Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 91
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Cell Rep ; 43(5): 114257, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38761373

RESUMEN

Spiny projection neurons (SPNs) of the striatum are critical in integrating neurochemical information to coordinate motor and reward-based behavior. Mutations in the regulatory transcription factors expressed in SPNs can result in neurodevelopmental disorders (NDDs). Paralogous transcription factors Foxp1 and Foxp2, which are both expressed in the dopamine receptor 1 (D1) expressing SPNs, are known to have variants implicated in NDDs. Utilizing mice with a D1-SPN-specific loss of Foxp1, Foxp2, or both and a combination of behavior, electrophysiology, and cell-type-specific genomic analysis, loss of both genes results in impaired motor and social behavior as well as increased firing of the D1-SPNs. Differential gene expression analysis implicates genes involved in autism risk, electrophysiological properties, and neuronal development and function. Viral-mediated re-expression of Foxp1 into the double knockouts is sufficient to restore electrophysiological and behavioral deficits. These data indicate complementary roles between Foxp1 and Foxp2 in the D1-SPNs.


Asunto(s)
Cuerpo Estriado , Factores de Transcripción Forkhead , Animales , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Ratones , Cuerpo Estriado/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Ratones Noqueados , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D1/genética , Masculino , Neuronas/metabolismo , Ratones Endogámicos C57BL , Conducta Social
2.
Sci Adv ; 10(18): eadm7039, 2024 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-38701209

RESUMEN

Long-range glutamatergic inputs originating from the cortex and thalamus are indispensable for striatal development, providing the foundation for motor and cognitive functions. Despite their significance, transcriptional regulation governing these inputs remains largely unknown. We investigated the role of a transcription factor encoded by a high-risk autism-associated gene, FOXP1, in sculpting glutamatergic inputs onto spiny projection neurons (SPNs) within the striatum. We find a neuron subtype-specific role of FOXP1 in strengthening and maturing glutamatergic inputs onto dopamine receptor 2-expressing SPNs (D2 SPNs). We also find that FOXP1 promotes synaptically driven excitability in these neurons. Using single-nuclei RNA sequencing, we identify candidate genes that mediate these cell-autonomous processes through postnatal FOXP1 function at the post-synapse. Last, we demonstrate that postnatal FOXP1 reinstatement rescues electrophysiological deficits, cell type-specific gene expression changes, and behavioral phenotypes. Together, this study enhances our understanding of striatal circuit development and provides proof of concept for a therapeutic approach for FOXP1 syndrome and other neurodevelopmental disorders.


Asunto(s)
Cuerpo Estriado , Factores de Transcripción Forkhead , Neuronas , Receptores de Dopamina D2 , Proteínas Represoras , Animales , Factores de Transcripción Forkhead/metabolismo , Factores de Transcripción Forkhead/genética , Cuerpo Estriado/metabolismo , Cuerpo Estriado/citología , Ratones , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Proteínas Represoras/genética , Fenotipo , Sinapsis/metabolismo , Sinapsis/fisiología , Masculino
3.
bioRxiv ; 2023 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-37961477

RESUMEN

Long-range glutamatergic inputs from the cortex and thalamus are critical for motor and cognitive processing in the striatum. Transcription factors that orchestrate the development of these inputs are largely unknown. We investigated the role of a transcription factor and high-risk autism-associated gene, FOXP1, in the development of glutamatergic inputs onto spiny projection neurons (SPNs) in the striatum. We find that FOXP1 robustly drives the strengthening and maturation of glutamatergic input onto dopamine receptor 2-expressing SPNs (D2 SPNs) but has a comparatively milder effect on D1 SPNs. This process is cell-autonomous and is likely mediated through postnatal FOXP1 function at the postsynapse. We identified postsynaptic FOXP1-regulated transcripts as potential candidates for mediating these effects. Postnatal reinstatement of FOXP1 rescues electrophysiological deficits, reverses gene expression alterations resulting from embryonic deletion, and mitigates behavioral phenotypes. These results provide support for a possible therapeutic approach for individuals with FOXP1 syndrome.

4.
Nature ; 623(7986): 274-282, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37938705

RESUMEN

Neuroscience research has entered a phase of key discoveries in the realm of neurogenomics owing to strong financial and intellectual support for resource building and tool development. The previous challenge of tissue heterogeneity has been met with the application of techniques that can profile individual cells at scale. Moreover, the ability to perturb genes, gene regulatory elements and neuronal activity in a cell-type-specific manner has been integrated with gene expression studies to uncover the functional underpinnings of the genome at a systems level. Although these insights have necessarily been grounded in model systems, we now have the opportunity to apply these approaches in humans and in human tissue, thanks to advances in human genetics, brain imaging and tissue collection. We acknowledge that there will probably always be limits to the extent to which we can apply the genomic tools developed in model systems to human neuroscience; however, as we describe in this Perspective, the neuroscience field is now primed with an optimal foundation for tackling this ambitious challenge. The application of systems-level network analyses to these datasets will facilitate a deeper appreciation of human neurogenomics that cannot otherwise be achieved from directly observable phenomena.


Asunto(s)
Genómica , Neurociencias , Biología de Sistemas , Humanos , Encéfalo/metabolismo , Genómica/tendencias , Modelos Biológicos , Neurociencias/métodos , Neurociencias/tendencias , Biología de Sistemas/tendencias
5.
bioRxiv ; 2023 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-37745404

RESUMEN

DNA-based evolutionary comparisons of regulatory genomic elements enable insight into functional changes, overcoming tissue inaccessibility. Here, we harnessed adult and fetal cortex single-cell ATAC-seq datasets to uncover DNA substitutions specific to the human and human-ancestral lineages within apes. We found that fetal microglia identity is evolutionarily divergent in all lineages, whereas other cell types are conserved. Using multiomic datasets, we further identified genes linked to multiple lineage-divergent gene regulatory elements and implicated biological pathways associated with these divergent features. We also uncovered patterns of transcription factor binding site evolution across lineages and identified expansion of bHLH-PAS factor targets in human-hominin lineages, and MEF2 factor targets in the ape lineage. Finally, conserved features were more enriched in brain disease variants, whereas there was no distinct enrichment on the human lineage compared to its ancestral lineages. Our study identifies major evolutionary patterns in the human brain epigenome at cellular resolution.

6.
PLoS Biol ; 21(8): e3001852, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37540706

RESUMEN

During cortical development, human basal radial glial cells (bRGCs) are highly capable of sustained self-renewal and neurogenesis. Selective pressures on this cell type may have contributed to the evolution of the human neocortex, leading to an increase in cortical size. bRGCs have enriched expression for Forkhead Box P1 (FOXP1), a transcription factor implicated in neurodevelopmental disorders (NDDs) such as autism spectrum disorder. However, the cell type-specific roles of FOXP1 in bRGCs during cortical development remain unexplored. Here, we examine the requirement for FOXP1 gene expression regulation underlying the production of bRGCs using human brain organoids. We examine a developmental time point when FOXP1 expression is highest in the cortical progenitors, and the bRGCs, in particular, begin to actively produce neurons. With the loss of FOXP1, we show a reduction in the number of bRGCs, as well as reduced proliferation and differentiation of the remaining bRGCs, all of which lead to reduced numbers of excitatory cortical neurons over time. Using single-nuclei RNA sequencing and cell trajectory analysis, we uncover a role for FOXP1 in directing cortical progenitor proliferation and differentiation by regulating key signaling pathways related to neurogenesis and NDDs. Together, these results demonstrate that FOXP1 regulates human-specific features in early cortical development.


Asunto(s)
Factores de Transcripción Forkhead , Neocórtex , Neurogénesis , Proteínas Represoras , Humanos , Células Ependimogliales/metabolismo , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Proteínas Represoras/metabolismo
7.
bioRxiv ; 2023 Jun 26.
Artículo en Inglés | MEDLINE | ID: mdl-37425820

RESUMEN

Spiny projection neurons (SPNs) of the striatum are critical in integrating neurochemical information to coordinate motor and reward-based behavior. Mutations in the regulatory transcription factors expressed in SPNs can result in neurodevelopmental disorders (NDDs). Paralogous transcription factors Foxp1 and Foxp2, which are both expressed in the dopamine receptor 1 (D1) expressing SPNs, are known to have variants implicated in NDDs. Utilizing mice with a D1-SPN specific loss of Foxp1, Foxp2, or both and a combination of behavior, electrophysiology, and cell-type specific genomic analysis, loss of both genes results in impaired motor and social behavior as well as increased firing of the D1-SPNs. Differential gene expression analysis implicates genes involved in autism risk, electrophysiological properties, and neuronal development and function. Viral mediated re-expression of Foxp1 into the double knockouts was sufficient to restore electrophysiological and behavioral deficits. These data indicate complementary roles between Foxp1 and Foxp2 in the D1-SPNs.

8.
Nature ; 620(7972): 145-153, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37468639

RESUMEN

Human-specific genomic changes contribute to the unique functionalities of the human brain1-5. The cellular heterogeneity of the human brain6,7 and the complex regulation of gene expression highlight the need to characterize human-specific molecular features at cellular resolution. Here we analysed single-nucleus RNA-sequencing and single-nucleus assay for transposase-accessible chromatin with sequencing datasets for human, chimpanzee and rhesus macaque brain tissue from posterior cingulate cortex. We show a human-specific increase of oligodendrocyte progenitor cells and a decrease of mature oligodendrocytes across cortical tissues. Human-specific regulatory changes were accelerated in oligodendrocyte progenitor cells, and we highlight key biological pathways that may be associated with the proportional changes. We also identify human-specific regulatory changes in neuronal subtypes, which reveal human-specific upregulation of FOXP2 in only two of the neuronal subtypes. We additionally identify hundreds of new human accelerated genomic regions associated with human-specific chromatin accessibility changes. Our data also reveal that FOS::JUN and FOX motifs are enriched in the human-specifically accessible chromatin regions of excitatory neuronal subtypes. Together, our results reveal several new mechanisms underlying the evolutionary innovation of human brain at cell-type resolution.


Asunto(s)
Evolución Molecular , Giro del Cíngulo , Animales , Humanos , Núcleo Celular/metabolismo , Cromatina/genética , Cromatina/metabolismo , Conjuntos de Datos como Asunto , Genoma Humano/genética , Genómica , Giro del Cíngulo/citología , Giro del Cíngulo/metabolismo , Macaca mulatta/genética , Neuronas/clasificación , Neuronas/citología , Oligodendroglía/citología , Oligodendroglía/metabolismo , Pan troglodytes/genética , Análisis de Expresión Génica de una Sola Célula , Células Madre/citología , Transposasas/metabolismo , Ensamble y Desensamble de Cromatina
9.
Sci Rep ; 13(1): 9031, 2023 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-37270616

RESUMEN

The striatum integrates dense neuromodulatory inputs from many brain regions to coordinate complex behaviors. This integration relies on the coordinated responses from distinct striatal cell types. While previous studies have characterized the cellular and molecular composition of the striatum using single-cell RNA-sequencing at distinct developmental timepoints, the molecular changes spanning embryonic through postnatal development at the single-cell level have not been examined. Here, we combine published mouse striatal single-cell datasets from both embryonic and postnatal timepoints to analyze the developmental trajectory patterns and transcription factor regulatory networks within striatal cell types. Using this integrated dataset, we found that dopamine receptor-1 expressing spiny projection neurons have an extended period of transcriptional dynamics and greater transcriptional complexity over postnatal development compared to dopamine receptor-2 expressing neurons. Moreover, we found the transcription factor, FOXP1, exerts indirect changes to oligodendrocytes. These data can be accessed and further analyzed through an interactive website ( https://mouse-striatal-dev.cells.ucsc.edu ).


Asunto(s)
Cuerpo Estriado , Neuronas , Animales , Ratones , Neuronas/metabolismo , Cuerpo Estriado/metabolismo , Neostriado/metabolismo , Factores de Transcripción/metabolismo , Receptores Dopaminérgicos/metabolismo
10.
Cerebellum ; 22(6): 1166-1181, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36242761

RESUMEN

Essential tremor (ET) is a common, progressive neurological disease characterized by an 8-12-Hz kinetic tremor. Despite its high prevalence, the patho-mechanisms of tremor in ET are not fully known. Through comprehensive studies in postmortem brains, we identified major morphological changes in the ET cerebellum that reflect cellular damage in Purkinje cells (PCs), suggesting that PC damage is central to ET pathogenesis. We previously performed a transcriptome analysis in ET cerebellar cortex, identifying candidate genes and several dysregulated pathways. To directly target PCs, we purified RNA from PCs isolated by laser capture microdissection and performed the first ever PC-specific RNA-sequencing analysis in ET versus controls. Frozen postmortem cerebellar cortex from 24 ETs and 16 controls underwent laser capture microdissection, obtaining ≥2000 PCs per sample. RNA transcriptome was analyzed via differential gene expression, principal component analysis (PCA), and gene set enrichment analyses (GSEA). We identified 36 differentially expressed genes, encompassing multiple cellular processes. Some ET (13/24) had greater dysregulation of these genes and segregated from most controls and remaining ETs in PCA. Characterization of genes/pathways enriched in this PCA and GSEA identified multiple pathway dysregulations in ET, including RNA processing/splicing, synapse organization/ion transport, and oxidative stress/inflammation. Furthermore, a different set of pathways characterized marked heterogeneity among ET patients. Our data indicate a range of possible mechanisms for the pathogenesis of ET. Significant heterogeneity among ET combined with dysregulation of multiple cellular processes supports the notion that ET is a family of disorders rather than one disease entity.


Asunto(s)
Temblor Esencial , Células de Purkinje , Humanos , Células de Purkinje/metabolismo , Temblor Esencial/patología , Temblor/patología , Cerebelo/patología , Perfilación de la Expresión Génica , ARN/metabolismo , Rayos Láser
11.
Neuron ; 110(24): 4043-4056.e5, 2022 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-36240767

RESUMEN

Ambient RNA contamination in single-cell and single-nuclei RNA sequencing (snRNA-seq) is a significant problem, but its consequences are poorly understood. Here, we show that ambient RNAs in brain snRNA-seq datasets have a nuclear or non-nuclear origin with distinct gene set signatures. Both ambient RNA signatures are predominantly neuronal, and we find that some previously annotated neuronal cell types are distinguished by ambient RNA contamination. We detect pervasive neuronal ambient RNA contamination in all glial cell types unless glia and neurons are physically separated prior to sequencing. We demonstrate that this contamination can be removed in silico and show that previous single-nuclei RNA-seq-based annotations of immature oligodendrocytes are glial nuclei contaminated with ambient RNAs. After ambient RNA removal, we detect rare, committed oligodendrocyte progenitor cells not annotated in most previous adult human brain datasets. Together, these results provide an in-depth analysis of ambient RNA contamination in brain single-nuclei datasets.


Asunto(s)
Núcleo Celular , ARN , Adulto , Humanos , ARN/metabolismo , Núcleo Celular/metabolismo , Neuronas/metabolismo , Análisis de Secuencia de ARN/métodos , ARN Nuclear Pequeño/genética , ARN Nuclear Pequeño/metabolismo , Núcleo Solitario , Análisis de la Célula Individual/métodos
12.
Dev Neurobiol ; 82(6): 517-532, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35932776

RESUMEN

The neocortex (or pallium) consists of diverse cell types that are organized in a highly species-specific manner under strict spatiotemporal control during development. Many of the cell types are present transiently throughout development but contribute to permanent species-specific cortical features that are acquired through evolution. Therefore, capturing cell type-specific biological information has always been an important quest in the field of neurodevelopment. The progress in achieving fine cellular resolution has been slow due to technical challenges. However, with recent advancements in single-cell and multi-omics technologies, many laboratories have begun to successfully interrogate cellular and molecular mechanisms driving corticogenesis at single-cell resolution. In this review, we provide summarized results from many primary publications and several in-depth review articles that utilize or address single-cell genomics techniques to understand important topics, such as cellular and molecular mechanisms governing cortical progenitor proliferation, cell lineage progression, neuronal specification, and arealization, across multiple gyrencephalic (i.e., human and non-human primates) and lissencephalic species (i.e., mouse, reptiles, and songbirds). We also examine findings from recent studies involving epigenomic and posttranscriptional regulation of corticogenesis. In the discussion section, we provide our insights on the challenges the field currently faces as well as promising future applications of single cell technologies.


Asunto(s)
Corteza Cerebral , Neocórtex , Animales , Linaje de la Célula , Ratones , Neuronas/metabolismo
13.
Curr Opin Cell Biol ; 78: 102118, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35947942

RESUMEN

While there is extensive research on memory-related oscillations and brain gene expression, the relationship between oscillations and gene expression has rarely been studied. Recently, progress has been made to identify specific genes associated with oscillations that are correlated with episodic memory. Neocortical regions, in particular the temporal pole, have been examined in this line of research due to their accessibility during neurosurgical procedures. By harnessing this accessibility, a unique and powerful study design has allowed gene expression and intracranial oscillatory data to be sourced from the same human patients. These studies have identified a plethora of understudied gene targets that should be further characterized with respect to human brain function. Future work should extend to other brain regions to increase our understanding of the genetic signatures of oscillations and, ultimately, human cognition.


Asunto(s)
Ondas Encefálicas , Memoria Episódica , Encéfalo , Humanos
14.
Nat Commun ; 13(1): 3328, 2022 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-35680911

RESUMEN

Gene expression covaries with brain activity as measured by resting state functional magnetic resonance imaging (MRI). However, it is unclear how genomic differences driven by disease state can affect this relationship. Here, we integrate from the ABIDE I and II imaging cohorts with datasets of gene expression in brains of neurotypical individuals and individuals with autism spectrum disorder (ASD) with regionally matched brain activity measurements from fMRI datasets. We identify genes linked with brain activity whose association is disrupted in ASD. We identified a subset of genes that showed a differential developmental trajectory in individuals with ASD compared with controls. These genes are enriched in voltage-gated ion channels and inhibitory neurons, pointing to excitation-inhibition imbalance in ASD. We further assessed differences at the regional level showing that the primary visual cortex is the most affected region in ASD. Our results link disrupted brain expression patterns of individuals with ASD to brain activity and show developmental, cell type, and regional enrichment of activity linked genes.


Asunto(s)
Trastorno del Espectro Autista , Trastorno del Espectro Autista/diagnóstico por imagen , Trastorno del Espectro Autista/genética , Encéfalo/diagnóstico por imagen , Mapeo Encefálico/métodos , Expresión Génica , Humanos , Imagen por Resonancia Magnética/métodos , Vías Nerviosas
15.
Front Genet ; 12: 754198, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34795694

RESUMEN

Early life stress (ELS), such as abuse, neglect, and maltreatment, exhibits a strong impact on the brain and mental development of children. However, it is not fully understood how ELS affects social behaviors and social-associated behaviors as well as developing prefrontal cortex (PFC). In this study, we performed social isolation on weaned pre-adolescent mice until adolescence and investigated these behaviors and PFC characteristics in adolescent mice. We found the ELS induced social impairments in social novelty, social interaction, and social preference in adolescent mice. We also observed increases of anxiety-like behaviors in ELS mice. In histological analysis, we found a reduced number of neurons and an increased number of microglia in the PFC of ELS mice. To identify the gene associated with behavioral and histological features, we analyzed transcriptome in the PFC of ELS mice and identified 15 differentially expressed genes involved in transcriptional regulation, stress, and synaptic signaling. Our study demonstrates that ELS influences social behaviors, anxiety-like behaviors through cytoarchitectural and transcriptomic alterations in the PFC of adolescent mice.

16.
STAR Protoc ; 2(4): 100844, 2021 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-34585170

RESUMEN

Single-nucleus RNA sequencing (snRNA-seq), where nuclear transcriptomes are a proxy to cellular transcriptomes, has been used to profile human brain. snRNA-seq is sensitive to tissue processing, tissue quality, postmortem interval time, and cellular debris. This protocol outlines steps for the isolation of high-quality nuclei from surgically resected human brain tissue followed by a sucrose gradient yielding neuronal and non-neuronal nuclei enabling unbiased analysis of various cell types. For complete details on the use and execution of this protocol, please refer to Ayhan et al. (2021).


Asunto(s)
Núcleo Celular , Hipocampo , Núcleo Celular/metabolismo , Hipocampo/cirugía , Humanos , ARN Nuclear Pequeño/metabolismo , Análisis de Secuencia de ARN/métodos , Núcleo Solitario
17.
Elife ; 102021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34473622

RESUMEN

Concussion is associated with a myriad of deleterious immediate and long-term consequences. Yet the molecular mechanisms and genetic targets promoting the selective vulnerability of different neural subtypes to dysfunction and degeneration remain unclear. Translating experimental models of blunt force trauma in C. elegans to concussion in mice, we identify a conserved neuroprotective mechanism in which reduction of mitochondrial electron flux through complex IV suppresses trauma-induced degeneration of the highly vulnerable dopaminergic neurons. Reducing cytochrome C oxidase function elevates mitochondrial-derived reactive oxygen species, which signal through the cytosolic hypoxia inducing transcription factor, Hif1a, to promote hyperphosphorylation and inactivation of the pyruvate dehydrogenase, PDHE1α. This critical enzyme initiates the Warburg shunt, which drives energetic reallocation from mitochondrial respiration to astrocyte-mediated glycolysis in a neuroprotective manner. These studies demonstrate a conserved process in which glycolytic preconditioning suppresses Parkinson-like hypersensitivity of dopaminergic neurons to trauma-induced degeneration via redox signaling and the Warburg effect.


Concussion is a type of traumatic brain injury that results from a sudden blow or jolt to the head. Symptoms can include a passing headache, dizziness, confusion or sensitivity to light, but experiencing multiple concussions can have drastic repercussions in later life. Studies of professional athletes have shown that those who experience one or more concussions are prone to developing Alzheimer's and Parkinson's disease, two well-known neurodegenerative diseases. Both conditions involve the progressive loss or breakdown of nerve cells, called neurons. But exactly how this so-called neurodegeneration of brain cells stems from the original, physical injury remains unclear. Head trauma may cause damage to the structural support of a cell or disrupt the flow of electrical impulses through neurons. Energy use and production in damaged cells could shift into overdrive to repair the damage. The chemical properties of different types of brain cells could also make some more vulnerable to trauma than others. Besides neurons, star-shaped support cells in the brain called astrocytes, which may have some protective ability, could also be affected. To investigate which cells may be more susceptible to traumatic injuries, Solano Fonseca et al. modelled the impacts of concussion-like head trauma in roundworms (C. elegans) and mice. In both animals, one type of neuron was extremely vulnerable to cell death after trauma. Neurons that release dopamine, a chemical involved in cell-to-cell communication and the brain's reward system, showed signs of cell damage and deteriorated after injury. Dopaminergic cells, as these cells are called, are involved in motor coordination, and the loss of dopaminergic cells has been linked to both Alzheimer's and Parkinson's disease. Astrocytes, however, had a role in reducing the death of dopaminergic neurons after trauma. In experiments, astrocytes appeared to restore the balance of energy production to meet the increased energy demands of impacted neurons. Single-cell analyses showed that genes involved in metabolism were switched on in astrocytes to produce energy via an alternative pathway. This energetic shift facilitated via astrocytes may help mitigate against some damage to dopamine-producing neurons after trauma, reducing cell death. This work furthers our understanding of cellular changes in the concussed brain. More research will be required to better characterise how this immediate trauma to cells, and the subsequent loss of dopaminergic neurons, impacts brain health long-term. Efforts to design effective therapies to slow or reverse these changes could then follow.


Asunto(s)
Astrocitos , Lesiones Traumáticas del Encéfalo , Glucólisis/fisiología , Degeneración Nerviosa , Neuroprotección/fisiología , Animales , Astrocitos/citología , Astrocitos/metabolismo , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/fisiopatología , Caenorhabditis elegans , Células Cultivadas , Neuronas Dopaminérgicas/citología , Neuronas Dopaminérgicas/metabolismo , Células HEK293 , Humanos , Ratones , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/fisiopatología
18.
Neuron ; 109(20): 3268-3282.e6, 2021 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-34416169

RESUMEN

The suprachiasmatic nucleus (SCN) is the master circadian pacemaker in mammals and is entrained by environmental light. However, the molecular basis of the response of the SCN to light is not fully understood. We used RNA/chromatin immunoprecipitation/single-nucleus sequencing with circadian behavioral assays to identify mouse SCN cell types and explore their responses to light. We identified three peptidergic cell types that responded to light in the SCN: arginine vasopressin (AVP), vasoactive intestinal peptide (VIP), and cholecystokinin (CCK). In each cell type, light-responsive subgroups were enriched for expression of neuronal Per-Arnt-Sim (PAS) domain protein 4 (NPAS4) target genes. Further, mice lacking Npas4 had a longer circadian period under constant conditions, a damped phase response curve to light, and reduced light-induced gene expression in the SCN. Our data indicate that NPAS4 is necessary for normal transcriptional responses to light in the SCN and critical for photic phase-shifting of circadian behavior.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Ritmo Circadiano/genética , Luz , Neuronas/metabolismo , Núcleo Supraquiasmático/metabolismo , Animales , Arginina Vasopresina/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Colecistoquinina/metabolismo , Inmunoprecipitación de Cromatina , Ritmo Circadiano/efectos de la radiación , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Neuronas/efectos de la radiación , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Núcleo Supraquiasmático/citología , Núcleo Supraquiasmático/efectos de la radiación , Péptido Intestinal Vasoactivo/metabolismo
19.
Redox Biol ; 45: 102057, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34198071

RESUMEN

Methylglyoxal (MG) is a reactive and cytotoxic α-dicarbonyl byproduct of glycolysis. Our bodies have several bio-defense systems to detoxify MG, including an enzymatic system by glyoxalase (GLO) 1 and GLO2. We identified a subtype of schizophrenia patients with novel mutations in the GLO1 gene that results in reductions of enzymatic activity. Moreover, we found that vitamin B6 (VB6) levels in peripheral blood of the schizophrenia patients with GLO1 dysfunction are significantly lower than that of healthy controls. However, the effects of GLO1 dysfunction and VB6 deficiency on the pathophysiology of schizophrenia remains poorly understood. Here, we generated a novel mouse model for this subgroup of schizophrenia patients by feeding Glo1 knockout mice VB6-deficent diets (KO/VB6(-)) and evaluated the combined effects of GLO1 dysfunction and VB6 deficiency on brain function. KO/VB6(-) mice accumulated homocysteine in plasma and MG in the prefrontal cortex (PFC), hippocampus, and striatum, and displayed behavioral deficits, such as impairments of social interaction and cognitive memory and a sensorimotor deficit in the prepulse inhibition test. Furthermore, we found aberrant gene expression related to mitochondria function in the PFC of the KO/VB6(-) mice by RNA-sequencing and weighted gene co-expression network analysis (WGCNA). Finally, we demonstrated abnormal mitochondrial respiratory function and subsequently enhanced oxidative stress in the PFC of KO/VB6(-) mice in the PFC. These findings suggest that the combination of GLO1 dysfunction and VB6 deficiency may cause the observed behavioral deficits via mitochondrial dysfunction and oxidative stress in the PFC.


Asunto(s)
Lactoilglutatión Liasa , Esquizofrenia , Deficiencia de Vitamina B 6 , Animales , Humanos , Lactoilglutatión Liasa/genética , Lactoilglutatión Liasa/metabolismo , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Corteza Prefrontal/metabolismo , Esquizofrenia/genética
20.
Neuron ; 109(13): 2091-2105.e6, 2021 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-34051145

RESUMEN

The hippocampus supports many facets of cognition, including learning, memory, and emotional processing. Anatomically, the hippocampus runs along a longitudinal axis, posterior to anterior in primates. The structure, function, and connectivity of the hippocampus vary along this axis. In human hippocampus, longitudinal functional heterogeneity remains an active area of investigation, and structural heterogeneity has not been described. To understand the cellular and molecular diversity along the hippocampal long axis in human brain and define molecular signatures corresponding to functional domains, we performed single-nuclei RNA sequencing on surgically resected human anterior and posterior hippocampus from epilepsy patients, identifying differentially expressed genes at cellular resolution. We further identify axis- and cell-type-specific gene expression signatures that differentially intersect with human genetic signals, identifying cell-type-specific genes in the posterior hippocampus for cognitive function and the anterior hippocampus for mood and affect. These data are accessible as a public resource through an interactive website.


Asunto(s)
Expresión Génica , Hipocampo/metabolismo , Neuronas/metabolismo , Femenino , Perfilación de la Expresión Génica , Hipocampo/anatomía & histología , Humanos , Masculino , Microglía/metabolismo , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA