1
|
Yu C, Delatycki M, Hussain SM, McNeil JJ, Lacaze P, Olynyk JK. Haemochromatosis Genotypes and Incident Dementia in a Prospective Study of Older Adults. Neurology 2025; 104:e213743. [PMID: 40440589 PMCID: PMC12123751 DOI: 10.1212/wnl.0000000000213743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 04/03/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND AND OBJECTIVES Variants in the homeostatic iron regulator (HFE) gene are prevalent among individuals of European ancestry and have been linked to an increased risk of dementia. This study aimed to evaluate the effects of HFE p.Cys282Tyr and p.His63Asp variants on serum ferritin levels and the incidence of dementia in a cohort of initially healthy older adults. METHOD This prospective longitudinal study used data from the Aspirin in Reducing Events in the Elderly trial. Participants had no history of cardiovascular disease, dementia, or cognitive decline at enrollment. Genotyping for HFE p.Cys282Tyr and p.His63Asp variants was conducted using microarrays, and baseline serum ferritin concentrations were measured in peripheral blood samples. Dementia diagnoses were confirmed by an adjudication committee over a median follow-up of 6.4 years. Associations were evaluated using Cox proportional hazards models adjusted for related covariates. RESULTS The study included 12,174 unrelated, healthy participants of European ancestry aged 70 years or older, comprising 5,583 men (45.9%) and 6,591 women (54.1%). The median age was 73.7 years (interquartile range [IQR]: 71.6-76.9) for men and 73.9 years (IQR: 71.7-77.5) for women. Compared with the wild-type group, men with p.Cys282Tyr+/+ (p = 0.048) and p.Cys282Tyr+/p.His63Asp + genotypes (p < 0.001) had significantly higher baseline ferritin levels. Women with p.His63Asp+/+ (p = 0.015) and p.Cys282Tyr+/p.His63Asp+ (p < 0.001) genotypes also exhibited elevated ferritin levels. No significant association was observed between baseline serum ferritin levels and dementia risk. However, men with p.His63Asp+/+ genotype had a significantly higher risk of incident dementia (adjusted hazard ratio = 2.39, 95% CI 1.25-4.57, p = 0.009) compared with those without HFE variations. This association was not observed in women. DISCUSSION Among initially healthy older adults, HFE p.His63Asp homozygosity was associated with a higher risk of incident dementia in men but not women. These findings highlight a potential sex-specific genetic risk factor for dementia and warrant further research into the underlying mechanisms linking p.His63Asp and dementia.
Collapse
Affiliation(s)
- Chenglong Yu
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Martin Delatycki
- Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia
- Victorian Clinical Genetics Service, The Royal Children's Hospital, Parkville, VIC, Australia
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Parkville, VIC, Australia
| | - Sultana Monira Hussain
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - John James McNeil
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - Paul Lacaze
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC, Australia
| | - John K Olynyk
- Curtin Medical School, Curtin University, Bentley, Australia; and
- Department of Gastroenterology, Fiona Stanley Hospital, Murdoch, Australia
| |
Collapse
|
2
|
Beriault DR, Chen Y, Yip P, Blasutig I, Bhayana V, Rutledge AC, Parker M, Kinniburgh D, Thomas D, Colbourne P, Langman L, Delaney SR, Bennett M, Oleschuk C, Huang Y, Rodriguez-Capote K, Hauff K, Konforte D, Kalra J, Bouhtiauy I, El Hassan MA, Elnenaei M, Thorlacius L, Venner AA, Randell EW, Sohn KY, Leung F, Taher J, Lou A, Arnoldo S, CSCC Utilization Special Interest Group. Choosing Wisely Canada recommendations for clinical biochemistry: test ordering for sustainable and high-quality patient care. Clin Biochem 2025:110951. [PMID: 40409393 DOI: 10.1016/j.clinbiochem.2025.110951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 04/16/2025] [Accepted: 04/30/2025] [Indexed: 05/25/2025]
Abstract
Laboratory Medicine is growing at a rapid rate in both the breadth of unique tests and the total number of tests performed per year. Inappropriate overutilization of laboratory tests can lead to patient harm, excessive environmental waste and increased carbon emissions. A focus on reducing inefficiencies in healthcare is needed to ensure a robust and sustainable healthcare system. To promote laboratory sustainability, the Canadian Society of Clinical Chemists (CSCC) has developed ten recommendations related to medical tests within clinical biochemistry. These recommendations are designed as 'low-hanging fruit' that should be adopted by both hospital and community laboratories. By implementing automated strategies and/or educational approaches to reduce misuse of laboratory resources, clinical laboratories can move toward a more sustainable model that improves patient care. This list of recommendations, created for Choosing Wisely Canada, covers tests for diabetes, celiac disease, monoclonal gammopathies, iron disorders, liver disorders, kidney disorders, substance use disorders, and allergen testing.
Collapse
Affiliation(s)
- Daniel R Beriault
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine, Unity Health Toronto, Toronto, ON, Canada
| | - Yu Chen
- Department of Laboratory Medicine and Pathology, Dr. Everett Chalmers Regional Hospital, Horizon Health Network, Fredericton, NB, Canada; Department of Pathology, Dalhousie University, Halifax, NS, Canada; Laboratory Medicine, Faculty of Medicine, Memorial University, St John's, NL, Canada
| | - Paul Yip
- Precision Diagnostics & Therapeutics Program, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Ivan Blasutig
- Division of Biochemistry, Children's Hospital of Eastern Ontario Eastern Ontario Regional Laboratory Association (ELORA), Ottawa, ON, Canada; Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Vipin Bhayana
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON, Canada
| | - Angela C Rutledge
- Department of Pathology and Laboratory Medicine, London Health Sciences Centre, London, ON, Canada
| | - Michelle Parker
- Alberta Precision Laboratories, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - David Kinniburgh
- Department of Physiology and Pharmacology, Alberta Centre for Toxicology, University of Calgary, Calgary, AB, Canada
| | - Dylan Thomas
- Alberta Precision Laboratories, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Penny Colbourne
- Alberta Precision Laboratories, Edmonton, AB, Canada; Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Loralie Langman
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sarah R Delaney
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine, Unity Health Toronto, Toronto, ON, Canada
| | - Melissa Bennett
- Department of Physiology and Pharmacology, Alberta Centre for Toxicology, University of Calgary, Calgary, AB, Canada
| | - Curtis Oleschuk
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Yun Huang
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Karina Rodriguez-Capote
- Department of Pathology and Laboratory Medicine, Interior Health Authority, Kelowna, BC, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, BC, Canada
| | - Kristin Hauff
- Department of Pathology and Laboratory Medicine, Interior Health Authority, Kelowna, BC, Canada; Department of Pathology & Laboratory Medicine, University of British Columbia, BC, Canada
| | | | - Jay Kalra
- Department of Pathology and Lab Medicine, University of Saskatchewan and Saskatchewan Health Authority, Saskatoon, SK, Canada
| | - Ihssan Bouhtiauy
- Department of Biochemistry, Vitalite Health Network, and Universite de Moncton Campus Edmundston, NB, Canada
| | - Mohamed Abou El Hassan
- Department of Pathology, Dalhousie University, Halifax, NS, Canada; Lifelabs Medical Laboratory Services, ON, Canada
| | - Manal Elnenaei
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, NS, Canada
| | - Laurel Thorlacius
- Clinical Biochemistry, Diagnostic Services, Shared Health. Max Rady College of Medicine, University of Manitoba, MB, Canada
| | - Allison A Venner
- Alberta Precision Laboratories, Calgary, AB, Canada; Department of Pathology and Laboratory Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Edward W Randell
- Laboratory Medicine, Faculty of Medicine, Memorial University, St John's, NL, Canada
| | - Kun-Young Sohn
- Laboratory Medicine and Genetics Program, Trillium Health Partners, Mississauga, ON, Canada
| | - Felix Leung
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Jennifer Taher
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Pathology and Laboratory Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Amy Lou
- Department of Pathology and Laboratory Medicine, Dalhousie University, Halifax, NS, Canada
| | - Saranya Arnoldo
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada; Department of Laboratory Medicine, Unity Health Toronto, Toronto, ON, Canada
| | | |
Collapse
|
3
|
Mitchell NDP, Pierre TGS, Ramm LE, Ramm GA, Olynyk JK. Risk profiling for cirrhosis and hepatocellular carcinoma in HFE hemochromatosis using mobilizable iron stores and alcohol consumption. Sci Rep 2025; 15:16011. [PMID: 40341892 PMCID: PMC12062215 DOI: 10.1038/s41598-025-99672-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
HFE hemochromatosis (HH) may cause cirrhosis and hepatocellular carcinoma (HCC). Progression to these endpoints depends on the severity of iron overload and cofactors, such as alcohol. We evaluated alcohol and iron-related risk factors in relation to cirrhosis at diagnosis and future development of HCC in a retrospective analysis of 197 HH subjects. The proportion of subjects either with cirrhosis or who developed HCC during follow-up were 29/197 (14.7%) or 10/197 (5.1%), respectively. The median (IQR) follow-up time after diagnosis was 15.2 (4.6 to 22.1) years. The median mobilizable iron stores and daily alcohol consumption (IQR) were 6.0 (3.8-11.0) g and 20 (0-40) g, respectively. An optimal logistic regression model for the odds of cirrhosis was developed by adding candidate liver insult variables (mobilizable iron, alcohol consumption, and age as a surrogate for duration of exposure) in a forward stepwise strategy using area under the receiver operating characteristic curve (AUROC) analysis and the corrected Akaike information criterion. This model demonstrated an AUROC (95% CI) of 0.966 (0.935-0.996), with sensitivity 76 (58-88)% and specificity 97 (93-99) % for prediction of cirrhosis and had a negative predictive value of 99.4 (95% CI 96.7-99.97) % for development of HCC. Thus, future risk of HCC can be assessed from mobilizable iron stores and alcohol consumption of HH subjects.
Collapse
Affiliation(s)
| | - Timothy G St Pierre
- School of Physics, Mathematics, and Computing, The University of Western Australia, Perth, WA, Australia
| | - Louise E Ramm
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - Grant A Ramm
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, QLD, Australia
| | - John K Olynyk
- Department of Gastroenterology, Fiona Stanley Hospital, Murdoch, WA, Australia.
- Curtin Medical School, Curtin University, Kent St., Bentley, WA, 6102, Australia.
| |
Collapse
|
4
|
Lucas MR, Pilling LC, Atkins JL, Melzer D. Incidence of liver complications with hemochromatosis-associated HFE p.C282Y homozygosity: The role of central adiposity. Hepatology 2025; 81:1522-1534. [PMID: 39178373 PMCID: PMC11999091 DOI: 10.1097/hep.0000000000001056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 07/30/2024] [Indexed: 08/25/2024]
Abstract
BACKGROUND AND AIMS The HFE p.C282Y+/+ (homozygous) genotype and central adiposity both increase liver disease and diabetes risks, but the combined effects are unclear. We estimated waist-to-hip ratio (WHR) associations with incident clinical outcomes in routine care in p.C282Y+/+ participants in the UK Biobank community cohort. APPROACH AND RESULTS Baseline WHR data available in 1297 male and 1602 female p.C282Y+/+ with 13.3-year mean follow-up for diagnoses. Spline regressions and Cox proportional hazard models were adjusted for age and genetic principal components. Cumulative incidence was from age 40 to 80 years. In p.C282Y+/+ males, there were positive linear WHR relationships for hospital inpatient-diagnosed liver fibrosis/cirrhosis ( p = 2.4 × 10 -5 ), liver cancer ( p = 0.007), non-alcoholic fatty liver disease ( p = 7.7 × 10 -11 ), and type 2 diabetes ( p = 5.1 × 10 -16 ). The hazard ratio for high WHR in p.C282Y+/+ males (≥0.96; 33.9%) was 4.13 for liver fibrosis/cirrhosis (95% CI: 2.04-8.39, p = 8.4 × 10 -5 vs. normal WHR); cumulative age 80 incidence 15.0% (95% CI: 9.8%-22.6%) versus 3.9% (95% CI: 1.9%-7.6%); for liver cancer, cumulative incidence was 9.2% (95% CI: 5.7%-14.6%) versus 3.6% (95% CI: 1.9%-6.6%). Hemochromatosis was diagnosed in 23 (96%) of the 24 high WHR p.C282Y+/+ males with incident fibrosis/cirrhosis. High WHR (≥0.85; 30.0%) p.C282Y+/+ females had raised hazards for liver fibrosis/cirrhosis (hazard ratio = 9.17, 95% CI: 2.51-33.50, p = 3.8 × 10 -7 ) and Non-alcoholic fatty liver disease (hazard ratio = 5.17, 95% CI: 2.48-10.78, p = 1.2 × 10 -5 ). Fibrosis/cirrhosis associations were similar in the subset with additional primary care diagnoses. CONCLUSIONS In p.C282Y+/+ males and females, increasing WHR is associated with substantially higher risks of liver complications. Interventions to reduce central adiposity to improve these outcomes should be tested.
Collapse
|
5
|
Kouroumalis E, Tsomidis I, Voumvouraki A. HFE-Related Hemochromatosis May Be a Primary Kupffer Cell Disease. Biomedicines 2025; 13:683. [PMID: 40149659 PMCID: PMC11940282 DOI: 10.3390/biomedicines13030683] [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: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/08/2025] [Indexed: 03/29/2025] Open
Abstract
Iron overload can lead to increased deposition of iron and cause organ damage in the liver, the pancreas, the heart and the synovium. Iron overload disorders are due to either genetic or acquired abnormalities such as excess transfusions or chronic liver diseases. The most common genetic disease of iron deposition is classic hemochromatosis (HH) type 1, which is caused by mutations of HFE. Other rare forms of HH include type 2A with mutations at the gene hemojuvelin or type 2B with mutations in HAMP that encodes hepcidin. HH type 3, is caused by mutations of the gene that encodes transferrin receptor 2. Mutations of SLC40A1 which encodes ferroportin cause either HH type 4A or HH type 4B. In the present review, an overview of iron metabolism including absorption by enterocytes and regulation of iron by macrophages, liver sinusoidal endothelial cells (LSECs) and hepatocyte production of hepcidin is presented. Hereditary Hemochromatosis and the current pathogenetic model are analyzed. Finally, a new hypothesis based on published data was suggested. The Kupffer cell is the primary defect in HFE hemochromatosis (and possibly in types 2 and 3), while the hepcidin-relative deficiency, which is the common underlying abnormality in the three types of HH, is a secondary consequence.
Collapse
Affiliation(s)
- Elias Kouroumalis
- Department of Gastroenterology, PAGNI University Hospital, University of Crete Medical School, 71500 Heraklion, Greece
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Ioannis Tsomidis
- Laboratory of Gastroenterology and Hepatology, University of Crete Medical School, 71500 Heraklion, Greece;
| | - Argyro Voumvouraki
- 1st Department of Internal Medicine, AHEPA University Hospital, 54621 Thessaloniki, Greece;
| |
Collapse
|
6
|
De Vincentis S, Evangelisti S, Rossi B, Decaroli MC, Locaso M, Ansaloni A, Ferrara F, Corradini E, Pietrangelo A, Rochira V. Hypoparathyroidism in adults with iron overload diseases (IOD): evidence of a subclinical phenotype. Endocrine 2025; 87:1257-1266. [PMID: 39630327 DOI: 10.1007/s12020-024-04124-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 11/28/2024] [Indexed: 01/12/2025]
Abstract
PURPOSE To explore the prevalence of hypoparathyroidism (HPT), overt and subclinical, in a cohort of adults with Iron Overload Diseases (IOD). Secondary aim was to test the calcium (Ca)-to-phosphorus (P) ratio performance in identifying HPT. METHODS Single-center, prospective, case-control study. Sixty-five IOD, 40 with thalassemia major/intermedia (TMI) and 25 with hemochromatosis (HC), and 76 age-matched controls were included. Main outcomes (serum Ca, P, Ca/P ratio, intact parathyroid hormone (PTH), albumin) defined overt and subclinical HPT. RESULTS Albumin-adjusted Ca was lower (p = 0.004) and P higher (p = 0.002) comparing IOD to controls. Ca/P ratio was lower in IOD than controls (p < 0.001); PTH did not change. P was higher and Ca/P lower comparing TMI to HC and controls (p < 0.001); Ca did not change. A total of 28/65 IOD (43%) had HPT (9.2% overt, 33.8% subclinical) whose prevalence was higher in TMI than HC (p < 0.001). Ca/P ratio <2.32 had sensitivity 71.4% and specificity 83.9% in detecting overt/subclinical HPT. IOD with Ca/P ratio <2.32 (1.78 in SI) had an almost 12-fold increased likelihood to be affected by HPT (OR 12.92 [3.90-42.82]; p < 0.001). Ca/P (p = 0.002) was the only independent risk factor for HPT at multivariate analysis. CONCLUSIONS HPT, especially non-classical subclinical HPT, is common in adult IOD with higher prevalence in TMI than HC. Ca/P ratio <2.32 is accurate to screen for overt/subclinical HPT and should be periodically evaluated in IOD to early detect an unbalanced mineral metabolism, and to monitor a possible evolution from subclinical to overt HPT.
Collapse
Affiliation(s)
- Sara De Vincentis
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy.
| | - Selene Evangelisti
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria Policlinico di Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - Barbara Rossi
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria Policlinico di Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - Maria Chiara Decaroli
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Michela Locaso
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anna Ansaloni
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria Policlinico di Modena, Ospedale Civile di Baggiovara, Modena, Italy
| | - Francesca Ferrara
- Internal Medicine Unit and Centre for Hereditary Anemias, ERN-EuroBloodNet Center for Iron Disorders, Azienda Ospedaliero-Universitaria di Modena-Policlinico, Modena, Italy
| | - Elena Corradini
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, ERN-EuroBloodNet Center for Iron Disorders and ERN Rare-Liver for Hepatological Diseases, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Antonello Pietrangelo
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
- Internal Medicine and Centre for Hemochromatosis and Hereditary Liver Diseases, ERN-EuroBloodNet Center for Iron Disorders and ERN Rare-Liver for Hepatological Diseases, Azienda Ospedaliero-Universitaria Policlinico di Modena, Modena, Italy
| | - Vincenzo Rochira
- Endocrinology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- Unit of Endocrinology, Department of Medical Specialties, Azienda Ospedaliero-Universitaria Policlinico di Modena, Ospedale Civile di Baggiovara, Modena, Italy
| |
Collapse
|
7
|
Zhang JW, Zhang N, Lyu Y, Zhang XF. Influence of Sex in the Development of Liver Diseases. Semin Liver Dis 2025; 45:15-32. [PMID: 39809453 DOI: 10.1055/a-2516-0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The liver is a sexually dimorphic organ. Sex differences in prevalence, progression, prognosis, and treatment prevail in most liver diseases, and the mechanism of how liver diseases act differently among male versus female patients has not been fully elucidated. Biological sex differences in normal physiology and disease arise principally from sex hormones and/or sex chromosomes. Sex hormones contribute to the development and progression of most liver diseases, with estrogen- and androgen-mediated signaling pathways mechanistically involved. In addition, genetic factors in sex chromosomes have recently been found to contribute to the sex disparity of many liver diseases, which might explain, to some extent, the difference in gene expression pattern, immune response, and xenobiotic metabolism between men and women. Although increasing evidence suggests that sex is one of the most important modulators of disease prevalence and outcomes, at present, basic and clinical studies have long been sex unbalanced, with female subjects underestimated. As such, this review focuses on sex disparities of liver diseases and summarizes the current understanding of sex-specific mechanisms, including sex hormones, sex chromosomes, etc. We anticipate that understanding sex-specific pathogenesis will aid in promoting personalized therapies for liver disease among male versus female patients.
Collapse
Affiliation(s)
- Jie-Wen Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Nan Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Yi Lyu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| | - Xu-Feng Zhang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- Institute of Advanced Surgical Technology and Engineering, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
- National-Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi Province, People's Republic of China
| |
Collapse
|
8
|
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 2025; 130:179-189. [PMID: 39578337 PMCID: PMC11870931 DOI: 10.1007/s11547-024-01930-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [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
|
9
|
Barton JC, Barton JC, Acton RT. Factors related to mean corpuscular volume in HFE p.C282Y homozygotes. EJHAEM 2025; 6:e1063. [PMID: 39866924 PMCID: PMC11756985 DOI: 10.1002/jha2.1063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 11/15/2024] [Accepted: 11/22/2024] [Indexed: 01/28/2025]
Abstract
Introduction The aim of this study was to define the relationships between factors other than transferrin saturation (TS) to mean corpuscular volume (MCV) and macrocytosis (MCV > 100 fL) in HFE p.C282Y (rs1800562) homozygotes. Methods We studied white post-screening participants with p.C282Y homozygosity who did not have anemia, report cirrhosis or pregnancy, or use medications that increase MCV. We analyzed relations of MCV and macrocytosis with age, sex, diabetes reports, daily alcohol consumption, swollen or tender 2nd/3rd metacarpophalangeal (MCP) joints, TS, and serum ferritin (SF). Results There were 257 participants (110 men and 147 women). Median alcohol consumption, median TS, median SF, and macrocytosis prevalence were significantly greater in men. Relative risk of macrocytosis in men was 2.81. In men and women, there were significant positive Pearson's correlations of MCV versus age and Spearman's correlations of MCV versus alcohol consumption and TS. Mean MCV and macrocytosis prevalence were significantly greater in participants with than without swollen or tender 2nd/3rd MCP joints. Linear regression on MCV revealed positive associations: age (p < 0.0001), alcohol consumption (p = 0.0007), and TS (p < 0.0001). Logistic regression on macrocytosis revealed an odds ratio for age of 1.04 (95% confidence interval: 1.00, 1.07). Conclusions MCV in HFE p.C282Y homozygotes is positively related to age, daily alcohol consumption, TS, and swollen or tender 2nd/3rd MCP joints.
Collapse
Affiliation(s)
- James C. Barton
- Department of MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Southern Iron Disorders CenterBirminghamAlabamaUSA
| | | | - Ronald T. Acton
- Southern Iron Disorders CenterBirminghamAlabamaUSA
- Department of MicrobiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| |
Collapse
|
10
|
Palmer WC, Stancampiano FF. Hemochromatosis. Ann Intern Med 2025; 178:ITC17-ITC32. [PMID: 39928951 DOI: 10.7326/annals-24-03710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/12/2025] Open
Abstract
Hemochromatosis is an inheritable condition that mainly affects White populations of European descent. Most patients remain asymptomatic, but others develop advanced organ damage that reduces quality of life and long-term survival. Arthropathy, diabetes mellitus, cirrhosis, hypogonadotropic hypogonadism, and cardiomyopathy are key clinical manifestations. Primary care and hospital medicine physicians play an essential role in early identification of this disease, which can be accomplished via standard hematologic testing. Early diagnosis and therapeutic phlebotomy improve clinical outcomes.
Collapse
Affiliation(s)
- William C Palmer
- Professor of Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic Florida, Jacksonville, Florida (W.C.P.)
| | - Fernando F Stancampiano
- Associate Professor of Medicine, Division of Community Internal Medicine, Mayo Clinic Florida, Jacksonville, Florida (F.F.S.)
| |
Collapse
|
11
|
Cristancho LCQ, Urbano MA, Nati-Castillo HA, Obando MA, Gómez-Gutiérrez R, Izquierdo-Condoy JS. A decade of iron overload disorders and hemochromatosis: clinical and genetic findings from a specialized center in Colombia. Front Med (Lausanne) 2024; 11:1494527. [PMID: 39720661 PMCID: PMC11666385 DOI: 10.3389/fmed.2024.1494527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Background Iron overload disorders, including hereditary hemochromatosis (HH), are characterized by excessive iron accumulation, which can cause severe organ damage. HH is most associated with the C282Y mutation in Caucasian populations, but its prevalence and genetic profiles in Latin American populations remain underexplored. Objectives To describe the clinical manifestations, genetic profiles, and biochemical characteristics of patients with suspected iron overload disorders in a specialized hematology center in Cali, Colombia. Methods A retrospective observational study was conducted on 70 patients diagnosed with iron overload disorders between 2014 and 2024. Data on clinical presentation, laboratory results, imaging, and genetic mutations were collected. Statistical analyses, including chi-square tests and logistic regression, were used to evaluate factors associated with HH diagnosis. Results Male patients constituted 64.3% of the sample, with a mean age of 56.1 years at diagnosis. Fatigue (27.1%) and joint pain (17.1%) were the most common symptoms. Of the total sample, 32.9% were diagnosed with hemochromatosis. The H63D mutation was the most prevalent (52.2%), while the C282Y mutation was rare. A predominance of both slight (100.0%) and limitrophe (58.3%) iron overload was identified among patients with hemochromatosis (p = 0.036). Conclusion Colombian patients with iron overload disorders show clinical, epidemiological, and biochemical profiles consistent with global patterns, yet exhibit distinct genetic diversity. Notably, they have a low prevalence of the C282Y mutation and a higher prevalence of the H63D mutation, differing from European HH profiles. Despite elevated ferritin and transferrin saturation, no significant clinical symptoms were observed, suggesting potential delays in diagnosis. These findings highlight the need for early, region-specific diagnostic approaches to prevent complications like cirrhosis and underscore the importance of further genetic research across Latin America.
Collapse
Affiliation(s)
- L. C. Quiroga Cristancho
- Department of Internal Medicine, Interinstitutional Internal Medicine Group (GIMI 1), Universidad Libre, Cali, Colombia
| | - María Alejandra Urbano
- Department of Internal Medicine, Interinstitutional Internal Medicine Group (GIMI 1), Universidad Libre, Cali, Colombia
| | - H. A. Nati-Castillo
- Department of Internal Medicine, Interinstitutional Internal Medicine Group (GIMI 1), Universidad Libre, Cali, Colombia
| | | | - Rigoberto Gómez-Gutiérrez
- Department of Internal Medicine, Interinstitutional Internal Medicine Group (GIMI 1), Universidad Libre, Cali, Colombia
- Departamento de Hematología, Hemato Oncólogos S.A, Cali, Colombia
| | | |
Collapse
|
12
|
Mottelson M, Helby J, Nordestgaard BG, Ellervik C, Mandrup-Poulsen T, Petersen J, Bojesen SE, Glenthøj A. Mortality and risk of diabetes, liver disease, and heart disease in individuals with haemochromatosis HFE C282Y homozygosity and normal concentrations of iron, transferrin saturation, or ferritin: prospective cohort study. BMJ 2024; 387:e079147. [PMID: 39653412 PMCID: PMC11626441 DOI: 10.1136/bmj-2023-079147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/16/2024] [Indexed: 12/13/2024]
Abstract
OBJECTIVES To test whether haemochromatosis HFE C282Y homozygotes have increased risk of diabetes, liver disease, and heart disease even when they have normal plasma iron, transferrin saturation, or ferritin concentrations and to test whether C282Y homozygotes with diabetes, liver disease, or heart disease have increased mortality compared with non-carriers with these diseases. DESIGN Prospective cohort study. SETTING Three Danish general population cohorts: the Copenhagen City Heart Study, the Copenhagen General Population Study, and the Danish General Suburban Population Study. PARTICIPANTS 132 542 individuals genotyped for the HFE C282Y and H63D variants, 422 of whom were C282Y homozygotes, followed prospectively for up to 27 years after study enrolment. MAIN OUTCOME MEASURE Hospital contacts and deaths, retrieved from national registers, covering all hospitals and deaths in Denmark. RESULTS Comparing C282Y homozygotes with non-carriers, hazard ratios were 1.72 (95% confidence interval (CI) 1.24 to 2.39) for diabetes, 2.22 (1.40 to 3.54) for liver disease, and 1.01 (0.78 to 1.31) for heart disease. Depending on age group, the absolute five year risk of diabetes was 0.54-4.3% in C282Y homozygous women, 0.37-3.0% in non-carrier women, 0.86-6.8% in C282Y homozygous men, and 0.60-4.80% in non-carrier men. When studied according to levels of iron, transferrin saturation, and ferritin in a single blood sample obtained at study enrolment, risk of diabetes was increased in C282Y homozygotes with normal transferrin saturation (hazard ratio 2.00, 95% CI 1.04 to 3.84) or ferritin (3.76, 1.41 to 10.05) and in C282Y homozygotes with normal levels of both ferritin and transferrin saturation (6.49, 2.09 to 20.18). C282Y homozygotes with diabetes had a higher risk of death from any cause than did non-carriers with diabetes (hazard ratio 1.94, 95% CI 1.19 to 3.18), but mortality was not increased in C282Y homozygotes without diabetes. The percentage of all deaths among C282Y homozygotes that could theoretically be prevented if excess deaths in individuals with a specific disease were eliminated (the population attributable fraction) was 27.3% (95% CI 12.4% to 39.7%) for diabetes and 14.4% (3.1% to 24.3%) for liver disease. Risk of diabetes or liver disease was not increased in H63D heterozygotes, H63D homozygotes, C282Y heterozygotes, or C282Y/H63D compound heterozygotes. CONCLUSIONS Haemochromatosis C282Y homozygotes with normal transferrin saturation and/or ferritin, not recommended for HFE genotyping according to most guidelines, had increased risk of diabetes. Furthermore, C282Y homozygotes with diabetes had higher mortality than non-carriers with diabetes, and 27.3% of all deaths among C282Y homozygotes were potentially attributable to diabetes. These results indicate that prioritising detection and treatment of diabetes in C282Y homozygotes may be relevant.
Collapse
Affiliation(s)
- Mathis Mottelson
- Danish Red Blood Cell Center, Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Jens Helby
- Danish Red Blood Cell Center, Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Børge Grønne Nordestgaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Christina Ellervik
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Thomas Mandrup-Poulsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jesper Petersen
- Danish Red Blood Cell Center, Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Stig Egil Bojesen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Andreas Glenthøj
- Danish Red Blood Cell Center, Department of Hematology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
13
|
Singh P, Millson C, Driver R. Hereditary haemochromatosis: A review. J R Coll Physicians Edinb 2024; 54:340-347. [PMID: 39523455 DOI: 10.1177/14782715241298724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Hereditary haemochromatosis (HH) is the commonest genetic condition among populations of Northern European ancestry. Mutations to the HFE gene leads to uninhibited intestinal iron absorption followed by excess iron deposition in various organs such as the liver, pituitary gland, pancreas and heart. Due to variable biochemical and clinical penetrance, not all individuals with C282Y homozygosity will develop HH. Early diagnosis is crucial to prevent morbidity and mortality but is challenging with most patients not exhibiting any symptoms. Patients with HH should undergo clinical assessment to evaluate their symptoms, presence of organ damage and hepatic fibrosis using transient elastography. Patients who are negative for the HFE mutations but have significant liver iron loading seen on magnetic resonance imaging should be reviewed by a specialist and considered for genetic tests looking for the rarer non-HFE mutations. HH patients are predominantly treated with venesection which can improve symptoms, hepatic fibrosis and mortality.
Collapse
Affiliation(s)
- Prabhsimran Singh
- Hepatology Department, York and Scarborough Teaching Hospitals NHS Foundation Trust
- Hull York Medical School, UK
| | - Charles Millson
- Hepatology Department, York and Scarborough Teaching Hospitals NHS Foundation Trust
| | - Robert Driver
- Hepatology Department, York and Scarborough Teaching Hospitals NHS Foundation Trust
- Hull York Medical School, UK
| |
Collapse
|
14
|
Uguen K, Le Tertre M, Tchernitchko D, Elbahnsi A, Maestri S, Gourlaouen I, Férec C, Ka C, Callebaut I, Le Gac G. The dual loss and gain of function of the FPN1 iron exporter results in the ferroportin disease phenotype. HGG ADVANCES 2024; 5:100335. [PMID: 39039793 PMCID: PMC11343060 DOI: 10.1016/j.xhgg.2024.100335] [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: 03/13/2024] [Revised: 07/16/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024] Open
Abstract
Heterozygous mutations in SLC40A1, encoding a multi-pass membrane protein of the major facilitator superfamily known as ferroportin 1 (FPN1), are responsible for two distinct hereditary iron-overload diseases: ferroportin disease, which is associated with reduced FPN1 activity (i.e., decrease in cellular iron export), and SLC40A1-related hemochromatosis, which is associated with abnormally high FPN1 activity (i.e., resistance to hepcidin). Here, we report three SLC40A1 missense variants with opposite functional consequences. In cultured cells, the p.Arg40Gln and p.Ser47Phe substitutions partially reduced the ability of FPN1 to export iron and also partially reduced its sensitivity to hepcidin. The p.Ala350Val substitution had more profound effects, resulting in low FPN1 iron egress and weak FPN1/hepcidin interaction. Structural analyses helped to differentiate the first two substitutions, which are predicted to cause local instabilities, and the third, which is thought to prevent critical rigid-body movements that are essential to the iron transport cycle. The phenotypic traits observed in a total of 12 affected individuals are highly suggestive of ferroportin disease. Our findings dismantle the classical dualism of FPN1 loss versus gain of function, highlight some specific and unexpected functions of FPN1 transmembrane helices in the molecular mechanism of iron export and its regulation by hepcidin, and extend the spectrum of rare genetic variants that may cause ferroportin disease.
Collapse
Affiliation(s)
- Kevin Uguen
- University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France; CHU de Brest, 29200 Brest, France
| | - Marlène Le Tertre
- University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France; CHU de Brest, 29200 Brest, France
| | - Dimitri Tchernitchko
- CHU Paris Nord-Val de Seine - Hôpital Xavier Bichat-Claude Bernard, 75018 Paris, France; Centre de Recherche sur l'Inflammation, Inserm U1149, 75018 Paris, France
| | - Ahmad Elbahnsi
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France
| | - Sandrine Maestri
- University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France
| | | | - Claude Férec
- University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France; CHU de Brest, 29200 Brest, France; Association Gaétan Saleün, 29200 Brest, France
| | - Chandran Ka
- University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France; CHU de Brest, 29200 Brest, France; Laboratory of Excellence GR-Ex, 75015 Paris, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, 75005 Paris, France
| | - Gérald Le Gac
- University Brest, Inserm, EFS, UMR 1078, GGB, 29200 Brest, France; CHU de Brest, 29200 Brest, France; Laboratory of Excellence GR-Ex, 75015 Paris, France.
| |
Collapse
|
15
|
Horeau M, Delalande M, Ropert M, Leroyer P, Martin B, Orfila L, Loréal O, Derbré F. Sex similarities and divergences in systemic and muscle iron metabolism adaptations to extreme physical inactivity in rats. J Cachexia Sarcopenia Muscle 2024; 15:1989-1998. [PMID: 39049183 PMCID: PMC11446688 DOI: 10.1002/jcsm.13547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/15/2024] [Accepted: 06/26/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Previous data in humans suggest that extreme physical inactivity (EPI) affects iron metabolism differently between sexes. Our objective was to deepen the underlying mechanisms by studying rats of both sexes exposed to hindlimb unloading (HU), the reference experimental model mimicking EPI. METHODS Eight-week-old male and female Wistar rats were assigned to control (CTL) or hindlimb unloading (HU) conditions (n = 12/group). After 7 days of HU, serum, liver, spleen, and soleus muscle were removed. Iron parameters were measured in serum samples, and ICP-MS was used to quantify iron in tissues. Iron metabolism genes and proteins were analysed by RT-qPCR and Western blot. RESULTS Compared with control males, control females exhibited higher iron concentrations in serum (+43.3%, p < 0.001), liver (LIC; +198%, P < 0.001), spleen (SIC; +76.1%, P < 0.001), and transferrin saturation (TS) in serum (+53.3%, P < 0.001), contrasting with previous observations in humans. HU rat males, but not females, exhibited an increase of LIC (+54% P < 0.001) and SIC (+30.1%, P = 0.023), along with a rise of H-ferritin protein levels (+60.9% and +134%, respectively, in liver and spleen; P < 0.05) and a decrease of TFRC protein levels (-36%; -50%, respectively, P < 0.05). HU males also exhibited an increase of splenic HO-1 and NRF2 mRNA levels, (p < 0.001), as well as HU females (P < 0.001). Concomitantly to muscle atrophy observed in HU animals, the iron concentration increased in soleus in females (+26.7, P = 0.004) while only a trend is observed in males (+17.5%, P = 0.088). In addition, the H-ferritin and myoglobin protein levels in soleus were increased in males (+748%, P < 0.001, +22%, P = 0.011, respectively) and in females (+369%, P < 0.001, +21.9%, P = 0.007, respectively), whereas TFRC and ferroportin (FPN) protein levels were reduced in males (-68.9%, P < 0.001, -76.8%, P < 0.001, respectively) and females (-75.9%, P < 0.001, -62.9%, P < 0.001, respectively). Interestingly, in both sexes, heme exporter FLVCR1 mRNA increased in soleus, while protein levels decreased (-39.9% for males P = 0.010 and -49.1% for females P < 0.001). CONCLUSIONS Taken together, these data support that, in rats (1) extreme physical inactivity differently impacts the distribution of iron in both sexes, (2) splenic erythrophagocytosis could play a role in this iron misdistribution. The higher iron concentrations in atrophied soleus from both sexes are associated with a decoupling between the increase in iron storage proteins (i.e., ferritin and myoglobin) and the decrease in levels of iron export proteins (i.e., FPN and FLVCR1), thus supporting an iron sequestration in skeletal muscle under extreme physical inactivity.
Collapse
Affiliation(s)
- Mathieu Horeau
- Laboratory “Movement Sport and Health Sciences” EA7470University of Rennes/ENSRennesFrance
- INSERM, University of Rennes, INRAE, UMR 1317Nutrition Metabolisms and Cancer (NuMeCan) InstituteRennesFrance
| | - Melissa Delalande
- Laboratory “Movement Sport and Health Sciences” EA7470University of Rennes/ENSRennesFrance
| | - Martine Ropert
- INSERM, University of Rennes, INRAE, UMR 1317Nutrition Metabolisms and Cancer (NuMeCan) InstituteRennesFrance
- Elemental Analysis and Metabolism of Metals (AEM2) PlatformUniv Rennes CHU PontchaillouRennesFrance
| | - Patricia Leroyer
- INSERM, University of Rennes, INRAE, UMR 1317Nutrition Metabolisms and Cancer (NuMeCan) InstituteRennesFrance
| | - Brice Martin
- Laboratory “Movement Sport and Health Sciences” EA7470University of Rennes/ENSRennesFrance
| | - Luz Orfila
- Laboratory “Movement Sport and Health Sciences” EA7470University of Rennes/ENSRennesFrance
| | - Olivier Loréal
- INSERM, University of Rennes, INRAE, UMR 1317Nutrition Metabolisms and Cancer (NuMeCan) InstituteRennesFrance
- Elemental Analysis and Metabolism of Metals (AEM2) PlatformUniv Rennes CHU PontchaillouRennesFrance
| | - Frédéric Derbré
- Laboratory “Movement Sport and Health Sciences” EA7470University of Rennes/ENSRennesFrance
| |
Collapse
|
16
|
Świątczak M, Raczak A, Świątczak A, Młodziński K, Sikorska K, Jaźwińska A, Kaufmann D, Daniłowicz-Szymanowicz L. Fatigue Assessment in Patients with Hereditary Hemochromatosis: First Use of the Popular Diagnostic Tools. J Clin Med 2024; 13:5544. [PMID: 39337031 PMCID: PMC11432497 DOI: 10.3390/jcm13185544] [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: 08/19/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Background: Hereditary hemochromatosis (HH) is a genetic condition with fatigue as an essential but not precisely assessed symptom. While some well-specified scales for fatigue assessment in some pathologies exist, data on their usefulness in HH need to be collected. This research aimed to evaluate fatigue in HH using the Fatigue Assessment Scale (FAS), Fatigue Severity Scale (FSS), and Chalder Fatigue Scale (CFQ). Methodology: Seventy-nine HH patients underwent a questionnaire containing items about detailed medical history and the FAS, FSS, and CFQ scales. Twenty-five sex- and age-matched healthy persons constituted the control group (controls); additionally, thirty blood donors (donors) were compared. Results: The fatigue indices were significantly worse in the HH patients than in the controls and donors (HH vs. controls p-value: FAS = 0.003, FSS < 0.001, and CFQ = 0.003; HH vs. donors p-value: FAS = 0.025, FSS < 0.001, and CFQ = 0.041). There were no differences between the severity of fatigue and the specific genotype or the age of the patients. The HH women presented more severe fatigue than the men. High internal consistency and reliability for each scale were revealed: the Cronbach alpha values were as follows: FAS 0.92, FSS 0.95, and CFQ 0.93. Additionally, the construct validity and factorial validity of the implemented scales were confirmed. Conclusions: The HH patients exhibited significantly worse fatigue across all the scales. The FAS, FSS, and CFQ are simple and reliable diagnostic tools for assessing and quantifying fatigue for clinical and research purposes.
Collapse
Affiliation(s)
- Michał Świątczak
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland
| | - Alicja Raczak
- Clinical Psychology Department, Faculty of Health Sciences, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Agata Świątczak
- Department of Pediatrics, Hematology and Oncology, Faculty of Medicine, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Krzysztof Młodziński
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland
| | - Katarzyna Sikorska
- Department of Tropical Medicine and Epidemiology, Faculty of Health Sciences, Medical University of Gdańsk, 80-214 Gdańsk, Poland
| | - Anna Jaźwińska
- Regional Blood Donation and Hemotherapy Center, 80-210 Gdańsk, Poland
| | - Damian Kaufmann
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland
| | - Ludmiła Daniłowicz-Szymanowicz
- Department of Cardiology and Electrotherapy, Faculty of Medicine, Medical University of Gdańsk, Smoluchowskiego 17, 80-214 Gdańsk, Poland
| |
Collapse
|
17
|
Mottelson M, Glenthøj A, Nordestgaard BG, Ellervik C, Petersen J, Bojesen SE, Helby J. Iron, hemochromatosis genotypes, and risk of infections: a cohort study of 142 188 general population individuals. Blood 2024; 144:693-707. [PMID: 38728387 DOI: 10.1182/blood.2023022235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 04/08/2024] [Accepted: 05/01/2024] [Indexed: 05/12/2024] Open
Abstract
ABSTRACT It is unclear whether risk of infection is increased in individuals with hereditary hemochromatosis and in individuals with low or high plasma iron, transferrin saturation, or ferritin. Therefore, we tested whether high and low iron, transferrin saturation, and ferritin are associated with risk of infections observationally and genetically through HFE genotypes. We studied 142 188 Danish general population individuals. Iron, transferrin saturation, and ferritin were measured in 136 656, 136 599, and 38 020 individuals, respectively. HFE was genotyped for C282Y and H63D in 132 542 individuals. Median follow-up after study enrollment was 8 years (range, 0-38) for hospital and emergency room admissions with infections (n = 20 394) using the National Patient Register, covering all Danish hospitals. Hazard ratios for any infection were 1.20 (95% confidence interval [CI], 1.12-1.28) and 1.14 (95% CI, 1.07-1.22) in individuals with plasma iron ≤5th or ≥95th percentile compared with individuals with iron from 26th to 74th percentiles. Findings for transferrin saturation were similar, whereas infection risk was not increased in individuals with ferritin ≤5th or ≥95th percentile. Hazard ratios in C282Y homozygotes vs noncarriers were 1.40 (95% CI, 1.16-1.68) for any infection, 1.69 (95% CI, 1.05-2.73) for sepsis, and 2.34 (95% CI, 1.41-3.90) for death from infectious disease. Risk of infection was increased in C282Y homozygotes with normal plasma iron, transferrin saturation, or ferritin, and in C282Y homozygotes without liver disease, diabetes, and/or heart failure. In summary, low and high plasma iron and transferrin saturation were independently associated with increased infection risk. C282Y homozygotes had increased risk of any infection, sepsis, and death from infections. Even C282Y homozygotes with normal iron, transferrin saturation, or ferritin, not currently recommended for genotyping, had increased infection risk.
Collapse
Affiliation(s)
- Mathis Mottelson
- Department of Haematology, Danish Red Blood Cell Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Andreas Glenthøj
- Department of Haematology, Danish Red Blood Cell Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Børge Grønne Nordestgaard
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Christina Ellervik
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- Department of Production, Research, and Innovation, Region Zealand, Sorø, Denmark
- Department of Laboratory Medicine, Boston Children's Hospital, Boston, MA
- Department of Pathology, Harvard Medical School, Boston, MA
| | - Jesper Petersen
- Department of Haematology, Danish Red Blood Cell Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
| | - Stig Egil Bojesen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Biochemistry, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
- The Copenhagen City Heart Study, Copenhagen University Hospital - Bispebjerg and Frederiksberg, Copenhagen, Denmark
| | - Jens Helby
- Department of Haematology, Danish Red Blood Cell Centre, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| |
Collapse
|
18
|
Hrdina A, Serra Canales M, Arias-Rojas A, Frahm D, Iatsenko I. The endosymbiont Spiroplasma poulsonii increases Drosophila melanogaster resistance to pathogens by enhancing iron sequestration and melanization. mBio 2024; 15:e0093624. [PMID: 38940615 PMCID: PMC11323552 DOI: 10.1128/mbio.00936-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/28/2024] [Indexed: 06/29/2024] Open
Abstract
Facultative endosymbiotic bacteria, such as Wolbachia and Spiroplasma species, are commonly found in association with insects and can dramatically alter their host physiology. Many endosymbionts are defensive and protect their hosts against parasites or pathogens. Despite the widespread nature of defensive insect symbioses and their importance for the ecology and evolution of insects, the mechanisms of symbiont-mediated host protection remain poorly characterized. Here, we utilized the fruit fly Drosophila melanogaster and its facultative endosymbiont Spiroplasma poulsonii to characterize the mechanisms underlying symbiont-mediated host protection against bacterial and fungal pathogens. Our results indicate a variable effect of S. poulsonii on infection outcome, with endosymbiont-harboring flies being more resistant to Rhyzopus oryzae, Staphylococcus aureus, and Providencia alcalifaciens but more sensitive or as sensitive as endosymbiont-free flies to the infections with Pseudomonas species. Further focusing on the protective effect, we identified Transferrin-mediated iron sequestration induced by Spiroplasma as being crucial for the defense against R. oryzae and P. alcalifaciens. In the case of S. aureus, enhanced melanization in Spiroplasma-harboring flies plays a major role in protection. Both iron sequestration and melanization induced by Spiroplasma require the host immune sensor protease Persephone, suggesting a role of proteases secreted by the symbiont in the activation of host defense reactions. Hence, our work reveals a broader defensive range of Spiroplasma than previously appreciated and adds nutritional immunity and melanization to the defensive arsenal of symbionts. IMPORTANCE Defensive endosymbiotic bacteria conferring protection to their hosts against parasites and pathogens are widespread in insect populations. However, the mechanisms by which most symbionts confer protection are not fully understood. Here, we studied the mechanisms of protection against bacterial and fungal pathogens mediated by the Drosophila melanogaster endosymbiont Spiroplasma poulsonii. We demonstrate that besides the previously described protection against wasps and nematodes, Spiroplasma also confers increased resistance to pathogenic bacteria and fungi. We identified Spiroplasma-induced iron sequestration and melanization as key defense mechanisms. Our work broadens the known defense spectrum of Spiroplasma and reveals a previously unappreciated role of melanization and iron sequestration in endosymbiont-mediated host protection. We propose that the mechanisms we have identified here may be of broader significance and could apply to other endosymbionts, particularly to Wolbachia, and potentially explain their protective properties.
Collapse
Affiliation(s)
- Alexandra Hrdina
- Research group Genetics of Host-Microbe Interactions, Max Planck Institute for Infection Biology, Berlin, Germany
- Faculty of Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marina Serra Canales
- Research group Genetics of Host-Microbe Interactions, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Aranzazu Arias-Rojas
- Research group Genetics of Host-Microbe Interactions, Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Biology, Chemistry, and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Dagmar Frahm
- Research group Genetics of Host-Microbe Interactions, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Igor Iatsenko
- Research group Genetics of Host-Microbe Interactions, Max Planck Institute for Infection Biology, Berlin, Germany
| |
Collapse
|
19
|
Delatycki MB, Allen KJ. Population Screening for Hereditary Haemochromatosis-Should It Be Carried Out, and If So, How? Genes (Basel) 2024; 15:967. [PMID: 39202328 PMCID: PMC11353936 DOI: 10.3390/genes15080967] [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: 04/28/2024] [Revised: 06/22/2024] [Accepted: 07/22/2024] [Indexed: 09/03/2024] Open
Abstract
The Human Genome Project, completed in 2003, heralded a new era in precision medicine. Somewhat tempering the excitement of the elucidation of the human genome is the emerging recognition that there are fewer single gene disorders than first anticipated, with most diseases predicted to be polygenic or at least gene-environment modified. Hereditary haemochromatosis (HH) is an inherited iron overload disorder, for which the vast majority of affected individuals (>90%) have homozygosity for a single pathogenic variant in the HFE gene, resulting in p.Cys282Tyr. Further, there is significant benefit to an individual in identifying the genetic risk of HH, since the condition evolves over decades, and the opportunity to intervene and prevent disease is both simple and highly effective through regular venesection. Add to that the immediate benefit to society of an increased pool of ready blood donors (blood obtained from HH venesections can generally be used for donation), and the case for population screening to identify those genetically at risk for HH becomes more cogent. Concerns about genetic discrimination, creating a cohort of "worried well", antipathy to acting on medical advice to undertake preventive venesection or simply not understanding the genetic risk of the condition adequately have all been allayed by a number of investigations. So why then has HH population genetic screening not been routinely implemented anywhere in the world? The answer is complex, but in this article we explore the pros and cons of screening for HH and the different views regarding whether it should be phenotypic (screening for iron overload by serum ferritin and/or transferrin saturation) or genotypic (testing for HFE p.Cys282Tyr). We argue that now is the time to give this poster child for population genetic screening the due consideration required to benefit the millions of individuals at risk of HFE-related iron overload.
Collapse
Affiliation(s)
- Martin B. Delatycki
- Victorian Clinical Genetics Services, Parkville, VIC 3052, Australia
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
| | - Katrina J. Allen
- Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia;
| |
Collapse
|
20
|
Song J, Xu S, Chen Q, Gou Y, Zhao C, Jia C, Liu H, Zhang Z, Li B, Gao Y, Zhao Y, Ji E. Cardiomyocyte-specific overexpression of FPN1 diminishes cardiac hypertrophy induced by chronic intermittent hypoxia. J Cell Mol Med 2024; 28:e18543. [PMID: 39054575 PMCID: PMC11272608 DOI: 10.1111/jcmm.18543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/26/2024] [Accepted: 07/10/2024] [Indexed: 07/27/2024] Open
Abstract
The significance of iron in myocardial mitochondria function cannot be underestimated, because deviations in iron levels within cardiomyocytes may have profound detrimental effects on cardiac function. In this study, we investigated the effects of ferroportin 1 (FPN1) on cardiac iron levels and pathological alterations in mice subjected to chronic intermittent hypoxia (CIH). The cTNT-FPN1 plasmid was administered via tail vein injection to induce the mouse with FPN1 overexpression in the cardiomyocytes. CIH was established by exposing the mice to cycles of 21%-5% FiO2 for 3 min, 8 h per day. Subsequently, the introduction of hepcidin resulted in a reduction in FPN1 expression, and H9C2 cells were used to establish an IH model to further elucidate the role of FPN1. First, FPN1 overexpression ameliorated CIH-induced cardiac dysfunction, myocardial hypertrophy, mitochondrial damage and apoptosis. Second, FPN1 overexpression attenuated ROS levels during CIH. In addition, FPN1 overexpression mitigated CIH-induced cardiac iron accumulation. Moreover, the administration of hepcidin resulted in a reduction in FPN1 levels, further accelerating the CIH-induced levels of ROS, LIP and apoptosis in H9C2 cells. These findings indicate that the overexpression of FPN1 in cardiomyocytes inhibits CIH-induced cardiac iron accumulation, subsequently reducing ROS levels and mitigating mitochondrial damage. Conversely, the administration of hepcidin suppressed FPN1 expression and worsened cardiomyocyte iron toxicity injury.
Collapse
Affiliation(s)
- Ji‐Xian Song
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Shan Xu
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
- Hebei Key Laboratory of Turbidity Toxin SyndromeThe First Affiliated Hospital Hebei University of Chinese MedicineShijiazhuangChina
| | - Qi Chen
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Yujing Gou
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Chen‐Bing Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Cui‐Ling Jia
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Han Liu
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Zhi Zhang
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Bo‐liang Li
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Yuhui Gao
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| | - Yashuo Zhao
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
- Hebei Key Laboratory of Turbidity Toxin SyndromeThe First Affiliated Hospital Hebei University of Chinese MedicineShijiazhuangChina
| | - En‐Sheng Ji
- Hebei Technology Innovation Center of TCM Combined Hydrogen MedicineHebei University of Chinese MedicineShijiazhuangChina
- Department of Physiology, Institute of Basic MedicineHebei University of Chinese MedicineShijiazhuangChina
| |
Collapse
|
21
|
Szczerbinska A, Kasztelan-Szczerbinska B, Rycyk-Bojarzynska A, Kocki J, Cichoz-Lach H. Hemochromatosis-How Not to Overlook and Properly Manage "Iron People"-A Review. J Clin Med 2024; 13:3660. [PMID: 38999226 PMCID: PMC11242024 DOI: 10.3390/jcm13133660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 06/20/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024] Open
Abstract
Hemochromatosis (HC) is the main genetic disorder of iron overload and is regarded as metal-related human toxicosis. HC may result from HFE and rare non-HFE gene mutations, causing hepcidin deficiency or, sporadically, hepcidin resistance. This review focuses on HFE-related HC. The illness presents a strong biochemical penetrance, but its prevalence is low. Unfortunately, the majority of patients with HC remain undiagnosed at their disease-curable stage. The main aim of HC management is to prevent iron overload in its early phase and remove excess iron from the body by phlebotomy in its late stage. Raising global awareness of HC among health staff, teaching them how not to overlook early HC manifestations, and paying attention to careful patient monitoring remain critical management strategies for preventing treatment delays, upgrading its efficacy, and improving patient prognosis.
Collapse
Affiliation(s)
- Agnieszka Szczerbinska
- Faculty of Medicine, Medical University of Warsaw, 61 Zwirki i Wigury Street, 02-091 Warsaw, Poland
| | - Beata Kasztelan-Szczerbinska
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, 8 Jaczewski Street, 20-954 Lublin, Poland
| | - Anna Rycyk-Bojarzynska
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, 8 Jaczewski Street, 20-954 Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Street, 20-080 Lublin, Poland;
| | - Halina Cichoz-Lach
- Department of Gastroenterology with Endoscopy Unit, Medical University of Lublin, 8 Jaczewski Street, 20-954 Lublin, Poland
| |
Collapse
|
22
|
Sun H, Huang T, Alam MM, Li J, Jang DW, Wang T, Chen H, Ho YP, Gao Z. Minimizing Contact Resistance and Flicker Noise in Micro Graphene Hall Sensors Using Persistent Carbene Modified Gold Electrodes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:31473-31479. [PMID: 38850243 PMCID: PMC11194764 DOI: 10.1021/acsami.4c05451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/10/2024]
Abstract
Scalable micro graphene Hall sensors (μGHSs) hold tremendous potential for highly sensitive and label-free biomagnetic sensing in physiological solutions. To enhance the performance of these devices, it is crucial to optimize frequency-dependent flicker noise to reduce the limit of detection (LOD), but it remains a great challenge due to the large contact resistance at the graphene-metal contact. Here we present a surface modification strategy employing persistent carbene on gold electrodes to reduce the contact resistivity by a factor of 25, greatly diminishing μGHS flicker noise by a factor of 1000 to 3.13 × 10-14 V2/Hz while simultaneously lowering the magnetic LOD SB1/2 to 1440 nT/Hz1/2 at 1 kHz under a 100 μA bias current. To the best of our knowledge, this represents the lowest SB1/2 reported for scalable μGHSs fabricated through wafer-scale photolithography. The reduction in contact noise is attributed to the π-π stacking interaction between the graphene and the benzene rings of persistent carbene, as well as the decrease in the work function of gold as confirmed by Kelvin Probe Force Microscopy. By incorporating a microcoil into the μGHS, we have demonstrated the real-time detection of superparamagnetic nanoparticles (SNPs), achieving a remarkable LOD of ∼528 μg/L. This advancement holds great potential for the label-free detection of magnetic biomarkers, e.g., ferritin, for the early diagnosis of diseases associated with iron overload, such as hereditary hemochromatosis (HHC).
Collapse
Affiliation(s)
- Honglin Sun
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
| | - Ting Huang
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
| | - Md Masruck Alam
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
| | - Jingwei Li
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
- Department
of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon 999077, Hong Kong SAR, China
| | - Dong Wook Jang
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
| | - Tianle Wang
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
| | - Haohan Chen
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
- School
of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi 214122, China
| | - Yi-Ping Ho
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
- Centre
for Novel Biomaterials, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR, China
- Hong
Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics, The Chinese University of Hong Kong, Shatin, New Territories 999077, Hong Kong SAR, China
- State
Key
Laboratory of Marine Pollution, City University
of Hong Kong, Kowloon Tong, Kowloon 999077, Hong Kong
SAR, China
| | - Zhaoli Gao
- Department
of Biomedical Engineering, The Chinese University
of Hong Kong, Shatin,
New Territories 999077, Hong Kong SAR,
China
- Shun
Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, New Territories 999077, Hong Kong SAR, China
- CUHK
Shenzhen Research Institute, Nanshan, Shenzhen 518172, China
| |
Collapse
|
23
|
Konkwo C, Chowdhury S, Vilarinho S. Genetics of liver disease in adults. Hepatol Commun 2024; 8:e0408. [PMID: 38551385 PMCID: PMC10984672 DOI: 10.1097/hc9.0000000000000408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/30/2024] [Indexed: 04/02/2024] Open
Abstract
Chronic liver disease stands as a significant global health problem with an estimated 2 million annual deaths across the globe. Combining the use of next-generation sequencing technologies with evolving knowledge in the interpretation of genetic variation across the human genome is propelling our understanding, diagnosis, and management of both rare and common liver diseases. Here, we review the contribution of risk and protective alleles to common forms of liver disease, the rising number of monogenic diseases affecting the liver, and the role of somatic genetic variants in the onset and progression of oncological and non-oncological liver diseases. The incorporation of genomic information in the diagnosis and management of patients with liver disease is driving the beginning of a new era of genomics-informed clinical hepatology practice, facilitating personalized medicine, and improving patient care.
Collapse
Affiliation(s)
- Chigoziri Konkwo
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
| | - Shanin Chowdhury
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Silvia Vilarinho
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
24
|
Lucas MR, Atkins JL, Pilling LC, Shearman JD, Melzer D. HFE genotypes, haemochromatosis diagnosis and clinical outcomes at age 80 years: a prospective cohort study in the UK Biobank. BMJ Open 2024; 14:e081926. [PMID: 38479735 PMCID: PMC10936495 DOI: 10.1136/bmjopen-2023-081926] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 02/06/2024] [Indexed: 12/31/2024] Open
Abstract
OBJECTIVES HFE haemochromatosis genetic variants have an uncertain clinical penetrance, especially to older ages and in undiagnosed groups. We estimated p.C282Y and p.H63D variant cumulative incidence of multiple clinical outcomes in a large community cohort. DESIGN Prospective cohort study. SETTING 22 assessment centres across England, Scotland, and Wales in the UK Biobank (2006-2010). PARTICIPANTS 451 270 participants genetically similar to the 1000 Genomes European reference population, with a mean of 13.3-year follow-up through hospital inpatient, cancer registries and death certificate data. MAIN OUTCOME MEASURES Cox proportional HRs of incident clinical outcomes and mortality in those with HFE p.C282Y/p.H63D mutations compared with those with no variants, stratified by sex and adjusted for age, assessment centre and genetic stratification. Cumulative incidences were estimated from age 40 years to 80 years. RESULTS 12.1% of p.C282Y+/+ males had baseline (mean age 57 years) haemochromatosis diagnoses, with a cumulative incidence of 56.4% at age 80 years. 33.1% died vs 25.4% without HFE variants (HR 1.29, 95% CI: 1.12 to 1.48, p=4.7×10-4); 27.9% vs 17.1% had joint replacements, 20.3% vs 8.3% had liver disease, and there were excess delirium, dementia, and Parkinson's disease but not depression. Associations, including excess mortality, were similar in the group undiagnosed with haemochromatosis. 3.4% of women with p.C282Y+/+ had baseline haemochromatosis diagnoses, with a cumulative incidence of 40.5% at age 80 years. There were excess incident liver disease (8.9% vs 6.8%; HR 1.62, 95% CI: 1.27 to 2.05, p=7.8×10-5), joint replacements and delirium, with similar results in the undiagnosed. p.C282Y/p.H63D and p.H63D+/+ men or women had no statistically significant excess fatigue or depression at baseline and no excess incident outcomes. CONCLUSIONS Male and female p.C282Y homozygotes experienced greater excess morbidity than previously documented, including those undiagnosed with haemochromatosis in the community. As haemochromatosis diagnosis rates were low at baseline despite treatment being considered effective, trials of screening to identify people with p.C282Y homozygosity early appear justified.
Collapse
Affiliation(s)
- Mitchell R Lucas
- Epidemiology and Public Health Group, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Janice L Atkins
- Epidemiology and Public Health Group, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Luke C Pilling
- Epidemiology and Public Health Group, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Jeremy D Shearman
- Department of Gastroenterology, South Warwickshire University NHS Foundation Trust, Warwick, UK
| | - David Melzer
- Epidemiology and Public Health Group, Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| |
Collapse
|
25
|
Zhang Z, Li J, Zuo C, Cheng C. Hemochromatosis Affects the Distribution of 68 Ga-DOTATATE in the Liver and Spleen. Clin Nucl Med 2024; 49:e137-e138. [PMID: 38271250 DOI: 10.1097/rlu.0000000000005034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
ABSTRACT Hemosiderosis is a chronic condition characterized by abnormal iron accumulation in tissues. In a PET scan of a 37-year-old woman, we observed an irregular distribution of 68 Ga-DOTATATE in the liver and spleen. Specifically, 68 Ga-DOTATATE appeared to be concentrated primarily in the peripheral regions of these organs, creating a distinctive "shell-like" appearance. This peculiar pattern may be attributed to the substantial accumulation of hemosiderin in the liver and spleen.
Collapse
Affiliation(s)
- Zeyu Zhang
- From the Department of Nuclear Medicine, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | | | | | | |
Collapse
|
26
|
Schaefer B, Pammer LM, Pfeifer B, Neururer S, Troppmair MR, Panzer M, Wagner S, Pertler E, Gieger C, Kronenberg F, Lamina C, Tilg H, Zoller H. Penetrance, cancer incidence and survival in HFE haemochromatosis-A population-based cohort study. Liver Int 2024; 44:838-847. [PMID: 38263707 DOI: 10.1111/liv.15797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND AND AIMS Haemochromatosis is characterized by progressive iron overload affecting the liver and can cause cirrhosis and hepatocellular carcinoma. Most haemochromatosis patients are homozygous for p.C282Y in HFE, but only a minority of individuals with this genotype will develop the disease. The aim was to assess the penetrance of iron overload, fibrosis, hepatocellular carcinoma and life expectancy. METHODS A total of 8839 individuals from the Austrian region of Tyrol were genotyped for the p.C282Y variant between 1997 and 2021. Demographic, laboratory parameters and causes of death were assessed from health records. Penetrance, survival, and cancer incidence were ascertained from diagnosed cases, insurance- and cancer registry data. Outcomes were compared with a propensity score-matched control population. RESULTS Median age at diagnosis in 542 p.C282Y homozygous individuals was 47.8 years (64% male). At genotyping, the prevalence of iron overload was 55%. The cumulative penetrance of haemochromatosis defined as the presence of provisional iron overload was 24.2% in males and 10.5% in females aged 60 years or younger. Among p.C282Y homozygotes of the same ages, the cumulative proportion of individuals without fibrosis (FIB-4 score < 1.3) was 92.8% in males and 96.7% in females. Median life expectancy was reduced by 6.8 years in individuals homozygous for p.C282Y when compared with population-matched controls (p = .001). Hepatocellular carcinoma incidence was not significantly higher in p.C282Y homozygotes than in controls matched for age and sex. CONCLUSION Reduced survival and the observed age-dependent increase in penetrance among p.C282Y homozygotes call for earlier diagnosis of haemochromatosis to prevent complications.
Collapse
Affiliation(s)
- Benedikt Schaefer
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Lorenz M Pammer
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Bernhard Pfeifer
- Division for Digital Medicine and Telehealth, UMIT TIROL-Private University for Health Sciences and Health Technology, Hall (Tyrol), Austria
- Tyrolean Federal Institute for Integrated Care, Tirol Kliniken Gmbh, Innsbruck, Austria
| | - Sabrina Neururer
- Division for Digital Medicine and Telehealth, UMIT TIROL-Private University for Health Sciences and Health Technology, Hall (Tyrol), Austria
- Tyrolean Federal Institute for Integrated Care, Tirol Kliniken Gmbh, Innsbruck, Austria
| | - Maria R Troppmair
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Marlene Panzer
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sonja Wagner
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Elke Pertler
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Christian Gieger
- Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health, Neuherberg, Germany
| | - Florian Kronenberg
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Claudia Lamina
- Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Herbert Tilg
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
| | - Heinz Zoller
- Department of Medicine I, Gastroenterology, Hepatology and Endocrinology, Medical University of Innsbruck, Innsbruck, Austria
- Christian Doppler Laboratory for Iron and Phosphate Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
27
|
Shenoy G, Slagle-Webb B, Khunsriraksakul C, Pandya Shesh B, Luo J, Khristov V, Smith N, Mansouri A, Zacharia BE, Holder S, Lathia JD, Barnholtz-Sloan JS, Connor JR. Analysis of anemia and iron supplementation among glioblastoma patients reveals sex-biased association between anemia and survival. Sci Rep 2024; 14:2389. [PMID: 38287054 PMCID: PMC10825121 DOI: 10.1038/s41598-024-52492-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 01/19/2024] [Indexed: 01/31/2024] Open
Abstract
The association between anemia and outcomes in glioblastoma patients is unclear. We analyzed data from 1346 histologically confirmed adult glioblastoma patients in the TriNetX Research Network. Median hemoglobin and hematocrit levels were quantified for 6 months following diagnosis and used to classify patients as anemic or non-anemic. Associations of anemia and iron supplementation of anemic patients with median overall survival (median-OS) were then studied. Among 1346 glioblastoma patients, 35.9% of male and 40.5% of female patients were classified as anemic using hemoglobin-based WHO guidelines. Among males, anemia was associated with reduced median-OS compared to matched non-anemic males using hemoglobin (HR 1.24; 95% CI 1.00-1.53) or hematocrit-based cutoffs (HR 1.28; 95% CI 1.03-1.59). Among females, anemia was not associated with median-OS using hemoglobin (HR 1.00; 95% CI 0.78-1.27) or hematocrit-based cutoffs (HR: 1.10; 95% CI 0.85-1.41). Iron supplementation of anemic females trended toward increased median-OS (HR 0.61; 95% CI 0.32-1.19) although failing to reach statistical significance whereas no significant association was found in anemic males (HR 0.85; 95% CI 0.41-1.75). Functional transferrin-binding assays confirmed sexually dimorphic binding in resected patient samples indicating underlying differences in iron biology. Anemia among glioblastoma patients exhibits a sex-specific association with survival.
Collapse
Affiliation(s)
- Ganesh Shenoy
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - Becky Slagle-Webb
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | | | | | - Jingqin Luo
- Division of Public Health Sciences, Department of Surgery and Siteman Cancer Center Biostatistics Shared Resource, Washington University School of Medicine, St. Louis, MO, USA
| | - Vladimir Khristov
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - Nataliya Smith
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - Alireza Mansouri
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - Brad E Zacharia
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
| | - Sheldon Holder
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, USA
| | - Justin D Lathia
- Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jill S Barnholtz-Sloan
- Trans-Divisional Research Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, Bethesda, MD, USA
| | - James R Connor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA.
| |
Collapse
|
28
|
Sozen S, Shah A. Alcohol Use Unmasking Heterozygous Hereditary Hemochromatosis. Cureus 2024; 16:e52364. [PMID: 38361672 PMCID: PMC10868447 DOI: 10.7759/cureus.52364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2024] [Indexed: 02/17/2024] Open
Abstract
Hereditary hemochromatosis (HH) is an autosomal recessive disorder characterized by excess iron absorption in the body following a mutation in the HFE gene. Though prolonged iron deposition has been shown to cause clinical symptoms such as hyperpigmentation, arthralgias, and liver damage, many individuals remain asymptomatic and exhibit no signs of iron overload. Here, we present a case where a 34-year-old with a history of severe alcohol use disorder presented with high iron, ferritin and transferrin saturation levels indicative of iron overload. Further testing for HFE gene mutations revealed simple heterozygote C282Y status, confirming the diagnosis of hereditary hemochromatosis. Simple heterozygotes, however, typically do not present with any symptoms of iron overload. This patient was counseled on lifestyle modifications which included abstaining from alcohol and reducing iron and vitamin C intake. As a result, his iron panel parameters improved. Thus, our case highlights that excessive alcohol consumption can exacerbate hereditary hemochromatosis and risk for overload even among heterozygotes.
Collapse
Affiliation(s)
- Serra Sozen
- Medicine, University of Vermont, Burlington, USA
- Primary Care, Danbury Hospital, Danbury, USA
| | - Akash Shah
- Internal Medicine, Nuvance Health, Brookfield, USA
| |
Collapse
|
29
|
Liu Y, Fillebeen C, Forest A, Botta A, Varin TV, Marette A, Burelle Y, Des Rosiers C, Pantopoulos K, Sweeney G. Perturbations in lipid metabolism and gut microbiota composition precede cardiac dysfunction in a mouse model of thalassemia. FASEB J 2023; 37:e23257. [PMID: 37902616 DOI: 10.1096/fj.202301043r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 09/05/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023]
Abstract
Cardiomyopathy is a major complication of thalassemia, yet the precise underlying molecular mechanisms remain unclear. We examined whether altered lipid metabolism is an early driving factor in the development of cardiomyopathy using the Th3/+ mouse model of thalassemia. At age 20 weeks, male and female Th3/+ mice manifested anemia and iron overload; however, only males displayed metabolic defects and altered cardiac function. Untargeted lipidomics indicated that the circulating levels of 35 lipid species were significantly altered in Th3/+ mice compared to wild-type controls: triglycerides (TGs) with saturated fatty acids (FAs; TG42:0 and TG44:0) were elevated, while TGs with unsaturated FAs (TG(18:2_20:5_18:2 and TG54:8)) were reduced. Similarly, phosphatidylcholines (PCs) with long chain FAs (palmitic (16:0) or oleic (18:1)) were increased, while PCs with polyunsaturated FAs decreased. Circulating PC(16:0_14:0), GlcCer(d18:1/24:0) correlated significantly with iron overload and cardiac hypertrophy. 16S rRNA gene profiling revealed alterations in the intestinal microbiota of Th3/+ mice. Differentially abundant bacterial genera correlated with PC(39:6), PC(18:1_22:6), GlcCer(d18:1/24:1) and CE(14:0). These results provide new knowledge on perturbations in lipid metabolism and the gut microbiota of Th3/+ mice and identify specific factors which may represent early biomarkers or therapeutic targets to prevent development of cardiomyopathy in β-thalassemia.
Collapse
Affiliation(s)
- Ying Liu
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Carine Fillebeen
- Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Anik Forest
- Montreal Heart Institute Research Center, Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Amy Botta
- Department of Biology, York University, Toronto, Ontario, Canada
| | - Thibault V Varin
- Department of Medicine, Heart and lung Institute, University of Laval, Quebec City, Quebec, Canada
| | - André Marette
- Department of Medicine, Heart and lung Institute, University of Laval, Quebec City, Quebec, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Christine Des Rosiers
- Montreal Heart Institute Research Center, Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Kostas Pantopoulos
- Lady Davis Institute for Medical Research and Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Gary Sweeney
- Department of Biology, York University, Toronto, Ontario, Canada
| |
Collapse
|
30
|
Kowdley KV, Modi NB, Peltekian K, Vierling JM, Ferris C, Valone FH, Gupta S. Rusfertide for the treatment of iron overload in HFE-related haemochromatosis: an open-label, multicentre, proof-of-concept phase 2 trial. Lancet Gastroenterol Hepatol 2023; 8:1118-1128. [PMID: 37863080 DOI: 10.1016/s2468-1253(23)00250-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/23/2023] [Accepted: 07/27/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Hereditary haemochromatosis protein (HFE)-related haemochromatosis, an inherited iron overload disorder caused by insufficient hepcidin production, results in excessive iron absorption and tissue and organ injury, and is treated with first-line therapeutic phlebotomy. We aimed to investigate the efficacy and safety of rusfertide, a peptidic mimetic of hepcidin, in patients with HFE-related haemochromatosis. METHODS This open-label, multicentre, proof-of-concept phase 2 trial was done across nine academic and community centres in the USA and Canada. Adults (aged ≥18 years) with HFE-related haemochromatosis on a stable therapeutic phlebotomy regimen (maintenance phase) for at least 6 months before screening and who had a phlebotomy frequency of at least 0·25 per month (eg, at least three phlebotomies in 12 months or at least four phlebotomies in 15 months) and less than one phlebotomy per month, with serum ferritin of less than 300 ng/mL and haemoglobin of more than 11·5 g/dL, were eligible. Patients initiated 24 weeks of subcutaneous rusfertide treatment within 7 days of a scheduled phlebotomy at 10 mg once weekly. Rusfertide doses and dosing schedules could be adjusted to maintain serum transferrin iron saturation (TSAT) at less than 40%. During rusfertide treatment, investigators were to consider the need for phlebotomy when the serum ferritin and TSAT values exceeded the patient's individual pre-phlebotomy serum ferritin and TSAT values. No primary endpoint or testing hierarchy was prespecified. Prespecified efficacy endpoints included the change in the frequency of phlebotomies; the proportion of patients achieving phlebotomy independence; change in serum iron, TSAT, serum transferrin, serum ferritin, and liver iron concentration (LIC) as measured by MRI; and treatment-emergent adverse events (TEAEs). The key efficacy analyses for phlebotomy rate and LIC were conducted by use of paired t tests in the intention-to-treat population, defined as all patients who received any study drug and who had pretreatment and at least one post-dose measurement. We included all participants who received at least one dose of rusfertide in the safety analyses. This trial is closed and completed and is registered with ClinicalTrials.gov, NCT04202965. FINDINGS Between March 11, 2020, and April 23, 2021, 28 patients were screened and 16 (ten [63%] men and six [38%] women) were enrolled. 16 were included in analyses of phlebotomy endpoints and 14 for the LIC endpoint. 12 (75%) patients completed 24 weeks of treatment. The mean number of phlebotomies was significantly reduced during the 24-week rusfertide treatment (0·06 phlebotomies [95% CI -0·07 to 0·20]) compared with 24 weeks pre-study (2·31 phlebotomies [95% CI 1·77 to 2·85]; p<0·0001). 15 (94%) of 16 patients were phlebotomy-free during the treatment period. Mean LIC in the 14 patients in the intention-to-treat population was 1·4 mg iron per g dry liver weight (95% CI 1·0 to 1·8) at screening and 1·1 mg iron per g dry liver weight (95% CI 0·9 to 1·3) at the end of treatment (p=0·068). Mean TSAT was 45·3% (95% CI 33·2 to 57·3) at screening, 36·7% (24·2 to 49·2) after the pretreatment phlebotomy, 21·8% (15·8 to 27·9) 24 h after the first dose of rusfertide, 40·4% (27·1 to 53·8) at the end of treatment, and 32·6% (25·0 to 40·1) over the treatment duration. Mean serum iron was 24·6 μmol/L (95% CI 18·6 to 30·6), 20·1 μmol/L (14·8 to 25·3), 11·9 μmol/L (9·2 to 14·7), 22·5 μmol/L (15·9 to 29·1), and 19·0 μmol/L (15·3 to 22·6) at these same timepoints, respectively. Mean serum ferritin was 83·3 μg/L (52·2 to 114.4), 65·5 μg/L (32·1 to 98·9), 62·8 μg/L (33·8 to 91·9), 150·0 μg/L (86·6 to 213.3), and 94·3 μg/L (54·9 to 133.6) at these same timepoints, respectively. There were only minor changes in serum transferrin concentration. 12 (75%) patients had at least one TEAE, the most common of which was injection site pain (five [31%] patients). All TEAEs were mild or moderate in severity, except for a serious adverse event of pancreatic adenocarcinoma, which was considered severe and unrelated to treatment and was pre-existing and diagnosed 21 days after starting rusfertide treatment. INTERPRETATION Rusfertide prevents iron re-accumulation in the absence of phlebotomies and could be a viable therapeutic option for selected patients with haemochromatosis. FUNDING Protagonist Therapeutics.
Collapse
Affiliation(s)
- Kris V Kowdley
- Liver Institute Northwest, Seattle, WA, USA; Elson S Floyd College of Medicine, Washington State University, Seattle, WA, USA.
| | | | - Kevork Peltekian
- Division of Digestive Care and Endoscopy, Department of Medicine, Dalhousie University, Halifax, NS, Canada
| | - John M Vierling
- Section of Gastroenterology and Hepatology and Division of Abdominal Transplantation, Baylor College of Medicine, Houston, TX, USA
| | | | | | | |
Collapse
|
31
|
Stein D, Kars ME, Wu Y, Bayrak ÇS, Stenson PD, Cooper DN, Schlessinger A, Itan Y. Genome-wide prediction of pathogenic gain- and loss-of-function variants from ensemble learning of a diverse feature set. Genome Med 2023; 15:103. [PMID: 38037155 PMCID: PMC10688473 DOI: 10.1186/s13073-023-01261-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023] Open
Abstract
Gain-of-function (GOF) variants give rise to increased/novel protein functions whereas loss-of-function (LOF) variants lead to diminished protein function. Experimental approaches for identifying GOF and LOF are generally slow and costly, whilst available computational methods have not been optimized to discriminate between GOF and LOF variants. We have developed LoGoFunc, a machine learning method for predicting pathogenic GOF, pathogenic LOF, and neutral genetic variants, trained on a broad range of gene-, protein-, and variant-level features describing diverse biological characteristics. LoGoFunc outperforms other tools trained solely to predict pathogenicity for identifying pathogenic GOF and LOF variants and is available at https://itanlab.shinyapps.io/goflof/ .
Collapse
Affiliation(s)
- David Stein
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Meltem Ece Kars
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Yiming Wu
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- College of Life Science, China West Normal University, Nan Chong, Si Chuan, 637009, China
| | - Çiğdem Sevim Bayrak
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Peter D Stenson
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Avner Schlessinger
- Department of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| | - Yuval Itan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
32
|
Cooper KM, Delk M, Devuni D, Sarkar M. Sex differences in chronic liver disease and benign liver lesions. JHEP Rep 2023; 5:100870. [PMID: 37791378 PMCID: PMC10542645 DOI: 10.1016/j.jhepr.2023.100870] [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: 01/27/2023] [Revised: 06/23/2023] [Accepted: 07/01/2023] [Indexed: 10/05/2023] Open
Abstract
The epidemiology, natural history, and therapeutic responses of chronic liver diseases and liver lesions often vary by sex. In this review, we summarize available clinical and translational data on these aspects of the most common liver conditions encountered in clinical practice, including the potential contributions of sex hormones to the underlying pathophysiology of observed differences. We also highlight areas of notable knowledge gaps and discuss sex disparities in access to liver transplant and potential strategies to address these barriers. Given established sex differences in immune response, drug metabolism, and response to liver-related therapies, emerging clinical trials and epidemiological studies should prioritize dedicated analyses by sex to inform sex-specific approaches to liver-related care.
Collapse
Affiliation(s)
- Katherine M. Cooper
- UMass Chan Medical School, Department of Medicine, Division of Gastroenterology/Hepatology, Worcester, MA, United States
| | - Molly Delk
- University of California San Francisco, Department of Medicine, Division of Gastroenterology/Hepatology, San Francisco, CA, United States
| | - Deepika Devuni
- UMass Chan Medical School, Department of Medicine, Division of Gastroenterology/Hepatology, Worcester, MA, United States
| | - Monika Sarkar
- University of California San Francisco, Department of Medicine, Division of Gastroenterology/Hepatology, San Francisco, CA, United States
| |
Collapse
|
33
|
Zhu H, Wei Y, He Q, Song Y, Liu L, Sun Y, Zhang H, Guo H, Xu X, Wang B. Association of plasma iron with the risk of incident cancer in Chinese adults with hypertension: a nested case-control study. Front Oncol 2023; 13:1223579. [PMID: 37860192 PMCID: PMC10583576 DOI: 10.3389/fonc.2023.1223579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023] Open
Abstract
Background Iron is an essential element for organismal health but excessive iron is potentially toxic. However, few observational studies link plasma iron (PI) concentrations and cancer risk, and the results are inconsistent. Objective This study aimed to explore the associations of PI concentrations with cancer risk in Chinese adults with hypertension. Methods We conducted a nested, case-control study, including 223 pairs of incident cancer cases and matched controls from the China Stroke Primary Prevention Trial. The median time between blood sample collection and subsequent cancer event occurrence was 2.13 years. The odds ratio (OR) and 95% confidence interval (CI) for the risk of cancer by PI were estimated from multivariable conditional logistic regression models. Results There was a nonlinear association between PI concentrations and total cancer risk. When compared with participants in tertile 2 of PI, the ORs of total cancer were 2.17 (95%CI: 1.25-3.85) and 1.29 (95%CI: 0.77-2.19) in participants in PI tertiles 3 and 1, respectively. Furthermore, higher PI was associated with increased digestive system cancer risk (OR=3.25, 95%CI:1.29-8.90), while lower PI was associated with increased risk of non-digestive system cancer (OR=3.32, 95%CI: 1.39-8.71). In a sensitivity analysis, the increases in total cancer risk or digestive system cancer risk were still observed with higher PI after excluding cancer cases occurring within the first year. Conclusion Our results showed an increased risk of cancer related to higher PI or lower PI in Chinese adults with hypertension. Higher iron levels were linked to an increased risk of digestive system cancers, whereas lower iron levels were linked to an increased risk of non-digestive system cancers.
Collapse
Affiliation(s)
- Hehao Zhu
- School of Science, China Pharmaceutical University, Nanjing, China
| | - Yaping Wei
- College of Public Health, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Qiangqiang He
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
- Clinical Research Center, Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Yun Song
- Clinical Research Center, Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Lishun Liu
- Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
- Clinical Research Center, Shenzhen Evergreen Medical Institute, Shenzhen, China
| | - Yong Sun
- Department of Neurosurgery, People’s Hospital of Lianyungang City/The First Affiliated Hospital of Kangda College of Nanjing Medical University, Lianyungang, China
| | - Hao Zhang
- College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Huiyuan Guo
- College of Food Sciences and Nutritional Engineering, China Agricultural University, Beijing, China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xiping Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Binyan Wang
- Clinical Research Center, Shenzhen Evergreen Medical Institute, Shenzhen, China
| |
Collapse
|
34
|
Zeitoun T, El-Sohemy A. Using Mendelian Randomization to Study the Role of Iron in Health and Disease. Int J Mol Sci 2023; 24:13458. [PMID: 37686261 PMCID: PMC10487635 DOI: 10.3390/ijms241713458] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/09/2023] [Accepted: 08/25/2023] [Indexed: 09/10/2023] Open
Abstract
Iron has been shown to play a dual role in health and disease, with either a protective or harmful effect. Some of the contradictory findings from observational studies may be due to reverse causation, residual confounding, or small sample size. One approach that may overcome these limitations without the high cost of randomized control trials is the use of Mendelian randomization to examine the long-term role of iron in a variety of health outcomes. As there is emerging evidence employing Mendelian randomization as a method of assessing the role of micronutrients in health and disease, this narrative review will highlight recent Mendelian randomization findings examining the role of iron in cardiometabolic disorders, inflammation, neurological disorders, different cancers, and a number of other health-related outcomes.
Collapse
Affiliation(s)
| | - Ahmed El-Sohemy
- Department of Nutritional Sciences, Temerty Faculty of Medicine, University of Toronto, Medical Sciences Building, Room 5326A, 1 King’s College Circle, Toronto, ON M5S 1A8, Canada;
| |
Collapse
|
35
|
Kontoghiorghes GJ. Iron Load Toxicity in Medicine: From Molecular and Cellular Aspects to Clinical Implications. Int J Mol Sci 2023; 24:12928. [PMID: 37629109 PMCID: PMC10454416 DOI: 10.3390/ijms241612928] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Iron is essential for all organisms and cells. Diseases of iron imbalance affect billions of patients, including those with iron overload and other forms of iron toxicity. Excess iron load is an adverse prognostic factor for all diseases and can cause serious organ damage and fatalities following chronic red blood cell transfusions in patients of many conditions, including hemoglobinopathies, myelodyspasia, and hematopoietic stem cell transplantation. Similar toxicity of excess body iron load but at a slower rate of disease progression is found in idiopathic haemochromatosis patients. Excess iron deposition in different regions of the brain with suspected toxicity has been identified by MRI T2* and similar methods in many neurodegenerative diseases, including Alzheimer's disease and Parkinson's disease. Based on its role as the major biological catalyst of free radical reactions and the Fenton reaction, iron has also been implicated in all diseases associated with free radical pathology and tissue damage. Furthermore, the recent discovery of ferroptosis, which is a cell death program based on free radical generation by iron and cell membrane lipid oxidation, sparked thousands of investigations and the association of iron with cardiac, kidney, liver, and many other diseases, including cancer and infections. The toxicity implications of iron in a labile, non-protein bound form and its complexes with dietary molecules such as vitamin C and drugs such as doxorubicin and other xenobiotic molecules in relation to carcinogenesis and other forms of toxicity are also discussed. In each case and form of iron toxicity, the mechanistic insights, diagnostic criteria, and molecular interactions are essential for the design of new and effective therapeutic interventions and of future targeted therapeutic strategies. In particular, this approach has been successful for the treatment of most iron loading conditions and especially for the transition of thalassemia from a fatal to a chronic disease due to new therapeutic protocols resulting in the complete elimination of iron overload and of iron toxicity.
Collapse
Affiliation(s)
- George J Kontoghiorghes
- Postgraduate Research Institute of Science, Technology, Environment and Medicine, 3, Ammochostou Street, Limassol 3021, Cyprus
| |
Collapse
|
36
|
Hofmeister RJ, Ribeiro DM, Rubinacci S, Delaneau O. Accurate rare variant phasing of whole-genome and whole-exome sequencing data in the UK Biobank. Nat Genet 2023:10.1038/s41588-023-01415-w. [PMID: 37386248 DOI: 10.1038/s41588-023-01415-w] [Citation(s) in RCA: 70] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 05/04/2023] [Indexed: 07/01/2023]
Abstract
Phasing involves distinguishing the two parentally inherited copies of each chromosome into haplotypes. Here, we introduce SHAPEIT5, a new phasing method that quickly and accurately processes large sequencing datasets and applied it to UK Biobank (UKB) whole-genome and whole-exome sequencing data. We demonstrate that SHAPEIT5 phases rare variants with low switch error rates of below 5% for variants present in just 1 sample out of 100,000. Furthermore, we outline a method for phasing singletons, which, although less precise, constitutes an important step towards future developments. We then demonstrate that the use of UKB as a reference panel improves the accuracy of genotype imputation, which is even more pronounced when phased with SHAPEIT5 compared with other methods. Finally, we screen the UKB data for loss-of-function compound heterozygous events and identify 549 genes where both gene copies are knocked out. These genes complement current knowledge of gene essentiality in the human genome.
Collapse
Affiliation(s)
- Robin J Hofmeister
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Diogo M Ribeiro
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Simone Rubinacci
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Olivier Delaneau
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
37
|
Pühringer R, Muckenthaler M, Burtscher M. Association Between Ferritin Levels and Altitude-Dependent Cardiorespiratory Fitness in Mountain Guides. High Alt Med Biol 2023; 24:139-143. [PMID: 37134197 DOI: 10.1089/ham.2023.0016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023] Open
Abstract
Pühringer, Reinhard, Martina Muckenthaler, and Martin Burtscher. Association between ferritin levels and altitude-dependent cardiorespiratory fitness in mountain guides. High Alt Med Biol. 24:139-143, 2023. Background: Higher ferritin levels may be associated with lower cardiorespiratory fitness (CRF; i.e., maximal oxygen uptake, VO2max) and may represent early markers of cardiovascular risk but may also support high-altitude acclimatization. To evaluate these potential associations, data recordings from a large sample of male mountain guides have been analyzed. Methods: A total of 154 data sets (including anthropometric data, VO2max, blood lipids, hemoglobin, ferritin, and transferrin levels) of regularly physically active and well-acclimatized mountain guides were available for analyses. Participants performed equal incremental cycle ergometer tests to exhaustion at low (600 m) and (∼1 week later) at moderate altitude (2,000 m). Results: Ferritin levels were positively correlated with levels of hemoglobin (r = 0.29, p < 0.01), total cholesterol (r = 0.18, p < 0.05), triglycerides (r = 0.23, p < 0.01), and low-density lipoprotein (r = 0.22, p < 0.01), and negatively with high-density lipoprotein levels (r = -0.16, p < 0.05) and also with baseline (taken at low altitude) VO2max values (r = -0.19, p < 0.05). In contrast, higher ferritin levels were associated with less VO2max decline from low-to-moderate altitude (r = 0.26, p < 0.01). Conclusion: Higher ferritin levels in male mountain guides are weakly associated with lower CRF and higher prevalence of cardiovascular risk factors but with slightly less reduction in VO2max when acutely exposed to moderate altitude. The clinical relevance of these observations needs further investigation.
Collapse
Affiliation(s)
- Reinhard Pühringer
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
- Austrian Society for Alpine- and High-Altitude Medicine, Innsbruck, Austria
| | - Martina Muckenthaler
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pediatric Oncology, Hematology, Immunology and Pulmonology, Centre for Translational Biomedical Iron Research, Molecular Medicine Partnership Unit (MMPU) Group Leader, University Hospital Heidelberg, Heidelberg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, Heidelberg, Germany
| | - Martin Burtscher
- Department of Sport Science, University of Innsbruck, Innsbruck, Austria
- Austrian Society for Alpine- and High-Altitude Medicine, Innsbruck, Austria
| |
Collapse
|
38
|
Abstract
Haemochromatosis is one of the most common genetic diseases affecting patients of northern European ancestry. It is overdiagnosed in patients without iron overload and is underdiagnosed in many patients. Early diagnosis by genetic testing and therapy by periodic phlebotomy can prevent the most serious complications, which include liver cirrhosis, liver cancer, and death. This Seminar includes an update on the origins of haemochromatosis; and an overview pathophysiology, genetics, natural history, signs and symptoms, differential diagnoses, treatment with phlebotomy, outcomes, and future directions.
Collapse
Affiliation(s)
- Paul C Adams
- Department of Medicine, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada.
| | - Gary Jeffrey
- Medical School, University of Western Australia, Perth, WA, Australia
| | - John Ryan
- Royal College of Surgeons of Ireland, Dublin, Ireland
| |
Collapse
|
39
|
Kersting N, Fontana JC, Athayde FPD, Carlotto FM, Machado BA, Araújo CDSRD, Sekine L, Onsten TGH, Leistner-Segal S. Hereditary hemochromatosis beyond hyperferritinemia: Clinical and laboratory investigation of the patient's profile submitted to phlebotomy in two reference centers in southern Brazil. Genet Mol Biol 2023; 46:e20220230. [PMID: 37216649 DOI: 10.1590/1678-4685-gmb-2022-0230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Accepted: 01/23/2023] [Indexed: 05/24/2023] Open
Abstract
Hereditary Hemochromatosis is a disorder characterized by iron deposition in several organs and hyperferritinemia. The most studied variants are linked to the HFE gene. In Brazil, surveys that characterize this population are scarce, with no sampling in the state of Rio Grande do Sul. Our objective is to carry out a data collection focusing on the profile of this population and the influence of the most frequently HFE variants. Two centers were enrolled: Hospital de Clínicas de Porto Alegre and Hospital São Vicente de Paulo. Patients with hyperferritinemia and undergoing phlebotomy were invited. Clinical data were collected, including HFE investigation. Among the descriptive data, the allele frequency of the C282Y variant (0.252) stands out, which differs from the national scenario. Systemic arterial hypertension was the most cited comorbidity. Differences between centers were observed, highlighting higher frequency of H63D cases in HSVP (p<0.01). Genotypes were stratified according to deleterious effect of C282Y variant. Higher transferrin saturation and number of phlebotomies were observed in the C282Y/C282Y cases (p<0.001). Positive family history for hyperferritinemia was more prevalent in compound heterozygotes (p<0.01). The results presented confirm the importance of encouraging such studies and reiterate the need for greater attention to this population.
Collapse
Affiliation(s)
- Nathalia Kersting
- Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | - Juliana Cristine Fontana
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Genética Médica, Porto Alegre, RS, Brazil
| | | | | | | | | | - Leo Sekine
- Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil
- Hospital de Clínicas de Porto Alegre, Serviço de Hemoterapia, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul (UFRGS), Faculdade de Medicina (Famed), Departamento de Medicina Interna, Porto Alegre, RS, Brazil
| | - Tor Gunnar Hugo Onsten
- Hospital de Clínicas de Porto Alegre, Serviço de Hemoterapia, Porto Alegre, RS, Brazil
- Universidade Federal do Rio Grande do Sul (UFRGS), Faculdade de Medicina (Famed), Departamento de Medicina Interna, Porto Alegre, RS, Brazil
| | - Sandra Leistner-Segal
- Universidade Federal do Rio Grande do Sul (UFRGS), Programa de Pós-Graduação em Medicina: Ciências Médicas, Porto Alegre, RS, Brazil
- Hospital de Clínicas de Porto Alegre (HCPA), Serviço de Genética Médica, Porto Alegre, RS, Brazil
| |
Collapse
|
40
|
Guo Q, Qian C, Qian ZM. Iron metabolism and atherosclerosis. Trends Endocrinol Metab 2023:S1043-2760(23)00090-5. [PMID: 37210298 DOI: 10.1016/j.tem.2023.04.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 04/20/2023] [Accepted: 04/25/2023] [Indexed: 05/22/2023]
Abstract
Despite several decades of study, whether iron is involved in the development of atherosclerosis remains a controversial and unresolved issue. Here, we focus on the up-to-date advances in studies on role of iron in atherosclerosis and discuss possible reasons why patients with hereditary hemochromatosis (HH) do not show any increased incidence of atherosclerosis. In addition, we analyze conflicting results concerning the role of iron in atherogenesis from several epidemiological and animal studies. We argue that atherosclerosis is not observed in HH because iron homeostasis in the arterial wall, the actual location of atherosclerosis, is not significantly affected, and support a causal link between iron in the arterial wall and atherosclerosis.
Collapse
Affiliation(s)
- Qian Guo
- Institute of Translational & Precision Medicine, Nantong University, Nantong, JS 226001, China; School of Medicine, Shanghai University, Shanghai 200444, China
| | - Christopher Qian
- School of Biomedical Sciences and Gerald Choa Neuroscience Centre, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong
| | - Zhong-Ming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, JS 226001, China; Laboratory of Neuropharmacology, Fudan University School of Pharmacy, Shanghai 201203, China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China.
| |
Collapse
|
41
|
Olynyk JK, Grainger R, Currie H, Ramm LE, Ramm GA. The ancestral haplotype markers HLA -A3 and B7 do not influence the likelihood of advanced hepatic fibrosis or cirrhosis in HFE hemochromatosis. Sci Rep 2023; 13:7775. [PMID: 37179448 PMCID: PMC10183001 DOI: 10.1038/s41598-023-35028-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/11/2023] [Indexed: 05/15/2023] Open
Abstract
Advanced hepatic fibrosis occurs in up to 25% of individuals with C282Y homozygous hemochromatosis. Our aim was to determine whether human leukocyte antigen (HLA)-A3 and B7 alleles act as genetic modifiers of the likelihood of advanced hepatic fibrosis. Between 1972 and 2013, 133 HFE C282Y homozygous individuals underwent clinical and biochemical evaluation, HLA typing, liver biopsy for fibrosis staging and phlebotomy treatment. Hepatic fibrosis was graded according to Scheuer as F0-2 (low grade hepatic fibrosis), F3-4 (advanced hepatic fibrosis), and F4 cirrhosis. We analysed associations between the severity of fibrosis and HLA-A3 homozygosity, heterozygosity or absence, with or without the presence of HLA-B7 using categorical analysis. The mean age of HLA-A3 homozygotes (n = 24), heterozygotes (n = 65) and HLA-A3 null individuals (n = 44) was 40 years. There were no significant differences between the groups for mean(± SEM) serum ferritin levels (1320 ± 296, 1217 ± 124, 1348 ± 188 [Formula: see text]g/L), hepatic iron concentration (178 ± 26, 213 ± 22, 199 ± 29 [Formula: see text]mol/g), mobilizable iron stores (9.9 ± 1.5, 9.5 ± 1.5, 11.5 ± 1.7 g iron removed via phlebotomy), frequency of advanced hepatic fibrosis (5/24[12%], 13/63[19%], 10/42[19%]) or cirrhosis (3/24[21%], 12/63[21%], 4/42[24%]), respectively. The presence or absence of HLA-B7 did not influence the outcome. Thus, HLA-A3 and HLA-B7 alleles are not associated with the risk of advanced hepatic fibrosis or cirrhosis in C282Y hemochromatosis.
Collapse
Affiliation(s)
- John K Olynyk
- Medical School, Curtin University, Bentley, WA, Australia.
- Department of Gastroenterology, Fiona Stanley Fremantle Hospital Group, Murdoch, WA, Australia.
| | - Richard Grainger
- Department of Gastroenterology, Fiona Stanley Fremantle Hospital Group, Murdoch, WA, Australia
| | - Helen Currie
- Department of Gastroenterology, Fiona Stanley Fremantle Hospital Group, Murdoch, WA, Australia
| | - Louise E Ramm
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, Australia
| | - Grant A Ramm
- QIMR-Berghofer Medical Research Institute, Herston, QLD, Australia
| |
Collapse
|
42
|
Crawford DHG, Ramm GA, Bridle KR, Nicoll AJ, Delatycki MB, Olynyk JK. Clinical practice guidelines on hemochromatosis: Asian Pacific Association for the Study of the Liver. Hepatol Int 2023; 17:522-541. [PMID: 37067673 DOI: 10.1007/s12072-023-10510-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/28/2023] [Indexed: 04/18/2023]
Affiliation(s)
- Darrell H G Crawford
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Gallipoli Medical Research Foundation, Brisbane, Australia
| | - Grant A Ramm
- Faculty of Medicine, The University of Queensland, Brisbane, Australia
- Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kim R Bridle
- Faculty of Medicine, The University of Queensland, Brisbane, Australia.
- Gallipoli Medical Research Foundation, Brisbane, Australia.
| | - Amanda J Nicoll
- Department of Gastroenterology, Eastern Health, Box Hill, VIC, Australia
- Monash University, Melbourne, VIC, Australia
| | - Martin B Delatycki
- Bruce Lefroy Centre, Murdoch Children's Research Institute, Melbourne, VIC, Australia
- The University of Melbourne, Melbourne, VIC, Australia
- Victorian Clinical Genetics Services, Parkville, VIC, Australia
| | - John K Olynyk
- Department of Gastroenterology, Fiona Stanley Hospital, Murdoch, WA, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA, Australia
| |
Collapse
|
43
|
de Carvalho Machado C, Dinis-Oliveira RJ. Clinical and Forensic Signs Resulting from Exposure to Heavy Metals and Other Chemical Elements of the Periodic Table. J Clin Med 2023; 12:2591. [PMID: 37048674 PMCID: PMC10095087 DOI: 10.3390/jcm12072591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/19/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Several heavy metals and other chemical elements are natural components of the Earth's crust and their properties and toxicity have been recognized for thousands of years. Moreover, their use in industries presents a major source of environmental and occupational pollution. Therefore, this ubiquity in daily life may result in several potential exposures coming from natural sources (e.g., through food and water contamination), industrial processes, and commercial products, among others. The toxicity of most chemical elements of the periodic table accrues from their highly reactive nature, resulting in the formation of complexes with intracellular compounds that impair cellular pathways, leading to dysfunction, necrosis, and apoptosis. Nervous, gastrointestinal, hematopoietic, renal, and dermatological systems are the main targets. This manuscript aims to collect the clinical and forensic signs related to poisoning from heavy metals, such as thallium, lead, copper, mercury, iron, cadmium, and bismuth, as well as other chemical elements such as arsenic, selenium, and fluorine. Furthermore, their main sources of occupational and environmental exposure are highlighted in this review. The importance of rapid recognition is related to the fact that, through a high degree of suspicion, the clinician could rapidly initiate treatment even before the toxicological results are available, which can make a huge difference in these patients' outcomes.
Collapse
Affiliation(s)
- Carolina de Carvalho Machado
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Ricardo Jorge Dinis-Oliveira
- Department of Public Health and Forensic Sciences, and Medical Education, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
- TOXRUN—Toxicology Research Unit, University Institute of Health Sciences (IUCS), CESPU, 4585-116 Gandra, Portugal
- UCIBIO-REQUIMTE-Applied Molecular Biosciences Unit, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
- MTG Research and Development Lab, 4200-604 Porto, Portugal
| |
Collapse
|
44
|
Cathcart J, Mukhopadhya A. European Association for Study of the Liver (EASL) clinical practice guidelines on haemochromatosis. Frontline Gastroenterol 2023; 14:282-286. [PMID: 37409332 PMCID: PMC11138169 DOI: 10.1136/flgastro-2022-102296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 02/14/2023] [Indexed: 07/07/2023] Open
Abstract
The European Association for the Study of the Liver has recently updated guidance on haemochromatosis with a more extensive discussion on investigation and management.[ The new guidance focuses on non-invasive methods for fibrosis assessment and early diagnosis to include more extensive genetic testing if needed. Early diagnosis and treatment is vital as it reduces morbidity and mortality. We review this guideline and offer key updated messages with a focus on new developments since the last guidance and key aspects of current practice.
Collapse
|
45
|
Zhabyeyev P, Sadasivan C, Shah S, Wang F, Oudit GY. Amlodipine rescues advanced iron overload cardiomyopathy in hemojuvelin knockout murine model: Clinical implications. Front Cardiovasc Med 2023; 10:1129349. [PMID: 37153462 PMCID: PMC10160373 DOI: 10.3389/fcvm.2023.1129349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/28/2023] [Indexed: 05/09/2023] Open
Abstract
Background Iron overload cardiomyopathy (IOC) is a major co-morbidity of genetic hemochromatosis and secondary iron overload with limited therapeutic options. We aim to investigate mechanisms of rescue action of amlodipine in the murine model of iron overload, characterize changes in human cardiac tissue due to IOC, and compare them to the changes in the animal model of IOC. Methods and results As an animal model, we used male hemojuvelin knockout (HJVKO) mice, which lacked hemojuvelin (a co-receptor protein for hepcidin expression). The mice were fed a high-iron diet from 4 weeks to 1 year of age. As a rescue, iron-fed mice received the Ca2+ channel blocker, amlodipine, from 9 to 12 months. Iron overload resulted in systolic and diastolic dysfunctions and changes in the cardiac tissue similar to the changes in the explanted human heart with IOC. An IOC patient (β-thalassemia) with left-ventricular ejection fraction (LVEF) 25% underwent heart transplantation. The murine model and the explanted heart showed intra-myocyte iron deposition, fibrosis, hypertrophy, oxidative stress, remodeling of Ca2+ cycling proteins, and metabolic kinases typical of heart failure. Single-myocyte contractility and Ca2+ release were diminished in the murine model. The amlodipine-treated group exhibited normalization of cellular function and reversed fibrosis, hypertrophy, oxidative stress, and metabolic remodeling. We also report a clinical case of primary hemochromatosis successfully treated with amlodipine. Conclusions The aged HJVKO murine model on the iron-rich diet reproduced many features of the human case of IOC. The use of amlodipine in the murine model and clinical case reversed IOC remodeling, demonstrating that amlodipine is effective adjuvant therapy for IOC.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Chandu Sadasivan
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Saumya Shah
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
| | - Faqi Wang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
| | - Gavin Y. Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, AB, Canada
- MazankowskiAlberta Heart Institute, University of Alberta, Edmonton, AB, Canada
- Correspondence: Gavin Y. Oudit
| |
Collapse
|
46
|
Habib A, Shaaban A, Thompson J, Chinnakotla S, Martin CM, Vercellotti GM. Sudden Onset Iron Overload Cardiomyopathy After Liver Transplantation. J Investig Med High Impact Case Rep 2023; 11:23247096231159812. [PMID: 36914978 PMCID: PMC10017927 DOI: 10.1177/23247096231159812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/15/2023] Open
Abstract
Iron overload cardiomyopathy has been described in patients who develop acute heart failure after liver transplantation but few reports of this are available. We present a case of a patient with end-stage liver disease who underwent a deceased donor liver transplantation and developed acute onset systolic heart failure with reduced left ventricular ejection fraction. A cardiac magnetic resonance image demonstrated late gadolinium enhancement with diffuse enhancement globally and T1 mapping with severely decreased pre-contrast T1 values suggesting iron overload cardiomyopathy. The patient was treated with iron chelating therapy as well as heart failure guideline-directed medical therapy with subsequent improvement in cardiac function on follow-up magnetic resonance images. Despite our patient's diagnosis of iron overload cardiomyopathy, her iron studies showed normal serum iron and ferritin levels and no evidence of hepatic iron deposition in the transplanted liver.
Collapse
Affiliation(s)
- Alma Habib
- University of Minnesota, Minneapolis, MN, USA.,The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | | | | | | | | |
Collapse
|
47
|
Barton JC, Edwards CQ, Acton RT. HFE hemochromatosis in African Americans: Prevalence estimates of iron overload and iron overload-related disease. Am J Med Sci 2023; 365:31-36. [PMID: 36096187 DOI: 10.1016/j.amjms.2022.08.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 05/03/2022] [Accepted: 08/10/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Little is known about the prevalence of HFE (homeostatic iron regulator) hemochromatosis in African Americans (AA). METHODS We defined AA as self-identified AA, blacks, or non-Hispanic blacks. We defined hemochromatosis-associated HFE genotypes as p.C282Y/p.C282Y and p.C282Y/p.H63D. We compiled prevalences of these genotypes in AA using published population and cohort data and numbers of men and women ≥18 y in 2018 U.S. Census estimates. We defined iron overload (IO) and IO-related disease by genotype as previously reported in population and cohort studies of hemochromatosis in whites of European ancestry. We used these definitions to estimate prevalences and numbers of AA with IO and IO-related disease associated with hemochromatosis-associated HFE genotypes. RESULTS There were ∼16,287,599 men and ∼17,644,898 women. HFE genotypes and their respective prevalences were: p.C282Y/p.C282Y, 0.00017 (6/34,905) [95% confidence interval 0.000034, 0.00031] and p.C282Y/p.H63D, 0.0012 (41/33,596) [0.000084, 0.0016]. IO prevalences were: men 0.000076 [0.000072, 0.000081] and women 0.0000061 [0.0000050, 0.0000073]. IO-related disease prevalences were: men 0.000063 [0.000059, 0.000067] and women 0.0000021 [0.0000014, 0.0000027]. There were ∼1021 [961, 1091] men and ∼36 [25, 48] women with IO-related disease. CONCLUSIONS We conclude that ∼1/25,061 AA >18 y have a hemochromatosis-associated HFE genotype and IO and that ∼1/32,103 AA >18 y have a hemochromatosis-associated HFE genotype and IO-related disease.
Collapse
Affiliation(s)
- James C Barton
- Southern Iron Disorders Center, Birmingham, AL, USA; Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Corwin Q Edwards
- Department of Medicine, Intermountain Medical Center and University of Utah, Salt Lake City, UT, USA
| | - Ronald T Acton
- Southern Iron Disorders Center, Birmingham, AL, USA; Department of Microbiology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
48
|
Affiliation(s)
- John K Olynyk
- From the Department of Gastroenterology and Hepatology, Fiona Stanley Fremantle Hospital Group, Murdoch, and the School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA (J.K.O.); and the Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Herston, QLD (G.A.R.) - all in Australia
| | - Grant A Ramm
- From the Department of Gastroenterology and Hepatology, Fiona Stanley Fremantle Hospital Group, Murdoch, and the School of Medical and Health Sciences, Edith Cowan University, Joondalup, WA (J.K.O.); and the Hepatic Fibrosis Group, QIMR Berghofer Medical Research Institute, Herston, QLD (G.A.R.) - all in Australia
| |
Collapse
|
49
|
Infanti L, Leitner G, Moe MK, Pehlic V, Benkert P, Cattaneo M, Holbro A, Passweg J, Worel N, Buser A. Indices of iron homeostasis in asymptomatic subjects with HFE mutations and moderate ferritin elevation during iron removal treatment. Blood Cells Mol Dis 2022; 97:102689. [DOI: 10.1016/j.bcmd.2022.102689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 10/31/2022]
|
50
|
Belhomme N, Morcet J, Lescoat A, Robin F, Deugnier Y, Bardou-Jacquet E, Lainé F. Patient-reported outcomes and their relation with iron parameters in HFE haemochromatosis during maintenance therapy: A prospective cohort study. Liver Int 2022; 42:2473-2481. [PMID: 35727816 DOI: 10.1111/liv.15341] [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: 03/11/2022] [Revised: 06/16/2022] [Accepted: 06/18/2022] [Indexed: 02/13/2023]
Abstract
BACKGROUND & AIMS The standard of care for haemochromatosis is regular phlebotomy in order to maintain low ferritin levels. Many patients report fatigue or joint pain despite serum ferritin within the therapeutic targets. We evaluated Patient-Reported Outcomes, and their relation with iron parameters, in C282Y homozygous patients undergoing maintenance phlebotomy. METHODS Patients were prospectively enrolled in a French referral care centre. At each phlebotomy, patients completed a numeric fatigue scale, a joint pain questionnaire and SF-36 Mental Component Score (MCS) and Physical Component Score (PCS). Haemoglobin, iron, TS and ferritin were collected concomitantly. RESULTS About 701 visits were performed in 259 patients. The median fatigue score was 3/10; 171 (66%) patients reported joint pain. Age and worsening of joint pain were associated with fatigue (p < .0001 for both). Female gender (p < .037), age (p < .003), and a decrease of TS (p = .050) were associated with joint pain. Main features associated with PCS <50 were worsening of joint pain and age (p < .001 for both) and TS <20% (p < .02). CONCLUSIONS Fatigue was independent from iron parameters. The main factor impacting quality of life was joint pain, which was more severe in patients with low TS values. Then, a more precise monitoring of TS should be proposed during haemochromatosis maintenance therapy; while less stringent monitoring of serum ferritin levels could be tested.
Collapse
Affiliation(s)
- Nicolas Belhomme
- Service de Médecine Interne et Immunologie Clinique, CHU de Rennes, Rennes, France.,Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France.,Unité d'Investigation Clinique, CHU de Rennes, Rennes, France
| | - Jeff Morcet
- Unité d'Investigation Clinique, CHU de Rennes, Rennes, France
| | - Alain Lescoat
- Service de Médecine Interne et Immunologie Clinique, CHU de Rennes, Rennes, France.,Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail) - UMR_S 1085, Rennes, France.,Faculté de médecine, Université Rennes 1, Rennes, France
| | - François Robin
- Faculté de médecine, Université Rennes 1, Rennes, France.,Service de Rhumatologie, CHU de Rennes, Rennes, France.,INSERM UMR 991, Rennes, France
| | - Yves Deugnier
- Unité d'Investigation Clinique, CHU de Rennes, Rennes, France.,Centre de référence des surcharges génétiques en fer, service des maladies du foie, CHU de Rennes, France
| | - Edouard Bardou-Jacquet
- Unité d'Investigation Clinique, CHU de Rennes, Rennes, France.,Faculté de médecine, Université Rennes 1, Rennes, France.,Centre de référence des surcharges génétiques en fer, service des maladies du foie, CHU de Rennes, France
| | - Fabrice Lainé
- Unité d'Investigation Clinique, CHU de Rennes, Rennes, France.,Centre de référence des surcharges génétiques en fer, service des maladies du foie, CHU de Rennes, France
| |
Collapse
|