1
|
Alhamar G, Vinci C, Franzese V, Tramontana F, Le Goux N, Ludvigsson J, Nissim A, Strollo R. The role of oxidative post-translational modifications in type 1 diabetes pathogenesis. Front Immunol 2025; 16:1537405. [PMID: 40028329 PMCID: PMC11868110 DOI: 10.3389/fimmu.2025.1537405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
The pathogenesis of type 1 diabetes (T1D) involves a complex interplay of genetic predisposition, immune processes, and environmental factors, leading to the selective destruction of pancreatic beta-cells by the immune system. Emerging evidence suggests that intrinsic beta-cell factors, including oxidative stress and post-translational modifications (PTM) of beta-cell antigens, may also contribute to their immunogenicity, shedding new light on the multifaceted pathogenesis of T1D. Over the past 30 years, neoepitopes generated by PTMs have been hypothesized to play a role in T1D pathogenesis, but their involvement has only been systematically investigated in recent years. In this review, we explored the interplay between oxidative PTMs, neoepitopes, and T1D, highlighting oxidative stress as a pivotal factor in immune system dysfunction, beta-cell vulnerability, and disease onset.
Collapse
Affiliation(s)
- Ghadeer Alhamar
- Department of Immunology and Microbiology, Dasman Diabetes Institute, Dasman, Kuwait
| | - Chiara Vinci
- Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
| | - Valentina Franzese
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Rome, Italy
- Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
- Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Flavia Tramontana
- Department of Medicine, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Nelig Le Goux
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Johnny Ludvigsson
- Crown Princess Victoria Children’s Hospital and Division of Pediatrics, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | - Rocky Strollo
- Department for the Promotion of Human Science and Quality of Life, San Raffaele Open University, Rome, Italy
- Department of Medicine, Fondazione Policlinico Universitario Campus Bio-Medico di Roma, Rome, Italy
| |
Collapse
|
2
|
Vasudevan A, Venkatesan P. Association of organ iron levels with type 2 diabetes mellitus and glycemic traits: A bidirectional two-sample Mendelian randomization study. J Trace Elem Med Biol 2025; 87:127586. [PMID: 39754912 DOI: 10.1016/j.jtemb.2024.127586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 12/25/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
INTRODUCTION Observational studies have found that higher iron levels are associated with an increased risk of diabetes mellitus. Given the limitations of causal inferences from observational studies and the expensive and time-consuming nature of randomized controlled trials, Mendelian randomization analysis presents a reasonable alternative to study causal relationships. Previous MR analyses studying iron levels and diabetes have used indirect markers of iron levels, such as serum ferritin, and found conflicting results. In this study, we performed bidirectional Mendelian Randomization analyses using organ iron (liver, spleen, and pancreas) levels, which are more direct markers of iron status, to study the causal association of iron levels with type 2 diabetes mellitus and glycaemic traits. METHODS Two sample MR analyses were employed bi-directionally to study the causal effect of liver, spleen, and pancreas iron levels on type 2 diabetes and glycaemic traits and the causal effect of type 2 diabetes on organ iron levels, using summary data from genome-wide association studies (UK-Biobank, DIAGRAM, and MAGIC consortia). SNPs associated with organ iron levels with a cut-off of P < 5 × 10-7 were used as instrumental variables for the MR analyses of the effect of organ iron levels on type 2 diabetes/glycaemic traits, and SNPs associated with diabetes mellitus with a cut-off of P < 5 × 10-8 were used as instrumental variables for the MR analyses of the causal effect of type 2 diabetes on organ iron levels. Serum ferritin (GWAS meta-analysis of deCODE, UK INTERVAL, and Denmark studies) and haemoglobin (Blood Cell consortium) were used as positive controls for the MR analysis with liver iron as the exposure. Primary analyses used the inverse variance weighted means of Wald's ratio. Sensitivity analyses included inverse variance weighted median, weighted mode, and MR-Egger methods. RESULTS Our findings reveal no causal association between liver and pancreas iron levels with type 2 diabetes (Liver iron: OR = 1.02, P = 0.1, Pancreas iron: OR = 1.11, P = 0.5). This also holds for glycaemic traits, except for the negative causal effect of liver iron levels on HbA1c (OR = 0.93, P = 0.001). Spleen iron levels had a negative causal effect on type 2 diabetes (OR = 0.94, P = 0.049). However, these exceptions are likely due to possible pleiotropy, as these associations can be explained by the effect of the genetic variants on factors that falsely decrease HbA1c levels. No causal association was found for the effect of type 2 diabetes on organ iron levels. CONCLUSION Organ iron levels, which are relatively more direct indicators of iron status, showed no causal association with type 2 diabetes in the European population.
Collapse
Affiliation(s)
- Akshaya Vasudevan
- Department of Community Medicine, Christian Medical College, Vellore, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Chennai, India.
| | - Padmanaban Venkatesan
- Department of Biochemistry, Christian Medical College, Vellore, Tamil Nadu, India; Affiliated to The Tamil Nadu Dr. MGR Medical University, Chennai, India.
| |
Collapse
|
3
|
Conway RB, Pratte KA, Bowler RP, Young KA, Kinney GL, Austin E, Li Y, McClain D, Hokanson J, Crapo JD. Plasma Proteomic Markers of Iron and Risk of Diabetes in a Cohort of African American and White American Current and Former Smokers. Diabetes Metab Syndr Obes 2024; 17:4767-4776. [PMID: 39678225 PMCID: PMC11646377 DOI: 10.2147/dmso.s492124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024] Open
Abstract
Background Little information is available on iron with diabetes risk among African Americans, a population where both anemia and elevated ferritin are common. We tested whether plasma proteomic measurements of ferritin and transferrin were associated with increased diabetes risk in a cohort of current and former African American (NHB) and Non-Hispanic White (NHW) smokers. Methods NHB and NHW participants from the COPDGene study who were free of diabetes (n = 4693) at baseline were followed for incident diabetes. The SomaScan was used to determine the relative amounts of natural log-transformed ferritin, transferrin, and hepcidin. Findings During an average of 5.6 years of follow-up, diabetes incidence was 7.9%. Ferritin at follow-up was higher in NHB than NHW participants (p = <0.0001). Ferritin at follow-up was associated with increased diabetes risk (OR = 1.36, 95% CI = 1.08-1.70), while transferrin was associated with decreased risk (OR = 0.25, 95% CI = 0.08-0.77) controlling for age, sex, BMI, smoking pack-years, hepcidin, CRP, and Il-6. Race-specifically, increased risk associated with higher ferritin levels among NHB (OR = 1.56, 95% CI = 1.13-2.16) but not NHW (OR = 1.22, 95% CI = 0.89-1.68) participants. Sex-specifically, ferritin's relationship was similar among NHB men and women and NHW women (ORs ranging from 1.41-1.59); but not NHW men (OR = 0.98, 95% CI = 0.64-1.49). Similarly, transferrin ORs non-significantly ranged from 0.19-0.30 for NHB men and women and NHW women, but was significant for NHW men (OR = 0.07, 95% CI = 0.01-0.63). Interpretation Higher body iron stores is associated with increased diabetes risk among both NHB and NHW people. Unsuspected elevated iron stores may increase diabetes risk in NHB patients and should be monitored.
Collapse
Affiliation(s)
- Rebecca Baqiyyah Conway
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Katherine A Pratte
- Division of Biostatistics and Bioinformatics, National Jewish Health, Denver, CO, 80206, USA
| | - Russell Paul Bowler
- Department of Genomic Sciences, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
| | - Kendra A Young
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Gregory l Kinney
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Erin Austin
- Department of Mathematical and Statistical Sciences, Denver, University of Colorado, Denver, CO, 80204, USA
| | - Yisha Li
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Donald McClain
- Section of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - John Hokanson
- Department of Epidemiology, University of Colorado, Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - James D Crapo
- Department of Medicine, National Jewish Health, Denver, CO, 80206, USA
| |
Collapse
|
4
|
Tam E, Nguyen K, Sung HK, Sweeney G. MitoNEET preserves muscle insulin sensitivity during iron overload by regulating mitochondrial iron, reactive oxygen species and fission. FEBS J 2024; 291:4062-4075. [PMID: 38944692 DOI: 10.1111/febs.17214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/13/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
Iron overload (IO) is known to contribute to metabolic dysfunctions such as type 2 diabetes and insulin resistance. Using L6 skeletal muscle cells overexpressing the CDGSH iron-sulfur domain-containing protein 1 (CISD1, also known as mitoNEET) (mitoN) protein, we examined the potential role of MitoN in preventing IO-induced insulin resistance. In L6 control cells, IO resulted in insulin resistance which could be prevented by MitoN as demonstrated by western blot of p-Akt and Akt biosensor cells. Mechanistically, IO increased; mitochondrial iron accumulation, mitochondrial reactive oxygen species (ROS), Fis1-dependent mitochondrial fission, mitophagy, FUN14 domain-containing protein 1 (FUNDC1) expression, and decreased Parkin. MitoN overexpression was able to reduce increases in mitochondrial iron accumulation, mitochondrial ROS, mitochondrial fission, mitophagy and FUNDC1 upregulation due to IO. MitoN did not have any effect on the IO-induced downregulation of Parkin. MitoN alone also upregulated peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) protein levels, a master regulator of mitochondrial biogenesis. The use of mitochondrial antioxidant, Skq1, or fission inhibitor, Mdivi-1, prevented IO-induced insulin resistance implying both mitochondrial ROS and fission play a causal role in the development of insulin resistance. Taken together, MitoN is able to confer protection against IO-induced insulin resistance in L6 skeletal muscle cells through regulation of mitochondrial iron content, mitochondrial ROS, and mitochondrial fission.
Collapse
Affiliation(s)
- Eddie Tam
- Department of Biology, York University, Toronto, Canada
| | - Khang Nguyen
- Department of Biology, York University, Toronto, Canada
| | | | - Gary Sweeney
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
5
|
Feng J, Shan X, Wang L, Lu J, Cao Y, Yang L. Association of Body Iron Metabolism with Type 2 Diabetes Mellitus in Chinese Women of Childbearing Age: Results from the China Adult Chronic Disease and Nutrition Surveillance (2015). Nutrients 2023; 15:nu15081935. [PMID: 37111154 PMCID: PMC10141641 DOI: 10.3390/nu15081935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/15/2023] [Indexed: 04/29/2023] Open
Abstract
High iron stores have been reported to be associated with type 2 diabetes mellitus (T2DM). However, evidence for the associations of iron metabolism with T2DM is inconsistent, and whether there is a threshold effect remains controversial. In the present study, we aimed to examine the associations between various iron biomarkers and the risk of T2DM as well as impaired glucose metabolism (IGM) and hyperglycemia in Chinese women of childbearing age. A total of 1145 women were divided into three groups (normal blood glucose metabolism group; IGM group; T2DM group). Biomarkers of iron metabolism (serum ferritin (SF), transferrin, soluble transferrin receptor (sTfR), transferrin saturation, serum iron, total body iron, and sTfR-to-lgferritin index) were measured. After adjusting for various confounding risk factors, SF and sTfR were positively associated with the risk of IGM (fourth vs. first quartile: SF odds ratio (OR) = 1.93 (95% CI 1.17-3.20) and sTfR OR = 3.08 (95% CI 1.84-5.14)) and T2DM (SF OR = 2.39 (95% CI 1.40-4.06) and sTfR OR = 3.84 (95% CI 2.53-5.83)). There was a nonlinear relationship between SF and risk of T2DM and hyperglycemia (p for nonlinearity < 0.01). Our findings suggested that SF and sTfR could be independent predictors of T2DM risk.
Collapse
Affiliation(s)
- Jie Feng
- Key Laboratory of Trace Element Nutrition of National Health Committee, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Xiaoyun Shan
- Key Laboratory of Trace Element Nutrition of National Health Committee, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
- Hunan Key Laboratory of Typical Environmental Pollution and Health Hazards, School of Public Health, Hengyang Medical School, University of South China, Hengyang 241001, China
| | - Lijuan Wang
- Key Laboratory of Trace Element Nutrition of National Health Committee, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Jiaxi Lu
- Key Laboratory of Trace Element Nutrition of National Health Committee, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Yang Cao
- Key Laboratory of Trace Element Nutrition of National Health Committee, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| | - Lichen Yang
- Key Laboratory of Trace Element Nutrition of National Health Committee, National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing 100050, China
| |
Collapse
|
6
|
Dalle S, Abderrahmani A, Renard E. Pharmacological inhibitors of β-cell dysfunction and death as therapeutics for diabetes. Front Endocrinol (Lausanne) 2023; 14:1076343. [PMID: 37008937 PMCID: PMC10050720 DOI: 10.3389/fendo.2023.1076343] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 02/20/2023] [Indexed: 03/17/2023] Open
Abstract
More than 500 million adults suffer from diabetes worldwide, and this number is constantly increasing. Diabetes causes 5 million deaths per year and huge healthcare costs per year. β-cell death is the major cause of type 1 diabetes. β-cell secretory dysfunction plays a key role in the development of type 2 diabetes. A loss of β-cell mass due to apoptotic death has also been proposed as critical for the pathogenesis of type 2 diabetes. Death of β-cells is caused by multiple factors including pro-inflammatory cytokines, chronic hyperglycemia (glucotoxicity), certain fatty acids at high concentrations (lipotoxicity), reactive oxygen species, endoplasmic reticulum stress, and islet amyloid deposits. Unfortunately, none of the currently available antidiabetic drugs favor the maintenance of endogenous β-cell functional mass, indicating an unmet medical need. Here, we comprehensively review over the last ten years the investigation and identification of molecules of pharmacological interest for protecting β-cells against dysfunction and apoptotic death which could pave the way for the development of innovative therapies for diabetes.
Collapse
Affiliation(s)
- Stéphane Dalle
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
| | - Amar Abderrahmani
- Université Lille, Centre National de la Recherche Scientifique (CNRS), Centrale Lille, Polytechnique Hauts-de-France, UMR 8520, IEMN, Lille, France
| | - Eric Renard
- Institut de Génomique Fonctionnelle, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Institut National de la Santé et de la Recherche Médicale (INSERM), Montpellier, France
- Laboratoire de Thérapie Cellulaire du Diabète, Centre Hospitalier Universitaire, Montpellier, France
- Département d’Endocrinologie, Diabètologie, Centre Hospitalier Universitaire, Montpellier, France
| |
Collapse
|
7
|
Garbowski MW, Cabantchik I, Hershko C, Hider R, Porter JB. The clinical relevance of detectable plasma iron species in iron overload states and subsequent to intravenous iron-carbohydrate administration. Am J Hematol 2023; 98:533-540. [PMID: 36565452 DOI: 10.1002/ajh.26819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/20/2022] [Accepted: 11/26/2022] [Indexed: 12/25/2022]
Abstract
Many disorders of iron homeostasis (e.g., iron overload) are associated with the dynamic kinetic profiles of multiple non-transferrin bound iron (NTBI) species, chronic exposure to which is associated with deleterious end-organ effects. Here we discuss the chemical nature of NTBI species, challenges with measuring NTBI in plasma, and the clinical relevance of NTBI exposure based on source (iron overload disorder vs. intravenous iron-carbohydrate complex administration). NTBI is not a single entity but consists of multiple, often poorly characterized species, some of which are kinetically non-exchangeable while others are relatively exchangeable. Prolonged presence of plasma NTBI is associated with excessive tissue iron accumulation in susceptible tissues, with consequences, such as endocrinopathy and heart failure. In contrast, intravenous iron-carbohydrate nanomedicines administration leads only to transient NTBI appearance and lacks evidence for association with adverse clinical outcomes. Assays to measure plasma NTBI are typically technically complex and remain chiefly a research tool. There have been two general approaches to estimating NTBI: capture assays and redox-activity assays. Early assays could not avoid capturing some iron from transferrin, thus overestimating NTBI. By contrast, some later assays may have promoted the donation of NTBI species to transferrin during the assay procedure, potentially underestimating NTBI levels. The levels of transferrin saturation at which NTBI species have been detectable have varied between different methodologies and between patient populations studied.
Collapse
Affiliation(s)
- Maciej W Garbowski
- Cancer Institute Haematology Department, University College London, London, United Kingdom.,London Metallomics Consortium, London, United Kingdom
| | - Ioav Cabantchik
- Alexander Silberman Institute of Life Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Chaim Hershko
- Shaare Zedek Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Robert Hider
- London Metallomics Consortium, London, United Kingdom.,Institute of Pharmaceutical Science, King's College London, London, United Kingdom
| | - John B Porter
- Cancer Institute Haematology Department, University College London, London, United Kingdom
| |
Collapse
|
8
|
Qin Y, Huang Y, Li Y, Qin L, Wei Q, Chen X, Yang C, Zhang M. Association between systemic iron status and β-cell function and insulin sensitivity in patients with newly diagnosed type 2 diabetes. Front Endocrinol (Lausanne) 2023; 14:1143919. [PMID: 37077360 PMCID: PMC10107407 DOI: 10.3389/fendo.2023.1143919] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 03/14/2023] [Indexed: 04/05/2023] Open
Abstract
OBJECTIVE Abnormal iron metabolism is related to the risk of diabetes, but the underlying mechanism of this association remains uncertain. This study was conducted to evaluate the contributions of systemic iron status to β-cell function and insulin sensitivity of patients with newly diagnosed T2DM. METHODS A total of 162 patients with newly diagnosed T2DM and 162 healthy controls were enrolled in the study. Basic characteristics, biochemical indicators, and iron metabolism biomarkers, including serum iron (SI), ferritin (SF), transferrin (Trf), and transferrin saturation (TS), were collected. All patients underwent a 75 g oral glucose tolerance test. A series of parameters for assessing β-cell function and insulin sensitivity were calculated. The multivariate stepwise linear regression model was used to investigate the contributions of iron metabolism to β-cell function and insulin sensitivity. RESULTS Compared with healthy controls, patients with newly diagnosed T2DM had significantly higher levels of SF. Among the diabetic patients, the SI and TS levels were higher, and the percentage of Trf levels below normal values was lower in men than in women. In all diabetic patients, SF was the independent risk factor associated with impaired β-cell function. Further stratification analysis showed that Trf was an independent protective factor for β-cell function in male patients, while SF was an independent risk factor for impaired β-cell function in female patients. However, systemic iron status did not affect insulin sensitivity. CONCLUSION Elevated SF levels and decreased Trf levels had a profound effect on impaired β-cell function in Chinese patients with newly diagnosed T2DM.
Collapse
Affiliation(s)
- Yao Qin
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yiting Huang
- Department of Clinical Nutrition, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuxiao Li
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lu Qin
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qianying Wei
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xin Chen
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chuanhui Yang
- Department of Endocrinology, the First People’s Hospital of Lianyungang, Lianyungang, China
| | - Mei Zhang
- Department of Endocrinology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- *Correspondence: Mei Zhang,
| |
Collapse
|
9
|
Strollo R, Vinci C, Man YKS, Bruzzaniti S, Piemonte E, Alhamar G, Briganti SI, Malandrucco I, Tramontana F, Fanali C, Garnett J, Buccafusca R, Guyer P, Mamula M, James EA, Pozzilli P, Ludvigsson J, Winyard PG, Galgani M, Nissim A. Autoantibody and T cell responses to oxidative post-translationally modified insulin neoantigenic peptides in type 1 diabetes. Diabetologia 2023; 66:132-146. [PMID: 36207582 PMCID: PMC9729141 DOI: 10.1007/s00125-022-05812-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/28/2022] [Indexed: 12/14/2022]
Abstract
AIMS/HYPOTHESIS Antibodies specific to oxidative post-translational modifications (oxPTM) of insulin (oxPTM-INS) are present in most individuals with type 1 diabetes, even before the clinical onset. However, the antigenic determinants of such response are still unknown. In this study, we investigated the antibody response to oxPTM-INS neoepitope peptides (oxPTM-INSPs) and evaluated their ability to stimulate humoral and T cell responses in type 1 diabetes. We also assessed the concordance between antibody and T cell responses to the oxPTM-INS neoantigenic peptides. METHODS oxPTM-INS was generated by exposing insulin to various reactive oxidants. The insulin fragments resulting from oxPTM were fractionated by size-exclusion chromatography further to ELISA and LC-MS/MS analysis to identify the oxidised peptide neoepitopes. Immunogenic peptide candidates were produced and then modified in house or designed to incorporate in silico-oxidised amino acids during synthesis. Autoantibodies to the oxPTM-INSPs were tested by ELISA using sera from 63 participants with new-onset type 1 diabetes and 30 control participants. An additional 18 fresh blood samples from participants with recently diagnosed type 1 diabetes, five with established disease, and from 11 control participants were used to evaluate, in parallel, CD4+ and CD8+ T cell activation by oxPTM-INSPs. RESULTS We observed antibody and T cell responses to three out of six LC-MS/MS-identified insulin peptide candidates: A:12-21 (SLYQLENYCN, native insulin peptide 3 [Nt-INSP-3]), B:11-30 (LVEALYLVCGERGFFYTPKT, Nt-INSP-4) and B:21-30 (ERGFFYTPKT, Nt-INSP-6). For Nt-INSP-4 and Nt-INSP-6, serum antibody binding was stronger in type 1 diabetes compared with healthy control participants (p≤0.02), with oxidised forms of ERGFFYTPKT, oxPTM-INSP-6 conferring the highest antibody binding (83% binders to peptide modified in house by hydroxyl radical [●OH] and >88% to in silico-oxidised peptide; p≤0.001 vs control participants). Nt-INSP-4 induced the strongest T cell stimulation in type 1 diabetes compared with control participants for both CD4+ (p<0.001) and CD8+ (p=0.049). CD4+ response to oxPTM-INSP-6 was also commoner in type 1 diabetes than in control participants (66.7% vs 27.3%; p=0.039). Among individuals with type 1 diabetes, the CD4+ response to oxPTM-INSP-6 was more frequent than to Nt-INSP-6 (66.7% vs 27.8%; p=0.045). Overall, 44.4% of patients showed a concordant autoimmune response to oxPTM-INSP involving simultaneously CD4+ and CD8+ T cells and autoantibodies. CONCLUSIONS/INTERPRETATION Our findings support the concept that oxidative stress, and neoantigenic epitopes of insulin, may be involved in the immunopathogenesis of type 1 diabetes.
Collapse
Affiliation(s)
- Rocky Strollo
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Vinci
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Y K Stella Man
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Sara Bruzzaniti
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Biology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Erica Piemonte
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ghadeer Alhamar
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Silvia Irina Briganti
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Ilaria Malandrucco
- The UOSD of Endocrinology and Metabolic Diseases, Azienda Sanitaria Locale (ASL) Frosinone, Frosinone, Italy
| | - Flavia Tramontana
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Fanali
- Department of Science and Technology for Humans and the Environment, Università Campus Bio-Medico di Roma, Rome, Italy
| | - James Garnett
- Centre for Host-Microbiome Interactions, Dental Institute, King's College London, London, UK
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Roberto Buccafusca
- School of Biological and Chemical Sciences, Queen Mary University of London, London, UK
| | - Perrin Guyer
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Mark Mamula
- Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Eddie A James
- Program for Translational Immunology, Benaroya Research Institute, Seattle, WA, USA
| | - Paolo Pozzilli
- Department of Medicine, Unit of Endocrinology & Diabetes, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Johnny Ludvigsson
- Division of Pediatrics, Department of Biomedical and Clinical Sciences, Crown Princess Victoria Children's Hospital, Linköping University, Linköping, Sweden
| | - Paul G Winyard
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, St Luke's Campus, Exeter, UK
| | - Mario Galgani
- Institute for Experimental Endocrinology and Oncology 'G. Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, Università degli Studi di Napoli 'Federico II', Naples, Italy
| | - Ahuva Nissim
- Biochemical Pharmacology, William Harvey Research Institute, Queen Mary University of London, London, UK.
| |
Collapse
|
10
|
Fuentes-Lemus E, Reyes JS, López-Alarcón C, Davies MJ. Crowding modulates the glycation of plasma proteins: In vitro analysis of structural modifications to albumin and transferrin and identification of sites of modification. Free Radic Biol Med 2022; 193:551-566. [PMID: 36336230 DOI: 10.1016/j.freeradbiomed.2022.10.319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/20/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Protein modification occurs in biological milieus that are characterized by high concentrations of (macro)molecules (i.e. heterogeneous and packed environments). Recent data indicate that crowding can modulate the extent and rate of protein oxidation, however its effect on other post-translational modifications remains to be explored. In this work we hypothesized that crowding would affect the glycation of plasma proteins. Physiologically-relevant concentrations of albumin (35 mg mL-1) and transferrin (2 mg mL-1) were incubated with methylglyoxal and glyoxal (5 μM-5 mM), two α-oxoaldehyde metabolites that are elevated in the plasma of people with diabetes. Crowding was induced by adding dextran or ficoll polymers. Electrophoresis, electron microscopy, fluorescence spectroscopy and mass spectrometry were employed to investigate the structural consequences of glycation under crowded conditions. Our data demonstrate that crowding modulates the extent of formation of transferrin cross-links, and also the modification pathways in both albumin and transferrin. Arginine was the most susceptible residue to modification, with lysine and cysteine also affected. Loss of 0.48 and 7.28 arginine residues per protein molecule were determined on incubation with 500 μM methylglyoxal for albumin and transferrin, respectively. Crowding did not influence the extent of loss of arginine and lysine for either protein, but the sites of modification, detected by LC-MS, were different between dilute and crowded conditions. These data confirm the relevance of studying modification processes under conditions that closely mimic biological milieus. These data unveil additional factors that influence the pattern and extent of protein modification, and their structural consequences, in biological systems.
Collapse
Affiliation(s)
- Eduardo Fuentes-Lemus
- Department of Biomedical Sciences, Panum Institute, Blegdamsvej 3, University of Copenhagen, Copenhagen, 2200, Denmark.
| | - Juan S Reyes
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Camilo López-Alarcón
- Facultad de Química y de Farmacia, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, Blegdamsvej 3, University of Copenhagen, Copenhagen, 2200, Denmark.
| |
Collapse
|
11
|
Dos Santos L, Bertoli SR, Ávila RA, Marques VB. Iron overload, oxidative stress and vascular dysfunction: Evidences from clinical studies and animal models. Biochim Biophys Acta Gen Subj 2022; 1866:130172. [PMID: 35597504 DOI: 10.1016/j.bbagen.2022.130172] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 03/07/2022] [Accepted: 05/13/2022] [Indexed: 11/18/2022]
Abstract
Although iron is a metal involved in many in vital processes due to its redox capacity, body iron overloads lead to tissue damage, including the cardiovascular system. While cardiomyopathy was the focus since the 1960s, the impact on the vasculature was comparatively neglected for about 40 years, when clinical studies correlating iron overload, oxidative stress, endothelial dysfunction, arterial stiffness and atherosclerosis reinforced an "iron hypothesis". Due to controversial results from some epidemiological studies investigating atherosclerotic events and iron levels, well-controlled trials and animal studies provided essential data about the influence of iron, per se, on the vasculature. As a result, the pathophysiology of vascular dysfunction in iron overload have been revisited. This review summarizes the knowledge obtained from epidemiological studies, animal models and "in vitro" cellular systems in recent decades, highlighting a more harmful than innocent role of iron excess for the vascular homeostasis, which supports our proposal to hereafter denominate "iron overload vasculopathy". Additionally, evidence-based therapeutic targets are pointed out to be tested in pre-clinical research that may be useful in cardiovascular protection for patients with iron overload syndromes.
Collapse
Affiliation(s)
- Leonardo Dos Santos
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, ES, Brazil.
| | - Sabrina Rodrigues Bertoli
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, ES, Brazil; Faculdade Novo Milenio, Vila Velha, ES, Brazil
| | - Renata Andrade Ávila
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitória, ES, Brazil; Faculdades Integradas São Pedro (FAESA), Vitória, ES, Brazil
| | | |
Collapse
|
12
|
Jin Y, Kim D, Choi YJ, Song I, Chung YS. Gene Network Analysis for Osteoporosis, Sarcopenia, Diabetes, and Obesity in Human Mesenchymal Stromal Cells. Genes (Basel) 2022; 13:genes13030459. [PMID: 35328013 PMCID: PMC8953569 DOI: 10.3390/genes13030459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
The systemic gene interactions that occur during osteoporosis and their underlying mechanisms remain to be determined. To this end, mesenchymal stromal cells (MSCs) were analyzed from bone marrow samples collected from healthy individuals (n = 5) and patients with osteoporosis (n = 5). A total of 120 osteoporosis-related genes were identified using RNA-sequencing (RNA-seq) and Ingenuity Pathway Analysis (IPA) software. In order to analyze these genes, we constructed a heatmap of one-way hierarchical clustering and grouped the gene expression patterns of the samples. The MSCs from one control participant showed a similar expression pattern to that observed in the MSCs of three patients with osteoporosis, suggesting that the differentiating genes might be important genetic determinants of osteoporosis. Then, we selected the top 38 genes based on fold change and expression, excluding osteoporosis-related genes from the control participant. We identified a network among the top 38 genes related to osteoblast and osteoclast differentiation, bone remodeling, osteoporosis, and sarcopenia using the Molecule Activity Predictor program. Among them, 25 genes were essential systemic genes involved in osteoporosis. Furthermore, we identified 24 genes also associated with diabetes and obesity, among which 10 genes were involved in a network related to bone and energy metabolism. The study findings may have implications for the treatment and prevention of osteoporosis.
Collapse
Affiliation(s)
- Yilan Jin
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Korea; (Y.J.); (Y.J.C.); (I.S.)
- Ajou Institute on Aging, Ajou University Medical Center, Suwon 16499, Korea
| | - Dowan Kim
- Ajou Translational OMICS Center, Ajou University School of Medicine, Suwon 16499, Korea;
- Department of Medical Sciences, Ajou University Graduate School of Medicine, Suwon 16499, Korea
| | - Yong Jun Choi
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Korea; (Y.J.); (Y.J.C.); (I.S.)
- Ajou Institute on Aging, Ajou University Medical Center, Suwon 16499, Korea
| | - Insun Song
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Korea; (Y.J.); (Y.J.C.); (I.S.)
- Ajou Institute on Aging, Ajou University Medical Center, Suwon 16499, Korea
| | - Yoon-Sok Chung
- Department of Endocrinology and Metabolism, Ajou University School of Medicine, Suwon 16499, Korea; (Y.J.); (Y.J.C.); (I.S.)
- Ajou Institute on Aging, Ajou University Medical Center, Suwon 16499, Korea
- Correspondence:
| |
Collapse
|
13
|
He XC, Chen HY, Qiu Y, Tian L, Bao BS, Hao XP, Chen YH. Associations of iron status with breast cancer risk factors in adult women: Findings from National Health and Nutrition Examination Survey 2017-2018. J Trace Elem Med Biol 2021; 68:126867. [PMID: 34592676 DOI: 10.1016/j.jtemb.2021.126867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 08/24/2021] [Accepted: 09/22/2021] [Indexed: 11/25/2022]
Abstract
OBJECTIVE This study examined the association between iron status and a set of breast cancer risk factors among U.S. adult women aged 20-80 years. METHODS Data from National Health and Nutrition Examination Survey (2017-2018) were used to examine the relation between serum ferritin, serum iron and transferrin saturation with a set of breast cancer risk factors [body mass index (BMI), waist circumference, glycosylated hemoglobin (HbA1c), fasting plasma glucose, insulin and HOMA-IR]. The multivariable linear regressions were used controlling for age, race/ethnicity, menopause status, education level, smoking status, alcohol consumption, physical activity, high-sensitivity C-reactive protein (hsCRP) and total energy intake. RESULTS HbA1c, BMI and waist circumference data were available for 1902 women with a fasting sample (n = 913) for fasting plasma glucose, insulin and HOMA-IR. Transferrin saturation had significant, inverse associations with BMI, waist circumference and HbA1c. The size of difference observed were that participants in the fourth quartile of transferrin saturation had a 4.50 kg/m2 smaller BMI, a 9.36 cm smaller waist circumference and a 0.1 % lower HbA1c level than participants in the first quartile. Similarly, serum iron concentrations were inversely associated with BMI and waist circumference. In addition, serum iron had significant, inverse associations with insulin and HOMA-IR. Sensitivity analyses among men gave similar results. For serum ferritin, there was a trend towards a positive association between waist circumference, HbA1c and fasting plasma glucose with serum ferritin. However, the associations did not reach statistical significance among women. CONCLUSIONS Iron status may impact breast cancer risk via effects on adiposity or glucose metabolism. The findings should be confirmed with further prospective data.
Collapse
Affiliation(s)
- Xiao-Chong He
- Department of Nursing Administration, Army Medical University, Chongqing, 400038, China.
| | - Hong-Ye Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Yue Qiu
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Lin Tian
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Bao-Shi Bao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiao-Peng Hao
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| | - Yu-Hui Chen
- Department of General Surgery, The First Medical Center, Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
14
|
Bi Y, Ajoolabady A, Demillard LJ, Yu W, Hilaire ML, Zhang Y, Ren J. Dysregulation of iron metabolism in cardiovascular diseases: From iron deficiency to iron overload. Biochem Pharmacol 2021; 190:114661. [PMID: 34157296 DOI: 10.1016/j.bcp.2021.114661] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 11/19/2022]
Abstract
Iron deficiency and iron overload are the most prevalent and opposite forms of dysregulated iron metabolism that affect approximately 30 percent of the world population, in particularly, elderly and patients with chronic diseases. Both iron deficiency and overload are frequently observed in a wide range of cardiovascular diseases, contributing to the onset and progression of these diseases. One of the devastating seqeulae for iron overload is the induction of ferroptosis, a newly defined form of regulated cell death which heavily impacts cardiac function through ferroptotic cell death in cardiomyocytes. In this review, we will aim to evaluate iron deficiency and iron overload in cardiovascular diseases. We will summarize current therapeutic strategies to tackle iron deficiency and iron overload, major pitfalls of current studies, and future perspectives.
Collapse
Affiliation(s)
- Yaguang Bi
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Amir Ajoolabady
- School of Pharmacy and Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Laurie J Demillard
- School of Pharmacy and Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Wenjun Yu
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Michelle L Hilaire
- School of Pharmacy and Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY 82071, USA
| | - Yingmei Zhang
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai 200032, China; Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
15
|
Iron overload-induced oxidative stress in myelodysplastic syndromes and its cellular sequelae. Crit Rev Oncol Hematol 2021; 163:103367. [PMID: 34058341 DOI: 10.1016/j.critrevonc.2021.103367] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 03/30/2021] [Accepted: 05/03/2021] [Indexed: 12/14/2022] Open
Abstract
The myelodysplastic syndromes (MDS) are clonal hematopoietic stem cell disorders. MDS patients often require red blood cell transfusions, resulting in iron overload (IOL). IOL increases production of reactive oxygen species (ROS), oxygen free radicals. We review and illustrate how IOL-induced ROS influence cellular activities relevant to MDS pathophysiology. ROS damage lipids, nucleic acids in mitochondrial and nuclear DNA, structural proteins, transcription factors and enzymes. Cellular consequences include decreased metabolism and tissue and organ dysfunction. In hematopoietic stem cells (HSC), consequences of ROS include decreased glycolysis, shifting the cell from anaerobic to aerobic metabolism and causing HSC to exit the quiescent state, leading to HSC exhaustion or senescence. ROS oxidizes DNA bases, resulting in accumulation of mutations. Membrane oxidation alters fluidity and permeability. In summary, evidence indicates that IOL-induced ROS alters cellular signaling pathways resulting in toxicity to organs and hematopoietic cells, in keeping with adverse clinical outcomes in MDS.
Collapse
|
16
|
Conway RBN, Sudenga S, McClain D, Blot WJ. Diabetes and liver cancer risk: A stronger effect in Whites than Blacks? J Diabetes Complications 2021; 35:107816. [PMID: 33323327 PMCID: PMC8045414 DOI: 10.1016/j.jdiacomp.2020.107816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 11/10/2020] [Accepted: 11/11/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Both diabetes and liver cancer are overrepresented among African Americans, but limited information is available on the interrelationship of these two diseases among African Americans. We examined the association of diabetes with the incidence of liver cancer and whether this varied by participant self-reported race/ethnicity. METHODS Using the Southern Community Cohort Study, we conducted a cancer follow up (2002-2016) of a cohort of mostly low-income participants aged 40-79 with diabetes (n = 15,879) and without diabetes (n = 59,077) at study baseline. Cox regression was used to compute Hazard Ratios (HR) and 95% CIs for the risk of incident liver cancer. RESULTS With 790,132 person years of follow up, 320 incident cases of liver cancer were identified. In analyses controlling for age, sex, race, BMI, current and former smoking, total alcohol consumption, family history of liver cancer, any hepatitis infection, hyperlipidemia and socioeconomic factors, the association between diabetes and risk of liver cancer differed significantly (pinteraction = 0.0001) between participants identifying as Black/African American (AA) or White/European American (EA). Diabetes was associated with 5.3-fold increased cancer risk among EAs (HR 5.4, 95% CI 3.2-9.3) vs an 80% increase (HR 1.8, 95% CI 1.3-2.5) among AAs. Furthermore, controlling for diabetes greatly attenuated the higher risk of liver cancer among AAs; indeed, while the cancer risk among those without diabetes was twice as high among AAs than EAs (HR = 2.0, 95% CI = 1.4-2.9), no excess in AAs was observed among those with diabetes (HR = 0.7, 95% CI = 0.4-1.1). CONCLUSION While liver cancer risk in general is greater in AAs than EAs and diabetes increases this risk in both racial/ethnic groups, diabetes appears to impact liver cancer to a much greater extent among EAs. The findings raise the possibility of racially different mechanisms and impacts of diabetes on this often fatal cancer among AAs and EAs.
Collapse
Affiliation(s)
- Rebecca Baqiyyah N Conway
- School of Community and Rural Health, University of Texas Health Science Center at Tyler, Tyler, TX, United States of America.
| | - Staci Sudenga
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| | - Donald McClain
- Section of Endocrinology and Metabolism, Wake Forest School of Medicine, Winston-Salem, NC, United States of America
| | - William J Blot
- Division of Epidemiology, Vanderbilt University Medical Center, Nashville, TN, United States of America
| |
Collapse
|
17
|
Mitochondrial Carriers Regulating Insulin Secretion Profiled in Human Islets upon Metabolic Stress. Biomolecules 2020; 10:biom10111543. [PMID: 33198243 PMCID: PMC7697104 DOI: 10.3390/biom10111543] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/28/2020] [Accepted: 11/10/2020] [Indexed: 12/27/2022] Open
Abstract
Chronic exposure of β-cells to nutrient-rich metabolic stress impairs mitochondrial metabolism and its coupling to insulin secretion. We exposed isolated human islets to different metabolic stresses for 3 days: 0.4 mM oleate or 0.4 mM palmitate at physiological 5.5 mM glucose (lipotoxicity), high 25 mM glucose (glucotoxicity), and high 25 mM glucose combined with 0.4 mM oleate and/or palmitate (glucolipotoxicity). Then, we profiled the mitochondrial carriers and associated genes with RNA-Seq. Diabetogenic conditions, and in particular glucotoxicity, increased expression of several mitochondrial solute carriers in human islets, such as the malate carrier DIC, the α-ketoglutarate-malate exchanger OGC, and the glutamate carrier GC1. Glucotoxicity also induced a general upregulation of the electron transport chain machinery, while palmitate largely counteracted this effect. Expression of different components of the TOM/TIM mitochondrial protein import system was increased by glucotoxicity, whereas glucolipotoxicity strongly upregulated its receptor subunit TOM70. Expression of the mitochondrial calcium uniporter MCU was essentially preserved by metabolic stresses. However, glucotoxicity altered expression of regulatory elements of calcium influx as well as the Na+/Ca2+ exchanger NCLX, which mediates calcium efflux. Overall, the expression profile of mitochondrial carriers and associated genes was modified by the different metabolic stresses exhibiting nutrient-specific signatures.
Collapse
|
18
|
Vaquero MP, Martínez-Maqueda D, Gallego-Narbón A, Zapatera B, Pérez-Jiménez J. Relationship between iron status markers and insulin resistance: an exploratory study in subjects with excess body weight. PeerJ 2020; 8:e9528. [PMID: 32821534 PMCID: PMC7397981 DOI: 10.7717/peerj.9528] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background Controversy exists on the relationship between iron metabolism and cardiometabolic risk. The aim of this study was to determine if there is a link between dysmetabolic iron and cardiometabolic markers in subjects with excess body weight. Methods Cross-sectional study with fifty participants presenting overweight or obesity and at least another metabolic syndrome factor. Determinations: anthropometry, body composition, blood pressure, lipids, glucose, insulin, leptin, areas under the curve (AUC) for glucose and insulin after an oral glucose tolerance test, hs-C reactive protein (hs-CRP), blood count, ferritin, transferrin, transferrin saturation (TSAT), soluble transferrin receptor (sTfR). Gender-adjusted linear correlations and two independent samples t tests were used. Results Ferritin was positively correlated with insulin-AUC (r = 0.547, p = 0.008) and TSAT was negatively correlated with waist-hip ratio (r = − 0.385, p = 0.008), insulin (r = − 0.551, p < 0.001), and insulin resistance (HOMA-IR, r = − 0.586, p < 0.001). Subjects with TSAT ≤ 20% had higher insulin (p = 0.012) and HOMA-IR (p = 0.003) compared to those with TSAT > 20%. In conclusion, the observed results suggest that iron transport and storage are altered in subjects with overweight/obesity, at the same time that they exhibit the characteristic features of insulin resistance. Nevertheless, this occurs without iron overload or deficiency. These results should be validated in wider cohorts since they suggest that iron transport and storage should be assessed when performing the clinical evaluation of subjects with excess body weight.
Collapse
Affiliation(s)
- M Pilar Vaquero
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| | - Daniel Martínez-Maqueda
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain.,Madrid Institute for Rural, Agricultural and Food Research and Development (IMIDRA), Madrid, Spain
| | - Angélica Gallego-Narbón
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain.,Department of Biology, Universidad Autónoma de Madrid (UAM), Madrid, Spain, España
| | - Belén Zapatera
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| | - Jara Pérez-Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| |
Collapse
|
19
|
Carrivick S, Alfonso H, Golledge J, Clancy P, Flicker L, Gunton JE, Hankey GJ, Almeida OP, Norman PE, Yeap BB. Differential associations of ferritin and 25-hydroxyvitamin D with fasting glucose and diabetes risk in community dwelling older men. Diabetes Metab Res Rev 2019; 35:e3172. [PMID: 30997951 DOI: 10.1002/dmrr.3172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/23/2019] [Accepted: 04/07/2019] [Indexed: 12/29/2022]
Abstract
AIMS We examined associations of ferritin and 25-hydroxyvitamin D with fasting glucose and prevalent diabetes in older men. METHODS Cross-sectional analysis of 4153 community-dwelling men aged 70 to 89 years in Western Australia. Plasma ferritin, 25-hydroxyvitamin D, and glucose were assayed. Diabetes was ascertained from self-report, medications, and fasting glucose. RESULTS There were 577 men with diabetes (13.9%). In the whole cohort, ferritin was associated with fasting glucose (0.051 mmol/L per 1 SD increase in ferritin, P = .006) and 25-hydroxyvitamin D was inversely associated (-0.085 mmol/L per 1 SD, P < .001). Ferritin was not associated with prevalent diabetes (highest vs. lowest quartile; >225 vs <66 μg/L: adjusted odds ratio [OR] 0.97, 95% confidence interval [CI], 0.74-1.27, P = .83). Higher vitamin D was associated with decreased odds of prevalent diabetes (highest vs lowest quartile; >82 nmol/L vs <53 nmol/L: OR = 0.57, 95% CI = 0.43-0.75, P < .001). There was no interaction between ferritin and vitamin D on diabetes risk. CONCLUSIONS In older men, ferritin is associated with fasting glucose but not prevalent diabetes. Higher 25-hydroxyvitamin D concentrations are independently associated with lower fasting glucose and reduced risk of diabetes. Clinical trials are required to determine whether interventions, which raise vitamin D concentrations, would reduce incidence of diabetes in this expanding demographic group.
Collapse
Affiliation(s)
- Simon Carrivick
- Medical School, University of Western Australia, Perth, Australia
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia
| | - Helman Alfonso
- School of Public Health, Curtin University, Perth, Australia
| | - Jonathan Golledge
- Vascular Biology Unit, Queensland Research Centre for Peripheral Vascular Disease, School of Medicine, James Cook University, Townsville, Australia
- Department of Vascular and Endovascular Surgery, Townsville Hospital, Townsville, Australia
| | - Paula Clancy
- Health Practitioners And Researchers Together-Blood Endothelium And Tissue (HART-BEAT), Townsville Hospital, Townsville, Australia
| | - Leon Flicker
- Medical School, University of Western Australia, Perth, Australia
- Western Australian Centre for Health and Ageing, Centre for Medical Research, University of Western Australia, Perth, Australia
| | - Jenny E Gunton
- Westmead Hospital, The University of Sydney, Sydney, Australia
| | - Graeme J Hankey
- Medical School, University of Western Australia, Perth, Australia
| | - Osvaldo P Almeida
- Medical School, University of Western Australia, Perth, Australia
- Western Australian Centre for Health and Ageing, Centre for Medical Research, University of Western Australia, Perth, Australia
| | - Paul E Norman
- Medical School, University of Western Australia, Perth, Australia
| | - Bu B Yeap
- Medical School, University of Western Australia, Perth, Australia
- Department of Endocrinology and Diabetes, Fiona Stanley Hospital, Perth, Australia
| |
Collapse
|
20
|
Thorsen SU, Pipper CB, Ellervik C, Pociot F, Kyvsgaard JN, Svensson J. Association between Neonatal Whole Blood Iron Content and Cytokines, Adipokines, and Other Immune Response Proteins. Nutrients 2019; 11:nu11030543. [PMID: 30836628 PMCID: PMC6470999 DOI: 10.3390/nu11030543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 02/23/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022] Open
Abstract
(1) Background: High iron associates with inflammation and type 1 diabetes (T1D). Iron is essential not only for neonatal development but also for infectious microorganisms. The neonatal immune system is immature, and innate immunity prevails before immunocompetence develops. (2) Methods: In 398 newborns from the Danish Newborn Screening Biobank, we examined if whole blood iron (WB-Iron) content were associated with cytokines, adipokines, C-reactive protein (CRP), and mannose-binding lectin (MBL) in non-infected healthy neonates, and if these associations differed in newborns who later developed T1D (cases) (n = 199). WB-Iron was quantified using laser ablation inductively coupled plasma mass spectrometry on the neonatal dried blood spots. For each analyte, the relative change (RC) in the mean level was modeled by robust log-normal regression. (3) Results: A one unit increase in neonatal WB-Iron was associated with a 38% decrease in mean interleukin (IL)-6 levels (0.62; 95% CI: 0.40–0.95, p = 0.03), and a 37% decrease in mean MBL levels (0.63; 95% CI: 0.41–0.95, p = 0.03), but was not statistically significant after correction for multiple testing. (4) Conclusions: In summary, we found that higher neonatal WB-iron content was inversely associated with IL-6 and MBL, which may increase susceptibility to infections.
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, Copenhagen, Oester Farimagsgade 5, 1710 Copenhagen K, Denmark.
| | - Christina Ellervik
- Department of Production, Research, and Innovation, Region Zealand, Alleen 15, 4180 Sorø, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, 2200 Copenhagen N, Denmark.
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| | - 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.
- Steno Diabetes Center Copenhagen, Niels Steensensvej, 2820 Gentofte, Denmark.
| | - Julie N Kyvsgaard
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev Hospital, University of Copenhagen, Herlev Ringvej 75, 2730 Herlev, 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
|
21
|
Cejvanovic V, Kjær LK, Mørup Bergholdt HK, Henriksen T, Weimann A, Ellervik C, Poulsen HE. RNA oxidation and iron levels in patients with diabetes. Free Radic Biol Med 2018; 129:532-536. [PMID: 30339885 DOI: 10.1016/j.freeradbiomed.2018.10.420] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 09/10/2018] [Accepted: 10/09/2018] [Indexed: 12/11/2022]
Abstract
AIM The urinary biomarker for oxidative stress to RNA, 8-oxo-7,8-dihydro-guanosine (8-oxoGuo) is associated with mortality in patients with type 2 diabetes. Iron has also been linked to diabetes. In individuals with untreated hereditary iron overload it has been observed that 8-oxoGuo was higher compared to controls. In the current study, we hypothesized that 8-oxoGuo was associated with diagnosis of diabetes, and that iron confounded this association. METHODS Participants from a general Danish population were included in the study (n = 3567). UPLC-MS/MS method was used for 8-oxoGuo (nmol/mmol creatinine) measurement in spot urine. Iron biomarkers included total plasma iron, ferritin, transferrin saturation (TS) and transferrin. RESULTS 8-oxoGuo was 17% higher in diabetes patients (n = 208) compared to non-diabetes controls. Unadjusted logistic regression model showed an odds ratio of diabetes of 1.38 (95%CI:1.21-1.57, P < 0.0001) per unit increase of 8-oxoGuo. When the model was adjusted for possible confounders the odds ratio was 1.09 (95%CI:0.94-1.26, P = 0.24). When additional adjustment was performed including ferritin, TS, or transferrin, respectively, the OR were 1.14 (95%CI:0.97-1.33, P = 0.09), 1.10 (95%CI: 0.95-1.28, P = 0.18), and 1.17 (95%CI:1.01-1.38, P = 0.04). CONCLUSIONS Our study indicates that 8-oxoGuo is higher in diabetes patients. The lack of association between 8-oxoGuo and diabetes in the adjusted model may be due to the cross-sectional design including post-treatment bias. Our data did not show consistent effect of all iron biomarkers in relation to diabetes. Most likely, the iron biomarkers were affected by inflammation thus not reflecting true iron levels.
Collapse
Affiliation(s)
- Vanja Cejvanovic
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark; University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark.
| | - Laura Kofoed Kjær
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark; University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| | | | - Trine Henriksen
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Allan Weimann
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Christina Ellervik
- University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark; Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Production, Research and Innovation, Region Zealand, Denmark
| | - Henrik Enghusen Poulsen
- Department of Clinical Pharmacology, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark; University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen, Denmark
| |
Collapse
|
22
|
Hansen JB, Dos Santos LRB, Liu Y, Prentice KJ, Teudt F, Tonnesen M, Jonas JC, Wheeler MB, Mandrup-Poulsen T. Glucolipotoxic conditions induce β-cell iron import, cytosolic ROS formation and apoptosis. J Mol Endocrinol 2018; 61:69-77. [PMID: 30030388 DOI: 10.1530/jme-17-0262] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/16/2018] [Indexed: 12/28/2022]
Abstract
Type 2 diabetes (T2D) arises when the pancreatic beta-cell fails to compensate for increased insulin needs due to insulin resistance. Glucolipotoxicity (GLT) has been proposed to induce beta-cell dysfunction in T2D by formation of reactive oxygen species (ROS). Here, we examined if modeling glucolipotoxic conditions by high glucose-high free fatty acid (FFA) exposure (GLT) regulates beta-cell iron transport, by increasing the cytosolic labile iron pool (LIP). In isolated mouse islets, the GLT-induced increase in the LIP catalyzed cytosolic ROS formation and induced apoptosis. We show that GLT-induced ROS production is regulated by an increased LIP associated with elevated expression of genes regulating iron import. Using pharmacological and transgenic approaches, we show that iron reduction and decreased iron import protects from GLT-induced ROS production, prevents impairment of the mitochondrial membrane potential (MMP) and inhibits apoptosis. This study identifies a novel pathway underlying GLT-induced apoptosis involving increased iron import, generation of hydroxyl radicals from hydrogen peroxide through the Fenton reaction in the cytosolic compartment associated with dissipation of the MMP and beta-cell apoptosis.
Collapse
Affiliation(s)
- Jakob Bondo Hansen
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Laila Romagueira Bichara Dos Santos
- Université Catholique de Louvain, Institute of Experimental and Clinical Research, Pole of Endocrinology, Diabetes and Nutrition, Brussels, Belgium
| | - Ying Liu
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Kacey J Prentice
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Frederik Teudt
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Morten Tonnesen
- Department of Diabetes Complications Biology & Pharmacology, Novo Nordisk, Måløv, Denmark
| | - Jean-Christophe Jonas
- Université Catholique de Louvain, Institute of Experimental and Clinical Research, Pole of Endocrinology, Diabetes and Nutrition, Brussels, Belgium
| | - Michael B Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
23
|
Størdal K, McArdle HJ, Hayes H, Tapia G, Viken MK, Lund-Blix NA, Haugen M, Joner G, Skrivarhaug T, Mårild K, Njølstad PR, Eggesbø M, Mandal S, Page CM, London SJ, Lie BA, Stene LC. Prenatal iron exposure and childhood type 1 diabetes. Sci Rep 2018; 8:9067. [PMID: 29899542 PMCID: PMC5998022 DOI: 10.1038/s41598-018-27391-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/23/2018] [Indexed: 02/06/2023] Open
Abstract
Iron overload due to environmental or genetic causes have been associated diabetes. We hypothesized that prenatal iron exposure is associated with higher risk of childhood type 1 diabetes. In the Norwegian Mother and Child cohort study (n = 94,209 pregnancies, n = 373 developed type 1 diabetes) the incidence of type 1 diabetes was higher in children exposed to maternal iron supplementation than unexposed (36.8/100,000/year compared to 28.6/100,000/year, adjusted hazard ratio 1.33, 95%CI: 1.06-1.67). Cord plasma biomarkers of high iron status were non-significantly associated with higher risk of type 1 diabetes (ferritin OR = 1.05 [95%CI: 0.99-1.13] per 50 mg/L increase; soluble transferrin receptor: OR = 0.91 [95%CI: 0.81-1.01] per 0.5 mg/L increase). Maternal but not fetal HFE genotypes causing high/intermediate iron stores were associated with offspring diabetes (odds ratio: 1.45, 95%CI: 1.04, 2.02). Maternal anaemia or non-iron dietary supplements did not significantly predict type 1 diabetes. Perinatal iron exposures were not associated with cord blood DNA genome-wide methylation, but fetal HFE genotype was associated with differential fetal methylation near HFE. Maternal cytokines in mid-pregnancy of the pro-inflammatory M1 pathway differed by maternal iron supplements and HFE genotype. Our results suggest that exposure to iron during pregnancy may be a risk factor for type 1 diabetes in the offspring.
Collapse
Affiliation(s)
- Ketil Størdal
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway.
- Pediatric Department, Ostfold Hospital Trust, Fredrikstad, Norway.
| | - Harry J McArdle
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - Helen Hayes
- The Rowett Institute of Nutrition and Health, University of Aberdeen, Foresterhill, Aberdeen, Scotland, UK
| | - German Tapia
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Marte K Viken
- Department of Medical Genetics, University of Oslo, Oslo University Hospital, Ullevål, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Nicolai A Lund-Blix
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
- Department of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Margaretha Haugen
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Geir Joner
- Department of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Torild Skrivarhaug
- Department of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Karl Mårild
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
| | - Pål R Njølstad
- Department of Paediatrics and Adolescent Medicine, Haukeland University Hospital, Bergen, Norway
- KG Jebsen Center for Diabetes Research, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Merete Eggesbø
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Siddhartha Mandal
- Department of Environmental Exposure and Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Christian M Page
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
- Centre for Biostatistics and Epidemiology, Oslo University Hospital, Oslo, Norway
| | - Stephanie J London
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, Durham, NC, 27709, USA
| | - Benedicte A Lie
- Department of Medical Genetics, University of Oslo, Oslo University Hospital, Ullevål, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Lars C Stene
- Department of non-communicable diseases, Norwegian Institute of Public Health, Oslo, Norway
| |
Collapse
|
24
|
Iron Status and Gestational Diabetes-A Meta-Analysis. Nutrients 2018; 10:nu10050621. [PMID: 29762515 PMCID: PMC5986501 DOI: 10.3390/nu10050621] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 04/30/2018] [Accepted: 05/11/2018] [Indexed: 12/21/2022] Open
Abstract
A meta-analysis of the association of iron overload with gestational diabetes mellitus (GDM) may inform the health debate. We performed a meta-analysis investigating the association of iron biomarkers and dietary iron exposure with GDM. We identified 33 eligible studies (N = 44,110) published in 2001–2017. The standardized mean differences (SMD) in women who had GDM compared to pregnant women without were 0.25 µg/dL (95% CI: 0.001–0.50) for iron, 1.54 ng/mL (0.56–2.53) for ferritin, 1.05% (0.02 to 2.08) for transferrin saturation, and 0.81 g/dL (0.40–1.22) for hemoglobin. Adjusted odds ratio for GDM were 1.58 (95% CI: 1.20–2.08) for ferritin, 1.30 (1.01–1.67) for hemoglobin, and 1.48 (1.29–1.69) for dietary heme intake. We did not find any differences in TIBC or transferrin concentration in women with and without GDM. We also did not find any association of increased transferrin receptor or increased intake of total dietary iron, non-heme iron or supplemental iron, with increased odds ratios for GDM. Considerable heterogeneity was present among the studies (0–99%), but no evidence of publication bias. Accumulating evidence suggests that circulating and dietary iron biomarkers among pregnant women are associated with GDM, but the results should be interpreted with caution due to the high heterogeneity of analyses. Randomized trials investigating the benefits of iron reduction in women at high risk for GDM are warranted.
Collapse
|
25
|
Vela D, Sopi RB, Mladenov M. Low Hepcidin in Type 2 Diabetes Mellitus: Examining the Molecular Links and Their Clinical Implications. Can J Diabetes 2018; 42:179-187. [DOI: 10.1016/j.jcjd.2017.04.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 04/18/2017] [Accepted: 04/21/2017] [Indexed: 01/14/2023]
|
26
|
Heltberg A, Andersen JS, Sandholdt H, Siersma V, Kragstrup J, Ellervik C. Predictors of undiagnosed prevalent type 2 diabetes - The Danish General Suburban Population Study. Prim Care Diabetes 2018; 12:13-22. [PMID: 28964672 DOI: 10.1016/j.pcd.2017.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/26/2017] [Accepted: 08/29/2017] [Indexed: 02/01/2023]
Abstract
AIMS To investigate how self-reported risk factors (including socioeconomic status) predict undiagnosed, prevalent type 2 diabetes mellitus (T2DM). To externally validate Leicester Risk Assessment Score (LRAS), Finnish Diabetes Risk Score (FINDRISC) and Danish Diabetes Risk Score (DDRS), and to investigate how these predict a European Heart SCORE≥5% in a Danish population study. METHODS We included 21,205 adults from the Danish General Suburban Population Study. We used relative importance calculations of self-reported variables in prediction of undiagnosed T2DM. We externally validated established prediction models reporting ROC-curves for undiagnosed T2DM, pre-diabetes and SCORE. RESULTS More than 20% of people with T2DM were undiagnosed. The 7 most important self-rated predictors in sequential order were high BMI, antihypertensive-therapy, age, cardiovascular disease, waist-circumference, fitness compared to peers and family disposition for T2DM. The Area Under the Curve for prediction of undiagnosed T2DM was 77.1 for LRAS; 75.4 for DDRS and 67.9 for FINDRISC. AUCs for SCORE was 75.1 for LRAS; 62.3 for DDRS and 54.3 for FINDRISC. CONCLUSIONS BMI and self-reported cardiovascular disease are important risk factors for undiagnosed T2DM. LRAS performed better than DDRS and FINDRISC in prediction of undiagnosed T2DM and SCORE≥5%. SCORE performed best in predicting pre-diabetes.
Collapse
Affiliation(s)
- Andreas Heltberg
- Section of General Practice, Department of Public Health and Research Unit for General Practice, University of Copenhagen, Denmark.
| | - John Sahl Andersen
- Section of General Practice, Department of Public Health and Research Unit for General Practice, University of Copenhagen, Denmark
| | - Håkon Sandholdt
- Section of General Practice, Department of Public Health and Research Unit for General Practice, University of Copenhagen, Denmark
| | - Volkert Siersma
- Section of General Practice, Department of Public Health and Research Unit for General Practice, University of Copenhagen, Denmark
| | - Jakob Kragstrup
- Section of General Practice, Department of Public Health and Research Unit for General Practice, University of Copenhagen, Denmark
| | - Christina Ellervik
- Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark; Department of Laboratory Medicine, Boston Children's Hospital & Harvard Medical School, Boston, MA, USA; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Denmark
| |
Collapse
|
27
|
Kyvsgaard JN, Overgaard AJ, Thorsen SU, Hansen TH, Pipper CB, Mortensen HB, Pociot F, Svensson J. High Neonatal Blood Iron Content Is Associated with the Risk of Childhood Type 1 Diabetes Mellitus. Nutrients 2017; 9:nu9111221. [PMID: 29113123 PMCID: PMC5707693 DOI: 10.3390/nu9111221] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 11/01/2017] [Accepted: 11/03/2017] [Indexed: 12/30/2022] Open
Abstract
(1) Background: Iron requirement increases during pregnancy and iron supplementation is therefore recommended in many countries. However, excessive iron intake may lead to destruction of pancreatic β-cells. Therefore, we aim to test if higher neonatal iron content in blood is associated with the risk of developing type 1 diabetes mellitus (T1D) in childhood; (2) Methods: A case-control study was conducted, including 199 children diagnosed with T1D before the age of 16 years from 1991 to 2005 and 199 controls matched on date of birth. Information on confounders was available in 181 cases and 154 controls. Iron was measured on a neonatal single dried blood spot sample and was analyzed by laser ablation inductively coupled plasma mass spectrometry. Multivariate logistic regression was used to evaluate if iron content in whole blood was associated with the risk of T1D; (3) Results: A doubling of iron content increased the odds of developing T1D more than two-fold (odds ratio (95% CI), 2.55 (1.04; 6.24)). Iron content increased with maternal age (p = 0.04) and girls had higher content than boys (p = 0.01); (4) Conclusions: Higher neonatal iron content associates to an increased risk of developing T1D before the age of 16 years. Iron supplementation during early childhood needs further investigation, including the causes of high iron in neonates.
Collapse
Affiliation(s)
- Julie Nyholm Kyvsgaard
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev University Hospital, 2730 Herlev, Denmark.
| | - Anne Julie Overgaard
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev University Hospital, 2730 Herlev, Denmark.
| | - Steffen Ullitz Thorsen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev University Hospital, 2730 Herlev, Denmark.
| | - Thomas Hesselhøj Hansen
- Department of Plant and Environmental Sciences, Faculty of Science, University of Copenhagen, 2000 Frederiksberg, Denmark.
| | - Christian Bressen Pipper
- Section of Biostatistics, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, 2099 Copenhagen, Denmark.
| | - Henrik Bindesbøl Mortensen
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev University Hospital, 2730 Herlev, Denmark.
| | - Flemming Pociot
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev University Hospital, 2730 Herlev, Denmark.
| | - Jannet Svensson
- Copenhagen Diabetes Research Center (CPH-DIRECT), Department of Paediatrics, Herlev University Hospital, 2730 Herlev, Denmark.
| |
Collapse
|
28
|
Coates TD, Carson S, Wood JC, Berdoukas V. Management of iron overload in hemoglobinopathies: what is the appropriate target iron level? Ann N Y Acad Sci 2017; 1368:95-106. [PMID: 27186942 DOI: 10.1111/nyas.13060] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/04/2016] [Accepted: 03/09/2016] [Indexed: 01/19/2023]
Abstract
Patients with thalassemia become iron overloaded from increased absorption of iron, ineffective erythropoiesis, and chronic transfusion. Before effective iron chelation became available, thalassemia major patients died of iron-related cardiac failure in the second decade of life. Initial treatment goals for chelation therapy were aimed at levels of ferritin and liver iron concentrations associated with prevention of adverse cardiac outcomes and avoidance of chelator toxicity. Cardiac deaths were greatly reduced and survival was much longer. Epidemiological data from the general population draw clear associations between increased transferrin saturation (and, by inference, labile iron) and early death, diabetes, and malignant transformation. The rate of cancers now seems to be significantly higher in thalassemia than in the general population. Reduction in iron can reverse many of these complications and reduce the risk of malignancy. As toxicity can result from prolonged exposure to even low levels of excess iron, and survival in thalassemia patients is now many decades, it would seem prudent to refocus attention on prevention of long-term complications of iron overload and to maintain labile iron and total body iron levels within a normal range, if expertise and resources are available to avoid complications of overtreatment.
Collapse
Affiliation(s)
- Thomas D Coates
- Section of Hematology, Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation
| | - Susan Carson
- Section of Hematology, Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation
| | - John C Wood
- Division of Cardiology, Children's Hospital Los Angeles, University of Southern California, Keck School of Medicine, Los Angeles, California
| | - Vasilios Berdoukas
- Section of Hematology, Children's Center for Cancer, Blood Diseases and Bone Marrow Transplantation
| |
Collapse
|
29
|
Heltberg A, Andersen JS, Kragstrup J, Siersma V, Sandholdt H, Ellervik C. Social disparities in diabetes care: a general population study in Denmark. Scand J Prim Health Care 2017; 35:54-63. [PMID: 28277046 PMCID: PMC5361420 DOI: 10.1080/02813432.2017.1288702] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
OBJECTIVE We investigated the association between socioeconomic factors and the attainment of treatment goals and pharmacotherapy in patients with type 2 diabetes in Denmark. DESIGN A cross-sectional population study. SETTING The municipality of Naestved, Denmark. SUBJECTS We studied 907 patients with type 2 diabetes identified from a random sample of 21,205 Danish citizens. MAIN OUTCOME MEASURES The proportion of patients who were not achieving goals for diabetes care based on their HbA1c, LDL-cholesterol, blood pressure, and lifestyle, and the proportion of patients who were treated with antihypertensive and cholesterol- and glucose-lowering medication. METHODS We investigated the association of the socioeconomic factors such as age, gender, education, occupation, income, and civil status and attainment of treatment goals and pharmacotherapy in logistic regression analyses. We investigated effect modification of cardiovascular disease and kidney disease. RESULTS Middle age (40-65 years), low education level (i.e. basic schooling), and low household income (i.e. less than 21,400 € per year) were associated with nonattainment of goals for diabetes care. The association of socioeconomic factors with attainment of individual treatment goals varied. Patients with low socioeconomic status were more often obese, physically inactive, smoking, and had elevated blood pressure. Socioeconomic factors were not associated with treatment goals for hyperglycemia. Socioeconomic factors were inconsistently associated with pharmacotherapy. There was no difference in contacts to general practitioners according to SES. CONCLUSIONS In a country with free access to health care, the socioeconomic factors such as middle age, low education, and low income were associated with nonattainment of goals for diabetes care. KEY POINTS Middle age, low education, and low income were associated with nonattainment of goals for diabetes care, especially for lifestyle goals. Patients with low socioeconomic status were more often obese, physically inactive, smoking, and had elevated blood pressure. Association of socioeconomic factors with pharmacotherapy was inconsistent.
Collapse
Affiliation(s)
- Andreas Heltberg
- Section of General Practice, Department of Public Health and Research Unit for General Practice, Copenhagen University, Copenhagen, Denmark;
- The General Population Study, Nykøbing Falster Hospital, Nykøbing Falster, Denmark;
- CONTACT Andreas Heltberg Center for Research and Education in General Practice, Institute of Public Health, Faculty of Medicine, Copenhagen University, Denmark
| | - John Sahl Andersen
- Section of General Practice, Department of Public Health and Research Unit for General Practice, Copenhagen University, Copenhagen, Denmark;
| | - Jakob Kragstrup
- Section of General Practice, Department of Public Health and Research Unit for General Practice, Copenhagen University, Copenhagen, Denmark;
| | - Volkert Siersma
- Section of General Practice, Department of Public Health and Research Unit for General Practice, Copenhagen University, Copenhagen, Denmark;
| | - Håkon Sandholdt
- Section of General Practice, Department of Public Health and Research Unit for General Practice, Copenhagen University, Copenhagen, Denmark;
| | - Christina Ellervik
- The General Population Study, Nykøbing Falster Hospital, Nykøbing Falster, Denmark;
- Department of Laboratory Medicine, Boston Children’s Hospital Study, Boston, MA, USA;
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
- Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark
| |
Collapse
|
30
|
Iron Profile and Glycaemic Control in Patients with Type 2 Diabetes Mellitus. Med Sci (Basel) 2016; 4:medsci4040022. [PMID: 29083385 PMCID: PMC5635795 DOI: 10.3390/medsci4040022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/01/2016] [Accepted: 12/05/2016] [Indexed: 12/31/2022] Open
Abstract
Iron overload is increasingly being connected to insulin resistance in Type 2 Diabetes Mellitus (T2DM) patients. Free iron causes the assembly of reactive oxygen species that invariably steer the body’s homeostasis towards oxidative stress-mediated diabetic complications. This study aims to assess the serum iron, total iron binding capacity (TIBC), and percentage transferrin saturation (Tsat) of 150 subjects divided into three groups (I,II,III) of 50. Healthy individuals (controls) constituted Group I. Group II consisted of T2DM patients with optimal glycaemic control. T2DM patients with suboptimal glycaemic control formed group III. Mean serum free iron concentration was 105.34 ± 3.5, 107.33 ± 3.45, and 125.58 ± 3.45 μg/dL in Group I, Group II, and Group III, respectively. Mean serum TIBC concentration in Group I, Group II, and Group III was 311.39 ± 5.47, 309.63 ± 6.1, and 284.2 ± 3.18μg/dL, respectively. Mean serum transferrin saturation (%) in Group I, Group II, and Group III was 34.17 ± 1.21, 35.02 ± 1.2, and 44.39 ± 1.07, respectively. The difference between TIBC, mean serum free iron concentration, and transferrin saturation between Group I and Group III (for all, p values <0.001), as well as between Group II and Group III (p values 0.0012, 0.0015, and <0.0001, respectively) was statistically significant. The fasting plasma glucose values of Groups II and III were significantly higher than those of Group I, (p < 0.0001). Glycated haemoglobin (HbA1c) values were also shown to increase from Group I to II and then III, and the increase was highly significant (all p values <0.0001). Thus, decreased glycaemic control and an increase in the glycation of haemoglobin was the key to elevation in serum iron values and alterations in other parameters. However, a significant correlation was absent between serum iron and HbA1c (r = 0.05) and transferrin saturation (r = 0.0496) in Group III.
Collapse
|
31
|
Krisai P, Leib S, Aeschbacher S, Kofler T, Assadian M, Maseli A, Todd J, Estis J, Risch M, Risch L, Conen D. Relationships of iron metabolism with insulin resistance and glucose levels in young and healthy adults. Eur J Intern Med 2016; 32:31-7. [PMID: 27113814 DOI: 10.1016/j.ejim.2016.03.017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 02/21/2016] [Accepted: 03/18/2016] [Indexed: 12/09/2022]
Abstract
AIMS Several biomarkers within the iron metabolism pathway have been related to the occurrence of diabetes mellitus, but underlying mechanisms are unknown. The aim of our study was to investigate the differential relationships of iron metabolism with a broad range of diabetes markers in young and healthy adults. DESIGN 2160 participants aged 25 to 41years were enrolled in a population-based study. Established cardiovascular disease, diabetes or a body mass index >35kg/m(2) were exclusion criteria. Multivariable linear regression models were built to assess the associations of ferritin and transferrin saturation (TSAT) with blood levels of glucagon-like peptide-1 (GLP-1), insulin, homeostatic model assessment-insulin resistance (HOMA-IR), fasting plasma glucose (FPG) and hemoglobin A1c (HbA1c). RESULTS Median (interquartile range) age was 37 (31, 40) years. In multivariable linear regression analyses, β-coefficients (95% confidence intervals) per 1-SD increase in ferritin were 0.04 (0.02; 0.07, p=0.0008) for GLP-1, 0.06 (0.04; 0.08, p<0.0001) for insulin, 0.07 (0.04; 0.09, p<0.0001) for HOMA-IR, 0.004 (-0.00; 0.01, p=0.07) for FPG and -0.003 (-0.01; -0.00, p=0.07) for HbA1c. β-coefficients (95% CI) per 1-SD increase in TSAT were -0.07 (-0.09; -0.05, p<0.0001) for GLP-1, -0.06 (-0.08; -0.04, p<0.0001) for insulin, -0.07(-0.09; -0.05, p<0.0001) for HOMA-IR, -0.01 (-0.01; -0.00, p<0.0001) for FPG and -0.01 (-0.01; -0.00, p=0.0004) for HbA1c. CONCLUSIONS Markers of insulin resistance are strongly related with markers of iron metabolism in healthy subjects. These relationships were inconsistent and weaker for short-term and long-term glucose levels. These results may provide insights in the relationships between iron metabolism and diabetes occurrence.
Collapse
Affiliation(s)
- Philipp Krisai
- Department of Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland
| | - Stefanie Leib
- Department of Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland
| | | | - Thomas Kofler
- Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland
| | - Mustafa Assadian
- Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland
| | - Anna Maseli
- Department of Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland
| | - John Todd
- Singulex, Inc., 1701 Harbor Bay Parkway Suite 200, Alameda, CA 94502, USA
| | - Joel Estis
- Singulex, Inc., 1701 Harbor Bay Parkway Suite 200, Alameda, CA 94502, USA
| | - Martin Risch
- Labormedizinisches Zentrum Dr. Risch, Schaan, Liechtenstein; Division of Laboratory Medicine, Kantonsspital Graubünden, Chur, Switzerland
| | - Lorenz Risch
- Labormedizinisches Zentrum Dr. Risch, Schaan, Liechtenstein; Division of Clinical Biochemistry, Medical University Innsbruck, Austria; Private University, Triesen, Liechtenstein
| | - David Conen
- Department of Medicine, University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland; Cardiovascular Research Institute Basel, University Hospital Basel, Switzerland.
| |
Collapse
|
32
|
Abstract
Iron is an essential element for several metabolic pathways and physiological processes. The maintenance of iron homeostasis within the human body requires a dynamic and highly sophisticated interplay of several proteins, as states of iron deficiency or excess are both potentially deleterious to health. Among these is plasma transferrin, which is central to iron metabolism not only through iron transport between body tissues in a soluble nontoxic form but also through its protective scavenger role in sequestering free toxic iron. The transferrin saturation (TSAT), an index that takes into account both plasma iron and its main transport protein, is considered an important biochemical marker of body iron status. Its increasing use in many health systems is due to the increased availability of measurement methods, such as calorimetry, turbidimetry, nephelometry, and immunochemistry to estimate its value. However, despite its frequent use in clinical practice to detect states of iron deficiency or iron overload, careful attention should be paid to the inherent limitations of the test especially in certain settings such as inflammation in order to avoid misinterpretation and erroneous conclusions. Beyond its usual clinical use, an emerging body of evidence has linked TSAT levels to major clinical outcomes such as cardiovascular mortality. This has the potential to extend the utility of TSAT index to risk stratification and prognostication. However, most of the current evidence is mainly driven by observational studies where the risk of residual confounding cannot be fully eliminated. Indeed, future efforts are required to fully explore this capability in well-designed clinical trials or prospective large-scale cohorts.
Collapse
Affiliation(s)
- M E Elsayed
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland; University Hospital Limerick, Limerick, Ireland
| | - M U Sharif
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland; University Hospital Limerick, Limerick, Ireland
| | - A G Stack
- Graduate Entry Medical School, University of Limerick, Limerick, Ireland; University Hospital Limerick, Limerick, Ireland; Health Research Institute, University of Limerick, Limerick, Ireland.
| |
Collapse
|
33
|
Backe MB, Moen IW, Ellervik C, Hansen JB, Mandrup-Poulsen T. Iron Regulation of Pancreatic Beta-Cell Functions and Oxidative Stress. Annu Rev Nutr 2016; 36:241-73. [PMID: 27146016 DOI: 10.1146/annurev-nutr-071715-050939] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dietary advice is the cornerstone in first-line treatment of metabolic diseases. Nutritional interventions directed at these clinical conditions mainly aim to (a) improve insulin resistance by reducing energy-dense macronutrient intake to obtain weight loss and (b) reduce fluctuations in insulin secretion through avoidance of rapidly absorbable carbohydrates. However, even in the majority of motivated patients selected for clinical trials, massive efforts using this approach have failed to achieve lasting efficacy. Less attention has been given to the role of micronutrients in metabolic diseases. Here, we review the evidence that highlights (a) the importance of iron in pancreatic beta-cell function and dysfunction in diabetes and (b) the integrative pathophysiological effects of tissue iron levels in the interactions among the beta cell, gut microbiome, hypothalamus, innate and adaptive immune systems, and insulin-sensitive tissues. We propose that clinical trials are warranted to clarify the impact of dietary or pharmacological iron reduction on the development of metabolic disorders.
Collapse
Affiliation(s)
- Marie Balslev Backe
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark;
| | - Ingrid Wahl Moen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark;
| | - Christina Ellervik
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, Massachusetts 02115
| | - Jakob Bondo Hansen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark;
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark;
| |
Collapse
|
34
|
Podmore C, Meidtner K, Schulze MB, Scott RA, Ramond A, Butterworth AS, Di Angelantonio E, Danesh J, Arriola L, Barricarte A, Boeing H, Clavel-Chapelon F, Cross AJ, Dahm CC, Fagherazzi G, Franks PW, Gavrila D, Grioni S, Gunter MJ, Gusto G, Jakszyn P, Katzke V, Key TJ, Kühn T, Mattiello A, Nilsson PM, Olsen A, Overvad K, Palli D, Quirós JR, Rolandsson O, Sacerdote C, Sánchez-Cantalejo E, Slimani N, Sluijs I, Spijkerman AMW, Tjonneland A, Tumino R, van der A DL, van der Schouw YT, Feskens EJM, Forouhi NG, Sharp SJ, Riboli E, Langenberg C, Wareham NJ. Association of Multiple Biomarkers of Iron Metabolism and Type 2 Diabetes: The EPIC-InterAct Study. Diabetes Care 2016; 39:572-81. [PMID: 26861925 PMCID: PMC5058436 DOI: 10.2337/dc15-0257] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 01/10/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Observational studies show an association between ferritin and type 2 diabetes (T2D), suggesting a role of high iron stores in T2D development. However, ferritin is influenced by factors other than iron stores, which is less the case for other biomarkers of iron metabolism. We investigated associations of ferritin, transferrin saturation (TSAT), serum iron, and transferrin with T2D incidence to clarify the role of iron in the pathogenesis of T2D. RESEARCH DESIGN AND METHODS The European Prospective Investigation into Cancer and Nutrition-InterAct study includes 12,403 incident T2D cases and a representative subcohort of 16,154 individuals from a European cohort with 3.99 million person-years of follow-up. We studied the prospective association of ferritin, TSAT, serum iron, and transferrin with incident T2D in 11,052 cases and a random subcohort of 15,182 individuals and assessed whether these associations differed by subgroups of the population. RESULTS Higher levels of ferritin and transferrin were associated with a higher risk of T2D (hazard ratio [HR] [95% CI] in men and women, respectively: 1.07 [1.01-1.12] and 1.12 [1.05-1.19] per 100 μg/L higher ferritin level; 1.11 [1.00-1.24] and 1.22 [1.12-1.33] per 0.5 g/L higher transferrin level) after adjustment for age, center, BMI, physical activity, smoking status, education, hs-CRP, alanine aminotransferase, and γ-glutamyl transferase. Elevated TSAT (≥45% vs. <45%) was associated with a lower risk of T2D in women (0.68 [0.54-0.86]) but was not statistically significantly associated in men (0.90 [0.75-1.08]). Serum iron was not associated with T2D. The association of ferritin with T2D was stronger among leaner individuals (Pinteraction < 0.01). CONCLUSIONS The pattern of association of TSAT and transferrin with T2D suggests that the underlying relationship between iron stores and T2D is more complex than the simple link suggested by the association of ferritin with T2D.
Collapse
Affiliation(s)
- Clara Podmore
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, U.K.
| | - Karina Meidtner
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Matthias B Schulze
- Department of Molecular Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Anna Ramond
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Adam S Butterworth
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | | | - John Danesh
- Department of Public Health and Primary Care, University of Cambridge, Cambridge, U.K
| | - Larraitz Arriola
- Public Health Division of Gipuzkoa, Basque Government, San Sebastian, Spain Instituto BIO-Donostia, Basque Government, San Sebastian, Spain Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública), Madrid, Spain
| | - Aurelio Barricarte
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública), Madrid, Spain Navarre Public Health Institute, Pamplona, Navarra, Spain
| | - Heiner Boeing
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbruecke, Nuthetal, Germany
| | - Françoise Clavel-Chapelon
- INSERM, CESP Centre for Research in Epidemiology and Population Health, Villejuif, France University Paris-Sud, Villejuif, France
| | - Amanda J Cross
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, U.K
| | - Christina C Dahm
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark
| | - Guy Fagherazzi
- INSERM, CESP Centre for Research in Epidemiology and Population Health, Villejuif, France University Paris-Sud, Villejuif, France
| | - Paul W Franks
- Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital, Lund University, Malmö, Sweden Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Diana Gavrila
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública), Madrid, Spain Department of Epidemiology, Murcia Regional Health Council, Murcia, Spain
| | - Sara Grioni
- Fondazione IRCCS Istituto Nazionale dei Tumori Milan, Milan, Italy
| | - Marc J Gunter
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, U.K
| | - Gaelle Gusto
- INSERM, CESP Centre for Research in Epidemiology and Population Health, Villejuif, France University Paris-Sud, Villejuif, France
| | - Paula Jakszyn
- Nutrition, Environment and Cancer Unit, Department of Epidemiology, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Verena Katzke
- Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Timothy J Key
- Cancer Epidemiology Unit, Nuffield Department of Population Health, University of Oxford, Oxford, U.K
| | - Tilman Kühn
- Division of Cancer Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - Amalia Mattiello
- Dipartimento di Medicina Clinica e Chirurgia, Federico II University, Naples, Italy
| | - Peter M Nilsson
- Department of Clinical Sciences, Clinical Research Center, Skåne University Hospital, Lund University, Malmö, Sweden
| | - Anja Olsen
- Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Kim Overvad
- Department of Public Health, Section for Epidemiology, Aarhus University, Aarhus, Denmark Department of Cardiology, Aalborg University Hospital, Aalborg, Denmark
| | - Domenico Palli
- Molecular and Nutritional Epidemiology Unit, Cancer Research and Prevention Institute (ISPO), Florence, Italy
| | - J Ramón Quirós
- Consejería de Sanidad, Public Health Directorate, Oviedo-Asturias, Spain
| | - Olov Rolandsson
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Carlotta Sacerdote
- Unit of Cancer Epidemiology, AO Citta' della Salute e della Scienza Hospital-University of Turin and Center for Cancer Prevention (CPO), Turin, Italy Human Genetics Foundation (HuGeF), Turin, Italy
| | - Emilio Sánchez-Cantalejo
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBER Epidemiología y Salud Pública), Madrid, Spain Escuela Andaluza de Salud Pública, Instituto de Investigación Biosanitaria ibs.Granada, Hospitales Universitarios de Granada/Universidad de Granada, Granada, Spain
| | - Nadia Slimani
- International Agency for Research on Cancer, Dietary Exposure Assessment Group (DEX), Lyon, France
| | - Ivonne Sluijs
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | | | - Rosario Tumino
- Cancer Registry and Histopathology Unit, "Civile - M.P. Arezzo" Hospital, Ragusa, Italy Associazone Iblea per la Ricerca Epidemiologica - Onlus, Ragusa, Italy
| | - Daphne L van der A
- National Institute for Public Health and the Environment (RIVM), Bilthoven, the Netherlands
| | - Yvonne T van der Schouw
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition, Section of Nutrition and Epidemiology, Wageningen University, Wageningen, the Netherlands
| | - Nita G Forouhi
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Stephen J Sharp
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Elio Riboli
- Department of Epidemiology & Biostatistics, School of Public Health, Imperial College London, London, U.K
| | - Claudia Langenberg
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| | - Nicholas J Wareham
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, U.K
| |
Collapse
|
35
|
Pechlaner R, Weiss G, Bansal S, Mayr M, Santer P, Pallhuber B, Notdurfter M, Bonora E, Willeit J, Kiechl S. Inadequate hepcidin serum concentrations predict incident type 2 diabetes mellitus. Diabetes Metab Res Rev 2016; 32:187-92. [PMID: 26378394 DOI: 10.1002/dmrr.2711] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Revised: 06/15/2015] [Accepted: 08/17/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) is closely associated with elevated body iron stores. The hormone hepcidin is the key regulator of iron homeostasis. Inadequately low hepcidin levels were recently reported in subjects with manifest T2DM. We investigated whether alterations of hepcidin levels precede the manifestation of T2DM and predict T2DM development independently of established risk conditions. METHODS This prospective population-based study included 675 subjects aged 50-89 years, 51.9% of whom were female. Hepcidin levels were measured by gold standard tandem mass spectrometry. Diabetes was diagnosed according to American Diabetes Association criteria, and incident diabetes was recorded between baseline in 2000 and 2010. RESULTS The baseline hepcidin-to-ferritin ratio in subjects that subsequently developed diabetes during follow-up was reduced on average by 29.8% as compared with subjects with normal glucose tolerance (95% confidence interval, -50.7% to -0.2%; p = 0.049). After adjustment for age, sex, and serum ferritin, higher hepcidin levels were associated with reduced risk of incident diabetes (hazard ratio per 1-unit higher log2 hepcidin, 0.80; 95% confidence interval, 0.64-0.98; p = 0.035; 33 events). Additional adjustment for established diabetes risk factors and determinants of hepcidin concentration did not appreciably change these results (HR, 0.81; 95% CI, 0.66-0.99). Likewise, inadequately low hepcidin levels were also detected in subjects with prevalent T2DM (n = 76). CONCLUSIONS Hepcidin levels that are inadequately low in relation to body iron stores are an independent predictor for incident T2DM and may contribute to diabetes-related tissue iron overload. Copyright © 2015 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Raimund Pechlaner
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Günter Weiss
- Department of Internal Medicine VI, Medical University of Innsbruck, Innsbruck, Austria
| | - Sukhvinder Bansal
- Institute of Pharmaceutical Science, King's College London, London, UK
| | - Manuel Mayr
- King's British Heart Foundation Centre, King's College London, London, UK
| | - Peter Santer
- Department of Laboratory Medicine, Hospital of Bruneck, Bruneck, Italy
| | - Barbara Pallhuber
- Department of Internal Medicine, Hospital of Bruneck, Bruneck, Italy
| | | | - Enzo Bonora
- Division of Endocrinology, Diabetes and Metabolic Diseases, University and Hospital Trust of Verona, Verona, Italy
| | - Johann Willeit
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Stefan Kiechl
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
36
|
Abstract
Humans have evolved to retain iron in the body and are exposed to a high risk of iron overload and iron-related toxicity. Excess iron in the blood, in the absence of increased erythropoietic needs, can saturate the buffering capacity of serum transferrin and result in non-transferrin-bound highly reactive forms of iron that can cause damage, as well as promote fibrogenesis and carcinogenesis in the parenchymatous organs. A number of hereditary or acquired diseases are associated with systemic or local iron deposition or iron misdistribution in organs or cells. Two of these, the HFE- and non-HFE hemochromatosis syndromes represent the paradigms of genetic iron overload. They share common clinical features and the same pathogenic basis, in particular, a lack of synthesis or activity of hepcidin, the iron hormone. Before hepcidin was discovered, the liver was simply regarded as the main site of iron storage and, as such, the main target of iron toxicity. Now, as the main source of hepcidin, it appears that the loss of the hepcidin-producing liver mass or genetic and acquired factors that repress hepcidin synthesis in the liver may also lead to iron overload. Usually, there is low-grade excess iron which, through oxidative stress, is sufficient to worsen the course of the underlying liver disease or other chronic diseases that are apparently unrelated to iron, such as chronic metabolic and cardiovascular diseases. In the future, modulation of hepcidin synthesis and activity or hepcidin hormone-replacing strategies may become therapeutic options to cure iron-related disorders.
Collapse
Affiliation(s)
- Antonello Pietrangelo
- Division of Internal Medicine 2 and Center for Hemochromatosis, University Hospital of Modena, Modena, Italy
| |
Collapse
|
37
|
Huth C, Beuerle S, Zierer A, Heier M, Herder C, Kaiser T, Koenig W, Kronenberg F, Oexle K, Rathmann W, Roden M, Schwab S, Seissler J, Stöckl D, Meisinger C, Peters A, Thorand B. Biomarkers of iron metabolism are independently associated with impaired glucose metabolism and type 2 diabetes: the KORA F4 study. Eur J Endocrinol 2015; 173:643-53. [PMID: 26294793 DOI: 10.1530/eje-15-0631] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Accepted: 08/20/2015] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Iron has been suggested to play a role in the etiology of type 2 diabetes mellitus (T2DM). Except for ferritin, evidence is sparse for other markers of iron metabolism that are regulated differently and might act through independent pathways. We therefore investigated the associations of serum ferritin, transferrin, soluble transferrin receptor (sTfR), transferrin saturation (TSAT), sTfR-to-log10ferritin (sTfR-F) index, and iron with impaired glucose metabolism (IGM/'prediabetes'), T2DM, and four continuous glucose and insulin traits. DESIGN AND METHODS Data from 2893 participants of the population-based Cooperative Health Research in the Region of Augsburg (KORA) F4 study (Germany) was investigated through regression analysis. The results were adjusted for socio-demographic, life-style, and obesity measures as well as metabolic, inflammatory, and other iron biomarkers following a step-wise approach. Non-linearity was tested by adding a non-linear spline component to the model. RESULTS Ferritin and transferrin were positively associated with IGM (fourth vs first sex-specific quartile: ferritin odds ratio (OR)=2.08 (95% CI 1.43-3.04) and transferrin OR=1.89 (95% CI 1.32-2.70)), T2DM (ferritin OR=1.98 (95% CI 1.22-3.22) and transferrin OR=2.42 (95% CI 1.54-3.81)), and fasting as well as 2-h glucose. TSAT (OR=0.55 (95% CI 0.34-0.88)) and iron (OR=0.61 (95% CI 0.38-0.97)) were inversely associated with T2DM, sTfR-F-index was inversely associated with IGM (OR=0.67 (95% CI 0.48-0.95)). There was no strong evidence for non-linear relationships. CONCLUSIONS The observed associations of several markers of iron metabolism with hyperglycemia and insulin resistance suggest that iron stores as well as iron-related metabolic pathways contribute to the pathogenesis of IGM and T2DM. Moreover, TSAT levels are decreased in T2DM patients.
Collapse
Affiliation(s)
- Cornelia Huth
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Simon Beuerle
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Astrid Zierer
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Margit Heier
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Christian Herder
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Thorsten Kaiser
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Wolfgang Koenig
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Florian Kronenberg
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Konrad Oexle
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Wolfgang Rathmann
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Michael Roden
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Sigrid Schwab
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany
| | - Jochen Seissler
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Doris Stöckl
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Christa Meisinger
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Annette Peters
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| | - Barbara Thorand
- Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyGerman Center for Diabetes Research (DZD)Partner Düsseldorf, GermanyInstitute of Laboratory MedicineClinical Chemistry and Molecular Diagnostics, University Leipzig, Leipzig, GermanyDepartment of Internal Medicine II - CardiologyUniversity of Ulm Medical Center, Ulm, GermanyDivision of Genetic EpidemiologyDepartment of Medical Genetics, Molecular and Clinical Pharmacology, Medical University of Innsbruck, Innsbruck, AustriaInstitute of Human GeneticsKlinikum Rechts der Isar, Technische Universität München, Munich, GermanyInstitute of Biometrics and EpidemiologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyDepartment of Endocrinology and DiabetologyMedical Faculty, Heinrich Heine University Düsseldorf, Düsseldorf, GermanyMedizinische Klinik und Poliklinik IVDiabetes Zentrum - Campus Innenstadt, Klinikum der Ludwig-Maximilians-Universität, München, GermanyClinical Cooperation Group DiabetesLudwig-Maximilians-Universität München and Helmholtz Zentrum München, München, Germany Institute of Epidemiology IIHelmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstraße 1, D-85764 Neuherberg, GermanyGerman Center for Diabetes Research (DZD)Partner Neuherberg, GermanyMONICA/KORA Myocardial Infarction RegistryCentral Hospital of Augsburg, Augsburg, GermanyInstitute for Clinical DiabetologyGerman Diabetes Center, Leibniz Center for Diabetes Research, Heinrich Heine University
| |
Collapse
|
38
|
Pietrangelo A. Pathogens, Metabolic Adaptation, and Human Diseases--An Iron-Thrifty Genetic Model. Gastroenterology 2015; 149:834-8. [PMID: 26291901 DOI: 10.1053/j.gastro.2015.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
39
|
Park RJ, Moon JD. Low transferrin saturation is associated with impaired fasting glucose and insulin resistance in the South Korean adults: the 2010 Korean National Health and Nutrition Examination Survey. Diabet Med 2015; 32:673-8. [PMID: 25444086 DOI: 10.1111/dme.12643] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/25/2014] [Indexed: 12/12/2022]
Abstract
AIMS The associations of transferrin saturation with diabetes have not been well evaluated and conflicting results have been reported. The purpose of this study is to examine the association of iron indices (serum ferritin and transferrin saturation) with risk of impaired fasting glucose and insulin resistance. METHODS We conducted a cross-sectional study in 2413 individuals (1150 men and 1263 women) aged 20-50 years who participated in the 2010 Korean National Health and Nutrition Examination Survey. Participants were free of diabetes, malignancy, liver cirrhosis, chronic renal failure, anaemia, pregnancy and menopause. Fasting plasma glucose, insulin and the homeostasis model assessment of insulin resistance (HOMA-IR) were measured as the outcomes. RESULTS Impaired fasting glucose was more prevalent in the highest compared with the lowest serum ferritin quartile among men (odds ratio [OR], 1.97; 95% confidence interval [CI], 1.20-3.24) after adjustment for multiple covariates. Following the same adjustment, impaired fasting glucose was less prevalent in the highest compared with the lowest transferrin saturation quartile among men (OR, 0.45; 95% CI, 0.25-0.80) and women (OR, 0.33; 95% CI, 0.14-0.77). Moreover, a higher ferritin level was significantly associated with higher HOMA-IR after adjusting for confounders in men. Lower transferrin saturation was also significantly associated with higher insulin levels and HOMA-IR in both sexes. CONCLUSIONS Lower transferrin saturations were associated with an increased risk of impaired fasting glucose and insulin resistance among general South Korean population.
Collapse
Affiliation(s)
- R J Park
- Department of Occupational and Environmental Medicine, Gwangyang Sarang General Hospital, Gwangyang-si, Jeollanam-do, Republic of Korea; Department of Medicine, Graduate School of Chonnam National University, Gwangju, Republic of Korea
| | | |
Collapse
|
40
|
Yeap BB, Divitini ML, Gunton JE, Olynyk JK, Beilby JP, McQuillan B, Hung J, Knuiman MW. Higher ferritin levels, but not serum iron or transferrin saturation, are associated with Type 2 diabetes mellitus in adult men and women free of genetic haemochromatosis. Clin Endocrinol (Oxf) 2015; 82:525-32. [PMID: 24953981 DOI: 10.1111/cen.12529] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Revised: 04/14/2014] [Accepted: 06/06/2014] [Indexed: 12/28/2022]
Abstract
CONTEXT Iron overload predisposes to diabetes and higher ferritin levels have been associated with diabetes. However, it is unclear whether ferritin reflects differences in iron-related parameters between diabetic and nondiabetic persons. We examined associations of serum ferritin, iron and transferrin saturation with Type 2 diabetes in adults without genetic predisposition to iron overload. DESIGN, PARTICIPANTS AND MEASUREMENTS Cross-sectional analysis of community-dwelling men and women aged 17-97 years from the Busselton Health Survey, Western Australia. Men and women carrying genotypes associated with haemochromatosis (C282Y/C282Y or C282Y/H63D) were excluded. Serum ferritin, iron and transferrin saturation were assayed. RESULTS There were 1834 men (122 with diabetes, 6·6%) and 2351 women (141 with diabetes, 6%). In men, higher serum ferritin was associated with diabetes after adjusting for age, smoking, alcohol, cardiovascular history, body mass index (BMI), waist, blood pressure, lipids, C-reactive protein (CRP), adiponectin, alanine transaminase (ALT) and gamma-glutamyl transpeptidase (GGT) [odds ratio (OR): 1·29 per 1 unit increase log ferritin, 95% confidence interval (CI) = 1·01-1·65, P = 0·043]. In women, higher serum ferritin was associated with diabetes [fully adjusted OR: 1·31 per 1 unit increase log ferritin, 95% CI = 1·04-1·63, P = 0·020; 1·84 for tertile (T) 3 vs T1, 95% CI = 1·09-3·11]. Neither iron levels nor transferrin saturation were associated with diabetes risk in men or women. Higher ferritin was not associated with insulin resistance in nondiabetic adults. CONCLUSIONS In adults, higher ferritin levels are independently associated with prevalent diabetes while iron and transferrin saturation are not. Ferritin is a robust biomarker for diabetes risk, but further investigation is needed to clarify whether this relationship is mediated via iron metabolism.
Collapse
Affiliation(s)
- Bu B Yeap
- School of Medicine and Pharmacology, University of Western Australia, Perth, WA, Australia; Department of Endocrinology and Diabetes, Fremantle Hospital, Fremantle, WA, Australia
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Helicobacter pylori seropositivity's association with markers of iron, 1-carbon metabolism, and antioxidant status among US adults: a structural equations modeling approach. PLoS One 2015; 10:e0121390. [PMID: 25815731 PMCID: PMC4376857 DOI: 10.1371/journal.pone.0121390] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 01/30/2015] [Indexed: 12/20/2022] Open
Abstract
Objectives We tested a model in which Helicobacter pylori seropositivity (Hps) predicted iron status, which in turn acted as a predictor for markers of 1-C metabolism that were then allowed to predict antioxidant status. Methods National Health and Nutrition Examination Surveys (NHANES 1999–2000) cross-sectional data among adults aged 20–85 y were analyzed (n = 3,055). Markers of Hps, iron status (serum ferritin and transferrin saturation (TS)); 1-C metabolism (serum folate (FOLserum), B-12, total homocysteine (tHcy), methylmalonic acid (MMA)) and antioxidant status (vitamins A and E) were entered into a structural equations model (SEM). Results Predictors of Hps included older age, lower education and income, racial/ethnic groups (lowest among Non-Hispanic Whites), and lifetime cigarette smoking. SEM modeling indicated that Hps had a direct inverse relationship with iron status (combining serum ferritin and TS) which in turn was positively related to 1-C metabolites (higher serum folate, B-12 or lower tHcy/MMA) that were positively associated with antioxidant status (combining serum vitamins A and E). Another pathway that was found bypassed 1-C metabolites (Hps → Iron_st → Antiox). The sum of all indirect effects from Hps combining both pathways and the other indirect pathways in the model (Hps → Iron_st → OneCarbon; Hps →OneCarbon →Antiox) was estimated at β = -0.006±0.003, p<0.05. Conclusions In sum, of the total effect of H. pylori seropositivity on antioxidant status, two significant indirect pathways through Iron status and 1-Carbon metabolites were found. Randomized controlled trials should be conducted to uncover the concomitant causal effect of H. pylori eradication on improving iron status, folate, B-12 and antioxidant status among H. pylori seropositive individuals.
Collapse
|
42
|
Puliyel M, Mainous AG, Berdoukas V, Coates TD. Iron toxicity and its possible association with treatment of Cancer: lessons from hemoglobinopathies and rare, transfusion-dependent anemias. Free Radic Biol Med 2015; 79:343-51. [PMID: 25463277 DOI: 10.1016/j.freeradbiomed.2014.10.861] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 10/22/2014] [Accepted: 10/30/2014] [Indexed: 01/19/2023]
Abstract
Exposure to elevated levels of iron causes tissue damage and organ failure, and increases the risk of cancer. The toxicity of iron is mediated through generation of oxidants. There is also solid evidence indicating that oxidant stress plays a significant role in a variety of human disease states, including malignant transformation. Iron toxicity is the main focus when managing thalassemia. However, the short- and long-term toxicities of iron have not been extensively considered in children and adults treated for malignancy, and only recently have begun to draw oncologists' attention. The treatment of malignancy can markedly increase exposure of patients to elevated toxic iron species without the need for excess iron input from transfusion. This under-recognized exposure likely enhances organ toxicity and may contribute to long-term development of secondary malignancy and organ failure. This review discusses the current understanding of iron metabolism, the mechanisms of production of toxic free iron species in humans, and the relation of the clinical marker, transferrin saturation (TS), to the presence of toxic free iron. We will present epidemiological data showing that high TS is associated with poor outcomes and development of cancer, and that lowering free iron may improve outcomes. Finally, we will discuss the possible relation between some late complications seen in survivors of cancer and those due to iron toxicity.
Collapse
Affiliation(s)
- Mammen Puliyel
- Section of Hematology, Childrens Center for Cancer, Blood Disease and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles California, USA
| | - Arch G Mainous
- Department of Health Services Research, Management and Policy, University of Florida, Gainesville, Fla. USA
| | - Vasilios Berdoukas
- Section of Hematology, Childrens Center for Cancer, Blood Disease and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles California, USA
| | - Thomas D Coates
- Section of Hematology, Childrens Center for Cancer, Blood Disease and Bone Marrow Transplantation, Children's Hospital Los Angeles, University of Southern California Keck School of Medicine, Los Angeles California, USA.
| |
Collapse
|
43
|
Ellervik C, Marott JL, Tybjærg-Hansen A, Schnohr P, Nordestgaard BG. Total and cause-specific mortality by moderately and markedly increased ferritin concentrations: general population study and metaanalysis. Clin Chem 2014; 60:1419-28. [PMID: 25156997 DOI: 10.1373/clinchem.2014.229013] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Previous population-based studies of plasma ferritin concentration have not revealed a relationship with total mortality. We tested the possible association of increased ferritin concentrations with increased risk of total and cause-specific mortality in the general population. METHODS We examined total and cause-specific mortality according to baseline plasma ferritin concentrations in a Danish population-based study (the Copenhagen City Heart Study) of 8988 individuals, 6364 of whom died (median follow-up 23 years). We also included a metaanalysis of total mortality comprising population-based studies according to ferritin quartiles or tertiles. RESULTS Multifactorially adjusted hazard ratios (HRs) for total mortality for individuals with ferritin ≥200 vs <200 μg/L were 1.1 (95% CI 1.1-1.2; P = 0.0008) overall, 1.1 (1.0-1.2; P = 0.02) in men, and 1.2 (1.0-1.3; P = 0.03) in women. Stepwise increasing concentrations of ferritin were associated with a stepwise increased risk of premature death overall (log rank, P = 2 × 10(-22)), with median survival of 55 years at ferritin concentrations ≥600 μg/L, 72 years at 400-599 μg/L, 76 years at 200-399 μg/L, and 79 years at ferritin <200 μg/L. The corresponding HR for total overall mortality for ferritin ≥600 vs <200 μg/L was 1.5 (1.2-1.8; P = 0.00008). Corresponding adjusted HRs for ferritin ≥600 vs <200 μg/L were 1.6 (1.1-2.3; P = 0.01) for cancer mortality, 2.9 (1.7-5.0; P = 0.0001) for endocrinological mortality, and 1.5 (1.1-2.0; P = 0.01) for cardiovascular mortality. The metaanalysis random effects odds ratio for total mortality for ferritin upper vs reference quartile or tertile was 1.0 (0.9-1.1; P = 0.3) (P heterogeneity = 0.5). CONCLUSIONS Moderately to markedly increased ferritin concentrations represent a biological biomarker predictive of early death in a dose-dependent linear manner in the general population.
Collapse
Affiliation(s)
- Christina Ellervik
- Department of Research, Nykøbing Falster Hospital, Nykøbing Falster, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Jacob Louis Marott
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Anne Tybjærg-Hansen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Peter Schnohr
- The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Copenhagen City Heart Study, Frederiksberg Hospital, Copenhagen University Hospital, Copenhagen, Denmark; Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark
| |
Collapse
|
44
|
Orban E, Schwab S, Thorand B, Huth C. Association of iron indices and type 2 diabetes: a meta-analysis of observational studies. Diabetes Metab Res Rev 2014; 30:372-94. [PMID: 24327370 DOI: 10.1002/dmrr.2506] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Revised: 10/29/2013] [Accepted: 10/31/2013] [Indexed: 02/06/2023]
Abstract
The literature on the role of body iron status in the development of type 2 diabetes (T2D) in humans is inconsistent. We aimed to assess the association between iron indices and T2D by a meta-analysis of previously published studies. A systematic literature search was conducted in PubMed and EMBASE. Observational studies on the association of ferritin (when controlled for age and sex), transferrin saturation, soluble transferrin receptor and transferrin with T2D were included. Pooled association estimates were calculated using a random effects model. Forty-six eligible studies were identified. The pooled multivariable adjusted relative risks of T2D in the highest versus lowest quartile of ferritin levels were significantly elevated in both cross-sectional as well as prospective studies and after restriction to inflammation-adjusted studies [overall: 1.67 (95% CI 1.41-1.99)]. The mean difference indicated 43.54 ng/mL (95% CI 28.14-58.94) higher ferritin levels in type 2 diabetic individuals. The relative risk for a transferrin saturation ≥ 50% was 1.59 (95% CI 1.28-1.97), the mean difference was -1.92% [95% CI -2.99-(-0.85)]. Study-specific results of soluble transferrin receptor and transferrin levels were extremely heterogeneous. Ferritin and clinically elevated transferrin saturation were strongly associated with an increased risk of T2D, overall and in prospective studies. Ferritin was also significantly associated after multivariable adjustment including inflammation. Thus, the current evidence hints at a causal effect; however, publication bias and unmeasured confounding cannot be excluded.
Collapse
|
45
|
Hansen JB, Moen IW, Mandrup-Poulsen T. Iron: the hard player in diabetes pathophysiology. Acta Physiol (Oxf) 2014; 210:717-32. [PMID: 24521359 DOI: 10.1111/apha.12256] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/09/2014] [Accepted: 02/03/2014] [Indexed: 12/14/2022]
Abstract
The interest in the role of ferrous iron in diabetes pathophysiology has been revived by recent evidence of iron as an important determinant of pancreatic islet inflammation and as a biomarker of diabetes risk and mortality. The iron metabolism in the β-cell is complex. Excess free iron is toxic, but at the same time, iron is required for normal β-cell function and thereby glucose homeostasis. In the pathogenesis of diabetes, iron generates reactive oxygen species (ROS) by participating in the Fenton chemistry, which can induce oxidative damage and apoptosis. The aim of this review is to present and discuss recent evidence, suggesting that iron is a key pathogenic factor in both type 1 and type 2 diabetes with a focus on inflammatory pathways. Pro-inflammatory cytokine-induced β-cell death is not fully understood, but may include iron-induced ROS formation resulting in dedifferentiation by activation of transcription factors, activation of the mitochondrial apoptotic machinery or of other cell death mechanisms. The pro-inflammatory cytokine IL-1β facilitates divalent metal transporter 1 (DMT1)-induced β-cell iron uptake and consequently ROS formation and apoptosis, and we propose that this mechanism provides the relay between inflammation and oxidative β-cell damage. Iron chelation may be a potential therapeutic approach to reduce disease severity and mortality among diabetes patients. However, the therapeutic effect and safety of iron reduction need to be tested in clinical trials before dietary interventions or the use of iron chelation therapy titrated to avoid anaemia.
Collapse
Affiliation(s)
- J. B. Hansen
- Section for Endocrinological Research; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Physiology; University of Toronto; Toronto ON Canada
| | - I. W. Moen
- Section for Endocrinological Research; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
| | - T. Mandrup-Poulsen
- Section for Endocrinological Research; Department of Biomedical Sciences; University of Copenhagen; Copenhagen Denmark
- Department of Molecular Medicine and Surgery; Karolinska Institutet; Stockholm Sweden
| |
Collapse
|
46
|
Ellervik C, Mandrup-Poulsen T, Tybjærg-Hansen A, Nordestgaard BG. Total and cause-specific mortality by elevated transferrin saturation and hemochromatosis genotype in individuals with diabetes: two general population studies. Diabetes Care 2014; 37:444-52. [PMID: 24130348 DOI: 10.2337/dc13-1198] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Mortality is increased in patients with hereditary hemochromatosis, in individuals from the general population with increased transferrin saturation (TS), and also in patients with type 1 diabetes and increased TS from a highly specialized diabetes clinic. Thus, we have recommended targeted screening for TS in specialized diabetes clinics. Whether mortality is also increased in individuals from the general population with diabetes and increased TS is unknown. RESEARCH DESIGN AND METHODS In two Danish population studies (N = 84,865), we examined mortality according to baseline levels of TS and hemochromatosis genotype (HFE) G → A substitution at nucleotide 845 in codon 282 (C282Y/C282Y) in individuals with diabetes (type 1, N = 118; type 2, N = 3,228; total, N = 3,346). RESULTS The cumulative survival rate was reduced in individuals with diabetes with TS ≥50% vs. <50% (log-rank; P < 0.0001), with median survival ages of 66 and 79 years, respectively. The hazard ratio (HR) for TS ≥50% vs. <50% was 2.0 (95% CI 1.3-2.8; P = 0.0004) for total mortality overall (and similar for men and women separately); 2.6 (1.3-5.4; P = 0.008) for neoplasms; and 3.4 (2.0-6.0; P = 0.00002) for endocrinological causes. A stepwise increased risk of total mortality was observed for stepwise increasing TS (log-rank test, P = 0.0001), with an HR for TS ≥70% vs. TS <20% of 4.8 (2.0-12; P = 0.0006). The HR for total mortality in individuals with diabetes for C282Y/C282Y versus wild type/wild type was 3.3 (1.04-10; P = 0.04), and for C282Y/C282Y and TS ≥50% versus wild type/wild type and TS <50% was 6.0 (1.5-24; P = 0.01). Six percent of these premature deaths can possibly be avoided by early screening for TS or HFE genotype. CONCLUSIONS Individuals with diabetes, ascertained in the general population, with increased TS or HFE genotype have a twofold to sixfold increased risk of premature death.
Collapse
|
47
|
Mainous AG, Wright RU, Hulihan MM, Twal WO, McLaren CE, Diaz VA, McLaren GD, Argraves WS, Grant AM. Elevated transferrin saturation, health-related quality of life and telomere length. Biometals 2013; 27:135-41. [PMID: 24337410 DOI: 10.1007/s10534-013-9693-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 12/07/2013] [Indexed: 12/26/2022]
Abstract
We sought to examine the relationship between elevated transferrin saturation (TS) and measures of health status (telomere length and patient-reported health-related quality of life) to assess whether elevated TS is associated with negative patient outcomes beyond increased risk for morbidity and mortality, using a cross-sectional analysis of the Hemochromatosis and Iron Overload Screening Study supplemented with assays for leukocyte telomere length in adults ≥25 years old (n = 669). Among individuals with elevated TS (≥45 % for women and ≥50 % for men), who also had a usual source of care, only 5.2 % reported ever being told by a doctor that they had an elevated iron condition. In a fully adjusted general linear regression model controlling for demographic characteristics as well as health conditions associated with iron overload, elevated TS versus non-elevated TS was associated with worse general health status (60.4 vs. 63.8, P < 0.05), mental health status (76.5 vs. 82.2, P < 0.0001) and shorter telomere length (241.4 vs. 261.3, P < 0.05). Increased surveillance of elevated TS may be in order as elevated TS is associated with decreased health status and very few patients with elevated TS are aware of their condition.
Collapse
Affiliation(s)
- Arch G Mainous
- Departments of Health Services Research, Management & Policy, and Community Health and Family Medicine, Health Science Center, University of Florida, PO Box 100195, Gainesville, FL, 32610, USA,
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Ellervik C, Andersen HU, Tybjærg-Hansen A, Frandsen M, Birgens H, Nordestgaard BG, Mandrup-Poulsen T. Total mortality by elevated transferrin saturation in patients with diabetes. Diabetes Care 2013; 36:2646-54. [PMID: 23801727 PMCID: PMC3747880 DOI: 10.2337/dc12-2032] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE It is not known to what extent iron overload predicts prognosis in patients with diabetes after diagnosis or whether iron overload is a risk factor independent of the HFE genotype. We investigated total and cause-specific mortality according to increased transferrin saturation (≥ 50 vs. <50%), whether mortality is driven by the HFE genotype, and whether early measurement of transferrin saturation helps to predict mortality outcome. RESEARCH DESIGN AND METHODS Cohort 1 included patients with late-onset type 1 diabetes (n = 716) with a cross-sectional measurement of transferrin saturation and HFE genotype. Cohort 2 included consecutively recruited patients with any diabetes (n = 6,120), transferrin saturation measurement at referral, and HFE genotype if transferrin saturation was above 50%. RESULTS In cohort 1, the hazard ratio for total mortality was 2.3 (95% CI 1.3-3.9; P = 0.002) and for cause-specific mortality by neoplasms was 5.8 (2.4-14; P = 0.00007) in patients with transferrin saturation ≥ 50 vs. <50%. Excluding genotypes C282Y/C282Y and C282Y/H63D gave similar results. The hazard ratio for total mortality was 4.0 (1.2-13; P = 0.01) and for cause-specific mortality by neoplasms was 13 (3.6-49; P = 0.0001) in patients with C282Y/C282Y versus wild type. In cohort 2, total mortality was not different in patients with transferrin saturation ≥ 50 vs. <50%. In patients with late-onset type 1 diabetes and transferrin saturation ≥ 50%, the hazard ratio for total mortality was 0.4 (0.2-0.9; P = 0.03) in cohort 2 versus cohort 1. CONCLUSIONS Increased transferrin saturation and HFE genotype C282Y/C282Y predict total mortality in patients with late-onset type 1 diabetes, and increased transferrin saturation after diagnosis is an independent risk factor. Early measurement of transferrin saturation in these patients leading to early intervention improves life expectancy.
Collapse
Affiliation(s)
- Christina Ellervik
- Department of Clinical Biochemistry, Herlev Hospital, Copenhagen University Hospital, Herlev, Denmark.
| | | | | | | | | | | | | |
Collapse
|
49
|
Lundh M, Scully SS, Mandrup-Poulsen T, Wagner BK. Small-molecule inhibition of inflammatory β-cell death. Diabetes Obes Metab 2013; 15 Suppl 3:176-84. [PMID: 24003935 PMCID: PMC3777666 DOI: 10.1111/dom.12158] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 04/15/2013] [Indexed: 01/09/2023]
Abstract
With the worldwide increase in diabetes prevalence there is a pressing unmet need for novel antidiabetic therapies. Insufficient insulin production due to impaired β-cell function and apoptotic reduction of β-cell mass is a common denominator in the pathogenesis of diabetes. Current treatments are directed at improving insulin sensitivity, and stimulating insulin secretion or replacing the hormone, but do not target progressive apoptotic β-cell loss. Here we review the current development of small-molecule inhibitors designed to rescue β-cells from apoptosis. Several distinct classes of small molecules have been identified that protect β-cells from inflammatory, oxidative and/or metabolically induced apoptosis. Although none of these have yet reached the clinic, β-cell protective small molecules alone or in combination with current therapies provide exciting opportunities for the development of novel treatments for diabetes.
Collapse
Affiliation(s)
- Morten Lundh
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Department of Biomedical Sciences, University of Copenhagen, Denmark
| | - Stephen S. Scully
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, University of Copenhagen, Denmark
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Bridget K. Wagner
- Chemical Biology Program, Broad Institute of Harvard and MIT, Cambridge, MA
- Corresponding author: ; Broad Institute, 7 Cambridge Center, Cambridge, MA 02142; Tel: (617) 714-7363, Fax (617) 714-8943
| |
Collapse
|
50
|
High ferritin and low transferrin saturation are associated with pre-diabetes among a national representative sample of U.S. adults. Clin Nutr 2012; 32:1055-60. [PMID: 23312547 DOI: 10.1016/j.clnu.2012.11.024] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 11/19/2012] [Accepted: 11/26/2012] [Indexed: 12/22/2022]
Abstract
BACKGROUND & AIMS Iron overload is known to cause diabetes. However, the underlying mechanism is poorly understood. We therefore studied the association of different markers of iron metabolism, namely ferritin, erythrocyte protoporphyrin and transferrin saturation (TSAT, as defined by a percentage of transferrin that is saturated with iron) with pre-diabetes (preDM) in US adults without chronic kidney disease, anemia, and iron deficiency. METHODS Data on 2575 participants of the National Health and Nutrition Examination Survey (NHANES) 1999-2002 who were free of diabetes, chronic kidney disease, iron deficiency, and anemia were analyzed. Data on 3876 participants of the NHANES III (1988-1994) were used as replication. Homeostasis model assessment of insulin resistance (HOMA-IR), blood glycosylated hemoglobin level (HbA1C), fasting glucose, insulin, and preDM (defined as a fasting plasma glucose 100-125 mg/dl or an HBA1C value 5.7-6.4%) were measured as the outcomes. RESULTS Logistic regression analyses indicated independent associations of high ferritin (Ptrend = 0.028) and low TSAT (P(trend) = 0.029) with preDM after adjusting for sociodemographics, physical activity (active/sedementary), metabolic and inflammatory markers (triglycerides, total cholesterol, HDL cholesterol, mean arterial pressure, CRP, white cell count, and albumin), and liver enzymes (GGT, Alk phos, AST, and ALT). The NHANES III data showed similar associations. Combining the results showed a more significant association for high ferritin (P(meta) = 0.016) and low TSAT (P(meta) = 0.002). Moreover, TSAT was associated with HbA1C, fasting glucose, insulin, and HOMA-IR (P(meta) ≤ 0.001). CONCLUSIONS Higher ferritin and lower TSAT are associated with higher risk of preDM in a general population without confounding diseases. Further research is needed to examine the underlying mechanism of these two indices, especially TSAT, in the pathophysiology of preDM.
Collapse
|