Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
1.
Acta Neuropathol Commun ; 12(1): 139, 2024 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-39217398

RESUMEN

CSF1R-related disorder (CSF1R-RD) is a neurodegenerative condition that predominantly affects white matter due to genetic alterations in the CSF1R gene, which is expressed by microglia. We studied an elderly man with a hereditary, progressive dementing disorder of unclear etiology. Standard genetic testing for leukodystrophy and other neurodegenerative conditions was negative. Brain autopsy revealed classic features of adult-onset leukoencephalopathy with axonal spheroids and pigmented glia (ALSP), including confluent white matter degeneration with axonal spheroids and pigmented glial cells in the affected white matter, consistent with CSF1R-RD. Subsequent long-read sequencing identified a novel deletion in CSF1R that was not detectable with short-read exome sequencing. To gain insight into potential mechanisms underlying white matter degeneration in CSF1R-RD, we studied multiple brain regions exhibiting varying degrees of white matter pathology. We found decreased CSF1R transcript and protein across brain regions, including intact white matter. Single nuclear RNA sequencing (snRNAseq) identified two disease-associated microglial cell states: lipid-laden microglia (expressing GPNMB, ATG7, LGALS1, LGALS3) and inflammatory microglia (expressing IL2RA, ATP2C1, FCGBP, VSIR, SESN3), along with a small population of CD44+ peripheral monocyte-derived macrophages exhibiting migratory and phagocytic signatures. GPNMB+ lipid-laden microglia with ameboid morphology represented the end-stage disease microglia state. Disease-associated oligodendrocytes exhibited cell stress signatures and dysregulated apoptosis-related genes. Disease-associated oligodendrocyte precursor cells (OPCs) displayed a failure in their differentiation into mature myelin-forming oligodendrocytes, as evidenced by upregulated LRP1, PDGFRA, SOX5, NFIA, and downregulated NKX2-2, NKX6.2, SOX4, SOX8, TCF7L2, YY1, ZNF488. Overall, our findings highlight microglia-oligodendroglia crosstalk in demyelination, with CSF1R dysfunction promoting phagocytic and inflammatory microglia states, an arrest in OPC differentiation, and oligodendrocyte depletion.


Asunto(s)
Neuroglía , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Humanos , Masculino , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Neuroglía/patología , Neuroglía/metabolismo , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Leucoencefalopatías/metabolismo , Anciano , Microglía/patología , Microglía/metabolismo , Perfilación de la Expresión Génica , Transcriptoma , Sustancia Blanca/patología , Sustancia Blanca/metabolismo , Encéfalo/patología , Encéfalo/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos
2.
Proc Natl Acad Sci U S A ; 121(21): e2321496121, 2024 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-38753517

RESUMEN

RNASET2-deficient leukodystrophy is a rare infantile white matter disorder mimicking a viral infection and resulting in severe psychomotor impairments. Despite its severity, there is little understanding of cellular mechanisms of pathogenesis and no treatments. Recent research using the rnaset2 mutant zebrafish model has suggested that microglia may be the drivers of the neuropathology, due to their failure to digest apoptotic debris during neurodevelopment. Therefore, we developed a strategy for microglial replacement through transplantation of adult whole kidney marrow-derived macrophages into embryonic hosts. Using live imaging, we revealed that transplant-derived macrophages can engraft within host brains and express microglia-specific markers, suggesting the adoption of a microglial phenotype. Tissue-clearing strategies revealed the persistence of transplanted cells in host brains beyond embryonic stages. We demonstrated that transplanted cells clear apoptotic cells within the brain, as well as rescue overactivation of the antiviral response otherwise seen in mutant larvae. RNA sequencing at the point of peak transplant-derived cell engraftment confirms that transplantation can reduce the brain-wide immune response and particularly, the antiviral response, in rnaset2-deficient brains. Crucially, this reduction in neuroinflammation resulted in behavioral rescue-restoring rnaset2 mutant motor activity to wild-type (WT) levels in embryonic and juvenile stages. Together, these findings demonstrate the role of microglia as the cellular drivers of neuropathology in rnaset2 mutants and that macrophage transplantation is a viable strategy for microglial replacement in the zebrafish. Therefore, microglia-targeted interventions may have therapeutic benefits in RNASET2-deficient leukodystrophy.


Asunto(s)
Encéfalo , Macrófagos , Microglía , Animales , Encéfalo/patología , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Leucoencefalopatías/metabolismo , Macrófagos/metabolismo , Microglía/metabolismo , Microglía/patología , Pez Cebra , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/metabolismo
3.
Cytotherapy ; 26(2): 185-193, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38054911

RESUMEN

BACKGROUND AIMS: White matter diseases are commonly associated with microglial activation and neuroinflammation. Mesenchymal stromal cells (MSCs) have immunomodulatory properties and thus have the potential to be developed as cell therapy for white matter disease. MSCs interact with resident macrophages to alter the trajectory of inflammation; however, the impact MSCs have on central nervous system macrophages and the effect this has on the progression of white matter disease are unclear. METHODS: In this study, we utilized numerous assays of varying complexity to model different aspects of white matter disease. These assays ranged from an in vivo spinal cord acute demyelination model to a simple microglial cell line activation assay. Our goal was to investigate the influence of human umbilical cord tissue MSCs on the activation of microglia. RESULTS: MSCs reduced the production of tumor necrosis factor (TNF) by microglia and decreased demyelinated lesions in the spinal cord after acute focal injury. To determine if MSCs could directly suppress the activation of microglia and to develop an efficient potency assay, we utilized isolated primary microglia from mouse brains and the Immortalized MicroGlial Cell Line (IMG). MSCs suppressed the activation of microglia and the release of TNF after stimulation with lipopolysaccharide, a toll-like receptor agonist. CONCLUSIONS: In this study, we demonstrated that MSCs altered the immune response after acute injury in the spinal cord. In numerous assays, MSCs suppressed activation of microglia and release of the pro-inflammatory cytokine TNF. Of these assays, IMG could be standardized and used as an effective potency assay to determine the efficacy of MSCs for treating white matter disease or other neuroinflammatory conditions associated with microglial activation.


Asunto(s)
Leucoencefalopatías , Células Madre Mesenquimatosas , Ratones , Animales , Humanos , Microglía/metabolismo , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Leucoencefalopatías/metabolismo
4.
Pediatr Dev Pathol ; 27(2): 198-204, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37903135

RESUMEN

Caseinolytic peptidase B homolog (CLPB) is a mitochondrial protein which is highly expressed in brain. Its deficiency may be associated with severe neonatal encephalopathy. This report describes a case of fatal neonatal encephalopathy associated with biallelic stop-gain mutation in CLPB (NM_001258392.3:c.1159C>T/p.Arg387*). Neurologic disorder encompasses pre- and post-natal features including polyhydramnios, intrauterine growth restriction, respiratory insufficiency, lethargy, excessive startle reflex, generalized hypertonia, and epileptic seizures. Brain macroscopic examination demonstrates frontal severe periventricular cystic leukoencephalopathy, along with mild ex-vacuo tri-ventricular dilatation. The most striking immunohistopathologic features are striato-thalamic neurodegeneration and deep white matter loss associated with strong reactive astrogliosis. This report supports that CLPB deficiency should be considered among the neurometabolic disorders associated with severe prenatal-onset neurologic impairment that may result from cystic leukoencephalopathy.


Asunto(s)
Epilepsia , Enfermedades del Recién Nacido , Leucoencefalopatías , Recién Nacido , Femenino , Embarazo , Humanos , Endopeptidasa Clp/genética , Endopeptidasa Clp/metabolismo , Encéfalo/patología , Epilepsia/metabolismo , Leucoencefalopatías/diagnóstico , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Codón sin Sentido/metabolismo , Enfermedades del Recién Nacido/patología
5.
Glia ; 72(2): 375-395, 2024 02.
Artículo en Inglés | MEDLINE | ID: mdl-37909242

RESUMEN

White matter abnormalities, related to poor cerebral perfusion, are a core feature of small vessel cerebrovascular disease, and critical determinants of vascular cognitive impairment and dementia. Despite this importance there is a lack of treatment options. Proliferation of microglia producing an expanded, reactive population and associated neuroinflammatory alterations have been implicated in the onset and progression of cerebrovascular white matter disease, in patients and in animal models, suggesting that targeting microglial proliferation may exert protection. Colony-stimulating factor-1 receptor (CSF1R) is a key regulator of microglial proliferation. We found that the expression of CSF1R/Csf1r and other markers indicative of increased microglial abundance are significantly elevated in damaged white matter in human cerebrovascular disease and in a clinically relevant mouse model of chronic cerebral hypoperfusion and vascular cognitive impairment. Using the mouse model, we investigated long-term pharmacological CSF1R inhibition, via GW2580, and demonstrated that the expansion of microglial numbers in chronic hypoperfused white matter is prevented. Transcriptomic analysis of hypoperfused white matter tissue showed enrichment of microglial and inflammatory gene sets, including phagocytic genes that were the predominant expression modules modified by CSF1R inhibition. Further, CSF1R inhibition attenuated hypoperfusion-induced white matter pathology and rescued spatial learning impairments and to a lesser extent cognitive flexibility. Overall, this work suggests that inhibition of CSF1R and microglial proliferation mediates protection against chronic cerebrovascular white matter pathology and cognitive deficits. Our study nominates CSF1R as a target for the treatment of vascular cognitive disorders with broader implications for treatment of other chronic white matter diseases.


Asunto(s)
Trastornos Cerebrovasculares , Trastornos del Conocimiento , Disfunción Cognitiva , Leucoencefalopatías , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos , Sustancia Blanca , Animales , Ratones , Trastornos Cerebrovasculares/metabolismo , Trastornos Cerebrovasculares/patología , Trastornos del Conocimiento/etiología , Trastornos del Conocimiento/patología , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Ratones Endogámicos C57BL , Microglía/metabolismo , Receptores del Factor Estimulante de Colonias/metabolismo , Sustancia Blanca/patología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo
6.
Int J Mol Sci ; 22(23)2021 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-34884611

RESUMEN

Promoting oligodendrocyte viability has been proposed as a therapeutic strategy for alleviating many neuronal diseases, such as multiple sclerosis and stroke. However, molecular pathways critical for oligodendrocyte survival under various stresses are still not well known. p53 is a strong tumor suppressor and regulates cell cycle, DNA repair and cell death. Our previous studies have shown that p53 plays an important role in promoting neuronal survival after insults, but its specific role in oligodendrocyte survival is not known. Here, we constructed the mice with oligodendrocyte-specific p53 loss by crossing TRP53flox/flox mice and CNP-cre mice, and found that p53 was dispensable for oligodendrocyte differentiation and myelin formation under physiological condition. In the experimental autoimmune encephalomyelitis (EAE) model, p53 loss of function, specifically in oligodendrocytes, did not affect the EAE disease severity and had no effect on demyelination in the spinal cord of the mice. Interestingly, p53 deficiency in oligodendrocytes significantly attenuated the demyelination of corpus callosum and alleviated the functional impairment of motor coordination and spatial memory in the cuprizone demyelination model. Moreover, the oligodendrocyte-specific loss of p53 provided protection against subcortical white matter damage and mitigated recognition memory impairment in mice in the white matter stroke model. These results suggest that p53 plays different roles in the brain and spinal cord or in response to various stresses. Thus, p53 may be a therapeutic target for oligodendrocyte prevention in specific brain injuries, such as white matter stroke and multiple sclerosis.


Asunto(s)
Cuprizona/toxicidad , Encefalomielitis Autoinmune Experimental/prevención & control , Leucoencefalopatías/prevención & control , Trastornos de la Memoria/prevención & control , Oligodendroglía/citología , Accidente Cerebrovascular/prevención & control , Proteína p53 Supresora de Tumor/fisiología , Animales , Quelantes/toxicidad , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Femenino , Leucoencefalopatías/etiología , Leucoencefalopatías/metabolismo , Leucoencefalopatías/patología , Masculino , Trastornos de la Memoria/etiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oligodendroglía/metabolismo , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología
7.
Nat Commun ; 12(1): 6530, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34764281

RESUMEN

Infantile-onset RNaseT2 deficient leukoencephalopathy is characterised by cystic brain lesions, multifocal white matter alterations, cerebral atrophy, and severe psychomotor impairment. The phenotype is similar to congenital cytomegalovirus brain infection and overlaps with type I interferonopathies, suggesting a role for innate immunity in its pathophysiology. To date, pathophysiological studies have been hindered by the lack of mouse models recapitulating the neuroinflammatory encephalopathy found in patients. In this study, we generated Rnaset2-/- mice using CRISPR/Cas9-mediated genome editing. Rnaset2-/- mice demonstrate upregulation of interferon-stimulated genes and concurrent IFNAR1-dependent neuroinflammation, with infiltration of CD8+ effector memory T cells and inflammatory monocytes into the grey and white matter. Single nuclei RNA sequencing reveals homeostatic dysfunctions in glial cells and neurons and provide important insights into the mechanisms of hippocampal-accentuated brain atrophy and cognitive impairment. The Rnaset2-/- mice may allow the study of CNS damage associated with RNaseT2 deficiency and may be used for the investigation of potential therapies.


Asunto(s)
Endorribonucleasas/metabolismo , Leucoencefalopatías/metabolismo , Leucoencefalopatías/patología , Animales , Linfocitos T CD8-positivos/metabolismo , Disfunción Cognitiva/genética , Disfunción Cognitiva/metabolismo , Modelos Animales de Enfermedad , Endorribonucleasas/genética , Femenino , Citometría de Flujo , Genotipo , Humanos , Inmunohistoquímica , Leucoencefalopatías/genética , Imagen por Resonancia Magnética , Masculino , Células T de Memoria/metabolismo , Ratones , Ratones Noqueados , Neuroglía/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa
8.
Cells ; 10(8)2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34440627

RESUMEN

Translation initiation factor 2B (eIF2B) is a master regulator of global protein synthesis in all cell types. The mild genetic Eif2b5(R132H) mutation causes a slight reduction in eIF2B enzymatic activity which leads to abnormal composition of mitochondrial electron transfer chain complexes and impaired oxidative phosphorylation. Previous work using primary fibroblasts isolated from Eif2b5(R132H/R132H) mice revealed that owing to increased mitochondrial biogenesis they exhibit normal cellular ATP level. In contrast to fibroblasts, here we show that primary astrocytes isolated from Eif2b5(R132H/R132H) mice are unable to compensate for their metabolic impairment and exhibit chronic state of low ATP level regardless of extensive adaptation efforts. Mutant astrocytes are hypersensitive to oxidative stress and to further energy stress. Moreover, they show migration deficit upon exposure to glucose starvation. The mutation in Eif2b5 prompts reactive oxygen species (ROS)-mediated inferior ability to stimulate the AMP-activated protein kinase (AMPK) axis, due to a requirement to increase the mammalian target of rapamycin complex-1 (mTORC1) signalling in order to enable oxidative glycolysis and generation of specific subclass of ROS-regulating proteins, similar to cancer cells. The data disclose the robust impact of eIF2B on metabolic and redox homeostasis programs in astrocytes and point at their hyper-sensitivity to mutated eIF2B. Thereby, it illuminates the central involvement of astrocytes in Vanishing White Matter Disease (VWMD), a genetic neurodegenerative leukodystrophy caused by homozygous hypomorphic mutations in genes encoding any of the 5 subunits of eIF2B.


Asunto(s)
Adenosina Trifosfato/metabolismo , Astrocitos/metabolismo , Encéfalo/metabolismo , Metabolismo Energético , Factor 2B Eucariótico de Iniciación/metabolismo , Leucoencefalopatías/metabolismo , Mitocondrias/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Apoptosis , Astrocitos/patología , Encéfalo/patología , Movimiento Celular , Células Cultivadas , Modelos Animales de Enfermedad , Factor 2B Eucariótico de Iniciación/genética , Leucoencefalopatías/genética , Leucoencefalopatías/patología , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Ratones Mutantes , Mitocondrias/genética , Mitocondrias/patología , Mutación , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal
9.
Hum Mol Genet ; 30(17): 1649-1665, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34100078

RESUMEN

Megalencephalic Leukoencephalopathy with subcortical Cysts (MLC) is a type of vacuolating leukodystrophy, which is mainly caused by mutations in MLC1 or GLIALCAM. The two MLC-causing genes encode for membrane proteins of yet unknown function that have been linked to the regulation of different chloride channels such as the ClC-2 and VRAC. To gain insight into the role of MLC proteins, we have determined the brain GlialCAM interacting proteome. The proteome includes different transporters and ion channels known to be involved in the regulation of brain homeostasis, proteins related to adhesion or signaling as several G protein-coupled receptors (GPCRs), including the orphan GPRC5B and the proposed prosaposin receptor GPR37L1. Focusing on these two GPCRs, we could validate that they interact directly with MLC proteins. The inactivation of Gpr37l1 in mice upregulated MLC proteins without altering their localization. Conversely, a reduction of GPRC5B levels in primary astrocytes downregulated MLC proteins, leading to an impaired activation of ClC-2 and VRAC. The interaction between the GPCRs and MLC1 was dynamically regulated upon changes in the osmolarity or potassium concentration. We propose that GlialCAM and MLC1 associate with different integral membrane proteins modulating their functions and acting as a recruitment site for various signaling components as the GPCRs identified here. We hypothesized that the GlialCAM/MLC1 complex is working as an adhesion molecule coupled to a tetraspanin-like molecule performing regulatory effects through direct binding or influencing signal transduction events.


Asunto(s)
Quistes/genética , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/genética , Receptores Acoplados a Proteínas G/genética , Animales , Astrocitos/metabolismo , Encéfalo/metabolismo , Moléculas de Adhesión Celular Neurona-Glia/genética , Moléculas de Adhesión Celular Neurona-Glia/metabolismo , Proteínas de Ciclo Celular/genética , Canales de Cloruro/genética , Quistes/metabolismo , Células HEK293 , Células HeLa , Enfermedades Desmielinizantes del Sistema Nervioso Central Hereditarias/metabolismo , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Mutación , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Transporte de Proteínas , Receptores Acoplados a Proteínas G/metabolismo
10.
Nat Commun ; 12(1): 2344, 2021 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-33879794

RESUMEN

Direct determination of RNA structures and interactions in living cells is critical for understanding their functions in normal physiology and disease states. Here, we present PARIS2, a dramatically improved method for RNA duplex determination in vivo with >4000-fold higher efficiency than previous methods. PARIS2 captures ribosome binding sites on mRNAs, reporting translation status on a transcriptome scale. Applying PARIS2 to the U8 snoRNA mutated in the neurological disorder LCC, we discover a network of dynamic RNA structures and interactions which are destabilized by patient mutations. We report the first whole genome structure of enterovirus D68, an RNA virus that causes polio-like symptoms, revealing highly dynamic conformations altered by antiviral drugs and different pathogenic strains. We also discover a replication-associated asymmetry on the (+) and (-) strands of the viral genome. This study establishes a powerful technology for efficient interrogation of the RNA structurome and interactome in human diseases.


Asunto(s)
Enfermedades Transmisibles/genética , Enfermedades Transmisibles/metabolismo , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/metabolismo , Fotoquímica/métodos , ARN/química , ARN/metabolismo , Calcinosis/genética , Calcinosis/metabolismo , Quistes del Sistema Nervioso Central/genética , Quistes del Sistema Nervioso Central/metabolismo , Reactivos de Enlaces Cruzados , Enterovirus Humano D/genética , Furocumarinas , Genoma Viral , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Procesos Fotoquímicos , ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Nucleolar Pequeño/química , ARN Nucleolar Pequeño/genética , ARN Nucleolar Pequeño/metabolismo , ARN Viral/química , ARN Viral/genética
11.
Cerebrovasc Dis ; 50(3): 347-355, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33730735

RESUMEN

INTRODUCTION: Sodium MRI (23Na MRI) derived biomarkers such as tissue sodium concentration (TSC) provide valuable information on cell function and brain tissue viability and has become a reliable tool for the assessment of brain tumors and ischemic stroke beyond pathoanatomical morphology. Patients with major stroke often suffer from different degrees of underlying white matter lesions (WMLs) attributed to chronic small vessel disease. This study aimed to evaluate the WM TSC in patients with an acute ischemic stroke and to correlate the TSC with the extent of small vessel disease. Furthermore, the reliability of relative TSC (rTSC) compared to absolute TSC in these patients was analyzed. METHODOLOGY: We prospectively examined 62 patients with acute ischemic stroke (73 ± 13 years) between November 2016 and August 2019 from which 18 patients were excluded and thus 44 patients were evaluated. A 3D 23Na MRI was acquired in addition to a T2-TIRM and a diffusion-weighted image. Coregistration and segmentation were performed with SPM 12 based on the T2-TIRM image. The extension of WM T2 hyperintense lesions in each patient was classified using the Fazekas scale of WMLs. The absolute TSC in the WM region was correlated to the Fazekas grades. The stroke region was manually segmented on the coregistered absolute diffusion coefficient image and absolute, and rTSC was calculated in the stroke region and compared to nonischemic WM region. Statistical significance was evaluated using the Student t-test. RESULTS: For patients with Fazekas grade I (n = 25, age: 68.5 ± 15.1 years), mean TSC in WM was 55.57 ± 7.43 mM, and it was not statistically significant different from patients with Fazekas grade II (n = 7, age: 77.9 ± 6.4 years) with a mean TSC in WM of 53.9 ± 6.4 mM, p = 0.58. For patients with Fazekas grade III (n = 9, age: 81.4 ± 7.9 years), mean TSC in WM was 68.7 ± 10.5 mM, which is statistically significantly higher than the TSC in patients with Fazekas grade I and II (p < 0.001 and p = 0.05, respectively). There was a positive correlation between the TSC in WM and the Fazekas grade with r = 0.48 p < 0.001. The rTSC in the stroke region was statistically significant difference between low (0 and I) and high (2 and 3) Fazekas grades (p = 0.0353) whereas there was no statistically significant difference in absolute TSC in the stroke region between low (0 and I) and high (2 and 3) Fazekas grades. CONCLUSION: The significant difference in absolute TSC in WM in patients with severe small vessel disease; Fazekas grade 3 can lead to inaccuracies using rTSC quantification for evaluation of acute ischemic stroke using 23 Na MRI. The study, therefore, emphasizes the importance of absolute tissue sodium quantification.


Asunto(s)
Enfermedades de los Pequeños Vasos Cerebrales/diagnóstico por imagen , Accidente Cerebrovascular Isquémico/diagnóstico por imagen , Leucoencefalopatías/diagnóstico por imagen , Imagen por Resonancia Magnética , Isótopos de Sodio/metabolismo , Sustancia Blanca/diagnóstico por imagen , Anciano , Anciano de 80 o más Años , Enfermedades de los Pequeños Vasos Cerebrales/metabolismo , Imagen de Difusión por Resonancia Magnética , Femenino , Humanos , Interpretación de Imagen Asistida por Computador , Accidente Cerebrovascular Isquémico/metabolismo , Leucoencefalopatías/metabolismo , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Estudios Prospectivos , Reproducibilidad de los Resultados , Sustancia Blanca/metabolismo
13.
Hum Mol Genet ; 30(5): 331-342, 2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33517449

RESUMEN

Leukodystrophy with vanishing white matter (VWM), also called Childhood Ataxia with Central Nervous System Hypomyelination, is caused by mutations in the subunits of the eukaryotic translation initiation factor, EIF2B1, EIF2B2, EIF2B3, EIF2B4 or EIF2B5. However, little is known regarding the underlying pathogenetic mechanisms, and there is no curative treatment for VWM. In this study, we established the first EIF2B3 animal model for VWM disease in vertebrates by CRISPR mutagenesis of the highly conserved zebrafish ortholog eif2b3. Using CRISPR, we generated two mutant alleles in zebrafish eif2b3, 10- and 16-bp deletions, respectively. The eif2b3 mutants showed defects in myelin development and glial cell differentiation, and increased expression of genes in the induced stress response pathway. Interestingly, we also found ectopic angiogenesis and increased VEGF expression. Ectopic angiogenesis in the eif2b3 mutants was reduced by the administration of VEGF receptor inhibitor SU5416. Using the eif2b3 mutant zebrafish model together with in silico protein modeling analysis, we demonstrated the pathogenicity of 18 reported mutations in EIF2B3, as well as of a novel variant identified in a 19-month-old female patient: c.503 T > C (p.Leu168Pro). In summary, our zebrafish mutant model of eif2b3 provides novel insights into VWM pathogenesis and offers rapid functional analysis of human EIF2B3 gene variants.


Asunto(s)
Factor 2B Eucariótico de Iniciación/genética , Regulación del Desarrollo de la Expresión Génica , Leucoencefalopatías/genética , Vaina de Mielina/genética , Neovascularización Fisiológica , Pez Cebra/genética , Pez Cebra/metabolismo , Alelos , Animales , Diferenciación Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Modelos Animales de Enfermedad , Factor 2B Eucariótico de Iniciación/química , Femenino , Técnicas de Inactivación de Genes , Humanos , Lactante , Leucoencefalopatías/metabolismo , Modelos Moleculares , Vaina de Mielina/metabolismo , Neovascularización Fisiológica/genética , Conformación Proteica , Eliminación de Secuencia , Estrés Fisiológico , Factor A de Crecimiento Endotelial Vascular/metabolismo
14.
J Thorac Cardiovasc Surg ; 160(2): e55-e66, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32689704

RESUMEN

OBJECTIVES: This study aims to evaluate the protective effects of progesterone on white matter injury and brain immaturity in neonatal rats with chronic hypoxia. METHODS: Three-day old Sprague-Dawley rats were randomly divided into 3 groups: (1) control (n = 48), rats were exposed to normoxia (fraction of inspired oxygen: 21% ± 0%); (2) chronic hypoxia (n = 48), rats were exposed to hypoxia (fraction of inspired oxygen: 10.5% ± 1.0%); and (3) progesterone (n = 48), rats were exposed to hypoxia and administrated with progesterone (8 mg/kg/d). Hematoxylin-eosin staining, immunohistochemistry, real-time quantitative polymerase chain reaction, and Western blot analyses were compared on postnatal day 14 in different groups. Motor skill and coordination abilities of rats were assessed via rotation experiments. RESULTS: Increased brain weights (P < .05), narrowed ventricular sizes (P < .01), and rotarod experiment scores (P < .01) were better in the progesterone group than in the chronic hypoxia group. The number of mature oligodendrocytes and myelin basic protein expression increased in the progesterone group compared with the chronic hypoxia group (P < .01). The polarization of M1 microglia cells in the corpus callosum of chronic hypoxia-induced hypomyelination rats was significantly increased, whereas there were fewer M2 microglia cells. Conversely, progesterone therapy had an opposite effect and caused an increase in M2 microglia polarization versus a reduction in M1 microglia cells. CONCLUSIONS: Progesterone could prevent white matter injury and improve brain maturation in a neonatal hypoxic rat model; this may be associated with inducing a switch from M1 to M2 in microglia.


Asunto(s)
Encéfalo/efectos de los fármacos , Hipoxia/tratamiento farmacológico , Leucoencefalopatías/prevención & control , Fármacos Neuroprotectores/farmacología , Progesterona/farmacología , Sustancia Blanca/efectos de los fármacos , Animales , Animales Recién Nacidos , Conducta Animal/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Plasticidad de la Célula/efectos de los fármacos , Enfermedad Crónica , Modelos Animales de Enfermedad , Femenino , Hipoxia/metabolismo , Hipoxia/patología , Hipoxia/fisiopatología , Leucoencefalopatías/metabolismo , Leucoencefalopatías/patología , Leucoencefalopatías/fisiopatología , Masculino , Microglía/efectos de los fármacos , Microglía/metabolismo , Microglía/patología , Actividad Motora/efectos de los fármacos , Proteína Básica de Mielina/metabolismo , Oligodendroglía/efectos de los fármacos , Oligodendroglía/metabolismo , Oligodendroglía/patología , Ratas Sprague-Dawley , Sustancia Blanca/metabolismo , Sustancia Blanca/patología , Sustancia Blanca/fisiopatología
15.
Clin Nucl Med ; 45(10): e439-e440, 2020 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-32657880

RESUMEN

A 69-year-old woman presented with cognitive impairment related to attentive, executive, and mnemonic functions; progressive worsening of walking, speaking, writing, and reading ability; and double sphincter incontinence. Leukoencephalopathy, cystic lesions, and calcifications, suspected for Labrune syndrome, were observed at MRI and CT brain images. Generalized wave abnormalities were also visible at electroencephalogram. Functional brain imaging performed with F-FDG PET/CT demonstrated a decreased glucose metabolism in impaired brain regions, in accordance with MRI findings. Genetic testing confirmed a mutation of SNORD118.


Asunto(s)
Calcinosis/diagnóstico por imagen , Quistes del Sistema Nervioso Central/diagnóstico por imagen , Fluorodesoxiglucosa F18 , Leucoencefalopatías/diagnóstico por imagen , Tomografía Computarizada por Tomografía de Emisión de Positrones , Anciano , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Calcinosis/genética , Calcinosis/metabolismo , Quistes del Sistema Nervioso Central/genética , Quistes del Sistema Nervioso Central/metabolismo , Glucosa/metabolismo , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Imagen por Resonancia Magnética , Masculino , Mutación , ARN Nucleolar Pequeño/genética
16.
Stroke ; 51(6): 1750-1757, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32397933

RESUMEN

Background and Purpose- Distribution patterns of iron deposition in deep gray matter and their association with clinical characteristics in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) remain unclear. We aimed to evaluate iron deposition in deep gray matter in patients with CADASIL using 7.0-T susceptibility-weighted imaging and mapping and to explore its correlations with clinical characteristics. Methods- Thirty-nine patients with CADASIL, confirmed via genetic analysis or skin biopsy, were enrolled. We examined patients using the Mini-Mental State Examination, modified Rankin Scale, and brain 7.0-T magnetic resonance imaging and obtained magnetic resonance imaging lesion loads, small vessel disease scores, and susceptibility mapping. The following regions of interest were selected: caudate nucleus, putamen, globus pallidus, thalamus, substantia nigra, and red nucleus. The quantitative differences in the susceptibility of deep gray matter between the CADASIL and control groups and the correlations between deep gray matter susceptibility and clinical characteristics were identified. Results- Compared with the control group, the CADASIL group showed significantly increased susceptibility of caudate nucleus, putamen, thalamus, substantia nigra, and red nucleus. The susceptibility of deep gray matter in basal ganglia region, including caudate nucleus, putamen, and thalamus, significantly increased with age or disease duration and positively correlated with small vessel disease scores in patients with CADASIL. Moreover, the susceptibility of thalamus positively correlated with modified Rankin Scale scores after adjusting for age and disease duration and that of putamen negatively correlated with Mini-Mental State Examination scores in patients with CADASIL after adjusting for age. Conclusions- Our findings indicate an association between abnormal iron deposition in deep gray matter of patients with CADASIL and their clinical characteristics. Therefore, excess iron deposition in deep gray matter, as indicated by 7.0-T susceptibility-weighted imaging and mapping, might not only be a novel magnetic resonance imaging feature but also a potential biomarker for CADASIL severity.


Asunto(s)
Alopecia/diagnóstico por imagen , Alopecia/metabolismo , Infarto Cerebral/diagnóstico por imagen , Infarto Cerebral/metabolismo , Sustancia Gris , Hierro/metabolismo , Leucoencefalopatías/diagnóstico por imagen , Leucoencefalopatías/metabolismo , Imagen por Resonancia Magnética , Enfermedades de la Columna Vertebral/diagnóstico por imagen , Enfermedades de la Columna Vertebral/metabolismo , Adulto , Alopecia/genética , Infarto Cerebral/genética , Femenino , Sustancia Gris/diagnóstico por imagen , Sustancia Gris/metabolismo , Humanos , Leucoencefalopatías/genética , Masculino , Persona de Mediana Edad , Enfermedades de la Columna Vertebral/genética
17.
Biol Open ; 9(5)2020 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-32295832

RESUMEN

Human infantile-onset RNASET2-deficient cystic leukoencephalopathy is a Mendelian mimic of in utero cytomegalovirus brain infection with prenatally developing inflammatory brain lesions. We used an RNASET2-deficient zebrafish model to elucidate the underlying disease mechanisms. Mutant and wild-type zebrafish larvae brain development between 2 and 5 days post fertilization (dpf) was examined by confocal live imaging in fluorescent reporter lines of the major types of brain cells. In contrast to wild-type brains, RNASET2-deficient larvae displayed increased numbers of microglia with altered morphology, often containing inclusions of neurons. Furthermore, lysosomes within distinct populations of the myeloid cell lineage including microglia showed increased lysosomal staining. Neurons and oligodendrocyte precursor cells remained unaffected. This study provides a first look into the prenatal onset pathomechanisms of human RNASET2-deficient leukoencephalopathy, linking this inborn lysosomal disease to the innate immune system and other immune-related childhood encephalopathies like Aicardi-Goutières syndrome (AGS).


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Leucoencefalopatías/diagnóstico , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Microglía/metabolismo , Fenotipo , Ribonucleasas/deficiencia , Proteínas Supresoras de Tumor/deficiencia , Animales , Apoptosis , Susceptibilidad a Enfermedades , Estudios de Asociación Genética/métodos , Humanos , Larva , Neuronas/metabolismo , Organogénesis/genética , Rombencéfalo/anomalías , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Pez Cebra
18.
Orphanet J Rare Dis ; 14(1): 184, 2019 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-31349848

RESUMEN

BACKGROUND: Ribonucleases (RNases) are crucial for degradation of ribosomal RNA (rRNA). RNASET2 as a subtype of RNASEs is a 256 amino acid protein, encoded by RNASET2 gene located on chromosome six. Defective RNASET2 leads to RNASET2-deficient leukoencephalopathy, a rare autosomal recessive neurogenetic disorder with psychomotor delay as its main clinical symptom. The clinical findings can be similar to congenital cytomegalovirus (CMV) infection and Aicardi-Goutieres syndrome (AGS). METHODS: Herein, we presented a patient with motor delay, neurological regression, infrequent seizures and microcephaly at 5 months of age. Brain imaging showed white matter involvement, calcification and anterior temporal cysts. Basic metabolic tests, serum and urine CMV polymerase chain reaction (PCR) were requested. According to clinical and imaging findings, screening of RNASET2 and RMND1 genes were performed. The clinical data and magnetic resonance imaging (MRI) findings of previous reported individuals with RNASET2-deficient leukodystrophy were also reviewed and compared to the findings of our patient. RESULTS: Brain MRI findings were suggestive of RNASET2-deficient leukoencephalopathy, AGS and CMV infection. Basic metabolic tests were normal and CMV PCR was negative. Molecular study revealed a novel homozygous variant of c.233C > A; p.Ser78Ter in exon 4 of RNASET2 gene compatible with the diagnosis of RNASET2-deficient leukoencephalopathy. CONCLUSIONS: RNASET2-deficiency is a possible diagnosis in an infant presented with a static leukoencephalopathy and white matter involvement without megalencephaly. Due to overlapping clinical and radiologic features of RNASET2-deficient leukoencephalopathy, AGS and congenital CMV infections, molecular study as an important and helpful diagnostic tool should be considered to avoid misdiagnosis.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso/diagnóstico , Infecciones por Citomegalovirus/diagnóstico , Leucoencefalopatías/diagnóstico , Malformaciones del Sistema Nervioso/diagnóstico , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Encéfalo/diagnóstico por imagen , Encéfalo/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Preescolar , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/metabolismo , Femenino , Humanos , Leucoencefalopatías/genética , Leucoencefalopatías/metabolismo , Imagen por Resonancia Magnética , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Embarazo , Ribonucleasas/genética , Ribonucleasas/metabolismo , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
19.
Brain ; 141(8): 2289-2298, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-30010796

RESUMEN

Defects in iron-sulphur [Fe-S] cluster biogenesis are increasingly recognized as causing neurological disease. Mutations in a number of genes that encode proteins involved in mitochondrial [Fe-S] protein assembly lead to complex neurological phenotypes. One class of proteins essential in the early cluster assembly are ferredoxins. FDX2 is ubiquitously expressed and is essential in the de novo formation of [2Fe-2S] clusters in humans. We describe and genetically define a novel complex neurological syndrome identified in two Brazilian families, with a novel homozygous mutation in FDX2. Patients were clinically evaluated, underwent MRI, nerve conduction studies, EMG and muscle biopsy. To define the genetic aetiology, a combination of homozygosity mapping and whole exome sequencing was performed. We identified six patients from two apparently unrelated families with autosomal recessive inheritance of a complex neurological phenotype involving optic atrophy and nystagmus developing by age 3, followed by myopathy and recurrent episodes of cramps, myalgia and muscle weakness in the first or second decade of life. Sensory-motor axonal neuropathy led to progressive distal weakness. MRI disclosed a reversible or partially reversible leukoencephalopathy. Muscle biopsy demonstrated an unusual pattern of regional succinate dehydrogenase and cytochrome c oxidase deficiency with iron accumulation. The phenotype was mapped in both families to the same homozygous missense mutation in FDX2 (c.431C > T, p.P144L). The deleterious effect of the mutation was validated by real-time reverse transcription polymerase chain reaction and western blot analysis, which demonstrated normal expression of FDX2 mRNA but severely reduced expression of FDX2 protein in muscle tissue. This study describes a novel complex neurological phenotype with unusual MRI and muscle biopsy features, conclusively mapped to a mutation in FDX2, which encodes a ubiquitously expressed mitochondrial ferredoxin essential for early [Fe-S] cluster biogenesis.


Asunto(s)
Ferredoxinas/genética , Ferredoxinas/fisiología , Adolescente , Adulto , Brasil , Niño , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Homocigoto , Humanos , Hierro/metabolismo , Proteínas Hierro-Azufre/genética , Proteínas Hierro-Azufre/fisiología , Leucoencefalopatías/metabolismo , Masculino , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Enfermedades Musculares/genética , Mialgia/genética , Atrofia Óptica/genética , Linaje , Fenotipo , Succinato Deshidrogenasa/metabolismo , Síndrome , Secuenciación del Exoma
20.
Am J Med Genet B Neuropsychiatr Genet ; 177(1): 101-109, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29152901

RESUMEN

White matter lesions have been described in patients with PTEN hamartoma tumor syndrome (PHTS). How these lesions correlate with the neurocognitive features associated with PTEN mutations, such as autism spectrum disorder (ASD) or developmental delay, has not been well established. We report nine patients with PTEN mutations and white matter changes on brain magnetic resonance imaging (MRI), eight of whom were referred for reasons other than developmental delay or ASD. Their clinical presentations ranged from asymptomatic macrocephaly with normal development/intellect, to obsessive compulsive disorder, and debilitating neurological disease. To our knowledge, this report constitutes the first detailed description of PTEN-related white matter changes in adult patients and in children with normal development and intelligence. We present a detailed assessment of the neuropsychological phenotype of our patients and discuss the relationship between the wide array of neuropsychiatric features and observed white matter findings in the context of these individuals.


Asunto(s)
Síndrome de Hamartoma Múltiple/fisiopatología , Leucoencefalopatías/metabolismo , Fosfohidrolasa PTEN/metabolismo , Adolescente , Adulto , Trastorno del Espectro Autista/genética , Niño , Preescolar , Discapacidades del Desarrollo , Femenino , Síndrome de Hamartoma Múltiple/genética , Humanos , Inteligencia , Leucoencefalopatías/genética , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Fenotipo , Sustancia Blanca/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA