Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.052
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 187(17): 4733-4750.e26, 2024 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-38971152

RESUMEN

We identify a population of Protogenin-positive (PRTG+ve) MYChigh NESTINlow stem cells in the four-week-old human embryonic hindbrain that subsequently localizes to the ventricular zone of the rhombic lip (RLVZ). Oncogenic transformation of early Prtg+ve rhombic lip stem cells initiates group 3 medulloblastoma (Gr3-MB)-like tumors. PRTG+ve stem cells grow adjacent to a human-specific interposed vascular plexus in the RLVZ, a phenotype that is recapitulated in Gr3-MB but not in other types of medulloblastoma. Co-culture of Gr3-MB with endothelial cells promotes tumor stem cell growth, with the endothelial cells adopting an immature phenotype. Targeting the PRTGhigh compartment of Gr3-MB in vivo using either the diphtheria toxin system or chimeric antigen receptor T cells constitutes effective therapy. Human Gr3-MBs likely arise from early embryonic RLVZ PRTG+ve stem cells inhabiting a specific perivascular niche. Targeting the PRTGhigh compartment and/or the perivascular niche represents an approach to treat children with Gr3-MB.


Asunto(s)
Meduloblastoma , Células Madre Neoplásicas , Humanos , Meduloblastoma/patología , Meduloblastoma/metabolismo , Animales , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Ratones , Rombencéfalo/metabolismo , Rombencéfalo/embriología , Neoplasias Cerebelosas/metabolismo , Neoplasias Cerebelosas/patología , Células Endoteliales/metabolismo , Nicho de Células Madre , Células Madre/metabolismo , Técnicas de Cocultivo , Estructuras Embrionarias , Metencéfalo/embriología
2.
Cell ; 186(14): 3062-3078.e20, 2023 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-37343561

RESUMEN

Seemingly simple behaviors such as swatting a mosquito or glancing at a signpost involve the precise coordination of multiple body parts. Neural control of coordinated movements is widely thought to entail transforming a desired overall displacement into displacements for each body part. Here we reveal a different logic implemented in the mouse gaze system. Stimulating superior colliculus (SC) elicits head movements with stereotyped displacements but eye movements with stereotyped endpoints. This is achieved by individual SC neurons whose branched axons innervate modules in medulla and pons that drive head movements with stereotyped displacements and eye movements with stereotyped endpoints, respectively. Thus, single neurons specify a mixture of endpoints and displacements for different body parts, not overall displacement, with displacements for different body parts computed at distinct anatomical stages. Our study establishes an approach for unraveling motor hierarchies and identifies a logic for coordinating movements and the resulting pose.


Asunto(s)
Fijación Ocular , Movimientos Sacádicos , Animales , Ratones , Movimientos Oculares , Neuronas/fisiología , Colículos Superiores/fisiología , Rombencéfalo , Movimientos de la Cabeza/fisiología
3.
Cell ; 186(1): 14-16, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36608650

RESUMEN

How the neocortex modulates hindbrain and spinal circuits is of fundamental interest for understanding motor control and adaptive behaviors. New work from Yang, Kanodia, and Arber demonstrates that there is an exquisite anatomical organization and functional modulation from the anterior (motor) cortex on downstream medulla populations during forelimb behaviors in mice.


Asunto(s)
Miembro Anterior , Neocórtex , Animales , Ratones , Corteza Motora/fisiología , Rombencéfalo/fisiología , Columna Vertebral/fisiología
4.
Cell ; 185(26): 5011-5027.e20, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36563666

RESUMEN

To track and control self-location, animals integrate their movements through space. Representations of self-location are observed in the mammalian hippocampal formation, but it is unknown if positional representations exist in more ancient brain regions, how they arise from integrated self-motion, and by what pathways they control locomotion. Here, in a head-fixed, fictive-swimming, virtual-reality preparation, we exposed larval zebrafish to a variety of involuntary displacements. They tracked these displacements and, many seconds later, moved toward their earlier location through corrective swimming ("positional homeostasis"). Whole-brain functional imaging revealed a network in the medulla that stores a memory of location and induces an error signal in the inferior olive to drive future corrective swimming. Optogenetically manipulating medullary integrator cells evoked displacement-memory behavior. Ablating them, or downstream olivary neurons, abolished displacement corrections. These results reveal a multiregional hindbrain circuit in vertebrates that integrates self-motion and stores self-location to control locomotor behavior.


Asunto(s)
Neuronas , Pez Cebra , Animales , Pez Cebra/fisiología , Neuronas/fisiología , Rombencéfalo/fisiología , Encéfalo/fisiología , Natación/fisiología , Homeostasis , Mamíferos
5.
Cell ; 182(6): 1589-1605.e22, 2020 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-32841600

RESUMEN

Hunger and thirst have distinct goals but control similar ingestive behaviors, and little is known about neural processes that are shared between these behavioral states. We identify glutamatergic neurons in the peri-locus coeruleus (periLCVGLUT2 neurons) as a polysynaptic convergence node from separate energy-sensitive and hydration-sensitive cell populations. We develop methods for stable hindbrain calcium imaging in free-moving mice, which show that periLCVGLUT2 neurons are tuned to ingestive behaviors and respond similarly to food or water consumption. PeriLCVGLUT2 neurons are scalably inhibited by palatability and homeostatic need during consumption. Inhibition of periLCVGLUT2 neurons is rewarding and increases consumption by enhancing palatability and prolonging ingestion duration. These properties comprise a double-negative feedback relationship that sustains food or water consumption without affecting food- or water-seeking. PeriLCVGLUT2 neurons are a hub between hunger and thirst that specifically controls motivation for food and water ingestion, which is a factor that contributes to hedonic overeating and obesity.


Asunto(s)
Regulación del Apetito/fisiología , Ingestión de Líquidos/fisiología , Ingestión de Alimentos/fisiología , Locus Coeruleus/citología , Red Nerviosa/fisiología , Neuronas/fisiología , Rombencéfalo/fisiología , Análisis de la Célula Individual/métodos , Animales , Apetito/fisiología , Escala de Evaluación de la Conducta , Retroalimentación , Conducta Alimentaria/fisiología , Femenino , Glutamina/metabolismo , Glutamina/fisiología , Homeostasis/fisiología , Hambre/fisiología , Masculino , Ratones , Ratones Noqueados , Motivación/fisiología , Neuronas/efectos de los fármacos , Proteínas Recombinantes , Recompensa , Rombencéfalo/citología , Rombencéfalo/diagnóstico por imagen , Gusto/fisiología , Sed/fisiología
6.
Cell ; 180(3): 536-551.e17, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-31955849

RESUMEN

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


Asunto(s)
Cerebelo/fisiología , Toma de Decisiones/fisiología , Tiempo de Reacción/fisiología , Pez Cebra/fisiología , Animales , Conducta Animal/fisiología , Mapeo Encefálico/métodos , Cerebro/fisiología , Cognición/fisiología , Condicionamiento Operante/fisiología , Objetivos , Habénula/fisiología , Calor , Larva/fisiología , Actividad Motora/fisiología , Movimiento , Neuronas/fisiología , Desempeño Psicomotor/fisiología , Rombencéfalo/fisiología
7.
Cell ; 183(7): 1913-1929.e26, 2020 12 23.
Artículo en Inglés | MEDLINE | ID: mdl-33333020

RESUMEN

Neurons in the cerebral cortex connect through descending pathways to hindbrain and spinal cord to activate muscle and generate movement. Although components of this pathway have been previously generated and studied in vitro, the assembly of this multi-synaptic circuit has not yet been achieved with human cells. Here, we derive organoids resembling the cerebral cortex or the hindbrain/spinal cord and assemble them with human skeletal muscle spheroids to generate 3D cortico-motor assembloids. Using rabies tracing, calcium imaging, and patch-clamp recordings, we show that corticofugal neurons project and connect with spinal spheroids, while spinal-derived motor neurons connect with muscle. Glutamate uncaging or optogenetic stimulation of cortical spheroids triggers robust contraction of 3D muscle, and assembloids are morphologically and functionally intact for up to 10 weeks post-fusion. Together, this system highlights the remarkable self-assembly capacity of 3D cultures to form functional circuits that could be used to understand development and disease.


Asunto(s)
Corteza Cerebral/fisiología , Corteza Motora/fisiología , Organoides/fisiología , Animales , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Vértebras Cervicales , Regulación de la Expresión Génica , Glutamatos/metabolismo , Humanos , Células Madre Pluripotentes Inducidas/citología , Ratones , Músculos/fisiología , Mioblastos/metabolismo , Red Nerviosa/fisiología , Optogenética , Organoides/ultraestructura , Rombencéfalo/fisiología , Esferoides Celulares/citología , Médula Espinal/citología
8.
Nature ; 632(8025): 585-593, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38987598

RESUMEN

The most successful obesity therapeutics, glucagon-like peptide-1 receptor (GLP1R) agonists, cause aversive responses such as nausea and vomiting1,2, effects that may contribute to their efficacy. Here, we investigated the brain circuits that link satiety to aversion, and unexpectedly discovered that the neural circuits mediating these effects are functionally separable. Systematic investigation across drug-accessible GLP1R populations revealed that only hindbrain neurons are required for the efficacy of GLP1-based obesity drugs. In vivo two-photon imaging of hindbrain GLP1R neurons demonstrated that most neurons are tuned to either nutritive or aversive stimuli, but not both. Furthermore, simultaneous imaging of hindbrain subregions indicated that area postrema (AP) GLP1R neurons are broadly responsive, whereas nucleus of the solitary tract (NTS) GLP1R neurons are biased towards nutritive stimuli. Strikingly, separate manipulation of these populations demonstrated that activation of NTSGLP1R neurons triggers satiety in the absence of aversion, whereas activation of APGLP1R neurons triggers strong aversion with food intake reduction. Anatomical and behavioural analyses revealed that NTSGLP1R and APGLP1R neurons send projections to different downstream brain regions to drive satiety and aversion, respectively. Importantly, GLP1R agonists reduce food intake even when the aversion pathway is inhibited. Overall, these findings highlight NTSGLP1R neurons as a population that could be selectively targeted to promote weight loss while avoiding the adverse side effects that limit treatment adherence.


Asunto(s)
Fármacos Antiobesidad , Reacción de Prevención , Receptor del Péptido 1 Similar al Glucagón , Vías Nerviosas , Rombencéfalo , Respuesta de Saciedad , Animales , Femenino , Masculino , Ratones , Fármacos Antiobesidad/efectos adversos , Fármacos Antiobesidad/farmacología , Área Postrema/metabolismo , Área Postrema/efectos de los fármacos , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Ingestión de Alimentos/efectos de los fármacos , Ingestión de Alimentos/fisiología , Péptido 1 Similar al Glucagón/metabolismo , Receptor del Péptido 1 Similar al Glucagón/agonistas , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Ratones Endogámicos C57BL , Vías Nerviosas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/fisiología , Neuronas/efectos de los fármacos , Obesidad/metabolismo , Rombencéfalo/citología , Rombencéfalo/efectos de los fármacos , Rombencéfalo/metabolismo , Rombencéfalo/fisiología , Respuesta de Saciedad/efectos de los fármacos , Respuesta de Saciedad/fisiología , Núcleo Solitario/citología , Núcleo Solitario/efectos de los fármacos , Núcleo Solitario/metabolismo , Núcleo Solitario/fisiología , Alimentos
9.
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
10.
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
11.
Annu Rev Neurosci ; 42: 67-86, 2019 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-30699050

RESUMEN

The genetic approach, based on the study of inherited forms of deafness, has proven to be particularly effective for deciphering the molecular mechanisms underlying the development of the peripheral auditory system, the cochlea and its afferent auditory neurons, and how this system extracts the physical parameters of sound. Although this genetic dissection has provided little information about the central auditory system, scattered data suggest that some genes may have a critical role in both the peripheral and central auditory systems. Here, we review the genes controlling the development and function of the peripheral and central auditory systems, focusing on those with demonstrated intrinsic roles in both systems and highlighting the current underappreciation of these genes. Their encoded products are diverse, from transcription factors to ion channels, as are their roles in the central auditory system, mostly evaluated in brainstem nuclei. We examine the ontogenetic and evolutionary mechanisms that may underlie their expression at different sites.


Asunto(s)
Vías Auditivas/fisiología , Regulación del Desarrollo de la Expresión Génica , Genes , Neurogénesis/genética , Animales , Vías Auditivas/crecimiento & desarrollo , Evolución Biológica , Cóclea/embriología , Cóclea/crecimiento & desarrollo , Cóclea/fisiología , Ontología de Genes , Células Ciliadas Auditivas/citología , Células Ciliadas Auditivas/fisiología , Trastornos de la Audición/genética , Humanos , Canales Iónicos/genética , Canales Iónicos/fisiología , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/fisiología , Rombencéfalo/embriología , Rombencéfalo/crecimiento & desarrollo , Rombencéfalo/fisiología , Células Receptoras Sensoriales/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología
12.
Development ; 151(4)2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38251863

RESUMEN

The interplay between neural progenitors and stem cells (NPSCs), and their extracellular matrix (ECM) is a crucial regulatory mechanism that determines their behavior. Nonetheless, how the ECM dictates the state of NPSCs remains elusive. The hindbrain is valuable to examine this relationship, as cells in the ventricular surface of hindbrain boundaries (HBs), which arise between any two neighboring rhombomeres, express the NPSC marker Sox2, while being surrounded with the membrane-bound ECM molecule chondroitin sulphate proteoglycan (CSPG), in chick and mouse embryos. CSPG expression was used to isolate HB Sox2+ cells for RNA-sequencing, revealing their distinguished molecular properties as typical NPSCs, which express known and newly identified genes relating to stem cells, cancer, the matrisome and cell cycle. In contrast, the CSPG- non-HB cells, displayed clear neural-differentiation transcriptome. To address whether CSPG is significant for hindbrain development, its expression was manipulated in vivo and in vitro. CSPG manipulations shifted the stem versus differentiation state of HB cells, evident by their behavior and altered gene expression. These results provide further understanding of the uniqueness of hindbrain boundaries as repetitive pools of NPSCs in-between the rapidly growing rhombomeres, which rely on their microenvironment to maintain their undifferentiated state during development.


Asunto(s)
Células-Madre Neurales , Proteoglicanos , Ratones , Animales , Proteoglicanos/metabolismo , Sulfatos de Condroitina , Proteoglicanos Tipo Condroitín Sulfato , Matriz Extracelular/metabolismo , Rombencéfalo/metabolismo , Células-Madre Neurales/metabolismo
13.
Development ; 151(13)2024 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-38860486

RESUMEN

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


Asunto(s)
Cerebelo , Células-Madre Neurales , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/citología , Cerebelo/citología , Cerebelo/metabolismo , Regulación del Desarrollo de la Expresión Génica , Neuronas/metabolismo , Neuronas/citología , Receptores Notch/metabolismo , Receptores Notch/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Transducción de Señal , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Rombencéfalo/metabolismo , Rombencéfalo/citología , Nicho de Células Madre , Proteínas del Tejido Nervioso/metabolismo , Proteínas del Tejido Nervioso/genética
14.
Nature ; 599(7883): 131-135, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34646010

