Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Más filtros










Intervalo de año de publicación
1.
Hematology ; 26(1): 896-903, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34789084

RESUMEN

ABSTRACTObjectives: Hereditary hyperferritinaemia cataract syndrome (HHCS) is an autosomal dominant disease characterized by high serum ferritin levels and juvenile bilateral cataracts. It is often caused by mutations in the iron response element (IRE) of the ferritin L-subunit (FTL) gene. Here, we report a 73-year-old woman who presented to clinic with persistently elevated serum ferritin and family history of juvenile bilateral cataracts in four generations.Methods: Exome sequencing was used to identify the mutation of the FTL gene. Moreover, Sanger sequencing was performed to validate the mutation in the proband. We also reviewed the FLT gene mutations in published HHCS cases to provide experience for accurate diagnosis of similar patients.Results: A heterozygous mutation at position +33 (c.-167C > T, chr19:49468598) of the FTL gene was identified in the patient.Discussion: HHCS should be considered in the differential diagnosis of hyperferritinemia, especially in the presence of normal serum iron concentration and transferrin saturation.Conclusion: For patients with unexplained hyperferritinemia and bilateral cataracts who have experienced early vision loss, the establishment of genetic counseling is essential to diagnose other family members who are at risk in time.Abbreviations: FTL: ferritin L-subunit; HHCS: hereditary hyperferritinaemia cataract syndrome; IDT: integrated DNA technologies; IRE: iron response element; IRP: iron regulatory proteins; MRI: magnetic resonance imaging; SNV: single nucleotide variant; UTR: untranslated region.


Asunto(s)
Apoferritinas/genética , Catarata/congénito , Trastornos del Metabolismo del Hierro/congénito , Mutación , Anciano , Alelos , Apoferritinas/sangre , Biomarcadores , Catarata/diagnóstico , Catarata/genética , Catarata/metabolismo , Catarata/terapia , Análisis Mutacional de ADN , Femenino , Genotipo , Humanos , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/metabolismo , Trastornos del Metabolismo del Hierro/terapia , Linaje , Elementos de Respuesta/genética , Evaluación de Síntomas
2.
Biochem Biophys Res Commun ; 553: 114-118, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33765555

RESUMEN

Major depressive disorder (MDD) is a debilitating mental illness that can cause significant emotional disturbances and severe socioeconomic burdens. Rodent and nonhuman primate-based depression models have been studied, such as brain-derived neurotrophic factor (BDNF) and monoamine acid disorder hypotheses, as well as peripheral microbiota disturbances causing MDD; however, the pathogenesis is still largely unknown. This study aims to explore the relationship between ferritin and MDD. First, alterations in ferritin, including ferritin light chain (FTL) and ferritin heavy chain (FTH), in MDD patient plasma compared with healthy control (HC) plasma were detected using ELISA. Then, serum ferritin expression in cLPS-depressed mice was measured by ELISA. The existence of FTH in the hippocampus was validated by immunofluorescence, and the change in FTH levels in the hippocampus of mice injected with cLPS was detected by western blotting. FTL levels in MDD patients were decreased compared with those in HCs. In cLPS-depressed mice, serum ferritin was not different from that in the control group, while the expression of FTH in the hippocampus was significantly reduced in depressed mice. Our findings demonstrate the alteration of ferritin expression in MDD and provide new insight into the pathogenesis of MDD.


Asunto(s)
Apoferritinas/sangre , Apoferritinas/metabolismo , Trastorno Depresivo Mayor/sangre , Trastorno Depresivo Mayor/metabolismo , Hipocampo/metabolismo , Animales , Apoferritinas/deficiencia , Apoferritinas/genética , Biomarcadores/sangre , Biomarcadores/metabolismo , Estudios de Casos y Controles , Trastorno Depresivo Mayor/genética , Trastorno Depresivo Mayor/patología , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Femenino , Hipocampo/efectos de los fármacos , Hipocampo/patología , Humanos , Lipopolisacáridos/farmacología , Masculino , Ratones , Adulto Joven
3.
Clin Chem Lab Med ; 57(12): 1837-1845, 2019 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-31211687

RESUMEN

Background In hereditary hyperferritinaemia-cataract syndrome (HHCS), single nucleic acid alterations in the ferritin light chain (L-ferritin) iron response element (IRE) constitutively derepress ferritin synthesis, resulting in hyperferritinaemia, L-ferritin deposits in the lens of the eye and early bilateral cataract onset. Methods In this study, six German families with putative HHCS were analysed. Clinical diagnosis of HHCS was based on medical history, evaluation of ferritin serum levels, transferrin saturation and clinical ophthalmological examination. Diagnosis was confirmed by polymerase chain reaction (PCR)-based DNA sequencing of the L-ferritin IRE. Results Genetic analysis of the L-ferritin IRE revealed relevant single nucleic acid alterations in each of the affected families. Variants c.-168G > A, c.-168G > U and c.-167C > U were located in the C-bulge region; and variants c.-161C > U and c.-157G > A were located in the hexanucleotide loop of the L-ferritin IRE. Conclusions Family history of hyperferritinaemia and juvenile cataracts are strong indicators of HHCS. Genetic analysis of the L-ferritin IRE is a straightforward procedure to confirm the diagnosis. Accurate diagnosis of hyperferritinaemia can avoid unnecessary treatment by venesection, and focus attention on early cataract detection in offspring at risk.


Asunto(s)
Apoferritinas/genética , Catarata/congénito , Trastornos del Metabolismo del Hierro/congénito , Adulto , Anciano , Anciano de 80 o más Años , Apoferritinas/análisis , Apoferritinas/sangre , Secuencia de Bases/genética , Catarata/diagnóstico , Catarata/epidemiología , Familia , Femenino , Ferritinas/genética , Pruebas Genéticas/métodos , Alemania/epidemiología , Humanos , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/epidemiología , Masculino , Persona de Mediana Edad , Mutación/genética , Linaje
4.
Genes (Basel) ; 10(6)2019 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-31208055

RESUMEN

Iron is essential for physical activity due to its role in energy production pathways and oxygen transportation via hemoglobin and myoglobin. Changes in iron-related biochemical parameters after physical exercise in athletes are of substantial research interest, but molecular mechanisms such as gene expression are still rarely tested in sports. In this paper, we evaluated the mRNA levels of genes related to iron metabolism (PCBP1, PCBP2, FTL, FTH, and TFRC) in leukocytes of 24 amateur runners at four time points: before, immediately after, 3 h after, and 24 h after a marathon. We measured blood morphology as well as serum concentrations of iron, ferritin, and C-reactive protein (CRP). Our results showed significant changes in gene expression (except for TFRC), serum iron, CRP, and morphology after the marathon. However, the alterations in mRNA and protein levels occurred at different time points (immediately and 3 h post-run, respectively). The levels of circulating ferritin remained stable, whereas the number of transcripts in leukocytes differed significantly. We also showed that running pace might influence mRNA expression. Our results indicated that changes in the mRNA of genes involved in iron metabolism occurred independently of serum iron and ferritin concentrations.


Asunto(s)
Atletas , Ferritinas/sangre , Hierro/sangre , Carrera/fisiología , Adulto , Apoferritinas/sangre , Proteína C-Reactiva/metabolismo , Proteínas de Unión al ADN/sangre , Expresión Génica/genética , Hemoglobinas/metabolismo , Humanos , Leucocitos/metabolismo , Masculino , Persona de Mediana Edad , Mioglobina/sangre , Oxidorreductasas/sangre , ARN Mensajero/sangre , Proteínas de Unión al ARN/sangre
5.
J Clin Endocrinol Metab ; 104(3): 688-696, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30260393

RESUMEN

Context: Iron overload has been associated with greater adipose tissue (AT) depots. We retrospectively studied the potential interactions between iron and AT during an experimental overfeeding in participants without obesity. Methods: Twenty-six participants (mean body mass index ± SD, 24.7 ± 3.1 kg/m2) underwent a 56-day overfeeding (+760 kcal/d). Serum iron biomarkers (ELISA), subcutaneous AT (SAT) gene expression, and abdominal AT distribution assessed by MRI were analyzed at the beginning and the end of the intervention. Results: Before intervention: SAT mRNA expression of the iron transporter transferrin (Tf) was positively correlated with the expression of genes related to lipogenesis (lipin 1, ACSL1) and lipid storage (SCD). SAT expression of the ferritin light chain (FTL) gene, encoding ferritin (FT), an intracellular iron storage protein, was negatively correlated to SREBF1, a gene related to lipogenesis. Serum FT (mean, 92 ± 57 ng/mL) was negatively correlated with the expression of SAT genes linked to lipid storage (SCD, DGAT2) and to lipogenesis (SREBF1, ACSL1). After intervention: Overfeeding led to a 2.3 ± 1.3-kg weight gain. In parallel to increased expression of lipid storage-related genes (mitoNEET, SCD, DGAT2, SREBF1), SAT Tf, SLC40A1 (encoding ferroportin 1, a membrane iron export channel) and hephaestin mRNA levels increased, whereas SAT FTL mRNA decreased, suggesting increased AT iron requirement. Serum FT decreased to 67 ± 43 ng/mL. However, no significant associations between serum iron biomarkers and AT distribution or expansion were observed. Conclusion: In healthy men, iron metabolism gene expression in SAT is associated with lipid storage and lipogenesis genes expression and is modulated during a 56-day overfeeding diet.


Asunto(s)
Adiposidad/fisiología , Hierro/metabolismo , Lipogénesis/fisiología , Hipernutrición/fisiopatología , Grasa Subcutánea/metabolismo , Adulto , Apoferritinas/sangre , Apoferritinas/metabolismo , Biomarcadores/sangre , Proteínas de Transporte de Catión/metabolismo , Regulación de la Expresión Génica/fisiología , Voluntarios Sanos , Humanos , Imagen por Resonancia Magnética , Masculino , Proteínas de la Membrana/metabolismo , Hipernutrición/etiología , Estudios Retrospectivos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Grasa Subcutánea/diagnóstico por imagen , Aumento de Peso/fisiología , Adulto Joven
6.
Eur J Haematol ; 102(2): 131-142, 2019 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-30325535

RESUMEN

OBJECTIVES: We previously reported the prognostic value of serum ferritin in younger patients with intermediate-risk acute myeloid leukemia (AML). The aims of this study were to confirm this finding in a larger cohort regardless of age and prognostic subgroups, to explore the expression and functional role of ferritin in AML cells as well as the regulation of serum ferritin levels in AML patients. PATIENTS/MATERIALS/METHODS: Serum ferritin levels at diagnosis were collected in a cohort of 525 patients treated by intensive chemotherapy. In silico, in vitro, and in vivo analyses were conducted to assess the pattern of expression and functional role of FTH1 and FTL in AML. RESULTS: We confirmed the independent prognostic value of serum ferritin. In transcriptomic databases, FTH1 and FTL were overexpressed in AML and leukemic stem cells compared to normal hematopoietic stem cells. The gene signature designed from AML patients overexpressing FTH1 revealed a significant enrichment in genes of the immune and inflammatory response including Nf-KB pathway, oxidative stress, or iron pathways. This gene signature was enriched in cytarabine-resistant AML cells in a patient-derived xenograft model. FTH1 protein was also overexpressed in patient's samples and correlated with the in vitro cytotoxic activity of cytarabine. Lastly, we demonstrated that chemotherapy induced an inflammatory response including a significant increase in serum ferritin levels between day 1 and 8 of induction chemotherapy that was blocked by dexamethasone. CONCLUSION: Ferritin is deregulated in most AML patients likely through inflammation, associated with chemoresistance, and could represent a new therapeutic target.


Asunto(s)
Apoferritinas/genética , Ferritinas/sangre , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/sangre , Leucemia Mieloide Aguda/genética , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Apoferritinas/sangre , Biomarcadores , Terapia Combinada , Resistencia a Antineoplásicos , Femenino , Ferritinas/genética , Perfilación de la Expresión Génica , Humanos , Mediadores de Inflamación , Estimación de Kaplan-Meier , Leucemia Mieloide Aguda/mortalidad , Leucemia Mieloide Aguda/terapia , Masculino , Persona de Mediana Edad , Oportunidad Relativa , Oxidorreductasas , Pronóstico , Modelos de Riesgos Proporcionales , Resultado del Tratamiento
7.
Blood Adv ; 2(11): 1191-1197, 2018 06 12.
Artículo en Inglés | MEDLINE | ID: mdl-29844204

RESUMEN

Red cell distribution width (RDW) is associated with venous thromboembolism (VTE), but the underlying mechanism(s) is unclear. Iron deficiency is associated with high RDW, and studies suggest an association between iron deficiency and VTE. To assess whether iron deficiency is a risk factor for VTE that explains the association between RDW and VTE, we conducted a nested case-control study of 390 patients with VTE and 802 age- and sex-matched controls selected from the population-based cohort of the Tromsø Study. Physical measurements and blood samples were collected from 1994 to 1995. Logistic regression models were used to calculate odds ratios (ORs) with 95% confidence intervals (CIs) for VTE by RDW, hepcidin, and ferritin light chain (FtL). RDW was inversely associated with hepcidin, FtL, and hemoglobin. The risk of VTE increased linearly across categories of higher plasma hepcidin levels. Participants with hepcidin in the highest quartile had an OR for VTE of 1.32 (95% CI, 1.00-2.42), and those in the >90% percentile had an OR for VTE of 1.66 (95% CI, 1.14-2.42) compared with the reference group (quartiles 2 and 3). The risk estimates remained similar after adjustment for C-reactive protein. The risk of VTE increased by categories of higher RDW and was strengthened after inclusion of hepcidin and FtL in the multivariable model. Our findings reject the hypothesis that iron deficiency explains the association between RDW and VTE and suggest, in contrast, that high body iron levels might increase the risk of VTE.


Asunto(s)
Índices de Eritrocitos , Hepcidinas/sangre , Modelos Cardiovasculares , Tromboembolia Venosa/sangre , Adulto , Anciano , Apoferritinas/sangre , Femenino , Humanos , Hierro/sangre , Deficiencias de Hierro , Masculino , Persona de Mediana Edad , Estudios Prospectivos , Factores de Riesgo
8.
J Pediatr Hematol Oncol ; 40(8): 620-624, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-29432304

RESUMEN

INTRODUCTION: Despite anemia in acquired heart disease being a common problem, little is known in patients with congenital heart disease (CHD). METHODS: In total, 544 consecutive stable noncyanotic CHD patients were studied to determine demographic, clinical, and analytic parameters. Anemia was defined as a condition in which hemoglobin concentration was <13 g/dL in male individuals and <12 g/dL in female individuals. RESULTS: In total, 49 (9%) CHD patients had anemia. Patients with complex anatomy had the highest prevalence of anemia (33%). The median hemoglobin concentration was 14.4 (13.5 to 15.6) mg/dL. Of the total anemic CHD patients, 21 of 49 (43%) were microcytic (mean corpuscular volume <84 fL) and 46 of 49 (94%) had a mean corpuscular volume under 95 fL. Oral anticoagulation, oral antiaggregation, diuretic treatment, and having valve prostheses or cardiovascular risk factors, such as arterial hypertension or diabetes mellitus, did not reach statistical significance between anemic and nonanemic CHD patients. Multivariate analyses determined as risk factors for anemia a worse New York Heart Association functional class (patients in class >II/IV) (odds ratio [OR], 8.37; 95% confidence interval [CI], 1.69-41.35), N-terminal proB-type natriuretic peptide levels >125 pg/mL (OR, 7.90; 95% CI, 2.88-21.69), and apoferritn levels below 15 ng/mL (OR, 0.21; 95% CI, 0.09-0.50). The Kaplan-Meier survival analysis showed no significant differences in mortality between anemic and nonanemic CHD patients (P=0.143). CONCLUSIONS: The incidence of anemia in CHD patients is similar to that of the normal population and iron deficiency anemia accounts for most of the cases. There were no significant differences in mortality between CHD patients with and without anemia.


Asunto(s)
Anemia Ferropénica , Cardiopatías Congénitas , Adolescente , Adulto , Anemia Ferropénica/sangre , Anemia Ferropénica/mortalidad , Anemia Ferropénica/patología , Apoferritinas/sangre , Supervivencia sin Enfermedad , Femenino , Estudios de Seguimiento , Cardiopatías Congénitas/sangre , Cardiopatías Congénitas/mortalidad , Cardiopatías Congénitas/patología , Hemoglobinas/metabolismo , Humanos , Masculino , Péptido Natriurético Encefálico/sangre , Fragmentos de Péptidos/sangre , Factores de Riesgo , Tasa de Supervivencia
9.
Einstein (Säo Paulo) ; 15(4): 492-495, Oct.-Dec. 2017. tab, graf
Artículo en Inglés | LILACS | ID: biblio-891420

RESUMEN

ABSTRACT Hereditary hyperferritinemia-cataract syndrome is an autosomal dominant genetic disorder associated with mutations in the 5'UTR region of the ferritin light chain gene. These mutations cause the ferritin levels to increase even in the absence of iron overload. Patients also develop bilateral cataract early due to accumulation of ferritin in the lens, and many are misdiagnosed as having hemochromatosis and thus not properly treated. The first cases were described in 1995 and several mutations have already been identified. However, this syndrome is still a poorly understood. We report two cases of unrelated Brazilian families with clinical suspicion of the syndrome, which were treated in our department. For the definitive diagnosis, the affected patients, their parents and siblings were submitted to Sanger sequencing of the 5'UTR region for detection of the ferritin light gene mutation. Single nucleotide polymorphism-like mutations were found in the affected patients, previously described. The test assisted in making the accurate diagnosis of the disease, and its description is important so that the test can be incorporated into clinical practice.


RESUMO A síndrome hereditária hiperferritinemia-catarata é uma doença genética autossômica dominante associada a mutações na região 5'UTR do gene da cadeia leve da ferritina. Estas mutações elevam os níveis de ferritina, mesmo na ausência de sobrecarga de ferro. Os pacientes também desenvolvem catarata bilateral precocemente, devido ao acúmulo de ferritina no cristalino, e muitos são erroneamente diagnosticados como portadores de hemocromatose, sendo tratados de maneira inadequada. Os primeiros casos foram descritos em 1995, e diversas mutações já foram identificadas. Entretanto, essa síndrome ainda é pouco conhecida. Relatamos dois casos de famílias brasileiras, não relacionadas, com suspeita clínica da síndrome, que foram atendidas em nosso serviço. Para o diagnóstico definitivo, os pacientes afetados, seus pais e irmãos foram submetidos à pesquisa de mutação do gene ferritina, por sequenciamento de Sanger da região 5'UTR. Foram encontradas mutações do tipo polimorfismo de nucleotídeo único nos pacientes afetados, já descritas anteriormente. O teste auxiliou no diagnóstico preciso da doença e é importante ser divulgado, para ser incorporado na prática clínica.


Asunto(s)
Humanos , Masculino , Preescolar , Niño , Apoferritinas/sangre , Catarata/congénito , Trastornos del Metabolismo del Hierro/congénito , Hierro/sangre , Síndrome , Catarata/genética , Catarata/sangre , Brasil , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/sangre , Mutación/genética
10.
Einstein (Sao Paulo) ; 15(4): 492-495, 2017.
Artículo en Inglés, Portugués | MEDLINE | ID: mdl-28746593

RESUMEN

Hereditary hyperferritinemia-cataract syndrome is an autosomal dominant genetic disorder associated with mutations in the 5'UTR region of the ferritin light chain gene. These mutations cause the ferritin levels to increase even in the absence of iron overload. Patients also develop bilateral cataract early due to accumulation of ferritin in the lens, and many are misdiagnosed as having hemochromatosis and thus not properly treated. The first cases were described in 1995 and several mutations have already been identified. However, this syndrome is still a poorly understood. We report two cases of unrelated Brazilian families with clinical suspicion of the syndrome, which were treated in our department. For the definitive diagnosis, the affected patients, their parents and siblings were submitted to Sanger sequencing of the 5'UTR region for detection of the ferritin light gene mutation. Single nucleotide polymorphism-like mutations were found in the affected patients, previously described. The test assisted in making the accurate diagnosis of the disease, and its description is important so that the test can be incorporated into clinical practice.


Asunto(s)
Apoferritinas/sangre , Catarata/congénito , Trastornos del Metabolismo del Hierro/congénito , Hierro/sangre , Brasil , Catarata/sangre , Catarata/genética , Niño , Preescolar , Humanos , Trastornos del Metabolismo del Hierro/sangre , Trastornos del Metabolismo del Hierro/genética , Masculino , Mutación/genética , Síndrome
11.
Br J Haematol ; 166(2): 279-91, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24750026

RESUMEN

Ferritin plays important roles in iron metabolism and controls iron absorption in the intestine. The ferritin subunits ferritin heavy chain (Fth1) and ferritin light chain (Ftl1) are tightly regulated at both the transcriptional and post-transcriptional levels. However, mechanisms of maintaining stable, basal expression of Fth1 are poorly understood. Here, we show that global deletion of Mbd5 in mice induces an iron overload phenotype. Liver and serum iron levels in Mbd5(-/-) mice were 3·2-fold and 1·5-fold higher respectively, than wild-type littermates; moreover, serum ferritin was increased >5-fold in the Mbd5(-/-) mice. Mbd5 encodes a member of the methyl-CpG binding domain family; however, the precise function of this gene is poorly understood. Here, we found that intestinal Fth1 mRNA levels were decreased in Mbd5(-/-) mice. Loss of Fth1 expression in the intestine could lead to iron over-absorption. Furthermore, deleting Mbd5 specifically in the intestine resulted in a phenotype similar to that of conditional deletion of Fth1 mice. An Fth1 promoter-report luciferase assay indicated that overexpression of Mbd5 enhanced Fth1 transcription in a dose-dependent manner. Histone H4 acetylation of the Fth1 promoter was reduced in the intestine of Mbd5(-/-) mice and further analysis showed that histone acetyltransferase KAT2A was essential for MBD5-induced Fth1 transcription.


Asunto(s)
Apoferritinas/genética , Histona Acetiltransferasas/fisiología , Sobrecarga de Hierro/metabolismo , Hierro/metabolismo , Proteína 2 de Unión a Metil-CpG/fisiología , Acetilación , Animales , Apoferritinas/sangre , Metilación de ADN , Eliminación de Gen , Regulación de la Expresión Génica , Marcación de Gen , Hemocromatosis/sangre , Hemocromatosis/genética , Hemocromatosis/metabolismo , Hepcidinas/biosíntesis , Histonas/metabolismo , Absorción Intestinal/fisiología , Sobrecarga de Hierro/sangre , Sobrecarga de Hierro/genética , Hígado/metabolismo , Proteína 2 de Unión a Metil-CpG/deficiencia , Proteína 2 de Unión a Metil-CpG/genética , Ratones , Ratones Noqueados , Motivos de Nucleótidos , Fenotipo , Regiones Promotoras Genéticas , ARN Mensajero/genética , Transcripción Genética
12.
Mol Vis ; 19: 835-44, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23592921

RESUMEN

PURPOSE: Cataract is a clinically and genetically heterogeneous disorder of the ocular lens and an important cause of visual impairment. The aim of this study was to map and identify the gene underlying autosomal dominant cataract segregating in a four-generation family, determine the lens expression profile of the identified gene, and test for its association with age-related cataract in a case-control cohort. METHODS: Genomic DNA was prepared from blood leukocytes, and genotyping was performed by means of single-nucleotide polymorphism markers and microsatellite markers. Linkage analyses were performed using the GeneHunter and MLINK programs, and mutation detection was achieved by dideoxy cycle sequencing. Lens expression studies were performed using reverse-transcription polymerase chain reaction (RT-PCR) and in situ hybridization. RESULTS: Genome-wide linkage analysis with single nucleotide polymorphism markers in the family identified a likely disease-haplotype interval on chromosome 19q (rs888861-[~17Mb]-rs8111640) that encompassed the microsatellite marker D19S879 (logarithm of the odds score [Z]=2.03, recombination distance [θ]=0). Mutation profiling of positional-candidate genes detected a heterozygous, noncoding G-to-T transversion (c.-168G>T) located in the iron response element (IRE) of the gene coding for ferritin light chain (FTL) that cosegregated with cataract in the family. Serum ferritin levels were found to be abnormally elevated (~fourfold), without evidence of iron overload, in an affected family member; this was consistent with a diagnosis of hereditary hyperferritinemia-cataract syndrome. No sequence variations located within the IRE were detected in a cohort of 197 cases with age-related cataract and 102 controls with clear lenses. Expression studies of human FTL, and its mouse counterpart FTL1, in the lens detected RT-PCR amplicons containing full-length protein-coding regions, and strong in situ localization of FTL1 transcripts to the lens equatorial epithelium and peripheral cortex. CONCLUSIONS: The data are consistent with robust transcription of FTL in the lens, and suggest that whereas variations clustered in the IRE of the FTL gene are directly associated with hereditary hyperferritinemia-cataract syndrome, such IRE variations are unlikely to play a significant role in the genetic etiology of age-related cataract.


Asunto(s)
Envejecimiento/genética , Apoferritinas/genética , Catarata/genética , ADN Intergénico/genética , Mutación/genética , Adolescente , Adulto , Envejecimiento/patología , Animales , Apoferritinas/sangre , Secuencia de Bases , Catarata/sangre , Niño , Preescolar , Femenino , Genes Dominantes/genética , Ligamiento Genético , Humanos , Lactante , Hierro/sangre , Cristalino/metabolismo , Cristalino/patología , Escala de Lod , Masculino , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Linaje , Elementos de Respuesta/genética , Adulto Joven
13.
Shock ; 39(6): 520-6, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23524846

RESUMEN

Ferritin L (FTL) and ferritin H (FTH) subunits are responsible for intracellular iron storage. Serum ferritin levels are not only dependant on body iron stores. Aims of the present study are to demonstrate nature, source, and major regulatory mediators of serum ferritin in an animal model of acute-phase (AP) response. Animals (rats, wild-type [WT] mice, and interleukin [IL]-6ko mice) were injected with turpentine oil (TO) intra-muscularity to induce a sterile abscess and sacrificed at different time points afterward. Rat hepatocytes were isolated for cell culture and, after reaching confluence, stimulated with major AP cytokines to induce AP conditions. We found a significantly increased expression of both ferritin subunits in liver at mRNA and protein levels during AP response. In the serum of both control and TO-injected rats, only FTL was detectable by Western blotting, whereas no increase in serum FTL was measured by Western blot or enzyme-linked immunosorbent assay. An increase in protein expression of FTL and FTH was observed in lysates of rat hepatocytes after treatment with IL-6, IL-1ß, and tumor necrosis factor-α; however, only FTL was increasingly released into supernatant. In both TO-injected rats and WT mice, a dramatic increase in serum IL-6 levels was observed, along with an increased amount of hepatic ferritin subunits. However, an increase of hepatic FTL but not of FTH protein expression was observed in IL-6ko mice after TO injection. Our data demonstrate that FTL is the only rat serum ferritin whose release into circulation from the hepatocytes is increased by the effect of AP cytokines (e.g., IL-6). In contrast, FTH expression is intracellular in both under physiological and AP conditions.


Asunto(s)
Proteínas de Fase Aguda/metabolismo , Reacción de Fase Aguda/metabolismo , Apoferritinas/metabolismo , Hígado/metabolismo , Reacción de Fase Aguda/inducido químicamente , Reacción de Fase Aguda/patología , Animales , Apoferritinas/sangre , Apoferritinas/genética , Células Cultivadas , Citocinas/fisiología , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Interleucina-6/deficiencia , Interleucina-6/genética , Interleucina-6/fisiología , Masculino , Ratones , Ratones Noqueados , ARN Mensajero/genética , Ratas , Ratas Wistar , Trementina
14.
Congenit Heart Dis ; 8(2): 117-23, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-22891845

RESUMEN

INTRODUCTION: Iron deficiency anemia is the most common single cause of anemia worldwide. The purpose of our study was to estimate the prevalence of anemia in adult congenital heart disease (ACHD) patients, compare different hematology parameters between hypoxemic and nonhypoxemic ACHD patients, and determine which parameters detect iron deficiency anemia in hypoxemic ACHD patients. METHODS: ACHD patients were studied and blood samples collected for determination of hemoglobin, derived red cell indices, serum iron, apoferritin, total iron-binding capacity, transferrin saturation index, C-reactive protein (CRP), and N-terminal proB-type natriuretic peptide (NT-proBNP) levels. RESULTS: Two hundred seventy-eight ACHD patients, mean age 31.6 ± 14.3 years old, were studied. One hundred sixty-seven (60%) patients were male. Two hundred forty-five patients were nonhypoxemic and 33 patients were hypoxemic. Hypoxemic ACHD patients had significant higher hemoglobin concentration (g/dL) (17.5 ± 3.5 vs. 14.6 ± 1.7, P <.001), red cell distribution width (RDW) (%) (17.0 ± 3.3 vs. 14.1 ± 7.6, P <.034), apoferritin (ng/mL) (19.8 [4.1-147.2] vs. 38.0 [6.7-191.2], P =.019), CRP (mg/dL) (0.50 [0.0-3.8] vs. 0.12 [0.0-1.4], P <.001), and NT-proBNP (pg/mL) (409.3 [33.3-9830.8] vs. 5.2 [0.0-1068.4], P <.001) levels than nonhypoxemic ACHD patients. Serum iron, total iron-binding capacity, and transferrin saturation index were not statistically significant between hypoxemic and nonhypoxemic ACHD patients. In the hypoxemic group, 15 (45%) patients had apoferritin levels <20 ng/mL and eight (24%) patients developed microcytosis and hypochromia. A RDW above the normal range (>14.5%) in hypoxemic ACHD patients allowed the detection of an apoferritin level <20 ng/mL with a sensitivity of 93%. CONCLUSIONS: RDW seems to be a useful and economic tool to detect low serum apoferritin levels in hypoxemic ACHD patients.


Asunto(s)
Anemia Ferropénica/diagnóstico , Anemia Ferropénica/epidemiología , Cardiopatías Congénitas/epidemiología , Hipoxia/epidemiología , Adolescente , Adulto , Anemia Ferropénica/sangre , Apoferritinas/sangre , Biomarcadores/sangre , Proteína C-Reactiva/análisis , Distribución de Chi-Cuadrado , Regulación hacia Abajo , Índices de Eritrocitos , Femenino , Cardiopatías Congénitas/sangre , Hemoglobinas/análisis , Humanos , Hipoxia/sangre , Hierro/sangre , Masculino , Persona de Mediana Edad , Péptido Natriurético Encefálico/sangre , Fragmentos de Péptidos/sangre , Valor Predictivo de las Pruebas , Prevalencia , España/epidemiología , Adulto Joven
15.
PLoS One ; 7(9): e46153, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23029420

RESUMEN

Glioblastomas (GBMs) are the most common and lethal primary tumors of the central nervous system with high level of recurrence despite aggressive therapy. Tumor-associated proteins/peptides may appear in the plasma of these patients as a result of disruption of the blood-brain barrier in them, raising the scope for development of plasma-based tests for diagnosis and monitoring the disease. With this objective, we analyzed the levels of proteins present in the plasma from GBM patients using an iTRAQ based LC-MS/MS approach. Analysis with pooled plasma specimens from the patient and healthy control samples revealed high confidence identification of 296 proteins, of which 61 exhibited a fold-change ≥1.5 in the patient group. Forty-eight of them contained signal sequence. A majority have been reported in the differentially expressed transcript or protein profile of GBM tissues; 6 have been previously studied as plasma biomarkers for GBM and 16 for other types of cancers. Altered levels of three representative proteins-ferritin light chain (FTL), S100A9, and carnosinase 1 (CNDP1)-were verified by ELISA in a test set of ten individual plasma specimens. FTL is an inflammation marker also implicated in cancer, S100A9 is an important member of the Ca(2+) signaling cascade reported to be altered in GBM tissue, and CNDP1 has been reported for its role in the regulation of the levels of carnosine, implicated as a potential drug for GBM. These and other proteins in the dataset may form useful starting points for further clinical investigations for the development of plasma-based biomarker panels for GBM.


Asunto(s)
Apoferritinas/genética , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Calgranulina B/genética , Dipeptidasas/genética , Regulación Neoplásica de la Expresión Génica , Glioblastoma/genética , Proteínas de Neoplasias/genética , Apoferritinas/sangre , Biomarcadores de Tumor/sangre , Neoplasias Encefálicas/sangre , Neoplasias Encefálicas/diagnóstico , Calcio/metabolismo , Calgranulina B/sangre , Carnosina/metabolismo , Cromatografía Liquida , Dipeptidasas/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Glioblastoma/sangre , Glioblastoma/diagnóstico , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Neoplasias/sangre , Proteómica/métodos , Transducción de Señal , Espectrometría de Masas en Tándem
16.
Parkinsonism Relat Disord ; 18(8): 909-15, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22818529

RESUMEN

Neuroferritinopathy is an autosomal dominantly inherited disorder caused by mutations in the gene encoding the ferritin light chain polypeptide. It leads to iron deposition particularly in the cerebellum, basal ganglia and motor cortex. The disease becomes clinically apparent in adulthood mainly with extrapyramidal signs and progresses slowly over decades. Patients usually have intact cognition until the very late stages of this disorder. Neuroimaging is the most helpful investigation and shows a very distinctive picture. So far no medication has been shown to have a disease-modifying effect. We present five new cases of this condition and review the current understanding of the pathogenesis and its clinical findings.


Asunto(s)
Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/genética , Distrofias Neuroaxonales/diagnóstico , Distrofias Neuroaxonales/genética , Apoferritinas/sangre , Apoferritinas/genética , Apoferritinas/metabolismo , Encéfalo/metabolismo , Encéfalo/patología , Humanos , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/patología , Masculino , Persona de Mediana Edad , Mutación/genética , Distrofias Neuroaxonales/patología , Neuronas/metabolismo , Neuronas/patología , Linaje
17.
Ann Clin Biochem ; 49(Pt 3): 302-5, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22535864

RESUMEN

Investigating persistent hyperferritinaemia without apparent iron overload is challenging. Even when inflammation, cirrhosis, Still's disease, fatty liver and malignancy are excluded, there remains a group of patients with unexplained hyperferritinaemia for whom rare forms of haemochromatosis (ferroportin disease) are a consideration. Preliminary results suggest that abnormal percentage glycosylation of serum ferritin is seen in some cases of genetically determined hyperferritinaemia. Serum ferritin is normally 50-81% glycosylated, but low glycosylation (20-42%) prevails in hereditary hyperferritinaemia cataract syndrome. This contrasts with hyperglycosylation (>90%) associated with the benign hyperferritinaemia related to missense L ferritin (p.Thr30Ile) mutation. Here, we describe two novel missense L ferritin variants also associated with hyperglycosylation, p.Gln26Ile and p.Ala27Val. Ferritin glycosylation, a comparatively simple measurement, can identify patients for DNA sequencing as hyperglycosylation (>90%) is associated with benign hyperferritinaemia and mutant L ferritin chain.


Asunto(s)
Apoferritinas/genética , Ferritinas/genética , Trastornos del Metabolismo del Hierro/genética , Mutación Missense , Anciano , Secuencia de Aminoácidos , Apoferritinas/sangre , Ferritinas/sangre , Glicosilación , Humanos , Trastornos del Metabolismo del Hierro/sangre , Trastornos del Metabolismo del Hierro/diagnóstico , Masculino , Persona de Mediana Edad , Modelos Moleculares , Datos de Secuencia Molecular , Sistemas de Lectura Abierta , Análisis de Secuencia de ADN
18.
Mol Vis ; 17: 1011-5, 2011 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-21541272

RESUMEN

PURPOSE: To date, more than thirty nine genetic loci have been associated with congenital cataracts. Despite this progress, current diagnostic techniques are insufficient for unraveling the underlying genetic defect in sporadic patients and small families. In the present manuscript we demonstrate the contribution of routine laboratory tests in the search for genetic defects of childhood cataracts. METHODS: Two families with congenital cataracts and hematologic findings that included hyperferritinemia and the "ii" blood type underwent detailed ophthalmologic and clinical examinations. Mutation analysis of the ferritin light chain (FTL) and glucosaminyl (N-acetyl) transferase 2, I-branching enzyme (GCNT2) genes was performed in the two families, respectively. RESULTS: In the family with the "ii" blood group we found a novel GCNT2 mutation c.G935A (p.G312D) in the cataract patients, while in the family with hyperferritinemia cataract syndrome we identified a G→C heterozygous mutation at position +32 of FTL. CONCLUSIONS: Hematologic biomarkers may simplify the search for the underlying molecular defect in families with congenital cataract.


Asunto(s)
Apoferritinas/genética , Catarata/congénito , Catarata/genética , Trastornos del Metabolismo del Hierro/congénito , N-Acetilglucosaminiltransferasas/genética , Adulto , Apoferritinas/sangre , Biomarcadores/sangre , Antígenos de Grupos Sanguíneos/análisis , Antígenos de Grupos Sanguíneos/genética , Catarata/sangre , Catarata/complicaciones , Catarata/metabolismo , Catarata/patología , Niño , Consanguinidad , Análisis Mutacional de ADN , Femenino , Ligamiento Genético , Predisposición Genética a la Enfermedad , Humanos , Trastornos del Metabolismo del Hierro/complicaciones , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/metabolismo , Israel , Cristalino/metabolismo , Cristalino/patología , Masculino , Mutación , N-Acetilglucosaminiltransferasas/sangre , Linaje
19.
Am J Physiol Gastrointest Liver Physiol ; 298(4): G525-9, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20110460

RESUMEN

Hereditary hemochromatosis (HH), a widespread hereditary iron metabolism disorder, is characterized by an excessive absorption of dietary iron, resulting in increased body iron stores. Some studies indicate a sex difference in disease expression, with women showing a slower disease progression and a less severe clinical profile. This is usually attributed to iron loss during menstruation and pregnancy. However, this link has not been clearly demonstrated. The Hfe-/- mouse model recapitulates key aspects of HH, including an iron overload phenotype similar to that observed in human patients. In this study, we use it to test the impact of multiple pregnancies in the iron stores. One-year-old nulliparous and pluriparous (averaging 29 weaned pups per female) C57BL/6 (B6) and Hfe-/- mice were euthanized, and blood and tissues were collected. Several serological and erythroid parameters were evaluated, as well as tissue nonheme iron content and serum ferritin. Hepcidin 1, hepcidin 2, and bone morphogenetic protein 6 (BMP6) expressions in the liver were determined by real-time PCR. No significant differences were observed for many serological and erythroid parameters although differences occurred in transferrin saturation and mean corpuscular volume in Hfe-/- mice and total iron-binding capacity in B6 mice. Hepatic iron concentration was similar for nulliparous and pluriparous mice of both genotypes, but total iron per organ (liver, spleen, heart, and pancreas) was higher overall in pluriparous females than nulliparous. Hepcidin 1 and 2 and BMP6 expressions were significantly decreased in pluriparous females, when compared with nulliparous, in both genotypes. In conclusion, multiple pregnancies do not reduce body iron stores in Hfe-/- mice.


Asunto(s)
Hemocromatosis/complicaciones , Hemocromatosis/metabolismo , Antígenos de Histocompatibilidad Clase I/genética , Hierro/metabolismo , Proteínas de la Membrana/genética , Paridad/fisiología , Complicaciones del Embarazo/metabolismo , Animales , Péptidos Catiónicos Antimicrobianos/genética , Apoferritinas/sangre , Proteína Morfogenética Ósea 6/genética , Recuento de Eritrocitos , Índices de Eritrocitos/genética , Femenino , Expresión Génica/genética , Hematócrito , Hemocromatosis/sangre , Hemocromatosis/genética , Proteína de la Hemocromatosis , Hemoglobinas/análisis , Hemoglobinas/metabolismo , Hepcidinas , Hierro/análisis , Hierro/sangre , Hígado/química , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miocardio/química , Miocardio/metabolismo , Páncreas/química , Páncreas/metabolismo , Embarazo , Complicaciones del Embarazo/sangre , Complicaciones del Embarazo/genética , Bazo/química , Bazo/metabolismo , Transferrina/química , Transferrina/metabolismo
20.
Blood Rev ; 23(3): 95-104, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18835072

RESUMEN

Ferritin, a major iron storage protein, is essential to iron homeostasis and is involved in a wide range of physiologic and pathologic processes. In clinical medicine, ferritin is predominantly utilized as a serum marker of total body iron stores. In cases of iron deficiency and overload, serum ferritin serves a critical role in both diagnosis and management. Elevated serum and tissue ferritin are linked to coronary artery disease, malignancy, and poor outcomes following stem cell transplantation. Ferritin is directly implicated in less common but potentially devastating human diseases including sideroblastic anemias, neurodegenerative disorders, and hemophagocytic syndrome. Additionally, recent research describes novel functions of ferritin independent of iron storage.


Asunto(s)
Ferritinas/sangre , Ferritinas/metabolismo , Trastornos del Metabolismo del Hierro/metabolismo , Hierro/metabolismo , Quininógenos/metabolismo , Neoplasias/metabolismo , Apoferritinas/sangre , Apoferritinas/química , Apoferritinas/metabolismo , Ferritinas/química , Homeostasis/fisiología , Humanos , Hierro/sangre , Trastornos del Metabolismo del Hierro/terapia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...