Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 98
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Stem Cell Reports ; 18(12): 2418-2433, 2023 12 12.
Artigo em Inglês | MEDLINE | ID: mdl-37995703

RESUMO

Although adult subependymal zone (SEZ) neural stem cells mostly generate GABAergic interneurons, a small progenitor population expresses the proneural gene Neurog2 and produces glutamatergic neurons. Here, we determined whether Neurog2 could respecify SEZ neural stem cells and their progeny toward a glutamatergic fate. Retrovirus-mediated expression of Neurog2 induced the glutamatergic lineage markers TBR2 and TBR1 in cultured SEZ progenitors, which differentiated into functional glutamatergic neurons. Likewise, Neurog2-transduced SEZ progenitors acquired glutamatergic neuron hallmarks in vivo. Intriguingly, they failed to migrate toward the olfactory bulb and instead differentiated within the SEZ or the adjacent striatum, where they received connections from local neurons, as indicated by rabies virus-mediated monosynaptic tracing. In contrast, lentivirus-mediated expression of Neurog2 failed to reprogram early SEZ neurons, which maintained GABAergic identity and migrated to the olfactory bulb. Our data show that NEUROG2 can program SEZ progenitors toward a glutamatergic identity but fails to reprogram their neuronal progeny.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Células-Tronco Neurais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Neurônios/metabolismo , Células-Tronco Neurais/metabolismo , Diferenciação Celular , Bulbo Olfatório/metabolismo , Neurogênese/fisiologia
2.
Neuron ; 111(15): 2367-2382.e6, 2023 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-37279750

RESUMO

The central nervous system regulates systemic immune responses by integrating the physiological and behavioral constraints faced by an individual. Corticosterone (CS), the release of which is controlled in the hypothalamus by the paraventricular nucleus (PVN), is a potent negative regulator of immune responses. Using the mouse model, we report that the parabrachial nucleus (PB), an important hub linking interoceptive afferent information to autonomic and behavioral responses, also integrates the pro-inflammatory cytokine IL-1ß signal to induce the CS response. A subpopulation of PB neurons, directly projecting to the PVN and receiving inputs from the vagal complex (VC), responds to IL-1ß to drive the CS response. Pharmacogenetic reactivation of these IL-1ß-activated PB neurons is sufficient to induce CS-mediated systemic immunosuppression. Our findings demonstrate an efficient brainstem-encoded modality for the central sensing of cytokines and the regulation of systemic immune responses.


Assuntos
Citocinas , Núcleos Parabraquiais , Animais , Camundongos , Corticosterona , Retroalimentação , Hipotálamo , Núcleo Hipotalâmico Paraventricular/fisiologia
3.
Sci Adv ; 8(23): eabg9445, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35687687

RESUMO

Cell transplantation is a promising approach for the reconstruction of neuronal circuits after brain damage. Transplanted neurons integrate with remarkable specificity into circuitries of the mouse cerebral cortex affected by neuronal ablation. However, it remains unclear how neurons perform in a local environment undergoing reactive gliosis, inflammation, macrophage infiltration, and scar formation, as in traumatic brain injury (TBI). To elucidate this, we transplanted cells from the embryonic mouse cerebral cortex into TBI-injured, inflamed-only, or intact cortex of adult mice. Brain-wide quantitative monosynaptic rabies virus (RABV) tracing unraveled graft inputs from correct regions across the brain in all conditions, with pronounced quantitative differences: scarce in intact and inflamed brain versus exuberant after TBI. In the latter, the initial overshoot is followed by pruning, with only a few input neurons persisting at 3 months. Proteomic profiling identifies candidate molecules for regulation of the synaptic yield, a pivotal parameter to tailor for functional restoration of neuronal circuits.

4.
Sci Adv ; 8(23): eabg9287, 2022 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-35687689

RESUMO

Transplantation is a clinically relevant approach for brain repair, but much remains to be understood about influences of the disease environment on transplant connectivity. To explore the effect of amyloid pathology in Alzheimer's disease (AD) and aging, we examined graft connectivity using monosynaptic rabies virus tracing in APP/PS1 mice and in 16- to 18-month-old wild-type (WT) mice. Transplanted neurons differentiated within 4 weeks and integrated well into the host visual cortex, receiving input from the appropriate brain regions for this area. Unexpectedly, we found a prominent several-fold increase in local inputs, in both amyloid-loaded and aged environments. State-of-the-art deep proteome analysis using mass spectrometry highlights complement system activation as a common denominator of environments promoting excessive local input connectivity. These data therefore reveal the key role of the host pathology in shaping the input connectome, calling for caution in extrapolating results from one pathological condition to another.

5.
Nat Commun ; 13(1): 2659, 2022 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-35551446

RESUMO

Traumatic brain injury (TBI) results in deficits that are often followed by recovery. The contralesional cortex can contribute to this process but how distinct contralesional neurons and circuits respond to injury remains to be determined. To unravel adaptations in the contralesional cortex, we used chronic in vivo two-photon imaging. We observed a general decrease in spine density with concomitant changes in spine dynamics over time. With retrograde co-labeling techniques, we showed that callosal neurons are uniquely affected by and responsive to TBI. To elucidate circuit connectivity, we used monosynaptic rabies tracing, clearing techniques and histology. We demonstrate that contralesional callosal neurons adapt their input circuitry by strengthening ipsilateral connections from pre-connected areas. Finally, functional in vivo two-photon imaging demonstrates that the restoration of pre-synaptic circuitry parallels the restoration of callosal activity patterns. Taken together our study thus delineates how callosal neurons structurally and functionally adapt following a contralateral murine TBI.


Assuntos
Lesões Encefálicas Traumáticas , Corpo Caloso , Animais , Córtex Cerebral , Corpo Caloso/fisiologia , Camundongos , Neurônios/fisiologia
6.
Neuron ; 110(12): 2024-2040.e10, 2022 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-35452606

RESUMO

General anesthetics induce loss of consciousness, a global change in behavior. However, a corresponding global change in activity in the context of defined cortical cell types has not been identified. Here, we show that spontaneous activity of mouse layer 5 pyramidal neurons, but of no other cortical cell type, becomes consistently synchronized in vivo by different general anesthetics. This heightened neuronal synchrony is aperiodic, present across large distances, and absent in cortical neurons presynaptic to layer 5 pyramidal neurons. During the transition to and from anesthesia, changes in synchrony in layer 5 coincide with the loss and recovery of consciousness. Activity within both apical and basal dendrites is synchronous, but only basal dendrites' activity is temporally locked to somatic activity. Given that layer 5 is a major cortical output, our results suggest that brain-wide synchrony in layer 5 pyramidal neurons may contribute to the loss of consciousness during general anesthesia.


Assuntos
Anestésicos Gerais , Células Piramidais , Anestesia Geral , Anestésicos Gerais/farmacologia , Animais , Dendritos/fisiologia , Camundongos , Células Piramidais/fisiologia , Inconsciência
7.
Nat Methods ; 19(1): 81-89, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34949807

RESUMO

CD4+ T cells are central mediators of adaptive and innate immune responses and constitute a major reservoir for human immunodeficiency virus (HIV) in vivo. Detailed investigations of resting human CD4+ T cells have been precluded by the absence of efficient approaches for genetic manipulation limiting our understanding of HIV replication and restricting efforts to find a cure. Here we report a method for rapid, efficient, activation-neutral gene editing of resting, polyclonal human CD4+ T cells using optimized cell cultivation and nucleofection conditions of Cas9-guide RNA ribonucleoprotein complexes. Up to six genes, including HIV dependency and restriction factors, were knocked out individually or simultaneously and functionally characterized. Moreover, we demonstrate the knock in of double-stranded DNA donor templates into different endogenous loci, enabling the study of the physiological interplay of cellular and viral components at single-cell resolution. Together, this technique allows improved molecular and functional characterizations of HIV biology and general immune functions in resting CD4+ T cells.


Assuntos
Linfócitos T CD4-Positivos/fisiologia , Sistemas CRISPR-Cas/genética , Edição de Genes/métodos , Infecções por HIV/genética , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/virologia , Proteína 9 Associada à CRISPR/genética , Movimento Celular/genética , Células Cultivadas , DNA , Técnicas de Inativação de Genes , Infecções por HIV/metabolismo , Humanos , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Resistência a Myxovirus/genética , Proteínas de Resistência a Myxovirus/metabolismo , RNA Guia de Cinetoplastídeos , Proteína 1 com Domínio SAM e Domínio HD/genética , Transgenes , Fatores de Poliadenilação e Clivagem de mRNA/genética , Fatores de Poliadenilação e Clivagem de mRNA/metabolismo
8.
Nat Commun ; 12(1): 6855, 2021 11 25.
Artigo em Inglês | MEDLINE | ID: mdl-34824253

RESUMO

The bat sarbecovirus RaTG13 is a close relative of SARS-CoV-2, the cause of the COVID-19 pandemic. However, this bat virus was most likely unable to directly infect humans since its Spike (S) protein does not interact efficiently with the human ACE2 receptor. Here, we show that a single T403R mutation increases binding of RaTG13 S to human ACE2 and allows VSV pseudoparticle infection of human lung cells and intestinal organoids. Conversely, mutation of R403T in the SARS-CoV-2 S reduces pseudoparticle infection and viral replication. The T403R RaTG13 S is neutralized by sera from individuals vaccinated against COVID-19 indicating that vaccination might protect against future zoonoses. Our data suggest that a positively charged amino acid at position 403 in the S protein is critical for efficient utilization of human ACE2 by S proteins of bat coronaviruses. This finding could help to better predict the zoonotic potential of animal coronaviruses.


Assuntos
Enzima de Conversão de Angiotensina 2/química , Ligação Proteica , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Animais , COVID-19/virologia , Vacinas contra COVID-19 , Células CACO-2 , Clonagem Molecular , Células HEK293 , Humanos , Simulação de Dinâmica Molecular , Mutação , Replicon , Especificidade da Espécie , Células-Tronco , Zoonoses
9.
Cell Stem Cell ; 28(12): 2104-2121.e10, 2021 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-34592167

RESUMO

Reprogramming brain-resident glial cells into clinically relevant induced neurons (iNs) is an emerging strategy toward replacing lost neurons and restoring lost brain functions. A fundamental question is now whether iNs can promote functional recovery in pathological contexts. We addressed this question in the context of therapy-resistant mesial temporal lobe epilepsy (MTLE), which is associated with hippocampal seizures and degeneration of hippocampal GABAergic interneurons. Using a MTLE mouse model, we show that retrovirus-driven expression of Ascl1 and Dlx2 in reactive hippocampal glia in situ, or in cortical astroglia grafted in the epileptic hippocampus, causes efficient reprogramming into iNs exhibiting hallmarks of interneurons. These induced interneurons functionally integrate into epileptic networks and establish GABAergic synapses onto dentate granule cells. MTLE mice with GABAergic iNs show a significant reduction in both the number and cumulative duration of spontaneous recurrent hippocampal seizures. Thus glia-to-neuron reprogramming is a potential disease-modifying strategy to reduce seizures in therapy-resistant epilepsy.


Assuntos
Epilepsia do Lobo Temporal , Animais , Neurônios GABAérgicos , Hipocampo , Interneurônios , Camundongos , Neuroglia , Convulsões
10.
Viruses ; 13(9)2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34578307

RESUMO

Lyssaviruses are neurotropic rhabdoviruses thought to be restricted to mammalian hosts, and to originate from bats. The identification of lyssavirus sequences from amphibians and reptiles by metatranscriptomics thus comes as a surprise and challenges the mammalian origin of lyssaviruses. The novel sequences of the proposed American tree frog lyssavirus (ATFLV) and anole lizard lyssavirus (ALLV) reveal substantial phylogenetic distances from each other and from bat lyssaviruses, with ATFLV being the most distant. As virus isolation has not been successful yet, we have here studied the functionality of the authentic ATFLV- and ALLV-encoded glycoproteins in the context of rabies virus pseudotype particles. Cryogenic electron microscopy uncovered the incorporation of the plasmid-encoded G proteins in viral envelopes. Infection experiments revealed the infectivity of ATFLV and ALLV G-coated RABV pp for a broad spectrum of cell lines from humans, bats, and reptiles, demonstrating membrane fusion activities. As presumed, ATFLV and ALLV G RABV pp escaped neutralization by human rabies immune sera. The present findings support the existence of contagious lyssaviruses in poikilothermic animals, and reveal a broad cell tropism in vitro, similar to that of the rabies virus.


Assuntos
Anfíbios/virologia , Glicoproteínas/genética , Lyssavirus/patogenicidade , Mamíferos/virologia , Répteis/virologia , Animais , Linhagem Celular , Glicoproteínas/imunologia , Células HEK293 , Especificidade de Hospedeiro , Humanos , Lyssavirus/química , Lyssavirus/classificação , Lyssavirus/imunologia , Testes de Neutralização , Filogenia , Vírus da Raiva/imunologia , Vírus da Raiva/patogenicidade , Zoonoses Virais/transmissão
11.
Sci Rep ; 11(1): 10400, 2021 05 17.
Artigo em Inglês | MEDLINE | ID: mdl-34002008

RESUMO

The lateral hypothalamus (LH) is critically involved in the regulation of homeostatic energy balance. Some neurons in the LH express receptors for leptin (LepRb), a hormone known to increase energy expenditure and decrease energy intake. However, the neuroanatomical inputs to LepRb-expressing LH neurons remain unknown. We used rabies virus tracing technology to map these inputs, but encountered non-specific tracing. To optimize this technology for a minor cell population (LepRb is not ubiquitously expressed in LH), we used LepRb-Cre mice and assessed how different titers of the avian tumor virus receptor A (TVA) helper virus affected rabies tracing efficiency and specificity. We found that rabies expression is dependent on TVA receptor expression, and that leakiness of TVA receptors is dependent on the titer of TVA virus used. We concluded that a titer of 1.0-3.0 × 107 genomic copies per µl of the TVA virus is optimal for rabies tracing. Next, we successfully applied modified rabies virus tracing technology to map inputs to LepRb-expressing LH neurons. We discovered that other neurons in the LH itself, the periventricular hypothalamic nucleus (Pe), the posterior hypothalamic nucleus (PH), the bed nucleus of the stria terminalis (BNST), and the paraventricular hypothalamic nucleus (PVN) are the most prominent input areas to LepRb-expressing LH neurons.


Assuntos
Conectoma/métodos , Hipotálamo/diagnóstico por imagem , Imagem Molecular/métodos , Neurônios/metabolismo , Receptores para Leptina/análise , Animais , Proteínas Aviárias/genética , Feminino , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Vírus Auxiliares/genética , Hipotálamo/citologia , Hipotálamo/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Microscopia de Fluorescência , Vírus da Raiva/genética , Receptores para Leptina/metabolismo , Receptores Virais/genética , Núcleos Septais/citologia , Núcleos Septais/diagnóstico por imagem , Núcleos Septais/metabolismo , Técnicas Estereotáxicas
12.
Cell Rep ; 35(7): 109126, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-33974846

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) evades most innate immune responses but may still be vulnerable to some. Here, we systematically analyze the impact of SARS-CoV-2 proteins on interferon (IFN) responses and autophagy. We show that SARS-CoV-2 proteins synergize to counteract anti-viral immune responses. For example, Nsp14 targets the type I IFN receptor for lysosomal degradation, ORF3a prevents fusion of autophagosomes and lysosomes, and ORF7a interferes with autophagosome acidification. Most activities are evolutionarily conserved. However, SARS-CoV-2 Nsp15 antagonizes IFN signaling less efficiently than the orthologs of closely related RaTG13-CoV and SARS-CoV-1. Overall, SARS-CoV-2 proteins counteract autophagy and type I IFN more efficiently than type II or III IFN signaling, and infection experiments confirm potent inhibition by IFN-γ and -λ1. Our results define the repertoire and selected mechanisms of SARS-CoV-2 innate immune antagonists but also reveal vulnerability to type II and III IFN that may help to develop safe and effective anti-viral approaches.


Assuntos
COVID-19/virologia , SARS-CoV-2/imunologia , Proteínas Virais/imunologia , Animais , Antivirais/farmacologia , Autofagossomos/imunologia , Autofagia/imunologia , COVID-19/imunologia , Linhagem Celular , Chlorocebus aethiops , Exorribonucleases/imunologia , Células HEK293 , Células HeLa , Humanos , Evasão da Resposta Imune , Imunidade Inata , Interferon Tipo I/metabolismo , Interferons/metabolismo , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptor de Interferon alfa e beta/imunologia , SARS-CoV-2/patogenicidade , Células Vero , Proteínas não Estruturais Virais/imunologia
13.
PLoS Pathog ; 17(4): e1009064, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33882114

RESUMO

Vaccines of outstanding efficiency, safety, and public acceptance are needed to halt the current SARS-CoV-2 pandemic. Concerns include potential side effects caused by the antigen itself and safety of viral DNA and RNA delivery vectors. The large SARS-CoV-2 spike (S) protein is the main target of current COVID-19 vaccine candidates but can induce non-neutralizing antibodies, which might cause vaccination-induced complications or enhancement of COVID-19 disease. Besides, encoding of a functional S in replication-competent virus vector vaccines may result in the emergence of viruses with altered or expanded tropism. Here, we have developed a safe single round rhabdovirus replicon vaccine platform for enhanced presentation of the S receptor-binding domain (RBD). Structure-guided design was employed to build a chimeric minispike comprising the globular RBD linked to a transmembrane stem-anchor sequence derived from rabies virus (RABV) glycoprotein (G). Vesicular stomatitis virus (VSV) and RABV replicons encoding the minispike not only allowed expression of the antigen at the cell surface but also incorporation into the envelope of secreted non-infectious particles, thus combining classic vector-driven antigen expression and particulate virus-like particle (VLP) presentation. A single dose of a prototype replicon vaccine complemented with VSV G, VSVΔG-minispike-eGFP (G), stimulated high titers of SARS-CoV-2 neutralizing antibodies in mice, equivalent to those found in COVID-19 patients, and protected transgenic K18-hACE2 mice from COVID-19-like disease. Homologous boost immunization further enhanced virus neutralizing activity. The results demonstrate that non-spreading rhabdovirus RNA replicons expressing minispike proteins represent effective and safe alternatives to vaccination approaches using replication-competent viruses and/or the entire S antigen.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Vacinas contra COVID-19/administração & dosagem , COVID-19/prevenção & controle , Imunização/métodos , SARS-CoV-2/imunologia , Vacinas de Partículas Semelhantes a Vírus/administração & dosagem , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , COVID-19/imunologia , COVID-19/virologia , Feminino , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL
14.
J Neurosci ; 40(46): 8870-8882, 2020 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-33051345

RESUMO

The emergence of genetic tools has provided new means of mapping functionality in central amygdala (CeA) neuron populations based on their molecular profiles, response properties, and importantly, connectivity patterns. While abundant evidence indicates that neuronal signals arrive in the CeA eliciting both aversive and appetitive behaviors, our understanding of the anatomy of the underlying long-range CeA network remains fragmentary. In this study, we combine viral tracings, electrophysiological, and optogenetic approaches to establish in male mice, a wiring chart between the insula cortex (IC), a major sensory input region of the lateral and capsular part of the CeA (CeL/C), and four principal output streams of this nucleus. We found that retrogradely labeled output neurons occupy discrete and likely strategic locations in the CeL/C, and that they are disproportionally controlled by the IC. We identified a direct line of connection between the IC and the lateral hypothalamus (LH), which engages numerous LH-projecting CeL/C cells whose activity can be strongly upregulated on firing of IC neurons. In comparison, CeL/C neurons projecting to the bed nucleus of the stria terminalis (BNST) are also frequently contacted by incoming IC axons, but the strength of this connection is weak. Our results provide a link between long-range inputs and outputs of the CeA and pave the way to a better understanding of how internal, external, and experience dependent information may impinge on action selection by the CeA.SIGNIFICANCE STATEMENT Our current knowledge of the circuit organization within the central amygdala (CeA), a critical regulator of emotional states, includes independent information about its long-range efferents and afferents. We do not know how incoming sensory information is appraised and routed through the CeA to the different output channels. We address this issue by using three different techniques to investigate how a sensory region, the insula cortex (IC), connects with the motor, physiological and autonomic output centers of the CeA. We uncover a strong connection between the IC and the lateral hypothalamus (LH) with a monosynaptic relay in the CeA and shed new light on the previously described functions of IC and CeA through direct projections to the LH.


Assuntos
Núcleo Central da Amígdala/fisiologia , Córtex Cerebral/fisiologia , Animais , Axônios/fisiologia , Fenômenos Eletrofisiológicos , Região Hipotalâmica Lateral/fisiologia , Técnicas In Vitro , Interneurônios/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Vias Neurais/fisiologia , Optogenética , Núcleos Septais/fisiologia
15.
Elife ; 92020 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-32940600

RESUMO

The insular cortex (IC) plays key roles in emotional and regulatory brain functions and is affected across psychiatric diseases. However, the brain-wide connections of the mouse IC have not been comprehensively mapped. Here, we traced the whole-brain inputs and outputs of the mouse IC across its rostro-caudal extent. We employed cell-type-specific monosynaptic rabies virus tracings to characterize afferent connections onto either excitatory or inhibitory IC neurons, and adeno-associated viral tracings to label excitatory efferent axons. While the connectivity between the IC and other cortical regions was highly bidirectional, the IC connectivity with subcortical structures was often unidirectional, revealing prominent cortical-to-subcortical or subcortical-to-cortical pathways. The posterior and medial IC exhibited resembling connectivity patterns, while the anterior IC connectivity was distinct, suggesting two major functional compartments. Our results provide insights into the anatomical architecture of the mouse IC and thus a structural basis to guide investigations into its complex functions.


Assuntos
Mapeamento Encefálico , Córtex Cerebral/anatomia & histologia , Camundongos/anatomia & histologia , Neurônios/citologia , Animais , Feminino , Masculino
16.
Viruses ; 12(9)2020 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-32872471

RESUMO

Rhabdoviruses, as single-stranded, negative-sense RNA viruses within the order Mononegavirales, are characterised by bullet-shaped or bacteroid particles that contain a helical ribonucleoprotein complex (RNP). Here, we review the components of the RNP and its higher-order structural assembly.


Assuntos
Rhabdoviridae/química , Ribonucleoproteínas/química , Proteínas Virais/química , Proteínas do Nucleocapsídeo/química , Proteínas do Nucleocapsídeo/ultraestrutura , Conformação Proteica , Rhabdoviridae/genética , Ribonucleoproteínas/ultraestrutura , Proteínas da Matriz Viral/química , Proteínas da Matriz Viral/ultraestrutura , Proteínas Virais/ultraestrutura , Proteínas do Complexo da Replicase Viral/química , Proteínas do Complexo da Replicase Viral/ultraestrutura , Vírion/química
17.
Sci Rep ; 10(1): 10953, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616790

RESUMO

Anatomically incomplete spinal cord injuries can be followed by functional recovery mediated, in part, by the formation of intraspinal detour circuits. Here, we show that adult mice recover tactile and proprioceptive function following a unilateral dorsal column lesion. We therefore investigated the basis of this recovery and focused on the plasticity of the dorsal column-medial lemniscus pathway. We show that ascending dorsal root ganglion (DRG) axons branch in the spinal grey matter and substantially increase the number of these collaterals following injury. These sensory fibers exhibit synapsin-positive varicosities, indicating their integration into spinal networks. Using a monosynaptic circuit tracing with rabies viruses injected into the cuneate nucleus, we show the presence of spinal cord neurons that provide a detour pathway to the original target area of DRG axons. Notably the number of contacts between DRG collaterals and those spinal neurons increases by more than 300% after injury. We then characterized these interneurons and showed that the lesion triggers a remodeling of the connectivity pattern. Finally, using re-lesion experiments after initial remodeling of connections, we show that these detour circuits are responsible for the recovery of tactile and proprioceptive function. Taken together our study reveals that detour circuits represent a common blueprint for axonal rewiring after injury.


Assuntos
Gânglios Espinais/fisiologia , Regeneração Nervosa , Vias Neurais , Neurônios/fisiologia , Recuperação de Função Fisiológica , Células Receptoras Sensoriais/fisiologia , Traumatismos da Medula Espinal/prevenção & controle , Animais , Comportamento Animal , Gânglios Espinais/citologia , Camundongos , Camundongos Endogâmicos C57BL , Plasticidade Neuronal , Neurônios/citologia , Traumatismos da Medula Espinal/etiologia , Traumatismos da Medula Espinal/patologia
18.
Cell Metab ; 31(4): 791-808.e8, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32220306

RESUMO

Astrocytes have emerged for playing important roles in brain tissue repair; however, the underlying mechanisms remain poorly understood. We show that acute injury and blood-brain barrier disruption trigger the formation of a prominent mitochondrial-enriched compartment in astrocytic endfeet, which enables vascular remodeling. Integrated imaging approaches revealed that this mitochondrial clustering is part of an adaptive response regulated by fusion dynamics. Astrocyte-specific conditional deletion of Mitofusin 2 (Mfn2) suppressed perivascular mitochondrial clustering and disrupted mitochondria-endoplasmic reticulum (ER) contact sites. Functionally, two-photon imaging experiments showed that these structural changes were mirrored by impaired mitochondrial Ca2+ uptake leading to abnormal cytosolic transients within endfeet in vivo. At the tissue level, a compromised vascular complexity in the lesioned area was restored by boosting mitochondrial-ER perivascular tethering in MFN2-deficient astrocytes. These data unmask a crucial role for mitochondrial dynamics in coordinating astrocytic local domains and have important implications for repairing the injured brain.


Assuntos
Lesões Encefálicas/metabolismo , Encéfalo/irrigação sanguínea , Retículo Endoplasmático/metabolismo , Mitocôndrias/metabolismo , Remodelação Vascular , Animais , Astrócitos , Células Cultivadas , Feminino , GTP Fosfo-Hidrolases/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL
19.
Neuron ; 104(5): 972-986.e6, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31761708

RESUMO

How neural circuits develop in the human brain has remained almost impossible to study at the neuronal level. Here, we investigate human cortical neuron development, plasticity, and function using a mouse/human chimera model in which xenotransplanted human cortical pyramidal neurons integrate as single cells into the mouse cortex. Combined neuronal tracing, electrophysiology, and in vivo structural and functional imaging of the transplanted cells reveal a coordinated developmental roadmap recapitulating key milestones of human cortical neuron development. The human neurons display a prolonged developmental timeline, indicating the neuron-intrinsic retention of juvenile properties as an important component of human brain neoteny. Following maturation, human neurons in the visual cortex display tuned, decorrelated responses to visual stimuli, like mouse neurons, demonstrating their capacity for physiological synaptic integration in host cortical circuits. These findings provide new insights into human neuronal development and open novel avenues for the study of human neuronal function and disease. VIDEO ABSTRACT.


Assuntos
Neurogênese/fisiologia , Células Piramidais/citologia , Células Piramidais/fisiologia , Células Piramidais/transplante , Animais , Diferenciação Celular/fisiologia , Xenoenxertos , Humanos , Camundongos , Córtex Visual/citologia , Córtex Visual/fisiologia
20.
Cell Rep ; 28(13): 3450-3461.e8, 2019 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-31553913

RESUMO

Parvalbumin (PV)-expressing GABAergic neurons are the largest class of inhibitory neocortical cells. We visualize brain-wide, monosynaptic inputs to PV neurons in mouse barrel cortex. We develop intersectional rabies virus tracing to specifically target GABAergic PV cells and exclude a small fraction of excitatory PV cells from our starter population. Local inputs are mainly from layer (L) IV and excitatory cells. A small number of inhibitory inputs originate from LI neurons, which connect to LII/III PV neurons. Long-range inputs originate mainly from other sensory cortices and the thalamus. In visual cortex, most transsynaptically labeled neurons are located in LIV, which contains a molecularly mixed population of projection neurons with putative functional similarity to LIII neurons. This study expands our knowledge of the brain-wide circuits in which PV neurons are embedded and introduces intersectional rabies virus tracing as an applicable tool to dissect the circuitry of more clearly defined cell types.


Assuntos
Córtex Cerebral/metabolismo , Neurônios GABAérgicos/metabolismo , Vias Neurais/metabolismo , Parvalbuminas/metabolismo , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...