1
|
Walker SL, Leete P, Boldison J. Tissue Resident and Infiltrating Immune Cells: Their Influence on the Demise of Beta Cells in Type 1 Diabetes. Biomolecules 2025; 15:441. [PMID: 40149976 PMCID: PMC11939886 DOI: 10.3390/biom15030441] [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: 02/10/2025] [Revised: 03/11/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Type 1 diabetes (T1D) is an organ-specific autoimmune disease that results in the selective loss of pancreatic beta cells and an eventual deficit in insulin production to maintain glucose homeostasis. It is now increasingly accepted that this dynamic disease process is multifactorial; involves a variety of immune cells which contribute to an inflamed pancreatic microenvironment; and that the condition is heterogenous, resulting in variable rates of subsequent beta cell damage. In this review, we will explore the current understanding of the cellular interactions between both resident and infiltrating immune cells within the pancreatic environment, highlighting key mechanisms which may promote the beta cell destruction and islet damage associated with T1D.
Collapse
Affiliation(s)
| | | | - Joanne Boldison
- Department of Clinical and Biomedical Sciences, University of Exeter, RILD Building (Level 4), Barrack Road, Exeter EX2 5DW, UK; (S.L.W.); (P.L.)
| |
Collapse
|
2
|
Adebekun J, Nadig A, Saarah P, Asgari S, Kachuri L, Alagpulinsa DA. Genetic relations between type 1 diabetes, coronary artery disease and leukocyte counts. Diabetologia 2024; 67:2518-2529. [PMID: 39141130 DOI: 10.1007/s00125-024-06247-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 06/19/2024] [Indexed: 08/15/2024]
Abstract
AIMS/HYPOTHESIS Type 1 diabetes is associated with excess coronary artery disease (CAD) risk even when known cardiovascular risk factors are accounted for. Genetic perturbation of haematopoiesis that alters leukocyte production is a novel independent modifier of CAD risk. We examined whether there are shared genetic determinants and causal relationships between type 1 diabetes, CAD and leukocyte counts. METHODS Genome-wide association study summary statistics were used to perform pairwise linkage disequilibrium score regression and heritability estimation from summary statistics (ρ-HESS) to respectively estimate the genome-wide and local genetic correlations, and two-sample Mendelian randomisation to estimate the causal relationships between leukocyte counts (335,855 healthy individuals), type 1 diabetes (18,942 cases, 501,638 control individuals) and CAD (122,733 cases, 424,528 control individuals). A latent causal variable (LCV) model was performed to estimate the genetic causality proportion of the genetic correlation between type 1 diabetes and CAD. RESULTS There was significant genome-wide genetic correlation (rg) between type 1 diabetes and CAD (rg=0.088, p=8.60 × 10-3) and both diseases shared significant genome-wide genetic determinants with eosinophil count (rg for type 1 diabetes [rg(T1D)]=0.093, p=7.20 × 10-3, rg for CAD [rg(CAD)]=0.092, p=3.68 × 10-6) and lymphocyte count (rg(T1D)=-0.052, p=2.76 × 10-2, rg(CAD)=0.176, p=1.82 × 10-15). Sixteen independent loci showed stringent Bonferroni significant local genetic correlations between leukocyte counts, type 1 diabetes and/or CAD. Cis-genetic regulation of the expression levels of genes within shared loci between type 1 diabetes and CAD was associated with both diseases as well as leukocyte counts, including SH2B3, CTSH, MORF4L1, CTRB1, CTRB2, CFDP1 and IFIH1. Genetically predicted lymphocyte, neutrophil and eosinophil counts were associated with type 1 diabetes and CAD (lymphocyte OR for type 1 diabetes [ORT1D]=0.67, p=2.02-19, ORCAD=1.09, p=2.67 × 10-6; neutrophil ORT1D=0.82, p=5.63 × 10-5, ORCAD=1.17, p=5.02 × 10-14; and eosinophil ORT1D=1.67, p=5.45 × 10-25, ORCAD=1.07, p=2.03 × 10-4. The genetic causality proportion between type 1 diabetes and CAD was 0.36 ± 0.16 (pLCV=1.30 × 10-2), suggesting a possible intermediary causal variable. CONCLUSIONS/INTERPRETATION This study sheds light on shared genetic mechanisms underlying type 1 diabetes and CAD, which may contribute to their co-occurrence through regulation of gene expression and leukocyte counts and identifies cellular and molecular targets for further investigation for disease prediction and potential drug discovery.
Collapse
Affiliation(s)
- Jolade Adebekun
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Ajay Nadig
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Priscilla Saarah
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Samira Asgari
- Institute for Genomic Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Linda Kachuri
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - David A Alagpulinsa
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA.
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
3
|
Luo J, Wang J, Xiang Y, Wang N, Zhao X, Liu G, Liu L, Chang H. Unveiling the influence of circulating immune cells count on type 1 diabetes: Insight from bidirectional Mendelian randomization. Medicine (Baltimore) 2024; 103:e39842. [PMID: 39331871 PMCID: PMC11441873 DOI: 10.1097/md.0000000000039842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 09/03/2024] [Indexed: 09/29/2024] Open
Abstract
Observational studies have demonstrated an association between circulating immune cell and type 1 diabetes (T1D) risk. However, it is unknown whether this relationship is causal. Herein, we adopted a 2-sample bidirectional Mendelian randomization study to figure out whether circulating immune cell profiles causally impact T1D liability. Summary statistical data were obtained from genome-wide association study (GWAS) to investigate the causal relationship between white cell (WBC) count, 5 specific WBC count, and lymphocyte subtypes cell count and T1D risk. After false discovery rate (FDR) correction, the results indicated that lower lymphocyte cell count (odds ratio [OR] per 1 standard deviation [SD] decrease = 0.746, 95% confidence interval (CI): 0.673-0.828, PFDR = 0.036), and basophil cell count (OR per 1 SD decrease = 0.808, 95% CI: 0.700-0.932, PFDR = 0.010) were causally associated with T1D susceptibility. However, the absolute count of WBC, monocyte, neutrophil, eosinophil, and lymphocyte subtypes cell had no statistically significant effect on T1D risk. Taken together, this study indicates suggestive association between circulating immune cell count and T1D. Moreover, lower numbers of circulating lymphocyte and basophil cell were associated with the increased risk of T1D, which confirmed the immunity predisposition for T1D.
Collapse
Affiliation(s)
- Jia Luo
- Changsha Blood Center, Changsha, Hunan Province, China
| | - Jing Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yukun Xiang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ningning Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xin Zhao
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - GengYan Liu
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan Province, China
| | - Lihui Liu
- Changsha Blood Center, Changsha, Hunan Province, China
| | - Haoxiao Chang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
4
|
Abbam G, Mensah K, Appiah SK, Nkansah C, Daud S, Aikins CN, Osei-Afoakwa AN, Osei-Boakye F, Derigubah CA, Mohammed S, Tandoh S, Bani SB. Complete Blood Count Reference Intervals for Children Aged Less Than 1 to 12 Years in the Northern Region of Ghana. BIOMED RESEARCH INTERNATIONAL 2024; 2024:6607281. [PMID: 38764534 PMCID: PMC11101252 DOI: 10.1155/2024/6607281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 04/11/2024] [Accepted: 04/29/2024] [Indexed: 05/21/2024]
Abstract
Reliable laboratory diagnostic results are key for evaluating and improving children's health. To interpret these results, child-specific reference intervals (RIs), which account for constant biological changes and physiological development with sex and age, are required, as recommended by the Clinical and Laboratory Standards Institute (CLSI). This study presents age- and sex-specific reference intervals for complete blood count (CBC) parameters in children (<1-12 years old) in the Northern Region of Ghana. In this cross-sectional study, 600 healthy children from randomly sampled schools in Tamale (the Northern Region) were recruited and screened. Data from 388 eligible children were used to nonparametrically determine the reference intervals of CBC parameters at the 2.5th and 97.5th percentiles. The CBC reference intervals were compared for variations in sex and age groups using the Wilcoxon rank-sum test. There were no statistically significant differences in most CBC parameters by sex (RBC, Hb, HCT, MCH, RDW (CV/SD), WBC, LYM#, MON#(%) NEU#(%), EOS#(%), and BAS#(%); p > 0.05) and age group (RBC, MCV, RDW (CV/SD), WBC, LYM#, MON#(%) NEU#(%), EOS#(%), and BAS%; p > 0.05). However, there were observable differences between this locally established CBC reference interval and that used for children at Tamale Teaching Hospital (manufacturer's RIs). This study emphasises the importance of determining reference intervals representative of the local child population and incorporating them into the current reporting system of laboratories in the Northern Region to ensure the provision of effective and efficient healthcare services.
Collapse
Affiliation(s)
- Gabriel Abbam
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Kofi Mensah
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
| | - Samuel Kwasi Appiah
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
| | - Charles Nkansah
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
| | - Samira Daud
- Department of Haematology, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Cheryl Namusoke Aikins
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Akua Nyarko Osei-Afoakwa
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| | - Felix Osei-Boakye
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
- Department of Medical Laboratory Technology, Faculty of Applied Science and Technology, Sunyani Technical University, Sunyani, Ghana
| | - Charles Angnataa Derigubah
- Department of Medical Laboratory Science, Faculty of Health Science and Technology, Ebonyi State University, Abakaliki, Nigeria
- Department of Medical Laboratory Technology, School of Applied Science and Arts, Bolgatanga Technical University, Bolgatanga, Ghana
| | - Sanda Mohammed
- Systems Solutions Geospatial Research Services, Accra, Ghana
- Research Department, SSNIT, Accra, Ghana
| | - Samuel Tandoh
- University Clinic Laboratory, University of Education, Winneba, Ghana
| | - Simon Bannison Bani
- Department of Biomedical Laboratory Sciences, School of Allied Health Sciences, University for Development Studies, Tamale, Ghana
| |
Collapse
|
5
|
Dashti M, Nizam R, Jacob S, Al-Kandari H, Al Ozairi E, Thanaraj TA, Al-Mulla F. Association between alleles, haplotypes, and amino acid variations in HLA class II genes and type 1 diabetes in Kuwaiti children. Front Immunol 2023; 14:1238269. [PMID: 37638053 PMCID: PMC10457110 DOI: 10.3389/fimmu.2023.1238269] [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: 06/11/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Type 1 diabetes (T1D) is a complex autoimmune disorder that is highly prevalent globally. The interactions between genetic and environmental factors may trigger T1D in susceptible individuals. HLA genes play a significant role in T1D pathogenesis, and specific haplotypes are associated with an increased risk of developing the disease. Identifying risk haplotypes can greatly improve the genetic scoring for early diagnosis of T1D in difficult to rank subgroups. This study employed next-generation sequencing to evaluate the association between HLA class II alleles, haplotypes, and amino acids and T1D, by recruiting 95 children with T1D and 150 controls in the Kuwaiti population. Significant associations were identified for alleles at the HLA-DRB1, HLA-DQA1, and HLA-DQB1 loci, including DRB1*03:01:01, DQA1*05:01:01, and DQB1*02:01:01, which conferred high risk, and DRB1*11:04:01, DQA1*05:05:01, and DQB1*03:01:01, which were protective. The DRB1*03:01:01~DQA1*05:01:01~DQB1*02:01:01 haplotype was most strongly associated with the risk of developing T1D, while DRB1*11:04-DQA1*05:05-DQB1*03:01 was the only haplotype that rendered protection against T1D. We also identified 66 amino acid positions across the HLA-DRB1, HLA-DQA1, and HLA-DQB1 genes that were significantly associated with T1D, including novel associations. These results validate and extend our knowledge on the associations between HLA genes and T1D in Kuwaiti children. The identified risk alleles, haplotypes, and amino acid variations may influence disease development through effects on HLA structure and function and may allow early intervention via population-based screening efforts.
Collapse
Affiliation(s)
- Mohammed Dashti
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Rasheeba Nizam
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Sindhu Jacob
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| | - Hessa Al-Kandari
- Department of Population Health, Dasman Diabetes Institute, Dasman, Kuwait
- Department of Pediatrics, Farwaniya Hospital, Ministry of Health, Sabah Al Nasser, Kuwait
| | - Ebaa Al Ozairi
- Clinical Care Research and Trials, Dasman Diabetes Institute, Dasman, Kuwait
- Faculty of Medicine, Kuwait University, Jabriya, Kuwait
| | | | - Fahd Al-Mulla
- Department of Genetics and Bioinformatics, Dasman Diabetes Institute, Dasman, Kuwait
| |
Collapse
|
6
|
Brenu EW, Harris M, Hamilton-Williams EE. Circulating biomarkers during progression to type 1 diabetes: A systematic review. Front Endocrinol (Lausanne) 2023; 14:1117076. [PMID: 36817583 PMCID: PMC9935596 DOI: 10.3389/fendo.2023.1117076] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 01/25/2023] [Indexed: 02/05/2023] Open
Abstract
AIM Progression to type 1 diabetes (T1D) is defined in stages and clinical disease is preceded by a period of silent autoimmunity. Improved prediction of the risk and rate of progression to T1D is needed to reduce the prevalence of diabetic ketoacidosis at presentation as well as for staging participants for clinical trials. This systematic review evaluates novel circulating biomarkers associated with future progression to T1D. METHODS PubMed, Ovid, and EBSCO databases were used to identify a comprehensive list of articles. The eligibility criteria included observational studies that evaluated the usefulness of circulating markers in predicting T1D progression in at-risk subjects <20 years old. RESULTS Twenty-six studies were identified, seventeen were cohort studies and ten were case control studies. From the 26 studies, 5 found evidence for protein and lipid dysregulation, 11 identified molecular markers while 12 reported on changes in immune parameters during progression to T1D. An increased risk of T1D progression was associated with the presence of altered gene expression, immune markers including regulatory T cell dysfunction and higher short-lived effector CD8+ T cells in progressors. DISCUSSION Several circulating biomarkers are dysregulated before T1D diagnosis and may be useful in predicting either the risk or rate of progression to T1D. Further studies are required to validate these biomarkers and assess their predictive accuracy before translation into broader use. SYSTEMATIC REVIEW REGISTRATION https://www.crd.york.ac.uk/prospero, identifier (CRD42020166830).
Collapse
Affiliation(s)
- Ekua W. Brenu
- School of Medicine, University of Notre Dame, Sydney, NSW, Australia
| | - Mark Harris
- Endocrinology Department, Queensland Children’s Hospital, South Brisbane, QLD, Australia
| | - Emma E. Hamilton-Williams
- Frazer Institute, The University of Queensland, Woolloongabba, QLD, Australia
- *Correspondence: Emma E. Hamilton-Williams,
| |
Collapse
|
7
|
Uribe-Querol E, Rosales C. Neutrophils Actively Contribute to Obesity-Associated Inflammation and Pathological Complications. Cells 2022; 11:1883. [PMID: 35741012 PMCID: PMC9221045 DOI: 10.3390/cells11121883] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 02/01/2023] Open
Abstract
Obesity is characterized by an increase in body weight associated with an exaggerated enlargement of the adipose tissue. Obesity has serious negative effects because it is associated with multiple pathological complications such as type 2 diabetes mellitus, cardiovascular diseases, cancer, and COVID-19. Nowadays, 39% of the world population is obese or overweight, making obesity the 21st century epidemic. Obesity is also characterized by a mild, chronic, systemic inflammation. Accumulation of fat in adipose tissue causes stress and malfunction of adipocytes, which then initiate inflammation. Next, adipose tissue is infiltrated by cells of the innate immune system. Recently, it has become evident that neutrophils, the most abundant leukocytes in blood, are the first immune cells infiltrating the adipose tissue. Neutrophils then get activated and release inflammatory factors that recruit macrophages and other immune cells. These immune cells, in turn, perpetuate the inflammation state by producing cytokines and chemokines that can reach other parts of the body, creating a systemic inflammatory condition. In this review, we described the recent findings on the role of neutrophils during obesity and the initiation of inflammation. In addition, we discuss the involvement of neutrophils in the generation of obesity-related complications using diabetes as a prime example.
Collapse
Affiliation(s)
- Eileen Uribe-Querol
- Laboratorio de Biología del Desarrollo, División de Estudios de Posgrado e Investigación, Facultad de Odontología, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | - Carlos Rosales
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
8
|
Long-Term GAD-alum Treatment Effect on Different T-Cell Subpopulations in Healthy Children Positive for Multiple Beta Cell Autoantibodies. J Immunol Res 2022; 2022:3532685. [PMID: 35664355 PMCID: PMC9159828 DOI: 10.1155/2022/3532685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/14/2022] [Accepted: 05/06/2022] [Indexed: 11/21/2022] Open
Abstract
Objective The objective of this study was to explore whether recombinant GAD65 conjugated hydroxide (GAD-alum) treatment affected peripheral blood T-cell subpopulations in healthy children with multiple beta cell autoantibodies. Method The Diabetes Prevention–Immune Tolerance 2 (DiAPREV-IT 2) clinical trial enrolled 26 children between 4 and 13 years of age, positive for glutamic acid decarboxylase autoantibody (GADA) and at least one other autoantibody (insulin, insulinoma antigen-2, or zinc transporter 8 autoantibody (IAA, IA-2A, or ZnT8A)) at baseline. The children were randomized to two doses of subcutaneously administered GAD-alum treatment or placebo, 30 days apart. Complete blood count (CBC) and immunophenotyping of T-cell subpopulations by flow cytometry were performed regularly during the 24 months of follow-up posttreatment. Cross-sectional analyses were performed comparing lymphocyte and T-cell subpopulations between GAD-alum and placebo-treated subjects. Results GAD-alum-treated children had lower levels of lymphocytes (109 cells/L) (p = 0.006), T-cells (103 cells/μL) (p = 0.008), T-helper cells (103 cells/μL) (p = 0.014), and cytotoxic T-cells (103 cells/μL) (p = 0.023) compared to the placebo-treated children 18 months from first GAD-alum injection. This difference remained 24 months after the first treatment for lymphocytes (p = 0.027), T-cells (p = 0.022), T-helper cells (p = 0.048), and cytotoxic T-cells (p = 0.018). Conclusion Our findings suggest that levels of total T-cells and T-cell subpopulations declined 18 and 24 months after GAD-alum treatment in healthy children with multiple beta-cell autoantibodies including GADA.
Collapse
|
9
|
Popp SK, Vecchio F, Brown DJ, Fukuda R, Suzuki Y, Takeda Y, Wakamatsu R, Sarma MA, Garrett J, Giovenzana A, Bosi E, Lafferty AR, Brown KJ, Gardiner EE, Coupland LA, Thomas HE, Chong BH, Parish CR, Battaglia M, Petrelli A, Simeonovic CJ. Circulating platelet-neutrophil aggregates characterize the development of type 1 diabetes in humans and NOD mice. JCI Insight 2022; 7:153993. [PMID: 35076023 PMCID: PMC8855805 DOI: 10.1172/jci.insight.153993] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 12/01/2021] [Indexed: 12/19/2022] Open
Abstract
Platelet-neutrophil aggregates (PNAs) facilitate neutrophil activation and migration and could underpin the recruitment of neutrophils to the pancreas during type 1 diabetes (T1D) pathogenesis. PNAs, measured by flow cytometry, were significantly elevated in the circulation of autoantibody-positive (Aab+) children and new-onset T1D children, as well as in pre-T1D (at 4 weeks and 10–12 weeks) and T1D-onset NOD mice, compared with relevant controls, and PNAs were characterized by activated P-selectin+ platelets. PNAs were similarly increased in pre-T1D and T1D-onset NOD isolated islets/insulitis, and immunofluorescence staining revealed increased islet-associated neutrophil extracellular trap (NET) products (myeloperoxidase [MPO] and citrullinated histones [CitH3]) in NOD pancreata. In vitro, cell-free histones and NETs induced islet cell damage, which was prevented by the small polyanionic drug methyl cellobiose sulfate (mCBS) that binds to histones and neutralizes their pathological effects. Elevated circulating PNAs could, therefore, act as an innate immune and pathogenic biomarker of T1D autoimmunity. Platelet hyperreactivity within PNAs appears to represent a previously unrecognized hematological abnormality that precedes T1D onset. In summary, PNAs could contribute to the pathogenesis of T1D and potentially function as a pre-T1D diagnostic.
Collapse
Affiliation(s)
- Sarah K. Popp
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| | - Federica Vecchio
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Debra J. Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| | - Riho Fukuda
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Yuri Suzuki
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Yuma Takeda
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Rikako Wakamatsu
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Tokyo Medical and Dental University, Bunkyo City, Tokyo, Japan
| | - Mahalakshmi A. Sarma
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| | - Jessica Garrett
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Anna Giovenzana
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Emanuele Bosi
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
- San Raffaele Vita Salute University, Milan, Italy
| | - Antony R.A. Lafferty
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Department of Pediatrics, The Canberra Hospital, Canberra, Australia
| | - Karen J. Brown
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
- Department of Pediatrics, The Canberra Hospital, Canberra, Australia
| | - Elizabeth E. Gardiner
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Lucy A. Coupland
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Helen E. Thomas
- St. Vincent’s Institute of Medical Research, Melbourne, Australia
| | - Beng H. Chong
- Hematology Research Unit, St. George and Sutherland Clinical School, University of New South Wales, Sydney, Australia
| | - Christopher R. Parish
- ACRF Department of Cancer Biology and Therapeutics, The John Curtin School of Medical Research, ANU, Canberra, Australia
| | - Manuela Battaglia
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Alessandra Petrelli
- San Raffaele Diabetes Research Institute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Charmaine J. Simeonovic
- Department of Immunology and Infectious Disease, The John Curtin School of Medical Research, The Australian National University (ANU), Canberra, Australia
| |
Collapse
|
10
|
Zhong J, Mao X, Li H, Shen G, Cao X, He N, Wang J, Xu L, Chen J, Song X, Liu S, Zhang X, Shen Y, Wang LL, Xiang C, Chen YY. Single-cell RNA sequencing analysis reveals the relationship of bone marrow and osteopenia in STZ-induced type 1 diabetic mice. J Adv Res 2022; 41:145-158. [PMID: 36328744 PMCID: PMC9637485 DOI: 10.1016/j.jare.2022.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/30/2021] [Accepted: 01/14/2022] [Indexed: 11/30/2022] Open
Abstract
scRNA-seq analysis reveals the profiles of bone marrow cells in STZ-induced T1D mice. scRNA-seq analysis reveals the heterogeneity of bone marrow immune cells in STZ-induced T1D mice. The ratio of BM-neutrophils to B lymphocytes is increased in the bone marrow of STZ-induced T1D mice. Osteopenia is occurred in STZ-induced T1D mice. This increased ratio is negatively correlated with osteopenia in STZ-induced T1D mice.
Introduction Type 1 diabetes (T1D) is a multifactorial autoimmune disease. Broad knowledge about the genetics, epidemiology and clinical management of T1D has been achieved, but understandings about the cell varieties in the bone marrow during T1D remain limited. Objectives We aimed to present a profile of the bone marrow cells and reveal the relationship of bone marrow and osteopenia in streptozotocin (STZ)-induced T1D mice. Methods The whole bone marrow cells from the femurs and tibias of healthy (group C) and STZ-induced T1D mice (group D) were collected for single-cell RNA sequencing analysis. Single-cell flow cytometry and immunohistochemistry were performed to confirm the proportional changes among bone marrow neutrophils (BM-neutrophils) (Cxcr2+, Ly6g+) and B lymphocytes (Cd19+). X-ray and micro-CT were performed to detect bone mineral density. The correlation between the ratio of BM-neutrophils/B lymphocytes and osteopenia in STZ-induced T1D mice was analyzed by nonparametric Spearman correlation analysis. Results The bone marrow cells in groups C and D were divided into 12 clusters, and 249 differentially expressed genes were found. The diversity of CD45+ immune cells between groups C and D were greatly affected: the proportion of BM-neutrophils showed a significant increase while the proportion of B lymphocytes in group D showed a significant decrease. X-ray and micro-CT analyses confirmed that osteopenia occurred in group D mice. In addition, the results of single-cell flow cytometry and correlation analysis showed that the ratio of BM-neutrophils/B lymphocytes negatively correlated with osteopenia in STZ-induced T1D mice. Conclusion A single-cell RNA sequencing analysis revealed the profile and heterogeneity of bone marrow immune cells in STZ-induced T1D mice for the first time. The ratio of BM-neutrophils/B lymphocytes negatively correlated with osteopenia in STZ-induced T1D mice, which may enhance understanding for treating T1D and preventing T1D-induced osteopenia.
Collapse
Affiliation(s)
- Jinjie Zhong
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xingjia Mao
- Department of Orthopedic, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Heyangzi Li
- Department of Basic Medicine Sciences, and Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Gerong Shen
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xi Cao
- Department of Basic Medicine Sciences, and Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Ning He
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jingyu Wang
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Lintao Xu
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jun Chen
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xinghui Song
- Core Facilities, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Shuangshuang Liu
- Core Facilities, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoming Zhang
- Department of Basic Medicine Sciences, and Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yueliang Shen
- Department of Basic Medicine Sciences, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lin-Lin Wang
- Department of Basic Medicine Sciences, and Department of Orthopaedics of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Chuan Xiang
- Department of Orthopedic, The Second Hospital of Shanxi Medical University, Taiyuan 030001, China.
| | - Ying-Ying Chen
- Department of Basic Medicine Sciences, and Department of Obstetrics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
11
|
Huang J, Pearson JA, Wong FS, Wen L, Zhou Z. Innate immunity in latent autoimmune diabetes in adults. Diabetes Metab Res Rev 2022; 38:e3480. [PMID: 34156143 PMCID: PMC8813511 DOI: 10.1002/dmrr.3480] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 11/06/2022]
Abstract
Latent autoimmune diabetes in adults (LADA) is an autoimmune disease that shares some genetic, immunological and clinical features with both type 1 diabetes and type 2 diabetes. Immune cells including CD4+ T cells, CD8+ T cells, B cells, macrophages and dendritic cells (DCs) have been detected in the pancreas of patients with LADA and a rat model of LADA. Therefore, similar to type 1 diabetes, the pathogenesis of LADA may be caused by interactions between islet β-cells and innate and adaptive immune cells. However, the role of the immunity in the initiation and progression of LADA remains largely unknown. In this review, we have summarized the potential roles of innate immunity and immune-modulators in LADA development. Furthermore, we have examined the evidence and discussed potential innate immunological reasons for the slower development of LADA compared with type 1 diabetes. More in-depth mechanistic studies are needed to fully elucidate the roles of innate immune-associated genes, molecules and cells in their contributions to LADA pathogenesis. Undertaking these studies will greatly enhance the development of new strategies and optimization of current strategies for the diagnosis and treatment of the disease.
Collapse
Affiliation(s)
- Juan 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, Central South University, Changsha, Hunan, China
- Department of Internal Medicine, Section of Endocrinology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | | | - F. Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Li Wen
- Department of Internal Medicine, Section of Endocrinology, School of Medicine, Yale University, New Haven, Connecticut, USA
| | - 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, Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Brænne I, Onengut-Gumuscu S, Chen R, Manichaikul AW, Rich SS, Chen WM, Farber CR. Dynamic changes in immune gene co-expression networks predict development of type 1 diabetes. Sci Rep 2021; 11:22651. [PMID: 34811390 PMCID: PMC8609030 DOI: 10.1038/s41598-021-01840-z] [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] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 11/01/2021] [Indexed: 01/13/2023] Open
Abstract
Significant progress has been made in elucidating genetic risk factors influencing Type 1 diabetes (T1D); however, features other than genetic variants that initiate and/or accelerate islet autoimmunity that lead to the development of clinical T1D remain largely unknown. We hypothesized that genetic and environmental risk factors can both contribute to T1D through dynamic alterations of molecular interactions in physiologic networks. To test this hypothesis, we utilized longitudinal blood transcriptomic profiles in The Environmental Determinants of Diabetes in the Young (TEDDY) study to generate gene co-expression networks. In network modules that contain immune response genes associated with T1D, we observed highly dynamic differences in module connectivity in the 600 days (~ 2 years) preceding clinical diagnosis of T1D. Our results suggest that gene co-expression is highly plastic and that connectivity differences in T1D-associated immune system genes influence the timing and development of clinical disease.
Collapse
Affiliation(s)
- Ingrid Brænne
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA
| | - Suna Onengut-Gumuscu
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
| | - Ruoxi Chen
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA
| | - Ani W Manichaikul
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
| | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Wei-Min Chen
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA
| | - Charles R Farber
- Center for Public Health Genomics, University of Virginia, P.O. Box 800717, Charlottesville, VA, 22908, USA.
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, 22908, USA.
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
13
|
Huang J, Tan Q, Tai N, Pearson JA, Li Y, Chao C, Zhang L, Peng J, Xing Y, Zhang L, Hu Y, Zhou Z, Wong FS, Wen L. IL-10 Deficiency Accelerates Type 1 Diabetes Development via Modulation of Innate and Adaptive Immune Cells and Gut Microbiota in BDC2.5 NOD Mice. Front Immunol 2021; 12:702955. [PMID: 34394099 PMCID: PMC8362616 DOI: 10.3389/fimmu.2021.702955] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/05/2021] [Indexed: 01/02/2023] Open
Abstract
Type 1 diabetes is an autoimmune disease caused by T cell-mediated destruction of insulin-producing β cells. BDC2.5 T cells in BDC2.5 CD4+ T cell receptor transgenic Non-Obese Diabetic (NOD) mice (BDC2.5+ NOD mice) can abruptly invade the pancreatic islets resulting in severe insulitis that progresses rapidly but rarely leads to spontaneous diabetes. This prevention of diabetes is mediated by T regulatory (Treg) cells in these mice. In this study, we investigated the role of interleukin 10 (IL-10) in the inhibition of diabetes in BDC2.5+ NOD mice by generating Il-10-deficient BDC2.5+ NOD mice (BDC2.5+Il-10-/- NOD mice). Our results showed that BDC2.5+Il-10-/- NOD mice displayed robust and accelerated diabetes development. Il-10 deficiency in BDC2.5+ NOD mice promoted the generation of neutrophils in the bone marrow and increased the proportions of neutrophils in the periphery (blood, spleen, and islets), accompanied by altered intestinal immunity and gut microbiota composition. In vitro studies showed that the gut microbiota from BDC2.5+Il-10-/- NOD mice can expand neutrophil populations. Moreover, in vivo studies demonstrated that the depletion of endogenous gut microbiota by antibiotic treatment decreased the proportion of neutrophils. Although Il-10 deficiency in BDC2.5+ NOD mice had no obvious effects on the proportion and function of Treg cells, it affected the immune response and activation of CD4+ T cells. Moreover, the pathogenicity of CD4+ T cells was much increased, and this significantly accelerated the development of diabetes when these CD4+ T cells were transferred into immune-deficient NOD mice. Our study provides novel insights into the role of IL-10 in the modulation of neutrophils and CD4+ T cells in BDC2.5+ NOD mice, and suggests important crosstalk between gut microbiota and neutrophils in type 1 diabetes development.
Collapse
Affiliation(s)
- Juan 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, Central South University, Changsha, China
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Qiyuan Tan
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Department of Endocrinology and Metabolism, Shanghai Jiaotong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Ningwen Tai
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - James Alexander Pearson
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Yangyang Li
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Department of Endocrinology, The Second Hospital of Jilin University, Changchun, China
| | - Chen Chao
- 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, Central South University, Changsha, China
| | - Lucy Zhang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Jian Peng
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - Yanpeng Xing
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Luyao Zhang
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
- Department of Gastrointestinal Surgery, The First Hospital of Jilin University, Changchun, China
| | - Youjia Hu
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| | - 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, Central South University, Changsha, China
| | - F. Susan Wong
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Li Wen
- Section of Endocrinology, Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT, United States
| |
Collapse
|
14
|
Salami F, N.Tamura R, Elding Larsson H, Lernmark Å, Törn C. Complete blood counts with red blood cell determinants associate with reduced beta-cell function in seroconverted Swedish TEDDY children. Endocrinol Diabetes Metab 2021; 4:e00251. [PMID: 34277975 PMCID: PMC8279594 DOI: 10.1002/edm2.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To investigate whether changes in complete blood count (CBC) in islet autoantibody positive children with increased genetic risk for type 1 diabetes are associated with oral glucose tolerance tests (OGTT) and HbA1c over time. METHODS The Environmental Determinants of Diabetes in the Young (TEDDY) study follows children with increased risk for type 1 diabetes in the United States, Germany, Sweden and Finland. In the current study, 89 Swedish TEDDY children (median age 8.8 years) positive for one or multiple islet autoantibodies were followed up to 5 (median 2.3) years for CBC, OGTT and HbA1c. A statistical mixed effect model was used to investigate the association between CBC and OGTT or HbA1c. RESULTS HbA1c over time increased by the number of autoantibodies (p < .001). Reduction in mean corpuscular haemoglobin (MCH) and mean cell volume (MCV) was both associated with an increase in HbA1c (p < .001). A reduction in red blood cell (RBC) counts (p = .003), haemoglobin (p = .002) and haematocrit (p = .006) levels was associated with increased fasting glucose. Increased red blood cells, haemoglobin, haematocrit and MCH but decreased levels of red blood cell distribution widths (RDW) were all associated with increased fasting insulin. CONCLUSIONS The decrease in RBC indices with increasing HbA1c and the decrease in RBC and its parameters with increasing fasting glucose in seroconverted children may reflect an insidious deterioration in glucose metabolism associated with islet beta-cell autoimmunity.
Collapse
Affiliation(s)
- Falastin Salami
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Roy N.Tamura
- Health Informatics InstituteDepartment of PediatricsUniversity of South FloridaTampaFloridaUSA
| | - Helena Elding Larsson
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Åke Lernmark
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Carina Törn
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | | |
Collapse
|
15
|
Neutrophil elastase triggers the development of autoimmune diabetes by exacerbating innate immune responses in pancreatic islets of non-obese diabetic mice. Clin Sci (Lond) 2020; 134:1679-1696. [DOI: 10.1042/cs20200021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 06/16/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022]
Abstract
Abstract
Type 1 diabetes is an autoimmune disease resulted from self-destruction of insulin-producing pancreatic β cells. However, the pathological pathways that trigger the autoimmune destruction remain poorly understood. Clinical studies have demonstrated close associations of neutrophils and neutrophil elastase (NE) with β-cell autoimmunity in patients with Type 1 diabetes. The present study aims to investigate the impact of NE inhibition on development of autoimmune diabetes in NOD mice. NE pharmacological inhibitor (sivelestat) or biological inhibitor (elafin) was supplemented into NOD mice to evaluate their effects on islet inflammation and diabetogenesis. The impact of NE inhibition on innate and adaptive immune cells was measured with flow cytometry and immunohistochemistry. A significant but transient increase in neutrophil infiltration accompanied with elevated NE activity was observed in the neonatal period of NOD mice. Treatment of NOD mice with sivelestat or elafin at the early age led to a marked reduction in spontaneous development of insulitis and autoimmune diabetes. Mechanistically, inhibition of NE significantly attenuated infiltration of macrophages and islet inflammation, thus ameliorating cytotoxic T cell-mediated autoimmune attack of pancreatic β cells. In vitro studies showed that NE directly induced inflammatory responses in both min6 β cells and RAW264.7 macrophages, and promoted macrophage migration. These findings support an important role of NE in triggering the onset and progression of β-cell autoimmunity, and suggest that pharmacological inhibition of NE may represent a promising therapeutic strategy for treatment of autoimmune diabetes.
Collapse
|
16
|
Nigi L, Maccora C, Dotta F, Sebastiani G. From immunohistological to anatomical alterations of human pancreas in type 1 diabetes: New concepts on the stage. Diabetes Metab Res Rev 2020; 36:e3264. [PMID: 31850667 DOI: 10.1002/dmrr.3264] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 12/01/2019] [Accepted: 12/11/2019] [Indexed: 12/14/2022]
Abstract
The histological analysis of human pancreatic samples in type 1 diabetes (T1D) has been proven essential to move forward in the evaluation of in situ events characterizing T1D. Increasing availability of pancreatic tissues collected from diabetic multiorgan donors by centralized biorepositories, which have shared tissues among researchers in the field, has allowed a deeper understanding of T1D pathophysiology, using novel immunohistological and high-throughput methods. In this review, we provide a comprehensive update of the main recent advancements in the characterization of cellular and molecular events involving endocrine and exocrine pancreas as well as the immune system in the onset and progression of T1D. Additionally, we underline novel elements, which provide evidence that T1D pathological changes affect not only islet β-cells but also the entire pancreas.
Collapse
Affiliation(s)
- Laura Nigi
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Carla Maccora
- UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Francesco Dotta
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
- UOC Diabetologia, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Guido Sebastiani
- Diabetes Unit, Department of Medicine, Surgery and Neurosciences, University of Siena, Siena, Italy
| |
Collapse
|
17
|
HLA class II genotyping of admixed Brazilian patients with type 1 diabetes according to self-reported color/race in a nationwide study. Sci Rep 2020; 10:6628. [PMID: 32313169 PMCID: PMC7170860 DOI: 10.1038/s41598-020-63322-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 03/20/2020] [Indexed: 11/22/2022] Open
Abstract
The HLA region is responsible for almost 50% of the genetic risk of type 1 diabetes (T1D). However, haplotypes and their effects on risk or protection vary among different ethnic groups, mainly in an admixed population. We aimed to evaluate the HLA class II genetic profile of Brazilian individuals with T1D and its relationship with self-reported color/race. This was a nationwide multicenter study conducted in 10 Brazilian cities. We included 1,019 T1D individuals and 5,116 controls matched for the region of birth and self-reported color/race. Control participants belonged to the bone marrow transplant donor registry of Brazil (REDOME). HLA-class II alleles (DRB1, DQA1, and DQB1) were genotyped using the SSO and NGS methods. The most frequent risk and protection haplotypes were HLA~DRB1*03:01~DQA1*05:01 g~DQB1*02:01 (OR 5.8, p < 0.00001) and HLA~DRB1*07:01~DQA1*02:01~DQB1*02:02 (OR 0.54, p < 0.0001), respectively, regardless of self-reported color/race. Haplotypes HLA~DRB1*03:01~DQA1*05:01 g~DQB1*02:01 and HLA~DRB1*04:02~DQA1*03:01 g~DQB1*03:02 were more prevalent in the self-reported White group than in the Black group (p = 0.04 and p = 0.02, respectively). The frequency of haplotype HLA~DRB1*09:01~DQA1*03:01 g~DQB1*02:02 was higher in individuals self-reported as Black than White (p = <0.00001). No difference between the Brazilian geographical regions was found. Individuals with T1D presented differences in frequencies of haplotypes within self-reported color/race, but the more prevalent haplotypes, regardless of self-reported color/race, were the ones described previously in Europeans. We hypothesize that, in the T1D population of Brazil, although highly admixed, the disease risk alleles come mostly from Europeans as a result of centuries of colonization and migration.
Collapse
|
18
|
Battaglia M, Petrelli A, Vecchio F. Neutrophils and type 1 diabetes: current knowledge and suggested future directions. Curr Opin Endocrinol Diabetes Obes 2019; 26:201-206. [PMID: 31157631 DOI: 10.1097/med.0000000000000485] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE OF REVIEW Purpose of this review is to describe the most recent human studies on neutrophils in type 1 diabetes (T1D) and to focus on the key questions that still need to be addressed. RECENT FINDINGS Recent evidences demonstrate that neutrophils have marked abnormalities in phenotype and function and play a central role in initiation and perpetuation of aberrant immune responses and organ damage in various systemic autoimmune diseases such as lupus erythematosus and rheumatoid arthritis. In T1D, we have recently demonstrated that reduced circulating neutrophil numbers precede and accompany the disease and that neutrophils infiltrate the pancreas and extrude neutrophil extracellular traps already before the onset of clinical symptoms. However, few other evidences of alterations in neutrophil phenotype and function have been reported in humans, especially in the T1D presymptomatic phases. SUMMARY Dissecting the pathogenic role of these cells in human T1D is crucial for a better understanding of the disease and to open new therapeutic opportunities.
Collapse
Affiliation(s)
- Manuela Battaglia
- San Raffaele Diabetes Research Institute, IRCCS Ospedale, San Raffaele, Milan, Italy
| | | | | |
Collapse
|