RESUMEN

Oestrogen depletion in rodents and humans leads to inactivity, fat accumulation and diabetes1,2, underscoring the conserved metabolic benefits of oestrogen that inevitably decrease with age. In rodents, the preovulatory surge in 17ß-oestradiol (E2) temporarily increases energy expenditure to coordinate increased physical activity with peak sexual receptivity. Here we report that a subset of oestrogen-sensitive neurons in the ventrolateral ventromedial hypothalamic nucleus (VMHvl)3-7 projects to arousal centres in the hippocampus and hindbrain, and enables oestrogen to rebalance energy allocation in female mice. Surges in E2 increase melanocortin-4 receptor (MC4R) signalling in these VMHvl neurons by directly recruiting oestrogen receptor-α (ERα) to the Mc4r gene. Sedentary behaviour and obesity in oestrogen-depleted female mice were reversed after chemogenetic stimulation of VMHvl neurons expressing both MC4R and ERα. Similarly, a long-term increase in physical activity is observed after CRISPR-mediated activation of this node. These data extend the effect of MC4R signalling - the most common cause of monogenic human obesity8 - beyond the regulation of food intake and rationalize reported sex differences in melanocortin signalling, including greater disease severity of MC4R insufficiency in women9. This hormone-dependent node illuminates the power of oestrogen during the reproductive cycle in motivating behaviour and maintaining an active lifestyle in women.


Asunto(s)
Encéfalo/fisiología , Estrógenos/metabolismo , Esfuerzo Físico/fisiología , Receptor de Melanocortina Tipo 4/metabolismo , Transducción de Señal , Animales , Sistemas CRISPR-Cas , Metabolismo Energético , Receptor alfa de Estrógeno/metabolismo , Estrógenos/deficiencia , Femenino , Edición Génica , Hipocampo/metabolismo , Masculino , Melanocortinas/metabolismo , Ratones , Neuronas/metabolismo , Obesidad/metabolismo , Rombencéfalo/metabolismo , Conducta Sedentaria , Caracteres Sexuales , Núcleo Hipotalámico Ventromedial/citología , Núcleo Hipotalámico Ventromedial/fisiología
15.
Development ; 150(18)2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37642135

RESUMEN

Developing tissues are sequentially patterned by extracellular signals that are turned on and off at specific times. In the zebrafish hindbrain, fibroblast growth factor (Fgf) signalling has different roles at different developmental stages: in the early hindbrain, transient Fgf3 and Fgf8 signalling from rhombomere 4 is required for correct segmentation, whereas later, neuronal Fgf20 expression confines neurogenesis to specific spatial domains within each rhombomere. How the switch between these two signalling regimes is coordinated is not known. We present evidence that the Zbtb16 transcription factor is required for this transition to happen in an orderly fashion. Zbtb16 expression is high in the early anterior hindbrain, then gradually upregulated posteriorly and confined to neural progenitors. In mutants lacking functional Zbtb16, fgf3 expression fails to be downregulated and persists until a late stage, resulting in excess and more widespread Fgf signalling during neurogenesis. Accordingly, the spatial pattern of neurogenesis is disrupted in Zbtb16 mutants. Our results reveal how the distinct stage-specific roles of Fgf signalling are coordinated in the zebrafish hindbrain.


Asunto(s)
Neurogénesis , Pez Cebra , Animales , Neurogénesis/genética , Rombencéfalo , Transducción de Señal/genética , Factores de Transcripción/genética , Pez Cebra/genética
16.
Development ; 150(13)2023 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-37381820

RESUMEN

Cerebellar granule neurons (CGNs) are the most abundant neurons in the human brain. Dysregulation of their development underlies movement disorders and medulloblastomas. It is suspected that these disorders arise in progenitor states of the CGN lineage, for which human models are lacking. Here, we have differentiated human hindbrain neuroepithelial stem (hbNES) cells to CGNs in vitro using soluble growth factors, recapitulating key progenitor states in the lineage. We show that hbNES cells are not lineage committed and retain rhombomere 1 regional identity. Upon differentiation, hbNES cells transit through a rhombic lip (RL) progenitor state at day 7, demonstrating human specific sub-ventricular cell identities. This RL state is followed by an ATOH1+ CGN progenitor state at day 14. By the end of a 56-day differentiation procedure, we obtain functional neurons expressing CGN markers GABAARα6 and vGLUT2. We show that sonic hedgehog promotes GABAergic lineage specification and CGN progenitor proliferation. Our work presents a new model with which to study development and diseases of the CGN lineage in a human context.


Asunto(s)
Cerebelo , Proteínas Hedgehog , Humanos , Proteínas Hedgehog/metabolismo , Rombencéfalo/metabolismo , Diferenciación Celular/fisiología , Neurogénesis , Células Madre
17.
PLoS Pathog ; 20(7): e1012384, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39024393

RESUMEN

Interbacterial competition is known to shape the microbial communities found in the host, however the interplay between this competition and host defense are less clear. Here, we use the zebrafish hindbrain ventricle (HBV) as an in vivo platform to investigate host responses to defined bacterial communities with distinct forms of interbacterial competition. We found that antibacterial activity of the type VI secretion system (T6SS) from both Vibrio cholerae and Acinetobacter baylyi can induce host inflammation and sensitize the host to infection independent of any individual effector. Chemical suppression of inflammation could resolve T6SS-dependent differences in host survival, but the mechanism by which this occurred differed between the two bacterial species. By contrast, colicin-mediated antagonism elicited by an avirulent strain of Shigella sonnei induced a negligible host response despite being a more potent bacterial killer, resulting in no impact on A. baylyi or V. cholerae virulence. Altogether, these results provide insight into how different modes of interbacterial competition in vivo affect the host in distinct ways.


Asunto(s)
Sistemas de Secreción Tipo VI , Vibrio cholerae , Pez Cebra , Animales , Pez Cebra/microbiología , Sistemas de Secreción Tipo VI/metabolismo , Vibrio cholerae/patogenicidad , Acinetobacter , Virulencia , Interacciones Huésped-Patógeno , Antibiosis/fisiología , Rombencéfalo/microbiología , Rombencéfalo/metabolismo
18.
Nature ; 578(7795): 413-418, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051589

RESUMEN

The mammalian claustrum, owing to its widespread connectivity with other forebrain structures, has been hypothesized to mediate functions that range from decision-making to consciousness1. Here we report that a homologue of the claustrum, identified by single-cell transcriptomics and viral tracing of connectivity, also exists in a reptile-the Australian bearded dragon Pogona vitticeps. In Pogona, the claustrum underlies the generation of sharp waves during slow-wave sleep. The sharp waves, together with superimposed high-frequency ripples2, propagate to the entire neighbouring pallial dorsal ventricular ridge (DVR). Unilateral or bilateral lesions of the claustrum suppress the production of sharp-wave ripples during slow-wave sleep in a unilateral or bilateral manner, respectively, but do not affect the regular and rapidly alternating sleep rhythm that is characteristic of sleep in this species3. The claustrum is thus not involved in the generation of the sleep rhythm itself. Tract tracing revealed that the reptilian claustrum projects widely to a variety of forebrain areas, including the cortex, and that it receives converging inputs from, among others, areas of the mid- and hindbrain that are known to be involved in wake-sleep control in mammals4-6. Periodically modulating the concentration of serotonin in the claustrum, for example, caused a matching modulation of sharp-wave production there and in the neighbouring DVR. Using transcriptomic approaches, we also identified a claustrum in the turtle Trachemys scripta, a distant reptilian relative of lizards. The claustrum is therefore an ancient structure that was probably already present in the brain of the common vertebrate ancestor of reptiles and mammals. It may have an important role in the control of brain states owing to the ascending input it receives from the mid- and hindbrain, its widespread projections to the forebrain and its role in sharp-wave generation during slow-wave sleep.


Asunto(s)
Claustro/anatomía & histología , Claustro/fisiología , Lagartos/anatomía & histología , Lagartos/fisiología , Sueño/fisiología , Animales , Claustro/citología , Claustro/lesiones , Masculino , Mamíferos/fisiología , Mesencéfalo/citología , Mesencéfalo/fisiología , Vías Nerviosas , RNA-Seq , Rombencéfalo/citología , Rombencéfalo/fisiología , Serotonina/metabolismo , Análisis de la Célula Individual , Transcriptoma , Tortugas/anatomía & histología , Tortugas/fisiología
19.
J Neurosci ; 44(31)2024 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-38811166

RESUMEN

Neurons in the caudal nucleus of the solitary tract (cNTS) and intermediate reticular nucleus (IRt) that express the glucagon gene (Gcg) give rise to glucagon-like peptide 1 (GLP1)-immunopositive axons in the spinal cord and many subcortical brain regions. Central GLP1 receptor signaling contributes to motivated behavior and stress responses in rats and mice, in which hindbrain GLP1 neurons are activated to express c-Fos in a metabolic state-dependent manner. The present study examined whether GLP1 inputs to distinct brain regions arise from distinct subsets of Gcg-expressing neurons, and mapped the distribution of axon collaterals arising from projection-defined GLP1 neural populations. Using our Gcg-Cre knock-in rat model, Cre-dependent adeno-associated virus (AAV) tracing was conducted in adult male and female rats to compare axonal projections of IRt versus cNTS GLP1 neurons. Overlapping projections were observed in all brain regions that receive GLP1 input, with the caveat that cNTS injections produced Cre-dependent labeling of some IRt neurons, and vice versa. In additional experiments, specific diencephalic or limbic forebrain nuclei were microinjected with Cre-dependent retrograde AAVs (AAVrg) that expressed reporters to fully label the axon collaterals of transduced GLP1 neurons. AAVrg injected into each forebrain site labeled Gcg-expressing neurons in both the cNTS and IRt. The collective axon collaterals of labeled neurons entered the spinal cord and every brain region previously reported to contain GLP1-positive axons. These results indicate that the axons of GLP1 neural populations that innervate the thalamic paraventricular nucleus, paraventricular nucleus of the hypothalamus, and/or bed nucleus of the stria terminalis collectively innervate all central regions that receive GLP1 axonal input.


Asunto(s)
Axones , Péptido 1 Similar al Glucagón , Neuronas , Rombencéfalo , Animales , Masculino , Femenino , Ratas , Péptido 1 Similar al Glucagón/metabolismo , Péptido 1 Similar al Glucagón/genética , Neuronas/metabolismo , Axones/metabolismo , Rombencéfalo/metabolismo , Vías Nerviosas/metabolismo , Ratas Sprague-Dawley , Hipotálamo/metabolismo , Hipotálamo/citología , Prosencéfalo/metabolismo , Sistema Límbico/metabolismo , Núcleo Solitario/metabolismo , Receptor del Péptido 1 Similar al Glucagón/genética , Receptor del Péptido 1 Similar al Glucagón/metabolismo
20.
Development ; 149(22)2022 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-36325991

RESUMEN

In the developing hindbrain, facial branchiomotor (FBM) neurons migrate caudally from rhombomere 4 (r4) to r6 to establish the circuit that drives jaw movements. Although the mechanisms regulating initiation of FBM neuron migration are well defined, those regulating directionality are not. In mutants lacking the Wnt/planar cell polarity (PCP) component Celsr1, many FBM neurons inappropriately migrate rostrally into r3. We hypothesized that Celsr1 normally blocks inappropriate rostral migration of FBM neurons by suppressing chemoattraction towards Wnt5a in r3 and successfully tested this model. First, FBM neurons in Celsr1; Wnt5a double mutant embryos never migrated rostrally, indicating that inappropriate rostral migration in Celsr1 mutants results from Wnt5a-mediated chemoattraction, which is suppressed in wild-type embryos. Second, FBM neurons migrated rostrally toward Wnt5a-coated beads placed in r3 of wild-type hindbrain explants, suggesting that excess Wnt5a chemoattractant can overcome endogenous Celsr1-mediated suppression. Third, rostral migration of FBM neurons was greatly enhanced in Celsr1 mutants overexpressing Wnt5a in r3. These results reveal a novel role for a Wnt/PCP component in regulating neuronal migration through suppression of chemoattraction.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Neuronas Motoras , Neuronas Motoras/fisiología , Rombencéfalo , Polaridad Celular , Movimiento Celular/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA