Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 14(1): 8367, 2024 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-38600221

RESUMO

Post-traumatic epilepsy (PTE) stands as one of the numerous debilitating consequences that follow traumatic brain injury (TBI). Despite its impact on many individuals, the current landscape offers only a limited array of reliable treatment options, and our understanding of the underlying mechanisms and susceptibility factors remains incomplete. Among the potential contributors to epileptogenesis, astrocytes, a type of glial cell, have garnered substantial attention as they are believed to promote hyperexcitability and the development of seizures in the brain following TBI. The current study evaluated the transcriptomic changes in cortical astrocytes derived from animals that developed seizures as a result of severe focal TBI. Using RNA-Seq and ingenuity pathway analysis (IPA), we unveil a distinct gene expression profile in astrocytes, including alterations in genes supporting inflammation, early response modifiers, and neuropeptide-amidating enzymes. The findings underscore the complex molecular dynamics in astrocytes during PTE development, offering insights into therapeutic targets and avenues for further exploration.


Assuntos
Lesões Encefálicas Traumáticas , Epilepsia Pós-Traumática , Humanos , Animais , Epilepsia Pós-Traumática/etiologia , Astrócitos/metabolismo , Transcriptoma , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/metabolismo , Convulsões , Perfilação da Expressão Gênica , Modelos Animais de Doenças
2.
Commun Biol ; 7(1): 373, 2024 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-38548965

RESUMO

Astrocytes in the retrotrapezoid nucleus (RTN) stimulate breathing in response to CO2/H+, however, it is not clear how these cells detect changes in CO2/H+. Considering Kir4.1/5.1 channels are CO2/H+-sensitive and important for several astrocyte-dependent processes, we consider Kir4.1/5.1 a leading candidate CO2/H+ sensor in RTN astrocytes. To address this, we show that RTN astrocytes express Kir4.1 and Kir5.1 transcripts. We also characterized respiratory function in astrocyte-specific inducible Kir4.1 knockout mice (Kir4.1 cKO); these mice breathe normally under room air conditions but show a blunted ventilatory response to high levels of CO2, which could be partly rescued by viral mediated re-expression of Kir4.1 in RTN astrocytes. At the cellular level, astrocytes in slices from astrocyte-specific inducible Kir4.1 knockout mice are less responsive to CO2/H+ and show a diminished capacity for paracrine modulation of respiratory neurons. These results suggest Kir4.1/5.1 channels in RTN astrocytes contribute to respiratory behavior.


Assuntos
Astrócitos , Dióxido de Carbono , Camundongos , Animais , Astrócitos/fisiologia , Respiração , Neurônios/fisiologia , Camundongos Knockout
3.
Int J Mol Sci ; 25(5)2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38474127

RESUMO

Traumatic brain injury (TBI) can lead to post-traumatic epilepsy (PTE). Blast TBI (bTBI) found in Veterans presents with several complications, including cognitive and behavioral disturbances and PTE; however, the underlying mechanisms that drive the long-term sequelae are not well understood. Using an unbiased proteomics approach in a mouse model of repeated bTBI (rbTBI), this study addresses this gap in the knowledge. After rbTBI, mice were monitored using continuous, uninterrupted video-EEG for up to four months. Following this period, we collected cortex and hippocampus tissues from three groups of mice: those with post-traumatic epilepsy (PTE+), those without epilepsy (PTE-), and the control group (sham). Hundreds of differentially expressed proteins were identified in the cortex and hippocampus of PTE+ and PTE- relative to sham. Focusing on protein pathways unique to PTE+, pathways related to mitochondrial function, post-translational modifications, and transport were disrupted. Computational metabolic modeling using dysregulated protein expression predicted mitochondrial proton pump dysregulation, suggesting electron transport chain dysregulation in the epileptic tissue relative to PTE-. Finally, data mining enabled the identification of several novel and previously validated TBI and epilepsy biomarkers in our data set, many of which were found to already be targeted by drugs in various phases of clinical testing. These findings highlight novel proteins and protein pathways that may drive the chronic PTE sequelae following rbTBI.


Assuntos
Lesões Encefálicas Traumáticas , Epilepsia Pós-Traumática , Epilepsia , Camundongos , Animais , Epilepsia Pós-Traumática/complicações , Proteômica , Epilepsia/complicações , Córtex Cerebral
4.
J Neurosci ; 44(12)2024 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-38360749

RESUMO

While originally identified as an antiviral pathway, recent work has implicated that cyclic GMP-AMP-synthase-Stimulator of Interferon Genes (cGAS-STING) signaling is playing a critical role in the neuroinflammatory response to traumatic brain injury (TBI). STING activation results in a robust inflammatory response characterized by the production of inflammatory cytokines called interferons, as well as hundreds of interferon stimulated genes (ISGs). Global knock-out (KO) mice inhibiting this pathway display neuroprotection with evidence that this pathway is active days after injury; yet, the early neuroinflammatory events stimulated by STING signaling remain understudied. Furthermore, the source of STING signaling during brain injury is unknown. Using a murine controlled cortical impact (CCI) model of TBI, we investigated the peripheral immune and microglial response to injury utilizing male chimeric and conditional STING KO animals, respectively. We demonstrate that peripheral and microglial STING signaling contribute to negative outcomes in cortical lesion volume, cell death, and functional outcomes postinjury. A reduction in overall peripheral immune cell and neutrophil infiltration at the injury site is STING dependent in these models at 24 h. Transcriptomic analysis at 2 h, when STING is active, reveals that microglia drive an early, distinct transcriptional program to elicit proinflammatory genes including interleukin 1-ß (IL-1ß), which is lost in conditional knock-out mice. The upregulation of alternative innate immune pathways also occurs after injury in these animals, which supports a complex relationship between brain-resident and peripheral immune cells to coordinate the proinflammatory response and immune cell influx to damaged tissue after injury.


Assuntos
Lesões Encefálicas Traumáticas , Microglia , Animais , Masculino , Camundongos , Lesões Encefálicas Traumáticas/patologia , Citocinas/metabolismo , Interferons/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microglia/metabolismo , Transdução de Sinais
5.
J Neuroinflammation ; 21(1): 41, 2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38310257

RESUMO

Monocytes represent key cellular elements that contribute to the neurological sequela following brain injury. The current study reveals that trauma induces the augmented release of a transcriptionally distinct CD115+/Ly6Chi monocyte population into the circulation of mice pre-exposed to clodronate depletion conditions. This phenomenon correlates with tissue protection, blood-brain barrier stability, and cerebral blood flow improvement. Uniquely, this shifted the innate immune cell profile in the cortical milieu and reduced the expression of pro-inflammatory Il6, IL1r1, MCP-1, Cxcl1, and Ccl3 cytokines. Monocytes that emerged under these conditions displayed a morphological and gene profile consistent with a subset commonly seen during emergency monopoiesis. Single-cell RNA sequencing delineated distinct clusters of monocytes and revealed a key transcriptional signature of Ly6Chi monocytes enriched for Apoe and chitinase-like protein 3 (Chil3/Ym1), commonly expressed in pro-resolving immunoregulatory monocytes, as well as granule genes Elane, Prtn3, MPO, and Ctsg unique to neutrophil-like monocytes. The predominate shift in cell clusters included subsets with low expression of transcription factors involved in monocyte conversion, Pou2f2, Na4a1, and a robust enrichment of genes in the oxidative phosphorylation pathway which favors an anti-inflammatory phenotype. Transfer of this monocyte assemblage into brain-injured recipient mice demonstrated their direct role in neuroprotection. These findings reveal a multifaceted innate immune response to brain injury and suggest targeting surrogate monocyte subsets may foster tissue protection in the brain.


Assuntos
Lesões Encefálicas , Monócitos , Camundongos , Animais , Monócitos/metabolismo , Neutrófilos/metabolismo , Lesões Encefálicas/metabolismo , Encéfalo/metabolismo , Perfilação da Expressão Gênica , Catepsina G/metabolismo
6.
Artigo em Inglês | MEDLINE | ID: mdl-38316554

RESUMO

In addition to their many functions in the healthy central nervous system (CNS), astrocytes respond to CNS damage and disease through a process called "reactivity." Recent evidence reveals that astrocyte reactivity is a heterogeneous spectrum of potential changes that occur in a context-specific manner. These changes are determined by diverse signaling events and vary not only with the nature and severity of different CNS insults but also with location in the CNS, genetic predispositions, age, and potentially also with "molecular memory" of previous reactivity events. Astrocyte reactivity can be associated with both essential beneficial functions as well as with harmful effects. The available information is rapidly expanding and much has been learned about molecular diversity of astrocyte reactivity. Emerging functional associations point toward central roles for astrocyte reactivity in determining the outcome in CNS disorders.

7.
Brain ; 147(5): 1856-1870, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38146224

RESUMO

Alterations in the extracellular matrix are common in patients with epilepsy and animal models of epilepsy, yet whether they are the cause or consequence of seizures and epilepsy development is unknown. Using Theiler's murine encephalomyelitis virus (TMEV) infection-induced model of acquired epilepsy, we found de novo expression of chondroitin sulfate proteoglycans (CSPGs), a major extracellular matrix component, in dentate gyrus (DG) and amygdala exclusively in mice with acute seizures. Preventing the synthesis of CSPGs specifically in DG and amygdala by deletion of the major CSPG aggrecan reduced seizure burden. Patch-clamp recordings from dentate granule cells revealed enhanced intrinsic and synaptic excitability in seizing mice that was significantly ameliorated by aggrecan deletion. In situ experiments suggested that dentate granule cell hyperexcitability results from negatively charged CSPGs increasing stationary cations on the membrane, thereby depolarizing neurons, increasing their intrinsic and synaptic excitability. These results show increased expression of CSPGs in the DG and amygdala as one of the causal factors for TMEV-induced acute seizures. We also show identical changes in CSPGs in pilocarpine-induced epilepsy, suggesting that enhanced CSPGs in the DG and amygdala may be a common ictogenic factor and potential therapeutic target.


Assuntos
Tonsila do Cerebelo , Proteoglicanas de Sulfatos de Condroitina , Giro Denteado , Convulsões , Animais , Giro Denteado/metabolismo , Tonsila do Cerebelo/metabolismo , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Camundongos , Convulsões/metabolismo , Masculino , Theilovirus , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Camundongos Knockout , Agrecanas/metabolismo , Neurônios/metabolismo
8.
bioRxiv ; 2023 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-37292901

RESUMO

Alterations in the extracellular matrix (ECM) are common in epilepsy, yet whether they are cause or consequence of disease is unknow. Using Theiler's virus infection model of acquired epilepsy we find de novo expression of chondroitin sulfate proteoglycans (CSPGs), a major ECM component, in dentate gyrus (DG) and amygdala exclusively in mice with seizures. Preventing synthesis of CSPGs specifically in DG and amygdala by deletion of major CSPG aggrecan reduced seizure burden. Patch-clamp recordings from dentate granule cells (DGCs) revealed enhanced intrinsic and synaptic excitability in seizing mice that was normalized by aggrecan deletion. In situ experiments suggest that DGCs hyperexcitability results from negatively charged CSPGs increasing stationary cations (K+, Ca2+) on the membrane thereby depolarizing neurons, increasing their intrinsic and synaptic excitability. We show similar changes in CSPGs in pilocarpine-induced epilepsy suggesting enhanced CSPGs in the DG and amygdala may be a common ictogenic factor and novel therapeutic potential.

9.
Cells ; 12(9)2023 04 25.
Artigo em Inglês | MEDLINE | ID: mdl-37174647

RESUMO

BACKGROUND: Traumatic brain injury (TBI) remains a significant risk factor for post-traumatic epilepsy (PTE). The pathophysiological mechanisms underlying the injury-induced epileptogenesis are under investigation. The dentate gyrus-a structure that is highly susceptible to injury-has been implicated in the evolution of seizure development. METHODS: Utilizing the murine unilateral focal control cortical impact (CCI) injury, we evaluated seizure onset using 24/7 EEG video analysis at 2-4 months post-injury. Cellular changes in the dentate gyrus and hilus of the hippocampus were quantified by unbiased stereology and Imaris image analysis to evaluate Prox1-positive cell migration, astrocyte branching, and morphology, as well as neuronal loss at four months post-injury. Isolation of region-specific astrocytes and RNA-Seq were performed to determine differential gene expression in animals that developed post-traumatic epilepsy (PTE+) vs. those animals that did not (PTE-), which may be associated with epileptogenesis. RESULTS: CCI injury resulted in 37% PTE incidence, which increased with injury severity and hippocampal damage. Histological assessments uncovered a significant loss of hilar interneurons that coincided with aberrant migration of Prox1-positive granule cells and reduced astroglial branching in PTE+ compared to PTE- mice. We uniquely identified Cst3 as a PTE+-specific gene signature in astrocytes across all brain regions, which showed increased astroglial expression in the PTE+ hilus. CONCLUSIONS: These findings suggest that epileptogenesis may emerge following TBI due to distinct aberrant cellular remodeling events and key molecular changes in the dentate gyrus of the hippocampus.


Assuntos
Lesões Encefálicas Traumáticas , Epilepsia Pós-Traumática , Camundongos , Animais , Epilepsia Pós-Traumática/etiologia , Epilepsia Pós-Traumática/patologia , Gliose/complicações , Lesões Encefálicas Traumáticas/complicações , Convulsões , Interneurônios/metabolismo
10.
JCI Insight ; 7(15)2022 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-35737458

RESUMO

Circulating monocytes have emerged as key regulators of the neuroinflammatory milieu in a number of neuropathological disorders. Ephrin type A receptor 4 (Epha4) receptor tyrosine kinase, a prominent axon guidance molecule, has recently been implicated in the regulation of neuroinflammation. Using a mouse model of brain injury and a GFP BM chimeric approach, we found neuroprotection and a lack of significant motor deficits marked by reduced monocyte/macrophage cortical infiltration and an increased number of arginase-1+ cells in the absence of BM-derived Epha4. This was accompanied by a shift in monocyte gene profile from pro- to antiinflammatory that included increased Tek (Tie2 receptor) expression. Inhibition of Tie2 attenuated enhanced expression of M2-like genes in cultured Epha4-null monocytes/macrophages. In Epha4-BM-deficient mice, cortical-isolated GFP+ monocytes/macrophages displayed a phenotypic shift from a classical to an intermediate subtype, which displayed reduced Ly6chi concomitant with increased Ly6clo- and Tie2-expressing populations. Furthermore, clodronate liposome-mediated monocyte depletion mimicked these effects in WT mice but resulted in attenuation of phenotype in Epha4-BM-deficient mice. This demonstrates that monocyte polarization not overall recruitment dictates neural tissue damage. Thus, coordination of monocyte proinflammatory phenotypic state by Epha4 is a key regulatory step mediating brain injury.


Assuntos
Lesões Encefálicas , Monócitos , Humanos , Lesões Encefálicas/metabolismo , Efrinas/metabolismo , Monócitos/metabolismo , Fenótipo , Receptor EphB2/metabolismo , Animais , Camundongos
11.
Curr Opin Neurobiol ; 74: 102550, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35544965

RESUMO

Astrocytes represent an abundant type of glial cell involved in nearly every aspect of central nervous system (CNS) function, including synapse formation and maturation, ion and neurotransmitter homeostasis, blood-brain barrier maintenance, as well as neuronal metabolic support. These various functions are enabled by the morphological complexity that astrocytes adopt. Recent experimental advances in genetic and viral labeling, lineage tracing, and live- and ultrastructural imaging of miniscule astrocytic sub-compartments reveal a complex morphological heterogeneity that is based on the origin, local function, and environmental context in which astrocytes reside. In this minireview, we highlight recent findings that reveal the plastic nature of astrocytes in the healthy brain, particularly at the synapse, and emerging technologies that have advanced our understanding of these morphologically complex cells.


Assuntos
Astrócitos , Encéfalo , Astrócitos/metabolismo , Encéfalo/fisiologia , Neurogênese , Neuroglia , Sinapses
12.
Curr Neuropharmacol ; 20(1): 5-15, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33588730

RESUMO

General anesthetics are a mainstay of modern medicine, and although much progress has been made towards identifying molecular targets of anesthetics and neural networks contributing to endpoints of general anesthesia, our understanding of how anesthetics work remains unclear. Reducing this knowledge gap is of fundamental importance to prevent unwanted and life-threatening side-effects associated with general anesthesia. General anesthetics are chemically diverse, yet they all have similar behavioral endpoints, and so for decades, research has sought to identify a single underlying mechanism to explain how anesthetics work. However, this effort has given way to the 'multiple target hypothesis' as it has become clear that anesthetics target many cellular proteins, including GABAA receptors, glutamate receptors, voltage-independent K+ channels, and voltagedependent K+, Ca2+ and Na+ channels, to name a few. Yet, despite evidence that astrocytes are capable of modulating multiple aspects of neural function and express many anesthetic target proteins, they have been largely ignored as potential targets of anesthesia. The purpose of this brief review is to highlight the effects of anesthetic on astrocyte processes and identify potential roles of astrocytes in behavioral endpoints of anesthesia (hypnosis, amnesia, analgesia, and immobilization).


Assuntos
Anestésicos Gerais , Astrócitos , Anestesia Geral , Anestésicos Gerais/efeitos adversos , Humanos , Receptores de GABA-A
13.
Mol Cell Proteomics ; 21(1): 100180, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34808356

RESUMO

Alexander disease (AxD) is a rare and fatal neurodegenerative disorder caused by mutations in the gene encoding glial fibrillary acidic protein (GFAP). In this report, a mouse model of AxD (GFAPTg;Gfap+/R236H) was analyzed that contains a heterozygous R236H point mutation in murine Gfap as well as a transgene with a GFAP promoter to overexpress human GFAP. Using label-free quantitative proteomic comparisons of brain tissue from GFAPTg;Gfap+/R236H versus wild-type mice confirmed upregulation of the glutathione metabolism pathway and indicated proteins were elevated in the peroxisome proliferator-activated receptor (PPAR) signaling pathway, which had not been reported previously in AxD. Relative protein-level differences were confirmed by a targeted proteomics assay, including proteins related to astrocytes and oligodendrocytes. Of particular interest was the decreased level of the oligodendrocyte protein, 2-hydroxyacylsphingosine 1-beta-galactosyltransferase (Ugt8), since Ugt8-deficient mice exhibit a phenotype similar to GFAPTg;Gfap+/R236H mice (e.g., tremors, ataxia, hind-limb paralysis). In addition, decreased levels of myelin-associated proteins were found in the GFAPTg;Gfap+/R236H mice, consistent with the role of Ugt8 in myelin synthesis. Fabp7 upregulation in GFAPTg;Gfap+/R236H mice was also selected for further investigation due to its uncharacterized association to AxD, critical function in astrocyte proliferation, and functional ability to inhibit the anti-inflammatory PPAR signaling pathway in models of amyotrophic lateral sclerosis (ALS). Within Gfap+ astrocytes, Fabp7 was markedly increased in the hippocampus, a brain region subjected to extensive pathology and chronic reactive gliosis in GFAPTg;Gfap+/R236H mice. Last, to determine whether the findings in GFAPTg;Gfap+/R236H mice are present in the human condition, AxD patient and control samples were analyzed by Western blot, which indicated that Type I AxD patients have a significant fourfold upregulation of FABP7. However, immunohistochemistry analysis showed that UGT8 accumulates in AxD patient subpial brain regions where abundant amounts of Rosenthal fibers are located, which was not observed in the GFAPTg;Gfap+/R236H mice.


Assuntos
Doença de Alexander , Doença de Alexander/genética , Doença de Alexander/metabolismo , Doença de Alexander/patologia , Animais , Astrócitos/metabolismo , Modelos Animais de Doenças , Gliose/metabolismo , Gliose/patologia , Humanos , Camundongos , Camundongos Transgênicos , Mutação , Proteômica
14.
Glia ; 69(2): 310-325, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32865323

RESUMO

Astrocyte heterogeneity is an emerging concept in which astrocytes within or between brain regions show variable morphological and/or gene expression profiles that presumably reflect different functional roles. Recent evidence indicates that retrotrapezoid nucleus (RTN) astrocytes sense changes in tissue CO2/ H+ to regulate respiratory activity; however, mechanism(s) by which they do so remain unclear. Alterations in inward K+ currents represent a potential mechanism by which CO2 /H+ signals may be conveyed to neurons. Here, we use slice electrophysiology in rats of either sex to show that RTN astrocytes intrinsically respond to CO2 /H+ by inhibition of an inward rectifying potassium (Kir ) conductance and depolarization of the membrane, while cortical astrocytes do not exhibit such CO2 /H+ -sensitive properties. Application of Ba2+ mimics the effect of CO2 /H+ on RTN astrocytes as measured by reductions in astrocyte Kir -like currents and increased RTN neuronal firing. These CO2 /H+ -sensitive currents increase developmentally, in parallel to an increased expression in Kir 4.1 and Kir 5.1 in the brainstem. Finally, the involvement of Kir 5.1 in the CO2 /H+ -sensitive current was verified using a Kir5.1 KO rat. These data suggest that Kir inhibition by CO2 /H+ may govern the degree to which astrocytes mediate downstream chemoreceptive signaling events through cell-autonomous mechanisms. These results identify Kir channels as potentially important regional CO2 /H+ sensors early in development, thus expanding our understanding of how astrocyte heterogeneity may uniquely support specific neural circuits and behaviors.


Assuntos
Astrócitos , Animais , Encéfalo , Dióxido de Carbono/farmacologia , Células Quimiorreceptoras , Neurônios , Canais de Potássio Corretores do Fluxo de Internalização , Ratos , Canal Kir5.1
15.
J Neurophysiol ; 124(3): 740-749, 2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32727273

RESUMO

All inhalation anesthetics used clinically including isoflurane can suppress breathing; since this unwanted side effect can persist during the postoperative period and complicate patient recovery, there is a need to better understand how isoflurane affects cellular and molecular elements of respiratory control. Considering that astrocytes in a brainstem region known as the retrotrapezoid nucleus (RTN) contribute to the regulation of breathing in response to changes in CO2/H+ (i.e., function as respiratory chemoreceptors), and astrocytes in other brain regions are highly sensitive to isoflurane, we wanted to determine whether and how RTN astrocytes respond to isoflurane. We found that RTN astrocytes in slices from neonatal rat pups (7-12 days postnatal) respond to clinically relevant levels of isoflurane by inhibition of a CO2/H+-sensitive Kir4.1/5.1-like conductance [50% effective concentration (EC50) = 0.8 mM or ~1.7%]. We went on to confirm that similar levels of isoflurane (EC50 = 0.53 mM or 1.1%) inhibit recombinant Kir4.1/5.1 channels but not homomeric Kir4.1 channels expressed in HEK293 cells. We also found that exposure to CO2/H+ occluded subsequent effects of isoflurane on both native and recombinant Kir4.1/5.1 currents. These results identify Kir4.1/5.1 channels in astrocytes as novel targets of isoflurane. These results suggest astrocyte Kir4.1/5.1 channels contribute to certain aspects of general anesthesia including altered respiratory control.NEW & NOTEWORTHY An unwanted side effect of isoflurane anesthesia is suppression of breathing. Despite this clinical significance, effects of isoflurane on cellular and molecular elements of respiratory control are not well understood. Here, we show that isoflurane inhibits heteromeric Kir4.1/5.1 channels in a mammalian expression system and a Kir4.1/5.1-like conductance in astrocytes in a brainstem respiratory center. These results identify astrocyte Kir4.1/5.1 channels as novel targets of isoflurane and potential substrates for altered respiratory control during isoflurane anesthesia.


Assuntos
Anestésicos Inalatórios/farmacologia , Astrócitos/efeitos dos fármacos , Tronco Encefálico/efeitos dos fármacos , Células Quimiorreceptoras/efeitos dos fármacos , Isoflurano/farmacologia , Canais de Potássio Corretores do Fluxo de Internalização/efeitos dos fármacos , Fenômenos Fisiológicos Respiratórios/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Células HEK293 , Humanos , Ratos , Proteínas Recombinantes , Canal Kir5.1
16.
Glia ; 68(7): 1495-1512, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32068308

RESUMO

Kir4.1, a glial-specific inwardly rectifying potassium channel, is implicated in astrocytic maintenance of K+ homeostasis. Underscoring the role of Kir4.1 in central nervous system (CNS) functioning, genetic mutations in KCNJ10, the gene which encodes Kir4.1, causes seizures, ataxia and developmental disability in humans. Kir4.1 protein and mRNA loss are consistently observed in CNS injury and neurological diseases linked to hyperexcitability and neuronal dysfunction, leading to the notion that Kir4.1 represents an attractive therapeutic target. Despite this, little is understood regarding the mechanisms that underpin this downregulation. Previous work by our lab revealed that DNA hypomethylation of the Kcnj10 gene functions to regulate mRNA levels during astrocyte maturation whereas hypermethylation in vitro led to decreased promoter activity. In the present study, we utilized two vastly different injury models with known acute and chronic loss of Kir4.1 protein and mRNA to evaluate the methylation status of Kcnj10 as a candidate molecular mechanism for reduced transcription and subsequent protein loss. Examining whole hippocampal tissue and isolated astrocytes, in a lithium-pilocarpine model of epilepsy, we consistently identified hypermethylation of CpG island two, which resides in the large intronic region spanning the Kcnj10 gene. Strikingly similar results were observed using the second injury paradigm, a fifth cervical (C5) vertebral hemi-contusion model of spinal cord injury. Our previous work indicates the same gene region is significantly hypomethylated when transcription increases during astrocyte maturation. Our results suggest that DNA methylation can bidirectionally modulate Kcnj10 transcription and may represent a targetable molecular mechanism for the restoring astroglial Kir4.1 expression following CNS insult.


Assuntos
Sistema Nervoso Central/metabolismo , Metilação de DNA/fisiologia , Canais de Potássio Corretores do Fluxo de Internalização/metabolismo , Traumatismos da Medula Espinal/patologia , Animais , Astrócitos/metabolismo , Epilepsia/metabolismo , Neuroglia/metabolismo , Neurônios/citologia , Ratos Sprague-Dawley , Convulsões/metabolismo , Traumatismos da Medula Espinal/metabolismo
17.
J Cell Biol ; 218(12): 3888-3889, 2019 12 02.
Artigo em Inglês | MEDLINE | ID: mdl-31723008

RESUMO

The electrical properties of neuronal cells rely on gradients of ions across their membranes and the extracellular fluid (ECF) in which they are bathed. Little is known regarding how the ECF volume and content is maintained. In this issue, Li et al. (2019. J. Cell Biol. https://doi.org/10.1083/jcb.201907138) identify the kinase SIK3 in glia as a key signal transduction regulator in ion and volume homeostasis in Drosophila peripheral nerves.


Assuntos
Drosophila , Água , Animais , Homeostase , Íons , Neuroglia
18.
Int J Mol Sci ; 20(15)2019 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-31387202

RESUMO

Rett syndrome (RTT) is a rare, X-linked neurodevelopmental disorder typically affecting females, resulting in a range of symptoms including autistic features, intellectual impairment, motor deterioration, and autonomic abnormalities. RTT is primarily caused by the genetic mutation of the Mecp2 gene. Initially considered a neuronal disease, recent research shows that glial dysfunction contributes to the RTT disease phenotype. In the following manuscript, we review the evidence regarding glial dysfunction and its effects on disease etiology.


Assuntos
Estudos de Associação Genética , Predisposição Genética para Doença , Proteína 2 de Ligação a Metil-CpG/deficiência , Neuroglia/metabolismo , Síndrome de Rett/genética , Síndrome de Rett/metabolismo , Animais , Astrócitos/metabolismo , Metabolismo Energético , Estudos de Associação Genética/métodos , Humanos , Oligodendroglia/metabolismo , Fenótipo , Síndrome de Rett/diagnóstico
19.
Elife ; 82019 08 21.
Artigo em Inglês | MEDLINE | ID: mdl-31433295

RESUMO

Brain-derived neurotrophic factor (BDNF) is a critical growth factor involved in the maturation of the CNS, including neuronal morphology and synapse refinement. Herein, we demonstrate astrocytes express high levels of BDNF's receptor, TrkB (in the top 20 of protein-coding transcripts), with nearly exclusive expression of the truncated isoform, TrkB.T1, which peaks in expression during astrocyte morphological maturation. Using a novel culture paradigm, we show that astrocyte morphological complexity is increased in the presence of BDNF and is dependent upon BDNF/TrkB.T1 signaling. Deletion of TrkB.T1, globally and astrocyte-specifically, in mice revealed morphologically immature astrocytes with significantly reduced volume, as well as dysregulated expression of perisynaptic genes associated with mature astrocyte function. Indicating a role for functional astrocyte maturation via BDNF/TrkB.T1 signaling, TrkB.T1 KO astrocytes do not support normal excitatory synaptogenesis or function. These data suggest a significant role for BDNF/TrkB.T1 signaling in astrocyte morphological maturation, a critical process for CNS development.


Assuntos
Astrócitos/citologia , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diferenciação Celular , Glicoproteínas de Membrana/metabolismo , Morfogênese , Proteínas Tirosina Quinases/metabolismo , Transdução de Sinais , Animais , Células Cultivadas , Glicoproteínas de Membrana/deficiência , Camundongos , Camundongos Knockout , Isoformas de Proteínas/metabolismo , Proteínas Tirosina Quinases/deficiência
20.
Curr Protoc Neurosci ; 88(1): e71, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31216394

RESUMO

Interest in evaluating individual cellular populations in the central nervous system has prompted the development of several techniques enabling the enrichment of single-cell populations. Herein we detail a relatively inexpensive method to specifically isolate neurons, astrocytes, and microglia from a mixed homogenate utilizing magnetic beads conjugated to cell-type specific antibodies. We have used this technique to isolate astrocytes across development and into late adulthood. Finally, we detail the utilization of this technique in novel astrocyte and astrocyte/neuron co-culture paradigms. © 2019 by John Wiley & Sons, Inc.


Assuntos
Astrócitos , Encéfalo/citologia , Separação Celular/métodos , Fenômenos Magnéticos , Microglia , Neurônios , Animais , Astrócitos/fisiologia , Encéfalo/fisiologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/fisiologia , Neurônios/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...