Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 682
Filtrar
1.
Genes (Basel) ; 15(4)2024 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-38674338

RESUMEN

Microribonucleic acids (miRNAs) comprising miR-23a/b clusters, specifically miR-23a and miR-27a, are recognized for their divergent roles in myelination within the central nervous system. However, cluster-specific miRNA functions remain controversial as miRNAs within the same cluster have been suggested to function complementarily. This study aims to clarify the role of miR-23a/b clusters in myelination using mice with a miR-23a/b cluster deletion (KO mice), specifically in myelin expressing proteolipid protein (PLP). Inducible conditional KO mice were generated by crossing miR-23a/b clusterflox/flox mice with PlpCre-ERT2 mice; the offspring were injected with tamoxifen at 10 days or 10 weeks of age to induce a myelin-specific miR-23a/b cluster deletion. Evaluation was performed at 10 weeks or 12 months of age and compared with control mice that were not treated with tamoxifen. KO mice exhibit impaired motor function and hypoplastic myelin sheaths in the brain and spinal cord at 10 weeks and 12 months of age. Simultaneously, significant decreases in myelin basic protein (MBP) and PLP expression occur in KO mice. The percentages of oligodendrocyte precursors and mature oligodendrocytes are consistent between the KO and control mice. However, the proportion of oligodendrocytes expressing MBP is significantly lower in KO mice. Moreover, changes in protein expression occur in KO mice, with increased leucine zipper-like transcriptional regulator 1 expression, decreased R-RAS expression, and decreased phosphorylation of extracellular signal-regulated kinases. These findings highlight the significant influence of miR-23a/b clusters on myelination during postnatal growth and aging.


Asunto(s)
Envejecimiento , MicroARNs , Vaina de Mielina , Animales , MicroARNs/genética , MicroARNs/metabolismo , Ratones , Vaina de Mielina/metabolismo , Vaina de Mielina/genética , Envejecimiento/genética , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/crecimiento & desarrollo , Ratones Noqueados , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Médula Espinal/metabolismo , Médula Espinal/crecimiento & desarrollo , Proteína Básica de Mielina/metabolismo , Proteína Básica de Mielina/genética , Oligodendroglía/metabolismo , Encéfalo/metabolismo , Encéfalo/crecimiento & desarrollo
2.
Trends Mol Med ; 30(5): 459-470, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38582621

RESUMEN

Pelizaeus-Merzbacher disease (PMD) is caused by mutations in the proteolipid protein 1 (PLP1) gene encoding proteolipid protein (PLP). As a major component of myelin, mutated PLP causes progressive neurodegeneration and eventually death due to severe white matter deficits. Medical care has long been limited to symptomatic treatments, but first-in-class PMD therapies with novel mechanisms now stand poised to enter clinical trials. Here, we review PMD disease mechanisms and outline rationale for therapeutic interventions, including PLP1 suppression, cell transplantation, iron chelation, and intracellular stress modulation. We discuss available preclinical data and their implications on clinical development. With several novel treatments on the horizon, PMD is on the precipice of a new era in the diagnosis and treatment of patients suffering from this debilitating disease.


Asunto(s)
Proteína Proteolipídica de la Mielina , Vaina de Mielina , Enfermedad de Pelizaeus-Merzbacher , Enfermedad de Pelizaeus-Merzbacher/genética , Enfermedad de Pelizaeus-Merzbacher/terapia , Enfermedad de Pelizaeus-Merzbacher/diagnóstico , Enfermedad de Pelizaeus-Merzbacher/patología , Humanos , Vaina de Mielina/metabolismo , Vaina de Mielina/patología , Animales , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Mutación
3.
Brain Res ; 1834: 148912, 2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38575106

RESUMEN

Multiple system atrophy (MSA) is a rare, neurodegenerative disorder with rapid motor and non-motor symptom progression. MSA is characterized by protein aggregations of α-synuclein found in the cytoplasm of oligodendrocytes. Despite this pathological hallmark, there is still little known about the cause of this disease, resulting in poor treatment options and quality of life post-diagnosis. In this study, we investigated differentially expressed genes (DEGs) via RNA-sequencing of brain samples from a validated PLP-α-synuclein transgenic mouse model, identifying a total of 40 DEGs in the PLP group compared to wild-type (WT), with top detected genes being Gm15446, Mcm6, Aldh7a1 and Gm3435. We observed a significant enrichment of immune pathways and endothelial cell genes among the upregulated genes, whereas downregulated genes were significantly enriched for oligodendrocyte and neuronal genes. We then calculated possible overlap of these DEGs with previously profiled human MSA RNA, resulting in the identification of significant downregulation of the Tsr2 gene. Identifying key gene expression profiles specific to MSA patients is crucial to further understanding the cause, and possible prevention, of this rapidly progressive neurodegenerative disorder.


Asunto(s)
Modelos Animales de Enfermedad , Ratones Transgénicos , Atrofia de Múltiples Sistemas , Transcriptoma , alfa-Sinucleína , Animales , Humanos , Ratones , alfa-Sinucleína/genética , alfa-Sinucleína/metabolismo , Encéfalo/metabolismo , Ratones Endogámicos C57BL , Atrofia de Múltiples Sistemas/genética , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/metabolismo , Masculino , Femenino
4.
Stem Cell Res ; 74: 103276, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-38104430

RESUMEN

Genetic alterations in the PLP1 gene, i.e. point mutations and duplications, are associated with demyelinating disease Pelizaeus-Merzbacher. Here, we describe the generation of a human iPSC line harboring a PLP1 variant in codon 33 which leads to an amino acid change from cysteine to tyrosine. The established PLP1C33Y iPSC line enables the study of PMD pathophysiology by investigating various cell types and -characteristics in our developed protocol for bioengineered neuronal organoids (BENOs)1.


Asunto(s)
Células Madre Pluripotentes Inducidas , Enfermedad de Pelizaeus-Merzbacher , Humanos , Enfermedad de Pelizaeus-Merzbacher/genética , Proteína Proteolipídica de la Mielina/genética , Células Madre Pluripotentes Inducidas/metabolismo , Edición Génica , Sistemas CRISPR-Cas/genética , Mutación/genética
5.
Sci Immunol ; 8(88): eadl0618, 2023 10 13.
Artículo en Inglés | MEDLINE | ID: mdl-37801515

RESUMEN

Curated expression of proteolipid protein 1 (PLP1) is essential for multiple sclerosis-derived autoantibody recognition.


Asunto(s)
Esclerosis Múltiple , Proteína Proteolipídica de la Mielina , Humanos , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo
6.
Ann Clin Transl Neurol ; 10(9): 1590-1602, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37475517

RESUMEN

OBJECTIVES: Hereditary spastic paraplegia (HSP) is a genetically heterogeneous disease caused by over 70 genes, with a significant number of patients still genetically unsolved. In this study, we recruited a suspected HSP family characterized by spasticity, developmental delay, ataxia and hypomyelination, and intended to reveal its molecular etiology by whole exome sequencing (WES) and long-read sequencing (LRS) analyses. METHODS: WES was performed on 13 individuals of the family to identify the causative mutations, including analyses of SNVs (single-nucleotide variants) and CNVs (copy number variants). Accurate circular consensus (CCS) long-read sequencing (LRS) was used to verify the findings of CNV analysis from WES. RESULTS: SNVs analysis identified a missense variant c.195G>T (p.E65D) of MORF4L2 at Xq22.2 co-segregating in this family from WES data. Further CNVs analysis revealed a microdeletion, which was adjacent to the MORF4L2 gene, also co-segregating in this family. LRS verified this microdeletion and confirmed the deletion range (chrX: 103,690,507-103,715,018, hg38) with high resolution at nucleotide level accuracy. INTERPRETATIONS: In this study, we identified an Xq22.2 microdeletion (about 24.5 kb), which contains distal enhancers of the PLP1 gene, as a likely cause of SPG2 in this family. The lack of distal enhancers may result in transcriptional repression of PLP1 in oligodendrocytes, potentially affecting its role in the maintenance of myelin, and causing SPG2 phenotype. This study has highlighted the importance of noncoding genomic alterations in the genetic etiology of SPG2.


Asunto(s)
Paraplejía Espástica Hereditaria , Humanos , Paraplejía Espástica Hereditaria/genética , Proteína Proteolipídica de la Mielina/genética , Mutación , Mutación Missense , Fenotipo , Factores de Transcripción/genética
7.
Prenat Diagn ; 43(3): 304-313, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36797813

RESUMEN

OBJECTIVE: Xq chromosome duplication with complex rearrangements is generally acknowledged to be associated with neurodevelopmental disorders, such as Pelizaeus-Merzbacher disease (PMD) and MECP2 duplication syndrome. For couples who required a PGT-M (pre-implantation genetic testing for monogenic disease) for these disorders, junction-specific PCR is useful to directly detect pathogenic variants. Therefore, pre-clinical workup for PGT-M requires the identification of the junction of duplicated segments in PMD and MECP2 duplication syndrome, which is generally difficult. METHODS: In this report, we used nanopore long-read sequencing targeting the X chromosome using an adaptive sampling method to identify breakpoint junctions in disease-causing triplications. RESULTS: By long-read sequencing, we successfully identified breakpoint junctions in one PMD case with PLP1 triplication and in another MECP2 triplication case in a single sequencing run. Surprisingly, the duplicated region involving MECP2 was inserted 45 Mb proximal to the original position. This inserted region was confirmed by FISH analysis. With the help of precise mapping of the pathogenic variant, we successfully re-established STR haplotyping for PGT-M and avoided any potential misinterpretation of the pathogenic allele due to recombination. CONCLUSION: Long-read sequencing with adaptive sampling in a PGT-M pre-clinical workup is a beneficial method for identifying junctions of chromosomal complex structural rearrangements.


Asunto(s)
Secuenciación de Nanoporos , Enfermedad de Pelizaeus-Merzbacher , Diagnóstico Preimplantación , Femenino , Embarazo , Humanos , Proteína Proteolipídica de la Mielina/genética , Duplicación de Gen , Pruebas Genéticas/métodos , Enfermedad de Pelizaeus-Merzbacher/genética , Cromosomas , Diagnóstico Preimplantación/métodos
8.
Ann Clin Transl Neurol ; 10(3): 328-338, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36622199

RESUMEN

OBJECTIVE: Spastic paraplegia type 2 (SPG2) is an X-linked recessive (XLR) form of hereditary spastic paraplegia (HSP) caused by mutations in proteolipid protein 1 (PLP1) gene. We described the clinical and genetic features of three unrelated families with PLP1 mutations and reviewed PLP1-related cases worldwide to summarize the genotype-phenotype correlations. METHODS: The three probands were 23, 26, and 27 years old, respectively, with progressively aggravated walking difficulty as well as lower limb spasticity. Detailed physical examination showed elevated muscle tone, hyperreflexia, and Babinski signs in lower limbs. Brain MRI examinations were investigated for all cases. PLP1 mutations were identified by whole exome sequencing, followed by Sanger sequencing, family co-segregation, and phenotypic reevaluation. RESULTS: A total of eight patients with SPG2 were identified in these three families. The probands additionally had cognitive impairment, urinary or fecal incontinence, ataxia, and white matter lesions (WML) in periventricular regions, with or without kinetic tremor. Three hemizygous mutations in PLP1 were identified, including c.453+159G>A, c.834A>T (p.*278C), and c.434G>A (p.W145*), of which c.834A>T was first associated with HSP. INTERPRETATION: We identified three families with complicated SPG2 due to three PLP1 mutations. Our study supports the clinically inter-and intra-family heterogeneity of SPG2. The periventricular region WML and cognitive impairment are the most common characteristics. The kinetic tremor in upper limbs was observed in 2/3 families, suggesting the spectrum of PLP1-related disorders is still expanding.


Asunto(s)
Paraplejía Espástica Hereditaria , Humanos , Paraplejía Espástica Hereditaria/genética , Paraplejía Espástica Hereditaria/patología , Proteína Proteolipídica de la Mielina/genética , Temblor , Mutación
9.
Glia ; 71(3): 509-523, 2023 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36354016

RESUMEN

Healthy myelin sheaths consist of multiple compacted membrane layers closely encasing the underlying axon. The ultrastructure of CNS myelin requires specialized structural myelin proteins, including the transmembrane-tetraspan proteolipid protein (PLP) and the Ig-CAM myelin-associated glycoprotein (MAG). To better understand their functional relevance, we asked to what extent the axon/myelin-units display similar morphological changes if PLP or MAG are lacking. We thus used focused ion beam-scanning electron microscopy (FIB-SEM) to re-investigate axon/myelin-units side-by-side in Plp- and Mag-null mutant mice. By three-dimensional reconstruction and morphometric analyses, pathological myelin outfoldings extend up to 10 µm longitudinally along myelinated axons in both models. More than half of all assessed outfoldings emerge from internodal myelin. Unexpectedly, three-dimensional reconstructions demonstrated that both models displayed complex axonal pathology underneath the myelin outfoldings, including axonal sprouting. Axonal anastomosing was additionally observed in Plp-null mutant mice. Importantly, normal-appearing axon/myelin-units displayed significantly increased axonal diameters in both models according to quantitative assessment of electron micrographs. These results imply that healthy CNS myelin sheaths facilitate normal axonal diameters and shape, a function that is impaired when structural myelin proteins PLP or MAG are lacking.


Asunto(s)
Sistema Nervioso Central , Proteína Proteolipídica de la Mielina , Vaina de Mielina , Glicoproteína Asociada a Mielina , Animales , Ratones , Axones/metabolismo , Sistema Nervioso Central/metabolismo , Ratones Noqueados , Microscopía Electrónica de Rastreo , Proteínas de la Mielina/metabolismo , Vaina de Mielina/metabolismo , Glicoproteína Asociada a Mielina/genética , Proteína Proteolipídica de la Mielina/genética
10.
Eur J Paediatr Neurol ; 41: 71-79, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36368233

RESUMEN

BACKGROUND: The clinical spectrum of Pelizaeus-Merzbacher disease (PMD), a common hypomyelinating leukodystrophy, ranges between severe neonatal onset and a relatively stable presentation with later onset and mainly lower limb spasticity. In view of emerging treatment options and in order to grade severity and progression, we developed a PMD myelination score. METHODS: Myelination was scored in 15 anatomic sites (items) on conventional T2-and T1w images in controls (n = 328) and 28 PMD patients (53 MRI; n = 5 connatal, n = 3 transitional, n = 10 classic, n = 3 intermediate, n = 2 PLP0, n = 3 SPG2, n = 2 female). Items included in the score were selected based on interrater variability, practicability of scoring and importance of scoring items for discrimination between patients and controls and between patient subgroups. Bicaudate ratio, maximal sagittal pons diameter, and visual assessment of midsagittal corpus callosum were separately recorded. RESULTS: The resulting myelination score consisting of 8 T2-and 5 T1-items differentiates patients and controls as well as patient subgroups at first MRI. There was very little myelin and early loss in severely affected connatal and transitional patients, more, though still severely deficient myelin in classic PMD, ongoing myelination during childhood in classic and intermediate PMD. Atrophy, present in 50% of patients, increased with age at imaging. CONCLUSIONS: The proposed myelination score allows stratification of PMD patients and standardized assessment of follow-up. Loss of myelin in severely affected and PLP0 patients and progressing myelination in classic and intermediate PMD must be considered when evaluating treatment efficacy.


Asunto(s)
Enfermedad de Pelizaeus-Merzbacher , Recién Nacido , Humanos , Femenino , Proteína Proteolipídica de la Mielina/genética , Mutación , Imagen por Resonancia Magnética , Cuerpo Calloso/diagnóstico por imagen
11.
Orphanet J Rare Dis ; 17(1): 137, 2022 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-35346287

RESUMEN

BACKGROUND: The natural history and genotype-phenotype correlation of Pelizaeus-Merzbacher disease (PMD) of Chinese patients has been rarely reported. METHOD: Patients who met the criteria for PMD were enrolled in our study. Genomic analysis was conducted by multiplex ligation probe amplification (MLPA) and Sanger or whole-exome sequencing (WES). Natural history differences and genotype-phenotype correlations were analyzed. RESULT: A total of 111 patients were enrolled in our follow-up study. The median follow-up interval was 53 m (1185). Among PMD patients, developmental delay was the most common sign, and nystagmus and hypotonia were the most common initial symptoms observed. A total of 78.4% of the patients were able to control their head, and 72.1% could speak words. However, few of the patients could stand (9.0%) or walk (4.5%) by themselves. Nystagmus improved in more than half of the patients, and hypotonia sometimes deteriorated to movement disorders. More PLP1 point mutations patients were categorized into severe group, while more patients with PLP1 duplications were categorized into mild group (p < 0.001). Compared to patients in mild groups, those in the severe group had earlier disease onset and had acquired fewer skills at a later age. CONCLUSION: PMD patients have early disease onset with nystagmus and hypotonia followed by decreased nystagmus and movement disorders, such as spasticit. Patients with PLP1 duplication were more likely to be categorized into the mild group, whereas patients with point mutations were more likely to be categorized into the severe group.


Asunto(s)
Enfermedad de Pelizaeus-Merzbacher , China , Estudios de Seguimiento , Estudios de Asociación Genética , Humanos , Proteína Proteolipídica de la Mielina/genética , Enfermedad de Pelizaeus-Merzbacher/diagnóstico , Enfermedad de Pelizaeus-Merzbacher/genética
12.
Eur J Hum Genet ; 30(7): 860-864, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35217805

RESUMEN

Leukodystrophies are a heterogenous group of genetic disorders, characterised by abnormal development of cerebral white matter. Pelizaeus-Merzbacher disease is caused by mutations in PLP1, encoding major myelin-resident protein required for myelin sheath assembly. We report a missense variant p.(Ala109Asp) in MAL as causative for a rare, hypomyelinating leukodystrophy similar to Pelizaeus-Merzbacher disease. MAL encodes a membrane proteolipid that directly interacts with PLP1, ensuring correct distribution during myelin assembly. In contrast to wild-type MAL, mutant MAL was retained in the endoplasmic reticulum but was released following treatment with 4-phenylbutyrate. Proximity-dependent identification of wild-type MAL interactants implicated post-Golgi vesicle-mediated protein transport and protein localisation to membranes, whereas mutant MAL interactants suggested unfolded protein responses. Our results suggest that mislocalisation of MAL affects PLP1 distribution, consistent with known pathomechanisms for hypomyelinating leukodystrophies.


Asunto(s)
Enfermedades Neurodegenerativas , Enfermedad de Pelizaeus-Merzbacher , Humanos , Mutación , Mutación Missense , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Enfermedad de Pelizaeus-Merzbacher/genética , Transporte de Proteínas
13.
Neuroscience ; 476: 60-71, 2021 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-34506833

RESUMEN

Among the hypomyelinating leukodystrophies, Pelizaeus-Merzbacher disease (PMD) is a representative disorder. The disease is caused by different types of PLP1 mutations, among which PLP1 duplication accounts for ∼70% of the mutations. Previous studies have shown that PLP1 duplications lead to PLP1 retention in the endoplasmic reticulum (ER); in parallel, recent studies have demonstrated that PLP1 duplication can also lead to mitochondrial dysfunction. As such, the respective roles and interactions of the ER and mitochondria in the pathogenesis of PLP1 duplication are not clear. In both PLP1 patients' and healthy fibroblasts, we measured mitochondrial respiration with a Seahorse XF Extracellular Analyzer and examined the interactions between the ER and mitochondria with super-resolution microscopy (spinning-disc pinhole-based structured illumination microscopy, SD-SIM). For the first time, we demonstrated that PLP1 duplication mutants had closer ER-mitochondrion interfaces mediated through structural and morphological changes in both the ER and mitochondria-associated membranes (MAMs). These changes in both the ER and mitochondria then led to mitochondrial dysfunction, as reported previously. This work highlights the roles of MAMs in bridging PLP1 expression in the ER and pathogenic dysfunction in mitochondria, providing novel insight into the pathogenicity of mitochondrial dysfunction resulting from PLP1 duplication. These findings suggest that interactions between the ER and mitochondria may underlie pathogenic mechanisms of hypomyelinating leukodystrophies diseases at the organelle level.


Asunto(s)
Proteína Proteolipídica de la Mielina , Enfermedad de Pelizaeus-Merzbacher , Retículo Endoplásmico , Humanos , Mitocondrias , Mutación , Proteína Proteolipídica de la Mielina/genética , Enfermedad de Pelizaeus-Merzbacher/genética , Virulencia
14.
PLoS One ; 16(8): e0256207, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34403440

RESUMEN

Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Larva/genética , Vaina de Mielina/genética , Tiroxina/deficiencia , Triyodotironina/deficiencia , Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Antígenos/genética , Antígenos/metabolismo , Embrión no Mamífero , Desarrollo Embrionario , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Ácido Yopanoico/farmacología , Larva/citología , Larva/efectos de los fármacos , Larva/crecimiento & desarrollo , Mesencéfalo/citología , Mesencéfalo/efectos de los fármacos , Mesencéfalo/crecimiento & desarrollo , Mesencéfalo/metabolismo , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/efectos de los fármacos , Vaina de Mielina/metabolismo , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Factor de Transcripción 2 de los Oligodendrocitos/genética , Factor de Transcripción 2 de los Oligodendrocitos/metabolismo , Oligodendroglía/citología , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Prosencéfalo/citología , Prosencéfalo/efectos de los fármacos , Prosencéfalo/crecimiento & desarrollo , Prosencéfalo/metabolismo , Proteoglicanos/genética , Proteoglicanos/metabolismo , Rombencéfalo/citología , Rombencéfalo/efectos de los fármacos , Rombencéfalo/crecimiento & desarrollo , Rombencéfalo/metabolismo , Factores de Transcripción SOXE/genética , Factores de Transcripción SOXE/metabolismo , Médula Espinal/citología , Médula Espinal/efectos de los fármacos , Médula Espinal/crecimiento & desarrollo , Médula Espinal/metabolismo , Triyodotironina/farmacología , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
15.
Hum Mol Genet ; 30(23): 2225-2239, 2021 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-34230963

RESUMEN

Oligodendrocytes (OLs) produce myelin in the central nervous system (CNS), which accelerates the propagation of action potentials and supports axonal integrity. As a major component of CNS myelin, proteolipid protein 1 (Plp1) is indispensable for the axon-supportive function of myelin. Notably, this function requires the continuous high-level expression of Plp1 in OLs. Equally important is the controlled expression of Plp1, as illustrated by Pelizaeus-Merzbacher disease for which the most common cause is PLP1 overexpression. Despite a decade-long search, promoter-distal OL enhancers that govern Plp1 remain elusive. We have recently developed an innovative method that maps promoter-distal enhancers to genes in a principled manner. Here, we applied it to Plp1, uncovering two OL enhancers for it (termed Plp1-E1 and Plp1-E2). Remarkably, clustered regularly interspaced short palindromic repeats (CRISPR) interference epigenome editing showed that Plp1-E1 and Plp1-E2 do not regulate two genes in their vicinity, highlighting their exquisite specificity to Plp1. Assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and chromatin immunoprecipitation with high-throughput sequencing (ChIP-seq) data show that Plp1-E1 and Plp1-E2 are OL-specific enhancers that are conserved among human, mouse and rat. Hi-C data reveal that the physical interactions between Plp1-E1/2 and PLP1 are among the strongest in OLs and specific to OLs. We also show that Myrf, a master regulator of OL development, acts on Plp1-E1 and Plp1-E2 to promote Plp1 expression.


Asunto(s)
Elementos de Facilitación Genéticos , Regulación de la Expresión Génica , Proteína Proteolipídica de la Mielina/genética , Oligodendroglía/metabolismo , Animales , Secuencia de Bases , Sitios de Unión , Sistemas CRISPR-Cas , Secuenciación de Inmunoprecipitación de Cromatina , Mapeo Cromosómico , Proteínas de Unión al ADN/metabolismo , Humanos , Proteínas de la Membrana/metabolismo , Ratones , Proteína Proteolipídica de la Mielina/metabolismo , Motivos de Nucleótidos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
16.
JCI Insight ; 6(7)2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33661767

RESUMEN

Here, we report on the identification of Itga7-expressing muscle-resident glial cells activated by loss of neuromuscular junction (NMJ) integrity. Gene expression analysis at the bulk and single-cell level revealed that these cells are distinct from Itga7-expressing muscle satellite cells. We show that a selective activation and expansion of Itga7+ glial cells occur in response to muscle nerve lesion. Upon activation, muscle glial-derived progenies expressed neurotrophic genes, including nerve growth factor receptor, which enables their isolation by FACS. We show that activated muscle glial cells also expressed genes potentially implicated in extracellular matrix remodeling at NMJs. We found that tenascin C, which was highly expressed by muscle glial cells, activated upon nerve injury and preferentially localized to NMJ. Interestingly, we observed that the activation of muscle glial cells by acute nerve injury was reversible upon NMJ repair. By contrast, in a mouse model of ALS, in which NMJ degeneration is progressive, muscle glial cells steadily increased over the course of the disease. However, they exhibited an impaired neurotrophic activity, suggesting that pathogenic activation of glial cells may be implicated in ALS progression.


Asunto(s)
Esclerosis Amiotrófica Lateral/patología , Músculo Esquelético/citología , Neuroglía/fisiología , Traumatismos de la Médula Espinal/patología , Animales , Antígenos CD/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica , Cadenas alfa de Integrinas/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Neuroglía/citología , Unión Neuromuscular/citología , Receptor de Factor de Crecimiento Nervioso/genética , Receptores Colinérgicos/metabolismo , Nervio Ciático/lesiones , Análisis de la Célula Individual , Superóxido Dismutasa-1/genética
17.
J Hum Genet ; 66(10): 1035-1037, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-33785861

RESUMEN

Monoallelic mutations on TMEM63A have been recently reported as cause of a previously unrecognized disorder named "infantile-onset transient hypomyelination". Clinical and neuroradiological presentation is described as highly similar to Pelizaeus-Merzbacher Disease but evolution over time was surprisingly benign with a progressive spontaneous improving course. We report on a new TMEM63A-mutated girl. The clinical picture was similar to the one already described except for the presence of recurrent episodes of unilateral eyelid twitching, and for the evidence of spinal cord involvement on MRI. These are interesting findings helping in distinguishing this condition from classic PMD since early disease stages. However, additional observations are needed to confirm if these are common features of this condition.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteínas de la Membrana/genética , Proteína Proteolipídica de la Mielina/genética , Enfermedad de Pelizaeus-Merzbacher/genética , Médula Espinal/diagnóstico por imagen , Femenino , Humanos , Lactante , Imagen por Resonancia Magnética , Mutación/genética , Enfermedad de Pelizaeus-Merzbacher/diagnóstico por imagen , Enfermedad de Pelizaeus-Merzbacher/metabolismo , Enfermedad de Pelizaeus-Merzbacher/patología , Médula Espinal/metabolismo , Médula Espinal/patología
18.
Ann Clin Transl Neurol ; 8(2): 456-470, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33440071

RESUMEN

OBJECTIVE: To determine whether animals with Japanese macaque encephalomyelitis (JME), a spontaneous demyelinating disease similar to multiple sclerosis (MS), harbor myelin-specific T cells in their central nervous system (CNS) and periphery. METHODS: Mononuclear cells (MNCs) from CNS lesions, cervical lymph nodes (LNs) and peripheral blood of Japanese macaques (JMs) with JME, and cervical LN and blood MNCs from healthy controls or animals with non-JME conditions were analyzed for the presence of myelin-specific T cells and changes in interleukin 17 (IL-17) and interferon gamma (IFNγ) expression. RESULTS: Demyelinating JME lesions contained CD4+ T cells and CD8+ T cells specific to myelin oligodendrocyte glycoprotein (MOG), myelin basic protein (MBP), and/or proteolipid protein (PLP). CD8+ T-cell responses were absent in JME peripheral blood, and in age- and sex-matched controls. However, CD4+ Th1 and Th17 responses were detected in JME peripheral blood versus controls. Cervical LN MNCs from eight of nine JME animals had CD3+ T cells specific for MOG, MBP, and PLP that were not detected in controls. Mapping myelin epitopes revealed a heterogeneity in responses among JME animals. Comparison of myelin antigen sequences with those of JM rhadinovirus (JMRV), which is found in JME lesions, identified six viral open reading frames (ORFs) with similarities to myelin antigen sequences. Overlapping peptides to these JMRV ORFs did not induce IFNγ responses. INTERPRETATIONS: JME possesses an immune-mediated component that involves both CD4+ and CD8+ T cells specific for myelin antigens. JME may shed new light on inflammatory demyelinating disease pathogenesis linked to gamma-herpesvirus infection.


Asunto(s)
Enfermedades Desmielinizantes/diagnóstico por imagen , Enfermedades Desmielinizantes/patología , Encefalomielitis/diagnóstico por imagen , Encefalomielitis/patología , Vaina de Mielina/inmunología , Linfocitos T/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Desmielinizantes/virología , Encefalomielitis/virología , Ensayo de Inmunoadsorción Enzimática , Mapeo Epitopo , Epítopos/genética , Epítopos/inmunología , Femenino , Infecciones por Herpesviridae/inmunología , Interferón gamma/análisis , Interleucina-17/análisis , Macaca fuscata , Masculino , Enfermedades de los Monos , Proteína Básica de Mielina/genética , Proteína Básica de Mielina/inmunología , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/inmunología , Vaina de Mielina/patología , Glicoproteína Mielina-Oligodendrócito/genética , Glicoproteína Mielina-Oligodendrócito/inmunología , Rhadinovirus/genética , Rhadinovirus/inmunología
19.
Aging (Albany NY) ; 13(1): 1488-1497, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33429367

RESUMEN

A family with a history of Pelizaeus-Merzbacher disease (PMD) received prenatal diagnosis of PLP1 gene duplication in a fetus using a single nucleotide polymorphism (SNP) array. A 27-year-old pregnant woman was referred for genetic counseling due to her four-year-old son being diagnosed with a suspected classic type of PMD. Amniocentesis was performed at 18 and 3/7 weeks of gestation, and the SNP array was carried out on DNA from the mother, her affected son, and fetus, then further confirmed by multiplex ligation-dependent probe amplification (MLPA). Cytogenetic analysis of the fetus showed 46,XY. SNP array analysis revealed that the male fetus did not carry PLP1 gene duplication but the affected boy did, and the mother was a carrier for the duplication of the PLP1 gene. All SNP array results were further confirmed by MLPA. SNP array and MLPA analyses of peripheral blood verified the nonduplication of the PLP1 gene in the infant after birth. At present, the child (without PLP1 duplication) is developing normally. This study preliminarily suggests that SNP array is a sensitive and accurate technology for identifying PLP1 duplication and is feasible for reliable diagnosis, including for the prenatal diagnosis of PMD resulting from PLP1 duplication.


Asunto(s)
Amniocentesis , Técnicas Genéticas , Proteína Proteolipídica de la Mielina/genética , Enfermedad de Pelizaeus-Merzbacher/diagnóstico , Enfermedad de Pelizaeus-Merzbacher/genética , Adulto , Femenino , Duplicación de Gen , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Embarazo
20.
PLoS Biol ; 18(11): e3000943, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33196637

RESUMEN

In several neurodegenerative disorders, axonal pathology may originate from impaired oligodendrocyte-to-axon support of energy substrates. We previously established transgenic mice that allow measuring axonal ATP levels in electrically active optic nerves. Here, we utilize this technique to explore axonal ATP dynamics in the Plpnull/y mouse model of spastic paraplegia. Optic nerves from Plpnull/y mice exhibited lower and more variable basal axonal ATP levels and reduced compound action potential (CAP) amplitudes, providing a missing link between axonal pathology and a role of oligodendrocytes in brain energy metabolism. Surprisingly, when Plpnull/y optic nerves are challenged with transient glucose deprivation, both ATP levels and CAP decline slower, but recover faster upon reperfusion of glucose. Structurally, myelin sheaths display an increased frequency of cytosolic channels comprising glucose and monocarboxylate transporters, possibly facilitating accessibility of energy substrates to the axon. These data imply that complex metabolic alterations of the axon-myelin unit contribute to the phenotype of Plpnull/y mice.


Asunto(s)
Adenosina Trifosfato/metabolismo , Vaina de Mielina/metabolismo , Paraplejía/metabolismo , Potenciales de Acción , Animales , Axones/metabolismo , Modelos Animales de Enfermedad , Metabolismo Energético , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Microscopía Inmunoelectrónica , Proteína Proteolipídica de la Mielina/deficiencia , Proteína Proteolipídica de la Mielina/genética , Vaina de Mielina/patología , Nervio Óptico/metabolismo , Nervio Óptico/patología , Paraplejía/genética , Paraplejía/patología , Fenotipo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...