Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Brain Struct Funct ; 223(2): 851-872, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28993883

RESUMO

Massive corticothalamic afferents originating from layer 6a of primary sensory cortical areas modulate sensory responsiveness of thalamocortical neurons and are pivotal for shifting neuronal firing between burst and tonic modes. The influence of the corticothalamic pathways on the firing mode and sensory gain of thalamic neurons has only been extensively examined in anesthetized animals, but has yet to be established in the awake state. We made lesions of the rat barrel cortex and on the following day recorded responses of single thalamocortical and thalamic reticular neurons to a single vibrissal deflection in the somatosensory system during wakefulness. Our results showed that the cortical lesions shifted the response of thalamic neurons towards bursting, elevated the response probability and the gain of thalamocortical neurons, predominantly of recurring responses. In addition, after the lesions, the spontaneous activities of the vibrissa-responsive thalamic neurons, but not those of vibrissa-unresponsive cells, were typified by waxing-and-waning spindle-like rhythmic spiking with frequent bursting. In awake rats with intact cortex, identified layer 6a corticothalamic neurons responded to a single vibrissal deflection with short latencies that matched those of layer 4 neurons, strongly suggesting the existence of an immediate corticothalamic feedback. The present results show the importance of corticothalamic neurons in shaping thalamic activities during wakefulness.


Assuntos
Vias Neurais/fisiologia , Neurônios/fisiologia , Córtex Somatossensorial/fisiologia , Tálamo/citologia , Vigília/fisiologia , Potenciais de Ação/fisiologia , Animais , Estimulação Elétrica , Eletrocardiografia , Compostos Heterocíclicos com 3 Anéis/farmacocinética , Estimulação Física , Ratos , Ratos Sprague-Dawley , Tempo de Reação/fisiologia , Rodaminas , Córtex Somatossensorial/citologia , Córtex Somatossensorial/lesões , Núcleos Ventrais do Tálamo/lesões , Vibrissas/inervação , Privação de Água/fisiologia
2.
Acta Neurobiol Exp (Wars) ; 77(1): 1-17, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28379212

RESUMO

A number of neurological disorders such as epidural hematoma can cause compression of cerebral cortex. We here tested the hypothesis that sustained compression of primary somatosensory cortex may affect stellate neurons and thalamocortical afferent (TCA) fibers. A rat model with barrel cortex subjected to bead epidural compression was used. Golgi-Cox staining analyses showed the shrinkage of dendritic arbors and the stripping of dendritic spines of stellate neurons for at least 3 months post-lesion. Anterograde tracing analyses exhibited a progressive decline of TCA fiber density in barrel field for 6 months post-lesion. Due to the abrupt decrease of TCA fiber density at 3 days after compression, we further used electron microscopy to investigate the ultrastructure of TCA fibers at this time. Some TCA fiber terminal profiles with dissolved or darkened mitochondria and fewer synaptic vesicles were distorted and broken. Furthermore, the disruption of mitochondria and myelin sheath was observed in some myelinated TCA fibers. In addition, expressions of oxidative markers 3-nitrotyrosine and 4-hydroxynonenal were elevated in barrel field post-lesion. Treatment of antioxidant ascorbic acid or apocynin was able to reverse the increase of oxidative stress and the decline of TCA fiber density, rather than the shrinkage of dendrites and the stripping of dendritic spines of stellate neurons post-lesion. Together, these results indicate that sustained epidural compression of primary somatosensory cortex affects the TCA fibers and the dendrites of stellate neurons for a prolonged period. In addition, oxidative stress is responsible for the reduction of TCA fiber density in barrels rather than the shrinkage of dendrites and the stripping of dendritic spines of stellate neurons.


Assuntos
Vias Aferentes/patologia , Lesões Encefálicas/patologia , Espaço Epidural , Neurônios/patologia , Córtex Somatossensorial/patologia , Tálamo/patologia , Acetofenonas/uso terapêutico , Aldeídos/metabolismo , Animais , Antioxidantes/uso terapêutico , Ácido Ascórbico/uso terapêutico , Biotina/análogos & derivados , Biotina/farmacocinética , Lesões Encefálicas/tratamento farmacológico , Dendritos/patologia , Dendritos/ultraestrutura , Dextranos/farmacocinética , Modelos Animais de Doenças , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Espaço Epidural/fisiologia , Lateralidade Funcional , Masculino , Neurônios/ultraestrutura , Estresse Oxidativo/fisiologia , Ratos , Córtex Somatossensorial/lesões , Tálamo/ultraestrutura , Fatores de Tempo , Tirosina/análogos & derivados , Tirosina/metabolismo
3.
J Neurosci Methods ; 201(2): 296-306, 2011 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-21871490

RESUMO

Traumatic brain injury (TBI) is a major cause of death and disability for which there is no cure. One of the issues inhibiting clinical trial success is the lack of targeting specific patient populations due to inconsistencies between clinical diagnostic tools and underlying pathophysiology. The development of reliable, noninvasive markers of TBI severity and injury mechanisms may better identify these populations, thereby improving clinical trial design. Magnetic resonance elastography (MRE), by assessing tissue mechanical properties, can potentially provide such marker. MRE synchronizes mechanical excitations with a phase contrast imaging pulse sequence to noninvasively register shear wave propagation, from which local values of tissue viscoelastic properties can be deduced. The working hypothesis of this study is that TBI involves a compression of brain tissue large enough to bring the material out of its elastic range, sufficiently altering mechanical properties to generate contrast on MRE measurements. To test this hypothesis, we combined microscopic MRE with brain tissue collected from adult male rats subjected to a controlled cortical impact injury. Measurements were made in different regions of interest (somatosensory cortex, hippocampus, and thalamus), and at different time points following the injury (immediate, 24 h, 7 days, 28 days). Values of stiffness in the somatosensory cortex were found to be 23-32% lower in the injured hemisphere than in the healthy one, when no significant difference was observed in the case of sham brains. A preliminary in vivo experiment is also presented, as well as alternatives to improve the faithfulness of stiffness recovery.


Assuntos
Lesões Encefálicas/fisiopatologia , Técnicas de Imagem por Elasticidade/métodos , Miniaturização/métodos , Animais , Lesões Encefálicas/patologia , Modelos Animais de Doenças , Técnicas de Imagem por Elasticidade/instrumentação , Hipocampo/lesões , Hipocampo/patologia , Hipocampo/fisiopatologia , Masculino , Camundongos , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/lesões , Córtex Somatossensorial/patologia , Córtex Somatossensorial/fisiopatologia , Tálamo/lesões , Tálamo/patologia , Tálamo/fisiopatologia
4.
J Neurotrauma ; 27(11): 1971-82, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20809786

RESUMO

Traumatic brain injury (TBI) results in enduring functional deficits. Strategies aimed at promoting plasticity within the injured brain may aid in enhancing functional outcome. We have previously shown that spontaneous pericontusional axon sprouting occurs within 7-14 days after controlled cortical impact injury in the adult rat, but ultimately fails due to an increasingly growth-inhibitory environment. We therefore sought to determine whether acute infusion of chondroitinase ABC into the site of the cortical contusion, to further reduce pericontusional growth-inhibitory chondroitin sulfate proteoglycans (CSPGs), would enhance and prolong the sprouting response. We also wanted to determine if chondroitinase-enhanced sprouting would ameliorate the behavioral deficits in forelimb function that occur in this model. Acute chondroitinase infusion decreased intact CSPGs and significantly increased pericontusional cortical grey and white matter growth-associated protein 43 (GAP43)-positive axon sprouting at 7 days post-injury. A return of intact CSPGs at later time points likely contributed to the absence of persistently increased levels of axon sprouting by 14-21 days post-injury. There was no overall benefit on forelimb function during the time of maximal sprouting or at any subsequent times in three of four behavioral outcome measures. However, there was a chondroitinase-induced improvement in recovery from unskilled limb use deficits on the staircase forelimb reaching test toward sham-injured values at 28 days, which was not achieved by the vehicle-treated rats, indicating that there is some minor functional benefit of the increased sprouting induced by chondroitinase treatment. The current results, together with data from spinal cord injury models after chondroitinase intervention, suggest that a combinatorial approach with the addition of neurotrophins and rehabilitation would result in more robust axon sprouting and consequently improve behavioral outcome.


Assuntos
Axônios/efeitos dos fármacos , Comportamento Animal/fisiologia , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/patologia , Condroitina ABC Liase/farmacologia , Animais , Atrofia , Lesões Encefálicas/psicologia , Contagem de Células , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Proteína GAP-43/metabolismo , Processamento de Imagem Assistida por Computador , Imuno-Histoquímica , Masculino , Córtex Motor/lesões , Córtex Motor/patologia , Procedimentos Neurocirúrgicos , Desempenho Psicomotor/fisiologia , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/lesões , Córtex Somatossensorial/patologia , Caminhada/fisiologia
5.
J Neurosci Res ; 87(13): 2937-50, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19437549

RESUMO

Axonal injury is a major hallmark of traumatic brain injury (TBI), and it seems likely that therapies directed toward enhancing axon repair could potentially improve functional outcomes. One potential target is chondroitin sulfate proteoglycans (CSPGs), which are major axon growth inhibitory molecules that are generally, but not always, up-regulated after central nervous system injury. The current study was designed to determine temporal changes in cerebral cortical mRNA or protein expression levels of CSPGs and to determine their regional localization and cellular association by using immunohistochemistry in a controlled cortical impact model of TBI. The results showed significant increases in versican mRNA at 4 and 14 days after TBI but no change in neurocan, aggrecan, or phosphacan. Semiquantitative Western blot (WB) analysis of cortical CSPG protein expression revealed a significant ipsilateral decrease of all CSPGs at 1 day after TBI. Lower CSPG protein levels were sustained until at least 14 days, after which the levels began to normalize. Immunohistochemistry data confirm previous reports of regional increases in CSPG proteins after CNS injury, seen primarily within the developing glial scar after TBI, but also corroborate the WB data by revealing wide areas of pericontusional tissue that are deficient in both extracellular and perineuronal net-associated CSPGs. Given the evidence that CSPGs are largely inhibitory to axonal growth, we interpret these data to indicate a potential for regional spontaneous plasticity after TBI. If this were the case, the gradual normalization of CSPG proteins over time postinjury would suggest that this may be temporally as well as regionally limited.


Assuntos
Lesões Encefálicas/metabolismo , Proteoglicanas de Sulfatos de Condroitina/biossíntese , Regulação da Expressão Gênica , Proteínas do Tecido Nervoso/biossíntese , Animais , Lesões Encefálicas/genética , Lesões Encefálicas/patologia , Proteoglicanas de Sulfatos de Condroitina/genética , Cicatriz/etiologia , Cicatriz/genética , Cicatriz/metabolismo , Cicatriz/patologia , Gliose/etiologia , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Masculino , Córtex Motor/lesões , Córtex Motor/metabolismo , Proteínas do Tecido Nervoso/genética , Plasticidade Neuronal , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/lesões , Córtex Somatossensorial/metabolismo , Fatores de Tempo , Versicanas/biossíntese , Versicanas/genética , Cicatrização
6.
Neuroscience ; 150(4): 970-83, 2007 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-18022327

RESUMO

Early postnatal freeze-lesions to the cortical plate result in malformations resembling human microgyria. Microgyria in primary somatosensory cortex (S1) of rats are associated with a reduced behavioral detection of rapid auditory transitions and the loss of large cells in the thalamic nucleus projecting to primary auditory cortex (A1). Detection of slow transitions in sound is intact in animals with S1 microgyria, suggesting dissociation between responding to slow versus rapid transitions and a possible dissociation between levels of auditory processing affected. We hypothesized that neuronal responses in primary auditory cortex (A1) would be differentially reduced for rapid sound repetitions but not for slow sound sequences in animals with S1 microgyria. We assessed layer IV cortical responses in primary auditory cortex (A1) to single pure-tones and periodic noise bursts (PNB) in rats with and without S1 microgyria. We found that responses to both types of acoustic stimuli were reduced in magnitude in animals with microgyria. Furthermore, spectral resolution was degraded in animals with microgyria. The cortical selectivity and temporal precision were then measured with conventional methods for PNB and tone-stimuli, but no significant changes were observed between microgyric and control animals. Surprisingly, the observed spike rate reduction was similar for rapid and slow temporal modulations of PNB stimuli. These results suggest that acoustic processing in A1 is indeed altered with early perturbations of neighboring cortex. However, the type of deficit does not affect the temporal dynamics of the cortical output. Instead, acoustic processing is altered via a systematic reduction in the driven spike rate output and spectral integration resolution in A1. This study suggests a novel form of plasticity, whereas early postnatal lesions of one sensory cortex can have a functional impact on processing in neighboring sensory cortex.


Assuntos
Córtex Auditivo/fisiopatologia , Percepção Auditiva/fisiologia , Lesões Encefálicas/patologia , Potenciais Evocados Auditivos/fisiologia , Córtex Somatossensorial/lesões , Estimulação Acústica/métodos , Acústica , Animais , Animais Recém-Nascidos , Mapeamento Encefálico , Período Crítico Psicológico , Relação Dose-Resposta à Radiação , Feminino , Lateralidade Funcional , Masculino , Gravidez , Ratos , Ratos Wistar , Tempo de Reação , Córtex Somatossensorial/fisiopatologia
7.
Acta Neurol Belg ; 101(4): 221-3, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11851029
8.
Acta Neuropathol ; 99(2): 147-53, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10672321

RESUMO

The temporospatial relationship between microglial and astrocytic reactions and delayed thalamic cell death was examined 1-7 days following a traumatic cold lesion of the rat sensorimotor cortex using immunocytochemistry in combination with terminal deoxynucleotidyltransferase-mediated biotinylated dUTP nick end labeling (TUNEL) of nuclear DNA fragmentation. No or only occasional TUNEL-positive cells were found in the thalamic relay nuclei up to 3 days after trauma. After 7 days, on the other hand, a considerable number of TUNEL-positive cells were seen in the ventrobasal, the ventrolateral and posterior thalamic nuclei. Already 3 days after trauma, i.e., before cell injury was detectable, many protoplasmic astrocytes, which were reactive for glial fibrillary acidic protein, and ramified microglia, which were positive for complement receptor type 3b (CR3b) but negative for major histocompatibility complex (MHC) class II antigen, were noticed in the thalamus. The number of labeled astro- and microglia further increased after 7 days, when DNA fragmentation became evident. At this time, the morphology of microglia shifted towards bushy and rod-like cells, and microglia became also reactive for MHC class II antigen. Clusters of CR3b- and MHC class II-positive microglia were found in the ventrobasal thalamus. The present findings demonstrate that trauma-induced microglial and astrocytic reactions appear in the thalamus prior the onset of cell damage.


Assuntos
Apoptose , Astrócitos/patologia , Lesões Encefálicas/patologia , Microglia/patologia , Córtex Somatossensorial/patologia , Tálamo/patologia , Animais , Proteína Glial Fibrilar Ácida/análise , Antígenos de Histocompatibilidade Classe II/análise , Marcação In Situ das Extremidades Cortadas , Complexo Principal de Histocompatibilidade , Masculino , Microglia/imunologia , Ratos , Ratos Sprague-Dawley , Receptores de Complemento 3b/análise , Córtex Somatossensorial/imunologia , Córtex Somatossensorial/lesões , Núcleos Talâmicos/imunologia , Núcleos Talâmicos/patologia , Tálamo/imunologia
9.
Cereb Cortex ; 7(5): 453-64, 1997.
Artigo em Inglês | MEDLINE | ID: mdl-9261574

RESUMO

Induction of microgyria by freezing injury to the developing somatosensory cortex of neonatal rats causes a defect in fast auditory processing in males, but not in females. It was speculated that early damage to the cortex has sexually dimorphic cascading effects on other brain regions mediating auditory processing, which can lead to the observed behavioral deficits. In the current series of experiments, bilateral microgyri were induced by placement of a freezing probe on the skulls of newborn male and female rats, and these animals were tested in adulthood for auditory temporal processing. Control animals received sham surgery. The brains from these animals were embedded in celloidin, cut in the coronal plane and the following morphometric measures assessed: microgyric volume, medial geniculate nucleus (MGN) volume, cell number, and cell size, and, as a control, dorsal lateral geniculate nucleus (dLGN) volume, cell number and cell size. There were no sex differences in the cortical pathology of lesioned animals. However, microgyric males had more small and fewer large neurons in the MGN than their sham-operated counterparts, whereas there was no difference between lesioned and sham-operated females. There was no effect on dLGN cell size distribution in either sex. Microgyric males were significantly impaired in fast auditory temporal processing when compared to control males, whereas lesioned females exhibited no behavioral deficits. These results suggest that early injury to the cerebral cortex may have different effects on specific thalamic nuclei in males and females, with corresponding differences in behavioral effects.


Assuntos
Percepção Auditiva/fisiologia , Córtex Somatossensorial/patologia , Tálamo/patologia , Animais , Comportamento Animal/fisiologia , Contagem de Células , Tamanho Celular/fisiologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Congelamento , Corpos Geniculados/fisiologia , Masculino , Degeneração Neural , Ratos , Ratos Wistar , Caracteres Sexuais , Córtex Somatossensorial/lesões , Córtex Somatossensorial/fisiologia , Tálamo/fisiologia
10.
Neuroscience ; 59(4): 975-92, 1994 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-8058130

RESUMO

Local cerebral glucose utilization was assessed during whisker stimulation by 2-deoxyglucose autoradiography. Whisker stimulation increased local cerebral glucose utilization in brainstem, thalamus and whisker sensory cortex in normal rats. Whereas whisker stimulation increased glucose metabolism in brainstem, whisker stimulation failed to increase glucose metabolism in thalamus of rats that had whisker sensory cortex ablated 5 h to five weeks previously. The failure of whisker stimulation to activate thalamus after cortical ablations was probably not due to decreased cortical input to thalamus because whisker stimulation activated thalamus after large cortical tetrodotoxin injections. Failure of whisker stimulation to activate thalamus at early times (5 h and one day) after cortical ablations was not due to thalamic neuronal death, since it takes days to weeks for axotomized thalamic neurons to die. The failure of whisker stimulation to activate thalamus at early times after cortical ablations was likely due to the failure of trigeminal brainstem neurons that project to thalamus to activate axotomized thalamic neurons. This might occur because of synaptic retraction, glial stripping or inhibition of trigeminal brainstem synapses onto thalamic neurons. The thalamic neuronal death that occurs over the days and weeks following cortical ablations was associated with thalamic hypometabolism. This is consistent with the idea that the thalamic neurons die because of the absence of a cortically derived trophic factor, since the excitotoxic thalamic cell death that occurs following cortical kainate injections is associated with thalamic hypermetabolism. The glucose metabolism of parts of the host thalamus was higher and the glucose metabolism in surrounding nuclei lower than the normal side of thalamus in rats that sat quietly and had fetal cortex transplants placed into cavities in whisker sensory cortex five to 16 weeks previously. Whisker stimulation in these subjects activated the contralateral host thalamus and fetal cortical transplants. This was accomplished using a double-label 2-deoxyglucose method to assess brain glucose metabolism in the same rat while it was resting and during whisker stimulation. The high glucose metabolism of parts of host thalamus ipsilateral to the fetal cortical transplants is consistent with prolonged survival of some axotomized thalamic neurons. The finding that whisker stimulation activates portions of host thalamus further suggests that the cortical transplants maintained survival of the host thalamic neurons and that synaptic connections between whisker brainstem and thalamic neurons were functional.


Assuntos
Tronco Encefálico/fisiologia , Transplante de Tecido Encefálico , Descorticação Cerebral , Transplante de Tecido Fetal , Córtex Somatossensorial/fisiologia , Tálamo/fisiologia , Vibrissas/fisiologia , Animais , Morte Celular , Desoxiglucose/metabolismo , Metabolismo Energético , Feminino , Degeneração Neural , Ratos , Ratos Sprague-Dawley , Córtex Somatossensorial/lesões , Córtex Somatossensorial/transplante , Estresse Mecânico , Tetrodotoxina/farmacologia , Tetrodotoxina/toxicidade , Tálamo/efeitos dos fármacos , Nervo Trigêmeo/fisiologia , Núcleos do Trigêmeo/fisiologia , Núcleos do Trigêmeo/ultraestrutura
11.
Acta Physiol Pol ; 39(5-6): 475-83, 1988.
Artigo em Inglês | MEDLINE | ID: mdl-3257060

RESUMO

Ipsi- and contralateral cortical somatosensory evoked potentials (SEP) were recorded following median nerve stimulation in 12 patients with unilateral brain lesions and in 5 healthy subjects. Computed tomographic scans of brain were performed on admission. In all patients with lesions of the claustrum there was absence of SEP contralateral to the side of the lesion and ipsilateral to the stimulated nerve. This phenomenon did not appear in our material following lesions involving other structures e.g. thalamus or somatosensory cortex. Our observations suggest that the claustrum may influence deeply the contralateral somatosensory cortex. This may be due to the fact that a large part of the claustrum is involved in transmission of the sensory information from receptors to the somatosensory cortex.


Assuntos
Lesões Encefálicas/fisiopatologia , Potenciais Somatossensoriais Evocados , Córtex Somatossensorial/lesões , Tálamo/lesões , Eletroencefalografia , Feminino , Lateralidade Funcional , Humanos , Masculino , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA