Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 974
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 624(7991): 403-414, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092914

RESUMEN

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.


Asunto(s)
Encéfalo , Perfilación de la Expresión Génica , Vías Nerviosas , Neuronas , Médula Espinal , Animales , Ratones , Hipotálamo , Neuronas/metabolismo , Neuropéptidos , Médula Espinal/citología , Médula Espinal/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Neurotransmisores , Mesencéfalo/citología , Formación Reticular/citología , Electrofisiología , Cerebelo/citología , Corteza Cerebral/citología
2.
Nature ; 624(7991): 333-342, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092915

RESUMEN

The function of the mammalian brain relies upon the specification and spatial positioning of diversely specialized cell types. Yet, the molecular identities of the cell types and their positions within individual anatomical structures remain incompletely known. To construct a comprehensive atlas of cell types in each brain structure, we paired high-throughput single-nucleus RNA sequencing with Slide-seq1,2-a recently developed spatial transcriptomics method with near-cellular resolution-across the entire mouse brain. Integration of these datasets revealed the cell type composition of each neuroanatomical structure. Cell type diversity was found to be remarkably high in the midbrain, hindbrain and hypothalamus, with most clusters requiring a combination of at least three discrete gene expression markers to uniquely define them. Using these data, we developed a framework for genetically accessing each cell type, comprehensively characterized neuropeptide and neurotransmitter signalling, elucidated region-specific specializations in activity-regulated gene expression and ascertained the heritability enrichment of neurological and psychiatric phenotypes. These data, available as an online resource ( www.BrainCellData.org ), should find diverse applications across neuroscience, including the construction of new genetic tools and the prioritization of specific cell types and circuits in the study of brain diseases.


Asunto(s)
Encéfalo , Perfilación de la Expresión Génica , Animales , Ratones , Encéfalo/anatomía & histología , Encéfalo/citología , Encéfalo/metabolismo , Perfilación de la Expresión Génica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Hipotálamo/citología , Hipotálamo/metabolismo , Mesencéfalo/citología , Mesencéfalo/metabolismo , Neuropéptidos/metabolismo , Neurotransmisores/metabolismo , Fenotipo , Rombencéfalo/citología , Rombencéfalo/metabolismo , Análisis de Expresión Génica de una Sola Célula , Transcriptoma/genética
3.
Nature ; 624(7991): 355-365, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38092919

RESUMEN

Single-cell analyses parse the brain's billions of neurons into thousands of 'cell-type' clusters residing in different brain structures1. Many cell types mediate their functions through targeted long-distance projections allowing interactions between specific cell types. Here we used epi-retro-seq2 to link single-cell epigenomes and cell types to long-distance projections for 33,034 neurons dissected from 32 different regions projecting to 24 different targets (225 source-to-target combinations) across the whole mouse brain. We highlight uses of these data for interrogating principles relating projection types to transcriptomics and epigenomics, and for addressing hypotheses about cell types and connections related to genetics. We provide an overall synthesis with 926 statistical comparisons of discriminability of neurons projecting to each target for every source. We integrate this dataset into the larger BRAIN Initiative Cell Census Network atlas, composed of millions of neurons, to link projection cell types to consensus clusters. Integration with spatial transcriptomics further assigns projection-enriched clusters to smaller source regions than the original dissections. We exemplify this by presenting in-depth analyses of projection neurons from the hypothalamus, thalamus, hindbrain, amygdala and midbrain to provide insights into properties of those cell types, including differentially expressed genes, their associated cis-regulatory elements and transcription-factor-binding motifs, and neurotransmitter use.


Asunto(s)
Encéfalo , Epigenómica , Vías Nerviosas , Neuronas , Animales , Ratones , Amígdala del Cerebelo , Encéfalo/citología , Encéfalo/metabolismo , Secuencia de Consenso , Conjuntos de Datos como Asunto , Perfilación de la Expresión Génica , Hipotálamo/citología , Mesencéfalo/citología , Vías Nerviosas/citología , Neuronas/metabolismo , Neurotransmisores/metabolismo , Secuencias Reguladoras de Ácidos Nucleicos , Rombencéfalo/citología , Análisis de la Célula Individual , Tálamo/citología , Factores de Transcripción/metabolismo
4.
Planta Med ; 89(11): 1087-1096, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37044130

RESUMEN

Over the last years, Sideritis extracts were shown to improve memory. However, their potential to promote the generation of new neurons, starting with the neuronal differentiation of neural stem cells, remains unexplored. Therefore, the present study aimed to evaluate the neurogenic effects of different Sideritis infusions in neural stem and precursor cells and their impact on cell viability. Moreover, the metabolic fingerprints were recorded using LC-DAD, LC-HRESIMS, and GC-MS. The neurogenic potential of infusions of the eight Sideritis taxa tested was as potent as the classical neuronal inducer combination of retinoic acid and valproic acid. Further cytotoxicity assays revealed that the IC50 values of the extracts were between 163 and 322 µg/mL. Hierarchical cluster analyses of the metabolic fingerprints unveiled that the two Sideritis taxa with the lowest IC50 values were the most divergent in the analytical techniques used. As the analysis focused on polyphenols, it is reasonable to assume that these compounds are responsible for the effect on the cell viability of SH-SY5Y neuroblastoma cells. This study is the first report on the neurogenic potential of Sideritis taxa and might support the use of Sideritis herbal preparations in the context of neurodegenerative diseases.


Asunto(s)
Neurogénesis , Extractos Vegetales , Sideritis , Sideritis/química , Sideritis/clasificación , Extractos Vegetales/farmacología , Neurogénesis/efectos de los fármacos , Animales , Ratones , Estructuras Embrionarias/citología , Neuronas/efectos de los fármacos , Línea Celular Tumoral , Encéfalo/citología , Especificidad de la Especie
5.
Nature ; 616(7955): 132-136, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36949189

RESUMEN

While motor cortical circuits contain information related to specific movement parameters1, long-range inputs also have a critical role in action execution2,3. Thalamic projections can shape premotor activity2-6 and have been suggested7 to mediate the selection of short, stereotyped actions comprising more complex behaviours8. However, the mechanisms by which thalamus interacts with motor cortical circuits to execute such movement sequences remain unknown. Here we find that thalamic drive engages a specific subpopulation of premotor neurons within the zebra finch song nucleus HVC (proper name) and that these inputs are critical for the progression between vocal motor elements (that is, 'syllables'). In vivo two-photon imaging of thalamic axons in HVC showed robust song-related activity, and online perturbations of thalamic function caused song to be truncated at syllable boundaries. We used thalamic stimulation to identify a sparse set of thalamically driven neurons within HVC, representing ~15% of the premotor neurons within that network. Unexpectedly, this population of putative thalamorecipient neurons is robustly active immediately preceding syllable onset, leading to the possibility that thalamic input can initiate individual song components through selectively targeting these 'starter cells'. Our findings highlight the motor thalamus as a director of cortical dynamics in the context of an ethologically relevant behavioural sequence.


Asunto(s)
Cortejo , Pinzones , Tálamo , Vocalización Animal , Animales , Pinzones/fisiología , Neuronas/fisiología , Tálamo/citología , Tálamo/fisiología , Vocalización Animal/fisiología , Corteza Motora/citología , Corteza Motora/fisiología , Vías Nerviosas/fisiología , Encéfalo/citología , Encéfalo/fisiología , Masculino
6.
J Neurosci ; 43(13): 2338-2348, 2023 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-36849414

RESUMEN

Photoaffinity ligands are best known as tools used to identify the specific binding sites of drugs to their molecular targets. However, photoaffinity ligands have the potential to further define critical neuroanatomic targets of drug action. In the brains of WT male mice, we demonstrate the feasibility of using photoaffinity ligands in vivo to prolong anesthesia via targeted yet spatially restricted photoadduction of azi-m-propofol (aziPm), a photoreactive analog of the general anesthetic propofol. Systemic administration of aziPm with bilateral near-ultraviolet photoadduction in the rostral pons, at the border of the parabrachial nucleus and locus coeruleus, produced a 20-fold increase in the duration of sedative and hypnotic effects compared with control mice without UV illumination. Photoadduction that missed the parabrachial-coerulean complex also failed to extend the sedative or hypnotic actions of aziPm and was indistinguishable from nonadducted controls. Paralleling the prolonged behavioral and EEG consequences of on target in vivo photoadduction, we conducted electrophysiologic recordings in rostral pontine brain slices. Using neurons within the locus coeruleus to further highlight the cellular consequences of irreversible aziPm binding, we demonstrate transient slowing of spontaneous action potentials with a brief bath application of aziPm that becomes irreversible on photoadduction. Together, these findings suggest that photochemistry-based strategies are a viable new approach for probing CNS physiology and pathophysiology.SIGNIFICANCE STATEMENT Photoaffinity ligands are drugs capable of light-induced irreversible binding, which have unexploited potential to identify the neuroanatomic sites of drug action. We systemically administer a centrally acting anesthetic photoaffinity ligand in mice, conduct localized photoillumination within the brain to covalently adduct the drug at its in vivo sites of action, and successfully enrich irreversible drug binding within a restricted 250 µm radius. When photoadduction encompassed the pontine parabrachial-coerulean complex, anesthetic sedation and hypnosis was prolonged 20-fold, thus illustrating the power of in vivo photochemistry to help unravel neuronal mechanisms of drug action.


Asunto(s)
Anestésicos Intravenosos , Encéfalo , Hipnosis , Hipnóticos y Sedantes , Ligandos , Etiquetas de Fotoafinidad , Propofol , Animales , Masculino , Ratones , Neuronas Adrenérgicas/efectos de los fármacos , Anestesia Intravenosa , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/efectos de la radiación , Electrocorticografía , Electroencefalografía , Hipnosis/métodos , Hipnóticos y Sedantes/administración & dosificación , Hipnóticos y Sedantes/química , Hipnóticos y Sedantes/farmacología , Hipnóticos y Sedantes/efectos de la radiación , Locus Coeruleus/citología , Locus Coeruleus/efectos de los fármacos , Locus Coeruleus/metabolismo , Locus Coeruleus/efectos de la radiación , Ratones Endogámicos C57BL , Núcleos Parabraquiales/efectos de los fármacos , Núcleos Parabraquiales/metabolismo , Núcleos Parabraquiales/efectos de la radiación , Etiquetas de Fotoafinidad/química , Etiquetas de Fotoafinidad/efectos de la radiación , Propofol/administración & dosificación , Propofol/análogos & derivados , Propofol/farmacología , Propofol/efectos de la radiación , Factores de Tiempo , Rayos Ultravioleta , Anestésicos Intravenosos/administración & dosificación , Anestésicos Intravenosos/química , Anestésicos Intravenosos/farmacología , Anestésicos Intravenosos/efectos de la radiación
7.
Math Biosci Eng ; 19(3): 2592-2615, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-35240798

RESUMEN

Neural stem cells (NSCs) offer a potential solution to treating brain tumors. This is because NSCs can circumvent the blood-brain barrier and migrate to areas of damage in the central nervous system, including tumors, stroke, and wound injuries. However, for successful clinical application of NSC treatment, a sufficient number of viable cells must reach the diseased or damaged area(s) in the brain, and evidence suggests that it may be affected by the paths the NSCs take through the brain, as well as the locations of tumors. To study the NSC migration in brain, we develop a mathematical model of therapeutic NSC migration towards brain tumor, that provides a low cost platform to investigate NSC treatment efficacy. Our model is an extension of the model developed in Rockne et al. (PLoS ONE 13, e0199967, 2018) that considers NSC migration in non-tumor bearing naive mouse brain. Here we modify the model in Rockne et al. in three ways: (i) we consider three-dimensional mouse brain geometry, (ii) we add chemotaxis to model the tumor-tropic nature of NSCs into tumor sites, and (iii) we model stochasticity of migration speed and chemosensitivity. The proposed model is used to study migration patterns of NSCs to sites of tumors for different injection strategies, in particular, intranasal and intracerebral delivery. We observe that intracerebral injection results in more NSCs arriving at the tumor site(s), but the relative fraction of NSCs depends on the location of injection relative to the target site(s). On the other hand, intranasal injection results in fewer NSCs at the tumor site, but yields a more even distribution of NSCs within and around the target tumor site(s).


Asunto(s)
Neoplasias Encefálicas , Encéfalo , Glioma , Modelos Neurológicos , Células-Madre Neurales , Animales , Encéfalo/citología , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Movimiento Celular/fisiología , Glioma/patología , Glioma/terapia , Ratones , Células-Madre Neurales/citología , Células-Madre Neurales/trasplante
8.
Food Chem Toxicol ; 158: 112693, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34801652

RESUMEN

The use of opioids during pregnancy has recently dramatically increased presenting major health problems, especially on the developing neonatal nervous system development. Nalufin is considered one of the most used opioid analgesics for treatment of moderate to severe pain, especially during pregnancy. The aim of the present study was firstly to assess the possible neurotoxic effects of nalufin injection during the organogenesis period of chick embryos, and second to investigate the ameliorative effects of selenium as a supplement. Fertilized chicken eggs were in ovo injected with 0.2ml of either nalufin (20 mg/kg egg) or selenium (0.1 mg/kg egg) or both. Nalufin injection resulted in cerebral cortical layer disruption, increase of Caspase-3 immunoexpression and chromatolytic nuclei, degenerated organelles, rarefied cytoplasm and hemorrhage. On the molecular levels, nalufin induced DNA fragmentation, cell cycle arrest and increased the percentage of apoptosis of the neuronal cells. Selenium combined treatment restored the three-layered structure of the cerebral cortex, decreased caspase-3 immuno-expression, improved ultrastructure and recovered cell cycle arrest, decreased apoptosis, and DNA degradation. In conclusion, nalufin treatment during pregnancy imposes great concerns and should not be used during embryonic development, on the other hands, selenium appears to be a promising neuroprotective agent against nalufin-induced neurotoxicity.


Asunto(s)
Apoptosis/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Nalbufina/toxicidad , Neurotoxinas/toxicidad , Selenio/farmacología , Animales , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Embrión de Pollo , Fármacos Neuroprotectores/farmacología
9.
Molecules ; 26(20)2021 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-34684776

RESUMEN

Oxidative stress in aquatic organisms might suppress the immune system and propagate infectious diseases. This study aimed to investigate the protective effect of polyphenolic extracts from spent coffee grounds (SCG) against oxidative stress, induced by H2O2, in C. viridis brain cells, through an in vitro model. Hydrophilic extracts from SCG are rich in quinic, ferulic and caffeic acids and showed antioxidant capacity in DPPH, ORAC and FRAP assays. Furthermore, pretreatment of C. viridis brain cells with the polyphenolic extracts from SCG (230 and 460 µg/mL) for 24 h prior to 100 µM H2O2 exposure (1 h) significantly increased antioxidant enzymes activity (superoxide dismutase and catalase) and reduced lipid peroxidation (measured by MDA levels). These results suggest that polyphenols found in SCG extracts exert an antioxidative protective effect against oxidative stress in C. viridis brain cells by stimulating the activity of SOD and CAT.


Asunto(s)
Antioxidantes/química , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Café/química , Perciformes/metabolismo , Animales , Antioxidantes/aislamiento & purificación , Antioxidantes/farmacología , Encéfalo/citología , Catalasa/metabolismo , Células Cultivadas , Coffea/química , Proteínas de Peces/metabolismo , Explotaciones Pesqueras , Depuradores de Radicales Libres/química , Depuradores de Radicales Libres/aislamiento & purificación , Depuradores de Radicales Libres/farmacología , Peróxido de Hidrógeno/farmacología , Peroxidación de Lípido/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Extractos Vegetales/farmacología , Polifenoles , Superóxido Dismutasa/metabolismo
10.
Nature ; 595(7867): 409-414, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34194038

RESUMEN

Social interactions among animals mediate essential behaviours, including mating, nurturing, and defence1,2. The gut microbiota contribute to social activity in mice3,4, but the gut-brain connections that regulate this complex behaviour and its underlying neural basis are unclear5,6. Here we show that the microbiome modulates neuronal activity in specific brain regions of male mice to regulate canonical stress responses and social behaviours. Social deviation in germ-free and antibiotic-treated mice is associated with elevated levels of the stress hormone corticosterone, which is primarily produced by activation of the hypothalamus-pituitary-adrenal (HPA) axis. Adrenalectomy, antagonism of glucocorticoid receptors, or pharmacological inhibition of corticosterone synthesis effectively corrects social deficits following microbiome depletion. Genetic ablation of glucocorticoid receptors in specific brain regions or chemogenetic inactivation of neurons in the paraventricular nucleus of the hypothalamus that produce corticotrophin-releasing hormone (CRH) reverse social impairments in antibiotic-treated mice. Conversely, specific activation of CRH-expressing neurons in the paraventricular nucleus induces social deficits in mice with a normal microbiome. Via microbiome profiling and in vivo selection, we identify a bacterial species, Enterococcus faecalis, that promotes social activity and reduces corticosterone levels in mice following social stress. These studies suggest that specific gut bacteria can restrain the activation of the HPA axis, and show that the microbiome can affect social behaviours through discrete neuronal circuits that mediate stress responses in the brain.


Asunto(s)
Encéfalo/citología , Encéfalo/fisiología , Microbioma Gastrointestinal/fisiología , Neuronas/metabolismo , Conducta Social , Estrés Psicológico , Animales , Corticosterona/sangre , Hormona Liberadora de Corticotropina/metabolismo , Enterococcus faecalis/metabolismo , Vida Libre de Gérmenes , Glucocorticoides/metabolismo , Hipotálamo/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Glucocorticoides/metabolismo , Transducción de Señal
11.
Neurosci Lett ; 760: 136078, 2021 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-34161823

RESUMEN

Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases, characterized by the accumulation of abnormal tau proteins within neurons and amyloid plaques in the brain parenchyma, which leads to progressive loss of neurons in the brain. While the detailed mechanism of the pathogenesis of AD is still unknown, evidence suggests that mitochondrial dysfunction likely plays a fundamental role in the pathogenesis of this disease. Due to the relevance of mitochondrial alterations in AD, recent works have suggested the therapeutic potential of mitochondrial-targeted lithium. Lithium has been shown to possess neuroprotective and neurotrophic properties that could also be related to the upregulation of mitochondrial function. In the current work, we perform a comprehensive investigation of the significance of mitochondrial dysfunction in AD and pharmacological treatment with lithium as imperative in this pathology, through a brief review of the major findings on the effects of lithium as a therapeutic approach targeting mitochondria in the context of AD.


Asunto(s)
Enfermedad de Alzheimer/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Compuestos de Litio/uso terapéutico , Mitocondrias/efectos de los fármacos , Enfermedad de Alzheimer/patología , Encéfalo/citología , Encéfalo/patología , Línea Celular , Ensayos Clínicos como Asunto , Evaluación Preclínica de Medicamentos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Compuestos de Litio/farmacología , Mitocondrias/metabolismo , Mitocondrias/patología , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos
12.
Methods Mol Biol ; 2277: 133-142, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34080149

RESUMEN

Mitochondria are targets of newly synthesized drugs and being tested for the treatment of various diseases caused or accompanied by disruption of cellular bioenergetics. In drug development, it is necessary to test for drug-induced changes in mitochondrial enzyme activity that may be related to therapeutic or adverse drug effects. Measurement of drug effect on mitochondrial oxygen consumption kinetics and/or protective effects of drugs against calcium-induced inhibition of the mitochondrial respiration can be used for the study mitochondrial toxicity and neuroprotective effects of drugs. Supposing that the drug-induced inhibition of the mitochondrial respiratory rate and/or individual mitochondrial complexes is associated with adverse drug effects, the effects of drugs on mitochondrial respiration in isolated mitochondria allow selection of novel molecules that are relatively safe for mitochondrial toxicity.


Asunto(s)
Evaluación Preclínica de Medicamentos/métodos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Animales , Encéfalo/citología , Evaluación Preclínica de Medicamentos/instrumentación , Complejo I de Transporte de Electrón/metabolismo , Complejo III de Transporte de Electrones/metabolismo , Porcinos
13.
Nat Commun ; 12(1): 2643, 2021 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-33976118

RESUMEN

Prediction of future sensory input based on past sensory information is essential for organisms to effectively adapt their behavior in dynamic environments. Humans successfully predict future stimuli in various natural settings. Yet, it remains elusive how the brain achieves effective prediction despite enormous variations in sensory input rate, which directly affect how fast sensory information can accumulate. We presented participants with acoustic sequences capturing temporal statistical regularities prevalent in nature and investigated neural mechanisms underlying predictive computation using MEG. By parametrically manipulating sequence presentation speed, we tested two hypotheses: neural prediction relies on integrating past sensory information over fixed time periods or fixed amounts of information. We demonstrate that across halved and doubled presentation speeds, predictive information in neural activity stems from integration over fixed amounts of information. Our findings reveal the neural mechanisms enabling humans to robustly predict dynamic stimuli in natural environments despite large sensory input rate variations.


Asunto(s)
Adaptación Fisiológica/fisiología , Algoritmos , Encéfalo/fisiología , Modelos Neurológicos , Red Nerviosa/fisiología , Sensación/fisiología , Estimulación Acústica , Adulto , Encéfalo/citología , Femenino , Humanos , Magnetoencefalografía/métodos , Masculino , Neuronas/fisiología , Desempeño Psicomotor/fisiología , Adulto Joven
14.
J Cereb Blood Flow Metab ; 41(11): 3052-3068, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34027687

RESUMEN

Brain mural cells, including pericytes and vascular smooth muscle cells, are important for vascular development, blood-brain barrier function, and neurovascular coupling, but the molecular characteristics of human brain mural cells are incompletely characterized. Single cell RNA-sequencing (scRNA-seq) is increasingly being applied to assess cellular diversity in the human brain, but the scarcity of mural cells in whole brain samples has limited their molecular profiling. Here, we leverage the combined power of multiple independent human brain scRNA-seq datasets to build a transcriptomic database of human brain mural cells. We use this combined dataset to determine human-mouse species differences in mural cell transcriptomes, culture-induced dedifferentiation of human brain pericytes, and human mural cell organotypicity, with several key findings validated by RNA fluorescence in situ hybridization. Together, this work improves knowledge regarding the molecular constituents of human brain mural cells, serves as a resource for hypothesis generation in understanding brain mural cell function, and will facilitate comparative evaluation of animal and in vitro models.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/citología , Miocitos del Músculo Liso/metabolismo , Pericitos/metabolismo , Transcriptoma/genética , Animales , Barrera Hematoencefálica/fisiología , Humanos , Hibridación Fluorescente in Situ/métodos , Medicina Integrativa/métodos , Ratones , Acoplamiento Neurovascular/fisiología , ARN Citoplasmático Pequeño/genética , RNA-Seq/métodos
15.
Life Sci ; 278: 119614, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-34022200

RESUMEN

AIMS: Sodium butyrate (SB) is a major product of gut microbiota with signaling activity in the human body. It has become a dietary supplement in the treatment of intestinal disorders. However, the toxic effect of overdosed SB and treatment strategy remain unknown. The two issues are addressed in current study. MATERIALS AND METHODS: SB (0.3-2.5 g/kg) was administrated through a single peritoneal injection in mice. The core body temperature and mitochondrial function in the brown adipose tissue and brain were monitored. Pharmacodynamics, targeted metabolomics, electron microscope, oxygen consumption rate and gene knockdown were employed to dissect the mechanism for the toxic effect. KEY FINDINGS: The temperature was reduced by SB (1.2-2.5 g/kg) in a dose-dependent manner in mice for 2-4 h. In the brain, the effect was associated with SB elevation and neurotransmitter reduction. Metabolites changes were seen in the glycolysis, TCA cycle and pentose phosphate pathways. Adenine nucleotide translocase (ANT) was activated by butyrate for proton transportation leading to a transient potential collapse through proton leak. The SB activity was attenuated by ANT inhibition from gene knockdown or pharmacological blocker. ROS was elevated by SB for the increased ANT activity in proton leak in Neuro-2a. SIGNIFICANCE: Excessive SB generated an immediate and reversible toxic effect for inhibition of body temperature through transient mitochondrial dysfunction in the brain. The mechanism was quick activation of ANT proteins for potential collapse in mitochondria. ROS may be a factor in the ANT activation by SB.


Asunto(s)
Ácido Butírico/farmacología , Antagonistas de los Receptores Histamínicos/farmacología , Mitocondrias/efectos de los fármacos , Neuronas/efectos de los fármacos , Animales , Temperatura Corporal/efectos de los fármacos , Encéfalo/citología , Encéfalo/efectos de los fármacos , Ácido Butírico/administración & dosificación , Ácido Butírico/efectos adversos , Células Cultivadas , Relación Dosis-Respuesta a Droga , Antagonistas de los Receptores Histamínicos/administración & dosificación , Antagonistas de los Receptores Histamínicos/efectos adversos , Masculino , Ratones Endogámicos C57BL , Mitocondrias/metabolismo , Neuronas/metabolismo , Protones
16.
Commun Biol ; 4(1): 586, 2021 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-33990685

RESUMEN

Oxytocin (OXT) and arginine vasopressin (AVP) support a broad range of behaviors and homeostatic functions including sex-specific and context-appropriate social behaviors. Although the alterations of these systems have been linked with social-related disorders such as autism spectrum disorder, their formation and developmental dynamics remain largely unknown. Using novel brain clearing techniques and 3D imaging, we have reconstructed the specification of oxytocinergic and vasopressinergic circuits in the developing mouse brain with unprecedented cellular resolution. A systematic quantification indicates that OXT and AVP neurons in the hypothalamus display distinctive developmental dynamics and high cellular plasticity from embryonic to early postnatal stages. Our findings reveal new insights into the specification and consolidation of neuropeptidergic systems in the developing CNS.


Asunto(s)
Encéfalo/citología , Encéfalo/metabolismo , Hipotálamo/metabolismo , Vías Nerviosas , Oxitocina/metabolismo , Vasopresinas/metabolismo , Animales , Femenino , Hipotálamo/citología , Masculino , Ratones , Ratones Endogámicos ICR , Conducta Social
17.
Sci Rep ; 11(1): 10411, 2021 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-34002002

RESUMEN

TRPM4 is a calcium-activated non-selective monovalent cation channel implicated in diseases such as stroke. Lack of potent and selective inhibitors remains a major challenge for studying TRPM4. Using a polypeptide from rat TRPM4, we have generated a polyclonal antibody M4P which could alleviate reperfusion injury in a rat model of stroke. Here, we aim to develop a monoclonal antibody that could block human TRPM4 channel. Two mouse monoclonal antibodies M4M and M4M1 were developed to target an extracellular epitope of human TRPM4. Immunohistochemistry and western blot were used to characterize the binding of these antibodies to human TRPM4. Potency of inhibition was compared using electrophysiological methods. We further evaluated the therapeutic potential on a rat model of middle cerebral artery occlusion. Both M4M and M4M1 could bind to human TRPM4 channel on the surface of live cells. Prolonged incubation with TRPM4 blocking antibody internalized surface TRPM4. Comparing to M4M1, M4M is more effective in blocking human TRPM4 channel. In human brain microvascular endothelial cells, M4M successfully inhibited TRPM4 current and ameliorated hypoxia-induced cell swelling. Using wild type rats, neither antibody demonstrated therapeutic potential on stroke. Human TRPM4 channel can be blocked by a monoclonal antibody M4M targeting a key antigenic sequence. For future clinical translation, the antibody needs to be humanized and a transgenic animal carrying human TRPM4 sequence is required for in vivo characterizing its therapeutic potential.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Daño por Reperfusión/prevención & control , Canales Catiónicos TRPM/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales/uso terapéutico , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/patología , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Células HEK293 , Humanos , Infarto de la Arteria Cerebral Media/complicaciones , Masculino , Técnicas de Placa-Clamp , Ratas , Daño por Reperfusión/etiología , Daño por Reperfusión/patología , Canales Catiónicos TRPM/metabolismo
18.
Cell Rep ; 35(2): 108985, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852843

RESUMEN

Decreased cognitive performance is a hallmark of brain aging, but the underlying mechanisms and potential therapeutic avenues remain poorly understood. Recent studies have revealed health-protective and lifespan-extending effects of dietary spermidine, a natural autophagy-promoting polyamine. Here, we show that dietary spermidine passes the blood-brain barrier in mice and increases hippocampal eIF5A hypusination and mitochondrial function. Spermidine feeding in aged mice affects behavior in homecage environment tasks, improves spatial learning, and increases hippocampal respiratory competence. In a Drosophila aging model, spermidine boosts mitochondrial respiratory capacity, an effect that requires the autophagy regulator Atg7 and the mitophagy mediators Parkin and Pink1. Neuron-specific Pink1 knockdown abolishes spermidine-induced improvement of olfactory associative learning. This suggests that the maintenance of mitochondrial and autophagic function is essential for enhanced cognition by spermidine feeding. Finally, we show large-scale prospective data linking higher dietary spermidine intake with a reduced risk for cognitive impairment in humans.


Asunto(s)
Envejecimiento/genética , Proteína 7 Relacionada con la Autofagia/genética , Disfunción Cognitiva/genética , Suplementos Dietéticos , Proteínas Quinasas/genética , Espermidina/farmacología , Ubiquitina-Proteína Ligasas/genética , Envejecimiento/metabolismo , Animales , Proteína 7 Relacionada con la Autofagia/metabolismo , Encéfalo/citología , Encéfalo/efectos de los fármacos , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Cognición/efectos de los fármacos , Cognición/fisiología , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/fisiopatología , Disfunción Cognitiva/prevención & control , Drosophila melanogaster/efectos de los fármacos , Drosophila melanogaster/genética , Drosophila melanogaster/crecimiento & desarrollo , Drosophila melanogaster/metabolismo , Femenino , Regulación de la Expresión Génica , Humanos , Aprendizaje/efectos de los fármacos , Aprendizaje/fisiología , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/genética , Mitocondrias/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Fosforilación Oxidativa/efectos de los fármacos , Proteínas Quinasas/metabolismo , Transducción de Señal , Memoria Espacial/efectos de los fármacos , Memoria Espacial/fisiología , Ubiquitina-Proteína Ligasas/metabolismo
19.
Int J Mol Sci ; 22(5)2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33800815

RESUMEN

In the last decade, different research groups in the academic setting have developed induced pluripotent stem cell-based protocols to generate three-dimensional, multicellular, neural organoids. Their use to model brain biology, early neural development, and human diseases has provided new insights into the pathophysiology of neuropsychiatric and neurological disorders, including microcephaly, autism, Parkinson's disease, and Alzheimer's disease. However, the adoption of organoid technology for large-scale drug screening in the industry has been hampered by challenges with reproducibility, scalability, and translatability to human disease. Potential technical solutions to expand their use in drug discovery pipelines include Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) to create isogenic models, single-cell RNA sequencing to characterize the model at a cellular level, and machine learning to analyze complex data sets. In addition, high-content imaging, automated liquid handling, and standardized assays represent other valuable tools toward this goal. Though several open issues still hamper the full implementation of the organoid technology outside academia, rapid progress in this field will help to prompt its translation toward large-scale drug screening for neurological disorders.


Asunto(s)
Descubrimiento de Drogas/métodos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Organoides/efectos de los fármacos , Animales , Automatización , Encéfalo/citología , Sistemas CRISPR-Cas , Técnicas de Cultivo de Célula , Colágeno , Combinación de Medicamentos , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/organización & administración , Predicción , Ensayos Analíticos de Alto Rendimiento , Humanos , Células Madre Pluripotentes Inducidas/citología , Laminina , Aprendizaje Automático , Microscopía/métodos , Enfermedades del Sistema Nervioso/patología , Proteoglicanos , RNA-Seq , Reproducibilidad de los Resultados , Análisis de la Célula Individual
20.
Nat Methods ; 18(4): 374-377, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33795878

RESUMEN

SNT is an end-to-end framework for neuronal morphometry and whole-brain connectomics that supports tracing, proof-editing, visualization, quantification and modeling of neuroanatomy. With an open architecture, a large user base, community-based documentation, support for complex imagery and several model organisms, SNT is a flexible resource for the broad neuroscience community. SNT is both a desktop application and multi-language scripting library, and it is available through the Fiji distribution of ImageJ.


Asunto(s)
Encéfalo/anatomía & histología , Neuronas/citología , Animales , Encéfalo/citología , Conectoma , Humanos , Análisis de la Célula Individual
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA