Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 259
Filtrar
1.
Zhonghua Bing Li Xue Za Zhi ; 53(5): 452-457, 2024 May 08.
Artículo en Chino | MEDLINE | ID: mdl-38678325

RESUMEN

Objective: To investigate the role of RNA m6A methylation in mediating cerebellar dysplasia through analyzing the phenotypes of the mouse cerebella and the expression of several key m6A regulators upon hypobaric hypoxia treatment. Methods: Five-day old C57/BL6 mice were exposed to hypobaric hypoxia for 9 days. The status of mouse cerebellar development was analyzed by comparing the body weights, brain weights and histological features. Immunostaining of cell-type-specific markers was performed to analyze the cerebellar morphology. Real-time PCR, Western blot and immunohistochemical staining were performed to detect the expression of key m6A regulators in the mouse cerebella. Results: Compared with the control, the body weights, brain weights and cerebellar volumes of hypobaric hypoxic mice were significantly reduced (P<0.01). The expression of specific markers in different cells, including NeuN (mature neuron), Calbindin-D28K (Purkinje cell) and GFAP (astrocyte), was decreased in hypobaric hypoxic mouse cerebella (P<0.01), accompanied with disorganized cellular structure. The expression of methyltransferase METTL3 was significantly down-regulated in the cerebella of hypobaric hypoxic mice (P<0.05). Conclusions: Hypobaric hypoxia stimulation causes mouse cerebellar dysplasia, with structural abnormalities in mature granular neurons, Purkinje cells and astrocytes. Expression of METTL3 is decreased in hypobaric hypoxic mice cerebellum compared with that of normobaric normoxic mice, suggesting that its mediated RNA m6A methylation may play an important role in hypobaric hypoxia-induced mouse cerebellar dysplasia.


Asunto(s)
Calbindinas , Cerebelo , Proteínas de Unión al ADN , Hipoxia , Metiltransferasas , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso , Células de Purkinje , Animales , Ratones , Cerebelo/metabolismo , Hipoxia/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Células de Purkinje/metabolismo , Células de Purkinje/patología , Calbindinas/metabolismo , Calbindinas/genética , Metiltransferasas/metabolismo , Metiltransferasas/genética , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Astrocitos/metabolismo , Regulación hacia Abajo , Metilación , Adenosina/metabolismo , Adenosina/análogos & derivados , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/genética
2.
Mol Psychiatry ; 29(3): 566-579, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38129659

RESUMEN

Three Prime Repair Exonuclease 1 (TREX1) gene mutations have been associated with Aicardi-Goutières Syndrome (AGS) - a rare, severe pediatric autoimmune disorder that primarily affects the brain and has a poorly understood etiology. Microglia are brain-resident macrophages indispensable for brain development and implicated in multiple neuroinflammatory diseases. However, the role of TREX1 - a DNase that cleaves cytosolic nucleic acids, preventing viral- and autoimmune-related inflammatory responses - in microglia biology remains to be elucidated. Here, we leverage a model of human embryonic stem cell (hESC)-derived engineered microglia-like cells, bulk, and single-cell transcriptomics, optical and transmission electron microscopy, and three-month-old assembloids composed of microglia and oligodendrocyte-containing organoids to interrogate TREX1 functions in human microglia. Our analyses suggest that TREX1 influences cholesterol metabolism, leading to an active microglial morphology with increased phagocytosis in the absence of TREX1. Notably, regulating cholesterol metabolism with an HMG-CoA reductase inhibitor, FDA-approved atorvastatin, rescues these microglial phenotypes. Functionally, TREX1 in microglia is necessary for the transition from gliogenic intermediate progenitors known as pre-oligodendrocyte precursor cells (pre-OPCs) to precursors of the oligodendrocyte lineage known as OPCs, impairing oligodendrogenesis in favor of astrogliogenesis in human assembloids. Together, these results suggest routes for therapeutic intervention in pathologies such as AGS based on microglia-specific molecular and cellular mechanisms.


Asunto(s)
Diferenciación Celular , Colesterol , Exodesoxirribonucleasas , Homeostasis , Microglía , Oligodendroglía , Fosfoproteínas , Humanos , Exodesoxirribonucleasas/metabolismo , Exodesoxirribonucleasas/genética , Microglía/metabolismo , Diferenciación Celular/fisiología , Oligodendroglía/metabolismo , Colesterol/metabolismo , Fosfoproteínas/metabolismo , Homeostasis/fisiología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/genética , Encéfalo/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Organoides/metabolismo
3.
Zhonghua Yu Fang Yi Xue Za Zhi ; 57(6): 923-928, 2023 Jun 06.
Artículo en Chino | MEDLINE | ID: mdl-37357214

RESUMEN

To establish and identify induced pluripotent stem cells (iPSCs) derived from patients with Aicardi-Goutières syndrome (AGS) with TREX1 gene 667G>A mutation, and obtain a specific induced pluripotent stem cell model for Aicardi-Goutières syndrome (AGS-iPSCs). A 3-year-old male child with Aicardi-Goutieres syndrome was admitted to Zhongshan People's Hospital in December 2020. After obtaining the informed consent of the patient's family members, 5 ml peripheral blood samples from the patient were collected, and mononuclear cells were isolated. Then,the peripheral blood mononuclear cells(PBMCs) were transduced with OCT3/4, SOX2, c-Myc and Klf4 by using Sendai virus, and PBMCs were reprogrammed into iPSCs. The pluripotency and differentiation ability of the cells were identified by cellular morphological analysis, real-time PCR, alkaline phosphatase staining (AP), immunofluorescence, teratoma formation experiments in mice. The results showed that the induced pluripotent stem cell line of Aicardi-Goutieres syndrome was successfully constructed and showed typical embryonic stem-like morphology after stable passage, RT-PCR showed mRNA expression of stem cell markers, AP staining was positive, OCT4, SOX2, NANOG, SSEA4, TRA-1-81 and TRA-1-60 pluripotency marker proteins were strongly expressed. In vivo teratoma formation experiments showed that iPSCs differentiate into the ectoderm (neural tube like tissue), mesoderm (vascular wall tissue) and endoderm (glandular tissue). Karyotype analysis also confirmed that iPSCs still maintained the original karyotype (46, XY). In conclusion, induced pluripotent stem cell line for Aicardi-Goutières syndrome was successfully established using Sendai virus, which provided an important model platform for studying the pathogenesis of the disease and for drug screening.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Malformaciones del Sistema Nervioso , Animales , Preescolar , Humanos , Masculino , Ratones , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/patología , Diferenciación Celular , Línea Celular , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Leucocitos Mononucleares , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/patología
4.
Front Immunol ; 14: 1100967, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36949945

RESUMEN

Aicardi-Goutières syndrome (AGS1-9) is a genetically determined encephalopathy that falls under the type I interferonopathy disease class, characterized by excessive type I interferon (IFN-I) activity, coupled with upregulation of IFN-stimulated genes (ISGs), which can be explained by the vital role these proteins play in self-non-self-discrimination. To date, few mouse models fully replicate the vast clinical phenotypes observed in AGS patients. Therefore, we investigated the use of zebrafish as an alternative species for generating a clinically relevant model of AGS. Using CRISPR-cas9 technology, we generated a stable mutant zebrafish line recapitulating AGS5, which arises from recessive mutations in SAMHD1. The resulting homozygous mutant zebrafish larvae possess a number of neurological phenotypes, exemplified by variable, but increased expression of several ISGs in the head region, a significant increase in brain cell death, microcephaly and locomotion deficits. A link between IFN-I signaling and cholesterol biosynthesis has been highlighted by others, but not previously implicated in the type I interferonopathies. Through assessment of neurovascular integrity and qPCR analysis we identified a significant dysregulation of cholesterol biosynthesis in the zebrafish model. Furthermore, dysregulation of cholesterol biosynthesis gene expression was also observed through RNA sequencing analysis of AGS patient whole blood. From this novel finding, we hypothesize that cholesterol dysregulation may play a role in AGS disease pathophysiology. Further experimentation will lend critical insight into the molecular pathophysiology of AGS and the potential links involving aberrant type I IFN signaling and cholesterol dysregulation.


Asunto(s)
Enfermedades Autoinmunes del Sistema Nervioso , Interferón Tipo I , Malformaciones del Sistema Nervioso , Animales , Ratones , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Interferón Tipo I/genética , Interferón Tipo I/metabolismo , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Proteína 1 que Contiene Dominios SAM y HD/genética , Pez Cebra/genética , Pez Cebra/metabolismo
5.
Sci Adv ; 8(13): eabl9156, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35363520

RESUMEN

Recent studies using single-cell RNA-sequencing have revealed cellular heterogeneity in the developing mammalian cerebellum, yet the regulatory logic underlying this cellular diversity remains to be elucidated. Using integrated single-cell RNA and ATAC analyses, we resolved developmental trajectories of cerebellar progenitors and identified putative trans- and cis-elements that control cell state transition. We reverse engineered gene regulatory networks (GRNs) of each cerebellar cell type. Through in silico simulations and in vivo experiments, we validated the efficacy of GRN analyses and uncovered the molecular control of a posterior transitory zone (PTZ), a distinct progenitor zone residing immediately anterior to the morphologically defined rhombic lip (RL). We showed that perturbing cell fate specification in the PTZ and RL causes posterior cerebellar vermis hypoplasia, the most common cerebellar birth defect in humans. Our study provides a foundation for comprehensive studies of developmental programs of the mammalian cerebellum.


Asunto(s)
Malformaciones del Sistema Nervioso , Transcriptoma , Animales , Diferenciación Celular/genética , Cerebelo/metabolismo , Epigénesis Genética , Mamíferos/genética , Ratones , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo
6.
Hum Genet ; 141(8): 1423-1429, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35107634

RESUMEN

Glycosylphosphatidylinositol (GPI) functions to anchor certain proteins to the cell surface. Although defects in GPI biosynthesis can result in a wide range of phenotypes, most affected patients present with neurological abnormalities and their diseases are grouped as inherited-GPI deficiency disorders. We present two siblings with global developmental delay, brain anomalies, hypotonia, and contractures. Exome sequencing revealed a homozygous variant, NM_001035005.4:c.90dupC (p.Phe31Leufs*3) in C18orf32, a gene not previously associated with any disease in humans. The encoded protein is known to be important for GPI-inositol deacylation. Knockout of C18orf32 in HEK293 cells followed by a transfection rescue assay revealed that the PIPLC (Phosphatidylinositol-Specific Phospholipase C) sensitivity of GPI-APs (GPI-anchored proteins) was restored only by the wild type and not the mutant C18orf32. Immunofluorescence revealed that the mutant C18orf32 was localized to the endoplasmic reticulum and was also found as aggregates in the nucleus. In conclusion, we identified a pathogenic variant in C18orf32 as the cause of a novel autosomal recessive neurodevelopmental disorder with hypotonia and contractures. Our results demonstrate the importance of C18orf32 in the biosynthesis of GPI-anchors, the molecular impact of the variant on the protein function, and add a novel candidate gene to the existing repertoire of genes implicated in neurodevelopmental disorders.


Asunto(s)
Contractura , Hipotonía Muscular , Malformaciones del Sistema Nervioso , Trastornos del Neurodesarrollo , Contractura/genética , Contractura/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Células HEK293 , Humanos , Hipotonía Muscular/genética , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Trastornos del Neurodesarrollo/genética , Trastornos del Neurodesarrollo/metabolismo
7.
Nat Commun ; 13(1): 16, 2022 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-35013230

RESUMEN

Primary microcephaly and megalencephaly are severe brain malformations defined by reduced and increased brain size, respectively. Whether these two pathologies arise from related alterations at the molecular level is unclear. Microcephaly has been largely associated with centrosomal defects, leading to cell death. Here, we investigate the consequences of WDR81 loss of function, which causes severe microcephaly in patients. We show that WDR81 regulates endosomal trafficking of EGFR and that loss of function leads to reduced MAP kinase pathway activation. Mouse radial glial progenitor cells knocked-out for WDR81 exhibit reduced proliferation rate, subsequently leading to reduced brain size. These proliferation defects are rescued in vivo by expressing a megalencephaly-causing mutant form of Cyclin D2. Our results identify the endosomal machinery as an important regulator of proliferation rates and brain growth, demonstrating that microcephaly and megalencephaly can be caused by opposite effects on the proliferation rate of radial glial progenitors.


Asunto(s)
Proliferación Celular , Microcefalia , Proteínas del Tejido Nervioso/metabolismo , Células-Madre Neurales/metabolismo , Vesículas Transportadoras , Animales , Encéfalo/embriología , Encéfalo/metabolismo , Células Cultivadas , Endosomas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Sistema de Señalización de MAP Quinasas , Megalencefalia/etiología , Megalencefalia/metabolismo , Megalencefalia/patología , Ratones , Microcefalia/etiología , Microcefalia/metabolismo , Microcefalia/patología , Malformaciones del Sistema Nervioso/etiología , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/patología , Neuroglía/metabolismo , Transporte de Proteínas/fisiología , Vesículas Transportadoras/metabolismo , Vesículas Transportadoras/patología
8.
Immunity ; 54(9): 1948-1960.e5, 2021 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-34343497

RESUMEN

The RNA deaminase ADAR1 is an essential negative regulator of the RNA sensor MDA5, and loss of ADAR1 function triggers inappropriate activation of MDA5 by self-RNAs. Mutations in ADAR, the gene that encodes ADAR1, cause human immune diseases, including Aicardi-Goutières syndrome (AGS). However, the mechanisms of MDA5-dependent disease pathogenesis in vivo remain unknown. Here we generated mice with a single amino acid change in ADAR1 that models the most common human ADAR AGS mutation. These Adar mutant mice developed lethal disease that required MDA5, the RIG-I-like receptor LGP2, type I interferons, and the eIF2α kinase PKR. A small-molecule inhibitor of the integrated stress response (ISR) that acts downstream of eIF2α phosphorylation prevented immunopathology and rescued the mice from mortality. These findings place PKR and the ISR as central components of immunopathology in vivo and identify therapeutic targets for treatment of human diseases associated with the ADAR1-MDA5 axis.


Asunto(s)
Adenosina Desaminasa/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/patología , Malformaciones del Sistema Nervioso/patología , Estrés Fisiológico/fisiología , eIF-2 Quinasa/metabolismo , Células A549 , Animales , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Modelos Animales de Enfermedad , Células HEK293 , Humanos , Ratones , Ratones Mutantes , Mutación , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo
9.
J Neuroinflammation ; 18(1): 169, 2021 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-34332594

RESUMEN

BACKGROUND: Aicardi-Goutières syndrome (AGS) is a severe infant or juvenile-onset autoimmune disease characterized by inflammatory encephalopathy with an elevated type 1 interferon-stimulated gene (ISG) expression signature in the brain. Mutations in seven different protein-coding genes, all linked to DNA/RNA metabolism or sensing, have been identified in AGS patients, but none of them has been demonstrated to activate the IFN pathway in the brain of an animal. The molecular mechanism of inflammatory encephalopathy in AGS has not been well defined. Adenosine Deaminase Acting on RNA 1 (ADAR1) is one of the AGS-associated genes. It carries out A-to-I RNA editing that converts adenosine to inosine at double-stranded RNA regions. Whether an AGS-associated mutation in ADAR1 activates the IFN pathway and causes autoimmune pathogenesis in the brain is yet to be determined. METHODS: Mutations in the ADAR1 gene found in AGS patients were introduced into the mouse genome via CRISPR/Cas9 technology. Molecular activities of the specific p.K999N mutation were investigated by measuring the RNA editing levels in brain mRNA substrates of ADAR1 through RNA sequencing analysis. IFN pathway activation in the brain was assessed by measuring ISG expression at the mRNA and protein level through real-time RT-PCR and Luminex assays, respectively. The locations in the brain and neural cell types that express ISGs were determined by RNA in situ hybridization (ISH). Potential AGS-related brain morphologic changes were assessed with immunohistological analysis. Von Kossa and Luxol Fast Blue staining was performed on brain tissue to assess calcification and myelin, respectively. RESULTS: Mice bearing the ADAR1 p.K999N were viable though smaller than wild type sibs. RNA sequencing analysis of neuron-specific RNA substrates revealed altered RNA editing activities of the mutant ADAR1 protein. Mutant mice exhibited dramatically elevated levels of multiple ISGs within the brain. RNA ISH of brain sections showed selective activation of ISG expression in neurons and microglia in a patchy pattern. ISG-15 mRNA was upregulated in ADAR1 mutant brain neurons whereas CXCL10 mRNA was elevated in adjacent astroglia. No calcification or gliosis was detected in the mutant brain. CONCLUSIONS: We demonstrated that an AGS-associated mutation in ADAR1, specifically the p.K999N mutation, activates the IFN pathway in the mouse brain. The ADAR1 p.K999N mutant mouse replicates aspects of the brain interferonopathy of AGS. Neurons and microglia express different ISGs. Basal ganglia calcification and leukodystrophy seen in AGS patients were not observed in K999N mutant mice, indicating that development of the full clinical phenotype may need an additional stimulus besides AGS mutations. This mutant mouse presents a robust tool for the investigation of AGS and neuroinflammatory diseases including the modeling of potential "second hits" that enable severe phenotypes of clinically variable diseases.


Asunto(s)
Adenosina Desaminasa/genética , Enfermedades Autoinmunes del Sistema Nervioso/genética , Encéfalo/inmunología , Inmunidad Innata/genética , Mutación , Malformaciones del Sistema Nervioso/genética , Animales , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Quimiocinas/metabolismo , Citocinas/metabolismo , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Ratones , Malformaciones del Sistema Nervioso/inmunología , Malformaciones del Sistema Nervioso/metabolismo , Edición de ARN
10.
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
11.
J Biol Chem ; 296: 100389, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33561442

RESUMEN

The c-RET proto-oncogene encodes a receptor-tyrosine kinase. Loss-of-function mutations of RET have been shown to be associated with Hirschsprung disease and Down's syndrome (HSCR-DS) in humans. DS is known to involve cerebellar hypoplasia, which is characterized by reduced cerebellar size. Despite the fact that c-Ret has been shown to be associated with HSCR-DS in humans and to be expressed in Purkinje cells (PCs) in experimental animals, there is limited information about the role of activity of c-Ret/c-RET kinase in cerebellar hypoplasia. We found that a loss-of-function mutation of c-Ret Y1062 in PCs causes cerebellar hypoplasia in c-Ret mutant mice. Wild-type mice had increased phosphorylation of c-Ret in PCs during postnatal development, while c-Ret mutant mice had postnatal hypoplasia of the cerebellum with immature neurite outgrowth in PCs and granule cells (GCs). c-Ret mutant mice also showed decreased numbers of glial fibers and mitogenic sonic hedgehog (Shh)-positive vesicles in the external germinal layer of PCs. c-Ret-mediated cerebellar hypoplasia was rescued by subcutaneous injection of a smoothened agonist (SAG) as well as by reduced expression of Patched1, a negative regulator for Shh. Our results suggest that the loss-of-function mutation of c-Ret Y1062 results in the development of cerebellar hypoplasia via impairment of the Shh-mediated development of GCs and glial fibers in mice with HSCR-DS.


Asunto(s)
Cerebelo/anomalías , Síndrome de Down/genética , Enfermedad de Hirschsprung/genética , Mutación con Pérdida de Función , Malformaciones del Sistema Nervioso/genética , Proteínas Proto-Oncogénicas c-ret/genética , Animales , Cerebelo/metabolismo , Cerebelo/patología , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/metabolismo , Discapacidades del Desarrollo/patología , Modelos Animales de Enfermedad , Síndrome de Down/complicaciones , Síndrome de Down/metabolismo , Síndrome de Down/patología , Técnicas de Sustitución del Gen/métodos , Proteínas Hedgehog/metabolismo , Enfermedad de Hirschsprung/complicaciones , Enfermedad de Hirschsprung/metabolismo , Enfermedad de Hirschsprung/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/patología , Neuroglía/metabolismo , Neuroglía/patología , Fosforilación , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-ret/metabolismo , Células de Purkinje/metabolismo , Células de Purkinje/patología
12.
Semin Cell Dev Biol ; 111: 23-31, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-32718852

RESUMEN

Human brain organoids are self-organizing three-dimensional structures that emerge from human pluripotent stem cells and mimic aspects of the cellular composition and functionality of the developing human brain. Despite their impressive self-organizing capacity, organoids lack the stereotypic structural anatomy of their in vivo counterpart, making conventional analysis techniques underpowered to assess cellular composition and gene network regulation in organoids. Advances in single cell transcriptomics have recently allowed characterization and improvement of organoid protocols, as they continue to evolve, by enabling identification of cell types and states along with their developmental origins. In this review, we summarize recent approaches, progresses and challenges in resolving brain organoid's complexity through single-cell transcriptomics. We then discuss emerging technologies that may complement single-cell RNA sequencing by providing additional readouts of cellular states to generate an organ-level view of developmental processes. Altogether, these integrative technologies will allow monitoring of global gene regulation in thousands of individual cells and will offer an unprecedented opportunity to investigate features of human brain development and disease across multiple cellular modalities and with cell-type resolution.


Asunto(s)
Encéfalo/metabolismo , Proteínas del Tejido Nervioso/genética , Malformaciones del Sistema Nervioso/genética , Organoides/metabolismo , Análisis de la Célula Individual/métodos , Transcriptoma , Encéfalo/patología , Diferenciación Celular , Linaje de la Célula/genética , Células Ependimogliales/citología , Células Ependimogliales/metabolismo , Regulación de la Expresión Génica , Humanos , Modelos Biológicos , Mutación , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/patología , Malformaciones del Sistema Nervioso/fisiopatología , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neuronas/citología , Neuronas/metabolismo , Organoides/patología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Análisis de Secuencia de ARN
13.
Hum Mol Genet ; 29(23): 3757-3764, 2021 02 04.
Artículo en Inglés | MEDLINE | ID: mdl-33205811

RESUMEN

Congenital hydrocephalus is a potentially devastating, highly heterogeneous condition whose genetic subset remains incompletely known. We here report a consanguineous family where three fetuses presented with brain ventriculomegaly and limb contractures and shared a very rare homozygous variant of KIDINS220, consisting of an in-frame deletion of three amino acids adjacent to the fourth transmembrane domain. Fetal brain imaging and autopsy showed major ventriculomegaly, reduced brain mass, and with no histomorphologic abnormalities. We demonstrate that the binding of KIDINS220 to TrkA is diminished by the deletion mutation. This family is the second that associates a KIDINS220 genetic variant with human ventriculomegaly and limb contractures, validating causality of the gene and indicating TrkA as a likely mediator of the phenotype.


Asunto(s)
Feto/patología , Hidrocefalia/patología , Proteínas de la Membrana/genética , Mutación , Proteínas del Tejido Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Receptor trkA/metabolismo , Femenino , Feto/metabolismo , Homocigoto , Humanos , Hidrocefalia/etiología , Hidrocefalia/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Malformaciones del Sistema Nervioso/etiología , Malformaciones del Sistema Nervioso/metabolismo , Linaje , Receptor trkA/genética
14.
JCI Insight ; 5(18)2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32938829

RESUMEN

Hydrocephalus is a serious condition that impacts patients of all ages. The standards of care are surgical options to divert, or inhibit production of, cerebrospinal fluid; to date, there are no effective pharmaceutical treatments, to our knowledge. The causes vary widely, but one commonality of this condition is aberrations in salt and fluid balance. We have used a genetic model of hydrocephalus to show that ventriculomegaly can be alleviated by inhibition of the transient receptor potential vanilloid 4, a channel that is activated by changes in osmotic balance, temperature, pressure and inflammatory mediators. The TRPV4 antagonists do not appear to have adverse effects on the overall health of the WT or hydrocephalic animals.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Modelos Animales de Enfermedad , Hidrocefalia/tratamiento farmacológico , Morfolinas/farmacología , Malformaciones del Sistema Nervioso/tratamiento farmacológico , Pirroles/farmacología , Canales Catiónicos TRPV/antagonistas & inhibidores , Animales , Corteza Cerebral/patología , Hidrocefalia/metabolismo , Hidrocefalia/patología , Malformaciones del Sistema Nervioso/metabolismo , Malformaciones del Sistema Nervioso/patología , Ratas
15.
Nat Commun ; 11(1): 3165, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32576829

RESUMEN

SAMHD1 regulates cellular 2'-deoxynucleoside-5'-triphosphate (dNTP) homeostasis by catalysing the hydrolysis of dNTPs into 2'-deoxynucleosides and triphosphate. In CD4+ myeloid lineage and resting T-cells, SAMHD1 blocks HIV-1 and other viral infections by depletion of the dNTP pool to a level that cannot support replication. SAMHD1 mutations are associated with the autoimmune disease Aicardi-Goutières syndrome and hypermutated cancers. Furthermore, SAMHD1 sensitises cancer cells to nucleoside-analogue anti-cancer therapies and is linked with DNA repair and suppression of the interferon response to cytosolic nucleic acids. Nevertheless, despite its requirement in these processes, the fundamental mechanism of SAMHD1-catalysed dNTP hydrolysis remained unknown. Here, we present structural and enzymological data showing that SAMHD1 utilises an active site, bi-metallic iron-magnesium centre that positions a hydroxide nucleophile in-line with the Pα-O5' bond to catalyse phosphoester bond hydrolysis. This precise molecular mechanism for SAMHD1 catalysis, reveals how SAMHD1 down-regulates cellular dNTP and modulates the efficacy of nucleoside-based anti-cancer and anti-viral therapies.


Asunto(s)
Nucleósido-Trifosfatasa/química , Proteína 1 que Contiene Dominios SAM y HD/química , Agua/química , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , VIH-1/genética , VIH-1/fisiología , Humanos , Hidrólisis , Interferones , Modelos Moleculares , Mutación , Malformaciones del Sistema Nervioso/metabolismo , Polifosfatos , Conformación Proteica , Proteína 1 que Contiene Dominios SAM y HD/genética , Replicación Viral/fisiología
16.
Epilepsy Behav ; 108: 107068, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32375098

RESUMEN

Increasing awareness of the congenital and developmental risks associated with the use of sodium valproate (VPA) has led to recent European guidelines designed to avoid the use of this drug in pregnancy if effective alternative treatments are available. In the general population, it is well established that periconceptual folic acid reduces the risk of neural tube defects (NTDs) and possibly other congenital abnormalities. We here review the evidence 1) that VPA interferes with one-carbon metabolism, including the transport of methylfolate into the brain and the placenta by targeting folate receptors; 2) that VPA effects on the folate metabolic system contribute to congenital and developmental problems associated with VPA exposure; and 3) that genetic factors, notably polymorphisms related to one-carbon metabolism, contribute to the vulnerability to these VPA-induced risks. Based on these facts, we propose that the standard periconceptual use of 400 µg of folic acid may not adequately protect against VPA or other antiepileptic drug (AED)-induced congenital or developmental risks. Pending definitive studies to determine appropriate dose, we recommend up to 5 mg of folic acid periconceptually in at-risk women with the caveat that the addition of supplementary vitamin B12 may also be prudent because vitamin B12 deficiency is common in pregnancy in some countries and is an additional risk factor for developmental abnormalities.


Asunto(s)
Anticonvulsivantes/efectos adversos , Ácido Fólico/uso terapéutico , Malformaciones del Sistema Nervioso/prevención & control , Trastornos del Neurodesarrollo/prevención & control , Ácido Valproico/efectos adversos , Complejo Vitamínico B/uso terapéutico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Ácido Fólico/metabolismo , Ácido Fólico/farmacología , Humanos , Malformaciones del Sistema Nervioso/inducido químicamente , Malformaciones del Sistema Nervioso/metabolismo , Defectos del Tubo Neural/inducido químicamente , Defectos del Tubo Neural/metabolismo , Defectos del Tubo Neural/prevención & control , Trastornos del Neurodesarrollo/inducido químicamente , Trastornos del Neurodesarrollo/metabolismo , Embarazo , Complejo Vitamínico B/metabolismo , Complejo Vitamínico B/farmacología
17.
Proc Natl Acad Sci U S A ; 117(19): 10565-10574, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32345721

RESUMEN

Numerous mutations that impair retrograde membrane trafficking between endosomes and the Golgi apparatus lead to neurodegenerative diseases. For example, mutations in the endosomal retromer complex are implicated in Alzheimer's and Parkinson's diseases, and mutations of the Golgi-associated retrograde protein (GARP) complex cause progressive cerebello-cerebral atrophy type 2 (PCCA2). However, how these mutations cause neurodegeneration is unknown. GARP mutations in yeast, including one causing PCCA2, result in sphingolipid abnormalities and impaired cell growth that are corrected by treatment with myriocin, a sphingolipid synthesis inhibitor, suggesting that alterations in sphingolipid metabolism contribute to cell dysfunction and death. Here we tested this hypothesis in wobbler mice, a murine model with a homozygous partial loss-of-function mutation in Vps54 (GARP protein) that causes motor neuron disease. Cytotoxic sphingoid long-chain bases accumulated in embryonic fibroblasts and spinal cords from wobbler mice. Remarkably, chronic treatment of wobbler mice with myriocin markedly improved their wellness scores, grip strength, neuropathology, and survival. Proteomic analyses of wobbler fibroblasts revealed extensive missorting of lysosomal proteins, including sphingolipid catabolism enzymes, to the Golgi compartment, which may contribute to the sphingolipid abnormalities. Our findings establish that altered sphingolipid metabolism due to GARP mutations contributes to neurodegeneration and suggest that inhibiting sphingolipid synthesis might provide a useful strategy for treating these disorders.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Esfingolípidos/metabolismo , Animales , Modelos Animales de Enfermedad , Endosomas/metabolismo , Ácidos Grasos Monoinsaturados/farmacología , Femenino , Fibroblastos/metabolismo , Aparato de Golgi/metabolismo , Masculino , Ratones , Ratones Mutantes Neurológicos , Enfermedad de la Neurona Motora/genética , Enfermedad de la Neurona Motora/metabolismo , Enfermedad de la Neurona Motora/patología , Neuronas Motoras/metabolismo , Células Madre Embrionarias de Ratones , Mutación , Malformaciones del Sistema Nervioso/metabolismo , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/fisiopatología , Transporte de Proteínas , Proteómica , Proteínas de Transporte Vesicular/metabolismo
18.
Clin Pharmacol Ther ; 108(2): 358-367, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32243572

RESUMEN

Ataxia telangiectasia (AT) and Aicardi-Goutières syndrome (AGS) are inherited disorders of immunity with prevalent neurological phenotype. Available treatments are only partially effective, and the prognosis is poor. Induced pluripotent stem cells (iPSCs) are obtained by reprogramming patient somatic cells, preserving the donor individual genetic heritage and creating patient-specific disease models, useful to investigate pathogenesis and drug effects and to develop precision therapies. The aim is to investigate the cytotoxicity of a panel of immunomodulators using iPSCs of patients with AT or different forms of AGS (AGS1, AGS2, and AGS7). iPSCs were obtained by reprogramming AT and AGS patients' cells and, as a control, the BJ normal human fibroblast line, using Sendai virus. Cytotoxic effects of two drugs proposed to treat respectively AT and AGS (dexamethasone and mepacrine) were tested by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay after 72 hours' exposure. Data were obtained also for other immunomodulatory drugs (thioguanine, mercaptopurine, thalidomide, and lenalidomide). Relative expression of genes involved in the tested drug pathways was analyzed. AGS7-derived iPSCs displayed altered viability when treated with a low dose of mepacrine and higher expression of cyclic guanosine monophosphate-adenosine monophosphate synthase, which is the main target for mepacrine action. AGS7-derived iPSCs were also more sensitive to thioguanine, while AGS2 and AT iPSCs were less sensitive to this medication than the BJ-iPSC. All iPSCs were equally sensitive to mercaptopurine and resistant to dexamethasone, thalidomide, and lenalidomide. This work establishes an innovative in vitro model that is useful to investigate the mechanisms of drugs potentially effective in AT and AGS.


Asunto(s)
Ataxia Telangiectasia/tratamiento farmacológico , Enfermedades Autoinmunes del Sistema Nervioso/tratamiento farmacológico , Factores Inmunológicos/farmacología , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Malformaciones del Sistema Nervioso/tratamiento farmacológico , Medicina de Precisión , Ataxia Telangiectasia/genética , Ataxia Telangiectasia/inmunología , Ataxia Telangiectasia/metabolismo , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Biomarcadores/metabolismo , Línea Celular , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Toma de Decisiones Clínicas , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Resistencia a Medicamentos , Predisposición Genética a la Enfermedad , Humanos , Células Madre Pluripotentes Inducidas/inmunología , Células Madre Pluripotentes Inducidas/metabolismo , Lenalidomida/farmacología , Mercaptopurina/farmacología , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/inmunología , Malformaciones del Sistema Nervioso/metabolismo , Fenotipo , Valor Predictivo de las Pruebas , Quinacrina/farmacología , Talidomida/farmacología , Tioguanina/farmacología
19.
J Immunol Methods ; 480: 112751, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31982420

RESUMEN

Intracellular DNA triggers interferon release during the innate immune response. Cyclic GMP-AMP synthase (cGAS) senses intracellular double-stranded DNA not only in response to viral infection but also under autoimmune conditions. Measuring the levels of cyclic GMP-AMP (cGAMP) as a second messenger of cGAS activation is important to elucidate the physiological and pathological roles of cGAS. Therefore, we generated monoclonal antibodies against cGAMP using hybridoma technology to test antibody specificity and establish methods to detect intracellular cGAMP. The resulting cGAMP-specific antibody enabled the development of a time-resolved fluorescence energy transfer assay with a quantifiable range of 0.1 nM to 100 nM cGAMP. Using this assay, we detected cellular and tissue cGAMP. We confirmed that the cGAMP antibody successfully targeted intracellular cGAMP through immunocytochemical analyses. These results demonstrated that the cGAMP antibody is a powerful tool that allows determining cGAS involvement in autoimmunity and disease pathology at the cell and tissue levels.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Enfermedades Autoinmunes del Sistema Nervioso/metabolismo , Transferencia Resonante de Energía de Fluorescencia , Inmunohistoquímica , Neoplasias/metabolismo , Malformaciones del Sistema Nervioso/metabolismo , Nucleótidos Cíclicos/metabolismo , Nucleotidiltransferasas/metabolismo , Animales , Especificidad de Anticuerpos , Enfermedades Autoinmunes del Sistema Nervioso/genética , Enfermedades Autoinmunes del Sistema Nervioso/inmunología , Autoinmunidad , Biomarcadores/metabolismo , Células CACO-2 , Modelos Animales de Enfermedad , Activación Enzimática , Exodesoxirribonucleasas/deficiencia , Exodesoxirribonucleasas/genética , Células HEK293 , Células HL-60 , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/inmunología , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/inmunología , Nucleótidos Cíclicos/inmunología , Nucleotidiltransferasas/genética , Fosfoproteínas/deficiencia , Fosfoproteínas/genética , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
20.
Brain ; 143(2): 491-502, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31851307

RESUMEN

Primary familial brain calcification is a monogenic disease characterized by bilateral calcifications in the basal ganglia and other brain regions, and commonly presents motor, psychiatric, and cognitive symptoms. Currently, four autosomal dominant (SLC20A2, PDGFRB, PDGFB, XPR1) and one autosomal recessive (MYORG) causative genes have been identified. Compared with patients with autosomal dominant primary familial brain calcification, patients with the recessive form of the disease present with more severe clinical and imaging phenotypes, and deserve more clinical and research attention. Biallelic mutations in MYORG cannot explain all autosomal recessive primary familial brain calcification cases, indicating the existence of novel autosomal recessive genes. Using homozygosity mapping and whole genome sequencing, we detected a homozygous frameshift mutation (c.140delT, p.L48*) in the JAM2 gene in a consanguineous family with two affected siblings diagnosed with primary familial brain calcification. Further genetic screening in a cohort of 398 probands detected a homozygous start codon mutation (c.1A>G, p.M1?) and compound heterozygous mutations [c.504G>C, p.W168C and c.(67+1_68-1)_(394+1_395-1), p.Y23_V131delinsL], respectively, in two unrelated families. The clinical phenotypes of the four patients included parkinsonism (3/4), dysarthria (3/4), seizures (1/4), and probable asymptomatic (1/4), with diverse onset ages. All patients presented with severe calcifications in the cortex in addition to extensive calcifications in multiple brain areas (lenticular nuclei, caudate nuclei, thalamus, cerebellar hemispheres, ± brainstem; total calcification scores: 43-77). JAM2 encodes junctional adhesion molecule 2, which is highly expressed in neurovascular unit-related cell types (endothelial cells and astrocytes) and is predominantly localized on the plasma membrane. It may be important in cell-cell adhesion and maintaining homeostasis in the CNS. In Chinese hamster ovary cells, truncated His-tagged JAM2 proteins were detected by western blot following transfection of p.Y23_V131delinsL mutant plasmid, while no protein was detected following transfection of p.L48* or p.1M? mutant plasmids. In immunofluorescence experiments, the p.W168C mutant JAM2 protein failed to translocate to the plasma membrane. We speculated that mutant JAM2 protein resulted in impaired cell-cell adhesion functions and reduced integrity of the neurovascular unit. This is similar to the mechanisms of other causative genes for primary familial brain calcification or brain calcification syndromes (e.g. PDGFRB, PDGFB, MYORG, JAM3, and OCLN), all of which are highly expressed and functionally important in the neurovascular unit. Our study identifies a novel causative gene for primary familial brain calcification, whose vital function and high expression in the neurovascular unit further supports impairment of the neurovascular unit as the root of primary familial brain calcification pathogenesis.


Asunto(s)
Encefalopatías/genética , Encéfalo/metabolismo , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Células Endoteliales/metabolismo , Adulto , Encéfalo/patología , Encefalopatías/metabolismo , Calcinosis/genética , Femenino , Genes Recesivos/genética , Humanos , Masculino , Persona de Mediana Edad , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/metabolismo , Linaje , Fenotipo , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptor de Retrovirus Xenotrópico y Politrópico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...