Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
1.
J Neurosci ; 42(47): 8780-8794, 2022 11 23.
Artigo em Inglês | MEDLINE | ID: mdl-36202615

RESUMO

The mammalian brain contains numerous neurons distributed across forebrain, midbrain, and hindbrain that project axons to the lower spinal cord and work in concert to control movement and achieve homeostasis. Extensive work has mapped the anatomic location of supraspinal cell types and continues to establish specific physiological functions. The patterns of gene expression that typify and distinguish these disparate populations, however, are mostly unknown. Here, using adult mice of mixed sex, we combined retrograde labeling of supraspinal cell nuclei with fluorescence-activated nuclei sorting and single-nuclei RNA sequencing analyses to transcriptionally profile neurons that project axons from the brain to lumbar spinal cord. We identified 14 transcriptionally distinct cell types and used a combination of established and newly identified marker genes to assign an anatomic location to each. To validate the putative marker genes, we visualized selected transcripts and confirmed selective expression within lumbar-projecting neurons in discrete supraspinal regions. Finally, we illustrate the potential utility of these data by examining the expression of transcription factors that distinguish different supraspinal cell types and by surveying the expression of receptors for growth and guidance cues that may be present in the spinal cord. Collectively, these data establish transcriptional differences between anatomically defined supraspinal populations, identify a new set of marker genes of use in future experiments, and provide insight into potential differences in cellular and physiological activity across the supraspinal connectome.SIGNIFICANCE STATEMENT The brain communicates with the body through a wide variety of neuronal populations with distinct functions and differential sensitivity to damage and disease. We have used single-nuclei RNA sequencing technology to distinguish patterns of gene expression within a diverse set of neurons that project axons from the mouse brain to the lumbar spinal cord. The results reveal transcriptional differences between populations previously defined on the basis of anatomy, provide new marker genes to facilitate rapid identification of cell type in future work, and suggest distinct responsiveness of different supraspinal populations to external growth and guidance cues.


Assuntos
Axônios , Medula Espinal , Animais , Camundongos , Medula Espinal/fisiologia , Axônios/fisiologia , Núcleo Solitário , Neurônios , Mamíferos
2.
Proc Natl Acad Sci U S A ; 117(48): 30710-30721, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33208539

RESUMO

Although ubiquitous in biological studies, the enhanced green and yellow fluorescent proteins (EGFP and EYFP) were not specifically optimized for neuroscience, and their underwhelming brightness and slow expression in brain tissue limits the fidelity of dendritic spine analysis and other indispensable techniques for studying neurodevelopment and plasticity. We hypothesized that EGFP's low solubility in mammalian systems must limit the total fluorescence output of whole cells, and that improving folding efficiency could therefore translate into greater brightness of expressing neurons. By introducing rationally selected combinations of folding-enhancing mutations into GFP templates and screening for brightness and expression rate in human cells, we developed mGreenLantern, a fluorescent protein having up to sixfold greater brightness in cells than EGFP. mGreenLantern illuminates neurons in the mouse brain within 72 h, dramatically reducing lag time between viral transduction and imaging, while its high brightness improves detection of neuronal morphology using widefield, confocal, and two-photon microscopy. When virally expressed to projection neurons in vivo, mGreenLantern fluorescence developed four times faster than EYFP and highlighted long-range processes that were poorly detectable in EYFP-labeled cells. Additionally, mGreenLantern retains strong fluorescence after tissue clearing and expansion microscopy, thereby facilitating superresolution and whole-brain imaging without immunohistochemistry. mGreenLantern can directly replace EGFP/EYFP in diverse systems due to its compatibility with GFP filter sets, recognition by EGFP antibodies, and excellent performance in mouse, human, and bacterial cells. Our screening and rational engineering approach is broadly applicable and suggests that greater potential of fluorescent proteins, including biosensors, could be unlocked using a similar strategy.


Assuntos
Expressão Gênica , Proteínas de Fluorescência Verde/genética , Imagem Molecular , Neurônios/metabolismo , Animais , Encéfalo/metabolismo , Imunofluorescência , Genes Reporter , Proteínas de Fluorescência Verde/química , Camundongos , Microscopia de Fluorescência , Imagem Molecular/métodos , Mutação , Estabilidade Proteica , Proteólise , Solubilidade , Análise Espectral
3.
J Neurosci ; 38(49): 10566-10581, 2018 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-30341180

RESUMO

The brain communicates with the spinal cord through numerous axon tracts that arise from discrete nuclei, transmit distinct functions, and often collateralize to facilitate the coordination of descending commands. This complexity presents a major challenge to interpreting functional outcomes from therapies that target supraspinal connectivity after injury or disease, while the wide distribution of supraspinal nuclei complicates the delivery of therapeutics. Here we harness retrograde viral vectors to overcome these challenges. We demonstrate that injection of AAV2-Retro to the cervical spinal cord of adult female mice results in highly efficient transduction of supraspinal populations throughout the brainstem, midbrain, and cortex. Some supraspinal populations, including corticospinal and rubrospinal neurons, were transduced with >90% efficiency, with robust transgene expression within 3 d of injection. In contrast, propriospinal and raphe spinal neurons showed much lower rates of retrograde transduction. Using tissue clearing and light-sheet microscopy we present detailed visualizations of descending axons tracts and create a mesoscopic projectome for the spinal cord. Moreover, chemogenetic silencing of supraspinal neurons with retrograde vectors resulted in complete and reversible forelimb paralysis, illustrating effective modulation of supraspinal function. Retrograde vectors were also highly efficient when injected after spinal injury, highlighting therapeutic potential. These data provide a global view of supraspinal connectivity and illustrate the potential of retrograde vectors to parse the functional contributions of supraspinal inputs.SIGNIFICANCE STATEMENT The complexity of descending inputs to the spinal cord presents a major challenge in efforts deliver therapeutics to widespread supraspinal systems, and to interpret their functional effects. Here we demonstrate highly effective gene delivery to diverse supraspinal nuclei using a retrograde viral approach and combine it with tissue clearing and 3D microscopy to map the descending projectome from brain to spinal cord. These data highlight newly developed retrograde viruses as therapeutic and research tools, while offering new insights into supraspinal connectivity.


Assuntos
Encéfalo/diagnóstico por imagem , Imageamento Tridimensional/métodos , Rede Nervosa/diagnóstico por imagem , Tratos Piramidais/diagnóstico por imagem , Animais , Encéfalo/fisiologia , Química Encefálica/fisiologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Rede Nervosa/química , Rede Nervosa/fisiologia , Propriocepção/fisiologia , Tratos Piramidais/química , Tratos Piramidais/fisiologia , Transdução de Sinais/fisiologia , Medula Espinal
4.
Neurobiol Dis ; 89: 10-22, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26804026

RESUMO

NG2 cells, also known as oligodendrocyte progenitors or polydendrocytes, are a major component of the glial scar that forms after spinal cord injury. NG2 cells react to injury by proliferating around the lesion site and differentiating into oligodendrocytes and astrocytes, but the molecular mechanism is poorly understood. In this study, we tested the role of the transcription factor STAT3, and its suppressor SOCS3, in NG2 cell proliferation and differentiation after spinal cord injury. Using knockout mice in which STAT3 or SOCS3 are genetically deleted specifically in NG2 cells, we found that deletion of STAT3 led to a reduction in oligodendrogenesis, while deletion of SOCS3 led to enhanced proliferation of NG2 cells within the glial scar after spinal cord injury. Additionally, STAT3 and SOCS3 were not required for astrogliogenesis from NG2 cells after spinal cord injury. Interestingly, genetic deletion of STAT3 and SOCS3 did not have opposing effects, suggesting that SOCS3 may have targets other than the STAT3 pathway in NG2 cells after spinal cord injury. Altogether, our data show that both STAT3 and SOCS3 play important, yet unexpected, roles in NG2 cell proliferation and differentiation after spinal cord injury.


Assuntos
Diferenciação Celular , Proliferação de Células , Oligodendroglia/fisiologia , Fator de Transcrição STAT3/fisiologia , Traumatismos da Medula Espinal/fisiopatologia , Proteína 3 Supressora da Sinalização de Citocinas/fisiologia , Animais , Astrócitos/fisiologia , Contagem de Células , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligodendroglia/metabolismo , Fator de Transcrição STAT3/genética , Células-Tronco/fisiologia , Proteína 3 Supressora da Sinalização de Citocinas/genética
5.
Eur J Neurosci ; 44(10): 2858-2870, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27564458

RESUMO

Astrocytes are a morphologically and functionally heterogeneous population of cells that play critical roles in neurodevelopment and in the regulation of central nervous system homeostasis. Studies of human astrocytes have been hampered by the lack of specific molecular markers and by the difficulties associated with purifying and culturing astrocytes from adult human brains. Human neural progenitor cells (NPCs) with self-renewal and multipotent properties represent an appealing model system to gain insight into the developmental genetics and function of human astrocytes, but a comprehensive molecular characterization that confirms the validity of this cellular system is still missing. Here we used an unbiased transcriptomic analysis to characterize in vitro culture of human NPCs and to define the gene expression programs activated during the differentiation of these cells into astrocytes using FBS or the combination of CNTF and BMP4. Our results demonstrate that in vitro cultures of human NPCs isolated during the gliogenic phase of neurodevelopment mainly consist of radial glial cells (RGCs) and glia-restricted progenitor cells. In these cells the combination of CNTF and BMP4 activates the JAK/STAT and SMAD signaling cascades, leading to the inhibition of oligodendrocytes lineage commitment and activation of astrocytes differentiation. On the other hand, FBS-derived astrocytes have properties of reactive astrocytes. Our work suggests that in vitro culture of human NPCs represents a valuable cellular system to study human disorders characterized by impairment of astrocytes development and function. Our datasets represent an important resource for researchers studying human astrocytes development and might set the basis for the discovery of novel human-specific astrocyte markers.


Assuntos
Astrócitos/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Neurais/metabolismo , Transcriptoma , Astrócitos/citologia , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Células Cultivadas , Fator Neurotrófico Ciliar/genética , Fator Neurotrófico Ciliar/metabolismo , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , Células-Tronco Neurais/citologia , Neurogênese , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo
6.
Hum Mol Genet ; 22(8): 1601-14, 2013 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-23314018

RESUMO

Axon degeneration is a common and often early feature of neurodegeneration that correlates with the clinical manifestations and progression of neurological disease. Nicotinamide mononucleotide adenylytransferase (NMNAT) is a neuroprotective factor that delays axon degeneration following injury and in models of neurodegenerative diseases suggesting a converging molecular pathway of axon self-destruction. The underlying mechanisms have been under intense investigation and recent reports suggest a central role for axonal mitochondria in both degeneration and NMNAT/WLD(S) (Wallerian degeneration slow)-mediated protection. We used dorsal root ganglia (DRG) explants and Drosophila larval motor neurons (MNs) as models to address the role of mitochondria in Wallerian degeneration (WD). We find that expression of Drosophila NMNAT delays WD in human DRG neurons demonstrating evolutionary conservation of NMNAT function. Morphological comparison of mitochondria from WLD(S)-protected axons demonstrates that mitochondria shrink post-axotomy, though analysis of complex IV activity suggests that they retain their functional capacity despite this morphological change. To determine whether mitochondria are a critical site of regulation for WD, we genetically ablated mitochondria from Drosophila MN axons via the mitochondria trafficking protein milton. Milton loss-of-function did not induce axon degeneration in Drosophila larval MNs, and when axotomized WD proceeded stereotypically in milton distal axons although with a mild, but significant delay. Remarkably, the protective effects of NMNAT/WLD(S) were also maintained in axons devoid of mitochondria. These experiments unveil an axon self-destruction cascade governing WD that is not initiated by axonal mitochondria and for the first time illuminate a mitochondria-independent mechanism(s) regulating WD and NMNAT/WLD(S)-mediated axon protection.


Assuntos
Axônios/metabolismo , Mitocôndrias/metabolismo , Neurônios Motores/metabolismo , Nicotinamida-Nucleotídeo Adenililtransferase/metabolismo , Degeneração Walleriana/genética , Animais , Animais Geneticamente Modificados , Axônios/patologia , Axotomia , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Demência Frontotemporal/genética , Demência Frontotemporal/patologia , Gânglios Espinais/metabolismo , Gânglios Espinais/fisiopatologia , Humanos , Camundongos , Mitocôndrias/patologia , Neurônios Motores/patologia , Proteínas do Tecido Nervoso/genética , Doenças Neurodegenerativas/genética , Doenças Neurodegenerativas/patologia , Nicotinamida-Nucleotídeo Adenililtransferase/genética , Degeneração Walleriana/metabolismo , Degeneração Walleriana/patologia
7.
J Neurosci ; 33(34): 13882-7, 2013 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-23966707

RESUMO

Injury to the CNS leads to formation of scar tissue, which is important in sealing the lesion and inhibiting axon regeneration. The fibrotic scar that comprises a dense extracellular matrix is thought to originate from meningeal cells surrounding the CNS. However, using transgenic mice, we demonstrate that perivascular collagen1α1 cells are the main source of the cellular composition of the fibrotic scar after contusive spinal cord injury in which the dura remains intact. Using genetic lineage tracing, light sheet fluorescent microscopy, and antigenic profiling, we identify collagen1α1 cells as perivascular fibroblasts that are distinct from pericytes. Our results identify collagen1α1 cells as a novel source of the fibrotic scar after spinal cord injury and shift the focus from the meninges to the vasculature during scar formation.


Assuntos
Cicatriz/etiologia , Fibroblastos/patologia , Pericitos/patologia , Traumatismos da Medula Espinal/complicações , Análise de Variância , Animais , Antígenos/genética , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Antígenos CD13/metabolismo , Contagem de Células , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Modelos Animais de Doenças , Progressão da Doença , Feminino , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Fluorescência Verde/genética , Lectinas , Antígenos Comuns de Leucócito , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pericitos/metabolismo , Piperidinas/metabolismo , Proteoglicanas/genética , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Traumatismos da Medula Espinal/patologia , Fatores de Tempo , Uracila/análogos & derivados , Uracila/metabolismo
8.
Exp Neurol ; 377: 114810, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38714284

RESUMO

Most projection neurons, including retinal ganglion cells (RGCs), undergo cell death after axotomy proximal to the cell body. Specific RGC subtypes, such as ON-OFF direction selective RGCs (ooDSGCs) are particularly vulnerable, whereas intrinsically photosensitive RGCs (ipRGCs) exhibit resilience to axonal injury. Through the application of RNA sequencing and fluorescent in situ hybridization, we show that the expression of chloride intracellular channel protein 1 and 4 (Clic1 and Clic4) are highly increased in the ooDSGCs after axonal injury. Toward determining a gene's role in RGCs, we optimized the utility and efficacy of adenovirus associated virus (AAV)-retro expressing short hairpin RNA (shRNA). Injection of AAV2-retro into the superior colliculus results in efficient shRNA expression in RGCs. Incorporating histone H2B gene fused with mGreenLantern results in bright nuclear reporter expression, thereby enhancing single RGC identification and cell quantitation in live retinas. Lastly, we demonstrate that AAV2-retro mediated knockdown of both Clic1 and Clic4 promotes RGC survival after injury. Our findings establish an integrated use of AAV2-retro-shRNA and real-time fundus imaging and reveal CLICs' contribution to RGC death.


Assuntos
Morte Celular , Canais de Cloreto , Dependovirus , Células Ganglionares da Retina , Animais , Células Ganglionares da Retina/metabolismo , Dependovirus/genética , Canais de Cloreto/genética , Canais de Cloreto/metabolismo , Morte Celular/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Masculino , RNA Interferente Pequeno/genética
9.
bioRxiv ; 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38854133

RESUMO

The ability of neurons to sense and respond to damage is fundamental to homeostasis and nervous system repair. For some cell types, notably dorsal root ganglia (DRG) and retinal ganglion cells (RGCs), extensive profiling has revealed a large transcriptional response to axon injury that determines survival and regenerative outcomes. In contrast, the injury response of most supraspinal cell types, whose limited regeneration constrains recovery from spinal injury, is mostly unknown. Here we employed single-nuclei sequencing in mice to profile the transcriptional responses of diverse supraspinal cell types to spinal injury. Surprisingly, thoracic spinal injury triggered only modest changes in gene expression across all populations, including corticospinal tract (CST) neurons. Moreover, CST neurons also responded minimally to cervical injury but much more strongly to intracortical axotomy, including upregulation of numerous regeneration and apoptosis-related transcripts shared with injured DRG and RGC neurons. Thus, the muted response of CST neuron to spinal injury is linked to the injury's distal location, rather than intrinsic cellular characteristics. More broadly, these findings indicate that a central challenge for enhancing regeneration after a spinal injury is the limited sensing of distant injuries and the subsequent modest baseline neuronal response.

10.
J Biol Chem ; 286(42): 36492-9, 2011 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-21865157

RESUMO

Acetylcholinesterase (AChE) is highly expressed at sites of nerve-muscle contact where it is regulated at both the transcriptional and post-transcriptional levels. Our understanding of the molecular mechanisms underlying its regulation is incomplete, but they appear to involve both translational and post-translational events as well. Here, we show that Pumilio-2 (PUM2), an RNA binding translational repressor, is highly localized at the neuromuscular junction where AChE mRNA concentrates. Immunoprecipitation of muscle cell extracts with a PUM2 specific antibody precipitated AChE mRNA, suggesting that PUM2 binds to the AChE transcripts in a complex. Gel shift assays using a bacterially expressed PUM2 RNA binding domain showed specific binding using wild type AChE 3'-UTR RNA segment that was abrogated by mutation of the consensus recognition site. Transfecting skeletal muscle cells with shRNAs specific for PUM2 up-regulated AChE expression, whereas overexpression of PUM2 decreased AChE activity. We conclude that PUM2 binds to AChE mRNA and regulates AChE expression translationally at the neuromuscular synapse. Finally, we found that PUM2 is regulated by the motor nerve suggesting a trans-synaptic mechanism for locally regulating translation of specific proteins involved in modulating synaptic transmission, analogous to CNS synapses.


Assuntos
Regiões 3' não Traduzidas/fisiologia , Acetilcolinesterase/biossíntese , Músculo Esquelético/metabolismo , Junção Neuromuscular/metabolismo , Biossíntese de Proteínas/fisiologia , Proteínas de Ligação a RNA/metabolismo , Transmissão Sináptica/fisiologia , Acetilcolinesterase/genética , Animais , Regulação Enzimológica da Expressão Gênica/fisiologia , Camundongos , Junção Neuromuscular/genética , Ligação Proteica , Codorniz , Proteínas de Ligação a RNA/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Regulação para Cima/fisiologia
11.
eNeuro ; 9(3)2022.
Artigo em Inglês | MEDLINE | ID: mdl-35610024

RESUMO

Because of their ease of use, adeno-associated viruses (AAVs) are indispensable tools for much of neuroscience. Yet AAVs have been used relatively little to study the identities and connectivity of peripheral sensory neurons, principally because methods to selectively target peripheral neurons have been limited. The introduction of the AAV-PHP.S capsid with enhanced tropism for peripheral neurons (Chan et al., 2017) offered a solution, which we further elaborate here. Using AAV-PHP.S with GFP or mScarlet fluorescent proteins, we show that the mouse sensory ganglia for cranial nerves V, VII, IX, and X are targeted. Pseudounipolar neurons of both somatic and visceral origin, but not satellite glia, express the reporters. One week after virus injection, ≈66% of geniculate ganglion neurons were transduced. Fluorescent reporters were transported along the central and peripheral axons of these sensory neurons, permitting visualization of terminals at high resolution, and in intact, cleared brain using light sheet microscopy. Further, using a Cre-dependent reporter, we demonstrate by anatomic and functional criteria, that expression is in a cell type-selective manner. Finally, we integrate earlier neuroanatomical and molecular data with in vivo Ca2+ imaging to demonstrate the sensory characteristics of geniculate ganglion auricular neurons, which were previously undocumented. Our analyses suggest that the AAV-PHP.S serotype will be a powerful tool for anatomically and functionally mapping the receptive fields and circuits of the expanding numbers of molecular subtypes of many somatosensory and viscerosensory neurons that continue to be defined via single-cell RNA sequencing.


Assuntos
Dependovirus , Gânglios Espinais , Animais , Dependovirus/genética , Gânglios Sensitivos , Gânglios Espinais/metabolismo , Vetores Genéticos , Camundongos , Regiões Promotoras Genéticas , Células Receptoras Sensoriais
12.
Elife ; 112022 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-35838234

RESUMO

The supraspinal connectome is essential for normal behavior and homeostasis and consists of numerous sensory, motor, and autonomic projections from brain to spinal cord. Study of supraspinal control and its restoration after damage has focused mostly on a handful of major populations that carry motor commands, with only limited consideration of dozens more that provide autonomic or crucial motor modulation. Here, we assemble an experimental workflow to rapidly profile the entire supraspinal mesoconnectome in adult mice and disseminate the output in a web-based resource. Optimized viral labeling, 3D imaging, and registration to a mouse digital neuroanatomical atlas assigned tens of thousands of supraspinal neurons to 69 identified regions. We demonstrate the ability of this approach to clarify essential points of topographic mapping between spinal levels, measure population-specific sensitivity to spinal injury, and test the relationships between region-specific neuronal sparing and variability in functional recovery. This work will spur progress by broadening understanding of essential but understudied supraspinal populations.


Assuntos
Conectoma , Traumatismos da Medula Espinal , Traumatismos da Coluna Vertebral , Animais , Encéfalo , Camundongos , Recuperação de Função Fisiológica , Medula Espinal
13.
Exp Neurol ; 355: 114147, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35738417

RESUMO

Following injury in the central nervous system, a population of astrocytes occupy the lesion site, form glial bridges and facilitate axon regeneration. These astrocytes originate primarily from resident astrocytes or NG2+ oligodendrocyte progenitor cells. However, the extent to which these cell types give rise to the lesion-filling astrocytes, and whether the astrocytes derived from different cell types contribute similarly to optic nerve regeneration remain unclear. Here we examine the distribution of astrocytes and NG2+ cells in an optic nerve crush model. We show that optic nerve astrocytes partially fill the injury site over time after a crush injury. Viral mediated expression of a growth-promoting factor, ciliary neurotrophic factor (CNTF), in retinal ganglion cells (RGCs) promotes axon regeneration without altering the lesion size or the degree of lesion-filling GFAP+ cells. Strikingly, using inducible NG2CreER driver mice, we found that CNTF overexpression in RGCs increases the occupancy of NG2+ cell-derived astrocytes in the optic nerve lesion. An EdU pulse-chase experiment shows that the increase in NG2 cell-derived astrocytes is not due to an increase in cell proliferation. Lastly, we performed RNA-sequencing on the injured optic nerve and reveal that CNTF overexpression in RGCs results in significant changes in the expression of distinct genes, including those that encode chemokines, growth factor receptors, and immune cell modulators. Even though CNTF-induced axon regeneration has long been recognized, this is the first evidence of this procedure affecting glial cell fate at the optic nerve crush site. We discuss possible implication of these results for axon regeneration.


Assuntos
Traumatismos do Nervo Óptico , Traumatismos do Sistema Nervoso , Animais , Astrócitos/metabolismo , Axônios/patologia , Fator Neurotrófico Ciliar , Citocinas/metabolismo , Camundongos , Regeneração Nervosa/fisiologia , Traumatismos do Nervo Óptico/patologia , Células Ganglionares da Retina/metabolismo , Traumatismos do Sistema Nervoso/metabolismo
14.
Exp Neurol ; 346: 113862, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34520726

RESUMO

The supraspinal connectome consists of dozens of neuronal populations that project axons from the brain to the spinal cord to influence a wide range of motor, autonomic, and sensory functions. The complexity and wide distribution of supraspinal neurons present significant technical challenges, leading most spinal cord injury research to focus on a handful of major pathways such as the corticospinal, rubrospinal, and raphespinal. Much less is known about many additional populations that carry information to modulate or compensate for these main pathways, or which carry pre-autonomic and other information of high value to individuals with spinal injury. A confluence of technical developments, however, now enables a whole-connectome study of spinal cord injury. Improved viral labeling, tissue clearing, and automated registration to 3D atlases can quantify supraspinal neurons throughout the murine brain, offering a practical means to track responses to injury and treatment on an unprecedented scale. Here we discuss the need for expanded connectome-wide analyses in spinal injury research, illustrate the potential by discussing a new web-based resource for brain-wide study of supraspinal neurons, and highlight future prospects for connectome analyses.


Assuntos
Pesquisa Biomédica/tendências , Conectoma/tendências , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/metabolismo , Medula Espinal/metabolismo , Transcriptoma/fisiologia , Animais , Pesquisa Biomédica/métodos , Conectoma/métodos , Humanos , Medula Espinal/patologia
15.
J Exp Med ; 218(8)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34132743

RESUMO

The wound healing process that occurs after spinal cord injury is critical for maintaining tissue homeostasis and limiting tissue damage, but eventually results in a scar-like environment that is not conducive to regeneration and repair. A better understanding of this dichotomy is critical to developing effective therapeutics that target the appropriate pathobiology, but a major challenge has been the large cellular heterogeneity that results in immensely complex cellular interactions. In this study, we used single-cell RNA sequencing to assess virtually all cell types that comprise the mouse spinal cord injury site. In addition to discovering novel subpopulations, we used expression values of receptor-ligand pairs to identify signaling pathways that are predicted to regulate specific cellular interactions during angiogenesis, gliosis, and fibrosis. Our dataset is a valuable resource that provides novel mechanistic insight into the pathobiology of not only spinal cord injury but also other traumatic disorders of the CNS.


Assuntos
Comunicação Celular , Análise de Célula Única , Traumatismos da Medula Espinal/patologia , Angiopoietinas/metabolismo , Animais , Astrócitos/metabolismo , Quimiotaxia , Feminino , Fibroblastos/metabolismo , Fibrose , Gliose/complicações , Gliose/patologia , Inflamação/patologia , Interleucina-6/metabolismo , Ligantes , Macrófagos/patologia , Camundongos Endogâmicos C57BL , Células Mieloides/patologia , Neuroglia/patologia , Oncostatina M/metabolismo , Receptores de Oncostatina M/metabolismo , Transdução de Sinais , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/imunologia , Fatores de Tempo , Transcriptoma/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Biochem Biophys Res Commun ; 393(4): 812-7, 2010 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-20171177

RESUMO

Neutrotrophin-3 (NT3) plays a protective role in injured central nervous system tissues through interaction with trk receptors. To enhance the regeneration of damaged tissue, a combination therapy with cell transplantation and neurotrophins has been under development. We examined whether the transplantation of neural progenitor cells (NPCs) secreting NT3/D15A, a multi-neurotrophin with the capacity to bind both trkB and trkC, would enhance the repair of damaged tissues and the functional recovery in a chronic phase of spinal cord injury. The cultured NPCs with lentiviral vector containing either GFP or NT3/D15A were transplanted into the contused spinal cord at 6 weeks after the initial thoracic injury. Eight weeks after the transplantation, the NT3/D15A transplants displayed better survival than the GFP transplants, and they exhibited enhanced myelin formation and partial improvement of hindlimb function. Our study revealed that NT3/D15A produced positive effects in injured spinal cords even in the chronic phase. These effects suggest an enhanced neurotrophin-trk signaling by NT3/D15A.


Assuntos
Bainha de Mielina/metabolismo , Neurônios/metabolismo , Neurônios/transplante , Neurotrofina 3/biossíntese , Traumatismos da Medula Espinal/cirurgia , Transplante de Células-Tronco/métodos , Células-Tronco/metabolismo , Animais , Encéfalo/metabolismo , Encéfalo/fisiopatologia , Sobrevivência de Enxerto , Membro Posterior/fisiologia , Humanos , Regeneração Nervosa , Neurotrofina 3/genética , Ratos , Ratos Endogâmicos F344 , Medula Espinal/metabolismo , Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/fisiopatologia , Coloração e Rotulagem , Transfecção
17.
Restor Neurol Neurosci ; 25(1): 65-76, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17473396

RESUMO

PURPOSE: A proliferation of stem/progenitor cells is observed after brain injury. We examined the regional and temporal profile of mitotically active cells to determine whether traumatic brain injury (TBI) would increase neurogenesis in selective brain regions. METHODS: Male Sprague-Dawley rats received injections (IP) of 5-bromo-deoxyuridine (BrdU), a compound used to detect mitotic cells, before and after fluid-percussion brain injury. At 3 hr, 1, 2, 3, 7, and 14 days after moderate fluid percussion, brains were processed for immunocytochemical and confocal analysis. Sections were double-labeled for markers selective for neurons (NeuN), astrocytes (GFAP), olidgodendrocytes (CNPase and MBP) and macrophage/microglia (ED1). RESULTS: At 3 hr post-trauma, the majority of BrdU labeled cells were associated with the subventricular zone of the traumatized hemisphere. At later time points, a significant increase in BrdU positive cells was observed throughout the traumatized cerebral cortex, hippocampus, white matter structures, and some contralateral regions. BrdU labeled cells were observed as late as 14 days post-injury. Double-label studies with confocal microscopy demonstrated that cell phenotypes including astrocytes, macrophage/microglia, oligodendrocytes, and neurons were BrdU positive with the majority of cells appearing glial in nature. Evidence for neurogenesis was seen in the granular cell layer of the hippocampus. CONCLUSION: These findings indicate that TBI stimulates widespread cellular proliferation for days after injury and results in focal neurogenesis in the dentate gyrus of the hippocampus. These cellular responses to injury may participate in brain repair and functional recovery.


Assuntos
Lesões Encefálicas/patologia , Lesões Encefálicas/fisiopatologia , Proliferação de Células , Neurônios/fisiologia , Organogênese/fisiologia , Análise de Variância , Animais , Encéfalo/patologia , Bromodesoxiuridina/metabolismo , Contagem de Células , Modelos Animais de Doenças , Ectodisplasinas/metabolismo , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Proteína Básica da Mielina/metabolismo , Fosfopiruvato Hidratase/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo
18.
eNeuro ; 4(4)2017.
Artigo em Inglês | MEDLINE | ID: mdl-28856242

RESUMO

Retinal ganglion cells (RGCs), the sole output cells of the retina, are a heterogeneous population of neurons that project axons to visual targets in the brain. Like most CNS neurons, RGCs are considered incapable of mounting long distance axon regeneration. Using immunolabeling-enabled 3D imaging of solvent-cleared organs (iDISCO) in transgenic mice, we tracked the entire paths of individual RGC axons and show that adult RGCs are highly capable of spontaneous long-distance regeneration, even without any treatment. Our results show that the Thy1-H-YFP mouse sparsely labels RGCs, consisting predominantly of regeneration-competent α-type RGCs (αRGCs). Following optic nerve crush, many of the YFP-labeled RGC axons extend considerable distances proximal to the injury site with only a few penetrating through the lesion. This tortuous axon growth proximal to the lesion site is even more striking with intravitreal ciliary neurotrophic factor (CNTF) treatment. We further demonstrate that despite traveling more than 5 mm (i.e., a distance equal to the length of mouse optic nerve), many of these circuitous axons are confined to the injury area and fail to reach the brain. Our results re-evaluate the view that RGCs are naturally incapable of re-extending long axons, and shift the focus from promoting axon elongation, to understanding factors that prevent direct growth of axons through the lesion and the injured nerve.


Assuntos
Regeneração Nervosa , Células Ganglionares da Retina/patologia , Animais , Axônios/fisiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Crescimento Celular , Fator Neurotrófico Ciliar/administração & dosagem , Fator Neurotrófico Ciliar/metabolismo , Feminino , Imageamento Tridimensional , Imuno-Histoquímica , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Masculino , Camundongos Transgênicos , Regeneração Nervosa/fisiologia , Nervo Óptico/patologia , Nervo Óptico/fisiopatologia , Traumatismos do Nervo Óptico/patologia , Traumatismos do Nervo Óptico/fisiopatologia , Células Ganglionares da Retina/fisiologia
19.
Nat Commun ; 8(1): 1963, 2017 12 06.
Artigo em Inglês | MEDLINE | ID: mdl-29213073

RESUMO

Neural circuitry in the lumbar spinal cord governs two principal features of locomotion, rhythm and pattern, which reflect intra- and interlimb movement. These features are functionally organized into a hierarchy that precisely controls stepping in a stereotypic, speed-dependent fashion. Here, we show that a specific component of the locomotor pattern can be independently manipulated. Silencing spinal L2 interneurons that project to L5 selectively disrupts hindlimb alternation allowing a continuum of walking to hopping to emerge from the otherwise intact network. This perturbation, which is independent of speed and occurs spontaneously with each step, does not disrupt multi-joint movements or forelimb alternation, nor does it translate to a non-weight-bearing locomotor activity. Both the underlying rhythm and the usual relationship between speed and spatiotemporal characteristics of stepping persist. These data illustrate that hindlimb alternation can be manipulated independently from other core features of stepping, revealing a striking freedom in an otherwise precisely controlled system.


Assuntos
Membro Posterior/inervação , Membro Posterior/fisiologia , Interneurônios/fisiologia , Rede Nervosa/fisiologia , Medula Espinal/fisiologia , Animais , Fenômenos Biomecânicos , Contagem de Células , Eletromiografia , Feminino , Membro Anterior/inervação , Membro Anterior/fisiologia , Locomoção/fisiologia , Modelos Animais , Neurônios Motores/fisiologia , Músculo Esquelético/inervação , Músculo Esquelético/fisiopatologia , Vias Neurais/fisiologia , Ratos , Ratos Sprague-Dawley , Análise Espaço-Temporal , Traumatismos da Medula Espinal/fisiopatologia , Sinapses/fisiologia , Caminhada/fisiologia , Velocidade de Caminhada/fisiologia
20.
PLoS One ; 12(10): e0186091, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29049317

RESUMO

In jawed vertebrates, oligodendrocytes (OLs) are the myelin-producing glial cells responsible for ensheathment of axons within the central nervous system and are also crucial for remyelination following injury or disease. Olig2 is a crucial factor in the specification and differentiation of oligodendrocyte precursor cells (OPCs) that give rise to mature, myelin-producing OLs in the developing and postnatal CNS; however, its role in adulthood is less well understood. To investigate the role Olig2 plays in regulating gene expression in the adult OL lineage in a physiologically-relevant context, we performed chromatin immunoprecipitation followed by next generation sequencing analysis (ChIP-Seq) using whole spinal cord tissue harvested from adult mice. We found that many of the Olig2-bound sites were associated with genes with biological processes corresponding to OL differentiation (Nkx2.2, Nkx6.2, and Sip1), myelination and ensheathment (Mbp, Cldn11, and Mobp), as well as cell cycle and cytoskeletal regulation. This suggests Olig2 continues to play a critical role in processes related to OL differentiation and myelination well into adulthood.


Assuntos
Genoma , Fator de Transcrição 2 de Oligodendrócitos/genética , Medula Espinal/metabolismo , Animais , Imunoprecipitação da Cromatina , Proteína Homeobox Nkx-2.2 , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Bainha de Mielina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA