract
We used the LEW1.WR1 rat model of Kilham Rat Virus (KRV)-induced type 1 diabetes (T1D) to test the hypothesis that disease mechanisms are linked with beta cell infection and intra-islet immune activation prior to insulitis. KRV induces genes involved in type I and type II interferon pathways in islet cell lines in vitro and in islets from day-5-infected animals in vivo via mechanisms that do not involve insulitis, beta cell apoptosis, or impaired insulin expression. Immunohistochemistry studies indicated that KRV protein is expressed in beta cells 5 days following infection. KRV induces the phosphorylation of Janus Kinase 1/2 (JAK1/2) and signal transducer and activator of transcription 1 (STAT-1) in islet cells via a mechanism that could involve TLR9 and NF-κB pathways. These data demonstrate for the first time that KRV-induced islet destruction is associated with beta cell infection and intra-islet innate immune upregulation early in the disease process.
Collapse
Affiliation(s)
- Aimon K Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO 80045, United States
| | - Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO 80045, United States
| | - Roberto Gianani
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO 80045, United States
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO 80045, United States.
| |
Collapse
|
12
|
Gottlieb PA, Alkanani AK, Michels AW, Lewis EC, Shapiro L, Dinarello CA, Zipris D. α1-Antitrypsin therapy downregulates toll-like receptor-induced IL-1β responses in monocytes and myeloid dendritic cells and may improve islet function in recently diagnosed patients with type 1 diabetes. J Clin Endocrinol Metab 2014; 99:E1418-26. [PMID: 24527714 PMCID: PMC4121034 DOI: 10.1210/jc.2013-3864] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CONTEXT Recent studies have implicated proinflammatory responses in the mechanism of type 1 diabetes (T1D). OBJECTIVE Our objective was to evaluate the safety and effects of therapy with the anti-inflammatory serum protein α1-antitrypsin (AAT) on islet function and innate immunity in recent-onset patients. DESIGN AND SETTING This was an open-label phase I trial at the Barbara Davis Center for Childhood Diabetes, University of Colorado Denver. PATIENTS Twelve recently diagnosed subjects with T1D with detectable C-peptides were included in the study. INTERVENTION Eight consecutive weekly infusions of 80 mg/kg of AAT were given. MAIN OUTCOME MEASURES PATIENTS were monitored for adverse effects of AAT therapy, C-peptide responses to a mixed-meal tolerance test, and toll-like receptor (TLR)-induced cellular IL-1β in monocytes and myeloid dendritic cells (mDCs). RESULTS No adverse effects were detected. AAT led to increased, unchanged, or moderately reduced levels of C-peptide responses compared with baseline in 5 patients. The total content of TLR4-induced cellular IL-1β in monocytes at 12 months after AAT therapy was 3-fold reduced compared with baseline (P < .05). Furthermore, at baseline, 82% of monocytes produced IL-1β, but at 12 months after therapy, the level decreased to 42%. Similar reductions were observed using TLR7/8 and TLR3 agonists in monocytes and mDCs. Unexpectedly, the reduction in cellular IL-1β was observed only 9 and 12 months after treatment but not in untreated diabetics. Improved β-cell function in the 5 AAT-treated individuals correlated with lower frequencies of monocytes and mDCs producing IL-1β compared with subjects without improvement of islet function (P < .04 and P < .02, respectively). CONCLUSIONS We hypothesize that AAT may have a beneficial effect on T1D in recently diagnosed patients that is associated with downmodulation of IL-1β.
Collapse
Affiliation(s)
- Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes (P.A.G., A.K.A., A.W.M., D.Z.) and Division of Infectious Diseases (C.A.D.), University of Colorado Denver, Aurora, Colorado 80045; Department of Clinical Biochemistry and Pharmacology (E.C.L.), Faculty of Health Sciences, Ben-Gurion University of the Negev, 84105 Beer-Sheva, Israel; and Department of Medicine (L.S.), Division of Infectious Diseases, Veterans Affairs Medical Center and University of Colorado Denver, Denver, Colorado 80202
| | | | | | | | | | | | | |
Collapse
|
13
|
Yang C, Jurczyk A, diIorio P, Norowski E, Brehm MA, Grant CW, Guberski DL, Greiner DL, Bortell R. Salicylate prevents virus-induced type 1 diabetes in the BBDR rat. PLoS One 2013; 8:e78050. [PMID: 24147110 PMCID: PMC3797740 DOI: 10.1371/journal.pone.0078050] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2013] [Accepted: 09/09/2013] [Indexed: 12/15/2022] Open
Abstract
Epidemiologic and clinical evidence suggests that virus infection plays an important role in human type 1 diabetes pathogenesis. We used the virus-inducible BioBreeding Diabetes Resistant (BBDR) rat to investigate the ability of sodium salicylate, a non-steroidal anti-inflammatory drug (NSAID), to modulate development of type 1 diabetes. BBDR rats treated with Kilham rat virus (KRV) and polyinosinic:polycytidylic acid (pIC, a TLR3 agonist) develop diabetes at nearly 100% incidence by ~2 weeks. We found distinct temporal profiles of the proinflammatory serum cytokines, IL-1β, IL-6, IFN-γ, IL-12, and haptoglobin (an acute phase protein) in KRV+pIC treated rats. Significant elevations of IL-1β and IL-12, coupled with sustained elevations of haptoglobin, were specific to KRV+pIC and not found in rats co-treated with pIC and H1, a non-diabetogenic virus. Salicylate administered concurrently with KRV+pIC inhibited the elevations in IL-1β, IL-6, IFN-γ and haptoglobin almost completely, and reduced IL-12 levels significantly. Salicylate prevented diabetes in a dose-dependent manner, and diabetes-free animals had no evidence of insulitis. Our data support an important role for innate immunity in virus-induced type 1 diabetes pathogenesis. The ability of salicylate to prevent diabetes in this robust animal model demonstrates its potential use to prevent or attenuate human autoimmune diabetes.
Collapse
Affiliation(s)
- Chaoxing Yang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Agata Jurczyk
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Philip diIorio
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Elaine Norowski
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Michael A. Brehm
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Christian W. Grant
- Biomedical Research Models, Worcester, Massachusetts, United States of America
| | - Dennis L. Guberski
- Biomedical Research Models, Worcester, Massachusetts, United States of America
| | - Dale L. Greiner
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - Rita Bortell
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- *E-mail:
| |
Collapse
|
14
|
Hara N, Alkanani AK, Dinarello CA, Zipris D. Modulation of virus-induced innate immunity and type 1 diabetes by IL-1 blockade. Innate Immun 2013; 20:574-84. [PMID: 24062197 DOI: 10.1177/1753425913502242] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/30/2013] [Indexed: 12/27/2022] Open
Abstract
We used the LEW1.WR1 model of Kilham rat virus (KRV)-induced type 1 diabetes (T1D) to test the hypothesis that blocking IL-1 pathways early in the course of the disease can modulate virus-induced innate immunity and prevent disease progression. Administering KRV plus IL-1 receptor antagonist (Anakinra) for 14 d prevented insulitis and T1D. Anakinra reversed the KRV-induced systemic inflammation evidenced by the accumulation of T cells in the spleen and pancreatic lymph nodes on d 5 post-infection. Blocking IL-1 modulated the level of IRF-7 and IL-6 gene expression in the spleen and the p40 subunit of IL-12 and IL-23 in the serum. Anakinra did not interfere with the ability of LEW1.WR1 rats to clear the virus from the spleen, pancreatic lymph nodes or serum. Consistent with these data, normal levels of KRV-specific adaptive immune responses were detected in in the spleen and peripheral blood of the treated animals. Finally, blocking IL-1 pathways reversed the KRV-induced modulation of gut bacterial communities. The data may imply that IL-1 pathways are directly linked with early mechanisms whereby KRV infection leads to islet destruction, raising the hypothesis that blocking IL-1 pathways early in the course of the disease could be a useful therapeutic approach for disease prevention.
Collapse
Affiliation(s)
- Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, USA
| | - Aimon K Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, USA
| | - Charles A Dinarello
- Division of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, USA
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
15
|
Histone deacetylase inhibitor suppresses virus-induced proinflammatory responses and type 1 diabetes. J Mol Med (Berl) 2013; 92:93-102. [PMID: 23982318 DOI: 10.1007/s00109-013-1078-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2013] [Revised: 07/02/2013] [Accepted: 08/14/2013] [Indexed: 01/04/2023]
Abstract
UNLABELLED Microbial infections are hypothesized to play a key role in the mechanism leading to type 1 diabetes (T1D). We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced islet destruction to better understand how virus infection triggers T1D. Inoculation of the LEW1.WR1 rat with KRV results in systemic inflammation followed by insulitis and islet destruction 2-4 weeks post-infection. In this study, we evaluated the effect of treatment with the anti-inflammatory histone deacetylase inhibitor (HDACi) ITF-2357 on KRV-induced immunity and disease progression. Administering LEW1.WR1 rats with KRV plus ITF-2357 on 14 consecutive days beginning on the day of infection protected animals from islet infiltration and T1D. ITF-2357 reversed KRV-induced T and B cell accumulation in the spleen or pancreatic lymph nodes on day 5 following infection. Moreover, ITF-2357 reduced the expression level of KRV-induced p40 subunit of IL-12/IL-23 in spleen cells in vitro and in the peripheral blood in vivo. ITF-2357 suppressed the KRV-induced expression of transcripts for IRF-7 in the rat INS-1 beta cell line. ITF-2357 increased the virus-induced IL-6 gene expression in the spleen, but did not alter the ability of LEW1.WR1 rats to develop normal KRV-specific humoral and cellular immune responses and clear the virus from the pancreatic lymph nodes, spleen, and serum. Finally, ITF-2357 reversed virus-induced modulation of bacterial communities in the intestine early following infection. The data suggest that targeting innate immune pathways with inhibitors of HDAC might represent an efficient therapeutic strategy for preventing T1D. KEY MESSAGE Microbial infections have been implicated in triggering type 1 diabetes in humans and animal models. The LEW1.WR1 rat develops inflammation and T1D following infection with Kilham rat virus. The histone deacetylase inhibitor ITF-2357 suppresses virus-induced inflammation and prevents diabetes. ITF-2357 prevents T1D without altering virus-specific adaptive immunity or virus clearance. ITF-2357 therapy may be an efficient approach to prevent T1D in genetically susceptible individuals.
Collapse
|
16
|
Bortell R, Yang C. The BB rat as a model of human type 1 diabetes. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2013; 933:31-44. [PMID: 22893399 DOI: 10.1007/978-1-62703-068-7_3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The BB rat is an important rodent model of human type 1 diabetes (T1D) and has been used to study mechanisms of diabetes pathogenesis as well as to investigate potential intervention therapies for clinical trials. The Diabetes-Prone BB (BBDP) rat spontaneously develops autoimmune T1D between 50 and 90 days of age. The Diabetes-Resistant BB (BBDR) rat has similar diabetes-susceptible genes as the BBDP, but does not become diabetic in viral antibody-free conditions. However, the BBDR rat can be induced to develop T1D in response to certain treatments such as regulatory T cell (T(reg)) depletion, toll-like receptor ligation, or virus infection. These diabetes-inducible rats develop hyperglycemia under well-controlled circumstances and within a short, predictable time frame (14-21 days), thus facilitating their utility for investigations of specific stages of diabetes development. Therefore, these rat strains are invaluable models for studying autoimmune diabetes and the role of environmental factors in its development, of particular importance due to the influx of studies associating virus infection and human T1D.
Collapse
Affiliation(s)
- Rita Bortell
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, USA.
| | | |
Collapse
|
17
|
Abstract
Type 1 diabetes mellitus (T1DM) is a multi-factorial autoimmune disease determined by the interaction of genetic, environmental and immunologic factors. One of the environmental risk factors identified by a series of independent studies is represented by viral infection, with strong evidence showing that viruses can indeed infect pancreatic beta cells with consequent effects ranging from functional damage to cell death. In this chapter we review the data obtained both in man and in experimental animal models in support of the potential participation of viral infections to Type 1 diabetes pathogenesis, with a particular emphasis on virus-triggered islet inflammation, beta-cell dysfunction and autoimmunity.
Collapse
|
18
|
Hara N, Alkanani AK, Ir D, Robertson CE, Wagner BD, Frank DN, Zipris D. The role of the intestinal microbiota in type 1 diabetes. Clin Immunol 2012; 146:112-9. [PMID: 23314185 DOI: 10.1016/j.clim.2012.12.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2012] [Revised: 11/30/2012] [Accepted: 12/01/2012] [Indexed: 02/07/2023]
Abstract
The digestive tract hosts trillions of bacteria that interact with the immune system and can influence the balance between pro-inflammatory and regulatory immune responses. Recent studies suggest that alterations in the composition of the intestinal microbiota may be linked with the development of type 1 diabetes (T1D). Data from the biobreeding diabetes prone (BBDP) and the LEW1.WR1 models of T1D support the hypothesis that intestinal bacteria may be involved in early disease mechanisms. The data indicate that cross-talk between the gut microbiota and the innate immune system may be involved in islet destruction. Whether a causal link between intestinal microbiota and T1D exists, the identity of the bacteria and the mechanism whereby they promote the disease remain to be examined. A better understanding of the interplay between microbes and innate immune pathways in early disease stages holds promise for the design of immune interventions and disease prevention in genetically susceptible individuals.
Collapse
Affiliation(s)
- Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
Alkanani AK, Rewers M, Dong F, Waugh K, Gottlieb PA, Zipris D. Dysregulated Toll-like receptor-induced interleukin-1β and interleukin-6 responses in subjects at risk for the development of type 1 diabetes. Diabetes 2012; 61:2525-33. [PMID: 22751696 PMCID: PMC3447890 DOI: 10.2337/db12-0099] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
We tested the hypothesis that altered Toll-like receptor (TLR) signaling may be involved in early stages of type 1 diabetes (T1D). To do so, we analyzed TLR-induced interleukin (IL)-1β and IL-6 responses in freshly isolated peripheral blood mononuclear cells (PBMNCs) from seropositive compared with seronegative subjects. Similar frequencies of myeloid dendritic cells (mDCs), plasmacytoid DCs (pDCs), and monocytes were observed in seropositive and seronegative subjects. Subjects with autoantibodies had increased proportions of monocytes expressing IL-1β ex vivo. Activating PBMNCs with TLR3, TLR4, or TLR7/8 agonists in vitro led to increased percentages of IL-1β-expressing monocytes and mDCs from seropositive versus seronegative subjects. TLR ligation also resulted in a diminished IL-6 response in seropositive individuals as lower frequencies of IL-6-expressing monocytes and mDCs were induced. The dysregulated TLR-induced IL-1β and IL-6 pathways were more readily detectable in children aged <11 years and from 11 to <21 years, respectively, and did not involve altered HbA(1c) or the presence of one or more autoantibodies. Finally, subjects with autoantibodies had lower amounts of serum chemokine (C-X-C motif) ligand 10 compared with autoantibody-negative subjects. Our data may imply that alterations in innate immune pathways are detectable in genetically susceptible individuals and could be linked with the early course of T1D.
Collapse
|
20
|
Hara N, Alkanani AK, Ir D, Robertson CE, Wagner BD, Frank DN, Zipris D. Prevention of virus-induced type 1 diabetes with antibiotic therapy. THE JOURNAL OF IMMUNOLOGY 2012; 189:3805-14. [PMID: 22988033 DOI: 10.4049/jimmunol.1201257] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Microbes were hypothesized to play a key role in the progression of type 1 diabetes (T1D). We used the LEW1.WR1 rat model of Kilham rat virus (KRV)-induced T1D to test the hypothesis that the intestinal microbiota is involved in the mechanism leading to islet destruction. Treating LEW1.WR1 rats with KRV and a combination of trimethoprim and sulfamethoxazole (Sulfatrim) beginning on the day of infection protected the rats from insulitis and T1D. Pyrosequencing of bacterial 16S rRNA and quantitative RT-PCR indicated that KRV infection resulted in a transient increase in the abundance of Bifidobacterium spp. and Clostridium spp. in fecal samples from day 5- but not day 12-infected versus uninfected animals. Similar alterations in the gut microbiome were observed in the jejunum of infected animals on day 5. Treatment with Sulfatrim restored the level of intestinal Bifidobacterium spp. and Clostridium spp. We also observed that virus infection induced the expression of KRV transcripts and the rapid upregulation of innate immune responses in Peyer's patches and pancreatic lymph nodes. However, antibiotic therapy reduced the virus-induced inflammation as reflected by the presence of lower amounts of proinflammatory molecules in both the Peyer's patches and pancreatic lymph nodes. Finally, Sulfatrim treatment reduced the number of B cells in Peyer's patches and downmodulated adaptive immune responses to KRV, but did not interfere with antiviral Ab responses or viral clearance from the spleen, pancreatic lymph nodes, and serum. The data suggest that gut microbiota may be involved in promoting virus-induced T1D in the LEW1.WR1 rat model.
Collapse
Affiliation(s)
- Naoko Hara
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
BACKGROUND Rat models of diabetes have emerged as a powerful experimental tool for addressing the role of microbial pathogens in the mechanism of autoimmune diabetes. We have used the biobreeding diabetes resistant and LEW1.WR1 rat models to identify the role of virus-induced innate immunity in the mechanism of type 1 diabetes. METHODS Groups of rats 21-25 days of age were left untreated, injected i.p. with 1×10(7) PFU of Kilham rat virus (KRV) only, or with 1-3 µg/g body-weight-purified toll-like receptor agonists on three consecutive days and infected with 1×10(7) PFU of KRV on the following day. Spleens and pancreatic lymph nodes were recovered 5 days after infection and used for gene array analysis. To test the role of inflammation in diabetes, rats injected with KRV only or Poly(I:C) plus KRV were also administered with 2 or 0.2 µg/g body weight of dexamethasone and followed for diabetes for 40 days. RESULTS KRV induced the expression of a vast array of proinflammatory genes in pancreatic lymph nodes on day 5 following infection. Brief dexamethasone therapy downmodulated inflammation and completely blocked diabetes. CONCLUSIONS Our data suggest a strong association between early virus-induced proinflammatory responses and islet destruction and raise the possibility that targeting innate immune pathways in the early stages of diabetes may be a useful strategy for disease prevention.
Collapse
Affiliation(s)
- Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, CO 80045, USA.
| |
Collapse
|
22
|
Navratil V, de Chassey B, Combe CR, Lotteau V. When the human viral infectome and diseasome networks collide: towards a systems biology platform for the aetiology of human diseases. BMC SYSTEMS BIOLOGY 2011; 5:13. [PMID: 21255393 PMCID: PMC3037315 DOI: 10.1186/1752-0509-5-13] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2010] [Accepted: 01/21/2011] [Indexed: 12/15/2022]
Abstract
Background Comprehensive understanding of molecular mechanisms underlying viral infection is a major challenge towards the discovery of new antiviral drugs and susceptibility factors of human diseases. New advances in the field are expected from systems-level modelling and integration of the incessant torrent of high-throughput "-omics" data. Results Here, we describe the Human Infectome protein interaction Network, a novel systems virology model of a virtual virus-infected human cell concerning 110 viruses. This in silico model was applied to comprehensively explore the molecular relationships between viruses and their associated diseases. This was done by merging virus-host and host-host physical protein-protein interactomes with the set of genes essential for viral replication and involved in human genetic diseases. This systems-level approach provides strong evidence that viral proteomes target a wide range of functional and inter-connected modules of proteins as well as highly central and bridging proteins within the human interactome. The high centrality of targeted proteins was correlated to their essentiality for viruses' lifecycle, using functional genomic RNAi data. A stealth-attack of viruses on proteins bridging cellular functions was demonstrated by simulation of cellular network perturbations, a property that could be essential in the molecular aetiology of some human diseases. Networking the Human Infectome and Diseasome unravels the connectivity of viruses to a wide range of diseases and profiled molecular basis of Hepatitis C Virus-induced diseases as well as 38 new candidate genetic predisposition factors involved in type 1 diabetes mellitus. Conclusions The Human Infectome and Diseasome Networks described here provide a unique gateway towards the comprehensive modelling and analysis of the systems level properties associated to viral infection as well as candidate genes potentially involved in the molecular aetiology of human diseases.
Collapse
Affiliation(s)
- Vincent Navratil
- Université de Lyon, IFR128 BioSciences Lyon-Gerland, Lyon 69007, France.
| | | | | | | |
Collapse
|
23
|
Mori K, Yoshida K. Viral infection in induction of Hashimoto's thyroiditis: a key player or just a bystander? Curr Opin Endocrinol Diabetes Obes 2010; 17:418-24. [PMID: 20625285 DOI: 10.1097/med.0b013e32833cf518] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Viral infection activates both the innate and adaptive immunity and is implicated as a trigger of autoimmune diseases including Hashimoto's thyroiditis. This review summarizes our knowledge respecting the role of viral infection in the cause of Hashimoto's thyroiditis. RECENT FINDINGS Components of several viruses such as hepatitis C virus, human parvovirus B19, coxsackie virus and herpes virus are detected in the thyroid of Hashimoto's thyroiditis patients. Bystander activation of autoreactive T cells may be involved in triggering intrathyroidal inflammation. Signaling molecules associated with antiviral responses including Toll-like receptors may participate in Hashimoto's thyroiditis induction. However, studies have provided insufficient direct evidence for the viral hypothesis in Hashimoto's thyroiditis. SUMMARY Despite interesting circumstantial evidence, whether viral infection is responsible for Hashimoto's thyroiditis remains unclear. Studies addressing this issue are required to substantiate a contribution from viral infection to Hashimoto's thyroiditis and, consequently, the prospect for developing preventive modalities for Hashimoto's thyroiditis.
Collapse
Affiliation(s)
- Kouki Mori
- Division of Nephrology, Endocrinology and Vascular Medicine, Tohoku University Graduate School of Medicine, Aoba-ku, Sendai, Japan. mail:
| | | |
Collapse
|
24
|
Thomas R. The balancing act of autoimmunity: central and peripheral tolerance versus infection control. Int Rev Immunol 2010; 29:211-33. [PMID: 20367141 DOI: 10.3109/08830180903434219] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Genetic associations with autoimmune disease are enriched in immune response regulators. The immune system in individuals at genetic risk of autoimmunity must balance pressures on the innate and adaptive immune system, most notably infection control, with those of maintaining self-tolerance or controlling autoimmune inflammation. In spite of multiple tolerance mechanisms, inflammation becomes chronic in autoimmune disease, and complete resolution is difficult. This article proposes a perspective on the pathogenesis of autoimmunity-focusing on rheumatoid arthritis and type 1 diabetes-integrating clinical advances and animal models with the role that colonizing micro-organisms play in the balance between tolerance and autoimmunity.
Collapse
Affiliation(s)
- Ranjeny Thomas
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, University of Queensland, Princess Alexandra Hospital, Australia.
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Type 1 diabetes (T1D) is an autoimmune disease typically believed to result from malfunctions in adaptive immune response signaling which result in activation of self-reactive T cells. However, recent research has indicated components of the innate immune response as having a key role in the initiation of the autoimmune process of T1D. This review will highlight recent studies which examined the role of innate immune response signaling and the connections to T1D pathogenesis. RECENT FINDINGS Investigations indicate that components of innate immunity, including inflammation and Toll-like receptor signaling, are involved in pancreatic islet infiltration and insulitis. Recent studies examining the role of viral infections in T1D development also implicate innate immune response signaling in disease pathogenesis. SUMMARY Current research indicates that components of innate immune response signaling are involved in the initiation of the autoimmune process which results in the eventual destruction of beta cells during T1D pathogenesis. Continuing efforts by researchers to uncover the molecular pathways of innate immunity linked to T1D development could potentially lead to therapeutics capable of preventing and curing the autoimmune disease.
Collapse
Affiliation(s)
| | | | - Rita Bortell
- Correspondence to: Rita Bortell, PhD; Diabetes Division, Suite 218, 373 Plantation Street, Worcester, MA 01605, Tel: 508-856-3788; fax. 508-856-4093;
| |
Collapse
|
26
|
Current world literature. Curr Opin Endocrinol Diabetes Obes 2010; 17:177-85. [PMID: 20190584 DOI: 10.1097/med.0b013e3283382286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
27
|
Londono P, Komura A, Hara N, Zipris D. Brief dexamethasone treatment during acute infection prevents virus-induced autoimmune diabetes. Clin Immunol 2010; 135:401-11. [PMID: 20167539 DOI: 10.1016/j.clim.2010.01.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2009] [Revised: 01/19/2010] [Accepted: 01/19/2010] [Indexed: 10/19/2022]
Abstract
We used the LEW1.WR1 rat to test the hypothesis that Kilham rat virus-induced innate immune activation is involved in the mechanism of autoimmune diabetes. Animals were treated with dexamethasone, an anti-inflammatory glucocorticoid, beginning on the day of infection. Administering dexamethasone on five consecutive days completely blocked the disease. Strikingly, a single dose of dexamethasone was sufficient to prevent islet destruction. Dexamethasone downmodulated inflammation and restored normal ratios between CD8(+) and CD4(+)CD25(+)Foxp3(+) cells in the spleen. Finally, dexamethasone therapy lowered the frequency of splenic anti-virus CD8(+) T cells, but did not interfere with the ability of the host to generate anti-KRV antibodies and eliminate the virus from the spleen. Our data demonstrate a strong association between early virus-induced proinflammatory responses and islet destruction and raise the possibility that targeting innate immune pathways in the early stages of diabetes may be a useful strategy for disease prevention.
Collapse
Affiliation(s)
- Pilar Londono
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
28
|
Abstract
The immune system plays an important role in the development of personalized medicine for a variety of diseases including cancer, autoimmune diseases, and infectious diseases. Immunoinformatics, or computational immunology, is an emerging area that provides fundamental methodologies in the study of immunomics, that is, immune-related genomics and proteomics. The integration of immunoinformatics with systems biology approaches may lead to a better understanding of immune-related diseases at various systems levels. Such methods can contribute to translational studies that bring scientific discoveries of the immune system into better clinical practice. One of the most intensely studied areas of the immune system is immune epitopes. Epitopes are important for disease understanding, host-pathogen interaction analyses, antimicrobial target discovery, and vaccine design. The information about genetic diversity of the immune system may help define patient subgroups for individualized vaccine or drug development. Cellular pathways and host immune-pathogen interactions have a crucial impact on disease pathogenesis and immunogen design. Epigenetic studies may help understand how environmental changes influence complex immune diseases such as allergy. High-throughput technologies enable the measurements and catalogs of genes, proteins, interactions, and behavior. Such perception may contribute to the understanding of the interaction network among humans, vaccines, and drugs, to enable new insights of diseases and therapeutic responses. The integration of immunomics information may ultimately lead to the development of optimized vaccines and drugs tailored to personalized prevention and treatment. An immunoinformatics portal containing relevant resources is available at http://immune.pharmtao.com.
Collapse
|