1
|
Tam E, Nguyen K, Sung HK, Sweeney G. MitoNEET preserves muscle insulin sensitivity during iron overload by regulating mitochondrial iron, reactive oxygen species and fission. FEBS J 2024; 291:4062-4075. [PMID: 38944692 DOI: 10.1111/febs.17214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/13/2024] [Accepted: 06/20/2024] [Indexed: 07/01/2024]
Abstract
Iron overload (IO) is known to contribute to metabolic dysfunctions such as type 2 diabetes and insulin resistance. Using L6 skeletal muscle cells overexpressing the CDGSH iron-sulfur domain-containing protein 1 (CISD1, also known as mitoNEET) (mitoN) protein, we examined the potential role of MitoN in preventing IO-induced insulin resistance. In L6 control cells, IO resulted in insulin resistance which could be prevented by MitoN as demonstrated by western blot of p-Akt and Akt biosensor cells. Mechanistically, IO increased; mitochondrial iron accumulation, mitochondrial reactive oxygen species (ROS), Fis1-dependent mitochondrial fission, mitophagy, FUN14 domain-containing protein 1 (FUNDC1) expression, and decreased Parkin. MitoN overexpression was able to reduce increases in mitochondrial iron accumulation, mitochondrial ROS, mitochondrial fission, mitophagy and FUNDC1 upregulation due to IO. MitoN did not have any effect on the IO-induced downregulation of Parkin. MitoN alone also upregulated peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α) protein levels, a master regulator of mitochondrial biogenesis. The use of mitochondrial antioxidant, Skq1, or fission inhibitor, Mdivi-1, prevented IO-induced insulin resistance implying both mitochondrial ROS and fission play a causal role in the development of insulin resistance. Taken together, MitoN is able to confer protection against IO-induced insulin resistance in L6 skeletal muscle cells through regulation of mitochondrial iron content, mitochondrial ROS, and mitochondrial fission.
Collapse
Affiliation(s)
- Eddie Tam
- Department of Biology, York University, Toronto, Canada
| | - Khang Nguyen
- Department of Biology, York University, Toronto, Canada
| | | | - Gary Sweeney
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
2
|
Li D, Li J, Zhang H, Zhu Q, Wang T, Zhao W, Zhao S, Li W. Hereditary hemochromatosis caused by a C282Y/H63D mutation in the HFE gene: A case report. Heliyon 2024; 10:e28046. [PMID: 38560130 PMCID: PMC10979142 DOI: 10.1016/j.heliyon.2024.e28046] [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: 11/08/2023] [Revised: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Hereditary hemochromatosis (HH) is a disease characterized by disordered iron metabolism. It often involves mutations of the HFE gene, which encodes the homeostatic iron regulator protein (HFE), as well as mutations affecting hepcidin antimicrobial peptide, hemojuvelin, or transferrin receptor 2. Historically, HH has been observed primarily in European and European diaspora populations, while classical HH is rare in Asian populations, including in China. In this article, we report a rare case of HH in a Chinese man that could be attributed to a heterozygous C282Y/H63D HFE mutation. Based on clinical examination, liver biopsy, and genetic testing results, the patient was diagnosed with HH. Clinical signs and symptoms and serum iron-related test results were recorded for a period of two years after the patient began treatment. Over this observation period, the patient was subjected to 25 phlebotomies (accounting for a total blood loss of 10.2 L). His serum ferritin levels decreased from 1550 μg/L to 454 μg/L, his serum iron concentration decreased from 40 μmol/L to 24.6 μmol/L, and his transferrin saturation decreased from 97.5% to 55.1%. Early diagnosis is essential for patients with HH to obtain good outcomes. Regular phlebotomy after diagnosis can improve HH symptoms and delay HH disease progression.
Collapse
Affiliation(s)
- Dongdong Li
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| | - Jinfeng Li
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| | - Hongkun Zhang
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| | - Qiuyu Zhu
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| | - Teng Wang
- Yiwu Central Hospital, 322000 Yiwu, Zhejiang, China
| | - Wen Zhao
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| | - Shousong Zhao
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| | - Wei Li
- Department of Infectious Diseases, First Affiliated Hospital of Bengbu Medical College, 233000 Bengbu, Anhui, China
- National Clinical Research Center for Infectious Diseases, China
| |
Collapse
|
3
|
Romero‐Cortadellas L, Venturi V, Martín‐Sánchez JC, Toska K, Prince D, Butzeck B, Porto G, Milman NT, Committee HIS, Sánchez M. Haemochromatosis patients' research priorities: Towards an improved quality of life. Health Expect 2023; 26:2293-2301. [PMID: 37503783 PMCID: PMC10632644 DOI: 10.1111/hex.13830] [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: 05/15/2023] [Revised: 06/16/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Chronic diseases are associated with a range of functional and psychosocial consequences that can adversely affect patients' quality of life (QoL). Haemochromatosis (HC) is a genetically heterogeneous disorder characterized by chronic iron overload that can ultimately lead to multiple organ dysfunction. Clinical diagnosis remains challenging due to the nonspecificity of symptoms and a lack of confirmatory genotyping in a substantial proportion of patients. Illness perception among HC patients has not been extensively investigated, lacking relevant information on how to improve their QoL. METHODS We present the results of the first worldwide survey conducted in nearly 1500 HC respondents, in which we collected essential demographic information and identified the aspects that concern HC patients the most. RESULTS Out of all the participants, 45.3% (n = 676) voiced their concern about physical and psychological consequences such as HC-related arthropathies, which can ultimately affect their social functioning. A similar proportion of patients (n = 635, 42.5%) also consider that better-informed doctors are key for improved HC disease management. Taking a patient-centred approach, we expose differences in patients' disease perspective by social and economic influences. CONCLUSIONS We identify potential targets to improve patients' health-related QoL and reflect on strategic measures to foster gender equity in access to health resources. Finally, we make a call for a highly coordinated effort across a range of public policy areas to empower participants in the HC research process and design. PATIENT OR PUBLIC CONTRIBUTION Nearly 1500 patients with hereditary HC responded to an anonymized online survey in which research and clinical priorities were addressed regarding this chronic and rare disease.
Collapse
Affiliation(s)
- Lídia Romero‐Cortadellas
- Department of Basic Sciences, Iron metabolism: Regulation and DiseasesUniversitat Internacional de Catalunya (UIC)Sant Cugat del VallèsBarcelonaSpain
| | - Veronica Venturi
- Department of Basic Sciences, Iron metabolism: Regulation and DiseasesUniversitat Internacional de Catalunya (UIC)Sant Cugat del VallèsBarcelonaSpain
| | - Juan Carlos Martín‐Sánchez
- Group of Evaluation of Health Determinants and Health Policies, Department of Basic SciencesUniversitat Internacional de CatalunyaSant Cugat del VallèsSpain
| | - Ketil Toska
- Norwegian Haemochromatosis AssociationBergenNorway
| | - Dianne Prince
- Haemochromatosis AustraliaMeridan PlainsQueenslandAustralia
| | - Barbara Butzeck
- Hämochromatose‐Vereinigung Deutschland e.V. HVDEuropean Federation of Associations of Patients with Haemochromatosis (EFAPH)HattingenGermany
| | - Graça Porto
- i3S—Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar.Universidade do PortoPortoPortugal
| | | | | | - Mayka Sánchez
- Department of Basic Sciences, Iron metabolism: Regulation and DiseasesUniversitat Internacional de Catalunya (UIC)Sant Cugat del VallèsBarcelonaSpain
- BloodGenetics S.L. Diagnostics in Inherited Blood DiseasesEsplugues de LlobregatSpain
| |
Collapse
|
4
|
Heilmeier U, Burghardt AJ, Tse JJ, Kapoor P, Stok KS, Manske S, Voll RE, Schett G, Finzel S. Analysis of Hand Joint Space Morphology in Women and Men with Hereditary Hemochromatosis. Calcif Tissue Int 2023; 112:440-451. [PMID: 36738308 PMCID: PMC10025180 DOI: 10.1007/s00223-022-01050-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 11/25/2022] [Indexed: 02/05/2023]
Abstract
Hereditary hemochromatosis (HH) causes unbalanced iron deposition in many organs including the joints leading to severe cartilage loss and bone damage in the metacarpophalangeal joints (MCPJ). High-resolution peripheral quantitative computed tomography (HR-pQCT) and its joint space width (JSW) quantification algorithm quantifies in vivo 3D joint morphology. We therefore aimed to (i) determine feasibility and performance of the JSW algorithm in HH, (ii) quantify joint space morphology, and (iii) investigate the relationship between morphological and clinical parameters in HH. Here, we performed an exploratory study on 24 HH patients and sex- and age-matched controls using HR-pQCT imaging of MCPJ. Mineralized bone structure was automatically segmented from the grayscale image data and periosteal surface bone masks and joint space masks were generated. Mean, minimal, and maximal joint space width (JSW; JSW.MIN; JSW.MAX), JSW heterogeneity (JSW.SD), JSW asymmetry (JSW.AS), and joint space volume (JSV) were computed. Demographics and, for HH patients, disease-specific parameters were recorded. Segmentation of JS was very good with 79.7% of MCPJs successfully segmented at first attempt and 20.3% requiring semi-manual correction. HH men showed larger JSV at all MCPs (+ 25.4% < JSV < + 41.8%, p < 0.05), larger JSW.MAX at MCP 3-4 (+ 14%, 0.006 < p < 0.062), and wider JSW (+ 13%, p = 0.043) at MCP 4 relative to HH women. Compared to controls, both HH men and HH women showed larger JSW.AS and smaller JSW.MIN at all MCP levels, reaching significance for HH men at MCP 2 and 3 (JSW.AS: + 323% < JSW.AS < + 359%, 0.020 < p < 0.043; JSW.MIN: - 216% < JSW.MIN < - 225%, p < 0.043), and for women at MCP 3 (JSW.AS: + 180%, p = 0.025; JSW.MIN: - 41.8%, p = 0.022). Time since HH diagnosis was correlated positively with MCP 4 JSW.AS and JSW.SD (0.463 < ρ < 0.499, p < 0.040), and the number of phlebotomies since diagnosis was correlated with JSW.SD at all MCPs (0.432 < ρ < 0.535, p < 0.050). HR-pQCT-based JSW quantification in MCPJ of HH patients is feasible, performs well even in narrow JS, and allows to define the microstructural joint burden of HH.
Collapse
Affiliation(s)
- Ursula Heilmeier
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Freiburg - Medical Center, Hugstetterstraße 55, 79106, Freiburg, Germany.
- Musculoskeletal Quantitative Imaging Research Group, University of California San Francisco, 185 Berry Street, San Francisco, CA, 94158, USA.
| | - Andrew J Burghardt
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry Street, San Francisco, CA, 94158, USA
| | - Justin J Tse
- Department of Radiology, Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| | - Puneet Kapoor
- Musculoskeletal Quantitative Imaging Research Group, University of California San Francisco, 185 Berry Street, San Francisco, CA, 94158, USA
| | - Kathryn S Stok
- Department of Biomedical Engineering, The University of Melbourne, Parkville, VIC, Australia
| | - Sarah Manske
- Department of Radiology, Cumming School of Medicine, McCaig Institute for Bone and Joint Health, University of Calgary, 3280 Hospital Drive NW, Calgary, AB, T2N 4Z6, Canada
| | - Reinhard E Voll
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Freiburg - Medical Center, Hugstetterstraße 55, 79106, Freiburg, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Stephanie Finzel
- Department of Rheumatology and Clinical Immunology, Faculty of Medicine, University of Freiburg - Medical Center, Hugstetterstraße 55, 79106, Freiburg, Germany
| |
Collapse
|
5
|
Abstract
The cardiovascular system requires iron to maintain its high energy demands and metabolic activity. Iron plays a critical role in oxygen transport and storage, mitochondrial function, and enzyme activity. However, excess iron is also cardiotoxic due to its ability to catalyze the formation of reactive oxygen species and promote oxidative damage. While mammalian cells have several redundant iron import mechanisms, they are equipped with a single iron-exporting protein, which makes the cardiovascular system particularly sensitive to iron overload. As a result, iron levels are tightly regulated at many levels to maintain homeostasis. Iron dysregulation ranges from iron deficiency to iron overload and is seen in many types of cardiovascular disease, including heart failure, myocardial infarction, anthracycline-induced cardiotoxicity, and Friedreich's ataxia. Recently, the use of intravenous iron therapy has been advocated in patients with heart failure and certain criteria for iron deficiency. Here, we provide an overview of systemic and cellular iron homeostasis in the context of cardiovascular physiology, iron deficiency, and iron overload in cardiovascular disease, current therapeutic strategies, and future perspectives.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Adam De Jesus
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
| | - Hossein Ardehali
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, IL 60611
- Division of Cardiology, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| |
Collapse
|
6
|
Fernandez M, Lokan J, Leung C, Grigg A. A critical evaluation of the role of iron overload in fatty liver disease. J Gastroenterol Hepatol 2022; 37:1873-1883. [PMID: 35906772 DOI: 10.1111/jgh.15971] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 07/16/2022] [Accepted: 07/27/2022] [Indexed: 12/09/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has been associated with a condition known as the dysmetabolic iron overload syndrome, but the frequency and severity of iron overload in NAFLD is not well described. There is emerging evidence that mild to moderate excess hepatic iron can aggravate the risk of progression of NAFLD to nonalcoholic steatohepatitis and eventually cirrhosis. Mechanisms are postulated to be via reactive oxygen species, inflammatory cytokines, lipid oxidation, and oxidative stress. The aim of this review is to assess the evidence for true hepatic iron overload in NAFLD, to discuss the pathogenesis by which excess iron may be toxic, and to critically evaluate the studies designed to deplete iron by regular venesection. In brief, the studies are inconclusive due to heterogeneity in eligibility criteria, sample size, randomization, hepatic iron measurement, serial histological endpoints, target ferritin levels, length of venesection, and degree of confounding lifestyle intervention. We propose a trial designed to overcome the limitations of these studies.
Collapse
Affiliation(s)
- Monique Fernandez
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia
| | - Julie Lokan
- Department of Anatomical Pathology, Austin Health, Heidelberg, Victoria, Australia
| | - Christopher Leung
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.,Departments of Gastroenterology and Hepatology, Austin Health, Heidelberg, Victoria, Australia
| | - Andrew Grigg
- Department of Medicine, University of Melbourne, Parkville, Victoria, Australia.,Department of Clinical Haematology, Austin Health, Heidelberg, Victoria, Australia
| |
Collapse
|
7
|
Kroll DS, McPherson KL, Manza P, Schwandt ML, Shen PH, Goldman D, Diazgranados N, Wang GJ, Wiers CE, Volkow ND. Elevated transferrin saturation in individuals with alcohol use disorder: Association with HFE polymorphism and alcohol withdrawal severity. Addict Biol 2022; 27:e13144. [PMID: 35229939 PMCID: PMC9373047 DOI: 10.1111/adb.13144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 11/07/2021] [Accepted: 12/21/2021] [Indexed: 11/26/2022]
Abstract
Iron loading has been consistently reported in those with alcohol use disorder (AUD), but its effect on the clinical course of the disease is not yet fully understood. Here, we conducted a cohort study to examine whether peripheral iron measures, genetic variation in HFE rs1799945 and their interaction differed between 594 inpatient participants with alcohol use disorder (AUD) undergoing detoxification and 472 healthy controls (HC). We also assessed whether HFE rs1799945 was associated with elevated peripheral iron and can serve as a predictor of withdrawal severity. AUD patients showed significantly higher serum transferrin saturation than HC. Within the AUD group, transferrin saturation significantly predicted withdrawal symptoms (CIWA-Ar) and cumulative dose of benzodiazepine treatment during the first week of detoxification, which is an indicator of withdrawal severity. HFE rs1799945 minor allele carriers showed elevated transferrin saturation compared to non-carriers, both in AUD and healthy controls. Exploratory analyses indicated that, within the AUD cohort, HFE rs1799945 predicted CIWA withdrawal scores, and this relationship was significantly mediated by transferrin saturation. We provide evidence that serum transferrin saturation predicts alcohol withdrawal severity in AUD. Moreover, our findings replicated previous studies on elevated serum transferrin saturation in AUD and an involvement of HFE rs1799945 in serum transferrin saturation levels in both AUD and healthy controls. Future studies may use transferrin saturation measures as predictors for treatment or potentially treat iron overload to ameliorate withdrawal symptoms.
Collapse
Affiliation(s)
- Danielle S. Kroll
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Katherine L. McPherson
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Peter Manza
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Melanie L. Schwandt
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Pei-Hong Shen
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - David Goldman
- Laboratory of Neurogenetics, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Nancy Diazgranados
- Office of the Clinical Director, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Gene-Jack Wang
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
| | - Corinde E. Wiers
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nora D. Volkow
- Laboratory of Neuroimaging, National Institute on Alcohol Abuse and Alcoholism, Bethesda, Maryland
- National Institute on Drug Abuse, Bethesda, Maryland
| |
Collapse
|
8
|
Acevedo LAU, Alvarenga AM, Fonseca PFS, da Silva NK, Cançado RD, Naoum FA, Dinardo CL, Pereira AC, Brissot P, Santos PCJL. Quality of Life Scores Remained Different among the Genotypic Groups of Patients with Suspected Hemochromatosis, Even after Treatment Period. Genes (Basel) 2022; 13:genes13010118. [PMID: 35052458 PMCID: PMC8774363 DOI: 10.3390/genes13010118] [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: 12/02/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Hemochromatosis is a genetic condition of iron overload caused by deficiency of hepcidin. In a previous stage of this study, patients with suspected hemochromatosis had their quality of life (QL) measured. We observed that QL scores differed among genotypic groups of patients. In this reported final phase of the study, the aims were to compare QL scores after a treatment period of approximately 3 years and to analyze a possible association of the serum ferritin values with QL scores. Methods: Sixty-five patients were enrolled in this final phase and divided into group 1 (patients that showed primary iron overload and homozygous genotype for the HFE p.Cys282Tyr mutation) and group 2 (other kinds of genotypes). Short Form 36 (SF-36) was performed and consisted of eight domains with a physical and also a mental component. Results: Both groups had a significant decrease in serum ferritin concentrations: group 1 had a variation from 1844 ± 1313 ng/mL to 281 ± 294 ng/mL, and group 2 had a variation from 1216 ± 631 ng/mL to 236 ± 174 ng/mL. Group 1 had a smaller mean value for these six SF-36 domains compared with group 2, indicating a worse QL. Conclusions: In this final stage, six domains demonstrated a difference among genotypic groups (role emotional and mental health, adding to the four of the initial phase), reassuring the impact of the identified genotype on the QL of hemochromatosis patients. Furthermore, despite that both patient groups demonstrated similar and significant decreases in serum ferritin values, no association was found between the decrease in this biological parameter and the SF-36 domains.
Collapse
Affiliation(s)
- Luis Alfredo Utria Acevedo
- Department of Pharmacology—Escola Paulista de Medicina, Universidade Federal de Sao Paulo (EPM-Unifesp), Sao Paulo 05403-904, Brazil; (L.A.U.A.); (A.M.A.); (N.K.d.S.)
| | - Aline Morgan Alvarenga
- Department of Pharmacology—Escola Paulista de Medicina, Universidade Federal de Sao Paulo (EPM-Unifesp), Sao Paulo 05403-904, Brazil; (L.A.U.A.); (A.M.A.); (N.K.d.S.)
| | - Paula Fernanda Silva Fonseca
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo 05403-904, Brazil; (P.F.S.F.); (A.C.P.)
| | - Nathália Kozikas da Silva
- Department of Pharmacology—Escola Paulista de Medicina, Universidade Federal de Sao Paulo (EPM-Unifesp), Sao Paulo 05403-904, Brazil; (L.A.U.A.); (A.M.A.); (N.K.d.S.)
| | | | | | - Carla Luana Dinardo
- Fundação Pró-Sangue, Hemocentro de São Paulo, São Paulo, Brazil, Universidade de São Paulo (USP), Sao Paulo 05403-904, Brazil;
| | - Alexandre Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo 05403-904, Brazil; (P.F.S.F.); (A.C.P.)
| | - Pierre Brissot
- Institut NuMeCan, Inserm U-1241, Univ Rennes 1, 35000 Rennes, France;
| | - Paulo Caleb Junior Lima Santos
- Department of Pharmacology—Escola Paulista de Medicina, Universidade Federal de Sao Paulo (EPM-Unifesp), Sao Paulo 05403-904, Brazil; (L.A.U.A.); (A.M.A.); (N.K.d.S.)
- Correspondence:
| |
Collapse
|
9
|
Postoperative Complications in Patients With Hereditary Hemochromatosis Undergoing Total Joint Arthroplasty: A Matched Cohort Analysis. J Am Acad Orthop Surg 2022; 30:e99-e107. [PMID: 34932508 DOI: 10.5435/jaaos-d-21-00105] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 07/14/2021] [Indexed: 02/01/2023] Open
Abstract
AIMS The purpose of this study is to determine differences in the rates of 90-day postoperative complications and 2- and 5-year surgical outcomes between patients with and without hereditary hemochromatosis (HH) after total joint arthroplasty (TJA). METHODS Patients who underwent primary total hip arthroplasty (THA) or total knee arthroplasty (TKA) from 2010 to 2018 were identified in a national database (PearlDiver Technologies) using Current Procedural Terminology and International Classification of Diseases-9/10 codes. Patients with a history of HH were identified within the THA and TKA cohorts and matched with non-HH patients based on age, sex, Charlson Comorbidity Index, smoking status, and obesity (body mass index > 30). Ninety-day medical complications assessed included renal failure, arrhythmia, bleeding complications, blood transfusion, pneumonia, stroke, deep vein thrombosis, liver failure, heart failure, pulmonary embolism, sepsis, surgical site infection, wound dehiscence, readmission rate, and death. Two- and 5-year surgical complications assessed included all-cause revision, prosthetic joint infection, implant loosening, joint stiffness, and manipulation under anesthesia. All complications were analyzed using bivariate analysis and logistic regression, with significance set at P < 0.05. RESULTS Compared with non-HH patients, patients with HH had higher rates of stiffness at 2 and 5 years after THA (all, P < 0.001), as well as higher rates of aseptic loosening at 5 years after TKA (P = 0.036). However, patients with HH undergoing THA and TKA had no notable difference in 90-day postoperative complications when compared with non-HH patients. DISCUSSION Compared with non-HH patients, patients with HH undergoing TJA were shown to have worse 2- and 5-year surgical outcomes, without any increased risk of 90-day medical complications. These findings may be useful for surgical decision making for patients with HH undergoing TJA. CLINICAL RELEVANCE This study addresses a paucity in the current literature concerning the complication profile in HH patients with destructive joint arthropathy undergoing joint arthroplasty surgery.
Collapse
|
10
|
Distribution and Associated Factors of Hepatic Iron-A Population-Based Imaging Study. Metabolites 2021; 11:metabo11120871. [PMID: 34940629 PMCID: PMC8705957 DOI: 10.3390/metabo11120871] [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: 12/01/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 11/17/2022] Open
Abstract
Hepatic iron overload can cause severe organ damage; therefore, an early diagnosis and the identification of potential risk factors is crucial. We aimed to investigate the sex-specific distribution of hepatic iron content (HIC) in a population-based cohort and identify relevant associated factors from a panel of markers. We analyzed N = 353 participants from a cross-sectional sample (KORA FF4) who underwent whole-body magnetic resonance imaging. HIC was assessed by single-voxel spectroscopy with a high-speed T2-corrected multi-echo technique. A large panel of markers, including anthropometric, genetic, and laboratory values, as well as behavioral risk factors were assessed. Relevant factors associated with HIC were identified by variable selection based on LASSO regression with bootstrap resampling. HIC in the study sample (mean age at examination: 56.0 years, 58.4% men) was significantly lower in women (mean ± SD: 39.2 ± 4.1 s-1) than in men (41.8 ± 4.7 s-1, p < 0.001). Relevant factors associated with HIC were HbA1c as well as prediabetes for men and visceral adipose tissue as well as age for women. Hepatic fat, alcohol consumption, and genetic risk score for iron levels were associated with HIC in both sexes. In conclusion, there are sex-specific associations of HIC with markers of body composition, glucose metabolism, and alcohol consumption.
Collapse
|
11
|
Hruby M, Martínez IIS, Stephan H, Pouckova P, Benes J, Stepanek P. Chelators for Treatment of Iron and Copper Overload: Shift from Low-Molecular-Weight Compounds to Polymers. Polymers (Basel) 2021; 13:3969. [PMID: 34833268 PMCID: PMC8618197 DOI: 10.3390/polym13223969] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/09/2021] [Accepted: 11/15/2021] [Indexed: 12/18/2022] Open
Abstract
Iron and copper are essential micronutrients needed for the proper function of every cell. However, in excessive amounts, these elements are toxic, as they may cause oxidative stress, resulting in damage to the liver and other organs. This may happen due to poisoning, as a side effect of thalassemia infusion therapy or due to hereditary diseases hemochromatosis or Wilson's disease. The current golden standard of therapy of iron and copper overload is the use of low-molecular-weight chelators of these elements. However, these agents suffer from severe side effects, are often expensive and possess unfavorable pharmacokinetics, thus limiting the usability of such therapy. The emerging concepts are polymer-supported iron- and copper-chelating therapeutics, either for parenteral or oral use, which shows vivid potential to keep the therapeutic efficacy of low-molecular-weight agents, while avoiding their drawbacks, especially their side effects. Critical evaluation of this new perspective polymer approach is the purpose of this review article.
Collapse
Affiliation(s)
- Martin Hruby
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic Heyrovského Náměstí 2, 162 06 Prague, Czech Republic;
| | - Irma Ivette Santana Martínez
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400, 01328 Dresden, Germany; (I.I.S.M.); (H.S.)
| | - Holger Stephan
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research Bautzner Landstraße 400, 01328 Dresden, Germany; (I.I.S.M.); (H.S.)
| | - Pavla Pouckova
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Salmovska 1, 120 00 Prague, Czech Republic; (P.P.); (J.B.)
| | - Jiri Benes
- Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Salmovska 1, 120 00 Prague, Czech Republic; (P.P.); (J.B.)
| | - Petr Stepanek
- Institute of Macromolecular Chemistry, Academy of Sciences of the Czech Republic Heyrovského Náměstí 2, 162 06 Prague, Czech Republic;
| |
Collapse
|
12
|
Ying H, Shen Z, Wang J, Zhou B. Role of iron homeostasis in the heart : Heart failure, cardiomyopathy, and ischemia-reperfusion injury. Herz 2021; 47:141-149. [PMID: 33978777 DOI: 10.1007/s00059-021-05039-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/15/2020] [Accepted: 04/09/2021] [Indexed: 02/07/2023]
Abstract
As an essential trace mineral in mammals and the second most abundant metal in the Earth's crust, iron acts as a double-edged sword in humans. Iron plays important beneficial roles in numerous biological processes ranging from deoxyribonucleic acid biosynthesis and protein function to cell cycle progression. However, iron metabolism disruption leads to widespread tissue degeneration and organ dysfunction. An increasing number of studies have focused on iron regulation pathways and have explored the relationship between iron and cardiovascular diseases. Ferroptosis, an iron-dependent form of programmed cell death, was first described in cancer cells and has recently been linked to heart diseases, including cardiac ischemia-reperfusion injury and doxorubicin-induced myocardiopathy. Here, we summarize recent advances in our understanding of iron homeostasis and heart diseases and discuss potential relationships between ferroptosis and cardiac ischemia-reperfusion injury and cardiomyopathy.
Collapse
Affiliation(s)
- Hangying Ying
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Zhejiang, Hangzhou, China
| | - Zhida Shen
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Zhejiang, Hangzhou, China
| | - Jiacheng Wang
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Zhejiang, Hangzhou, China
| | - Binquan Zhou
- Department of Cardiology, Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016, Zhejiang, Hangzhou, China.
| |
Collapse
|
13
|
Zaky S, Alboraie M, El Badry M, Metwally MA, Abdelaziz A, Fouad Y, Abd-Elsalam S, Mahmoud A, Shiha G, Baki AA, El Kassas M, Esmat G. Management of liver disease patients in different clinical situations during COVID-19 pandemic. EGYPTIAN LIVER JOURNAL 2021; 11:21. [PMID: 34777868 PMCID: PMC7994958 DOI: 10.1186/s43066-021-00091-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/10/2021] [Indexed: 02/06/2023] Open
Abstract
Chronic liver diseases are common worldwide, especially in developing countries. The rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)/(COVID-19) leads to the infection of many patients with underlying chronic liver diseases. As a relatively new disease, management of COVID-19, in the context of chronic liver disease, is mainly based on the experience of the treating physician and the available data. In this review, we summarize the available evidence about the management of liver disease patients, in the context of COVID-19 infection, which can increase the severity of viral hepatitis B. Also, its clearance in HBV patients is delayed. A sixfold increased severity of COVID-19 was reported in obese patients with metabolic associated fatty liver disease (MAFDL). In patients with autoimmune liver disease (AILD), it is not recommended to change their immunosuppressive therapy (as long as they are not infected with COVID-19), in order to avoid a flare of liver disease. However, immunosuppressant drugs should be modified, in the case of infection with COVID-19. To date, no data suggest an increased risk or severity in metabolic liver diseases, such as hemochromatosis, Wilson's disease, or alpha-1 antitrypsin deficiency. Patients with liver cirrhosis should be carefully managed with minimum exposure to healthcare facilities. Basic investigations for follow-up can be scheduled at wider intervals; if patients need admission, this should be in COVID-19-clean areas. Patients with hepatocellular carcinomas may have a poor prognosis according to preliminary reports from China. The course of COVID-19 in liver transplant recipients on immunosuppression seems to have a benign course, based on few reports in children and adults. The hepatotoxicity of COVID-19 drugs ranges from mild liver enzyme elevation to a flare of underlying liver diseases. Therefore, the decision should be customized. Telemedicine can minimize the exposure of healthcare workers and patients to infection with COVID-19 and decrease the consumption of personal protective equipment.
Collapse
Affiliation(s)
- Samy Zaky
- Hepatogastroenterology and Infectious Diseases Department, Al-Azhar University, Cairo, Egypt
| | - Mohamed Alboraie
- Department of Internal Medicine, Al-Azhar University, Cairo, Egypt
| | - Mohamed El Badry
- Endemic Medicine Department, Faculty of Medicine, Helwan University, 2-Ahmed Elzomor Street, Nasr City, Cairo Egypt
| | - Mohamed A. Metwally
- Hepatology, Gastroenterology and Infectious Diseases Department, Benha University, Benha, Egypt
| | - Ahmed Abdelaziz
- Hepatogastroenterology and Infectious Diseases, Al-Azhar University, Demiatta, Egypt
| | - Yasser Fouad
- Tropical Medicine Department, Minia Faculty of Medicine, Minia University, Minia, Egypt
| | | | - Abdelmajeed Mahmoud
- Tropical Medicine and Gastroenterology Department, Aswan University, Aswan, Egypt
| | - Gamal Shiha
- Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Amin Abdel Baki
- Department of Hepatology, National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Mohamed El Kassas
- Endemic Medicine Department, Faculty of Medicine, Helwan University, 2-Ahmed Elzomor Street, Nasr City, Cairo Egypt
| | - Gamal Esmat
- Endemic Medicine and Hepatology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Chidambaranathan-Reghupaty S, Fisher PB, Sarkar D. Hepatocellular carcinoma (HCC): Epidemiology, etiology and molecular classification. Adv Cancer Res 2020; 149:1-61. [PMID: 33579421 PMCID: PMC8796122 DOI: 10.1016/bs.acr.2020.10.001] [Citation(s) in RCA: 505] [Impact Index Per Article: 101.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC), the primary malignancy of hepatocytes, is a diagnosis with bleak outcome. According to National Cancer Institute's SEER database, the average five-year survival rate of HCC patients in the US is 19.6% but can be as low as 2.5% for advanced, metastatic disease. When diagnosed at early stages, it is treatable with locoregional treatments including surgical resection, Radio-Frequency Ablation, Trans-Arterial Chemoembolization or liver transplantation. However, HCC is usually diagnosed at advanced stages when the tumor is unresectable, making these treatments ineffective. In such instances, systemic therapy with tyrosine kinase inhibitors (TKIs) becomes the only viable option, even though it benefits only 30% of patients, provides only a modest (~3months) increase in overall survival and causes drug resistance within 6months. HCC, like many other cancers, is highly heterogeneous making a one-size fits all option problematic. The selection of liver transplantation, locoregional treatment, TKIs or immune checkpoint inhibitors as a treatment strategy depends on the disease stage and underlying condition(s). Additionally, patients with similar disease phenotype can have different molecular etiology making treatment responses different. Stratification of patients at the molecular level would facilitate development of the most effective treatment option. With the increase in efficiency and affordability of "omics"-level analysis, considerable effort has been expended in classifying HCC at the molecular, metabolic and immunologic levels. This review examines the results of these efforts and the ways they can be leveraged to develop targeted treatment options for HCC.
Collapse
Affiliation(s)
- Saranya Chidambaranathan-Reghupaty
- C. Kenneth and Dianne Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, United States
| | - Paul B Fisher
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States
| | - Devanand Sarkar
- Department of Human and Molecular Genetics, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Institute of Molecular Medicine, Virginia Commonwealth University, School of Medicine, Richmond, VA, United States; VCU Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
15
|
Nichols L, Rowley L, Lall A. Disseminated cryptococcosis and hemochromatosis: clues to diagnosis. AUTOPSY AND CASE REPORTS 2020; 10:e2020180. [PMID: 33344301 PMCID: PMC7703462 DOI: 10.4322/acr.2020.180] [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] [Indexed: 11/23/2022] Open
Abstract
Hepatic cirrhosis, diabetes mellitus and iron overload can each independently predispose to cryptococcosis. Hereditary hemochromatosis leads to all three of these predispositions. This report is the case of a patient with chronic hepatitis B virus infection and cirrhosis, who had markedly elevated serum ferritin and 99% transferrin saturation, and developed a leukemoid reaction. Autopsy revealed disseminated cryptococcosis for which the leukemoid reaction was a clue and possible hereditary hemochromatosis of which elevated ferritin and transferrin saturation can be clues. Hereditary hemochromatosis is an important diagnosis clinicians should never miss because early treatment with phlebotomy can be life-saving. Disseminated cryptococcosis can be rapidly diagnosed with serum cryptococcal antigen test and is treatable.
Collapse
Affiliation(s)
- Larry Nichols
- Mercer University School of Medicine, Department of Pathology. Macon, GA, USA
| | | | - Ashley Lall
- Mercer University School of Medicine. Macon, GA, USA
| |
Collapse
|
16
|
Limberg AK, Chalmers BP, Perry KI, Lewallen DG, Berry DJ, Abdel MP. Contemporary Total Hip and Total Knee Arthroplasty Results in Patients with Hemochromatosis. J Arthroplasty 2020; 35:976-980. [PMID: 31870583 DOI: 10.1016/j.arth.2019.11.038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/21/2019] [Accepted: 11/26/2019] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Hemochromatosis can result in metabolic bone pathology (due to excessive iron absorption) and degenerative joint disease, leading to total joint arthroplasties. The aim of this study is to analyze the survivorship, complications, radiographic results, and clinical outcomes of patients with hemochromatosis who received either a total hip arthroplasty (THA) or a total knee arthroplasty (TKA). METHODS We identified 34 lower extremity arthroplasties in 29 patients with hemochromatosis performed between 2000 and 2016. There were 17 primary THAs in 15 patients and 17 primary TKAs in 14 patients. Mean age at arthroplasty was 63 years with 76% being male. The mean body mass index was 28 kg/m2. Mean follow-up was 5 years. RESULTS The survivorship free from any revision for THAs was 94% at 10 years. One patient was revised for aseptic loosening of the femoral stem at 6 months. In THA patients, no infections, no other complications, and no radiographic evidence of aseptic loosening were identified. Harris Hip Scores improved from a mean of 55 preoperatively to 94 postoperatively (P < .001). The survivorship free from any revision for TKAs was 100% at 10 years. Two patients (12%) developed acquired idiopathic stiffness postoperatively; no infections were identified. There was no radiographic evidence of aseptic loosening in any TKA. Knee Society Scores improved from a mean of 61 preoperatively to 94 postoperatively (P < .001). CONCLUSION This study found excellent survivorship, significant improvements in clinical outcomes, and a very low complication profile for both THA and TKA in patients with hemochromatosis.
Collapse
Affiliation(s)
- Afton K Limberg
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | - Kevin I Perry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | | | - Daniel J Berry
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| | - Matthew P Abdel
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN
| |
Collapse
|
17
|
Chen VL, Chen Y, Du X, Handelman SK, Speliotes EK. Genetic variants that associate with cirrhosis have pleiotropic effects on human traits. Liver Int 2020; 40:405-415. [PMID: 31815349 PMCID: PMC7395656 DOI: 10.1111/liv.14321] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 11/16/2019] [Accepted: 12/03/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND AND AIMS Cirrhosis is characterized by extensive fibrosis of the liver and is a major cause of liver-related mortality. Cirrhosis is partially heritable but genetic contributions to cirrhosis have not been systemically explored. Here, we carry out association analyses with cirrhosis in two large biobanks and determine the effects of cirrhosis associated variants on multiple human disease/traits. METHODS We carried out a genome-wide association analysis of cirrhosis as a diagnosis in UK BioBank (UKBB; 1088 cases vs. 407 873 controls) and then tested top-associating loci for replication with cirrhosis in a hospital-based cohort from the Michigan Genomics Initiative (MGI; 875 cases of cirrhosis vs. 30 346 controls). For replicating variants or variants previously associated with cirrhosis that also affected cirrhosis in UKBB or MGI, we determined single nucleotide polymorphism effects on all other diagnoses in UKBB (PheWAS), common metabolic traits/diseases and serum/plasma metabolites. RESULTS Unbiased genome-wide association study identified variants in/near PNPLA3 and HFE, and candidate variant analysis identified variants in/near TM6SF2, MBOAT7, SERPINA1, HSD17B13, STAT4 and IFNL4 that reproducibly affected cirrhosis. Most affected liver enzyme concentrations and/or aspartate transaminase-to-platelet ratio index. PheWAS, metabolic trait and serum/plasma metabolite association analyses revealed effects of these variants on lipid, inflammatory and other processes including new effects on many human diseases and traits. CONCLUSIONS We identified eight loci that reproducibly associate with population-based cirrhosis and define their diverse effects on human diseases and traits.
Collapse
Affiliation(s)
- Vincent L. Chen
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yanhua Chen
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Xiaomeng Du
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA
| | - Samuel K. Handelman
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Elizabeth K. Speliotes
- Division of Gastroenterology and Hepatology, University of Michigan Health System, Ann Arbor, MI, USA,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
18
|
Harro CC, Smedley RC, Buchweitz JP, Langlois DK. Hepatic copper and other trace mineral concentrations in dogs with hepatocellular carcinoma. J Vet Intern Med 2019; 33:2193-2199. [PMID: 31493348 PMCID: PMC6766484 DOI: 10.1111/jvim.15619] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Accepted: 08/28/2019] [Indexed: 12/14/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary liver tumor in dogs. Abnormalities in hepatic copper, iron, zinc, and selenium concentrations increase risk for HCC development in other species, but trace mineral concentrations have not been evaluated in dogs with HCC. Objectives To investigate hepatic trace mineral concentrations in dogs with HCC. Animals Archived liver specimens from 85 dogs with HCC and 85 control dogs. Methods Retrospective case‐control study. A histopathology database was searched to identify dogs with HCC (test population) and an age‐matched control population. Demographic information was retrieved, and H&E and rhodanine stained slides were reviewed for all cases. Copper, iron, zinc, and selenium concentrations were determined in noncancerous liver tissues (test and control population) and in HCC tissues (test population) using inductively coupled plasma mass spectrometry. Results Hepatic copper concentrations (non‐neoplastic hepatic tissue) were greater in test population dogs (median, IQR; 294.9 μg/g, 233.5‐475.9 μg/g) than in control dogs (202.8 μg/g, 135.0‐295.3 μg/g; P < .001). Hepatic zinc concentrations in test (132.1 μg/g,108.6‐163.2 μg/g) and control dogs (151.5 μg/g, 117.1‐184.5 μg/g) also were different (P = .03). Within test population dogs, all trace mineral concentrations were decreased in the HCC tissue as compared to the non‐neoplastic hepatic tissue (all P < .001). Conclusions and Clinical Importance Hepatic copper accumulation and other abnormalities in hepatic trace mineral concentrations could be involved in the pathogenesis of HCC in some dogs.
Collapse
Affiliation(s)
- Cailin C Harro
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Rebecca C Smedley
- Veterinary Diagnostic Laboratory, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - John P Buchweitz
- Department of Pathobiology and Diagnostic Investigation, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| | - Daniel K Langlois
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan
| |
Collapse
|
19
|
Abstract
Hereditary hemochromatosis (HH) is one of the most common genetic disorders among persons of northern European descent. There have been recent advances in the diagnosis, management, and treatment of HH. The availability of molecular diagnostic testing for HH has made possible confirmation of the diagnosis for most patients. Several genotype-phenotype correlation studies have clarified the differences in clinical features between patients with the C282Y homozygous genotypes and other HFE mutation patterns. The increasing use of noninvasive tests such as MRI T2* has made quantification of hepatic iron deposition easier and eliminated the need for liver biopsy in most patients. Serum ferritin of <1,000 ng/mL at diagnosis remains an important diagnostic test to identify patients with a low risk of advanced hepatic fibrosis and should be used routinely as part of the initial diagnostic evaluation. Genetic testing for other types of HH is available but is expensive and generally not useful in most clinical settings. Serum ferritin may be elevated among patients with nonalcoholic fatty liver disease and in those with alcoholic liver disease. These diagnoses are more common than HH among patients with elevated serum ferritin who are not C282Y homozygotes or C282Y/H63D compound heterozygotes. A secondary cause for liver disease should be excluded among patients with suspected iron overload who are not C282Y homozygotes. Phlebotomy remains the mainstay of therapy, but emerging novel therapies such as new chelating agents may have a role for selected patients.
Collapse
|
20
|
Excessive Reactive Iron Impairs Hematopoiesis by Affecting Both Immature Hematopoietic Cells and Stromal Cells. Cells 2019; 8:cells8030226. [PMID: 30857202 PMCID: PMC6468739 DOI: 10.3390/cells8030226] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 03/01/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Iron overload is the accumulation of excess iron in the body that may occur as a result of various genetic disorders or as a consequence of repeated blood transfusions. The surplus iron is then stored in the liver, pancreas, heart and other organs, which may lead to chronic liver disease or cirrhosis, diabetes and heart disease, respectively. In addition, excessive iron may impair hematopoiesis, although the mechanisms of this deleterious effect is not entirely known. In this study, we found that ferrous ammonium sulfate (FeAS), induced growth arrest and apoptosis in immature hematopoietic cells, which was mediated via reactive oxygen species (ROS) activation of p38MAPK and JNK pathways. In in vitro hematopoiesis derived from embryonic stem cells (ES cells), FeAS enhanced the development of dysplastic erythroblasts but inhibited their terminal differentiation; in contrast, it had little effect on the development of granulocytes, megakaryocytes, and B lymphocytes. In addition to its directs effects on hematopoietic cells, iron overload altered the expression of several adhesion molecules on stromal cells and impaired the cytokine production profile of these cells. Therefore, excessive iron would affect whole hematopoiesis by inflicting vicious effects on both immature hematopoietic cells and stromal cells.
Collapse
|
21
|
Karlsson M, Ekstedt M, Dahlström N, Forsgren MF, Ignatova S, Norén B, Dahlqvist Leinhard O, Kechagias S, Lundberg P. Liver R2* is affected by both iron and fat: A dual biopsy-validated study of chronic liver disease. J Magn Reson Imaging 2019; 50:325-333. [PMID: 30637926 DOI: 10.1002/jmri.26601] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 11/21/2018] [Accepted: 11/21/2018] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Liver iron content (LIC) in chronic liver disease (CLD) is currently determined by performing an invasive liver biopsy. MRI using R2* relaxometry is a noninvasive alternative for estimating LIC. Fat accumulation in the liver, or proton density fat fraction (PDFF), may be a possible confounder of R2* measurements. Previous studies of the effect of PDFF on R2* have not used quantitative LIC measurement. PURPOSE To assess the associations between R2*, LIC, PDFF, and liver histology in patients with suspected CLD. STUDY TYPE Prospective. POPULATION Eighty-one patients with suspected CLD. FIELD STRENGTH/SEQUENCE 1.5 T. Multiecho turbo field echo to quantify R2*. PRESS MRS to quantify PDFF. ASSESSMENT Each patient underwent an MR examination, followed by two needle biopsies immediately following the MR examination. The first biopsy was used for conventional histological assessment of LIC, whereas the second biopsy was used to quantitatively measure LIC using mass spectrometry. R2* was correlated with both LIC and PDFF. A correction for the influence of fat on R2* was calculated. STATISTICAL TESTS Pearson correlation, linear regression, and area under the receiver operating curve. RESULTS There was a positive linear correlation between R2* and PDFF (R = 0.69), after removing data from patients with elevated iron levels, as defined by LIC. R2*, corrected for PDFF, was the best method for identifying patients with elevated iron levels, with a correlation of R = 0.87 and a sensitivity and specificity of 87.5% and 98.6%, respectively. DATA CONCLUSION PDFF increases R2*. LEVEL OF EVIDENCE 2 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;50:325-333.
Collapse
Affiliation(s)
- Markus Karlsson
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Mattias Ekstedt
- Department of Gastroenterology and Hepatology, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Nils Dahlström
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Radiology, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Mikael F Forsgren
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Wolfram MathCore AB and Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Simone Ignatova
- Department of Clinical Pathology and Clinical Genetics, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Bengt Norén
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Olof Dahlqvist Leinhard
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden.,Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - Stergios Kechagias
- Department of Gastroenterology and Hepatology, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Peter Lundberg
- Center for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden.,Department of Radiation Physics, Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
22
|
|