Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Exp Med ; 220(2)2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36520518

RESUMO

Microglia are critical regulators of brain development that engulf synaptic proteins during postnatal synapse remodeling. However, the mechanisms through which microglia sense the brain environment are not well defined. Here, we characterized the regulatory program downstream of interleukin-33 (IL-33), a cytokine that promotes microglial synapse remodeling. Exposing the developing brain to a supraphysiological dose of IL-33 altered the microglial enhancer landscape and increased binding of stimulus-dependent transcription factors including AP-1/FOS. This induced a gene expression program enriched for the expression of pattern recognition receptors, including the scavenger receptor MARCO. CNS-specific deletion of IL-33 led to increased excitatory/inhibitory synaptic balance, spontaneous absence-like epileptiform activity in juvenile mice, and increased seizure susceptibility in response to chemoconvulsants. We found that MARCO promoted synapse engulfment, and Marco-deficient animals had excess thalamic excitatory synapses and increased seizure susceptibility. Taken together, these data define coordinated epigenetic and functional changes in microglia and uncover pattern recognition receptors as potential regulators of postnatal synaptic refinement.


Assuntos
Interleucina-33 , Microglia , Animais , Camundongos , Microglia/metabolismo , Interleucina-33/metabolismo , Sinapses/metabolismo , Encéfalo/metabolismo , Convulsões/metabolismo , Camundongos Endogâmicos C57BL
2.
Sci Transl Med ; 14(652): eabj4310, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35857628

RESUMO

Inflammatory processes induced by brain injury are important for recovery; however, when uncontrolled, inflammation can be deleterious, likely explaining why most anti-inflammatory treatments have failed to improve neurological outcomes after brain injury in clinical trials. In the thalamus, chronic activation of glial cells, a proxy of inflammation, has been suggested as an indicator of increased seizure risk and cognitive deficits that develop after cortical injury. Furthermore, lesions in the thalamus, more than other brain regions, have been reported in patients with viral infections associated with neurological deficits, such as SARS-CoV-2. However, the extent to which thalamic inflammation is a driver or by-product of neurological deficits remains unknown. Here, we found that thalamic inflammation in mice was sufficient to phenocopy the cellular and circuit hyperexcitability, enhanced seizure risk, and disruptions in cortical rhythms that develop after cortical injury. In our model, down-regulation of the GABA transporter GAT-3 in thalamic astrocytes mediated this neurological dysfunction. In addition, GAT-3 was decreased in regions of thalamic reactive astrocytes in mouse models of cortical injury. Enhancing GAT-3 in thalamic astrocytes prevented seizure risk, restored cortical states, and was protective against severe chemoconvulsant-induced seizures and mortality in a mouse model of traumatic brain injury, emphasizing the potential of therapeutically targeting this pathway. Together, our results identified a potential therapeutic target for reducing negative outcomes after brain injury.


Assuntos
Lesões Encefálicas , COVID-19 , Animais , Astrócitos/metabolismo , Modelos Animais de Doenças , Proteínas da Membrana Plasmática de Transporte de GABA/metabolismo , Inflamação/patologia , Camundongos , Polímeros , Roedores/metabolismo , SARS-CoV-2 , Convulsões , Tálamo/metabolismo , Tálamo/patologia
3.
J Comp Neurol ; 530(7): 998-1019, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-34633669

RESUMO

While cortical injuries, such as traumatic brain injury (TBI) and neocortical stroke, acutely disrupt the neocortex, most of their consequent disabilities reflect secondary injuries that develop over time. Thalamic neuroinflammation has been proposed to be a biomarker of cortical injury and of the long-term cognitive and neurological deficits that follow. However, the extent to which thalamic neuroinflammation depends on the type of cortical injury or its location remains unknown. Using two mouse models of focal neocortical injury that do not directly damage subcortical structures-controlled cortical impact and photothrombotic ischemic stroke-we found that chronic neuroinflammation in the thalamic region mirrors the functional connections with the injured cortex, and that sensory corticothalamic regions may be more likely to sustain long-term damage than nonsensory circuits. Currently, heterogeneous clinical outcomes complicate treatment. Understanding how thalamic inflammation depends on the injury site can aid in predicting features of subsequent deficits and lead to more effective, customized therapies.


Assuntos
Lesões Encefálicas Traumáticas , Acidente Vascular Cerebral , Animais , Lesões Encefálicas Traumáticas/complicações , Modelos Animais de Doenças , Camundongos , Microglia , Doenças Neuroinflamatórias , Acidente Vascular Cerebral/complicações , Tálamo
4.
Science ; 373(6560): eabj2685, 2021 Sep 10.
Artigo em Inglês | MEDLINE | ID: mdl-34516796

RESUMO

Although traumatic brain injury (TBI) acutely disrupts the cortex, most TBI-related disabilities reflect secondary injuries that accrue over time. The thalamus is a likely site of secondary damage because of its reciprocal connections with the cortex. Using a mouse model of mild TBI (mTBI), we found a chronic increase in C1q expression specifically in the corticothalamic system. Increased C1q expression colocalized with neuron loss and chronic inflammation and correlated with disruption in sleep spindles and emergence of epileptic activities. Blocking C1q counteracted these outcomes, suggesting that C1q is a disease modifier in mTBI. Single-nucleus RNA sequencing demonstrated that microglia are a source of thalamic C1q. The corticothalamic circuit could thus be a new target for treating TBI-related disabilities.


Assuntos
Lesões Encefálicas/complicações , Complemento C1q/fisiologia , Fases do Sono , Transtornos do Sono-Vigília/etiologia , Transtornos do Sono-Vigília/fisiopatologia , Tálamo/fisiopatologia , Animais , Lesões Encefálicas/fisiopatologia , Complemento C1q/genética , Modelos Animais de Doenças , Epilepsia/fisiopatologia , Camundongos , Microglia/metabolismo , Tálamo/metabolismo
5.
Elife ; 102021 04 12.
Artigo em Inglês | MEDLINE | ID: mdl-33843585

RESUMO

Visual perception in natural environments depends on the ability to focus on salient stimuli while ignoring distractions. This kind of selective visual attention is associated with gamma activity in the visual cortex. While the nucleus reticularis thalami (nRT) has been implicated in selective attention, its role in modulating gamma activity in the visual cortex remains unknown. Here, we show that somatostatin- (SST) but not parvalbumin-expressing (PV) neurons in the visual sector of the nRT preferentially project to the dorsal lateral geniculate nucleus (dLGN), and modulate visual information transmission and gamma activity in primary visual cortex (V1). These findings pinpoint the SST neurons in nRT as powerful modulators of the visual information encoding accuracy in V1 and represent a novel circuit through which the nRT can influence representation of visual information.


Assuntos
Ritmo Gama/fisiologia , Neurônios/fisiologia , Núcleos Talâmicos/fisiologia , Córtex Visual/fisiologia , Percepção Visual/fisiologia , Animais , Feminino , Masculino , Camundongos , Somatostatina/metabolismo
6.
Elife ; 92020 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-32452758

RESUMO

​Maf (c-Maf) and Mafb transcription factors (TFs) have compensatory roles in repressing somatostatin (SST+) interneuron (IN) production in medial ganglionic eminence (MGE) secondary progenitors in mice. Maf and Mafb conditional deletion (cDKO) decreases the survival of MGE-derived cortical interneurons (CINs) and changes their physiological properties. Herein, we show that (1) Mef2c and Snap25 are positively regulated by Maf and Mafb to drive IN morphological maturation; (2) Maf and Mafb promote Mef2c expression which specifies parvalbumin (PV+) INs; (3) Elmo1, Igfbp4 and Mef2c are candidate markers of immature PV+ hippocampal INs (HIN). Furthermore, Maf/Mafb neonatal cDKOs have decreased CINs and increased HINs, that express Pnoc, an HIN specific marker. Our findings not only elucidate key gene targets of Maf and Mafb that control IN development, but also identify for the first time TFs that differentially regulate CIN vs. HIN production.


Assuntos
Regulação da Expressão Gênica , Interneurônios/metabolismo , Fator de Transcrição MafB/fisiologia , Proteínas Proto-Oncogênicas c-maf/fisiologia , Animais , Feminino , Fatores de Transcrição MEF2/metabolismo , Camundongos , Doenças do Sistema Nervoso/etiologia , Gravidez , Precursores de Proteínas/genética , Receptores CXCR4/metabolismo , Receptores Opioides/genética , Análise de Célula Única , Proteína 25 Associada a Sinaptossoma/metabolismo , Transcriptoma
7.
Cell Rep ; 26(5): 1157-1173.e5, 2019 01 29.
Artigo em Inglês | MEDLINE | ID: mdl-30699346

RESUMO

Mafb and c-Maf transcription factor (TF) expression is enriched in medial ganglionic eminence (MGE) lineages, beginning in late-secondary progenitors and continuing into mature parvalbumin (PV+) and somatostatin (SST+) interneurons. However, the functions of Maf TFs in MGE development remain to be elucidated. Herein, Mafb and c-Maf were conditionally deleted, alone and together, in the MGE and its lineages. Analyses of Maf mutant mice revealed redundant functions of Mafb and c-Maf in secondary MGE progenitors, where they repress the generation of SST+ cortical and hippocampal interneurons. By contrast, Mafb and c-Maf have distinct roles in postnatal cortical interneuron (CIN) morphological maturation, synaptogenesis, and cortical circuit integration. Thus, Mafb and c-Maf have redundant and opposing functions at different steps in CIN development.


Assuntos
Linhagem da Célula , Córtex Cerebral/metabolismo , Interneurônios/metabolismo , Fator de Transcrição MafB/metabolismo , Proteínas Proto-Oncogênicas c-maf/metabolismo , Potenciais de Ação , Animais , Animais Recém-Nascidos , Apoptose , Membrana Celular/metabolismo , Movimento Celular , Proliferação de Células , Hipocampo/metabolismo , Eminência Mediana/metabolismo , Camundongos Knockout , Neuritos/metabolismo , Neurogênese , Parvalbuminas/metabolismo , Somatostatina/metabolismo , Sinapses/metabolismo
9.
Cell Rep ; 26(1): 54-64.e6, 2019 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-30605686

RESUMO

Loss of function in the Scn1a gene leads to a severe epileptic encephalopathy called Dravet syndrome (DS). Reduced excitability in cortical inhibitory neurons is thought to be the major cause of DS seizures. Here, in contrast, we show enhanced excitability in thalamic inhibitory neurons that promotes the non-convulsive seizures that are a prominent yet poorly understood feature of DS. In a mouse model of DS with a loss of function in Scn1a, reticular thalamic cells exhibited abnormally long bursts of firing caused by the downregulation of calcium-activated potassium SK channels. Our study supports a mechanism in which loss of SK activity causes the reticular thalamic neurons to become hyperexcitable and promote non-convulsive seizures in DS. We propose that reduced excitability of inhibitory neurons is not global in DS and that non-GABAergic mechanisms such as SK channels may be important targets for treatment.


Assuntos
Epilepsias Mioclônicas/fisiopatologia , Convulsões/fisiopatologia , Tálamo/fisiopatologia , Animais , Modelos Animais de Doenças , Humanos , Camundongos
10.
Science ; 359(6381): 1269-1273, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29420261

RESUMO

Neuronal synapse formation and remodeling are essential to central nervous system (CNS) development and are dysfunctional in neurodevelopmental diseases. Innate immune signals regulate tissue remodeling in the periphery, but how this affects CNS synapses is largely unknown. Here, we show that the interleukin-1 family cytokine interleukin-33 (IL-33) is produced by developing astrocytes and is developmentally required for normal synapse numbers and neural circuit function in the spinal cord and thalamus. We find that IL-33 signals primarily to microglia under physiologic conditions, that it promotes microglial synapse engulfment, and that it can drive microglial-dependent synapse depletion in vivo. These data reveal a cytokine-mediated mechanism required to maintain synapse homeostasis during CNS development.


Assuntos
Astrócitos/metabolismo , Sistema Nervoso Central/crescimento & desenvolvimento , Interleucina-33/metabolismo , Microglia/fisiologia , Rede Nervosa/crescimento & desenvolvimento , Neurogênese , Sinapses/fisiologia , Animais , Sistema Nervoso Central/metabolismo , Homeostase , Interleucina-33/genética , Camundongos , Camundongos Knockout , Córtex Sensório-Motor/crescimento & desenvolvimento , Córtex Sensório-Motor/fisiologia , Tálamo/anormalidades
11.
Cell Rep ; 19(10): 2130-2142, 2017 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-28591583

RESUMO

Integrative brain functions depend on widely distributed, rhythmically coordinated computations. Through its long-ranging connections with cortex and most senses, the thalamus orchestrates the flow of cognitive and sensory information. Essential in this process, the nucleus reticularis thalami (nRT) gates different information streams through its extensive inhibition onto other thalamic nuclei, however, we lack an understanding of how different inhibitory neuron subpopulations in nRT function as gatekeepers. We dissociated the connectivity, physiology, and circuit functions of neurons within rodent nRT, based on parvalbumin (PV) and somatostatin (SOM) expression, and validated the existence of such populations in human nRT. We found that PV, but not SOM, cells are rhythmogenic, and that PV and SOM neurons are connected to and modulate distinct thalamocortical circuits. Notably, PV, but not SOM, neurons modulate somatosensory behavior and disrupt seizures. These results provide a conceptual framework for how nRT may gate incoming information to modulate brain-wide rhythms.


Assuntos
Ondas Encefálicas , Córtex Cerebral/metabolismo , Neurônios/metabolismo , Núcleos Talâmicos/metabolismo , Animais , Córtex Cerebral/citologia , Feminino , Humanos , Masculino , Camundongos , Neurônios/citologia , Parvalbuminas/biossíntese , Somatostatina/biossíntese , Núcleos Talâmicos/citologia
12.
Sci Rep ; 6: 34097, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27671211

RESUMO

Traumatic brain injury (TBI) is a major cause of mortality and morbidity with limited therapeutic options. Traumatic axonal injury (TAI) is an important component of TBI pathology. It is difficult to reproduce TAI in animal models of closed head injury, but in vitro stretch injury models reproduce clinical TAI pathology. Existing in vitro models employ primary rodent neurons or human cancer cell line cells in low throughput formats. This in vitro neuronal stretch injury model employs human induced pluripotent stem cell-derived neurons (hiPSCNs) in a 96 well format. Silicone membranes were attached to 96 well plate tops to create stretchable, culture substrates. A custom-built device was designed and validated to apply repeatable, biofidelic strains and strain rates to these plates. A high content approach was used to measure injury in a hypothesis-free manner. These measurements are shown to provide a sensitive, dose-dependent, multi-modal description of the response to mechanical insult. hiPSCNs transition from healthy to injured phenotype at approximately 35% Lagrangian strain. Continued development of this model may create novel opportunities for drug discovery and exploration of the role of human genotype in TAI pathology.

13.
BMC Res Notes ; 9: 160, 2016 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-26969621

RESUMO

BACKGROUND: Brain edema is a significant challenge facing clinicians managing severe traumatic brain injury (TBI) in the acute period. If edema reaches a critical point, it leads to runaway intracranial hypertension that, in turn, leads to severe morbidity or death if left untreated. Clinical data on the efficacy of standard interventions is mixed. The goal of this study was to validate a novel therapeutic strategy for reducing post-traumatic brain edema in a mouse model. Prior in vitro work reported that the brain swells due to coupled electrostatic and osmotic forces generated by large, negatively charged, immobile molecules in the matrix that comprises brain tissue. Chondroitinase ABC (ChABC) digests chondroitin sulfate proteoglycan, a molecule that contributes to this negative charge. Therefore, we administered ChABC by intracerebroventricular (ICV) injection after controlled cortical impact TBI in the mouse and measured associated changes in edema. RESULTS: Almost half of the edema induced by injury was eliminated by ChABC treatment. CONCLUSIONS: ICV administration of ChABC may be a novel and effective method of treating post-traumatic brain edema in the acute period.


Assuntos
Edema Encefálico/complicações , Edema Encefálico/tratamento farmacológico , Lesões Encefálicas Traumáticas/complicações , Lesões Encefálicas Traumáticas/tratamento farmacológico , Condroitina ABC Liase/administração & dosagem , Condroitina ABC Liase/uso terapêutico , Doença Aguda , Animais , Injeções Intraventriculares , Camundongos Endogâmicos C57BL , Água/metabolismo
14.
J Neurotrauma ; 33(13): 1202-11, 2016 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-26414212

RESUMO

An increasing number of studies have reported blood-brain barrier (BBB) dysfunction after blast-induced traumatic brain injury (bTBI). Despite this evidence, there is limited quantitative understanding of the extent of BBB opening and the time course of damage after blast injury. In addition, many studies do not report kinematic parameters of head motion, making it difficult to separate contributions of primary and tertiary blast-loading. Detailed characterization of blast-induced BBB damage may hold important implications for serum constituents that may potentially cross the compromised barrier and contribute to neurotoxicity, neuroinflammation, and persistent neurologic deficits. Using an in vivo bTBI model, systemic administration of sodium fluorescein (NaFl; 376 Da), Evans blue (EB; 69 kDa when bound to serum albumin), and dextrans (3-500 kDa) was used to estimate the pore size of BBB opening and the time required for recovery. Exposure to blast with 272 ± 6 kPa peak overpressure, 0.69 ± 0.01 ms duration, and 65 ± 1 kPa*ms impulse resulted in significant acute extravasation of NaFl, 3 kDa dextran, and EB. However, there was no significant acute extravasation of 70 kDa or 500 kDa dextrans, and minimal to no extravasation of NaFl, dextrans, or EB 1 day after exposure. This study presents a detailed analysis of the time course and pore size of BBB opening after bTBI, supported by a characterization of kinematic parameters associated with blast-induced head motion.


Assuntos
Traumatismos por Explosões/complicações , Barreira Hematoencefálica/lesões , Barreira Hematoencefálica/fisiopatologia , Lesões Encefálicas Traumáticas/complicações , Modelos Animais de Doenças , Animais , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL
15.
J Cereb Blood Flow Metab ; 35(7): 1191-8, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25757751

RESUMO

Owing to the frequent incidence of blast-induced traumatic brain injury (bTBI) in recent military conflicts, there is an urgent need to develop effective therapies for bTBI-related pathologies. Blood-brain barrier (BBB) breakdown has been reported to occur after primary blast exposure, making restoration of BBB function and integrity a promising therapeutic target. We tested the hypothesis that treatment with dexamethasone (DEX) after primary blast injury potentiates recovery of an in vitro BBB model consisting of mouse brain endothelial cells (bEnd.3). DEX treatment resulted in complete recovery of transendothelial electrical resistance and hydraulic conductivity 1 day after injury, compared with 3 days for vehicle-treated injured cultures. Administration of RU486 (mifepristone) inhibited effects of DEX, confirming that barrier restoration was mediated by glucocorticoid receptor signaling. Potentiated recovery with DEX treatment was accompanied by stronger zonula occludens (ZO)-1 tight junction immunostaining and expression, suggesting that increased ZO-1 expression was a structural correlate to BBB recovery after blast. Interestingly, augmented ZO-1 protein expression was associated with specific upregulation of the α(+) isoform but not the α(-) isoform. This is the first study to provide a mechanistic basis for potentiated functional recovery of an in vitro BBB model because of glucocorticoid treatment after primary blast injury.


Assuntos
Traumatismos por Explosões/tratamento farmacológico , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/fisiopatologia , Dexametasona/uso terapêutico , Glucocorticoides/uso terapêutico , Proteína da Zônula de Oclusão-1/metabolismo , Animais , Traumatismos por Explosões/metabolismo , Traumatismos por Explosões/fisiopatologia , Barreira Hematoencefálica/metabolismo , Linhagem Celular , Camundongos , Proteína da Zônula de Oclusão-1/análise
16.
J Neural Eng ; 10(3): 036001, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23548462

RESUMO

OBJECTIVE: Intraspinal microstimulation (ISMS) is a promising method for reanimating paralyzed limbs following neurological injury. ISMS within the cervical and lumbar spinal cord is capable of evoking a variety of highly-functional movements prior to injury, but the ability of ISMS to evoke forelimb movements after cervical spinal cord injury is unknown. Here we examine the forelimb movements and muscles activated by cervical ISMS both before and after contusion injury. APPROACH: We documented the forelimb muscles activated and movements evoked via systematic stimulation of the rodent cervical spinal cord both before injury and three, six and nine weeks following a moderate C4/C5 lateralized contusion injury. Animals were anesthetized with isoflurane to permit construction of somatotopic maps of evoked movements and quantify evoked muscle synergies between cervical segments C3 and T1. MAIN RESULTS: When ISMS was delivered to the cervical spinal cord, a variety of responses were observed at 68% of locations tested, with a spatial distribution that generally corresponded to the location of motor neuron pools. Stimulus currents required to achieve movement and the number of sites where movements could be evoked were unchanged by spinal cord injury. A transient shift toward extension-dominated movements and restricted muscle synergies were observed at three and six weeks following injury, respectively. By nine weeks after injury, however, ISMS-evoked patterns were similar to spinally-intact animals. SIGNIFICANCE: The results demonstrate the potential for cervical ISMS to reanimate hand and arm function following spinal cord injury. Robust forelimb movements can be evoked both before and during the chronic stages of recovery from a clinically relevant and sustained cervical contusion injury.


Assuntos
Membro Anterior/inervação , Membro Anterior/fisiopatologia , Músculo Esquelético/fisiopatologia , Paralisia/fisiopatologia , Paralisia/reabilitação , Traumatismos da Medula Espinal/fisiopatologia , Traumatismos da Medula Espinal/reabilitação , Animais , Vértebras Cervicais/fisiopatologia , Feminino , Contração Muscular , Ratos , Ratos Long-Evans , Traumatismos da Medula Espinal/complicações , Estimulação da Medula Espinal , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...