Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 11.446
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-38733989

RESUMEN

The vasculature of the central nervous system is a 3D lattice composed of laminar vascular beds interconnected by penetrating vessels. The mechanisms controlling 3D lattice network formation remain largely unknown. Combining viral labeling, genetic marking, and single-cell profiling in the mouse retina, we discovered a perivascular neuronal subset, annotated as Fam19a4/Nts-positive retinal ganglion cells (Fam19a4/Nts-RGCs), directly contacting the vasculature with perisomatic endfeet. Developmental ablation of Fam19a4/Nts-RGCs led to disoriented growth of penetrating vessels near the ganglion cell layer (GCL), leading to a disorganized 3D vascular lattice. We identified enriched PIEZO2 expression in Fam19a4/Nts-RGCs. Piezo2 loss from all retinal neurons or Fam19a4/Nts-RGCs abolished the direct neurovascular contacts and phenocopied the Fam19a4/Nts-RGC ablation deficits. The defective vascular structure led to reduced capillary perfusion and sensitized the retina to ischemic insults. Furthermore, we uncovered a Piezo2-dependent perivascular granule cell subset for cerebellar vascular patterning, indicating neuronal Piezo2-dependent 3D vascular patterning in the brain.


Asunto(s)
Cerebelo , Neuronas , Retina , Animales , Femenino , Masculino , Ratones , Cerebelo/metabolismo , Cerebelo/irrigación sanguínea , Cerebelo/citología , Canales Iónicos/metabolismo , Ratones Endogámicos C57BL , Neuronas/metabolismo , Retina/citología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Vasos Retinianos/metabolismo
2.
Cell ; 177(3): 669-682.e24, 2019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30929904

RESUMEN

Throughout mammalian neocortex, layer 5 pyramidal (L5) cells project via the pons to a vast number of cerebellar granule cells (GrCs), forming a fundamental pathway. Yet, it is unknown how neuronal dynamics are transformed through the L5→GrC pathway. Here, by directly comparing premotor L5 and GrC activity during a forelimb movement task using dual-site two-photon Ca2+ imaging, we found that in expert mice, L5 and GrC dynamics were highly similar. L5 cells and GrCs shared a common set of task-encoding activity patterns, possessed similar diversity of responses, and exhibited high correlations comparable to local correlations among L5 cells. Chronic imaging revealed that these dynamics co-emerged in cortex and cerebellum over learning: as behavioral performance improved, initially dissimilar L5 cells and GrCs converged onto a shared, low-dimensional, task-encoding set of neural activity patterns. Thus, a key function of cortico-cerebellar communication is the propagation of shared dynamics that emerge during learning.


Asunto(s)
Cerebelo/metabolismo , Neocórtex/metabolismo , Animales , Conducta Animal , Calcio/metabolismo , Miembro Anterior/fisiología , Ratones , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Neocórtex/patología , Opsinas/genética , Opsinas/metabolismo , Células Piramidales/metabolismo
3.
Cell ; 167(5): 1385-1397.e11, 2016 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-27863250

RESUMEN

The association of histone modification changes with autism spectrum disorder (ASD) has not been systematically examined. We conducted a histone acetylome-wide association study (HAWAS) by performing H3K27ac chromatin immunoprecipitation sequencing (ChIP-seq) on 257 postmortem samples from ASD and matched control brains. Despite etiological heterogeneity, ≥68% of syndromic and idiopathic ASD cases shared a common acetylome signature at >5,000 cis-regulatory elements in prefrontal and temporal cortex. Similarly, multiple genes associated with rare genetic mutations in ASD showed common "epimutations." Acetylome aberrations in ASD were not attributable to genetic differentiation at cis-SNPs and highlighted genes involved in synaptic transmission, ion transport, epilepsy, behavioral abnormality, chemokinesis, histone deacetylation, and immunity. By correlating histone acetylation with genotype, we discovered >2,000 histone acetylation quantitative trait loci (haQTLs) in human brain regions, including four candidate causal variants for psychiatric diseases. Due to the relative stability of histone modifications postmortem, we anticipate that the HAWAS approach will be applicable to multiple diseases.


Asunto(s)
Trastorno del Espectro Autista/genética , Cerebelo/metabolismo , Código de Histonas , Corteza Prefrontal/metabolismo , Sitios de Carácter Cuantitativo , Lóbulo Temporal/metabolismo , Acetilación , Trastorno del Espectro Autista/metabolismo , Autopsia , Inmunoprecipitación de Cromatina , Elementos de Facilitación Genéticos , Humanos , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo
4.
Genes Dev ; 35(21-22): 1445-1460, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34711653

RESUMEN

Joubert syndrome (JS) is a recessive ciliopathy in which all affected individuals have congenital cerebellar vermis hypoplasia. Here, we report that CEP120, a JS-associated protein involved in centriole biogenesis and cilia assembly, regulates timely neuronal differentiation and the departure of granule neuron progenitors (GNPs) from their germinal zone during cerebellar development. Our results show that depletion of Cep120 perturbs GNP cell cycle progression, resulting in a delay of cell cycle exit in vivo. To dissect the potential mechanism, we investigated the association between CEP120 interactome and the JS database and identified KIAA0753 (a JS-associated protein) as a CEP120-interacting protein. Surprisingly, we found that CEP120 recruits KIAA0753 to centrioles, and that loss of this interaction induces accumulation of GNPs in the germinal zone and impairs neuronal differentiation. Importantly, the replenishment of wild-type CEP120 rescues the above defects, whereas expression of JS-associated CEP120 mutants, which hinder KIAA0753 recruitment, does not. Together, our data reveal a close interplay between CEP120 and KIAA0753 for the germinal zone exit and timely neuronal differentiation of GNPs during cerebellar development, and mutations in CEP120 and KIAA0753 may participate in the heterotopia and cerebellar hypoplasia observed in JS patients.


Asunto(s)
Centriolos , Enfermedades Renales Quísticas , Anomalías Múltiples , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Centriolos/genética , Centriolos/metabolismo , Cerebelo/anomalías , Cerebelo/metabolismo , Anomalías del Ojo , Humanos , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Proteínas Asociadas a Microtúbulos , Retina/anomalías
5.
Cell ; 152(5): 1065-76, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23452854

RESUMEN

Medulloblastoma is the most common pediatric malignant brain tumor. Although current therapies improve survival, these regimens are highly toxic and are associated with significant morbidity. Here, we report that placental growth factor (PlGF) is expressed in the majority of medulloblastomas, independent of their subtype. Moreover, high expression of PlGF receptor neuropilin 1 (Nrp1) correlates with poor overall survival in patients. We demonstrate that PlGF and Nrp1 are required for the growth and spread of medulloblastoma: PlGF/Nrp1 blockade results in direct antitumor effects in vivo, resulting in medulloblastoma regression, decreased metastasis, and increased mouse survival. We reveal that PlGF is produced in the cerebellar stroma via tumor-derived Sonic hedgehog (Shh) and show that PlGF acts through Nrp1-and not vascular endothelial growth factor receptor 1-to promote tumor cell survival. This critical tumor-stroma interaction-mediated by Shh, PlGF, and Nrp1 across medulloblastoma subtypes-supports the development of therapies targeting PlGF/Nrp1 pathway.


Asunto(s)
Neoplasias Cerebelosas/patología , Cerebelo/metabolismo , Meduloblastoma/patología , Neuropilina-1/metabolismo , Proteínas Gestacionales/metabolismo , Transducción de Señal , Animales , Células Cultivadas , Neoplasias Cerebelosas/metabolismo , Humanos , Meduloblastoma/metabolismo , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Comunicación Paracrina , Factor de Crecimiento Placentario , Trasplante Heterólogo , Receptor 1 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Am J Hum Genet ; 111(5): 913-926, 2024 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-38626762

RESUMEN

Expanded CAG repeats in coding regions of different genes are the most common cause of dominantly inherited spinocerebellar ataxias (SCAs). These repeats are unstable through the germline, and larger repeats lead to earlier onset. We measured somatic expansion in blood samples collected from 30 SCA1, 50 SCA2, 74 SCA3, and 30 SCA7 individuals over a mean interval of 8.5 years, along with postmortem tissues and fetal tissues from SCA1, SCA3, and SCA7 individuals to examine somatic expansion at different stages of life. We showed that somatic mosaicism in the blood increases over time. Expansion levels are significantly different among SCAs and correlate with CAG repeat lengths. The level of expansion is greater in individuals with SCA7 who manifest disease compared to that of those who do not yet display symptoms. Brain tissues from SCA individuals have larger expansions compared to the blood. The cerebellum has the lowest mosaicism among the studied brain regions, along with a high expression of ATXNs and DNA repair genes. This was the opposite in cortices, with the highest mosaicism and lower expression of ATXNs and DNA repair genes. Fetal cortices did not show repeat instability. This study shows that CAG repeats are increasingly unstable during life in the blood and the brain of SCA individuals, with gene- and tissue-specific patterns.


Asunto(s)
Mosaicismo , Ataxias Espinocerebelosas , Expansión de Repetición de Trinucleótido , Humanos , Ataxias Espinocerebelosas/genética , Expansión de Repetición de Trinucleótido/genética , Femenino , Masculino , Adulto , Persona de Mediana Edad , Cerebelo/metabolismo , Cerebelo/patología , Anciano , Encéfalo/metabolismo , Encéfalo/patología , Ataxina-1/genética
7.
Development ; 151(14)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-38912572

RESUMEN

The neurons of the three cerebellar nuclei (CN) are the primary output neurons of the cerebellum. The excitatory neurons (e) of the medial (m) CN (eCNm) were recently divided into molecularly defined subdomains in the adult; however, how they are established during development is not known. We define molecular subdomains of the mouse embryonic eCNm using single-cell RNA-sequencing and spatial expression analysis, showing that they evolve during embryogenesis to prefigure the adult. Furthermore, eCNm are transcriptionally divergent from cells in the other nuclei by embryonic day 14.5. We previously showed that loss of the homeobox genes En1 and En2 leads to loss of approximately half of the embryonic eCNm. We demonstrate that mutation of En1/2 in the embryonic eCNm results in death of specific posterior eCNm molecular subdomains and downregulation of TBR2 (EOMES) in an anterior embryonic subdomain, as well as reduced synaptic gene expression. We further reveal a similar function for EN1/2 in mediating TBR2 expression, neuron differentiation and survival in the other excitatory neurons (granule and unipolar brush cells). Thus, our work defines embryonic eCNm molecular diversity and reveals conserved roles for EN1/2 in the cerebellar excitatory neuron lineage.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Neuronas , Animales , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Ratones , Neuronas/metabolismo , Neuronas/citología , Supervivencia Celular/genética , Diferenciación Celular/genética , Cerebelo/embriología , Cerebelo/metabolismo , Cerebelo/citología , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/genética , Núcleos Cerebelosos/metabolismo , Núcleos Cerebelosos/embriología , Núcleos Cerebelosos/citología , Análisis de la Célula Individual , Proteínas del Tejido Nervioso
8.
Development ; 151(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38860486

RESUMEN

Cerebellar granule neuron progenitors (GNPs) originate from the upper rhombic lip (URL), a germinative niche in which developmental defects produce human diseases. T-cell factor (TCF) responsiveness and Notch dependence are hallmarks of self-renewal in neural stem cells. TCF activity, together with transcripts encoding proneural gene repressors hairy and enhancer of split (Hes/Hey), are detected in the URL; however, their functions and regulatory modes are undeciphered. Here, we established amphibian as a pertinent model for studying vertebrate URL development. The amphibian long-lived URL is TCF active, whereas the external granular layer (EGL) is non-proliferative and expresses hes4 and hes5 genes. Using functional and transcriptomic approaches, we show that TCF activity is necessary for URL emergence and maintenance. We establish that the transcription factor Barhl1 controls GNP exit from the URL, acting partly through direct TCF inhibition. Identification of Barhl1 target genes suggests that, besides TCF, Barhl1 inhibits transcription of hes5 genes independently of Notch signaling. Observations in amniotes suggest a conserved role for Barhl in maintenance of the URL and/or EGL via co-regulation of TCF, Hes and Hey genes.


Asunto(s)
Cerebelo , Células-Madre Neurales , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Cerebelo/citología , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Neuronas/citología , Receptores Notch/metabolismo , Receptores Notch/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Rombencéfalo/metabolismo , Rombencéfalo/citología , Nicho de Células Madre , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética
9.
Cell ; 148(1-2): 296-308, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22265417

RESUMEN

Although uridine-rich small nuclear RNAs (U-snRNAs) are essential for pre-mRNA splicing, little is known regarding their function in the regulation of alternative splicing or of the biological consequences of their dysfunction in mammals. Here, we demonstrate that mutation of Rnu2-8, one of the mouse multicopy U2 snRNA genes, causes ataxia and neurodegeneration. Coincident with the observed pathology, the level of mutant U2 RNAs was highest in the cerebellum and increased after granule neuron maturation. Furthermore, neuron loss was strongly dependent on the dosage of mutant and wild-type snRNA genes. Comprehensive transcriptome analysis identified a group of alternative splicing events, including the splicing of small introns, which were disrupted in the mutant cerebellum. Our results suggest that the expression of mammalian U2 snRNA genes, previously presumed to be ubiquitous, is spatially and temporally regulated, and dysfunction of a single U2 snRNA causes neuron degeneration through distortion of pre-mRNA splicing.


Asunto(s)
Empalme Alternativo , ARN Nuclear Pequeño/genética , Animales , Ataxia/genética , Secuencia de Bases , Cerebelo/citología , Cerebelo/metabolismo , Perfilación de la Expresión Génica , Ratones , Datos de Secuencia Molecular , Mutagénesis , Mutación , Enfermedades Neurodegenerativas/genética , Alineación de Secuencia
10.
Cell ; 151(7): 1417-30, 2012 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-23260135

RESUMEN

The high level of 5-hydroxymethylcytosine (5hmC) present in neuronal genomes suggests that mechanisms interpreting 5hmC in the CNS may differ from those present in embryonic stem cells. Here, we present quantitative, genome-wide analysis of 5hmC, 5-methylcytosine (5mC), and gene expression in differentiated CNS cell types in vivo. We report that 5hmC is enriched in active genes and that, surprisingly, strong depletion of 5mC is observed over these regions. The contribution of these epigenetic marks to gene expression depends critically on cell type. We identify methyl-CpG-binding protein 2 (MeCP2) as the major 5hmC-binding protein in the brain and demonstrate that MeCP2 binds 5hmC- and 5mC-containing DNA with similar high affinities. The Rett-syndrome-causing mutation R133C preferentially inhibits 5hmC binding. These findings support a model in which 5hmC and MeCP2 constitute a cell-specific epigenetic mechanism for regulation of chromatin structure and gene expression.


Asunto(s)
Cerebelo/metabolismo , Citosina/análogos & derivados , Epigénesis Genética , Proteína 2 de Unión a Metil-CpG/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Cerebelo/citología , Cromatina/metabolismo , Citosina/metabolismo , Humanos , Ratones , Ratones Noqueados , Neuroglía/metabolismo , Neuronas/metabolismo , Células de Purkinje/metabolismo , Síndrome de Rett/metabolismo
11.
Proc Natl Acad Sci U S A ; 121(34): e2405901121, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39150780

RESUMEN

Astrotactin 2 (ASTN2) is a transmembrane neuronal protein highly expressed in the cerebellum that functions in receptor trafficking and modulates cerebellar Purkinje cell (PC) synaptic activity. Individuals with ASTN2 mutations exhibit neurodevelopmental disorders, including autism spectrum disorder (ASD), attention-deficit/hyperactivity disorder (ADHD), learning difficulties, and language delay. To provide a genetic model for the role of the cerebellum in ASD-related behaviors and study the role of ASTN2 in cerebellar circuit function, we generated global and PC-specific conditional Astn2 knockout (KO and cKO, respectively) mouse lines. Astn2 KO mice exhibit strong ASD-related behavioral phenotypes, including a marked decrease in separation-induced pup ultrasonic vocalization calls, hyperactivity, repetitive behaviors, altered behavior in the three-chamber test, and impaired cerebellar-dependent eyeblink conditioning. Hyperactivity and repetitive behaviors are also prominent in Astn2 cKO animals, but they do not show altered behavior in the three-chamber test. By Golgi staining, Astn2 KO PCs have region-specific changes in dendritic spine density and filopodia numbers. Proteomic analysis of Astn2 KO cerebellum reveals a marked upregulation of ASTN2 family member, ASTN1, a neuron-glial adhesion protein. Immunohistochemistry and electron microscopy demonstrate a significant increase in Bergmann glia volume in the molecular layer of Astn2 KO animals. Electrophysiological experiments indicate a reduced frequency of spontaneous excitatory postsynaptic currents (EPSCs), as well as increased amplitudes of both spontaneous EPSCs and inhibitory postsynaptic currents in the Astn2 KO animals, suggesting that pre- and postsynaptic components of synaptic transmission are altered. Thus, ASTN2 regulates ASD-like behaviors and cerebellar circuit properties.


Asunto(s)
Trastorno del Espectro Autista , Cerebelo , Ratones Noqueados , Células de Purkinje , Animales , Ratones , Trastorno del Espectro Autista/metabolismo , Trastorno del Espectro Autista/genética , Trastorno del Espectro Autista/fisiopatología , Células de Purkinje/metabolismo , Cerebelo/metabolismo , Conducta Animal/fisiología , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética , Modelos Animales de Enfermedad , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Masculino
12.
Proc Natl Acad Sci U S A ; 121(31): e2323050121, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39042684

RESUMEN

Cerebellar injury in preterm infants with central nervous system (CNS) hemorrhage results in lasting neurological deficits and an increased risk of autism. The impact of blood-induced pathways on cerebellar development remains largely unknown, so no specific treatments have been developed to counteract the harmful effects of blood after neurovascular damage in preterm infants. Here, we show that fibrinogen, a blood-clotting protein, plays a central role in impairing neonatal cerebellar development. Longitudinal MRI of preterm infants revealed that cerebellar bleeds were the most critical factor associated with poor cerebellar growth. Using inflammatory and hemorrhagic mouse models of neonatal cerebellar injury, we found that fibrinogen increased innate immune activation and impeded neurogenesis in the developing cerebellum. Fibrinogen inhibited sonic hedgehog (SHH) signaling, the main mitogenic pathway in cerebellar granule neuron progenitors (CGNPs), and was sufficient to disrupt cerebellar growth. Genetic fibrinogen depletion attenuated neuroinflammation, promoted CGNP proliferation, and preserved normal cerebellar development after neurovascular damage. Our findings suggest that fibrinogen alters the balance of SHH signaling in the neurovascular niche and may serve as a therapeutic target to mitigate developmental brain injury after CNS hemorrhage.


Asunto(s)
Barrera Hematoencefálica , Cerebelo , Fibrinógeno , Proteínas Hedgehog , Transducción de Señal , Proteínas Hedgehog/metabolismo , Animales , Fibrinógeno/metabolismo , Cerebelo/metabolismo , Ratones , Barrera Hematoencefálica/metabolismo , Humanos , Animales Recién Nacidos , Recién Nacido , Neurogénesis , Femenino , Masculino , Modelos Animales de Enfermedad
13.
Hum Mol Genet ; 33(4): 299-317, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-37862125

RESUMEN

Spinocerebellar ataxia type 3 (SCA3) is an autosomal dominant hereditary disorder, caused by an expansion of polyglutamine in the ataxin-3 protein. SCA3 symptoms include progressive motor decline caused by an atrophy of the cerebellum and brainstem. However, it was recently reported that SCA3 patients also suffer from the cerebellar cognitive affective syndrome. The majority of SCA3 patients exhibit cognitive decline and approximately half of them suffer from depression and anxiety. The necessity to find a combined therapy for both motor and cognitive deficits in a SCA3 mouse model is required for the development of SCA3 treatment. Here, we demonstrated that the SCA3-84Q transgenic mice exhibited anxiety over the novel brightly illuminated environment in the open field, novelty suppressed feeding, and light-dark place preference tests. Moreover, SCA3-84Q mice also suffered from a decline in recognition memory during the novel object recognition test. SCA3-84Q mice also demonstrated floating behavior during the Morris water maze that can be interpreted as a sign of low mood and aversion to activity, i.e. depressive-like state. SCA3-84Q mice also spent more time immobile during the forced swimming and tail suspension tests which is also evidence for depressive-like behavior. Therefore, the SCA3-84Q mouse model may be used as a model system to test the possible treatments for both ataxia and non-motor symptoms including depression, anxiety, and memory loss.


Asunto(s)
Enfermedad de Machado-Joseph , Humanos , Ratones , Animales , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/metabolismo , Depresión/genética , Cerebelo/metabolismo , Ataxina-3/genética , Ataxina-3/metabolismo , Ratones Transgénicos , Ansiedad/genética
14.
Hum Mol Genet ; 33(16): 1406-1419, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38727562

RESUMEN

Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease, is reported to be the most common type of autosomal dominant cerebellar ataxia (ADCA). SCA3 patients suffer from a progressive decline in motor coordination and other disease-associated symptoms. Moreover, recent studies have reported that SCA3 patients also exhibit symptoms of cerebellar cognitive affective syndrome (CCAS). We previously observed signs of CCAS in mouse model of SCA3. Particularly, SCA3-84Q mice suffer from anxiety, recognition memory decline, and also exhibit signs of low mood and aversion to activity. Here we studied the effect of long-term injections of SK channels activator chlorzoxazone (CHZ) together and separately with the folic acid (FA) on the cerebellar Purkinje cell (PC) firing and histology, and also on the motor and cognitive functions as well as mood alterations in SCA3-84Q hemizygous transgenic mice. We realized that both CHZ and CHZ-FA combination had similar positive effect on pure cerebellum impairments including PC firing precision, PC histology, and motor performance in SCA3-84Q mice. However, only the CHZ-FA combination, but not CHZ, had significantly ameliorated the signs of anxiety and depression, and also noticeably improved recognition memory in SCA3-84Q mice. Our results suggest that the combination therapy for both ataxia and non-motor symptoms is required for the complex treatment of ADCA.


Asunto(s)
Ansiedad , Clorzoxazona , Depresión , Modelos Animales de Enfermedad , Ácido Fólico , Enfermedad de Machado-Joseph , Ratones Transgénicos , Animales , Ratones , Ansiedad/tratamiento farmacológico , Ansiedad/fisiopatología , Depresión/tratamiento farmacológico , Depresión/genética , Depresión/fisiopatología , Ácido Fólico/farmacología , Ácido Fólico/administración & dosificación , Enfermedad de Machado-Joseph/tratamiento farmacológico , Enfermedad de Machado-Joseph/genética , Enfermedad de Machado-Joseph/fisiopatología , Enfermedad de Machado-Joseph/patología , Clorzoxazona/farmacología , Células de Purkinje/efectos de los fármacos , Células de Purkinje/metabolismo , Células de Purkinje/patología , Memoria/efectos de los fármacos , Humanos , Cerebelo/efectos de los fármacos , Cerebelo/metabolismo , Masculino , Ataxina-3/genética , Ataxina-3/metabolismo
15.
Hum Mol Genet ; 33(16): 1442-1453, 2024 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-38751342

RESUMEN

Primary cilia are antenna-like structures protruding from the surface of various eukaryotic cells, and have distinct protein compositions in their membranes. This distinct protein composition is maintained by the presence of the transition zone (TZ) at the ciliary base, which acts as a diffusion barrier between the ciliary and plasma membranes. Defects in cilia and the TZ are known to cause a group of disorders collectively called the ciliopathies, which demonstrate a broad spectrum of clinical features, such as perinatally lethal Meckel syndrome (MKS), relatively mild Joubert syndrome (JBTS), and nonsyndromic nephronophthisis (NPHP). Proteins constituting the TZ can be grouped into the MKS and NPHP modules. The MKS module is composed of several transmembrane proteins and three soluble proteins. TMEM218 was recently reported to be mutated in individuals diagnosed as MKS and JBTS. However, little is known about how TMEM218 mutations found in MKS and JBTS affect the functions of cilia. In this study, we found that ciliary membrane proteins were not localized to cilia in TMEM218-knockout cells, indicating impaired barrier function of the TZ. Furthermore, the exogenous expression of JBTS-associated TMEM218 variants but not MKS-associated variants in TMEM218-knockout cells restored the localization of ciliary membrane proteins. In particular, when expressed in TMEM218-knockout cells, the TMEM218(R115H) variant found in JBTS was able to restore the barrier function of cells, whereas the MKS variant TMEM218(R115C) could not. Thus, the severity of symptoms of MKS and JBTS individuals appears to correlate with the degree of their ciliary defects at the cellular level.


Asunto(s)
Anomalías Múltiples , Cilios , Ciliopatías , Encefalocele , Anomalías del Ojo , Enfermedades Renales Quísticas , Proteínas de la Membrana , Mutación , Retina , Cilios/metabolismo , Cilios/genética , Cilios/patología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ciliopatías/genética , Ciliopatías/metabolismo , Ciliopatías/patología , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/patología , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/patología , Anomalías Múltiples/genética , Anomalías Múltiples/metabolismo , Anomalías Múltiples/patología , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Anomalías del Ojo/metabolismo , Retina/metabolismo , Retina/anomalías , Retina/patología , Cerebelo/anomalías , Cerebelo/metabolismo , Cerebelo/patología , Enfermedades Cerebelosas/genética , Enfermedades Cerebelosas/metabolismo , Enfermedades Cerebelosas/patología , Animales , Membrana Celular/metabolismo , Ratones , Trastornos de la Motilidad Ciliar , Enfermedades Renales Poliquísticas , Retinitis Pigmentosa
16.
Annu Rev Neurosci ; 41: 119-137, 2018 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-29618285

RESUMEN

The noncoding portion of the genome, including microRNAs, has been fertile evolutionary soil for cortical development in primates. A major contribution to cortical expansion in primates is the generation of novel precursor cell populations. Because miRNA expression profiles track closely with cell identity, it is likely that numerous novel microRNAs have contributed to cellular diversity in the brain. The tools to determine the genomic context within which novel microRNAs emerge and how they become integrated into molecular circuitry are now in hand.


Asunto(s)
Evolución Biológica , Cerebelo/metabolismo , Corteza Cerebral/metabolismo , MicroARNs/metabolismo , Vías Nerviosas/metabolismo , Animales , Cerebelo/crecimiento & desarrollo , Corteza Cerebral/crecimiento & desarrollo , Humanos , MicroARNs/genética , Vías Nerviosas/crecimiento & desarrollo , Neurogénesis/genética
17.
J Cell Sci ; 137(13)2024 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-38841887

RESUMEN

Centrosomal proteins play pivotal roles in orchestrating microtubule dynamics, and their dysregulation leads to disorders, including cancer and ciliopathies. Understanding the multifaceted roles of centrosomal proteins is vital to comprehend their involvement in disease development. Here, we report novel cellular functions of CEP41, a centrosomal and ciliary protein implicated in Joubert syndrome. We show that CEP41 is an essential microtubule-associated protein with microtubule-stabilizing activity. Purified CEP41 binds to preformed microtubules, promotes microtubule nucleation and suppresses microtubule disassembly. When overexpressed in cultured cells, CEP41 localizes to microtubules and promotes microtubule bundling. Conversely, shRNA-mediated knockdown of CEP41 disrupts the interphase microtubule network and delays microtubule reassembly, emphasizing its role in microtubule organization. Further, we demonstrate that the association of CEP41 with microtubules relies on its conserved rhodanese homology domain (RHOD) and the N-terminal region. Interestingly, a disease-causing mutation in the RHOD domain impairs CEP41-microtubule interaction. Moreover, depletion of CEP41 inhibits cell proliferation and disrupts cell cycle progression, suggesting its potential involvement in cell cycle regulation. These insights into the cellular functions of CEP41 hold promise for unraveling the impact of its mutations in ciliopathies.


Asunto(s)
Proliferación Celular , Microtúbulos , Humanos , Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Asociadas a Microtúbulos/genética , Centrosoma/metabolismo , Retina/metabolismo , Retina/patología , Retina/anomalías , Ciliopatías/metabolismo , Ciliopatías/genética , Ciliopatías/patología , Cerebelo/metabolismo , Cerebelo/anomalías , Cerebelo/patología , Enfermedades Renales Quísticas/metabolismo , Enfermedades Renales Quísticas/genética , Enfermedades Renales Quísticas/patología , Cilios/metabolismo , Cilios/patología , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Animales , Anomalías Múltiples/metabolismo , Anomalías Múltiples/genética , Anomalías Múltiples/patología , Anomalías del Ojo/metabolismo , Anomalías del Ojo/genética , Anomalías del Ojo/patología , Unión Proteica , Ciclo Celular/genética , Células HEK293
18.
Cell ; 147(7): 1601-14, 2011 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-22196734

RESUMEN

The assembly of synapses and neuronal circuits relies on an array of molecular recognition events and their modification by neuronal activity. Neurexins are a highly polymorphic family of synaptic receptors diversified by extensive alternative splicing. Neurexin variants exhibit distinct isoform-specific biochemical interactions and synapse assembly functions, but the mechanisms governing splice isoform choice are not understood. We demonstrate that Nrxn1 alternative splicing is temporally and spatially controlled in the mouse brain. Neuronal activity triggers a shift in Nrxn1 splice isoform choice via calcium/calmodulin-dependent kinase IV signaling. Activity-dependent alternative splicing of Nrxn1 requires the KH-domain RNA-binding protein SAM68 that associates with RNA response elements in the Nrxn1 pre-mRNA. Our findings uncover SAM68 as a key regulator of dynamic control of Nrxn1 molecular diversity and activity-dependent alternative splicing in the central nervous system.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Empalme Alternativo , Cerebelo/metabolismo , Moléculas de Adhesión de Célula Nerviosa/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Secuencia de Bases , Proteínas de Unión al Calcio , Cerebelo/citología , Humanos , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Neuronas/metabolismo , Proteínas de Unión al ARN/genética , Elementos de Respuesta
19.
J Neurosci ; 44(15)2024 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-38383496

RESUMEN

Almost all medulloblastomas (MB) of the Wingless/Int-1 (WNT) type are characterized by hotspot mutations in CTNNB1, and mouse models have convincingly demonstrated the tumor-initiating role of these mutations. Additional alterations in SMARCA4 are detected in ∼20% of WNT MB, but their functional role is mostly unknown. We, therefore, amended previously described brain lipid binding protein (Blbp)-cre::Ctnnb1(ex3)fl/wt mice by the introduction of floxed Smarca4 alleles. Unexpectedly, mutated and thereby stabilized ß-catenin on its own induced severe developmental phenotypes in male and female Blbp-cre::Ctnnb1(ex3)fl/wt mice in our hands, including a thinned cerebral cortex, hydrocephalus, missing cerebellar layering, and cell accumulations in the brainstem and cerebellum. An additional loss of SMARCA4 even resulted in prenatal death for most mice. Respective Blbp-cre::Ctnnb1(ex3)fl/wt::Smarca4fl/rec mutants (male and female) developed large proliferative lesions in the cerebellum evolving from E13.5 to E16.5. Histological and molecular analysis of these lesions by DNA methylation profiling and single-cell RNA sequencing suggested an origin in early undifferentiated SOX2-positive cerebellar progenitors. Furthermore, upregulated WNT signaling, altered actin/cytoskeleton organization, and reduced neuronal differentiation were evident in mutant cells. In vitro, cells harboring alterations in both Ctnnb1 and Smarca4 were negatively selected and did not show tumorigenic potential after transplantation in adult female recipient mice. However, in cerebellar explant cultures, mutant cells displayed significantly increased proliferation, suggesting an important role of the embryonic microenvironment in the development of lesions. Altogether, these results represent an important first step toward the unraveling of tumorigenic mechanisms induced by aberrant WNT signaling and SMARCA4 deficiency.


Asunto(s)
Neoplasias Cerebelosas , Meduloblastoma , Animales , Femenino , Masculino , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Cerebelo/metabolismo , Mutación/genética , Transducción de Señal , Microambiente Tumoral
20.
J Neurosci ; 44(11)2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38331582

RESUMEN

Cerebellum has been implicated in drug addiction; however, its underlying cellular populations and neuronal circuitry remain largely unknown. In the current study, we identified a neural pathway from tyrosine hydroxylase (TH)-positive Purkinje cells (PCTH+) in cerebellar lobule VI to calcium/calmodulin-dependent protein kinase II (CaMKII)-positive glutamatergic neurons in the medial cerebellar nucleus (MedCaMKII), forming the lobule VI PCTH+-MedCaMKII pathway in male mice. In naive male mice, inhibition of PCTH+ neurons activated Med neurons. During conditioned place preference (CPP) training, exposure to methamphetamine (METH) inhibited lobule VI PCTH+ neurons while excited MedCaMKII neurons in mice. Silencing MedCaMKII using a tetanus toxin light chain (tettox) suppressed the acquisition of METH CPP in mice but resulted in motor coordination deficits in naive mice. In contrast, activating lobule VI PCTH+ terminals within Med inhibited the activity of Med neurons and subsequently blocked the acquisition of METH CPP in mice without affecting motor coordination, locomotor activity, and sucrose reinforcements in naive mice. Our findings identified a novel lobule VI PCTH+-MedCaMKII pathway within the cerebellum and explored its role in mediating the acquisition of METH-preferred behaviors.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Metanfetamina , Animales , Masculino , Ratones , Metanfetamina/farmacología , Tirosina 3-Monooxigenasa/metabolismo , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Refuerzo en Psicología , Cerebelo/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA