Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 76
Filtrar
1.
Neuropediatrics ; 51(2): 160-163, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31766059

RESUMEN

CASE: We are reporting the third unrelated case of cerebral aspartate-glutamate carrier isoform 1 (AGC1) deficiency. Patient is a 21-month-old Yemeni male who presented with refractory seizure disorder and developmental arrest. Neuroimaging showed cerebral volume loss and diminished N-acetylaspartate (NAA) peak. Whole exome sequencing revealed a homozygous novel missense variant in the SLC25A12 gene. Patient's seizure frequency abated drastically following initiation of ketogenic diet. DISCUSSION AND CONCLUSION: Cerebral AGC1 deficiency results in dysfunction of mitochondrial malate aspartate shuttle, thereby prohibiting myelin synthesis. There are significant phenotypic commonalities between our patient and previously reported cases including intractable epilepsy, psychomotor delay, cerebral atrophy, and diminished NAA peak. Our report also provides evidence regarding beneficial effect of ketogenic diet in this rare neurometabolic epilepsy.


Asunto(s)
Sistemas de Transporte de Aminoácidos Acídicos/deficiencia , Antiportadores/deficiencia , Dieta Cetogénica , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico , Enfermedades Mitocondriales/diagnóstico , Proteínas de Transporte de Membrana Mitocondrial/genética , Trastornos Psicomotores/diagnóstico , Adulto , Sistemas de Transporte de Aminoácidos Acídicos/genética , Antiportadores/genética , Epilepsia Refractaria/dietoterapia , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/dietoterapia , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Humanos , Masculino , Enfermedades Mitocondriales/dietoterapia , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/fisiopatología , Mutación Missense , Isoformas de Proteínas , Trastornos Psicomotores/dietoterapia , Trastornos Psicomotores/genética , Trastornos Psicomotores/fisiopatología , Secuenciación del Exoma , Adulto Joven
2.
Clin Genet ; 93(5): 1087-1092, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29388673

RESUMEN

Hypomyelinating leukodystrophies (HLDs) affect the white matter of the central nervous system and manifest as neurological disorders. They are genetically heterogeneous. Very recently, biallelic variants in NKX6-2 have been suggested to cause a novel form of autosomal recessive HLD. Using whole-exome or whole-genome sequencing, we identified the previously reported c.196delC and c.487C>G variants in NKX6-2 in 3 and 2 unrelated index cases, respectively; the novel c.608G>A variant was identified in a sixth patient. All variants were homozygous in affected family members only. Our patients share a primary diagnosis of psychomotor delay, and they show spastic quadriparesis, nystagmus and hypotonia. Seizures and dysmorphic features (observed in 2 families each) represent an addition to the phenotype, while developmental regression (observed in 3 families) appears to be a notable and previously underestimated clinical feature. Our findings extend the clinical and mutational spectra associated with this novel form of HLD. Comparative analysis of our 10 patients and the 15 reported previously did, however, not reveal clear evidence for a genotype-phenotype correlation.


Asunto(s)
Predisposición Genética a la Enfermedad , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas de Homeodominio/genética , Convulsiones/genética , Adolescente , Niño , Preescolar , Exoma/genética , Femenino , Estudios de Asociación Genética , Heterogeneidad Genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Homocigoto , Humanos , Lactante , Masculino , Mutación , Fenotipo , Convulsiones/fisiopatología , Sustancia Blanca/patología , Secuenciación del Exoma , Secuenciación Completa del Genoma
3.
Rev Invest Clin ; 70(2): 68-75, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29718010

RESUMEN

BACKGROUND: Retinal vasculopathy with cerebral leukodystrophy (RVCL) is an adult-onset, autosomal dominant disease involving microvessels of the brain and eye resulting in central nervous system degeneration with visual disturbances, stroke, motor impairment, and cognitive decline. Frameshift mutations at the C-terminus of TREX1 gene are the molecular cause of this disorder. OBJECTIVES: The objective of this study is to present the different clinical manifestations of RVCL in three-related patients and to investigate the presence of TREX1 mutation in the extended genealogy. METHODS: Multidisciplinary testing was performed in three related patients. Based on their family history, the study was extended to 34 relatives from the same small community. Neurological evaluation, sequencing of TREX1, and presymptomatic diagnosis were offered to all participants. RESULTS: The patients exhibited the heterozygous TREX1 mutation p.V235Gfs*6, but with phenotypic variability. In addition, 15 relatives were identified as pre-manifest mutation carriers. The remaining participants did not carry the mutation. CONCLUSIONS: This is the figrst report of a large Mexican genealogy with RVCL, where the same TREX1 mutation causes a variation in organ involvement and clinical progression. The early identification and follow-up of individuals at risk may help provide insights into the basis for this variability in presentation.


Asunto(s)
Variación Biológica Poblacional , Exodesoxirribonucleasas/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Fosfoproteínas/genética , Enfermedades de la Retina/fisiopatología , Enfermedades Vasculares/fisiopatología , Femenino , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Heterocigoto , Humanos , Masculino , México , Persona de Mediana Edad , Mutación , Enfermedades de la Retina/diagnóstico , Enfermedades de la Retina/genética , Enfermedades Vasculares/diagnóstico , Enfermedades Vasculares/genética
4.
Glia ; 65(3): 489-501, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28063167

RESUMEN

The tumor overexpressed gene (TOG) protein is present in RNA granules that transport myelin basic protein (MBP) mRNA in oligodendrocyte processes to the myelin compartment. Its role was investigated by conditionally knocking it out (KO) in myelinating glia in vivo. TOG KO mice have severe motor deficits that are already apparent at the time of weaning. This phenotype correlates with a paucity of myelin in several CNS regions, the most severe being in the spinal cord. In the TOG KO optic nerve <30% of axons are myelinated. The number of oligodendrocytes in the corpus callosum, cerebellum, and cervical spinal cord is normal. In the absence of TOG, the most patent biochemical change is a large reduction in MBP content, yet normal amounts of MBP transcripts are found in the brain of affected animals. MBP transcripts are largely confined to the cell body of the oligodendrocytes in the TOG KO in contrast to the situation in wild type mice where they are found in the processes of the oligodendrocytes and in the myelin compartment. These findings indicate that MBP gene expression involves a post-transcriptional TOG-dependent step. TOG may be necessary for MBP mRNA assembly into translation permissive granules, and/or for transport to preferred sites of translation. GLIA 2017;65:489-501.


Asunto(s)
Regulación de la Expresión Génica/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas Asociadas a Microtúbulos/deficiencia , Oligodendroglía/patología , Animales , Células Cultivadas , Corteza Cerebral/citología , Modelos Animales de Enfermedad , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica de Transmisión , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Actividad Motora/genética , Proteínas de la Mielina/genética , Proteínas de la Mielina/metabolismo , Oligodendroglía/ultraestructura , Equilibrio Postural/genética
5.
Ann Neurol ; 77(1): 114-31, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25382142

RESUMEN

OBJECTIVE: Megalencephalic leukoencephalopathy with cysts (MLC) is a genetic disease characterized by infantile onset white matter edema and delayed onset neurological deterioration. Loss of MLC1 function causes MLC. MLC1 is involved in ion-water homeostasis, but its exact role is unknown. We generated Mlc1-null mice for further studies. METHODS: We investigated which brain cell types express MLC1, compared developmental expression in mice and men, and studied the consequences of loss of MLC1 in Mlc1-null mice. RESULTS: Like humans, mice expressed MLC1 only in astrocytes, especially those facing fluid-brain barriers. In mice, MLC1 expression increased until 3 weeks and then stabilized. In humans, MLC1 expression was highest in the first year, decreased, and stabilized from approximately 5 years. Mlc1-null mice had early onset megalencephaly and increased brain water content. From 3 weeks, abnormal astrocytes were present with swollen processes abutting fluid-brain barriers. From 3 months, widespread white matter vacuolization with intramyelinic edema developed. Mlc1-null astrocytes showed slowed regulatory volume decrease and reduced volume-regulated anion currents, which increased upon MLC1 re-expression. Mlc1-null astrocytes showed reduced expression of adhesion molecule GlialCAM and chloride channel ClC-2, but no substantial changes in other known MLC1-interacting proteins. INTERPRETATION: Mlc1-null mice replicate early stages of the human disease with early onset intramyelinic edema. The cellular functional defects, described for human MLC, were confirmed. The earliest change was astrocytic swelling, substantiating that in MLC the primary defect is in volume regulation by astrocytes. MLC1 expression affects expression of GlialCAM and ClC-2. Abnormal interplay between these proteins is part of the pathomechanisms of MLC.


Asunto(s)
Quistes/genética , Quistes/patología , Quistes/fisiopatología , Regulación del Desarrollo de la Expresión Génica/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Adolescente , Adulto , Factores de Edad , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Astrocitos/patología , Edema Encefálico/etiología , Cerebelo/patología , Corteza Cerebral/citología , Corteza Cerebral/patología , Niño , Preescolar , Quistes/metabolismo , Modelos Animales de Enfermedad , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Humanos , Lactante , Recién Nacido , Potenciales de la Membrana/genética , Proteínas de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/genética , Equilibrio Postural/genética , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Trastornos de la Sensación/genética , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Sustancia Blanca/ultraestructura , Adulto Joven
6.
Hum Mol Genet ; 22(21): 4405-16, 2013 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23793458

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy caused by mutations in either MLC1 or GLIALCAM genes and is associated with myelin and astrocyte vacuolation. It has been suggested that MLC is caused by impaired cell volume regulation as a result of defective activation of astrocytic volume-regulated anion currents (VRAC). GlialCAM brings MLC1 and the ClC-2 Cl(-) channel to cell-cell junctions, even though the role of ClC-2 in MLC disease remains incompletely understood. To gain insights into the biological role of GlialCAM in the pathogenesis of MLC disease, here we analyzed the gain- and loss-of-function phenotypes of GlialCAM in Hela cells and primary astrocytes, focusing on its interaction with the MLC1 protein. Unexpectedly, GlialCAM ablation provoked intracellular accumulation and reduced expression of MLC1 at the plasma membrane. Conversely, over-expression of GlialCAM increased the cellular stability of mutant MLC1 variants. Reduction in GlialCAM expression resulted in defective activation of VRAC and augmented vacuolation, phenocopying MLC1 mutations. Importantly, over-expression of GlialCAM together with MLC1 containing MLC-related mutations was able to reactivate VRAC currents and to reverse the vacuolation caused in the presence of mutant MLC1. These results indicate a previously unrecognized role of GlialCAM in facilitating the biosynthetic maturation and cell surface expression of MLC1, and suggest that pharmacological strategies aimed to increase surface expression of MLC1 and/or VRAC activity may be beneficial for MLC patients.


Asunto(s)
Astrocitos/fisiología , Canales de Cloruro/fisiología , Quistes/fisiopatología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Proteínas/genética , Proteínas/metabolismo , Animales , Astrocitos/ultraestructura , Canales de Cloruro CLC-2 , Proteínas de Ciclo Celular , Quistes/genética , Variación Genética , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutación , Fenotipo , Estabilidad Proteica , Interferencia de ARN , Ratas , Vacuolas/fisiología
8.
Genet Couns ; 26(2): 233-6, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26349194

RESUMEN

MLC or Van der Knaap disease is a rare entity, a rare and genetically heterogeneous cerebral white matter disease. It is characterized by the presence of macrocephaly, epilepsy and a slowly progressive spastic cerebellar syndrome. It is an autosomal recessive disease caused from mutations of MLC1 gene. In the current case report, a case with MLC who had a homozygous mutation (c.448delC, p.Leul50 ser fsX11) on exon 6 of MLC1 gene is presented.


Asunto(s)
Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas de la Membrana/genética , Preescolar , Quistes/patología , Quistes/fisiopatología , Exones/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Homocigoto , Humanos , Masculino , Mutación
9.
J Neurochem ; 124(3): 347-62, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23216354

RESUMEN

The mitochondrial transporter of aspartate-glutamate Aralar/AGC1 is a regulatory component of the malate-aspartate shuttle. Aralar deficiency in mouse and human causes a shutdown of brain shuttle activity and global cerebral hypomyelination. A lack of neurofilament-labeled processes is detected in the cerebral cortex, but whether different types of neurons are differentially affected by Aralar deficiency is still unknown. We have now found that Aralar-knockout (Aralar-KO) post-natal mice show hyperactivity, anxiety-like behavior, and hyperreactivity with a decrease of dopamine (DA) in terminal-rich regions. The striatum is the brain region most affected in terms of size, amino acid and monoamine content. We find a decline in vesicular monoamine transporter-2 (VMAT2) levels associated with increased DA metabolism through MAO activity (DOPAC/DA ratio) in Aralar-KO striatum. However, no decrease in DA or in the number of nigral tyrosine hydroxylase-positive cells was detected in Aralar-KO brainstem. Adult Aralar-hemizygous mice presented also increased DOPAC/DA ratio in striatum and enhanced sensitivity to amphetamine. Our results suggest that Aralar deficiency causes a fall in GSH/GSSG ratio and VMAT2 in striatum that might be related to a failure to produce mitochondrial NADH and to an increase of reactive oxygen species (ROS) in the cytosol. The results indicate that the nigrostriatal dopaminergic system is a target of Aralar deficiency.


Asunto(s)
Ácido Aspártico/metabolismo , Cuerpo Estriado/metabolismo , Dopamina/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Malatos/metabolismo , Enfermedades Mitocondriales/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Trastornos Psicomotores/metabolismo , Sustancia Negra/metabolismo , Sistemas de Transporte de Aminoácidos Acídicos/deficiencia , Sistemas de Transporte de Aminoácidos Acídicos/genética , Sistemas de Transporte de Aminoácidos Acídicos/metabolismo , Animales , Antiportadores/deficiencia , Antiportadores/genética , Antiportadores/metabolismo , Ácido Aspártico/fisiología , Cuerpo Estriado/citología , Dopamina/deficiencia , Dopamina/genética , Emociones/fisiología , Conducta Exploratoria/fisiología , Femenino , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Masculino , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/fisiopatología , Proteínas de Transporte de Membrana Mitocondrial/deficiencia , Proteínas de Transporte de Membrana Mitocondrial/genética , Trastornos de la Destreza Motora/genética , Trastornos de la Destreza Motora/metabolismo , Vías Nerviosas/citología , Vías Nerviosas/metabolismo , Vías Nerviosas/fisiopatología , Estrés Oxidativo/fisiología , Embarazo , Trastornos Psicomotores/genética , Trastornos Psicomotores/fisiopatología , Sustancia Negra/citología
12.
Eur J Paediatr Neurol ; 45: 29-35, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37267771

RESUMEN

BACKGROUND: Leukodystrophies are monogenic disorders primarily affecting the white matter. We aimed to evaluate the utility of genetic testing and time-to-diagnosis in a retrospective cohort of children with suspected leukodystrophy. METHODS: Medical records of patients who attended the leukodystrophy clinic at the Dana-Dwek Children's Hospital between June 2019 and December 2021 were retrieved. Clinical, molecular, and neuroimaging data were reviewed, and the diagnostic yield was compared across genetic tests. RESULTS: Sixty-seven patients (Female/Male ratio 35/32) were included. Median age at symptom onset was 9 months (interquartile range (IQR) 3-18 months), and median length of follow-up was 4.75 years (IQR 3-8.5). Time from symptom onset to a confirmed genetic diagnosis was 15months (IQR 11-30). Pathogenic variants were identified in 60/67 (89.6%) patients; classic leukodystrophy (55/67, 82.1%), leukodystrophy mimics (5/67, 7.5%). Seven patients (10.4%) remained undiagnosed. Exome sequencing showed the highest diagnostic yield (34/41, 82.9%), followed by single-gene sequencing (13/24, 54%), targeted panels (3/9, 33.3%) and chromosomal microarray (2/25, 8%). Familial pathogenic variant testing confirmed the diagnosis in 7/7 patients. A comparison between patients who presented before (n = 31) and after (n = 21) next-generation sequencing (NGS) became clinically available in Israel revealed that the time-to-diagnosis was shorter in the latter group with a median of 12months (IQR 3.5-18.5) vs. a median of 19 months (IQR 13-51) (p = 0.005). CONCLUSIONS: NGS carries the highest diagnostic yield in children with suspected leukodystrophy. Access to advanced sequencing technologies accelerates speed to diagnosis, which is increasingly crucial as targeted treatments become available.


Asunto(s)
Pruebas Genéticas , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias , Preescolar , Femenino , Humanos , Lactante , Masculino , Secuenciación del Exoma , Secuenciación de Nucleótidos de Alto Rendimiento , Estudios Retrospectivos , Sustancia Blanca/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Niño , Adolescente , Judíos/genética , Imagen por Resonancia Magnética , Efecto Fundador
14.
Brain ; 134(Pt 11): 3342-54, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22006981

RESUMEN

Megalencephalic leucoencephalopathy with subcortical cysts is a genetic brain disorder with onset in early childhood. Affected infants develop macrocephaly within the first year of life, after several years followed by slowly progressive, incapacitating cerebellar ataxia and spasticity. From early on, magnetic resonance imaging shows diffuse signal abnormality and swelling of the cerebral white matter, with evidence of highly increased white matter water content. In most patients, the disease is caused by mutations in the gene MLC1, which encodes a plasma membrane protein almost exclusively expressed in brain and at lower levels in leucocytes. Within the brain, MLC1 is mainly located in astrocyte-astrocyte junctions adjacent to the blood-brain and cereborspinal fluid-brain barriers. Thus far, the function of MLC1 has remained unknown. We tested the hypothesis that MLC1 mutations cause a defect in ion currents involved in water and ion homeostasis, resulting in cerebral white matter oedema. Using whole-cell patch clamp studies we demonstrated an association between MLC1 expression and anion channel activity in different cell types, most importantly astrocytes. The currents were absent in chloride-free medium and in cells with disease-causing MLC1 mutations. MLC1-dependent currents were greatly enhanced by hypotonic pretreatment causing cell swelling, while ion channel blockers, including Tamoxifen, abolished the currents. Down regulation of endogenous MLC1 expression in astrocytes by small interfering RNA greatly reduced the activity of this channel, which was rescued by overexpression of normal MLC1. The current-voltage relationship and the pharmacological profiles of the currents indicated that the channel activated by MLC1 expression is a volume-regulated anion channel. Such channels are involved in regulatory volume decrease. We showed that regulatory volume decrease was hampered in lymphoblasts from patients with megalencephalic leucoencephalopathy. A similar trend was observed in astrocytes with decreased MLC1 expression; this effect was rescued by overexpression of normal MLC1. In the present study, we show that absence or mutations of the MLC1 protein negatively impact both volume-regulated anion channel activity and regulatory volume decrease, indicating that megalencephalic leucoencephalopathy is caused by a disturbance of cell volume regulation mediated by chloride transport.


Asunto(s)
Astrocitos/patología , Cloruros/metabolismo , Quistes/fisiopatología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Transporte Iónico/fisiología , Proteínas de la Membrana/genética , Astrocitos/metabolismo , Tamaño de la Célula , Quistes/metabolismo , Quistes/patología , Células HEK293 , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Humanos , Proteínas de la Membrana/metabolismo
15.
Neurobiol Dis ; 43(1): 228-38, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21440627

RESUMEN

Megalencephalic leukoencephalopathy with subcortical cysts (MLC) is a rare type of leukodystrophy, in the majority of cases caused by mutations in the MLC1 gene. MRI from MLC patients shows diffuse cerebral white matter signal abnormality and swelling, with evidence of increased water content. Histopathology in a MLC patient shows vacuolation of myelin, which causes the cerebral white matter swelling. MLC1 protein is expressed in astrocytic processes that are part of blood- and cerebrospinal fluid-brain barriers. We aimed to create an astrocyte cell model of MLC disease. The characterization of rat astrocyte cultures revealed MLC1 localization in cell-cell contacts, which contains other proteins described typically in tight and adherent junctions. MLC1 localization in these contacts was demonstrated to depend on the actin cytoskeleton; it was not altered when disrupting the microtubule or the GFAP networks. In human tissues, MLC1 and the protein Zonula Occludens 1 (ZO-1), which is linked to the actin cytoskeleton, co-localized by EM immunostaining and were specifically co-immunoprecipitated. To create an MLC cell model, knockdown of MLC1 in primary astrocytes was performed. Reduction of MLC1 expression resulted in the appearance of intracellular vacuoles. This vacuolation was reversed by the co-expression of human MLC1. Re-examination of a human brain biopsy from an MLC patient revealed that vacuoles were also consistently present in astrocytic processes. Thus, vacuolation of astrocytes is also a hallmark of MLC disease.


Asunto(s)
Astrocitos/metabolismo , Quistes/genética , Quistes/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/genética , Vacuolas/genética , Adolescente , Animales , Astrocitos/patología , Células Cultivadas , Quistes/fisiopatología , Regulación hacia Abajo/genética , Líquido Extracelular/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Humanos , Proteínas de la Membrana/fisiología , Ratones , Ratas , Ratas Sprague-Dawley , Vacuolas/patología
16.
Am J Hum Genet ; 83(1): 30-42, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18571143

RESUMEN

Hypomyelinating leukodystrophies (HMLs) are disorders involving aberrant myelin formation. The prototype of primary HMLs is the X-linked Pelizaeus-Merzbacher disease (PMD) caused by mutations in PLP1. Recently, homozygous mutations in GJA12 encoding connexin 47 were found in patients with autosomal-recessive Pelizaeus-Merzbacher-like disease (PMLD). However, many patients of both genders with PMLD carry neither PLP1 nor GJA12 mutations. We report a consanguineous Israeli Bedouin kindred with clinical and radiological findings compatible with PMLD, in which linkage to PLP1 and GJA12 was excluded. Using homozygosity mapping and mutation analysis, we have identified a homozygous missense mutation (D29G) not previously described in HSPD1, encoding the mitochondrial heat-shock protein 60 (Hsp60) in all affected individuals. The D29G mutation completely segregates with the disease-associated phenotype. The pathogenic effect of D29G on Hsp60-chaperonin activity was verified by an in vivo E. coli complementation assay, which demonstrated compromised ability of the D29G-Hsp60 mutant protein to support E. coli survival, especially at high temperatures. The disorder, which we have termed MitCHAP-60 disease, can be distinguished from spastic paraplegia 13 (SPG13), another Hsp60-associated autosomal-dominant neurodegenerative disorder, by its autosomal-recessive inheritance pattern, as well as by its early-onset, profound cerebral involvement and lethality. Our findings suggest that Hsp60 defects can cause neurodegenerative pathologies of varying severity, not previously suspected on the basis of the SPG13 phenotype. These findings should help to clarify the important role of Hsp60 in myelinogenesis and neurodegeneration.


Asunto(s)
Chaperonina 60/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Proteínas Mitocondriales/genética , Enfermedades Neurodegenerativas/genética , Secuencia de Aminoácidos , Estudios de Casos y Controles , Chaperonina 60/análisis , Chaperonina 60/química , Chaperonina 60/metabolismo , Cromosomas Humanos Par 2 , Consanguinidad , Secuencia Conservada , Análisis Mutacional de ADN , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Femenino , Genes Letales , Genes Recesivos , Ligamiento Genético , Marcadores Genéticos , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico por imagen , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Humanos , Lactante , Masculino , Repeticiones de Microsatélite , Datos de Secuencia Molecular , Mutación , Enfermedades Neurodegenerativas/diagnóstico , Enfermedades Neurodegenerativas/diagnóstico por imagen , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Linaje , Mapeo Físico de Cromosoma , Polimorfismo de Longitud del Fragmento de Restricción , Radiografía , Homología de Secuencia de Aminoácido
17.
Pediatr Neurol ; 121: 11-19, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34111619

RESUMEN

BACKGROUND: A recurrent homozygous missense variant, c.160G>C;p.(Val54Leu) in HIKESHI, was found to cause a hypomyelinating leukodystrophy with high frequency in the Ashkenazi Jewish population. We provide extended phenotypic classification of this disorder based on clinical history of a further seven affected individuals, assess carrier frequency in the Ashkenazi Jewish population, and provide a neuropathological study. METHODS: Clinical information, neuroimaging, and biosamples were collected. Brain autopsy was performed for one case. RESULTS: Individuals with HIKESHI-related disease share common clinical features: early axial hypotonia evolving to dystonia or with progressive spasticity, hyperreflexia and clonus, feeding difficulties with poor growth, and nystagmus. Severe morbidity or death during febrile illness occurred in five of the nine affected individuals. Magnetic resonance images of seven patients were analyzed and demonstrated diffuse hypomyelination and thin corpus callosum. Genotyping data of more than 125,000 Ashkenazi Jewish individuals revealed a carrier frequency of 1 in 216. Gross pathology examination in one case revealed abnormal white matter. Microscopically, there was a near-total absence of myelin with a relative preservation of axons. The cerebral white matter showed several reactive astrocytes and microglia. CONCLUSIONS: We provide pathologic evidence for a primary disorder of the myelin in HIKESHI-related leukodystrophy. These findings are consistent with the hypomyelination seen in brain magnetic resonance imaging and with the clinical features of early-onset spastic/dystonic quadriplegia and nystagmus. The high carrier rate of the recurrent variant seen in the Ashkenazi Jewish population requires increased attention to screening and diagnosis of this condition, particularly in this population.


Asunto(s)
Proteínas Portadoras/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/patología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Niño , Cuerpo Calloso/diagnóstico por imagen , Cuerpo Calloso/patología , Humanos , Judíos/genética , Imagen por Resonancia Magnética , Secuenciación Completa del Genoma
18.
Curr Opin Neurol ; 23(3): 234-41, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20216214

RESUMEN

PURPOSE OF REVIEW: Knowledge of the metabolic and genetic basis of known and previously unknown leukodystrophies is constantly increasing, opening new treatment options such as enzyme replacement or cell-based therapies. This brief review highlights some recent work, particularly emphasizing results from studies in adulthood leukodystrophies. RECENT FINDINGS: Evidence from recent studies suggests increasing importance of metabolic dysfunctions, for example, in peroxisomal lipid metabolism or energy homeostasis, influencing axonal integrity and oligodendrocyte function and leading to white matter demyelination. In addition, diagnostic and therapeutic progress in metachromatic leukodystrophy, X-linked adrenoleukodystrophy, Krabbe diseases and other rare leukodystrophies with late onset are summarized. SUMMARY: Better understanding of leukodystrophies in neurological routine practice is of crucial importance for differentiating between other white matter diseases such as toxic, inflammatory or vascular leukoencephalopathies. Many leukodystrophies are particularly important to recognize because specific treatments already exist or are currently under investigation. The article also provides an overview of currently known leukodystrophies in adulthood.


Asunto(s)
Encefalopatías Metabólicas Innatas/metabolismo , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/fisiopatología , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Fibras Nerviosas Mielínicas/metabolismo , Adrenoleucodistrofia/genética , Adrenoleucodistrofia/metabolismo , Adrenoleucodistrofia/fisiopatología , Edad de Inicio , Animales , Encefalopatías Metabólicas Innatas/genética , Sistema Nervioso Central/patología , Metabolismo Energético/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Humanos , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/fisiopatología , Leucodistrofia Metacromática/genética , Leucodistrofia Metacromática/metabolismo , Leucodistrofia Metacromática/fisiopatología , Trastornos del Metabolismo de los Lípidos/genética , Trastornos del Metabolismo de los Lípidos/metabolismo , Fibras Nerviosas Mielínicas/patología
19.
Drug News Perspect ; 23(5): 287-94, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20603652

RESUMEN

Multiple sclerosis (MS) is a chronic demyelinating autoimmune disease of the central nervous system. Different therapeutic options are available, but all require parenteral application and some expose patients to potentially lethal adverse effects. The response to treatment remains variable and often incomplete. Thus, the search for novel therapeutic agents is ongoing and relevant. The chimeric compound quinpramine-generated from its precursor substances imipramine and quinacrine-has recently demonstrated clinical efficacy in experimental autoimmune encephalomyelitis (EAE), an animal model for MS. Mechanistic considerations and recent experimental data suggest that intracellular redistribution of antigen-presenting molecules and cholesterol-rich membrane domains may account for the clinical efficacy of this drug. This article summarizes available treatment options in MS and explains why the candidate compound quinpramine may transfer from bench to bedside in the near future.


Asunto(s)
Acridinas/uso terapéutico , Bencilaminas/uso terapéutico , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/tratamiento farmacológico , Acridinas/efectos adversos , Animales , Bencilaminas/efectos adversos , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/terapia , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Humanos , Esclerosis Múltiple/tratamiento farmacológico , Esclerosis Múltiple/fisiopatología , Priones/fisiología
20.
Brain ; 132(Pt 8): 2161-9, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19625339

RESUMEN

Mutations in one of the five eukaryotic initiation factor 2B genes (EIF2B1-5) were first described in childhood ataxia with cerebral hypomyelination--vanishing white matter syndrome. The syndrome is characterized by (i) cerebellar and pyramidal signs in children aged 2-5 years; (ii) extensive cavitating leucoencephalopathy; and (iii) episodes of rapid deterioration following stress. Since then a broad clinical spectrum from congenital to adult-onset forms has been reported, leading to the concept of eIF2B-related disorders. Our aim was to describe clinical and brain magnetic resonance imaging characteristics, genetic findings and natural history of patients with adult-onset eIF2B-related disorders (after age 16). The inclusion criteria were based on the presence of eIF2B mutations and a disease onset after the age of 16 years. One patient with an asymptomatic diagnosis (age 16 years) was also included. Clinical and magnetic resonance findings were retrospectively recorded in all patients. All patients were examined to assess clinical evolution, using functional, pyramidal, cerebellar and cognitive scales. This multi-centric study included 16 patients from 14 families. A sex ratio imbalance was noted (male/female = 3/13). The mean age of onset was 31.1 years (range 16-62). Initial symptoms were neurologic (n = 11), psychiatric (n = 2) and ovarian failure (n = 2). Onset of the symptoms was linked to a precipitating factor in 13% of cases that included minor head trauma and delivery. During follow-up (mean: 11.2 years, range 2-22 years) 12.5% of the patients died. Of the 14 survivors, 62% showed a decline in their cognitive functions, and 79% were severely handicapped or bedridden. One case remained asymptomatic. Stress worsened clinical symptoms in 38% of the patients. Magnetic resonance imaging findings consist of constant cerebral atrophy, extensive cystic leucoencephalopathy (81%), corpus callosum (69%) and cerebellar (38%) T2-weighted hyperintensities. All families except one showed mutations in the EIF2B5 gene. The recurrent p.Arg113His-eIF2Bepsilon mutation was found in 79% of the 14 eIF2B-mutated families, mainly at a homozygous state. The family with a mutation in EIF2B2 had the relatively prevalent p.Glu213Gly mutation. eIF2B-related disorder is probably underestimated as an adult-onset inherited leucoencephalopathy. In this late-onset form, presentation ranges from neurologic symptoms to psychiatric manifestations or primary ovarian failure. Cerebral atrophy is constant, whereas the typical vanishing of the white matter can be absent. Functional and/or cognitive prognosis remains severe. Molecular diagnosis is facilitated for these forms by the screening of the two recurrent p.Arg113His-eIF2Bepsilon and p.Glu213Gly-eIF2Bbeta mutations, positive in 86% of cases.


Asunto(s)
Factor 2B Eucariótico de Iniciación/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Mutación , Adolescente , Adulto , Edad de Inicio , Encéfalo/patología , Trastornos del Conocimiento/etiología , Traumatismos Craneocerebrales/complicaciones , Progresión de la Enfermedad , Femenino , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/complicaciones , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/diagnóstico , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/fisiopatología , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Insuficiencia Ovárica Primaria/etiología , Pronóstico , Estudios Retrospectivos , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA