1
|
Hu Y, Huang J, Wang S, Sun X, Wang X, Yu H. Deciphering Autoimmune Diseases: Unveiling the Diagnostic, Therapeutic, and Prognostic Potential of Immune Repertoire Sequencing. Inflammation 2025; 48:676-695. [PMID: 38914737 DOI: 10.1007/s10753-024-02079-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/31/2024] [Accepted: 06/08/2024] [Indexed: 06/26/2024]
Abstract
Autoimmune diseases (AIDs) are immune system disorders where the body exhibits an immune response to its own antigens, causing damage to its own tissues and organs. The pathogenesis of AIDs is incompletely understood. However, recent advances in immune repertoire sequencing (IR-seq) technology have opened-up a new avenue to study the IR. These studies have revealed the prevalence in IR alterations, potentially inducing AIDs by disrupting immune tolerance and thereby contributing to our comprehension of AIDs. IR-seq harbors significant potential for the clinical diagnosis, personalized treatment, and prognosis of AIDs. This article reviews the application and progress of IR-seq in diseases, such as multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, and type 1 diabetes, to enhance our understanding of the pathogenesis of AIDs and offer valuable references for the diagnosis and treatment of AIDs.
Collapse
Affiliation(s)
- Yuelin Hu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Jialing Huang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Shuqing Wang
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Sun
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Xin Wang
- School of Basic Medical Sciences, Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China
| | - Hongsong Yu
- Department of Immunology, Special Key Laboratory of Ocular Diseases of Guizhou Province, Zunyi Medical University, Zunyi, Guizhou, P.R. China.
| |
Collapse
|
2
|
Du M, Li S, Jiang J, Ma X, Liu L, Wang T, Zhang J, Niu D. Advances in the Pathogenesis and Treatment Strategies for Type 1 Diabetes Mellitus. Int Immunopharmacol 2025; 148:114185. [PMID: 39893858 DOI: 10.1016/j.intimp.2025.114185] [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: 10/17/2024] [Revised: 01/26/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Type 1 diabetes (T1D) is a complex autoimmune disorder distinguished by the infiltration of immune cells into pancreatic islets, primarily resulting in damage to pancreatic β-cells. Despite extensive research, the precise pathogenesis of T1D remains elusive, with its etiology linked to a complex interplay of genetic, immune, and environmental factors. While genetic predispositions, such as HLA and other susceptibility genes, are necessary, they do not fully account for disease development. Environmental influences such as viral infections and dietary factors may contribute to the disease by affecting the immune system and epigenetic modifications. Additionally, endogenous retroviruses (ERVs) might play a role in T1D pathogenesis. Current therapeutic approaches, including insulin replacement therapy, immune omodulatory therapy, autoantigen immunotherapy, organ transplantation, and genetic modification, offer potential to alter disease progression but are still constrained by limitations. This review presents updated knowledge on T1D, with a focus on risk factors, predisposing hypotheses, and recent advancements in therapeutic strategies.
Collapse
Affiliation(s)
- Meiheng Du
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, Zhejiang 311300, China
| | - Sihong Li
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, Zhejiang 311300, China
| | - Jun Jiang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, Zhejiang 311300, China
| | - Xiang Ma
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, Zhejiang 311300, China
| | - Lu Liu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, Zhejiang 311300, China
| | - Tao Wang
- Nanjing Kgene Genetic Engineering Co., Ltd, Nanjing, Jiangsu 211300, China
| | - Jufang Zhang
- Department of Plastic Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| | - Dong Niu
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Provincial Engineering Research Center for Animal Health Diagnostics & Advanced Technology, Zhejiang International Science and Technology Cooperation Base for Veterinary Medicine and Health Management, China Australia Joint Laboratory for Animal Health Big Data Analytics, Hangzhou, Zhejiang 311300, China.
| |
Collapse
|
3
|
Vandewalle J, Desouter AK, Van der Auwera BJ, Chapaza KB, Nobels F, Abrams P, Lebrethon MC, Lapauw B, Keymeulen B, Gorus FK, Van de Casteele M. The stage- and subgroup-specific impact of non-HLA polymorphisms on preclinical type 1 diabetes progression. Heliyon 2025; 11:e42156. [PMID: 40196768 PMCID: PMC11947702 DOI: 10.1016/j.heliyon.2025.e42156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/28/2024] [Accepted: 01/20/2025] [Indexed: 04/09/2025] Open
Abstract
Besides variation within the HLA gene complex determining a major part of genetic susceptibility to Type 1 diabetes, genome-wide association studies have identified over 60 non-HLA loci also contributing to disease risk. While individual single nucleotide polymorphisms (SNPs) have limited predictive power, genetic risk scores (GRS) can identify at-risk individuals. However, current models do not fully capture the heterogeneous progression of asymptomatic islet autoimmunity, especially in autoantibody-positive subjects. In this study, we investigated the additional stage-specific impact of 17 non-HLA loci on previously established prediction models in 448 persistently autoantibody-positive first-degree relatives. Cox regression and Kaplan Meier survival analysis were used to assess their influence on progression from single to multiple autoantibody-positivity, and from there to clinical onset. FUT2 and CTSH significantly accelerated progression of single to multiple autoAb-positivity, but only in presence of insulin autoantibodies and HLA-DQ2/DQ8, respectively. At the stage of multiple autoantibody-positivity, progression to clinical onset was impacted by various non-HLA SNPs either as independent predictors (GLIS3, CENPW, IL2, GSDM, MEG3A, and NRP-1) or through interaction with HLA class I alleles (CLEC16A, NRP-1, TCF7L2), maternal diabetes status (CTSH), or a high-risk autoantibody-profile (CD226). Our data indicate that, unlike for GRS, the weight of distinct non-HLA polymorphisms varies significantly among individuals at risk, depending on disease stage and other stage-specific risk factors. They refine our previous stage-specific prediction models including age, autoantibody-profile, HLA genotype, and other non-HLA SNPs, and emphasize the importance of stratifying accordingly to personalize time-to-event prediction in risk groups, or for preparing or interpreting prevention trials.
Collapse
Affiliation(s)
- Julie Vandewalle
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Aster K. Desouter
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetes and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | | | - Kaven B. Chapaza
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Frank Nobels
- Department of Endocrinology, OLV Hospital Aalst, Aalst, Belgium
| | - Pascale Abrams
- Department of Endocrinology and Diabetology, GZA Hospitals Antwerp, Wilrijk, Antwerp, Belgium
| | | | - Bruno Lapauw
- Department of Endocrinology, University Hospital Ghent–UGent, Ghent, Belgium
| | - Bart Keymeulen
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetes and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Frans K. Gorus
- Diabetes Research Center, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetes and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | | | | |
Collapse
|
4
|
Ratajczak W, Jones AG, Atkinson SD, Kelly C. Type 1 Diabetes Risk Variants Reduce Beta Cell Function. Genes (Basel) 2025; 16:172. [PMID: 40004501 PMCID: PMC11855905 DOI: 10.3390/genes16020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/16/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
INTRODUCTION The variants rs10517086 and rs1534422 are predictive of type 1 diabetes mellitus (T1DM) development and poor residual β cell function within the first year of diagnosis. However, the mechanism by which risk is conferred is unknown. We explored the impact of both variants on β cell function in vitro and assessed their relationship with C-peptide in people with T1DM and type 2 diabetes mellitus (T2DM). METHODS Using CRISPR/Cas9, the variants were introduced into a β cell line (BRIN-BD11) and a T cell line (Jurkat cells) from which the conditioned media was applied to otherwise healthy β cells to model the inflammatory environment associated with these variants. RESULTS Both variants significantly reduced glucose-stimulated insulin secretion, increased production of pro-inflammatory cytokines and reduced expression of several β cell markers and transcription factors (KCNJ11, KCNQ1, SCL2A2, GCK, NKX6.1, Pdx1 NGN3). However, HNF1A was significantly upregulated in the presence of both variants. We subsequently silenced HNF1A in variant expressing BRIN-BD11 cells using siRNA and found that gene expression profiles were normalised. Induction of each variant significantly increased expression of the lncRNAs they encode, which was normalised upon HNF1A silencing. Analysis of the DARE (Diabetes Alliance for Research in England) study revealed an association of rs10517086_A genotype with C-peptide in 153 individuals with T1DM, but not in 417 people with T2DM. CONCLUSIONS These data suggest that rs1534422 and rs10517086 exert multiple insults on the β cell through excessive upregulation of HNF1A and induction of pro-inflammatory cytokines, and highlight their utility as prognostic markers of β cell function.
Collapse
Affiliation(s)
- Wiktoria Ratajczak
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Hospital Campus, Glenshane Road, Derry BT48 7JL, UK;
| | - Angus G. Jones
- University of Exeter Medical School, RILD Building, RD&E Hospital Wonford, Barrack Road, Exeter EX1 2LU, UK;
| | - Sarah D. Atkinson
- School of Biomedical Sciences, Ulster University, Cromore Road, Coleraine BT52 1SA, UK;
| | - Catriona Kelly
- Personalised Medicine Centre, School of Medicine, Ulster University, C-TRIC Building, Altnagelvin Hospital Campus, Glenshane Road, Derry BT48 7JL, UK;
| |
Collapse
|
5
|
Alvarez F, Acuff NV, La Muraglia GM, Sabri N, Milla ME, Mooney JM, Mackey MF, Peakman M, Piccirillo CA. The IL-2 SYNTHORIN molecule promotes functionally adapted Tregs in a preclinical model of type 1 diabetes. JCI Insight 2024; 9:e182064. [PMID: 39704171 DOI: 10.1172/jci.insight.182064] [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: 04/19/2024] [Accepted: 10/25/2024] [Indexed: 12/21/2024] Open
Abstract
Deficits in IL-2 signaling can precipitate autoimmunity by altering the function and survival of FoxP3+ regulatory T cells (Tregs) while high concentrations of IL-2 fuel inflammatory responses. Recently, we showed that the non-beta IL-2 SYNTHORIN molecule SAR444336 (SAR'336) can bypass the induction of autoimmune and inflammatory responses by increasing its reliance on IL-2 receptor α chain subunit (CD25) to provide a bona fide IL-2 signal selectively to Tregs, making it an attractive approach for the control of autoimmunity. In this report, we further demonstrate that SAR'336 can support non-beta IL-2 signaling in murine Tregs and limit NK and CD8+ T cells' proliferation and function. Using a murine model of spontaneous type 1 diabetes, we showed that the administration of SAR'336 slows the development of disease in mice by decreasing the degree of insulitis through the expansion of antigen-specific Tregs over Th1 cells in pancreatic islets. Specifically, SAR'336 promoted the differentiation of IL-33-responsive (ST2+), IL-10-producing GATA3+ Tregs over other Treg subsets in the pancreas, demonstrating the ability of this molecule to further orchestrate Treg adaptation. These results offer insight into the capacity of SAR'336 to generate highly specialized, tissue-localized Tregs that promote restoration of homeostasis during ongoing autoimmune disease.
Collapse
Affiliation(s)
- Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Program in Infectious Diseases and Immunology in Global Health, the Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), RI-MUHC, Montreal, Quebec, Canada
| | | | | | - Nazila Sabri
- Synthorx, a Sanofi company, La Jolla, California, USA
| | | | - Jill M Mooney
- Synthorx, a Sanofi company, La Jolla, California, USA
| | | | | | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Program in Infectious Diseases and Immunology in Global Health, the Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), RI-MUHC, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Nishimoto Y, Hashimoto N, Kido N, Irahara A, Takeuchi T, Takabe M, Ishihara S, Kinoshita Y, Ohara T. Prevalence of celiac disease in patients with type 1 diabetes mellitus: a single-center cross-sectional cohort study. J Clin Biochem Nutr 2024; 75:213-216. [PMID: 39583973 PMCID: PMC11579853 DOI: 10.3164/jcbn.24-39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/04/2024] [Indexed: 11/26/2024] Open
Abstract
Type 1 diabetes mellitus (T1DM) may be associated with other autoimmune diseases. Celiac disease (CD), another autoimmune disorder that mainly affects the small intestine, is caused by intolerance to gluten ingestion. CD has a higher prevalence in patients with T1DM than in the general population. However, the prevalence of CD in patients with T1DM in Japan is unknown. This study investigated the prevalence of CD in Japanese patients with T1DM. We included 115 patients with T1DM treated at Hyogo Brain and Heart Center from December 2020 to April 2021. A questionnaire survey about dietary habits and abdominal symptoms was administered, and serum anti-tissue transglutaminase (TTG) antibody titers were determined for all participants. A CD (CD-seropositive) diagnosis was based on TTG levels >10 U/ml. Fifty-eight patients (50.4%) had some abdominal symptoms (such as constipation, diarrhea, and abdominal pain). The average TTG-IgA antibody titer was 0.75 ± 0.49 U/ml and negative (<10 U/ml) in all patients. In conclusion, the prevalence of CD among patients with T1DM at our hospital was 0%. Thus, the prevalence of CD in Japan is low compared to that in other countries, even among patients with T1DM, who are considered to have high comorbidity rates.
Collapse
Affiliation(s)
- Yuki Nishimoto
- Department of Diabetes and Endocrinology, Hyogo Prefectural Harima-Himeji General Medical Center, 3-264, Kamiya-cho, Himeji-shi, Hyogo 670-8560, Japan
| | - Naoko Hashimoto
- Department of Diabetes and Endocrinology, Hyogo Prefectural Harima-Himeji General Medical Center, 3-264, Kamiya-cho, Himeji-shi, Hyogo 670-8560, Japan
| | - Nozomi Kido
- Department of Diabetes and Endocrinology, Kobe University Hospital, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe-shi, Hyogo 650-0017, Japan
| | - Aya Irahara
- Department of Diabetes and Endocrinology, Hyogo Prefectural Harima-Himeji General Medical Center, 3-264, Kamiya-cho, Himeji-shi, Hyogo 670-8560, Japan
| | - Takehito Takeuchi
- Department of Diabetes and Endocrinology, Hyogo Prefectural Harima-Himeji General Medical Center, 3-264, Kamiya-cho, Himeji-shi, Hyogo 670-8560, Japan
| | - Michinori Takabe
- Takabe Diabetes Clinic, 2F Miyanishi building, 4-7-1 Miyanishi-cho, Himeji-shi, Hyogo 670-0837, Japan
| | - Shunji Ishihara
- Department of Gastroenterology and Hepatology, Shimane University School of Medicine, 89-1 Enya-cho, Izumo-shi, Shimane 693-8501, Japan
| | - Yoshikazu Kinoshita
- Director of Hospital, Hyogo Prefectural Harima-Himeji General Medical Center, 3-264 Kamiya-cho, Himeji-shi, Hyogo 670-8560, Japan
| | - Takeshi Ohara
- Department of Diabetes and Endocrinology, Hyogo Prefectural Harima-Himeji General Medical Center, 3-264, Kamiya-cho, Himeji-shi, Hyogo 670-8560, Japan
| |
Collapse
|
7
|
Golden GJ, Wu VH, Hamilton JT, Amses KR, Shapiro MR, Japp AS, Liu C, Pampena MB, Kuri-Cervantes L, Knox JJ, Gardner JS, Atkinson MA, Brusko TM, Prak ETL, Kaestner KH, Naji A, Betts MR. Immune perturbations in human pancreas lymphatic tissues prior to and after type 1 diabetes onset. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590798. [PMID: 39345402 PMCID: PMC11429609 DOI: 10.1101/2024.04.23.590798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Autoimmune destruction of pancreatic β cells results in type 1 diabetes (T1D), with pancreatic immune infiltrate representing a key feature in this process. Studies of human T1D immunobiology have predominantly focused on circulating immune cells in the blood, while mouse models suggest diabetogenic lymphocytes primarily reside in pancreas-draining lymph nodes (pLN). A comprehensive study of immune cells in human T1D was conducted using pancreas draining lymphatic tissues, including pLN and mesenteric lymph nodes, and the spleen from non-diabetic control, β cell autoantibody positive non-diabetic (AAb+), and T1D organ donors using complementary approaches of high parameter flow cytometry and CITEseq. Immune perturbations suggestive of a proinflammatory environment were specific for T1D pLN and AAb+ pLN. In addition, certain immune populations correlated with high T1D genetic risk independent of disease state. These datasets form an extensive resource for profiling human lymphatic tissue immune cells in the context of autoimmunity and T1D.
Collapse
Affiliation(s)
- Gregory J Golden
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Vincent H Wu
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jacob T Hamilton
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Kevin R Amses
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Melanie R Shapiro
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
| | - Alberto Sada Japp
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Chengyang Liu
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Maria Betina Pampena
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Leticia Kuri-Cervantes
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - James J Knox
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Jay S Gardner
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, College of Medicine, Gainesville, FL 32610, USA
- Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Eline T Luning Prak
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Klaus H Kaestner
- Department of Genetics and Institute for Diabetes, Obesity, and Metabolism, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Ali Naji
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Michael R Betts
- Department of Microbiology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
- Institute for Immunology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
8
|
Paul RK, Raza K. Natural hypoglycaemic bioactives: Newer avenues and newer possibilities. Phytother Res 2024; 38:4428-4452. [PMID: 38990182 DOI: 10.1002/ptr.8281] [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: 01/09/2024] [Revised: 06/09/2024] [Accepted: 06/13/2024] [Indexed: 07/12/2024]
Abstract
The incidences of endocrine and metabolic disorders like diabetes have increased worldwide. Several proposed molecular pathways mechanisms for the management of diabetes have been identified, but glycaemic control is still a challenging task in the drug discovery process. Most of the drug discovery processes lead to numerous scaffolds that are prominent in natural products. The review deals with the natural bioactives as an α-amylase inhibitors, α-glucosidase inhibitors, protein tyrosine phosphatase-1B inhibitors, dipeptidyl peptidase-IV inhibitors, G-protein coupled receptors-40 agonists, PPAR-γ agonists and the activators of 5'-adenosine monophosphate-activated protein kinase and glucokinase. So, in this review, we focused on the hypoglycaemic bioactives, which will assist scientific developers, traditional medicinal practitioners, and readers to discover some potent antidiabetic molecules. Strategies like chemometric approaches, scaffold hopping, and total synthesis of natural products by group modification or ring opening/closing mechanism could be useful for the development of novel hit/lead antidiabetic molecules. The study concludes that each phyto molecule inherits a potential to get explored by repurposing techniques for various antidiabetic targets and offer an alternative antidiabetic therapeutic medicinal potential.
Collapse
Affiliation(s)
- Rakesh Kumar Paul
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Kaisar Raza
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
9
|
Ansari MA, Chauhan W, Shoaib S, Alyahya SA, Ali M, Ashraf H, Alomary MN, Al-Suhaimi EA. Emerging therapeutic options in the management of diabetes: recent trends, challenges and future directions. Int J Obes (Lond) 2023; 47:1179-1199. [PMID: 37696926 DOI: 10.1038/s41366-023-01369-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 07/04/2023] [Accepted: 08/17/2023] [Indexed: 09/13/2023]
Abstract
Diabetes is a serious health issue that causes a progressive dysregulation of carbohydrate metabolism due to insufficient insulin hormone, leading to consistently high blood glucose levels. According to the epidemiological data, the prevalence of diabetes has been increasing globally, affecting millions of individuals. It is a long-term condition that increases the risk of various diseases caused by damage to small and large blood vessels. There are two main subtypes of diabetes: type 1 and type 2, with type 2 being the most prevalent. Genetic and molecular studies have identified several genetic variants and metabolic pathways that contribute to the development and progression of diabetes. Current treatments include gene therapy, stem cell therapy, statin therapy, and other drugs. Moreover, recent advancements in therapeutics have also focused on developing novel drugs targeting these pathways, including incretin mimetics, SGLT2 inhibitors, and GLP-1 receptor agonists, which have shown promising results in improving glycemic control and reducing the risk of complications. However, these treatments are often expensive, inaccessible to patients in underdeveloped countries, and can have severe side effects. Peptides, such as glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), are being explored as a potential therapy for diabetes. These peptides are postprandial glucose-dependent pancreatic beta-cell insulin secretagogues and have received much attention as a possible treatment option. Despite these advances, diabetes remains a major health challenge, and further research is needed to develop effective treatments and prevent its complications. This review covers various aspects of diabetes, including epidemiology, genetic and molecular basis, and recent advancements in therapeutics including herbal and synthetic peptides.
Collapse
Affiliation(s)
- Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute for Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam, 31441, Saudi Arabia.
| | - Waseem Chauhan
- Department of Hematology, Duke University, Durham, NC, 27710, USA
| | - Shoaib Shoaib
- Department of Biochemistry, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Sami A Alyahya
- Wellness and Preventive Medicine Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia
| | - Mubashshir Ali
- USF Health Byrd Alzheimer's Center and Neuroscience Institute, Department of Molecular Medicine, Tampa, FL, USA
| | - Hamid Ashraf
- Rajiv Gandhi Center for Diabetes and Endocrinology, Faculty of Medicine, Aligarh Muslim University, Aligarh, Uttar Pradesh, India
| | - Mohammad N Alomary
- Advanced Diagnostic and Therapeutic Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, 11442, Saudi Arabia.
| | - Ebtesam A Al-Suhaimi
- King Abdulaziz & his Companions Foundation for Giftedness & Creativity, Riyadh, Saudi Arabia.
| |
Collapse
|
10
|
Urrutia I, Martínez R, Calvo B, Saso-Jiménez L, González P, Fernández-Rubio E, Martín-Nieto A, Aguayo A, Rica I, Gaztambide S, Castano L. Autoimmune Diabetes From Childhood to Adulthood: The Role of Pancreatic Autoantibodies and HLA-DRB1 Genotype. J Clin Endocrinol Metab 2023; 108:e1341-e1346. [PMID: 37207452 DOI: 10.1210/clinem/dgad277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 05/15/2023] [Accepted: 05/15/2023] [Indexed: 05/21/2023]
Abstract
CONTEXT Autoimmune diabetes can develop at any age, but unlike early-onset diabetes, adult onset is less well documented. We aimed to compare, over a wide age range, the most reliable predictive biomarkers for this pathology: pancreatic-autoantibodies and HLA-DRB1 genotype. METHODS A retrospective study of 802 patients with diabetes (aged 11 months to 66 years) was conducted. Pancreatic autoantibodies at diagnosis: insulin autoantibodies (IAA), glutamate decarboxylase autoantibodies (GADA), islet tyrosine phosphatase 2 autoantibodies (IA2A), and zinc transporter-8 autoantibodies (ZnT8A) and HLA-DRB1 genotype were analyzed. RESULTS Compared with early-onset patients, adults had a lower frequency of multiple autoantibodies, with GADA being the most common. At early onset, IAA was the most frequent in those younger than 6 years and correlated inversely with age; GADA and ZnT8A correlated directly and IA2A remained stable.The absence of HLA-DRB1 risk genotype was associated with higher age at diabetes onset (27.5 years; interquartile range [IQR], 14.3-35.7), whereas the high-risk HLA-DR3/DR4 was significantly more common at lower age (11.9 years; IQR, 7.1-21.6). ZnT8A was associated with DR4/non-DR3 (odds ratio [OR], 1.91; 95% CI, 1.15-3.17), GADA with DR3/non-DR4 (OR, 2.97; 95% CI, 1.55-5.71), and IA2A with DR4/non-DR3 and DR3/DR4 (OR, 3.89; 95% CI, 2.28-6.64, and OR, 3.08; 95% CI, 1.83-5.18, respectively). No association of IAA with HLA-DRB1 was found. CONCLUSION Autoimmunity and HLA-DRB1 genotype are age-dependent biomarkers. Adult-onset autoimmune diabetes is associated with lower genetic risk and lower immune response to pancreatic islet cells compared with early-onset diabetes.
Collapse
Affiliation(s)
- Inés Urrutia
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Rosa Martínez
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Begona Calvo
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Medical Oncology, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Laura Saso-Jiménez
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Pedro González
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Elsa Fernández-Rubio
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Alicia Martín-Nieto
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Anibal Aguayo
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| | - Itxaso Rica
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
- Department of Pediatric Endocrinology, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Sonia Gaztambide
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
- Department of Endocrinology and Nutrition, Hospital Universitario Cruces, 48903 Barakaldo, Spain
| | - Luis Castano
- Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Spain
- CIBERDEM, CIBERER, UPV-EHU, Endo-ERN, 48903 Barakaldo, Spain
| |
Collapse
|
11
|
Hebbar P, Nizam R, John SE, Antony D, Dashti M, Channanath A, Shaltout A, Al-Khandari H, Koistinen HA, Tuomilehto J, Alsmadi O, Thanaraj TA, Al-Mulla F. Linkage analysis using whole exome sequencing data implicates SLC17A1, SLC17A3, TATDN2 and TMEM131L in type 1 diabetes in Kuwaiti families. Sci Rep 2023; 13:14978. [PMID: 37696853 PMCID: PMC10495342 DOI: 10.1038/s41598-023-42255-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/07/2023] [Indexed: 09/13/2023] Open
Abstract
Type 1 diabetes (T1D) is characterized by the progressive destruction of pancreatic β-cells, leading to insulin deficiency and lifelong dependency on exogenous insulin. Higher estimates of heritability rates in monozygotic twins, followed by dizygotic twins and sib-pairs, indicate the role of genetics in the pathogenesis of T1D. The incidence and prevalence of T1D are alarmingly high in Kuwait. Consanguineous marriages account for 50-70% of all marriages in Kuwait, leading to an excessive burden of recessive allele enrichment and clustering of familial disorders. Thus, genetic studies from this Arab region are expected to lead to the identification of novel gene loci for T1D. In this study, we performed linkage analyses to identify the recurrent genetic variants segregating in high-risk Kuwaiti families with T1D. We studied 18 unrelated Kuwaiti native T1D families using whole exome sequencing data from 86 individuals, of whom 37 were diagnosed with T1D. The study identified three potential loci with a LOD score of ≥ 3, spanning across four candidate genes, namely SLC17A1 (rs1165196:pT269I), SLC17A3 (rs942379: p.S370S), TATDN2 (rs394558:p.V256I), and TMEM131L (rs6848033:p.R190R). Upon examination of missense variants from these genes in the familial T1D dataset, we observed a significantly increased enrichment of the genotype homozygous for the minor allele at SLC17A3 rs56027330_p.G279R accounting for 16.2% in affected children from 6 unrelated Kuwaiti T1D families compared to 1000 genomes Phase 3 data (0.9%). Data from the NephQTL database revealed that the rs1165196, rs942379, rs394558, and rs56027330 SNPs exhibited genotype-based differential expression in either glomerular or tubular tissues. Data from the GTEx database revealed rs942379 and rs394558 as QTL variants altering the expression of TRIM38 and IRAK2 respectively. Global genome-wide association studies indicated that SLC17A1 rs1165196 and other variants from SLC17A3 are associated with uric acid concentrations and gout. Further evidence from the T1D Knowledge portal supported the role of shortlisted variants in T1D pathogenesis and urate metabolism. Our study suggests the involvement of SLC17A1, SLC17A3, TATDN2, and TMEM131L genes in familial T1D in Kuwait. An enrichment selection of genotype homozygous for the minor allele is observed at SLC17A3 rs56027330_p.G279R variant in affected members of Kuwaiti T1D families. Future studies may focus on replicating the findings in a larger T1D cohort and delineate the mechanistic details of the impact of these novel candidate genes on the pathophysiology of T1D.
Collapse
Affiliation(s)
- Prashantha Hebbar
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Rasheeba Nizam
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Sumi Elsa John
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Dinu Antony
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Mohammad Dashti
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Arshad Channanath
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait
| | - Azza Shaltout
- Department of Population Health, Dasman Diabetes Institute, Kuwait City, Kuwait
| | - Hessa Al-Khandari
- Department of Population Health, Dasman Diabetes Institute, Kuwait City, Kuwait
- Department of Pediatrics, Farwaniya Hospital, Ministry of Health, Kuwait City, Kuwait
| | - Heikki A Koistinen
- Department of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Minerva Foundation Institute for Medical Research, Helsinki, Finland
| | - Jaakko Tuomilehto
- Department of Public Health and Welfare, Finnish Institute for Health and Welfare, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Diabetes Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | | | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, 15462, Kuwait City, Kuwait.
| |
Collapse
|
12
|
Jensen ASH, Ytting H, Winther-Sørensen M, Burisch J, Bergquist A, Gluud LL, Wewer Albrechtsen NJ. Autoimmune liver diseases and diabetes. Eur J Gastroenterol Hepatol 2023; 35:938-947. [PMID: 37505973 DOI: 10.1097/meg.0000000000002594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/30/2023]
Abstract
Autoimmune liver diseases include autoimmune hepatitis, primary biliary cholangitis, and primary sclerosing cholangitis. They are chronic, heterogenous diseases affecting the liver which is a key metabolic organ that ensures glucose homeostasis. It is well known that patients with other chronic liver diseases such as cirrhosis and nonalcoholic fatty liver disease (NAFLD) display glucose disturbances like insulin resistance and have an increased risk of diabetes. Previous evidence on glucose disturbances in patients with autoimmune liver disease is scarce but does point towards a potentially increased risk of type 1 diabetes and type 2 diabetes. The underlying mechanisms are unknown but may reflect genetic predisposition, concurrent NAFLD and or cirrhosis development, and treatment (steroid) related impairment of glucose homeostasis. Therefore, increased awareness and surveillance of diabetes development in patients with autoimmune liver disease may be important. Overall, detection and treatment of diabetes generally follow the usual diabetes guidelines; however, in patients with advanced liver cirrhosis, HbA1c may not be a reliable marker of average glucose levels, and treatment with insulin is generally recommended. In addition, it has recently been suggested that sodium-glucose cotransporter 2 inhibitors may be beneficial in treating refractory ascites. Further research on diabetes risk in autoimmune liver disease is warranted.
Collapse
Affiliation(s)
- Anne-Sofie H Jensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen
- Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre Hospital, Hvidovre
| | - Henriette Ytting
- Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre Hospital, Hvidovre
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Rigshospitalet
- Institute for Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen
| | - Marie Winther-Sørensen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen
| | - Johan Burisch
- Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre Hospital, Hvidovre
| | - Annika Bergquist
- Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre Hospital, Hvidovre
- Department of Upper GI Diseases, Karolinska University Hospital, Department of Medicine, Karolinska Institutet, Stockholm
| | - Lise Lotte Gluud
- Gastro Unit, Copenhagen University Hospital - Amager and Hvidovre Hospital, Hvidovre
- Institute for Clinical Medicine, Faculty of Health and Medical Sciences, Copenhagen University, Copenhagen
| | - Nicolai J Wewer Albrechtsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen
- Department of Clinical Biochemistry, Copenhagen University Hospital - Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| |
Collapse
|
13
|
Heath KE, Feduska JM, Taylor JP, Houp JA, Botta D, Lund FE, Mick GJ, McGwin G, McCormick KL, Tse HM. GABA and Combined GABA with GAD65-Alum Treatment Alters Th1 Cytokine Responses of PBMCs from Children with Recent-Onset Type 1 Diabetes. Biomedicines 2023; 11:1948. [PMID: 37509587 PMCID: PMC10377053 DOI: 10.3390/biomedicines11071948] [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: 05/19/2023] [Revised: 06/19/2023] [Accepted: 06/29/2023] [Indexed: 07/30/2023] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune disease culminating in the destruction of insulin-producing pancreatic cells. There is a need for the development of novel antigen-specific strategies to delay cell destruction, including combinatorial strategies that do not elicit systemic immunosuppression. Gamma-aminobutyric acid (GABA) is expressed by immune cells, β-cells, and gut bacteria and is immunomodulatory. Glutamic-acid decarboxylase 65 (GAD65), which catalyzes GABA from glutamate, is a T1D autoantigen. To test the efficacy of combinatorial GABA treatment with or without GAD65-immunization to dampen autoimmune responses, we enrolled recent-onset children with T1D in a one-year clinical trial (ClinicalTrials.gov NCT02002130) and examined T cell responses. We isolated peripheral blood mononuclear cells and evaluated cytokine responses following polyclonal activation and GAD65 rechallenge. Both GABA alone and GABA/GAD65-alum treatment inhibited Th1 cytokine responses over the 12-month study with both polyclonal and GAD65 restimulation. We also investigated whether patients with HLA-DR3-DQ2 and HLA-DR4-DQ8, the two highest-risk human leukocyte antigen (HLA) haplotypes in T1D, exhibited differences in response to GABA alone and GABA/GAD65-alum. HLA-DR4-DQ8 patients possessed a Th1-skewed response compared to HLA-DR3-DQ2 patients. We show that GABA and GABA/GAD65-alum present an attractive immunomodulatory treatment for children with T1D and that HLA haplotypes should be considered.
Collapse
Affiliation(s)
- Katie E. Heath
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA (J.M.F.); (J.P.T.); (D.B.); (F.E.L.)
| | - Joseph M. Feduska
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA (J.M.F.); (J.P.T.); (D.B.); (F.E.L.)
| | - Jared P. Taylor
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA (J.M.F.); (J.P.T.); (D.B.); (F.E.L.)
| | - Julie A. Houp
- Department of Surgery, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Davide Botta
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA (J.M.F.); (J.P.T.); (D.B.); (F.E.L.)
| | - Frances E. Lund
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL 35294, USA (J.M.F.); (J.P.T.); (D.B.); (F.E.L.)
| | - Gail J. Mick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (G.J.M.); (K.L.M.)
| | - Gerald McGwin
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Kenneth L. McCormick
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Alabama at Birmingham, Birmingham, AL 35294, USA; (G.J.M.); (K.L.M.)
| | - Hubert M. Tse
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Mail Stop 3029, 1012 Wahl Hall West, 3901 Rainbow Boulevard, Kansas City, KS 66160, USA
| |
Collapse
|
14
|
Ziegler AG. The countdown to type 1 diabetes: when, how and why does the clock start? Diabetologia 2023:10.1007/s00125-023-05927-2. [PMID: 37231274 DOI: 10.1007/s00125-023-05927-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 03/27/2023] [Indexed: 05/27/2023]
Abstract
'The clock to type 1 diabetes has started when islet antibodies are first detected', commented George Eisenbarth with regard to the pathogenesis of type 1 diabetes. This review focuses on 'starting the clock', i.e. the initiation of pre-symptomatic islet autoimmunity/the first appearance of islet autoantibodies. In particular, this review addresses why susceptibility to developing islet autoimmunity is greatest in the first 2 years of life and why beta cells are a frequent target of the immune system during this fertile period. A concept for the development of beta cell autoimmunity in childhood is discussed and three factors are highlighted that contribute to this early predisposition: (1) high beta cell activity and potential vulnerability to stress; (2) high rates of and first exposures to infection; and (3) a heightened immune response, with a propensity for T helper type 1 (Th1) immunity. Arguments are presented that beta cell injury, accompanied by activation of an inflammatory immune response, precedes the initiation of autoimmunity. Finally, the implications for strategies aimed at primary prevention for a world without type 1 diabetes are discussed.
Collapse
Affiliation(s)
- Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Munich, German Center for Environmental Health, Munich, Germany.
- Forschergruppe Diabetes, School of Medicine, Klinikum rechts der Isar, Technical University Munich, Munich, Germany.
- Forschergruppe Diabetes e.V. at Helmholtz Munich, German Research Center for Environmental Health, Munich, Germany.
| |
Collapse
|
15
|
Devi MB, Sarma HK, Mukherjee AK, Khan MR. Mechanistic Insights into Immune-Microbiota Interactions and Preventive Role of Probiotics Against Autoimmune Diabetes Mellitus. Probiotics Antimicrob Proteins 2023:10.1007/s12602-023-10087-1. [PMID: 37171690 DOI: 10.1007/s12602-023-10087-1] [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] [Accepted: 05/04/2023] [Indexed: 05/13/2023]
Abstract
Recent studies on genetically susceptible individuals and animal models revealed the potential role of the intestinal microbiota in the pathogenesis of type 1 diabetes (T1D) through complex interactions with the immune system. T1D incidence has been increasing exponentially with modern lifestyle altering normal microbiota composition, causing dysbiosis characterized by an imbalance in the gut microbial community. Dysbiosis has been suggested to be a potential contributing factor in T1D. Moreover, several studies have shown the potential role of probiotics in regulating T1D through various mechanisms. Current T1D therapies target curative measures; however, preventive therapeutics are yet to be proven. This review highlights immune microbiota interaction and the immense role of probiotics and postbiotics as important immunological interventions for reducing the risk of T1D.
Collapse
Affiliation(s)
- M Bidyarani Devi
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
- Department of Biotechnology, Gauhati University, Guwahati, Assam, India
| | | | - Ashis K Mukherjee
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Mojibur R Khan
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India.
| |
Collapse
|
16
|
Papadimitriou DT, Dermitzaki E, Christopoulos P, Papagianni M, Kleanthous K, Marakaki C, Papadimitriou A, Mastorakos G. Secondary Prevention of Diabetes Type 1 with Oral Calcitriol and Analogs, the PRECAL Study. CHILDREN (BASEL, SWITZERLAND) 2023; 10:862. [PMID: 37238410 PMCID: PMC10217040 DOI: 10.3390/children10050862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 05/04/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023]
Abstract
Screening for Type 1 Diabetes (T1D, incidence 1:300) with T1D autoantibodies (T1Ab) at ages 2 and 6, while sensitive, lacks a preventive strategy. Cholecalciferol 2000 IU daily since birth reduced T1D by 80% at 1 year. T1D-associated T1Ab negativized within 0.6 years with oral calcitriol in 12 children. To further investigate secondary prevention of T1D with calcitriol and its less calcemic analog, paricalcitol, we initiated a prospective interventional non-randomized clinical trial, the PRECAL study (ISRCTN17354692). In total, 50 high-risk children were included: 44 were positive for T1Ab, and 6 had predisposing for T1D HLA genotypes. Nine T1Ab+ patients had variable impaired glucose tolerance (IGT), four had pre-T1D (3 T1Ab+, 1 HLA+), nine had T1Ab+ new-onset T1D not requiring insulin at diagnosis. T1Ab, thyroid/anti-transglutaminase Abs, glucose/calcium metabolism were determined prior and q3-6 months on calcitriol, 0.05 mcg/Kg/day, or paricalcitol 1-4 mcg × 1-3 times/day p.o. while on cholecalciferol repletion. Available data on 42 (7 dropouts, 1 follow-up < 3 months) patients included: all 26 without pre-T1D/T1D followed for 3.06 (0.5-10) years negativized T1Ab (15 +IAA, 3 IA2, 4 ICA, 2 +GAD, 1 +IAA/+GAD, 1 +ICA/+GAD) within 0.57 (0.32-1.3) years or did not develop to T1D (5 +HLA, follow-up 3 (1-4) years). From four pre-T1D cases, one negativized T1Ab (follow-up 1 year), one +HLA did not progress to T1D (follow-up 3.3 years) and two +T1Ab patients developed T1D in 6 months/3 years. Three out of nine T1D cases progressed immediately to overt disease, six underwent complete remission for 1 year (1 month-2 years). Five +T1Ab patients relapsed and negativized again after resuming therapy. Four (aged <3 years) negativized anti-TPO/TG, and two anti-transglutaminase-IgA. Eight presented mild hypercalciuria/hypercalcemia, resolving with dose titration/discontinuation. Secondary prevention of T1D with calcitriol and paricalcitol seems possible and reasonably safe, if started soon enough after seroconversion.
Collapse
Affiliation(s)
- Dimitrios T. Papadimitriou
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Eleni Dermitzaki
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Panagiotis Christopoulos
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Maria Papagianni
- Department of Nutrition and Dietetics, University of Thessaly, 42132 Trikala, Greece
- Unit of Endocrinology, Diabetes and Metabolism, Third Department of Pediatrics, Aristotle University of Thessaloniki, Hippokrateion Hospital of Thessaloniki, 54642 Thessaloniki, Greece
| | - Kleanthis Kleanthous
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Chrysanthi Marakaki
- Department of Pediatric-Adolescent Endocrinology and Diabetes, Athens Medical Center, 15125 Marousi, Greece
| | - Anastasios Papadimitriou
- Pediatric Endocrinology Unit, Attikon University Hospital, National and Kapodistrian University of Athens, 12462 Haidari, Greece
| | - George Mastorakos
- Second Department of Obstetrics and Gynecology, Aretaieion University Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
17
|
Vandewalle J, Desouter AK, Van der Auwera BJ, Tenoutasse S, Gillard P, De Block C, Keymeulen B, Gorus FK, Van de Casteele M. CTLA4, SH2B3, and CLEC16A diversely affect the progression of early islet autoimmunity in relatives of Type 1 diabetes patients. Clin Exp Immunol 2023; 211:224-232. [PMID: 36622793 PMCID: PMC10038324 DOI: 10.1093/cei/uxad002] [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: 10/06/2022] [Revised: 12/12/2022] [Accepted: 01/05/2023] [Indexed: 01/10/2023] Open
Abstract
The HLA region is the major genetic risk determinant of Type 1 diabetes. How non-HLA loci contribute to the genetic risk is incompletely understood, but there are indications that at least some impact progression of asymptomatic autoimmunity. We examined whether SNPs in 7 susceptibility loci (INS, SH2B3, PTPN2, PTPN22, CTLA4, CLEC16A, and IL2RA) could improve prediction of the progression from single to multiple autoantibody positivity, and from there on to diagnosis. SNPs were genotyped in persistently autoantibody positive relatives by allelic discrimination qPCR and disease progression was studied by multivariate Cox regression analysis. In our cohort, only the CTLA4 GA genotype (rs3087243, P = 0.002) and the CLEC16A AA genotype (rs12708716, P = 0.021) were associated with accelerated progression from single to multiple autoantibody positivity, but their effects were restricted to presence of HLA-DQ2/DQ8, and IAA as first autoantibody, respectively. The interaction of CTLA4 and HLA-DQ2/DQ8 overruled the effect of DQ2/DQ8 alone. The HLA-DQ2/DQ8-mediated risk of progression to multiple autoantibodies nearly entirely depended on heterozygosity for CTLA4. The SH2B3 TT genotype (rs3184504) was protective for HLA-DQ8 positive subjects (P = 0.003). At the stage of multiple autoantibodies, only the CTLA4 GA genotype was a minor independent risk factor for progression towards clinical diabetes (P = 0.034). Our study shows that non-HLA polymorphisms impact progression of islet autoimmunity in a subgroup-, stage- and SNP-specific way, suggesting distinct mechanisms. If confirmed, these findings may help refine risk assessment, follow-up, and prevention trials in risk groups.
Collapse
Affiliation(s)
- Julie Vandewalle
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Aster K Desouter
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetology and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Bart J Van der Auwera
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Sylvie Tenoutasse
- Department of Diabetology, Hôpital Universitaire des Enfants Reine Fabiola, HUDERF, Université Libre De Bruxelles, Brussels, Belgium
| | - Pieter Gillard
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetology and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
- Department of Endocrinology, University Hospitals Leuven, Leuven, Belgium
| | - Christophe De Block
- Department of Endocrinology, Diabetology and Metabolism, Universitair Ziekenhuis Antwerpen, Edegem, Belgium
| | - Bart Keymeulen
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetology and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Frans K Gorus
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
- Department of Diabetology and Endocrinology, Universitair Ziekenhuis Brussel (UZ Brussel), Brussels, Belgium
| | - Mark Van de Casteele
- Department of Diabetes Pathology and Therapy, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | | |
Collapse
|
18
|
Ballou C, Barton F, Payne EH, Berney T, Villard J, Meier RPH, Baidal D, Alejandro R, Robien M, Eggerman TL, Kamoun M, Muller YD. Matching for HLA-DR excluding diabetogenic HLA-DR3 and HLA-DR4 predicts insulin independence after pancreatic islet transplantation. Front Immunol 2023; 14:1110544. [PMID: 37026004 PMCID: PMC10070978 DOI: 10.3389/fimmu.2023.1110544] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/16/2023] [Indexed: 04/08/2023] Open
Abstract
Introduction In pancreatic islet transplantation, the exact contribution of human leukocyte antigen (HLA) matching to graft survival remains unclear. Islets may be exposed to allogenic rejection but also the recurrence of type 1 diabetes (T1D). We evaluated the HLA-DR matching, including the impact of diabetogenic HLA-DR3 or HLA-DR4 matches. Methods We retrospectively examined the HLA profile in 965 transplant recipients and 2327 islet donors. The study population was obtained from patients enrolled in the Collaborative Islet Transplant Registry. We then identified 87 recipients who received a single-islet infusion. Islet-kidney recipients, 2nd islet infusion, and patients with missing data were excluded from the analysis (n=878). Results HLA-DR3 and HLA-DR4 were present in 29.7% and 32.6% of T1D recipients and 11.6% and 15.8% of the donors, respectively. We identified 52 T1D islet recipients mismatched for HLA-DR (group A), 11 with 1 or 2 HLA-DR-matches but excluding HLA-DR3 and HLA- DR4 (group B), and 24 matched for HLA-DR3 or HLA-DR4 (group C). Insulin-independence was maintained in a significantly higher percentage of group B recipients from year one through five post-transplantation (p<0.01). At five-year post-transplantation, 78% of group B was insulin-independent compared to 24% (group A) and 35% (group C). Insulin-independence correlated with significantly better glycemic control (HbA1c <7%), fasting blood glucose, and reduced severe hypoglycemic events. Matching HLA-A-B-DR (≥3) independently of HLA- DR3 or HLA-DR4 matching did not improve graft survival. Conclusion This study suggests that matching HLA-DR but excluding the diabetogenic HLA-DR3 and/or 4 is a significant predictor for long-term islet survival.
Collapse
Affiliation(s)
- Cassandra Ballou
- Collaborative Islet Transplant Registry Coordinating Center, The EMMES Company, LLC, Rockville, MD, United States
- *Correspondence: Yannick D. Muller, ; Cassandra Ballou,
| | - Franca Barton
- Collaborative Islet Transplant Registry Coordinating Center, The EMMES Company, LLC, Rockville, MD, United States
| | - Elizabeth H. Payne
- Collaborative Islet Transplant Registry Coordinating Center, The EMMES Company, LLC, Rockville, MD, United States
| | - Thierry Berney
- Division of Transplantation, Department of Surgery, Geneva University Hospitals, Geneva, Switzerland
| | - Jean Villard
- Department of Genetic, Laboratory and Pathology Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Raphael P. H. Meier
- Department of Surgery, School of Medicine, University of Maryland, Baltimore, MD, United States
| | - David Baidal
- Department of Medicine and the Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Rodolfo Alejandro
- Department of Medicine and the Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Mark Robien
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Thomas L. Eggerman
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Malek Kamoun
- Immunology and Histocompatibility Testing Laboratory, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
| | - Yannick D. Muller
- Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- *Correspondence: Yannick D. Muller, ; Cassandra Ballou,
| |
Collapse
|
19
|
Harsini S, Rezaei N. Autoimmune diseases. Clin Immunol 2023. [DOI: 10.1016/b978-0-12-818006-8.00001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
20
|
Pokrovskaya EV, Zhgun ES, Shestakova EA, Sklyanik IA, Fedushkina IV, Olekhnovich EI, Konanov DN, Kardonsky DA, Kislun YV, Sorokina EA, Zilberman LI, Zaytseva NV, Ilina EN, Govorun VM, Shestakova MV. Feсal microbiota transplantation in the format of complex therapy in obesive siblings: clinical case. DIABETES MELLITUS 2022. [DOI: 10.14341/dm12893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Obesity and associated metabolic diseases are often accompanied by changes in the gut microbiota leading to metagenome gene diversity decrease. Fecal microbiota transplantation (FMT) is one of the most effective methods for correcting the intestinal microflora. FMT obtained from healthy donors has been proven to be an effective treatment of infections caused by Clostridium difficile. The use of FMT for correction of metabolic disorders is promising, however, data on its application is limited and has contradictory results. In our work, two patients (siblings) presented with obesity grade II and various types of diabetes mellitus (DM): the older brother (44 years old) with diabetes mellitus type 2 (DM 2), a younger brother (39 years old) with diabetes mellitus type 1 (DM 1). Both patients underwent FMT as part of complex antidiabetic therapy. During the course of treatment, a decrease in body weight was noted in both patients (4–5 kg for the first month of observation, then -1–2 kg per month). One year after FMT, a patient with type 2 diabetes showed a decrease in the severity of insulin resistance (IR), measured by the hyperinsulinemic euglycemic clamp test (initial M-index 2.42 mg/kg*min, after 1 year — 3.83 mg/kg* min) as well as the maintenance of satisfactory carbohydrate metabolism compensation against the diminishing the hypoglycemic therapy. In a patient with DM 1, no significant dynamics of carbohydrate exchange indices, including detected glycated hemoglobin (HbA1c), insulin dose and IR were during the observation period. Metagenomic sequencing of stool samples (n = 20) collected from both patients before and within 1 year after FMT showed no significant changes in the taxonomic profile of the microbiota at the level of microbial families. Metabolomic analysis of the composition of feces showed no directed changes in the composition of metabolites after the FMT procedure, the nature of changes within the samples from each patient during the entire study period was random. Thus, FMT had no effect on the course of DM1, but served as a starting point for weight loss and improvement glucose profile in DM2. However, convincing data confirming a causal correlation between FMT and improvement in the course of T2DM have not been obtained.
Collapse
Affiliation(s)
| | - E. S. Zhgun
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | | | | | - I. V. Fedushkina
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | | | - D. N. Konanov
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | - D. A. Kardonsky
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | - Yu. V. Kislun
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | - E. A. Sorokina
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | | | | | - E. N. Ilina
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | - V. M. Govorun
- Federal Research and Clinical Center of Physical-Chemical Medicine
| | | |
Collapse
|
21
|
Dwyer JR, Racine JJ, Chapman HD, Quinlan A, Presa M, Stafford GA, Schmitz I, Serreze DV. Nfkbid Overexpression in Nonobese Diabetic Mice Elicits Complete Type 1 Diabetes Resistance in Part Associated with Enhanced Thymic Deletion of Pathogenic CD8 T Cells and Increased Numbers and Activity of Regulatory T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:227-237. [PMID: 35760520 PMCID: PMC9365269 DOI: 10.4049/jimmunol.2100558] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 05/13/2022] [Indexed: 06/15/2023]
Abstract
Type 1 diabetes (T1D) in both humans and NOD mice is caused by T cell-mediated autoimmune destruction of pancreatic β cells. Increased frequency or activity of autoreactive T cells and failures of regulatory T cells (Tregs) to control these pathogenic effectors have both been implicated in T1D etiology. Due to the expression of MHC class I molecules on β cells, CD8 T cells represent the ultimate effector population mediating T1D. Developing autoreactive CD8 T cells normally undergo extensive thymic negative selection, but this process is impaired in NOD mice and also likely T1D patients. Previous studies identified an allelic variant of Nfkbid, a NF-κB signal modulator, as a gene strongly contributing to defective thymic deletion of autoreactive CD8 T cells in NOD mice. These previous studies found ablation of Nfkbid in NOD mice using the clustered regularly interspaced short palindromic repeats system resulted in greater thymic deletion of pathogenic CD8 AI4 and NY8.3 TCR transgenic T cells but an unexpected acceleration of T1D onset. This acceleration was associated with reductions in the frequency of peripheral Tregs. In this article, we report transgenic overexpression of Nfkbid in NOD mice also paradoxically results in enhanced thymic deletion of autoreactive CD8 AI4 T cells. However, transgenic elevation of Nfkbid expression also increased the frequency and functional capacity of peripheral Tregs, in part contributing to the induction of complete T1D resistance. Thus, future identification of a pharmaceutical means to enhance Nfkbid expression might ultimately provide an effective T1D intervention approach.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Ingo Schmitz
- Department of Molecular Immunology, Ruhr-University, Bochum, Germany
| | | |
Collapse
|
22
|
Colclough K, van Heugten R, Patel K. An update on the diagnosis and management of monogenic diabetes. PRACTICAL DIABETES 2022. [DOI: 10.1002/pdi.2410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Kevin Colclough
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust Exeter UK
| | - Rachel van Heugten
- Exeter Genomics Laboratory, Royal Devon & Exeter NHS Foundation Trust Exeter UK
| | - Kashyap Patel
- Institute of Biomedical and Clinical Science University of Exeter Medical School Exeter UK
| |
Collapse
|
23
|
Oelen R, de Vries DH, Brugge H, Gordon MG, Vochteloo M, Ye CJ, Westra HJ, Franke L, van der Wijst MGP. Single-cell RNA-sequencing of peripheral blood mononuclear cells reveals widespread, context-specific gene expression regulation upon pathogenic exposure. Nat Commun 2022; 13:3267. [PMID: 35672358 PMCID: PMC9174272 DOI: 10.1038/s41467-022-30893-5] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 05/24/2022] [Indexed: 12/15/2022] Open
Abstract
The host's gene expression and gene regulatory response to pathogen exposure can be influenced by a combination of the host's genetic background, the type of and exposure time to pathogens. Here we provide a detailed dissection of this using single-cell RNA-sequencing of 1.3M peripheral blood mononuclear cells from 120 individuals, longitudinally exposed to three different pathogens. These analyses indicate that cell-type-specificity is a more prominent factor than pathogen-specificity regarding contexts that affect how genetics influences gene expression (i.e., eQTL) and co-expression (i.e., co-expression QTL). In monocytes, the strongest responder to pathogen stimulations, 71.4% of the genetic variants whose effect on gene expression is influenced by pathogen exposure (i.e., response QTL) also affect the co-expression between genes. This indicates widespread, context-specific changes in gene expression level and its regulation that are driven by genetics. Pathway analysis on the CLEC12A gene that exemplifies cell-type-, exposure-time- and genetic-background-dependent co-expression interactions, shows enrichment of the interferon (IFN) pathway specifically at 3-h post-exposure in monocytes. Similar genetic background-dependent association between IFN activity and CLEC12A co-expression patterns is confirmed in systemic lupus erythematosus by in silico analysis, which implies that CLEC12A might be an IFN-regulated gene. Altogether, this study highlights the importance of context for gaining a better understanding of the mechanisms of gene regulation in health and disease.
Collapse
Affiliation(s)
- Roy Oelen
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Dylan H de Vries
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Harm Brugge
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - M Grace Gordon
- Biological and Medical Informatics Graduate Program, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA, USA
- UCSF Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Martijn Vochteloo
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Chun J Ye
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- UCSF Division of Rheumatology, Department of Medicine, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Harm-Jan Westra
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Oncode Institute, Utrecht, The Netherlands
| | - Lude Franke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| | - Monique G P van der Wijst
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.
- Oncode Institute, Utrecht, The Netherlands.
| |
Collapse
|
24
|
Çiçekli İ, Durusoy R. Breastfeeding, nutrition and type 1 diabetes: a case-control study in Izmir, Turkey. Int Breastfeed J 2022; 17:42. [PMID: 35624486 PMCID: PMC9145075 DOI: 10.1186/s13006-022-00470-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 03/27/2022] [Indexed: 01/11/2023] Open
Abstract
Background The relationship between infant breastfeeding and type 1 diabetes mellitus (DM) is unclear but it has been suggested that there may be a link between many environmental factors, including dietary antigens affecting diabetes epidemiology. The main objective of this study is to investigate nutritional risk factors, especially breastfeeding early in life that may be associated with the development of type 1 DM and to determine the relationship these factors have with the disease. Methods This research is a case-control study and was carried out in Ege University Children’s Hospital in İzmir, Turkey between 13 January 2020 and 5 March 2020. A total of 246 children aged between 4 and 14 years were included in the study. The case group consisted of patients diagnosed with type 1 DM followed-up by Ege University Children’s Hospital’s Endocrinology Unit and the control group included non-diabetic children attending the same hospital’s General Pediatric Outpatient Clinic. A structured questionnaire was created by the researchers after reviewing the literature related to nutritional and other risk factors for type 1 DM. The questionnaire was administered by interviewing the parents and it was related to the child, mother and family of the child. In this study, breastfeeding duration was defined as the total duration of breastfeeding and exclusive breastfeeding meant that the child received only breast milk from the mother. Results The mean age at diagnosis was 6.30 ± 4.03 years for cases and 7.48 ± 2.56 years for controls. We found that each monthly increase in exclusive breastfeeding duration provided a 0.83-fold (95% CI 0.72, 0.96) decrease in the risk of type 1 DM. Introduction of cereals in the diet at the sixth month or earlier was associated with a 2.58-fold (95% CI 1.29, 5.16) increased risk. Conclusions Determining the contribution of exclusive breastfeeding to the disease is important in establishing preventive policies. A longer duration of exclusive breastfeeding may be an important role in preventing the disease. This free intervention that truly works will be cost-effective. Future studies are needed to clarify the role of both exclusive and non-exclusive breastfeeding on the development of type 1 DM. Supplementary Information The online version contains supplementary material available at 10.1186/s13006-022-00470-z.
Collapse
Affiliation(s)
- İpek Çiçekli
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acibadem University, İstanbul, Turkey.
| | - Raika Durusoy
- Department of Public Health, Faculty of Medicine, Ege University, İzmir, Turkey
| |
Collapse
|
25
|
Al Yafei Z, Mack SJ, Alvares M, Ali BR, Afandi B, Beshyah SA, Sharma C, Osman W, Mirghani R, Nasr A, Al Remithi S, Al Jubeh J, Almawi WY, AlKaabi J, ElGhazali G. HLA-DRB1 and -DQB1 Alleles, Haplotypes and Genotypes in Emirati Patients with Type 1 Diabetes Underscores the Benefits of Evaluating Understudied Populations. Front Genet 2022; 13:841879. [PMID: 35419034 PMCID: PMC8997289 DOI: 10.3389/fgene.2022.841879] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/31/2022] [Indexed: 02/05/2023] Open
Abstract
Background: HLA class II (DR and DQ) alleles and antigens have historically shown strong genetic predisposition to type 1 diabetes (T1D). This study evaluated the association of DRB1 and DQB1 alleles, genotypes, and haplotypes with T1D in United Arab Emirates. Materials and Methods: Study subjects comprised 149 patients with T1D, and 147 normoglycemic control subjects. Cases and controls were Emiratis and were HLA-DRB1 and -DQB1 genotyped using sequence-based typing. Statistical analysis was performed using Bridging Immunogenomic Data-Analysis Workflow Gaps R package. Results: In total, 15 DRB1 and 9 DQB1 alleles were identified in the study subjects, of which the association of DRB1*03:01, DRB1*04:02, DRB1*11:01, DRB1*16:02, and DQB1*02:01, DQB1*03:02, DQB1*03:01, and DQB1*06:01 with altered risk of T1D persisted after correcting for multiple comparisons. Two-locus haplotype analysis identified DRB1*03:01∼DQB1*02:01 [0.44 vs. 0.18, OR (95% CI) = 3.44 (2.33-5.1), Pc = 3.48 × 10-10]; DRB1*04:02∼DQB1*03:02 [0.077 vs. 0.014, OR = 6.06 (2.03-24.37), Pc = 2.3 × 10-3] and DRB1*04:05∼DQB1*03:02 [0.060 vs. 0.010, OR = 6.24 (1.79-33.34), Pc = 0.011] as positively associated, and DRB1*16:02∼DQB1*05:02 [0.024 vs. 0.075, OR = 0.3 (0.11-0.74), Pc = 0.041] as negatively associated with T1D, after applying Bonferroni correction. Furthermore, the highest T1D risk was observed for DR3/DR4 [0.104 vs. 0.006, OR = 25.03 (8.23-97.2), Pc = 2.6 × 10-10], followed by DR3/DR3 [0.094 vs. 0.010, OR = 8.72 (3.17-25.32), Pc = 3.18 × 10-8] diplotypes. Conclusion: While DRB1 and DQB1 alleles and haplotypes associated with T1D in Emiratis showed similarities to Caucasian and non-Caucasian populations, several alleles and haplotypes associated with T1D in European, African, and Asian populations, were not observed. This underscores the contribution of ethnic diversity and possible diverse associations between DRB1 and DQB1 and T1D across different populations.
Collapse
Affiliation(s)
- Zain Al Yafei
- Sheikh Khalifa Medical City, Purehealth, Abu Dhabi, United Arab Emirates
| | - Steven J. Mack
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Marion Alvares
- Sheikh Khalifa Medical City, Purehealth, Abu Dhabi, United Arab Emirates
| | - Bassam R. Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Bachar Afandi
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
| | - Salem A. Beshyah
- Department of Medicine, Dubai Medical College, Dubai, United Arab Emirates
| | - Charu Sharma
- Department of Internal Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Wael Osman
- College of Arts and Sciences, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Rajaa Mirghani
- Higher College of Technology, Abu Dhabi, United Arab Emirates
| | - Amre Nasr
- Department of Basic Medical Sciences, College of Medicine, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Sareea Al Remithi
- Sheikh Khalifa Medical City, Purehealth, Abu Dhabi, United Arab Emirates
| | - Jamal Al Jubeh
- Sheikh Khalifa Medical City, Purehealth, Abu Dhabi, United Arab Emirates
| | - Wasim Y. Almawi
- Department of Biomedical Sciences, Nazarbayev University School of Medicine, Astana, Kazakhstan
| | - Juma AlKaabi
- Department of Internal Medicine, Tawam Hospital, Al Ain, United Arab Emirates
- Department of Internal Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Gehad ElGhazali
- Sheikh Khalifa Medical City, Purehealth, Abu Dhabi, United Arab Emirates
- Department of Internal Medicine, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
26
|
Thomas S, Ouhtit A, Al Khatib HA, Eid AH, Mathew S, Nasrallah GK, Emara MM, Al Maslamani MA, Yassine HM. Burden and Disease Pathogenesis of Influenza and Other Respiratory Viruses in Diabetic Patients. J Infect Public Health 2022; 15:412-424. [DOI: 10.1016/j.jiph.2022.03.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/24/2022] [Accepted: 03/07/2022] [Indexed: 02/07/2023] Open
|
27
|
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
|
28
|
Brodnicki TC. A Role for lncRNAs in Regulating Inflammatory and Autoimmune Responses Underlying Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:97-118. [DOI: 10.1007/978-3-030-92034-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
29
|
Abou El Ella SS, Tawfik MA, Mohammed ZS, Barseem NF. PTPN22 gene and IL2RA rs11594656, rs2104286 gene variants: additional insights of polygenic single-nucleotide polymorphisms’ pattern among Egyptian children with type 1 diabetes. EGYPTIAN PEDIATRIC ASSOCIATION GAZETTE 2021. [DOI: 10.1186/s43054-021-00079-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Type 1 diabetes mellitus (T1D) results from environmental and genetic factors.
We aimed to investigate the distribution of PTPN22, IL2RA rs11594656, and rs2104286 variants and its association with T1D in children.
A case-control study conducted on 100 diabetic patients and 100 control children. PTPN22 gene, IL2RA rs11594656, and rs2104286 polymorphisms study were done by PCR followed by restriction fragment length polymorphism (RFLP) assay.
Results
T allele of PTPN22 gene was presented more frequently 47% in patient group versus 30% in controls, while C allele was 53% in the diabetic group versus 70% in controls showing a statistically significant difference between patient and control groups. Similarly, TT 1858 genotype was found in higher frequency with a statistically significant difference in favor of T1D patients (p = 0.038), OR (CI 95% 3.16 (1.28–7.09).
For IL2RA rs11594656 polymorphism, the frequency of TT, TA, and AA in patients at percentages of 20%, 60%, and 20% versus 4%, 60%, and 36% in controls respectively showed significant difference (p = 0.045). Also, T allele was detected more in patients group as evidenced by p = 0.059, OR (95% CI) of 2.38(1.49–6.12). Whereas, IL2RA rs2104286 polymorphism revealed a difference of otherwise non-statistical significance (p = 0.091). Those who harbored homozygous pattern of both IL2RA polymorphisms frequently had DKA and high mean HbA1C values.
Conclusion
PTPN22 (C1858T) and IL2RA rs11594656 polymorphisms increased the risk of T1DM development, while IL2RA rs2104286 polymorphism did not display any significant association among children with T1D. Having more than one risk allele could affect progression and control of T1D.
Collapse
|
30
|
Frørup C, Mirza AH, Yarani R, Nielsen LB, Mathiesen ER, Damm P, Svare J, Engelbrekt C, Størling J, Johannesen J, Mortensen HB, Pociot F, Kaur S. Plasma Exosome-Enriched Extracellular Vesicles From Lactating Mothers With Type 1 Diabetes Contain Aberrant Levels of miRNAs During the Postpartum Period. Front Immunol 2021; 12:744509. [PMID: 34691048 PMCID: PMC8531745 DOI: 10.3389/fimmu.2021.744509] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/10/2021] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes is an immune-driven disease, where the insulin-producing beta cells from the pancreatic islets of Langerhans becomes target of immune-mediated destruction. Several studies have highlighted the implication of circulating and exosomal microRNAs (miRNAs) in type 1 diabetes, underlining its biomarker value and novel therapeutic potential. Recently, we discovered that exosome-enriched extracellular vesicles carry altered levels of both known and novel miRNAs in breast milk from lactating mothers with type 1 diabetes. In this study, we aimed to characterize exosomal miRNAs in the circulation of lactating mothers with and without type 1 diabetes, hypothesizing that differences in type 1 diabetes risk in offspring from these groups are reflected in the circulating miRNA profile. We performed small RNA sequencing on exosome-enriched extracellular vesicles extracted from plasma of 52 lactating mothers around 5 weeks postpartum (26 with type 1 diabetes and 26 age-matched controls), and found a total of 2,289 miRNAs in vesicles from type 1 diabetes and control libraries. Of these, 176 were differentially expressed in plasma from mothers with type 1 diabetes (167 upregulated; 9 downregulated, using a cut-off of abs(log2FC) >1 and FDR adjusted p-value <0.05). Extracellular vesicles were verified by nanoparticle tracking analysis, transmission electron microscopy and immunoblotting. Five candidate miRNAs were selected based on their involvement in diabetes and immune modulation/beta-cell functions: hsa-miR-127-3p, hsa-miR-146a-5p, hsa-miR-26a-5p, hsa-miR-24-3p and hsa-miR-30d-5p. Real-time qPCR validation confirmed that hsa-miR-146a-5p, hsa-miR-26a-5p, hsa-miR-24-3p, and hsa-miR-30d-5p were significantly upregulated in lactating mothers with type 1 diabetes as compared to lactating healthy mothers. To determine possible target genes and affected pathways of the 5 miRNA candidates, computational network-based analyses were carried out with TargetScan, mirTarBase, QIAGEN Ingenuity Pathway Analysis and PantherDB database. The candidates showed significant association with inflammatory response and cytokine and chemokine mediated signaling pathways. With this study, we detect aberrant levels of miRNAs within plasma extracellular vesicles from lactating mothers with type 1 diabetes during the postpartum period, including miRNAs with associations to disease pathogenesis and inflammatory responses.
Collapse
Affiliation(s)
- Caroline Frørup
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Aashiq H Mirza
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Pharmacology, Weill Cornell Medical, New York, NY, United States
| | - Reza Yarani
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Lotte B Nielsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Elisabeth R Mathiesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
| | - Peter Damm
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Center for Pregnant Women with Diabetes, Rigshospitalet, Copenhagen, Denmark.,Department of Obstetrics, Rigshospitalet, Copenhagen, Denmark
| | - Jens Svare
- Department of Obstetrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | | | - Joachim Størling
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Johannesen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Henrik B Mortensen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Pediatrics, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Simranjeet Kaur
- Translational Type 1 Diabetes Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| |
Collapse
|
31
|
Sisk LJ, Patel RK, Stevens KK. A descriptive analysis of non-human leukocyte antigens present in renal transplant donor-recipient pairs. Transpl Immunol 2021; 69:101474. [PMID: 34582968 DOI: 10.1016/j.trim.2021.101474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022]
Abstract
INTRODUCTION End stage renal disease (ESRD) is the irreversible deterioration of renal function requiring renal replacement therapy by dialysis or transplant. Human leucocyte antigens (HLA) have been well examined however research still is required into the non-HLA antibodies. Antibody mediated rejection (AMR) can be seen in the absence of HLA antibodies on biopsies of patients who have received identical transplants; anti-endothelial cell antibodies may explain this. Investigation into endothelial cell antigens on donor and recipient endothelium may elucidate and stratify the degree of risk of any given transplant and may guide towards the best matched donor. METHODS Protein array analysis was carried out on 8 patient pairs using nitro-cellulose membranes and biotinylated detection antibodies. The fluorescence emitted was captured by X-Ray film and results were recorded with ImageJ software. A fold increase of more than 2 was considered to be positive. RESULTS 11 proteins identified had a fold increase of increase ≥2 and were present in ≥2 patient pairs which may point to potential clinical utility. Nectin2/CD112 may be measured in order analyse graft survival time in transplant recipients. Prognosticating renal failure has clinical importance and potential markers that have been identified to aid which include MEPE, CRELD2, and TIMP-4. Novel pharmacological therapies for specific biomarkers identified in this study include JAM-A, E-Selectin, CD147, Galectin-3, JAM-C, PAR-1, and TNFR2. CONCLUSION Protein analysis showed differences in expression of antigens between patients with and without Chronic Kidney Disease (CKD). This information could be used at the matching stage of renal transplantation and also in the treatment of rejection episodes. The results highlight biomarkers that potentially prognosticate and pharmacological therapies that may ameliorate kidney disease and rejection in ESRD and transplant recipients.
Collapse
Affiliation(s)
- Louis J Sisk
- University of Glasgow, United Kingdom; British Heart Foundation Cardiovascular Research Centre, University of Glasgow, United Kingdom.
| | - Rajan K Patel
- University of Glasgow, United Kingdom; British Heart Foundation Cardiovascular Research Centre, University of Glasgow, United Kingdom
| | - Kathryn K Stevens
- University of Glasgow, United Kingdom; British Heart Foundation Cardiovascular Research Centre, University of Glasgow, United Kingdom
| |
Collapse
|
32
|
Clinical features, epidemiology, autoantibody status, HLA haplotypes and genetic mechanisms of type 1 diabetes mellitus among children in Qatar. Sci Rep 2021; 11:18887. [PMID: 34556755 PMCID: PMC8460652 DOI: 10.1038/s41598-021-98460-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 09/03/2021] [Indexed: 11/08/2022] Open
Abstract
To describe the clinical features, epidemiology, autoantibody status, HLA haplotypes and genetic mechanisms of type 1 diabetes mellitus (T1DM). Patients (0-18 years) with diabetes were recruited. Clinical data was collected, autoantibodies and c-peptide were measured. Whole Genome Sequencing was performed. Genomic data analysis was compared with the known genes linked with T1DM and HLA alleles were studied. 1096 patients had one or more antibody positivity. The incidence of T1DM in 2020 was 38.05 per 100,000 children and prevalence was 249.73. GADA was the most common autoantibody followed by IAA. Variants in GSTCD, SKAP2, SLC9B1, BANK1 were most prevalent. An association of HLA haplotypes DQA1*03:01:01G (OR = 2.46, p value = 0.011) and DQB1*03:02:01G (OR = 2.43, p value = 0.022) was identified. The incidence of T1DM in Qatar is the fourth highest in the world, IA2 autoantibody was the most specific with some patients only having ZnT8 or IA2 autoantibodies thus underlining the necessity of profiling all 4 autoantibodies. The genes associated with T1DM in the Arab population were different from those that are common in the Caucasian population. HLA-DQ was enriched in the Qatari patients suggesting that it can be considered a major risk factor at an early age.
Collapse
|
33
|
Bauer W, Gyenesei A, Krętowski A. The Multifactorial Progression from the Islet Autoimmunity to Type 1 Diabetes in Children. Int J Mol Sci 2021; 22:7493. [PMID: 34299114 PMCID: PMC8305179 DOI: 10.3390/ijms22147493] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/04/2021] [Accepted: 07/09/2021] [Indexed: 12/12/2022] Open
Abstract
Type 1 Diabetes (T1D) results from autoimmune destruction of insulin producing pancreatic ß-cells. This disease, with a peak incidence in childhood, causes the lifelong need for insulin injections and necessitates careful monitoring of blood glucose levels. However, despite the current insulin therapies, it still shortens life expectancy due to complications affecting multiple organs. Recently, the incidence of T1D in childhood has increased by 3-5% per year in most developed Western countries. The heterogeneity of the disease process is supported by the findings of follow-up studies started early in infancy. The development of T1D is usually preceded by the appearance of autoantibodies targeted against antigens expressed in the pancreatic islets. The risk of T1D increases significantly with an increasing number of positive autoantibodies. The order of autoantibody appearance affects the disease risk. Genetic susceptibility, mainly defined by the human leukocyte antigen (HLA) class II gene region and environmental factors, is important in the development of islet autoimmunity and T1D. Environmental factors, mainly those linked to the changes in the gut microbiome as well as several pathogens, especially viruses, and diet are key modulators of T1D. The aim of this paper is to expand the understanding of the aetiology and pathogenesis of T1D in childhood by detailed description and comparison of factors affecting the progression from the islet autoimmunity to T1D in children.
Collapse
Affiliation(s)
- Witold Bauer
- Clinical Research Centre, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (A.G.); (A.K.)
| | - Attila Gyenesei
- Clinical Research Centre, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (A.G.); (A.K.)
- Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, 7624 Pécs, Hungary
| | - Adam Krętowski
- Clinical Research Centre, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland; (A.G.); (A.K.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Białystok, Marii Skłodowskiej-Curie 24a, 15-276 Białystok, Poland
| |
Collapse
|
34
|
The dark side of insulin: A primary autoantigen and instrument of self-destruction in type 1 diabetes. Mol Metab 2021; 52:101288. [PMID: 34242821 PMCID: PMC8513143 DOI: 10.1016/j.molmet.2021.101288] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/27/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Background Since its discovery 100 years ago, insulin, as the ‘cure’ for type 1 diabetes, has rescued the lives of countless individuals. As the century unfolded and the autoimmune nature of type 1 diabetes was recognised, a darker side of insulin emerged. Autoimmunity to insulin was found to be an early marker of risk for type 1 diabetes in young children. In humans, it remains unclear if autoimmunity to insulin is primarily due to a defect in the beta cell itself or to dysregulated immune activation. Conversely, it may be secondary to beta-cell damage from an environmental agent (e.g., virus). Nevertheless, direct, interventional studies in non-obese diabetic (NOD) mouse models of type 1 diabetes point to a critical role for (pro)insulin as a primary autoantigen that drives beta cell pathology. Scope of review Modelled on Koch's postulates for the pathogenicity of an infectious agent, evidence for a pathogenic role of (pro)insulin as an autoantigen in type 1 diabetes, particularly applicable to the NOD mouse model, is reviewed. Evidence in humans remains circumstantial. Additionally, as (pro)insulin is a target of autoimmunity in type 1 diabetes, its application as a therapeutic tool to elicit antigen-specific immune tolerance is assessed. Major conclusions Paradoxically, insulin is both a ‘cure’ and a potential ‘cause’ of type 1 diabetes, actively participating as an autoantigen to drive autoimmune destruction of beta cells - the instrument of its own destruction.
Collapse
|
35
|
Torun A, Hupalowska A, Trzonkowski P, Kierkus J, Pyrzynska B. Intestinal Microbiota in Common Chronic Inflammatory Disorders Affecting Children. Front Immunol 2021; 12:642166. [PMID: 34163468 PMCID: PMC8215716 DOI: 10.3389/fimmu.2021.642166] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/24/2021] [Indexed: 12/12/2022] Open
Abstract
The incidence and prevalence rate of chronic inflammatory disorders is on the rise in the pediatric population. Recent research indicates the crucial role of interactions between the altered intestinal microbiome and the immune system in the pathogenesis of several chronic inflammatory disorders in children, such as inflammatory bowel disease (IBD) and autoimmune diseases, such as type 1 diabetes mellitus (T1DM) and celiac disease (CeD). Here, we review recent knowledge concerning the pathogenic mechanisms underlying these disorders, and summarize the facts suggesting that the initiation and progression of IBD, T1DM, and CeD can be partially attributed to disturbances in the patterns of composition and abundance of the gut microbiota. The standard available therapies for chronic inflammatory disorders in children largely aim to treat symptoms. Although constant efforts are being made to maximize the quality of life for children in the long-term, sustained improvements are still difficult to achieve. Additional challenges are the changing physiology associated with growth and development of children, a population that is particularly susceptible to medication-related adverse effects. In this review, we explore new promising therapeutic approaches aimed at modulation of either gut microbiota or the activity of the immune system to induce a long-lasting remission of chronic inflammatory disorders. Recent preclinical studies and clinical trials have evaluated new approaches, for instance the adoptive transfer of immune cells, with genetically engineered regulatory T cells expressing antigen-specific chimeric antigen receptors. These approaches have revolutionized cancer treatments and have the potential for the protection of high-risk children from developing autoimmune diseases and effective management of inflammatory disorders. The review also focuses on the findings of studies that indicate that the responses to a variety of immunotherapies can be enhanced by strategic manipulation of gut microbiota, thus emphasizing on the importance of proper interaction between the gut microbiota and immune system for sustained health benefits and improvement of the quality of life of pediatric patients.
Collapse
Affiliation(s)
- Anna Torun
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| | - Anna Hupalowska
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdansk, Gdansk, Poland
| | - Jaroslaw Kierkus
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Beata Pyrzynska
- Chair and Department of Biochemistry, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
36
|
George MN, Leavens KF, Gadue P. Genome Editing Human Pluripotent Stem Cells to Model β-Cell Disease and Unmask Novel Genetic Modifiers. Front Endocrinol (Lausanne) 2021; 12:682625. [PMID: 34149620 PMCID: PMC8206553 DOI: 10.3389/fendo.2021.682625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 05/13/2021] [Indexed: 01/21/2023] Open
Abstract
A mechanistic understanding of the genetic basis of complex diseases such as diabetes mellitus remain elusive due in large part to the activity of genetic disease modifiers that impact the penetrance and/or presentation of disease phenotypes. In the face of such complexity, rare forms of diabetes that result from single-gene mutations (monogenic diabetes) can be used to model the contribution of individual genetic factors to pancreatic β-cell dysfunction and the breakdown of glucose homeostasis. Here we review the contribution of protein coding and non-protein coding genetic disease modifiers to the pathogenesis of diabetes subtypes, as well as how recent technological advances in the generation, differentiation, and genome editing of human pluripotent stem cells (hPSC) enable the development of cell-based disease models. Finally, we describe a disease modifier discovery platform that utilizes these technologies to identify novel genetic modifiers using induced pluripotent stem cells (iPSC) derived from patients with monogenic diabetes caused by heterozygous mutations.
Collapse
Affiliation(s)
- Matthew N. George
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Karla F. Leavens
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Division of Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Paul Gadue
- Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| |
Collapse
|
37
|
Heller S, Melzer MK, Azoitei N, Julier C, Kleger A. Human Pluripotent Stem Cells Go Diabetic: A Glimpse on Monogenic Variants. Front Endocrinol (Lausanne) 2021; 12:648284. [PMID: 34079523 PMCID: PMC8166226 DOI: 10.3389/fendo.2021.648284] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 04/13/2021] [Indexed: 12/17/2022] Open
Abstract
Diabetes, as one of the major diseases in industrial countries, affects over 350 million people worldwide. Type 1 (T1D) and type 2 diabetes (T2D) are the most common forms with both types having invariable genetic influence. It is accepted that a subset of all diabetes patients, generally estimated to account for 1-2% of all diabetic cases, is attributed to mutations in single genes. As only a subset of these genes has been identified and fully characterized, there is a dramatic need to understand the pathophysiological impact of genetic determinants on β-cell function and pancreatic development but also on cell replacement therapies. Pluripotent stem cells differentiated along the pancreatic lineage provide a valuable research platform to study such genes. This review summarizes current perspectives in applying this platform to study monogenic diabetes variants.
Collapse
Affiliation(s)
- Sandra Heller
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Michael Karl Melzer
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
- Department of Urology, Ulm University Hospital, Ulm, Germany
| | - Ninel Azoitei
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| | - Cécile Julier
- Université de Paris, Institut Cochin, INSERM U1016, CNRS UMR-8104, Paris, France
| | - Alexander Kleger
- Department of Internal Medicine I, Ulm University Hospital, Ulm, Germany
| |
Collapse
|
38
|
Type I interferons as key players in pancreatic β-cell dysfunction in type 1 diabetes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2021; 359:1-80. [PMID: 33832648 DOI: 10.1016/bs.ircmb.2021.02.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease characterized by pancreatic islet inflammation (insulitis) and specific pancreatic β-cell destruction by an immune attack. Although the precise underlying mechanisms leading to the autoimmune assault remain poorly understood, it is well accepted that insulitis takes place in the context of a conflicting dialogue between pancreatic β-cells and the immune cells. Moreover, both host genetic background (i.e., candidate genes) and environmental factors (e.g., viral infections) contribute to this inadequate dialogue. Accumulating evidence indicates that type I interferons (IFNs), cytokines that are crucial for both innate and adaptive immune responses, act as key links between environmental and genetic risk factors in the development of T1D. This chapter summarizes some relevant pathways involved in β-cell dysfunction and death, and briefly reviews how enteroviral infections and genetic susceptibility can impact insulitis. Moreover, we present the current evidence showing that, in β-cells, type I IFN signaling pathway activation leads to several outcomes, such as long-lasting major histocompatibility complex (MHC) class I hyperexpression, endoplasmic reticulum (ER) stress, epigenetic changes, and induction of posttranscriptional as well as posttranslational modifications. MHC class I overexpression, when combined with ER stress and posttranscriptional/posttranslational modifications, might lead to sustained neoantigen presentation to immune system and β-cell apoptosis. This knowledge supports the concept that type I IFNs are implicated in the early stages of T1D pathogenesis. Finally, we highlight the promising therapeutic avenues for T1D treatment directed at type I IFN signaling pathway.
Collapse
|
39
|
Pang H, Xia Y, Luo S, Huang G, Li X, Xie Z, Zhou Z. Emerging roles of rare and low-frequency genetic variants in type 1 diabetes mellitus. J Med Genet 2021; 58:289-296. [PMID: 33753534 PMCID: PMC8086251 DOI: 10.1136/jmedgenet-2020-107350] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 12/12/2022]
Abstract
Type 1 diabetes mellitus (T1DM) is defined as an autoimmune disorder and has enormous complexity and heterogeneity. Although its precise pathogenic mechanisms are obscure, this disease is widely acknowledged to be precipitated by environmental factors in individuals with genetic susceptibility. To date, the known susceptibility loci, which have mostly been identified by genome-wide association studies, can explain 80%–85% of the heritability of T1DM. Researchers believe that at least a part of its missing genetic component is caused by undetected rare and low-frequency variants. Most common variants have only small to modest effect sizes, which increases the difficulty of dissecting their functions and restricts their potential clinical application. Intriguingly, many studies have indicated that rare and low-frequency variants have larger effect sizes and play more significant roles in susceptibility to common diseases, including T1DM, than common variants do. Therefore, better recognition of rare and low-frequency variants is beneficial for revealing the genetic architecture of T1DM and for providing new and potent therapeutic targets for this disease. Here, we will discuss existing challenges as well as the great significance of this field and review current knowledge of the contributions of rare and low-frequency variants to T1DM.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ying Xia
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shuoming Luo
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Xia Li
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| |
Collapse
|
40
|
Sharma C, R Ali B, Osman W, Afandi B, Aburawi EH, Beshyah SA, Al-Mahayri Z, Al-Rifai RH, Al Yafei Z, ElGhazali G, Alkaabi J. Association of variants in PTPN22, CTLA-4, IL2-RA, and INS genes with type 1 diabetes in Emiratis. Ann Hum Genet 2021; 85:48-57. [PMID: 32970831 DOI: 10.1111/ahg.12406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 08/07/2020] [Accepted: 09/04/2020] [Indexed: 02/05/2023]
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease with a complex interrelation of genetic and environmental factors. Genetic studies have reported HLA and non-HLA loci as significant contributors to T1D. However, the genetic basis of T1D among Emiratis is unexplored. This study aims to determine the contribution of four genes PTPN22, CTLA-4, IL2-RA, and INS to T1D risk among Emiratis. The association between variants in PTPN22 (rs2476601, rs1310182), CTLA-4 (rs11571316, rs231775, rs3087243, rs1427676, and rs231727), IL2-RA (rs7090530), and INS (rs7111341) with T1D was tested in 310 Emiratis (139 T1D patients and 171 controls). A significant association was found at rs1310182, and rs2476601 both in PTPN22, rs3087243, and rs231775 both in CTLA-4, and rs12251307 in IL2-RA. Moreover, a haplotype constituted from GG and AG genotypes at rs231727 and rs231775, respectively, in CTLA-4 was significantly associated with an increased T1D risk. The cumulative effects of risk alleles for all significantly associated SNPs showed 11.8 higher relative risk for T1D for those who carry 5-6 compared to 0-1 risk alleles. This study illustrated that PTPN22, CTLA-4, and IL2-RA gene variants could confer risk alleles for T1D among the Emirati population.
Collapse
Affiliation(s)
- Charu Sharma
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, UAE
| | - Bassam R Ali
- Department of Genetics and Genomics, College of Medicine and Health Sciences, United Arab Emirates University, UAE
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, UAE
| | - Wael Osman
- College of Arts and Sciences, Khalifa University, Abu Dhabi, UAE
| | | | - Elhadi H Aburawi
- Department of Pediatrics, College of Medicine and Health Sciences, United Arab Emirates University, UAE
| | | | - Zeina Al-Mahayri
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, UAE
| | - Rami H Al-Rifai
- Institute of Public Health, College of Medicine and Health Sciences, United Arab Emirates University, UAE
| | - Zain Al Yafei
- Department of Immunology, PLMS, Sheikh Khalifa Medical City, Abu Dhabi, UAE
| | - Gehad ElGhazali
- Department of Immunology, PLMS, Sheikh Khalifa Medical City, Abu Dhabi, UAE
| | - Juma Alkaabi
- Department of Internal Medicine, College of Medicine and Health Sciences, United Arab Emirates University, UAE
| |
Collapse
|
41
|
Milluzzo A, Falorni A, Brozzetti A, Pezzino G, Tomaselli L, Tumminia A, Frittitta L, Vigneri R, Sciacca L. Risk for Coexistent Autoimmune Diseases in Familial and Sporadic Type 1 Diabetes is Related to Age at Diabetes Onset. Endocr Pract 2021; 27:110-117. [PMID: 33616044 DOI: 10.1016/j.eprac.2020.09.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 07/31/2020] [Accepted: 09/04/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE Type 1 diabetes (T1D) is frequently associated with other autoimmune diseases (AIDs). Although most of T1D patients are sporadic cases (S-T1D), 10% to 15% have a familial form (F-T1D) involving 2 or more first-degree relatives. This study evaluated the effect of T1D family aggregation and age onset on AIDs occurrence. METHODS In this observational, cross-sectional, case-control, single center study, we enrolled 115 F-T1D and 115 S-T1D patients matched for gender, age, T1D age onset, and duration. With respect to T1D age onset (before or after 18 years), both groups were further subdivided into young- or adult-onset F-T1D and young- or adult-onset S-T1D. The presence of organ-specific antibodies and/or overt AIDs was evaluated. RESULTS The F-T1D group had a higher percentage of AIDs (29.8% vs 18.4%, P = .04) and a significant earlier onset of AIDs at Cox regression analysis (P = .04) than the S-T1D group. Based on multivariate analysis, the adult-onset F-T1D subgroup had the highest prevalence of both additional organ-specific antibodies (60.5%) and overt AIDs (34.9%), whereas the adult S-T1D subgroup was the least frequently involved (29.1% and 12.7%, respectively). In F-T1D patients, offsprings develop T1D and AIDs earlier than their parents do. CONCLUSIONS In T1D patients, familial aggregation and adult-onset of T1D increase the risk for coexistent AIDs. These clinical predictors could guide clinicians to address T1D patients for the screening of T1D-related AIDs.
Collapse
Affiliation(s)
- Agostino Milluzzo
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Catania, Italy
| | - Alberto Falorni
- Department of Medicine, University of Perugia, Perugia, Italy
| | | | - Giulia Pezzino
- Endocrinology, Azienda Ospedaliera di Rilievo Nazionale ad Alta Specializzazione Garibaldi, Catania, Italy
| | - Letizia Tomaselli
- Endocrinology, Azienda Ospedaliera di Rilievo Nazionale ad Alta Specializzazione Garibaldi, Catania, Italy
| | - Andrea Tumminia
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Catania, Italy
| | - Lucia Frittitta
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Catania, Italy
| | - Riccardo Vigneri
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Catania, Italy; Institute of Crystallography, Catania Section, National Research Council, Catania, Italy
| | - Laura Sciacca
- Department of Clinical and Experimental Medicine, Endocrinology Section, University of Catania Medical School, Catania, Italy.
| |
Collapse
|
42
|
Mannering SI, Bhattacharjee P. Insulin's other life: an autoantigen in type 1 diabetes. Immunol Cell Biol 2021; 99:448-460. [PMID: 33524197 DOI: 10.1111/imcb.12442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/21/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022]
Abstract
One hundred years ago, Frederick Banting, John Macleod, Charles Best and James Collip, and their collaborators, discovered insulin. This discovery paved the way to saving countless lives and ushered in the "Insulin Era." Since the discovery of insulin, we have made enormous strides in understanding its role in metabolism and diabetes. Insulin has played a dramatic role in the treatment of people with diabetes; particularly type 1 diabetes (T1D). Insulin replacement is a life-saving therapy for people with T1D and some with type 2 diabetes. T1D is an autoimmune disease caused by the T-cell-mediated destruction of the pancreatic insulin-producing beta cells that leads to a primary insulin deficiency. It has become increasingly clear that insulin, and its precursors preproinsulin (PPI) and proinsulin (PI), can play another role-not as a hormone but as an autoantigen in T1D. Here we review the role played by the products of the INS gene as autoantigens in people with T1D. From many elegant animal studies, it is clear that T-cell responses to insulin, PPI and PI are essential for T1D to develop. Here we review the evidence that autoimmune responses to insulin and PPI arise in people with T1D and discuss the recently described neoepitopes derived from the products of the insulin gene. Finally, we look forward to new approaches to deliver epitopes derived from PPI, PI and insulin that may allow immune tolerance to pancreatic beta cells to be restored in people with, or at risk of, T1D.
