1
|
Liu X, Wang N, Gu S, He Z. Changes of RNA m 6A/m 5C Modification Regulatory Molecules in Ferroptosis of T2DM Rat Pancreas. Cell Biochem Biophys 2024; 82:1279-1289. [PMID: 38709441 DOI: 10.1007/s12013-024-01282-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2024] [Indexed: 05/07/2024]
Abstract
N6-methyladenine (m6A) and 5-methylcytosine (m5C) are two common forms of RNA methylation that play an important role in the epigenetics of type 2 diabetes mellitus (T2DM). One type of cell death, ferroptosis, has been implicated in islet β-cell damage in T2DM. Notably, RNA methylation, an upstream regulatory mechanism of mRNAs, can regulate the expression of ferroptosis signaling molecules, thereby affecting cell proliferation and death. Here, we found that the ferroptosis signaling pathway was activated in pancreas of T2DM rats, followed by significant changes in m6A/m5C modification regulatory molecules. These detection data together with the prediction results that m6A and m5C exist in the mRNAs of ferroptosis molecules, we speculate that m6A and m5C are probably involved in pancreatic cell damage by modifying of ferroptosis signaling molecules. In short, our findings provide a new research idea for future studies on the molecular mechanisms of pancreatic cell damage and point to a new direction for exploring the mechanisms of ferroptosis from the perspective of RNA methylation modification.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China
| | - Nan Wang
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China
| | - Shiyan Gu
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China.
| | - Zuoshun He
- Institute of Preventive Medicine, School of Public Health, Dali University, Dali, Yunnan, China.
| |
Collapse
|
2
|
Yin X, Ni G, Zhang X, Fu S, Li H, Gao Z. Tyrosine nitration of glucagon impairs its function: Extending the role of heme in T2D pathogenesis. J Inorg Biochem 2024; 255:112519. [PMID: 38507994 DOI: 10.1016/j.jinorgbio.2024.112519] [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: 11/18/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
New studies raise the possibility that the higher glucagon (GCG) level present in type 2 diabetes (T2D) is a compensatory mechanism to enhance β-cell function, rather than induce dysregulated glucose homeostasis, due to an important role for GCG that acts directly within the pancreas on insulin secretion by intra-islet GCG signaling. However, in states of poorly controlled T2D, pancreatic α cell mass increases (overproduced GCG) in response to insufficient insulin secretion, indicating decreased local GCG activity. The reason for this decrease is not clear. Recent evidence has uncovered a new role of heme in cellular signal transduction, and its mechanism involves reversible binding of heme to proteins. Considering that protein tyrosine nitration in diabetic islets increases and glucose-stimulated insulin secretion (GSIS) decreases, we speculated that heme modulates GSIS by transient interaction with GCG and catalyzing its tyrosine nitration, and the tyrosine nitration may impair GCG activity, leading to loss of intra-islet GCG signaling and markedly impaired insulin secretion. Data presented here elucidate a novel role for heme in disrupting local GCG signaling in diabetes. Heme bound to GCG and induced GCG tyrosine nitration. Two tyrosine residues in GCG were both sensitive to the nitrating species. Further, GCG was also demonstrated to be a preferred target peptide for tyrosine nitration by co-incubation with BSA. Tyrosine nitration impaired GCG stimulated cAMP-dependent signaling in islet β cells and decreased insulin release. Our results provided a new role of heme for impaired GSIS in the pathological process of diabetes.
Collapse
Affiliation(s)
- Xiaoying Yin
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Guoqi Ni
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Xuan Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Shitao Fu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China.
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Wuhan 430074, PR China; School of Chemistry and Chemical Engineering, Huazhong University of Science & Technology, Wuhan 430074, PR China.
| |
Collapse
|
3
|
Wang M, Liu Y, Gui H, Ma G, Li B, Zhang Z, Yu G, Wu A, Xu X, Zhang D. ED-71 ameliorates bone regeneration in type 2 diabetes by reducing ferroptosis in osteoblasts via the HIF1α pathway. Eur J Pharmacol 2024; 969:176303. [PMID: 38211715 DOI: 10.1016/j.ejphar.2023.176303] [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: 09/03/2023] [Revised: 12/15/2023] [Accepted: 12/21/2023] [Indexed: 01/13/2024]
Abstract
Eldecalcitol (ED-71), a novel active form of vitamin D, shows potential in treating osteoporosis. However, its underlying mechanisms of action remain to be determined. This study aimed to investigate the effect of ED-71 on bone regeneration and to illustrate its mode of action. The in-vitro model was developed using rat primary osteoblasts cultured under high-glucose conditions, and these cells were treated with ED-71. Additionally, an in vivo model of cranial bone defects was established in type 2 diabetic rats, and ED-71 was administered by gavage. The results demonstrated that ED-71 prevented osteoblast cell death, enhanced rat primary osteoblasts' osteogenic capacity, and attenuated the overexpression of hypoxia-inducible factor 1α (HIF1α) induced by high glucose levels. Furthermore, ED-71 increased glutathione peroxidase 4 (GPX4) levels and inhibited ferroptosis in response to hyperglycemic stimulation. Notably, interference with the HIF1α activator and ferroptosis activator Erastin significantly reduced the therapeutic effects of edetate osteolysis. These findings were further tested in vivo experiments. These results suggest that ED-71 activates the HIF1α pathway in vivo and in vitro, effectively relieving the ferroptosis induced by high glucose. Significantly, ED-71 may improve osteogenic disorders caused by diabetes.
Collapse
Affiliation(s)
- Maoshan Wang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Yingxue Liu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Houda Gui
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Gaoqiang Ma
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Binyang Li
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Zhanwei Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Gyeonghwi Yu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Ailin Wu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Xin Xu
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| | - Dongjiao Zhang
- Department of Implantology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Laboratory for Dental Materials and Oral Tissue Regeneration, No. 44-1 Wenhua Road West, Jinan, 250012, China.
| |
Collapse
|
4
|
Li W, Lv BM, Quan Y, Zhu Q, Zhang HY. Associations between Serum Mineral Nutrients, Gut Microbiota, and Risk of Neurological, Psychiatric, and Metabolic Diseases: A Comprehensive Mendelian Randomization Study. Nutrients 2024; 16:244. [PMID: 38257137 PMCID: PMC10818407 DOI: 10.3390/nu16020244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
Recent observational studies have reported associations between serum mineral nutrient levels, gut microbiota composition, and neurological, psychiatric, and metabolic diseases. However, the causal effects of mineral nutrients on gut microbiota and their causal associations with diseases remain unclear and require further investigation. This study aimed to identify the associations between serum mineral nutrients, gut microbiota, and risk of neurological, psychiatric, and metabolic diseases using Mendelian randomization (MR). We conducted an MR study using the large-scale genome-wide association study (GWAS) summary statistics of 5 serum mineral nutrients, 196 gut microbes at the phylum, order, family, and genus levels, and a variety of common neurological, psychiatric, and metabolic diseases. Initially, the independent causal associations of mineral nutrients and gut microbiota with diseases were examined by MR. Subsequently, the causal effect of mineral nutrients on gut microbiota was estimated to investigate whether specific gut microbes mediated the association between mineral nutrients and diseases. Finally, we performed sensitivity analyses to assess the robustness of the study results. After correcting for multiple testing, we identified a total of 33 causal relationships among mineral nutrients, gut microbiota, and diseases. Specifically, we found 4 causal relationships between 3 mineral nutrition traits and 3 disease traits, 15 causal associations between 14 gut microbiota traits and 6 disease traits, and 14 causal associations involving 4 mineral nutrition traits and 15 gut microbiota traits. Meanwhile, 118 suggestive associations were identified. The current study reveals multiple causal associations between serum mineral nutrients, gut microbiota, risk of neurological, psychiatric, and metabolic diseases, and potentially provides valuable insights for subsequent nutritional therapies.
Collapse
Affiliation(s)
- Wang Li
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
| | - Bo-Min Lv
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
- Human Phenome Institute, Fudan University, Shanghai 200438, China
| | - Yuan Quan
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
| | - Qiang Zhu
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
- Key Laboratory of Smart Farming for Agricultural Animals, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China; (W.L.); (B.-M.L.); (Y.Q.); (H.-Y.Z.)
| |
Collapse
|
5
|
Liu Y, Fillebeen C, Forest A, Botta A, Varin TV, Marette A, Burelle Y, Des Rosiers C, Pantopoulos K, Sweeney G. Perturbations in lipid metabolism and gut microbiota composition precede cardiac dysfunction in a mouse model of thalassemia. FASEB J 2023; 37:e23257. [PMID: 37902616 DOI: 10.1096/fj.202301043r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/05/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023]
Abstract
Cardiomyopathy is a major complication of thalassemia, yet the precise underlying molecular mechanisms remain unclear. We examined whether altered lipid metabolism is an early driving factor in the development of cardiomyopathy using the Th3/+ mouse model of thalassemia. At age 20 weeks, male and female Th3/+ mice manifested anemia and iron overload; however, only males displayed metabolic defects and altered cardiac function. Untargeted lipidomics indicated that the circulating levels of 35 lipid species were significantly altered in Th3/+ mice compared to wild-type controls: triglycerides (TGs) with saturated fatty acids (FAs; TG42:0 and TG44:0) were elevated, while TGs with unsaturated FAs (TG(18:2_20:5_18:2 and TG54:8)) were reduced. Similarly, phosphatidylcholines (PCs) with long chain FAs (palmitic (16:0) or oleic (18:1)) were increased, while PCs with polyunsaturated FAs decreased. Circulating PC(16:0_14:0), GlcCer(d18:1/24:0) correlated significantly with iron overload and cardiac hypertrophy. 16S rRNA gene profiling revealed alterations in the intestinal microbiota of Th3/+ mice. Differentially abundant bacterial genera correlated with PC(39:6), PC(18:1_22:6), GlcCer(d18:1/24:1) and CE(14:0). These results provide new knowledge on perturbations in lipid metabolism and the gut microbiota of Th3/+ mice and identify specific factors which may represent early biomarkers or therapeutic targets to prevent development of cardiomyopathy in β-thalassemia.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Carine Fillebeen
- Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Anik Forest
- Montreal Heart Institute Research Center, Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Amy Botta
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Thibault V Varin
- Department of Medicine, Heart and lung Institute, University of Laval, Quebec City, Quebec, Canada
| | - André Marette
- Department of Medicine, Heart and lung Institute, University of Laval, Quebec City, Quebec, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Christine Des Rosiers
- Montreal Heart Institute Research Center, Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Qiu F, Wu L, Yang G, Zhang C, Liu X, Sun X, Chen X, Wang N. The role of iron metabolism in chronic diseases related to obesity. Mol Med 2022; 28:130. [PMID: 36335331 PMCID: PMC9636637 DOI: 10.1186/s10020-022-00558-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/14/2022] [Indexed: 11/08/2022] Open
Abstract
Obesity is one of the major public health problems threatening the world, as well as a potential risk factor for chronic metabolic diseases. There is growing evidence that iron metabolism is altered in obese people, however, the highly refined regulation of iron metabolism in obesity and obesity-related complications is still being investigated. Iron accumulation can affect the body’s sensitivity to insulin, Type 2 diabetes, liver disease and cardiovascular disease. This review summarized the changes and potential mechanisms of iron metabolism in several chronic diseases related to obesity, providing new clues for future research.
Collapse
|
7
|
Li N, Liao Y, Huang H, Fu S. Co-regulation of hepatic steatosis by ferritinophagy and unsaturated fatty acid supply. Hepatol Commun 2022; 6:2640-2653. [PMID: 35861547 PMCID: PMC9512465 DOI: 10.1002/hep4.2040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/26/2022] [Accepted: 06/28/2022] [Indexed: 11/06/2022] Open
Abstract
Both iron overload and iron deficiency have been reported in obesity and metabolic syndromes. Due to the presence of multiple intracellular iron pools and the dynamic nature of iron mobilization and use, the actual status and contribution of free and metabolically active iron toward metabolic syndrome remain to be established. The discovery of nuclear receptor coactivator 4 (NCOA4) as a ferritinophagy receptor provides an opening to address the connection between iron and metabolic diseases. This study aims to specifically dissect the role of hepatic ferritinophagy in lipid metabolism and hepatic steatosis. We conducted a series of Ncoa4 gain- and loss-of-function experiments to examine how ferritinophagy affects lipid metabolism through phenotypic and lipidomic analyses both in vitro and in vivo. We show that ferritinophagy is required to release iron from ferritin cages for biological use, and is induced by lipid loading in vitro and during the development of obesity in vivo. Ncoa4 knockdown impairs mitochondrial morphology and reduces palmitate-induced lipid droplet formation in cultured cells and the development of hepatic steatosis in obese mice models. Importantly, the effect of Ncoa4 deficiency on mitochondrial morphology and lipid accumulation is specifically linked to lipidomic reductions in unsaturated fatty acid content in triglycerides and cardiolipins, and an external supply of unsaturated fatty acids reverses these phenotypes. Conclusion: This study shows that ferritinophagy-derived iron supports fatty acid desaturation and the synthesis of unsaturated fatty acid-rich lipids to reduce lipotoxicity. However, the continuous activation of ferritinophagy contributes to the development of hepatic steatosis and liver damage in obesity.
Collapse
Affiliation(s)
- Ning Li
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Yilie Liao
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Haipeng Huang
- School of Life SciencesTsinghua UniversityBeijingChina
| | - Suneng Fu
- Department of Basic ResearchGuangzhou LaboratoryGuangdongChina
| |
Collapse
|
8
|
Wang M, Wang Z, Lee Y, Lai HTM, de Oliveira Otto MC, Lemaitre RN, Fretts A, Sotoodehnia N, Budoff M, DiDonato JA, McKnight B, Tang WHW, Psaty BM, Siscovick DS, Hazen SL, Mozaffarian D. Dietary Meat, Trimethylamine N-Oxide-Related Metabolites, and Incident Cardiovascular Disease Among Older Adults: The Cardiovascular Health Study. Arterioscler Thromb Vasc Biol 2022; 42:e273-e288. [PMID: 35912635 PMCID: PMC9420768 DOI: 10.1161/atvbaha.121.316533] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 05/24/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Effects of animal source foods (ASF) on atherosclerotic cardiovascular disease (ASCVD) and underlying mechanisms remain controversial. We investigated prospective associations of different ASF with incident ASCVD and potential mediation by gut microbiota-generated trimethylamine N-oxide, its L-carnitine-derived intermediates γ-butyrobetaine and crotonobetaine, and traditional ASCVD risk pathways. METHODS Among 3931 participants from a community-based US cohort aged 65+ years, ASF intakes and trimethylamine N-oxide-related metabolites were measured serially over time. Incident ASCVD (myocardial infarction, fatal coronary heart disease, stroke, other atherosclerotic death) was adjudicated over 12.5 years median follow-up. Cox proportional hazards models with time-varying exposures and covariates examined ASF-ASCVD associations; and additive hazard models, mediation proportions by different risk pathways. RESULTS After multivariable-adjustment, higher intakes of unprocessed red meat, total meat, and total ASF associated with higher ASCVD risk, with hazard ratios (95% CI) per interquintile range of 1.15 (1.01-1.30), 1.22 (1.07-1.39), and 1.18 (1.03-1.34), respectively. Trimethylamine N-oxide-related metabolites together significantly mediated these associations, with mediation proportions (95% CI) of 10.6% (1.0-114.5), 7.8% (1.0-32.7), and 9.2% (2.2-44.5), respectively. Processed meat intake associated with a nonsignificant trend toward higher ASCVD (1.11 [0.98-1.25]); intakes of fish, poultry, and eggs were not significantly associated. Among other risk pathways, blood glucose, insulin, and C-reactive protein, but not blood pressure or blood cholesterol, each significantly mediated the total meat-ASCVD association. CONCLUSIONS In this large, community-based cohort, higher meat intake associated with incident ASCVD, partly mediated by microbiota-derived metabolites of L-carnitine, abundant in red meat. These novel findings support biochemical links between dietary meat, gut microbiome pathways, and ASCVD.
Collapse
Affiliation(s)
- Meng Wang
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Zeneng Wang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Yujin Lee
- Department of Food and Nutrition, Myongji University, Yongin, South Korea 17055
| | - Heidi TM Lai
- Imperial College London, Department of Primary Care and Public Health, London, SW7 2AZ, UK
| | - Marcia C. de Oliveira Otto
- Division of Epidemiology, Human Genetics and Environmental Sciences, The University of Texas Health Science Center at Houston (UTHealth) School of Public Health, Houston, TX
| | - Rozenn N. Lemaitre
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Amanda Fretts
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Matthew Budoff
- Los Angeles BioMedical Research Institute, Harbor UCLA Medical Center, CA
| | - Joseph A. DiDonato
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Barbara McKnight
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Biostatistics, University of Washington, Seattle, WA
| | - W. H. Wilson Tang
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
| | | | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular Medicine, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH
| | - Dariush Mozaffarian
- Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| |
Collapse
|
9
|
Ye H, Wang R, Wei J, Wang Y, Zhang X, Wang L. Bioinformatics Analysis Identifies Potential Ferroptosis Key Gene in Type 2 Diabetic Islet Dysfunction. Front Endocrinol (Lausanne) 2022; 13:904312. [PMID: 35898457 PMCID: PMC9309693 DOI: 10.3389/fendo.2022.904312] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/10/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND Islet β cells dysfunction (IBCD) is a cortical component in pathogenesis of type 2 diabetic mellitus (T2DM). However, the relationship of ferroptosis and IBCD remains unknown. This study was aimed to screen potential ferroptosis key genes to reveal latent physiological and pathological process of IBCD in T2DM. METHODS Firstly, T2DM key genes were screened by combining with differentially expressed genes (DEGs) analysis and WGCNA. Then, ferroptosis-related genes (FRGs) in IBCD of T2DM were identified by taking the intersection between T2DM key genes and FRGs. Finally, T2DM-FRGs were validated in another T2DM dataset as well as islet single-cell RNA sequencing dataset and the miRNA regulated T2DM-FRG was predicted by using four miRNA databases. RESULTS 89 T2DM key genes were identified between DEGs and WGCNA. Then, 3 T2DM-FRGs were screened by taking the intersection of T2DM key genes and FRGs, namely ITGA6, MGST1 and ENO2. At last, MGST1 were validated as the T2DM-FRG in another T2DM islet issues dataset and islet single-cell RNA sequencing dataset. CONCLUSION MGST1 may be the potential ferroptosis key gene of IBCD in T2DM.
Collapse
Affiliation(s)
- Haowen Ye
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ruxin Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Jinjing Wei
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ying Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiaofang Zhang
- Clinical Experimental Center, First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Xiaofang Zhang, ; Lihong Wang,
| | - Lihong Wang
- Department of Endocrinology and Metabolism, First Affiliated Hospital of Jinan University, Guangzhou, China
- *Correspondence: Xiaofang Zhang, ; Lihong Wang,
| |
Collapse
|
10
|
Marku A, Galli A, Marciani P, Dule N, Perego C, Castagna M. Iron Metabolism in Pancreatic Beta-Cell Function and Dysfunction. Cells 2021; 10:2841. [PMID: 34831062 PMCID: PMC8616520 DOI: 10.3390/cells10112841] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/26/2022] Open
Abstract
Iron is an essential element involved in a variety of physiological functions. In the pancreatic beta-cells, being part of Fe-S cluster proteins, it is necessary for the correct insulin synthesis and processing. In the mitochondria, as a component of the respiratory chain, it allows the production of ATP and reactive oxygen species (ROS) that trigger beta-cell depolarization and potentiate the calcium-dependent insulin release. Iron cellular content must be finely tuned to ensure the normal supply but also to prevent overloading. Indeed, due to the high reactivity with oxygen and the formation of free radicals, iron excess may cause oxidative damage of cells that are extremely vulnerable to this condition because the normal elevated ROS production and the paucity in antioxidant enzyme activities. The aim of the present review is to provide insights into the mechanisms responsible for iron homeostasis in beta-cells, describing how alteration of these processes has been related to beta-cell damage and failure. Defects in iron-storing or -chaperoning proteins have been detected in diabetic conditions; therefore, the control of iron metabolism in these cells deserves further investigation as a promising target for the development of new disease treatments.
Collapse
Affiliation(s)
| | | | | | | | - Carla Perego
- Department of Excellence Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Trentacoste, 22134 Milano, Italy; (A.M.); (A.G.); (P.M.); (N.D.)
| | - Michela Castagna
- Department of Excellence Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Trentacoste, 22134 Milano, Italy; (A.M.); (A.G.); (P.M.); (N.D.)
| |
Collapse
|
11
|
Wang X, Fang X, Zheng W, Zhou J, Song Z, Xu M, Min J, Wang F. Genetic Support of A Causal Relationship Between Iron Status and Type 2 Diabetes: A Mendelian Randomization Study. J Clin Endocrinol Metab 2021; 106:e4641-e4651. [PMID: 34147035 PMCID: PMC8530720 DOI: 10.1210/clinem/dgab454] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Indexed: 12/15/2022]
Abstract
CONTEXT Iron overload is a known risk factor for type 2 diabetes (T2D); however, iron overload and iron deficiency have both been associated with metabolic disorders in observational studies. OBJECTIVE Using mendelian randomization (MR), we assessed how genetically predicted systemic iron status affected T2D risk. METHODS A 2-sample MR analysis was used to obtain a causal estimate. We selected genetic variants strongly associated (P < 5 × 10-8) with 4 biomarkers of systemic iron status from a study involving 48 972 individuals performed by the Genetics of Iron Status consortium and applied these biomarkers to the T2D case-control study (74 124 cases and 824 006 controls) performed by the Diabetes Genetics Replication and Meta-analysis consortium. The simple median, weighted median, MR-Egger, MR analysis using mixture-model, weighted allele scores, and MR based on a Bayesian model averaging approaches were used for the sensitivity analysis. RESULTS Genetically instrumented serum iron (odds ratio [OR]: 1.07; 95% CI, 1.02-1.12), ferritin (OR: 1.19; 95% CI, 1.08-1.32), and transferrin saturation (OR: 1.06; 95% CI, 1.02-1.09) were positively associated with T2D. In contrast, genetically instrumented transferrin, a marker of reduced iron status, was inversely associated with T2D (OR: 0.91; 95% CI, 0.87-0.96). CONCLUSION Genetic evidence supports a causal link between increased systemic iron status and increased T2D risk. Further studies involving various ethnic backgrounds based on individual-level data and studies regarding the underlying mechanism are warranted for reducing the risk of T2D.
Collapse
Affiliation(s)
- Xinhui Wang
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xuexian Fang
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Wanru Zheng
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jiahui Zhou
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Zijun Song
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai 200030, China
| | - Junxia Min
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fudi Wang
- The Fourth Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| |
Collapse
|
12
|
Divalent Metal Transporter 1 Knock-Down Modulates IL-1β Mediated Pancreatic Beta-Cell Pro-Apoptotic Signaling Pathways through the Autophagic Machinery. Int J Mol Sci 2021; 22:ijms22158013. [PMID: 34360779 PMCID: PMC8348373 DOI: 10.3390/ijms22158013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 12/21/2022] Open
Abstract
Pro-inflammatory cytokines promote cellular iron-import through enhanced divalent metal transporter-1 (DMT1) expression in pancreatic β-cells, consequently cell death. Inhibition of β-cell iron-import by DMT1 silencing protects against apoptosis in animal models of diabetes. However, how alterations of signaling networks contribute to the protective action of DMT1 knock-down is unknown. Here, we performed phosphoproteomics using our sequential enrichment strategy of mRNA, protein, and phosphopeptides, which enabled us to explore the concurrent molecular events in the same set of wildtype and DMT1-silenced β-cells during IL-1β exposure. Our findings reveal new phosphosites in the IL-1β-induced proteins that are clearly reverted by DMT1 silencing towards their steady-state levels. We validated the levels of five novel phosphosites of the potential protective proteins using parallel reaction monitoring. We also confirmed the inactivation of autophagic flux that may be relevant for cell survival induced by DMT1 silencing during IL-1β exposure. Additionally, the potential protective proteins induced by DMT1 silencing were related to insulin secretion that may lead to improving β-cell functions upon exposure to IL-1β. This global profiling has shed light on the signal transduction pathways driving the protection against inflammation-induced cell death in β-cells after DMT1 silencing.
Collapse
|
13
|
Wang D, Mao Y, Wang T, Xiong T, Yang X. Letter by Wang et al Regarding Article, "Treatment With Treprostinil and Metformin Normalizes Hyperglycemia and Improves Cardiac Function in Pulmonary Hypertension Associated With Heart Failure With Preserved Ejection Fraction". Arterioscler Thromb Vasc Biol 2020; 40:e260-e261. [PMID: 32845776 DOI: 10.1161/atvbaha.120.314939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Daxin Wang
- From the Clinical Medical College, Yangzhou University, Jiangsu, China (D.W., T.W., X.Y.)
| | - Yongqing Mao
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.M., T.X., X.Y.)
| | - Ti Wang
- From the Clinical Medical College, Yangzhou University, Jiangsu, China (D.W., T.W., X.Y.)
| | - Ting Xiong
- Department of Cardiology, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China (Y.M., T.X., X.Y.)
| | - Xinquan Yang
- From the Clinical Medical College, Yangzhou University, Jiangsu, China (D.W., T.W., X.Y.)
| |
Collapse
|
14
|
Abstract
The effect of dietary fats on cardiometabolic diseases, including cardiovascular diseases and type 2 diabetes mellitus, has generated tremendous interest. Many earlier investigations focused on total fat and conventional fat classes (such as saturated and unsaturated fats) and their influence on a limited number of risk factors. However, dietary fats comprise heterogeneous molecules with diverse structures, and growing research in the past two decades supports correspondingly complex health effects of individual dietary fats. Moreover, health effects of dietary fats might be modified by additional factors, such as accompanying nutrients and food-processing methods, emphasizing the importance of the food sources. Accordingly, the rapidly increasing scientific findings on dietary fats and cardiometabolic diseases have generated debate among scientists, caused confusion for the general public and present challenges for translation into dietary advice and policies. This Review summarizes the evidence on the effects of different dietary fats and their food sources on cell function and on risk factors and clinical events of cardiometabolic diseases. The aim is not to provide an exhaustive review but rather to focus on the most important evidence from randomized controlled trials and prospective cohort studies and to highlight current areas of controversy and the most relevant future research directions for understanding how to improve the prevention and management of cardiometabolic diseases through optimization of dietary fat intake.
Collapse
|
15
|
Kellon EM, Gustafson KM. Possible dysmetabolic hyperferritinemia in hyperinsulinemic horses. Open Vet J 2020; 9:287-293. [PMID: 32042647 PMCID: PMC6971364 DOI: 10.4314/ovj.v9i4.2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/15/2019] [Indexed: 01/02/2023] Open
Abstract
Background Hyperinsulinemia associated with equine metabolic syndrome and pituitary pars intermedia dysfunction is a risk factor for laminitis. Research in other species has shown elevated body iron levels as both a predictor and consequence of insulin resistance. In humans, this is known as dysmetabolic hyperferritinemia. Aim To explore the relationship between equine hyperinsulinemia and body iron levels. Methods We reviewed case histories and laboratory results from an open access database maintained by the Equine Cushing's and Insulin Resistance Group Inc. (ECIR). We identified 33 horses with confirmed hyperinsulinemia and laboratory results for serum iron, total iron binding capacity, and ferritin. Pearson correlation was used to test the relationship between insulin and iron indices. Additionally, we performed a secondary analysis of a previously reported controlled trial that was originally designed to test the correlation between iron status and the insulin response in horses. Here, we used a t-test to compare the mean values of insulin and ferritin between horses we categorized as normal or hyperinsulinemic based on their response to an oral challenge. Results Serum ferritin exceeded published reference range in 100% of the horses identified from the ECIR database. There were no statistically significant associations between insulin indices (RISQI, log insulin) and iron indices (log serum iron, log TSI%, log ferritin). There were trends for a negative association between RISQI and log iron [r(31) = -0.33, p = 0.058] and a positive association between age and ferritin [r(30) = 0.34, p = 0.054]. From the secondary data analysis of published data, we found significantly elevated ferritin (p = 0.05) in horses considered hyperinsulinemic by dynamic insulin testing compared to horses with a normal response. Conclusion These results suggest the potential for iron overload in hyperinsulinemic horses, a feature documented in other species and should stimulate further study into the relationship between insulin and iron dysregulation in the horse.
Collapse
Affiliation(s)
- Eleanor M. Kellon
- Equine Cushing’s and Insulin Resistance Group, Inc, 2307 Rural Road, Tempe, AZ 85282, USA
| | - Kathleen M. Gustafson
- Equine Cushing’s and Insulin Resistance Group, Inc, 2307 Rural Road, Tempe, AZ 85282, USA
| |
Collapse
|
16
|
Abdellatif AM, Jensen Smith H, Harms RZ, Sarvetnick NE. Human Islet Response to Selected Type 1 Diabetes-Associated Bacteria: A Transcriptome-Based Study. Front Immunol 2019; 10:2623. [PMID: 31781116 PMCID: PMC6857727 DOI: 10.3389/fimmu.2019.02623] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Accepted: 10/22/2019] [Indexed: 12/12/2022] Open
Abstract
Type 1 diabetes (T1D) is a chronic autoimmune disease that results from destruction of pancreatic β-cells. T1D subjects were recently shown to harbor distinct intestinal microbiome profiles. Based on these findings, the role of gut bacteria in T1D is being intensively investigated. The mechanism connecting intestinal microbial homeostasis with the development of T1D is unknown. Specific gut bacteria such as Bacteroides dorei (BD) and Ruminococcus gnavus (RG) show markedly increased abundance prior to the development of autoimmunity. One hypothesis is that these bacteria might traverse the damaged gut barrier, and their constituents elicit a response from human islets that causes metabolic abnormalities and inflammation. We have tested this hypothesis by exposing human islets to BD and RG in vitro, after which RNA-Seq analysis was performed. The bacteria altered expression of many islet genes. The commonly upregulated genes by these bacteria were cytokines, chemokines and enzymes, suggesting a significant effect of gut bacteria on islet antimicrobial and biosynthetic pathways. Additionally, each bacteria displayed a unique set of differentially expressed genes (DEGs). Ingenuity pathway analysis of DEGs revealed that top activated pathways and diseases included TREM1 signaling and inflammatory response, illustrating the ability of bacteria to induce islet inflammation. The increased levels of selected factors were confirmed using immunoblotting and ELISA methods. Our data demonstrate that islets produce a complex anti-bacterial response. The response includes both symbiotic and pathogenic aspects. Both oxidative damage and leukocyte recruitment factors were prominent, which could induce beta cell damage and subsequent autoimmunity.
Collapse
Affiliation(s)
- Ahmed M. Abdellatif
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Heather Jensen Smith
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, United States
- Eppley Institute for Research in Cancer, University of Nebraska Medical Center, Omaha, NE, United States
| | - Robert Z. Harms
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| | - Nora E. Sarvetnick
- Department of Surgery-Transplant, University of Nebraska Medical Center, Omaha, NE, United States
- Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
17
|
Jiang L, Wang K, Lo K, Zhong Y, Yang A, Fang X, Akezhuoli H, Song Z, Chen L, An P, Xu M, Min J, Wang F. Sex-Specific Association of Circulating Ferritin Level and Risk of Type 2 Diabetes: A Dose-Response Meta-Analysis of Prospective Studies. J Clin Endocrinol Metab 2019; 104:4539-4551. [PMID: 31074789 DOI: 10.1210/jc.2019-00495] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/06/2019] [Indexed: 12/15/2022]
Abstract
CONTEXT Although the role of iron in the development of type 2 diabetes (T2D) has long been a concern, prospective studies directly linking body iron stores to T2D risk in a sex-dependent context have been inconsistent. OBJECTIVE A systematic meta-analysis was conducted to explore the sex-specific association of circulating ferritin with T2D risk. DATA SOURCES We searched PubMed, Web of Science, and EMBASE databases to identify available prospective studies through 1 August 2018. RESULTS Fifteen prospective studies comprising 77,352 participants and 18,404 patients with T2D, aged 20 to 80 years, and with ∼3 to 17 years of follow-up were identified. For each 100-μg/L increment in ferritin levels of overall participants, T2D risk increased by 22% (RR, 1.22; 95% CI, 1.14 to 1.31). Of note, major heterogeneities by sex were identified, with increased ferritin level having an apparently greater effect on T2D risk in women (RR, 1.53; 95% CI, 1.29 to 1.82) than in men (RR, 1.21; 95% CI, 1.15 to 1.27) after exclusion of a study with high heterogeneity (41,512 men and 6974 women for sex-specific analyses; P = 0.020 for sex difference). Further nonlinear analysis between circulating ferritin and T2D risk also showed sex-dimorphic association in that the T2D risk of women was twice as strong in magnitude as that of men at the same ferritin level. CONCLUSIONS Greater circulating ferritin levels were independently associated with increased T2D risk, which appeared stronger among women than men. Our findings provide prospective evidence for further testing of the utility of ferritin levels in predicting T2D risk in a sex-specific manner.
Collapse
Affiliation(s)
- Li Jiang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- The First Affiliated Hospital, School of Public Health, Zhengzhou University, Zhengzhou, China
| | - Kai Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Kenneth Lo
- Departments of Cardiology and Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Centre for Global Cardiometabolic Health, Department of Epidemiology, Brown University, Providence, Rhode Island
| | - Yueyang Zhong
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Aimin Yang
- Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Kong Kong SAR, China
| | - Xuexian Fang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Hailati Akezhuoli
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zijun Song
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Liyun Chen
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Peng An
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Mingqing Xu
- Bio-X Institutes, Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Shanghai Jiao Tong University, Shanghai, China
| | - Junxia Min
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fudi Wang
- The First Affiliated Hospital, School of Public Health, Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
- The First Affiliated Hospital, School of Public Health, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
18
|
Ma W, Feng Y, Jia L, Li S, Li J, Wang Z, Chen X, Du H. Dietary Iron Modulates Glucose and Lipid Homeostasis in Diabetic Mice. Biol Trace Elem Res 2019; 189:194-200. [PMID: 30027366 DOI: 10.1007/s12011-018-1446-3] [Citation(s) in RCA: 23] [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: 06/17/2018] [Accepted: 07/12/2018] [Indexed: 12/21/2022]
Abstract
Imbalance of iron homeostasis has been involved in clinical courses of metabolic diseases such as type 2 diabetes mellitus, obesity, and nonalcoholic fatty liver, through mechanisms not yet fully elucidated. Herein, we evaluated the effect of dietary iron on the development of diabetic syndromes in genetically obese db/db mice. Mice (aged 7 weeks) were fed with high-iron (HI) diets (1000 mg/kg chow) or low-iron (LI) diets (12 mg/kg) for 9 weeks. HI diets increased hepatic iron threefold and led to fourfold higher mRNA levels of hepcidin. HI also induced a 60% increase in fasting glucose due to insulin resistance, as confirmed by decreased hepatic glycogen deposition eightfold and a 21% decrease of serum adiponectin level. HI-fed mice had lower visceral adipose tissue mass estimated by epididymal and inguinal fat pad, associated with iron accumulation and smaller size of adipocytes. Gene expression analysis of liver showed that HI diet upregulated gluconeogenesis and downregulated lipogenesis. These results suggested that excess dietary iron leads to reduced mass, increased fasting glucose, decreased adiponectin level, and enhancement of insulin resistance, which indicated a multifactorial role of excess iron in the development of diabetes in the setting of obesity.
Collapse
Affiliation(s)
- Wan Ma
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Yunfei Feng
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Medical School of Zhejiang University, Hangzhou, China
| | - Li Jia
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Shuhui Li
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Jiahui Li
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Zhenjie Wang
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Xiaoyun Chen
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China
- College of Animal Science, Zhejiang University, Hangzhou, China
| | - Huahua Du
- Key Laboratory of Animal Nutrition and Feed Science (Eastern of China), Ministry of Agriculture, Zhejiang University, Hangzhou, China.
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, Zhejiang University, Hangzhou, China.
- College of Animal Science, Zhejiang University, Hangzhou, China.
| |
Collapse
|
19
|
Zhao J, Wu J, Yang Z, Ouyang L, Zhu L, Gao Z, Li H. Nitration of hIAPP promotes its toxic oligomer formation and exacerbates its toxicity towards INS-1 cells. Nitric Oxide 2019; 87:23-30. [PMID: 30849493 DOI: 10.1016/j.niox.2019.02.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 01/09/2023]
Abstract
Amyloid formation of human islet amyloid polypeptide (hIAPP) is one of the most common pathological features of type 2 diabetes (T2D). Increasing evidences have shown that the overproduction of reactive oxygen species (ROS) and reactive nitrogen species (RNS) play an important role in the development of the T2D. Interestingly, our previous studies indicated that heme could bind to hIAPP, and the complex might induce the nitration of tyrosine residue (Y37) of hIAPP in the presence of hydrogen peroxide and nitrite. However, it remains unclear about effect of the nitration on the implicated function of hIAPP in the development of T2D. In this study, fluorescent assays, transmission electron microscopy (TEM), atomic force microscope (AFM) were used to demonstrate that nitration of hIAPP significantly decreased its fibril formation. But the decreased fibril formation was not through the diminished aggregation of hIAPP monomer as suggested by the results of circular dichroism spectroscopy (CD) and gel electrophoresis assay. Surface-enhanced raman spectroscopy (SERS) indicated that nitration of hIAPP impaired the intermolecular hydrogen bonding. On the basis of these results, we hypothesize that nitration of hIAPP may block the intermolecular hydrogen bonding, leading to the inhibition of its fibril formation. In addition, cytotoxicity study of native and modified hIAPP was also performed on INS-1 cells, which revealed exacerbated toxicity of hIAPP by its nitration. The findings in this study that nitration of hIAPP promotes its oligomer formation and thus exacerbates its cytotoxicity suggests a possible link between the nitrite (or the sum of nitrite and nitrate) levels and T2D, and ameliorated nitration of hIAPP by diminishing nitrative stress might be a promising therapeutic strategy for T2D.
Collapse
Affiliation(s)
- Jie Zhao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Jinming Wu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Zhen Yang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China; Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, 77030, United States
| | - Lei Ouyang
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Lihua Zhu
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China
| | - Zhonghong Gao
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China.
| | - Hailing Li
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan, 430074, People's Republic of China.
| |
Collapse
|
20
|
Wang H, Cade BE, Sofer T, Sands SA, Chen H, Browning SR, Stilp AM, Louie TL, Thornton TA, Johnson WC, Below JE, Conomos MP, Evans DS, Gharib SA, Guo X, Wood AC, Mei H, Yaffe K, Loredo JS, Ramos AR, Barrett-Connor E, Ancoli-Israel S, Zee PC, Arens R, Shah NA, Taylor KD, Tranah GJ, Stone KL, Hanis CL, Wilson JG, Gottlieb DJ, Patel SR, Rice K, Post WS, Rotter JI, Sunyaev SR, Cai J, Lin X, Purcell SM, Laurie CC, Saxena R, Redline S, Zhu X. Admixture mapping identifies novel loci for obstructive sleep apnea in Hispanic/Latino Americans. Hum Mol Genet 2019; 28:675-687. [PMID: 30403821 PMCID: PMC6360325 DOI: 10.1093/hmg/ddy387] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/30/2018] [Accepted: 11/05/2018] [Indexed: 01/11/2023] Open
Abstract
Obstructive sleep apnea (OSA) is a common disorder associated with increased risk of cardiovascular disease and mortality. Its prevalence and severity vary across ancestral background. Although OSA traits are heritable, few genetic associations have been identified. To identify genetic regions associated with OSA and improve statistical power, we applied admixture mapping on three primary OSA traits [the apnea hypopnea index (AHI), overnight average oxyhemoglobin saturation (SaO2) and percentage time SaO2 < 90%] and a secondary trait (respiratory event duration) in a Hispanic/Latino American population study of 11 575 individuals with significant variation in ancestral background. Linear mixed models were performed using previously inferred African, European and Amerindian local genetic ancestry markers. Global African ancestry was associated with a lower AHI, higher SaO2 and shorter event duration. Admixture mapping analysis of the primary OSA traits identified local African ancestry at the chromosomal region 2q37 as genome-wide significantly associated with AHI (P < 5.7 × 10-5), and European and Amerindian ancestries at 18q21 suggestively associated with both AHI and percentage time SaO2 < 90% (P < 10-3). Follow-up joint ancestry-SNP association analyses identified novel variants in ferrochelatase (FECH), significantly associated with AHI and percentage time SaO2 < 90% after adjusting for multiple tests (P < 8 × 10-6). These signals contributed to the admixture mapping associations and were replicated in independent cohorts. In this first admixture mapping study of OSA, novel associations with variants in the iron/heme metabolism pathway suggest a role for iron in influencing respiratory traits underlying OSA.
Collapse
Affiliation(s)
- Heming Wang
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Brian E Cade
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Scott A Sands
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
| | - Han Chen
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Precision Health, School of Public Health & School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Sharon R Browning
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Adrienne M Stilp
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Tin L Louie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | | | - W Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Department of Medical Genetics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Matthew P Conomos
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Daniel S Evans
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Sina A Gharib
- Computational Medicine Core, Center for Lung Biology, UW Medicine Sleep Center, Division of Pulmonary, Critical Care and Sleep Medicine, University of Washington, Seattle, WA, USA
| | - Xiuqing Guo
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Alexis C Wood
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Yaffe
- Departments of Psychiatry and Neurology, University of California, San Francisco, San Francisco, CA, USA
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA, USA
| | - Jose S Loredo
- Division of Pulmonary Critical Care and Sleep Medicine, Department of Medicine, UC San Diego School of Medicine, La Jolla, CA, USA
| | - Alberto R Ramos
- Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | | | - Sonia Ancoli-Israel
- Departments of Medicine and Psychiatry, University of California, San Diego, CA, USA
- Department of Veterans Affairs, San Diego Center of Excellence for Stress and Mental Health, San Diego, CA, USA
| | - Phyllis C Zee
- Department of Neurology and Sleep Medicine Center, Northwestern University, Chicago, IL, USA
| | - Raanan Arens
- The Children’s Hospital at Montefiore, Division of Respiratory and Sleep Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Neomi A Shah
- Division of Pulmonary, Critical Care and Sleep Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Gregory J Tranah
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Katie L Stone
- California Pacific Medical Center Research Institute, San Francisco, CA, USA
| | - Craig L Hanis
- Human Genetics Center, Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - James G Wilson
- Physiology and Biophysics, University of Mississippi, Jackson, MS, USA
| | - Daniel J Gottlieb
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- VA Boston Healthcare System, Boston, MA, USA
| | - Sanjay R Patel
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ken Rice
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Wendy S Post
- Division of Cardiology, Johns Hopkins University, Baltimore, MD, USA
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute and Department of Pediatrics at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Shamil R Sunyaev
- Broad Institute, Cambridge, MA 02142, USA
- Division of Genetics, Brigham and Women’s Hospital, Boston, MA, USA
- Division of Medical Sciences, Harvard Medical School, Boston, MA, USA
| | - Jianwen Cai
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Shaun M Purcell
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Broad Institute, Cambridge, MA 02142, USA
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Cathy C Laurie
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Richa Saxena
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute, Cambridge, MA 02142, USA
- Center for Genomic Medicine and Department of Anesthesia, Pain, and Critical Care Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA , USA
- Division of Sleep Medicine, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary, Critical Care, and Sleep Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Xiaofeng Zhu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
21
|
Iron regulatory protein 2 deficiency may correlate with insulin resistance. Biochem Biophys Res Commun 2019; 510:191-197. [PMID: 30685084 DOI: 10.1016/j.bbrc.2019.01.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022]
Abstract
Iron is known to be a crucial regulator of glucose, and several studies have demonstrated that iron overload is one of the risk factors for insulin resistance and diabetes; however, the mechanism has not yet been clarified. To investigate the effect of iron overload on glucose metabolism and the underlying mechanism, Irp2 knockout (Irp2-/-) mice (endogenous iron overload model) were used. We found that Irp2-/- mice exhibited hyperglycemia and iron overload in the liver and skeletal muscle. Increased MDA, decreased SOD levels, and increased cell apoptosis were also found in the liver and muscle of Irp2-/- mice. Glucose concentrations were significantly higher in Irp2-/- mice in insulin tolerance tests. However, early-phase insulin secretion was not altered in Irp2-/- mice. The expression of hepatic IRS2 and muscle GLUT4 was declined in Irp2-/- mice at both mRNA and protein levels when compared with those of wild-type control. In conclusions, Irp2-/- mice showed hyperglycemia, which might due to insulin resistance rather than due to impaired insulin secretion.
Collapse
|
22
|
Dziegala M, Josiak K, Kasztura M, Kobak K, von Haehling S, Banasiak W, Anker SD, Ponikowski P, Jankowska E. Iron deficiency as energetic insult to skeletal muscle in chronic diseases. J Cachexia Sarcopenia Muscle 2018; 9:802-815. [PMID: 30178922 PMCID: PMC6204587 DOI: 10.1002/jcsm.12314] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 04/05/2018] [Accepted: 04/22/2018] [Indexed: 12/19/2022] Open
Abstract
Specific skeletal myopathy constitutes a common feature of heart failure, chronic obstructive pulmonary disease, and type 2 diabetes mellitus, where it can be characterized by the loss of skeletal muscle oxidative capacity. There is evidence from in vitro and animal studies that iron deficiency affects skeletal muscle functioning mainly in the context of its energetics by limiting oxidative metabolism in favour of glycolysis and by alterations in both carbohydrate and fat catabolic processing. In this review, we depict the possible molecular pathomechanisms of skeletal muscle energetic impairment and postulate iron deficiency as an important factor causally linked to loss of muscle oxidative capacity that contributes to skeletal myopathy seen in patients with heart failure, chronic obstructive pulmonary disease, and type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Magdalena Dziegala
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
| | - Krystian Josiak
- Centre for Heart DiseasesMilitary Hospital50‐981WroclawPoland
- Department of Heart DiseasesWroclaw Medical University50‐367WroclawPoland
| | - Monika Kasztura
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
| | - Kamil Kobak
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity Medicine Göttingen (UMG)37075GöttingenGermany
| | | | - Stefan D. Anker
- Department of Cardiology and PneumologyUniversity Medicine Göttingen (UMG)37075GöttingenGermany
- Division of Cardiology and MetabolismCharité Universitätsmedizin10117BerlinGermany
- Department of Cardiology (CVK)Charité Universitätsmedizin10117BerlinGermany
- Berlin‐Brandenburg Center for Regenerative Therapies (BCRT)Charité Universitätsmedizin10117BerlinGermany
- German Centre for Cardiovascular Research (DZHK) partner site BerlinCharité Universitätsmedizin10117BerlinGermany
| | - Piotr Ponikowski
- Centre for Heart DiseasesMilitary Hospital50‐981WroclawPoland
- Department of Heart DiseasesWroclaw Medical University50‐367WroclawPoland
| | - Ewa Jankowska
- Laboratory for Applied Research on Cardiovascular System, Department of Heart DiseasesWroclaw Medical University50‐981WroclawPoland
- Centre for Heart DiseasesMilitary Hospital50‐981WroclawPoland
| |
Collapse
|
23
|
Pokorski M, Poździk M, Mazzatenta A. Antioxidant treatment for impaired hypoxic ventilatory responses in experimental diabetes in the rat. Respir Physiol Neurobiol 2018; 255:30-38. [DOI: 10.1016/j.resp.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Revised: 05/05/2018] [Accepted: 05/08/2018] [Indexed: 12/19/2022]
|
24
|
Kell DB, Pretorius E. No effects without causes: the Iron Dysregulation and Dormant Microbes hypothesis for chronic, inflammatory diseases. Biol Rev Camb Philos Soc 2018; 93:1518-1557. [PMID: 29575574 PMCID: PMC6055827 DOI: 10.1111/brv.12407] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 02/12/2018] [Accepted: 02/15/2018] [Indexed: 12/11/2022]
Abstract
Since the successful conquest of many acute, communicable (infectious) diseases through the use of vaccines and antibiotics, the currently most prevalent diseases are chronic and progressive in nature, and are all accompanied by inflammation. These diseases include neurodegenerative (e.g. Alzheimer's, Parkinson's), vascular (e.g. atherosclerosis, pre-eclampsia, type 2 diabetes) and autoimmune (e.g. rheumatoid arthritis and multiple sclerosis) diseases that may appear to have little in common. In fact they all share significant features, in particular chronic inflammation and its attendant inflammatory cytokines. Such effects do not happen without underlying and initially 'external' causes, and it is of interest to seek these causes. Taking a systems approach, we argue that these causes include (i) stress-induced iron dysregulation, and (ii) its ability to awaken dormant, non-replicating microbes with which the host has become infected. Other external causes may be dietary. Such microbes are capable of shedding small, but functionally significant amounts of highly inflammagenic molecules such as lipopolysaccharide and lipoteichoic acid. Sequelae include significant coagulopathies, not least the recently discovered amyloidogenic clotting of blood, leading to cell death and the release of further inflammagens. The extensive evidence discussed here implies, as was found with ulcers, that almost all chronic, infectious diseases do in fact harbour a microbial component. What differs is simply the microbes and the anatomical location from and at which they exert damage. This analysis offers novel avenues for diagnosis and treatment.
Collapse
Affiliation(s)
- Douglas B. Kell
- School of ChemistryThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- The Manchester Institute of BiotechnologyThe University of Manchester, 131 Princess StreetManchesterLancsM1 7DNU.K.
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| | - Etheresia Pretorius
- Department of Physiological SciencesStellenbosch University, Stellenbosch Private Bag X1Matieland7602South Africa
| |
Collapse
|
25
|
Varghese J, James J, Vaulont S, Mckie A, Jacob M. Increased intracellular iron in mouse primary hepatocytes in vitro causes activation of the Akt pathway but decreases its response to insulin. Biochim Biophys Acta Gen Subj 2018; 1862:1870-1882. [PMID: 29859963 DOI: 10.1016/j.bbagen.2018.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 05/17/2018] [Accepted: 05/28/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND An iron-overloaded state has been reported to be associated with insulin resistance. On the other hand, conditions such as classical hemochromatosis (where iron overload occurs primarily in the liver) have been reported to be associated with increased insulin sensitivity. The reasons for these contradictory findings are unclear. In this context, the effects of increased intracellular iron per se on insulin signaling in hepatocytes are not known. METHODS Mouse primary hepatocytes were loaded with iron in vitro by incubation with ferric ammonium citrate (FAC). Intracellular events related to insulin signaling, as well as changes in gene expression and hepatocyte glucose production (HGP), were studied in the presence and absence of insulin and/or forskolin (a glucagon mimetic). RESULTS In vitro iron-loading of hepatocytes resulted in phosphorylation-mediated activation of Akt and AMP-activated protein kinase. This was associated with decreased basal and forskolin-stimulated HGP. Iron attenuated forskolin-mediated induction of the key gluconeogenic enzyme, glucose-6-phosphatase. It also attenuated activation of the Akt pathway in response to insulin, which was associated with decreased protein levels of insulin receptor substrates 1 and 2, constituting insulin resistance. CONCLUSIONS Increased intracellular iron has dual effects on insulin sensitivity in hepatocytes. It increased basal activation of the Akt pathway, but decreased activation of this pathway in response to insulin. GENERAL SIGNIFICANCE These findings may help explain why both insulin resistance and increased sensitivity have been observed in iron-overloaded states. They are of relevance to a variety of disease conditions characterized by hepatic iron overload and increased risk of diabetes.
Collapse
Affiliation(s)
- Joe Varghese
- Department of Biochemistry, Christian Medical College, Vellore 632002, India(1).
| | - Jithu James
- Department of Biochemistry, Christian Medical College, Vellore 632002, India(1)
| | | | - Andrew Mckie
- Diabetes and Nutritional Sciences Division, School of Medicine, King's College, London, UK
| | - Molly Jacob
- Department of Biochemistry, Christian Medical College, Vellore 632002, India(1)
| |
Collapse
|
26
|
Schindler C, Birkenfeld AL, Hanefeld M, Schatz U, Köhler C, Grüneberg M, Tschöpe D, Blüher M, Hasslacher C, Bornstein SR. Intravenous Ferric Carboxymaltose in Patients with Type 2 Diabetes Mellitus and Iron Deficiency: CLEVER Trial Study Design and Protocol. Diabetes Ther 2018; 9:37-47. [PMID: 29134606 PMCID: PMC5801218 DOI: 10.1007/s13300-017-0330-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Indexed: 01/31/2023] Open
Abstract
INTRODUCTION HbA1c is the gold standard for glycemic control in pre-diabetes and diabetes. However, its validity has been questioned, especially in the presence of imbalanced iron homeostasis. The CLEVER trial aims to evaluate the relationship between iron deficiency and HbA1c (a biomarker for the diagnosis and therapeutic monitoring of type 2 diabetes) in a randomized, placebo-controlled, multicenter clinical trial. METHODS The CLEVER (intravenous ferric CarboxymaLtosE for improVement of mEtabolic parameters in type 2 diabetes patients with iRon deficiency) trial is a randomized, single-blind, proof-of-concept study with two treatment arms. 140 men and women diagnosed with type 2 diabetes and iron deficiency will receive either placebo or ferric carboxymaltose (500 or 1000 mg) as intravenous infusions. The primary outcome measure is the change in HbA1c level between baseline and after 12 weeks of treatment. Secondary endpoints include change of iron status and metabolic markers as well as treatment safety and tolerability. Furthermore, the potential clinical improvement in quality of life and the reliability of HbA1c measurement in patients with type 2 diabetes and iron deficiency will be investigated. RESULTS Both excessive iron and iron deficiency are associated with metabolic disorders; excessive iron is a risk factor for the development of diabetes, whereas iron deficiency is associated with obesity and insulin resistance. It has been suggested that iron increases insulin secretion in pancreatic beta-cells. CLEVER is the first study to investigate the hypothesis that intravenous substitution with ferric carboxymaltose reduces HbA1c levels in patients with type 2 diabetes and iron deficiency, thereby improving metabolic status and quality of life. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov (NCT01513369). FUNDING GWT-TUD GmbH acts as sponsor of the clinical trial. Financial support is provided by Vifor Pharma.
Collapse
Affiliation(s)
- Christoph Schindler
- Clinical Research Center Hannover and Center for Pharmacology and Toxicology, Hannover Medical School, Hannover, Germany.
| | - Andreas L Birkenfeld
- Medical Clinic and Policlinic III at the University Hospital Dresden, Dresden, Germany
- Centre for Metabolic Vascular Medicine, GWT-TUD, Dresden, Germany
| | - Markolf Hanefeld
- Centre for Metabolic Vascular Medicine, GWT-TUD, Dresden, Germany
| | - Ulrike Schatz
- Medical Clinic and Policlinic III at the University Hospital Dresden, Dresden, Germany
| | | | | | - Diethelm Tschöpe
- Herz- und Diabeteszentrum Nordrhein-Westfalen, Ruhr-Universität Bochum, Bad Oeynhausen, Germany
| | - Matthias Blüher
- Department of Medicine at the University Hospital Leipzig, Leipzig, Germany
| | - Christoph Hasslacher
- Diabetesinstitut Heidelberg and Department of Clinical Studies, St. Josefskrankenhaus Heidelberg, Heidelberg, Germany
| | - Stefan R Bornstein
- Medical Clinic and Policlinic III at the University Hospital Dresden, Dresden, Germany
| |
Collapse
|
27
|
Lee G, Jun Y, Jang H, Yoon J, Lee J, Hong M, Chung S, Kim DH, Lee S. Enhanced oxygen permeability in membrane-bottomed concave microwells for the formation of pancreatic islet spheroids. Acta Biomater 2018; 65:185-196. [PMID: 29101017 DOI: 10.1016/j.actbio.2017.10.045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Revised: 10/22/2017] [Accepted: 10/30/2017] [Indexed: 12/20/2022]
Abstract
Oxygen availability is a critical factor in regulating cell viability that ultimately contributes to the normal morphogenesis and functionality of human tissues. Among various cell culture platforms, construction of 3D multicellular spheroids based on microwell arrays has been extensively applied to reconstitute in vitro human tissue models due to its precise control of tissue culture conditions as well as simple fabrication processes. However, an adequate supply of oxygen into the spheroidal cellular aggregation still remains one of the main challenges to producing healthy in vitro spheroidal tissue models. Here, we present a novel design for controlling the oxygen distribution in concave microwell arrays. We show that oxygen permeability into the microwell is tightly regulated by varying the poly-dimethylsiloxane (PDMS) bottom thickness of the concave microwells. Moreover, we validate the enhanced performance of the engineered microwell arrays by culturing non-proliferated primary rat pancreatic islet spheroids on varying bottom thickness from 10 μm to 1050 μm. Morphological and functional analyses performed on the pancreatic islet spheroids grown for 14 days prove the long-term stability, enhanced viability, and increased hormone secretion under the sufficient oxygen delivery conditions. We expect our results could provide knowledge on oxygen distribution in 3-dimensional spheroidal cell structures and critical design concept for tissue engineering applications. STATEMENT OF SIGNIFICANCE In this study, we present a noble design to control the oxygen distribution in concave microwell arrays for the formation of highly functional pancreatic islet spheroids by engineering the bottom of the microwells. Our new platform significantly enhanced oxygen permeability that turned out to improve cell viability and spheroidal functionality compared to the conventional thick-bottomed 3-D culture system. Therefore, we believe that this could be a promising medical biotechnology platform to further develop high-throughput tissue screening system as well as in vivo-mimicking customised 3-D tissue culture systems.
Collapse
|
28
|
Heme Oxygenase Induction Suppresses Hepatic Hepcidin and Rescues Ferroportin and Ferritin Expression in Obese Mice. J Nutr Metab 2017; 2017:4964571. [PMID: 29062571 PMCID: PMC5618758 DOI: 10.1155/2017/4964571] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/08/2017] [Indexed: 12/17/2022] Open
Abstract
Hepcidin, a phase II reactant secreted by hepatocytes, regulates cellular iron levels by increasing internalization of ferroportin-a transmembrane protein facilitating egress of cellular iron. Chronic low-grade inflammatory states, such as obesity, have been shown to increase oxidative stress and enhance hepcidin secretion from hepatocytes and macrophages. Heme-heme oxygenase (HO) is a stress response system which reduces oxidative stress. We investigated the effects of HO-1 induction on hepatic hepcidin levels and on iron homeostasis in hepatic tissues from lean and obese mice. Obese mice exhibited hyperglycemia (p < 0.05); increased levels of proinflammatory cytokines (MCP-1, IL-6, p < 0.05); oxidative stress (p < 0.05); and increased hepatic hepcidin levels (p < 0.05). Enhancement of hepcidin was reflected in the reduced expression of ferroportin in obese mice (p < 0.05). However, this effect is accompanied by a significant decline in ferritin expression. Additionally, there are reduced insulin receptor phosphorylation and attenuation of metabolic regulators pAMPK, pAKT, and pLKB1. Cobalt protoporphyrin- (CoPP-) induced HO-1 upregulation in obese mice reversed these alterations (p < 0.05), while attenuating hepatic hepcidin levels. These effects of CoPP were prevented in obese mice concurrently exposed to an inhibitor of HO (SnMP) (p < 0.05). Our results highlight a modulatory effect of HO on iron homeostasis mediated through the suppression of hepatic hepcidin.
Collapse
|
29
|
Huang Y, Yang Z, Xu H, Zhang P, Gao Z, Li H. Insulin enhances the peroxidase activity of heme by forming heme-insulin complex: Relevance to type 2 diabetes mellitus. Int J Biol Macromol 2017; 102:1009-1015. [DOI: 10.1016/j.ijbiomac.2017.04.113] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Revised: 04/26/2017] [Accepted: 04/28/2017] [Indexed: 12/12/2022]
|
30
|
Wu J, Zhao J, Yang Z, Li H, Gao Z. Strong Inhibitory Effect of Heme on hIAPP Fibrillation. Chem Res Toxicol 2017; 30:1711-1719. [DOI: 10.1021/acs.chemrestox.7b00170] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jinming Wu
- School
of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People’s Republic of China
| | - Jie Zhao
- School
of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People’s Republic of China
| | - Zhen Yang
- School
of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People’s Republic of China
- Department
of Chemical and Biomolecular Engineering, University of Houston, Houston, Texas 77004, United States
| | - Hailing Li
- School
of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People’s Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, People’s Republic of China
| | - Zhonghong Gao
- School
of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, People’s Republic of China
- Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, Huazhong University of Science and Technology, 1037 Luoyu Road, Wuhan 430074, People’s Republic of China
| |
Collapse
|
31
|
Cohen N, Breker M, Bakunts A, Pesek K, Chas A, Argemí J, Orsi A, Gal L, Chuartzman S, Wigelman Y, Jonas F, Walter P, Ernst R, Aragón T, van Anken E, Schuldiner M. Iron affects Ire1 clustering propensity and the amplitude of endoplasmic reticulum stress signaling. J Cell Sci 2017; 130:3222-3233. [PMID: 28794014 PMCID: PMC5665437 DOI: 10.1242/jcs.201715] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 08/02/2017] [Indexed: 01/10/2023] Open
Abstract
The unfolded protein response (UPR) allows cells to adjust secretory pathway capacity according to need. Ire1, the endoplasmic reticulum (ER) stress sensor and central activator of the UPR is conserved from the budding yeast Saccharomyces cerevisiae to humans. Under ER stress conditions, Ire1 clusters into foci that enable optimal UPR activation. To discover factors that affect Ire1 clustering, we performed a high-content screen using a whole-genome yeast mutant library expressing Ire1–mCherry. We imaged the strains following UPR induction and found 154 strains that displayed alterations in Ire1 clustering. The hits were enriched for iron and heme effectors and binding proteins. By performing pharmacological depletion and repletion, we confirmed that iron (Fe3+) affects UPR activation in both yeast and human cells. We suggest that Ire1 clustering propensity depends on membrane composition, which is governed by heme-dependent biosynthesis of sterols. Our findings highlight the diverse cellular functions that feed into the UPR and emphasize the cross-talk between organelles required to concertedly maintain homeostasis. Highlighted Article: To respond to folding stress in the ER, cells activate the conserved sensor Ire1. We show that iron is required for optimal Ire1 activation and suggest this is because iron is required for ergosterol biosynthesis.
Collapse
Affiliation(s)
- Nir Cohen
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Michal Breker
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel.,The Rockefeller University, New York, NY 10065, USA
| | - Anush Bakunts
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Ospedale San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Kristina Pesek
- Institute of Biochemistry and Buchmann Institute for Molecular Life Sciences, Goethe University Frankfurt, Max-von-Laue Str. 15, 60438 Frankfurt, Germany
| | - Ainara Chas
- Center for Applied Medical Research, Department of Gene Therapy and Regulation of Gene Expression. University of Navarra, 55 Pio XII St. 31008 Pamplona, Spain
| | - Josepmaria Argemí
- Center for Applied Medical Research, Department of Gene Therapy and Regulation of Gene Expression. University of Navarra, 55 Pio XII St. 31008 Pamplona, Spain
| | - Andrea Orsi
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Ospedale San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Lihi Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Silvia Chuartzman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Yoav Wigelman
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Felix Jonas
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Peter Walter
- Department of Biochemistry & Biophysics, University of California San Francisco and Howard Hughes Medical Institute, San Francisco, CA 94143, USA
| | - Robert Ernst
- Center for Molecular Signaling, Institute of Medical Biochemistry and Molecular Biology, Saarland University, 66421 Homburg, Germany
| | - Tomás Aragón
- Center for Applied Medical Research, Department of Gene Therapy and Regulation of Gene Expression. University of Navarra, 55 Pio XII St. 31008 Pamplona, Spain
| | - Eelco van Anken
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Ospedale San Raffaele, Via Olgettina 58, 20132, Milan, Italy
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
32
|
Bazi A, Sharifi-Rad J, Rostami D, Sargazi-Aval O, Safa A. Diabetes Mellitus in Thalassaemia Major Patients: A Report from the Southeast of Iran. J Clin Diagn Res 2017; 11:BC01-BC04. [PMID: 28658748 DOI: 10.7860/jcdr/2017/24762.9806] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 01/03/2017] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Diabetes Mellitus (DM) represents a major concern in Thalassaemia Major (TM) patients. AIM The present study was conducted to evaluate the frequency of Impaired Fasting Glucose (IFG) and DM in TM patients in Southeast of Iran. MATERIALS AND METHODS Fasting Blood Glucose (FBS) was determined using fasting blood samples in 148 TM patients. Demographical data was collected by a questionnaire. Clinical and laboratory variables including cell blood counts, pre-transfusion Haemoglobin (Hb) level, and five-year ferritin were extracted from medical records. Statistical analysis was performed in SPSS19.0 software using chi-square, student t-test and logistic regression. RESULTS Females and males comprised 83 (56.1%) and 65 (43.9%) subjects respectively. The mean age and mean five-year ferritin were 17.3±6.1 year-old and 5060.6±2395 ng/ml respectively. Overall, 39 (26.4%) patients had IFG, while 13 (8.8%) were diagnosed with DM. Significant differences were identified in the mean age, volume of transfused blood per occasion, and mean five-years ferritin between the patients with IFG or DM and the patients with normal fasting glucose level. Patients with age >25-year-old had an increased risk of both IFG (OR=4.7,95% CI: 1.3-17, p=0.01) and DM (OR= 7.1, 95% CI: 1-49.2, p=0.04). In addition, splenectomized patients showed a higher risk for IFG (OR=4.3, 95% CI: 1.5-12.1, p=0.005), and ferritin value >6000 ng/ml were associated with an elevated risk of DM (OR=7, 95% CI: 0.8-60.1, p=0.07). CONCLUSION Our results indicated that higher age, mean five-years ferritin, volume of blood transfused per occasion, as well as splenectomy were risk factors of IFG and DM in TM patients.
Collapse
Affiliation(s)
- Ali Bazi
- Lecturer, Clinical Research Development Unit, Amir-Al-Momenin Hospital, Zabol University Medical Science, Zabol, Iran
| | - Javad Sharifi-Rad
- Research Scholar, Department of Biochemistry, Phytochemistry Research Scholar, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Daryoush Rostami
- Associate Professor, Department of Hematology, Faculty of Allied Medical Science, Zabol University Medical Science, Zabol, Iran
| | - Omolbanin Sargazi-Aval
- Lecturer, Department of Hematology, Faculty of Allied Medical Science, Zabol University Medical Science, Zabol, Iran
| | - Amin Safa
- Lecturer, Department of Immunology, Faculty of Allied Medical Science, Zabol University Medical Science, Zabol, Iran
| |
Collapse
|
33
|
Vinokur V, Weksler-Zangen S, Berenshtein E, Eliashar R, Chevion M. The Loss of Myocardial Benefit following Ischemic Preconditioning Is Associated with Dysregulation of Iron Homeostasis in Diet-Induced Diabetes. PLoS One 2016; 11:e0159908. [PMID: 27458721 PMCID: PMC4961428 DOI: 10.1371/journal.pone.0159908] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 07/11/2016] [Indexed: 11/18/2022] Open
Abstract
Whether the diabetic heart benefits from ischemic preconditioning (IPC), similar to the non-diabetic heart, is a subject of controversy. We recently proposed new roles for iron and ferritin in IPC-protection in Type 1-like streptozotocin-induced diabetic rat heart. Here, we investigated iron homeostasis in Cohen diabetic sensitive rat (CDs) that develop hyperglycemia when fed on a high-sucrose/low-copper diet (HSD), but maintain normoglycemia on regular-diet (RD). Control Cohen-resistant rats (CDr) maintain normoglycemia on either diet. The IPC procedure improved the post-ischemic recovery of normoglycemic hearts (CDr-RD, CDr-HSD and CDs-RD). CDs-HSD hearts failed to show IPC-associated protection. The recovery of these CDs-HSD hearts following I/R (without prior IPC) was better than their RD controls. During IPC ferritin levels increased in normoglycemic hearts, and its level was maintained nearly constant during the subsequent prolonged ischemia, but decayed to its baseline level during the reperfusion phase. In CDs-HSD hearts the baseline levels of ferritin and ferritin-saturation with iron were notably higher than in the controls, and remained unchanged during the entire experiment. This unique and abnormal pattern of post-ischemic recovery of CDs-HSD hearts is associated with marked changes in myocardial iron homeostasis, and suggests that iron and iron-proteins play a causative role/s in the etiology of diabetes-associated cardiovascular disorders.
Collapse
Affiliation(s)
- Vladimir Vinokur
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
- Department of Otolaryngology-Head and Neck Surgery, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Sarah Weksler-Zangen
- The Diabetes Unit, Department of Internal Medicine, Hadassah-Hebrew University Medical Centre, Jerusalem, Israel
| | - Eduard Berenshtein
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ron Eliashar
- Department of Otolaryngology-Head and Neck Surgery, Hadassah-Hebrew University Hospital, Jerusalem, Israel
| | - Mordechai Chevion
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
34
|
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
|
35
|
Hepcidin and iron metabolism in non-diabetic obese and type 2 diabetic rats. ACTA ACUST UNITED AC 2015; 35:851-857. [DOI: 10.1007/s11596-015-1517-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 10/15/2015] [Indexed: 01/12/2023]
|
36
|
Yin Y, Han W, Wang Y, Zhang Y, Wu S, Zhang H, Jiang L, Wang R, Zhang P, Yu Y, Li B. Identification of Risk Factors Affecting Impaired Fasting Glucose and Diabetes in Adult Patients from Northeast China. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:12662-78. [PMID: 26473900 PMCID: PMC4626992 DOI: 10.3390/ijerph121012662] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2015] [Revised: 09/17/2015] [Accepted: 10/08/2015] [Indexed: 12/18/2022]
Abstract
BACKGROUND Besides genetic factors, the occurrence of diabetes is influenced by lifestyles and environmental factors as well as trace elements in diet materials. Subjects with impaired fasting glucose (IFG) have an increased risk of developing diabetes mellitus (DM). This study aimed to explore risk factors affecting IFG and diabetes in patients from Northeast China. METHODS A population-based, cross-sectional survey of chronic diseases and related risk factors was conducted in Jilin Province of Northeast China. All adult residents, aged 18-79, were invited to participate in this survey using the method of multistage stratified random cluster sampling. One hundred thirty-four patients with IFG or DM and 391 healthy control subjects were recruited. We compared demographic factors, body size measurements, healthy-related behaviors, and hair metallic element contents between IFG/diabetes patients and healthy individuals. RESULTS IFG/diabetes patients had a greater weight, waist, hip, and body mass index (BMI) than control subjects. Significant differences in the content of zinc (Zn), potassium (K), copper (Ca), and sodium (Na) as well as Cu/Zn ratios between IFG or DM patients and control subjects (p < 0.05) were also observed. Hair Cu, selenium (Se), and Na contents were positively correlated with blood glucose levels (Cu: rs = 0.135, p = 0.002; Se: rs = 0.110, p = 0.012; Na: rs = 0.091, p = 0.038). Polytomous logistic regression adjusting for age, sex, family history of diabetes and BMI, showed that subjects with high BMI were more likely to develop IFG and DM (IFG: OR = 1.15, OR 95% CI = 1.02-1.29; DM: OR = 1.15, OR 95% CI = 1.01-1.33). Moreover, rarely or never eating fruits was a risk factor for DM (OR = 5.46, OR 95% CI = 1.87-15.98) but not for IFG (OR = 1.70, OR 95% CI = 0.72-4.02). Subjects with abdominal obesity or DM history were more susceptible to DM (abdominal obesity: OR = 2.99, OR 95% CI = 1.07-8.37; DM history: OR = 2.69, OR 95% CI = 1.01-7.20). We found that subjects living in Changling County had a significantly lower chance to suffer from IFG (OR and 95% CI for OR: 0.25, 0.08-0.74). Men and 60-69 years old subjects were at increased risk for IFG (male: OR = 3.51, OR 95% CI = 1.34-9.18; age 60-69: OR = 6.64, OR 95% CI = 1.36-32.47). We did not find significant associations of IFG or DM with certain lifestyles (such as eating more meat, exercise or physical activity, smoking, or alcohol drinking) or the content of some metallic elements (such as iron (Fe), Zn , K, calcium (Ca), Na, or magnesium (Mg)). CONCLUSIONS This study demonstrated that less or no fruit eating, DM family history, abdominal obesity conferred vulnerability to DM. Living in Changling County, men and 60-69 years old subjects were found to be risk factors for IFG. Subjects with high BMI were more likely to develop IFG and DM.
Collapse
Affiliation(s)
- Yutian Yin
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Weiqing Han
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Yuhan Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Yue Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Shili Wu
- Administration Bureau of Changbai Mountain Natural Mineral Water Source Protection Areas, Jilin 130021, China.
| | - Huiping Zhang
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT 06511, USA.
| | - Lingling Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Rui Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Peng Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Yaqin Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| | - Bo Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, 1163 Xinmin Street, Changchun, Jilin 130021, China.
| |
Collapse
|