Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 13.436
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 39: 369-393, 2021 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-33561366

RESUMEN

Classically, skin was considered a mere structural barrier protecting organisms from a diversity of environmental insults. In recent decades, the cutaneous immune system has become recognized as a complex immunologic barrier involved in both antimicrobial immunity and homeostatic processes like wound healing. To sense a variety of chemical, mechanical, and thermal stimuli, the skin harbors one of the most sophisticated sensory networks in the body. However, recent studies suggest that the cutaneous nervous system is highly integrated with the immune system to encode specific sensations into evolutionarily conserved protective behaviors. In addition to directly sensing pathogens, neurons employ novel neuroimmune mechanisms to provide host immunity. Therefore, given that sensation underlies various physiologies through increasingly complex reflex arcs, a much more dynamic picture is emerging of the skin as a truly systemic organ with highly coordinated physical, immunologic, and neural functions in barrier immunology.


Asunto(s)
Sistema Inmunológico , Neuroinmunomodulación , Animales , Humanos , Sistema Nervioso
2.
Cell ; 187(8): 1971-1989.e16, 2024 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-38521060

RESUMEN

Amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD) share many clinical, pathological, and genetic features, but a detailed understanding of their associated transcriptional alterations across vulnerable cortical cell types is lacking. Here, we report a high-resolution, comparative single-cell molecular atlas of the human primary motor and dorsolateral prefrontal cortices and their transcriptional alterations in sporadic and familial ALS and FTLD. By integrating transcriptional and genetic information, we identify known and previously unidentified vulnerable populations in cortical layer 5 and show that ALS- and FTLD-implicated motor and spindle neurons possess a virtually indistinguishable molecular identity. We implicate potential disease mechanisms affecting these cell types as well as non-neuronal drivers of pathogenesis. Finally, we show that neuron loss in cortical layer 5 tracks more closely with transcriptional identity rather than cellular morphology and extends beyond previously reported vulnerable cell types.


Asunto(s)
Esclerosis Amiotrófica Lateral , Degeneración Lobar Frontotemporal , Corteza Prefrontal , Animales , Humanos , Ratones , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Esclerosis Amiotrófica Lateral/patología , Demencia Frontotemporal/genética , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Degeneración Lobar Frontotemporal/patología , Perfilación de la Expresión Génica , Neuronas/metabolismo , Corteza Prefrontal/metabolismo , Corteza Prefrontal/patología , Análisis de Expresión Génica de una Sola Célula
3.
Cell ; 187(7): 1785-1800.e16, 2024 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-38552614

RESUMEN

To understand biological processes, it is necessary to reveal the molecular heterogeneity of cells by gaining access to the location and interaction of all biomolecules. Significant advances were achieved by super-resolution microscopy, but such methods are still far from reaching the multiplexing capacity of proteomics. Here, we introduce secondary label-based unlimited multiplexed DNA-PAINT (SUM-PAINT), a high-throughput imaging method that is capable of achieving virtually unlimited multiplexing at better than 15 nm resolution. Using SUM-PAINT, we generated 30-plex single-molecule resolved datasets in neurons and adapted omics-inspired analysis for data exploration. This allowed us to reveal the complexity of synaptic heterogeneity, leading to the discovery of a distinct synapse type. We not only provide a resource for researchers, but also an integrated acquisition and analysis workflow for comprehensive spatial proteomics at single-protein resolution.


Asunto(s)
Proteómica , Imagen Individual de Molécula , ADN , Microscopía Fluorescente/métodos , Neuronas , Proteínas
4.
Cell ; 186(18): 3826-3844.e26, 2023 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-37536338

RESUMEN

Previous studies have identified topologically associating domains (TADs) as basic units of genome organization. We present evidence of a previously unreported level of genome folding, where distant TAD pairs, megabases apart, interact to form meta-domains. Within meta-domains, gene promoters and structural intergenic elements present in distant TADs are specifically paired. The associated genes encode neuronal determinants, including those engaged in axonal guidance and adhesion. These long-range associations occur in a large fraction of neurons but support transcription in only a subset of neurons. Meta-domains are formed by diverse transcription factors that are able to pair over long and flexible distances. We present evidence that two such factors, GAF and CTCF, play direct roles in this process. The relative simplicity of higher-order meta-domain interactions in Drosophila, compared with those previously described in mammals, allowed the demonstration that genomes can fold into highly specialized cell-type-specific scaffolds that enable megabase-scale regulatory associations.


Asunto(s)
Cromosomas de Insectos , Drosophila , Animales , Cromatina/genética , Empaquetamiento del ADN , Drosophila/genética , Mamíferos/genética , Neurogénesis , Neuronas , Factores de Transcripción , Proteínas de Drosophila , Genoma de los Insectos , Regulación de la Expresión Génica
5.
Cell ; 186(3): 607-620.e17, 2023 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-36640762

RESUMEN

Tissue immunity and responses to injury depend on the coordinated action and communication among physiological systems. Here, we show that, upon injury, adaptive responses to the microbiota directly promote sensory neuron regeneration. At homeostasis, tissue-resident commensal-specific T cells colocalize with sensory nerve fibers within the dermis, express a transcriptional program associated with neuronal interaction and repair, and promote axon growth and local nerve regeneration following injury. Mechanistically, our data reveal that the cytokine interleukin-17A (IL-17A) released by commensal-specific Th17 cells upon injury directly signals to sensory neurons via IL-17 receptor A, the transcription of which is specifically upregulated in injured neurons. Collectively, our work reveals that in the context of tissue damage, preemptive immunity to the microbiota can rapidly bridge biological systems by directly promoting neuronal repair, while also identifying IL-17A as a major determinant of this fundamental process.


Asunto(s)
Interleucina-17 , Microbiota , Regeneración Nerviosa , Células Th17 , Axones , Regeneración Nerviosa/fisiología , Células Receptoras Sensoriales , Animales , Ratones , Células Th17/citología
6.
Cell ; 186(9): 2002-2017.e21, 2023 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-37080201

RESUMEN

Paired mapping of single-cell gene expression and electrophysiology is essential to understand gene-to-function relationships in electrogenic tissues. Here, we developed in situ electro-sequencing (electro-seq) that combines flexible bioelectronics with in situ RNA sequencing to stably map millisecond-timescale electrical activity and profile single-cell gene expression from the same cells across intact biological networks, including cardiac and neural patches. When applied to human-induced pluripotent stem-cell-derived cardiomyocyte patches, in situ electro-seq enabled multimodal in situ analysis of cardiomyocyte electrophysiology and gene expression at the cellular level, jointly defining cell states and developmental trajectories. Using machine-learning-based cross-modal analysis, in situ electro-seq identified gene-to-electrophysiology relationships throughout cardiomyocyte development and accurately reconstructed the evolution of gene expression profiles based on long-term stable electrical measurements. In situ electro-seq could be applicable to create spatiotemporal multimodal maps in electrogenic tissues, potentiating the discovery of cell types and gene programs responsible for electrophysiological function and dysfunction.


Asunto(s)
Electrónica , Análisis de Secuencia de ARN , Humanos , Diferenciación Celular , Células Madre Pluripotentes Inducidas/fisiología , Miocitos Cardíacos/metabolismo , Análisis de la Célula Individual , Transcriptoma , Electrónica/métodos
7.
Cell ; 186(4): 803-820.e25, 2023 02 16.
Artículo en Inglés | MEDLINE | ID: mdl-36738734

RESUMEN

Complex diseases often involve the interplay between genetic and environmental factors. Charcot-Marie-Tooth type 2 neuropathies (CMT2) are a group of genetically heterogeneous disorders, in which similar peripheral neuropathology is inexplicably caused by various mutated genes. Their possible molecular links remain elusive. Here, we found that upon environmental stress, many CMT2-causing mutant proteins adopt similar properties by entering stress granules (SGs), where they aberrantly interact with G3BP and integrate into SG pathways. For example, glycyl-tRNA synthetase (GlyRS) is translocated from the cytoplasm into SGs upon stress, where the mutant GlyRS perturbs the G3BP-centric SG network by aberrantly binding to G3BP. This disrupts SG-mediated stress responses, leading to increased stress vulnerability in motoneurons. Disrupting this aberrant interaction rescues SG abnormalities and alleviates motor deficits in CMT2D mice. These findings reveal a stress-dependent molecular link across diverse CMT2 mutants and provide a conceptual framework for understanding genetic heterogeneity in light of environmental stress.


Asunto(s)
Enfermedad de Charcot-Marie-Tooth , Proteínas con Motivos de Reconocimiento de ARN , Gránulos de Estrés , Animales , Ratones , Enfermedad de Charcot-Marie-Tooth/genética , Enfermedad de Charcot-Marie-Tooth/metabolismo , Enfermedad de Charcot-Marie-Tooth/patología , Citoplasma , Neuronas Motoras , Proteínas con Motivos de Reconocimiento de ARN/metabolismo
8.
Cell ; 185(2): 328-344.e26, 2022 01 20.
Artículo en Inglés | MEDLINE | ID: mdl-35063074

RESUMEN

Locomotion is a complex behavior required for animal survival. Vertebrate locomotion depends on spinal interneurons termed the central pattern generator (CPG), which generates activity responsible for the alternation of flexor and extensor muscles and the left and right side of the body. It is unknown whether multiple or a single neuronal type is responsible for the control of mammalian locomotion. Here, we show that ventral spinocerebellar tract neurons (VSCTs) drive generation and maintenance of locomotor behavior in neonatal and adult mice. Using mouse genetics, physiological, anatomical, and behavioral assays, we demonstrate that VSCTs exhibit rhythmogenic properties and neuronal circuit connectivity consistent with their essential role in the locomotor CPG. Importantly, optogenetic activation and chemogenetic silencing reveals that VSCTs are necessary and sufficient for locomotion. These findings identify VSCTs as critical components for mammalian locomotion and provide a paradigm shift in our understanding of neural control of complex behaviors.


Asunto(s)
Locomoción/fisiología , Mamíferos/fisiología , Neuronas Motoras/citología , Tractos Espinocerebelares/citología , Animales , Axones/fisiología , Fenómenos Electrofisiológicos , Uniones Comunicantes/metabolismo , Silenciador del Gen , Ácido Glutámico/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de Homeodominio/metabolismo , Interneuronas/fisiología , Vértebras Lumbares/metabolismo , Ratones , Propiocepción , Natación , Sinapsis/fisiología , Factores de Transcripción/metabolismo
9.
Cell ; 185(22): 4170-4189.e20, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36240781

RESUMEN

Nociceptive pain is a hallmark of many chronic inflammatory conditions including inflammatory bowel diseases (IBDs); however, whether pain-sensing neurons influence intestinal inflammation remains poorly defined. Employing chemogenetic silencing, adenoviral-mediated colon-specific silencing, and pharmacological ablation of TRPV1+ nociceptors, we observed more severe inflammation and defective tissue-protective reparative processes in a murine model of intestinal damage and inflammation. Disrupted nociception led to significant alterations in the intestinal microbiota and a transmissible dysbiosis, while mono-colonization of germ-free mice with Gram+Clostridium spp. promoted intestinal tissue protection through a nociceptor-dependent pathway. Mechanistically, disruption of nociception resulted in decreased levels of substance P, and therapeutic delivery of substance P promoted tissue-protective effects exerted by TRPV1+ nociceptors in a microbiota-dependent manner. Finally, dysregulated nociceptor gene expression was observed in intestinal biopsies from IBD patients. Collectively, these findings indicate an evolutionarily conserved functional link between nociception, the intestinal microbiota, and the restoration of intestinal homeostasis.


Asunto(s)
Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Ratones , Animales , Microbioma Gastrointestinal/fisiología , Nociceptores/fisiología , Sustancia P , Disbiosis , Inflamación
10.
Cell ; 185(22): 4190-4205.e25, 2022 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-36243004

RESUMEN

Neuroepithelial crosstalk is critical for gut physiology. However, the mechanisms by which sensory neurons communicate with epithelial cells to mediate gut barrier protection at homeostasis and during inflammation are not well understood. Here, we find that Nav1.8+CGRP+ nociceptor neurons are juxtaposed with and signal to intestinal goblet cells to drive mucus secretion and gut protection. Nociceptor ablation led to decreased mucus thickness and dysbiosis, while chemogenetic nociceptor activation or capsaicin treatment induced mucus growth. Mouse and human goblet cells expressed Ramp1, receptor for the neuropeptide CGRP. Nociceptors signal via the CGRP-Ramp1 pathway to induce rapid goblet cell emptying and mucus secretion. Notably, commensal microbes activated nociceptors to control homeostatic CGRP release. In the absence of nociceptors or epithelial Ramp1, mice showed increased epithelial stress and susceptibility to colitis. Conversely, CGRP administration protected nociceptor-ablated mice against colitis. Our findings demonstrate a neuron-goblet cell axis that orchestrates gut mucosal barrier protection.


Asunto(s)
Colitis , Células Caliciformes , Ratones , Humanos , Animales , Células Caliciformes/metabolismo , Nociceptores/metabolismo , Péptido Relacionado con Gen de Calcitonina/metabolismo , Colitis/metabolismo , Moco/metabolismo , Proteína 1 Modificadora de la Actividad de Receptores/metabolismo
11.
Cell ; 185(16): 2899-2917.e31, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35914528

RESUMEN

Glioblastomas are incurable tumors infiltrating the brain. A subpopulation of glioblastoma cells forms a functional and therapy-resistant tumor cell network interconnected by tumor microtubes (TMs). Other subpopulations appear unconnected, and their biological role remains unclear. Here, we demonstrate that whole-brain colonization is fueled by glioblastoma cells that lack connections with other tumor cells and astrocytes yet receive synaptic input from neurons. This subpopulation corresponds to neuronal and neural-progenitor-like tumor cell states, as defined by single-cell transcriptomics, both in mouse models and in the human disease. Tumor cell invasion resembled neuronal migration mechanisms and adopted a Lévy-like movement pattern of probing the environment. Neuronal activity induced complex calcium signals in glioblastoma cells followed by the de novo formation of TMs and increased invasion speed. Collectively, superimposing molecular and functional single-cell data revealed that neuronal mechanisms govern glioblastoma cell invasion on multiple levels. This explains how glioblastoma's dissemination and cellular heterogeneity are closely interlinked.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Animales , Astrocitos/patología , Encéfalo/patología , Neoplasias Encefálicas/patología , Glioblastoma/genética , Glioblastoma/patología , Humanos , Ratones , Invasividad Neoplásica , Neuronas/fisiología
12.
Cell ; 184(2): 441-459.e25, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33333021

RESUMEN

Barrier tissue immune responses are regulated in part by nociceptors. Nociceptor ablation alters local immune responses at peripheral sites and within draining lymph nodes (LNs). The mechanisms and significance of nociceptor-dependent modulation of LN function are unknown. Using high-resolution imaging, viral tracing, single-cell transcriptomics, and optogenetics, we identified and functionally tested a sensory neuro-immune circuit that is responsive to lymph-borne inflammatory signals. Transcriptomics profiling revealed that multiple sensory neuron subsets, predominantly peptidergic nociceptors, innervate LNs, distinct from those innervating surrounding skin. To uncover LN-resident cells that may interact with LN-innervating sensory neurons, we generated a LN single-cell transcriptomics atlas and nominated nociceptor target populations and interaction modalities. Optogenetic stimulation of LN-innervating sensory fibers triggered rapid transcriptional changes in the predicted interacting cell types, particularly endothelium, stromal cells, and innate leukocytes. Thus, a unique population of sensory neurons monitors peripheral LNs and may locally regulate gene expression.


Asunto(s)
Inmunomodulación , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/inervación , Células Receptoras Sensoriales/inmunología , Potenciales de Acción , Animales , Inflamación/patología , Ratones , Nociceptores/metabolismo , Optogenética , Péptidos/metabolismo , Piel/inervación , Sistema Nervioso Simpático/fisiología , Receptores Toll-Like/agonistas , Receptores Toll-Like/metabolismo
13.
Cell ; 184(15): 4048-4063.e32, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34233165

RESUMEN

Microglia, the resident immune cells of the brain, have emerged as crucial regulators of synaptic refinement and brain wiring. However, whether the remodeling of distinct synapse types during development is mediated by specialized microglia is unknown. Here, we show that GABA-receptive microglia selectively interact with inhibitory cortical synapses during a critical window of mouse postnatal development. GABA initiates a transcriptional synapse remodeling program within these specialized microglia, which in turn sculpt inhibitory connectivity without impacting excitatory synapses. Ablation of GABAB receptors within microglia impairs this process and leads to behavioral abnormalities. These findings demonstrate that brain wiring relies on the selective communication between matched neuronal and glial cell types.


Asunto(s)
Microglía/metabolismo , Inhibición Neural/fisiología , Ácido gamma-Aminobutírico/metabolismo , Animales , Animales Recién Nacidos , Conducta Animal , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Parvalbúminas/metabolismo , Fenotipo , Receptores de GABA-B/metabolismo , Sinapsis/fisiología , Transcripción Genética
14.
Cell ; 184(12): 3222-3241.e26, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34004146

RESUMEN

The isocortex and hippocampal formation (HPF) in the mammalian brain play critical roles in perception, cognition, emotion, and learning. We profiled ∼1.3 million cells covering the entire adult mouse isocortex and HPF and derived a transcriptomic cell-type taxonomy revealing a comprehensive repertoire of glutamatergic and GABAergic neuron types. Contrary to the traditional view of HPF as having a simpler cellular organization, we discover a complete set of glutamatergic types in HPF homologous to all major subclasses found in the six-layered isocortex, suggesting that HPF and the isocortex share a common circuit organization. We also identify large-scale continuous and graded variations of cell types along isocortical depth, across the isocortical sheet, and in multiple dimensions in hippocampus and subiculum. Overall, our study establishes a molecular architecture of the mammalian isocortex and hippocampal formation and begins to shed light on its underlying relationship with the development, evolution, connectivity, and function of these two brain structures.


Asunto(s)
Hipocampo/citología , Neocórtex/citología , Transcriptoma/genética , Animales , Neuronas GABAérgicas/citología , Neuronas GABAérgicas/metabolismo , Ácido Glutámico/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos
15.
Cell ; 184(16): 4329-4347.e23, 2021 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-34237253

RESUMEN

We have produced gene expression profiles of all 302 neurons of the C. elegans nervous system that match the single-cell resolution of its anatomy and wiring diagram. Our results suggest that individual neuron classes can be solely identified by combinatorial expression of specific gene families. For example, each neuron class expresses distinct codes of ∼23 neuropeptide genes and ∼36 neuropeptide receptors, delineating a complex and expansive "wireless" signaling network. To demonstrate the utility of this comprehensive gene expression catalog, we used computational approaches to (1) identify cis-regulatory elements for neuron-specific gene expression and (2) reveal adhesion proteins with potential roles in process placement and synaptic specificity. Our expression data are available at https://cengen.org and can be interrogated at the web application CengenApp. We expect that this neuron-specific directory of gene expression will spur investigations of underlying mechanisms that define anatomy, connectivity, and function throughout the C. elegans nervous system.


Asunto(s)
Caenorhabditis elegans/metabolismo , Sistema Nervioso/metabolismo , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Colorantes Fluorescentes/metabolismo , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros , Larva/metabolismo , Neuronas/metabolismo , Neuropéptidos/genética , Neuropéptidos/metabolismo , Motivos de Nucleótidos/genética , RNA-Seq , Secuencias Reguladoras de Ácidos Nucleicos/genética , Transducción de Señal/genética , Factores de Transcripción/metabolismo , Transcripción Genética
16.
Cell ; 184(4): 912-930.e20, 2021 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-33571430

RESUMEN

Electrical stimulation is a promising tool for modulating brain networks. However, it is unclear how stimulation interacts with neural patterns underlying behavior. Specifically, how might external stimulation that is not sensitive to the state of ongoing neural dynamics reliably augment neural processing and improve function? Here, we tested how low-frequency epidural alternating current stimulation (ACS) in non-human primates recovering from stroke interacted with task-related activity in perilesional cortex and affected grasping. We found that ACS increased co-firing within task-related ensembles and improved dexterity. Using a neural network model, we found that simulated ACS drove ensemble co-firing and enhanced propagation of neural activity through parts of the network with impaired connectivity, suggesting a mechanism to link increased co-firing to enhanced dexterity. Together, our results demonstrate that ACS restores neural processing in impaired networks and improves dexterity following stroke. More broadly, these results demonstrate approaches to optimize stimulation to target neural dynamics.


Asunto(s)
Potenciales de Acción/fisiología , Accidente Cerebrovascular/fisiopatología , Animales , Conducta Animal/fisiología , Fenómenos Biomecánicos/fisiología , Estimulación Eléctrica , Haplorrinos , Corteza Motora/fisiopatología , Redes Neurales de la Computación , Neuronas/fisiología , Análisis y Desempeño de Tareas , Factores de Tiempo
17.
Cell ; 184(24): 5869-5885.e25, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34758294

RESUMEN

RTN4-binding proteins were widely studied as "NoGo" receptors, but their physiological interactors and roles remain elusive. Similarly, BAI adhesion-GPCRs were associated with numerous activities, but their ligands and functions remain unclear. Using unbiased approaches, we observed an unexpected convergence: RTN4 receptors are high-affinity ligands for BAI adhesion-GPCRs. A single thrombospondin type 1-repeat (TSR) domain of BAIs binds to the leucine-rich repeat domain of all three RTN4-receptor isoforms with nanomolar affinity. In the 1.65 Å crystal structure of the BAI1/RTN4-receptor complex, C-mannosylation of tryptophan and O-fucosylation of threonine in the BAI TSR-domains creates a RTN4-receptor/BAI interface shaped by unusual glycoconjugates that enables high-affinity interactions. In human neurons, RTN4 receptors regulate dendritic arborization, axonal elongation, and synapse formation by differential binding to glial versus neuronal BAIs, thereby controlling neural network activity. Thus, BAI binding to RTN4/NoGo receptors represents a receptor-ligand axis that, enabled by rare post-translational modifications, controls development of synaptic circuits.


Asunto(s)
Inhibidores de la Angiogénesis/metabolismo , Encéfalo/metabolismo , Neurogénesis , Neuronas/metabolismo , Proteínas Nogo/metabolismo , Receptores Nogo/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Adipoquinas/metabolismo , Secuencia de Aminoácidos , Animales , Axones/metabolismo , Adhesión Celular , Moléculas de Adhesión Celular Neuronal/metabolismo , Complemento C1q/metabolismo , Dendritas/metabolismo , Glicosilación , Células HEK293 , Células Madre Embrionarias Humanas/metabolismo , Humanos , Ligandos , Ratones Endogámicos C57BL , Red Nerviosa/metabolismo , Polisacáridos/metabolismo , Unión Proteica , Dominios Proteicos , Eliminación de Secuencia , Sinapsis/metabolismo , Transmisión Sináptica/fisiología
18.
Cell ; 184(2): 489-506.e26, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33338423

RESUMEN

Single-cell transcriptomics has been widely applied to classify neurons in the mammalian brain, while systems neuroscience has historically analyzed the encoding properties of cortical neurons without considering cell types. Here we examine how specific transcriptomic types of mouse prefrontal cortex (PFC) projection neurons relate to axonal projections and encoding properties across multiple cognitive tasks. We found that most types projected to multiple targets, and most targets received projections from multiple types, except PFC→PAG (periaqueductal gray). By comparing Ca2+ activity of the molecularly homogeneous PFC→PAG type against two heterogeneous classes in several two-alternative choice tasks in freely moving mice, we found that all task-related signals assayed were qualitatively present in all examined classes. However, PAG-projecting neurons most potently encoded choice in cued tasks, whereas contralateral PFC-projecting neurons most potently encoded reward context in an uncued task. Thus, task signals are organized redundantly, but with clear quantitative biases across cells of specific molecular-anatomical characteristics.


Asunto(s)
Cognición/fisiología , Neuronas/fisiología , Corteza Prefrontal/fisiología , Análisis y Desempeño de Tareas , Animales , Calcio/metabolismo , Conducta de Elección , Señales (Psicología) , Imagenología Tridimensional , Integrasas/metabolismo , Ratones Endogámicos C57BL , Odorantes , Optogenética , Sustancia Gris Periacueductal/fisiología , Recompensa , Análisis de la Célula Individual , Transcriptoma/genética
19.
Cell ; 184(21): 5465-5481.e16, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34582787

RESUMEN

In vivo cell fate conversions have emerged as potential regeneration-based therapeutics for injury and disease. Recent studies reported that ectopic expression or knockdown of certain factors can convert resident astrocytes into functional neurons with high efficiency, region specificity, and precise connectivity. However, using stringent lineage tracing in the mouse brain, we show that the presumed astrocyte-converted neurons are actually endogenous neurons. AAV-mediated co-expression of NEUROD1 and a reporter specifically and efficiently induces reporter-labeled neurons. However, these neurons cannot be traced retrospectively to quiescent or reactive astrocytes using lineage-mapping strategies. Instead, through a retrograde labeling approach, our results reveal that endogenous neurons are the source for these viral-reporter-labeled neurons. Similarly, despite efficient knockdown of PTBP1 in vivo, genetically traced resident astrocytes were not converted into neurons. Together, our results highlight the requirement of lineage-tracing strategies, which should be broadly applied to studies of cell fate conversions in vivo.


Asunto(s)
Astrocitos/citología , Diferenciación Celular , Linaje de la Célula , Neuronas/citología , Animales , Astrocitos/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Encéfalo/patología , Lesiones Encefálicas/patología , Línea Celular Tumoral , Reprogramación Celular , Dependovirus/metabolismo , Regulación hacia Abajo , Regulación de la Expresión Génica , Genes Reporteros , Proteína Ácida Fibrilar de la Glía/genética , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Integrasas/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuronas/metabolismo , Proteína de Unión al Tracto de Polipirimidina/metabolismo , Regiones Promotoras Genéticas/genética , Factores de Transcripción/metabolismo
20.
Cell ; 184(24): 5932-5949.e15, 2021 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-34798069

RESUMEN

Anosmia, the loss of smell, is a common and often the sole symptom of COVID-19. The onset of the sequence of pathobiological events leading to olfactory dysfunction remains obscure. Here, we have developed a postmortem bedside surgical procedure to harvest endoscopically samples of respiratory and olfactory mucosae and whole olfactory bulbs. Our cohort of 85 cases included COVID-19 patients who died a few days after infection with SARS-CoV-2, enabling us to catch the virus while it was still replicating. We found that sustentacular cells are the major target cell type in the olfactory mucosa. We failed to find evidence for infection of olfactory sensory neurons, and the parenchyma of the olfactory bulb is spared as well. Thus, SARS-CoV-2 does not appear to be a neurotropic virus. We postulate that transient insufficient support from sustentacular cells triggers transient olfactory dysfunction in COVID-19. Olfactory sensory neurons would become affected without getting infected.


Asunto(s)
Autopsia/métodos , COVID-19/mortalidad , COVID-19/virología , Bulbo Olfatorio/virología , Mucosa Olfatoria/virología , Mucosa Respiratoria/virología , Anciano , Anosmia , COVID-19/fisiopatología , Endoscopía/métodos , Femenino , Glucuronosiltransferasa/biosíntesis , Humanos , Inmunohistoquímica , Hibridación in Situ , Masculino , Microscopía Fluorescente , Persona de Mediana Edad , Trastornos del Olfato , Neuronas Receptoras Olfatorias/metabolismo , Sistema Respiratorio , SARS-CoV-2 , Olfato
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA