1
|
Pušeljić M, Stadlbauer V, Ahmadova N, Pohl M, Kopetzky M, Kaufmann-Bühler AK, Watzinger N, Igrec J, Fuchsjäger M, Talakić E. Impact of body fat composition on liver iron overload severity in hemochromatosis: a retrospective MRI analysis. LA RADIOLOGIA MEDICA 2024:10.1007/s11547-024-01930-8. [PMID: 39578337 DOI: 10.1007/s11547-024-01930-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 11/08/2024] [Indexed: 11/24/2024]
Abstract
PURPOSE To evaluate the correlation between ectopic adipose tissue and iron overload severity in patients with hemochromatosis. MATERIAL AND METHODS A retrospective cohort of 52 patients who underwent liver iron concentration quantification from January 2015 to October 2023 using a 3.0T MRI scanner. R2* relaxation times and proton density fat fraction (PDFF) were assessed for the entire liver volume and a specific region of interest (ROI) placed in the right lobe. Total body fat (TF), subcutaneous fat (SCF), intermuscular fat (IMF), and visceral fat (VSF) percentages were calculated from a single axial slice at the level of the third lumbar vertebra. Additionally, ratios of IMF-to-VSF, IMF-to-SCF, and SCF-to-VSF were calculated. Standard iron laboratory parameters were collected at least one month prior to MRI. Pearson correlation coefficient was used for correlation analysis. RESULTS The mean age of participants was 53.9 ± 19.6 years. IMF positively correlated with R2* values in the ROI (p = 0.005, rs = 0.382) and entire liver (p = 0.016, rs = 0.332). Conversely, VSF negatively correlated with R2* values from the ROI (p = < 0.001, rs = - 0.488) and entire liver (p = < 0.001, rs = - 0.459). Positive correlations were also found between IMF-to-VSF and R2* of the ROI (p = 0.003, rs = 0.400) and whole liver (p = 0.008, rs = 0.364). Ferritin levels positively correlated with R2* values calculated from ROI (p = 0.002, rs = 0.417) and whole liver volume (p = 0.004, rs = 0.397). A positive correlation was noted between PDFF of the entire liver and TF (p = 0.024, rs = 0.313). CONCLUSION The percentage of Intermuscular and visceral adipose tissues correlates with the severity of liver iron overload in hemochromatosis patients.
Collapse
Affiliation(s)
- Marijan Pušeljić
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Vanessa Stadlbauer
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Auenbruggerplatz 15, 8036, Graz, Austria
- Center of Biomarker Research in Medicine (CBmed), Stiftingtalstrasse 5, 8010, Graz, Austria
| | - Nigar Ahmadova
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Maximilian Pohl
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Michaela Kopetzky
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Ann-Katrin Kaufmann-Bühler
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Nikolaus Watzinger
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Jasminka Igrec
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Michael Fuchsjäger
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria
| | - Emina Talakić
- Division of General Radiology, Department of Radiology, Medical University of Graz, Auenbruggerplatz 9, 8036, Graz, Austria.
| |
Collapse
|
2
|
Connolly BJ, Saxton SN. Recent updates on the influence of iron and magnesium on vascular, renal, and adipose inflammation and possible consequences for hypertension. J Hypertens 2024; 42:1848-1861. [PMID: 39258532 PMCID: PMC11451934 DOI: 10.1097/hjh.0000000000003829] [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: 02/06/2024] [Revised: 06/26/2024] [Accepted: 07/22/2024] [Indexed: 09/12/2024]
Abstract
The inflammatory status of the kidneys, vasculature, and perivascular adipose tissue (PVAT) has a significant influence on blood pressure and hypertension. Numerous micronutrients play an influential role in hypertension-driving inflammatory processes, and recent reports have provided bases for potential targeted modulation of these micronutrients to reduce hypertension. Iron overload in adipose tissue macrophages and adipocytes engenders an inflammatory environment and may contribute to impaired anticontractile signalling, and thus a treatment such as chelation therapy may hold a key to reducing blood pressure. Similarly, magnesium intake has proven to greatly influence inflammatory signalling and concurrent hypertension in both healthy animals and in a model for chronic kidney disease, demonstrating its potential clinical utility. These findings highlight the importance of further research to determine the efficacy of micronutrient-targeted treatments for the amelioration of hypertension and their potential translation into clinical application.
Collapse
Affiliation(s)
- Benjamin J Connolly
- Divison of Cardiovascular Sciences, The University of Manchester, Manchester, UK
| | | |
Collapse
|
3
|
Zoller H, Tilg H. Ferritin-a promising biomarker in MASLD. Gut 2024; 73:720-721. [PMID: 38538068 DOI: 10.1136/gutjnl-2023-331848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/07/2024]
Affiliation(s)
- Heinz Zoller
- Department of Medicine I, Gastroenterology, Hepatology, Endocrinology and Metabolism, Medical University of Innsbruck, Innsbruck, Tirol, Austria
- Christian Doppler Laboratory on Iron and Phosphate Biology, Christian Doppler Forschungsgesellschaft, Innsbruck, Tirol, Austria
| | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medizinische Universitat Innsbruck, Innsbruck, Tirol, Austria
| |
Collapse
|
4
|
Lobbes H, Dalle C, Pereira B, Ruivard M, Mazur A, Gladine C. Eicosanoids and Oxylipin Signature in Hereditary Hemochromatosis Patients Are Similar to Dysmetabolic Iron Overload Syndrome Patients but Are Impacted by Dietary Iron Absorption. ANNALS OF NUTRITION & METABOLISM 2024; 80:117-127. [PMID: 38354712 DOI: 10.1159/000536657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
INTRODUCTION Oxylipins are mediators of oxidative stress. To characterize the underlying inflammatory processes and phenotype effect of iron metabolism disorders, we investigated the oxylipin profile in hereditary hemochromatosis (HH) and dysmetabolic iron overload syndrome (DIOS) patients. METHODS An LC-MS/MS-based method was performed to quantify plasma oxylipins in 20 HH and 20 DIOS patients in fasting conditions and 3 h after an iron-rich meal in HH patients. RESULTS Principal component analysis showed no separation between HH and DIOS, suggesting that the clinical phenotype has no direct impact on oxylipin metabolism. 20-HETE was higher in DIOS and correlated with hypertension (p = 0.03). Different oxylipin signatures were observed in HH before and after the iron-rich meal. Discriminant oxylipins include epoxy fatty acids derived from docosahexaenoic acid and arachidonic acid as well as 13-HODE and 9-HODE. Mediation analysis found no major contribution of dietary iron absorption for 16/22 oxylipins significantly affected by the meal. DISCUSSION The oxylipin profiles of HH and DIOS seemed similar except for 20-HETE, possibly reflecting different hypertension prevalence between the two groups. Oxylipins were significantly affected by the iron-rich meal, but the specific contribution of iron was not clear. Although iron may contribute to oxidative stress and inflammation in HH and DIOS, this does not seem to directly affect oxylipin metabolism.
Collapse
Affiliation(s)
- Hervé Lobbes
- Médecine Interne, Hôpital Estaing, 1 Place Lucie et Raymond Aubrac, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France,
- Institut Pascal, UMR 6602, Centre National de la Recherche Scientifique, Université Clermont Auvergne, Clermont-Ferrand, France,
| | - Céline Dalle
- Unité de Nutrition Humaine, UMR 1019, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| | - Bruno Pereira
- Unité de biostatistiques, Direction de la Recherche Clinique et de l'Innovation, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
| | - Marc Ruivard
- Médecine Interne, Hôpital Estaing, 1 Place Lucie et Raymond Aubrac, Centre Hospitalier Universitaire de Clermont-Ferrand, Clermont-Ferrand, France
- Institut Pascal, UMR 6602, Centre National de la Recherche Scientifique, Université Clermont Auvergne, Clermont-Ferrand, France
| | - Andrzej Mazur
- Unité de Nutrition Humaine, UMR 1019, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| | - Cécile Gladine
- Unité de Nutrition Humaine, UMR 1019, Institut national de recherche pour l'agriculture, l'alimentation et l'environnement (INRAE), Paris, France
| |
Collapse
|
5
|
Cheng W, Zhou Y, Chen H, Wu Q, Li Y, Wang H, Feng Y, Wang Y. The iron matters: Aged microplastics disrupted the iron homeostasis in the liver organoids. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 906:167529. [PMID: 37788777 DOI: 10.1016/j.scitotenv.2023.167529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/27/2023] [Accepted: 09/29/2023] [Indexed: 10/05/2023]
Abstract
Plastic products undergo artificial and unintentional aging during daily use, causing the presence of aged microplastics (aMP). Humans are inevitably exposed to aMP. Liver is one of the critical target organs of MP through oral intake, however, limited research has focused on the hepatic toxicity of aMP compared to pristine MP (pMP). We utilized the human pluripotent stem cells-derived liver organoids (LOs) to compare the cytotoxicity of pristine polystyrene microplastics (pPS) (1 μm, carbonyl index 0.08) and aged polystyrene microplastics (aPS) (1 μm, carbonyl index 0.20) ranged from 20 to 200 ng/mL. Our findings indicate that aPS was more cytotoxic than pPS. We explored the disrupted iron homeostasis in terms of the [Fe2+] and [Fe3+] levels, iron storage and transport. A "vector-like effect" induced by aPS has been preliminarily suggested by the correlated change in total iron level and co-localization of PS and ferritin light chain (FTL) in the LOs following exposure to aPS and ferric ammonium citrate (FAC) individually and combinedly. In addition, we observed abnormal mitochondrial morphology, elevated lipid peroxidation, and declined GSH peroxidase activity, together with the declined expression of transferrin receptor (TFRC) and elevated expressions of SLC7A11, FTL. The gene handled iron transport and iron use were disrupted by aPS. Moreover, we employed FAC to introduce iron overload and Nacetylcysteine (NAC) to protect the lipid peroxidation. In aPS + FAC group, aggravated effects could be observed in aspects of [Fe2+] level, lipid peroxidation, and compromised expression levels of iron homeostasis-related markers, in contrast, in aPS + NAC group, most of changes recovered but the hepatocytoxicity remained. Specifically, a dimorphic change in elevated FTL and decreased ferritin heavy chain (FTH1) caused by 50 ng/mL aMP (57.33 ± 3.57 items/mL, equivalent to human intake level), indicated a specific response to low-dose aMP.
Collapse
Affiliation(s)
- Wei Cheng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yue Zhou
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Hange Chen
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qian Wu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Li
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hui Wang
- State Key Laboratory of Oncogenes and Related Genes, Center for Single-Cell Omics, School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Feng
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Yan Wang
- The Ninth People's Hospital of Shanghai Jiao Tong University School of Medicine, School of Public Health, Collaborative Innovation Center for Clinical and Translational Science by Ministry of Education & Shanghai, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
6
|
Ruivard M, Lobbes H. [Diagnosis and treatment of iron overload]. Rev Med Interne 2023; 44:656-661. [PMID: 37507250 DOI: 10.1016/j.revmed.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Etiological investigation of hyperferritinemia includes a full clinical examination, with the measurement of waist circumference, and simple biological tests including transferrin saturation. The classification between hyperferritinemia without iron overload (inflammation, excessive alcohol intake, cytolysis, L-ferritin mutation) or with iron overload is then relatively easy. Dysmetabolic iron overload syndrome is the most common iron overload disease and is defined by an unexplained serum ferritin level elevation associated with various metabolic syndrome criteria and mild hepatic iron content increase assessed by magnetic resonance imaging. Bloodlettings are often poorly tolerated without clear benefit. Type 1 genetic hemochromatosis (homozygous C282Y mutation on the HFE gene) leads to iron accumulation through an increase of dietary iron absorption due to hypohepcidinemia. More than 95% of hemochromatosis are type 1 hemochromatosis but the phenotypic expression is highly variable. Elastography is recommended to identify advanced hepatic fibrosis when serum ferritin exceeds 1000μg/L. Life expectancy is normal when bloodlettings are started early. Ferroportin gene mutation is an autosomal dominant disease with generally moderate iron overload. Chelators are used in iron overload associated with anaemia (myelodysplastic syndromes or transfusion-dependent thalassemia). Chelation is initiated when hepatic iron content exceeds 120μmol/g. Deferasirox is often used as first-line therapy, but deferiprone may be of interest despite haematological toxicity (neutropenia). Deferoxamine (parenteral route) is the treatment of choice for severe iron overload or emergency conditions.
Collapse
Affiliation(s)
- M Ruivard
- Service médecine Interne, CHU de Clermont-Ferrand, CHU d'Estaing, 63003 Clermont-Ferrand, France; UMR 6602 UCA/CNRS/SIGMA, thérapies guidées par l'image (TGI), université Clermont Auvergne, 63000 Clermont-Ferrand, France.
| | - H Lobbes
- Service médecine Interne, CHU de Clermont-Ferrand, CHU d'Estaing, 63003 Clermont-Ferrand, France; UMR 6602 UCA/CNRS/SIGMA, thérapies guidées par l'image (TGI), université Clermont Auvergne, 63000 Clermont-Ferrand, France.
| |
Collapse
|
7
|
Valenti L, Corradini E, Adams LA, Aigner E, Alqahtani S, Arrese M, Bardou-Jacquet E, Bugianesi E, Fernandez-Real JM, Girelli D, Hagström H, Henninger B, Kowdley K, Ligabue G, McClain D, Lainé F, Miyanishi K, Muckenthaler MU, Pagani A, Pedrotti P, Pietrangelo A, Prati D, Ryan JD, Silvestri L, Spearman CW, Stål P, Tsochatzis EA, Vinchi F, Zheng MH, Zoller H. Consensus Statement on the definition and classification of metabolic hyperferritinaemia. Nat Rev Endocrinol 2023; 19:299-310. [PMID: 36805052 PMCID: PMC9936492 DOI: 10.1038/s41574-023-00807-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/19/2023]
Abstract
Hyperferritinaemia is a common laboratory finding that is often associated with metabolic dysfunction and fatty liver. Metabolic hyperferritinaemia reflects alterations in iron metabolism that facilitate iron accumulation in the body and is associated with an increased risk of cardiometabolic and liver diseases. Genetic variants that modulate iron homeostasis and tissue levels of iron are the main determinants of serum levels of ferritin in individuals with metabolic dysfunction, raising the hypothesis that iron accumulation might be implicated in the pathogenesis of insulin resistance and the related organ damage. However, validated criteria for the non-invasive diagnosis of metabolic hyperferritinaemia and the staging of iron overload are still lacking, and there is no clear evidence of a benefit for iron depletion therapy. Here, we provide an overview of the literature on the relationship between hyperferritinaemia and iron accumulation in individuals with metabolic dysfunction, and on the associated clinical outcomes. We propose an updated definition and a provisional staging system for metabolic hyperferritinaemia, which has been agreed on by a multidisciplinary global panel of expert researchers. The goal is to foster studies into the epidemiology, genetics, pathophysiology, clinical relevance and treatment of metabolic hyperferritinaemia, for which we provide suggestions on the main unmet needs, optimal design and clinically relevant outcomes.
Collapse
Affiliation(s)
- Luca Valenti
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy.
- Biological Resource Center and Precision Medicine Lab, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy.
| | - Elena Corradini
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy.
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy.
| | - Leon A Adams
- Medical School, University of Western Australia, Perth, Australia
| | - Elmar Aigner
- First Department of Medicine, University Clinic Salzburg, Paracelsus Medical University Salzburg, Salzburg, Austria
| | - Saleh Alqahtani
- Royal Clinics and Gastroenterology and Hepatology, King Faisal Specialist Hospital & Research Centre, Riyadh, Kingdom of Saudi Arabia
- Division of Gastroenterology and Hepatology, Johns Hopkins University, Baltimore, MD, USA
| | - Marco Arrese
- Department of Gastroenterology, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Edouard Bardou-Jacquet
- University of Rennes, UMR1241, CHU Rennes, National Reference Center for Hemochromatosis and iron metabolism disorder, INSERM CIC1414, Rennes, France
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology, University of Turin, Turin, Italy
| | - Jose-Manuel Fernandez-Real
- Department of Diabetes, Endocrinology and Nutrition, Dr Josep Trueta University Hospital, Girona, Spain
- Department of Medical Sciences, Faculty of Medicine, Girona University, Girona, Spain
- Nutrition, Eumetabolism and Health Group, Girona Biomedical Research Institute (IdibGi), Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain
| | - Domenico Girelli
- Section of Internal Medicine, Department of Medicine, University of Verona, Policlinico Giambattista Rossi, Verona, Italy
| | - Hannes Hagström
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Benjamin Henninger
- Department of Radiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Kris Kowdley
- Liver Institute Northwest, Seattle, WA, USA
- Elson S. Floyd College of Medicine, Washington State University, Seattle, WA, USA
| | - Guido Ligabue
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Division of Radiology, Ospedale di Sassuolo S.p.A, Sassuolo, Modena, Italy
| | - Donald McClain
- Wake Forest School of Medicine, Winston Salem, NC, USA
- Department of Veterans Affairs, Salisbury, NC, USA
| | - Fabrice Lainé
- INSERM CIC1414, Liver Unit, CHU Rennes, Rennes, France
| | - Koji Miyanishi
- Department of Medical Oncology, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Martina U Muckenthaler
- Department of Paediatric Hematology, Oncology and Immunology, University of Heidelberg, Heidelberg, Germany
- Center for Molecular Translational Iron Research, Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Heidelberg, Germany
| | - Alessia Pagani
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Patrizia Pedrotti
- Laboratorio di RM Cardiaca Cardiologia 4, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences, Università degli Studi di Modena e Reggio Emilia, Modena, Italy
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy
| | - Daniele Prati
- Department of Transfusion Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico Milano, Milan, Italy
| | - John D Ryan
- Hepatology Unit, Beaumont Hospital, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Laura Silvestri
- Regulation of Iron Metabolism Unit, Division of Genetics and Cell Biology, IRCCS Ospedale San Raffaele, Milan, Italy
| | - C Wendy Spearman
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Per Stål
- Division of Hepatology, Department of Upper GI Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Emmanuel A Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital and UCL, London, UK
| | - Francesca Vinchi
- Iron Research Laboratory, Lindsley F.Kimball Research Institute, New York Blood Center, New York, NY, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Ming-Hua Zheng
- NAFLD Research Center, Department of Hepatology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Diagnosis and Treatment for the Development of Chronic Liver Disease in Zhejiang Province, Wenzhou, China
| | - Heinz Zoller
- Department of Medicine I, Medical University of Innsbruck, Innsbruck, Austria
- Doppler Laboratory on Iron and Phosphate Biology, Innsbruck, Austria
| |
Collapse
|
8
|
Magnetic Resonance Liver Iron Concentration Can Guide Venesection Decision-Making in Hyperferritinemia. Dig Dis Sci 2023; 68:2704-2709. [PMID: 36929239 DOI: 10.1007/s10620-023-07873-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 02/09/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The clinical benefit of venesection in suspected iron overload can be unclear and serum ferritin may overestimate the degree of iron overload. AIMS To help inform practice, we examined magnetic resonance liver iron concentration (MRLIC) in a cohort investigated for haemochromatosis. METHODS One hundred and six subjects with suspected haemochromatosis underwent HFE genotyping and MRLIC with time-matched serum ferritin and transferrin saturation values. For those treated with venesection, volume of blood removed was calculated as a measure of iron overload. RESULTS Forty-seven C282Y homozygotes had median ferritin 937 µg/l and MRLIC 4.83 mg/g; MRLIC was significantly higher vs non-homozygotes for any given ferritin concentration. No significant difference in MRLIC was observed between homozygotes with and without additional risk factors for hyperferritinemia. Thirty-three compound heterozygotes (C282Y/H63D) had median ferritin 767 µg/l and MRLIC 2.58 mg/g; ferritin < 750 µg/l showed 100% specificity for lack of significant iron overload (< 3.2 mg/g). 79% of C282Y/H63D had additional risk factors-mean MRLIC was significantly lower in this sub-group (2.4 mg/g vs 3.23 mg/g). 26 C282Y heterozygous or wild-type had median ferritin 1226 µg/l and MRLIC 2.13 mg/g; 69% with additional risk factors had significantly higher ferritin concentrations (with comparable MRLIC) and ferritin < 1000 µg/l showed 100% specificity for lack of significant iron overload. In 31 patients (26 homozygotes, 5 C282Y/H63D) venesected to ferritin < 100 µg/l, MRLIC and total venesection volume correlated strongly (r = 0.749), unlike MRLIC and serum ferritin. CONCLUSION MRLIC is an accurate marker of iron overload in haemochromatosis. We propose serum ferritin thresholds in non-homozygotes which, if validated, could tailor cost-effective use of MRLIC in venesection decision-making.
Collapse
|
9
|
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
|
10
|
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
|
11
|
Qiu F, Wu L, Yang G, Zhang C, Liu X, Sun X, Chen X, Wang N. The role of iron metabolism in chronic diseases related to obesity. Mol Med 2022; 28:130. [PMID: 36335331 PMCID: PMC9636637 DOI: 10.1186/s10020-022-00558-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 10/14/2022] [Indexed: 11/08/2022] Open
Abstract
Obesity is one of the major public health problems threatening the world, as well as a potential risk factor for chronic metabolic diseases. There is growing evidence that iron metabolism is altered in obese people, however, the highly refined regulation of iron metabolism in obesity and obesity-related complications is still being investigated. Iron accumulation can affect the body’s sensitivity to insulin, Type 2 diabetes, liver disease and cardiovascular disease. This review summarized the changes and potential mechanisms of iron metabolism in several chronic diseases related to obesity, providing new clues for future research.
Collapse
|
12
|
Hanna HWZ, Baz HN, Al-Kzayer LFY, El Haddad HE, El-Mougy F. Assessment of plasma catecholamines in patients with dysmetabolic iron overload syndrome. J Appl Biomed 2022; 20:141-145. [PMID: 36708719 DOI: 10.32725/jab.2022.016] [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: 06/21/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND Dysmetabolic iron overload syndrome (DIOS) is characterized by hyperferritinemia and normal transferrin saturation level with components of metabolic syndrome (MS). Among cases of MS, we determined those with DIOS and their characterizations, then we evaluated the association between plasma catecholamines status and hypertension in DIOS. METHODS We compared 101 hypertensive patients with 50 healthy participants (control group). Iron (iron, transferrin, and ferritin), insulin, and plasma catecholamine (adrenaline, noradrenaline, and dopamine), profiles were measured for both groups. Homeostasis model assessment of insulin resistance index and transferrin saturation were also calculated. RESULTS Out of 101 hypertensive patients, 64 were diagnosed with MS, and 6 of the latter met the DIOS diagnostic criteria. Significantly, DIOS patients were older and had lower body mass index (BMI) compared with hypertensive non-DIOS patients with p-values of (0.026), and (0.033), respectively. Adrenaline, noradrenaline, and dopamine levels did not differ significantly between DIOS and non-DIOS patients. CONCLUSIONS Of the MS patients, 9.3% were diagnosed with DIOS. Accordingly, complete iron profiling should be performed routinely in the cases of MS for early diagnosis of DIOS, to prevent future complications. Further studies are required to test the hypothesis linking older age and lower BMI with the pathogenesis of DIOS.
Collapse
Affiliation(s)
| | - Heba N Baz
- Cairo University, Kasr Al Ainy, Faculty of Medicine, Department of Clinical and Chemical Pathology, Cairo, Egypt
| | | | - Hemmat E El Haddad
- Cairo University, Kasr Al Ainy, Faculty of Medicine, Department of Internal Medicine, Cairo, Egypt
| | - Fatma El-Mougy
- Cairo University, Kasr Al Ainy, Faculty of Medicine, Department of Clinical and Chemical Pathology, Cairo, Egypt
| |
Collapse
|
13
|
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
|
14
|
Fernandez M, Lokan J, Leung C, Grigg A. A critical evaluation of the role of iron overload in fatty liver disease. J Gastroenterol Hepatol 2022; 37:1873-1883. [PMID: 35906772 DOI: 10.1111/jgh.15971] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/16/2022] [Accepted: 07/27/2022] [Indexed: 12/09/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been associated with a condition known as the dysmetabolic iron overload syndrome, but the frequency and severity of iron overload in NAFLD is not well described. There is emerging evidence that mild to moderate excess hepatic iron can aggravate the risk of progression of NAFLD to nonalcoholic steatohepatitis and eventually cirrhosis. Mechanisms are postulated to be via reactive oxygen species, inflammatory cytokines, lipid oxidation, and oxidative stress. The aim of this review is to assess the evidence for true hepatic iron overload in NAFLD, to discuss the pathogenesis by which excess iron may be toxic, and to critically evaluate the studies designed to deplete iron by regular venesection. In brief, the studies are inconclusive due to heterogeneity in eligibility criteria, sample size, randomization, hepatic iron measurement, serial histological endpoints, target ferritin levels, length of venesection, and degree of confounding lifestyle intervention. We propose a trial designed to overcome the limitations of these studies.
Collapse
Affiliation(s)
- Monique Fernandez
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Julie Lokan
- Department of Anatomical Pathology, Austin Health, Heidelberg, Victoria, Australia
| | - Christopher Leung
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.,Departments of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Andrew Grigg
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.,Department of Clinical Haematology, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
15
|
Zoller H, Schaefer B, Vanclooster A, Griffiths B, Bardou-Jacquet E, Corradini E, Porto G, Ryan J, Cornberg M. EASL Clinical Practice Guidelines on haemochromatosis. J Hepatol 2022; 77:479-502. [PMID: 35662478 DOI: 10.1016/j.jhep.2022.03.033] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/15/2022]
Abstract
Haemochromatosis is characterised by elevated transferrin saturation (TSAT) and progressive iron loading that mainly affects the liver. Early diagnosis and treatment by phlebotomy can prevent cirrhosis, hepatocellular carcinoma, diabetes, arthropathy and other complications. In patients homozygous for p.Cys282Tyr in HFE, provisional iron overload based on serum iron parameters (TSAT >45% and ferritin >200 μg/L in females and TSAT >50% and ferritin >300 μg/L in males and postmenopausal women) is sufficient to diagnose haemochromatosis. In patients with high TSAT and elevated ferritin but other HFE genotypes, diagnosis requires the presence of hepatic iron overload on MRI or liver biopsy. The stage of liver fibrosis and other end-organ damage should be carefully assessed at diagnosis because they determine disease management. Patients with advanced fibrosis should be included in a screening programme for hepatocellular carcinoma. Treatment targets for phlebotomy are ferritin <50 μg/L during the induction phase and <100 μg/L during the maintenance phase.
Collapse
|
16
|
Jaccard E, Seyssel K, Gouveia A, Vergely C, Baratali L, Gubelmann C, Froissart M, Favrat B, Marques-Vidal P, Tappy L, Waeber G. Effect of acute iron infusion on insulin secretion: A randomized, double-blind, placebo-controlled trial. EClinicalMedicine 2022; 48:101434. [PMID: 35706490 PMCID: PMC9092517 DOI: 10.1016/j.eclinm.2022.101434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/01/2022] [Accepted: 04/14/2022] [Indexed: 11/29/2022] Open
Abstract
Background Chronic exposure to high iron levels increases diabetes risk partly by inducing oxidative stress, but the consequences of acute iron administration on beta cells are unknown. We tested whether the acute administration of iron for the correction of iron deficiency influenced insulin secretion and the production of reactive oxygen species. Methods Single-center, double-blinded, randomized controlled trial conducted between June 2017 and March 2020. 32 women aged 18 to 47 years, displaying symptomatic iron deficiency without anaemia, were recruited from a community setting and randomly allocated (1:1) to a single infusion of 1000 mg intravenous ferric carboxymaltose (iron) or saline (placebo). The primary outcome was the between group mean difference from baseline to day 28 in first and second phase insulin secretion, assessed by a two-step hyperglycaemic clamp. All analyses were performed by intention to treat. This trial was registered in ClinicalTrials.gov NCT03191201. Findings Iron infusion did not affect first and second phase insulin release. For first phase, the between group mean difference from baseline to day 28 was 0 μU × 10 min/mL [95% CI, -22 to 22, P = 0.99]. For second phase, it was -5 μUx10min/mL [95% CI, -161 to 151; P = 0.95] at the first plateau of the clamp and -249 μUx10min/mL [95% CI, -635 to 137; P = 0.20] at the second plateau. Iron infusion increased serum ascorbyl/ascorbate ratio, a marker of plasma oxidative stress, at day 14, with restoration of normal ratio at day 28 relative to placebo. Finally, high-sensitive C-reactive protein levels remained similar among groups. Interpretation In iron deficient women without anaemia, intravenous administration of 1000 mg of iron in a single sitting did not impair glucose-induced insulin secretion despite a transient increase in the levels of circulating reactive oxygen species. Funding The Swiss National Science Foundation, University of Lausanne and Leenaards, Raymond-Berger and Placide Nicod Foundations.
Collapse
Affiliation(s)
- Evrim Jaccard
- Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, rue du Bugnon 46, Lausanne 1011, Switzerland
| | - Kévin Seyssel
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, rue du Bugnon 7a, Lausanne 1005, Switzerland
| | - Alexandre Gouveia
- Center for Primary Care and Public Health, University of Lausanne, rue du Bugnon 44, Lausanne, Switzerland
| | - Catherine Vergely
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (PEC2, EA7460),UFR des Sciences de Santé, University of Bourgogne Franche-Comté, 7 boulevard Jeanne d’ Arc, Dijon 21079, France
| | - Laila Baratali
- Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, rue du Bugnon 46, Lausanne 1011, Switzerland
| | - Cédric Gubelmann
- Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, rue du Bugnon 46, Lausanne 1011, Switzerland
| | - Marc Froissart
- Clinical Research Center, CHUV, University of Lausanne, Switzerland
| | - Bernard Favrat
- Center for Primary Care and Public Health, University of Lausanne, rue du Bugnon 44, Lausanne, Switzerland
| | - Pedro Marques-Vidal
- Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, rue du Bugnon 46, Lausanne 1011, Switzerland
| | - Luc Tappy
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, rue du Bugnon 7a, Lausanne 1005, Switzerland
| | - Gérard Waeber
- Department of Medicine, Lausanne University Hospital (CHUV) and University of Lausanne, rue du Bugnon 46, Lausanne 1011, Switzerland
| |
Collapse
|
17
|
Le Gac G, Scotet V, Gourlaouen I, L'Hostis C, Merour MC, Karim Z, Deugnier Y, Bardou-Jacquet E, Lefebvre T, Assari S, Ferec C. Prevalence of HFE-related haemochromatosis and secondary causes of hyperferritinaemia and their association with iron overload in 1059 French patients treated by venesection. Aliment Pharmacol Ther 2022; 55:1016-1027. [PMID: 35122291 DOI: 10.1111/apt.16775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/02/2021] [Accepted: 01/04/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Venesection is the key therapy in haemochromatosis, but it remains controversial in hyperferritinaemia with moderate iron accumulation. There is substantial evidence that the results of HFE genotyping are routinely misinterpreted, while elevated serum ferritin has become more frequent in recent years in white adult populations following the increase of obesity and metabolic traits. AIMS To examine the reasons for prescribing venesection in 1,059 French patients during the period 2012-2015, determine the true prevalence of HFE-related haemochromatosis, and compare iron overload profiles between haemochromatosis and non-haemochromatosis patients. RESULTS Only 258 of the 488 patients referred for haemochromatosis had the p.[Cys282Tyr];[Cys282Tyr] disease causative genotype (adjusted prevalence: 24.4%). Of the 801 remaining patients, 112 (14.0%) had the debated p.[Cys282Tyr];[His63Asp] compound heterozygote genotype, 643 (80.3%) had central obesity, 475 (59.3%) had metabolic syndrome (MetS) and 93 (11.6%) were heavy drinkers. The non-haemochromatosis patients started therapeutic venesection 9 years later than haemochromatosis patients (P < 0.001). Despite similar serum ferritin values, they had lower transferrin saturation (41.1% vs 74.3%; P < 0.001), lower amounts of iron removed by venesection (1.7 vs 3.2 g; P < 0.001) and lower hepatic iron concentrations (107 vs 237 µmol/g; P < 0.001). CONCLUSIONS Haemochromatosis is over-diagnosed and is no longer the main reason for therapeutic venesection in France. Obesity and other metabolic abnormalities are frequently associated with mild elevation of serum ferritin, the MetS is confirmed in ~50% of treated patients. There is a minimal relationship between serum ferritin and iron overload in non-p.Cys282Tyr homozygotes. Our observations raise questions about venesection indications in non-haemochromatosis patients.
Collapse
|
18
|
A Pilot Study on the Prevalence of Micronutrient Imbalances in a Dutch General Population Cohort and the Effects of a Digital Lifestyle Program. Nutrients 2022; 14:nu14071426. [PMID: 35406037 PMCID: PMC9003341 DOI: 10.3390/nu14071426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/27/2022] [Accepted: 03/28/2022] [Indexed: 02/06/2023] Open
Abstract
Maintaining an adequate micronutrient status can be achieved by following a complete, diverse diet. Yet, food trends in Western countries show suboptimal consumption of healthy nutrients. In this study, we explored the prevalence of vitamin and mineral imbalances in a general population cohort of Dutch adults and evaluated the effect of a digital lifestyle program on the nutritional status and nutrition health behaviors of these individuals. A micronutrient panel was measured in 348 participants, alongside a dietary assessment. One hundred users subsequently underwent a remeasurement. We identified at least one nutritional imbalance in 301 individuals (86.5%). A total of 80% improved and normalized B6, 67% improved folate, 70% improved B12, and 86% improved vitamin D. Iron abnormalities were corrected in 75% of the participants. In conclusion, this study found that micronutrient deficiencies of easily obtainable vitamins through diet or supplementation such as B vitamins and vitamin D were more prevalent than expected in a Dutch population. This can partly be explained by insufficient consumption of food groups rich in B vitamins. Our preliminary results in those remeasured after a digitally enabled lifestyle intervention show these imbalances can be corrected with adequate behavioral support complemented with supplementation where needed.
Collapse
|
19
|
Bardou-Jacquet E, Hamdi-Roze H, Paisant A, Decraecker M, Bourlière M, Ganne-Carrié N, de Lédinghen V, Bureau C. Non-invasive diagnosis and follow-up of hyperferritinaemia. Clin Res Hepatol Gastroenterol 2022; 46:101762. [PMID: 34332132 DOI: 10.1016/j.clinre.2021.101762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Accepted: 07/23/2021] [Indexed: 02/04/2023]
Abstract
Increased serum ferritin is a very frequent cause of referral for which thorough evaluation is required to avoid unnecessary exploration and inaccurate diagnosis. Clinicians must thus know factors and tools that are relevant in this setting. Several biochemical and radiological tools drastically improved the diagnosis work-up of increased serum ferritin. Because serum ferritin value can be altered by many cofounding factors, scrutiny in the initial clinical evaluation is crucial. Alcohol consumption, and the metabolic syndrome are the most frequent causes of secondary increased ferritin. Serum transferrin saturation level is a pivotal test, and if increased prompt testing for HFE C282Y patients in Caucasian population. In most cases further tests are require to establish whether increased ferritin is associated or not to iron overload. Magnetic resonance imaging is the reference method allowing to accurately establish liver iron content which indirectly reflect body iron load. Second line genetic testing for rare forms of iron overload or increased serum ferritin are available in reference center and should be discussed if diagnosis is equivocal or remain uncertain after careful evaluation. Definite genetic diagnosis is worthwhile as it allows family screening and refining long term management of the patient. Liver biopsy remains seldom useful to assess liver fibrosis, mostly in patients with severe iron overload.
Collapse
Affiliation(s)
- Edouard Bardou-Jacquet
- Edouard Bardou-Jacquet, Service des maladies du foie, CHU Pontchaillou, 2 rue Henri le Guilloux, 35033 Rennes Cedex 9, France.
| | - Houda Hamdi-Roze
- Laboratoire de génétique moléculaire et génomique médicale, CHU Rennes, Rennes, France
| | - Anita Paisant
- Département de radiologie, CHU Angers, Angers, France
| | - Marie Decraecker
- Service d'hépato-gastroentérologie, Hôpital Haut-Lévêque, CHU Bordeaux, Pessac, France
| | - Marc Bourlière
- Service d'hépato-gastroentérologie, Hôpital Saint Joseph, Marseille, France
| | - Nathalie Ganne-Carrié
- Service d'hépatologie, Hôpital Avicenne, CHU Paris Seine-Saint-Denis, APHP, Bobigny, France
| | | | - Christophe Bureau
- Service d'hépatologie, Hôpital Rangueil, CHU Toulouse, Toulouse, France
| |
Collapse
|
20
|
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
|
21
|
Li X, Han J, Liu Y, Liang H. Lactobacillus casei relieves liver injury by regulating immunity and suppression of the enterogenic endotoxin-induced inflammatory response in rats cotreated with alcohol and iron. Food Sci Nutr 2021; 9:5391-5401. [PMID: 34646510 PMCID: PMC8497841 DOI: 10.1002/fsn3.2486] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 06/28/2021] [Accepted: 07/09/2021] [Indexed: 11/30/2022] Open
Abstract
Excessive alcohol and iron intake can reportedly cause liver damage. In the present study, we investigated the effect of Lactobacillus casei on liver injury in rats co-exposed to alcohol and iron and evaluated its possible mechanism. Sixty male Wistar rats were randomly divided into three groups for 12 weeks: the Control group (administered normal saline by gavage and provided a normal diet); alcohol +iron group (Model group, treated with alcohol [3.5-5.3 g/kg/day] by gavage and dietary iron [1,500 mg/kg]); Model group supplemented with L. casei (8 × 108 CFU kg-1 day-1) (L. casei group). Using hematoxylin and eosin (HE) staining and transmission electron microscopy, we observed that L. casei supplementation could alleviate disorders associated with lipid metabolism, inflammation, and intestinal mucosal barrier injury. Moreover, levels of serum alanine aminotransferase, gamma-glutamyl transferase, triglyceride (TG), and hepatic TG were significantly increased in the model group; however, these levels were significantly decreased following the 12-week L. casei supplementation. In addition, we observed notable improvements in intestinal mucosal barrier function and alterations in T lymphocyte subsets and natural killer cells in L. casei-treated rats when compared with the model group. Furthermore, L. casei intervention alleviated serum levels of tumor necrosis factor-α and interleukin-1β, accompanied by decreased serum endotoxin levels and downregulated expression of toll-like receptor 4 and its related molecules MyD88, nuclear factor kappa-B p65, and TNF-α. Accordingly, supplementation with L. casei could effectively improve liver injury induced by the synergistic interaction between alcohol and iron. The underlying mechanism for this improvement may be related to immune regulation and inhibition of enterogenic endotoxin-mediated inflammation.
Collapse
Affiliation(s)
- Xuelong Li
- Department of Human NutritionCollege of Public HealthQingdao UniversityQingdaoChina
- Department of Clinical NutritionThe Affiliated Yantai Yuhuangding Hospital of Qingdao UniversityYantaiChina
| | - Jianmin Han
- Department of Human NutritionCollege of Public HealthQingdao UniversityQingdaoChina
| | - Ying Liu
- Basic Medical CollegeQingdao UniversityQingdaoChina
| | - Hui Liang
- Department of Human NutritionCollege of Public HealthQingdao UniversityQingdaoChina
| |
Collapse
|
22
|
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
|
23
|
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
|
24
|
Iron depletion attenuates steatosis in a mouse model of non-alcoholic fatty liver disease: Role of iron-dependent pathways. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166142. [PMID: 33839281 DOI: 10.1016/j.bbadis.2021.166142] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Iron has been proposed as influencing the progression of liver disease in subjects with non-alcoholic fatty liver disease (NAFLD). We have previously shown that, in the Hfe-/- mouse model of hemochromatosis, feeding of a high-calorie diet (HCD) leads to increased liver injury. In this study we investigated whether the feeding of an iron deficient/HCD to Hfe-/- mice influenced the development of NAFLD. METHODS Liver histology was assessed in Hfe-/- mice fed a standard iron-containing or iron-deficient diet plus or minus a HCD. Hepatic iron concentration, serum transferrin saturation and free fatty acid were measured. Expression of genes implicated in iron regulation and fatty liver disease was determined by quantitative real-time PCR (qRT-PCR). RESULTS Standard iron/HCD-fed mice developed severe steatosis whereas NAS score was reduced in mice fed iron-deficient HCD. Mice fed iron-deficient HCD had lower liver weights, lower transferrin saturation and decreased ferroportin and hepcidin gene expression than HCD-fed mice. Serum non-esterified fatty acids were increased in iron-deficient HCD-fed mice compared with standard iron HCD. Expression analysis indicated that genes involved in fatty-acid binding and mTOR pathways were regulated by iron depletion. CONCLUSIONS Our results indicate that decreasing iron intake attenuates the development of steatosis resulting from a high calorie diet. These results also suggest that human studies of agents that modify iron balance in patients with NAFLD should be revisited.
Collapse
|
25
|
Abstract
Low-grade chronic adipose tissue (AT) inflammation is now recognized as a pivotal driver of the multi-organ dysfunction associated with obesity-related complications; and adipose tissue macrophages (ATMs) are key to the development of this inflammatory milieu. Along with their role in immunosurveillance, ATMs are central regulators of AT iron homeostasis. Under optimal conditions, ATMs maintain a proper homeostatic balance of iron in adipocytes; however, during obesity, this relationship is altered, and iron is repartitioned into adipocytes as opposed to ATMs. This adipocyte iron overload leads to systemic IR and the mechanism for these effects is still under investigation. Here, we comment on the most recent findings addressing the interplay between adipocyte and ATM iron handling, and metabolic dysfunction.
Collapse
|
26
|
Vaquero MP, Martínez-Maqueda D, Gallego-Narbón A, Zapatera B, Pérez-Jiménez J. Relationship between iron status markers and insulin resistance: an exploratory study in subjects with excess body weight. PeerJ 2020; 8:e9528. [PMID: 32821534 PMCID: PMC7397981 DOI: 10.7717/peerj.9528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
Background Controversy exists on the relationship between iron metabolism and cardiometabolic risk. The aim of this study was to determine if there is a link between dysmetabolic iron and cardiometabolic markers in subjects with excess body weight. Methods Cross-sectional study with fifty participants presenting overweight or obesity and at least another metabolic syndrome factor. Determinations: anthropometry, body composition, blood pressure, lipids, glucose, insulin, leptin, areas under the curve (AUC) for glucose and insulin after an oral glucose tolerance test, hs-C reactive protein (hs-CRP), blood count, ferritin, transferrin, transferrin saturation (TSAT), soluble transferrin receptor (sTfR). Gender-adjusted linear correlations and two independent samples t tests were used. Results Ferritin was positively correlated with insulin-AUC (r = 0.547, p = 0.008) and TSAT was negatively correlated with waist-hip ratio (r = − 0.385, p = 0.008), insulin (r = − 0.551, p < 0.001), and insulin resistance (HOMA-IR, r = − 0.586, p < 0.001). Subjects with TSAT ≤ 20% had higher insulin (p = 0.012) and HOMA-IR (p = 0.003) compared to those with TSAT > 20%. In conclusion, the observed results suggest that iron transport and storage are altered in subjects with overweight/obesity, at the same time that they exhibit the characteristic features of insulin resistance. Nevertheless, this occurs without iron overload or deficiency. These results should be validated in wider cohorts since they suggest that iron transport and storage should be assessed when performing the clinical evaluation of subjects with excess body weight.
Collapse
Affiliation(s)
- M Pilar Vaquero
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| | - Daniel Martínez-Maqueda
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain.,Madrid Institute for Rural, Agricultural and Food Research and Development (IMIDRA), Madrid, Spain
| | - Angélica Gallego-Narbón
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain.,Department of Biology, Universidad Autónoma de Madrid (UAM), Madrid, Spain, España
| | - Belén Zapatera
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| | - Jara Pérez-Jiménez
- Department of Metabolism and Nutrition, Institute of Food Science and Technology and Nutrition (ICTAN-CSIC), Madrid, Spain
| |
Collapse
|
27
|
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
|
28
|
Martin-Rodriguez J, Gonzalez-Cantero J, Gonzalez-Cantero A, Martí-Bonmatí L, Alberich-Bayarri Á, Gonzalez-Cejudo T, Gonzalez-Calvin J. Insulin resistance and NAFLD: Relationship with intrahepatic iron and serum TNF-α using 1H MR spectroscopy and MRI. DIABETES & METABOLISM 2019; 45:473-479. [PMID: 30660761 DOI: 10.1016/j.diabet.2019.01.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 12/18/2018] [Accepted: 01/09/2019] [Indexed: 02/07/2023]
|
29
|
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
|
30
|
Buzzetti E, Petta S, Manuguerra R, Luong TV, Cabibi D, Corradini E, Craxì A, Pinzani M, Tsochatzis E, Pietrangelo A. Evaluating the association of serum ferritin and hepatic iron with disease severity in non-alcoholic fatty liver disease. Liver Int 2019; 39:1325-1334. [PMID: 30851216 DOI: 10.1111/liv.14096] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/25/2019] [Accepted: 02/28/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Hyperferritinemia, with or without increased hepatic iron, represents a common finding in non-alcoholic fatty liver disease (NAFLD). However, it is unclear whether it reflects hepatic inflammation or true iron-overload and, in case the latter is confirmed, whether this influences disease progression. We therefore explored the association between serum ferritin, degree and pattern of hepatic iron deposition and liver disease severity in patients with NAFLD. METHODS We selected 468 patients with biopsy-proven NAFLD from 2 European centres. Iron, hepatic and metabolic parameters were collected at the time of liver biopsy. Iron deposits in hepatocytes and reticuloendothelial cells were assessed and graded. Diagnosis of non-alcoholic steatohepatitis (NASH) and fibrosis staging were performed. RESULTS A total of 122 (26%) patients had hyperferritinemia, whereas stainable hepatic iron was found in 116 (25%) patients (38% predominantly in hepatocytes, 20% in reticuloendothelial cells and 42% in both). Subjects with stainable hepatic iron, particularly those with a mixed pattern, had higher serum ferritin and transaminases but only a mixed pattern of iron deposition was among the variables significantly associated with presence of NASH. Serum ferritin was not associated with presence of NASH, however it increased with worsening fibrosis stage (F3 compared to F0-F1), and significantly decreased in stage F4. CONCLUSIONS A mixed pattern of hepatic iron deposition is associated with the presence of steatohepatitis, while serum ferritin increases with worsening fibrosis up to pre-cirrhotic stage. In individual NAFLD patients, serum ferritin could be evaluated as part of non-invasive diagnostic panels but not on its own.
Collapse
Affiliation(s)
- Elena Buzzetti
- Division of Internal Medicine 2 and Center for Hemochromatosis, University of Modena and Reggio Emilia, Modena, Italy.,UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Salvatore Petta
- Section of Gastroenterology and Hepatology, Di.Bi.M.I.S., University of Palermo, Palermo, Italy
| | | | - Tu Vinh Luong
- Department of Cellular Pathology, Royal Free Hospital, London, UK
| | - Daniela Cabibi
- Department of Sciences for the Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, Palermo, Italy
| | - Elena Corradini
- Division of Internal Medicine 2 and Center for Hemochromatosis, University of Modena and Reggio Emilia, Modena, Italy
| | - Antonio Craxì
- Section of Gastroenterology and Hepatology, Di.Bi.M.I.S., University of Palermo, Palermo, Italy
| | - Massimo Pinzani
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Emmanuel Tsochatzis
- UCL Institute for Liver and Digestive Health, Royal Free Hospital, London, UK
| | - Antonello Pietrangelo
- Division of Internal Medicine 2 and Center for Hemochromatosis, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
31
|
Effect of procyanidin on dietary iron absorption in hereditary hemochromatosis and in dysmetabolic iron overload syndrome: A crossover double-blind randomized controlled trial. Clin Nutr 2019; 39:97-103. [PMID: 30792142 DOI: 10.1016/j.clnu.2019.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/31/2019] [Accepted: 02/01/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND & AIMS Type I hereditary hemochromatosis (HH) and dysmetabolic iron overload syndrome (DIOS) are the two most prevalent iron overload diseases. Although many food components, particularly polyphenols, reduce iron bioavailability, there is no clinically validated nutritional strategy to reduce food-iron absorption in patients with these diseases. We aimed to determine whether supplementation with 100 mg of procyanidins during a meal reduces dietary iron absorption in patients with HH or DIOS. METHODS 20 HH and 20 DIOS patients were enrolled in a double-blind three-period crossover randomized study. Basal serum iron level was measured following an overnight fast. Each patient consumed a standardized test iron-rich meal containing 43 mg of iron with two capsules of placebo or procyanidin supplementation. Each period was separated by a 3-day wash-out period. The primary objective was a reduction of dietary iron absorption, assessed by a reduction of serum-iron area under the curve (AUC) corrected for baseline serum iron. RESULTS All patients completed the study. The meal and the procyanidin supplements were well tolerated. In both HH and DIOS patients, the iron-rich meal induced a significant increase of serum iron compared with baseline at 120, 180, 240 min, from 8 to 9.1% (p = 0.002, 0.001 and 0.003, respectively) in DIOS and from 15.8 to 25.7% (p < 0.001) in HH. Iron absorption was 3.5-fold higher in HH than in DIOS (p < 0.001). Procyanidin supplementation did not significantly modify iron absorption in DIOS (AUC of added iron 332.87 ± 649.55 vs 312.61 ± 678.61 μmol.h/L, p = 0.916) or in HH (1168.62 ± 652.87 vs 1148.54 μmol.h/L ± 1290.05, p = 0.917). CONCLUSIONS An iron-rich test meal led to a marked increase in iron absorption in HH but a mild increase in DIOS. Procyanidin supplementation does not significantly reduce dietary iron absorption in either disease. CLINICAL TRIAL REGISTRY: clinicaltrials.gov (NCT03453918).
Collapse
|
32
|
Mehta KJ, Farnaud SJ, Sharp PA. Iron and liver fibrosis: Mechanistic and clinical aspects. World J Gastroenterol 2019; 25:521-538. [PMID: 30774269 PMCID: PMC6371002 DOI: 10.3748/wjg.v25.i5.521] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/02/2019] [Accepted: 01/10/2019] [Indexed: 02/06/2023] Open
Abstract
Liver fibrosis is characterised by excessive deposition of extracellular matrix that interrupts normal liver functionality. It is a pathological stage in several untreated chronic liver diseases such as the iron overload syndrome hereditary haemochromatosis, viral hepatitis, alcoholic liver disease, non-alcoholic fatty liver disease, non-alcoholic steatohepatitis and diabetes. Interestingly, regardless of the aetiology, iron-loading is frequently observed in chronic liver diseases. Excess iron can feed the Fenton reaction to generate unquenchable amounts of free radicals that cause grave cellular and tissue damage and thereby contribute to fibrosis. Moreover, excess iron can induce fibrosis-promoting signals in the parenchymal and non-parenchymal cells, which accelerate disease progression and exacerbate liver pathology. Fibrosis regression is achievable following treatment, but if untreated or unsuccessful, it can progress to the irreversible cirrhotic stage leading to organ failure and hepatocellular carcinoma, where resection or transplantation remain the only curative options. Therefore, understanding the role of iron in liver fibrosis is extremely essential as it can help in formulating iron-related diagnostic, prognostic and treatment strategies. These can be implemented in isolation or in combination with the current approaches to prepone detection, and halt or decelerate fibrosis progression before it reaches the irreparable stage. Thus, this review narrates the role of iron in liver fibrosis. It examines the underlying mechanisms by which excess iron can facilitate fibrotic responses. It describes the role of iron in various clinical pathologies and lastly, highlights the significance and potential of iron-related proteins in the diagnosis and therapeutics of liver fibrosis.
Collapse
Affiliation(s)
- Kosha J Mehta
- School of Population Health and Environmental Sciences, Faculty of Life Sciences and Medicine, King’s College London, London SE1 1UL, United Kingdom
- Division of Human Sciences, School of Applied Sciences, London South Bank University, London SE1 0AA, United Kingdom
| | - Sebastien Je Farnaud
- Faculty Research Centre for Sport, Exercise and Life Sciences, Coventry University, Coventry CV1 2DS, United Kingdom
| | - Paul A Sharp
- Department of Nutritional Sciences, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King's College London, London SE1 9NH, United Kingdom
| |
Collapse
|
33
|
Golfeyz S, Lewis S, Weisberg IS. Hemochromatosis: pathophysiology, evaluation, and management of hepatic iron overload with a focus on MRI. Expert Rev Gastroenterol Hepatol 2018; 12:767-778. [PMID: 29966105 DOI: 10.1080/17474124.2018.1496016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hereditary hemochromatosis (HH) is an autosomal recessive disorder that occurs in approximately 1 in 200-250 individuals. Mutations in the HFE gene lead to excess iron absorption. Excess iron in the form of non-transferrin-bound iron (NTBI) causes injury and is readily uptaken by cardiomyocytes, pancreatic islet cells, and hepatocytes. Symptoms greatly vary among patients and include fatigue, abdominal pain, arthralgias, impotence, decreased libido, diabetes, and heart failure. Untreated hemochromatosis can lead to chronic liver disease, fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Many invasive and noninvasive diagnostic tests are available to aid in diagnosis and treatment. MRI has emerged as the reference standard imaging modality for the detection and quantification of hepatic iron deposition, as ultrasound (US) is unable to detect iron overload and computed tomography (CT) findings are nonspecific and influenced by multiple confounding variables. If caught and treated early, HH disease progression can significantly be altered. Area covered: The data on Hemochromatosis, iron overload, and MRI were gathered by searching PubMed. Expert commentary: MRI is a great tool for diagnosis and management of iron overload. It is safe, effective, and a standard protocol should be included in diagnostic algorithms of future treatment guidelines.
Collapse
Affiliation(s)
- Shmuel Golfeyz
- a Department of Internal Medicine , Mount Sinai Beth Israel , New York , NY , USA
| | - Sara Lewis
- b Department of Radiology , Icahn School of Medicine at Mount Sinai , New York , NY , USA.,c Translational and Molecular Imaging Institute , Icahn School of Medicine at Mount Sinai , New York , NY , USA
| | - Ilan S Weisberg
- d Department of Digestive Diseases and Hepatology , Mount Sinai Beth Israel , New York , NY , USA
| |
Collapse
|
34
|
Britton L, Bridle K, Reiling J, Santrampurwala N, Wockner L, Ching H, Stuart K, Subramaniam VN, Jeffrey G, St Pierre T, House M, Gummer J, Trengove R, Olynyk J, Crawford D, Adams L. Hepatic iron concentration correlates with insulin sensitivity in nonalcoholic fatty liver disease. Hepatol Commun 2018; 2:644-653. [PMID: 29881816 PMCID: PMC5983226 DOI: 10.1002/hep4.1190] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 03/23/2018] [Accepted: 04/01/2018] [Indexed: 01/01/2023] Open
Abstract
Rodent and cell‐culture models support a role for iron‐related adipokine dysregulation and insulin resistance in the pathogenesis of nonalcoholic fatty liver disease (NAFLD); however, substantial human data are lacking. We examined the relationship between measures of iron status, adipokines, and insulin resistance in patients with NAFLD in the presence and absence of venesection. This study forms part of the Impact of Iron on Insulin Resistance and Liver Histology in Nonalcoholic Steatohepatitis (IIRON2) study, a prospective randomized controlled trial of venesection for adults with NAFLD. Paired serum samples at baseline and 6 months (end of treatment) in controls (n = 28) and patients who had venesection (n = 23) were assayed for adiponectin, leptin, resistin, retinol binding protein‐4, tumor necrosis factor α, and interleukin‐6, using a Quantibody, customized, multiplexed enzyme‐linked immunosorbent assay array. Hepatic iron concentration (HIC) was determined using MR FerriScan. Unexpectedly, analysis revealed a significant positive correlation between baseline serum adiponectin concentration and HIC, which strengthened after correction for age, sex, and body mass index (rho = 0.36; P = 0.007). In addition, there were significant inverse correlations between HIC and measures of insulin resistance (adipose tissue insulin resistance (Adipo‐IR), serum insulin, serum glucose, homeostasis model assessment of insulin resistance, hemoglobin A1c, and hepatic steatosis), whereas a positive correlation was noted with the insulin sensitivity index. Changes in serum adipokines over 6 months did not differ between the control and venesection groups. Conclusion: HIC positively correlates with serum adiponectin and insulin sensitivity in patients with NAFLD. Further study is required to establish causality and mechanistic explanations for these associations and their relevance in the pathogenesis of insulin resistance and NAFLD. (Hepatology Communications 2018;2:644‐653)
Collapse
Affiliation(s)
- Laurence Britton
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia.,Department of Gastroenterology Princess Alexandra Hospital Woolloongabba Australia.,QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Kim Bridle
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia
| | - Janske Reiling
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia.,Department of Surgery, NUTRIM School of Nutrition and Translational Research in Metabolism Maastricht University Maastricht the Netherlands
| | - Nishreen Santrampurwala
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia.,QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Leesa Wockner
- QIMR Berghofer Medical Research Institute Brisbane Australia
| | - Helena Ching
- Medical School, Faculty of Health Sciences University of Western Australia Crawley Australia
| | - Katherine Stuart
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,Department of Gastroenterology Princess Alexandra Hospital Woolloongabba Australia
| | - V Nathan Subramaniam
- QIMR Berghofer Medical Research Institute Brisbane Australia.,Institute of Health and Biomedical Innovation and School of Biomedical Sciences Queensland University of Technology Kelvin Grove Australia
| | - Gary Jeffrey
- Medical School, Faculty of Health Sciences University of Western Australia Crawley Australia.,Department of Hepatology Sir Charles Gairdner Hospital Perth Australia
| | - Tim St Pierre
- School of Physics University of Western Australia Crawley Australia
| | - Michael House
- School of Physics University of Western Australia Crawley Australia
| | - Joel Gummer
- Separation Science and Metabolomics Laboratory (Metabolomics Australia, Western Australia node) Murdoch University Murdoch Australia
| | - Robert Trengove
- Separation Science and Metabolomics Laboratory (Metabolomics Australia, Western Australia node) Murdoch University Murdoch Australia
| | - John Olynyk
- Department of Gastroenterology Fiona Stanley and Fremantle Hospital Group Murdoch Australia.,School of Health and Medical Sciences Edith Cowan University Joondalup Australia
| | - Darrell Crawford
- Gallipoli Medical Research Institute Greenslopes Private Hospital Greenslopes Australia.,University of Queensland Herston Australia
| | - Leon Adams
- Medical School, Faculty of Health Sciences University of Western Australia Crawley Australia.,Department of Hepatology Sir Charles Gairdner Hospital Perth Australia
| |
Collapse
|
35
|
Murali AR, Gupta A, Brown K. Systematic review and meta-analysis to determine the impact of iron depletion in dysmetabolic iron overload syndrome and non-alcoholic fatty liver disease. Hepatol Res 2018; 48:E30-E41. [PMID: 28593739 DOI: 10.1111/hepr.12921] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Revised: 05/23/2017] [Accepted: 06/01/2017] [Indexed: 12/12/2022]
Abstract
AIMS Iron reduction has been proposed as treatment for dysmetabolic iron overload syndrome (DIOS) and non-alcoholic fatty liver disease (NAFLD), but results of published trials are conflicting. We undertook a systematic review and meta-analysis to determine the impact of phlebotomy in DIOS and NAFLD. METHODS We searched multiple databases systematically for studies evaluating the impact of phlebotomy in DIOS and NAFLD. We calculated weighted summary estimates using the inverse variance method. Study quality was assessed using the Cochrane collaboration tool. RESULTS We identified nine studies with 820 patients (427 had phlebotomy, 393 lifestyle changes alone). Iron depletion did not improve the Homeostasis Model Assessment (HOMA) index (mean difference [MD] -0.6; confidence interval (CI), -1.7, 0.5; P = 0.3), insulin level (MD -0.8 mU/L; CI, -5.3, 3.7; P = 0.73), or aspartate aminotransferase (AST) (MD -0.7 IU/L; CI, -3.2, 1.8; P = 0.6) in DIOS and/or NAFLD patients as compared to lifestyle changes alone (five studies, 626 patients). There was mild improvement in alanine aminotransferase (ALT) (MD -6.6 IU/L; CI, -11, -2.1); P < 0.01), but the effect size was very small (Cohen's d, 0.15; r statistic, 0.07). Even in the subgroup of patients with NAFLD and hyperferritinemia, phlebotomy did not improve the HOMA index, insulin level, ALT, or AST. Additionally, no study showed significant improvement in liver inflammation or fibrosis with iron reduction. CONCLUSIONS Phlebotomy does not bring about significant improvement in indices of insulin resistance, liver enzymes, or liver histology in patients with DIOS and/or NAFLD compared to lifestyle changes alone. Current evidence does not support the use of phlebotomy in patients with DIOS or NAFLD.
Collapse
Affiliation(s)
- Arvind R Murali
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Administration Medical Center, Iowa City, Iowa
| | | | - Kyle Brown
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa.,Iowa City Veterans Administration Medical Center, Iowa City, Iowa.,Program in Free Radical and Radiation Biology, Department of Radiation Oncology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| |
Collapse
|
36
|
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
|
37
|
FOXO1/3: Potential suppressors of fibrosis. Ageing Res Rev 2018; 41:42-52. [PMID: 29138094 DOI: 10.1016/j.arr.2017.11.002] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 10/07/2017] [Accepted: 11/08/2017] [Indexed: 02/06/2023]
Abstract
Fibrosis is a universally age-related disease that involves nearly all organs. It is typically initiated by organic injury and eventually results in organ failure. There are still few effective therapeutic strategy targets for fibrogenesis. Forkhead box proteins O1 and O3 (FOXO1/3) have been shown to have favorable inhibitory effects on fibroblast activation and subsequent extracellular matrix production and can ameliorate fibrosis levels in numerous organs, including the heart, liver, lung, and kidney; they are therefore promising targets for anti-fibrosis therapy. Moreover, we can develop appropriate strategies to make the best use of FOXO1/3's anti-fibrosis properties. The information reviewed here should be significant for understanding the roles of FOXO1/3 in fibrosis and should contribute to the design of further studies related to FOXO1/3 and the fibrotic response and shed light on a potential treatment for fibrosis.
Collapse
|
38
|
Castiella A, Alústiza JM, Zapata E, Zubiaurre L, Otazua P, Emparanza JI. Dysmetabolic iron overload syndrome: The need for an accurate liver iron concentration determination by magnetic resonance imaging. Hepatology 2017; 65:2119. [PMID: 28066930 DOI: 10.1002/hep.29030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
| | | | - Eva Zapata
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain
| | | | - Pedro Otazua
- Gastroenterology Service, Mondragon Hospital, Mondragon, Spain
| | - José I Emparanza
- Clinical Epidemiology Unit, CASPe, CIBER-ESP, Donostia University Hospital, Donostia, Spain
| |
Collapse
|
39
|
Lainé F, Bardou-Jacquet E, Paisant A, Gandon Y, Deugnier Y. Reply. Hepatology 2017; 65:2119-2120. [PMID: 28066931 DOI: 10.1002/hep.29032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Affiliation(s)
- Fabrice Lainé
- INSERM CIC 1414, Rennes, France.,Liver Unit, CHU Pontchaillou, Rennes, France
| | - Edouard Bardou-Jacquet
- INSERM CIC 1414, Rennes, France.,Liver Unit, CHU Pontchaillou, Rennes, France.,University of Rennes, Rennes, France
| | - Anita Paisant
- INSERM CIC 1414, Rennes, France.,University of Rennes, Rennes, France.,Department of Abdominal Imaging, CHU Pontchaillou, Rennes, France
| | - Yves Gandon
- University of Rennes, Rennes, France.,Department of Abdominal Imaging, CHU Pontchaillou, Rennes, France
| | - Yves Deugnier
- INSERM CIC 1414, Rennes, France.,Liver Unit, CHU Pontchaillou, Rennes, France.,University of Rennes, Rennes, France
| |
Collapse
|