1
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Ferroptosis-A Shared Mechanism for Parkinson's Disease and Type 2 Diabetes. Int J Mol Sci 2024; 25:8838. [PMID: 39201524 PMCID: PMC11354749 DOI: 10.3390/ijms25168838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are the two most frequent age-related chronic diseases. There are many similarities between the two diseases: both are chronic diseases; both are the result of a decrease in a specific substance-insulin in T2D and dopamine in PD; and both are caused by the destruction of specific cells-beta pancreatic cells in T2D and dopaminergic neurons in PD. Recent epidemiological and experimental studies have found that there are common underlying mechanisms in the pathophysiology of T2D and PD: chronic inflammation, mitochondrial dysfunction, impaired protein handling and ferroptosis. Epidemiological research has indicated that there is a higher risk of PD in individuals with T2D. Moreover, clinical studies have observed that the symptoms of Parkinson's disease worsen significantly after the onset of T2D. This article provides an up-to-date review on the intricate interplay between oxidative stress, reactive oxygen species (ROS) and ferroptosis in PD and T2D. By understanding the shared molecular pathways and how they can be modulated, we can develop more effective therapies, or we can repurpose existing drugs to improve patient outcomes in both disorders.
Collapse
|
2
|
de Souza GO, Dos Santos WO, Donato J. Ironing out obesity. Trends Endocrinol Metab 2024; 35:456-458. [PMID: 38599900 DOI: 10.1016/j.tem.2024.04.001] [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: 03/21/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/12/2024]
Abstract
Obesity is associated with dysfunctions in hypothalamic neurons that regulate metabolism, including agouti-related protein (AgRP)-expressing neurons. In a recent article, Zhang et al. demonstrated that either diet- or genetically induced obesity promoted iron accumulation specifically in AgRP neurons. Preventing iron overload in AgRP neurons mitigated diet-induced obesity and related comorbidities in male mice.
Collapse
Affiliation(s)
- Gabriel O de Souza
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Willian O Dos Santos
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil.
| |
Collapse
|
3
|
Zhou D, Lu P, Mo X, Yang B, Chen T, Yao Y, Xiong T, Yue L, Yang X. Ferroptosis and metabolic syndrome and complications: association, mechanism, and translational applications. Front Endocrinol (Lausanne) 2024; 14:1248934. [PMID: 38260171 PMCID: PMC10800994 DOI: 10.3389/fendo.2023.1248934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/11/2023] [Indexed: 01/24/2024] Open
Abstract
Metabolic syndrome is a medical condition characterized by several metabolic disorders in the body. Long-term metabolic disorders raise the risk of cardiovascular disease (CVD) and type 2 diabetes mellitus (T2DM). Therefore, it is essential to actively explore the aetiology of metabolic syndrome (MetS) and its comorbidities to provide effective treatment options. Ferroptosis is a new form of cell death that is characterized by iron overload, lipid peroxide accumulation, and decreased glutathione peroxidase 4(GPX4) activity, and it involves the pathological processes of a variety of diseases. Lipid deposition caused by lipid diseases and iron overload is significant in metabolic syndrome, providing the theoretical conditions for developing ferroptosis. Recent studies have found that the major molecules of ferroptosis are linked to common metabolic syndrome consequences, such as T2DM and atherosclerosis. In this review, we first discussed the mechanics of ferroptosis, the regulatory function of inducers and inhibitors of ferroptosis, and the significance of iron loading in MetS. Next, we summarized the role of ferroptosis in the pathogenesis of MetS, such as obesity, type 2 diabetes, and atherosclerosis. Finally, we discussed relevant ferroptosis-targeted therapies and raised some crucial issues of concern to provide directions for future Mets-related treatments and research.
Collapse
Affiliation(s)
- Dongmei Zhou
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Peipei Lu
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Xianglai Mo
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Bing Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Ting Chen
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - You Yao
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Tian Xiong
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| | - Lin Yue
- School of Nursing, Hunan University of Medicine, Huaihua, China
| | - Xi Yang
- Department of Endocrinology, Geriatric Endocrinology and Metabolism, Guangxi Key Laboratory of Precision Medicine in Cardio-Cerebrovascular Diseases Control and Prevention, Guangxi Clinical Research Center for Cardio-Cerebrovascular Diseases, First Affiliated Hospital, Guangxi Medical University, Nanning, China
| |
Collapse
|
4
|
Sam RM, Shetty SS, Kumari N S, KP S, Bhandary P. Association between iron profile status and insulin resistance in patients with type 2 diabetes mellitus. J Diabetes Metab Disord 2023; 22:1453-1458. [PMID: 37975116 PMCID: PMC10638270 DOI: 10.1007/s40200-023-01268-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 07/19/2023] [Indexed: 11/19/2023]
Abstract
Introduction Individuals' burden of insulin resistance and metabolic syndrome, such as type 2 diabetes mellitus, is increasing. This indicates to intrigue into various facets of prevention, early screening, prognostication and feasible treatment alternatives in this arena. Aim This study targets to evaluate iron profile status among people diagnosed with type 2 diabetes mellitus and normoglycemic in order to deduce association between iron parameters and insulin resistance, if any exist. Methodology A case-control study of total 123 subjects, comprising males and females in the age group of 30 - 70 years were recruited for the study. Case group constituted 81 participants who were diagnosed with type 2 diabetes mellitus and control group constituted 42 healthy individuals who attended routine health check-ups in the hospital. Iron profile parameters including Serum Iron, Serum Ferritin, Total Iron binding Capacity and Glycemic profile parameter like fasting blood glucose, serum insulin were estimated. Transferrin saturation and HOMA-IR were calculated. Result Ferritin and Transferrin saturation was found to be higher in cases than in controls with significance of p = 0.003 and p = 0.021 respectively and TIBC (total iron-binding capacity) was lesser in cases with p = 0.031. Comparison of Serum Iron values did not yield a significant result. Correlation study between ferritin and insulin resistance parameters yielded a satisfactory result in the cases (p<0.05) and controls (p<0.01) separately. Conclusion This study implies that there is a clear link between iron profile status, notably ferritin, and the emergence of insulin resistance, and hence insulin production. This study supports the function of the micronutrient iron in the etiology of type 2 diabetes and its consequences.
Collapse
Affiliation(s)
- Reeba Maryam Sam
- KS Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, India
| | - Shilpa S Shetty
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, India
| | - Suchetha Kumari N
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, India
- Department of Biochemistry, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, India
| | - Sharmila KP
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, India
| | - Prajna Bhandary
- Central Research Laboratory, KS Hegde Medical Academy, Nitte (Deemed to be University), Mangalore, India
| |
Collapse
|
5
|
Xie D, Li K, Feng R, Xiao M, Sheng Z, Xie Y. Ferroptosis and Traditional Chinese Medicine for Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes 2023; 16:1915-1930. [PMID: 37398945 PMCID: PMC10312342 DOI: 10.2147/dmso.s412747] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/03/2023] [Indexed: 07/04/2023] Open
Abstract
Ferroptosis, an emerging form of regulated programmed cell death, has garnered significant attention in the past decade. It is characterized by the accumulation of lipid peroxides and subsequent damage to cellular membranes, which is dependent on iron. Ferroptosis has been implicated in the pathogenesis of various diseases, including tumors and diabetes mellitus. Traditional Chinese medicine (TCM) has unique advantages in preventing and treating type 2 diabetes mellitus (T2DM) due to its anti-inflammatory, antioxidant, immunomodulatory, and intestinal flora-regulating functions. Recent studies have determined that TCM may exert therapeutic effects on T2DM and its complications by modulating the ferroptosis-related pathways. Therefore, a comprehensive and systematic understanding of the role of ferroptosis in the pathogenesis and TCM treatment of T2DM is of great significance for developing therapeutic drugs for T2DM and enriching the spectrum of effective T2DM treatment with TCM. In this review, we review the concept, mechanism, and regulatory pathways of ferroptosis and the ferroptosis mechanism of action involved in the development of T2DM. Also, we develop a search strategy, establish strict inclusion and exclusion criteria, and summarize and analyze the application of the ferroptosis mechanism in TCM studies related to T2DM and its complications. Finally, we discuss the shortcomings of current studies and propose a future research focus.
Collapse
Affiliation(s)
- Dandan Xie
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, People’s Republic of China
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, Health Management Center, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
- Department of Clinical Nutrition, the First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Kai Li
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Ruxue Feng
- Department of Stomatology, Geriatric Hospital of Hainan, Haikou, Hainan, People’s Republic of China
| | - Man Xiao
- Department of Biochemistry and Molecular Biology, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| | - Zhifeng Sheng
- National Clinical Research Center for Metabolic Diseases, Hunan Provincial Key Laboratory of Metabolic Bone Diseases, Department of Metabolism and Endocrinology, Health Management Center, the Second Xiangya Hospital of Central South University, Changsha, Hunan, People’s Republic of China
| | - Yiqiang Xie
- College of Traditional Chinese Medicine, Hainan Medical University, Haikou, Hainan, People’s Republic of China
| |
Collapse
|
6
|
Pathak AK, Tiwari V, Kulshrestha MR, Singh S, Singh S, Singh V. Impact of essential metals on insulin sensitivity and fetuin in obesity-related type 2 diabetes pathogenesis. J Diabetes Metab Disord 2023; 22:703-712. [PMID: 37255834 PMCID: PMC10225454 DOI: 10.1007/s40200-023-01193-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 01/26/2023] [Indexed: 06/01/2023]
Abstract
Purpose Essential metals may be crucial in obesity and type 2 diabetes (T2DM); diabesity pathogenesis and consequences. This study aimed to determine the metal levels in obese and non-obese patients with and without T2DM and their relationships with fetuin-A(Fet-A) levels, insulin sensitivity, and insulin resistance. Methods A total of 314 participants were enrolled, with 160 newly diagnosed T2DM patients and 154 non-T2DM subjects categorized into diabetic obese (n = 57), diabetic non-obese (n = 103), non-diabetic obese (n = 48), and non-diabetic non-obese (n = 106) subgroups. Fet-A, insulin sensitivity (QUCKI)/resistance (HOMA-IR), fasting glucose, and body mass index (BMI) were assessed. The essential metals were measured using inductively coupled plasma mass spectroscopy (ICP-MS). Results Fet-A levels were 3-fold higher (1391.4 ± 839.8 ng/ml) in T2DM patients than in non-T2DM (2165.6 ± 651.9 vs. 424.3 ± 219.1 ng/ml, p < 0.0001). Fet-A levels were 2.3-fold higher in the diabetic obese group than in the diabetic non-obese group (p < 0.0001). Fet-A levels were 2.0-fold higher in the diabetic non-obese group than in the non-diabetic obese group (p < 0.0001). Fet-A levels were positively correlated with insulin resistance (HOMA-IR) (r = 0.34, p < 0.0001) and negatively correlated with insulin sensitivity (QUIKI) (r = -0.41, p < 0.0001).Cu, Se, Zn, and Fe levels were significantly lower in diabetic patients than in non-diabetic patients (p < 0.05). Se and Zn were significantly correlated with Fet-A (r = -0.41, p = 0.049 and r = -0.42, p = 0.001, respectively). Se and Zn were also correlated with insulin resistance (HOMA-IR) (r = -0.45, p = 0.049 and r = -0.36, p = 0.012, respectively) and insulin sensitivity (QUIKI) (r = 0.49, p = 0.042 and r = 0.30, p = 0.003, respectively). Similarly, Fe was negatively correlated with insulin levels (r = -0.33, p = 0.04) and insulin sensitivity (r = -0.34, p = 0.30). However, Mn was significantly correlated with Fet-A (r = 0.37, p = 0.001) and insulin resistance/sensitivity (r = 0.24, p = 0.026 and r = -0.24, p = 0.041) respectively in the diabetic obese group. Mg was an independent predictor of diabesity. Conclusions Mg play a significant role in obesity-related T2DM pathogenesis and complications via Fet-A, insulin sensitivity, and resistance modifications.
Collapse
Affiliation(s)
- Anumesh K. Pathak
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, 226010 India
| | - Vandana Tiwari
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, 226010 India
| | - Manish Raj Kulshrestha
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, 226010 India
| | - Shivani Singh
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, 226010 India
| | - Shefali Singh
- Department of Biochemistry, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, 226010 India
| | - Vikram Singh
- Department of General Medicine, Dr. Ram Manohar Lohia Institute of Medical Sciences, Lucknow, India
| |
Collapse
|
7
|
Valenti L, Corradini E, Adams LA, Aigner E, Alqahtani S, Arrese M, Bardou-Jacquet E, Bugianesi E, Fernandez-Real JM, Girelli D, Hagström H, Henninger B, Kowdley K, Ligabue G, McClain D, Lainé F, Miyanishi K, Muckenthaler MU, Pagani A, Pedrotti P, Pietrangelo A, Prati D, Ryan JD, Silvestri L, Spearman CW, Stål P, Tsochatzis EA, Vinchi F, Zheng MH, Zoller H. Consensus Statement on the definition and classification of metabolic hyperferritinaemia. Nat Rev Endocrinol 2023; 19:299-310. [PMID: 36805052 PMCID: PMC9936492 DOI: 10.1038/s41574-023-00807-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/19/2023]
Abstract
Hyperferritinaemia is a common laboratory finding that is often associated with metabolic dysfunction and fatty liver. Metabolic hyperferritinaemia reflects alterations in iron metabolism that facilitate iron accumulation in the body and is associated with an increased risk of cardiometabolic and liver diseases. Genetic variants that modulate iron homeostasis and tissue levels of iron are the main determinants of serum levels of ferritin in individuals with metabolic dysfunction, raising the hypothesis that iron accumulation might be implicated in the pathogenesis of insulin resistance and the related organ damage. However, validated criteria for the non-invasive diagnosis of metabolic hyperferritinaemia and the staging of iron overload are still lacking, and there is no clear evidence of a benefit for iron depletion therapy. Here, we provide an overview of the literature on the relationship between hyperferritinaemia and iron accumulation in individuals with metabolic dysfunction, and on the associated clinical outcomes. We propose an updated definition and a provisional staging system for metabolic hyperferritinaemia, which has been agreed on by a multidisciplinary global panel of expert researchers. The goal is to foster studies into the epidemiology, genetics, pathophysiology, clinical relevance and treatment of metabolic hyperferritinaemia, for which we provide suggestions on the main unmet needs, optimal design and clinically relevant outcomes.
Collapse
Affiliation(s)
- Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Biological Resource Center and Precision Medicine Lab, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
| | - Elena Corradini
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy.
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy.
| | - Leon A Adams
- Medical School, University of Western Australia, Perth, Australia
| | - Elmar Aigner
- First Department of Medicine, University Clinic Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Saleh Alqahtani
- Royal Clinics and Gastroenterology and Hepatology, King Faisal Specialist Hospital & Research Centre, Riyadh, Kingdom of Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Edouard Bardou-Jacquet
- University of Rennes, UMR1241, CHU Rennes, National Reference Center for Hemochromatosis and iron metabolism disorder, INSERM CIC1414, Rennes, France
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Turin, Italy
| | - Jose-Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Domenico Girelli
- Section of Internal Medicine, Department of Medicine, University of Verona, Policlinico Giambattista Rossi, Verona, Italy
| | - Hannes Hagström
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kris Kowdley
- Liver Institute Northwest, Seattle, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| | - Guido Ligabue
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Division of Radiology, Ospedale di Sassuolo S.p.A, Sassuolo, Modena, Italy
| | - Donald McClain
- Wake Forest School of Medicine, Winston Salem, NC, USA
- Department of Veterans Affairs, Salisbury, NC, USA
| | - Fabrice Lainé
- INSERM CIC1414, Liver Unit, CHU Rennes, Rennes, France
| | - Koji Miyanishi
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Martina U Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
- Center for Molecular Translational Iron Research, Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Heidelberg, Germany
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Pedrotti
- Laboratorio di RM Cardiaca Cardiologia 4, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy
| | - Daniele Prati
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - John D Ryan
- Hepatology Unit, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - C Wendy Spearman
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Per Stål
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley F.Kimball Research Institute, New York Blood Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Heinz Zoller
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
- Doppler Laboratory on Iron and Phosphate Biology, Innsbruck, Austria
| |
Collapse
|
8
|
Abstract
High iron is a risk factor for type 2 diabetes mellitus (T2DM) and affects most of its cardinal features: decreased insulin secretion, insulin resistance, and increased hepatic gluconeogenesis. This is true across the normal range of tissue iron levels and in pathologic iron overload. Because of iron's central role in metabolic processes (e.g., fuel oxidation) and metabolic regulation (e.g., hypoxia sensing), iron levels participate in determining metabolic rates, gluconeogenesis, fuel choice, insulin action, and adipocyte phenotype. The risk of diabetes related to iron is evident in most or all tissues that determine diabetes phenotypes, with the adipocyte, beta cell, and liver playing central roles. Molecular mechanisms for these effects are diverse, although there may be integrative pathways at play. Elucidating these pathways has implications not only for diabetes prevention and treatment, but also for the pathogenesis of other diseases that are, like T2DM, associated with aging, nutrition, and iron.
Collapse
Affiliation(s)
- Alexandria V Harrison
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
| | - Felipe Ramos Lorenzo
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
- Department of Veterans Affairs, W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina, USA
| | - Donald A McClain
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
- Department of Veterans Affairs, W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina, USA
| |
Collapse
|
9
|
The Effect of Dietary Protein Intake on the Risk of Gestational Diabetes. J FOOD QUALITY 2022. [DOI: 10.1155/2022/8368113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Background. The results of epidemiological studies on the association between dietary protein intake and gestational diabetes mellitus (GDM) are controversial. Thus, this systematic review and meta-analysis of cohort studies were established to attain comprehensive findings regarding the association between dietary protein and the risk of GDM. Methods. Bibliographic databases including PubMed, Scopus, Web of Science, and Google Scholar were searched to discover papers related to dietary protein and the risk of GDM. The summary relative risks with 95% confidence intervals (CIs) were estimated through a random effect model for the analysis of the highest versus the lowest categories of dietary proteins. Results. A significantly increased risk of GDM among women who consumed the highest amount of animal protein was observed (summarized risk estimate: 1.52; 95% CI: 1.07, 2.17; I2 = 50.8%). No significant associations were identified regarding vegetable protein (summarized risk estimate:0.99, 95% CI: 0.80 to 1.23, I2 = 63.8%) and total protein (summarized risk estimate: 1.12; 95% CI: 0.88, 1.41; I2 = 35.4%). Conclusion. This review revealed that total protein intake had no relationship with the risk of GDM, while animal protein increases this risk. Further larger prospective cohort studies are required to confirm our results.
Collapse
|
10
|
Quee FA, Peffer K, ter Braake AD, van den Hurk K. Cardiovascular benefits for blood donors? A systematic review. Transfus Med Rev 2022; 36:143-151. [DOI: 10.1016/j.tmrv.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 10/18/2022]
|
11
|
Hamad M, Mohammed AK, Hachim MY, Mukhopadhy D, Khalique A, Laham A, Dhaiban S, Bajbouj K, Taneera J. Heme Oxygenase-1 (HMOX-1) and inhibitor of differentiation proteins (ID1, ID3) are key response mechanisms against iron-overload in pancreatic β-cells. Mol Cell Endocrinol 2021; 538:111462. [PMID: 34547407 DOI: 10.1016/j.mce.2021.111462] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 09/08/2021] [Accepted: 09/16/2021] [Indexed: 12/13/2022]
Abstract
Iron overload promotes the generation of reactive oxygen species (ROS). Pancreatic β-cells can counter oxidative stress through multiple anti-oxidant responses. Herein, RNA-sequencing was used to describe the expression profile of iron regulatory genes in human islets with or without diabetes. Functional experiments including siRNA silencing, qPCR, western blotting, cell viability, ELISA and RNA-sequencing were performed as means of identifying the genetic signature of the protective response following iron overload-induced stress in human islets and INS-1. FTH1 and FTL genes were highly expressed in human islets and INS-1 cells, while hepcidin (HAMP) was low. FXN, DMT1 and FTHL1 genes were differentially expressed in diabetic islets compared to control. Silencing of Hamp in INS-1 cells impaired insulin secretion and influenced the expression of β-cell key genes. RNA-sequencing analysis in iron overloaded INS-1 cells identified Id1 and Id3 as the top down-regulated genes, while Hmox1 was the top upregulated. Expression of ID1, ID3 and HMOX1 was validated at the protein level in INS-1 cells and human islets. Differentially expressed genes (DEGs) were enriched for TGF-β, regulating stem cells, ferroptosis, and HIF-1 signaling. Hmox1-silenced cells treated with FAC elevated the expression of Id1 and Id3 expression than untreated cells. Our findings suggest that HMOX1, ID1 and ID3 define the response mechanism against iron-overload-induced stress in β-cells.
Collapse
Affiliation(s)
- Mawieh Hamad
- Department of Medical Lab. Sciences, College of Health Sciences, University of Sharjah, 27272, Sharjah, United Arab Emirates; Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Abdul Khader Mohammed
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Mahmood Y Hachim
- College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai, United Arab Emirates
| | - Debasmita Mukhopadhy
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Anila Khalique
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Amina Laham
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Sarah Dhaiban
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Khuloud Bajbouj
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates
| | - Jalal Taneera
- Sharjah Institute for Medical Research, University of Sharjah, 27272, Sharjah, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
12
|
Baum P, Toyka KV, Blüher M, Kosacka J, Nowicki M. Inflammatory Mechanisms in the Pathophysiology of Diabetic Peripheral Neuropathy (DN)-New Aspects. Int J Mol Sci 2021; 22:10835. [PMID: 34639176 PMCID: PMC8509236 DOI: 10.3390/ijms221910835] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/30/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022] Open
Abstract
The pathogenesis of diabetic neuropathy is complex, and various pathogenic pathways have been proposed. A better understanding of the pathophysiology is warranted for developing novel therapeutic strategies. Here, we summarize recent evidence from experiments using animal models of type 1 and type 2 diabetes showing that low-grade intraneural inflammation is a facet of diabetic neuropathy. Our experimental data suggest that these mild inflammatory processes are a likely common terminal pathway in diabetic neuropathy associated with the degeneration of intraepidermal nerve fibers. In contrast to earlier reports claiming toxic effects of high-iron content, we found the opposite, i.e., nutritional iron deficiency caused low-grade inflammation and fiber degeneration while in normal or high non-heme iron nutrition no or only extremely mild inflammatory signs were identified in nerve tissue. Obesity and dyslipidemia also appear to trigger mild inflammation of peripheral nerves, associated with neuropathy even in the absence of overt diabetes mellitus. Our finding may be the experimental analog of recent observations identifying systemic proinflammatory activity in human sensorimotor diabetic neuropathy. In a rat model of type 1 diabetes, a mild neuropathy with inflammatory components could be induced by insulin treatment causing an abrupt reduction in HbA1c. This is in line with observations in patients with severe diabetes developing a small fiber neuropathy upon treatment-induced rapid HbA1c reduction. If the inflammatory pathogenesis could be further substantiated by data from human tissues and intervention studies, anti-inflammatory compounds with different modes of action may become candidates for the treatment or prevention of diabetic neuropathy.
Collapse
Affiliation(s)
- Petra Baum
- Department of Neurology, University of Leipzig, Liebigstraße 20, D-04103 Leipzig, Germany;
| | - Klaus V. Toyka
- Department of Neurology, University of Würzburg, Josef-Schneider-Str. 11, D-97080 Würzburg, Germany;
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Liebigstraße 20, D-04103 Leipzig, Germany;
| | - Joanna Kosacka
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig, D-04103 Leipzig, Germany;
| | - Marcin Nowicki
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, D-04103 Leipzig, Germany
| |
Collapse
|
13
|
Winn NC, Wolf EM, Cottam MA, Bhanot M, Hasty AH. Myeloid-specific deletion of ferroportin impairs macrophage bioenergetics but is disconnected from systemic insulin action in adult mice. Am J Physiol Endocrinol Metab 2021; 321:E376-E391. [PMID: 34338042 PMCID: PMC8461794 DOI: 10.1152/ajpendo.00116.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/28/2021] [Accepted: 07/25/2021] [Indexed: 12/28/2022]
Abstract
Tissue iron overload is associated with insulin resistance and mitochondrial dysfunction in rodents and humans; however, the mechanisms or cell types that mediate this phenotype are not completely understood. Macrophages (Mɸs) are known to contribute to iron handling; thus, we hypothesized that perturbed iron handling by Mɸs impairs mitochondrial energetics and evokes systemic insulin resistance in mice. Male and female mice with myeloid-targeted (LysMCre) deletion of the canonical iron exporter, ferroportin (Fpn, encoded by Slc40a1), floxed littermates, and C57BL/6J wild-type mice were used to test our hypotheses. Myeloid-targeted deletion of Fpn evoked multitissue iron accumulation and reduced mitochondrial respiration in bone marrow-derived Mɸs, liver leukocytes, and Mɸ-enriched populations from adipose tissue (AT). In addition, a single bolus of exogenous iron administered to C57BL/6J mice phenocopied the loss of Fpn, resulting in a reduction in maximal and mitochondrial reserve capacity in Mɸ-enriched cellular fractions from liver and AT. In vivo exogenous iron chelation restored mitochondrial reserve capacity in liver leukocytes from Fpn LysMCre mice, but had no effect in AT myeloid populations. However, despite the impairments in mitochondrial respiration, neither loss of myeloid-specific Fpn nor exogenous iron overload perturbed glucose homeostasis or systemic insulin action in lean or obese mice, whereas aging coupled with lifelong loss of Fpn unmasked glucose intolerance. Together these data demonstrate that iron handling is critical for the maintenance of macrophage mitochondrial function, but perturbing myeloid iron flux via the loss of Fpn action is not sufficient to evoke systemic insulin resistance in young adult mice. These findings also suggest that if Mɸs are capable of storing iron properly, they have a pronounced ability to withstand iron excess without evoking overt collateral damage and associated insulin resistance that may be age dependent.NEW & NOTEWORTHY We used myeloid-specific knockout of ferroportin to determine whether macrophage iron enrichment alters systemic metabolism. We found that macrophages in several tissues showed mitochondrial defects such as a reduction in mitochondrial reserve capacity. However, insulin action in the mice was preserved. These findings also suggest that Mɸs have a pronounced ability to withstand iron excess without evoking overt collateral damage and associated insulin resistance, which appears to be age dependent.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Elysa M Wolf
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Matthew A Cottam
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Monica Bhanot
- Division of Diabetes, Endocrinology and Metabolism, Vanderbilt Medical Center, Nashville, Tennessee
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
- VA Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
14
|
Venkatesan P, Varghese J, Arthi TS, James JV, Anura A, Prasad J, Jacob M. Evidence of dysregulated iron homeostasis in newly diagnosed diabetics, but not in pre-diabetics. J Diabetes Complications 2021; 35:107977. [PMID: 34217587 DOI: 10.1016/j.jdiacomp.2021.107977] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 05/22/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022]
Abstract
AIM Diabetes mellitus has been reported to be associated with increased serum levels of ferritin. The basis of this association is unclear. It is also not precisely known whether other iron-related parameters, including hepcidin (the central regulator of systemic iron homeostasis), are affected under these circumstances. This study attempted to determine this. METHODS Adult men (normoglycemic or newly diagnosed with diabetes or pre-diabetes) were recruited. Anthropometric, metabolic, and hematological and iron-related parameters in blood were measured. Indices of insulin resistance (HOMA-IR) and pancreatic beta cell function (HOMA-β) were calculated. RESULTS Subjects in the 3 groups were similar in age, and anthropometric and hematological parameters. Serum ferritin and hepcidin levels were higher in diabetics, than in pre-diabetics and in control subjects. These elevations seen were not linked to the presence of inflammation. HOMA-IR was higher in diabetics, and HOMA-β lower in diabetics and pre-diabetics, than in control subjects. HOMA-IR and serum ferritin were positively correlated with one another. CONCLUSION Elevated levels of serum ferritin and hepcidin in newly diagnosed diabetics (but not pre-diabetics) indicate dysregulated iron homeostasis, with the former positively associated with insulin resistance in these patients.
Collapse
Affiliation(s)
- Padmanaban Venkatesan
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamil Nadu, India.
| | - Joe Varghese
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamil Nadu, India.
| | - T S Arthi
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamil Nadu, India
| | - Jithu V James
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamil Nadu, India
| | - Anji Anura
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamil Nadu, India
| | - Jasmin Prasad
- Department of Community Medicine, Christian Medical College, Vellore 632002, Tamil Nadu, India.
| | - Molly Jacob
- Department of Biochemistry, Christian Medical College, Vellore 632002, Tamil Nadu, India.
| |
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: 13] [Impact Index Per Article: 3.3] [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
|
Dahyaleh K, Sung HK, Prioriello M, Rengasamy P, Lam NH, Kim JB, Gross S, Sweeney G. Iron overload reduces adiponectin receptor expression via a ROS/FOXO1-dependent mechanism leading to adiponectin resistance in skeletal muscle cells. J Cell Physiol 2021; 236:5339-5351. [PMID: 33432609 DOI: 10.1002/jcp.30240] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 11/11/2022]
Abstract
Iron overload (IO) is a common yet underappreciated finding in metabolic syndrome (MetS) patients. With the prevalence of MetS continuing to rise, it is imperative to further elucidate cellular mechanisms leading to metabolic dysfunction. Adiponectin has many beneficial effects and is a therapeutic target for the treatment of MetS and cardiovascular diseases. IO positively correlates with reduced circulating adiponectin levels yet the impact of IO on adiponectin action is unknown. Here, we established a model of IO in L6 skeletal muscle cells and found that IO-induced adiponectin resistance. This was shown via reduced p38 mitogen-activated protein kinase phosphorylation in response to the small molecule adiponectin receptor (AdipoR) agonist, AdipoRon, in presence of IO. This correlated with reduced messenger RNA and protein levels of AdipoR1 and its facilitative signaling binding partner, APPL1. IO caused phosphorylation, nuclear extrusion, and thus inhibition of FOXO1, a known transcription factor regulating AdipoR1 expression. The antioxidant N-acetyl cystine attenuated the production of reactive oxygen species (ROS) by IO, and blunted its effect on FOXO1 phosphorylation and removal from the nucleus, as well as subsequent adiponectin resistance. In conclusion, our study identifies a ROS/FOXO1/AdipoR1 axis as a cause of skeletal muscle adiponectin resistance in response to IO. This new knowledge provides insight into a cellular mechanism with potential relevance to disease pathophysiology in MetS patients with IO.
Collapse
Affiliation(s)
| | - Hye K Sung
- Department of Biology, York University, Toronto, Canada
| | | | | | - Nhat H Lam
- Department of Biology, York University, Toronto, Canada
| | - Jae B Kim
- School of Biological Sciences, Seoul National University, Seoul, South Korea
| | - Sean Gross
- Department of Biomedical Engineering, OHSU Center for Spatial Systems Biomedicine, Knight Cancer Institute, Oregon Health and Sciences University, Portland, Oregon, USA
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
17
|
Abstract
Low-grade chronic adipose tissue (AT) inflammation is now recognized as a pivotal driver of the multi-organ dysfunction associated with obesity-related complications; and adipose tissue macrophages (ATMs) are key to the development of this inflammatory milieu. Along with their role in immunosurveillance, ATMs are central regulators of AT iron homeostasis. Under optimal conditions, ATMs maintain a proper homeostatic balance of iron in adipocytes; however, during obesity, this relationship is altered, and iron is repartitioned into adipocytes as opposed to ATMs. This adipocyte iron overload leads to systemic IR and the mechanism for these effects is still under investigation. Here, we comment on the most recent findings addressing the interplay between adipocyte and ATM iron handling, and metabolic dysfunction.
Collapse
|
18
|
Olgun NS, Morris AM, Bowers LN, Stefaniak AB, Friend SA, Reznik SE, Leonard SS. Mild steel and stainless steel welding fumes elicit pro-inflammatory and pro-oxidant effects in first trimester trophoblast cells. Am J Reprod Immunol 2020; 83:e13221. [PMID: 31943498 PMCID: PMC7079021 DOI: 10.1111/aji.13221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/18/2019] [Accepted: 12/07/2019] [Indexed: 12/17/2022] Open
Abstract
Problem As more women join the skilled‐trade workforce, the effects of workplace exposures on pregnancy need to be explored. This study aims to identify the effects of mild steel and stainless steel welding fume exposures on cultured placental trophoblast cells. Method of study Welding fumes (mild steel and stainless steel) were generously donated by Lincoln Electric. Electron microscopy was used to characterize welding fume particle size and the ability of particles to enter extravillous trophoblast cells (HTR‐8/SVneo). Cellular viability, free radical production, cytokine production, and ability of cells to maintain invasive properties were analyzed, respectively, by WST‐1, electron paramagnetic resonance, DCFH‐DA, V‐plex MULTI‐SPOT assay system, and a matrix gel invasion assay. Results For all three welding fume types, average particle size was <210 nm. HTR‐8/SVneo cells internalized welding particles, and nuclear condensation was observed. Cellular viability was significantly decreased at the high dose of 100 µg/mL for all three welding fumes, and stainless steel generated the greatest production of the hydroxyl radical, and intracellular reactive oxygen species. Production of the cytokines IL‐1β and TNFα were not observed in response to welding fume exposure, but IL‐6 and IL‐8 were. Finally, the invasive capability of cells was decreased upon exposure to both mild steel and stainless steel welding fumes. Conclusion Welding fumes are cytotoxic to extravillous trophoblasts, as is evident by the production of free radicals, pro‐inflammatory cytokines, and the observed decrease in invasive capabilities.
Collapse
Affiliation(s)
- Nicole S Olgun
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia
| | - Anna M Morris
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia
| | - Lauren N Bowers
- Respiratory Health Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia
| | - Aleksandr B Stefaniak
- Respiratory Health Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia
| | - Sherri A Friend
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia
| | - Sandra E Reznik
- Department of Pharmaceutical Sciences, St. John's University, Queens, New York
| | - Stephen S Leonard
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, West Virginia
| |
Collapse
|
19
|
Peffer K, den Heijer M, de Kort WLAM, Verbeek ALM, Atsma F. Cardiovascular risk in 159 934 frequent blood donors while addressing the healthy donor effect. Heart 2019; 105:1260-1265. [PMID: 30872386 DOI: 10.1136/heartjnl-2018-314138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/30/2019] [Accepted: 02/08/2019] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE To investigate whether regular blood donation decreases cardiovascular risk. METHODS All 159 934 Dutch whole-blood donors with an active donation career of at least 10 years were categorised into sex-specific donation tertiles based on the number of donations during this 10-year qualification period. Cardiovascular endpoints were based on hospital discharge diagnoses and death certificates from Dutch Hospital Data and Statistics Netherlands and occurring after the 10-year qualification period. Cox regression was used to estimate the age-adjusted and starting year-adjusted hazard rate ratio (HRR). RESULTS Female high-frequency blood donors had a reduced cardiovascular morbidity (HRR=0.91, 95% CI 0.85 to 0.98) compared with low-frequency blood donors. No effect was observed in men (HRR=1.00, 95% CI 0.95 to 1.05). To rule out a residual healthy donor effect (HDE), additional sensitivity analyses using a 5-year qualification period were conducted. The results supported the absence of a residual HDE. CONCLUSIONS This study showed a protective effect of long-term, high-frequency blood donation against cardiovascular disease. This effect was only observed in women and not in men.
Collapse
Affiliation(s)
- Karlijn Peffer
- Department of Donor Medicine Research - Donor Studies, Sanquin Research, Amsterdam, The Netherlands
| | - Martin den Heijer
- Department of Internal Medicine, VU University Medical Centre, Amsterdam, The Netherlands
| | - Wim L A M de Kort
- Department of Donor Medicine Research - Donor Studies, Sanquin Research, Amsterdam, The Netherlands.,Department of Public Health, Academic Medical Center Amsterdam UMC - Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - André L M Verbeek
- Department for Health Evidence, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Femke Atsma
- Department of Donor Medicine Research - Donor Studies, Sanquin Research, Amsterdam, The Netherlands.,Radboud Institute for Health Sciences, Scientific Center for Quality of Healthcare, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
20
|
Zhou Q, Guo W, Jia Y, Xu J. Comparison of Chromium and Iron Distribution in Serum and Urine among Healthy People and Prediabetes and Diabetes Patients. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3801639. [PMID: 30915353 PMCID: PMC6409039 DOI: 10.1155/2019/3801639] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 02/10/2019] [Indexed: 12/23/2022]
Abstract
The effect of chromium (Cr) and iron (Fe) on prevalence of diabetes has received great attention. This study investigated serum and urinary Cr and Fe levels among patients with impaired fasting glucose (IFG), impaired glucose tolerance (IGT), type 1 diabetes (T1D), and type 2 diabetes (T2D) in the Northeast Chinese population. From January 2010 to October 2011, patients with IFG (n=12), IGT (n=15), T1D (n=25), T2D (n=137) and healthy controls (n=50) were enrolled in the First Hospital of Jilin University. Trace elements were detected using an inductively coupled plasma spectrometer. Serum Cr levels decreased in T2D without complications, diabetic retinopathy (DR), diabetic peripheral neuropathy (DPN), and diabetic nephropathy (DN) (P<0.05). The urinary Cr level in T1D was the highest of all, which significantly exceeded those of the T2D groups with and without complications. No significant differences of serum Fe levels were found among all groups. The urinary Fe level of T1D was significantly increased (P<0.05). The correlation between serum Cr and serum Fe in T2D was obviously positive (P<0.05). One month of simvastatin therapy exerted no effects on serum or urinary Cr and Fe levels. These results suggest the potential role of Cr and Fe in diabetes should receive attention.
Collapse
Affiliation(s)
- Qi Zhou
- Department of Pediatrics, First Hospital of Jilin University, Changchun 130021, China
| | - Wenjia Guo
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Yanan Jia
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Jiancheng Xu
- Department of Laboratory Medicine, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
21
|
Serum ferritin levels are associated with insulin resistance in Chinese men and post-menopausal women: the Shanghai Changfeng study. Br J Nutr 2018; 120:863-871. [PMID: 30189905 DOI: 10.1017/s0007114518002167] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Associations between ferritin and insulin sensitivity have been described in recent studies. The possible association showed conflicting results by sex and menopausal status. We aimed to investigate the cross-sectional association of ferritin levels with insulin resistance and β-cell function. A total of 2518 participants (1033 men, 235 pre-menopausal women and 1250 post-menopausal women) were enrolled from the Changfeng Study. A standard interview was conducted, as well as anthropometric measurements and laboratory analyses, for each participant. The serum ferritin level was measured using electrochemiluminescence immunoassay. Insulin resistance and β-cell function indices were derived from a homeostasis model assessment. The results showed that the serum ferritin levels were 250·4 (sd 165·2), 94·6 (sd 82·0) and 179·8 (sd 126·6) ng/ml in the men, pre-menopausal and post-menopausal women, respectively. In fully adjusted models (adjusting for age, current smoking, BMI, waist:hip ratio, systolic blood pressure, diastolic blood pressure, TAG, HDL-cholesterol, LDL-cholesterol, log urine albumin:creatinine ratio, leucocytes, alanine aminotransferase, aspartate aminotransferase and γ-glutamyl transpeptidase), serum ferritin concentrations are significantly associated with insulin resistance in men and post-menopausal females, and the null association was observed in pre-menopausal females. Interestingly, an increased β-cell function associated with higher ferritin was observed in post-menopausal participants, but not in male participants. In conclusion, these results suggested that elevated serum ferritin levels were associated with surrogate measures of insulin resistance among the middle-aged and elderly male and post-menopausal women, but not in pre-menopausal women.
Collapse
|
22
|
Prion protein modulates glucose homeostasis by altering intracellular iron. Sci Rep 2018; 8:6556. [PMID: 29700330 PMCID: PMC5919926 DOI: 10.1038/s41598-018-24786-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 03/20/2018] [Indexed: 11/08/2022] Open
Abstract
The prion protein (PrPC), a mainly neuronal protein, is known to modulate glucose homeostasis in mouse models. We explored the underlying mechanism in mouse models and the human pancreatic β-cell line 1.1B4. We report expression of PrPC on mouse pancreatic β-cells, where it promoted uptake of iron through divalent-metal-transporters. Accordingly, pancreatic iron stores in PrP knockout mice (PrP-/-) were significantly lower than wild type (PrP+/+) controls. Silencing of PrPC in 1.1B4 cells resulted in significant depletion of intracellular (IC) iron, and remarkably, upregulation of glucose transporter GLUT2 and insulin. Iron overloading, on the other hand, resulted in downregulation of GLUT2 and insulin in a PrPC-dependent manner. Similar observations were noted in the brain, liver, and neuroretina of iron overloaded PrP+/+ but not PrP-/- mice, indicating PrPC-mediated modulation of insulin and glucose homeostasis through iron. Peripheral challenge with glucose and insulin revealed blunting of the response in iron-overloaded PrP+/+ relative to PrP-/- mice, suggesting that PrPC-mediated modulation of IC iron influences both secretion and sensitivity of peripheral organs to insulin. These observations have implications for Alzheimer's disease and diabetic retinopathy, known complications of type-2-diabetes associated with brain and ocular iron-dyshomeostasis.
Collapse
|
23
|
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]
|
24
|
Wankanit S, Chuansumrit A, Poomthavorn P, Khlairit P, Pongratanakul S, Mahachoklertwattana P. Acute Effects of Blood Transfusion on Insulin Sensitivity and Pancreatic β-Cell Function in Children with β-Thalassemia/Hemoglobin E Disease. J Clin Res Pediatr Endocrinol 2018; 10:1-7. [PMID: 28739553 PMCID: PMC5838366 DOI: 10.4274/jcrpe.4774] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE To assess the acute effects of blood transfusion on insulin sensitivity and pancreatic β-cell function in thalassemia patients. METHODS Fifty children and adolescents with β-thalassemia/HbE disease were enrolled in a prospective cohort study. Hemoglobin, serum ferritin and oral glucose tolerance test (OGTT) were performed prior to, and one week after blood transfusion. Insulin sensitivity indices [homeostatic model assessment (HOMA) of insulin resistance (HOMA-IR), whole body insulin sensitivity index (WBISI)] and β-cell function indices [HOMA of β-cell function (HOMA-β), insulinogenic index (IGI), and disposition index (DI)] were calculated from glucose and insulin levels obtained during the OGTT. RESULTS Following blood transfusion, hemoglobin and serum ferritin increased significantly; 8.5 to 10.1 g/dL (p<0.001) and 1764 to 2160 ng/mL (p<0.001), respectively. β-Cell function indices also increased significantly [median HOMA-β: 74.3 vs. 82.7 (p=0.033); median IGI: 59.6 vs. 79.3 (p=0.003); median DI: 658 vs. 794 (p=0.01)]. However, the insulin sensitivity index (WBISI) tended to decrease and the insulin resistance index (HOMA-IR) tended to increase although this did not reach significance. Multivariate analysis showed that pre-transfusion serum ferritin was the major factor negatively associated with WBISI and positively associated with HOMA-IR, but pre-transfusion hemoglobin had no significant association with insulin sensitivity indices post-transfusion. CONCLUSION This study demonstrated that acute increases in serum ferritin and hemoglobin following blood transfusion in patients with thalassemia might contribute to an increase in insulin secretion and to a trend towards increased insulin resistance.
Collapse
Affiliation(s)
- Somboon Wankanit
- Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Endocrinology and Metabolism, Bangkok, Thailand
| | - Ampaiwan Chuansumrit
- Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Hemato-Oncology, Bangkok, Thailand
| | - Preamrudee Poomthavorn
- Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Endocrinology and Metabolism, Bangkok, Thailand
| | - Patcharin Khlairit
- Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Endocrinology and Metabolism, Bangkok, Thailand
| | - Sarunyu Pongratanakul
- Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Endocrinology and Metabolism, Bangkok, Thailand
| | - Pat Mahachoklertwattana
- Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Endocrinology and Metabolism, Bangkok, Thailand,* Address for Correspondence: Mahidol University Faculty of Medicine, Ramathibodi Hospital, Department of Pediatrics, Divisions of Endocrinology and Metabolism, Bangkok, Thailand Phone: +662-201-1394 E-mail:
| |
Collapse
|
25
|
Machado MV, Diehl AM. Pathogenesis of Nonalcoholic Fatty Liver Disease. ZAKIM AND BOYER'S HEPATOLOGY 2018:369-390.e14. [DOI: 10.1016/b978-0-323-37591-7.00025-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Wilson MS, Thompson EW. Venesection is an effective non-pharmacological treatment for patients with high serum ferritin type 2 diabetes. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2017. [DOI: 10.1016/j.jecr.2016.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
27
|
Fernández-Cao JC, Arija V, Aranda N, Basora J, Diez-Espino J, Estruch R, Fitó M, Corella D, Salas-Salvadó J. Soluble transferrin receptor and risk of type 2 diabetes in the obese and nonobese. Eur J Clin Invest 2017; 47:221-230. [PMID: 28075490 DOI: 10.1111/eci.12725] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 01/09/2017] [Indexed: 12/20/2022]
Abstract
BACKGROUND Studies evaluating the relationship between soluble transferrin receptor (sTfR), a biomarker inversely related to body iron stores, and risk of type 2 diabetes mellitus (T2DM) are scarce and inconclusive. Furthermore, sTfR concentrations have been observed to be significantly higher in obese than in nonobese individuals. Therefore, the aim of this study was to assess the relationship between sTfR and the risk of T2DM in obese and nonobese subjects. DESIGN A nested case-control study of 153 cases of newly diagnosed diabetic subjects, 73 obese and 80 nonobese, and 306 individually matched controls, 138 obese and 166 nonobese, who did not develop T2DM for a median 6-year follow-up (interquartile range: 3·9-6·5) was conducted using data from the PREvention with MEDiterranean Diet (PREDIMED) cohort (http://www.controlled-trials.com/ISRCTN35739639). Cases and controls were matched for age (≤ 67 vs. > 67 years), gender, dietary intervention group and BMI (≤ 27 vs. > 27 kg/m2 ). RESULTS Waist circumference is the main determinant of sTfR concentrations in the whole sample (β = 0·476, P < 0·001), in the obese (β = 0·802, P < 0·001) and the nonobese (β = 0·455, P = 0·003). Furthermore, sTfR is directly associated with the risk of T2DM in obese individuals (OR = 2·79; 95% CI: 1·35-5·77, P = 0·005) and inversely associated in nonobese individuals (OR = 0·40; 95% CI: 0·20-0·79, P = 0·015). CONCLUSIONS The association between sTfR levels and risk of T2DM in a population at high cardiovascular risk depend on the presence or absence of obesity. While in nonobese subjects elevated sTfR levels are associated with a decreased risk of developing T2DM, in obese subjects the risk increases. This suggests that obesity alters the relationship between sTfR and T2DM incidence.
Collapse
Affiliation(s)
- José C Fernández-Cao
- Nutrition and Public Health Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Department of Nutrition and Dietetics, Faculty of Health Sciences, University of Atacama, Copiapó, Chile
| | - Victoria Arija
- Nutrition and Public Health Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Reus-Altebrat Primary Care, Institut d'Investigació en Atenció Primària (IDIAP), Jordi Gol i Gurina, Reus, Spain.,Pere i Virgili, Health Research Institute (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Núria Aranda
- Nutrition and Public Health Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,Pere i Virgili, Health Research Institute (IISPV), Universitat Rovira i Virgili, Reus, Spain
| | - Josep Basora
- Reus-Altebrat Primary Care, Institut d'Investigació en Atenció Primària (IDIAP), Jordi Gol i Gurina, Reus, Spain.,Pere i Virgili, Health Research Institute (IISPV), Universitat Rovira i Virgili, Reus, Spain.,Human Nutrition Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| | - Javier Diez-Espino
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,Primary Health Care Centre of Tafalla, Servicio Navarro de Salud-Osasunbidea, Navarra, Spain
| | - Ramón Estruch
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,Department of Internal Medicine, Hospital Clínic, Institut d'Investigació Biomédica August Pi I Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Montse Fitó
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,Cardiovascular Risk and Nutrition Research Group, Municipal Institute for Medical Research (IMIM-Hospital del Mar), Barcelona, Spain
| | - Dolores Corella
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain.,Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
| | - Jordi Salas-Salvadó
- Pere i Virgili, Health Research Institute (IISPV), Universitat Rovira i Virgili, Reus, Spain.,Human Nutrition Unit, Faculty of Medicine and Health Sciences, Universitat Rovira i Virgili, Reus, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Barton JC, Acton RT. Diabetes in HFE Hemochromatosis. J Diabetes Res 2017; 2017:9826930. [PMID: 28331855 PMCID: PMC5346371 DOI: 10.1155/2017/9826930] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/12/2016] [Accepted: 01/04/2017] [Indexed: 02/08/2023] Open
Abstract
Diabetes in whites of European descent with hemochromatosis was first attributed to pancreatic siderosis. Later observations revealed that the pathogenesis of diabetes in HFE hemochromatosis is multifactorial and its clinical manifestations are heterogeneous. Increased type 2 diabetes risk in HFE hemochromatosis is associated with one or more factors, including abnormal iron homeostasis and iron overload, decreased insulin secretion, cirrhosis, diabetes in first-degree relatives, increased body mass index, insulin resistance, and metabolic syndrome. In p.C282Y homozygotes, serum ferritin, usually elevated at hemochromatosis diagnosis, largely reflects body iron stores but not diabetes risk. In persons with diabetes type 2 without hemochromatosis diagnoses, serum ferritin levels are higher than those of persons without diabetes, but most values are within the reference range. Phlebotomy therapy to achieve iron depletion does not improve diabetes control in all persons with HFE hemochromatosis. The prevalence of type 2 diabetes diagnosed today in whites of European descent with and without HFE hemochromatosis is similar. Routine iron phenotyping or HFE genotyping of patients with type 2 diabetes is not recommended. Herein, we review diabetes in HFE hemochromatosis and the role of iron in diabetes pathogenesis in whites of European descent with and without HFE hemochromatosis.
Collapse
Affiliation(s)
- James C. Barton
- Southern Iron Disorders Center, Birmingham, AL 35209, USA
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ronald T. Acton
- Southern Iron Disorders Center, Birmingham, AL 35209, USA
- Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
29
|
De Sanctis V, Soliman AT, Elsedfy H, Yaarubi SAL, Skordis N, Khater D, El Kholy M, Stoeva I, Fiscina B, Angastiniotis M, Daar S, Kattamis C. The ICET-A Recommendations for the Diagnosis and Management of Disturbances of Glucose Homeostasis in Thalassemia Major Patients. Mediterr J Hematol Infect Dis 2016; 8:e2016058. [PMID: 27872738 PMCID: PMC5111521 DOI: 10.4084/mjhid.2016.058] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Accepted: 09/20/2016] [Indexed: 01/19/2023] Open
Abstract
Iron overload in patients with thalassemia major (TM) affects glucose regulation and is mediated by several mechanisms. The pathogenesis of glycaemic abnormalities in TM is complex and multifactorial. It has been predominantly attributed to a combination of reduced insulin secretory capacity and insulin resistance. The exact mechanisms responsible for progression from norm glycaemia to overt diabetes in these patients are still poorly understood but are attributed mainly to insulin deficiency resulting from the toxic effects of iron deposited in the pancreas and insulin resistance. A group of endocrinologists, haematologists and paediatricians, members of the International Network of Clinicians for Endocrinopathies in Thalassemia and Adolescence Medicine (ICET-A) convened to formulate recommendations for the diagnosis and management of abnormalities of glucose homeostasis in thalassemia major patients on the basis of available evidence from clinical and laboratory data and consensus practice. The results of their work and discussions are described in this article.
Collapse
Affiliation(s)
| | - Ashraf T. Soliman
- Department of Pediatrics, Division of Endocrinology, Alexandria University Children’s Hospital, Alexandria, Egypt
| | - Heba Elsedfy
- Department of Pediatrics, Ain Shams University, Cairo, Egypt
| | - Saif AL Yaarubi
- Pediatric Endocrine Unit, Department of Child Health, Sultan Qaboos University Hospital, Al-Khoud, Sultanate of Oman
| | - Nicos Skordis
- Division of Pediatric and Adolescent Endocrinology, Paedi Center for Specialized Pediatrics, St. George’s University Medical School at the University of Nicosia, Cyprus
| | - Doaa Khater
- Department of Pediatrics, Endocrinology Unit, Alexandria University Children’s Hospital, Egypt, and Child Health Department, Sultan Qaboos University Hospital, Muscat, Oman
| | | | - Iva Stoeva
- Paediatric Endocrinologist,”Screening and Functional Endocrine Diagnostics” SBALDB. Professor Ivan Mitev, Medical University Sofia, Bulgaria
| | | | | | - Shahina Daar
- Department of Hematology, College of Medicine and Health Sciences Sultan Qaboos University Oman, Sultanate of Oman & Visiting Scholar, Stellenbosch Institute for Advanced Study (STIAS), Wallenberg Research Centre at Stellenbosch University, Stellenbosch 7600, South Africa
| | - Christos Kattamis
- First Department of Paediatrics, University of Athens, Athens, Greece
| |
Collapse
|
30
|
Shimodaira M, Okaniwa S, Nakayama T. Investigation of the relationship between hemoglobin and serum iron levels and early-phase insulin secretion in non-diabetic subjects. Acta Diabetol 2016; 53:783-9. [PMID: 27311688 DOI: 10.1007/s00592-016-0873-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 05/30/2016] [Indexed: 12/17/2022]
Abstract
AIMS Recent biological and epidemiological studies have found that insulin resistance is linked to iron overload. However, little is known about the association between hemoglobin and/or serum iron levels and pancreatic β-cell function. In this gender-separated cross-sectional study, we aimed to investigate the association of hemoglobin and serum iron levels with early-phase insulin secretion in non-diabetic subjects. METHODS A total of 804 non-diabetic Japanese subjects (482 males and 322 females) aged over 30 years old were enrolled in the study. Early-phase insulin secretion was estimated using the insulinogenic index (IGI [ΔInsulin(30-0 min)/ΔGlucose(30-0 min)]) during a 75-g oral glucose tolerance test. RESULTS Simple linear regression analysis showed that IGI negatively correlated with hemoglobin levels in male but not in female subjects. However, IGI did not correlate with serum iron levels in either gender. Multivariate linear regression analysis in male subjects revealed that hemoglobin levels were predictors of IGI, responsible for 3.0 % of IGI variation (P = 0.008). The association was independent of age, BMI, fasting glucose and insulin levels, and lipid profiles. In non-diabetic Japanese males, hemoglobin levels significantly and negatively correlated with early-phase insulin secretion. CONCLUSIONS Our finding suggests that elevated hemoglobin levels may have a gender-specific impact on β-cell function and could be an independent predictor of β-cell dysfunction.
Collapse
Affiliation(s)
- Masanori Shimodaira
- Department of Internal Medicine, Iida Municipal Hospital, 438 Yawata-machi, Iida, Nagano, 395-8502, Japan.
- Division of Companion Diagnostics, Department of Pathology of Microbiology, Nihon University School of Medicine, 30-1 Ooyaguchi-kamimachi, Itabashi-ku, Tokyo, 173-8610, Japan.
| | - Shinji Okaniwa
- Department of Internal Medicine, Iida Municipal Hospital, 438 Yawata-machi, Iida, Nagano, 395-8502, Japan
| | - Tomohiro Nakayama
- Division of Companion Diagnostics, Department of Pathology of Microbiology, Nihon University School of Medicine, 30-1 Ooyaguchi-kamimachi, Itabashi-ku, Tokyo, 173-8610, Japan
| |
Collapse
|
31
|
González-Villalva A, Colín-Barenque L, Bizarro-Nevares P, Rojas-Lemus M, Rodríguez-Lara V, García-Pelaez I, Ustarroz-Cano M, López-Valdez N, Albarrán-Alonso JC, Fortoul TI. Pollution by metals: Is there a relationship in glycemic control? ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2016; 46:337-343. [PMID: 27552445 DOI: 10.1016/j.etap.2016.06.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 06/06/2023]
Abstract
There are evidences of environmental pollution and health effects. Metals are pollutants implicated in systemic toxicity. One of the least studied effects, but which is currently becoming more important, is the effect of metals on glycemic control. Metals have been implicated as causes of chronic inflammation and oxidative stress and are associated to obesity, hyperglycemia and even diabetes. Arsenic, iron, mercury, lead, cadmium and nickel have been studied as a risk factor for hyperglycemia and diabetes. There is another group of metals that causes hypoglycemia such as vanadium, chromium, zinc and magnesium by different mechanisms. Zinc, magnesium and chromium deficiency is associated with increased risk of diabetes. This review summarizes some metals involved in glycemic control and pretends to alert health professionals about considering environmental metals as an important factor that could explain the poor glycemic control in patients. Further studies are needed to understand this poorly assessed problem.
Collapse
Affiliation(s)
- Adriana González-Villalva
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Laura Colín-Barenque
- Laboratorio de Neuromorfología, FES Iztacala, UNAM CP 54090 Edo. de México, Mexico.
| | - Patricia Bizarro-Nevares
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Marcela Rojas-Lemus
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Vianey Rodríguez-Lara
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Isabel García-Pelaez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Martha Ustarroz-Cano
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Nelly López-Valdez
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Juan Carlos Albarrán-Alonso
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| | - Teresa I Fortoul
- Departamento de Biología Celular y Tisular, Facultad de Medicina, Universidad Nacional Autonoma de Mexico (UNAM), Mexico City, Mexico.
| |
Collapse
|
32
|
Shu XB, Zhang L, Huang J, Ji G. Relationship between iron overload and nonalcoholic fatty liver disease: An update. Shijie Huaren Xiaohua Zazhi 2016; 24:3398-3403. [DOI: 10.11569/wcjd.v24.i22.3398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
As a component of the multiple hits, iron overload plays an important role in the development of nonalcoholic fatty liver disease (NAFLD). Iron overload could affect glucose, lipid, energy metabolism and inflammatory reaction. This paper reviews the relationship between iron overload and nonalcoholic fatty liver disease, in order to clarify how and why iron overload influences the progression of NAFLD.
Collapse
|
33
|
Yang A, Zhao J, Lu M, Gu Y, Zhu Y, Chen D, Fu J. Expression of Hepcidin and Ferroportin in the Placenta, and Ferritin and Transferrin Receptor 1 Levels in Maternal and Umbilical Cord Blood in Pregnant Women with and without Gestational Diabetes. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:E766. [PMID: 27483296 PMCID: PMC4997452 DOI: 10.3390/ijerph13080766] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 07/05/2016] [Accepted: 07/19/2016] [Indexed: 02/05/2023]
Abstract
BACKGROUND Regulation of iron transfer from mother to fetus via the placenta is not fully understood and the relationship between stored iron status in the mothers' serum and gestational diabetes (GDM) in case-control studies is controversial. The present study aimed to detect circulating soluble transferrin receptor (sTfR) and ferritin levels in maternal and umbilical cord blood. We also examined the expression of hepcidin (Hep), transferrin receptor (TfR1), and ferroportin (FPN) in the placenta in pregnant women with and without GDM at full term. METHODS Eighty-two women participated (42 with GDM and 40 without GDM [controls]). Maternal samples were collected at 37-39 weeks' gestation. Umbilical cord blood was collected at birth. Ferritin and sTfR levels in maternal serum and umbilical cord blood, and Hep, TfR1, and FPN protein expression in plac enta were compared between the GDM and non-GDM groups. Serum ferritin (SF) was measured by electrochemiluminescence assay and sTfR was measured by ELISA. Hep, TfR1, and FPN expression was measured by immunohistochemistry. RESULTS Maternal serum sTfR levels were significantly elevated in the GDM group compared with the non-GDM group (p = 0.003). SF levels in cord blood in the GDM group were significantly higher than those in the non-GDM group (p = 0.003). However, maternal hemoglobin and SF, and umbilical cord sTfR levels were not different between the groups. In placental tissue, FPN expression was higher and hepcidin expression was lower in the GDM group compared with the non-GDM group (p = 0.000 and p = 0.044, respectively). There was no significant difference in TfR1 between the groups (p = 0.898). CONCLUSIONS Women with GDM transport iron more actively than those without GDM at term pregnancy. Maternal iron metabolism in GDM may play a role in fetal/placental iron demand and in the overall outcome of pregnancy.
Collapse
Affiliation(s)
- Anqiang Yang
- Department of Pathology, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Jun Zhao
- Clinical Laboratory, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Minhua Lu
- Department of Pathology, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Ying Gu
- Department of Obstetrics, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Yunlong Zhu
- Department of Obstetrics, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Daozhen Chen
- Clinical Laboratory, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| | - Jinyan Fu
- Department of Pathology, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
- Department of Obstetrics, Wuxi Maternity and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China.
| |
Collapse
|
34
|
Pisano G, Lombardi R, Fracanzani AL. Vascular Damage in Patients with Nonalcoholic Fatty Liver Disease: Possible Role of Iron and Ferritin. Int J Mol Sci 2016; 17:ijms17050675. [PMID: 27164079 PMCID: PMC4881501 DOI: 10.3390/ijms17050675] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 04/20/2016] [Accepted: 04/26/2016] [Indexed: 02/06/2023] Open
Abstract
Non Alcoholic Fatty Liver Disease (NAFLD) is the most common chronic liver disease in Western countries. Recent data indicated that NAFLD is a risk factor by itself contributing to the development of cardiovascular disease independently of classical known risk factors. Hyperferritinemia and mild increased iron stores are frequently observed in patients with NAFLD and several mechanisms have been proposed to explain the role of iron, through oxidative stress and interaction with insulin metabolism, in the development of vascular damage. Moreover, iron depletion has been shown to decrease atherogenesis in experimental models and in humans. This review presents the recent evidence on epidemiology, pathogenesis, and the possible explanation of the role of iron and ferritin in the development of cardiovascular damage in patients with NAFLD, and discusses the possible interplay between metabolic disorders associated with NAFLD and iron in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Giuseppina Pisano
- Department of Pathophysiology and Transplantation, Ca' Granda IRCCS Foundation, Policlinico Hospital, University of Milan, Centre of the Study of Metabolic and Liver Diseases, Via Francesco Sforza 35, 20122 Milan, Italy.
| | - Rosa Lombardi
- Department of Pathophysiology and Transplantation, Ca' Granda IRCCS Foundation, Policlinico Hospital, University of Milan, Centre of the Study of Metabolic and Liver Diseases, Via Francesco Sforza 35, 20122 Milan, Italy.
| | - Anna Ludovica Fracanzani
- Department of Pathophysiology and Transplantation, Ca' Granda IRCCS Foundation, Policlinico Hospital, University of Milan, Centre of the Study of Metabolic and Liver Diseases, Via Francesco Sforza 35, 20122 Milan, Italy.
| |
Collapse
|
35
|
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: 63] [Impact Index Per Article: 7.0] [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
|
36
|
De Sanctis V, Soliman AT, Elsedfy H, Pepe A, Kattamis C, El Kholy M, Yassin M. Diabetes and Glucose Metabolism in Thalassemia Major: An Update. Expert Rev Hematol 2016; 9:401-8. [DOI: 10.1586/17474086.2016.1136209] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
37
|
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
|
38
|
Schreinemachers DM, Ghio AJ. Effects of Environmental Pollutants on Cellular Iron Homeostasis and Ultimate Links to Human Disease. ENVIRONMENTAL HEALTH INSIGHTS 2016; 10:35-43. [PMID: 26966372 PMCID: PMC4782969 DOI: 10.4137/ehi.s36225] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/20/2016] [Accepted: 01/21/2016] [Indexed: 05/04/2023]
Abstract
Chronic disease has increased in the past several decades, and environmental pollutants have been implicated. The magnitude and variety of diseases may indicate the malfunctioning of some basic mechanisms underlying human health. Environmental pollutants demonstrate a capability to complex iron through electronegative functional groups containing oxygen, nitrogen, or sulfur. Cellular exposure to the chemical or its metabolite may cause a loss of requisite functional iron from intracellular sites. The cell is compelled to acquire further iron critical to its survival by activation of iron-responsive proteins and increasing iron import. Iron homeostasis in the exposed cells is altered due to a new equilibrium being established between iron-requiring cells and the inappropriate chelator (the pollutant or its catabolite). Following exposure to environmental pollutants, the perturbation of functional iron homeostasis may be the mechanism leading to adverse biological effects. Understanding the mechanism may lead to intervention methods for this major public health concern.
Collapse
|
39
|
Kilani N, Vollenweider P, Waeber G, Marques-Vidal P. Iron metabolism and incidence of metabolic syndrome. Nutr Metab Cardiovasc Dis 2015; 25:1025-1032. [PMID: 26315622 DOI: 10.1016/j.numecd.2015.07.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 07/10/2015] [Accepted: 07/13/2015] [Indexed: 12/26/2022]
Abstract
BACKGROUND AND AIMS Whether iron metabolism affects metabolic syndrome (METS) is debated. We assessed the association between several markers of iron metabolism and incidence of METS. METHODS AND RESULTS Data from 3271 participants (1870 women, 51.3 ± 10.4 years), free of METS at baseline and followed for 5.5 years. The association of serum iron, ferritin and transferrin with incident METS was assessed separately by gender. Incidence of METS was 22.6% in men and 16.5% in women (p < 0.001). After multivariate adjustment, a positive association was found between transferrin and incident METS in men: odds ratio (OR) and 95% confidence interval for the fourth relative to the first quartile 1.55 (1.04-2.31), p for trend = 0.03, while no association was found for iron OR = 0.81 (0.53-1.24), p for trend = 0.33 and ferritin OR = 1.30 (0.88-1.92), p for trend = 0.018. In women, a negative association was found between iron and incident METS: OR for the fourth relative to the first quartile 0.51 (0.33-0.80), p for trend<0.03; the association between transferrin and incident METS was borderline significant: OR = 1.45 (0.97-2.17), p for trend = 0.07 and no association was found for ferritin: OR = 1.11 (0.76-1.63), p for trend = 0.58. CONCLUSION Transferrin, not ferritin, is independently associated with an increased risk of incident METS; the protective effect of iron in women should be further explored.
Collapse
Affiliation(s)
- N Kilani
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland.
| | - P Vollenweider
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland.
| | - G Waeber
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland.
| | - P Marques-Vidal
- Department of Internal Medicine, Internal Medicine, Lausanne University Hospital, Lausanne, Switzerland.
| |
Collapse
|
40
|
Moore WT, Bowser SM, Fausnacht DW, Staley LL, Suh KS, Liu D. Beta Cell Function and the Nutritional State: Dietary Factors that Influence Insulin Secretion. Curr Diab Rep 2015; 15:76. [PMID: 26294335 DOI: 10.1007/s11892-015-0650-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Approximately 366 million people worldwide have been diagnosed with type-2 diabetes (T2D). Chronic insulin resistance, decreased functional β-cell mass, and elevated blood glucose are defining characteristics of T2D. Great advances have been made in understanding the pathogenesis of T2D with respect to the effects of dietary macronutrient composition and energy intake on β-cell physiology and glucose homeostasis. It has been further established that obesity is a leading pathogenic factor for developing insulin resistance. However, insulin resistance may not progress to T2D unless β-cells are unable to secret an adequate amount of insulin to compensate for decreased insulin sensitivity. Therefore, pancreatic β-cell dysfunction plays an important role in the development of overt diabetes. This paper reviews recent research findings on the effects of several micronutrients (zinc, vitamin D, iron, vitamin A), leucine, and the phytochemical, genistein on pancreatic β-cell physiology with emphasis on their effects on insulin secretion, specifically in the context of T2D.
Collapse
Affiliation(s)
- William T Moore
- Department of Human Nutrition, Foods and Exercises, College of Agricultural and Life Sciences, Virginia Tech Corporate Research Center, 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | | | | | | | | | | |
Collapse
|
41
|
Borai A, Livingstone C, Farzal A, Baljoon D, Al Sofyani A, Bahijri S, Kadam I, Hafiz K, Abdelaal M, Ferns G. Changes in metabolic indices in response to whole blood donation in male subjects with normal glucose tolerance. Clin Biochem 2015; 49:51-6. [PMID: 26320016 DOI: 10.1016/j.clinbiochem.2015.08.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2015] [Revised: 08/19/2015] [Accepted: 08/24/2015] [Indexed: 11/30/2022]
Abstract
OBJECTIVES Previous studies have investigated the impact of venesection upon different metabolic indices in patients with various conditions (e.g., type 2 diabetes and iron overload). We aimed to investigate the changes on different metabolic indices including glycemic, iron, lipids and inflammatory markers at different time points after blood donation in male subjects with normal glucose tolerance. DESIGN AND METHODS 42 male subjects were recruited to the study. Glucose tolerance was assessed by oral glucose tolerance test before (visit A) and after the blood donation (1day, visit B; 1week, visit C; 3weeks, visit D; and 3months, visit E). Fasting glucose, HbA1c, insulin, lipids, uric acid, C-reactive protein, iron stores and insulin resistance (HOMA-IR, ISI-gly) indices were measured. A repeated measures ANOVA was used for comparisons of quantitative variables between different visits. RESULTS All subjects had normal glucose tolerance according to WHO criteria. Fasting glucose, insulin and HOMA-IR were significantly higher (~2%, p<0.05; ~21%, p<0.01; and ~11%, p<0.05 respectively) at visit B following donation. At visit D, the mean±SE for HbA1c (5.28±0.06%) was significantly lower with a difference in percentage of ~-3% and p<0.05 compared to visit A (5.44±0.06%). Ferritin decreased significantly at visits B, C, D and E (~-8%, p<0.01, ~-24%, p<0.001, ~-39%, p<0.001 and ~-29%, p<0.01 respectively), when compared to visit A. CONCLUSIONS At different time points after blood donation, glycemic status and iron stores are affected significantly in male blood donors with normal glucose tolerance. The changes were particularly evident three weeks after donation. Hence, the interpretation of these parameters in male blood donors needs to take this into account, and the mechanisms resulting in these effects need to be clarified.
Collapse
Affiliation(s)
- Anwar Borai
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Callum Livingstone
- Faculty of Health & Medical Sciences, University of Surrey, GU2 7XH Guildford, United Kingdom.
| | - Anwar Farzal
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Dalal Baljoon
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Abeer Al Sofyani
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Suhad Bahijri
- Department of Clinical Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Ibrahim Kadam
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Khalid Hafiz
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Mohamed Abdelaal
- King Abdullah International Medical Research Center, King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City, P.O. Box 9515, Jeddah 21423, Saudi Arabia.
| | - Gordon Ferns
- Division of Medical Education, Brighton and Sussex Medical School, Mayfield House, Falmer, Brighton BN1 9PH, United Kingdom.
| |
Collapse
|
42
|
Aregbesola A, Voutilainen S, Virtanen JK, Aregbesola A, Tuomainen TP. Serum hepcidin concentrations and type 2 diabetes. World J Diabetes 2015; 6:978-982. [PMID: 26185605 PMCID: PMC4499531 DOI: 10.4239/wjd.v6.i7.978] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2014] [Revised: 12/22/2014] [Accepted: 03/20/2015] [Indexed: 02/05/2023] Open
Abstract
Hepcidin is a peptide hormone with both paracrine and endocrine functions that help in maintaining body iron stores. Type 2 diabetes (T2D) is one of the sequelae of excess body iron stores; thus, iron regulatory hormone hepcidin may have a direct or at least an indirect role in the aetiopathogenesis of T2D. Both human and animal studies at molecular and genetic levels have attempted to establish a role for hepcidin in the development of T2D, and a few epidemiologic studies have also showed a link between hepcidin and T2D at population level, but the findings are still inconclusive. Recent data have suggested different pathways in which hepcidin could be associated with T2D with much emphasis on its primary or secondary role in insulin resistance. Some of the suggested pathways are via transcription modulator of hepcidin (STAT3); ferroportin 1 expression on the cells involved in iron transport; transmembrane protease 6 enzyme; and pro-inflammatory cytokines, interleukin (IL)-1, IL-6, tumor necrosis factor-α and IL-10. This review briefly reports the existing evidence on the possible links between hepcidin and T2D and concludes that more data are needed to confirm or refute hepcidin’s role in the development of T2D. Examining this role could provide a further evidence base for iron in the aetiopathogenesis of T2D.
Collapse
|
43
|
Kim Y, Keogh J, Clifton P. A review of potential metabolic etiologies of the observed association between red meat consumption and development of type 2 diabetes mellitus. Metabolism 2015; 64:768-79. [PMID: 25838035 DOI: 10.1016/j.metabol.2015.03.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/02/2015] [Accepted: 03/15/2015] [Indexed: 01/12/2023]
Abstract
Epidemiological studies suggest that red and processed meat consumption is related to an increased risk of type 2 diabetes. However, it is not clearly understood which components of red and processed meat contribute to this increased risk. This review examines potential mechanisms addressing the role of saturated fatty acid, sodium, advanced glycation end products (AGEs), nitrates/nitrites, heme iron, trimethylamine N-oxide (TMAO), branched amino acids (BCAAs) and endocrine disruptor chemicals (EDCs) in the development of type 2 diabetes based on data from published clinical trials and animal models. TMAO which is derived from dietary carnitine and choline by the action of bacterial enzymes followed by oxidation in the liver may be a strong candidate molecule mediating the risk of type 2 diabetes. BCAAs may induce insulin resistance via the mammalian target of rapamycin complex 1 (mTORC1) and ribosomal protein S6 kinase β 1 (S6k1)-associated pathways. The increased risk associated with processed meat compared with red meat suggests that there are interactions between the saturated fat, salt, and nitrates in processed meat and iron, AGEs and TMAO. Intervention studies are required to clarify potential mechanisms and explore interactions among components, in order to make firm recommendations on red and processed meat consumption.
Collapse
Affiliation(s)
- Yoona Kim
- School of Pharmacy and Medical Science, University of South Australia, Australia
| | - Jennifer Keogh
- School of Pharmacy and Medical Science, University of South Australia, Australia
| | - Peter Clifton
- School of Pharmacy and Medical Science, University of South Australia, Australia.
| |
Collapse
|
44
|
Guglielmi V, D'Adamo M, Bellia A, Ciotto RT, Federici M, Lauro D, Sbraccia P. Iron status in obesity: An independent association with metabolic parameters and effect of weight loss. Nutr Metab Cardiovasc Dis 2015; 25:541-547. [PMID: 25843660 DOI: 10.1016/j.numecd.2015.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 02/23/2015] [Accepted: 02/23/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND AND AIMS Growing evidence has shown that ferritin concentrations are associated with obesity and insulin resistance, and with nonalcoholic fatty liver disease. However, it is unclear whether ferritin is simply an inflammatory marker, or it may directly contribute to the pathogenesis of obesity-related metabolic alterations. The aim of our study was to investigate the independent associations of ferritin levels with metabolic parameters in overweight/obese subjects before and after hypocaloric diet-induced weight changes. METHODS AND RESULTS A sample study of 48 premenopausal, 39 postmenopausal women and 50 men was retrospectively analyzed. Clinical, bioimpedentiometry and biochemical data from baseline evaluations and after 3, 6 and 12 months of hypocaloric diet were collected. In the whole sample study, the baseline values of ferritin concentrations were positively correlated with body mass index (BMI) (r = 0.21, p < 0.05) and mass body fat (MBF) (r = 0.26, p < 0.05), whereas the serum iron level was negatively correlated with MBF (r = -0.29, p < 0.05). In premenopausal women, BMI-adjusted ferritin concentrations were negatively associated with high-density lipoprotein-cholesterol and positively related with triglycerides and aspartate aminotransferase. Moreover, the quantitative ferritin reduction at 12 months was positively associated with the relative reduction of BMI (r = 0.34, p < 0.05). Finally, the association between changes of alanine aminotransferase and ferritin levels at 12 months from baseline turned out to be independent of respective BMI changes (β = 0.31, p < 0.05). CONCLUSION In obesity, ferritin, putatively entailing increased iron storage, is independently associated with lipid derangements and transaminase levels, and the association with the latter persists after weight changes.
Collapse
Affiliation(s)
- V Guglielmi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - M D'Adamo
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - A Bellia
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - R T Ciotto
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy
| | - M Federici
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - D Lauro
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - P Sbraccia
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy; Obesity Center, University Hospital Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
45
|
Adediran A, Uche E, Akinbami A, Dada A, Wakama T, Damulak D, Ajibola S, Okwegbuna O. Hemoglobin and Ferritin Concentrations in Subjects with Metabolic Syndrome. Nutr Metab Insights 2015; 8:15-9. [PMID: 26056471 PMCID: PMC4445885 DOI: 10.4137/nmi.s23302] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Metabolic syndrome (MetS), a clinical condition characterized by insulin resistance, glucose intolerance, dyslipidemia, hypertension, and obesity, has been linked with raised levels of serum ferritin (Sfr) concentrations. OBJECTIVES This study was carried out to compare hemoglobin (Hb) and Sfr concentrations in patients with MetS, regular donors and first-time donors. MATERIALS AND METHODS A total of 102 subjects who were between 18 and 60 years were enrolled for the study. They were divided into three groups. The first group (n = 20) was made up of 5 males and 15 females, all who met the criteria that define MetS. The second group (n = 52; M = 34, F = 18) were regular donors, while the last group (n = 30; M = 16, F = 14) were first-time donors or those who had not donated before. Following an overnight fast, 20 mL of venous blood was drawn from each subject. About 5 mL of this was put into sodium ethylenediaminetetraacetate (EDTA) specimen bottles for the full blood count parameters with Sysmex KX-21N hematology analyzer (made in Japan). The remaining 15 mL had serum separated for Sfr assay using enzyme-linked immunosorbent assay (ELISA) with a commercial assay kit manufactured by Teco Diagnostics. RESULTS Significant difference was found in the mean Sfr concentration of subjects with MetS (163 ± 136.92 ng/mL) and regular donors (41.46 ± 40.33 ng/mL), P = 0.001. The mean Sfr concentrations of subjects with MetS (163 ± 136.92 ng/mL) were also higher than that of first-time donors (102.46 ± 80.26 ng/mL), but it was not statistically significant, P = 0.053. The Hb concentrations of the three groups were not significantly different. CONCLUSION Sfr concentrations of regular donors were lower than that of subjects with MetS and first-time donors. The difference between regular donors and subjects with MetS was statistically significant. However, there is no significant difference in the Hb concentrations in the three groups. MetS is not associated with anemia or hyperferritinemia.
Collapse
Affiliation(s)
- Adewumi Adediran
- Department of Haematology and Blood Transfusion, College of Medicine, University of Lagos, Lagos, Nigeria
| | - Ebele Uche
- Department of Haematology and Blood Transfusion, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Akinsegun Akinbami
- Department of Haematology and Blood Transfusion, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Akin Dada
- Department of Medicine, Lagos State University Teaching Hospital, Ikeja, Lagos, Nigeria
| | - Tamunomieibi Wakama
- Department of Haematology and Blood Transfusion, National Hospital, Abuja, Nigeria
| | - Dapus Damulak
- Department of Haematology and Blood Transfusion, Jos University Teaching Hospital, Jos, Nigeria
| | - Sarah Ajibola
- Department of Haematology, Ben Carson School of Medicine, Babcock University, Ilisan-Remo, Ogun State, Nigeria
| | - Oluwakemi Okwegbuna
- Department of Haematology and Blood Transfusion, Lagos University Teaching Hospital, Lagos, Nigeria
| |
Collapse
|
46
|
Association of serum ferritin concentrations with prevalence of prediabetes, type 2 diabetes mellitus, and metabolic syndrome in a Chinese population from Sichuan. Int J Diabetes Dev Ctries 2015. [DOI: 10.1007/s13410-015-0308-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
|
47
|
Aigner E, Weiss G, Datz C. Dysregulation of iron and copper homeostasis in nonalcoholic fatty liver. World J Hepatol 2015; 7:177-188. [PMID: 25729473 PMCID: PMC4342600 DOI: 10.4254/wjh.v7.i2.177] [Citation(s) in RCA: 81] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 12/12/2014] [Accepted: 12/31/2014] [Indexed: 02/06/2023] Open
Abstract
Elevated iron stores as indicated by hyperferritinemia with normal or mildly elevated transferrin saturation and mostly mild hepatic iron deposition are a characteristic finding in subjects with non-alcoholic fatty liver disease (NAFLD). Excess iron is observed in approximately one third of NAFLD patients and is commonly referred to as the “dysmetabolic iron overload syndrome”. Clinical evidence suggests that elevated body iron stores aggravate the clinical course of NAFLD with regard to liver-related and extrahepatic disease complications which relates to the fact that excess iron catalyses the formation of toxic hydroxyl-radicals subsequently resulting in cellular damage. Iron removal improves insulin sensitivity, delays the onset of type 2 diabetes mellitus, improves pathologic liver function tests and likewise ameliorates NAFLD histology. Several mechanisms contribute to pathologic iron accumulation in NAFLD. These include impaired iron export from hepatocytes and mesenchymal Kupffer cells as a consequence of imbalances in the concentrations of iron regulatory factors, such as hepcidin, cytokines, copper or other dietary factors. This review summarizes the knowledge about iron homeostasis in NAFLD and the rationale for its therapeutic implications.
Collapse
|
48
|
Hubler MJ, Peterson KR, Hasty AH. Iron homeostasis: a new job for macrophages in adipose tissue? Trends Endocrinol Metab 2015; 26:101-9. [PMID: 25600948 PMCID: PMC4315734 DOI: 10.1016/j.tem.2014.12.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Revised: 12/09/2014] [Accepted: 12/16/2014] [Indexed: 12/29/2022]
Abstract
Elevated serum ferritin and increased cellular iron concentrations are risk factors for diabetes; however, the etiology of this association is unclear. Metabolic tissues such as pancreas, liver, and adipose tissue (AT), as well as the immune cells resident in these tissues, may be involved. Recent studies demonstrate that the polarization status of macrophages has important relevance to their iron-handling capabilities. Furthermore, a subset of macrophages in AT have elevated iron concentrations and a gene expression profile indicative of iron handling, a capacity diminished in obesity. Because iron overload in adipocytes increases systemic insulin resistance, iron handling by AT macrophages may have relevance not only to adipocyte iron stores but also to local and systemic insulin sensitivity.
Collapse
Affiliation(s)
- Merla J Hubler
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Kristin R Peterson
- Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN 37232, USA; Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN 37212, USA.
| |
Collapse
|
49
|
Moreno-Navarrete JM, Ortega F, Moreno M, Ricart W, Fernández-Real JM. Fine-tuned iron availability is essential to achieve optimal adipocyte differentiation and mitochondrial biogenesis. Diabetologia 2014; 57:1957-67. [PMID: 24973963 DOI: 10.1007/s00125-014-3298-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 05/27/2014] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Adipose tissue from obese and insulin-resistant individuals showed altered expression of several iron-related genes in a recent study, suggesting that iron might have an important role in adipogenesis. To investigate this possible role, we aimed to characterise the effects of iron on adipocyte differentiation. METHODS Intracellular iron deficiency was achieved using two independent approaches: deferoxamine administration (20 and 100 μmol/l) and transferrin knockdown (TF KD). The effects of added FeSO4, holo-transferrin and palmitate were studied during human and 3T3-L1 adipocyte differentiation. Finally, the relationship between iron-related and mitochondrial-related genes was investigated in human adipose tissue. RESULTS Most adipose tissue iron-related genes were predominantly expressed in adipocytes compared with stromal vascular cells. Of note, transferrin gene and protein expression increased significantly during adipocyte differentiation. Both deferoxamine and TF KD severely blunted adipocyte differentiation in parallel with increased inflammatory mRNAs. These effects were reversed in a dose-dependent manner after iron supplementation. Palmitate administration also led to a state of functional intracellular iron deficiency, with decreased Tf gene expression and iron uptake during adipocyte differentiation, which was reversed with transferrin co-treatment. On the other hand, iron in excess impaired differentiation, but this antiadipogenic effect was less pronounced than under iron chelation. Of interest, expression of several genes involved in mitochondrial biogenesis occurred in parallel with expression of iron-related genes both during adipogenesis and in human adipose tissue. CONCLUSIONS/INTERPRETATION Precise and fine-tuned iron availability is essential to achieve optimal adipocyte differentiation, possibly modulating adipocyte mitochondrial biogenesis.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Hospital of Girona 'Dr Josep Trueta', Girona, Spain
| | | | | | | | | |
Collapse
|
50
|
Espinoza A, Morales S, Arredondo M. Effects of acute dietary iron overload in pigs (Sus scrofa) with induced type 2 diabetes mellitus. Biol Trace Elem Res 2014; 158:342-52. [PMID: 24699828 DOI: 10.1007/s12011-014-9944-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Accepted: 03/12/2014] [Indexed: 01/29/2023]
Abstract
Epidemiological studies have reported an association between high iron (Fe) levels and elevated risk of developing type 2 diabetes mellitus (T2D). It is believed that the formation of Fe-catalyzed hydroxyl radicals may contribute to the development of diabetes. Our goal was to determine the effect of a diet with a high Fe content on type 2 diabetic pigs. Four groups of piglets were studied: (1) control group, basal diet; (2) Fe group, basal diet with 3,000 ppm ferrous sulfate; (3) diabetic group (streptozotocin-induced type 2 diabetes) with basal diet; (4) diabetic/Fe group, diabetic animals/3,000 ppm ferrous sulfate. For 2 months, biochemical and hematological parameters were evaluated. Tissue samples of liver and duodenum were obtained to determine mRNA relative abundance of DMT1, ferroportin (Fpn), ferritin (Fn), hepcidin (Hpc), and transferrin receptor by qRT-PCR. Fe group presented increased levels of hematological (erythrocytes, hematocrit, and hemoglobin) and iron parameters. Diabetic/Fe group showed similar behavior as Fe group but in lesser extent. The relative abundance of different genes in the four study groups yielded a different expression pattern. DMT1 showed a lower expression in the two iron groups compared with control and diabetic animals, and Hpc showed an increased on its expression in Fe and diabetic/Fe groups. Diabetic/Fe group presents greater expression of Fn and Fpn. These results suggest that there is an interaction between Fe nutrition, inflammation, and oxidative stress in the diabetes development.
Collapse
MESH Headings
- Analysis of Variance
- Animals
- Blood Glucose/metabolism
- C-Reactive Protein/metabolism
- Cation Transport Proteins/genetics
- Cholesterol/blood
- Cholesterol, LDL/blood
- Diabetes Mellitus, Experimental/blood
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/metabolism
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/metabolism
- Diet
- Duodenum/drug effects
- Duodenum/metabolism
- Ferritins/genetics
- Ferrous Compounds/administration & dosage
- Ferrous Compounds/metabolism
- Gene Expression/drug effects
- Hepcidins/genetics
- Iron Overload/metabolism
- Iron Overload/physiopathology
- Iron, Dietary/administration & dosage
- Iron, Dietary/metabolism
- Liver/drug effects
- Liver/metabolism
- Male
- Receptors, Transferrin/genetics
- Reverse Transcriptase Polymerase Chain Reaction
- Sus scrofa
- Time Factors
- Triglycerides/blood
Collapse
Affiliation(s)
- A Espinoza
- Micronutrient laboratory, Nutrition Institute and Food Technology (INTA), University of Chile, El Líbano 5524, Macul, Santiago, Chile
| | | | | |
Collapse
|