Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46.390
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 187(11): 2767-2784.e23, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38733989

RESUMO

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.


Assuntos
Cerebelo , Neurônios , Retina , Animais , Feminino , Masculino , Camundongos , Cerebelo/metabolismo , Cerebelo/irrigação sanguínea , Cerebelo/citologia , Canais Iônicos/metabolismo , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Retina/citologia , Retina/metabolismo , Células Ganglionares da Retina/metabolismo , Vasos Retinianos/metabolismo
2.
Cell ; 184(14): 3731-3747.e21, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34214470

RESUMO

In motor neuroscience, state changes are hypothesized to time-lock neural assemblies coordinating complex movements, but evidence for this remains slender. We tested whether a discrete change from more autonomous to coherent spiking underlies skilled movement by imaging cerebellar Purkinje neuron complex spikes in mice making targeted forelimb-reaches. As mice learned the task, millimeter-scale spatiotemporally coherent spiking emerged ipsilateral to the reaching forelimb, and consistent neural synchronization became predictive of kinematic stereotypy. Before reach onset, spiking switched from more disordered to internally time-locked concerted spiking and silence. Optogenetic manipulations of cerebellar feedback to the inferior olive bi-directionally modulated neural synchronization and reaching direction. A simple model explained the reorganization of spiking during reaching as reflecting a discrete bifurcation in olivary network dynamics. These findings argue that to prepare learned movements, olivo-cerebellar circuits enter a self-regulated, synchronized state promoting motor coordination. State changes facilitating behavioral transitions may generalize across neural systems.


Assuntos
Movimento/fisiologia , Rede Nervosa/fisiologia , Potenciais de Ação/fisiologia , Animais , Cálcio/metabolismo , Cerebelo/fisiologia , Sincronização Cortical , Membro Anterior/fisiologia , Interneurônios/fisiologia , Aprendizagem , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Neurológicos , Atividade Motora/fisiologia , Núcleo Olivar/fisiologia , Optogenética , Células de Purkinje/fisiologia , Comportamento Estereotipado , Análise e Desempenho de Tarefas
3.
Cell ; 180(3): 536-551.e17, 2020 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-31955849

RESUMO

Goal-directed behavior requires the interaction of multiple brain regions. How these regions and their interactions with brain-wide activity drive action selection is less understood. We have investigated this question by combining whole-brain volumetric calcium imaging using light-field microscopy and an operant-conditioning task in larval zebrafish. We find global, recurring dynamics of brain states to exhibit pre-motor bifurcations toward mutually exclusive decision outcomes. These dynamics arise from a distributed network displaying trial-by-trial functional connectivity changes, especially between cerebellum and habenula, which correlate with decision outcome. Within this network the cerebellum shows particularly strong and predictive pre-motor activity (>10 s before movement initiation), mainly within the granule cells. Turn directions are determined by the difference neuroactivity between the ipsilateral and contralateral hemispheres, while the rate of bi-hemispheric population ramping quantitatively predicts decision time on the trial-by-trial level. Our results highlight a cognitive role of the cerebellum and its importance in motor planning.


Assuntos
Cerebelo/fisiologia , Tomada de Decisões/fisiologia , Tempo de Reação/fisiologia , Peixe-Zebra/fisiologia , Animais , Comportamento Animal/fisiologia , Mapeamento Encefálico/métodos , Cérebro/fisiologia , Cognição/fisiologia , Condicionamento Operante/fisiologia , Objetivos , Habenula/fisiologia , Temperatura Alta , Larva/fisiologia , Atividade Motora/fisiologia , Movimento , Neurônios/fisiologia , Desempenho Psicomotor/fisiologia , Rombencéfalo/fisiologia
4.
Cell ; 177(3): 669-682.e24, 2019 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-30929904

RESUMO

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.


Assuntos
Cerebelo/metabolismo , Neocórtex/metabolismo , Animais , Comportamento Animal , Cálcio/metabolismo , Membro Anterior/fisiologia , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência por Excitação Multifotônica , Neocórtex/patologia , Opsinas/genética , Opsinas/metabolismo , Células Piramidais/metabolismo
5.
Annu Rev Neurosci ; 47(1): 145-166, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-38663092

RESUMO

The cerebellum has a well-established role in controlling motor functions, including coordination, posture, and the learning of skilled movements. The mechanisms for how it carries out motor behavior remain under intense investigation. Interestingly though, in recent years the mechanisms of cerebellar function have faced additional scrutiny since nonmotor behaviors may also be controlled by the cerebellum. With such complexity arising, there is now a pressing need to better understand how cerebellar structure, function, and behavior intersect to influence behaviors that are dynamically called upon as an animal experiences its environment. Here, we discuss recent experimental work that frames possible neural mechanisms for how the cerebellum shapes disparate behaviors and why its dysfunction is catastrophic in hereditary and acquired conditions-both motor and nonmotor. For these reasons, the cerebellum might be the ideal therapeutic target.


Assuntos
Cerebelo , Aprendizagem , Movimento , Cerebelo/fisiologia , Animais , Humanos , Aprendizagem/fisiologia , Movimento/fisiologia
6.
Cell ; 167(5): 1385-1397.e11, 2016 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-27863250

RESUMO

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.


Assuntos
Transtorno do Espectro Autista/genética , Cerebelo/metabolismo , Código das Histonas , Córtex Pré-Frontal/metabolismo , Locos de Características Quantitativas , Lobo Temporal/metabolismo , Acetilação , Transtorno do Espectro Autista/metabolismo , Autopsia , Imunoprecipitação da Cromatina , Elementos Facilitadores Genéticos , Humanos , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
7.
Annu Rev Neurosci ; 45: 151-175, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35803588

RESUMO

The cerebellar cortex is an important system for relating neural circuits and learning. Its promise reflects the longstanding idea that it contains simple, repeated circuit modules with only a few cell types and a single plasticity mechanism that mediates learning according to classical Marr-Albus models. However, emerging data have revealed surprising diversity in neuron types, synaptic connections, and plasticity mechanisms, both locally and regionally within the cerebellar cortex. In light of these findings, it is not surprising that attempts to generate a holistic model of cerebellar learning across different behaviors have not been successful. While the cerebellum remains an ideal system for linking neuronal function with behavior, it is necessary to update the cerebellar circuit framework to achieve its great promise. In this review, we highlight recent advances in our understanding of cerebellar-cortical cell types, synaptic connections, signaling mechanisms, and forms of plasticity that enrich cerebellar processing.


Assuntos
Plasticidade Neuronal , Células de Purkinje , Córtex Cerebelar/fisiologia , Cerebelo , Aprendizagem/fisiologia , Plasticidade Neuronal/fisiologia , Células de Purkinje/fisiologia
8.
Annu Rev Neurosci ; 45: 515-531, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35440142

RESUMO

Developmental abnormalities of the cerebellum are among the most recognized structural brain malformations in human prenatal imaging. Yet reliable information regarding their cause in humans is sparse, and few outcome studies are available to inform prognosis. We know very little about human cerebellar development, in stark contrast to the wealth of knowledge from decades of research on cerebellar developmental biology of model organisms, especially mice. Recent studies show that multiple aspects of human cerebellar development significantly differ from mice and even rhesus macaques, a nonhuman primate. These discoveries challenge many current mouse-centric models of normal human cerebellar development and models regarding the pathogenesis of several neurodevelopmental phenotypes affecting the cerebellum, including Dandy-Walker malformation and medulloblastoma. Since we cannot model what we do not know, additional normative and pathological human developmental data are essential, and new models are needed.


Assuntos
Neoplasias Cerebelares , Transtornos do Neurodesenvolvimento , Animais , Cerebelo , Feminino , Humanos , Macaca mulatta , Camundongos , Transtornos do Neurodesenvolvimento/genética , Transtornos do Neurodesenvolvimento/patologia , Gravidez , Transcriptoma
9.
Nature ; 625(7996): 788-796, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38029793

RESUMO

The expansion of the neocortex, a hallmark of mammalian evolution1,2, was accompanied by an increase in cerebellar neuron numbers3. However, little is known about the evolution of the cellular programmes underlying the development of the cerebellum in mammals. In this study we generated single-nucleus RNA-sequencing data for around 400,000 cells to trace the development of the cerebellum from early neurogenesis to adulthood in human, mouse and the marsupial opossum. We established a consensus classification of the cellular diversity in the developing mammalian cerebellum and validated it by spatial mapping in the fetal human cerebellum. Our cross-species analyses revealed largely conserved developmental dynamics of cell-type generation, except for Purkinje cells, for which we observed an expansion of early-born subtypes in the human lineage. Global transcriptome profiles, conserved cell-state markers and gene-expression trajectories across neuronal differentiation show that cerebellar cell-type-defining programmes have been overall preserved for at least 160 million years. However, we also identified many orthologous genes that gained or lost expression in cerebellar neural cell types in one of the species or evolved new expression trajectories during neuronal differentiation, indicating widespread gene repurposing at the cell-type level. In sum, our study unveils shared and lineage-specific gene-expression programmes governing the development of cerebellar cells and expands our understanding of mammalian brain evolution.


Assuntos
Cerebelo , Evolução Molecular , Mamíferos , Neurogênese , Animais , Humanos , Camundongos , Linhagem da Célula/genética , Cerebelo/citologia , Cerebelo/embriologia , Cerebelo/crescimento & desenvolvimento , Feto/citologia , Feto/embriologia , Regulação da Expressão Gênica no Desenvolvimento , Neurogênese/genética , Neurônios/citologia , Neurônios/metabolismo , Gambás/embriologia , Gambás/crescimento & desenvolvimento , Células de Purkinje/citologia , Células de Purkinje/metabolismo , Análise da Expressão Gênica de Célula Única , Especificidade da Espécie , Transcriptoma , Mamíferos/embriologia , Mamíferos/crescimento & desenvolvimento
10.
Nature ; 632(8027): 1092-1100, 2024 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-39048016

RESUMO

Placebo effects are notable demonstrations of mind-body interactions1,2. During pain perception, in the absence of any treatment, an expectation of pain relief can reduce the experience of pain-a phenomenon known as placebo analgesia3-6. However, despite the strength of placebo effects and their impact on everyday human experience and the failure of clinical trials for new therapeutics7, the neural circuit basis of placebo effects has remained unclear. Here we show that analgesia from the expectation of pain relief is mediated by rostral anterior cingulate cortex (rACC) neurons that project to the pontine nucleus (rACC→Pn)-a precerebellar nucleus with no established function in pain. We created a behavioural assay that generates placebo-like anticipatory pain relief in mice. In vivo calcium imaging of neural activity and electrophysiological recordings in brain slices showed that expectations of pain relief boost the activity of rACC→Pn neurons and potentiate neurotransmission in this pathway. Transcriptomic studies of Pn neurons revealed an abundance of opioid receptors, further suggesting a role in pain modulation. Inhibition of the rACC→Pn pathway disrupted placebo analgesia and decreased pain thresholds, whereas activation elicited analgesia in the absence of placebo conditioning. Finally, Purkinje cells exhibited activity patterns resembling those of rACC→Pn neurons during pain-relief expectation, providing cellular-level evidence for a role of the cerebellum in cognitive pain modulation. These findings open the possibility of targeting this prefrontal cortico-ponto-cerebellar pathway with drugs or neurostimulation to treat pain.


Assuntos
Vias Neurais , Percepção da Dor , Dor , Efeito Placebo , Animais , Feminino , Masculino , Camundongos , Analgesia , Antecipação Psicológica/fisiologia , Sinalização do Cálcio , Cerebelo/citologia , Cerebelo/fisiologia , Cognição/fisiologia , Eletrofisiologia , Perfilação da Expressão Gênica , Giro do Cíngulo/citologia , Giro do Cíngulo/fisiologia , Camundongos Endogâmicos C57BL , Neurônios/fisiologia , Dor/fisiopatologia , Dor/prevenção & controle , Dor/psicologia , Manejo da Dor/métodos , Manejo da Dor/psicologia , Manejo da Dor/tendências , Percepção da Dor/fisiologia , Limiar da Dor/fisiologia , Limiar da Dor/psicologia , Ponte/citologia , Ponte/fisiologia , Córtex Pré-Frontal/citologia , Córtex Pré-Frontal/fisiologia , Células de Purkinje/fisiologia , Receptores Opioides/metabolismo , Transmissão Sináptica
11.
Annu Rev Neurosci ; 44: 475-493, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34236892

RESUMO

Social interactions involve processes ranging from face recognition to understanding others' intentions. To guide appropriate behavior in a given context, social interactions rely on accurately predicting the outcomes of one's actions and the thoughts of others. Because social interactions are inherently dynamic, these predictions must be continuously adapted. The neural correlates of social processing have largely focused on emotion, mentalizing, and reward networks, without integration of systems involved in prediction. The cerebellum forms predictive models to calibrate movements and adapt them to changing situations, and cerebellar predictive modeling is thought to extend to nonmotor behaviors. Primary cerebellar dysfunction can produce social deficits, and atypical cerebellar structure and function are reported in autism, which is characterized by social communication challenges and atypical predictive processing. We examine the evidence that cerebellar-mediated predictions and adaptation play important roles in social processes and argue that disruptions in these processes contribute to autism.


Assuntos
Doenças Cerebelares , Cerebelo , Emoções , Humanos , Comportamento Social , Meio Social
12.
Cell ; 157(3): 651-63, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24766810

RESUMO

Neurodegenerative diseases can occur so early as to affect neurodevelopment. From a cohort of more than 2,000 consanguineous families with childhood neurological disease, we identified a founder mutation in four independent pedigrees in cleavage and polyadenylation factor I subunit 1 (CLP1). CLP1 is a multifunctional kinase implicated in tRNA, mRNA, and siRNA maturation. Kinase activity of the CLP1 mutant protein was defective, and the tRNA endonuclease complex (TSEN) was destabilized, resulting in impaired pre-tRNA cleavage. Germline clp1 null zebrafish showed cerebellar neurodegeneration that was rescued by wild-type, but not mutant, human CLP1 expression. Patient-derived induced neurons displayed both depletion of mature tRNAs and accumulation of unspliced pre-tRNAs. Transfection of partially processed tRNA fragments into patient cells exacerbated an oxidative stress-induced reduction in cell survival. Our data link tRNA maturation to neuronal development and neurodegeneration through defective CLP1 function in humans.


Assuntos
Cerebelo/crescimento & desenvolvimento , Cerebelo/patologia , Fator de Especificidade de Clivagem e Poliadenilação/metabolismo , Proteínas Nucleares/genética , Fosfotransferases/genética , Splicing de RNA , RNA de Transferência/genética , Fatores de Transcrição/genética , Proteínas de Peixe-Zebra/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/patologia , Fator de Especificidade de Clivagem e Poliadenilação/genética , Feminino , Humanos , Masculino , Camundongos , Modelos Moleculares , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Proteínas Nucleares/metabolismo , Linhagem , Fosfotransferases/metabolismo , RNA de Transferência/metabolismo , Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/metabolismo , Peixe-Zebra , Proteínas de Peixe-Zebra/genética
13.
Nature ; 624(7991): 403-414, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38092914

RESUMO

The brain controls nearly all bodily functions via spinal projecting neurons (SPNs) that carry command signals from the brain to the spinal cord. However, a comprehensive molecular characterization of brain-wide SPNs is still lacking. Here we transcriptionally profiled a total of 65,002 SPNs, identified 76 region-specific SPN types, and mapped these types into a companion atlas of the whole mouse brain1. This taxonomy reveals a three-component organization of SPNs: (1) molecularly homogeneous excitatory SPNs from the cortex, red nucleus and cerebellum with somatotopic spinal terminations suitable for point-to-point communication; (2) heterogeneous populations in the reticular formation with broad spinal termination patterns, suitable for relaying commands related to the activities of the entire spinal cord; and (3) modulatory neurons expressing slow-acting neurotransmitters and/or neuropeptides in the hypothalamus, midbrain and reticular formation for 'gain setting' of brain-spinal signals. In addition, this atlas revealed a LIM homeobox transcription factor code that parcellates the reticulospinal neurons into five molecularly distinct and spatially segregated populations. Finally, we found transcriptional signatures of a subset of SPNs with large soma size and correlated these with fast-firing electrophysiological properties. Together, this study establishes a comprehensive taxonomy of brain-wide SPNs and provides insight into the functional organization of SPNs in mediating brain control of bodily functions.


Assuntos
Encéfalo , Perfilação da Expressão Gênica , Vias Neurais , Neurônios , Medula Espinal , Animais , Camundongos , Hipotálamo , Neurônios/metabolismo , Neuropeptídeos , Medula Espinal/citologia , Medula Espinal/metabolismo , Encéfalo/citologia , Encéfalo/metabolismo , Neurotransmissores , Mesencéfalo/citologia , Formação Reticular/citologia , Eletrofisiologia , Cerebelo/citologia , Córtex Cerebral/citologia
14.
Genes Dev ; 35(21-22): 1445-1460, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34711653

RESUMO

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.


Assuntos
Centríolos , Doenças Renais Císticas , Anormalidades Múltiplas , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Centríolos/genética , Centríolos/metabolismo , Cerebelo/anormalidades , Cerebelo/metabolismo , Anormalidades do Olho , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Proteínas Associadas aos Microtúbulos , Retina/anormalidades
15.
Cell ; 154(1): 118-33, 2013 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-23827678

RESUMO

The CACNA1A gene, encoding the voltage-gated calcium channel subunit α1A, is involved in pre- and postsynaptic Ca(2+) signaling, gene expression, and several genetic neurological disorders. We found that CACNA1A coordinates gene expression using a bicistronic mRNA bearing a cryptic internal ribosomal entry site (IRES). The first cistron encodes the well-characterized α1A subunit. The second expresses a transcription factor, α1ACT, which coordinates expression of a program of genes involved in neural and Purkinje cell development. α1ACT also contains the polyglutamine (polyQ) tract that, when expanded, causes spinocerebellar ataxia type 6 (SCA6). When expressed as an independent polypeptide, α1ACT-bearing an expanded polyQ tract-lacks transcription factor function and neurite outgrowth properties, causes cell death in culture, and leads to ataxia and cerebellar atrophy in transgenic mice. Suppression of CACNA1A IRES function in SCA6 may be a potential therapeutic strategy.


Assuntos
Canais de Cálcio/genética , Ataxias Espinocerebelares/genética , Fatores de Transcrição/genética , Animais , Canais de Cálcio/metabolismo , Morte Celular , Linhagem Celular Tumoral , Cerebelo/embriologia , Cerebelo/fisiopatologia , Regulação da Expressão Gênica , Humanos , Camundongos , Neuritos/metabolismo , Peptídeos/genética , Células de Purkinje/metabolismo , Ratos , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/fisiopatologia , Fatores de Transcrição/metabolismo , Transcrição Gênica
16.
Cell ; 152(5): 1065-76, 2013 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-23452854

RESUMO

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.


Assuntos
Neoplasias Cerebelares/patologia , Cerebelo/metabolismo , Meduloblastoma/patologia , Neuropilina-1/metabolismo , Proteínas da Gravidez/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Neoplasias Cerebelares/metabolismo , Humanos , Meduloblastoma/metabolismo , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Comunicação Parácrina , Fator de Crescimento Placentário , Transplante Heterólogo , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo
17.
Cell ; 154(3): 505-17, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23911318

RESUMO

Purine biosynthesis and metabolism, conserved in all living organisms, is essential for cellular energy homeostasis and nucleic acid synthesis. The de novo synthesis of purine precursors is under tight negative feedback regulation mediated by adenosine and guanine nucleotides. We describe a distinct early-onset neurodegenerative condition resulting from mutations in the adenosine monophosphate deaminase 2 gene (AMPD2). Patients have characteristic brain imaging features of pontocerebellar hypoplasia (PCH) due to loss of brainstem and cerebellar parenchyma. We found that AMPD2 plays an evolutionary conserved role in the maintenance of cellular guanine nucleotide pools by regulating the feedback inhibition of adenosine derivatives on de novo purine synthesis. AMPD2 deficiency results in defective GTP-dependent initiation of protein translation, which can be rescued by administration of purine precursors. These data suggest AMPD2-related PCH as a potentially treatable early-onset neurodegenerative disease.


Assuntos
AMP Desaminase/metabolismo , Atrofias Olivopontocerebelares/metabolismo , Purinas/biossíntese , AMP Desaminase/química , AMP Desaminase/genética , Animais , Tronco Encefálico/patologia , Cerebelo/patologia , Criança , Feminino , Guanosina Trifosfato/metabolismo , Humanos , Masculino , Camundongos , Camundongos Knockout , Mutação , Células-Tronco Neurais/metabolismo , Atrofias Olivopontocerebelares/genética , Atrofias Olivopontocerebelares/patologia , Biossíntese de Proteínas , Saccharomyces cerevisiae/enzimologia , Saccharomyces cerevisiae/metabolismo
18.
Nature ; 605(7911): 722-727, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35545673

RESUMO

Cellular diversification is critical for specialized functions of the brain including learning and memory1. Single-cell RNA sequencing facilitates transcriptomic profiling of distinct major types of neuron2-4, but the divergence of transcriptomic profiles within a neuronal population and their link to function remain poorly understood. Here we isolate nuclei tagged5 in specific cell types followed by single-nucleus RNA sequencing to profile Purkinje neurons and map their responses to motor activity and learning. We find that two major subpopulations of Purkinje neurons, identified by expression of the genes Aldoc and Plcb4, bear distinct transcriptomic features. Plcb4+, but not Aldoc+, Purkinje neurons exhibit robust plasticity of gene expression in mice subjected to sensorimotor and learning experience. In vivo calcium imaging and optogenetic perturbation reveal that Plcb4+ Purkinje neurons have a crucial role in associative learning. Integrating single-nucleus RNA sequencing datasets with weighted gene co-expression network analysis uncovers a learning gene module that includes components of FGFR2 signalling in Plcb4+ Purkinje neurons. Knockout of Fgfr2 in Plcb4+ Purkinje neurons in mice using CRISPR disrupts motor learning. Our findings define how diversification of Purkinje neurons is linked to their responses in motor learning and provide a foundation for understanding their differential vulnerability to neurological disorders.


Assuntos
Células de Purkinje , Transcriptoma , Animais , Cerebelo , Aprendizagem/fisiologia , Camundongos , Camundongos Knockout , Plasticidade Neuronal/genética , Neurônios/fisiologia , Células de Purkinje/metabolismo , Transcriptoma/genética
19.
Nature ; 612(7941): 787-794, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36450980

RESUMO

Medulloblastoma (MB) is the most common malignant childhood brain tumour1,2, yet the origin of the most aggressive subgroup-3 form remains elusive, impeding development of effective targeted treatments. Previous analyses of mouse cerebella3-5 have not fully defined the compositional heterogeneity of MBs. Here we undertook single-cell profiling of freshly isolated human fetal cerebella to establish a reference map delineating hierarchical cellular states in MBs. We identified a unique transitional cerebellar progenitor connecting neural stem cells to neuronal lineages in developing fetal cerebella. Intersectional analysis revealed that the transitional progenitors were enriched in aggressive MB subgroups, including group 3 and metastatic tumours. Single-cell multi-omics revealed underlying regulatory networks in the transitional progenitor populations, including transcriptional determinants HNRNPH1 and SOX11, which are correlated with clinical prognosis in group 3 MBs. Genomic and Hi-C profiling identified de novo long-range chromatin loops juxtaposing HNRNPH1/SOX11-targeted super-enhancers to cis-regulatory elements of MYC, an oncogenic driver for group 3 MBs. Targeting the transitional progenitor regulators inhibited MYC expression and MYC-driven group 3 MB growth. Our integrated single-cell atlases of human fetal cerebella and MBs show potential cell populations predisposed to transformation and regulatory circuitries underlying tumour cell states and oncogenesis, highlighting hitherto unrecognized transitional progenitor intermediates predictive of disease prognosis and potential therapeutic vulnerabilities.


Assuntos
Neoplasias Encefálicas , Transformação Celular Neoplásica , Feto , Meduloblastoma , Humanos , Neoplasias Encefálicas/patologia , Transformação Celular Neoplásica/patologia , Neoplasias Cerebelares/patologia , Cerebelo/citologia , Cerebelo/patologia , Feto/citologia , Feto/patologia , Meduloblastoma/patologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/patologia , Prognóstico
20.
Nature ; 609(7929): 1021-1028, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36131014

RESUMO

Medulloblastoma (MB) comprises a group of heterogeneous paediatric embryonal neoplasms of the hindbrain with strong links to early development of the hindbrain1-4. Mutations that activate Sonic hedgehog signalling lead to Sonic hedgehog MB in the upper rhombic lip (RL) granule cell lineage5-8. By contrast, mutations that activate WNT signalling lead to WNT MB in the lower RL9,10. However, little is known about the more commonly occurring group 4 (G4) MB, which is thought to arise in the unipolar brush cell lineage3,4. Here we demonstrate that somatic mutations that cause G4 MB converge on the core binding factor alpha (CBFA) complex and mutually exclusive alterations that affect CBFA2T2, CBFA2T3, PRDM6, UTX and OTX2. CBFA2T2 is expressed early in the progenitor cells of the cerebellar RL subventricular zone in Homo sapiens, and G4 MB transcriptionally resembles these progenitors but are stalled in developmental time. Knockdown of OTX2 in model systems relieves this differentiation blockade, which allows MB cells to spontaneously proceed along normal developmental differentiation trajectories. The specific nature of the split human RL, which is destined to generate most of the neurons in the human brain, and its high level of susceptible EOMES+KI67+ unipolar brush cell progenitor cells probably predisposes our species to the development of G4 MB.


Assuntos
Diferenciação Celular , Neoplasias Cerebelares , Meduloblastoma , Metencéfalo , Diferenciação Celular/genética , Linhagem da Célula , Neoplasias Cerebelares/classificação , Neoplasias Cerebelares/genética , Neoplasias Cerebelares/patologia , Cerebelo/embriologia , Cerebelo/patologia , Subunidades alfa de Fatores de Ligação ao Core/genética , Proteínas Hedgehog/metabolismo , Histona Desmetilases , Humanos , Antígeno Ki-67/metabolismo , Meduloblastoma/classificação , Meduloblastoma/genética , Meduloblastoma/patologia , Metencéfalo/embriologia , Metencéfalo/patologia , Proteínas Musculares , Mutação , Fatores de Transcrição Otx/deficiência , Fatores de Transcrição Otx/genética , Proteínas Repressoras , Proteínas com Domínio T/metabolismo , Fatores de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA