1
|
Amangurbanova M, Huang DQ, Noureddin N, Tesfai K, Bettencourt R, Siddiqi H, Lopez SJ, Cervantes V, Madamba E, Loomba R. A Prospective Study on the Prevalence of MASLD in Patients With Type 2 Diabetes and Hyperferritinaemia. Aliment Pharmacol Ther 2025; 61:456-464. [PMID: 39499168 DOI: 10.1111/apt.18377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/08/2024] [Accepted: 10/22/2024] [Indexed: 11/07/2024]
Abstract
BACKGROUND Elevated levels of serum ferritin, a marker of hepatic iron overload and inflammation, may be associated with metabolic dysfunction-associated steatotic liver disease (MASLD) and hepatic fibrosis. AIM To determine the prevalence of MASLD and significant hepatic fibrosis among patients with type 2 diabetes mellitus (T2DM) and hyperferritinaemia. METHODS This is a cross-sectional analysis of a prospective cohort of 523 adults (64% female) aged 50-80 with T2DM and without a diagnosis of haemochromatosis. MASLD and significant fibrosis were defined as magnetic resonance imaging-proton density fat fraction (MRI-PDFF) ≥ 5% and magnetic resonance elastography (MRE) ≥ 3.0 kPa, respectively. Hyperferritinaemia was defined as serum ferritin ≥ 200 ng/mL in females or ≥ 300 ng/mL in males. The primary objective was to determine the prevalence of MASLD and significant fibrosis in hyperferritinaemia. RESULTS The mean age and body mass index were 64.1 (±8.1) years and 31.5 (±5.9) kg/m2, respectively. The overall prevalence of hyperferritinaemia was 20.5% (n = 107). The prevalence of MASLD (78.5% vs. 62.1%, p = 0.001) and significant fibrosis (35.5% vs. 22.1%, p = 0.002) were higher in participants with hyperferritinaemia than those without. Hyperferritinaemia remained an independent predictor of MASLD (OR 2.01; 95% CI 1.19-3.39; p = 0.009) and significant fibrosis (OR 2.33; CI 1.43-3.77; p = 0.001), even after adjustment for age, sex, obesity and insulin use. CONCLUSION Approximately 80% of people with hyperferritinaemia and T2DM have MASLD, and more than a third have significant hepatic fibrosis. Hyperferritinaemia may be a useful biomarker for MASLD and significant fibrosis in people with T2DM.
Collapse
Affiliation(s)
- Maral Amangurbanova
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Daniel Q Huang
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, Department of Medicine, National University Health System, Singapore, Singapore
| | - Nabil Noureddin
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Kaleb Tesfai
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Richelle Bettencourt
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Harris Siddiqi
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Scarlett J Lopez
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Vanessa Cervantes
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Egbert Madamba
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
| | - Rohit Loomba
- Division of Gastroenterology, MASLD Research Center, University of California at San Diego, La Jolla, California, USA
- Division of Epidemiology, Department of Family Medicine and Public Health, University of California at San Diego, San Diego, California, USA
| |
Collapse
|
2
|
Schleh MW, Ameka MK, Rodriguez AS, Hasty AH. Deficiency of the Hemoglobin-Haptoglobin Receptor, CD163, Worsens Insulin Sensitivity in Obese Male Mice. Diabetes 2024; 73:1990-2002. [PMID: 39325576 PMCID: PMC11579413 DOI: 10.2337/db24-0405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/17/2024] [Indexed: 09/28/2024]
Abstract
Excessive iron accumulation in metabolic organs such as the adipose tissue, liver, and skeletal muscle is associated with increased diabetes risk. Tissue-resident macrophages serve multiple roles, including managing inflammatory tone and regulating parenchymal iron homeostasis, thus protecting against metabolic dysfunction upon iron overload. The scavenger receptor CD163 is uniquely present on tissue-resident macrophages and plays a significant role in iron homeostasis by clearing extracellular hemoglobin-haptoglobin complexes, thereby limiting oxidative damage caused by free hemoglobin in metabolic tissues. We show that the absence of CD163 exacerbates glucose intolerance and insulin resistance in male mice with obesity. Additionally, loss of CD163 reduced the expression of iron regulatory genes (Tfr1, Cisd1, Slc40a1) in adipose tissue macrophages and anti-inflammatory (M2-like) bone marrow-derived macrophages (BMDMs). Furthermore, CD163 deficiency mediated a proinflammatory shift and limited hemoglobin scavenging specifically in M2-like BMDMs. To this end, iron buffering was diminished in inguinal white adipose tissue (iWAT) macrophages in vivo, which culminated in iron spillover into adipocytes and CD45+ CD11B- nonmyeloid immune cells in iWAT. These findings show that CD163 on tissue-resident macrophages is critical for their anti-inflammatory and hemoglobin scavenging roles, and its absence results in impaired systemic insulin action in an obese setting. ARTICLE HIGHLIGHTS
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, Myelomonocytic/metabolism
- Antigens, Differentiation, Myelomonocytic/genetics
- Male
- Antigens, CD/metabolism
- Antigens, CD/genetics
- Insulin Resistance/physiology
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Mice
- Obesity/metabolism
- Macrophages/metabolism
- Haptoglobins/metabolism
- Haptoglobins/genetics
- Hemoglobins/metabolism
- Iron/metabolism
- Mice, Inbred C57BL
- Mice, Obese
- Receptors, Transferrin/metabolism
- Receptors, Transferrin/genetics
- Adipose Tissue, White/metabolism
- Mice, Knockout
- Glucose Intolerance/metabolism
- Glucose Intolerance/genetics
- Cation Transport Proteins/metabolism
- Cation Transport Proteins/genetics
Collapse
Affiliation(s)
- Michael W. Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Magdalene K. Ameka
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alec S. Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
| | - Alyssa H. Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN
- VA Tennessee Valley Healthcare System, Nashville, TN
| |
Collapse
|
3
|
Mohammadi S, Ghaderi S, Fatehi F. Iron accumulation/overload and Alzheimer's disease risk factors in the precuneus region: A comprehensive narrative review. Aging Med (Milton) 2024; 7:649-667. [PMID: 39507230 PMCID: PMC11535174 DOI: 10.1002/agm2.12363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that is characterized by amyloid plaques, neurofibrillary tangles, and neuronal loss. Early cerebral and body iron dysregulation and accumulation interact with AD pathology, particularly in the precuneus, a crucial functional hub in cognitive functions. Quantitative susceptibility mapping (QSM), a novel post-processing approach, provides insights into tissue iron levels and cerebral oxygen metabolism and reveals abnormal iron accumulation early in AD. Increased iron deposition in the precuneus can lead to oxidative stress, neuroinflammation, and accelerated neurodegeneration. Metabolic disorders (diabetes, non-alcoholic fatty liver disease (NAFLD), and obesity), genetic factors, and small vessel pathology contribute to abnormal iron accumulation in the precuneus. Therefore, in line with the growing body of literature in the precuneus region of patients with AD, QSM as a neuroimaging method could serve as a non-invasive biomarker to track disease progression, complement other imaging modalities, and aid in early AD diagnosis and monitoring.
Collapse
Affiliation(s)
- Sana Mohammadi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
| | - Sadegh Ghaderi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in MedicineTehran University of Medical SciencesTehranIran
| | - Farzad Fatehi
- Neuromuscular Research Center, Department of Neurology, Shariati HospitalTehran University of Medical SciencesTehranIran
- Neurology DepartmentUniversity Hospitals of Leicester NHS TrustLeicesterUK
| |
Collapse
|
4
|
Drzymała A. The Functions of SARS-CoV-2 Receptors in Diabetes-Related Severe COVID-19. Int J Mol Sci 2024; 25:9635. [PMID: 39273582 PMCID: PMC11394807 DOI: 10.3390/ijms25179635] [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: 08/01/2024] [Revised: 08/25/2024] [Accepted: 09/01/2024] [Indexed: 09/15/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is considered a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) receptor of high importance, but due to its non-ubiquitous expression, studies of other proteins that may participate in virus internalisation have been undertaken. To date, many alternative receptors have been discovered. Their functioning may provide an explanation for some of the events observed in severe COVID-19 that cannot be directly explained by the model in which ACE2 constitutes the central point of infection. Diabetes mellitus type 2 (T2D) can induce severe COVID-19 development. Although many mechanisms associated with ACE2 can lead to increased SARS-CoV-2 virulence in diabetes, proteins such as basigin (CD147), glucose-regulated protein 78 kDa (GRP78), cluster of differentiation 4 (CD4), transferrin receptor (TfR), integrins α5β1/αvβ3, or ACE2 co-receptors neuropilin 2 (NRP2), vimentin, and even syalilated gangliosides may also be responsible for worsening the COVID-19 course. On the other hand, some others may play protective roles. Understanding how diabetes-associated mechanisms can induce severe COVID-19 via modification of virus receptor functioning needs further extensive studies.
Collapse
Affiliation(s)
- Adam Drzymała
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medical Sciences, University of Opole, Oleska 48, 45-052 Opole, Poland
| |
Collapse
|
5
|
Wang F, Glenn AJ, Tessier AJ, Mei Z, Haslam DE, Guasch-Ferré M, Tobias DK, Eliassen AH, Manson JE, Clish C, Lee KH, Rimm EB, Wang DD, Sun Q, Liang L, Willett WC, Hu FB. Integration of epidemiological and blood biomarker analysis links haem iron intake to increased type 2 diabetes risk. Nat Metab 2024; 6:1807-1818. [PMID: 39138340 DOI: 10.1038/s42255-024-01109-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024]
Abstract
Dietary haem iron intake is linked to an increased risk of type 2 diabetes (T2D), but the underlying plasma biomarkers are not well understood. We analysed data from 204,615 participants (79% females) in three large US cohorts over up to 36 years, examining the associations between iron intake and T2D risk. We also assessed plasma metabolic biomarkers and metabolomic profiles in subsets of 37,544 (82% females) and 9,024 (84% females) participants, respectively. Here we show that haem iron intake but not non-haem iron is associated with a higher T2D risk, with a multivariable-adjusted hazard ratio of 1.26 (95% confidence interval 1.20-1.33; P for trend <0.001) comparing the highest to the lowest quintiles. Haem iron accounts for significant proportions of the T2D risk linked to unprocessed red meat and specific dietary patterns. Increased haem iron intake correlates with unfavourable plasma profiles of insulinaemia, lipids, inflammation and T2D-linked metabolites. We also identify metabolites, including L-valine and uric acid, potentially mediating the haem iron-T2D relationship, highlighting their pivotal role in T2D pathogenesis.
Collapse
Affiliation(s)
- Fenglei Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Andrea J Glenn
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Toronto 3D Knowledge Synthesis and Clinical Trials Unit, Clinical Nutrition and Risk Factor Modification Centre, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Anne-Julie Tessier
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Zhendong Mei
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Danielle E Haslam
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Marta Guasch-Ferré
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Public Health, Section of Epidemiology, University of Copenhagen, Copenhagen, Denmark
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Deirdre K Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - A Heather Eliassen
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - JoAnn E Manson
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Clary Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kyu Ha Lee
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Dong D Wang
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Liming Liang
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Walter C Willett
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| |
Collapse
|
6
|
Mai X, Liu Y, Fan J, Xiao L, Liao M, Huang Z, Chen Z, Huang S, Sun R, Jiang X, Huang L, Sun J, Xie L, Chen H. Iron supplementation and iron accumulation promote adipocyte thermogenesis through PGC1α-ATGL-mediated lipolysis. J Biol Chem 2024; 300:107690. [PMID: 39159807 PMCID: PMC11420453 DOI: 10.1016/j.jbc.2024.107690] [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: 01/09/2024] [Revised: 07/24/2024] [Accepted: 08/07/2024] [Indexed: 08/21/2024] Open
Abstract
Iron homeostasis is essential for maintaining metabolic health and iron disorder has been linked to chronic metabolic diseases. Increasing thermogenic capacity in adipose tissue has been considered as a potential approach to regulate energy homeostasis. Both mitochondrial biogenesis and mitochondrial function are iron-dependent and essential for adipocyte thermogenic capacity, but the underlying relationships between iron accumulation and adipose thermogenesis is unclear. Firstly, we confirmed that iron homeostasis and the iron regulatory markers (e.g., Tfr1 and Hfe) are involved in cold-induced thermogenesis in subcutaneous adipose tissues using RNA-seq and bioinformatic analysis. Secondly, an Hfe (Hfe-/-)-deficient mouse model, in which tissues become overloaded with iron, was employed. We found iron accumulation caused by Hfe deficiency enhanced mitochondrial respiratory chain expression in subcutaneous white adipose in vivo and resulted in enhanced tissue thermogenesis with upregulation of PGC-1α and adipose triglyceride lipase, mitochondrial biogenesis and lipolysis. To investigate the thermogenic capacity in vitro, stromal vascular fraction from adipose tissues was isolated, followed with adipogenic differentiation. Primary adipocyte from Hfe-/- mice exhibited higher cellular oxygen consumption, associated with enhanced expression of mitochondrial oxidative respiratory chain protein, while primary adipocytes or stromal vascular fractions from WT mice supplemented with iron citrate) exhibited similar effect in thermogenic capacity. Taken together, these findings indicate iron supplementation and iron accumulation (Hfe deficiency) can regulate adipocyte thermogenic capacity, suggesting a potential role for iron homeostasis in adipose tissues.
Collapse
Affiliation(s)
- Xudong Mai
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China; State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Yifan Liu
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Jigang Fan
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lanling Xiao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Miaomiao Liao
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Zhipeng Huang
- State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Zijian Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shaojun Huang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Rui Sun
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaowan Jiang
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liujing Huang
- Medical Affairs Department, Guangzhou Betrue Technology Co, Ltd, Guangzhou, China
| | - Jia Sun
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Liwei Xie
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China; State Key Laboratory of Applied Microbiology Southern China, Guangdong Provincial Key Laboratory of Microbial Culture Collection and Application, Guangdong Open Laboratory of Applied Microbiology, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China; College of Life and Health Sciences, Guangdong Industry Polytechnic, Guangzhou, Guangdong, China.
| | - Hong Chen
- Department of Endocrinology and Metabolism, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
7
|
Yang T, Chen S, Qiu L, Guo Q, Wang Z, Jiang Y, Bai H, Bi Y, Chang G. Effect of High Dietary Iron on Fat Deposition and Gut Microbiota in Chickens. Animals (Basel) 2024; 14:2254. [PMID: 39123780 PMCID: PMC11310990 DOI: 10.3390/ani14152254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
To meet the demand of consumers for chicken products, poultry breeders have made improvements to chickens. However, this has led to a new problem in the modern poultry industry, namely excessive fat deposition. This study aims to understand the effects of dietary iron supplementation on fat deposition and gut microbiota in chickens. In this study, we investigated the effects of iron on the growth performance, fat deposition, and gut microbiota of silky fowl black-bone chickens. A total of 75 7-week-old silky fowl black-bone chickens were randomly divided into three groups (five replicates per group, five chickens per replicate) and fed them for 28 days using a growing diet (control group), a growing diet + 10% tallow (high-fat diet group, HFD group), and a growing diet + 10% tallow + 500 mg/kg iron (HFDFe500 group), respectively. We detected the effects of iron on the growth performance, fat deposition, and gut microbiota of silky fowl black-bone chickens using the growth performance index test, oil red O staining, and HE staining, and found that the high-fat diet significantly increased liver and serum fat deposition and liver injury, while the addition of iron to the diet could reduce the fat deposition caused by the high-fat diet and alleviate liver injury. In addition, 16S rDNA sequencing was used to compare the relative abundance of gut microbiota in the cecal contents in different feeding groups. The results showed that the high-fat diet could induce gut microbiota imbalance in chickens, while the high-iron diet reversed the gut microbiota imbalance. PICRUSt functional prediction analysis showed that dietary iron supplementation affected amino acid metabolism, energy metabolism, cofactors, and vitamin metabolism pathways. In addition, correlation analysis showed that TG was significantly associated with Firmicutes and Actinobacteriota (p < 0.05). Overall, these results revealed high dietary iron (500 mg/kg) could reduce fat deposition and affect the gut microbiota of silky fowl black-bone chickens, suggesting that iron may regulate fat deposition by influencing the gut microbiota of chickens and provides a potential avenue that prevents excessive fat deposition in chickens by adding iron to the diet.
Collapse
Affiliation(s)
- Ting Yang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Shihao Chen
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Lingling Qiu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Qixin Guo
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Zhixiu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Yong Jiang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Hao Bai
- Institutes of Agricultural Science and Technology Development, Yangzhou University, Yangzhou 225009, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Yulin Bi
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| | - Guobin Chang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China
- Key Laboratory for Animal Genetics & Molecular Breeding of Jiangsu Province, Yangzhou University, Yangzhou 225009, China
| |
Collapse
|
8
|
Yang W, Arora M, Han HW, Jiang W, Kim DM, Ai W, Pan Q, Kumar MNVR, Brashear WA, Sun Y, Guo S. ZnPP-laden nanoparticles improve glucose homeostasis and chronic inflammation during obesity. Br J Pharmacol 2024; 181:2886-2904. [PMID: 38679457 DOI: 10.1111/bph.16356] [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: 07/18/2023] [Revised: 01/16/2024] [Accepted: 02/09/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND AND PURPOSE Chronic inflammation plays a pivotal role in the development of Type 2 diabetes mellitus (T2DM). Previous studies have shown that haem oxygenase-1 (HO-1) plays a proinflammatory role during metabolic stress, suggesting that HO-1 inhibition could be an effective strategy to treat T2DM. However, the application of HO-1 inhibitors is restricted due to solubility-limited bioavailability. In this study, we encapsulated the HO-1 inhibitor, zinc protoporphyrin IX (ZnPP), within nanoparticles and investigated their role in regulating glucose homeostasis and chronic inflammation during obesity. EXPERIMENTAL APPROACH We delivered DMSO-dissolved ZnPP (DMSO-ZnPP) and ZnPP-laden nanoparticles (Nano-ZnPP) to diet-induced obese male mice for 6 weeks. Glucose and insulin tolerance tests were carried out, liver and adipose tissue gene expression profiles analysed, and systemic inflammation analysed using flow cytometry. KEY RESULTS Nanoparticles significantly increased the delivery efficiency of ZnPP in both cells and mice. In mice with diet-induced obesity, inhibition of HO-1 by Nano-ZnPP significantly decreased adiposity, increased insulin sensitivity, and improved glucose tolerance. Moreover, Nano-ZnPP treatment attenuated both local and systemic inflammatory levels during obesity. Mechanistically, Nano-ZnPP significantly attenuated glucagon, TNF, and fatty acid synthesis signalling pathways in the liver. In white adipose tissue, the oxidative phosphorylation signalling pathway was enhanced and the inflammation signalling pathway diminished by Nano-ZnPP. Our results show that Nano-ZnPP has better effects on the improvement of glucose homeostasis and attenuation of chronic inflammation, than those of DMSO-dissolved ZnPP. CONCLUSIONS AND IMPLICATIONS These findings indicate that ZnPP-laden nanoparticles are potential therapeutic agents for treating T2DM.
Collapse
Affiliation(s)
- Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Meenakshi Arora
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama, USA
| | - Hye Won Han
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Wen Jiang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Da Mi Kim
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - M N V Ravi Kumar
- College of Community Health Sciences, The University of Alabama, Tuscaloosa, Alabama, USA
- The Center for Convergent Bioscience and Medicine (CCBM), The University of Alabama, Tuscaloosa, Alabama, USA
| | - Wesley A Brashear
- High Performance Research Computing, Texas A&M University, College Station, Texas, USA
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, Texas, USA
| |
Collapse
|
9
|
Schleh MW, Ameka M, Rodriguez A, Hasty AH. Deficiency of the hemoglobin-haptoglobin receptor, CD163, worsens insulin sensitivity in obese male mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.31.596887. [PMID: 38895370 PMCID: PMC11185572 DOI: 10.1101/2024.05.31.596887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Excessive iron accumulation in metabolic organs such as the adipose tissue, liver, and skeletal muscle is associated with increased diabetes risk. Tissue-resident macrophages serve multiple roles including managing inflammatory tone and regulating parachymal iron homeostasis; thus protecting against metabolic dysfunction upon iron overload. The scavenger receptor CD163 is uniquely present on tissue-resident macrophages, and plays a significant role in iron homeostasis by clearing extracellular hemoglobin-haptoglobin complexes, thereby limiting oxidative damage caused by free hemoglobin in metabolic tissues. We show that the absence of CD163 exacerbates glucose intolerance and insulin resistance in male mice with obesity. Additionally, loss of CD163 reduced the expression of iron regulatory genes (Tfr1, Cisd1, Slc40a1) in adipose tissue macrophages and anti-inflammatory (M2-like) bone marrow-derived macrophages (BMDMs). Further, CD163 deficiency mediated a pro-inflammatory shift and limited hemoglobin scavenging specifically in M2-like BMDMs. To this end, iron buffering was diminished in inguinal white adipose tissue (iWAT) macrophages in vivo, which culminated in iron spillover into adipocytes and CD45+CD11B- non-myeloid immune cells in iWAT. These findings show that CD163 on tissue-resident macrophages is critical for their anti-inflammatory and hemoglobin scavenging roles, and its absence results in impaired systemic insulin action in an obese setting.
Collapse
Affiliation(s)
- Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Magdalene Ameka
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Alec Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine; Nashville, TN 37232, USA
- VA Tennessee Valley Healthcare System; Nashville, TN 37212, USA
| |
Collapse
|
10
|
do Nascimento FV, de Freitas BS, Dos Passos MP, Kleverston L, de Souza Dos Santos C, Kist LW, Bogo MR, Bromberg E, Schröder N. A high fat diet potentiates neonatal iron overload-induced memory impairments in rats. Eur J Nutr 2024; 63:1163-1175. [PMID: 38358514 DOI: 10.1007/s00394-024-03333-x] [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: 06/06/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE The present study aimed at evaluating possible synergistic effects between two risk factors for cognitive decline and neurodegenerative disorders, i.e. iron overload and exposure to a hypercaloric/hyperlipidic diet, on cognition, insulin resistance, and hippocampal GLUT1, GLUT3, Insr mRNA expression, and AKT phosporylation. METHODS Male Wistar rats were treated with iron (30 mg/kg carbonyl iron) or vehicle (5% sorbitol in water) from 12 to 14th post-natal days. Iron-treated rats received a standard laboratory diet or a high fat diet from weaning to adulthood (9 months of age). Recognition and emotional memory, peripheral blood glucose and insulin levels were evaluated. Glucose transporters (GLUT 1 and GLUT3) and insulin signaling were analyzed in the hippocampus of rats. RESULTS Both iron overload and exposure to a high fat diet induced memory deficits. Remarkably, the association of iron with the high fat diet induced more severe cognitive deficits. Iron overload in the neonatal period induced higher insulin levels associated with significantly higher HOMA-IR, an index of insulin resistance. Long-term exposure to a high fat diet resulted in higher fasting glucose levels. Iron treatment induced changes in Insr and GLUT1 expression in the hippocampus. At the level of intracellular signaling, both iron treatment and the high fat diet decreased AKT phosphorylation. CONCLUSION The combination of iron overload with exposure to a high fat diet only led to synergistic deleterious effect on emotional memory, while the effects induced by iron and by the high fat diet on AKT phosphorylation were comparable. These findings indicate that there is, at least to some extent, an additive effect of iron combined with the diet. Further studies investigating the mechanisms associated to deleterious effects on cognition and susceptibility for the development of age-associated neurodegenerative disorders are warranted.
Collapse
Affiliation(s)
- Filipe Valvassori do Nascimento
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Betânia Souza de Freitas
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Maiara Priscila Dos Passos
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiza Kleverston
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristophod de Souza Dos Santos
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Luiza Wilges Kist
- Laboratory of Genomics and Molecular Biology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Excitotoxicity and Neuroprotection (INCT-EN), National Institute of Science and Technology for Brain Diseases, Porto Alegre, Brazil
| | - Maurício Reis Bogo
- Laboratory of Genomics and Molecular Biology, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- Excitotoxicity and Neuroprotection (INCT-EN), National Institute of Science and Technology for Brain Diseases, Porto Alegre, Brazil
| | - Elke Bromberg
- Neurobiology and Developmental Biology Laboratory, School of Health and Life Sciences, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil
| | - Nadja Schröder
- National Institute of Science and Technology for Translational Medicine (INCT-TM), Conselho Nacional de Desenvolvimento Cientifico e Tecnologico (CNPq), Brasília, Brazil.
- Department of Physiology, Institute for Basic Health Sciences, Federal University of Rio Grande do Sul, Rua Ramiro Barcelos, 2600, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
11
|
Aguree S, Owora A, Hawkins M, Gletsu-Miller N. Obesity modifies the association between diabetes and iron biomarkers and red cell indices in reproductive-aged women in the United States. J Investig Med 2024; 72:425-437. [PMID: 38445643 DOI: 10.1177/10815589241240059] [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] [Indexed: 03/07/2024]
Abstract
Obesity and diabetes are associated with impaired iron metabolism. We aimed to examine the independent relationship between diabetes and iron after controlling for body weight (or obesity) in women aged 20-49 years. The National Health and Nutrition Examination Survey data from 2015 to 2018 were used in this investigation. Body composition data, HbAc1, iron biomarkers (serum ferritin (SF), soluble transferrin receptor (sTfR), and body iron index (BII)), mean corpuscular volume (MCV), mean hemoglobin concentration (MCH), red cell distribution width (RDW), and hemoglobin were used. Linear regression models were used to examine how and to what extent body mass index (BMI) modified the relationship between diabetes and iron status biomarkers. A total of 1834 women aged 20-49 were included in the analysis with a mean (SD) age of 32 .2 ± 6.1 years and BMI of 29.5 ± 6.9 kg/m2. The mean SF (p = 0.014) and BII (p < 0.001) were lower, while sTfR (p < 0.001) was higher in women with diabetes than those with no diabetes. Mean estimates for MCV and MCH were lower, while RDW (p = 0.001) was higher in diabetes patients (all p < 0.001). Women with diabetes were more likely to have iron deficiency, anemia, and iron deficiency anemia than those without diabetes (18.1% vs 8.6%, p < 0.001), (24.4% vs 8.4%, p < 0.001), and (14.8% vs 5.2%, p < 0.001), respectively. Among women with obesity, those with diabetes had lower predicted ferritin (β = -0.19, p = 0.016), BII (β = -0.99, p = 0.016), and hemoglobin (β = -0.27, p = 0.042) than those without diabetes. The study shows that diabetes is linked to lower iron stores; this is exacerbated in those with obesity.
Collapse
Affiliation(s)
- Sixtus Aguree
- Department of Applied Health Science, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Arthur Owora
- Department of Pediatrics, School of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Indiana University, Bloomington, IN, USA
| | - Misty Hawkins
- Department of Health and Wellness Design, School of Public Health, Indiana University, Bloomington, IN, USA
| | - Nana Gletsu-Miller
- Department of Applied Health Science, Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| |
Collapse
|
12
|
Chen Y, Zhao W, Hu A, Lin S, Chen P, Yang B, Fan Z, Qi J, Zhang W, Gao H, Yu X, Chen H, Chen L, Wang H. Type 2 diabetic mellitus related osteoporosis: focusing on ferroptosis. J Transl Med 2024; 22:409. [PMID: 38693581 PMCID: PMC11064363 DOI: 10.1186/s12967-024-05191-x] [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: 01/21/2024] [Accepted: 04/12/2024] [Indexed: 05/03/2024] Open
Abstract
With the aging global population, type 2 diabetes mellitus (T2DM) and osteoporosis(OP) are becoming increasingly prevalent. Diabetic osteoporosis (DOP) is a metabolic bone disorder characterized by abnormal bone tissue structure and reduced bone strength in patients with diabetes. Studies have revealed a close association among diabetes, increased fracture risk, and disturbances in iron metabolism. This review explores the concept of ferroptosis, a non-apoptotic cell death process dependent on intracellular iron, focusing on its role in DOP. Iron-dependent lipid peroxidation, particularly impacting pancreatic β-cells, osteoblasts (OBs) and osteoclasts (OCs), contributes to DOP. The intricate interplay between iron dysregulation, which comprises deficiency and overload, and DOP has been discussed, emphasizing how excessive iron accumulation triggers ferroptosis in DOP. This concise overview highlights the need to understand the complex relationship between T2DM and OP, particularly ferroptosis. This review aimed to elucidate the pathogenesis of ferroptosis in DOP and provide a prospective for future research targeting interventions in the field of ferroptosis.
Collapse
Affiliation(s)
- Yili Chen
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wen Zhao
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - An Hu
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - Shi Lin
- Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - Ping Chen
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Bing Yang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Zhirong Fan
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ji Qi
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Wenhui Zhang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Huanhuan Gao
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Xiubing Yu
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Haiyun Chen
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Luyuan Chen
- Stomatology Center, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, 510086, China.
| | - Haizhou Wang
- Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China.
| |
Collapse
|
13
|
Lu B, Guo S, Zhao J, Wang X, Zhou B. Adipose knockout of H-ferritin improves energy metabolism in mice. Mol Metab 2024; 80:101871. [PMID: 38184276 PMCID: PMC10803945 DOI: 10.1016/j.molmet.2024.101871] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 12/12/2023] [Accepted: 01/02/2024] [Indexed: 01/08/2024] Open
Abstract
OBJECTIVE Ferritin, the principal iron storage protein, is essential to iron homeostasis. How iron homeostasis affects the adipose tissue is not well understood. We investigated the role of ferritin heavy chain in adipocytes in energy metabolism. METHODS We generated adipocyte-specific ferritin heavy chain (Fth, also known as Fth1) knockout mice, herein referred to as FthAKO. These mice were analyzed for iron homeostasis, oxidative stress, mitochondrial biogenesis and activity, adaptive thermogenesis, insulin sensitivity, and metabolic measurements. Mouse embryonic fibroblasts and primary mouse adipocytes were used for in vitro experiments. RESULTS In FthAKO mice, the adipose iron homeostasis was disrupted, accompanied by elevated expression of adipokines, dramatically induced heme oxygenase 1(Hmox1) expression, and a notable decrease in the mitochondrial ROS level. Cytosolic ROS elevation in the adipose tissue of FthAKO mice was very mild, and we only observed this in the brown adipose tissue (BAT) but not in the white adipose tissue (WAT). FthAKO mice presented an altered metabolic profile and showed increased insulin sensitivity, glucose tolerance, and improved adaptive thermogenesis. Interestingly, loss of ferritin resulted in enhanced mitochondrial respiration capacity and a preference for lipid metabolism. CONCLUSIONS These findings indicate that ferritin in adipocytes is indispensable to intracellular iron homeostasis and regulates systemic lipid and glucose metabolism.
Collapse
Affiliation(s)
- Binyu Lu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Shanshan Guo
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Jialin Zhao
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoting Wang
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Bing Zhou
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; Faculty of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| |
Collapse
|
14
|
Moreno-Navarrete JM, Fernández-Real JM. Iron: The silent culprit in your adipose tissue. Obes Rev 2024; 25:e13647. [PMID: 37789591 DOI: 10.1111/obr.13647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/27/2023] [Accepted: 09/09/2023] [Indexed: 10/05/2023]
Abstract
Iron plays a vital role in essential biological processes and requires precise regulation within the body. Dysregulation of iron homeostasis, characterized by increased serum ferritin levels and excessive accumulation of iron in the liver, adipose tissue, and skeletal muscle, is associated with obesity and insulin resistance. Notably, iron excess in adipose tissue promotes adipose tissue dysfunction. As optimal adipose tissue function is crucial for maintaining a healthy phenotype in obesity, a comprehensive understanding of iron homeostasis in adipose tissue is imperative for designing new therapeutic approaches to improve and prevent adipose tissue dysfunction. Here, we conducted a review of relevant studies, focusing on and providing valuable insights into the intricate interplay between iron and adipose tissue. It sheds light on the impact of iron on adipogenesis and the physiology of both white and brown adipose tissue. Furthermore, we highlight the critical role of key modulators, such as cytosolic aconitase, mitochondria, and macrophages, in maintaining iron homeostasis within adipose tissue.
Collapse
Affiliation(s)
- José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), Girona, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Medical Sciences, University of Girona, Girona, Spain
| |
Collapse
|
15
|
Vinokur V, Berenshtein E, Chevion M, Chevion D. A New Concept in Antidiabetic Therapeutics: A Concerted Removal of Labile Iron and Intracellular Deposition of Zinc. Diabetes Metab J 2024; 48:59-71. [PMID: 38173374 PMCID: PMC10850271 DOI: 10.4093/dmj.2022.0292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 04/10/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGRUOUND The inflammatory process is known to be an integral part of the pathophysiology of type 2 diabetes mellitus (T2DM). The "labile," redox-active iron, serving as a catalyst in Fenton reaction, producing the deleterious reactive oxygen species, triggering and maintaining inflammation, is hypothesized to play a causative role in this process. Concenter Biopharma continued the development of a new platform of iron chelators (Zygosids), first initiated at the Hebrew University of Jerusalem, Israel (HUJI), acting via the novel mechanism, based on a sequestration of the labile redox-active iron and its substitution by zinc or gallium. The mode of action of Zygosids is based on the higher affinity of the metal-binding moiety of the complex to Fe3+ in comparison to already bound ion, leading to rapid release of the ion of another metal and chelation of Fe3+. Concomitantly, zinc ion, released by the complex, is known for its antidiabetic and anti-inflammatory role. METHODS The therapeutic effect of zinc-desferrioxamine (Zygosid-50) and gallium-desferrioxamine, was tested on fat sand rat (Psammomys obesus) model of diet-induced T2DM and on Leprdb transgenic diabetic mice. RESULTS Zygosids demonstrated an ability to noticeably reduce blood glucose and insulin levels and improve the lipid profile. Moreover, an ability to mitigate insulin resistance by >90% was shown on the sand rat model. In addition, a potent anti-inflammatory effect, expressed as a diminishment of the proinflammatory cytokines in tissue levels, was demonstrated. CONCLUSION Zygosids demonstrated robust therapeutic efficacy in treatment of T2DM. Importantly, no adverse effects were detected, in all the experiments, indicating high safety profile.
Collapse
Affiliation(s)
- Vladimir Vinokur
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem (HUJI), Jerusalem, Israel
- Concenter Biopharma, Jerusalem, Israel
| | - Eduard Berenshtein
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem (HUJI), Jerusalem, Israel
| | - Mordechai Chevion
- Department of Biochemistry and Molecular Biology, Institute of Medical Research Israel-Canada, The Hebrew University of Jerusalem (HUJI), Jerusalem, Israel
| | | |
Collapse
|
16
|
Babayev M, Klaunig J, Silveyra P, Henschel B, Gletsu-Miller N. Impact on oxidative stress of oral, high-dose, iron supplementation for management of iron deficiency after bariatric surgery, a preliminary study. J Trace Elem Med Biol 2023; 80:127310. [PMID: 37801789 DOI: 10.1016/j.jtemb.2023.127310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 10/08/2023]
Abstract
OBJECTIVES High-dose oral iron supplementation for patients who develop iron deficiency after bariatric surgery may induce oxidative stress in the gastrointestine. The study's objective was to test this hypothesis by determining the impact of high-dose oral iron on systemic oxidative stress. METHODS We used archived plasma samples from a randomized controlled clinical trial (NCT02404012) comparing FeSO4 (195 mg/day, NatureMade®, West Hills, CA) with a heme iron polypeptide (HIP, 60.4 mg/day, Proferrin®, Colorado Biolabs, Lafayette, CO) for 8 weeks. Systemic oxidative stress was measured using malondialdehyde and total antioxidant capacity (MDA, Abcam, ab238537 and TAC, Abcam, ab65329 Cambridge, UK) assays. Data was log-transformed and presented as means and standard deviations; a mixed model was used to determine the effects of time (0, 2, 4, and 8 weeks) and treatment (FeSO4 versus HIP) on oxidative stress. RESULTS The FeSO4 (N = 8) and HIP (N = 5) participants were balanced in body mass index (35.0 ± 5.5 kg/m2), race (93 % White), time post-surgery (7.3 ± 3.3 years), as well as serum concentrations of iron (P > 0.05). The FeSO4 group tended to be older (44.3 ± 4.5 years) and they had lower concentrations of serum ferritin (6.5 ± 2.7 µg/mL) than the HIP (38.2 ± 9.3 years, and 12.9 ± 16.8 µg/mL) group (P = 0.080, and P = 0.017 respectively). We observed a larger increase in serum iron in the FeSO4 group during the 8 weeks of Fe supplementation, compared to that in the HIP group (p = 0.004). We observed a decreasing trend in MDA over the 8 weeks (p = 0.080) in the FeSO4 treatment group. There were no significant differences in TAC between and within FeSO4 and HIP groups over the 8 week supplementation period. CONCLUSIONS This preliminary study suggests that high-dose oral iron supplementation for iron deficiency does not adversely impact systemic oxidative stress in patients undergoing bariatric surgery.
Collapse
Affiliation(s)
- Maksat Babayev
- Department of Environmental and Occupational Health, Indiana University School of Public Health Bloomington, 1025 E 7th Street, Bloomington, IN 47405, United States
| | - James Klaunig
- Department of Environmental and Occupational Health, Indiana University School of Public Health Bloomington, 1025 E 7th Street, Bloomington, IN 47405, United States
| | - Patricia Silveyra
- Department of Environmental and Occupational Health, Indiana University School of Public Health Bloomington, 1025 E 7th Street, Bloomington, IN 47405, United States
| | - Beate Henschel
- Department of Epidemiology and Biostatistics, Indiana University School of Public Health Bloomington, 2719 E. 10th Street, Bloomington, IN 47405, United States
| | - Nana Gletsu-Miller
- Department of Applied Health Science, Indiana University School of Public Health Bloomington, 1025 E 7th Street, Bloomington, IN 47405, United States.
| |
Collapse
|
17
|
Zhu B, Wei Y, Zhang M, Yang S, Tong R, Li W, Long E. Metabolic dysfunction-associated steatotic liver disease: ferroptosis related mechanisms and potential drugs. Front Pharmacol 2023; 14:1286449. [PMID: 38027027 PMCID: PMC10665502 DOI: 10.3389/fphar.2023.1286449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/24/2023] [Indexed: 12/01/2023] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is considered a "multisystem" disease that simultaneously suffers from metabolic diseases and hepatic steatosis. Some may develop into liver fibrosis, cirrhosis, and even hepatocellular carcinoma. Given the close connection between metabolic diseases and fatty liver, it is urgent to identify drugs that can control metabolic diseases and fatty liver as a whole and delay disease progression. Ferroptosis, characterized by iron overload and lipid peroxidation resulting from abnormal iron metabolism, is a programmed cell death mechanism. It is an important pathogenic mechanism in metabolic diseases or fatty liver, and may become a key direction for improving MASLD. In this article, we have summarized the physiological and pathological mechanisms of iron metabolism and ferroptosis, as well as the connections established between metabolic diseases and fatty liver through ferroptosis. We have also summarized MASLD therapeutic drugs and potential active substances targeting ferroptosis, in order to provide readers with new insights. At the same time, in future clinical trials involving subjects with MASLD (especially with the intervention of the therapeutic drugs), the detection of serum iron metabolism levels and ferroptosis markers in patients should be increased to further explore the efficacy of potential drugs on ferroptosis.
Collapse
Affiliation(s)
- Baoqiang Zhu
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Yuankui Wei
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingming Zhang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shiyu Yang
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Rongsheng Tong
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Wenyuan Li
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Enwu Long
- Department of Pharmacy, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
- School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
18
|
Oliveras-Cañellas N, Latorre J, Santos-González E, Lluch A, Ortega F, Mayneris-Perxachs J, Fernández-Real JM, Moreno-Navarrete JM. Inflammatory response to bacterial lipopolysaccharide drives iron accumulation in human adipocytes. Biomed Pharmacother 2023; 166:115428. [PMID: 37677967 DOI: 10.1016/j.biopha.2023.115428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
The association among increased inflammation, disrupted iron homeostasis, and adipose tissue dysfunction in obesity has been widely recognized. However, the specific impact of inflammation on iron homeostasis during human adipogenesis and in adipocytes remains poorly understood. In this study, we investigated the effects of bacterial lipopolysaccharide (LPS) on iron homeostasis during human adipocyte differentiation, in fully differentiated adipocytes, and in human adipose tissue. We found that LPS-induced inflammation hindered adipogenesis and led to a gene expression profile indicative of intracellular iron accumulation. This was accompanied by increased expression of iron importers (TFRC and SLC11A2), markers of intracellular iron accumulation (FTH, CYBA, FTL, and LCN2), and decreased expression of iron exporter-related genes (SLC40A1), concomitant with elevated intracellular iron levels. Mechanistically, RNA-seq analysis and gene knockdown experiments revealed the significant involvement of iron importers SLC39A14, SLC39A8, and STEAP4 in LPS-induced intracellular iron accumulation in human adipocytes. Notably, markers of LPS signaling pathway-related inflammation were also associated with a gene expression pattern indicative of intracellular iron accumulation in human adipose tissue, corroborating the link between LPS-induced inflammation and iron accumulation at the tissue level. In conclusion, our findings demonstrate that induction of adipocyte inflammation disrupts iron homeostasis, resulting in adipocyte iron overload.
Collapse
Affiliation(s)
- Núria Oliveras-Cañellas
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - Jessica Latorre
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - Elena Santos-González
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - Aina Lluch
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - Francisco Ortega
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - Jordi Mayneris-Perxachs
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - José-Manuel Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain; Department of Medicine, Universitat de Girona, Girona, Spain.
| | - José María Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain.
| |
Collapse
|
19
|
Kong M, Zhong C, Gao Q, Zhou X, Chen R, Xiong G, Hao L, Yang X, Lu Z, Yang N. Association of elevated mid-pregnancy maternal plasma ferritin concentrations and triglyceride concentrations with the risk of gestational diabetes mellitus: A prospective cohort study. Diabetes Metab Res Rev 2023; 39:e3637. [PMID: 36958940 DOI: 10.1002/dmrr.3637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/18/2023] [Accepted: 02/13/2023] [Indexed: 03/25/2023]
Abstract
OBJECTIVE Ferritin levels are well known to be associated with gestational diabetes mellitus (GDM). However, the association of the combination of ferritin and triglyceride (TG) levels in early mid-pregnancy with GDM has not been studied in depth. We investigated the independent and combined relationships of plasma ferritin and TG concentrations with the risk of GDM as well as the mediation effect of TG on ferritin. METHODS We analysed 2071 pregnant women from the Tongji Maternal and Child Health Cohort who had their plasma ferritin and TG concentrations measured at 11-20 weeks of gestation. Associations between ferritin and TG concentrations and GDM risk were estimated using multivariable logistic regression models. Youden's index was calculated to find the cut-off values of ferritin and TG by ROC curve analysis. The mediation effect of the TG concentration on the ferritin level with GDM risk was explored by a mediation analysis. RESULTS A total of 264 (12.3%) participants developed GDM. The median and IQR of ferritin was 53.9 (30.5-92.7) ng/mL. After adjusting for potential confounders, the relative risks (RRs) and 95% confidence intervals of GDM were 2.19 (1.42, 3.39) for ferritin and 2.02 (1.37, 2.97) for TG. The adjusted RR for combination was 2.40 (1.62, 3.55). Moreover, we found that the TG concentration mediated 15.0% of the total effect of the ferritin concentration on the risk of GDM. CONCLUSIONS Women with a combination of both high plasma ferritin (˃55.7 ng/mL) and high TG (˃1.9 mmoL/L) were at the highest risk of GDM. Additionally, we have revealed for the first time that an elevated maternal TG concentration in early pregnancy mediates the relationship between ferritin concentration and GDM risk. TRIAL REGISTRATION This trial is registered at https://ClinicalTrials.gov as NCT03099837.
Collapse
Affiliation(s)
- Man Kong
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Chunrong Zhong
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qin Gao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuezhen Zhou
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Renjuan Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Guoping Xiong
- Department of Obstetrics, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Liping Hao
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xuefeng Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhongxin Lu
- Department of Medical Laboratory, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Nianhong Yang
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
20
|
Hilton C, Sabaratnam R, Drakesmith H, Karpe F. Iron, glucose and fat metabolism and obesity: an intertwined relationship. Int J Obes (Lond) 2023; 47:554-563. [PMID: 37029208 PMCID: PMC10299911 DOI: 10.1038/s41366-023-01299-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/08/2023] [Accepted: 03/16/2023] [Indexed: 04/09/2023]
Abstract
A bidirectional relationship exists between adipose tissue metabolism and iron regulation. Total body fat, fat distribution and exercise influence iron status and components of the iron-regulatory pathway, including hepcidin and erythroferrone. Conversely, whole body and tissue iron stores associate with fat mass and distribution and glucose and lipid metabolism in adipose tissue, liver, and muscle. Manipulation of the iron-regulatory proteins erythroferrone and erythropoietin affects glucose and lipid metabolism. Several lines of evidence suggest that iron accumulation and metabolism may play a role in the development of metabolic diseases including obesity, type 2 diabetes, hyperlipidaemia and non-alcoholic fatty liver disease. In this review we summarise the current understanding of the relationship between iron homoeostasis and metabolic disease.
Collapse
Affiliation(s)
- Catriona Hilton
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
| | - Rugivan Sabaratnam
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- Steno Diabetes Center Odense, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| |
Collapse
|
21
|
Wu Q, Li S, Zhang X, Dong N. Type II Transmembrane Serine Proteases as Modulators in Adipose Tissue Phenotype and Function. Biomedicines 2023; 11:1794. [PMID: 37509434 PMCID: PMC10376093 DOI: 10.3390/biomedicines11071794] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Adipose tissue is a crucial organ in energy metabolism and thermoregulation. Adipose tissue phenotype is controlled by various signaling mechanisms under pathophysiological conditions. Type II transmembrane serine proteases (TTSPs) are a group of trypsin-like enzymes anchoring on the cell surface. These proteases act in diverse tissues to regulate physiological processes, such as food digestion, salt-water balance, iron metabolism, epithelial integrity, and auditory nerve development. More recently, several members of the TTSP family, namely, hepsin, matriptase-2, and corin, have been shown to play a role in regulating lipid metabolism, adipose tissue phenotype, and thermogenesis, via direct growth factor activation or indirect hormonal mechanisms. In mice, hepsin deficiency increases adipose browning and protects from high-fat diet-induced hyperglycemia, hyperlipidemia, and obesity. Similarly, matriptase-2 deficiency increases fat lipolysis and reduces obesity and hepatic steatosis in high-fat diet-fed mice. In contrast, corin deficiency increases white adipose weights and cell sizes, suppresses adipocyte browning and thermogenic responses, and causes cold intolerance in mice. These findings highlight an important role of TTSPs in modifying cellular phenotype and function in adipose tissue. In this review, we provide a brief description about TTSPs and discuss recent findings regarding the role of hepsin, matriptase-2, and corin in regulating adipose tissue phenotype, energy metabolism, and thermogenic responses.
Collapse
Affiliation(s)
- Qingyu Wu
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Shuo Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Xianrui Zhang
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
| | - Ningzheng Dong
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Prevention, Soochow University, Suzhou 215123, China
- NHC Key Laboratory of Thrombosis and Hemostasis, Jiangsu Institute of Hematology, Soochow University, Suzhou 215006, China
| |
Collapse
|
22
|
Valenti L, Corradini E, Adams LA, Aigner E, Alqahtani S, Arrese M, Bardou-Jacquet E, Bugianesi E, Fernandez-Real JM, Girelli D, Hagström H, Henninger B, Kowdley K, Ligabue G, McClain D, Lainé F, Miyanishi K, Muckenthaler MU, Pagani A, Pedrotti P, Pietrangelo A, Prati D, Ryan JD, Silvestri L, Spearman CW, Stål P, Tsochatzis EA, Vinchi F, Zheng MH, Zoller H. Consensus Statement on the definition and classification of metabolic hyperferritinaemia. Nat Rev Endocrinol 2023; 19:299-310. [PMID: 36805052 PMCID: PMC9936492 DOI: 10.1038/s41574-023-00807-6] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/19/2023]
Abstract
Hyperferritinaemia is a common laboratory finding that is often associated with metabolic dysfunction and fatty liver. Metabolic hyperferritinaemia reflects alterations in iron metabolism that facilitate iron accumulation in the body and is associated with an increased risk of cardiometabolic and liver diseases. Genetic variants that modulate iron homeostasis and tissue levels of iron are the main determinants of serum levels of ferritin in individuals with metabolic dysfunction, raising the hypothesis that iron accumulation might be implicated in the pathogenesis of insulin resistance and the related organ damage. However, validated criteria for the non-invasive diagnosis of metabolic hyperferritinaemia and the staging of iron overload are still lacking, and there is no clear evidence of a benefit for iron depletion therapy. Here, we provide an overview of the literature on the relationship between hyperferritinaemia and iron accumulation in individuals with metabolic dysfunction, and on the associated clinical outcomes. We propose an updated definition and a provisional staging system for metabolic hyperferritinaemia, which has been agreed on by a multidisciplinary global panel of expert researchers. The goal is to foster studies into the epidemiology, genetics, pathophysiology, clinical relevance and treatment of metabolic hyperferritinaemia, for which we provide suggestions on the main unmet needs, optimal design and clinically relevant outcomes.
Collapse
Affiliation(s)
- Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Biological Resource Center and Precision Medicine Lab, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
| | - Elena Corradini
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy.
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy.
| | - Leon A Adams
- Medical School, University of Western Australia, Perth, Australia
| | - Elmar Aigner
- First Department of Medicine, University Clinic Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Saleh Alqahtani
- Royal Clinics and Gastroenterology and Hepatology, King Faisal Specialist Hospital & Research Centre, Riyadh, Kingdom of Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Edouard Bardou-Jacquet
- University of Rennes, UMR1241, CHU Rennes, National Reference Center for Hemochromatosis and iron metabolism disorder, INSERM CIC1414, Rennes, France
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Turin, Italy
| | - Jose-Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Domenico Girelli
- Section of Internal Medicine, Department of Medicine, University of Verona, Policlinico Giambattista Rossi, Verona, Italy
| | - Hannes Hagström
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kris Kowdley
- Liver Institute Northwest, Seattle, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| | - Guido Ligabue
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Division of Radiology, Ospedale di Sassuolo S.p.A, Sassuolo, Modena, Italy
| | - Donald McClain
- Wake Forest School of Medicine, Winston Salem, NC, USA
- Department of Veterans Affairs, Salisbury, NC, USA
| | - Fabrice Lainé
- INSERM CIC1414, Liver Unit, CHU Rennes, Rennes, France
| | - Koji Miyanishi
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Martina U Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
- Center for Molecular Translational Iron Research, Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Heidelberg, Germany
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Pedrotti
- Laboratorio di RM Cardiaca Cardiologia 4, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy
| | - Daniele Prati
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - John D Ryan
- Hepatology Unit, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - C Wendy Spearman
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Per Stål
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley F.Kimball Research Institute, New York Blood Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Heinz Zoller
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
- Doppler Laboratory on Iron and Phosphate Biology, Innsbruck, Austria
| |
Collapse
|
23
|
Iron metabolism and ferroptosis in type 2 diabetes mellitus and complications: mechanisms and therapeutic opportunities. Cell Death Dis 2023; 14:186. [PMID: 36882414 PMCID: PMC9992652 DOI: 10.1038/s41419-023-05708-0] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/18/2023] [Accepted: 02/22/2023] [Indexed: 03/09/2023]
Abstract
The maintenance of iron homeostasis is essential for proper endocrine function. A growing body of evidence suggests that iron imbalance is a key factor in the development of several endocrine diseases. Nowadays, ferroptosis, an iron-dependent form of regulated cell death, has become increasingly recognized as an important process to mediate the pathogenesis and progression of type 2 diabetes mellitus (T2DM). It has been shown that ferroptosis in pancreas β cells leads to decreased insulin secretion; and ferroptosis in the liver, fat, and muscle induces insulin resistance. Understanding the mechanisms concerning the regulation of iron metabolism and ferroptosis in T2DM may lead to improved disease management. In this review, we summarized the connection between the metabolic pathways and molecular mechanisms of iron metabolism and ferroptosis in T2DM. Additionally, we discuss the potential targets and pathways concerning ferroptosis in treating T2DM and analysis the current limitations and future directions concerning these novel T2DM treatment targets.
Collapse
|
24
|
Hinojosa-Moscoso A, Motger-Albertí A, De la Calle-Vargas E, Martí-Navas M, Biarnés C, Arnoriaga-Rodríguez M, Blasco G, Puig J, Luque-Córdoba D, Priego-Capote F, Moreno-Navarrete JM, Fernández-Real JM. The Longitudinal Changes in Subcutaneous Abdominal Tissue and Visceral Adipose Tissue Volumetries Are Associated with Iron Status. Int J Mol Sci 2023; 24:4750. [PMID: 36902180 PMCID: PMC10002479 DOI: 10.3390/ijms24054750] [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: 01/09/2023] [Revised: 02/20/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
Excess iron is known to trigger adipose tissue dysfunction and insulin resistance. Circulating markers of iron status have been associated with obesity and adipose tissue in cross-sectional studies. We aimed to evaluate whether iron status is linked to changes in abdominal adipose tissue longitudinally. Subcutaneous abdominal tissue (SAT) and visceral adipose tissue (VAT) and its quotient (pSAT) were assessed using magnetic resonance imaging (MRI), at baseline and after one year of follow-up, in 131 (79 in follow-up) apparently healthy subjects, with and without obesity. Insulin sensitivity (euglycemic- hyperinsulinemic clamp) and markers of iron status were also evaluated. Baseline serum hepcidin (p = 0.005 and p = 0.002) and ferritin (p = 0.02 and p = 0.01)) were associated with an increase in VAT and SAT over one year in all subjects, while serum transferrin (p = 0.01 and p = 0.03) and total iron-binding capacity (p = 0.02 and p = 0.04) were negatively associated. These associations were mainly observed in women and in subjects without obesity, and were independent of insulin sensitivity. After controlling for age and sex, serum hepcidin was significantly associated with changes in subcutaneous abdominal tissue index (iSAT) (β = 0.406, p = 0.007) and visceral adipose tissue index (iVAT) (β = 0.306, p = 0.04), while changes in insulin sensitivity (β = 0.287, p = 0.03) and fasting triglycerides (β = -0.285, p = 0.03) were associated with changes in pSAT. These data indicated that serum hepcidin are associated with longitudinal changes in SAT and VAT, independently of insulin sensitivity. This would be the first prospective study evaluating the redistribution of fat according to iron status and chronic inflammation.
Collapse
Affiliation(s)
- Alejandro Hinojosa-Moscoso
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
| | - Anna Motger-Albertí
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIEROBN), 28029 Madrid, Spain
| | - Elena De la Calle-Vargas
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
| | - Marian Martí-Navas
- Medical Imaging, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
| | - Carles Biarnés
- Medical Imaging, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
| | - María Arnoriaga-Rodríguez
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIEROBN), 28029 Madrid, Spain
| | - Gerard Blasco
- Medical Imaging, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Radiology (IDI), Dr. Josep Trueta University Hospital, 17007 Girona, Spain
| | - Josep Puig
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
- Medical Imaging, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Radiology (IDI), Dr. Josep Trueta University Hospital, 17007 Girona, Spain
| | - Diego Luque-Córdoba
- Department of Analytical Chemistry, University of Córdoba, Annex Marie Curie Building, Campus of Rabanales, 14014 Córdoba, Spain
- Consortium for Biomedical Research in Frailty & Healthy Ageing (CIBERFES), Carlos III Institute of Health, 28029 Madrid, Spain
| | - Feliciano Priego-Capote
- Department of Analytical Chemistry, University of Córdoba, Annex Marie Curie Building, Campus of Rabanales, 14014 Córdoba, Spain
- Consortium for Biomedical Research in Frailty & Healthy Ageing (CIBERFES), Carlos III Institute of Health, 28029 Madrid, Spain
| | - José María Moreno-Navarrete
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIEROBN), 28029 Madrid, Spain
| | - José Manuel Fernández-Real
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), 17007 Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, 17003 Girona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIEROBN), 28029 Madrid, Spain
| |
Collapse
|
25
|
Abstract
High iron is a risk factor for type 2 diabetes mellitus (T2DM) and affects most of its cardinal features: decreased insulin secretion, insulin resistance, and increased hepatic gluconeogenesis. This is true across the normal range of tissue iron levels and in pathologic iron overload. Because of iron's central role in metabolic processes (e.g., fuel oxidation) and metabolic regulation (e.g., hypoxia sensing), iron levels participate in determining metabolic rates, gluconeogenesis, fuel choice, insulin action, and adipocyte phenotype. The risk of diabetes related to iron is evident in most or all tissues that determine diabetes phenotypes, with the adipocyte, beta cell, and liver playing central roles. Molecular mechanisms for these effects are diverse, although there may be integrative pathways at play. Elucidating these pathways has implications not only for diabetes prevention and treatment, but also for the pathogenesis of other diseases that are, like T2DM, associated with aging, nutrition, and iron.
Collapse
Affiliation(s)
- Alexandria V Harrison
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
| | - Felipe Ramos Lorenzo
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
- Department of Veterans Affairs, W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina, USA
| | - Donald A McClain
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA;
- Department of Veterans Affairs, W.G. (Bill) Hefner Veterans Affairs Medical Center, Salisbury, North Carolina, USA
| |
Collapse
|
26
|
Barbalho SM, Laurindo LF, Tofano RJ, Flato UAP, Mendes CG, de Alvares Goulart R, Briguezi AMGM, Bechara MD. Dysmetabolic Iron Overload Syndrome: Going beyond the Traditional Risk Factors Associated with Metabolic Syndrome. ENDOCRINES 2023; 4:18-37. [DOI: 10.3390/endocrines4010002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025] Open
Abstract
Dysmetabolic iron overload syndrome (DIOS) corresponds to the increase in iron stores associated with components of metabolic syndrome (MtS) and in the absence of an identifiable cause of iron excess. The objective of this work was to review the main aspects of DIOS. PUBMED and EMBASE were consulted, and PRISMA guidelines were followed. DIOS is usually asymptomatic and can be diagnosed by investigating MtS and steatosis. About 50% of the patients present altered hepatic biochemical tests (increased levels of γ-glutamyl transpeptidase itself or associated with increased levels of alanine aminotransferase). The liver may present parenchymal and mesenchymal iron overload, but the excess of iron is commonly mild. Steatosis or steatohepatitis is observed in half of the patients. Fibrosis is observed in about 15% of patients. Hyperferritinemia may damage the myocardium, liver, and several other tissues, increasing morbidity and mortality. Furthermore, DIOS is closely related to oxidative stress, which is closely associated with several pathological conditions such as inflammatory diseases, hypertension, diabetes, heart failure, and cancer. DIOS is becoming a relevant finding in the general population and can be associated with high morbidity/mortality. For these reasons, investigation of this condition could be an additional requirement for the early prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília, São Paulo 17500-000, Brazil
| | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Ricardo José Tofano
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Department of Cardiology, Associação Beneficente Hospital Universitário (ABHU), Rua Dr. Próspero Cecílio Coimbra, 80, Marília, São Paulo 17525-160, Brazil
| | - Uri Adrian Prync Flato
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
- Department of Biochemistry and Nutrition, School of Food and Technology of Marília (FATEC), Avenida Castro Alves, 62, Marília, São Paulo 17500-000, Brazil
| | - Claudemir G. Mendes
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Ricardo de Alvares Goulart
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Ana Maria Gonçalves Milla Briguezi
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Avenida Hygino Muzzy Filho, 1001, Marília, São Paulo 17525-902, Brazil
| |
Collapse
|
27
|
Ying L, Yan L, Huimin Z, Min L, Xiaojuan Z, Zhanjian W, Yaru Z. Tea polyphenols improve glucose metabolism in ceruloplasmin knockout mice via decreasing hepatic iron deposition. CYTA - JOURNAL OF FOOD 2022. [DOI: 10.1080/19476337.2022.2112299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Lei Ying
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Liu Yan
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Zhou Huimin
- Department of Endocrinology, The First Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Li Min
- Department of Nephrology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Zhang Xiaojuan
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Wang Zhanjian
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| | - Zhou Yaru
- Department of Endocrinology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, P.R. China
| |
Collapse
|
28
|
Chen H. Iron metabolism in non-alcoholic fatty liver disease: A promising therapeutic target. LIVER RESEARCH 2022; 6:203-213. [PMID: 39957910 PMCID: PMC11791839 DOI: 10.1016/j.livres.2022.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/05/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022]
Abstract
Non-alcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease worldwide, and is closely associated with the increased risk of the prevalence of obesity and diabetes. NAFLD begins with the presence of >5% excessive lipid accumulation in the liver, and potentially develops into non-alcoholic steatohepatitis, fibrosis, cirrhosis and hepatocellular carcinoma. Therefore, insight into the pathogenesis of NAFLD is of key importance to its effective treatment. Iron is an essential element in the life of all mammalian organisms. However, the free iron deposition is positively associated with histological severity in NAFLD patients due to the production of reactive oxygen species via the Fenton reaction. Recently, several iron metabolism-targeted therapies, such as phlebotomy, iron chelators, nanotherapeutics. and ferroptosis, have shown their potential as a therapeutic option in the treatment of NAFLD and as a clinical strategy to intervene in the progression of NAFLD. Herein, we review the recent overall evidence on iron metabolism and provide the mechanism of hepatic iron overload-induced liver pathologies and the recent advances in iron metabolism-targeted therapeutics in the treatment of NAFLD.
Collapse
Affiliation(s)
- Hanqing Chen
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou Key Laboratory of Digestive Diseases, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Radakovich LB, Burton LH, Culver LA, Afzali MF, Marolf AJ, Olver CS, Santangelo KS. Systemic iron reduction via an iron deficient diet decreases the severity of knee cartilage lesions in the Dunkin-Hartley guinea pig model of osteoarthritis. Osteoarthritis Cartilage 2022; 30:1482-1494. [PMID: 36030059 PMCID: PMC9588731 DOI: 10.1016/j.joca.2022.08.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 08/15/2022] [Accepted: 08/17/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Iron accumulation is emerging as a player in aging-related disorders due to its propensity for generating reactive oxygen species (ROS). Studies investigating the role of iron in the pathogenesis of primary osteoarthritis (OA) are limited. We designed a proof-of-principle study to determine the effect of systemic iron deficiency, via an iron deficient diet, on knee OA in an animal model. METHODS Twelve-week-old male Hartley guinea pigs received the standard diet (n = 6) or a diet devoid of iron (n = 6) for 19-weeks. Iron levels were determined in the serum, liver, and articular cartilage. Knees were collected to assess structural changes related to OA (microcomputed tomography, histopathology). Immunohistochemistry was performed to evaluate the presence and distribution of a disintegrin and metalloproteinase with thrombospondin motifs 4 (ADAMTS4) and ROS-driven 4-hydroxynonenal (4-HNE)-induced protein adducts. Transcript expression was also assessed. RESULTS Relative to control animals, an iron deficient diet reduced the concentration of this mineral in serum, liver, and articular cartilage. Iron deficient animals had lower histologic OA scores; decreased subchondral bone mineral density was also noted. This reduction in knee joint pathology was accompanied by a decrease in: ADAMTS4 in synovium; and 4-HNE protein adducts from lipid peroxidation in both the menisci and articular cartilage of iron deficient animals. Expression of iron-related genes in these tissues was also altered in treated animals. CONCLUSIONS Results from this study suggest that systemic iron levels may play a role in knee OA pathogenesis, with a short-term deficit in dietary iron reducing the severity of knee cartilage lesions.
Collapse
Affiliation(s)
- L B Radakovich
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| | - L H Burton
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA.
| | - L A Culver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - M F Afzali
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - A J Marolf
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - C S Olver
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA
| | - K S Santangelo
- Department of Microbiology, Immunology, and Pathology, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
30
|
Regulation of a High-Iron Diet on Lipid Metabolism and Gut Microbiota in Mice. Animals (Basel) 2022; 12:ani12162063. [PMID: 36009656 PMCID: PMC9405328 DOI: 10.3390/ani12162063] [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: 07/14/2022] [Revised: 08/11/2022] [Accepted: 08/12/2022] [Indexed: 12/02/2022] Open
Abstract
Iron homeostasis disorder is associated with the imbalance of lipid metabolism, while the specific interaction remains unclear. In the present study, we investigated the effect of a high-iron diet on lipid metabolism in mice. The C57BL/6 mice were fed with a normal diet (WT) or a high-iron diet (WT + Fe) for 12 weeks. We found that mice in the WT + Fe group showed a significant decrease in body weight gain, body fat and lipid accumulation of liver when compared with mice in the WT group. Accordingly, serum total cholesterol and triglyceride levels were both reduced in mice with a high-iron diet. Moreover, mice in the WT + Fe group exhibited a significant decrease in expression of genes regulating adipogenesis and adipocyte differentiation, and a significant increase in expression of fat hydrolysis enzyme genes in both liver and adipose tissues, which was consistent with their dramatic reduction in adipocyte cell size. In addition, a high-iron diet decreased the relative abundance of beneficial bacteria (Akkermansia, Bifidobacterium and Lactobacillus) and increased the relative abundance of pathogenic bacteria (Romboutsia and Erysipelatoclostridium). Thus, our research revealed that a high-iron diet reduced lipid deposition by inhibiting adipogenesis and promoting lipolysis. Altered gut microbial composition induced by a high-iron diet may not play a critical role in regulating lipid metabolism, but might cause unwanted side effects such as intestinal inflammation and damaged villi morphology at the intestinal host–microbe interface. These findings provide new insights into the relationship among iron, lipid metabolism and gut microbiota.
Collapse
|
31
|
Dietary iron modulates hepatic glucose homeostasis via regulating gluconeogenesis. J Nutr Biochem 2022; 109:109104. [PMID: 35863586 DOI: 10.1016/j.jnutbio.2022.109104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022]
Abstract
Iron exerts significant influences on glucose metabolism. However, the regulatory mechanisms underlying disordered glucose response remains largely unclear. The aim of this study was to examine the impact of dietary iron on hepatic gluconeogenesis in mice and in rat liver-derived cells. High iron models of C57BL/6J mice were fed with 1.25 g Fe/kg diets for 9 weeks, and high-iron BRL-3A cell models were treated with 250 μmol/L FeSO4 for 12 h and 24 h. Our data showed that higher iron intake resulted in higher hepatic iron without iron toxicity, and reduced body weight gain with no difference of food intakes. High dietary iron significantly increased 61% of hepatic glycogen deposition, but exhibited impairment in glucose responses in mice. Moreover, high dietary iron suppressed hepatic gluconeogenesis by repressing the expression of key gluconeogenic enzymes, phosphoenolpyruvate carboxykinase and glucose-6-phosphatase. Meanwhile, mice fed with higher iron diets exhibited both decreased AMP-activated protein kinase (AMPK) activity and peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) protein levels. Furthermore, in BRL-3A cells, iron treatment increased cellular glucose uptake, and altered gluconeogenesis rhythmically by regulating the activation of AMPK and expression of PGC-1α successively. This study demonstrated that dietary high iron was able to increase hepatic glycogen deposition by enhancement of glucose uptake, and suppress hepatic gluconeogenesis by regulation of AMPK and PGC-1α.
Collapse
|
32
|
High Iron Exposure from the Fetal Stage to Adulthood in Mice Alters Lipid Metabolism. Nutrients 2022; 14:nu14122451. [PMID: 35745181 PMCID: PMC9227341 DOI: 10.3390/nu14122451] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 06/02/2022] [Accepted: 06/07/2022] [Indexed: 12/11/2022] Open
Abstract
Iron supplementation is recommended during pregnancy and fetal growth. However, excess iron exposure may increase the risk of abnormal fetal development. We investigated the potential side effects of high iron levels in fetuses and through their adult life. C57BL/6J pregnant mice from 2 weeks of gestation and their offspring until 30 weeks were fed a control (CTRL, FeSO4 0 g/1 kg) or high iron (HFe, FeSO4 9.9 g/1 kg) diets. HFe group showed higher iron accumulation in the liver with increased hepcidin, reduced TfR1/2 mRNAs, and lowered ferritin heavy chain (FTH) proteins in both liver and adipose tissues despite iron loading. HFe decreased body weight, fat weight, adipocyte size, and triglyceride levels in the blood and fat, along with downregulation of lipogenesis genes, including PPARγ, C/EBPα, SREBP1c, FASN, and SCD1, and fatty acid uptake and oxidation genes, such as CD36 and PPARα. UCP2, adiponectin, and mRNA levels of antioxidant genes such as GPX4, HO-1, and NQO1 were increased in the HFe group, while total glutathione was reduced. We conclude that prolonged exposure to high iron from the fetal stage to adulthood may decrease fat accumulation by altering ferritin expression, adipocyte differentiation, and triglyceride metabolism, resulting in an alteration in normal growth.
Collapse
|
33
|
Ikeda Y, Funamoto M, Tsuchiya K. The role of iron in obesity and diabetes. THE JOURNAL OF MEDICAL INVESTIGATION 2022; 69:1-7. [PMID: 35466128 DOI: 10.2152/jmi.69.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
Abstract
Iron is an essential trace metal for all life, but excess iron causes oxidative stress through catalyzing the toxic hydroxy-radical production via the Fenton reaction. The number of patients with obesity and diabetes has been increasing worldwide, and their onset and development are affected by diet. In both clinical and experimental studies, a high body iron content was associated with obesity and diabetes, and the reduction of body iron content to an appropriate level can ameliorate the status and development of obesity and diabetes. Macrophages play an essential role in the pathophysiology of obesity and diabetes, and in the metabolism and homeostasis of iron in the body. Recent studies demonstrated that macrophage polarization is related to adipocyte hypertrophy and insulin resistance through their capabilities of iron handling. Control of iron in macrophages is a potential therapeutic strategy for obesity and diabetes. J. Med. Invest. 69 : 1-7, February, 2022.
Collapse
Affiliation(s)
- Yasumasa Ikeda
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Masafumi Funamoto
- Department of Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| | - Koichiro Tsuchiya
- Department of Medical Pharmacology, Institute of Biomedical Sciences, Tokushima University Graduate School, Tokushima, Japan
| |
Collapse
|
34
|
Chaudhari UK, Newcomb JD, Jen KLC, Hansen BC. Iron deficiency, but not anemia, is identified in naturally occurring obesity and insulin resistance in male nonhuman primates. J Med Primatol 2022; 51:165-171. [PMID: 35229310 DOI: 10.1111/jmp.12572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Revised: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 11/30/2022]
Abstract
OBJECTIVES To understand the development of iron deficiency in obesity and its long-term impact on the profile of anemia in spontaneously obese nonhuman primates. METHODS The study included 69 adult male nonhuman primates, (NHPs, Macaca mulatta, rhesus monkeys), ranging from normal to obese, and type 2 diabetes (T2D) as defined for humans. RESULTS Iron deficiency was present in 31.9% and mild anemia in 13% of the rhesus monkey in the colony. Serum iron levels were significantly lower in obese (p < .01) and T2D (p < .01)) compared with normal NHP. Obese NHPs also had significantly higher hemoglobin (p < .05), and red blood cell count (p < .05) than normal weight NHPs, thus not related to anemia. CONCLUSIONS Iron deficiency with increased hemoglobin and red blood cells was significantly associated with increased adiposity, insulin resistance, and diabetes. Iron deficiency does not cause and is not related to anemia in obese and T2D NHPs.
Collapse
Affiliation(s)
- Uddhav K Chaudhari
- Department of Internal Medicine, Obesity Diabetes and Aging Research Center, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA.,ICMR-National Institute for Research in Reproductive and Child Health (ICMR-NIRRCH), Mumbai, India
| | - Jennifer D Newcomb
- Department of Internal Medicine, Obesity Diabetes and Aging Research Center, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| | - K-L Catherine Jen
- Department of Nutrition and Food Science, Wayne State University, Detroit, Michigan, USA
| | - Barbara C Hansen
- Department of Internal Medicine, Obesity Diabetes and Aging Research Center, Morsani College of Medicine, University of South Florida, Tampa, Florida, USA
| |
Collapse
|
35
|
Romero AR, Mu A, Ayres JS. Adipose triglyceride lipase mediates lipolysis and lipid mobilization in response to iron-mediated negative energy balance. iScience 2022; 25:103941. [PMID: 35265813 PMCID: PMC8899412 DOI: 10.1016/j.isci.2022.103941] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/23/2021] [Accepted: 02/14/2022] [Indexed: 11/09/2022] Open
Abstract
Maintenance of energy balance is essential for overall organismal health. Mammals have evolved complex regulatory mechanisms that control energy intake and expenditure. Traditionally, studies have focused on understanding the role of macronutrient physiology in energy balance. In the present study, we examined the role of the essential micronutrient iron in regulating energy balance. We found that a short course of dietary iron caused a negative energy balance resulting in a severe whole body wasting phenotype. This disruption in energy balance was because of impaired intestinal nutrient absorption. In response to dietary iron-induced negative energy balance, adipose triglyceride lipase (ATGL) was necessary for wasting of subcutaneous white adipose tissue and lipid mobilization. Fat-specific ATGL deficiency protected mice from fat wasting, but caused a severe cachectic response in mice when fed iron. Our work reveals a mechanism for micronutrient control of lipolysis that is necessary for regulating mammalian energy balance.
Collapse
Affiliation(s)
- Alicia R. Romero
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Gene Expression Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Division of Biological Sciences, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093, USA
| | - Andre Mu
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Gene Expression Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Gene Expression Lab, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Nomis Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Road, La Jolla, CA 92037, USA,Corresponding author
| |
Collapse
|
36
|
Effects of high dietary iron on the lipid metabolism in the liver and adipose tissue of male broiler chickens. Anim Feed Sci Technol 2021. [DOI: 10.1016/j.anifeedsci.2021.115131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
37
|
Paeschke S, Winter K, Bechmann I, Klöting N, Blüher M, Baum P, Kosacka J, Nowicki M. Leptin Receptor-Deficient db/db Mice Show Significant Heterogeneity in Response to High Non-heme Iron Diet. Front Nutr 2021; 8:741249. [PMID: 34646852 PMCID: PMC8503537 DOI: 10.3389/fnut.2021.741249] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 08/30/2021] [Indexed: 12/22/2022] Open
Abstract
Recent studies have shown an association between iron homeostasis, obesity and diabetes. In this work, we investigated the differences in the metabolic status and inflammation in liver, pancreas and visceral adipose tissue of leptin receptor-deficient db/db mice dependent on high iron concentration diet. 3-month-old male BKS-Leprdb/db/JOrlRj (db/db) mice were divided into two groups, which were fed with different diets containing high iron (29 g/kg, n = 57) or standard iron (0.178 g/kg; n = 42) concentrations for 4 months. As anticipated, standard iron-fed db/db mice developed obesity and diabetes. However, high iron-fed mice exhibited a wide heterogeneity. By dividing into two subgroups at the diabetes level, non-diabetic subgroup 1 (<13.5 mmol/l, n = 30) significantly differed from diabetic subgroup two (>13.5 mmol/l, n = 27). Blood glucose concentration, HbA1c value, inflammation markers interleukin six and tumor necrosis factor α and heme oxygenase one in visceral adipose tissue were reduced in subgroup one compared to subgroup two. In contrast, body weight, C-peptide, serum insulin and serum iron concentrations, pancreatic islet and signal ratio as well as cholesterol, LDL and HDL levels were enhanced in subgroup one. While these significant differences require further studies and explanation, our results might also explain the often-contradictory results of the metabolic studies with db/db mice.
Collapse
Affiliation(s)
- Sabine Paeschke
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Karsten Winter
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Ingo Bechmann
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Nora Klöting
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum Munchen at the University of Leipzig, Leipzig, Germany
| | - Matthias Blüher
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum Munchen at the University of Leipzig, Leipzig, Germany.,Department of Medicine, University of Leipzig, Leipzig, Germany
| | - Petra Baum
- Department of Neurology, University of Leipzig, Leipzig, Germany
| | - Joanna Kosacka
- Department of Medicine, University of Leipzig, Leipzig, Germany.,Applied Molecular Hepatology Lab, Department of Visceral, Transplant, Thoracic and Vascular Surgery, University of Leipzig Medical Center, Leipzig, Germany
| | - Marcin Nowicki
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| |
Collapse
|
38
|
Evaluation of the relationship between serum ferritin and insulin resistance and visceral adiposity index (VAI) in women with polycystic ovary syndrome. Eat Weight Disord 2021; 26:1581-1593. [PMID: 32772321 DOI: 10.1007/s40519-020-00980-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 07/30/2020] [Indexed: 01/08/2023] Open
Abstract
PURPOSE There is a relationship between polycystic ovary syndrome (PCOS) and adipose tissue dysfunction (ADD), but this relationship is not clear. It has been recently shown that iron accumulation in adipose tissue is among the causes of adipose tissue dysfunction. Data on adipose tissue dysfunction in women with PCOS are insufficient. In this study, we aimed to evaluate the relationship between serum ferritin levels (iron accumulation biomarker) and visceral adiposity index (an indicator of adipose tissue dysfunction). METHODS The study is a case-control study. Women with diagnosed PCOS with 2003 Rotterdam Diagnostic Criteria (n = 40) were compared with non-PCOS group (n = 40). In this study, the cholesterol ratios, the homeostatic model evaluation index for insulin resistance (HOMA-IR) and the quantitative insulin sensitivity control index were calculated using biochemical parameters, and the visceral adiposity index (VAI) and the lipid accumulation product (LAP) were calculated using both anthropometric and biochemical parameters. In this study, insulin resistance was evaluated by HOMA-IR and adipose tissue dysfunction was evaluated by VAI index. RESULTS According to the results of this study, women with PCOS have a worse metabolic status than women without PCOS. However, this has been shown only in overweight and obese women, not in women with normal weight. CONCLUSION As a result, the presence of obesity in women with PCOS exacerbates metabolic status. LEVEL OF EVIDENCE Level V, cross-sectional descriptive study.
Collapse
|
39
|
Ma W, Jia L, Xiong Q, Feng Y, Du H. The role of iron homeostasis in adipocyte metabolism. Food Funct 2021; 12:4246-4253. [PMID: 33876811 DOI: 10.1039/d0fo03442h] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Iron plays a vital role in the metabolism of adipose tissue. On the one hand, iron is essential for differentiation, endocrine, energy supply and other physiological functions of adipocytes. Iron homeostasis affects the progression of many chronic metabolic diseases such as obesity, type 2 diabetes mellitus, and non-alcoholic fatty liver disease. In adipose tissue, iron deficiency is associated with obesity, mainly due to inflammation. Nevertheless, excessive iron in adipose tissue leads to decreased insulin sensitivity owing to mitochondrial dysfunction and adipokine changes. On the other hand, iron has an effect on the thermogenesis of adipocytes. Iron deficiency affects the production of beige fat and the direction of the differentiation of brown fat. In this review, we summarize the current understanding of the crosstalk between iron homeostasis and metabolism in adipose tissue.
Collapse
Affiliation(s)
- Wan Ma
- Key Laboratory of Animal Feed and Nutrition of Zhejiang Province, College of Animal Science, Zhejiang University, Hangzhou, China.
| | | | | | | | | |
Collapse
|
40
|
Iron aggravates hepatic insulin resistance in the absence of inflammation in a novel db/db mouse model with iron overload. Mol Metab 2021; 51:101235. [PMID: 33872860 PMCID: PMC8131719 DOI: 10.1016/j.molmet.2021.101235] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/31/2021] [Accepted: 04/09/2021] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVE The molecular pathogenesis of late complications associated with type 2 diabetes mellitus (T2DM) is not yet fully understood. While high glucose levels indicated by increased HbA1c only poorly explain disease progression and late complications, a pro-inflammatory status, oxidative stress, and reactive metabolites generated by metabolic processes were postulated to be involved. Individuals with metabolic syndrome (MetS) frequently progress to T2DM, whereby 70% of patients with T2DM show non-alcoholic fatty liver disease (NAFLD), the hepatic manifestation of MetS, and insulin resistance (IR). Epidemiological studies have shown that T2DM and steatosis are associated with alterations in iron metabolism and hepatic iron accumulation. Excess free iron triggers oxidative stress and a switch towards a macrophage pro-inflammatory status. However, so far it remains unclear whether hepatic iron accumulation plays a causative role in the generation of IR and T2DM or whether it is merely a manifestation of altered hepatic metabolism. To address this open question, we generated and characterized a mouse model of T2DM with IR, steatosis, and iron overload. METHODS Leprdb/db mice hallmarked by T2DM, IR and steatosis were crossed with Fpnwt/C326S mice with systemic iron overload to generate Leprdb/db/Fpnwt/C326S mice. The resulting progeny was characterized for major diabetic and iron-related parameters. RESULTS We demonstrated that features associated with T2DM in Leprdb/db mice, such as obesity, steatosis, or IR, reduce the degree of tissue iron overload in Fpnwt/C326S mice, suggesting an 'iron resistance' phenotype. Conversely, we observed increased serum iron levels that strongly exceeded those in the iron-overloaded Fpnwt/C326S mice. Increased hepatic iron levels induced oxidative stress and lipid peroxidation and aggravated IR, as indicated by diminished IRS1 phosphorylation and AKT activation. Additionally, in the liver, we observed gene response patterns indicative of de novo lipogenesis and increased gluconeogenesis as well as elevated free glucose levels. Finally, we showed that iron overload in Leprdb/db/Fpnwt/C326S mice enhances microvascular complications observed in retinopathy, suggesting that iron accumulation can enhance diabetic late complications associated with the liver and the eye. CONCLUSION Taken together, our data show that iron causes the worsening of symptoms associated with the MetS and T2DM. These findings imply that iron depletion strategies together with anti-diabetic drugs may ameliorate IR and diabetic late complications.
Collapse
|
41
|
Tang Y, Wang D, Zhang H, Zhang Y, Wang J, Qi R, Yang J, Shen H, Xu Y, Li M. Rapid responses of adipocytes to iron overload increase serum TG level by decreasing adiponectin. J Cell Physiol 2021; 236:7544-7553. [PMID: 33855731 DOI: 10.1002/jcp.30391] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/14/2021] [Accepted: 03/30/2021] [Indexed: 12/13/2022]
Abstract
Iron overload is tightly connected with metabolic disorders. Excess iron in the adipose and its roles in dyslipidemia are of interest to be identified. In acute iron overload mice receiving intraperitoneal injection of 100 mg/kg/day dextran-iron for 5 days, the epididymis adipose showed a remarkable increase in iron. Serum triglyceride and low-density lipoprotein cholesterol (LDL-C) levels were increased and high-density lipoprotein cholesterol (HDL-C) level was decreased, while serum alkaline phosphatase, aspartate aminotransferase, glucose, and insulin were not affected. The serum-cytokine-microarray showed that adipocytokines, including adiponectin, leptin, and resistin were significantly decreased. Other serum cytokines, including pro-insulin cytokines, inflammatory cytokines, chemokines, and growth factors were not changed, except that ghrelin and chemokine RANTES were increased. Iron overload decreased expressions of adiponectin and leptin both in vivo and in vitro. Intraperitoneal injection of recombinant leptin at 1 μg/g in acute iron overload mice had no significant effects on serum levels of TC, TG, HDL-C, and LDL-C, while intraperitoneal injection of recombinant adiponectin at 3 μg/g partially restored serum TG level through improving activities of lipoprotein lipase and hepatic lipase, but abnormal serum LDL-C and HDL-C were not redressed, suggesting other mechanisms also existed. In conclusion, the adipose responds to iron overload at an early stage to interfere with lipid metabolism by secreting adipocytokines, which may further affect glucose metabolism, inflammation, and other iron overload-induced effects on the body.
Collapse
Affiliation(s)
- Yuxiao Tang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Dongyao Wang
- School of Pharmacy, Second Military Medical University, Shanghai, China.,Faculty of Pharmacy, Shanghai University, Shanghai, China
| | - Hongwei Zhang
- Department of Nutrition, Second Military Medical University, Shanghai, China.,Department of Clinical Nutrition, Zhumadian Second People's Hospital, Henan, China
| | - Yinyin Zhang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jie Wang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Ruirui Qi
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Jianxin Yang
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Hui Shen
- Department of Nutrition, Second Military Medical University, Shanghai, China
| | - Yan Xu
- Institute of International Medical Science and Technology, Sanda University, Shanghai, China
| | - Min Li
- Department of Nutrition, Second Military Medical University, Shanghai, China.,Institute of International Medical Science and Technology, Sanda University, Shanghai, China
| |
Collapse
|
42
|
Liao W, Yang W, Shen Z, Ai W, Pan Q, Sun Y, Guo S. Heme Oxygenase-1 Regulates Ferrous Iron and Foxo1 in Control of Hepatic Gluconeogenesis. Diabetes 2021; 70:696-709. [PMID: 33408127 PMCID: PMC7897351 DOI: 10.2337/db20-0954] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
The liver is a key player for maintaining glucose homeostasis. Excessive hepatic glucose production is considered to be a key for the onset of type 2 diabetes. The primary function of heme oxygenase-1 (HO1) is to catalyze the degradation of heme into biliverdin, ferrous iron, and carbon monoxide. Previous studies have demonstrated that the degradation of heme by HO1 in the liver results in mitochondrial dysfunction and drives insulin resistance. In this study, by overexpressing HO1 in hepatocytes and mice, we showed that HO1 promotes gluconeogenesis in a Foxo1-dependent manner. Importantly, HO1 overexpression increased the generation of ferrous iron in the liver, which further activates nuclear factor-κB and phosphorylates Foxo1 at Ser273 to enhance gluconeogenesis. We further assessed the role of HO1 in insulin-resistant liver-specific knockout of IRS1 and IRS2 genes (L-DKO) mice, which exhibit upregulation of HO1 in the liver and hepatic ferrous iron overload. HO1 knockdown by shRNA or treatment of iron chelator rescued the aberrant gluconeogenesis in L-DKO mice. In addition, we found that systemic iron overload promotes gluconeogenesis by activating the hepatic protein kinase A→Foxo1 axis. Thus, our results demonstrate the role of HO1 in regulating hepatic iron status and Foxo1 to control gluconeogenesis and blood glucose.
Collapse
Affiliation(s)
- Wang Liao
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Wanbao Yang
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Zheng Shen
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Weiqi Ai
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Quan Pan
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Yuxiang Sun
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| | - Shaodong Guo
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX
| |
Collapse
|
43
|
Zhou B, Liu S, Yuan G. Combined Associations of Serum Ferritin and Body Size Phenotypes With Cardiovascular Risk Profiles: A Chinese Population-Based Study. Front Public Health 2021; 9:550011. [PMID: 33659229 PMCID: PMC7917052 DOI: 10.3389/fpubh.2021.550011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 01/20/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Serum ferritin (SF) has been correlated with one or more metabolic syndrome features associated with an increased risk for cardiovascular disease (CVD). This study explored the associations between SF and CVD risk factors among different body size phenotypes that were based on metabolic status and body mass index (BMI) categories. Methods: A cross-sectional study was performed using a cohort of 7,549 Chinese adults from the China Health and Nutrition Survey. Participants did not exhibit acute inflammation, were not underweight and were stratified based on their metabolic status and BMI categories. The metabolically at-risk status was defined as having two or more criteria of the Adult Treatment Panel-III metabolic syndrome definition, excluding waist circumference. Results: Compared with individuals without high SF, subjects with high SF had an increased risk of diabetes in the metabolically at-risk normal-weight (MANW) and metabolically at-risk overweight/obesity (MAO) groups. The multivariate-adjusted odds ratios (ORs) were 1.52 [95% confidence interval (Cls): 1.02, 2.28] and 1.63 (95% Cls: 1.27, 2.09), respectively. Adjusted ORs for hyperuricemia from high SF in metabolically healthy normal-weight (MHNW), metabolically healthy overweight/obesity (MHO), MANW, and MAO phenotypes were 1.78 (95% Cls: 1.26, 2.53), 1.42 (95% Cls: 1.03, 1.95), 1.66 (95% Cls: 1.17, 2.36), and 1.42 (95% Cls: 1.17, 1.73), respectively. Similarly, positive correlations of high SF with triglycerides, non-high-density lipoprotein cholesterol, and apolipoprotein B100 were observed in all phenotypes. No association between high SF and elevated low-density lipoprotein cholesterol were observed among participants who were metabolically at-risk, regardless of their BMI categories. However, the ORs for elevated low-density lipoprotein cholesterol from high SF were 1.64 (95% Cls: 1.29, 2.08) in the MHNW group and 1.52 (95% Cls:1.22, 1.91) in the MHO group, significantly. This study demonstrated that the highest ORs were in MAO with a high SF group for all unfavorable CVD risk factors except low-density lipoprotein cholesterol (all p < 0.001). Conclusions: The associations of high SF with the prevalence of CVD risk factors, including diabetes, dyslipidemia, and hyperuricemia, vary in individuals among different body size phenotypes. In the MAO group, subjects with high SF levels exhibited worse CVD risk profiles than individuals without high SF.
Collapse
Affiliation(s)
| | | | - Gang Yuan
- Department of Endocrinology and Metabolism, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
44
|
Yoon N, Kim S, Sung HK, Dang TQ, Jeon JS, Sweeney G. Use of 2-dimensional cell monolayers and 3-dimensional microvascular networks on microfluidic devices shows that iron increases transendothelial adiponectin flux via inducing ROS production. Biochim Biophys Acta Gen Subj 2020; 1865:129796. [PMID: 33212230 DOI: 10.1016/j.bbagen.2020.129796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND Iron excess is a risk factor for cardiovascular diseases and it is important to understand the effect of iron on vascular permeability, particularly for the transport of large metabolic hormones such as adiponectin. METHODS We used 2-dimensional monolayers of cultured human dermal microvascular endothelial cells (HDMEC) and human umbilical vein endothelial cells (HUVEC) as well as 3-dimensional microvascular networks to measure transendothelial flux. RESULTS Iron supplementation reduced transendothelial electric resistance (TEER). Flux analysis indicated that under control conditions permeability of 70 kDa dextran and oligomeric forms of adiponectin were restricted in comparison with a 3 kDa dextran, however upon iron treatment permeability of the larger molecules was increased. The increased permeability and size-dependent trans-endothelial movement in response to iron was also observed in 3-dimensional microvascular networks. Mechanistically, the alteration in barrier functionality was associated with increased oxidative stress in response to iron since alterations in TEER and permeability were rescued when reactive oxygen species production was attenuated by pre-treatment with the antioxidant N-acetyl cysteine.]. CONCLUSIONS Iron supplementation induced ROS production resulting in increased transendothelial permeability. GENERAL SIGNIFICANCE Altogether, this suggests that the oxidative stress associated with iron excess could play an important role in the regulation of endothelial functionality, controlling hormone action in peripheral tissues by regulating the first rate-limiting step controlling hormone access to target tissues.
Collapse
Affiliation(s)
- Nanyoung Yoon
- Department of Biology, York University, Toronto, ON, Canada
| | - Seunggyu Kim
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | | | - Thanh Q Dang
- Department of Biology, York University, Toronto, ON, Canada
| | - Jessie S Jeon
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada.
| |
Collapse
|
45
|
Fillebeen C, Lam NH, Chow S, Botta A, Sweeney G, Pantopoulos K. Regulatory Connections between Iron and Glucose Metabolism. Int J Mol Sci 2020; 21:ijms21207773. [PMID: 33096618 PMCID: PMC7589414 DOI: 10.3390/ijms21207773] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/07/2020] [Accepted: 10/16/2020] [Indexed: 02/06/2023] Open
Abstract
Iron is essential for energy metabolism, and states of iron deficiency or excess are detrimental for organisms and cells. Therefore, iron and carbohydrate metabolism are tightly regulated. Serum iron and glucose levels are subjected to hormonal regulation by hepcidin and insulin, respectively. Hepcidin is a liver-derived peptide hormone that inactivates the iron exporter ferroportin in target cells, thereby limiting iron efflux to the bloodstream. Insulin is a protein hormone secreted from pancreatic β-cells that stimulates glucose uptake and metabolism via insulin receptor signaling. There is increasing evidence that systemic, but also cellular iron and glucose metabolic pathways are interconnected. This review article presents relevant data derived primarily from mouse models and biochemical studies. In addition, it discusses iron and glucose metabolism in the context of human disease.
Collapse
Affiliation(s)
- Carine Fillebeen
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC H3Y 1P3, Canada;
| | - Nhat Hung Lam
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Samantha Chow
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Amy Botta
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada; (N.H.L.); (S.C.); (A.B.); (G.S.)
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC H3Y 1P3, Canada;
- Correspondence: ; Tel.: +1-514-340-8260 (ext. 25293)
| |
Collapse
|
46
|
Affiliation(s)
- Grischa Y. Chen
- Molecular and Systems Physiology Lab, Gene Expression Lab, NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
| | - Janelle S. Ayres
- Molecular and Systems Physiology Lab, Gene Expression Lab, NOMIS Center for Immunobiology and Microbial Pathogenesis, The Salk Institute for Biological Studies, La Jolla, California, United States of America
- * E-mail:
| |
Collapse
|
47
|
Seeßle J, Gan-Schreier H, Kirchner M, Stremmel W, Chamulitrat W, Merle U. Plasma Lipidome, PNPLA3 polymorphism and hepatic steatosis in hereditary hemochromatosis. BMC Gastroenterol 2020; 20:230. [PMID: 32680469 PMCID: PMC7368730 DOI: 10.1186/s12876-020-01282-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 04/22/2020] [Indexed: 12/20/2022] Open
Abstract
Background Hereditary hemochromatosis (HH) is an autosomal recessive genetic disorder with increased intestinal iron absorption and therefore iron Overload. iron overload leads to increased levels of toxic non-transferrin bound iron which results in oxidative stress and lipid peroxidation. The impact of iron on lipid metabolism is so far not fully understood. The aim of this study was to investigate lipid metabolism including lipoproteins (HDL, LDL), neutral (triglycerides, cholesterol) and polar lipids (sphingo- and phospholipids), and PNPLA3 polymorphism (rs738409/I148M) in HH. Methods We conducted a cohort study of 54 subjects with HH and 20 healthy subjects. Patients were analyzed for their iron status including iron, ferritin, transferrin and transferrin saturation and serum lipid profile on a routine follow-up examination. Results HH group showed significantly lower serum phosphatidylcholine (PC) and significantly higher phosphatidylethanolamine (PE) compared to healthy control group. The ratio of PC/PE was clearly lower in HH group indicating a shift from PC to PE. Triglycerides were significantly higher in HH group. No differences were seen for HDL, LDL and cholesterol. Hepatic steatosis was significantly more frequent in HH. PNPLA3 polymorphism (CC vs. CG/GG) did not reveal any significant correlation with iron and lipid parameters including neutral and polar lipids, grade of steatosis and fibrosis. Conclusion Our study strengthens the hypothesis of altered lipid metabolism in HH and susceptibility to nonalcoholic fatty liver disease. Disturbed phospholipid metabolism may represent an important factor in pathogenesis of hepatic steatosis in HH.
Collapse
Affiliation(s)
- Jessica Seeßle
- Department of Gastroenterology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Hongying Gan-Schreier
- Department of Gastroenterology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Marietta Kirchner
- Institute of Medical Biometry and Informatics, University Hospital Heidelberg, Heidelberg, Germany
| | - Wolfgang Stremmel
- Department of Gastroenterology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Walee Chamulitrat
- Department of Gastroenterology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Uta Merle
- Department of Gastroenterology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
| |
Collapse
|
48
|
Dubey P, Thakur V, Chattopadhyay M. Role of Minerals and Trace Elements in Diabetes and Insulin Resistance. Nutrients 2020; 12:1864. [PMID: 32585827 PMCID: PMC7353202 DOI: 10.3390/nu12061864] [Citation(s) in RCA: 194] [Impact Index Per Article: 38.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022] Open
Abstract
Minerals and trace elements are micronutrients that are essential to the human body but present only in traceable amounts. Nonetheless, they exhibit well-defined biochemical functions. Deficiencies in these micronutrients are related to widespread human health problems. This review article is focused on some of these minerals and trace element deficiencies and their consequences in diabetes and insulin resistance. The levels of trace elements vary considerably among different populations, contingent on the composition of the diet. In several Asian countries, large proportions of the population are affected by a number of micronutrient deficiencies. Local differences in selenium, zinc, copper, iron, chromium and iodine in the diet occur in both developed and developing countries, largely due to malnutrition and dependence on indigenous nutrition. These overall deficiencies and, in a few cases, excess of essential trace elements may lead to imbalances in glucose homeostasis and insulin resistance. The most extensive problems affecting one billion people or more worldwide are associated with inadequate supply of a number of minerals and trace elements including iodine, selenium, zinc, calcium, chromium, cobalt, iron, boron and magnesium. This review comprises various randomized controlled trials, cohort and case-controlled studies, and observational and laboratory-based studies with substantial outcomes of micronutrient deficiencies on diabetes and insulin resistance in diverse racial inhabitants from parts of Asia, Africa, and North America. Changes in these micronutrient levels in the serum and urine of subjects may indicate the trajectory toward metabolic changes, oxidative stress and provide disease-relevant information.
Collapse
Affiliation(s)
- Pallavi Dubey
- Department of Obstetrics and Gynecology, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
| | - Vikram Thakur
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
| | - Munmun Chattopadhyay
- Department of Molecular and Translational Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA;
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA
| |
Collapse
|
49
|
Zhou B, Ren H, Zhou X, Yuan G. Associations of iron status with apolipoproteins and lipid ratios: a cross-sectional study from the China Health and Nutrition Survey. Lipids Health Dis 2020; 19:140. [PMID: 32546165 PMCID: PMC7298938 DOI: 10.1186/s12944-020-01312-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/04/2020] [Indexed: 12/23/2022] Open
Abstract
Background Iron overload has been found to be related with various cardiometabolic disorders, like dyslipidemia, metabolic syndrome, and diabetes. The disturbance of the iron status and lipid metabolism can contribute to organ damage such as atherosclerotic plaque growth and instability. An assessment on the associations of iron status with apolipoproteins and lipid ratios would be informative for maintenance of metabolic homeostasis and hinderance of disease progression. Hence, this study aims to establish the relationships of iron status with apolipoproteins and lipid ratios. Methods A cross-sectional study of 7540 adult participants from the China Health and Nutrition Survey 2009 was conducted. Logistic regression analyses were used to investigate the relationships between indicators of iron status and the prevalence of unfavorable apolipoprotein profiles. Multivariate linear regression models were constructed to assess the dose-response correlations between serum ferritin and lipid parameters. Results After adjustment for confounding factors, in both sexes, the subjects in the top quartile of ferritin had the highest prevalence of an elevated apolipoprotein B (men: odds ratio (OR) 1.97, 95% confidence interval (CI) 1.50–2.62; women: OR 2.13, 95% CI 1.53–2.97) and an elevated apolipoprotein B/apolipoprotein A1 ratio (men: OR 2.00, 95% CI 1.50–2.66; women: OR 1.41, 95% CI 1.04–1.92) when compared with individuals in the lowest quartile. Hemoglobin were also independently associated with unfavorable apolipoprotein B and apolipoprotein B/apolipoprotein A1 ratio both in men and women. However, transferrin (men: OR 0.74, 95% CI 0.56–0.99; women: OR 0.73, 95% CI 0.56–0.95) and soluble transferrin receptor (men: OR 0.75, 95% CI 0.57–0.99; women: OR 0.71, 95% CI 0.55–0.91) were found to be negatively associated with a decreased apolipoprotein A1. Moreover, after controlling for potential confounders, the ferritin concentrations were significantly associated with the levels of lipid ratios including TG/HDL-C, non-HDL-C/HDL-C, TC/HDL-C, apoB/apoA1, and LDL-C/HDL-C ratio in men (β coefficient = 0.147, 0.061, 0.043, 0.038, 0.032, respectively, all P values < 0.001) and in women (β coefficient = 0.074, 0.034, 0.025, 0.020, 0.018, respectively, all P values < 0.05). Conclusions The indicators of iron status are significantly associated with unfavorable apolipoprotein profiles. Serum ferritin concentrations are positively correlated with the levels of lipid ratios. The management on the modifiable iron status and lipid metabolism has a clinical significance. The atherosclerotic lipid profiles of the patients with iron overload deserve special clinical concerns.
Collapse
Affiliation(s)
- Bowen Zhou
- Department of Endocrinology and Metabolism, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Huihui Ren
- Department of Endocrinology and Metabolism, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Xinrong Zhou
- Department of Endocrinology and Metabolism, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China
| | - Gang Yuan
- Department of Endocrinology and Metabolism, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, 430030, Hubei, China.
| |
Collapse
|
50
|
Development of insulin resistance preceded major changes in iron homeostasis in mice fed a high-fat diet. J Nutr Biochem 2020; 84:108441. [PMID: 32629238 PMCID: PMC7115812 DOI: 10.1016/j.jnutbio.2020.108441] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 03/10/2020] [Accepted: 05/22/2020] [Indexed: 02/06/2023]
Abstract
Type 2 diabetes mellitus (T2DM) and insulin resistance (IR) have been associated with dysregulation of iron metabolism. The basis for this association is not completely understood. To attempt to investigate this, we studied temporal associations between onset of insulin resistance (IR) and dysregulated iron homeostasis, in a mouse model of T2DM. Male C57Bl/6 mice (aged 8 weeks) were fed a high-fat diet (HFD; 60% energy from fat) or a control diet (CD; 10% energy from fat) for 4, 8, 12, 16, 20 and 24 weeks. Development of IR was documented, and various metabolic, inflammatory and iron-related parameters were studied in these mice. HFD-feeding induced weight gain, hepato-steatosis and IR in the mice. Onset of IR occurred from 12 weeks onwards. Hepatic iron stores progressively declined from 16 weeks onwards. Accompanying changes included a decrease in hepatic hepcidin (Hamp1) mRNA expression and serum hepcidin levels and an increase in iron content in the epididymal white adipose tissue (eWAT). Iron content in the liver negatively correlated with that in the eWAT. Factors known to regulate hepatic Hamp1 expression (such as serum iron levels, systemic inflammation, and bone marrow-derived erythroid regulators) were not affected by HFD-feeding. In conclusion, the results show that the onset of IR in HFD-fed mice preceded dysregulation of iron homeostasis, evidence of which were found both in the liver and visceral adipose tissue.
Collapse
|