Collapse
Affiliation(s)
- Stuart I Mannering
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia.,Department of Medicine, St. Vincent's Hospital, University of Melbourne, Fitzroy, VIC, Australia
| | - Pushpak Bhattacharjee
- Immunology and Diabetes Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| |
Collapse
|
43
|
Battaglia M, Buckner JH, Levings MK, Richardson SJ, Wong FS, Tree TI. Identifying the 'Achilles heel' of type 1 diabetes. Clin Exp Immunol 2021; 204:167-178. [PMID: 33368173 DOI: 10.1111/cei.13570] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/16/2020] [Accepted: 11/18/2020] [Indexed: 12/22/2022] Open
Abstract
When Thetis dipped her son Achilles into the River Styx to make him immortal, she held him by the heel, which was not submerged, and thus created a weak spot that proved deadly for Achilles. Millennia later, Achilles heel is part of today's lexicon meaning an area of weakness or a vulnerable spot that causes failure. Also implied is that an Achilles heel is often missed, forgotten or under-appreciated until it is under attack, and then failure is fatal. Paris killed Achilles with an arrow 'guided by the Gods'. Understanding the pathogenesis of type 1 diabetes (T1D) in order to direct therapy for prevention and treatment is a major goal of research into T1D. At the International Congress of the Immunology of Diabetes Society, 2018, five leading experts were asked to present the case for a particular cell/element that could represent 'the Achilles heel of T1D'. These included neutrophils, B cells, CD8+ T cells, regulatory CD4+ T cells, and enteroviruses, all of which have been proposed to play an important role in the pathogenesis of type 1 diabetes. Did a single entity emerge as 'the' Achilles heel of T1D? The arguments are summarized here, to make this case.
Collapse
Affiliation(s)
- M Battaglia
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Hospital, Milan, Italy
| | - J H Buckner
- Translational Research Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - M K Levings
- BC Children's Hospital Research Institute, Vancouver, BC, Canada
| | - S J Richardson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, Exeter, UK
| | - F S Wong
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, UK
| | - T I Tree
- Department of Immunobiology, School of Immunology and Microbial Sciences (SIMS), King's College London, London, UK.,NIHR Biomedical Research Centre, Guy's and St Thomas' NHS Foundation Trust and King's College London, London, UK
| |
Collapse
|
44
|
Ke Q, Kroger CJ, Clark M, Tisch RM. Evolving Antibody Therapies for the Treatment of Type 1 Diabetes. Front Immunol 2021; 11:624568. [PMID: 33679717 PMCID: PMC7930374 DOI: 10.3389/fimmu.2020.624568] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 12/31/2020] [Indexed: 12/24/2022] Open
Abstract
Type 1 diabetes (T1D) is widely considered to be a T cell driven autoimmune disease resulting in reduced insulin production due to dysfunction/destruction of pancreatic β cells. Currently, there continues to be a need for immunotherapies that selectively reestablish persistent β cell-specific self-tolerance for the prevention and remission of T1D in the clinic. The utilization of monoclonal antibodies (mAb) is one strategy to target specific immune cell populations inducing autoimmune-driven pathology. Several mAb have proven to be clinically safe and exhibit varying degrees of efficacy in modulating autoimmunity, including T1D. Traditionally, mAb therapies have been used to deplete a targeted cell population regardless of antigenic specificity. However, this treatment strategy can prove detrimental resulting in the loss of acquired protective immunity. Nondepleting mAb have also been applied to modulate the function of immune effector cells. Recent studies have begun to define novel mechanisms associated with mAb-based immunotherapy that alter the function of targeted effector cell pools. These results suggest short course mAb therapies may have persistent effects for regaining and maintaining self-tolerance. Furthermore, the flexibility to manipulate mAb properties permits the development of novel strategies to target multiple antigens and/or deliver therapeutic drugs by a single mAb molecule. Here, we discuss current and potential future therapeutic mAb treatment strategies for T1D, and T cell-mediated autoimmunity.
Collapse
Affiliation(s)
- Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
45
|
Clark M, Kroger CJ, Ke Q, Tisch RM. The Role of T Cell Receptor Signaling in the Development of Type 1 Diabetes. Front Immunol 2021; 11:615371. [PMID: 33603744 PMCID: PMC7884625 DOI: 10.3389/fimmu.2020.615371] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 12/15/2020] [Indexed: 12/15/2022] Open
Abstract
T cell receptor (TCR) signaling influences multiple aspects of CD4+ and CD8+ T cell immunobiology including thymic development, peripheral homeostasis, effector subset differentiation/function, and memory formation. Additional T cell signaling cues triggered by co-stimulatory molecules and cytokines also affect TCR signaling duration, as well as accessory pathways that further shape a T cell response. Type 1 diabetes (T1D) is a T cell-driven autoimmune disease targeting the insulin producing β cells in the pancreas. Evidence indicates that dysregulated TCR signaling events in T1D impact the efficacy of central and peripheral tolerance-inducing mechanisms. In this review, we will discuss how the strength and nature of TCR signaling events influence the development of self-reactive T cells and drive the progression of T1D through effects on T cell gene expression, lineage commitment, and maintenance of pathogenic anti-self T cell effector function.
Collapse
Affiliation(s)
- Matthew Clark
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Charles J Kroger
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Qi Ke
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Roland M Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
46
|
Ergun-Longmire B, Clemente E, Vining-Maravolo P, Roberts C, Buth K, Greydanus DE. Diabetes education in pediatrics: How to survive diabetes. Dis Mon 2021; 67:101153. [PMID: 33541707 DOI: 10.1016/j.disamonth.2021.101153] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus is the most common abnormal carbohydrate metabolism disorder affecting millions of people worldwide. It is characterized by hyperglycemia as a result of ß-cell destruction or dysfunction by both genetic and environmental factors. Over time chronic hyperglycemia leads to microvascular (i.e., retinopathy, nephropathy and neuropathy) and macrovascular (i.e., ischemic heart disease, peripheral vascular disease, and cerebrovascular disease) complications of diabetes. Diabetes complication trials showed the importance of achieving near-normal glycemic control to prevent and/or reduce diabetes-related morbidity and mortality. There is a staggering rate of increased incidence of diabetes in youth, raising concerns for future generations' health, quality of life and its enormous economic burden. Despite advancements in the technology, diabetes management remains cumbersome. Training individuals with diabetes to gain life-long survival skills requires a comprehensive and ongoing diabetes education by a multidisciplinary team. Diabetes education and training start at the time of diagnosis of diabetes and should be continuous throughout the course of disease. The goal is to empower the individuals and families to gain diabetes self-management skills. Diabetes education must be individualized depending on the individual's age, education, family dynamics, and support. In this article, we review the history of diabetes, etiopathogenesis and clinical presentation of both type 1 and type 2 diabetes in children as well as adolescents. We then focus on diabetes management with education methods and materials.
Collapse
Affiliation(s)
- Berrin Ergun-Longmire
- Associate Professor, Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA.
| | - Ethel Clemente
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Patricia Vining-Maravolo
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Cheryl Roberts
- Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Koby Buth
- Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI, USA
| | - Donald E Greydanus
- Professor, Department of Pediatric and Adolescent Medicine, Western Michigan University, Homer Stryker M.D. School of Medicine, Kalamazoo, MI United States
| |
Collapse
|
47
|
Kohil A, Al-Asmakh M, Al-Shafai M, Terranegra A. The Interplay Between Diet and the Epigenome in the Pathogenesis of Type-1 Diabetes. Front Nutr 2021; 7:612115. [PMID: 33585535 PMCID: PMC7876257 DOI: 10.3389/fnut.2020.612115] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/22/2020] [Indexed: 12/12/2022] Open
Abstract
The autoimmune disease, Type 1 Diabetes Mellitus (T1DM), results in the destruction of pancreatic β-cells, and the International Diabetes Federation reports that its incidence is increasing worldwide. T1DM is a complex disease due to the interaction between genetic and environmental factors. Certain dietary patterns and nutrients are known to cause epigenetic modifications in physiological conditions and diseases. However, the interplay between diet and epigenetics is not yet well-understood in the context of T1DM. Several studies have described epigenetic mechanisms involved in the autoimmune reactions that destroy the β-cells, but few explored diet components as potential triggers for epigenetic modifications. Clarifying the link between diet and epigenome can provide new insights into the pathogenesis of T1DM, potentially leading to new diagnostic and therapeutic approaches. In this mini review, we shed light on the influence of the diet-epigenome axis on the pathophysiology of T1DM.
Collapse
Affiliation(s)
- Amira Kohil
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar
| | - Maha Al-Asmakh
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar.,Biomedical Research Center, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Mashael Al-Shafai
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | | |
Collapse
|
48
|
Reading between the (Genetic) Lines: How Epigenetics is Unlocking Novel Therapies for Type 1 Diabetes. Cells 2020; 9:cells9112403. [PMID: 33153010 PMCID: PMC7692667 DOI: 10.3390/cells9112403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 12/19/2022] Open
Abstract
Type 1 diabetes (T1D) is an autoimmune condition where the body’s immune cells destroy their insulin-producing pancreatic beta cells leading to dysregulated glycaemia. Individuals with T1D control their blood glucose through exogenous insulin replacement therapy, often using multiple daily injections or pumps. However, failure to accurately mimic intrinsic glucose regulation results in glucose fluctuations and long-term complications impacting key organs such as the heart, kidneys, and/or the eyes. It is well established that genetic and environmental factors contribute to the initiation and progression of T1D, but recent studies show that epigenetic modifications are also important. Here, we discuss key epigenetic modifications associated with T1D pathogenesis and discuss how recent research is finding ways to harness epigenetic mechanisms to prevent, reverse, or manage T1D.
Collapse
|
49
|
Sur S. In silico analysis reveals interrelation of enriched pathways and genes in type 1 diabetes. Immunogenetics 2020; 72:399-412. [PMID: 32860078 DOI: 10.1007/s00251-020-01177-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
Type 1 diabetes (T1D) is a multifactorial, polygenic complex autoimmune disease damaging pancreatic islet β cells. Numerous genes linked to T1D have been discovered through genetical studies, GWAS and polymorphisms. Most genetical studies focused on independent genes while others overemphasized on SNPs. Here, a collective analysis of documented T1D-associated genes was performed using bioinformatics tools. Enriched biological pathways, functions, enrichment clustering, networks and interactomes were analysed. Besides, meta-analyses of T1D-associated genes and T1D-related genes from SNPs were investigated to find common genes, pathways, enrichment and interrelationships. Notable enriched pathways comprised of cytokine-mediated signalling, cytokine production, interferon gamma production, myeloid leukocyte activation, activation of immune response, lymphocyte activation, adaptive immune response, Th17 cell differentiation etc. Enrichment analysis of T1D-associated genes emphasized the role of immune-linked machineries in metabolism, disease progression and aetiology of type 1 diabetes. Interactome analysis revealed overrepresentation of T1D-associated genes compared with T1D-related genes from SNPs. MCODE components highlighted the significance of pathways linked to vitamin D metabolism, signalling by interleukins, toll-like receptors, chemokines, PD-1, NOTCH, antigen processes etc. About 153 genes from MCODE complexes representing enriched pathways of T1D-associated genes and T1D-related genes from SNPs play a crucial role and may be important for further investigations. The information may be valuable for designing precision medicine-based therapeutics.
Collapse
Affiliation(s)
- Saubashya Sur
- Postgraduate Department of Botany, Life Sciences Block, Ramananda College, Bishnupur, West Bengal, 722122, India.
| |
Collapse
|
50
|
Sliz A, Locker KCS, Lampe K, Godarova A, Plas DR, Janssen EM, Jones H, Herr AB, Hoebe K. Gab3 is required for IL-2- and IL-15-induced NK cell expansion and limits trophoblast invasion during pregnancy. Sci Immunol 2020; 4:4/38/eaav3866. [PMID: 31375526 DOI: 10.1126/sciimmunol.aav3866] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 07/03/2019] [Indexed: 12/12/2022]
Abstract
The scaffolding protein Grb2-associated binding protein 3 (Gab3) is a member of the Gab family, whose functions have remained elusive. Here, we identify Gab3 as a key determinant of peripheral NK cell expansion. Loss of Gab3 resulted in impaired IL-2 and IL-15-induced NK cell priming and expansion due to a selective impairment in MAPK signaling but not STAT5 signaling. In vivo, we found that Gab3 is required for recognition and elimination of "missing-self" and tumor targets. Unexpectedly, our studies also revealed that Gab3 plays an important role during pregnancy. Gab3-deficient mice exhibited impaired uterine NK cell expansion associated with abnormal spiral artery remodeling and increased trophoblast invasion in the decidua basalis. This coincided with stillbirth, retained placenta, maternal hemorrhage, and undelivered fetoplacental units at term. Thus, Gab3 is a key component required for cytokine-mediated NK cell priming and expansion that is essential for antitumor responses and limits trophoblast cell invasion during pregnancy.
Collapse
Affiliation(s)
- Anna Sliz
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Kathryn C S Locker
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Kristin Lampe
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA
| | - Alzbeta Godarova
- Biomedical Informatics Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - David R Plas
- Vontz Center for Molecular Studies, University of Cincinnati, Cincinnati, OH 45267, USA
| | | | - Helen Jones
- Division of General Pediatric and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA.,Department of Surgery, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | - Andrew B Herr
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA.,Division of Immunobiology, Cincinnati Children's Hospital Research Foundation, Cincinnati, OH 45229, USA.,Immunology Graduate Program, Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine, Cincinnati, OH 45220, USA
| | | |
Collapse
|