Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
Mol Psychiatry ; 24(9): 1369-1382, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-30899091

RESUMEN

Alzheimer's disease (AD) is characterized by the presence of neuritic plaques in which dystrophic neurites (DNs) are typical constituents. We recently showed that DNs labeled by antibodies to the tubular endoplasmic reticulum (ER) protein reticulon-3 (RTN3) are enriched with clustered tubular ER. However, multi-vesicle bodies are also found in DNs, suggesting that different populations of DNs exist in brains of AD patients. To understand how different DNs evolve to surround core amyloid plaques, we monitored the growth of DNs in AD mouse brains (5xFAD and APP/PS1ΔE9 mice) by multiple approaches, including two-dimensional and three-dimensional (3D) electron microscopy (EM). We discovered that a pre-autophagosome protein ATG9A was enriched in DNs when a plaque was just beginning to develop. ATG9A-positive DNs were often closer to the core amyloid plaque, whereas RTN3 immunoreactive DNs were mostly located in the outer layers of ATG9A-positive DNs. Proteins such as RAB7 and LC3 appeared in DNs at later stages during plaque growth, likely accumulated as a part of large autophagy vesicles, and were distributed relatively furthest from the core amyloid plaque. Reconstructing the 3D structure of different morphologies of DNs revealed that DNs in AD mouse brains were constituted in three layers that are distinct by enriching different types of vesicles, as validated by immune-EM methods. Collectively, our results provide the first evidence that DNs evolve from dysfunctions of pre-autophagosomes, tubular ER, mature autophagosomes, and the ubiquitin proteasome system during plaque growth.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Neuritas/patología , Distrofias Neuroaxonales/patología , Enfermedad de Alzheimer/fisiopatología , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Proteínas Relacionadas con la Autofagia/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Retículo Endoplásmico/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Distrofias Neuroaxonales/clasificación , Distrofias Neuroaxonales/diagnóstico por imagen , Placa Amiloide/metabolismo , Proteínas de Transporte Vesicular/metabolismo
8.
Toxicol Pathol ; 48(5): 694-701, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32476609

RESUMEN

Axonal dystrophy (AD) is a common age-related neurohistological finding in vertebrates that can be congenital or induced by xenobiotics, vitamin E deficiency, or trauma/compression. To understand the incidence and location of AD as a background finding in Beagle dogs used in routine toxicity studies, we examined central nervous system (CNS) and selected peripheral nervous system (PNS) tissues in twenty 18- to 24-month-old and ten 4- to 5-year-old control males and females. Both sexes were equally affected. The cuneate, gracile, and cochlear nuclei and the cerebellar white matter (rostral vermis) were the most common locations for AD. Incidence of AD increased with age in the cuneate nucleus, cerebellar white matter (rostral vermis), trigeminal nuclei/tracts, and lumbar spinal cord. Axonal dystrophy in the CNS was not accompanied by neuronal degeneration/necrosis, nerve fiber degeneration, and/or glial reaction. Axonal dystrophy was not observed in the PNS (sciatic nerve, vagus nerve branches, or gastrointestinal mural autonomic plexuses).


Asunto(s)
Encéfalo/patología , Enfermedades de los Perros/patología , Distrofias Neuroaxonales/patología , Médula Espinal/patología , Animales , Sistema Nervioso Central/patología , Modelos Animales de Enfermedad , Perros , Femenino , Masculino , Bulbo Raquídeo/patología , Degeneración Nerviosa/patología
9.
Vet Pathol ; 57(1): 172-182, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31272300

RESUMEN

Genetically engineered mouse lines on a C57BL/6J background are widely employed as preclinical models to study neurodegenerative human disorders and brain tumors. However, because of the lack of comprehensive data on the spontaneous background neuropathology of the C57BL/6J strain, discriminating between naturally occurring changes and lesions caused by experimental mutations can be challenging. In this context, this study aims at defining the spectrum and frequency of spontaneous brain changes in a large cohort of C57BL/6J mice and their association with specific biological variables, including age and sex. Brains from 203 experimentally naive and clinically unremarkable C57BL/6J mice were collected and analyzed by means of histopathology and immunohistochemistry. Mice ranged in age from 3 to 110 weeks with 89 females, 111 males, and 3 unknowns. Sixteen different spontaneous lesion categories were described in this cohort. Age-related neurodegenerative and/or neuroinflammatory findings represented the most common pathologic changes and included (1) Hirano-like inclusions in the thalamic neurons, (2) neuroaxonal dystrophy in the medulla oblongata, (3) periodic acid-Schiff-positive granular deposits in the neuropil of the hippocampus, and (4) progressive neuroinflammation characterized by microgliosis and astrogliosis. Neoplastic conditions, developmental abnormalities, and circulatory disorders were rarely observed incidental findings. In conclusion, this study describes spontaneous age-related brain lesions of the C57BL/6J mouse and provides a reference for evaluating and interpreting the neuropathological phenotype in genetically engineered mouse models developed and maintained on this congenic background.


Asunto(s)
Envejecimiento/patología , Distrofias Neuroaxonales/veterinaria , Enfermedades Neurodegenerativas/veterinaria , Enfermedades de los Roedores/patología , Animales , Encéfalo/patología , Femenino , Cuerpos de Inclusión/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Distrofias Neuroaxonales/patología , Enfermedades Neurodegenerativas/patología , Neuronas/patología , Fenotipo
10.
Biochemistry ; 58(18): 2318-2325, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30986045

RESUMEN

Neuroferritinopathy is a rare, adult-onset, dominantly inherited movement disorder caused by mutations in the ferritin gene. A ferritin light-chain variant related to neuroferritinopathy, in which alanine 96 is replaced with threonine (A96T), was expressed in Escherichia coli, purified, and characterized. The circular dichroism, analytical ultracentrifugation, and small-angle X-ray scattering studies have shown that both the subunit structure and the assembly of A96T are the same as those of wild-type human ferritin light chain (HuFTL). The iron-incorporation ability was also comparable to that of HuFTL. Although the structural stability against heat, acid, and denaturant was reduced, the structure was sufficiently stable under physiological conditions. The most remarkable defects observed for A96T were a lower refolding efficiency and a stronger propensity to aggregate. The possible relationship between folding deficiency and disease is discussed.


Asunto(s)
Apoferritinas/química , Ferritinas/química , Trastornos del Metabolismo del Hierro/metabolismo , Distrofias Neuroaxonales/metabolismo , Agregación Patológica de Proteínas , Pliegue de Proteína , Apoferritinas/genética , Apoferritinas/metabolismo , Dicroismo Circular , Ferritinas/genética , Ferritinas/metabolismo , Humanos , Concentración de Iones de Hidrógeno , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Microscopía Electrónica de Transmisión , Mutación Missense , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Estabilidad Proteica , Dispersión del Ángulo Pequeño , Temperatura , Difracción de Rayos X
11.
J Cell Physiol ; 233(12): 9179-9190, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30076709

RESUMEN

Nuclear receptor coactivator 4 mediated ferritinophagy is an autophagic phenomenon that specifically involves ferritin to release intracellular free iron. Ferritinophagy is implicated in maintaining efficient erythropoiesis. Notably, ferritinophagy also plays a central role in driving some pathological processes, including Parkinson's disease (PD) and urinary tract infections. Some evidence has demonstrated that ferritinophagy is critical to induce ferroptosis. Ferroptosis is a newly nonapoptotic form of cell death, characterized by the accumulation of iron-based lipid reactive oxygen species. Ferroptosis plays an important role in inhibiting some types of cancers, such as hepatocellular carcinoma, pancreatic carcinoma, prostate cancer, and breast cancer. Conversely, the activation of ferroptosis accelerates neurodegeneration diseases, including PD and Alzheimer's disease. Therefore, in this review, we summarize the regulatory mechanisms related to ferritinophagy and ferroptosis. Moreover, the distinctive effects of ferritinophagy in human erythropoiesis and some pathologies, coupled with the promotive or inhibitory role of tumorous and neurodegenerative diseases mediated by ferroptosis, are elucidated. Obviously, activating or inhibiting ferroptosis could be exploited to achieve desirable therapeutic effects on diverse cancers and neurodegeneration diseases. Interrupting ferritinophagy to control iron level might provide a potentially therapeutic avenue to suppress urinary tract infections.


Asunto(s)
Ferritinas/metabolismo , Trastornos del Metabolismo del Hierro/metabolismo , Hierro/metabolismo , Neoplasias/metabolismo , Distrofias Neuroaxonales/metabolismo , Autofagia/genética , Eritropoyesis/genética , Ferritinas/efectos adversos , Humanos , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Neoplasias/clasificación , Neoplasias/etiología , Neoplasias/patología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Coactivadores de Receptor Nuclear/genética , Enfermedad de Parkinson/genética , Enfermedad de Parkinson/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Infecciones Urinarias/genética , Infecciones Urinarias/metabolismo
12.
IUBMB Life ; 69(6): 423-434, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28276141

RESUMEN

Ferroptosis is a recently described form of regulated necrotic cell death, which appears to contribute to a number of diseases, such as tissue ischemia/reperfusion injury, acute renal failure, and neurodegeneration. A hallmark of ferroptosis is iron-dependent lipid peroxidation, which can be inhibited by the key ferroptosis regulator glutathione peroxidase 4(Gpx4), radical trapping antioxidants and ferroptosis-specific inhibitors, such as ferrostatins and liproxstatins, as well as iron chelation. Although great strides have been made towards a better understanding of the proximate signals of distinctive lipid peroxides in ferroptosis, still little is known about the mechanistic implication of iron in the ferroptotic process. Hence, this review aims at summarizing recent advances in our understanding to what is known about enzymatic and nonenzymatic routes of lipid peroxidation, the involvement of iron in this process and the identification of novel players in ferroptotic cell death. Additionally, we review early works carried out long time before the term "ferroptosis" was actually introduced but which were instrumental in a better understanding of the role of ferroptosis in physiological and pathophysiological contexts. © 2017 IUBMB Life, 69(6):423-434, 2017.


Asunto(s)
Antioxidantes/farmacología , Glutatión Peroxidasa/antagonistas & inhibidores , Trastornos del Metabolismo del Hierro/metabolismo , Hierro/metabolismo , Distrofias Neuroaxonales/metabolismo , Insuficiencia Renal/metabolismo , Daño por Reperfusión/metabolismo , Animales , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Ciclohexilaminas/farmacología , Glutatión Peroxidasa/genética , Glutatión Peroxidasa/metabolismo , Humanos , Quelantes del Hierro/uso terapéutico , Trastornos del Metabolismo del Hierro/tratamiento farmacológico , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Peroxidación de Lípido/efectos de los fármacos , Necrosis/genética , Necrosis/metabolismo , Necrosis/patología , Distrofias Neuroaxonales/tratamiento farmacológico , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Fenilendiaminas/farmacología , Fosfolípido Hidroperóxido Glutatión Peroxidasa , Quinoxalinas/farmacología , Insuficiencia Renal/tratamiento farmacológico , Insuficiencia Renal/genética , Insuficiencia Renal/patología , Daño por Reperfusión/genética , Daño por Reperfusión/patología , Daño por Reperfusión/prevención & control , Compuestos de Espiro/farmacología
13.
J Med Genet ; 53(3): 180-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26668131

RESUMEN

BACKGROUND: Mutations in PLA2G6, which encodes the calcium-independent phospholipase A2 group VI, cause neurodegeneration and diffuse cortical Lewy body formation by a yet undefined mechanism. We assessed whether altered protein glycosylation due to abnormal Golgi morphology might be a factor in the pathology of this disease. METHODS: Three patients presented with PLA2G6-associated neurodegeneration (PLAN); two had infantile neuroaxonal dystrophy (INAD) and one had adult-onset dystonia-parkinsonism. We analysed protein N-linked and O-linked glycosylation in cerebrospinal fluid, plasma, urine, and cultured skin fibroblasts using high performance liquid chromatography (HPLC) and matrix-assisted laser desorption ionization--time of flight/mass spectrometry (MALDI-TOF/MS). We also assessed sialylation and Golgi morphology in cultured fibroblasts by immunofluorescence and performed rescue experiments using a lentiviral vector. RESULTS: The patients with INAD had PLA2G6 mutations NM_003560.2: c.[950G>T];[426-1077dup] and c.[1799G>A];[2221C>T] and the patient with dystonia-parkinsonism had PLA2G6 mutations NM_003560.2: c.[609G>A];[2222G>A]. All three patients had altered Golgi morphology and abnormalities of protein O-linked glycosylation and sialylation in cultured fibroblasts that were rescued by lentiviral overexpression of wild type PLA2G6. CONCLUSIONS: Our findings add altered Golgi morphology, O-linked glycosylation and sialylation defects to the phenotypical spectrum of PLAN; these pathways are essential for correct processing and distribution of proteins. Lewy body and Tau pathology, two neuropathological features of PLAN, could emerge from these defects. Therefore, Golgi morphology, O-linked glycosylation and sialylation may play a role in the pathogenesis of PLAN and perhaps other neurodegenerative disorders.


Asunto(s)
Trastornos Distónicos/metabolismo , Trastornos Distónicos/patología , Aparato de Golgi/ultraestructura , Fosfolipasas A2 Grupo VI/deficiencia , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología , Trastornos Parkinsonianos/metabolismo , Trastornos Parkinsonianos/patología , Adulto , Células Cultivadas , Trastornos Distónicos/genética , Femenino , Fibroblastos/metabolismo , Fibroblastos/ultraestructura , Glicosilación , Aparato de Golgi/metabolismo , Fosfolipasas A2 Grupo VI/genética , Fosfolipasas A2 Grupo VI/metabolismo , Humanos , Lactante , Masculino , Mutación , Distrofias Neuroaxonales/genética , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Trastornos Parkinsonianos/genética , Sialiltransferasas/metabolismo
14.
Vet Pathol ; 54(5): 832-837, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28578630

RESUMEN

A novel form of neuroaxonal dystrophy is described in 3 Chihuahua pups, 2 of which were from the same litter. It was characterized not only by accumulation of numerous and widely distributed axonal swellings (spheroids) but also by a severe cavitating leukoencephalopathy. The dogs presented with progressive neurological signs, including gait abnormalities and postural reaction deficits. Magnetic resonance images and gross examination at necropsy revealed dilation of lateral ventricles and cerebral atrophy, accompanied by cavitation of the subcortical white matter. Histopathologically, severe axonal degeneration with formation of large spheroids was found in the cerebral and cerebellar white matter, thalamus, and brainstem nuclei. Small-caliber spheroids were observed in the cerebral and cerebellar gray matter. The telencephalic white matter had severe myelin loss and cavitation with relative sparing of the U-fibers. Different from previously reported cases of canine neuroaxonal dystrophy, in these Chihuahuas the spheroid distribution predominantly involved the white matter with secondary severe leukoencephalopathy.


Asunto(s)
Enfermedades de los Perros/diagnóstico , Leucoencefalopatías/veterinaria , Distrofias Neuroaxonales/veterinaria , Animales , Atrofia/diagnóstico por imagen , Atrofia/patología , Atrofia/veterinaria , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Enfermedades de los Perros/diagnóstico por imagen , Enfermedades de los Perros/patología , Perros , Femenino , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/patología , Imagen por Resonancia Magnética/veterinaria , Masculino , Distrofias Neuroaxonales/diagnóstico por imagen , Distrofias Neuroaxonales/patología
15.
Neurobiol Dis ; 87: 59-68, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26707700

RESUMEN

Aberrant brain iron deposition is observed in both common and rare neurodegenerative disorders, including those categorized as Neurodegeneration with Brain Iron Accumulation (NBIA), which are characterized by focal iron accumulation in the basal ganglia. Two NBIA genes are directly involved in iron metabolism, but whether other NBIA-related genes also regulate iron homeostasis in the human brain, and whether aberrant iron deposition contributes to neurodegenerative processes remains largely unknown. This study aims to expand our understanding of these iron overload diseases and identify relationships between known NBIA genes and their main interacting partners by using a systems biology approach. We used whole-transcriptome gene expression data from human brain samples originating from 101 neuropathologically normal individuals (10 brain regions) to generate weighted gene co-expression networks and cluster the 10 known NBIA genes in an unsupervised manner. We investigated NBIA-enriched networks for relevant cell types and pathways, and whether they are disrupted by iron loading in NBIA diseased tissue and in an in vivo mouse model. We identified two basal ganglia gene co-expression modules significantly enriched for NBIA genes, which resemble neuronal and oligodendrocytic signatures. These NBIA gene networks are enriched for iron-related genes, and implicate synapse and lipid metabolism related pathways. Our data also indicates that these networks are disrupted by excessive brain iron loading. We identified multiple cell types in the origin of NBIA disorders. We also found unforeseen links between NBIA networks and iron-related processes, and demonstrate convergent pathways connecting NBIAs and phenotypically overlapping diseases. Our results are of further relevance for these diseases by providing candidates for new causative genes and possible points for therapeutic intervention.


Asunto(s)
Ganglios Basales/metabolismo , Perfilación de la Expresión Génica/métodos , Trastornos del Metabolismo del Hierro/metabolismo , Distrofias Neuroaxonales/metabolismo , Transcriptoma , Adolescente , Anciano , Anciano de 80 o más Años , Animales , Ganglios Basales/patología , Niño , Preescolar , Modelos Animales de Enfermedad , Femenino , Proteína de la Hemocromatosis , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Lactante , Hierro/metabolismo , Trastornos del Metabolismo del Hierro/patología , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Transgénicos , Distrofias Neuroaxonales/patología , Neuronas/metabolismo , Neuronas/patología , Receptores de Transferrina/genética , Receptores de Transferrina/metabolismo , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Adulto Joven
16.
Neuropediatrics ; 47(2): 123-7, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26859818

RESUMEN

Neurodegeneration with brain iron accumulation (NBIA) is a heterogeneous group of single gene disorders with distinguished clinical phenotypes and definitive imaging findings. Beta propeller protein-associated neurodegeneration (BPAN) is a subentity of NBIA with X linked dominant inheritance. In this report, we describe a girl with autistic regression, seizures, intracranial calcification, iron accumulation in substantia nigra, and globi pallidi, and diagnosis of BPAN was established based on the identification of previously described disease causing variant in WD repeat domain 45 (WDR45) gene encoding for ß propeller protein. This is the first genetically proven case from India. BPAN is an underrecognized disorder and must be considered as a differential diagnosis in children with atypical Rett features and should be enlisted among the causes for autistic regression and intracranial calcification. Pediatricians must be aware of this rare entity for establishing early diagnosis, prognostication, and genetic counseling. Treatment is usually supportive. More research is needed to explore drugs in the management of BPAN that can facilitate the autophagy and promotes cytoprotection.


Asunto(s)
Trastorno Autístico/etiología , Enfermedades de los Ganglios Basales/genética , Enfermedades de los Ganglios Basales/patología , Calcinosis/etiología , Proteínas Portadoras/genética , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Ganglios Basales/metabolismo , Ganglios Basales/patología , Enfermedades de los Ganglios Basales/complicaciones , Enfermedades de los Ganglios Basales/metabolismo , Preescolar , Femenino , Humanos , Trastornos del Metabolismo del Hierro/complicaciones , Trastornos del Metabolismo del Hierro/metabolismo , Distrofias Neuroaxonales/complicaciones , Distrofias Neuroaxonales/metabolismo , Sustancia Negra/metabolismo , Sustancia Negra/patología
17.
Neuroradiology ; 58(10): 1035-1042, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27516098

RESUMEN

INTRODUCTION: Infantile neuroaxonal dystrophy (INAD), an autosomal recessive neurodegenerative disorder due to PLA2G6 mutation, is classified both as a PLA2G6-associated neurodegeneration (PLAN) disorder and as one of the neurodegeneration with brain iron accumulation (NBIA) disorders. Age of onset and clinical presentation in INAD is variable. Typically described imaging features of cerebellar atrophy, cerebellar cortex bright FLAIR signal, and globus pallidus iron deposition are variable or late findings. We characterize clinical and neuroimaging phenotypes in nine children with confirmed PLA2G6 mutations and show a useful imaging feature, clava hypertrophy, which may aid in earlier identification of patients. Measurements of the clava confirm actual enlargement, rather than apparent enlargement due to volume loss of the other brain stem structures. METHODS: A retrospective clinical and MRI review was performed. Brain stem measurements were performed and compared with age-matched controls. RESULTS: We identified nine patients, all with novel PLA2G6 gene mutations. MRI, available in eight, showed clava hypertrophy, regardless of age or the absence of other more typically described neuroimaging findings. Brain autopsy in our cohort confirmed prominent spheroid bodies in the clava nuclei. CONCLUSION: Clava hypertrophy is an important early imaging feature which may aid in indentification of children who would benefit from specific testing for PLA2G6 mutations.


Asunto(s)
Biometría/métodos , Fosfolipasas A2 Grupo VI/genética , Imagen por Resonancia Magnética/métodos , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Preescolar , Diagnóstico Diferencial , Femenino , Predisposición Genética a la Enfermedad/genética , Humanos , Hipertrofia , Lactante , Masculino , Distrofias Neuroaxonales/diagnóstico por imagen , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
18.
Neurobiol Dis ; 81: 134-43, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25772441

RESUMEN

Neuroferritinopathy is a rare, late-onset, dominantly inherited movement disorder caused by mutations in L-ferritin gene. It is characterized by iron and ferritin aggregate accumulation in brain, normal or low serum ferritin levels and high variable clinical feature. To date, nine causative mutations have been identified and eight of them are frameshift mutations determined by nucleotide(s) insertion in the exon 4 of L-ferritin gene altering the structural conformation of the C-terminus of the L-ferritin subunit. Acting in a dominant negative manner, mutations are responsible for an impairment of the iron storage efficiency of ferritin molecule. Here, we review the main characteristics of neuroferritinopathy and present a computational analysis of some representative recently defined mutations with the purpose to gain new information about the pathogenetic mechanism of the disorder. This is particularly important as neuroferritinopathy can be considered an interesting model to study the relationship between iron, oxidative stress and neurodegeneration.


Asunto(s)
Ferritinas , Trastornos del Metabolismo del Hierro , Distrofias Neuroaxonales , Animales , Encéfalo/metabolismo , Encéfalo/patología , Biología Computacional , Ferritinas/química , Ferritinas/genética , Ferritinas/metabolismo , Humanos , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/metabolismo , Trastornos del Metabolismo del Hierro/patología , Modelos Moleculares , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/metabolismo , Distrofias Neuroaxonales/patología
19.
Neurobiol Dis ; 81: 119-33, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25447222

RESUMEN

Neuroferritinopathy is a rare genetic disease with a dominant autosomal transmission caused by mutations of the ferritin light chain gene (FTL). It belongs to Neurodegeneration with Brain Iron Accumulation, a group of disorders where iron dysregulation is tightly associated with neurodegeneration. We studied the 498-499InsTC mutation which causes the substitution of the last 9 amino acids and an elongation of extra 16 amino acids at the C-terminus of L-ferritin peptide. An analysis with cyclic voltammetry on the purified protein showed that this structural modification severely reduces the ability of the protein to store iron. In order to analyze the impact of the mutation in vivo, we generated mouse models for the some pathogenic human FTL gene in FVB and C57BL/6J strains. Transgenic mice in the FVB background showed high accumulation of the mutated ferritin in brain where it correlated with increased iron deposition with age, as scored by magnetic resonance imaging. Notably, the accumulation of iron-ferritin bodies was accompanied by signs of oxidative damage. In the C57BL/6 background, both the expression of the mutant ferritin and the iron levels were lower than in the FVB strain. Nevertheless, also these mice showed oxidative alterations in the brain. Furthermore, post-natal hippocampal neurons obtained from these mice experienced a marked increased cell death in response to chronic iron overload and/or acute oxidative stress, in comparison to wild-type neurons. Ultrastructural analyses revealed an accumulation of lipofuscin granules associated with iron deposits, particularly enriched in the cerebellum and striatum of our transgenic mice. Finally, experimental subjects were tested throughout development and aging at 2-, 8- and 18-months for behavioral phenotype. Rotarod test revealed a progressive impaired motor coordination building up with age, FTL mutant old mice showing a shorter latency to fall from the apparatus, according to higher accumulation of iron aggregates in the striatum. Our data show that our 498-499InsTC mouse models recapitulate early pathological and clinical traits of the human neuroferritinopathy, thus providing a valuable model for the study of the disease. Finally, we propose a mechanistic model of lipofuscine formation that can account for the etiopathogenesis of human neuroferritinopathy.


Asunto(s)
Apoferritinas/genética , Encéfalo/patología , Trastornos del Metabolismo del Hierro/etiología , Distrofias Neuroaxonales , Enfermedades Neurodegenerativas/etiología , Trastornos Psicomotores/etiología , Factores de Edad , Animales , Apoferritinas/metabolismo , Encéfalo/metabolismo , Muerte Celular/genética , Células Cultivadas , Daño del ADN/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Hipocampo/citología , Humanos , Trastornos del Metabolismo del Hierro/complicaciones , Trastornos del Metabolismo del Hierro/genética , Trastornos del Metabolismo del Hierro/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Moleculares , Distrofias Neuroaxonales/complicaciones , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Neuronas/efectos de los fármacos , Neuronas/metabolismo
20.
Eur J Neurol ; 22(1): 178-86, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25164370

RESUMEN

BACKGROUND AND PURPOSE: Mutations in the PLA2G6 gene are causative of PLA2G6-associated neurodegeneration (PLAN), a spectrum of neurodegenerative conditions including infantile, childhood and adult onset forms. METHODS: Seventeen North African patients with a clinical suspicion of infantile-onset PLAN underwent clinical, neurophysiological and neuroimaging examinations, and PLA2G6 sequencing. Haplotype analysis was performed to date the identified founder mutation. RESULTS: All patients carried biallelic mutations in PLA2G6. Sixteen children had the commonest form of infantile-onset PLAN, with early onset of psychomotor regression, hypotonia, pyramidal and cerebellar signs, and abnormal ocular movements. The phenotype was highly homogeneous, with rapid development of severe spastic tetraparesis, cognitive impairment and optic atrophy. Neuroimaging showed cerebellar atrophy and claval hypertrophy to be the commonest and earliest signs, whilst cerebellar cortex hyperintensity and pallidal iron deposition were later findings. Motor or sensory-motor neuropathy and electroencephalogram fast rhythms were also frequent. Nine patients from six families shared the same founder mutation (p.V691del) which probably arose by the late seventeenth century. Only one patient fitted the diagnosis of the much rarer childhood-onset PLAN. Despite the early onset (18 months), clinical progression was slower, with behavioral disturbances and dystonia. Typical features of infantile-onset PLAN such as hypotonia, nystagmus/strabismus, optic atrophy, electroencephalogram fast rhythms and motor neuropathy were absent. Cerebellar atrophy, claval hypertrophy and pallidal hypointensity were evident at brain magnetic resonance imaging. This patient carried a missense variant predicted to be less deleterious. CONCLUSIONS: The PLAN-associated phenotypes and the challenges of diagnosing the childhood-onset form are delineated, and a common North African founder mutation is identifed.


Asunto(s)
Edad de Inicio , Fosfolipasas A2 Grupo VI/genética , Mutación/genética , Distrofias Neuroaxonales/clasificación , Atrofia/patología , Niño , Preescolar , Electroencefalografía , Electromiografía , Femenino , Efecto Fundador , Humanos , Lactante , Libia , Imagen por Resonancia Magnética , Masculino , Distrofias Neuroaxonales/genética , Distrofias Neuroaxonales/patología , Distrofias Neuroaxonales/fisiopatología , Linaje , Fenotipo , Túnez
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA