1
|
de Melo Campos P, Toreli AC, de Albuquerque DM, Costa FF. Iron overload is not the same everywhere: Particularities of iron-metabolism gene mutations in Brazil and a proposal for the investigation and management of iron overload in this population. Hematol Transfus Cell Ther 2025; 47:103846. [PMID: 40378601 DOI: 10.1016/j.htct.2025.103846] [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: 02/05/2025] [Revised: 04/02/2025] [Accepted: 04/15/2025] [Indexed: 05/19/2025] Open
Abstract
There is no physiological mechanism for the excretion of iron in humans, and excess iron may lead to severe tissue damage if not adequately treated. Iron overload can be caused by genetic factors (hemochromatosis) or acquired conditions (e.g., ineffective erythropoiesis, transfusions, iatrogenic iron treatment, viral hepatitis, alcohol intake, severe liver disease, metabolic dysfunction), and, in many cases, by a conjunction of these factors. Historically, guidelines for the genetic investigation of patients with iron overload have been based on data obtained from Caucasian individuals in Europe and North America. However, due to the genetic heterogeneity of iron overload gene mutations worldwide, these recommendations might not be applicable to other ethnic groups. This study analyzed previously published genetic data obtained from Brazilian patients with iron overload and found a relevant but small prevalence of HFE C282Y/C282Y patients when compared to European populations, while mutations of the TFR2, SCL40A1, HJV, HAMP, BMP6 and SLC11A1 genes seem to be important. This study proposes an adapted algorithm for the investigation and management of iron overload in Brazil.
Collapse
Affiliation(s)
- Paula de Melo Campos
- Hemocentro, Universidade Estadual de Campinas - Unicamp, Campinas, São Paulo, Brazil.
| | - Ana Carolina Toreli
- Hemocentro, Universidade Estadual de Campinas - Unicamp, Campinas, São Paulo, Brazil
| | | | | |
Collapse
|
2
|
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
|
3
|
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
|
4
|
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
|
5
|
Uguen K, Ka C, Collod-Béroud G, Le Tertre M, Guellec J, Férec C, Béroud C, Callebaut I, Le Gac G. The Spectra of Disease-Causing Mutations in the Ferroportin 1 ( SLC40A1) Encoding Gene and Related Iron Overload Phenotypes (Hemochromatosis Type 4 and Ferroportin Disease). Hum Mutat 2023; 2023:5162256. [PMID: 40225168 PMCID: PMC11919020 DOI: 10.1155/2023/5162256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 05/17/2023] [Accepted: 05/26/2023] [Indexed: 04/15/2025]
Abstract
SLC40A1 is the sole iron export protein reported in mammals and is a key player in both cellular and systemic iron homeostasis. This unique iron exporter, which belongs to the major facilitator superfamily, is predominantly regulated by the hyposideremic hormone hepcidin. SLC40A1 dysfunction causes ferroportin disease, and autosomal dominant iron overload disorder characterized by cellular iron retention, principally in reticuloendothelial cells, correlating with high serum ferritin and low to normal transferrin saturation. Resistant to hepcidin, SLC40A1 mutations are rather associated with elevated plasma iron and parenchymal iron deposition, a condition that resembles HFE-related hemochromatosis and is associated with more clinical complications. With very few exceptions, only missense variations are reported at the SLC40A1 locus; this situation increasingly limits the establishment of pathogenicity. In this mutation update, we provide a comprehensive review of all the pathogenic or likely pathogenic variants, variants of unknown significance, and benign or likely benign SLC40A1 variants. The classification is essentially determined using functional, structural, segregation, and recurrence data. We furnish new information on genotype-phenotype correlations for loss-of-function, gain-of-function, and other SLC40A1 variants, confirming the existence of wide clinical heterogeneity and the potential for misdiagnosis. All information is recorded in a locus-specific online database.
Collapse
Affiliation(s)
- Kevin Uguen
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
| | - Chandran Ka
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Laboratory of Excellence GR-Ex, F-75015, France
| | | | - Marlène Le Tertre
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
| | - Julie Guellec
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Association Gaétan Saleün, F-29200, France
| | - Claude Férec
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Association Gaétan Saleün, F-29200, France
| | - Christophe Béroud
- Aix Marseille University, INSERM, Marseille Medical Genetics, F-13005, France
| | - Isabelle Callebaut
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS 7590, Institut de Minéralogie, de Physique des Matériaux et de Cosmochimie, IMPMC, F-75005, France
| | - Gérald Le Gac
- Univ Brest, Inserm, EFS, UMR1078, GGB F-29200, France
- CHRU de Brest, Service de Génétique Médicale et Biologie de la Reproduction, Laboratoire de Génétique Moléculaire et Histocompatibilité, F-29200, France
- Laboratory of Excellence GR-Ex, F-75015, France
| |
Collapse
|
6
|
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
|
7
|
A Novel Hepcidin Mutation. Transfus Clin Biol 2023:S1246-7820(23)00037-X. [PMID: 36925058 DOI: 10.1016/j.tracli.2023.03.001] [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: 02/04/2023] [Revised: 03/02/2023] [Accepted: 03/02/2023] [Indexed: 03/18/2023]
Abstract
BACKGROUND The bioactive peptide hormone hepcidin-25 regulates iron levels by inhibiting iron transport to plasma via ferroportin. Hepcidin-25 is synthesized in the liver where the 84 amino acids pro-hepcidin is cleaved into the bioactive hepcidin-25. A patient admitted to the hospital presented with infertility and fatigue. METHODS Genomic DNA was purified from whole blood using the Maxwell 16 system (Promega). MLPA analysis was performed to detect large genomic rearrangements using the SALSA MLPA kit # P347, Hemochromatosis (MRC Holland, Holland). Plasma hepcidin measurements were performed using liquid chromatography/tandem mass spectrometry (LC-MS/MS). RESULTS A novel HAMP mutation (homozygous one base deletion in c.215delG, p.Cys72Serfs*?) was detected. The deletion in nucleotide 215 causes a frameshift altering the predicted protein sequence from cysteine13 in mature peptide. Whether this leads to nonsense mediated decay of the mRNA or synthesis of an aberrant peptide in unknown, but bioactive hepcidin-25 was undetectable in plasma. The patient had massive iron overload with ferritin up to 8360 µg/L. He was anaemic with a Hb at 7.0 mmol/L (11.3 g/dL) and suffered from hypogonadotropic hypogonadism with a total testosterone of 1.2 nmol/l . Continued treatment with venesection and gonadotropins led to reduced fatigue, reduction in iron overload, a normalized Hb and improvement of semen quality. CONCLUSION A novel hepcidin mutation was detected in a patient with massive iron overload, fatigue and hypogonadotropic hypogonadism.
Collapse
|
8
|
DENND3 p.L708V activating variant is involved in the pathogenesis of hereditary hemochromatosis via the RAB12/TFR2 signaling pathway. Hepatol Int 2023; 17:648-661. [PMID: 36729283 DOI: 10.1007/s12072-022-10474-w] [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: 09/30/2022] [Accepted: 12/24/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE Pathogenic variants in HFE and non-HFE genes have been identified in hereditary hemochromatosis (HH) in different patient populations, but there are still a considerable proportion of patients with unexplained primary iron overload. We recently identified in Chinese patients with unexplained primary iron overload a recurrent p.L708V variant in the differentially expressed in normal and neoplastic cells domain 3 (DENND3) gene, functioning as a guanine nucleotide exchange factor for small GTpase Rab12 which down-regulates TfR expression in mice. We aim to investigate the pathogenicity and the underlying mechanism of the DENND3 p.L708V variant in HH patients. METHODS Patients with primary iron overload were analyzed for DENND3 p.L708V. TFR2 and hepcidin expression in livers were examined in HH patients harboring DENND3 p.L708V. The effects of DENND3 p.L708V on RAB12/TFR2 and downstream iron metabolic pathways were investigated in vitro and in vivo. RESULTS Six of 31 patients with HH (19.35%) harbored the DENND3 p.L708V variant. The expression of TFR2 and hepcidin was decreased in the liver of HH patients with DENND3 p.L708V. Cells transfected with the DENND3 p.L708V vector showed up-regulation of RAB12 expression and TFR2 degradation in lysosomes, and down-regulation of the pSMAD1/5 and hepcidin. Mice models infected with adeno-associated virus expressing DENND3 p.L708V variant showed higher total serum iron concentrations and decreased HAMP level, increased amount of iron accumulation and the down-regulated of TFR2 expression in the liver. CONCLUSIONS The DENND3 p.L708V activating variant down-regulates hepcidin expression through the DENND3/RAB12/TFR2 axis, which may represent a potential novel pathogenic factor of HH.
Collapse
|
9
|
Girelli D, Busti F, Brissot P, Cabantchik I, Muckenthaler MU, Porto G, on behalf of the Nomenclature Committee of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society). Hemochromatosis classification: update and recommendations by the BIOIRON Society. Blood 2022; 139:3018-3029. [PMID: 34601591 PMCID: PMC11022970 DOI: 10.1182/blood.2021011338] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 09/05/2021] [Indexed: 12/15/2022] Open
Abstract
Hemochromatosis (HC) is a genetically heterogeneous disorder in which uncontrolled intestinal iron absorption may lead to progressive iron overload (IO) responsible for disabling and life-threatening complications such as arthritis, diabetes, heart failure, hepatic cirrhosis, and hepatocellular carcinoma. The recent advances in the knowledge of pathophysiology and molecular basis of iron metabolism have highlighted that HC is caused by mutations in at least 5 genes, resulting in insufficient hepcidin production or, rarely, resistance to hepcidin action. This has led to an HC classification based on different molecular subtypes, mainly reflecting successive gene discovery. This scheme was difficult to adopt in clinical practice and therefore needs revision. Here we present recommendations for unambiguous HC classification developed by a working group of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society), including both clinicians and basic scientists during a meeting in Heidelberg, Germany. We propose to deemphasize the use of the molecular subtype criteria in favor of a classification addressing both clinical issues and molecular complexity. Ferroportin disease (former type 4a) has been excluded because of its distinct phenotype. The novel classification aims to be of practical help whenever a detailed molecular characterization of HC is not readily available.
Collapse
Affiliation(s)
- Domenico Girelli
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Fabiana Busti
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
| | - Pierre Brissot
- INSERM, Univ-Rennes, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1241, Institut NuMeCan, Rennes, France
| | - Ioav Cabantchik
- Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
| | - Martina U. Muckenthaler
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
| | - Graça Porto
- Institute for Molecular and Cell Biology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Clinical Hematology, Santo António Hospital, Porto University, Porto, Portugal
| | - on behalf of the Nomenclature Committee of the International Society for the Study of Iron in Biology and Medicine (BIOIRON Society)
- Department of Medicine, Section of Internal Medicine, EuroBloodNet Center, University of Verona and Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy
- INSERM, Univ-Rennes, Institut National de la Recherche Agronomique, Unité Mixte de Recherche 1241, Institut NuMeCan, Rennes, France
- Alexander Silberman Institute of Life Sciences, Hebrew University, Jerusalem, Israel
- Department of Pediatric Oncology, Hematology, and Immunology and Molecular Medicine Partnership Unit, University of Heidelberg, Heidelberg, Germany
- Molecular Medicine Partnership Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Translational Lung Research Center, German Center for Lung Research, Heidelberg, Germany
- German Centre for Cardiovascular Research, Partner Site Heidelberg, Mannheim, Germany
- Institute for Molecular and Cell Biology, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Clinical Hematology, Santo António Hospital, Porto University, Porto, Portugal
| |
Collapse
|
10
|
Cancado RD, Alvarenga AM, Santos PCJ. HFE hemochromatosis: an overview about therapeutic recommendations. Hematol Transfus Cell Ther 2021; 44:95-99. [PMID: 34824033 PMCID: PMC8885398 DOI: 10.1016/j.htct.2021.06.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 06/12/2021] [Accepted: 06/24/2021] [Indexed: 11/17/2022] Open
Abstract
Hemochromatosis is currently characterized by the iron overload caused by hepcidin deficiency. Large advances in the knowledge on the hemochromatosis pathophysiology have occurred due to a better understanding of the protein of the iron metabolism, the genetic basis of hemochromatosis and of other iron overload diseases or conditions which can lead to this phenotype. In the present review, the main aims are to show updates on hemochromatosis and to report a practical set of therapeutic recommendations for the human factors engineering protein (HFE) hemochromatosis for the p.Cys282Tyr (C282Y/C282Y) homozygous genotype, elaborated by the Haemochromatosis International Taskforce.
Collapse
Affiliation(s)
- Rodolfo D Cancado
- Irmandade da Santa Casa de Misericórdia de São Paulo, Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP, Brazil
| | | | - Paulo Caleb Jl Santos
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
11
|
Kang W, Barad A, Clark AG, Wang Y, Lin X, Gu Z, O'Brien KO. Ethnic Differences in Iron Status. Adv Nutr 2021; 12:1838-1853. [PMID: 34009254 PMCID: PMC8483971 DOI: 10.1093/advances/nmab035] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/02/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023] Open
Abstract
Iron is unique among all minerals in that humans have no regulatable excretory pathway to eliminate excess iron after it is absorbed. Iron deficiency anemia occurs when absorbed iron is not sufficient to meet body iron demands, whereas iron overload and subsequent deposition of iron in key organs occur when absorbed iron exceeds body iron demands. Over time, iron accumulation in the body can increase risk of chronic diseases, including cirrhosis, diabetes, and heart failure. To date, only ∼30% of the interindividual variability in iron absorption can be captured by iron status biomarkers or iron regulatory hormones. Much of the regulation of iron absorption may be under genetic control, but these pathways have yet to be fully elucidated. Genome-wide and candidate gene association studies have identified several genetic variants that are associated with variations in iron status, but the majority of these data were generated in European populations. The purpose of this review is to summarize genetic variants that have been associated with alterations in iron status and to highlight the influence of ethnicity on the risk of iron deficiency or overload. Using extant data in the literature, linear mixed-effects models were constructed to explore ethnic differences in iron status biomarkers. This approach found that East Asians had significantly higher concentrations of iron status indicators (serum ferritin, transferrin saturation, and hemoglobin) than Europeans, African Americans, or South Asians. African Americans exhibited significantly lower hemoglobin concentrations compared with other ethnic groups. Further studies of the genetic basis for ethnic differences in iron metabolism and on how it affects disease susceptibility among different ethnic groups are needed to inform population-specific recommendations and personalized nutrition interventions for iron-related disorders.
Collapse
Affiliation(s)
- Wanhui Kang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Alexa Barad
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Andrew G Clark
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA,Department of Computational Biology, Cornell University, Ithaca, NY, USA
| | - Yiqin Wang
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | - Xu Lin
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, China
| | - Zhenglong Gu
- Division of Nutritional Sciences, Cornell University, Ithaca, NY, USA
| | | |
Collapse
|
12
|
Noé V, Aubets E, Félix AJ, Ciudad CJ. Nucleic acids therapeutics using PolyPurine Reverse Hoogsteen hairpins. Biochem Pharmacol 2020; 189:114371. [PMID: 33338475 DOI: 10.1016/j.bcp.2020.114371] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/10/2020] [Accepted: 12/11/2020] [Indexed: 02/08/2023]
Abstract
PolyPurine Reverse Hoogsteen hairpins (PPRHs) are DNA hairpins formed by intramolecular reverse Hoogsteen bonds which can bind to polypyrimidine stretches in dsDNA by Watson:Crick bonds, thus forming a triplex and displacing the fourth strand of the DNA complex. PPRHs were first described as a gene silencing tool in vitro for DHFR, telomerase and survivin genes. Then, the effect of PPRHs directed against the survivin gene was also determined in vivo using a xenograft model of prostate cancer cells (PC3). Since then, the ability of PPRHs to inhibit gene expression has been explored in other genes involved in cancer (BCL-2, mTOR, topoisomerase, C-MYC and MDM2), in immunotherapy (SIRPα/CD47 and PD-1/PD-L1 tandem) or in replication stress (WEE1 and CHK1). Furthermore, PPRHs have the ability to target the complementary strand of a G-quadruplex motif as a regulatory element of the TYMS gene. PPRHs have also the potential to correct point mutations in the DNA as shown in two collections of CHO cell lines bearing mutations in either the dhfr or aprt loci. Finally, based on the capability of PPRHs to form triplexes, they have been incorporated as probes in biosensors for the determination of the DNA methylation status of PAX-5 in cancer and the detection of mtLSU rRNA for the diagnosis of Pneumocystis jirovecii. Of note, PPRHs have high stability and do not present immunogenicity, hepatotoxicity or nephrotoxicity in vitro. Overall, PPRHs constitute a new economical biotechnological tool with multiple biomedical applications.
Collapse
Affiliation(s)
- Véronique Noé
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, & IN2UB, University of Barcelona, 08028 Barcelona, Spain
| | - Eva Aubets
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, & IN2UB, University of Barcelona, 08028 Barcelona, Spain
| | - Alex J Félix
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, & IN2UB, University of Barcelona, 08028 Barcelona, Spain
| | - Carlos J Ciudad
- Department of Biochemistry and Physiology, School of Pharmacy and Food Sciences, & IN2UB, University of Barcelona, 08028 Barcelona, Spain.
| |
Collapse
|
13
|
Alvarenga AM, da Silva NK, Fonseca PFS, Oliveira TGM, da Silva Monteiro JB, Cançado RD, Naoum FA, Dinardo CL, Brissot P, Santos PCJL. Novel mutations in the bone morphogenetic protein 6 gene in patients with iron overload and non-homozygous genotype for the HFE p.Cys282Tyr mutation. Blood Cells Mol Dis 2020; 84:102444. [PMID: 32464486 DOI: 10.1016/j.bcmd.2020.102444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 05/03/2020] [Accepted: 05/04/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Five main genes are associated with hemochromatosis; however, current studies show that, in addition to these genes, others may be associated with primary iron overload (IO). One of these is the bone morphogenetic protein 6 (BMP6), which encodes a protein that modulates hepcidin synthesis and, consequently, iron homeostasis. AIM To identify BMP6 gene pathogenic variants in patients with IO and non-homozygous genotype for the HFE p.Cys282Tyr mutation. MATERIALS AND METHODS Fifty-three patients with primary IO and non-homozygous genotype for the HFE p.Cys282Tyr were selected. Subsequent bidirectional DNA sequencing of BMP6 exons was performed. RESULTS Two novel variants were found. One at homozygous state p.Gln158Ter (c.472C>T) was pathogenic, the other one at heterozygous state p.Val394Met (c.1180G>A) was of uncertain significance (VUS); the third variant at heterozygous state p.Arg257His (c.770G>A) has already been described and associated with IO. No BMP6 pathogenic variants that would explain iron overload phenotypes were detected in 94% of the studied patients. CONCLUSION Identification of the BMP6 pathogenic variants in Brazilian patients with primary IO might contribute to the genetic understanding of this phenotype.
Collapse
Affiliation(s)
- Aline Morgan Alvarenga
- Department of Pharmacology - Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, Brazil.
| | - Nathália Kozikas da Silva
- Department of Pharmacology - Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, Brazil.
| | - Paula Fernanda Silva Fonseca
- Department of Pharmacology - Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, Brazil.
| | - Theo G M Oliveira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil.
| | | | | | | | - Carla Luana Dinardo
- Fundação Pró-Sangue, Hemocentro de São Paulo, São Paulo, SP, Brazil; Universidade de São Paulo (USP), São Paulo, SP, Brazil.
| | - Pierre Brissot
- Institut NuMeCan, Inserm U-1241, Univ Rennes 1, Rennes, France.
| | - Paulo Caleb Junior Lima Santos
- Department of Pharmacology - Escola Paulista de Medicina, Universidade Federal de São Paulo (EPM-Unifesp), São Paulo, Brazil.
| |
Collapse
|
14
|
Vlasveld LT, Janssen R, Bardou-Jacquet E, Venselaar H, Hamdi-Roze H, Drakesmith H, Swinkels DW. Twenty Years of Ferroportin Disease: A Review or An Update of Published Clinical, Biochemical, Molecular, and Functional Features. Pharmaceuticals (Basel) 2019; 12:ph12030132. [PMID: 31505869 PMCID: PMC6789780 DOI: 10.3390/ph12030132] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/14/2019] [Accepted: 08/20/2019] [Indexed: 12/14/2022] Open
Abstract
Iron overloading disorders linked to mutations in ferroportin have diverse phenotypes in vivo, and the effects of mutations on ferroportin in vitro range from loss of function (LOF) to gain of function (GOF) with hepcidin resistance. We reviewed 359 patients with 60 ferroportin variants. Overall, macrophage iron overload and low/normal transferrin saturation (TSAT) segregated with mutations that caused LOF, while GOF mutations were linked to high TSAT and parenchymal iron accumulation. However, the pathogenicity of individual variants is difficult to establish due to the lack of sufficiently reported data, large inter-assay variability of functional studies, and the uncertainty associated with the performance of available in silico prediction models. Since the phenotypes of hepcidin-resistant GOF variants are indistinguishable from the other types of hereditary hemochromatosis (HH), these variants may be categorized as ferroportin-associated HH, while the entity ferroportin disease may be confined to patients with LOF variants. To further improve the management of ferroportin disease, we advocate for a global registry, with standardized clinical analysis and validation of the functional tests preferably performed in human-derived enterocytic and macrophagic cell lines. Moreover, studies are warranted to unravel the definite structure of ferroportin and the indispensable residues that are essential for functionality.
Collapse
Affiliation(s)
- L Tom Vlasveld
- Department of Internal Medicine, Haaglanden MC-Bronovo, 2597AX The Hague, The Netherlands
| | - Roel Janssen
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Edouard Bardou-Jacquet
- Liver Diseases Department, French Reference Centre for Rare Iron Overload Diseases of Genetic Origin, University Hospital Pontchaillou, 35033 Rennes, France
| | - Hanka Venselaar
- Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud, University Medical Center, P.O. Box 9191, 6500 HB Nijmegen, The Netherlands
| | - Houda Hamdi-Roze
- Molecular Genetics Department, French Reference Centre for Rare Iron Overload Diseases of Genetic Origin, University Hospital Pontchaillou, 35033 Rennes, France
| | - Hal Drakesmith
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX39DS, UK
| | - Dorine W Swinkels
- Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands.
| |
Collapse
|
15
|
Kong X, Xie L, Zhu H, Song L, Xing X, Yang W, Chen X. Genotypic and phenotypic spectra of hemojuvelin mutations in primary hemochromatosis patients: a systematic review. Orphanet J Rare Dis 2019; 14:171. [PMID: 31286966 PMCID: PMC6615163 DOI: 10.1186/s13023-019-1097-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Accepted: 05/19/2019] [Indexed: 12/12/2022] Open
Abstract
Hereditary hemochromatosis (HH) is a genetic disorder that causes excess absorption of iron and can lead to a variety of complications including liver cirrhosis, arthritis, abnormal skin pigmentation, cardiomyopathy, hypogonadism, and diabetes. Hemojuvelin (HJV) is the causative gene of a rare subtype of HH worldwide. This study aims to systematically review the genotypic and phenotypic spectra of HJV-HH in multiple ethnicities, and to explore the genotype-phenotype correlations. A comprehensive search of PubMed database was conducted. Data were extracted from 57 peer-reviewed original articles including 132 cases with HJV-HH of multiple ethnicities, involving 117 biallelic cases and 15 heterozygotes. Among the biallelic cases, male and female probands of Caucasian ancestry were equally affected, whereas males were more often affected among East Asians (P=1.72×10-2). Hepatic iron deposition and hypogonadism were the most frequently reported complications. Hypogonadism and arthropathy were more prevalent in Caucasians than in East Asians (P=9.30×10-3, 1.69×10-2). Among the recurrent mutations, G320V (45 unrelated cases) and L101P (7 unrelated cases) were detected most frequently and restricted to Caucasians. [Q6H; C321*] was predominant in Chinese patients (6 unrelated cases). I281T (Chinese and Greek), A310G (Brazilian and African American), and R385* (Italian and North African) were reported across different ethnicities. In genotype-phenotype correlation analyses, 91.30% of homozygotes with exon 2-3 mutations developed early-onset HH compared to 66.00% of those with exon 4 mutations (P=2.40×10-2). Hypogonadism occurred more frequently in homozygotes with missense mutations (72.55%) than in those with nonsense mutations (35.71%; P=2.43×10-2). Liver biopsy was accepted by more probands with frame-shift or missense mutations (85.71% and 60.78%, respectively) than by those with nonsense mutations (28.57%; P=2.37×10-2, 3.93×10-2). The present review suggests that patients' ethnicity, geographical region, and genetic predisposition should be considered in the diagnosis, prognosis and management of HJV-HH.
Collapse
Affiliation(s)
- Xiaomu Kong
- Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029 China
| | - Lingding Xie
- Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029 China
| | - Haiqing Zhu
- Department of Endocrinology and Metabolism, China Meitan General Hospital, No. 29 Xibahe Nanli, Chaoyang District, Beijing, 100029 China
| | - Lulu Song
- Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029 China
| | - Xiaoyan Xing
- Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029 China
| | - Wenying Yang
- Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029 China
| | - Xiaoping Chen
- Department of Endocrinology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing, 100029 China
| |
Collapse
|
16
|
Lv T, Zhang W, Xu A, Li Y, Zhou D, Zhang B, Li X, Zhao X, Wang Y, Wang X, Duan W, Wang Q, Xu H, Zheng J, Zhao R, Zhu L, Dong Y, Lu L, Chen Y, Long J, Zheng S, Wang W, You H, Jia J, Ou X, Huang J. Non- HFE mutations in haemochromatosis in China: combination of heterozygous mutations involving HJV signal peptide variants. J Med Genet 2018; 55:650-660. [PMID: 30166352 DOI: 10.1136/jmedgenet-2018-105348] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Revised: 06/22/2018] [Accepted: 07/08/2018] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Hereditary haemochromatosis (HH) caused by a homozygous p.C282Y mutation in haemochromatosis (HFE) gene has been well documented. However, less is known about the causative non-HFE mutation. We aimed to assess mutation patterns of haemochromatosis-related genes in Chinese patients with primary iron overload. METHODS Patients were preanalysed for mutations in the classic HH-related genes: HFE, HJV, HAMP, TFR2 and SLC40A1. Whole exome sequencing was conducted for cases with variants in HJV signal peptide region. Representative variants were analysed for biological function. RESULTS None of the cases analysed harboured the HFE p.C282Y; however, 21 of 22 primary iron-overload cases harboured at least one non-synonymous variant in the non-HFE genes. Specifically, p.E3D or p.Q6H variants in the HJV signal peptide region were identified in nine cases (40.9%). In two of three probands with the HJV p.E3D, exome sequencing identified accompanying variants in BMP/SMAD pathway genes, including TMPRSS6 p.T331M and BMP4 p.R269Q, and interestingly, SUGP2 p.R639Q was identified in all the three cases. Pedigree analysis showed a similar pattern of combination of heterozygous mutations in cases with HJV p.E3D or p.Q6H, with SUGP2 p.R639Q or HJV p.C321X being common mutation. In vitro siRNA interference of SUGP2 showed a novel role of downregulating the BMP/SMAD pathway. Site-directed mutagenesis of HJV p.Q6H/p.C321X in cell lines resulted in loss of membrane localisation of mutant HJV, and downregulation of p-SMAD1/5 and HAMP. CONCLUSION Compound heterozygous mutations of HJV or combined heterozygous mutations of BMP/SMAD pathway genes, marked by HJV variants in the signal peptide region, may represent a novel aetiological factor for HH.
Collapse
Affiliation(s)
- Tingxia Lv
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Wei Zhang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Anjian Xu
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yanmeng Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Donghu Zhou
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Bei Zhang
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaojin Li
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xinyan Zhao
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Yu Wang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaoming Wang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Weijia Duan
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Qianyi Wang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Hexiang Xu
- Department of Infectious Diseases, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Hefei, China
| | - JiShun Zheng
- Department of Infectious Diseases, The Third Affiliated Hospital of Anhui Medical University, The First People's Hospital of Hefei, Hefei, China
| | - Rongrong Zhao
- Department of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Longdong Zhu
- Department of Infectious Diseases, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuwei Dong
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Lungen Lu
- Department of Gastroenterology, School of Medicine, Shanghai General Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Yongpeng Chen
- Department of Infectious Diseases, Institute of Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiang Long
- Department of Oncology Minimally Invasive Interventional Radiology, Beijing You-An Hospital, Capital Medical University, Shanghai, China
| | - Sujun Zheng
- Artificial Liver Center, Beijing You-An Hospital, Capital Medical University, Beijing, China
| | - Wei Wang
- Department of Clinical Laboratory, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hong You
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jidong Jia
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Xiaojuan Ou
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jian Huang
- Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Experimental Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- Clinical Research Center for Rare Liver Disease, Capital Medical University, Beijing, China
- Liver Research Center, National Clinical Research Center for Digestive Diseases, Beijing, China
| |
Collapse
|
17
|
Phenotypic analysis of hemochromatosis subtypes reveals variations in severity of iron overload and clinical disease. Blood 2018; 132:101-110. [PMID: 29743178 DOI: 10.1182/blood-2018-02-830562] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 05/03/2018] [Indexed: 12/15/2022] Open
Abstract
The clinical progression of HFE-related hereditary hemochromatosis (HH) and its phenotypic variability has been well studied. Less is known about the natural history of non-HFE HH caused by mutations in the HJV, HAMP, or TFR2 genes. The purpose of this study was to compare the phenotypic and clinical presentations of hepcidin-deficient forms of HH. A literature review of all published cases of genetically confirmed HJV, HAMP, and TFR2 HH was performed. Phenotypic and clinical data from a total of 156 patients with non-HFE HH was extracted from 53 publications and compared with data from 984 patients with HFE-p.C282Y homozygous HH from the QIMR Berghofer Hemochromatosis Database. Analyses confirmed that non-HFE forms of HH have an earlier age of onset and a more severe clinical course than HFE HH. HJV and HAMP HH are phenotypically and clinically very similar and have the most severe presentation, with cardiomyopathy and hypogonadism being particularly prevalent findings. TFR2 HH is more intermediate in its age of onset and severity. All clinical outcomes analyzed were more prevalent in the juvenile forms of HH, with the exception of arthritis and arthropathy, which were more commonly seen in HFE HH. This is the first comprehensive analysis comparing the different phenotypic and clinical aspects of the genetic forms of HH, and the results will be valuable for the differential diagnosis and management of these conditions. Importantly, our analyses indicate that factors other than iron overload may be contributing to joint pathology in patients with HFE HH.
Collapse
|
18
|
Fonseca PFS, Cançado RD, Naoum FA, Dinardo CL, Fonseca GHH, Gualandro SFM, Krieger JE, Pereira AC, Brissot P, Santos PCJL. Quality of life scores differs between genotypic groups of patients with suspected hereditary hemochromatosis. BMC MEDICAL GENETICS 2018; 19:3. [PMID: 29301508 PMCID: PMC5755339 DOI: 10.1186/s12881-017-0513-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 12/18/2017] [Indexed: 12/15/2022]
Abstract
Background Hereditary hemochromatosis (HH) encompasses a group of autosomal recessive disorders mainly characterized by enhanced intestinal absorption of iron and its accumulation in parenchymal organs. HH diagnosis is based on iron biochemical and magnetic resonance imaging (MRI) assessment, and genetic testing. Questionnaires, such as SF-36 (short form health survey), have been increasingly used to assess the impact of diseases on the patient’s quality of life (QL). In addition, different genotypes are identified as results of genetic tests in patients with suspected primary iron overload. In the present study, our aim was to evaluate whether domains of QL are different according to genotypic groups in patients suspected of HH. Methods Seventy-nine patients with primary iron overload were included and two genotypic groups were formed (group 1: homozygous genotype for the HFE p.Cys282Tyr mutation; group 2: other genotypes). Results Group 1 had higher means of plasma transferrin saturation (86 ± 19%) and serum ferritin (1669 ± 1209 ng/mL) compared to group 2 (71 ± 12%, 1252 ± 750 ng/mL, respectively; p = 0.001). Four domains were significantly different among groups 1 and 2: physical functioning (p = 0.03), bodily pain (p = 0.03), vitality (p = 0.02) and social functioning (p = 0.01). Conclusions Our main finding was that patients with p.Cys282Tyr homozygosity had a worse QL scenario assessed by SF-36, compared with patients with iron overload without the same genotype. Being aware of this relationship between genotypes and QL might be helpful in the overall management of patients suspected of hereditary hemochromatosis. Electronic supplementary material The online version of this article (doi: 10.1186/s12881-017-0513-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paula Fernanda Silva Fonseca
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, Av. Doutor Enéas de Carvalho Aguiar, 44-Cerqueira César, São Paulo, 05403 900, Brazil
| | | | | | - Carla Luana Dinardo
- Fundação Pró-Sangue, Hemocentro de São Paulo, São Paulo, SP, Brazil.,Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | | | - Sandra Fatima Menosi Gualandro
- Hematology and Hemotherapy Discipline, Hospital das Clinicas, Medical School, University of São Paulo, São Paulo, Brazil
| | - José Eduardo Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, Av. Doutor Enéas de Carvalho Aguiar, 44-Cerqueira César, São Paulo, 05403 900, Brazil
| | - Alexandre Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, Av. Doutor Enéas de Carvalho Aguiar, 44-Cerqueira César, São Paulo, 05403 900, Brazil
| | - Pierre Brissot
- Liver Disease Unit, Pontchaillou University Hospital, University of Rennes, and National Reference Centre for Rare Iron Overload Diseases of Genetic Origin, Rennes, France
| | - Paulo Caleb Junior Lima Santos
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, Av. Doutor Enéas de Carvalho Aguiar, 44-Cerqueira César, São Paulo, 05403 900, Brazil. .,Department of Pharmacology, Universidade Federal de Sao Paulo - UNIFESP, São Paulo, Brazil.
| |
Collapse
|
19
|
Structure-function analysis of ferroportin defines the binding site and an alternative mechanism of action of hepcidin. Blood 2017; 131:899-910. [PMID: 29237594 DOI: 10.1182/blood-2017-05-786590] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023] Open
Abstract
Nonclassical ferroportin disease (FD) is a form of hereditary hemochromatosis caused by mutations in the iron transporter ferroportin (Fpn), resulting in parenchymal iron overload. Fpn is regulated by the hormone hepcidin, which induces Fpn endocytosis and cellular iron retention. We characterized 11 clinically relevant and 5 nonclinical Fpn mutations using stably transfected, inducible isogenic cell lines. All clinical mutants were functionally resistant to hepcidin as a consequence of either impaired hepcidin binding or impaired hepcidin-dependent ubiquitination despite intact hepcidin binding. Mapping the residues onto 2 computational models of the human Fpn structure indicated that (1) mutations that caused ubiquitination-resistance were positioned at helix-helix interfaces, likely preventing the hepcidin-induced conformational change, (2) hepcidin binding occurred within the central cavity of Fpn, (3) hepcidin interacted with up to 4 helices, and (4) hepcidin binding should occlude Fpn and interfere with iron export independently of endocytosis. We experimentally confirmed hepcidin-mediated occlusion of Fpn in the absence of endocytosis in multiple cellular systems: HEK293 cells expressing an endocytosis-defective Fpn mutant (K8R), Xenopus oocytes expressing wild-type or K8R Fpn, and mature human red blood cells. We conclude that nonclassical FD is caused by Fpn mutations that decrease hepcidin binding or hinder conformational changes required for ubiquitination and endocytosis of Fpn. The newly documented ability of hepcidin and its agonists to occlude iron transport may facilitate the development of broadly effective treatments for hereditary iron overload disorders.
Collapse
|
20
|
Lescano MA, Tavares LC, Santos PCJL. Juvenile hemochromatosis: HAMP mutation and severe iron overload treated with phlebotomies and deferasirox. World J Clin Cases 2017; 5:381-383. [PMID: 29085829 PMCID: PMC5649000 DOI: 10.12998/wjcc.v5.i10.381] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/23/2017] [Accepted: 08/02/2017] [Indexed: 02/05/2023] Open
Abstract
Juvenile hemochromatosis (JH) is a rare condition classified as an autosomal recessive disorder that leads to severe iron absorption. JH usually affects people under the age of 30 and presents symptoms such as chronic liver damage, hypogonadotropic hypogonadism, cardiac diseases and endocrine dysfunctions. The present case reports a 29-year-old Brazilian woman with JH condition due to HAMP mutation (g.47G>A), treated with phlebotomies and deferasirox. She presented symptoms such as weakness, skin hyperpigmentation, joint pain in the shoulders and hands and amenorrhea. First laboratory tests showed altered biochemical parameters [serum ferritin (SF): 5696 ng/mL, transferrin saturation (TS): 85%]. After sessions of phlebotomies (450 mL every 15 d), the patient presented partial symptomatic improvements and biochemical parameters (SF: 1000 ng/mL, Hb: 11 g/dL). One year later, deferasirox (15 mg/kg per day) was introduced to the treatment, and the patient showed total symptomatic improvement, with significant clearing of the skin, SF: 169 ng/mL, and TS: 50%. Furthermore, after the combined deferasirox-phlebotomy therapy, magnetic resonance imaging measurements revealed normalized level for liver iron (30 μmol/g; reference value < 36 μmol/g). In conclusion, combined deferasirox-phlebotomy treatment was able to normalize iron levels and improve symptoms.
Collapse
Affiliation(s)
- Manuel A Lescano
- Institute of Digestive Tract of Southwestern Bahia, Bahia, BA 45023-145, Brazil
| | - Letícia C Tavares
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School, São Paulo, SP 05403-900, Brazil
| | - Paulo C J L Santos
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School, São Paulo, SP 05403-900, Brazil
| |
Collapse
|
21
|
Xu A, Li Y, Chen W, Li X, Zhang W, Li S, Wu L, Wu Z, Zhang B, Ou X, Huang J. "Haemochromatotic" characteristics of the human BEL-7402 cell line. Br J Haematol 2017; 183:302-306. [PMID: 28961311 DOI: 10.1111/bjh.14952] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Anjian Xu
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Research Centre for Rare Liver Diseases, Capital Medical University, Beijing, China.,National Clinical Research Centre for Digestive Diseases, Beijing, China
| | - Yanmeng Li
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wei Chen
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojin Li
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Research Centre for Rare Liver Diseases, Capital Medical University, Beijing, China
| | - Wei Zhang
- Clinical Research Centre for Rare Liver Diseases, Capital Medical University, Beijing, China.,Liver Research Centre, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
| | - Siwen Li
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Lina Wu
- Liver Research Centre, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
| | - Zhen Wu
- Liver Research Centre, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
| | - Bei Zhang
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaojuan Ou
- Clinical Research Centre for Rare Liver Diseases, Capital Medical University, Beijing, China.,National Clinical Research Centre for Digestive Diseases, Beijing, China.,Liver Research Centre, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
| | - Jian Huang
- Experimental Centre, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Clinical Research Centre for Rare Liver Diseases, Capital Medical University, Beijing, China.,National Clinical Research Centre for Digestive Diseases, Beijing, China.,Liver Research Centre, Beijing Friendship Hospital, Capital Medical University; Beijing Key Laboratory of Translational Medicine on Liver Cirrhosis, Beijing, China
| |
Collapse
|
22
|
Ikuta K, Hatayama M, Addo L, Toki Y, Sasaki K, Tatsumi Y, Hattori A, Kato A, Kato K, Hayashi H, Suzuki T, Kobune M, Tsutsui M, Gotoh A, Aota Y, Matsuura M, Hamada Y, Tokuda T, Komatsu N, Kohgo Y. Iron overload patients with unknown etiology from national survey in Japan. Int J Hematol 2016; 105:353-360. [DOI: 10.1007/s12185-016-2141-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/06/2016] [Accepted: 11/06/2016] [Indexed: 12/21/2022]
|
23
|
Next-generation sequencing of hereditary hemochromatosis-related genes: Novel likely pathogenic variants found in the Portuguese population. Blood Cells Mol Dis 2016; 61:10-5. [PMID: 27667161 DOI: 10.1016/j.bcmd.2016.07.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 02/07/2023]
Abstract
Hereditary hemochromatosis (HH) is an autosomal recessive disorder characterized by excessive iron absorption resulting in pathologically increased body iron stores. It is typically associated with common HFE gene mutation (p.Cys282Tyr and p.His63Asp). However, in Southern European populations up to one third of HH patients do not carry the risk genotypes. This study aimed to explore the use of next-generation sequencing (NGS) technology to analyse a panel of iron metabolism-related genes (HFE, TFR2, HJV, HAMP, SLC40A1, and FTL) in 87 non-classic HH Portuguese patients. A total of 1241 genetic alterations were detected corresponding to 53 different variants, 13 of which were not described in the available public databases. Among them, five were predicted to be potentially pathogenic: three novel mutations in TFR2 [two missense (p.Leu750Pro and p.Ala777Val) and one intronic splicing mutation (c.967-1G>C)], one missense mutation in HFE (p.Tyr230Cys), and one mutation in the 5'-UTR of HAMP gene (c.-25G>A). The results reported here illustrate the usefulness of NGS for targeted iron metabolism-related gene panels, as a likely cost-effective approach for molecular genetics diagnosis of non-classic HH patients. Simultaneously, it has contributed to the knowledge of the pathophysiology of those rare iron metabolism-related disorders.
Collapse
|
24
|
Badar S, Busti F, Ferrarini A, Xumerle L, Bozzini P, Capelli P, Pozzi-Mucelli R, Campostrini N, De Matteis G, Marin Vargas S, Giorgetti A, Delledonne M, Olivieri O, Girelli D. Identification of novel mutations in hemochromatosis genes by targeted next generation sequencing in Italian patients with unexplained iron overload. Am J Hematol 2016; 91:420-5. [PMID: 26799139 DOI: 10.1002/ajh.24304] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/08/2016] [Accepted: 01/13/2016] [Indexed: 12/14/2022]
Abstract
Hereditary hemochromatosis, one of the commonest genetic disorder in Caucasians, is mainly associated to homozygosity for the C282Y mutation in the HFE gene, which is highly prevalent (allele frequency up to near 10% in Northern Europe) and easily detectable through a widely available "first level" molecular test. However, in certain geographical regions like the Mediterranean area, up to 30% of patients with a HH phenotype has a negative or non-diagnostic (i.e. simple heterozygosity) test, because of a known heterogeneity involving at least four other genes (HAMP, HJV, TFR2, and SLC40A1). Mutations in such genes are generally rare/private, making the diagnosis of atypical HH essentially a matter of exclusion in clinical practice (from here the term of "non-HFE" HH), unless cumbersome traditional sequencing is applied. We developed a Next Generation Sequencing (NGS)-based test targeting the five HH genes, and applied it to patients with clinically relevant iron overload (IO) and a non-diagnostic first level genetic test. We identified several mutations, some of which were novel (i.e. HFE W163X, HAMP R59X, and TFR2 D555N) and allowed molecular reclassification of "non-HFE" HH clinical diagnosis, particularly in some highly selected IO patients without concurring acquired risk factors. This NGS-based "second level" genetic test may represent a useful tool for molecular diagnosis of HH in patients in whom HH phenotype remains unexplained after the search of common HFE mutations.
Collapse
Affiliation(s)
- Sadaf Badar
- Department of Medicine; Section of Internal Medicine, University of Verona; Verona Italy
| | - Fabiana Busti
- Department of Medicine; Section of Internal Medicine, University of Verona; Verona Italy
| | | | - Luciano Xumerle
- Department of Biotechnology; University of Verona; Verona Italy
| | - Paolo Bozzini
- Department of Medicine; Section of Internal Medicine, University of Verona; Verona Italy
| | - Paola Capelli
- Unit of Pathology, Azienda Ospedaliera Universitaria Integrata Verona; Verona Italy
| | - Roberto Pozzi-Mucelli
- Department of Diagnostics and Public Health; Section of Radiology, University of Verona; Verona Italy
| | - Natascia Campostrini
- Department of Medicine; Section of Internal Medicine, University of Verona; Verona Italy
| | - Giovanna De Matteis
- Unit of Clinical Chemistry, Azienda Ospedaliera Universitaria Integrata Verona; Verona Italy
| | | | | | | | - Oliviero Olivieri
- Department of Medicine; Section of Internal Medicine, University of Verona; Verona Italy
| | - Domenico Girelli
- Department of Medicine; Section of Internal Medicine, University of Verona; Verona Italy
- Veneto Regional Referral Center for Iron Metabolism Disorders, GIMFer (Gruppo Interdisciplinare Sulle Malattie Del Ferro); Azienda Ospedaliera Uiversitaria Integrata Verona; Verona Italy
| |
Collapse
|
25
|
Fonseca PFS, Cançado RD, Uellendahl Lopes MM, Correia E, Lescano MA, Santos PCJL. HAMP Gene Mutation Associated with Juvenile Hemochromatosis in Brazilian Patients. Acta Haematol 2016; 135:228-31. [PMID: 27007796 DOI: 10.1159/000444119] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 01/17/2016] [Indexed: 12/15/2022]
Affiliation(s)
- Paula Fernanda Silva Fonseca
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sx00E3;o Paulo Medical School, Sx00E3;o Paulo, Brazil
| | | | | | | | | | | |
Collapse
|
26
|
Castiella A, Zapata E, De Juan MD, Zubiaurre L, Iribarren A, Otazua P, Fernandez J, Aragón L, Zuriarrain O, Gorostidi A. Analysis of HFE mutations and non-HFE gene mutations (TFR2 and SLC40A1) in patients with phenotypic hemochromatosis from the Basque Country. Int J Lab Hematol 2016; 38:e5-e7. [PMID: 26547814 DOI: 10.1111/ijlh.12444] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- A Castiella
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain.
| | - E Zapata
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain
| | - M D De Juan
- Immunology Service, Universitary Donostia Hospital, Donostia, Spain
| | - L Zubiaurre
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain
| | - A Iribarren
- Gastroenterology Service, Mendaro Hospital, Mendaro, Spain
| | - P Otazua
- Gastroenterology Service, Mondragon Hospital, Mondragon, Spain
| | - J Fernandez
- Gastroenterology Service, Galdakao Hospital, Galdakao, Spain
| | - L Aragón
- Immunology Service, Universitary Donostia Hospital, Donostia, Spain
| | - O Zuriarrain
- Genomics Platform, Biodonostia Institute, Donostia, Spain
| | - A Gorostidi
- Genomics Platform, Biodonostia Institute, Donostia, Spain
| |
Collapse
|
27
|
Wallace DF, Subramaniam VN. The global prevalence of HFE and non-HFE hemochromatosis estimated from analysis of next-generation sequencing data. Genet Med 2015; 18:618-26. [PMID: 26633544 DOI: 10.1038/gim.2015.140] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 08/27/2015] [Indexed: 12/31/2022] Open
Abstract
PURPOSE The prevalence of HFE-related hereditary hemochromatosis (HH) among European populations has been well studied. There are no prevalence data for atypical forms of HH caused by mutations in HFE2, HAMP, TFR2, or SLC40A1. The purpose of this study was to estimate the population prevalence of these non-HFE forms of HH. METHODS A list of HH pathogenic variants in publically available next-generation sequence (NGS) databases was compiled and allele frequencies were determined. RESULTS Of 161 variants previously associated with HH, 43 were represented among the NGS data sets; an additional 40 unreported functional variants also were identified. The predicted prevalence of HFE HH and the p.Cys282Tyr mutation closely matched previous estimates from similar populations. Of the non-HFE forms of iron overload, TFR2-, HFE2-, and HAMP-related forms are predicted to be rare, with pathogenic allele frequencies in the range of 0.00007 to 0.0005. Significantly, SLC40A1 variants that have been previously associated with autosomal-dominant ferroportin disease were identified in several populations (pathogenic allele frequency 0.0004), being most prevalent among Africans. CONCLUSION We have, for the first time, estimated the population prevalence of non-HFE HH. This methodology could be applied to estimate the population prevalence of a wide variety of genetic disorders.Genet Med 18 6, 618-626.
Collapse
Affiliation(s)
- Daniel F Wallace
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - V Nathan Subramaniam
- Membrane Transport Laboratory, QIMR Berghofer Medical Research Institute, Brisbane, Australia.,Faculty of Medicine and Biomedical Sciences, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
28
|
Analysis of Familial Tendencies in Transferrin Saturation in a Korean Population. Dig Dis Sci 2015; 60:3136-41. [PMID: 26003557 DOI: 10.1007/s10620-015-3720-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 05/13/2015] [Indexed: 12/09/2022]
Abstract
BACKGROUND Despite the high transferrin saturation (TS) level in Koreans, the p.Cys282Tyr and p.His63Asp mutations are markedly less frequent than in Caucasians. We aimed to determine TS levels and their familial tendencies in a Korean population using nationwide data from the Fifth Korea National Health and Nutrition Examination Survey (KNHANES V-1 2010). METHODS A total of 4904 subjects without a history of hepatitis B and C virus infection, or liver cirrhosis, and who were negative for anemia and hepatitis B antigen were enrolled. A familial tendency analysis was performed in 260 families. Parents were grouped into four quartiles based on their TS levels. Offspring were categorized according to the mean parental TS four quartile scores (1.0, 1.5, 2.0, 2.5, 3.0, 3.5, and 4.0). A familial tendency was evaluated by comparing the mean TS of offspring in seven parental groups. RESULTS The mean TS was 39.3 ± 15.6% for Korean males and 33.2 ± 12.9% for Korean females, and both were significantly higher than those of Caucasians reported in the HEIRS study (30.6 ± 11.0% for male, 25.6 ± 10.6% for female, P < 0.001). The 260 families showed statistically significant familial tendencies of TS values (P < 0.001). The mean TS of offspring in parental group 1.0, 1.5, 2.0, and 2.5 showed a lower value than that in higher group 3.0, 3.5, and 4.0. In contrast, there were no significant differences in age, daily dietary iron intake, and AST or ALT value among seven groups. CONCLUSIONS These findings suggest unidentified genetic variations on high TS in Koreans beyond the p.Cys282Tyr and p.His63Asp mutations commonly identified in Caucasians.
Collapse
|
29
|
Sousa L, Garcia IJP, Costa TGF, Silva LND, Renó CO, Oliveira ES, Tilelli CQ, Santos LL, Cortes VF, Santos HL, Barbosa LA. Effects of Iron Overload on the Activity of Na,K-ATPase and Lipid Profile of the Human Erythrocyte Membrane. PLoS One 2015. [PMID: 26197432 PMCID: PMC4510300 DOI: 10.1371/journal.pone.0132852] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Iron is an essential chemical element for human life. However, in some pathological conditions, such as hereditary hemochromatosis type 1 (HH1), iron overload induces the production of reactive oxygen species that may lead to lipid peroxidation and a change in the plasma-membrane lipid profile. In this study, we investigated whether iron overload interferes with the Na,K-ATPase activity of the plasma membrane by studying erythrocytes that were obtained from the whole blood of patients suffering from iron overload. Additionally, we treated erythrocytes of normal subjects with 0.8 mM H2O2 and 1 μM FeCl3 for 24 h. We then analyzed the lipid profile, lipid peroxidation and Na,K-ATPase activity of plasma membranes derived from these cells. Iron overload was more frequent in men (87.5%) than in women and was associated with an increase (446%) in lipid peroxidation, as indicated by the amount of the thiobarbituric acid reactive substances (TBARS) and an increase (327%) in the Na,K-ATPase activity in the plasma membrane of erythrocytes. Erythrocytes treated with 1 μM FeCl3 for 24 h showed an increase (132%) in the Na,K-ATPase activity but no change in the TBARS levels. Iron treatment also decreased the cholesterol and phospholipid content of the erythrocyte membranes and similar decreases were observed in iron overload patients. In contrast, erythrocytes treated with 0.8 mM H2O2 for 24 h showed no change in the measured parameters. These results indicate that erythrocytes from patients with iron overload exhibit higher Na,K-ATPase activity compared with normal subjects and that this effect is specifically associated with altered iron levels.
Collapse
Affiliation(s)
- Leilismara Sousa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Israel J. P. Garcia
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Tamara G. F. Costa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Lilian N. D. Silva
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Cristiane O. Renó
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Eneida S. Oliveira
- Laboratório de Biologia Molecular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Cristiane Q. Tilelli
- Laboratório de Estudos em Neurociências das Epilepsias e Comorbidades, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Luciana L. Santos
- Laboratório de Biologia Molecular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Vanessa F. Cortes
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Herica L. Santos
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
| | - Leandro A. Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindú, Av Sebastião Gonçalves Coelho 400, 35501–296, Divinópolis, Brazil
- * E-mail:
| |
Collapse
|
30
|
Ekanayake D, Roddick C, Powell LW. Recent advances in hemochromatosis: a 2015 update : a summary of proceedings of the 2014 conference held under the auspices of Hemochromatosis Australia. Hepatol Int 2015; 9:174-82. [PMID: 25788196 DOI: 10.1007/s12072-015-9608-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Accepted: 01/07/2015] [Indexed: 02/07/2023]
Abstract
This review focuses on iron metabolism, the genetics of hemochromatosis, current treatment protocols and various screening methods. Even though the most common form of hereditary hemochromatosis, C282Y gene mutations in the HFE gene, has been extensively studied, novel mutations in both HFE and non-HFE genes have been implicated in this disease. These have important implications for the Asia-Pacific region. In overload, deposition of iron in various body tissues leads to toxic damage. Patients commonly present with non-specific symptoms of malaise and lethargy. Biochemical, imaging and genetic testing can be carried out to confirm diagnosis. Venesection forms the mainstay of treatment and at present cascade screening of affected families is recommended over population-level screening.
Collapse
Affiliation(s)
- Dilum Ekanayake
- School of Medicine, The University of Queensland, Brisbane, QLD, 4029, Australia
| | | | | |
Collapse
|
31
|
HFE genotyping in patients with elevated serum iron indices and liver diseases. BIOMED RESEARCH INTERNATIONAL 2015; 2015:164671. [PMID: 25654085 PMCID: PMC4310263 DOI: 10.1155/2015/164671] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 09/10/2014] [Accepted: 09/18/2014] [Indexed: 12/31/2022]
Abstract
Iron abnormalities in chronic liver disease may be the result of genetic diseases or secondary factors. The present study aimed to identify subjects with HFE-HH in order to describe the frequency of clinical manifestations, identify risk factors for iron elevation, and compare the iron profile of HFE-HH to other genotypes in liver disease patients. A total of 108 individuals with hepatic disease, transferrin saturation (TS) > 45%, and serum ferritin (SF) > 350 ng/mL were tested for HFE mutations. Two groups were characterized: C282Y/C282Y or C282Y/H63D genotypes (n = 16) were the HFE hereditary hemochromatosis (HFE-HH) group; and C282Y and H63D single heterozygotes, the H63D/H63D genotype, and wild-type were considered group 2 (n = 92). Nonalcoholic liver disease, alcoholism, and chronic hepatitis C were detected more frequently in group 2, whereas arthropathy, hepatocarcinoma, diabetes, and osteoporosis rates were significantly higher in the HFE-HH group. TS > 82%, SF > 2685 ng/mL, and serum iron > 178 μg/dL were the cutoffs for diagnosis of HFE-HH in patients with liver disease. Thus, in non-Caucasian populations with chronic liver disease, HFE-HH diagnosis is more predictable in those with iron levels higher than those proposed in current guidelines for the general population.
Collapse
|
32
|
Moreno-Carralero MI, Muñoz-Muñoz JA, Cuadrado-Grande N, López-Rodríguez R, José Hernández-Alfaro M, del-Castillo-Rueda A, Enríquez-de-Salamanca R, Méndez M, Morán-Jiménez MJ. A novel mutation in the SLC40A1 gene associated with reduced iron export in vitro. Am J Hematol 2014; 89:689-94. [PMID: 24644245 DOI: 10.1002/ajh.23714] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Revised: 03/14/2014] [Accepted: 03/14/2014] [Indexed: 01/09/2023]
Abstract
Ferroportin disease is an inherited disorder of iron metabolism and is caused by mutations in the ferroportin gene (SLC40A1). We present a patient with hyperferritinemia, iron overload in the liver with reticuloendothelial distribution and also in the spleen, and under treatment with erythropheresis. A molecular study of the genes involved in iron metabolism (HFE, HJV, HAMP, TFR2, SLC40A1) was undertaken. In vitro functional studies of the novel mutation found in the SLC40A1 gene was performed. The patient was heterozygous for a novel mutation, c.386T>C (p.L129P) in the SLC40A1 gene; some of his relatives were also heterozygous for this mutation. In vitro functional studies of the L129P mutation on ferroportin showed it impairs its capacity to export iron from cells but does not alter its sensitivity to hepcidin. These findings and the iron overload phenotype of the patient suggest that the novel mutation c.386T>C (p.L129P) in the SLC40A1 gene has incomplete penetrance and causes the classical form of ferroportin disease.
Collapse
|
33
|
Core AB, Canali S, Babitt JL. Hemojuvelin and bone morphogenetic protein (BMP) signaling in iron homeostasis. Front Pharmacol 2014; 5:104. [PMID: 24860505 PMCID: PMC4026703 DOI: 10.3389/fphar.2014.00104] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Accepted: 04/21/2014] [Indexed: 12/13/2022] Open
Abstract
Mutations in hemojuvelin (HJV) are the most common cause of the juvenile-onset form of the iron overload disorder hereditary hemochromatosis. The discovery that HJV functions as a co-receptor for the bone morphogenetic protein (BMP) family of signaling molecules helped to identify this signaling pathway as a central regulator of the key iron hormone hepcidin in the control of systemic iron homeostasis. This review highlights recent work uncovering the mechanism of action of HJV and the BMP-SMAD signaling pathway in regulating hepcidin expression in the liver, as well as additional studies investigating possible extra-hepatic functions of HJV. This review also explores the interaction between HJV, the BMP-SMAD signaling pathway and other regulators of hepcidin expression in systemic iron balance.
Collapse
Affiliation(s)
- Amanda B Core
- Division of Nephrology, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Program in Anemia Signaling Research Boston, MA, USA
| | - Susanna Canali
- Division of Nephrology, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Program in Anemia Signaling Research Boston, MA, USA
| | - Jodie L Babitt
- Division of Nephrology, Program in Membrane Biology, Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Program in Anemia Signaling Research Boston, MA, USA
| |
Collapse
|
34
|
Li M, Wang L, Wang W, Qi XL, Tang ZY. Mutations in the HFE gene and sporadic amyotrophic lateral sclerosis risk: a meta-analysis of observational studies. ACTA ACUST UNITED AC 2014; 47:215-22. [PMID: 24604426 PMCID: PMC3982942 DOI: 10.1590/1414-431x20133296] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 11/19/2013] [Indexed: 12/12/2022]
Abstract
Iron homeostasis dysregulation has been regarded as an important mechanism in neurodegenerative diseases. The H63D and C282Y polymorphisms in the HFE gene may be involved in the development of sporadic amyotrophic lateral sclerosis (ALS) through the disruption of iron homeostasis. However, studies investigating the relationship between ALS and these two polymorphisms have yielded contradictory outcomes. We performed a meta-analysis to assess the roles of the H63D and C282Y polymorphisms of HFE in ALS susceptibility. PubMed, MEDLINE, EMBASE, and Cochrane Library databases were systematically searched to identify relevant studies. Strict selection criteria and exclusion criteria were applied. Odds ratios (ORs) with 95% confidence intervals (CIs) were used to assess the strength of associations. A fixed- or random-effect model was selected, depending on the results of the heterogeneity test. Fourteen studies were included in the meta-analysis (six studies with 1692 cases and 8359 controls for C282Y; 14 studies with 5849 cases and 13,710 controls for H63D). For the C282Y polymorphism, significant associations were observed in the allele model (Y vs C: OR=0.76, 95%CI=0.62-0.92, P=0.005) and the dominant model (YY+CY vs CC: OR=0.75, 95%CI=0.61-0.92, P=0.006). No associations were found for any genetic model for the H63D polymorphism. The C282Y polymorphism in HFE could be a potential protective factor for ALS in Caucasians. However, the H63D polymorphism does not appear to be associated with ALS.
Collapse
Affiliation(s)
- M Li
- Department of Neurology, The Second Hospital Affiliated to Nanchang University, Medical College, Nanchang University, Nanchang, China
| | - L Wang
- Department of Neurology, The Second Hospital Affiliated to Nanchang University, Medical College, Nanchang University, Nanchang, China
| | - W Wang
- Department of Neurology, The Second Hospital Affiliated to Nanchang University, Medical College, Nanchang University, Nanchang, China
| | - X L Qi
- Department of Neurology, The Second Hospital Affiliated to Nanchang University, Medical College, Nanchang University, Nanchang, China
| | - Z Y Tang
- Department of Neurology, The Second Hospital Affiliated to Nanchang University, Medical College, Nanchang University, Nanchang, China
| |
Collapse
|
35
|
Pretorius E, Bester J, Vermeulen N, Lipinski B, Gericke GS, Kell DB. Profound morphological changes in the erythrocytes and fibrin networks of patients with hemochromatosis or with hyperferritinemia, and their normalization by iron chelators and other agents. PLoS One 2014; 9:e85271. [PMID: 24416376 PMCID: PMC3887013 DOI: 10.1371/journal.pone.0085271] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 11/25/2013] [Indexed: 12/22/2022] Open
Abstract
It is well-known that individuals with increased iron levels are more prone to thrombotic diseases, mainly due to the presence of unliganded iron, and thereby the increased production of hydroxyl radicals. It is also known that erythrocytes (RBCs) may play an important role during thrombotic events. Therefore the purpose of the current study was to assess whether RBCs had an altered morphology in individuals with hereditary hemochromatosis (HH), as well as some who displayed hyperferritinemia (HF). Using scanning electron microscopy, we also assessed means by which the RBC and fibrin morphology might be normalized. An important objective was to test the hypothesis that the altered RBC morphology was due to the presence of excess unliganded iron by removing it through chelation. Very striking differences were observed, in that the erythrocytes from HH and HF individuals were distorted and had a much greater axial ratio compared to that accompanying the discoid appearance seen in the normal samples. The response to thrombin, and the appearance of a platelet-rich plasma smear, were also markedly different. These differences could largely be reversed by the iron chelator desferal and to some degree by the iron chelator clioquinol, or by the free radical trapping agents salicylate or selenite (that may themselves also be iron chelators). These findings are consistent with the view that the aberrant morphology of the HH and HF erythrocytes is caused, at least in part, by unliganded (‘free’) iron, whether derived directly via raised ferritin levels or otherwise, and that lowering it or affecting the consequences of its action may be of therapeutic benefit. The findings also bear on the question of the extent to which accepting blood donations from HH individuals may be desirable or otherwise.
Collapse
Affiliation(s)
- Etheresia Pretorius
- Department of Physiology, University of Pretoria, Arcadia, South Africa
- * E-mail:
| | - Janette Bester
- Department of Physiology, University of Pretoria, Arcadia, South Africa
| | - Natasha Vermeulen
- Department of Physiology, University of Pretoria, Arcadia, South Africa
| | - Boguslaw Lipinski
- Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, United States of America
| | | | - Douglas B. Kell
- School of Chemistry and The Manchester Institute of Biotechnology, The University of Manchester, Lancs, United Kingdom
| |
Collapse
|
36
|
|
37
|
Détivaud L, Island ML, Jouanolle AM, Ropert M, Bardou-Jacquet E, Le Lan C, Mosser A, Leroyer P, Deugnier Y, David V, Brissot P, Loréal O. Ferroportin diseases: functional studies, a link between genetic and clinical phenotype. Hum Mutat 2013; 34:1529-36. [PMID: 23943237 DOI: 10.1002/humu.22396] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 08/05/2013] [Indexed: 12/21/2022]
Abstract
Ferroportin (FPN) mediates iron export from cells and this function is modulated by serum hepcidin. Mutations in the FPN gene (SLC40A1) lead to autosomal dominant iron overload diseases related either to loss or to gain of function, and usually characterized by normal or low transferrin saturation versus elevated transferrin saturation, respectively. However, for the same mutation, the phenotypic expression may vary from one patient to another. Using in vitro overexpression of wild-type or mutant FPN proteins, we characterized the functional impact of five recently identified FPN gene mutations regarding FPN localization, cell iron status, and hepcidin sensitivity. Our aim was to integrate functional results and biological findings in probands and relatives. We show that while the p.Arg371Gln (R371Q) mutation had no impact on studied parameters, the p.Trp158Leu (W158L), p.Arg88Gly (R88G), and p.Asn185Asp (N185D) mutations caused an iron export defect and were classified as loss-of-function mutations. The p.Gly204Ser (G204S) mutation induced a gain of FPN function. Functional studies are useful to determine whether or not a FPN gene mutation found in an iron overloaded patient is deleterious and to characterize its biological impact, especially when family studies are not fully informative and/or additional confounding factors may affect bio-clinical expression.
Collapse
Affiliation(s)
- Lénaïck Détivaud
- CHU Rennes, French Reference Centre for Rare Iron Overload Diseases of Genetic Origin, Rennes, France; CHU Rennes, Department of Molecular Biology, Rennes, France; CHU Rennes, Department of Liver Diseases, Rennes, France; INSERM UMR 991, Equipe Fer et Foie, Rennes, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Tanaka Y, Ikeda T, Yamamoto K, Ogawa H, Kamisako T. Dysregulated expression of fatty acid oxidation enzymes and iron-regulatory genes in livers of Nrf2-null mice. J Gastroenterol Hepatol 2012; 27:1711-7. [PMID: 22591204 DOI: 10.1111/j.1440-1746.2012.07180.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIM Hepatic excessive iron may play a role in the pathogenesis of non-alcoholic steatohepatitis (NASH). Nrf2 is a master regulator of antioxidative responses. However, the role of Nrf2 in lipid and iron homeostasis remains unclear. Accordingly, it was examined how Nrf2 regulates lipid-related and iron-regulatory genes after feeding a high-fat diet (HFD) with iron. METHODS Wild-type and Nrf2-null mice were fed the following diets: (i) control diet (4% soybean oil) for 12 weeks, (ii) control diet for 8 weeks followed by control diet containing 0.5% carbonyl iron for 4 weeks, (iii) HFD (4% soybean oil and 16% lard) for 12 weeks, (iv) HFD for 8 weeks followed by HFD containing 0.5% carbonyl iron for 4 weeks. Blood and livers were removed after 12 weeks. RESULTS Nrf2-null control mice exhibited a tendency towards higher hepatic triglycerides compared to wild-type control mice. Hepatic malondialdehyde was higher and hepatic iron levels tended to be higher in Nrf2-null mice than wild-type counterparts while on a HFD. The HFD with iron synergistically induced mRNA expression of Pparα targets, including Acox and Cpt1 in wild-type mice, yet the induction was diminished in Nrf2-null mice. Hepatic hepcidin and ferroportin 1 mRNA expression were increased in wild-type mice after feeding a HFD with iron, but were unchanged in any group of Nrf2-null mice. CONCLUSIONS Nrf2 deletion dysregulates hepatic mRNA expression of β-oxidation enzymes and iron-related genes, possibly causing a trend for increased hepatic triglyceride and iron concentrations. Nrf2 may have roles in the progression of NASH.
Collapse
Affiliation(s)
- Yuji Tanaka
- Department of Clinical Laboratory Medicine, Kinki University Faculty of Medicine, Osakasayama, Japan.
| | | | | | | | | |
Collapse
|
39
|
Santos PCJDL, Dinardo CL, Cançado RD, Schettert IT, Krieger JE, Pereira AC. Non-HFE hemochromatosis. Rev Bras Hematol Hemoter 2012; 34:311-6. [PMID: 23049448 PMCID: PMC3460409 DOI: 10.5581/1516-8484.20120079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2012] [Accepted: 07/10/2012] [Indexed: 12/15/2022] Open
Abstract
Hereditary hemochromatosis (HH) is an autosomal recessive disorder classically related to HFE mutations. However, since 1996, it is known that HFE mutations explain about 80% of HH cases, with the remaining around 20% denominated non-HFE hemochromatosis. Nowadays, four main genes are implicated in the pathophysiology of clinical syndromes classified as non-HFE hemochromatosis: hemojuvelin (HJV, type 2Ajuvenile HH), hepcidin (HAMP, type 2B juvenile HH), transferrin receptor 2 (TFR2, type 3 HH) and ferroportin (SLC40A1, type 4 HH). The aim of this review is to explore molecular, clinical and management aspects of non-HFE hemochromatosis.
Collapse
|
40
|
Molecular diagnostic and pathogenesis of hereditary hemochromatosis. Int J Mol Sci 2012; 13:1497-1511. [PMID: 22408404 PMCID: PMC3291973 DOI: 10.3390/ijms13021497] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2011] [Revised: 01/12/2012] [Accepted: 01/13/2012] [Indexed: 12/15/2022] Open
Abstract
Hereditary hemochromatosis (HH) is an autosomal recessive disorder characterized by enhanced intestinal absorption of dietary iron. Without therapeutic intervention, iron overload leads to multiple organ damage such as liver cirrhosis, cardiomyopathy, diabetes, arthritis, hypogonadism and skin pigmentation. Most HH patients carry HFE mutant genotypes: homozygosity for p.Cys282Tyr or p.Cys282Tyr/p.His63Asp compound heterozygosity. In addition to HFE gene, mutations in the genes that encode hemojuvelin (HJV), hepcidin (HAMP), transferrin receptor 2 (TFR2) and ferroportin (SLC40A1) have been associated with regulation of iron homeostasis and development of HH. The aim of this review was to identify the main gene mutations involved in the pathogenesis of type 1, 2, 3 and 4 HH and their genetic testing indication. HFE testing for the two main mutations (p.Cys282Tyr and p.His63Asp) should be performed in all patients with primary iron overload and unexplained increased transferrin saturation and/or serum ferritin values. The evaluation of the HJV p.Gly320Val mutation must be the molecular test of choice in suspected patients with juvenile hemochromatosis with less than 30 years and cardiac or endocrine manifestations. In conclusion, HH is an example that genetic testing can, in addition to performing the differential diagnostic with secondary iron overload, lead to more adequate and faster treatment.
Collapse
|
41
|
Santos PCJL, Gagliardi ACM, Miname MH, Chacra AP, Santos RD, Krieger JE, Pereira AC. SLCO1B1 haplotypes are not associated with atorvastatin-induced myalgia in Brazilian patients with familial hypercholesterolemia. Eur J Clin Pharmacol 2011; 68:273-9. [PMID: 21928084 DOI: 10.1007/s00228-011-1125-1] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Accepted: 08/31/2011] [Indexed: 01/06/2023]
Abstract
PURPOSE Recent studies reported the association of SLCO1B1 haplotypes with the development of musculoskeletal side effects during simvastatin use. The aim was to evaluate the pharmacogenetic association of SLCO1B1 haplotypes with atorvastatin-induced myalgia in a sample of individuals on high-dose atorvastatin regimens. METHODS One hundred and forty-three patients with familial hypercholesterolemia were followed for at least 12 months while receiving atorvastatin. Genotypes for the rs2306283 (c.A388G) and rs4149056 (c.T521C) polymorphisms were detected by high-resolution melting analysis. These markers form four distinct haplotypes (*1A, *1B, *5 and *15). RESULTS During the follow-up period, 14 (9.8%) patients developed myalgia and 16 (11.2%) presented CK levels more than 3 times the upper limit of the normal range. No association of the SLCO1B1 rs2306283 and rs4149056 genotypes or haplotypes with the presence of myalgia or creatine kinase (CK) values was found. Presence of rs2306283 AG + GG genotypes was not associated with increased risks of myalgia or abnormal CK values (OR 2.08, 95% CI 0.62-7.00, p = 0.24 and OR 0.51, 95% CI 0.21-1.26, p = 0.15 respectively). The presence of rs4149056 TC + CC genotypes was also not associated with increased risk of myalgia or abnormal CK values (OR 2.24, 95% CI 0.47-10.72, p = 0.31 and OR 1.51, 95% CI 0.57-3.96, p = 0.41 respectively). CONCLUSIONS Our findings reaffirm that the SLCO1B1 genetic risk appears to be greater in those patients receiving simvastatin compared with those receiving atorvastatin. This suggests that the importance of SLCO1B1 haplotypes depends on the specific statin that has been used.
Collapse
Affiliation(s)
- Paulo Caleb Junior Lima Santos
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of Sao Paulo Medical School, Avenue Dr. Enéas de Carvalho Aguiar, 44 Cerqueira César, São Paulo, SP, Brazil CEP 05403-000.
| | | | | | | | | | | | | |
Collapse
|