Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 349
Filtrar
1.
Ann Clin Transl Neurol ; 11(5): 1359-1364, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38561955

RESUMEN

Neuroferritinopathy is a disorder of neurodegeneration with brain iron accumulation that has no proven disease-modifying treatments. Clinical trials require biomarkers of iron deposition. We examined brain iron accumulation in one presymptomatic FTL mutation carrier, two individuals with neuroferritinopathy and one healthy control using ultra-high-field 7T MRI. There was increased magnetic susceptibility, suggestive of iron deposition, in superficial and deep gray matter in both presymptomatic and symptomatic neuroferritinopathy. Cavitation of the putamen and globus pallidus increased with disease stage and at follow up. The widespread brain iron deposition in presymptomatic and early disease provides an opportunity for monitoring disease-modifying intervention.


Asunto(s)
Trastornos del Metabolismo del Hierro , Hierro , Imagen por Resonancia Magnética , Distrofias Neuroaxonales , Humanos , Distrofias Neuroaxonales/diagnóstico por imagen , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Trastornos del Metabolismo del Hierro/diagnóstico por imagen , Trastornos del Metabolismo del Hierro/metabolismo , Trastornos del Metabolismo del Hierro/genética , Hierro/metabolismo , Adulto , Masculino , Femenino , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Persona de Mediana Edad , Apoferritinas/metabolismo , Apoferritinas/genética
2.
Ann Hematol ; 103(6): 2173-2176, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38637332

RESUMEN

A 58-year-old female was found to have hyperferritinemia (Serum ferritin:1683 ng/mL) during work-up for mild normocytic anemia. Transferrin saturation(TSAT) was low-normal. Magnetic resonance imaging (MRI) abdomen showed evidence of hepatic iron deposition. Liver biopsy showed 4 + hepatic iron deposition without any evidence of steatosis or fibrosis. Quantitative liver iron was elevated at 348.3 µmol/g dry liver weight [Reference range(RR): 3-33 µmol/g dry liver weight]. She was presumptively diagnosed with tissue iron overload, cause uncertain. A diagnosis of ferroportin disease (FD) was considered, but the pattern of iron distribution in the liver, mainly within the hepatic parenchyma (rather than in the hepatic Kupffer cells seen in FD), and the presence of anemia (uncommon in FD) made this less likely. She was treated with intermittent phlebotomy for over a decade with poor tolerance due to worsening normocytic to microcytic anemia. A trial of deferasirox was done but it was discontinued after a month due to significant side effects. During the course of treatment, her ferritin level decreased. Over the past 1.5 years, she developed progressively worsening neurocognitive decline. MRI brain showed areas of susceptibility involving basal ganglia, midbrain and cerebellum raising suspicion for metabolic deposition disease. Neuroimaging findings led to testing for serum copper and ceruloplasmin levels which were both found to be severely low. Low serum copper, ceruloplasmin levels and neuroimaging findings led us to consider Wilson disease however prior liver biopsy showing elevated hepatic iron rather than hepatic copper excluded the diagnosis of Wilson disease. After shared decision making, ceruloplasmin gene analysis was not pursued due to patient's preference and prohibitive cost of testing. The diagnosis of aceruloplasminemia was ultimately made. The biochemical triad of hyperferritinemia, low-normal TSAT and microcytic anemia should raise the possibility of aceruloplasminemia. Since neurological manifestations are rare in most inherited iron overload syndromes, neurological symptoms in a patient with tissue iron overload should prompt consideration of aceruloplasminemia as a differential diagnosis.


Asunto(s)
Ceruloplasmina , Trastornos del Metabolismo del Hierro , Imagen por Resonancia Magnética , Humanos , Femenino , Persona de Mediana Edad , Ceruloplasmina/deficiencia , Ceruloplasmina/análisis , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/genética , Enfermedades Neurodegenerativas/diagnóstico , Hígado/patología , Hígado/metabolismo , Hígado/diagnóstico por imagen , Ferritinas/sangre , Hierro/metabolismo , Hierro/sangre , Diagnóstico Diferencial , Sobrecarga de Hierro/diagnóstico , Deferasirox/uso terapéutico
3.
Parkinsonism Relat Disord ; 123: 106103, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38582019

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) encompasses a clinically and genetically heterogeneous group of rare disorders. Here, we report clinical, neuroimaging and genetic studies in twenty three Brazilian NBIA patients. In thirteen subjects, deleterious variants were detected in known NBIA-causing genes (PANK2, PLA2G6, C9ORF12, WDR45 and FA2H), including previously unreported variants in PANK2 and PLA2G6. Two patients carried rare, likely pathogenic variants in genes not previously associated with NBIA: KMT2A c.11785A > C (p.Ile3929Leu), and TIMM8A c.127T > C (p.Cys43Arg), suggesting an expansion of their associated phenotypes to include NBIA. In eight patients the etiology remains unsolved, suggesting variants undetectable by the adopted methods, or the existence of additional NBIA-causing genes.


Asunto(s)
Neuroimagen , Humanos , Brasil , Femenino , Masculino , Adulto , Adolescente , Adulto Joven , Niño , Preescolar , Imagen por Resonancia Magnética , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Encéfalo/patología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/diagnóstico por imagen , Fosfotransferasas (Aceptor de Grupo Alcohol)/genética , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/diagnóstico por imagen , Fosfolipasas A2 Grupo VI
5.
HGG Adv ; 4(4): 100236, 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37660254

RESUMEN

Ferritin, the iron-storage protein, is composed of light- and heavy-chain subunits, encoded by FTL and FTH1, respectively. Heterozygous variants in FTL cause hereditary neuroferritinopathy, a type of neurodegeneration with brain iron accumulation (NBIA). Variants in FTH1 have not been previously associated with neurologic disease. We describe the clinical, neuroimaging, and neuropathology findings of five unrelated pediatric patients with de novo heterozygous FTH1 variants. Children presented with developmental delay, epilepsy, and progressive neurologic decline. Nonsense FTH1 variants were identified using whole-exome sequencing, with a recurrent variant (p.Phe171∗) identified in four unrelated individuals. Neuroimaging revealed diffuse volume loss, features of pontocerebellar hypoplasia, and iron accumulation in the basal ganglia. Neuropathology demonstrated widespread ferritin inclusions in the brain. Patient-derived fibroblasts were assayed for ferritin expression, susceptibility to iron accumulation, and oxidative stress. Variant FTH1 mRNA transcripts escape nonsense-mediated decay (NMD), and fibroblasts show elevated ferritin protein levels, markers of oxidative stress, and increased susceptibility to iron accumulation. C-terminal variants in FTH1 truncate ferritin's E helix, altering the 4-fold symmetric pores of the heteropolymer, and likely diminish iron-storage capacity. FTH1 pathogenic variants appear to act by a dominant, toxic gain-of-function mechanism. The data support the conclusion that truncating variants in the last exon of FTH1 cause a disorder in the spectrum of NBIA. Targeted knockdown of mutant FTH1 transcript with antisense oligonucleotides rescues cellular phenotypes and suggests a potential therapeutic strategy for this pediatric neurodegenerative disorder.


Asunto(s)
Apoferritinas , Trastornos del Metabolismo del Hierro , Distrofias Neuroaxonales , Humanos , Niño , Apoferritinas/genética , Trastornos del Metabolismo del Hierro/genética , Hierro/metabolismo , Ferritinas/genética , Oxidorreductasas/metabolismo
8.
Mol Genet Metab ; 137(1-2): 26-32, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35878504

RESUMEN

BACKGROUND: Beta-propeller protein-associated neurodegeneration (BPAN) is a rare neurodegenerative disorder characterized by iron accumulation in the brain with spectrum of neurodevelopmental and movement phenotypes. In anticipation of future clinical trials and to inform clinical care, there is an unmet need to capture the phenotypic diversity of this rare disorder and better define disease subtypes. METHODS: A total of 27 individuals with BPAN were included in our natural history study, from which traditional outcome measures were obtained in 18 subjects. Demographic and diagnostic information, along with acquisition of basic developmental skills and overall neurologic severity were extracted from the medical records. Functional outcome measures were administered at the time of the evaluation or applied retrospectively at the last clinical encounter for patients who were not able to travel for in person. Based on age and functional level, the following assessments were administered: Leiter-3, Gross Motor Function Measure (GMFM)-66 Item Sets, Vineland-3, and Peabody-2. RESULTS: Overall, cognitive function was more impaired compared to gross motor function. Onset of symptoms of BPAN within the first 6 months of life was associated with decreased gain of ambulation and gain of spoken language (ambulation: log-rank test p = 0.0015; gain of first word: p = 0.0015). There was no difference in age at seizure onset by age at initial symptom onset (p = 0.8823). Collection of prospective outcome measures was limited by attention and behavior in our patient population, reinforcing the complexity of phenotype assessment and inadequacy of available standardized tests. Overall, gross motor and adaptive behavior assessments were better able to capture the dynamic range of function across the BPAN population than the fine motor and non-verbal cognitive tests. Floor effects were noted across outcome measures in a subset of individuals for cognitive and adaptive behavior tests. CONCLUSION: Our data suggest the distinct phenotypes of BPAN: a severe, early onset form and an attenuated form with higher cognitive capabilities. Early age at onset was a key factor in predicting future neurologic impairment.


Asunto(s)
Trastornos del Metabolismo del Hierro , Humanos , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/genética , Psicometría , Estudios Prospectivos , Estudios Retrospectivos , Proteínas Portadoras/genética , Hierro/metabolismo , Evaluación de Resultado en la Atención de Salud
10.
EBioMedicine ; 77: 103869, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35180557

RESUMEN

BACKGROUND: Neurodegeneration with brain iron accumulation (NBIA) are a group of clinically and genetically heterogeneous diseases characterized by iron overload in basal ganglia and progressive neurodegeneration. Little is known about the epidemiology of NBIA disorders. In the absence of large-scale population-based studies, obtaining reliable epidemiological data requires innovative approaches. METHODS: All pathogenic variants were collected from the 13 genes associated with autosomal recessive NBIA (PLA2G6, PANK2, COASY, ATP13A2, CP, AP4M1, FA2H, CRAT, SCP2, C19orf12, DCAF17, GTPBP2, REPS1). The allele frequencies of these disease-causing variants were assessed in exome/genome collections: the Genome Aggregation Database (gnomAD) and our in-house database. Lifetime risks were calculated from the sum of allele frequencies in the respective genes under assumption of Hardy-Weinberg equilibrium. FINDINGS: The combined estimated lifetime risk of all 13 investigated NBIA disorders is 0.88 (95% confidence interval 0.70-1.10) per 100,000 based on the global gnomAD dataset (n = 282,912 alleles), 0.92 (0.65-1.29) per 100,000 in the European gnomAD dataset (n = 129,206), and 0.90 (0.48-1.62) per 100,000 in our in-house database (n = 44,324). Individually, the highest lifetime risks (>0.15 per 100,000) are found for disorders caused by variants in PLA2G6, PANK2 and COASY. INTERPRETATION: This population-genetic estimation on lifetime risks of recessive NBIA disorders reveals frequencies far exceeding previous population-based numbers. Importantly, our approach represents lifetime risks from conception, thus including prenatal deaths. Understanding the true lifetime risk of NBIA disorders is important in estimating disease burden, allocating resources and targeting specific interventions. FUNDING: This work was carried out in the framework of TIRCON ("Treat Iron-Related Childhood-Onset Neurodegeneration").


Asunto(s)
Trastornos del Metabolismo del Hierro , Distrofias Neuroaxonales , Enfermedades Neurodegenerativas , Encéfalo/patología , Proteínas de Unión al Calcio , Niño , Bases de Datos Genéticas , Humanos , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Proteínas Mitocondriales/genética , Distrofias Neuroaxonales/epidemiología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/epidemiología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/patología , Proteínas Nucleares , Complejos de Ubiquitina-Proteína Ligasa
11.
Neurol Sci ; 43(3): 1791-1797, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-34559338

RESUMEN

OBJECTIVE: Structural abnormalities in thalami and basal ganglia, in particular the globus pallidus (GP), are a neuroimaging hallmark of hereditary aceruloplasminemia (HA), yet few functional imaging data exit in HA carriers. This study investigated the iron-related structural and functional abnormalities in an Italian HA family. METHODS: Multimodal imaging was used including structural 3 T MRI, functional imaging (SPECT imaging with 123I-ioflupane (DAT-SPECT), cardiac 123I metaiodobenzylguanidine (123I-MIBG) scintigraphy, and 18F-fluorodeoxyglucose (18F-FDG)-PET imaging). In the proband, MRI and scintigraphic evaluations were performed at baseline, 2 and 4 years (structural imaging), and 2 years of follow-up period (functional imaging). RESULTS: We investigated two cousins carrying a novel splicing homozygous mutation in intron 6 (IVS6 + 1 G > A) of CP gene. Interestingly, MRI features in both subjects were characterized by marked iron accumulation in the thalami and basal ganglia nuclei, while GP was not affected. MRI performed in the proband at 2 and 4 years of follow-up confirmed progressive neurodegeneration of the thalami and basal ganglia without the involvement of GP. Functional imaging showed reduced putaminal DAT uptake in both cousins, whereas cardiac MIBG and FDG uptakes performed in the proband were normal. Longitudinal scintigraphic investigations did not show significant changes over the time. CONCLUSIONS: For HA carriers, our findings demonstrate that GP was spared by iron accumulation over the time. The nigrostriatal presynaptic dopaminergic system was damaged while the cardiac sympathetic system remained longitudinally preserved, thus expanding the imaging features of this rare inherited disorder.


Asunto(s)
Trastornos del Metabolismo del Hierro , Enfermedades Neurodegenerativas , 3-Yodobencilguanidina , Ceruloplasmina/deficiencia , Humanos , Trastornos del Metabolismo del Hierro/diagnóstico por imagen , Trastornos del Metabolismo del Hierro/genética , Imagen por Resonancia Magnética , Imagen Multimodal , Mutación , Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/genética , Tomografía de Emisión de Positrones/métodos , Radiofármacos
12.
Can J Neurol Sci ; 49(3): 338-351, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-34082843

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) is a term used for a group of hereditary neurological disorders with abnormal accumulation of iron in basal ganglia. It is clinically and genetically heterogeneous with symptoms such as dystonia, dysarthria, Parkinsonism, intellectual disability, and spasticity. The age at onset and rate of progression are variable among individuals. Current therapies are exclusively symptomatic and unable to hinder the disease progression. Approximately 16 genes have been identified and affiliated to such condition with different functions such as iron metabolism (only two genes: Ferritin Light Chain (FTL) Ceruloplasmin (CP)), lipid metabolism, lysosomal functions, and autophagy process, but some functions have remained unknown so far. Subgroups of NBIA are categorized based on the mutant genes. Although in the last 10 years, the development of whole-exome sequencing (WES) technology has promoted the identification of disease-causing genes, there seem to be some unknown genes and our knowledge about the molecular aspects and pathogenesis of NBIA is not complete yet. There is currently no comprehensive study about the NBIA in Iran; however, one of the latest discovered NBIA genes, GTP-binding protein 2 (GTPBP2), has been identified in an Iranian family, and there are some patients who have genetically remained unknown.


Asunto(s)
Trastornos del Metabolismo del Hierro , Ganglios Basales , Encéfalo/patología , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , Humanos , Irán , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/genética , Mutación
13.
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
14.
Sci Rep ; 11(1): 22568, 2021 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-34799629

RESUMEN

WDR45 plays an essential role in the early stage of autophagy. De novo heterozygous mutations in WDR45 have been known to cause ß-propeller protein-associated neurodegeneration (BPAN), a subtype of neurodegeneration with brain iron accumulation (NBIA). Although BPAN patients display global developmental delay with intellectual disability, the neurodevelopmental pathophysiology of BPAN remains largely unknown. In the present study, we analyzed the physiological role of Wdr45 and pathophysiological significance of the gene abnormality during mouse brain development. Morphological and biochemical analyses revealed that Wdr45 is expressed in a developmental stage-dependent manner in mouse brain. Wdr45 was also found to be located in excitatory synapses by biochemical fractionation. Since WDR45 mutations are thought to cause protein degradation, we conducted acute knockdown experiments by in utero electroporation in mice to recapitulate the pathophysiological conditions of BPAN. Knockdown of Wdr45 caused abnormal dendritic development and synaptogenesis during corticogenesis, both of which were significantly rescued by co-expression with RNAi-resistant version of Wdr45. In addition, terminal arbors of callosal axons were less developed in Wdr45-deficient cortical neurons of adult mouse when compared to control cells. These results strongly suggest a pathophysiological significance of WDR45 gene abnormalities in neurodevelopmental aspects of BPAN.


Asunto(s)
Encéfalo/metabolismo , Proteínas Portadoras/metabolismo , Trastornos del Metabolismo del Hierro/metabolismo , Degeneración Nerviosa , Distrofias Neuroaxonales/metabolismo , Neurogénesis , Animales , Axones/metabolismo , Axones/patología , Encéfalo/embriología , Células COS , Proteínas Portadoras/genética , Chlorocebus aethiops , Dendritas/metabolismo , Dendritas/patología , Sinapsis Eléctricas/metabolismo , Sinapsis Eléctricas/patología , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Edad Gestacional , Trastornos del Metabolismo del Hierro/embriología , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Ratones Endogámicos ICR , Distrofias Neuroaxonales/embriología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Transducción de Señal
15.
Metab Brain Dis ; 36(8): 2273-2281, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34347207

RESUMEN

Aceruloplasminemia (ACP) is a rare disorder of iron overload resulting from ceruloplasmin (CP) variants. Because of its rarity and heterogeneity, the diagnosis of ACP is often missed or misdiagnosed. Here, we aim to present a clinical spectrum of ACP and raise more attention to the early diagnosis. Whole exome sequencing (WES) was performed in a Chinese female patient suspected with ACP and her clinical data were collected in detail. The PubMed databases was searched for published ACP patients within the last decade, and we present a systematic review of their clinical features with data extracted from these researches. A novel pathogenic variant (c.2689delC) and a known pathogenic variant (c.606dupA) within ceruloplasmin gene were identified in our patient and confirmed the diagnosis of ACP. Then we reviewed 51 ACP patients including the case we reported here. A possible timeline of symptoms was discovered, anemia appears first (29.7 years old on average), followed by diabetes (37.3 years old) and finally neurological symptoms (50.7 years old). The delay in diagnosis was significantly shortened in patients without neurological symptoms. Biochemical triad including anemia, low to undetectable serum ceruloplasmin, low serum iron and/or hyperferritinemia, showed better sensitivity in diagnosis than clinical triad including diabetes, neurological symptoms, and retinal degeneration. Due to the variable symptom spectrum, patients with ACP often visit different departments, which can lead to misdiagnosis. Clinical attention needs to be paid to symptoms and tests that have a warning effect. Prompt diagnosis in the early stage of the disease can be beneficial.


Asunto(s)
Ceruloplasmina , Trastornos del Metabolismo del Hierro , Adulto , Ceruloplasmina/deficiencia , Ceruloplasmina/genética , Ceruloplasmina/metabolismo , China , Femenino , Humanos , Trastornos del Metabolismo del Hierro/diagnóstico , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Persona de Mediana Edad , Mutación/genética , Enfermedades Neurodegenerativas
16.
Parkinsonism Relat Disord ; 89: 146-147, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34298215

RESUMEN

Mitochondrial Membrane-protein Associated Neurodegeneration (MPAN) is a rare disease, caused by C19orf12 mutations and up to 29 different mutations have been described. We report a young woman presented with spastic paraparesis due to C19orf12 gene. MPAN presenting like Hereditary spastic paraplegia-43 is rare and the genetic mutation had been described only once in the literature.


Asunto(s)
Trastornos del Metabolismo del Hierro/genética , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , Distrofias Neuroaxonales/genética , Paraplejía Espástica Hereditaria/genética , Exones , Femenino , Humanos , Mutación Missense , Adulto Joven
17.
Int J Mol Sci ; 22(11)2021 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-34064225

RESUMEN

Hereditary hyperferritinemia-cataract syndrome (HHCS) is a rare disease characterized by high serum ferritin levels, congenital bilateral cataracts, and the absence of tissue iron overload. This disorder is produced by mutations in the iron responsive element (IRE) located in the 5' untranslated regions (UTR) of the light ferritin (FTL) gene. A canonical IRE is a mRNA structure that interacts with the iron regulatory proteins (IRP1 and IRP2) to post-transcriptionally regulate the expression of proteins related to iron metabolism. Ferritin L and H are the proteins responsible for iron storage and intracellular distribution. Mutations in the FTL IRE abrogate the interaction of FTL mRNA with the IRPs, and de-repress the expression of FTL protein. Subsequently, there is an overproduction of ferritin that accumulates in serum (hyperferritinemia) and excess ferritin precipitates in the lens, producing cataracts. To illustrate this disease, we report two new families affected with hereditary hyperferritinemia-cataract syndrome with previous known mutations. In the diagnosis of congenital bilateral cataracts, HHCS should be taken into consideration and, therefore, it is important to test serum ferritin levels in patients with cataracts.


Asunto(s)
Catarata/congénito , Ferritinas/genética , Trastornos del Metabolismo del Hierro/congénito , Adulto , Catarata/genética , Niño , Femenino , Humanos , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/genética , Proteínas Reguladoras del Hierro/genética , Mutación/genética
18.
Zhongguo Dang Dai Er Ke Za Zhi ; 23(6): 650-656, 2021 Jun.
Artículo en Chino | MEDLINE | ID: mdl-34130790

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) is a group of rare neurogenetic degenerative diseases caused by genetic mutations and characterized by iron deposition in the central nervous system, especially in the basal ganglia, with an overall incidence rate of 2/1 000 000-3/1 000 000. Major clinical manifestations are extrapyramidal symptoms. This disease is presently classified into 14 different subtypes based on different pathogenic genes, and its pathogenesis and treatment remain unclear. This article summarizes the research advances in the pathogenesis and treatment of NBIA, so as to help pediatricians understand this disease and provide a reference for subsequent research on treatment.


Asunto(s)
Enfermedades de los Ganglios Basales , Trastornos del Metabolismo del Hierro , Ganglios Basales , Encéfalo , Humanos , Hierro , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/terapia
19.
Parkinsonism Relat Disord ; 86: 45-47, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33839641

RESUMEN

L-2-hydroxiglutaric aciduria (L2HGA) is a rare, childhood-onset, organic aciduria, with characteristic clinical (cerebellar ataxia) and neuroimaging (subcortical leukodystrophy) features. Movement disorders in this condition are usually of hyperkinetic type. Herein is reported the case of two adult siblings with recent L2HGA diagnosis, presenting with dopa-responsive parkinsonism and MRI iron deposition.


Asunto(s)
Encefalopatías Metabólicas Innatas/complicaciones , Encéfalo/patología , Trastornos del Metabolismo del Hierro/genética , Trastornos Parkinsonianos/genética , Femenino , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Linaje
20.
Eur J Med Genet ; 64(3): 104159, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33548513

RESUMEN

Mutations in the gene for the ferritin light chain (FTL) often present with hypoferritinemia associated with progressive, late onset extrapyramidal dysfunction. However, it has been suggested that some FTL mutations may impact ferritin levels without any neurological manifestations. We report on a FTL mutation in a three generation family with autosomal dominant hypoferritinemia without neurodegeneration. The 4 year old proband was identified with longstanding history of hypoferritinemia without evidence of anemia. Brain MRI did not show any evidence of iron deposition. It was found that the patient's 19 month old sister, 30 year old mother and 58 year old maternal grandmother also had hypoferritinemia and normal iron levels. Over the next nine years, none of these persons had any evidence of neurological dysfunction, including movement disorders, gait disturbances, behavioral or psychiatric dysfunction. Whole exome sequencing revealed a heterozygous interstitial deletion of at least 5 kb within cytogenic band 19q13.33 involving exons 3 and 4 of FTL in all affected family members. This 3' FTL deletion is predicted to create a significantly truncated protein product. We conclude that haploinsufficiency of FTL may be associated with hypoferritinemia without neurological dysfunction.


Asunto(s)
Apoferritinas/genética , Deficiencias de Hierro , Trastornos del Metabolismo del Hierro/genética , Distrofias Neuroaxonales/genética , Fenotipo , Adulto , Enfermedades Asintomáticas , Preescolar , Femenino , Eliminación de Gen , Haploinsuficiencia , Humanos , Lactante , Trastornos del Metabolismo del Hierro/diagnóstico , Masculino , Persona de Mediana Edad , Mutación , Distrofias Neuroaxonales/diagnóstico , Linaje
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA