1
|
Tinkov AA, Korobeinikova TV, Morozova GD, Aschner M, Mak DV, Santamaria A, Rocha JBT, Sotnikova TI, Tazina SI, Skalny AV. Association between serum trace element, mineral, and amino acid levels with non-alcoholic fatty liver disease (NAFLD) in adult women. J Trace Elem Med Biol 2024; 83:127397. [PMID: 38290269 DOI: 10.1016/j.jtemb.2024.127397] [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: 11/30/2023] [Revised: 12/29/2023] [Accepted: 01/13/2024] [Indexed: 02/01/2024]
Abstract
The objective of the present study is assessment of serum trace element and amino acid levels in non-alcoholic fatty liver disease (NAFLD) patients with subsequent evaluation of its independent associations with markers of liver injury and metabolic risk. MATERIALS AND METHODS 140 women aged 20-90 years old with diagnosed NAFLD and 140 healthy women with a respective age range were enrolled in the current study. Analysis of serum and hair levels of trace elements and minerals was performed with inductively-coupled plasma mass-spectrometry (ICP-MS). Serum amino acid concentrations were evaluated by high-pressure liquid chromatography (HPLC) with UV-detection. In addition, routine biochemical parameters including liver damage markers, alanine aminotransferase (ALT) and gamma-glutamyltransferase (GGT), were assessed spectrophotometrically. RESULTS The findings demonstrated that patients with NAFLD were characterized by higher ALT, GGT, lactate dehydrogenase (LDH) and cholinesterase (CE) activity, as well as increased levels of total cholesterol, low-density lipoprotein cholesterol, triglycerides, and uric acid. NAFLD patients were characterized by reduced serum and hair Co, Se, and Zn levels, as well as hair Cu content and serum Mn concentrations in comparison to controls. Circulating Ala, Cit, Glu, Gly, Ile, Leu, Phe, and Tyr levels in NAFLD patients exceeded those in the control group. Multiple linear regression demonstrated that serum and hair trace element levels were significantly associated with circulating amino acid levels after adjustment for age, BMI, and metabolic parameters including liver damage markers. CONCLUSION It is proposed that altered trace element handling may contribute to NAFLD pathogenesis through modulation of amino acid metabolism.
Collapse
Affiliation(s)
- Alexey A Tinkov
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia; Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia.
| | - Tatiana V Korobeinikova
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Galina D Morozova
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 10461 Bronx, NY, USA
| | - Daria V Mak
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Abel Santamaria
- Faculty of Sciencies, National Autonomous University of Mexico, 04510 Mexico City, Mexico
| | - Joao B T Rocha
- Departamento de Bioquímica e Biologia Molecular, CCNE, Universidade Federal de Santa Maria, Santa Maria 97105-900 RS, Brazil
| | - Tatiana I Sotnikova
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; City Clinical Hospital n. a. S.P. Botkin of the Moscow City Health Department, 125284 Moscow, Russia
| | - Serafima Ia Tazina
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; City Clinical Hospital n. a. S.P. Botkin of the Moscow City Health Department, 125284 Moscow, Russia
| | - Anatoly V Skalny
- Center of Bioelementology and Human Ecology, and World-Class Research Center "Digital Biodesign and Personalized Healthcare", and Department of Therapy of the Institute of Postgraduate Education, IM Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, 150003 Yaroslavl, Russia; Department of Medical Elementology, Peoples' Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
2
|
Berger MM, Shenkin A, Dizdar OS, Amrein K, Augsburger M, Biesalski HK, Bischoff SC, Casaer MP, Gundogan K, Lepp HL, de Man AME, Muscogiuri G, Pietka M, Pironi L, Rezzi S, Schweinlin A, Cuerda C. ESPEN practical short micronutrient guideline. Clin Nutr 2024; 43:825-857. [PMID: 38350290 DOI: 10.1016/j.clnu.2024.01.030] [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: 01/10/2024] [Accepted: 01/27/2024] [Indexed: 02/15/2024]
Abstract
BACKGROUND Trace elements and vitamins, named together micronutrients (MNs), are essential for human metabolism. The importance of MNs in common pathologies is recognized by recent research, with deficiencies significantly impacting the outcome. OBJECTIVE This short version of the guideline aims to provide practical recommendations for clinical practice. METHODS An extensive search of the literature was conducted in the databases Medline, PubMed, Cochrane, Google Scholar, and CINAHL for the initial guideline. The search focused on physiological data, historical evidence (for papers published before PubMed release in 1996), and observational and/or randomized trials. For each MN, the main functions, optimal analytical methods, impact of inflammation, potential toxicity, and provision during enteral or parenteral nutrition were addressed. The SOP wording was applied for strength of recommendations. RESULTS The limited number of interventional trials prevented meta-analysis and led to a low level of evidence for most recommendations. The recommendations underwent a consensus process, which resulted in a percentage of agreement (%): strong consensus required of >90 % of votes. Altogether the guideline proposes 3 general recommendations and specific recommendations for the 26 MNs. Monitoring and management strategies are proposed. CONCLUSION This short version of the MN guideline should facilitate handling of the MNs in at-risk diseases, whilst offering practical advice on MN provision and monitoring during nutritional support.
Collapse
Affiliation(s)
- Mette M Berger
- Faculty of Biology & Medicine, Lausanne University, Lausanne, Switzerland.
| | - Alan Shenkin
- Institute of Aging and Chronic Disease, University of Liverpool, Liverpool, UK.
| | - Oguzhan Sıtkı Dizdar
- Department of Internal Medicine and Clinical Nutrition Unit, University of Health Sciences Kayseri City Training and Research Hospital, Kayseri, Turkey.
| | - Karin Amrein
- Medical University of Graz, Department of Internal Medicine, Division of Endocrinology and Diabetology, Austria.
| | - Marc Augsburger
- University Centre of Legal Medicine Lausanne-Geneva, Lausanne University Hospital and University of Lausanne, Geneva University Hospital and University of Geneva, Lausanne-Geneva, Switzerland.
| | | | - Stephan C Bischoff
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Michael P Casaer
- KU Leuven, Department of Cellular and Molecular Medicine, Laboratory of Intensive Care Medicine, Leuven, Belgium.
| | - Kursat Gundogan
- Division of Intensive Care Medicine, Department of Internal Medicine, Erciyes University School of Medicine, Kayseri, Turkey.
| | | | - Angélique M E de Man
- Department of Intensive Care Medicine, Research VUmc Intensive Care (REVIVE), Amsterdam Cardiovascular Science (ACS), Amsterdam Infection and Immunity Institute (AI&II), Amsterdam Medical Data Science (AMDS), Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1117, 1081 HV Amsterdam, the Netherlands.
| | - Giovanna Muscogiuri
- Dipartimento di Medicina Clinica e Chirurgia, Sezione di Endocrinologia, Università di Napoli (Federico II), Naples, Italy; United Nations Educational, Scientific and Cultural Organization (UNESCO) Chair for Health Education and Sustainable Development, Federico II University, Naples, Italy.
| | - Magdalena Pietka
- Pharmacy Department, Stanley Dudrick's Memorial Hospital, Skawina, Poland.
| | - Loris Pironi
- Department of Medical and Surgical Sciences, University of Bologna, Italy; Centre for Chronic Intestinal Failure, IRCCS AOUBO, Bologna, Italy.
| | - Serge Rezzi
- Swiss Nutrition and Health Foundation, Epalinges, Switzerland.
| | - Anna Schweinlin
- Institute of Nutritional Medicine, University of Hohenheim, Stuttgart, Germany.
| | - Cristina Cuerda
- Departamento de Medicina, Universidad Complutense de Madrid, Nutrition Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain.
| |
Collapse
|
3
|
Sharma R, Zhao W, Zafar Y, Murali AR, Brown KE. Serum hepcidin levels in chronic liver disease: a systematic review and meta-analysis. Clin Chem Lab Med 2024; 62:373-384. [PMID: 37540837 DOI: 10.1515/cclm-2023-0540] [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: 05/22/2023] [Accepted: 07/25/2023] [Indexed: 08/06/2023]
Abstract
OBJECTIVES Dysregulation of hepcidin-iron axis is presumed to account for abnormal iron status in patients with chronic liver disease (CLD). Our aim is to determine the effect of specific etiologies of CLD and of cirrhosis on serum hepcidin levels. METHODS PubMed, Embase, Web of Science were searched for studies comparing serum hepcidin levels in patients with CLD to that in controls using enzyme-linked immunosorbent assay. The study was conducted in accordance with the Preferred Reporting Items for Systematic Review and Meta-Analysis Guidelines. Statistical analysis was carried out with STATA using random effects model to calculate the mean difference (MD) between two groups. RESULTS Hepcidin levels were significantly lower in subjects with hepatitis C virus (16 studies) [MD -1.6 (95 % CI: -2.66 to -0.54), p<0.01] and alcoholic liver disease (3 studies) [MD -0.84 (95 % CI: -1.6 to -0.07), p=0.03] than controls. Serum hepcidin was significantly higher in subjects with non-alcoholic fatty liver disease (12 studies) [MD 0.62 (95 % CI: 0.21 to 1.03), p<0.01], but did not differ in subjects with hepatitis B and controls (eight studies) [MD -0.65 (95 % CI: -1.47 to 0.16), p=0.12]. Hepcidin levels were significantly lower in patients with cirrhosis of any etiology (four studies) [MD -1.02 (CI: -1.59 to -0.45), p<0.01] vs. controls (CI: confidence interval). CONCLUSIONS Serum hepcidin levels are altered in common forms of CLD albeit not in a consistent direction. Additional study is needed to determine how changes in hepcidin levels are related to dysregulation of iron metabolism in CLD.
Collapse
Affiliation(s)
- Ruchi Sharma
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | - Weidan Zhao
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Department of Gastroenterology-Hepatology, SUNY Downstate, Brooklyn, NY, USA
| | - Yousaf Zafar
- University of Mississippi Medical Center, Jackson, MS, USA
| | - Arvind R Murali
- Department of Internal Medicine, Division of Gastroenterology-Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Orlando Health, Orlando, FL, USA
| | - Kyle E Brown
- Department of Internal Medicine, Division of Gastroenterology-Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
- Iowa City Veterans Administration Medical Center, Iowa City, IA, USA
- Department of Radiation Oncology, Program in Free Radical and Radiation Biology, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| |
Collapse
|
4
|
Gensluckner S, Wernly B, Koutny F, Strebinger G, Zandanell S, Stechemesser L, Paulweber B, Iglseder B, Trinka E, Frey V, Langthaler P, Semmler G, Valenti L, Corradini E, Datz C, Aigner E. Prevalence and Characteristics of Metabolic Hyperferritinemia in a Population-Based Central-European Cohort. Biomedicines 2024; 12:207. [PMID: 38255312 PMCID: PMC10813305 DOI: 10.3390/biomedicines12010207] [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: 12/12/2023] [Revised: 01/07/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Hyperferritinemia (HF) is a common finding and can be considered as metabolic HF (MHF) in combination with metabolic diseases. The definition of MHF was heterogenous until a consensus statement was published recently. Our aim was to apply the definition of MHF to provide data on the prevalence and characteristics of MHF in a Central-European cohort. METHODS This study was a retrospective analysis of the Paracelsus 10,000 study, a population-based cohort study from the region of Salzburg, Austria. We included 8408 participants, aged 40-77. Participants with HF were divided into three categories according to their level of HF and evaluated for metabolic co-morbidities defined by the proposed criteria for MHF. RESULTS HF was present in 13% (n = 1111) with a clear male preponderance (n = 771, 69% of HF). Within the HF group, 81% (n = 901) of subjects fulfilled the metabolic criteria and were defined as MHF, of which 75% (n = 674) were characterized by a major criterion. In the remaining HF cohort, 52% (n = 227 of 437) of subjects were classified as MHF after application of the minor criteria. CONCLUSION HF is a common finding in the general middle-aged population and the majority of cases are classified as MHF. The new classification provides useful criteria for defining MHF.
Collapse
Affiliation(s)
- Sophie Gensluckner
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria; (S.G.)
- Obesity Research Unit, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Bernhard Wernly
- Department of Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, Paracelsusstraße 37, 5110 Oberndorf, Austria
| | - Florian Koutny
- Department of Internal Medicine 2, Gastroenterology and Hepatology and Rheumatology, University Hospital of St. Pölten, Karl Landsteiner University of Health Sciences, Dunant-Platz 1, Kremser Landstraße 40, 3100 St. Pölten, Austria
| | - Georg Strebinger
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria; (S.G.)
- Obesity Research Unit, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Stephan Zandanell
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria; (S.G.)
- Obesity Research Unit, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Lars Stechemesser
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria; (S.G.)
- Obesity Research Unit, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
| | - Bernhard Paulweber
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria; (S.G.)
| | - Bernhard Iglseder
- Department of Geriatric Medicine, Christian Doppler University Hospital, Paracelsus Medical University, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria
| | - Eugen Trinka
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Centre for Cognitive Neuroscience, Affiliated Member of the European Reference Network EpiCARE, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria
| | - Vanessa Frey
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Centre for Cognitive Neuroscience, Affiliated Member of the European Reference Network EpiCARE, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria
| | - Patrick Langthaler
- Department of Neurology, Christian Doppler University Hospital, Paracelsus Medical University and Centre for Cognitive Neuroscience, Affiliated Member of the European Reference Network EpiCARE, Ignaz-Harrer-Straße 79, 5020 Salzburg, Austria
| | - Georg Semmler
- Vienna Hepatic Hemodynamic Lab, Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Via Francesco Forza 35, 20122 Milan, Italy;
- Precision Medicine, Biological Resource Center Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Via Francesco Sforza, 35, 20122 Milan, Italy
| | - Elena Corradini
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy;
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena Policlinico, 41124 Modena, Italy
| | - Christian Datz
- Department of Medicine, General Hospital Oberndorf, Teaching Hospital of the Paracelsus Medical University, Paracelsusstraße 37, 5110 Oberndorf, Austria
| | - Elmar Aigner
- Department of Internal Medicine I, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria; (S.G.)
- Obesity Research Unit, Paracelsus Medical University, Müllner Hauptstrasse 48, 5020 Salzburg, Austria
| |
Collapse
|
5
|
Yue F, Shi Y, Wu S, Xing L, He D, Wei L, Qiu A, Russell R, Zhang D. Metformin alleviates hepatic iron overload and ferroptosis through AMPK-ferroportin pathway in HFD-induced NAFLD. iScience 2023; 26:108560. [PMID: 38089577 PMCID: PMC10711470 DOI: 10.1016/j.isci.2023.108560] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 10/21/2023] [Accepted: 11/20/2023] [Indexed: 01/04/2025] Open
Abstract
Metformin prevents progression of non-alcoholic fatty liver disease (NAFLD). However, the potential mechanism is not entirely understood. Ferroptosis, a recently recognized nonapoptotic form of regulated cell death, has been reported to be involved in the pathogenesis of NAFLD. Here, we investigated the effects of metformin on ferroptosis and its potential mechanism in NAFLD. We found that metformin prevented the progression of NAFLD, and alleviated hepatic iron overload (HIO), ferroptosis and upregulated ferroportin (FPN) expression in vivo and in vitro. Mechanically, metformin reduced the lysosomal degradation pathway of FPN through activation AMPK, thus upregulated the expression of FPN protein, alleviated HIO and ferroptosis, and prevented progression of NAFLD. These findings discover a mechanism of metformin, suggesting that targeting FPN may have the therapeutic potential for treating NAFLD and related disorders.
Collapse
Affiliation(s)
- Fangzhi Yue
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Key Laboratory of Diabetes Immunology, Ministry of Education, Central South University, Changsha 410011, Hunan, China
| | - Ying Shi
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Shanyu Wu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Lin Xing
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Dan He
- College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Lin Wei
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Anqi Qiu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Ryan Russell
- Department of Health and Human Performance, College of Health Professions, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Dongmei Zhang
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
- Hunan Engineering Research Center for Obesity and its Metabolic Complications, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
6
|
Moris W, Verbeek J, Bakers FC, Rombout-Sestrienkova E, Innocenti F, Masclee AAM, Koek GH, van Deursen CTBM. Hyperferritinemia and liver iron content determined with MRI: Reintroduction of the liver iron index. Clin Res Hepatol Gastroenterol 2023; 47:102224. [PMID: 37813276 DOI: 10.1016/j.clinre.2023.102224] [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: 01/09/2023] [Revised: 09/19/2023] [Accepted: 10/02/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Hyperferritinemia is found in around 12 % of the general population. Analyzing the cause can be difficult. In case of doubt about the presence of major iron overload most guidelines advice to perform a MRI as a reliable non-invasive marker to measure liver iron concentration (LIC). In general, a LIC of ≥ 36 µmol/g dw is considered the be elevated however in hyperferritinemia associated with, for example, obesity or alcohol (over)consumption the LIC can be ≥ 36 µmol/g dw in abscence of major iron overload. So, unfortunately a clear cut-off value to differentiate iron overload from normal iron content is lacking. Previously the liver iron index (LII) (LIC measured in liver biopsy (LIC-b)/age (years)), was introduced to differentiate between patients with major (LII ≥ 2) and minor or no iron overload (LII < 2). Based on the good correlation between the LIC-b and LIC determined with MRI (LIC-MRI), our goal was to investigate whether a LII_MRI ≥ 2 is a good indicator of major iron overload, reflected by a significantly higher amount of iron needed to be mobilized to reach iron depletion. METHODS We compared the amount of mobilized iron to reach depletion and inflammation-related characteristics in two groups: LII-MRI ≥ 2 versus LII-MRI <2 in 92 hyperferritinemia patients who underwent HFE genotyping and MRI-LIC determination. RESULTS Significantly more iron needed to be mobilized to reach iron depletion in the LII ≥ 2 group (mean 4741, SD ± 4135 mg) versus the LII-MRI <2 group (mean 1340, SD ± 533 mg), P < 0.001. Furthermore, hyperferritinemia in LII-MRI < 2 patients was more often related to components of the metabolic syndrome while hyperferritinemia in LII-MRI ≥ 2 patients was more often related to HFE mutations. ROC curve analysis showed good performance of LII =2 as cut-off value. However the calculations showed that the optimal cut-off for the LII = 3.4. CONCLUSION The LII-MRI with a cut-off value of 2 is an effective method to differentiate major from minor iron overload in patients with hyperferritinemia. But the LII-MRI = 3.4 seems a more promising diagnostic test for major iron overload.
Collapse
Affiliation(s)
- Wenke Moris
- Department of Internal Medicine Gastroenterology and Clinical Geriatrics, Zuyderland Medical Center, Sittard-Geleen, the Netherlands; Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM) University Maastricht, the Netherlands
| | - Jef Verbeek
- Department of Gastroenterology & Hepatology, University Hospitals KU Leuven, Leuven, Belgium
| | - Frans C Bakers
- Department of radiology and nuclear medicine, Maastricht University Medical Centre, Maastricht, the Netherlands
| | - Eva Rombout-Sestrienkova
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, the Netherlands; Department of Transfusion Medicine, Sanquin Blood Supply, Amsterdam, the Netherlands
| | - Francesco Innocenti
- Department of Methodology & Statistics, Care and Public Health Research Institute (CAPHRI), University Maastricht, the Netherlands
| | - Ad A M Masclee
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM) University Maastricht, the Netherlands
| | - Ger H Koek
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, the Netherlands; School of Nutrition and Translational Research in Metabolism (NUTRIM) University Maastricht, the Netherlands
| | - Cees Th B M van Deursen
- Department of Internal Medicine Gastroenterology and Clinical Geriatrics, Zuyderland Medical Center, Sittard-Geleen, the Netherlands; Department of Internal Medicine, Division of Gastroenterology and Hepatology, Maastricht University Medical Centre+, Maastricht, the Netherlands.
| |
Collapse
|
7
|
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: 30] [Impact Index Per Article: 15.0] [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
|
8
|
Charlebois E, Pantopoulos K. Nutritional Aspects of Iron in Health and Disease. Nutrients 2023; 15:2441. [PMID: 37299408 PMCID: PMC10254751 DOI: 10.3390/nu15112441] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Dietary iron assimilation is critical for health and essential to prevent iron-deficient states and related comorbidities, such as anemia. The bioavailability of iron is generally low, while its absorption and metabolism are tightly controlled to satisfy metabolic needs and prevent toxicity of excessive iron accumulation. Iron entry into the bloodstream is limited by hepcidin, the iron regulatory hormone. Hepcidin deficiency due to loss-of-function mutations in upstream gene regulators causes hereditary hemochromatosis, an endocrine disorder of iron overload characterized by chronic hyperabsorption of dietary iron, with deleterious clinical complications if untreated. The impact of high dietary iron intake and elevated body iron stores in the general population is not well understood. Herein, we summarize epidemiological data suggesting that a high intake of heme iron, which is abundant in meat products, poses a risk factor for metabolic syndrome pathologies, cardiovascular diseases, and some cancers. We discuss the clinical relevance and potential limitations of data from cohort studies, as well as the need to establish causality and elucidate molecular mechanisms.
Collapse
Affiliation(s)
- Edouard Charlebois
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC H3T 1E2, Canada;
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
9
|
Valenti L, Corradini E, Adams LA, Aigner E, Alqahtani S, Arrese M, Bardou-Jacquet E, Bugianesi E, Fernandez-Real JM, Girelli D, Hagström H, Henninger B, Kowdley K, Ligabue G, McClain D, Lainé F, Miyanishi K, Muckenthaler MU, Pagani A, Pedrotti P, Pietrangelo A, Prati D, Ryan JD, Silvestri L, Spearman CW, Stål P, Tsochatzis EA, Vinchi F, Zheng MH, Zoller H. Consensus Statement on the definition and classification of metabolic hyperferritinaemia. Nat Rev Endocrinol 2023; 19:299-310. [PMID: 36805052 PMCID: PMC9936492 DOI: 10.1038/s41574-023-00807-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/19/2023]
Abstract
Hyperferritinaemia is a common laboratory finding that is often associated with metabolic dysfunction and fatty liver. Metabolic hyperferritinaemia reflects alterations in iron metabolism that facilitate iron accumulation in the body and is associated with an increased risk of cardiometabolic and liver diseases. Genetic variants that modulate iron homeostasis and tissue levels of iron are the main determinants of serum levels of ferritin in individuals with metabolic dysfunction, raising the hypothesis that iron accumulation might be implicated in the pathogenesis of insulin resistance and the related organ damage. However, validated criteria for the non-invasive diagnosis of metabolic hyperferritinaemia and the staging of iron overload are still lacking, and there is no clear evidence of a benefit for iron depletion therapy. Here, we provide an overview of the literature on the relationship between hyperferritinaemia and iron accumulation in individuals with metabolic dysfunction, and on the associated clinical outcomes. We propose an updated definition and a provisional staging system for metabolic hyperferritinaemia, which has been agreed on by a multidisciplinary global panel of expert researchers. The goal is to foster studies into the epidemiology, genetics, pathophysiology, clinical relevance and treatment of metabolic hyperferritinaemia, for which we provide suggestions on the main unmet needs, optimal design and clinically relevant outcomes.
Collapse
Affiliation(s)
- Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Biological Resource Center and Precision Medicine Lab, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
| | - Elena Corradini
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy.
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy.
| | - Leon A Adams
- Medical School, University of Western Australia, Perth, Australia
| | - Elmar Aigner
- First Department of Medicine, University Clinic Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Saleh Alqahtani
- Royal Clinics and Gastroenterology and Hepatology, King Faisal Specialist Hospital & Research Centre, Riyadh, Kingdom of Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Edouard Bardou-Jacquet
- University of Rennes, UMR1241, CHU Rennes, National Reference Center for Hemochromatosis and iron metabolism disorder, INSERM CIC1414, Rennes, France
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Turin, Italy
| | - Jose-Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Domenico Girelli
- Section of Internal Medicine, Department of Medicine, University of Verona, Policlinico Giambattista Rossi, Verona, Italy
| | - Hannes Hagström
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kris Kowdley
- Liver Institute Northwest, Seattle, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| | - Guido Ligabue
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Division of Radiology, Ospedale di Sassuolo S.p.A, Sassuolo, Modena, Italy
| | - Donald McClain
- Wake Forest School of Medicine, Winston Salem, NC, USA
- Department of Veterans Affairs, Salisbury, NC, USA
| | - Fabrice Lainé
- INSERM CIC1414, Liver Unit, CHU Rennes, Rennes, France
| | - Koji Miyanishi
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Martina U Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
- Center for Molecular Translational Iron Research, Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Heidelberg, Germany
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Pedrotti
- Laboratorio di RM Cardiaca Cardiologia 4, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy
| | - Daniele Prati
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - John D Ryan
- Hepatology Unit, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - C Wendy Spearman
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Per Stål
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley F.Kimball Research Institute, New York Blood Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Heinz Zoller
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
- Doppler Laboratory on Iron and Phosphate Biology, Innsbruck, Austria
| |
Collapse
|
10
|
Reboucas P, Fillebeen C, Botta A, Cleverdon R, Steele AP, Richard V, Zahedi RP, Borchers CH, Burelle Y, Hawke TJ, Pantopoulos K, Sweeney G. Discovery-Based Proteomics Identify Skeletal Muscle Mitochondrial Alterations as an Early Metabolic Defect in a Mouse Model of β-Thalassemia. Int J Mol Sci 2023; 24:ijms24054402. [PMID: 36901833 PMCID: PMC10002226 DOI: 10.3390/ijms24054402] [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: 01/17/2023] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
Although metabolic complications are common in thalassemia patients, there is still an unmet need to better understand underlying mechanisms. We used unbiased global proteomics to reveal molecular differences between the th3/+ mouse model of thalassemia and wild-type control animals focusing on skeletal muscles at 8 weeks of age. Our data point toward a significantly impaired mitochondrial oxidative phosphorylation. Furthermore, we observed a shift from oxidative fibre types toward more glycolytic fibre types in these animals, which was further supported by larger fibre-type cross-sectional areas in the more oxidative type fibres (type I/type IIa/type IIax hybrid). We also observed an increase in capillary density in th3/+ mice, indicative of a compensatory response. Western blotting for mitochondrial oxidative phosphorylation complex proteins and PCR analysis of mitochondrial genes indicated reduced mitochondrial content in the skeletal muscle but not the hearts of th3/+ mice. The phenotypic manifestation of these alterations was a small but significant reduction in glucose handling capacity. Overall, this study identified many important alterations in the proteome of th3/+ mice, amongst which mitochondrial defects leading to skeletal muscle remodelling and metabolic dysfunction were paramount.
Collapse
Affiliation(s)
| | - Carine Fillebeen
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
| | - Amy Botta
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Riley Cleverdon
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Alexandra P. Steele
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Vincent Richard
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute, Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
| | - René P. Zahedi
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute, Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Christoph H. Borchers
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
- Segal Cancer Proteomics Centre, Lady Davis Institute, Gerald Bronfman Department of Oncology, Jewish General Hospital, McGill University, Montreal, QC H3T 1E2, Canada
- Manitoba Centre for Proteomics & Systems Biology, University of Manitoba, Winnipeg, MB R3E 3P4, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Thomas J. Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Department of Medicine, McGill University, Montreal, QC H3T 1E2, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
- Correspondence: ; Tel.: +1-416-736-2100 (ext. 66635)
| |
Collapse
|
11
|
Kouroumalis E, Tsomidis I, Voumvouraki A. Iron as a therapeutic target in chronic liver disease. World J Gastroenterol 2023; 29:616-655. [PMID: 36742167 PMCID: PMC9896614 DOI: 10.3748/wjg.v29.i4.616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 11/03/2022] [Accepted: 12/31/2022] [Indexed: 01/20/2023] Open
Abstract
It was clearly realized more than 50 years ago that iron deposition in the liver may be a critical factor in the development and progression of liver disease. The recent clarification of ferroptosis as a specific form of regulated hepatocyte death different from apoptosis and the description of ferritinophagy as a specific variation of autophagy prompted detailed investigations on the association of iron and the liver. In this review, we will present a brief discussion of iron absorption and handling by the liver with emphasis on the role of liver macrophages and the significance of the iron regulators hepcidin, transferrin, and ferritin in iron homeostasis. The regulation of ferroptosis by endogenous and exogenous mod-ulators will be examined. Furthermore, the involvement of iron and ferroptosis in various liver diseases including alcoholic and non-alcoholic liver disease, chronic hepatitis B and C, liver fibrosis, and hepatocellular carcinoma (HCC) will be analyzed. Finally, experimental and clinical results following interventions to reduce iron deposition and the promising manipulation of ferroptosis will be presented. Most liver diseases will be benefited by ferroptosis inhibition using exogenous inhibitors with the notable exception of HCC, where induction of ferroptosis is the desired effect. Current evidence mostly stems from in vitro and in vivo experimental studies and the need for well-designed future clinical trials is warranted.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Liver Research Laboratory, University of Crete Medical School, Heraklion 71003, Greece
| | - Ioannis Tsomidis
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| | - Argyro Voumvouraki
- First Department of Internal Medicine, AHEPA University Hospital, Thessaloniki 54621, Greece
| |
Collapse
|
12
|
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
|
13
|
Baldelli E, Lonardo A. Digging the metabolic roots of NASH up. LIFE METABOLISM 2022; 1:203-204. [DOI: 10.1093/lifemeta/loac036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Affiliation(s)
- Enrica Baldelli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia , Modena , Italy
| | - Amedeo Lonardo
- Azienda Ospedaliero-Universitaria di Modena—Department of Internal Medicine—Ospedale Civile di Baggiovara , Modena , Italy
| |
Collapse
|
14
|
He H, Liao S, Zeng Y, Liang L, Chen J, Tao C. Causal relationships between metabolic-associated fatty liver disease and iron status: Two-sample Mendelian randomization. Liver Int 2022; 42:2759-2768. [PMID: 36226474 DOI: 10.1111/liv.15455] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/10/2022] [Accepted: 10/12/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND & AIMS Dysregulated iron homeostasis plays an important role in the hepatic manifestation of metabolic-associated fatty liver disease (MAFLD). We investigated the causal effects of five iron metabolism markers, regular iron supplementation and MAFLD risk. METHODS Genetic summary statistics were obtained from open genome-wide association study databases. Two-sample bidirectional Mendelian randomization analysis was performed to estimate the causal effect between iron status and MAFLD, including Mendelian randomization inverse-variance weighted, weighted median methods and Mendelian randomization-Egger regression. The Mendelian randomization-PRESSO outlier test, Cochran's Q test and Mendelian randomization-Egger regression were used to assess outliers, heterogeneity and pleiotropy respectively. RESULTS Mendelian randomization inverse-variance weighted results showed that the genetically predicted per standard deviation increase in liver iron (Data set 2: odds ratio 1.193, 95% confidence interval [CI] 1.074-1.326, p = .001) was associated with an increased MAFLD risk, consistent with the weighted median estimates and Mendelian randomization-Egger regression, although Data set 1 was not significant. Mendelian randomization inverse-variance weighted analysis showed that genetically predicted MAFLD was significantly associated with increased serum ferritin levels in both datasets (Dataset 1: β = .038, 95% CI = .014 to .062, p = .002; Dataset 2: β = .081, 95% CI = .025 to .136, p = .004), and a similar result was observed with the weighted median methods for Dataset 2 instead of Mendelian randomization-Egger regression. CONCLUSIONS This study uncovered genetically predicted causal associations between iron metabolism status and MAFLD. These findings underscore the need for improved guidelines for managing MAFLD risk by emphasizing hepatic iron levels as a risk factor and ferritin levels as a prognostic factor.
Collapse
Affiliation(s)
- He He
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Shenling Liao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Yuping Zeng
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Libo Liang
- Department of International Medical Centre, West China Hospital, Sichuan University, Sichuan, China
| | - Jie Chen
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Sichuan, China
| | - Chuanmin Tao
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Sichuan, China
| |
Collapse
|
15
|
Elumalai S, Karunakaran U, Moon JS, Won KC. Ferroptosis Signaling in Pancreatic β-Cells: Novel Insights & Therapeutic Targeting. Int J Mol Sci 2022; 23:13679. [PMID: 36430158 PMCID: PMC9690757 DOI: 10.3390/ijms232213679] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/02/2022] [Accepted: 11/05/2022] [Indexed: 11/10/2022] Open
Abstract
Metabolic stress impairs pancreatic β-cell survival and function in diabetes. Although the pathophysiology of metabolic stress is complex, aberrant tissue damage and β-cell death are brought on by an imbalance in redox equilibrium due to insufficient levels of endogenous antioxidant expression in β-cells. The vulnerability of β-cells to oxidative damage caused by iron accumulation has been linked to contributory β-cell ferroptotic-like malfunction under diabetogenic settings. Here, we take into account recent findings on how iron metabolism contributes to the deregulation of the redox response in diabetic conditions as well as the ferroptotic-like malfunction in the pancreatic β-cells, which may offer insights for deciphering the pathomechanisms and formulating plans for the treatment or prevention of metabolic stress brought on by β-cell failure.
Collapse
Affiliation(s)
- Suma Elumalai
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
| | - Udayakumar Karunakaran
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
| | - Jun-Sung Moon
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
| | - Kyu-Chang Won
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu 42415, Korea
- Department of Internal Medicine, College of Medicine, Yeungnam University, Daegu 42415, Korea
| |
Collapse
|
16
|
Chen H, Zhao W, Yan X, Huang T, Yang A. Overexpression of Hepcidin Alleviates Steatohepatitis and Fibrosis in a Diet-induced Nonalcoholic Steatohepatitis. J Clin Transl Hepatol 2022; 10:577-588. [PMID: 36062292 PMCID: PMC9396326 DOI: 10.14218/jcth.2021.00289] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/22/2021] [Accepted: 09/28/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS Iron overload can contribute to the progression of nonalcoholic fatty liver disease (NAFLD) to nonalcoholic steatohepatitis (NASH). Hepcidin (Hamp), which is primarily synthesized in hepatocytes, is a key regulator of iron metabolism. However, the role of Hamp in NASH remains unclear. Therefore, we aimed to elucidate the role of Hamp in the pathophysiology of NASH. METHODS Male mice were fed a choline-deficient L-amino acid-defined (CDAA) diet for 16 weeks to establish the mouse NASH model. A choline-supplemented amino acid-defined (CSAA) diet was used as the control diet. Recombinant adeno-associated virus genome 2 serotype 8 vector expressing Hamp (rAAV2/8-Hamp) or its negative control (rAAV2/8-NC) was administered intravenously at week 8 of either the CDAA or CSAA diet. RESULTS rAAV2/8-Hamp treatment markedly decreased liver weight and improved hepatic steatosis in the CDAA-fed mice, accompanied by changes in lipogenesis-related genes and adiponectin expression. Compared with the control group, rAAV2/8-Hamp therapy attenuated liver damage, with mice exhibiting reduced histological NAFLD inflammation and fibrosis, as well as lower levels of liver enzymes. Moreover, α-smooth muscle actin-positive activated hepatic stellate cells (HSCs) and CD68-postive macrophages increased in number in the CDAA-fed mice, which was reversed by rAAV2/8-Hamp treatment. Consistent with the in vivo findings, overexpression of Hamp increased adiponectin expression in hepatocytes and Hamp treatment inhibited HSC activation. CONCLUSIONS Overexpression of Hamp using rAAV2/8-Hamp robustly attenuated liver steatohepatitis, inflammation, and fibrosis in an animal model of NASH, suggesting a potential therapeutic role for Hamp.
Collapse
Affiliation(s)
- Hui Chen
- Digestive Department, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Correspondence to: Hui Chen, Digestive Department, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan Road, Shijingshan District, Beijing 100043, China. Tel: +86-10-51718484, Fax: +86-10-83165944, E-mail: . Aiting Yang, Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China. ORCID: https://orcid.org/0000-0002-5671-696X. Tel: +86-10-63139311, Fax: +86-10-83165944, E-mail:
| | - Wenshan Zhao
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Xuzhen Yan
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Tao Huang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Aiting Yang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- National Clinical Research Center of Digestive Diseases, Beijing, China
- Beijing Clinical Medicine Institute, Beijing, China
- Correspondence to: Hui Chen, Digestive Department, Beijing Chaoyang Hospital, Capital Medical University, No. 5 Jingyuan Road, Shijingshan District, Beijing 100043, China. Tel: +86-10-51718484, Fax: +86-10-83165944, E-mail: . Aiting Yang, Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong’an Road, Xicheng District, Beijing 100050, China. ORCID: https://orcid.org/0000-0002-5671-696X. Tel: +86-10-63139311, Fax: +86-10-83165944, E-mail:
| |
Collapse
|
17
|
Kim HY, Yoo YH. The Role of STAMP2 in Pathogenesis of Chronic Diseases Focusing on Nonalcoholic Fatty Liver Disease: A Review. Biomedicines 2022; 10:biomedicines10092082. [PMID: 36140186 PMCID: PMC9495648 DOI: 10.3390/biomedicines10092082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 11/16/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a major health issue. NAFLD can progress from simple hepatic steatosis to nonalcoholic steatohepatitis (NASH). NASH can progress to cirrhosis or hepatocellular carcinoma. Unfortunately, there is no currently approved pharmacologic therapy for NAFLD patients. The six transmembrane protein of prostate 2 (STAMP2), a metalloreductase involved in iron and copper homeostasis, is well known for its critical role in the coordination of glucose/lipid metabolism and inflammation in metabolic tissues. We previously demonstrated that hepatic STAMP2 could be a suitable therapeutic target for NAFLD. In this review, we discuss the emerging role of STAMP2 in the dysregulation of iron metabolism events leading to NAFLD and suggest therapeutic strategies targeting STAMP2.
Collapse
|
18
|
Xiong H, Zhang C, Han L, Xu T, Saeed K, Han J, Liu J, Klaassen CD, Gonzalez FJ, Lu Y, Zhang Y. Suppressed farnesoid X receptor by iron overload in mice and humans potentiates iron-induced hepatotoxicity. Hepatology 2022; 76:387-403. [PMID: 34870866 DOI: 10.1002/hep.32270] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 11/23/2021] [Accepted: 12/04/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND AND AIMS Iron overload (IO) is a frequent finding in the general population. As the major iron storage site, the liver is subject to iron toxicity. Farnesoid X receptor (FXR) regulates bile acid metabolism and is implicated in various liver diseases. We aimed to determine whether FXR plays a role in regulating iron hepatotoxicity. APPROACH AND RESULTS Human and mouse hepatocytes were treated with ferric ammonium citrate or iron dextran (FeDx). Mice were orally administered ferrous sulfate or injected i.p. with FeDx. Wild-type and Fxr-/- mice were fed an iron-rich diet for 1 or 5 weeks. Mice fed an iron-rich diet were coadministered the FXR agonist, GW4064. Forced expression of FXR was carried out with recombinant adeno-associated virus 1 week before iron-rich diet feeding. Serum levels of bile acids and fibroblast growth factor 19 (FGF19) were quantified in adults with hyperferritinemia and children with β-thalassemia. The data demonstrated that iron suppressed FXR expression and signaling in human and mouse hepatocytes as well as in mouse liver and intestine. FXR deficiency potentiated iron hepatotoxicity, accompanied with hepatic steatosis as well as dysregulated iron and bile acid homeostasis. FXR negatively regulated iron-regulatory proteins 1 and 2 and prevented hepatic iron accumulation. Forced FXR expression and ligand activation significantly suppressed iron hepatotoxicity in iron-fed mice. The FXR agonist, GW4064, almost completely restored dysregulated bile acid signaling and metabolic syndrome in iron-fed mice. Conjugated primary bile acids were increased and FGF19 was decreased in serum of adults with hyperferritinemia and children with β-thalassemia. CONCLUSIONS FXR plays a pivotal role in regulating iron homeostasis and protects mice against iron hepatotoxicity. Targeting FXR may represent a therapeutic strategy for IO-associated chronic liver diseases.
Collapse
Affiliation(s)
- Hui Xiong
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Chunze Zhang
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Lifeng Han
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Tong Xu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Khawar Saeed
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jing Han
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jing Liu
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Curtis D Klaassen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Frank J Gonzalez
- Laboratory of Metabolism, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland, USA
| | - Yuanfu Lu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Youcai Zhang
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
19
|
Tsuchiya H. Iron-Induced Hepatocarcinogenesis—Preventive Effects of Nutrients. Front Oncol 2022; 12:940552. [PMID: 35832553 PMCID: PMC9271801 DOI: 10.3389/fonc.2022.940552] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/03/2022] [Indexed: 01/10/2023] Open
Abstract
The liver is a primary organ that stores body iron, and plays a central role in the regulation of iron homeostasis. Hepatic iron overload (HIO) is a prevalent feature among patients with chronic liver diseases (CLDs), including alcoholic/nonalcoholic liver diseases and hepatitis C. HIO is suggested to promote the progression toward hepatocellular carcinoma because of the pro-oxidant nature of iron. Iron metabolism is tightly regulated by various factors, such as hepcidin and ferroportin, in healthy individuals to protect the liver from such deteriorative effects. However, their intrinsic expressions or functions are frequently compromised in patients with HIO. Thus, various nutrients have been reported to regulate hepatic iron metabolism and protect the liver from iron-induced damage. These nutrients are beneficial in HIO-associated CLD treatment and eventually prevent iron-mediated hepatocarcinogenesis. This mini-review aimed to discuss the mechanisms and hepatocarcinogenic risk of HIO in patients with CLDs. Moreover, nutrients that hold the potential to prevent iron-induced hepatocarcinogenesis are summarized.
Collapse
|
20
|
Trasolini R, Cox B, Galts C, Yoshida EM, Marquez V. Elevated serum ferritin in non-alcoholic fatty liver disease is not predictive of fibrosis. CANADIAN LIVER JOURNAL 2022; 5:152-159. [PMID: 35991481 PMCID: PMC9236592 DOI: 10.3138/canlivj-2021-0002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 10/24/2021] [Accepted: 11/06/2021] [Indexed: 09/22/2023]
Abstract
BACKGROUND Non-alcoholic fatty liver disease (NAFLD) is common with widely ranging severity. Non-invasive risk scores for risk stratification are recommended but misclassify a significant proportion of patients. In situations where non-invasive risk scores do not provide guidance, referral is typically made to a Hepatologist for transient elastography or liver biopsy. Serum ferritin is elevated in many patients with NAFLD related to dysmetabolic and inflammatory hyperferritinemia. Ferritin is widely available and part of a standard workup for chronic liver disease. METHODS To explore the association of ferritin and risk of fibrosis in NAFLD, we reviewed patients diagnosed with NAFLD at the hepatology clinic of the Vancouver General Hospital between the years of 2015 and 2018. We collected data on 317 patients retrospectively assessing for a relationship between serum ferritin and elastography score. RESULTS Two hundred twenty-four patients were included in the final analysis. Median ferritin was 145 µg/L (IQR 62-311). Median liver stiffness was 5.2 kPa with 14.3% of patients having liver stiffness ≥8.7 kPa and 17.4% ≥ 8.0 kPa. ROC curve analysis using a liver stiffness ≥8.0 kPa as a cutoff for F2 fibrosis showed an AUROC of 0.54 for serum ferritin levels. At a cut-off of both 300 µg/L; and 450 µg/L median liver stiffness did not differ significantly in those with ferritin above the cutoff (ferritin ≥300 µg/L; p = 0.099, ferritin ≥450 µg/L; p = 0.12). Ferritin was significantly higher in male patients (198 versus 91 µg/L; p = 0.0001). There was a weak linear association between AST and ferritin levels. CONCLUSION In this cohort of 224 patients with NAFLD, serum ferritin was not predictive of significant liver fibrosis.
Collapse
Affiliation(s)
- Roberto Trasolini
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Gastroenterology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Ben Cox
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ciaran Galts
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Eric M Yoshida
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Gastroenterology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| | - Vladimir Marquez
- Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
- Division of Gastroenterology, Vancouver General Hospital, Vancouver, British Columbia, Canada
| |
Collapse
|
21
|
Quee FA, Peffer K, ter Braake AD, van den Hurk K. Cardiovascular benefits for blood donors? A systematic review. Transfus Med Rev 2022; 36:143-151. [DOI: 10.1016/j.tmrv.2022.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 10/18/2022]
|
22
|
Wang K, Yang F, Zhang P, Yang Y, Jiang L. Genetic effects of iron levels on liver injury and risk of liver diseases: A two-sample Mendelian randomization analysis. Front Nutr 2022; 9:964163. [PMID: 36185655 PMCID: PMC9523310 DOI: 10.3389/fnut.2022.964163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background and aims Although iron homeostasis has been associated with liver function in many observational studies, the causality in this relationship remains unclear. By using Mendelian Randomization analyses, we aimed to evaluate the genetic effects of increased systemic iron levels on the risk of liver injury and various liver diseases. Moreover, in light of the sex-dependent iron regulation in human beings, we further estimated the sex-specific effect of iron levels in liver diseases. Methods Independent single nucleotide polymorphisms associated with systemic iron status (including four indicators) at the genome-wide significance level from the Genetics of Iron Status (GIS) Consortium were selected as instrumental variables. Summary data for six liver function biomarkers and five liver diseases were obtained from the UK Biobank, the Estonian Biobank, the eMERGE network, and FinnGen consortium. Mendelian Randomization assessment of the effect of iron on liver function and liver diseases was conducted. Results Genetically predicted iron levels were positively and significantly associated with an increased risk of different dimensions of liver injury. Furthermore, increased iron status posed hazardous effects on non-alcoholic fatty liver disease, alcoholic liver disease, and liver fibrosis/cirrhosis. Sex-stratified analyses indicated that the hepatoxic role of iron might exist in NAFLD and liver fibrosis/cirrhosis development among men. No significantly causal relationship was found between iron status and viral hepatitis. Conclusion Our study adds to current knowledge on the genetic role of iron in the risk of liver injury and related liver diseases, which provides clinical and public health implications for liver disease prevention as iron status can be modified.
Collapse
Affiliation(s)
- Kai Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China.,Eye Center of the Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Zhejiang Provincial Key Lab of Ophthalmology, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Fangkun Yang
- Department of Cardiology, Ningbo First Hospital, School of Medicine, Zhejiang University, Ningbo, China
| | - Pengcheng Zhang
- Department of Gastroenterology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yang Yang
- Department of Radiation Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Li Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Hangzhou Normal University, Hangzhou, China
| |
Collapse
|
23
|
Viveiros A, Schaefer B, Panzer M, Henninger B, Plaikner M, Kremser C, Franke A, Franzenburg S, Hoeppner MP, Stauder R, Janecke A, Tilg H, Zoller H. MRI-Based Iron Phenotyping and Patient Selection for Next-Generation Sequencing of Non-Homeostatic Iron Regulator Hemochromatosis Genes. Hepatology 2021; 74:2424-2435. [PMID: 34048062 PMCID: PMC8596846 DOI: 10.1002/hep.31982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/13/2021] [Accepted: 05/13/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS High serum ferritin is frequent among patients with chronic liver disease and commonly associated with hepatic iron overload. Genetic causes of high liver iron include homozygosity for the p.Cys282Tyr variant in homeostatic iron regulator (HFE) and rare variants in non-HFE genes. The aims of the present study were to describe the landscape and frequency of mutations in hemochromatosis genes and determine whether patient selection by noninvasive hepatic iron quantification using MRI improves the diagnostic yield of next-generation sequencing (NGS) in patients with hyperferritinemia. APPROACH AND RESULTS A cohort of 410 unselected liver clinic patients with high serum ferritin (defined as ≥200 μg/L for women and ≥300 μg/L for men) was investigated by HFE genotyping and abdominal MRI R2*. Forty-one (10%) patients were homozygous for the p.Cys282Tyr variant in HFE. Of the remaining 369 patients, 256 (69%) had high transferrin saturation (TSAT; ≥45%) and 199 (53%) had confirmed hepatic iron overload (liver R2* ≥70 s-1 ). NGS of hemochromatosis genes was carried out in 180 patients with hepatic iron overload, and likely pathogenic variants were identified in 68 of 180 (38%) patients, mainly in HFE (79%), ceruloplasmin (25%), and transferrin receptor 2 (19%). Low spleen iron (R2* <50 s-1 ), but not TSAT, was significantly associated with the presence of mutations. In 167 patients (93%), no monogenic cause of hepatic iron overload could be identified. CONCLUSIONS In patients without homozygosity for p.Cys282Tyr, coincident pathogenic variants in HFE and non-HFE genes could explain hyperferritinemia with hepatic iron overload in a subset of patients. Unlike HFE hemochromatosis, this type of polygenic hepatic iron overload presents with variable TSAT. High ferritin in blood is an indicator of the iron storage disease, hemochromatosis. A simple genetic test establishes this diagnosis in the majority of patients affected. MRI of the abdomen can guide further genetic testing.
Collapse
Affiliation(s)
- André Viveiros
- Department of Medicine I and Christian Doppler Laboratory on Iron and Phosphate BiologyMedical University of InnsbruckInnsbruckAustria
| | - Benedikt Schaefer
- Department of Medicine I and Christian Doppler Laboratory on Iron and Phosphate BiologyMedical University of InnsbruckInnsbruckAustria
| | - Marlene Panzer
- Department of Medicine I and Christian Doppler Laboratory on Iron and Phosphate BiologyMedical University of InnsbruckInnsbruckAustria
| | | | - Michaela Plaikner
- Department of RadiologyMedical University of InnsbruckInnsbruckAustria
| | - Christian Kremser
- Department of RadiologyMedical University of InnsbruckInnsbruckAustria
| | - André Franke
- Institute of Clinical Molecular Biology (IKMB)Kiel UniversityKielGermany
| | - Sören Franzenburg
- Institute of Clinical Molecular Biology (IKMB)Kiel UniversityKielGermany
| | - Marc P. Hoeppner
- Institute of Clinical Molecular Biology (IKMB)Kiel UniversityKielGermany
| | - Reinhard Stauder
- Department of Medicine VMedical University of InnsbruckInnsbruckAustria
| | - Andreas Janecke
- Department of PediatricsMedical University of InnsbruckInnsbruckAustria
- Department of GeneticsMedical University of InnsbruckInnsbruckAustria
| | - Herbert Tilg
- Department of Medicine I and Christian Doppler Laboratory on Iron and Phosphate BiologyMedical University of InnsbruckInnsbruckAustria
| | - Heinz Zoller
- Department of Medicine I and Christian Doppler Laboratory on Iron and Phosphate BiologyMedical University of InnsbruckInnsbruckAustria
| |
Collapse
|
24
|
Marku A, Galli A, Marciani P, Dule N, Perego C, Castagna M. Iron Metabolism in Pancreatic Beta-Cell Function and Dysfunction. Cells 2021; 10:2841. [PMID: 34831062 PMCID: PMC8616520 DOI: 10.3390/cells10112841] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/15/2021] [Accepted: 10/19/2021] [Indexed: 12/26/2022] Open
Abstract
Iron is an essential element involved in a variety of physiological functions. In the pancreatic beta-cells, being part of Fe-S cluster proteins, it is necessary for the correct insulin synthesis and processing. In the mitochondria, as a component of the respiratory chain, it allows the production of ATP and reactive oxygen species (ROS) that trigger beta-cell depolarization and potentiate the calcium-dependent insulin release. Iron cellular content must be finely tuned to ensure the normal supply but also to prevent overloading. Indeed, due to the high reactivity with oxygen and the formation of free radicals, iron excess may cause oxidative damage of cells that are extremely vulnerable to this condition because the normal elevated ROS production and the paucity in antioxidant enzyme activities. The aim of the present review is to provide insights into the mechanisms responsible for iron homeostasis in beta-cells, describing how alteration of these processes has been related to beta-cell damage and failure. Defects in iron-storing or -chaperoning proteins have been detected in diabetic conditions; therefore, the control of iron metabolism in these cells deserves further investigation as a promising target for the development of new disease treatments.
Collapse
Affiliation(s)
| | | | | | | | - Carla Perego
- Department of Excellence Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Trentacoste, 22134 Milano, Italy; (A.M.); (A.G.); (P.M.); (N.D.)
| | - Michela Castagna
- Department of Excellence Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Via Trentacoste, 22134 Milano, Italy; (A.M.); (A.G.); (P.M.); (N.D.)
| |
Collapse
|
25
|
Lahaye C, Gladine C, Pereira B, Berger J, Chinetti-Gbaguidi G, Lainé F, Mazur A, Ruivard M. Does iron overload in metabolic syndrome affect macrophage profile? A case control study. J Trace Elem Med Biol 2021; 67:126786. [PMID: 34022567 DOI: 10.1016/j.jtemb.2021.126786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 04/28/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022]
Abstract
AIMS Dysmetabolic iron overload syndrome (DIOS) is common but the clinical relevance of iron overload is not understood. Macrophages are central cells in iron homeostasis and inflammation. We hypothesized that iron overload in DIOS could affect the phenotype of monocytes and impair macrophage gene expression. METHODS This study compared 20 subjects with DIOS to 20 subjects with metabolic syndrome (MetS) without iron overload, and 20 healthy controls. Monocytes were phenotyped by Fluorescence-Activated Cell Sorting (FACS) and differentiated into anti-inflammatory M2 macrophages in the presence of IL-4. The expression of 38 genes related to inflammation, iron metabolism and M2 phenotype was assessed by real-time PCR. RESULTS FACS showed no difference between monocytes across the three groups. The macrophagic response to IL-4-driven differentiation was altered in four of the five genes of M2 phenotype (MRC1, F13A1, ABCA1, TGM2 but not FABP4), in DIOS vs Mets and controls demonstrating an impaired M2 polarization. The expression profile of inflammatory genes was not different in DIOS vs MetS. Several genes of iron metabolism presented a higher expression in DIOS vs MetS: SCL11A2 (a free iron transporter, +76 %, p = 0.04), SOD1 (an antioxidant enzyme, +27 %, p = 0.02), and TFRC (the receptor 1 of transferrin, +59 %, p = 0.003). CONCLUSIONS In DIOS, macrophage polarization toward the M2 alternative phenotype is impaired but not associated with a pro-inflammatory profile. The up regulation of transferrin receptor 1 (TFRC) in DIOS macrophages suggests an adaptive role that may limit iron toxicity in DIOS.
Collapse
Affiliation(s)
- Clément Lahaye
- Université Clermont Auvergne, CHU Clermont-Ferrand, Service de Médecine interne Hôpital Estaing, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| | - Bruno Pereira
- Université Clermont Auvergne, CHU Clermont-Ferrand, Unité de biostatistiques, F-63000 Clermont-Ferrand, France.
| | - Juliette Berger
- Université Clermont Auvergne, CHU Clermont-Ferrand, Laboratoire d'Hématologie, Hôpital Estaing, F-63000 Clermont-Ferrand, France.
| | | | - Fabrice Lainé
- INSERM CIC 1414, and Liver Unit, CHU Rennes, 35000 Rennes, France.
| | - Andrzej Mazur
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, F-63000 Clermont-Ferrand, France.
| | - Marc Ruivard
- Université Clermont Auvergne, CHU Clermont-Ferrand, Service de Médecine interne Hôpital Estaing, F-63000 Clermont-Ferrand, France.
| |
Collapse
|
26
|
Botta A, Barra NG, Lam NH, Chow S, Pantopoulos K, Schertzer JD, Sweeney G. Iron Reshapes the Gut Microbiome and Host Metabolism. J Lipid Atheroscler 2021; 10:160-183. [PMID: 34095010 PMCID: PMC8159756 DOI: 10.12997/jla.2021.10.2.160] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/12/2021] [Accepted: 02/21/2021] [Indexed: 12/12/2022] Open
Abstract
Compelling studies have established that the gut microbiome is a modifier of metabolic health. Changes in the composition of the gut microbiome are influenced by genetics and the environment, including diet. Iron is a potential node of crosstalk between the host-microbe relationship and metabolic disease. Although iron is well characterized as a frequent traveling companion of metabolic disease, the role of iron is underappreciated because the mechanisms of iron's influence on host metabolism are poorly characterized. Both iron deficiency and excessive amounts leading to iron overload can have detrimental effects on cardiometabolic health. Optimal iron homeostasis is critical for regulation of host immunity and metabolism in addition to regulation of commensal and pathogenic enteric bacteria. In this article we review evidence to support the notion that altering composition of the gut microbiome may be an important route via which iron impacts cardiometabolic health. We discuss reshaping of the microbiome by iron, the physiological significance and the potential for therapeutic interventions.
Collapse
Affiliation(s)
- Amy Botta
- Department of Biology, York University, Toronto, ON, Canada
| | - Nicole G. Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Nhat Hung Lam
- Department of Biology, York University, Toronto, ON, Canada
| | - Samantha Chow
- Department of Biology, York University, Toronto, ON, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research, Jewish General Hospital and Department of Medicine, McGill University, Montreal, QC, Canada
| | - Jonathan D. Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, ON, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, ON, Canada
| |
Collapse
|
27
|
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
|
28
|
Rives C, Fougerat A, Ellero-Simatos S, Loiseau N, Guillou H, Gamet-Payrastre L, Wahli W. Oxidative Stress in NAFLD: Role of Nutrients and Food Contaminants. Biomolecules 2020; 10:E1702. [PMID: 33371482 PMCID: PMC7767499 DOI: 10.3390/biom10121702] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is often the hepatic expression of metabolic syndrome and its comorbidities that comprise, among others, obesity and insulin-resistance. NAFLD involves a large spectrum of clinical conditions. These range from steatosis, a benign liver disorder characterized by the accumulation of fat in hepatocytes, to non-alcoholic steatohepatitis (NASH), which is characterized by inflammation, hepatocyte damage, and liver fibrosis. NASH can further progress to cirrhosis and hepatocellular carcinoma. The etiology of NAFLD involves both genetic and environmental factors, including an unhealthy lifestyle. Of note, unhealthy eating is clearly associated with NAFLD development and progression to NASH. Both macronutrients (sugars, lipids, proteins) and micronutrients (vitamins, phytoingredients, antioxidants) affect NAFLD pathogenesis. Furthermore, some evidence indicates disruption of metabolic homeostasis by food contaminants, some of which are risk factor candidates in NAFLD. At the molecular level, several models have been proposed for the pathogenesis of NAFLD. Most importantly, oxidative stress and mitochondrial damage have been reported to be causative in NAFLD initiation and progression. The aim of this review is to provide an overview of the contribution of nutrients and food contaminants, especially pesticides, to oxidative stress and how they may influence NAFLD pathogenesis.
Collapse
Affiliation(s)
- Clémence Rives
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Anne Fougerat
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Sandrine Ellero-Simatos
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Nicolas Loiseau
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Hervé Guillou
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Laurence Gamet-Payrastre
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
| | - Walter Wahli
- Toxalim (Research Center in Food Toxicology), Université de Toulouse, INRA, EVT, INP-Purpan, UPS, 31300 Toulouse, France; (C.R.); (A.F.); (S.E.-S.); (N.L.); (H.G.)
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, 11 Mandalay Road, Singapore 308232, Singapore
- Center for Integrative Genomics, Université de Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
| |
Collapse
|
29
|
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: 19] [Impact Index Per Article: 3.8] [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
|
30
|
Eder SK, Feldman A, Strebinger G, Kemnitz J, Zandanell S, Niederseer D, Strasser M, Haufe H, Sotlar K, Stickel F, Paulweber B, Datz C, Aigner E. Mesenchymal iron deposition is associated with adverse long-term outcome in non-alcoholic fatty liver disease. Liver Int 2020; 40:1872-1882. [PMID: 32378295 PMCID: PMC7496452 DOI: 10.1111/liv.14503] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/29/2020] [Accepted: 05/01/2020] [Indexed: 12/29/2022]
Abstract
BACKGROUND & AIMS Approximately one-third of patients with non-alcoholic fatty liver disease (NAFLD) show signs of mild-to-moderate iron overload. The impact of histological iron deposition on the clinical course of patients with NAFLD has not been established. METHODS & RESULTS For this retrospective study, 299 consecutive patients with biopsy-proven NAFLD and a mean follow-up of 8.4 (±4.1; range: 0.3-18.0) years were allocated to one of four groups according to presence of hepatic iron in the reticuloendothelial system (RES) and/or hepatocytes (HC): 156 subjects (52%) showed no stainable iron (NONE), 58 (19%) exclusively reticuloendothelial (xRES), 19 (6%) exclusively hepatocellular (xHC) and 66 (22%) showed a mixed (HC/RES) pattern of iron deposition. A long-term analysis for overall survival, hepatic, cardiovascular or extrahepatic-malignant events was conducted. Based on multivariate Cox proportional hazards models any reticuloendothelial iron was associated with fatal and non-fatal hepatic events. Specifically, xRES showed a cause-specific hazard ratio (csHR) of 2.4 (95%-CI, 1.0-5.8; P = .048) for hepatic as well as cardiovascular fatal and non-fatal events combined (csHR 3.2; 95%-CI, 1.2-8.2; P = .015). Furthermore, the mixed HC/RES iron pattern showed a higher rate of combined hepatic fatal and non-fatal events (csHR 3.6; 95%-CI, 1.4-9.5; P = .010), while xHC iron deposition was not associated with any defined events. CONCLUSIONS The presence of reticuloendothelial-accentuated hepatic iron distribution patterns is associated with detrimental long-term outcomes reflected in a higher rate of both liver-related and cardiovascular fatal and non-fatal events.
Collapse
Affiliation(s)
- Sebastian K. Eder
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria,Department of Pediatrics and Adolescent MedicineSt. Anna Children's HospitalMedical University of ViennaViennaAustria
| | - Alexandra Feldman
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria
| | - Georg Strebinger
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria
| | - Jana Kemnitz
- Department of Imaging & Functional Musculoskeletal ResearchInstitute of Anatomy & Cell BiologyParacelsus Medical UniversitySalzburgAustria
| | - Stephan Zandanell
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria
| | - David Niederseer
- Department of Internal MedicineHospital OberndorfTeaching Hospital of the Paracelsus Medical University SalzburgOberndorfAustria,Department of CardiologyUniversity Heart Center ZurichUniversity Hospital ZurichZurichSwitzerland
| | - Michael Strasser
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria
| | - Heike Haufe
- Institute of PathologyParacelsus Medical UniversitySalzburgAustria
| | - Karl Sotlar
- Institute of PathologyParacelsus Medical UniversitySalzburgAustria
| | - Felix Stickel
- Department of Gastroenterology and HepatologyUniversity Hospital ZurichZurichSwitzerland
| | - Bernhard Paulweber
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria
| | - Christian Datz
- Department of Internal MedicineHospital OberndorfTeaching Hospital of the Paracelsus Medical University SalzburgOberndorfAustria
| | - Elmar Aigner
- First Department of MedicineParacelsus Medical UniversitySalzburgAustria
| |
Collapse
|
31
|
Kim HY, Kwon WY, Park JB, Lee MH, Oh YJ, Suh S, Baek YH, Jeong JS, Yoo YH. Hepatic STAMP2 mediates recombinant FGF21-induced improvement of hepatic iron overload in nonalcoholic fatty liver disease. FASEB J 2020; 34:12354-12366. [PMID: 32721044 DOI: 10.1096/fj.202000790r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
Abstract
Although previous studies have shown that the administration of fibroblast growth factor 21 (FGF21) reverses hepatic steatosis, the mechanism by which FGF21 exerts a therapeutic effect on nonalcoholic fatty liver disease (NAFLD) is not yet entirely understood. We previously demonstrated that hepatic six transmembrane protein of prostate 2 (STAMP2) may represent a suitable target for NAFLD. We investigated the mechanism underlying the therapeutic effect of recombinant FGF21 on NAFLD, focusing on the involvement of hepatic STAMP2. In this study, we used human nonalcoholic steatosis patient pathology samples, C57BL/6 mice for a high-fat diet (HFD)-induced in vivo NAFLD model, and used human primary hepatocytes and HepG2 cells for oleic acid (OA)-induced in vitro NAFLD model. We observed that recombinant FGF21 treatment ameliorated hepatic steatosis and insulin resistance through the upregulation of STAMP2 expression. We further observed hepatic iron overload (HIO) and reduced iron exporter, ferroportin expression in the liver samples obtained from human NAFLD patients, and HFD-induced NAFLD mice and in OA-treated HepG2 cells. Importantly, recombinant FGF21 improved HIO through the hepatic STAMP2-mediated upregulation of ferroportin expression. Our data suggest that hepatic STAMP2 may represent a suitable therapeutic intervention target for FGF21-induced improvement of NAFLD accompanying HIO.
Collapse
Affiliation(s)
- Hye Young Kim
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan, South Korea
| | - Woo Young Kwon
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan, South Korea
| | - Joon Beom Park
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan, South Korea
| | - Mi Hwa Lee
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan, South Korea
| | - Yoo Jin Oh
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan, South Korea
| | - SungHwan Suh
- Department of Endocrinology, Dong-A University College of Medicine, Busan, South Korea
| | - Yang Hyun Baek
- Department of Gastroenterology, Dong-A University College of Medicine, Busan, South Korea
| | - Jin Sook Jeong
- Department of Pathology, Dong-A University College of Medicine, Busan, South Korea
| | - Young Hyun Yoo
- Department of Anatomy and Cell Biology, Dong-A University College of Medicine, Busan, South Korea
| |
Collapse
|
32
|
Liu HF, Wang Q, Du YN, Zhu ZH, Li YF, Zou LQ, Xing W. Dynamic contrast-enhanced MRI with Gd-EOB-DTPA for the quantitative assessment of early-stage liver fibrosis induced by carbon tetrachloride in rabbits. Magn Reson Imaging 2020; 70:57-63. [PMID: 32325235 DOI: 10.1016/j.mri.2020.04.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/31/2020] [Accepted: 04/15/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE To explore quantitative parameters obtained by dynamic contrast-enhanced magnetic resonance imaging (DCE MRI) with Gd-EOB-DTPA in discriminating early-stage liver fibrosis (LF) in a rabbit model. MATERIALS AND METHODS LF was established in 60 rabbits by the injection of 50% CCl4 oil solution, whereas 30 rabbits served as the control group. All rabbits underwent pathological examination to determine the LF stage using the METAVIR classification system. DCE MRI was performed, and quantitative parameters, including Ktrans, Kep, Ve, Vp and Re were measured and evaluated among the different LF stages using spearman correlation coefficients and receiver operating characteristic curve. RESULTS In all, 24, 25, and 22 rabbits had stage F0, stage F1, and stage F2 LF, respectively. Ktrans (r = 0.803) increased, and Kep (r = -0.495) and Re (r = -0.701) decreased with LF stage progression (P < 0.001), while no significant correlation was found for Ve or Vp. Ktrans and Re were significantly different between all LF stage pairs compared (F0 vs. F1, F0 vs. F2, F1 vs. F2, F0 vs. F1-F2, P < 0.05). With the exception of F0 vs. F1, Kep differed significantly between stages (P < 0.05). The AUC of Ktrans was higher than that of other quantitative parameters, with an AUC of 0.92, 0.99, 0.94 and 0.92 for staging F0 vs. F1, F0 vs. F2, F1 vs. F2, and F0 vs. F1-F2, respectively. CONCLUSION Among quantitative parameters of Gd-EOB-DTPA DCE MRI, Ktrans was the best predictor for quantitatively differentiating early-stage LF.
Collapse
Affiliation(s)
- Hai-Feng Liu
- Department of Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou 213003, Jiangsu, China
| | - Qing Wang
- Department of Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou 213003, Jiangsu, China
| | - Ya-Nan Du
- Department of Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou 213003, Jiangsu, China
| | - Zu-Hui Zhu
- Department of Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou 213003, Jiangsu, China
| | - Yu-Feng Li
- Department of Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou 213003, Jiangsu, China
| | - Li-Qiu Zou
- Department of Radiology, The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, China.
| | - Wei Xing
- Department of Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People's Hospital, Changzhou 213003, Jiangsu, China
| |
Collapse
|
33
|
Castiella A, Urreta I, Zapata E, Zubiaurre L, Alústiza JM, Otazua P, Salvador E, Letamendi G, Arrizabalaga B, Rincón ML, Emparanza JI. Liver iron concentration in dysmetabolic hyperferritinemia: Results from a prospective cohort of 276 patients. Ann Hepatol 2020; 19:31-35. [PMID: 31587985 DOI: 10.1016/j.aohep.2019.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 06/30/2019] [Accepted: 07/02/2019] [Indexed: 02/08/2023]
Abstract
INTRODUCTION AND OBJECTIVES We aimed to study the liver iron concentration in patients referred for hyperferritinemia to six hospitals in the Basque Country and to determine if there were differences between patients with or without metabolic syndrome. PATIENTS AND METHODS Metabolic syndrome was defined by accepted criteria. Liver iron concentration was determined by magnetic resonance imaging. RESULTS We obtained the data needed to diagnose metabolic syndrome in 276 patients; a total of 135 patients (49%), 115/240 men (48%), and 20/36 women (55.6%) presented metabolic syndrome. In all 276 patients, an MRI for the determination of liver iron concentration (mean±SD) was performed. The mean liver iron concentration was 30.83±19.38 for women with metabolic syndrome, 38.84±25.50 for men with metabolic syndrome, and 37.66±24.79 (CI 95%; 33.44-41.88) for the whole metabolic syndrome group. In 141 patients (51%), metabolic syndrome was not diagnosed: 125/240 were men (52%) and 16/36 were women (44.4%). The mean liver iron concentration was 34.88±16.18 for women without metabolic syndrome, 44.48±38.16 for men without metabolic syndrome, and 43.39±36.43 (CI 95%, 37.32-49.46) for the whole non-metabolic syndrome group. Comparison of the mean liver iron concentration from both groups (metabolic syndrome vs non-metabolic syndrome) revealed no significant differences (p=0.12). CONCLUSIONS Patients with hyperferritinemia and metabolic syndrome presented a mildly increased mean liver iron concentration that was not significantly different to that of patients with hyperferritinemia and non-metabolic syndrome.
Collapse
Affiliation(s)
- Agustin Castiella
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain; Gastroenterology Service, Donostia University Hospital, Donostia, Spain.
| | - Iratxe Urreta
- Clinical Epidemiology Unit, CASPe, CIBER-ESP, Donostia University Hospital, Donostia, Spain
| | - Eva Zapata
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain; Gastroenterology Service, Donostia University Hospital, Donostia, Spain
| | | | | | - Pedro Otazua
- Gastroenterology Service, Mondragon Hospital, Mondragon, Spain
| | | | | | | | | | - José I Emparanza
- Clinical Epidemiology Unit, CASPe, CIBER-ESP, Donostia University Hospital, Donostia, Spain
| | | |
Collapse
|
34
|
Serfaty L. [Pharmacological treatment of NASH]. Presse Med 2019; 48:1489-1495. [PMID: 31757735 DOI: 10.1016/j.lpm.2019.09.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 09/16/2019] [Indexed: 11/25/2022] Open
Abstract
Lifestyle modifications, especially weight loss, are efficient on NASH liver injury, however rarely followed in clinical practice. The target population of pharmacologic treatments is represented by patients with NASH and fibrosis. Out of histological improvement, efficacy of treatments should be assessed through liver morbi-mortality benefit, but also on extrahepatic events, such as cardiovascular. Among anti-diabetic treatments, glitazones et GLP-1 agonists have shown efficacy on histological liver injury. Vitamin E is efficient on liver injury but at the cost of prostate cancer and stroke over risk. About 60 new molecules are under investigation in NASH and have 4 different types of mechanism of action: metabolic, oxidative stress/apoptosis, anti inflammatory and anti fibrotic. A phase 3 trial evaluating obeticholic acid have shown a 72 weeks duration treatment improved significantly fibrosis.
Collapse
Affiliation(s)
- Lawrence Serfaty
- Hôpital Hautepierre, service d'hépato-gastroentérologie, 1, avenue Molière, 67200 Strasbourg, France; Université Paris Sorbonne, hôpital Saint-Antoine, Inserm UMR-S938, 67000 Strasbourg, France.
| |
Collapse
|
35
|
Salaye L, Bychkova I, Sink S, Kovalic AJ, Bharadwaj MS, Lorenzo F, Jain S, Harrison AV, Davis AT, Turnbull K, Meegalla NT, Lee SH, Cooksey R, Donati GL, Kavanagh K, Bonkovsky HL, McClain DA. A Low Iron Diet Protects from Steatohepatitis in a Mouse Model. Nutrients 2019; 11:nu11092172. [PMID: 31510077 PMCID: PMC6769937 DOI: 10.3390/nu11092172] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Revised: 08/31/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
High tissue iron levels are a risk factor for multiple chronic diseases including type 2 diabetes mellitus (T2DM) and non-alcoholic fatty liver disease (NAFLD). To investigate causal relationships and underlying mechanisms, we used an established NAFLD model-mice fed a high fat diet with supplemental fructose in the water ("fast food", FF). Iron did not affect excess hepatic triglyceride accumulation in the mice on FF, and FF did not affect iron accumulation compared to normal chow. Mice on low iron are protected from worsening of markers for non-alcoholic steatohepatitis (NASH), including serum transaminases and fibrotic gene transcript levels. These occurred prior to the onset of significant insulin resistance or changes in adipokines. Transcriptome sequencing revealed the major effects of iron to be on signaling by the transforming growth factor beta (TGF-β) pathway, a known mechanistic factor in NASH. High iron increased fibrotic gene expression in vitro, demonstrating that the effect of dietary iron on NASH is direct. Conclusion: A lower tissue iron level prevents accelerated progression of NAFLD to NASH, suggesting a possible therapeutic strategy in humans with the disease.
Collapse
Affiliation(s)
- Lipika Salaye
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
| | - Ielizaveta Bychkova
- Department of Internal Medicine, University of Utah Medical Center, Salt Lake City, UT 84112, USA
| | - Sandy Sink
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
| | - Alexander J Kovalic
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Manish S Bharadwaj
- Sticht Center on Aging, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Agilent Technologies, 121 Hartwell Ave, Lexington, MA 02421, USA
| | - Felipe Lorenzo
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
| | - Shalini Jain
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
| | - Alexandria V Harrison
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
| | - Ashley T Davis
- Department of Comparative Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Katherine Turnbull
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
- Department of Comparative Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Nuwan T Meegalla
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Soh-Hyun Lee
- Department of Internal Medicine, University of Utah Medical Center, Salt Lake City, UT 84112, USA
| | - Robert Cooksey
- Department of Internal Medicine, University of Utah Medical Center, Salt Lake City, UT 84112, USA
- VA Medical Center, Salt Lake City, UT 84148, USA
| | - George L Donati
- Department of Chemistry, Wake Forest University, Winston-Salem, NC 27109, USA
| | - Kylie Kavanagh
- Department of Comparative Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
- Department of Biomedicine, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Herbert L Bonkovsky
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Donald A McClain
- Center on Diabetes, Obesity and Metabolism, Department of Internal Medicine, Wake Forest Baptist Medical Center, Winston-Salem, NC 27101, USA.
- VA Medical Center, Salt Lake City, UT 84148, USA.
- VA Medical Center, Salisbury, NC 28144, USA.
| |
Collapse
|
36
|
Atarashi M, Izawa T, Kuwamura M, Yamate J. [The role of iron overload in the progression of nonalcoholic steatohepatitis (NASH)]. Nihon Yakurigaku Zasshi 2019; 154:61-65. [PMID: 31406044 DOI: 10.1254/fpj.154.61] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Non-alcoholic steatohepatitis (NASH), one of the most common chronic liver diseases (CLD), is getting the most important cause of cirrhosis and hepatocellular carcinoma. Iron is an essential micronutrient for organisms. Once excess iron is accumulated in vital organs, dysfunctions of these organs can occur via the generation of reactive oxygen species. Hepatic iron overload is often seen in CLD patients. In NASH patients, iron accumulation in the liver is positively correlated with histological severity. Thus iron overload can contribute to progression of nonalcoholic fatty liver disease (NAFLD) to NASH. In a rat model of NASH, feeding of high-fat and high-iron diet increases hepatic inflammation with increased hepatic cytokine expression compared with feeding of high-fat diet only. In this model, iron is intensely accumulated in Kupffer cells/macrophages within the lesion, raising the possibility that iron-laden Kupffer cells/macrophages can play a key role in the enhancement of hepatic inflammation in NASH condition. On the other hand, in a rat model of liver cirrhosis, dietary iron overload clearly abrogates the development and progression of liver cirrhosis induced by repeated administration of thioacetamide (TAA). These findings suggest that iron overload can promote or suppress chronic liver diseases depending on the tissue microenvironment. Here we review and introduce the recent findings on the pathological roles of iron overload in the development and progression of NAFLD/NASH.
Collapse
Affiliation(s)
- Machi Atarashi
- Researcher Pharmacology dept. Research & Development center FUSO Pharm., Ltd.,Laboratory of Veterinary Pathology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Sciences
| | - Takeshi Izawa
- Laboratory of Veterinary Pathology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Sciences
| | - Mitsuru Kuwamura
- Laboratory of Veterinary Pathology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Sciences
| | - Jyoji Yamate
- Laboratory of Veterinary Pathology, Division of Veterinary Science, Osaka Prefecture University Graduate School of Life and Environmental Sciences
| |
Collapse
|
37
|
Abstract
Hereditary hemochromatosis (HH) is one of the most common genetic disorders among persons of northern European descent. There have been recent advances in the diagnosis, management, and treatment of HH. The availability of molecular diagnostic testing for HH has made possible confirmation of the diagnosis for most patients. Several genotype-phenotype correlation studies have clarified the differences in clinical features between patients with the C282Y homozygous genotypes and other HFE mutation patterns. The increasing use of noninvasive tests such as MRI T2* has made quantification of hepatic iron deposition easier and eliminated the need for liver biopsy in most patients. Serum ferritin of <1,000 ng/mL at diagnosis remains an important diagnostic test to identify patients with a low risk of advanced hepatic fibrosis and should be used routinely as part of the initial diagnostic evaluation. Genetic testing for other types of HH is available but is expensive and generally not useful in most clinical settings. Serum ferritin may be elevated among patients with nonalcoholic fatty liver disease and in those with alcoholic liver disease. These diagnoses are more common than HH among patients with elevated serum ferritin who are not C282Y homozygotes or C282Y/H63D compound heterozygotes. A secondary cause for liver disease should be excluded among patients with suspected iron overload who are not C282Y homozygotes. Phlebotomy remains the mainstay of therapy, but emerging novel therapies such as new chelating agents may have a role for selected patients.
Collapse
|
38
|
Yeo YH, Lai YC. Redox Regulation of Metabolic Syndrome: Recent Developments in Skeletal Muscle Insulin Resistance and Non-alcoholic Fatty Liver Disease (NAFLD). CURRENT OPINION IN PHYSIOLOGY 2019; 9:79-86. [PMID: 32818162 DOI: 10.1016/j.cophys.2019.05.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Several new discoveries over the past decade have shown that metabolic syndrome, a cluster of metabolic disorders, including increased visceral obesity, hyperglycemia, hypertension, dyslipidemia and low HDL-cholesterol, is commonly associated with skeletal muscle insulin resistance. More recently, non-alcoholic fatty liver disease (NAFLD) was recognized as an additional condition that is strongly associated with features of metabolic syndrome. While the pathogenesis of skeletal muscle insulin resistance and fatty liver is multifactorial, the role of dysregulated redox signaling has been clearly demonstrated in the regulation of skeletal muscle insulin resistance and NAFLD. In this review, we aim to provide recent updates on redox regulation with respect to (a) pro-oxidant enzymes (e.g. NAPDH oxidase and xanthine oxidase); (b) mitochondrial dysfunction; (c) endoplasmic reticulum (ER) stress; (d) iron metabolism derangements; and (e) gut-skeletal muscle or gut-liver connection in the development of skeletal muscle insulin resistance and NAFLD. Furthermore, we discuss promising new therapeutic strategies targeting redox regulation currently under investigation for the treatment of skeletal muscle insulin resistance and NAFLD.
Collapse
Affiliation(s)
- Yee-Hui Yeo
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Palo Alto, California, USA
| | - Yen-Chun Lai
- Division of Pulmonary, Critical Care, Sleep and Occupational Medicine, Department of Medicine, Indiana University School of Medicine; Indianapolis, IN, USA
| |
Collapse
|
39
|
Iron-Induced Liver Injury: A Critical Reappraisal. Int J Mol Sci 2019; 20:ijms20092132. [PMID: 31052166 PMCID: PMC6539962 DOI: 10.3390/ijms20092132] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/12/2022] Open
Abstract
Iron is implicated in the pathogenesis of a number of human liver diseases. Hereditary hemochromatosis is the classical example of a liver disease caused by iron, but iron is commonly believed to contribute to the progression of other forms of chronic liver disease such as hepatitis C infection and nonalcoholic fatty liver disease. In this review, we present data from cell culture experiments, animal models, and clinical studies that address the hepatotoxicity of iron. These data demonstrate that iron overload is only weakly fibrogenic in animal models and rarely causes serious liver damage in humans, calling into question the concept that iron overload is an important cause of hepatotoxicity. In situations where iron is pathogenic, iron-induced liver damage may be potentiated by coexisting inflammation, with the resulting hepatocyte necrosis an important factor driving the fibrogenic response. Based on the foregoing evidence that iron is less hepatotoxic than is generally assumed, claims that assign a causal role to iron in liver injury in either animal models or human liver disease should be carefully evaluated.
Collapse
|
40
|
Vela D. Low hepcidin in liver fibrosis and cirrhosis; a tale of progressive disorder and a case for a new biochemical marker. Mol Med 2018; 24:5. [PMID: 30134796 PMCID: PMC6016890 DOI: 10.1186/s10020-018-0008-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/13/2018] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is a precursor of liver cirrhosis, which is associated with increased mortality. Though liver biopsy remains the gold standard for the diagnosis of fibrosis, noninvasive biochemical methods are cost-effective, practical and are not linked with major risks of complications. In this respect, serum hepcidin, has emerged as a new marker of fibrosis and cirrhosis. In this review the discussion uncovers molecular links between hepcidin disturbance and liver fibrosis/cirrhosis. The discussion also expands on clinical studies that suggest that hepcidin can potentially be used as a biochemical parameter of fibrosis/cirrhosis and target of therapeutic strategies to treat liver diseases. The debatable issues such as the complicated nature of hepcidin disturbance in non-alcoholic liver disease, serum levels of hepcidin in acute hepatitis C virus infection, cause of hepcidin disturbance in autoimmune hepatitis and hepatic insulin resistance are discussed, with potential solutions unveiled in order to be studied by future research.
Collapse
Affiliation(s)
- Driton Vela
- Department of Physiology, Faculty of Medicine, University of Prishtina, Martyr's Boulevard n.n, Prishtina, 10000, Kosovo.
| |
Collapse
|
41
|
Ryan JD, Armitage AE, Cobbold JF, Banerjee R, Borsani O, Dongiovanni P, Neubauer S, Morovat R, Wang LM, Pasricha SR, Fargion S, Collier J, Barnes E, Drakesmith H, Valenti L, Pavlides M. Hepatic iron is the major determinant of serum ferritin in NAFLD patients. Liver Int 2018; 38:164-173. [PMID: 28679028 DOI: 10.1111/liv.13513] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Elevated serum ferritin is common in NAFLD, and is associated with more advanced disease and increased mortality. Hyperferritinaemia in NAFLD is often attributed to inflammation, while in other conditions ferritin closely reflects body iron stores. The aim of this study was to clarify the underlying cause of hyperferritinaemia in NAFLD. METHODS Ferritin levels were examined with markers of iron status, inflammation and liver injury across the clinical spectrum of NAFLD using blood, tissue and magnetic resonance (MR) imaging. A separate larger group of NAFLD patients with hepatic iron staining and quantification were used for validation. RESULTS Serum ferritin correlated closely with the iron regulatory hormone hepcidin, and liver iron levels determined by MR. Furthermore, ferritin levels reflected lower serum adiponectin, a marker of insulin resistance, and liver fat, but not cytokine or CRP levels. Ferritin levels differed according to fibrosis stage, increasing from early to moderate disease, and declining in cirrhosis. A similar pattern was found in the validation cohort of NAFLD patients, where ferritin levels were highest in those with macrophage iron deposition. Multivariate analysis revealed liver iron and hepcidin levels as the major determinants of serum ferritin. CONCLUSIONS While hyperferritinaemia is associated with markers of liver injury and insulin resistance, serum hepcidin and hepatic iron are the strongest predictors of ferritin levels. These findings highlight the role of disordered iron homeostasis in the pathogenesis of NAFLD, suggesting that therapies aimed at correcting iron metabolism may be beneficial.
Collapse
Affiliation(s)
- John D Ryan
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Andrew E Armitage
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Jeremy F Cobbold
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | | | - Oscar Borsani
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Paola Dongiovanni
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Stefan Neubauer
- Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| | - Reza Morovat
- Department of Biochemistry, John Radcliffe Hospital, Oxford, UK
| | - Lai Mun Wang
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sant-Rayn Pasricha
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Silvia Fargion
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Jane Collier
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Eleanor Barnes
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Luca Valenti
- Internal Medicine and Metabolic Diseases, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Michael Pavlides
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK.,Oxford Centre for Clinical Magnetic Resonance Research, University of Oxford, Oxford, UK
| |
Collapse
|