Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 167
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.
Stem Cell Res ; 69: 103122, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37209469

RESUMEN

Infantile neuroaxonal dystrophy (INAD) is a rare neurodegenerative disease caused mainly by homozygous or compound heterozygous mutations in the PLA2G6 gene. We generated a human induced pluripotent stem cell (hiPSC) line (ONHi001-A) using fibroblasts derived from a patient with INAD. The patient exhibited c.517C > T (p.Q173X) and c.1634A > G (p.K545R) compound heterozygous mutations in the PLA2G6 gene. This hiPSC line may be useful for studying the pathogenic mechanism underlying INAD.


Asunto(s)
Células Madre Pluripotentes Inducidas , Distrofias Neuroaxonales , Enfermedades Neurodegenerativas , Humanos , Células Madre Pluripotentes Inducidas/patología , Enfermedades Neurodegenerativas/genética , Mutación/genética , Homocigoto , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Fosfolipasas A2 Grupo VI/genética
3.
Elife ; 122023 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-36645408

RESUMEN

Infantile neuroaxonal dystrophy (INAD) is caused by recessive variants in PLA2G6 and is a lethal pediatric neurodegenerative disorder. Loss of the Drosophila homolog of PLA2G6, leads to ceramide accumulation, lysosome expansion, and mitochondrial defects. Here, we report that retromer function, ceramide metabolism, the endolysosomal pathway, and mitochondrial morphology are affected in INAD patient-derived neurons. We show that in INAD mouse models, the same features are affected in Purkinje cells, arguing that the neuropathological mechanisms are evolutionary conserved and that these features can be used as biomarkers. We tested 20 drugs that target these pathways and found that Ambroxol, Desipramine, Azoramide, and Genistein alleviate neurodegenerative phenotypes in INAD flies and INAD patient-derived neural progenitor cells. We also develop an AAV-based gene therapy approach that delays neurodegeneration and prolongs lifespan in an INAD mouse model.


Asunto(s)
Proteínas de Drosophila , Distrofias Neuroaxonales , Trastornos Parkinsonianos , Ratones , Animales , Neuronas/metabolismo , Trastornos Parkinsonianos/metabolismo , Drosophila/metabolismo , Ceramidas/metabolismo , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Fosfolipasas A2 Grupo VI/metabolismo , Proteínas del Ojo/metabolismo , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo
4.
Biomolecules ; 12(5)2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35625641

RESUMEN

Disruption of cerebral iron regulation appears to have a role in aging and in the pathogenesis of various neurodegenerative disorders. Possible unfavorable impacts of iron accumulation include reactive oxygen species generation, induction of ferroptosis, and acceleration of inflammatory changes. Whole-brain iron-sensitive magnetic resonance imaging (MRI) techniques allow the examination of macroscopic patterns of brain iron deposits in vivo, while modern analytical methods ex vivo enable the determination of metal-specific content inside individual cell-types, sometimes also within specific cellular compartments. The present review summarizes the whole brain, cellular, and subcellular patterns of iron accumulation in neurodegenerative diseases of genetic and sporadic origin. We also provide an update on mechanisms, biomarkers, and effects of brain iron accumulation in these disorders, focusing on recent publications. In Parkinson's disease, Friedreich's disease, and several disorders within the neurodegeneration with brain iron accumulation group, there is a focal siderosis, typically in regions with the most pronounced neuropathological changes. The second group of disorders including multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis shows iron accumulation in the globus pallidus, caudate, and putamen, and in specific cortical regions. Yet, other disorders such as aceruloplasminemia, neuroferritinopathy, or Wilson disease manifest with diffuse iron accumulation in the deep gray matter in a pattern comparable to or even more extensive than that observed during normal aging. On the microscopic level, brain iron deposits are present mostly in dystrophic microglia variably accompanied by iron-laden macrophages and in astrocytes, implicating a role of inflammatory changes and blood-brain barrier disturbance in iron accumulation. Options and potential benefits of iron reducing strategies in neurodegeneration are discussed. Future research investigating whether genetic predispositions play a role in brain Fe accumulation is necessary. If confirmed, the prevention of further brain Fe uptake in individuals at risk may be key for preventing neurodegenerative disorders.


Asunto(s)
Trastornos del Metabolismo del Hierro , Distrofias Neuroaxonales , Enfermedades Neurodegenerativas , Encéfalo/patología , Humanos , Hierro/farmacología , Trastornos del Metabolismo del Hierro/patología , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/patología
5.
PLoS One ; 17(2): e0263332, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35108319

RESUMEN

Dysregulation of calcium homeostasis has been hypothesized to play a role in Alzheimer's disease (AD) pathogenesis. Increased calcium levels can impair axonal transport, disrupt synaptic transmission, and ultimately lead to cell death. Given the potential role of calcium dyshomeostasis in AD, there is interest in testing the ability of already approved drugs targeting various calcium channels to affect amyloid pathology and other aspects of disease. The objective of this study was to test the effects of FDA-approved L-type calcium channel antagonist nimodipine on amyloid accumulation and dystrophic neurite formation in 5XFAD mice, a mouse model of amyloid pathology. 5XFAD transgenic mice and non-transgenic littermates were treated with vehicle or nimodipine-containing chow from two to eight months of age, then brains were harvested and amyloid pathology assessed by immunoblot and immunofluorescence microscopy analyses. Nimodipine was well tolerated and crossed the blood brain barrier, as expected, but there was no effect on Aß accumulation or on the relative amount of neuritic dystrophy, as assessed by either immunoblot, dot blot or immunofluorescence imaging of Aß42 and dystrophic neurite marker LAMP1. While we conclude that nimodipine treatment is not likely to improve amyloid pathology or decrease neuritic dystrophy in AD, it is worth noting that nimodipine did not worsen the phenotype suggesting its use is safe in AD patients.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Modelos Animales de Enfermedad , Neuritas/efectos de los fármacos , Distrofias Neuroaxonales/tratamiento farmacológico , Nimodipina/administración & dosificación , Placa Amiloide/tratamiento farmacológico , Administración Oral , Enfermedad de Alzheimer/patología , Animales , Bloqueadores de los Canales de Calcio/administración & dosificación , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Neuritas/patología , Distrofias Neuroaxonales/patología , Placa Amiloide/patología
6.
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
7.
Oxid Med Cell Longev ; 2022: 8096009, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35116093

RESUMEN

The loss of transient receptor potential mucolipin 1 (TRPML1), an endosomal and lysosomal Ca2+-releasing channel, has been implicated in neurodegenerative disorders. Mounting evidence have shown that TRPML1 could clear intraneuronal amyloid-ß (Aß), which triggers a hypothesis that TRPML1 activation may be beneficial for axonal transport in Alzheimer's disease (AD). In this work, the functional roles of TRPML1 were studied in the APP/PS1 transgenic mice and Aß1-42-stimulated hippocampal neurons HT22. We found that lentivirus-mediated overexpression of TRPML1 was shown to promote an accumulation of autolysosomes and increase brain-derived neurotrophic factor (BDNF) transportation to the nucleus, suggesting an axon-protective function. More importantly, we found that TRPML1 also increased p62 that interacted with dynein. Lentivirus-mediated knockdown of p62 or inhibition of dynein by ciliobrevin D stimulation was found to reduce autolysosome formation and nuclear accumulation of BDNF in HT22 cells with Aß1-42 stimulation. Inhibition of p62 by XRK3F2 stimulation was observed to promote the death of hippocampal neurons of the APP/PS1 transgenic mice. TRPML1 recruited dynein by interacting with p62 to promote the autophagosome-lysosome fusion to mediate BDNF nuclear translocation to impede axon dystrophy in mice with Alzheimer-like phenotypes. In summary, these results demonstrate the presence of a TRPML1/p62/dynein regulatory network in AD, and activation of TRPML1 is required for axon protection to prevent neuroaxonal dystrophy.


Asunto(s)
Enfermedad de Alzheimer/patología , Autofagosomas/metabolismo , Lisosomas/metabolismo , Proteína Sequestosoma-1/metabolismo , Canales de Potencial de Receptor Transitorio/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/farmacología , Animales , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Dineínas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fragmentos de Péptidos/farmacología , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Sequestosoma-1/antagonistas & inhibidores , Proteína Sequestosoma-1/genética , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Canales de Potencial de Receptor Transitorio/genética
8.
Neurobiol Dis ; 165: 105649, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35122944

RESUMEN

BACKGROUND: PLA2G6-Associated Neurodegeneration (PLAN) is a rare neurodegenerative disease with autosomal recessive inheritance, which belongs to the NBIA (Neurodegeneration with Brain Iron Accumulation) group. Although the pathogenesis of the disease remains largely unclear, lipid peroxidation seems to play a central role in the pathogenesis. Currently, there is no cure for the disease. OBJECTIVE: In this work, we examined the presence of lipid peroxidation, iron accumulation and mitochondrial dysfunction in two cellular models of PLAN, patients-derived fibroblasts and induced neurons, and assessed the effects of α-tocopherol (vitamin E) in correcting the pathophysiological alterations in PLAN cell cultures. METHODS: Pathophysiological alterations were examined in fibroblasts and induced neurons generated by direct reprograming. Iron and lipofuscin accumulation were assessed using light and electron microscopy, as well as biochemical analysis techniques. Reactive Oxygen species production, lipid peroxidation and mitochondrial dysfunction were measured using specific fluorescent probes analysed by fluorescence microscopy and flow cytometry. RESULTS: PLAN fibroblasts and induced neurons clearly showed increased lipid peroxidation, iron accumulation and altered mitochondrial membrane potential. All these pathological features were reverted with vitamin E treatment. CONCLUSIONS: PLAN fibroblasts and induced neurons reproduce the main pathological alterations of the disease and provide useful tools for disease modelling. The main pathological alterations were corrected by Vitamin E supplementation in both models, suggesting that blocking lipid peroxidation progression is a critical therapeutic target.


Asunto(s)
Distrofias Neuroaxonales , Enfermedades Neurodegenerativas , Fosfolipasas A2 Grupo VI/metabolismo , Humanos , Hierro/metabolismo , Peroxidación de Lípido , Mitocondrias/metabolismo , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/metabolismo , Vitamina E/metabolismo , Vitamina E/farmacología
9.
J Clin Lab Anal ; 36(3): e24253, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35092705

RESUMEN

BACKGROUND: Infantile neuroaxonal dystrophy is an autosomal recessive neurological disorder. Individuals with infantile neuroaxonal dystrophy experience progressive loss of vision, mental skills and muscular control, and other variable clinical signs. Pathogenic variants in the PLA2G6 gene, encoding phospholipase A2, are recognized to be the fundamental reason for infantile neuroaxonal dystrophy. This study aimed to detect pathogenic variant in a consanguine Iranian family with infantile neuroaxonal dystrophy. METHODS: The mutation screening was done by whole exome sequencing followed by direct Sanger sequencing. RESULTS: We identified a homozygous insertion mutation, NM_003560: c.1548_1549insCG (p.G517Rfs*29) in exon 10 of PLA2G6 in the patient. The parents were heterozygous for variant. CONCLUSIONS: Because of the clinical heterogeneity and rarity of infantile neuroaxonal dystrophy, whole exome sequencing is critical to confirm the diagnosis and is an excellent tool for INAD management.


Asunto(s)
Distrofias Neuroaxonales , Fosfolipasas A2 Grupo VI/genética , Homocigoto , Humanos , Irán , Mutagénesis Insercional , Mutación/genética , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología
10.
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
11.
Int J Mol Sci ; 22(15)2021 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-34360586

RESUMEN

Copper, manganese, and iron are vital elements required for the appropriate development and the general preservation of good health. Additionally, these essential metals play key roles in ensuring proper brain development and function. They also play vital roles in the central nervous system as significant cofactors for several enzymes, including the antioxidant enzyme superoxide dismutase (SOD) and other enzymes that take part in the creation and breakdown of neurotransmitters in the brain. An imbalance in the levels of these metals weakens the structural, regulatory, and catalytic roles of different enzymes, proteins, receptors, and transporters and is known to provoke the development of various neurological conditions through different mechanisms, such as via induction of oxidative stress, increased α-synuclein aggregation and fibril formation, and stimulation of microglial cells, thus resulting in inflammation and reduced production of metalloproteins. In the present review, the authors focus on neurological disorders with psychiatric signs associated with copper, iron, and manganese excess and the diagnosis and potential treatment of such disorders. In our review, we described diseases related to these metals, such as aceruloplasminaemia, neuroferritinopathy, pantothenate kinase-associated neurodegeneration (PKAN) and other very rare classical NBIA forms, manganism, attention-deficit/hyperactivity disorder (ADHD), ephedrone encephalopathy, HMNDYT1-SLC30A10 deficiency (HMNDYT1), HMNDYT2-SLC39A14 deficiency, CDG2N-SLC39A8 deficiency, hepatic encephalopathy, prion disease and "prion-like disease", amyotrophic lateral sclerosis, Huntington's disease, Friedreich's ataxia, and depression.


Asunto(s)
Ceruloplasmina/deficiencia , Cobre/efectos adversos , Trastornos del Metabolismo del Hierro/patología , Hierro/efectos adversos , Manganeso/efectos adversos , Enfermedades Metabólicas/patología , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/patología , Humanos , Trastornos del Metabolismo del Hierro/inducido químicamente , Trastornos del Metabolismo del Hierro/etiología , Intoxicación por Manganeso/complicaciones , Enfermedades Metabólicas/inducido químicamente , Metaloproteínas/metabolismo , Distrofias Neuroaxonales/inducido químicamente , Enfermedades Neurodegenerativas/etiología , Estrés Oxidativo
12.
Clin Neurol Neurosurg ; 206: 106637, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34022688

RESUMEN

Variants of the C19ORF12-gene have been described in patients with spastic paraplegia type 43 and in patients with mitochondrial membrane protein-associated neurodegeneration (MPAN), a subtype of neurodegeneration associated with brain iron accumulation (NBIA). In both subtypes optic atrophy and neuropathy have been frequently described. This case report describes a patient with bilateral optic atrophy and severe distal muscle weakness based on motor neuropathy without involvement of the central nervous system. Exome sequencing revealed a homozygous pathogenic missense variant (c.187G>C;p.Ala63Pro) of the C19ORF12-gene while iron deposits were absent on repeat MR-imaging of the brain, thus showing that peripheral neuropathy and optic neuropathy can be the sole manifestations of the C19ORF12-related disease spectrum whereby iron accumulation in the brain may be absent.


Asunto(s)
Proteínas Mitocondriales/genética , Debilidad Muscular/genética , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Atrofias Ópticas Hereditarias/genética , Enfermedades del Sistema Nervioso Periférico/genética , Adulto , Humanos , Masculino , Mutación Missense
13.
Parkinsonism Relat Disord ; 84: 98-104, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33607528

RESUMEN

INTRODUCTION: Mitochondrial membrane protein-associated neurodegeneration (MPAN) is a rare neurological syndrome caused by pathogenic variants in the C19orf12 and is characterized by iron deposition in the basal ganglia and substantia nigra. Only a limited number of cohort studies were published to date and the prevalence of MPAN remains uncertain. METHODS: Recruited subjects with MPAN in Russia were diagnosed by whole-exome sequencing or Sanger sequencing of the C19orf12 gene. Data of over 14000 whole exome sequencing analyses was used to calculate the estimated disease frequency. RNA analysis was performed by RT-PCR. QSVanalyzer software was used to quantify the allelic disbalance. RESULTS: We describe the clinical and molecular characterizations of 17 patients with MPAN. DNA analysis detected three previously undescribed pathogenic/likely pathogenic variants in the C19orf12 gene. The estimated disease frequency was calculated to be 1:619150. We describe unusual clinical observations in several cases. One patient showed severe neurogenic muscle weakness along with a lack of marked spasticity or optic nerve atrophy. In another mild clinical case with the NM_001031726.3:c.204_214del (p.(Gly69Argfs*10)) variant in a heterozygous state, a marked allelic disbalance was observed on the RNA level with reduced expression level of the wild-type allele. Thus, this case became the first one of a possible regulatory variant causing MPAN. CONCLUSION: We reported a detailed clinical and molecular characterization of the third-largest MPAN cohort. We expanded the mutational and clinical spectrum of MPAN. Moreover, we calculated the estimated MPAN frequency in the Russian population for the first time.


Asunto(s)
Globo Pálido/patología , Trastornos del Metabolismo del Hierro , Proteínas de la Membrana , Membranas Mitocondriales , Proteínas Mitocondriales , Distrofias Neuroaxonales , Sustancia Negra/patología , Adolescente , Adulto , Niño , Femenino , Globo Pálido/diagnóstico por imagen , Humanos , Trastornos del Metabolismo del Hierro/epidemiología , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Trastornos del Metabolismo del Hierro/fisiopatología , Imagen por Resonancia Magnética , Distrofias Neuroaxonales/epidemiología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Distrofias Neuroaxonales/fisiopatología , Estudios Retrospectivos , Federación de Rusia/epidemiología , Sustancia Negra/diagnóstico por imagen , Secuenciación del Exoma
14.
Cell Mol Life Sci ; 78(7): 3355-3367, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33439270

RESUMEN

Neuroferritinopathy is a rare autosomal dominant inherited movement disorder caused by alteration of the L-ferritin gene that results in the production of a ferritin molecule that is unable to properly manage iron, leading to the presence of free redox-active iron in the cytosol. This form of iron has detrimental effects on cells, particularly severe for neuronal cells, which are highly sensitive to oxidative stress. Although very rare, the disorder is notable for two reasons. First, neuroferritinopathy displays features also found in a larger group of disorders named Neurodegeneration with Brain Iron Accumulation (NBIA), such as iron deposition in the basal ganglia and extrapyramidal symptoms; thus, the elucidation of its pathogenic mechanism may contribute to clarifying the incompletely understood aspects of NBIA. Second, neuroferritinopathy shows the characteristic signs of an accelerated process of aging; thus, it can be considered an interesting model to study the progress of aging. Here, we will review the clinical and neurological features of neuroferritinopathy and summarize biochemical studies and data from cellular and animal models to propose a pathogenic mechanism of the disorder.


Asunto(s)
Apoferritinas/metabolismo , Trastornos del Metabolismo del Hierro/patología , Hierro/metabolismo , Distrofias Neuroaxonales/patología , Animales , Humanos , Trastornos del Metabolismo del Hierro/metabolismo , Distrofias Neuroaxonales/metabolismo
15.
Neuropathol Appl Neurobiol ; 47(1): 26-42, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-32464705

RESUMEN

AIMS: Neuroferritinopathy (NF) or hereditary ferritinopathy (HF) is an autosomal dominant movement disorder due to mutation in the light chain of the iron storage protein ferritin (FTL). HF is the only late-onset neurodegeneration with brain iron accumulation disorder and study of HF offers a unique opportunity to understand the role of iron in more common neurodegenerative syndromes. METHODS: We carried out pathological and biochemical studies of six individuals with the same pathogenic FTL mutation. RESULTS: CNS pathological changes were most prominent in the basal ganglia and cerebellar dentate, echoing the normal pattern of brain iron accumulation. Accumulation of ferritin and iron was conspicuous in cells with a phenotype suggesting oligodendrocytes, with accompanying neuronal pathology and neuronal loss. Neurons still survived, however, despite extensive adjacent glial iron deposition, suggesting neuronal loss is a downstream event. Typical age-related neurodegenerative pathology was not normally present. Uniquely, the extensive aggregates of ubiquitinated ferritin identified indicate that abnormal FTL can aggregate, reflecting the intrinsic ability of FTL to self-assemble. Ferritin aggregates were seen in neuronal and glial nuclei showing parallels with Huntington's disease. There was neither evidence of oxidative stress activation nor any significant mitochondrial pathology in the affected basal ganglia. CONCLUSIONS: HF shows hallmarks of a protein aggregation disorder, in addition to iron accumulation. Degeneration in HF is not accompanied by age-related neurodegenerative pathology and the lack of evidence of oxidative stress and mitochondrial damage suggests that these are not key mediators of neurodegeneration in HF, casting light on other neurodegenerative diseases characterized by iron deposition.


Asunto(s)
Apoferritinas/metabolismo , Encéfalo/efectos de los fármacos , Trastornos del Metabolismo del Hierro/metabolismo , Hierro/metabolismo , Distrofias Neuroaxonales/metabolismo , Animales , Apoferritinas/química , Apoferritinas/genética , Encéfalo/patología , Modelos Animales de Enfermedad , Ferritinas/química , Ferritinas/genética , Ferritinas/metabolismo , Humanos , Trastornos del Metabolismo del Hierro/patología , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mutación/genética , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/patología , Estrés Oxidativo/efectos de los fármacos , Agregado de Proteínas/fisiología
17.
Mol Genet Genomics ; 296(1): 235-242, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33159255

RESUMEN

Sporadic occurrences of neurodegenerative disorders including neuroaxonal dystrophy (NAD) have been previously reported in sheep. However, so far no causative genetic variant has been found for ovine NAD. The aim of this study was to characterize the phenotype and the genetic aetiology of an early-onset neurodegenerative disorder observed in several lambs of purebred Swaledale sheep, a native English breed. Affected lambs showed progressive ataxia and stiff gait and subsequent histopathological analysis revealed the widespread presence of axonal spheroid indicating neuronal degeneration. Thus, the observed clinical phenotype could be explained by a novel form of NAD. After SNP genotyping and subsequent linkage mapping within a paternal half-sib pedigree with a total of five NAD-affected lambs, we identified two loss-of-function variants by whole-genome sequencing in the ovine PLA2G6 gene situated in a NAD-linked genome region on chromosome 3. All cases were carriers of a compound heterozygous splice site variant in intron 2 and a nonsense variant in exon 8. Herein we present evidence for the occurrence of a familial novel form of recessively inherited NAD in sheep due to allelic heterogeneity at PLA2G6. This study reports two pathogenic variants in PLA2G6 causing a novel form of NAD in Swaledale sheep which enables selection against this fatal disorder.


Asunto(s)
Precursor de Proteína beta-Amiloide/genética , Fosfolipasas A2 Grupo VI/genética , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/veterinaria , Polimorfismo de Nucleótido Simple , Enfermedades de las Ovejas/genética , Empalme Alternativo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Secuencia de Bases , Mapeo Cromosómico , Cromosomas de los Mamíferos/química , Exones , Femenino , Expresión Génica , Ligamiento Genético , Fosfolipasas A2 Grupo VI/deficiencia , Heterocigoto , Intrones , Masculino , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Ovinos , Enfermedades de las Ovejas/metabolismo , Enfermedades de las Ovejas/patología , Oveja Doméstica , Secuenciación Completa del Genoma
19.
Ann Clin Transl Neurol ; 7(8): 1436-1442, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32767480

RESUMEN

FBXO7 is implicated in the ubiquitin-proteasome system and parkin-mediated mitophagy. FBXO7defects cause a levodopa-responsive parkinsonian-pyramidal syndrome(PPS). METHODS: We investigated the disease molecular bases in a child with PPS and brain iron accumulation. RESULTS: A novel homozygous c.368C>G (p.S123*) FBXO7 mutation was identified in a child with spastic paraplegia, epilepsy, cerebellar degeneration, levodopa nonresponsive parkinsonism, and brain iron deposition. Patient's fibroblasts assays demonstrated an absence of FBXO7 RNA expression leading to impaired proteasome degradation and accumulation of poly-ubiquitinated proteins. CONCLUSION: This novel FBXO7 phenotype associated with impaired proteasome activity overlaps with neurodegeneration with brain iron accumulation disorders.


Asunto(s)
Proteínas F-Box/genética , Trastornos del Metabolismo del Hierro , Distrofias Neuroaxonales , Trastornos Parkinsonianos , Complejo de la Endopetidasa Proteasomal/metabolismo , Adulto , Consanguinidad , Epilepsia/enzimología , Epilepsia/genética , Epilepsia/patología , Epilepsia/fisiopatología , Femenino , Humanos , Trastornos del Metabolismo del Hierro/enzimología , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Trastornos del Metabolismo del Hierro/fisiopatología , Distrofias Neuroaxonales/enzimología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Distrofias Neuroaxonales/fisiopatología , Paraplejía/enzimología , Paraplejía/genética , Paraplejía/patología , Paraplejía/fisiopatología , Trastornos Parkinsonianos/enzimología , Trastornos Parkinsonianos/genética , Trastornos Parkinsonianos/patología , Trastornos Parkinsonianos/fisiopatología , Degeneraciones Espinocerebelosas/enzimología , Degeneraciones Espinocerebelosas/genética , Degeneraciones Espinocerebelosas/patología , Degeneraciones Espinocerebelosas/fisiopatología , Síndrome , Adulto Joven
20.
Sci Rep ; 10(1): 9161, 2020 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-32514050

RESUMEN

Cerebrospinal fluid (CSF) biomarkers are useful in the diagnosis and the prediction of progression of several neurodegenerative diseases. Among them, CSF neurofilament light (NfL) protein has particular interest, as its levels reflect neuroaxonal degeneration, a common feature in various neurodegenerative diseases. In the present study, we analyzed NfL levels in the CSF of 535 participants of the SPIN (Sant Pau Initiative on Neurodegeneration) cohort including cognitively normal participants, patients with Alzheimer disease (AD), Down syndrome (DS), frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), dementia with Lewy bodies (DLB), progressive supranuclear palsy (PSP) and corticobasal syndrome (CBS). We evaluated the differences in CSF NfL accross groups and its association with other CSF biomarkers and with cognitive scales. All neurogenerative diseases showed increased levels of CSF NfL, with the highest levels in patients with ALS, FTD, CBS and PSP. Furthermore, we found an association of CSF NfL levels with cognitive impairment in patients within the AD and FTD spectrum and with AD pathology in DLB and DS patients. These results have implications for the use of NfL as a marker in neurodegenerative diseases.


Asunto(s)
Enfermedades Neurodegenerativas/diagnóstico , Proteínas de Neurofilamentos/líquido cefalorraquídeo , Anciano , Biomarcadores/líquido cefalorraquídeo , Estudios de Cohortes , Progresión de la Enfermedad , Diagnóstico Precoz , Femenino , Estudios de Seguimiento , Humanos , Masculino , Distrofias Neuroaxonales/diagnóstico , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...