1401
|
Martinell M, Pingel R, Hallqvist J, Dorkhan M, Groop L, Rosengren A, Storm P, Stålhammar J. Education, immigration and income as risk factors for hemoglobin A1c >70 mmol/mol when diagnosed with type 2 diabetes or latent autoimmune diabetes in adult: a population-based cohort study. BMJ Open Diabetes Res Care 2017; 5:e000346. [PMID: 28761648 PMCID: PMC5530247 DOI: 10.1136/bmjdrc-2016-000346] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/10/2017] [Accepted: 03/14/2017] [Indexed: 12/30/2022] Open
Abstract
OBJECTIVES The aim of this research is to study education, income and immigration as risk factors for high hemoglobin A1c (HbA1c >70 mmol/mol (8.6%)) when diagnosed with type 2 diabetes (T2D) or latent autoimmune diabetes in the adult (LADA). RESEARCH DESIGN AND METHODS Patients were included from the All New Diabetics in Scania study (2008-2013). Level of education, disposable income and immigration year were retrieved from the longitudinal integrated database for labour market research (LISA) register compiled by Statistics Sweden. Logistic regression models were used to estimate ORs for HbA1c >70 mmol/mol (8.6%) at diagnosis. RESULTS A total of 3794 patients with incident T2D (n=3 525) or LADA (n=269) were included. Patients with T2D with a low (≤9 years) or medium (10-12 years) levels of education were more likely to have high HbA1c at diagnosis compared with patients with T2D with a high (>12 years) level of education (OR 1.34, 95% CI 1.08 to1.66, OR 1.26, 95% CI 1.03 to 1.54). Low-income patients with T2D (<60% of median) were more likely to have high HbA1c at diagnosis compared with high-income patients withT2D (>150% of median) (OR 1.35, 95% CI 1.02 to 1.79). CONCLUSIONS Patients with lower levels of education or low income and are more likely to have HbA1c is >70 mmol/mol (8.6%) when diagnosed with T2D. An understanding of how socioeconomic position influences the clinical presentation at diagnosis may facilitate screening programs designed to target populations at risk for delayed diagnosis.
Collapse
Affiliation(s)
- Mats Martinell
- Family Medicine and Preventive Medicine, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Ronnie Pingel
- Family Medicine and Preventive Medicine, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
- Department of Statistics, Uppsala University, Uppsala, Sweden
| | - Johan Hallqvist
- Family Medicine and Preventive Medicine, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| | - Mozhgan Dorkhan
- Lund University Diabetes Centre, Lunds Universitet, Malm, Sweden
| | - Leif Groop
- Lund University Diabetes Centre, Lunds Universitet, Malm, Sweden
| | - Anders Rosengren
- Lund University Diabetes Centre, Lunds Universitet, Malm, Sweden
| | - Petter Storm
- Lund University Diabetes Centre, Lunds Universitet, Malm, Sweden
| | - Jan Stålhammar
- Family Medicine and Preventive Medicine, Department of Public Health and Caring Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
1402
|
Qaisar N, Lin S, Ryan G, Yang C, Oikemus SR, Brodsky MH, Bortell R, Mordes JP, Wang JP. A Critical Role for the Type I Interferon Receptor in Virus-Induced Autoimmune Diabetes in Rats. Diabetes 2017; 66:145-157. [PMID: 27999109 PMCID: PMC5204313 DOI: 10.2337/db16-0462] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/01/2016] [Indexed: 12/11/2022]
Abstract
The pathogenesis of human type 1 diabetes, characterized by immune-mediated damage of insulin-producing β-cells of pancreatic islets, may involve viral infection. Essential components of the innate immune antiviral response, including type I interferon (IFN) and IFN receptor-mediated signaling pathways, are candidates for determining susceptibility to human type 1 diabetes. Numerous aspects of human type 1 diabetes pathogenesis are recapitulated in the LEW.1WR1 rat model. Diabetes can be induced in LEW.1WR1 weanling rats challenged with virus or with the viral mimetic polyinosinic:polycytidylic acid (poly I:C). We hypothesized that disrupting the cognate type I IFN receptor (type I IFN α/β receptor [IFNAR]) to interrupt IFN signaling would prevent or delay the development of virus-induced diabetes. We generated IFNAR1 subunit-deficient LEW.1WR1 rats using CRISPR-Cas9 (clustered regularly interspaced short palindromic repeats-associated protein 9) genome editing and confirmed functional disruption of the Ifnar1 gene. IFNAR1 deficiency significantly delayed the onset and frequency of diabetes and greatly reduced the intensity of insulitis after poly I:C treatment. The occurrence of Kilham rat virus-induced diabetes was also diminished in IFNAR1-deficient animals. These findings firmly establish that alterations in innate immunity influence the course of autoimmune diabetes and support the use of targeted strategies to limit or prevent the development of type 1 diabetes.
Collapse
Affiliation(s)
- Natasha Qaisar
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Suvana Lin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Glennice Ryan
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Chaoxing Yang
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Sarah R Oikemus
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Michael H Brodsky
- Department of Molecular, Cell, and Cancer Biology, University of Massachusetts Medical School, Worcester, MA
| | - Rita Bortell
- Department of Molecular Medicine, University of Massachusetts Medical School, Worcester, MA
| | - John P Mordes
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| | - Jennifer P Wang
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
1403
|
Andrade EF, Lima ARV, Nunes IE, Orlando DR, Gondim PN, Zangeronimo MG, Alves FHF, Pereira LJ. Exercise and Beta-Glucan Consumption (Saccharomyces cerevisiae) Improve the Metabolic Profile and Reduce the Atherogenic Index in Type 2 Diabetic Rats (HFD/STZ). Nutrients 2016; 8:nu8120792. [PMID: 27999319 PMCID: PMC5188447 DOI: 10.3390/nu8120792] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Revised: 11/24/2016] [Accepted: 11/29/2016] [Indexed: 02/07/2023] Open
Abstract
Physical activity and the ingestion of dietary fiber are non-drug alternatives commonly used as adjuvants to glycemic control in diabetic individuals. Among these fibers, we can highlight beta-glucans. However, few studies have compared isolated and synergic effects of physical exercise and beta-glucan ingestion, especially in type 2 diabetic rats. Therefore, we evaluated the effects beta-glucan (Saccharomyces cerevisiae) consumption, associated or not to exercise, on metabolic parameters of diabetic Wistar rats. The diabetes mellitus (DM) was induced by high-fat diet (HFD) associated with a low dose of streptozotocin (STZ-35 mg/kg). Trained groups were submitted to eight weeks of exercise in aquatic environment. In the last 28 days of experiment, animals received 30 mg/kg/day of beta-glucan by gavage. Isolated use of beta-glucan decreased glucose levels in fasting, Glycated hemoglobin (HbA1c), triglycerides (TAG), total cholesterol (TC), low-density lipoprotein (LDL-C), the atherogenic index of plasma. Exercise alone also decreased blood glucose levels, HbA1c, and renal lesions. An additive effect for reducing the atherogenic index of plasma and renal lesions was observed when both treatments were combined. It was concluded that both beta-glucan and exercise improved metabolic parameters in type 2 (HFD/STZ) diabetic rats.
Collapse
MESH Headings
- Animals
- Atherosclerosis/blood
- Atherosclerosis/diagnosis
- Atherosclerosis/etiology
- Atherosclerosis/prevention & control
- Biomarkers/blood
- Blood Glucose/metabolism
- Combined Modality Therapy
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/complications
- Diabetes Mellitus, Experimental/diagnosis
- Diabetes Mellitus, Experimental/therapy
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/therapy
- Diabetic Angiopathies/blood
- Diabetic Angiopathies/diagnosis
- Diabetic Angiopathies/etiology
- Diabetic Angiopathies/prevention & control
- Diabetic Nephropathies/etiology
- Diabetic Nephropathies/prevention & control
- Diet, High-Fat
- Dietary Fiber/administration & dosage
- Dietary Supplements
- Exercise Therapy
- Glycated Hemoglobin/metabolism
- Lipids/blood
- Male
- Rats, Wistar
- Saccharomyces cerevisiae/metabolism
- Streptozocin
- beta-Glucans/administration & dosage
- beta-Glucans/isolation & purification
Collapse
Affiliation(s)
- Eric Francelino Andrade
- Department of Veterinary Medicine, Federal University of Lavras, Mail Box 3037, Lavras 37200-000, Brazil.
| | | | - Ingrid Edwiges Nunes
- Department of Animal Sciences, Federal University of Lavras, Mail Box 3037, Lavras 37200-000, Brazil.
| | - Débora Ribeiro Orlando
- Department of Agricultural Sciences, Federal University of Jequitinhonha and Mucuri Valleys, Rua Vereador João Narciso, 1380-Bairro Cachoeira, Unaí 3861-000, Brazil.
| | - Paula Novato Gondim
- Department of Veterinary Medicine, Federal University of Lavras, Mail Box 3037, Lavras 37200-000, Brazil.
| | | | | | - Luciano José Pereira
- Department of Health Sciences, Federal University of Lavras, Mail Box 3037, Lavras 37200-000, Brazil.
| |
Collapse
|
1404
|
Evia-Viscarra ML, Guardado-Mendoza R, Rodea-Montero ER. Clinical and Metabolic Characteristics among Mexican Children with Different Types of Diabetes Mellitus. PLoS One 2016; 11:e0168377. [PMID: 27992493 PMCID: PMC5161364 DOI: 10.1371/journal.pone.0168377] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 11/29/2016] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Current classification of diabetes mellitus (DM) is based on etiology and includes type 1 (T1DM), type 2 (T2DM), gestational, and other. Clinical and pathophysiological characteristics of T1DM and T2DM in the same patient have been designated as type 1.5 DM (T1.5DM). OBJECTIVES The aim of this study was to classify pediatric patients with DM based on pancreatic autoimmunity and the presence or absence of overweight/obesity, and to compare the clinical, anthropometric, and biochemical characteristics between children in the different classes of DM. METHODS A sample of 185 patients, recruited (March 2008-April 2015) as part of the Cohort of Mexican Children with DM (CMC-DM); ClinicalTrials.gov, identifier: NCT02722655. The DM classification was made considering pancreatic autoimmunity (via antibodies GAD-65, IAA, and AICA) and the presence or absence of overweight/obesity. Clinical, anthropometric and biochemical variables, grouped by type of DM were compared (Kruskal-Wallis or chi-squared test). RESULTS The final analysis included 140 children; 18.57% T1ADM, 46.43% T1BDM, 12.14% T1.5DM, and 22.86% T2DM. Fasting C-Peptide (FCP), and hs-CRP levels were higher in T1.5DM and T2DM, and the greatest levels were observed in T1.5DM (p<0.001 and 0.024 respectively). CONCLUSIONS We clearly identified that the etiologic mechanisms of T1DM and T2DM are not mutually exclusive, and we detailed why FCP levels are not critical for the classification system of DM in children. The findings of this study suggest that T1.5DM should be considered during the classification of pediatric DM and might facilitate more tailored approaches to treatment, clinical care and follow-up.
Collapse
Affiliation(s)
- María Lola Evia-Viscarra
- Department of Pediatric Endocrinology, Hospital Regional de Alta Especialidad del Bajío, León, Guanajuato, México
| | - Rodolfo Guardado-Mendoza
- Department of Research, Hospital Regional de Alta Especialidad del Bajío, León, Guanajuato, México
| | | |
Collapse
|
1405
|
Seay HR, Yusko E, Rothweiler SJ, Zhang L, Posgai AL, Campbell-Thompson M, Vignali M, Emerson RO, Kaddis JS, Ko D, Nakayama M, Smith MJ, Cambier JC, Pugliese A, Atkinson MA, Robins HS, Brusko TM. Tissue distribution and clonal diversity of the T and B cell repertoire in type 1 diabetes. JCI Insight 2016; 1:e88242. [PMID: 27942583 DOI: 10.1172/jci.insight.88242] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The adaptive immune repertoire plays a critical role in type 1 diabetes (T1D) pathogenesis. However, efforts to characterize B cell and T cell receptor (TCR) profiles in T1D subjects have been largely limited to peripheral blood sampling and restricted to known antigens. To address this, we collected pancreatic draining lymph nodes (pLN), "irrelevant" nonpancreatic draining lymph nodes, peripheral blood mononuclear cells (PBMC), and splenocytes from T1D subjects (n = 18) and control donors (n = 9) as well as pancreatic islets from 1 T1D patient; from these tissues, we collected purified CD4+ conventional T cells (Tconv), CD4+ Treg, CD8+ T cells, and B cells. By conducting high-throughput immunosequencing of the TCR β chain (TRB) and B cell receptor (BCR) immunoglobulin heavy chain (IGH) on these samples, we sought to analyze the molecular signature of the lymphocyte populations within these tissues and of T1D. Ultimately, we observed a highly tissue-restricted CD4+ repertoire, while up to 24% of CD8+ clones were shared among tissues. We surveyed our data set for previously described proinsulin- and glutamic acid decarboxylase 65-reactive (GAD65-reactive) receptors, and interestingly, we observed a TRB with homology to a known GAD65-reactive TCR (clone GAD4.13) present in 7 T1D donors (38.9%), representing >25% of all productive TRB within Tconv isolated from the pLN of 1 T1D subject. These data demonstrate diverse receptor signatures at the nucleotide level and enriched autoreactive clones at the amino acid level, supporting the utility of coupling immunosequencing data with knowledge of characterized autoreactive receptors.
Collapse
Affiliation(s)
- Howard R Seay
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Erik Yusko
- Adaptive Biotechnologies Corporation, Seattle, Washington, USA
| | - Stephanie J Rothweiler
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Lin Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Martha Campbell-Thompson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Marissa Vignali
- Adaptive Biotechnologies Corporation, Seattle, Washington, USA
| | - Ryan O Emerson
- Adaptive Biotechnologies Corporation, Seattle, Washington, USA
| | - John S Kaddis
- Department of Information Sciences, City of Hope National Medical Center, Duarte, California, USA
| | - Dave Ko
- Department of Information Sciences, City of Hope National Medical Center, Duarte, California, USA
| | | | - Mia J Smith
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alberto Pugliese
- Diabetes Research Institute and Departments of Medicine, Microbiology, and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| | - Harlan S Robins
- Adaptive Biotechnologies Corporation, Seattle, Washington, USA.,Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida Diabetes Institute, Gainesville, Florida, USA
| |
Collapse
|
1406
|
Haller MJ, Gitelman SE, Gottlieb PA, Michels AW, Perry DJ, Schultz AR, Hulme MA, Shuster JJ, Zou B, Wasserfall CH, Posgai AL, Mathews CE, Brusko TM, Atkinson MA, Schatz DA. Antithymocyte Globulin Plus G-CSF Combination Therapy Leads to Sustained Immunomodulatory and Metabolic Effects in a Subset of Responders With Established Type 1 Diabetes. Diabetes 2016; 65:3765-3775. [PMID: 27669730 PMCID: PMC5127248 DOI: 10.2337/db16-0823] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/18/2016] [Indexed: 12/17/2022]
Abstract
Low-dose antithymocyte globulin (ATG) plus pegylated granulocyte colony-stimulating factor (G-CSF) preserves β-cell function for at least 12 months in type 1 diabetes. Herein, we describe metabolic and immunological parameters 24 months following treatment. Patients with established type 1 diabetes (duration 4-24 months) were randomized to ATG and pegylated G-CSF (ATG+G-CSF) (N = 17) or placebo (N = 8). Primary outcomes included C-peptide area under the curve (AUC) following a mixed-meal tolerance test (MMTT) and flow cytometry. "Responders" (12-month C-peptide ≥ baseline), "super responders" (24-month C-peptide ≥ baseline), and "nonresponders" (12-month C-peptide < baseline) were evaluated for biomarkers of outcome. At 24 months, MMTT-stimulated AUC C-peptide was not significantly different in ATG+G-CSF (0.49 nmol/L/min) versus placebo (0.29 nmol/L/min). Subjects treated with ATG+G-CSF demonstrated reduced CD4+ T cells and CD4+/CD8+ T-cell ratio and increased CD16+CD56hi natural killer cells (NK), CD4+ effector memory T cells (Tem), CD4+PD-1+ central memory T cells (Tcm), Tcm PD-1 expression, and neutrophils. FOXP3+Helios+ regulatory T cells (Treg) were elevated in ATG+G-CSF subjects at 6, 12, and 18 but not 24 months. Immunophenotyping identified differential HLA-DR expression on monocytes and NK and altered CXCR3 and PD-1 expression on T-cell subsets. As such, a group of metabolic and immunological responders was identified. A phase II study of ATG+G-CSF in patients with new-onset type 1 diabetes is ongoing and may support ATG+G-CSF as a prevention strategy in high-risk subjects.
Collapse
Affiliation(s)
- Michael J Haller
- Department of Pediatrics, University of Florida, Gainesville, FL
| | - Stephen E Gitelman
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA
| | - Peter A Gottlieb
- Department of Pediatrics and Medicine, University of Colorado, Denver, CO
| | - Aaron W Michels
- Department of Pediatrics and Medicine, University of Colorado, Denver, CO
| | - Daniel J Perry
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Andrew R Schultz
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Maigan A Hulme
- Department of Biomedical Engineering, University of Florida, Gainesville, FL
| | - Jonathan J Shuster
- Department of Health Outcomes and Policy, University of Florida, Gainesville, FL
| | - Baiming Zou
- Department of Biostatistics, University of Florida, Gainesville, FL
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Amanda L Posgai
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Mark A Atkinson
- Department of Pediatrics, University of Florida, Gainesville, FL
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Desmond A Schatz
- Department of Pediatrics, University of Florida, Gainesville, FL
| |
Collapse
|
1407
|
Hughes JW, Riddlesworth TD, DiMeglio LA, Miller KM, Rickels MR, McGill JB. Autoimmune Diseases in Children and Adults With Type 1 Diabetes From the T1D Exchange Clinic Registry. J Clin Endocrinol Metab 2016; 101:4931-4937. [PMID: 27676394 PMCID: PMC7530541 DOI: 10.1210/jc.2016-2478] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Type 1 diabetes (T1D) is associated with other autoimmune diseases (AIDs), but the prevalence and associated predictive factors for these comorbidities of T1D across all age groups have not been fully characterized. MATERIALS AND METHODS Data obtained from 25 759 participants with T1D enrolled in the T1D Exchange Registry were used to analyze the types and frequency of AIDs as well as their relationships to gender, age, and race/ethnicity. Diagnoses of autoimmune diseases, represented as ordinal categories (0, 1, 2, 3, or more AIDs) were obtained from medical records of Exchange Registry participants. RESULTS Among the 25 759 T1D Exchange participants, 50% were female, 82% non-Hispanic white, mean age was 23.0 ± 16.9 years and mean duration of diabetes was 11 years. Of these participants, 6876 (27%) were diagnosed with at least one AID. Frequency of two or more AIDs increased from 4.3% in participants aged younger than 13 years to 10.4% in those aged 50 years or older. The most common AIDs were thyroid (6097, 24%), gastrointestinal (1530, 6%), and collagen vascular diseases (432, 2%). Addison's disease was rare (75, 0.3%). The prevalence of one or more AIDs was increased in females and non-Hispanic whites and with older age. CONCLUSIONS In the T1D Exchange Clinic Registry, a diagnosis of one or more AIDs in addition to T1D is common, particularly in women, non-Hispanic whites, and older individuals. Results of this study have implications for both primary care and endocrine practice and will allow clinicians to better anticipate and manage the additional AIDs that develop in patients with T1D.
Collapse
Affiliation(s)
- Jing W Hughes
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Tonya D Riddlesworth
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Linda A DiMeglio
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Kellee M Miller
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Michael R Rickels
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Janet B McGill
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | -
- Department of Medicine, Washington University School of Medicine (J.W.H., J.B.M.), St Louis, Missouri 63110; Jaeb Center for Health Research (T.D.R., K.M.M.), Tampa, Florida 33647; Department of Pediatrics, Indiana University School of Medicine (L.A.D.), Indianapolis, Indiana 46202; and Department of Medicine, Perelman School of Medicine (M.R.R.) University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
1408
|
Matsuhisa M, Koyama M, Cheng X, Sumi M, Riddle MC, Bolli GB, Hirose T. Sustained glycaemic control and less nocturnal hypoglycaemia with insulin glargine 300U/mL compared with glargine 100U/mL in Japanese adults with type 1 diabetes (EDITION JP 1 randomised 12-month trial including 6-month extension). Diabetes Res Clin Pract 2016; 122:133-140. [PMID: 27835765 DOI: 10.1016/j.diabres.2016.10.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/23/2016] [Accepted: 10/03/2016] [Indexed: 01/17/2023]
Abstract
AIMS To evaluate the efficacy and safety of insulin glargine 300U/mL (Gla-300) versus glargine 100U/mL (Gla-100) in adults with type 1 diabetes in Japan over 12months. METHODS EDITION JP 1 was a multicentre, randomised, open-label phase 3 study. Following a 6-month on-treatment period, participants continued to receive Gla-300 or Gla-100 once daily, plus mealtime insulin, over a 6-month open-label extension phase. HbA1c, hypoglycaemia, body weight and adverse events were assessed. RESULTS Overall, 114/122 (93%) and 114/121 (94%) of participants in the Gla-300 and Gla-100 group, respectively, completed the 6-month extension phase. Glycaemic control was sustained in both groups up to month 12 (mean HbA1c: Gla-300, 7.9% [62mmol/mol]; Gla-100, 7.8% [62mmol/mol]). Annualised rates of hypoglycaemia were lower with Gla-300 versus Gla-100; significantly for nocturnal confirmed (<3.0mmol/L [<54mg/dL]) or severe hypoglycaemia (2.39 and 3.85 events per participant-year; rate ratio: 0.62 [0.39-0.97]). No between-treatment differences in mean body weight change or adverse events were observed. CONCLUSION Over 12months' treatment, participants with type 1 diabetes receiving Gla-300 achieved sustained glycaemic control and experienced less nocturnal hypoglycaemia that was confirmed (<3.0mmol/L [<54mg/dL]) or severe compared with Gla-100, supporting the 6-month results.
Collapse
|
1409
|
Paul DS, Teschendorff AE, Dang MA, Lowe R, Hawa MI, Ecker S, Beyan H, Cunningham S, Fouts AR, Ramelius A, Burden F, Farrow S, Rowlston S, Rehnstrom K, Frontini M, Downes K, Busche S, Cheung WA, Ge B, Simon MM, Bujold D, Kwan T, Bourque G, Datta A, Lowy E, Clarke L, Flicek P, Libertini E, Heath S, Gut M, Gut IG, Ouwehand WH, Pastinen T, Soranzo N, Hofer SE, Karges B, Meissner T, Boehm BO, Cilio C, Elding Larsson H, Lernmark Å, Steck AK, Rakyan VK, Beck S, Leslie RD. Increased DNA methylation variability in type 1 diabetes across three immune effector cell types. Nat Commun 2016; 7:13555. [PMID: 27898055 PMCID: PMC5141286 DOI: 10.1038/ncomms13555] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023] Open
Abstract
The incidence of type 1 diabetes (T1D) has substantially increased over the past decade, suggesting a role for non-genetic factors such as epigenetic mechanisms in disease development. Here we present an epigenome-wide association study across 406,365 CpGs in 52 monozygotic twin pairs discordant for T1D in three immune effector cell types. We observe a substantial enrichment of differentially variable CpG positions (DVPs) in T1D twins when compared with their healthy co-twins and when compared with healthy, unrelated individuals. These T1D-associated DVPs are found to be temporally stable and enriched at gene regulatory elements. Integration with cell type-specific gene regulatory circuits highlight pathways involved in immune cell metabolism and the cell cycle, including mTOR signalling. Evidence from cord blood of newborns who progress to overt T1D suggests that the DVPs likely emerge after birth. Our findings, based on 772 methylomes, implicate epigenetic changes that could contribute to disease pathogenesis in T1D.
Collapse
Affiliation(s)
- Dirk S. Paul
- Medical Genomics, UCL Cancer Institute, University College London, London WC1E 6BT, UK
- Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, Cambridge CB1 8RN, UK
| | - Andrew E. Teschendorff
- CAS Key Lab of Computational Biology, CAS-MPG Partner Institute for Computational Biology, Shanghai Institute for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Statistical Cancer Genomics, UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Mary A.N. Dang
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Robert Lowe
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Mohammed I. Hawa
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Simone Ecker
- Medical Genomics, UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Huriya Beyan
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Stephanie Cunningham
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Alexandra R. Fouts
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Anita Ramelius
- Department of Clinical Sciences, Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Frances Burden
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Samantha Farrow
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Sophia Rowlston
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Karola Rehnstrom
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Mattia Frontini
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
| | - Kate Downes
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
| | - Stephan Busche
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Warren A. Cheung
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Bing Ge
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Marie-Michelle Simon
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - David Bujold
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Tony Kwan
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Guillaume Bourque
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Avik Datta
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Ernesto Lowy
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Laura Clarke
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Emanuele Libertini
- Medical Genomics, UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - Simon Heath
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 4, 08028 Barcelona, Spain
- Universitat Pompeu Fabra, Plaça de la Mercè 10, 08002 Barcelona, Spain
| | - Marta Gut
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 4, 08028 Barcelona, Spain
- Universitat Pompeu Fabra, Plaça de la Mercè 10, 08002 Barcelona, Spain
| | - Ivo G Gut
- CNAG-CRG, Centre for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Baldiri Reixac 4, 08028 Barcelona, Spain
- Universitat Pompeu Fabra, Plaça de la Mercè 10, 08002 Barcelona, Spain
| | - Willem H. Ouwehand
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- British Heart Foundation Centre of Excellence, Cambridge Biomedical Campus, Cambridge CB2 0QQ, UK
- Human Genetics, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Tomi Pastinen
- Department of Human Genetics, McGill University, Montreal, Québec, Canada H3A 0G1
- McGill University and Genome Quebec Innovation Centre, Montreal, Québec, Canada H3A 0G1
| | - Nicole Soranzo
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0PT, UK
- Human Genetics, Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge CB10 1SA, UK
| | - Sabine E. Hofer
- Department of Pediatrics, Medical University of Innsbruck, 6020 Innsbruck, Austria
| | - Beate Karges
- Division of Endocrinology and Diabetes, RWTH Aachen University, 52074 Aachen, Germany
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
| | - Thomas Meissner
- German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, University Children's Hospital, Heinrich Heine University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Bernhard O. Boehm
- Division of Endocrinology, Department of Internal Medicine I, Ulm University Medical Centre, 89081 Ulm, Germany
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore
- Imperial College London, London SW7 2AZ, UK
| | - Corrado Cilio
- Department of Clinical Sciences, Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Helena Elding Larsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Åke Lernmark
- Department of Clinical Sciences, Lund University, Skåne University Hospital, SE-20502 Malmö, Sweden
| | - Andrea K. Steck
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Vardhman K. Rakyan
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| | - Stephan Beck
- Medical Genomics, UCL Cancer Institute, University College London, London WC1E 6BT, UK
| | - R. David Leslie
- The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK
| |
Collapse
|
1410
|
Manzoor F, Johnson MC, Li C, Samulski RJ, Wang B, Tisch R. β-cell-specific IL-35 therapy suppresses ongoing autoimmune diabetes in NOD mice. Eur J Immunol 2016; 47:144-154. [PMID: 27859048 DOI: 10.1002/eji.201646493] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 10/03/2016] [Accepted: 11/03/2016] [Indexed: 12/30/2022]
Abstract
IL-35 is a recently identified cytokine exhibiting potent immunosuppressive properties. The therapeutic potential and effects of IL-35 on pathogenic T effector cells (Teff) and Foxp3+ Treg, however, are ill defined. We tested the capacity of IL-35 to suppress ongoing autoimmunity in NOD mice. For this purpose, an adeno-associated virus vector in which IL-35 transgene expression is selectively targeted to β cells via an insulin promoter (AAV8mIP-IL35) was used. AAV8mIP-IL35 vaccination of NOD mice at a late preclinical stage of type 1 diabetes (T1D) suppressed β-cell autoimmunity and prevented diabetes onset. Numbers of islet-resident conventional CD4+ and CD8+ T cells, and DCs were reduced within 4 weeks of AAV8mIP-IL35 treatment. The diminished islet T-cell pool correlated with suppressed proliferation, and a decreased frequency of IFN-γ-expressing Teff. Ectopic IL-35 also reduced islet Foxp3+ Treg numbers and proliferation, and protection was independent of induction/expansion of adaptive islet immunoregulatory T cells. These findings demonstrate that IL-35-mediated suppression is sufficiently robust to block established β-cell autoimmunity, and support the use of IL-35 to treat T1D and other T-cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
- Fatima Manzoor
- Department of Microbiology & Immunology, Chapel Hill, NC, USA
| | - Mark C Johnson
- Department of Microbiology & Immunology, Chapel Hill, NC, USA
| | - Chengwen Li
- Gene Therapy Center, Chapel Hill, NC, USA.,Department of Pharmacology, Chapel Hill, NC, USA
| | - R Jude Samulski
- Gene Therapy Center, Chapel Hill, NC, USA.,Department of Pharmacology, Chapel Hill, NC, USA
| | - Bo Wang
- Department of Microbiology & Immunology, Chapel Hill, NC, USA
| | - Roland Tisch
- Department of Microbiology & Immunology, Chapel Hill, NC, USA.,Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| |
Collapse
|
1411
|
Simmons KM, Alkanani AK, McDaniel KA, Goyne C, Miao D, Zhao Z, Yu L, Michels AW. Islet Autoantibody Measurements from Dried Blood Spots on Filter Paper Strongly Correlate to Serum Levels. PLoS One 2016; 11:e0166213. [PMID: 27846247 PMCID: PMC5112938 DOI: 10.1371/journal.pone.0166213] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 10/19/2016] [Indexed: 01/13/2023] Open
Abstract
Type 1 diabetes (T1D) is increasing in incidence and predictable with measurement of serum islet autoantibodies (iAb) years prior to clinical disease onset. Identifying iAb positive individuals reduces diabetic ketoacidosis and identifies individuals for T1D prevention trials. However, large scale screening for iAb remains challenging as assays have varying sensitivities and specificities, insulin autoantibodies remain difficult to measure and venipuncture is generally required to obtain serum. We developed an approach to reliably measure all four major iAb, including insulin autoantibodies, from dried blood spots (DBS) on filter-paper. By spiking iAb positive serum into iAb negative whole blood in a dose titration, we optimized the conditions for autoantibody elution from filter paper as measured by fluid phase radioimmunoassays. After assessing stability of measuring iAb from DBS over time, we then screened iAb from DBS and the corresponding serum in new-onset T1D (n = 52), and controls (n = 72) which included first-degree relatives of T1D patients. iAb measured from eluted DBS in new-onset T1D strongly correlated with serum measurements (R2 = 0.96 for mIAA, GADA = 0.94, IA-2A = 0.85, ZnT8A = 0.82, p<0.01 for each autoantibody). There were no false positives in control subjects, and 5/6 with previously unknown iAb positivity in sera were detected using DBS. With further validation, measuring iAb from DBS can be a reliable method to screen for T1D risk.
Collapse
Affiliation(s)
- Kimber M. Simmons
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
- * E-mail:
| | - Aimon K. Alkanani
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
| | - Kristen A. McDaniel
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
| | - Christopher Goyne
- Indiana University School of Medicine, Indianapolis, IN, 46202, United States of America
| | - Dongmei Miao
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
| | - Zhiyuan Zhao
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
| | - Aaron W. Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado, Aurora, CO, 80045, United States of America
| |
Collapse
|
1412
|
|
1413
|
Burke SJ, Karlstad MD, Eder AE, Regal KM, Lu D, Burk DH, Collier JJ. Pancreatic β-Cell production of CXCR3 ligands precedes diabetes onset. Biofactors 2016; 42:703-715. [PMID: 27325565 PMCID: PMC5177512 DOI: 10.1002/biof.1304] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 05/18/2016] [Indexed: 12/18/2022]
Abstract
Type 1 diabetes mellitus (T1DM) results from immune cell-mediated reductions in function and mass of the insulin-producing β-cells within the pancreatic islets. While the initial trigger(s) that initiates the autoimmune process is unknown, there is a leukocytic infiltration that precedes islet β-cell death and dysfunction. Herein, we demonstrate that genes encoding the chemokines CXCL9, 10, and 11 are primary response genes in pancreatic β-cells and are also elevated as part of the inflammatory response in mouse, rat, and human islets. We further established that STAT1 participates in the transcriptional control of these genes in response to the pro-inflammatory cytokines IL-1β and IFN-γ. STAT1 is phosphorylated within five minutes after β-cell exposure to IFN-γ, with subsequent occupancy at proximal and distal response elements within the Cxcl9 and Cxcl11 gene promoters. This increase in STAT1 binding is coupled to the rapid appearance of chemokine transcript. Moreover, circulating levels of chemokines that activate CXCR3 are elevated in non-obese diabetic (NOD) mice, consistent with clinical findings in human diabetes. We also report herein that mice with genetic deletion of CXCR3 (receptor for ligands CXCL9, 10, and 11) exhibit a delay in diabetes development after being injected with multiple low doses of streptozotocin. Therefore, we conclude that production of CXCL9, 10, and 11 from islet β-cells controls leukocyte migration and activity into pancreatic tissue, which ultimately influences islet β-cell mass and function. © 2016 BioFactors, 42(6):703-715, 2016.
Collapse
Affiliation(s)
- Susan J. Burke
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Michael D. Karlstad
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN
| | - Adrianna E. Eder
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN
| | - Kellie M. Regal
- Department of Surgery, Graduate School of Medicine, University of Tennessee Health Science Center, Knoxville, TN
| | - Danhong Lu
- Sarah W. Stedman Nutrition and Metabolism Center, Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, NC
| | - David H. Burk
- Cell Biology and Bioimaging Core Facility, Pennington Biomedical Research Center, Baton Rouge, LA
| | - J. Jason Collier
- Laboratory of Islet Biology and Inflammation, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
1414
|
Unnikrishnan R, Shah VN, Mohan V. Challenges in diagnosis and management of diabetes in the young. Clin Diabetes Endocrinol 2016; 2:18. [PMID: 28702252 PMCID: PMC5471766 DOI: 10.1186/s40842-016-0036-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 11/03/2016] [Indexed: 02/07/2023] Open
Abstract
The prevalence of diabetes in children and adolescents is increasing worldwide, with profound implications on the long-term health of individuals, societies, and nations. The diagnosis and management of diabetes in youth presents several unique challenges. Although type 1 diabetes is more common among children and adolescents, the incidence of type 2 diabetes in youth is also on the rise, particularly among certain ethnic groups. In addition, less common types of diabetes such as monogenic diabetes syndromes and diabetes secondary to pancreatopathy (in some parts of the world) need to be accurately identified to initiate the most appropriate treatment. A detailed patient history and physical examination usually provides clues to the diagnosis. However, specific laboratory and imaging tests are needed to confirm the diagnosis. The management of diabetes in children and adolescents is challenging in some cases due to age-specific issues and the more aggressive nature of the disease. Nonetheless, a patient-centered approach focusing on comprehensive risk factor reduction with the involvement of all concerned stakeholders (the patient, parents, peers and teachers) could help in ensuring the best possible level of diabetes control and prevention or delay of long-term complications.
Collapse
Affiliation(s)
- Ranjit Unnikrishnan
- Madras Diabetes Research Foundation & Dr Mohan’s Diabetes Specialties Centre, Who Collaborating Centre for Non-Communicable Diseases Prevention and Control, 4, Conran Smith Road, Gopalapuram, Chennai, 600 086 India
| | - Viral N. Shah
- Barbara Davis Center for Diabetes, University of Colorado Anschutz Campus, Aurora, CO USA
| | - Viswanathan Mohan
- Madras Diabetes Research Foundation & Dr Mohan’s Diabetes Specialties Centre, Who Collaborating Centre for Non-Communicable Diseases Prevention and Control, 4, Conran Smith Road, Gopalapuram, Chennai, 600 086 India
| |
Collapse
|
1415
|
Thabit H, Bally L, Hovorka R. Available at a flash: a new way to check glucose. Lancet 2016; 388:2213-2214. [PMID: 27634582 DOI: 10.1016/s0140-6736(16)31582-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Accepted: 08/26/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Hood Thabit
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Department of Diabetes and Endocrinology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lia Bally
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Department of Diabetes and Endocrinology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK; Division of Diabetes, Endocrinology, Clinical Nutrition and Metabolism, Inselspital, Bern University Hospital, University of Bern, Switzerland
| | - Roman Hovorka
- Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK; Department of Paediatrics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
1416
|
Yang D, Deng H, Luo G, Wu G, Lin S, Yuan L, Xv M, Li S, Zhang X, Wu J, Lang J, Liang G, Lin J, Chen D, Li L, Fang Y, Wu Y, Ou W, Li J, Weng J, Yan J. Demographic and clinical characteristics of patients with type 1 diabetes mellitus: A multicenter registry study in Guangdong, China. J Diabetes 2016; 8:847-853. [PMID: 26663759 DOI: 10.1111/1753-0407.12366] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/27/2015] [Accepted: 12/06/2015] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND A lack of demographic and clinical data hinders efforts of health care providers in China to support patients with type 1 diabetes mellitus (T1D). Therefore, the aim of the present retrospective study was to provide an overview of the demographic and clinical characteristics of Chinese patients with T1D. METHODS Hospital medical records of patients with T1D (diagnosed between January 2000 and December 2011) in 105 secondary and tertiary hospitals across Guangdong province were reviewed. Data were collected on patient age at diagnosis, presentations at onset, physical examination, and diabetes management. RESULTS In all, 3173 patients diagnosed with T1D between January 2000 and December 2011 were included in the study (46.2% female). The median age at diagnosis was 27.5 years (interquartile range [IQR] 18.0-38.0) years and the median body mass index (BMI) at onset was 19.6 kg/m2 (IQR 17.4-21.8 kg/m2 ). Among adult patients, 0.9% were obese, 6.6% were overweight, 62.3% were normal weight, and 30.3 % were underweight. The prevalence of diabetic ketoacidosis (DKA) at onset was 50.1%. The proportion of patients with retinopathy, nephropathy, and neuropathy was 8.1%, 20.7 %, and 11.1%, respectively. CONCLUSION The adult-onset form of T1D is not rare in China. The registry participants were characterized by older age at onset, lower BMI, and a higher prevalence of DKA at onset compared with those in regions with a high incidence of T1D, such as northern Europe. These findings contribute to a better understanding of the heterogeneity of T1D in different populations and so will help healthcare providers to develop management models that are more suitable for these patients.
Collapse
Affiliation(s)
- Daizhi Yang
- Department of Endocrinology & Metabolic Disease, The 3rd Affiliated Hospital of Sun Yat-sen University
- Guangdong Diabetes Center, Guangzhou
| | - Hongrong Deng
- Department of Endocrinology & Metabolic Disease, The 3rd Affiliated Hospital of Sun Yat-sen University
- Guangdong Diabetes Center, Guangzhou
| | - Guochun Luo
- The 1st Affiliated Hospital of Shenzhen University, Shenzen
| | - Ge Wu
- Affiliated Hospital of Guangdong Medical College, Zhanjiang
| | - Shaoda Lin
- The 1st Affiliated Hospital of Shantou University Medical College, Shantou
| | - Lin Yuan
- Zhuhai People's Hospital, Zhuhai
| | - Meilun Xv
- Huizhou Municipal Central Hosptial, Huizhou
| | | | | | | | | | | | | | - Dingyu Chen
- Guangzhou 1st Municipal People's Hospital, Guangzhou
| | - Lu Li
- Shenzhen People's Hospital, Shenzhen
| | | | | | - Wenxin Ou
- Maoming People's Hospital, Maoming, China
| | - Jin Li
- Department of Endocrinology & Metabolic Disease, The 3rd Affiliated Hospital of Sun Yat-sen University
- Guangdong Diabetes Center, Guangzhou
| | - Jianping Weng
- Department of Endocrinology & Metabolic Disease, The 3rd Affiliated Hospital of Sun Yat-sen University
- Guangdong Diabetes Center, Guangzhou
| | - Jinhua Yan
- Department of Endocrinology & Metabolic Disease, The 3rd Affiliated Hospital of Sun Yat-sen University.
- Guangdong Diabetes Center, Guangzhou.
| |
Collapse
|
1417
|
Menon RK, Thomas IH, Sperling MA. Childhood diabetes mellitus: Advances & challenges. Indian J Med Res 2016; 144:641-644. [PMID: 28361813 PMCID: PMC5393071 DOI: 10.4103/ijmr.ijmr_1768_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Indexed: 01/13/2023] Open
Affiliation(s)
- Ram K. Menon
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| | - Inas H. Thomas
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, USA
| | - Mark A. Sperling
- Department of Pediatrics, University of Pittsburgh, Pittsburgh, Ann Arbor, USA
- Department of Pediatrics, Mount Sinai School of Medicine, New York, USA
| |
Collapse
|
1418
|
Ziegler AG, Bonifacio E, Powers AC, Todd JA, Harrison LC, Atkinson MA. Type 1 Diabetes Prevention: A Goal Dependent on Accepting a Diagnosis of an Asymptomatic Disease. Diabetes 2016; 65:3233-3239. [PMID: 27959859 PMCID: PMC5860440 DOI: 10.2337/db16-0687] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/14/2016] [Indexed: 12/13/2022]
Abstract
Type 1 diabetes, a disease defined by absolute insulin deficiency, is considered a chronic autoimmune disorder resulting from the destruction of insulin-producing pancreatic β-cells. The incidence of childhood-onset type 1 diabetes has been increasing at a rate of 3%-5% per year globally. Despite the introduction of an impressive array of therapies aimed at improving disease management, no means for a practical "cure" exist. This said, hope remains high that any of a number of emerging technologies (e.g., continuous glucose monitoring, insulin pumps, smart algorithms), alongside advances in stem cell biology, cell encapsulation methodologies, and immunotherapy, will eventually impact the lives of those with recently diagnosed or established type 1 diabetes. However, efforts aimed at reversing insulin dependence do not address the obvious benefits of disease prevention. Hence, key "stretch goals" for type 1 diabetes research include identifying improved and increasingly practical means for diagnosing the disease at earlier stages in its natural history (i.e., early, presymptomatic diagnosis), undertaking such efforts in the population at large to optimally identify those with presymptomatic type 1 diabetes, and introducing safe and effective therapeutic options for prevention.
Collapse
Affiliation(s)
- Anette-G Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, Neuherberg, Germany
- Forschergruppe Diabetes, Klinikum rechts der Isar, Technische Universität München, München, Germany
| | - Ezio Bonifacio
- DFG-Center for Regenerative Therapies Dresden, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD), Technische Universität Dresden, Dresden, Germany
- Forschergruppe Diabetes e.V., Neuherberg, Germany
| | - Alvin C Powers
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- VA Tennessee Valley Healthcare System, Nashville, TN
| | - John A Todd
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, U.K
| | - Leonard C Harrison
- Walter and Eliza Hall Institute of Medical Research, Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Mark A Atkinson
- Departments of Pathology and Pediatrics, UF Diabetes Institute, University of Florida, Gainesville, FL
| |
Collapse
|
1419
|
Richardson SJ, Rodriguez-Calvo T, Gerling IC, Mathews CE, Kaddis JS, Russell MA, Zeissler M, Leete P, Krogvold L, Dahl-Jørgensen K, von Herrath M, Pugliese A, Atkinson MA, Morgan NG. Islet cell hyperexpression of HLA class I antigens: a defining feature in type 1 diabetes. Diabetologia 2016; 59:2448-2458. [PMID: 27506584 PMCID: PMC5042874 DOI: 10.1007/s00125-016-4067-4] [Citation(s) in RCA: 211] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 07/04/2016] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS Human pancreatic beta cells may be complicit in their own demise in type 1 diabetes, but how this occurs remains unclear. One potentially contributing factor is hyperexpression of HLA class I antigens. This was first described approximately 30 years ago, but has never been fully characterised and was recently challenged as artefactual. Therefore, we investigated HLA class I expression at the protein and RNA levels in pancreases from three cohorts of patients with type 1 diabetes. The principal aims were to consider whether HLA class I hyperexpression is artefactual and, if not, to determine the factors driving it. METHODS Pancreas samples from type 1 diabetes patients with residual insulin-containing islets (n = 26) from the Network for Pancreatic Organ donors with Diabetes (nPOD), Diabetes Virus Detection study (DiViD) and UK recent-onset type 1 diabetes collections were immunostained for HLA class I isoforms, signal transducer and activator of transcription 1 (STAT1), NLR family CARD domain containing 5 (NLRC5) and islet hormones. RNA was extracted from islets isolated by laser-capture microdissection from nPOD and DiViD samples and analysed using gene-expression arrays. RESULTS Hyperexpression of HLA class I was observed in the insulin-containing islets of type 1 diabetes patients from all three tissue collections, and was confirmed at both the RNA and protein levels. The expression of β2-microglobulin (a second component required for the generation of functional HLA class I complexes) was also elevated. Both 'classical' HLA class I isoforms (i.e. HLA-ABC) as well as a 'non-classical' HLA molecule, HLA-F, were hyperexpressed in insulin-containing islets. This hyperexpression did not correlate with detectable upregulation of the transcriptional regulator NLRC5. However, it was strongly associated with increased STAT1 expression in all three cohorts. Islet hyperexpression of HLA class I molecules occurred in the insulin-containing islets of patients with recent-onset type 1 diabetes and was also detectable in many patients with disease duration of up to 11 years, declining thereafter. CONCLUSIONS/INTERPRETATION Islet cell HLA class I hyperexpression is not an artefact, but is a hallmark in the immunopathogenesis of type 1 diabetes. The response is closely associated with elevated expression of STAT1 and, together, these occur uniquely in patients with type 1 diabetes, thereby contributing to their selective susceptibility to autoimmune-mediated destruction.
Collapse
Affiliation(s)
- Sarah J Richardson
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK.
| | | | - Ivan C Gerling
- Department of Medicine, University of Tennessee, Memphis, TN, USA
| | - Clayton E Mathews
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - John S Kaddis
- Department of Information Sciences, City of Hope, Duarte, CA, USA
| | - Mark A Russell
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK
| | - Marie Zeissler
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK
| | - Pia Leete
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK
| | - Lars Krogvold
- Paediatric Department, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Knut Dahl-Jørgensen
- Paediatric Department, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | - Alberto Pugliese
- Diabetes Research Institute, Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Miami Miller School of Medicine, Miami, FL, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mark A Atkinson
- Department of Pathology, University of Florida, Gainesville, FL, USA
| | - Noel G Morgan
- Islet Biology Exeter (IBEx), Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, RILD Building (Level 4), Barrack Road, Exeter, EX2 5DW, UK.
| |
Collapse
|
1420
|
Affiliation(s)
- Zhiyuan Zhao
- 1 Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine , Aurora, Colorado
- 2 Department of Endocrinology, Second Hospital of Jilin University , Changchun, Jilin, China
| | - Liping Yu
- 1 Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine , Aurora, Colorado
| |
Collapse
|
1421
|
Kettner LO, Matthiesen NB, Ramlau-Hansen CH, Kesmodel US, Bay B, Henriksen TB. Fertility treatment and childhood type 1 diabetes mellitus: a nationwide cohort study of 565,116 live births. Fertil Steril 2016; 106:1751-1756. [PMID: 27773424 DOI: 10.1016/j.fertnstert.2016.09.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 08/15/2016] [Accepted: 09/07/2016] [Indexed: 11/26/2022]
Abstract
OBJECTIVE To investigate the association between specific types of fertility treatment and childhood type 1 diabetes mellitus. DESIGN Nationwide birth cohort study. SETTING Not applicable. PATIENT(S) All pregnancies resulting in a live-born singleton child in Denmark from 1995 to 2003. INTERVENTION(S) Not applicable. MAIN OUTCOME MEASURE(S) Childhood type 1 diabetes mellitus identified from redeemed prescriptions for insulin until 2013. RESULT(S) The study included 565,116 singleton pregnancies. A total of 14,985 children were conceived by ovulation induction or intrauterine insemination, and 8,490 children were conceived by in vitro fertilization or intracytoplasmic sperm injection. During the follow-up period, 2,011 (0.4%) children developed type 1 diabetes mellitus. The primary analyses showed no association between fertility treatment and childhood type 1 diabetes mellitus. In secondary analyses, ovulation induction or intrauterine insemination with follicle-stimulating hormone was associated with an increased risk of type 1 diabetes mellitus (hazard ratio 3.22; 95% confidence interval 1.20 to 8.64). No clear associations were seen with other types of fertility treatment or with specific treatment indications. CONCLUSION(S) No association between fertility treatment and childhood type 1 diabetes mellitus was found. Ovulation induction or intrauterine insemination with follicle-stimulating hormone may be associated with an increased risk of childhood type 1 diabetes mellitus. However, this finding may be due to chance or to confounding by indication and thus requires further investigation.
Collapse
Affiliation(s)
- Laura Ozer Kettner
- Perinatal Epidemiology Research Unit and Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark.
| | | | | | - Ulrik Schiøler Kesmodel
- Fertility Clinic, Department of Obstetrics and Gynecology, Herlev University Hospital, Herlev, Denmark
| | - Bjørn Bay
- Fertility Clinic, Department of Obstetrics and Gynecology, Aarhus University, Regional Hospital Horsens, Horsens, Denmark
| | - Tine Brink Henriksen
- Perinatal Epidemiology Research Unit and Department of Pediatrics, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
1422
|
Feng J, Xing W, Xie L. Regulatory Roles of MicroRNAs in Diabetes. Int J Mol Sci 2016; 17:E1729. [PMID: 27763497 PMCID: PMC5085760 DOI: 10.3390/ijms17101729] [Citation(s) in RCA: 102] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Revised: 10/02/2016] [Accepted: 10/09/2016] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs), a class of endogenous small noncoding RNAs in eukaryotes, have been recognized as significant regulators of gene expression through post-transcriptional mechanisms. To date, >2000 miRNAs have been identified in the human genome, and they orchestrate a variety of biological and pathological processes. Disruption of miRNA levels correlates with many diseases, including diabetes mellitus, a complex multifactorial metabolic disorder affecting >400 million people worldwide. miRNAs are involved in the pathogenesis of diabetes mellitus by affecting pancreatic β-cell functions, insulin resistance, or both. In this review, we summarize the investigations of the regulatory roles of important miRNAs in diabetes, as well as the potential of circulating miRNAs as diagnostic markers for diabetes mellitus.
Collapse
Affiliation(s)
- Juan Feng
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China.
| | - Wanli Xing
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China.
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China.
| | - Lan Xie
- Medical Systems Biology Research Center, School of Medicine, Tsinghua University, Beijing 100084, China.
- National Engineering Research Center for Beijing Biochip Technology, Beijing 102206, China.
| |
Collapse
|
1423
|
Thorsen SU, Pipper CB, Mortensen HB, Pociot F, Johannesen J, Svensson J. No Contribution of GAD-65 and IA-2 Autoantibodies around Time of Diagnosis to the Increasing Incidence of Juvenile Type 1 Diabetes: A 9-Year Nationwide Danish Study. Int J Endocrinol 2016; 2016:8350158. [PMID: 27818684 PMCID: PMC5081463 DOI: 10.1155/2016/8350158] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 08/31/2016] [Accepted: 09/14/2016] [Indexed: 12/27/2022] Open
Abstract
Aims. A new perspective on autoantibodies as pivotal players in the pathogenesis of type 1 diabetes (T1D) has recently emerged. Our key objective was to examine whether increased levels of autoantibodies against the β-cell autoantigens glutamic acid decarboxylase (isoform 65) (GADA) and insulinoma associated antigen-2A (IA-2A) mirrored the 3.4% annual increase in incidence of T1D. Methods. From the Danish Childhood Diabetes Register, we randomly selected 500 patients and 500 siblings for GADA and IA-2A analysis (1997 through 2005). Blood samples were taken within three months after onset. A robust log-normal regression model was used. Nine hundred children and adolescents had complete records and were included in the analysis. Cochran-Armitage test for trend was used to evaluate changes in prevalence of autoantibody positivity by period. Results. No significant changes in levels of GADA and IA-2A were found over our 9-year study period. No trends in autoantibody positivity-in either patients or siblings-were found. Levels of GADA and IA-2A were significantly associated with HLA risk groups and GADA with age. Conclusion. The prevalence of positivity and the levels of GADA and IA-2A have not changed between 1997 and 2005 in newly diagnosed patients with T1D and their siblings without T1D.
Collapse
Affiliation(s)
- Steffen U. Thorsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
| | - Christian B. Pipper
- Department of Public Health, Section of Biostatistics, University of Copenhagen, Øster Farimagsgade 5, 1710 Copenhagen K, Denmark
| | - Henrik B. Mortensen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Flemming Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Jesper Johannesen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| | - Jannet Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark
| |
Collapse
|
1424
|
Slattery J, MacFabe DF, Frye RE. The Significance of the Enteric Microbiome on the Development of Childhood Disease: A Review of Prebiotic and Probiotic Therapies in Disorders of Childhood. Clin Med Insights Pediatr 2016; 10:91-107. [PMID: 27774001 PMCID: PMC5063840 DOI: 10.4137/cmped.s38338] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 08/30/2016] [Accepted: 09/05/2016] [Indexed: 02/07/2023] Open
Abstract
Recent studies have highlighted the fact that the enteric microbiome, the trillions of microbes that inhabit the human digestive tract, has a significant effect on health and disease. Methods for manipulating the enteric microbiome, particularly through probiotics and microbial ecosystem transplantation, have undergone some study in clinical trials. We review some of the evidence for microbiome alteration in relation to childhood disease and discuss the clinical trials that have examined the manipulation of the microbiome in an effort to prevent or treat childhood disease with a primary focus on probiotics, prebiotics, and/or synbiotics (ie, probiotics + prebiotics). Studies show that alterations in the microbiome may be a consequence of events occurring during infancy and/or childhood such as prematurity, C-sections, and nosocomial infections. In addition, certain childhood diseases have been associated with microbiome alterations, namely necrotizing enterocolitis, infantile colic, asthma, atopic disease, gastrointestinal disease, diabetes, malnutrition, mood/anxiety disorders, and autism spectrum disorders. Treatment studies suggest that probiotics are potentially protective against the development of some of these diseases. Timing and duration of treatment, the optimal probiotic strain(s), and factors that may alter the composition and function of the microbiome are still in need of further research. Other treatments such as prebiotics, fecal microbial transplantation, and antibiotics have limited evidence. Future translational work, in vitro models, long-term and follow-up studies, and guidelines for the composition and viability of probiotic and microbial therapies need to be developed. Overall, there is promising evidence that manipulating the microbiome with probiotics early in life can help prevent or reduce the severity of some childhood diseases, but further research is needed to elucidate biological mechanisms and determine optimal treatments.
Collapse
Affiliation(s)
- John Slattery
- Arkansas Children’s Research Institute, Little Rock, AR, USA
- Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Derrick F. MacFabe
- The Kilee Patchell-Evans Autism Research Group, Departments of Psychology (Neuroscience) and Psychiatry, Division of Developmental Disabilities, University of Western Ontario, London, ON, Canada
| | - Richard E. Frye
- Arkansas Children’s Research Institute, Little Rock, AR, USA
- Division of Neurology, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
1425
|
Abstract
The gastrointestinal system represents one of the largest interfaces between the human internal microenvironment and the external world. This system harbors trillions of commensal bacteria that reside in symbiosis with the host. Intestinal bacteria play a crucial role in maintaining systemic and intestinal immune and metabolic homeostasis because of their effect on nutrient absorption and immune development and function. Recently, altered gut bacterial composition (dysbiosis) was hypothesized to be involved in mechanisms through which islet autoimmunity is triggered. Evidence from animal models indicates that alterations in the gut bacterial composition precede disease onset, thus implicating a causal role for the gut microbiome in islet destruction. However, it remains unclear whether dysbiosis is directly linked to the mechanisms of human type 1 diabetes (T1D). In this review, we discuss data implicating the gut microbiota in disease progression with an emphasis on our recent studies performed in humans and in rodent models of T1D.
Collapse
Affiliation(s)
- James C Needell
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO, 80045, USA
| | - Danny Zipris
- Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, 1775 Aurora Ct., Mail Stop B-140, Aurora, CO, 80045, USA.
| |
Collapse
|
1426
|
Abstract
Type 1 diabetes (T1D) results from the immune-mediated destruction of insulin-producing β cells located within the pancreatic islets of Langerhans. The autoimmune process leads to a deficiency in insulin production and resultant hyperglycemia requiring lifelong treatment with insulin administration. T1D continues to dramatically increase in incidence, especially in young children. Substantial knowledge surrounding human disease pathogenesis exists, such that T1D is now predictable with the measurement of antibodies in the peripheral blood directed against insulin and other β cell proteins. With the ability to predict, it naturally follows that T1D should be preventable. As such, over the last two decades, numerous well-controlled clinical trials have been completed attempting to prevent diabetes onset or maintain residual β cell function after clinical onset, all providing relatively disappointing results. Here, we review the T1D prevention efforts, the current landscape of clinical therapies, and end with a discussion regarding the future outlook for preventing T1D.
Collapse
Affiliation(s)
- Kimber M Simmons
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Peter A Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Aaron W Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
1427
|
Maffeis C, Martina A, Corradi M, Quarella S, Nori N, Torriani S, Plebani M, Contreas G, Felis GE. Association between intestinal permeability and faecal microbiota composition in Italian children with beta cell autoimmunity at risk for type 1 diabetes. Diabetes Metab Res Rev 2016; 32:700-709. [PMID: 26891226 DOI: 10.1002/dmrr.2790] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Revised: 12/10/2015] [Accepted: 01/19/2016] [Indexed: 12/29/2022]
Abstract
BACKGROUND Pancreatic organ-specific autoimmunity in subjects at risk for type 1 diabetes (T1D) is associated with increased intestinal permeability and an aberrant gut microbiota, but these factors have not yet been simultaneously investigated in the same subjects. Thus, the aim of this study was to assess both intestinal permeability and gut microbiota composition in an Italian sample of children at risk for T1D. METHODS Ten Italian children with beta cell autoimmunity at risk for T1D and 10 healthy children were involved in a case-control study. The lactulose/mannitol test was used to assess intestinal permeability. Analysis of microbiota composition was performed using polymerase chain reaction followed by denaturing gradient gel electrophoresis, based on the 16S rRNA gene. RESULTS Intestinal permeability was significantly higher in children at risk for T1D than in healthy controls. Moreover, the gut microbiota of the former differed from that of the latter group: Three microorganisms were detected - Dialister invisus, Gemella sanguinis and Bifidobacterium longum - in association with the pre-pathologic state. CONCLUSIONS The results of this study validated the hypothesis that increased intestinal permeability together with differences in microbiota composition are contemporaneously associated with the pre-pathological condition of T1D in a sample of Italian children. Further studies are necessary to confirm the microbial markers identified in this sample of children as well as to clarify the involvement of microbiota modifications in the mechanisms leading to increased permeability and the autoimmune mechanisms that promote diabetes onset. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Claudio Maffeis
- Unit of Pediatric Diabetes and Metabolic Diseases, Regional Center for Pediatric Diabetes, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy.
| | - Alessia Martina
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Massimiliano Corradi
- Unit of Pediatric Diabetes and Metabolic Diseases, Regional Center for Pediatric Diabetes, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Sara Quarella
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Nicole Nori
- Unit of Pediatric Diabetes and Metabolic Diseases, Regional Center for Pediatric Diabetes, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Sandra Torriani
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Mario Plebani
- Department of Laboratory Medicine, University of Padua, Padua, Italy
| | - Giovanna Contreas
- Unit of Pediatric Diabetes and Metabolic Diseases, Regional Center for Pediatric Diabetes, Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, University of Verona, Verona, Italy
| | - Giovanna E Felis
- Department of Biotechnology, University of Verona, Verona, Italy
| |
Collapse
|
1428
|
Todd JA, Evangelou M, Cutler AJ, Pekalski ML, Walker NM, Stevens HE, Porter L, Smyth DJ, Rainbow DB, Ferreira RC, Esposito L, Hunter KMD, Loudon K, Irons K, Yang JH, Bell CJM, Schuilenburg H, Heywood J, Challis B, Neupane S, Clarke P, Coleman G, Dawson S, Goymer D, Anselmiova K, Kennet J, Brown J, Caddy SL, Lu J, Greatorex J, Goodfellow I, Wallace C, Tree TI, Evans M, Mander AP, Bond S, Wicker LS, Waldron-Lynch F. Regulatory T Cell Responses in Participants with Type 1 Diabetes after a Single Dose of Interleukin-2: A Non-Randomised, Open Label, Adaptive Dose-Finding Trial. PLoS Med 2016; 13:e1002139. [PMID: 27727279 PMCID: PMC5058548 DOI: 10.1371/journal.pmed.1002139] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 08/25/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Interleukin-2 (IL-2) has an essential role in the expansion and function of CD4+ regulatory T cells (Tregs). Tregs reduce tissue damage by limiting the immune response following infection and regulate autoreactive CD4+ effector T cells (Teffs) to prevent autoimmune diseases, such as type 1 diabetes (T1D). Genetic susceptibility to T1D causes alterations in the IL-2 pathway, a finding that supports Tregs as a cellular therapeutic target. Aldesleukin (Proleukin; recombinant human IL-2), which is administered at high doses to activate the immune system in cancer immunotherapy, is now being repositioned to treat inflammatory and autoimmune disorders at lower doses by targeting Tregs. METHODS AND FINDINGS To define the aldesleukin dose response for Tregs and to find doses that increase Tregs physiologically for treatment of T1D, a statistical and systematic approach was taken by analysing the pharmacokinetics and pharmacodynamics of single doses of subcutaneous aldesleukin in the Adaptive Study of IL-2 Dose on Regulatory T Cells in Type 1 Diabetes (DILT1D), a single centre, non-randomised, open label, adaptive dose-finding trial with 40 adult participants with recently diagnosed T1D. The primary endpoint was the maximum percentage increase in Tregs (defined as CD3+CD4+CD25highCD127low) from the baseline frequency in each participant measured over the 7 d following treatment. There was an initial learning phase with five pairs of participants, each pair receiving one of five pre-assigned single doses from 0.04 × 106 to 1.5 × 106 IU/m2, in order to model the dose-response curve. Results from each participant were then incorporated into interim statistical modelling to target the two doses most likely to induce 10% and 20% increases in Treg frequencies. Primary analysis of the evaluable population (n = 39) found that the optimal doses of aldesleukin to induce 10% and 20% increases in Tregs were 0.101 × 106 IU/m2 (standard error [SE] = 0.078, 95% CI = -0.052, 0.254) and 0.497 × 106 IU/m2 (SE = 0.092, 95% CI = 0.316, 0.678), respectively. On analysis of secondary outcomes, using a highly sensitive IL-2 assay, the observed plasma concentrations of the drug at 90 min exceeded the hypothetical Treg-specific therapeutic window determined in vitro (0.015-0.24 IU/ml), even at the lowest doses (0.040 × 106 and 0.045 × 106 IU/m2) administered. A rapid decrease in Treg frequency in the circulation was observed at 90 min and at day 1, which was dose dependent (mean decrease 11.6%, SE = 2.3%, range 10.0%-48.2%, n = 37), rebounding at day 2 and increasing to frequencies above baseline over 7 d. Teffs, natural killer cells, and eosinophils also responded, with their frequencies rapidly and dose-dependently decreased in the blood, then returning to, or exceeding, pretreatment levels. Furthermore, there was a dose-dependent down modulation of one of the two signalling subunits of the IL-2 receptor, the β chain (CD122) (mean decrease = 58.0%, SE = 2.8%, range 9.8%-85.5%, n = 33), on Tregs and a reduction in their sensitivity to aldesleukin at 90 min and day 1 and 2 post-treatment. Due to blood volume requirements as well as ethical and practical considerations, the study was limited to adults and to analysis of peripheral blood only. CONCLUSIONS The DILT1D trial results, most notably the early altered trafficking and desensitisation of Tregs induced by a single ultra-low dose of aldesleukin that resolves within 2-3 d, inform the design of the next trial to determine a repeat dosing regimen aimed at establishing a steady-state Treg frequency increase of 20%-50%, with the eventual goal of preventing T1D. TRIAL REGISTRATION ISRCTN Registry ISRCTN27852285; ClinicalTrials.gov NCT01827735.
Collapse
Affiliation(s)
- John A. Todd
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (FWL); (JAT)
| | - Marina Evangelou
- Department of Mathematics, Imperial College London, London, United Kingdom
| | - Antony J. Cutler
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Marcin L. Pekalski
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Neil M. Walker
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Helen E. Stevens
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Linsey Porter
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Deborah J. Smyth
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Daniel B. Rainbow
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Ricardo C. Ferreira
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Laura Esposito
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Kara M. D. Hunter
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Kevin Loudon
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Kathryn Irons
- National Institute for Health Research Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Jennie H. Yang
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King’s College London, National Institute of Health Research Biomedical Research Centre, Guy’s and St Thomas’ National Health Service Foundation Trust and King’s College London, London, United Kingdom
| | - Charles J. M. Bell
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Helen Schuilenburg
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - James Heywood
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Ben Challis
- Wellcome Trust/MRC Institute of Metabolic Science, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Sankalpa Neupane
- Wellcome Trust/MRC Institute of Metabolic Science, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Pamela Clarke
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Gillian Coleman
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Sarah Dawson
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Donna Goymer
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Katerina Anselmiova
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Jane Kennet
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Judy Brown
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Sarah L. Caddy
- Division of Virology, Department of Pathology, Addenbrooke’s Hospital, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Jia Lu
- Division of Virology, Department of Pathology, Addenbrooke’s Hospital, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Jane Greatorex
- Public Health England, Clinical Microbiology and Public Health Laboratory, Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Ian Goodfellow
- Division of Virology, Department of Pathology, Addenbrooke’s Hospital, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Chris Wallace
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
- MRC Biostatistics Unit Hub for Trials Methodology Research, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Tim I. Tree
- Department of Immunobiology, Faculty of Life Sciences & Medicine, King’s College London, National Institute of Health Research Biomedical Research Centre, Guy’s and St Thomas’ National Health Service Foundation Trust and King’s College London, London, United Kingdom
| | - Mark Evans
- Wellcome Trust/MRC Institute of Metabolic Science, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Adrian P. Mander
- MRC Biostatistics Unit Hub for Trials Methodology Research, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Simon Bond
- National Institute for Health Research Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge Biomedical Campus, Cambridge, United Kingdom
- MRC Biostatistics Unit Hub for Trials Methodology Research, Cambridge Institute of Public Health, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Linda S. Wicker
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
| | - Frank Waldron-Lynch
- JDRF/Wellcome Trust Diabetes and Inflammation Laboratory, Department of Medical Genetics, Cambridge Institute for Medical Research, National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, United Kingdom
- * E-mail: (FWL); (JAT)
| |
Collapse
|
1429
|
Abstract
The receptor for advanced glycation end products (RAGE) is a novel protein increasingly studied in the pathogenesis of type 1 diabetes (T1D). RAGE is expressed by several immune cell types, including T cells, antigen-presenting cells, endothelial cells, and the endocrine cells of the pancreatic islets. RAGE binds various ligands including advanced glycation end products (AGEs), high-mobility group box protein 1 (HMGB1), S100 proteins, β-amyloid, β-sheet fibrils, and lipopolysaccharide. AGEs are a particularly interesting ligand because their exogenous introduction into the body can be accelerated by the consumption of AGE-rich processed foods. This review will detail RAGE isoforms and its ligands and discuss how RAGE binding on the aforementioned cells could be linked to T1D pathogenesis.
Collapse
Affiliation(s)
- Sherman S Leung
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Josephine M Forbes
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia.
- Mater Clinical School, School of Medicine, The University of Queensland, Brisbane, Queensland, Australia.
| | - Danielle J Borg
- Glycation and Diabetes, Mater Research Institute, Translational Research Institute, The University of Queensland, 37 Kent St, Woolloongabba, Brisbane, Queensland, Australia
| |
Collapse
|
1430
|
Klee P, Bosco D, Guérardel A, Somm E, Toulotte A, Maechler P, Schwitzgebel VM. Activation of Nicotinic Acetylcholine Receptors Decreases Apoptosis in Human and Female Murine Pancreatic Islets. Endocrinology 2016; 157:3800-3808. [PMID: 27471776 DOI: 10.1210/en.2015-2057] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Type 1 diabetes (T1DM) results from destruction of most insulin-secreting pancreatic β-cells. The persistence of β-cells decades after the onset of the disease indicates that the resistance of individual cells to the autoimmune insult is heterogeneous and might depend on the metabolic status of a cell at a given moment. The aim of this study is to investigate whether activation of nicotinic acetylcholine receptors (nACh-Rs) could increase β-cell resistance against the adverse environment prevailing at the onset of T1DM. Here, we show that nACh-R activation by nicotine and choline, 2 agonists of the receptor, decreases murine and human β-cell apoptosis induced by proinflammatory cytokines known to be present in the islet environment at the onset of T1DM. The protective mechanism activated by nicotine and choline involves attenuation of mitochondrial outer membrane permeabilization via modulation of endoplasmic reticulum stress, of the activity of B-cell lymphoma 2 family proteins and cytoplasmic calcium levels. Local inflammation and endoplasmic reticulum stress being key determinants of β-cell death in T1DM, we conclude that pharmacological activation of nACh-R could represent a valuable therapeutic option in the modulation of β-cell death in T1DM.
Collapse
Affiliation(s)
- Philippe Klee
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Domenico Bosco
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Audrey Guérardel
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Emmanuel Somm
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Audrey Toulotte
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Pierre Maechler
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| | - Valérie M Schwitzgebel
- Service of Development and Growth (P.K., A.G., E.S., A.T., V.S.), Department of Pediatrics, University Hospital of Geneva and Diabetes Center, University of Geneva, 1211 Geneva, Switzerland; Cell Isolation and Transplantation Center (D.B.), Department of Surgery, University Hospital of Geneva and University of Geneva, 1205 Geneva, Switzerland; and Department of Cell Physiology and Metabolism (P.M.), Geneva University Medical Center, 1205 Geneva, Switzerland
| |
Collapse
|
1431
|
Fouts A, Pyle L, Yu L, Miao D, Michels A, Krischer J, Sosenko J, Gottlieb P, Steck AK. Do Electrochemiluminescence Assays Improve Prediction of Time to Type 1 Diabetes in Autoantibody-Positive TrialNet Subjects? Diabetes Care 2016; 39:1738-44. [PMID: 27456836 PMCID: PMC5033080 DOI: 10.2337/dc16-0302] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Accepted: 06/28/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To explore whether electrochemiluminescence (ECL) assays can help improve prediction of time to type 1 diabetes in the TrialNet autoantibody-positive population. RESEARCH DESIGN AND METHODS TrialNet subjects who were positive for one or more autoantibodies (microinsulin autoantibody, GAD65 autoantibody [GADA], IA-2A, and ZnT8A) with available ECL-insulin autoantibody (IAA) and ECL-GADA data at their initial visit were analyzed; after a median follow-up of 24 months, 177 of these 1,287 subjects developed diabetes. RESULTS Univariate analyses showed that autoantibodies by radioimmunoassays (RIAs), ECL-IAA, ECL-GADA, age, sex, number of positive autoantibodies, presence of HLA DR3/4-DQ8 genotype, HbA1c, and oral glucose tolerance test (OGTT) measurements were all significantly associated with progression to diabetes. Subjects who were ECL positive had a risk of progression to diabetes within 6 years of 58% compared with 5% for the ECL-negative subjects (P < 0.0001). Multivariate Cox proportional hazards models were compared, with the base model including age, sex, OGTT measurements, and number of positive autoantibodies by RIAs. The model with positivity for ECL-GADA and/or ECL-IAA was the best, and factors that remained significantly associated with time to diabetes were area under the curve (AUC) C-peptide, fasting C-peptide, AUC glucose, number of positive autoantibodies by RIAs, and ECL positivity. Adding ECL to the Diabetes Prevention Trial risk score (DPTRS) improved the receiver operating characteristic curves with AUC of 0.83 (P < 0.0001). CONCLUSIONS ECL assays improved the ability to predict time to diabetes in these autoantibody-positive relatives at risk for developing diabetes. These findings might be helpful in the design and eligibility criteria for prevention trials in the future.
Collapse
Affiliation(s)
- Alexandra Fouts
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Laura Pyle
- Department of Pediatrics, University of Colorado Denver, Aurora, CO Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO
| | - Liping Yu
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Dongmei Miao
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Aaron Michels
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Jeffrey Krischer
- Pediatrics Epidemiology Center, University of South Florida, Tampa, FL
| | - Jay Sosenko
- University of Miami School of Medicine, Miami, FL
| | - Peter Gottlieb
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Andrea K Steck
- Barbara Davis Center for Childhood Diabetes, University of Colorado School of Medicine, Aurora, CO
| | | |
Collapse
|
1432
|
Li J, Karunananthan J, Pelham B, Kandeel F. Imaging pancreatic islet cells by positron emission tomography. World J Radiol 2016; 8:764-774. [PMID: 27721939 PMCID: PMC5039672 DOI: 10.4329/wjr.v8.i9.764] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Revised: 04/15/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023] Open
Abstract
It was estimated that every year more than 30000 persons in the United States - approximately 80 people per day - are diagnosed with type 1 diabetes (T1D). T1D is caused by autoimmune destruction of the pancreatic islet (β cells) cells. Islet transplantation has become a promising therapy option for T1D patients, while the lack of suitable tools is difficult to directly evaluate of the viability of the grafted islet over time. Positron emission tomography (PET) as an important non-invasive methodology providing high sensitivity and good resolution, is able to accurate detection of the disturbed biochemical processes and physiological abnormality in living organism. The successful PET imaging of islets would be able to localize the specific site where transplanted islets engraft in the liver, and to quantify the level of islets remain alive and functional over time. This information would be vital to establishing and evaluating the efficiency of pancreatic islet transplantation. Many novel imaging agents have been developed to improve the sensitivity and specificity of PET islet imaging. In this article, we summarize the latest developments in carbon-11, fluorine-18, copper-64, and gallium-68 labeled radioligands for the PET imaging of pancreatic islet cells.
Collapse
|
1433
|
Insights from lncRNAs Profiling of MIN6 Beta Cells Undergoing Inflammation. Mediators Inflamm 2016; 2016:9275106. [PMID: 27698546 PMCID: PMC5028877 DOI: 10.1155/2016/9275106] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 07/12/2016] [Accepted: 07/27/2016] [Indexed: 01/19/2023] Open
Abstract
Type 1 diabetes mellitus (T1DM) is an organ-specific autoimmune disease characterized by chronic and progressive apoptotic destruction of pancreatic beta cells. During the initial phases of T1DM, cytokines and other inflammatory mediators released by immune cells progressively infiltrate islet cells, induce alterations in gene expression, provoke functional impairment, and ultimately lead to apoptosis. Long noncoding RNAs (lncRNAs) are a new important class of pervasive genes that have a variety of biological functions and play key roles in many diseases. However, whether they have a function in cytokine-induced beta cell apoptosis is still uncertain. In this study, lncRNA microarray technology was used to identify the differently expressed lncRNAs and mRNAs in MIN6 cells exposed to proinflammatory cytokines. Four hundred forty-four upregulated and 279 downregulated lncRNAs were detected with a set filter fold-change ≧2.0. To elucidate the potential functions of these lncRNAs, Gene Ontology (GO) and pathway analyses were used to evaluate the potential functions of differentially expressed lncRNAs. Additionally, a lncRNA-mRNA coexpression network was constructed to predict the interactions between the most strikingly regulated lncRNAs and mRNAs. This study may be utilized as a background or reference resource for future functional studies on lncRNAs related to the diagnosis and development of new therapies for T1DM.
Collapse
|
1434
|
Østergaard JA, Laugesen E, Leslie RD. Should There be Concern About Autoimmune Diabetes in Adults? Current Evidence and Controversies. Curr Diab Rep 2016; 16:82. [PMID: 27457237 DOI: 10.1007/s11892-016-0780-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Autoimmune diabetes has a heterogeneous phenotype. Although often considered a condition starting in childhood, a substantial proportion of type 1 diabetes presents in adult life. This holds important implications for our understanding of the factors that modify the rate of progression through the disease prodrome to clinical diabetes and for our management of the disease. When autoimmune diabetes develops in adulthood, insulin treatment is often not required at the time of diagnosis, and this autoimmune non-insulin requiring diabetes is generally termed latent autoimmune diabetes in adults (LADA). Patients with LADA are generally leaner, younger at diabetes onset; have a greater reduction in C-peptide; and have a greater likelihood of insulin treatment as compared with patients with type 2 diabetes. The LADA subset of patients with adult-onset autoimmune diabetes has highlighted many shortcomings in the classification of diabetes and invokes the case for more personalized data analysis in line with the move towards precision medicine. Perhaps most importantly, the issues highlight our persistent failure to engage with the heterogeneity within the most common form of autoimmune diabetes, that is adult-onset type 1 diabetes, both insulin-dependent and initially non-insulin requiring (LADA). This review discusses characteristics of autoimmune diabetes and specifically aims to illustrate the heterogeneity of the disease.
Collapse
Affiliation(s)
- Jakob Appel Østergaard
- Department of Clinical Medicine, Aarhus University & Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus & The Danish Diabetes Academy, Odense, Denmark
| | - Esben Laugesen
- Department of Clinical Medicine, Aarhus University & Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus & The Danish Diabetes Academy, Odense, Denmark
| | | |
Collapse
|
1435
|
Jacobsen R, Thorsen SU, Cohen AS, Lundqvist M, Frederiksen P, Pipper CB, Pociot F, Thygesen LC, Ascherio A, Svensson J, Heitmann BL. Neonatal vitamin D status is not associated with later risk of type 1 diabetes: results from two large Danish population-based studies. Diabetologia 2016; 59:1871-81. [PMID: 27241183 DOI: 10.1007/s00125-016-4002-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Accepted: 05/05/2016] [Indexed: 12/19/2022]
Abstract
AIMS/HYPOTHESIS The aim of this work was to assess whether neonatal levels of 25-hydroxyvitamin D (25(OH)D) are associated with risk of developing type 1 diabetes before the age of 18 years. METHODS Two large-scale studies with different designs-a case-cohort and a case-control-were conducted using Danish national register data and biobank material. Weighted Cox regression and conditional logistic regression were used to calculate HRs and ORs, respectively. The concentration of 25(OH)D was assessed from neonatal dried blood spots using highly sensitive liquid chromatography-tandem mass spectrometry. Quintiles of 25(OH)D3 were used in the main analyses. RESULTS The case-cohort study included 912 type 1 diabetes cases and 2866 individuals without type 1 diabetes born in Denmark between 1981 and 2002 and followed up until the end of 2012. The case-control study included 527 matched case-control pairs born between 1981 and 1999 and followed up until May 2004. Both studies found no association between 25(OH)D3 levels and later risk of developing type 1 diabetes. The neonatal total 25(OH)D levels in the studies were low: 46% (case-cohort study) and 51% (case-control study) of individuals had 25(OH)D levels <25 nmol/l. CONCLUSIONS/INTERPRETATION Our two large-scale national studies showed that 25(OH)D3 levels around the time of birth were not associated with later type 1 diabetes risk. Whether higher levels of 25(OH)D3 during pregnancy, acquired by higher doses of supplementation than are recommended today in most countries, could protect the offspring against type 1 diabetes cannot be ruled out by the present studies.
Collapse
Affiliation(s)
- Ramune Jacobsen
- Research Unit for Dietary Studies at The Parker Institute, Copenhagen University Hospital, Bispebjerg og Frederiksberg, Nordre Fasanvej 57, Hovedvejen, Entrance 5, Ground Floor, 2000, Frederiksberg, Denmark.
- The Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, Frederiksberg, Denmark.
| | - Steffen U Thorsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arieh S Cohen
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
| | - Marika Lundqvist
- Department of Congenital Disorders, Statens Serum Institute, Copenhagen, Denmark
| | - Peder Frederiksen
- Research Unit for Dietary Studies at The Parker Institute, Copenhagen University Hospital, Bispebjerg og Frederiksberg, Nordre Fasanvej 57, Hovedvejen, Entrance 5, Ground Floor, 2000, Frederiksberg, Denmark
| | - Christian B Pipper
- Department of Public Health, Section of Biostatistics, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lau C Thygesen
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
| | - Alberto Ascherio
- Department of Nutrition, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Jannet Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev, Denmark
- Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Berit L Heitmann
- Research Unit for Dietary Studies at The Parker Institute, Copenhagen University Hospital, Bispebjerg og Frederiksberg, Nordre Fasanvej 57, Hovedvejen, Entrance 5, Ground Floor, 2000, Frederiksberg, Denmark
- The Institute of Preventive Medicine, Bispebjerg and Frederiksberg Hospital, The Capital Region, Frederiksberg, Denmark
- National Institute of Public Health, University of Southern Denmark, Copenhagen, Denmark
- The Boden Institute of Obesity, Nutrition Exercise and Eating Disorders, Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- Institute of Public Health, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
1436
|
Affiliation(s)
| | - Guang Ning
- Ruijin Hospital, Shanghai Jiaotong University.
| |
Collapse
|
1437
|
Perakakis N, Mantzoros CS. Immune therapy in type 1 diabetes mellitus - Attempts to untie the Gordian knot? Metabolism 2016; 65:1278-1285. [PMID: 27506735 DOI: 10.1016/j.metabol.2016.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/25/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Nikolaos Perakakis
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215.
| | - Christos S Mantzoros
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; USA Section of Endocrinology, Boston VA Healthcare System, Harvard Medical School, Boston, MA 02130, USA
| |
Collapse
|
1438
|
Nie X, Wang N, Chen Y, Chen C, Han B, Zhu C, Chen Y, Xia F, Cang Z, Lu M, Meng Y, Jiang B, D Jensen M, Lu Y. Blood cadmium in Chinese adults and its relationships with diabetes and obesity. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2016; 23:18714-23. [PMID: 27312901 DOI: 10.1007/s11356-016-7078-2] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/13/2016] [Indexed: 05/18/2023]
Abstract
The aim of this study is to evaluate blood cadmium levels (BCLs) in Chinese adults and explore whether blood cadmium is associated with diabetes or obesity. This study included 5544 adults from a cross-sectional SPECT-China study. BCL and blood lead level (BLL) was measured by atomic absorption spectrometry. Fasting plasma glucose (FPG) was used to define prediabetes and diabetes. Overweight and obesity were defined by body mass index (BMI). The associations of BCL with prediabetes, diabetes, overweight, and obesity were analyzed by multinomial logistic regression analyses. Medians (interquartile range) of BCL were 1.97 μg/L (0.60-3.82) in men and 1.59 μg/L (0.54-3.51) in women. Subjects in low-economic-status areas and urban areas had significantly higher BCL. BCL in current smokers was significantly higher than in current non-smokers. In the adjusted model, a mild positive relationship between BCL and FPG was found. Meanwhile, the prevalence of prediabetes was increased according to the increase in BCL tertiles. Surprisingly, BCL had a negative relationship with prevalence of overweight. In conclusion, BCL in Chinese adults was much higher than in other developed countries and was influenced by gender, smoking, and residential area. BCL was positively related to prediabetes while negatively related to overweight.
Collapse
Affiliation(s)
- Xiaomin Nie
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ningjian Wang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chi Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Bing Han
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Chunfang Zhu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Yingchao Chen
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Fangzhen Xia
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Zhen Cang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Meng Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ying Meng
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Boren Jiang
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Michael D Jensen
- Endocrine Research Unit, Mayo Clinic, 5-194 Joseph, Rochester, MN55905, USA.
| | - Yingli Lu
- Institute and Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| |
Collapse
|
1439
|
Schistosome-Derived Molecules as Modulating Actors of the Immune System and Promising Candidates to Treat Autoimmune and Inflammatory Diseases. J Immunol Res 2016; 2016:5267485. [PMID: 27635405 PMCID: PMC5011209 DOI: 10.1155/2016/5267485] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 06/29/2016] [Accepted: 07/13/2016] [Indexed: 12/27/2022] Open
Abstract
It is long known that some parasite infections are able to modulate specific pathways of host's metabolism and immune responses. This modulation is not only important in order to understand the host-pathogen interactions and to develop treatments against the parasites themselves but also important in the development of treatments against autoimmune and inflammatory diseases. Throughout the life cycle of schistosomes the mammalian hosts are exposed to several biomolecules that are excreted/secreted from the parasite infective stage, named cercariae, from their tegument, present in adult and larval stages, and finally from their eggs. These molecules can induce the activation and modulation of innate and adaptive responses as well as enabling the evasion of the parasite from host defense mechanisms. Immunomodulatory effects of helminth infections and egg molecules are clear, as well as their ability to downregulate proinflammatory cytokines, upregulate anti-inflammatory cytokines, and drive a Th2 type of immune response. We believe that schistosomes can be used as a model to understand the potential applications of helminths and helminth-derived molecules against autoimmune and inflammatory diseases.
Collapse
|
1440
|
Rumpler M, Mader JK, Fischer JP, Thar R, Granger JM, Deliane F, Klimant I, Aberer F, Sinner F, Pieber TR, Hajnsek M. First application of a transcutaneous optical single-port glucose monitoring device in patients with type 1 diabetes mellitus. Biosens Bioelectron 2016; 88:240-248. [PMID: 27554063 DOI: 10.1016/j.bios.2016.08.039] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 07/29/2016] [Accepted: 08/13/2016] [Indexed: 11/17/2022]
Abstract
The combination of continuous glucose monitoring (CGM) and continuous subcutaneous insulin infusion can be used to improve the treatment of patients with diabetes. The aim of this study was to advance an existing preclinical single-port system for clinical application by integrating the sensors of a phosphorescence based CGM system into a standard insulin infusion set. The extracorporeal optical phase fluorimeter was miniaturised and is now comparable with commercial CGM systems regarding size, weight and wear comfort. Sensor chemistry was adapted to improve the adhesion of the sensor elements on the insulin infusion set. In-vitro tests showed a linear correlation of R2=0.998 between sensor values and reference glucose values in the range of 0-300mg/dl. Electrical and cytotoxicity tests showed no negative impact on human health. Two single-port devices were tested in each of 12 patients with type 1 diabetes mellitus in a clinical set-up for 12h. Without additional data processing, the overall median absolute relative difference (median ARD) was 22.5%. For some of the used devices the median ARD was even well below 10%. The present results show that individual glucose sensors performance of the single-port system is comparable with commercial CGM systems but further improvements are needed. The new system offers a high extent of safety and usability by combining insulin infusion and continuous glucose measurement in a single-port system which could become a central element in an artificial pancreas for an improved treatment of patients with type 1 diabetes mellitus.
Collapse
Affiliation(s)
- M Rumpler
- JOANNEUM RESEARCH Forschungsgesellschaft mbH, HEALTH - Institute of Biomedicine and Health Sciences, Graz, Austria
| | - J K Mader
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | | | - R Thar
- Pyro Science GmbH, Aachen, Germany
| | - J M Granger
- RESCOLL Société de Recherche, Pessac Cedex, France
| | - F Deliane
- RESCOLL Société de Recherche, Pessac Cedex, France
| | - I Klimant
- Graz University of Technology, Institute of Analytical Chemistry and Food Chemistry, Graz, Austria
| | - F Aberer
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | - F Sinner
- JOANNEUM RESEARCH Forschungsgesellschaft mbH, HEALTH - Institute of Biomedicine and Health Sciences, Graz, Austria; Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | - T R Pieber
- JOANNEUM RESEARCH Forschungsgesellschaft mbH, HEALTH - Institute of Biomedicine and Health Sciences, Graz, Austria; Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Graz, Austria
| | - M Hajnsek
- JOANNEUM RESEARCH Forschungsgesellschaft mbH, HEALTH - Institute of Biomedicine and Health Sciences, Graz, Austria.
| |
Collapse
|
1441
|
Drexhage HA, Dik WA, Leenen PJM, Versnel MA. The Immune Pathogenesis of Type 1 Diabetes: Not Only Thinking Outside the Cell but Also Outside the Islet and Out of the Box. Diabetes 2016; 65:2130-3. [PMID: 27456621 DOI: 10.2337/dbi16-0030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
- Hemmo A Drexhage
- Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Wim A Dik
- Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Pieter J M Leenen
- Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Marjan A Versnel
- Department of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
1442
|
Ramakrishnan P, Yui MA, Tomalka JA, Majumdar D, Parameswaran R, Baltimore D. Deficiency of Nuclear Factor-κB c-Rel Accelerates the Development of Autoimmune Diabetes in NOD Mice. Diabetes 2016; 65:2367-79. [PMID: 27217485 PMCID: PMC4955991 DOI: 10.2337/db15-1607] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 05/15/2016] [Indexed: 12/18/2022]
Abstract
The nuclear factor-κB protein c-Rel plays a critical role in controlling autoimmunity. c-Rel-deficient mice are resistant to streptozotocin-induced diabetes, a drug-induced model of autoimmune diabetes. We generated c-Rel-deficient NOD mice to examine the role of c-Rel in the development of spontaneous autoimmune diabetes. We found that both CD4(+) and CD8(+) T cells from c-Rel-deficient NOD mice showed significantly decreased T-cell receptor-induced IL-2, IFN-γ, and GM-CSF expression. Despite compromised T-cell function, c-Rel deficiency dramatically accelerated insulitis and hyperglycemia in NOD mice along with a substantial reduction in T-regulatory (Treg) cell numbers. Supplementation of isogenic c-Rel-competent Treg cells from prediabetic NOD mice reversed the accelerated diabetes development in c-Rel-deficient NOD mice. The results suggest that c-Rel-dependent Treg cell function is critical in suppressing early-onset autoimmune diabetogenesis in NOD mice. This study provides a novel natural system to study autoimmune diabetes pathogenesis and reveals a previously unknown c-Rel-dependent mechanistic difference between chemically induced and spontaneous diabetogenesis. The study also reveals a unique protective role of c-Rel in autoimmune diabetes, which is distinct from other T-cell-dependent autoimmune diseases such as arthritis and experimental autoimmune encephalomyelitis, where c-Rel promotes autoimmunity.
Collapse
Affiliation(s)
- Parameswaran Ramakrishnan
- Department of Pathology, School of Medicine, Case Western Reserve University, and University Hospitals Case Medical Center, Cleveland, OH
| | - Mary A Yui
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Jeffrey A Tomalka
- Department of Pathology, School of Medicine, Case Western Reserve University, and University Hospitals Case Medical Center, Cleveland, OH
| | - Devdoot Majumdar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| | - Reshmi Parameswaran
- Department of Pathology, School of Medicine, Case Western Reserve University, and University Hospitals Case Medical Center, Cleveland, OH
| | - David Baltimore
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA
| |
Collapse
|
1443
|
Affiliation(s)
- Isabelle Nel
- INSERM U1016 and Centre National de la Recherche Scientifique UMR8104, Institut Cochin, and Laboratoire d'Excellence INFLAMEX, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| | - Agnes Lehuen
- INSERM U1016 and Centre National de la Recherche Scientifique UMR8104, Institut Cochin, and Laboratoire d'Excellence INFLAMEX, Université Paris Descartes, Université Sorbonne Paris Cité, Paris, France
| |
Collapse
|
1444
|
Venkatesha SH, Dudics S, Astry B, Moudgil KD. Control of autoimmune inflammation by celastrol, a natural triterpenoid. Pathog Dis 2016; 74:ftw059. [PMID: 27405485 DOI: 10.1093/femspd/ftw059] [Citation(s) in RCA: 94] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2016] [Indexed: 12/19/2022] Open
Abstract
Celastrol is a bioactive compound derived from traditional Chinese medicinal herbs of the Celastraceae family. Celastrol is known to possess anti-inflammatory and anti-oxidant activities. Our studies have highlighted the immunomodulatory attributes of celastrol in adjuvant-induced arthritis (AA), an experimental model of human rheumatoid arthritis (RA). RA is an autoimmune disease characterized by chronic inflammation of the synovial lining of the joints, leading eventually to tissue damage and deformities. Identification of the molecular targets of celastrol such as the NF-κB pathway, MAPK pathway, JAK/STAT pathway and RANKL/OPG pathway has unraveled its strategic checkpoints in controlling arthritic inflammation and tissue damage in AA. The pathological events that are targeted and rectified by celastrol include increased production of pro-inflammatory cytokines; an imbalance between pathogenic T helper 17 and regulatory T cells; enhanced production of chemokines coupled with increased migration of immune cells into the joints; and increased release of mediators of osteoclastic bone damage. Accordingly, celastrol is a promising candidate for further testing in the clinic for RA therapy. Furthermore, the results of other preclinical studies suggest that celastrol might also be beneficial for the treatment of a few other autoimmune diseases besides arthritis.
Collapse
Affiliation(s)
- Shivaprasad H Venkatesha
- Department of Microbiology and Immunology, Division of Rheumatology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Steven Dudics
- Department of Microbiology and Immunology, Division of Rheumatology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Brian Astry
- Department of Microbiology and Immunology, Division of Rheumatology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA
| | - Kamal D Moudgil
- Department of Microbiology and Immunology, Division of Rheumatology, University of Maryland School of Medicine, 685 W. Baltimore Street, HSF-1, Suite 380, Baltimore, MD 21201, USA Department of Medicine, Division of Rheumatology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
1445
|
Veijola R, Koskinen M, Helminen O, Hekkala A. Dysregulation of glucose metabolism in preclinical type 1 diabetes. Pediatr Diabetes 2016; 17 Suppl 22:25-30. [PMID: 27411433 DOI: 10.1111/pedi.12392] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2016] [Accepted: 03/23/2016] [Indexed: 01/13/2023] Open
Abstract
Long-term prospective studies have provided valuable information about preclinical type 1 diabetes (T1D). Children who have seroconverted to positive for islet autoantibodies have also, in follow-up, had metabolic tests to understand the timing and development of abnormal glucose tolerance and declining insulin secretion before the clinical diagnosis of T1D. First phase insulin response (FPIR) in the intravenous glucose tolerance test (IVGTT) is lower in the progressors positive for multiple islet autoantibodies in all age groups and as early as 4-6 years before the diagnosis when compared with the non-progressors positive for only islet cell antibodies (ICA). An accelerated decline in FPIR is seen in the progressors during the last 1.5 years before the diagnosis. These results indicate that the progressors may have an early intrinsic defect in beta cell development or function. In the oral glucose tolerance test (OGTT) the peak C-peptide response is delayed in the progressors at least 2 years before diagnosis. Glucose levels and HbA1c are increasing about 2 years before clinical diagnosis. An increase in HbA1c and detection of abnormal glucose tolerance in OGTT are useful in the prediction of the timing of clinical onset of T1D. Continuous glucose monitoring (CGM) may be useful in the prediction of T1D as an early indicator of increased glycemic variability but more data from larger series are needed for confirmation. Children followed in the prospective studies are diagnosed earlier and have a decreased frequency of ketoacidosis at the diagnosis of T1D when compared with age-matched cases from the population.
Collapse
Affiliation(s)
- Riitta Veijola
- Department of Pediatrics, Research Unit for Pediatrics, Dermatology, Clinical Genetics, Gynecology and Obstetrics (PEDEGO), Medical Research Center (MRC) Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Maarit Koskinen
- Department of Pediatrics, University of Turku, Turku, Finland.,Department of Pediatrics, Turku University Hospital, Turku, Finland
| | - Olli Helminen
- Department of Pediatrics, Research Unit for Pediatrics, Dermatology, Clinical Genetics, Gynecology and Obstetrics (PEDEGO), Medical Research Center (MRC) Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| | - Anne Hekkala
- Department of Pediatrics, Research Unit for Pediatrics, Dermatology, Clinical Genetics, Gynecology and Obstetrics (PEDEGO), Medical Research Center (MRC) Oulu, University of Oulu, Oulu, Finland.,Department of Children and Adolescents, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
1446
|
Li X, Cheng J, Zhou Z. Revisiting multiple models of progression of β-cell loss of function in type 1 diabetes: Significance for prevention and cure. J Diabetes 2016; 8:460-9. [PMID: 26754489 DOI: 10.1111/1753-0407.12376] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 12/24/2015] [Accepted: 01/07/2016] [Indexed: 01/12/2023] Open
Abstract
Type 1 diabetes (T1D) results from a chronic autoimmune process that leads to β-cell destruction and exogenous insulin dependence. The natural history of T1D proposed by Eisenbarth suggested six relatively independent stages over the course of the entire disease process, which was considered to be linear and chronic. Based on this classical theory, immunotherapies aim to prevent or reverse all these periods of β-cell loss. Over the past 30 years, much novel information about the pathogenesis of T1D proved that there are complex metabolic changes occurring throughout the entire disease process. Therefore, new possible models for the natural history of the disease have been proposed; these models, in turn, may help facilitate fresh avenues for the prevention and cure of T1D. Herein, we briefly review recent findings in this field of research, with the aim of providing a better theoretical basis for clinical practice.
Collapse
Affiliation(s)
- Xia Li
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital and the Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan, China
| | - Jin Cheng
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital and the Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan, China
| | - Zhiguang Zhou
- Institute of Metabolism and Endocrinology, The Second Xiangya Hospital and the Diabetes Center, Key Laboratory of Diabetes Immunology, Ministry of Education, National Clinical Research Center for Metabolic Diseases, Central South University, Changsha, Hunan, China
| |
Collapse
|
1447
|
El Karib AO, Al-Ani B, Al-Hashem F, Dallak M, Bin-Jaliah I, El-Gamal B, Bashir SO, Eid RA, Haidara MA. Insulin and vanadium protect against osteoarthritis development secondary to diabetes mellitus in rats. Arch Physiol Biochem 2016; 122:148-154. [PMID: 26939846 DOI: 10.3109/13813455.2016.1159698] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVE Diabetic complications such as cardiovascular disease and osteoarthritis (OA) are among the common public health problems. The effect of insulin on OA secondary to diabetes has not been investigated before in animal models. Therefore, we sought to determine whether insulin and the insulin-mimicking agent, vanadium can protect from developing OA in diabetic rats. METHODS Type 1 diabetes mellitus (T1DM) was induced in Sprague-Dawley rats and treated with insulin and/or vanadium. Tissues harvested from the articular cartilage of the knee joint were examined by scanning electron microscopy, and blood samples were assayed for oxidative stress and inflammatory biomarkers. RESULTS Eight weeks following the induction of diabetes, a profound damage to the knee joint compared to the control non-diabetic group was observed. Treatment of diabetic rats with insulin and/or vanadium differentially protected from diabetes-induced cartilage damage and deteriorated fibrils of collagen fibers. The relative biological potencies were insulin + vanadium >> insulin > vanadium. Furthermore, there was about 2- to 5-fold increase in TNF-α (from 31.02 ± 1.92 to 60.5 ± 1.18 pg/ml, p < 0.0001) and IL-6 (from 64.67 ± 8.16 to 338.0 ± 38.9 pg/ml, p < 0.0001) cytokines and free radicals measured as TBARS (from 3.21 ± 0.37 to 11.48 ± 1.5 µM, p < 0.0001) in the diabetic group, which was significantly reduced with insulin and or vanadium. Meanwhile, SOD decreased (from 17.79 ± 8.9 to 8.250.29, p < 0.0001) and was increased with insulin and vanadium. The relative potencies of the treating agents on inflammatory and oxidative stress biomarkers were insulin + vanadium >> insulin > vanadium. CONCLUSION The present study demonstrates that co-administration of insulin and vanadium to T1DM rats protect against diabetes-induced OA possibly by lowering biomarkers of inflammation and oxidative stress.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Refaat A Eid
- c Department of Pathology , College of Medicine, King Khalid University , Abha , Saudi Arabia , and
| | - Mohamed A Haidara
- a Department of Physiology
- d Department of Physiology , Kasr al-Aini Faculty of Medicine, Cairo University , Cairo , Egypt
| |
Collapse
|
1448
|
Harlan DM. Islet Transplantation for Hypoglycemia Unawareness/Severe Hypoglycemia: Caveat Emptor. Diabetes Care 2016; 39:1072-4. [PMID: 27330121 DOI: 10.2337/dci16-0008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- David M Harlan
- Diabetes Center of Excellence, Department of Medicine, University of Massachusetts Medical School, Worcester, MA
| |
Collapse
|
1449
|
Galli E, Härkönen T, Sainio MT, Ustav M, Toots U, Urtti A, Yliperttula M, Lindahl M, Knip M, Saarma M, Lindholm P. Increased circulating concentrations of mesencephalic astrocyte-derived neurotrophic factor in children with type 1 diabetes. Sci Rep 2016; 6:29058. [PMID: 27356471 PMCID: PMC4928177 DOI: 10.1038/srep29058] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 06/10/2016] [Indexed: 12/17/2022] Open
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) was recently shown to be essential for the survival and proliferation of pancreatic β-cells in mice, where deletion of MANF resulted in diabetes. The current study aimed at determining whether the concentration of circulating MANF is associated with the clinical manifestation of human type 1 diabetes (T1D). MANF expression in T1D or MANF levels in serum have not been previously studied. We developed an enzyme-linked immunosorbent assay (ELISA) for MANF and measured serum MANF concentrations from 186 newly diagnosed children and adolescents and 20 adults with longer-term T1D alongside with age-matched controls. In healthy controls the mean serum MANF concentration was 7.0 ng/ml. High MANF concentrations were found in children 1–9 years of age close to the diagnosis of T1D. The increased MANF concentrations were not associated with diabetes-predictive autoantibodies and autoantibodies against MANF were extremely rare. Patients with conspicuously high MANF serum concentrations had lower C-peptide levels compared to patients with moderate MANF concentrations. Our data indicate that increased MANF concentrations in serum are associated with the clinical manifestation of T1D in children, but the exact mechanism behind the increase remains elusive.
Collapse
Affiliation(s)
- Emilia Galli
- Institute of Biotechnology, University of Helsinki, Finland.,Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Finland
| | - Taina Härkönen
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Finland
| | | | | | | | - Arto Urtti
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Finland
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Centre for Drug Research, University of Helsinki, Finland
| | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Finland
| | - Mikael Knip
- Children's Hospital, University of Helsinki and Helsinki University Central Hospital, Finland.,Research Programs Unit, Diabetes and Obesity, University of Helsinki, Finland.,Folkhälsan Research Center, Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, University of Helsinki, Finland
| | - Päivi Lindholm
- Institute of Biotechnology, University of Helsinki, Finland
| |
Collapse
|
1450
|
Yeste A, Takenaka MC, Mascanfroni ID, Nadeau M, Kenison JE, Patel B, Tukpah AM, Babon JAB, DeNicola M, Kent SC, Pozo D, Quintana FJ. Tolerogenic nanoparticles inhibit T cell-mediated autoimmunity through SOCS2. Sci Signal 2016; 9:ra61. [PMID: 27330188 DOI: 10.1126/scisignal.aad0612] [Citation(s) in RCA: 152] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Type 1 diabetes (T1D) is a T cell-dependent autoimmune disease that is characterized by the destruction of insulin-producing β cells in the pancreas. The administration to patients of ex vivo-differentiated FoxP3(+) regulatory T (Treg) cells or tolerogenic dendritic cells (DCs) that promote Treg cell differentiation is considered a potential therapy for T1D; however, cell-based therapies cannot be easily translated into clinical practice. We engineered nanoparticles (NPs) to deliver both a tolerogenic molecule, the aryl hydrocarbon receptor (AhR) ligand 2-(1'H-indole-3'-carbonyl)-thiazole-4-carboxylic acid methyl ester (ITE), and the β cell antigen proinsulin (NPITE+Ins) to induce a tolerogenic phenotype in DCs and promote Treg cell generation in vivo. NPITE+Ins administration to 8-week-old nonobese diabetic mice suppressed autoimmune diabetes. NPITE+Ins induced a tolerogenic phenotype in DCs, which was characterized by a decreased ability to activate inflammatory effector T cells and was concomitant with the increased differentiation of FoxP3(+) Treg cells. The induction of a tolerogenic phenotype in DCs by NPs was mediated by the AhR-dependent induction of Socs2, which resulted in inhibition of nuclear factor κB activation and proinflammatory cytokine production (properties of tolerogenic DCs). Together, these data suggest that NPs constitute a potential tool to reestablish tolerance in T1D and potentially other autoimmune disorders.
Collapse
Affiliation(s)
- Ada Yeste
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Maisa C Takenaka
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ivan D Mascanfroni
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Meghan Nadeau
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jessica E Kenison
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Bonny Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ann-Marcia Tukpah
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Jenny Aurielle B Babon
- Department of Medicine, Diabetes Division, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Megan DeNicola
- Department of Medicine, Diabetes Division, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Sally C Kent
- Department of Medicine, Diabetes Division, Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - David Pozo
- CABIMER-Andalusian Center for Molecular Biology and Regenerative Medicine (Consejo Superior de Investigaciones Científicas-University of Seville-Universidad Pablo de Olavide), Seville 41092, Spain. Department of Medical Biochemistry, Molecular Biology and Immunology, University of Seville Medical School, Seville 41009, Spain
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA. Broad Institute of Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142, USA.
| |
Collapse
|