Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 13(1): 282, 2023 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-36609414

RESUMO

Neuroinflammation is a hallmark of hypoxic-ischemic injury and can be characterized by the activation of glial cells and the expression of inflammatory cytokines and chemokines. Interleukin (IL)-1ß and tumor necrosis factor (TNF)α are among the best-characterized early response cytokines and are often expressed concurrently. Several types of central nervous system cells secrete IL-1ß and TNFα, including microglia, astrocytes, and neurons, and these cytokines convey potent pro-inflammatory actions. Chemokines also play a central role in neuroinflammation by controlling inflammatory cell trafficking. Our aim was to characterise the evolution of early neuroinflammation in the neonatal piglet model of hypoxic-ischemic encephalopathy (HIE). Piglets (< 24 h old) were exposed to HI insult, and recovered to 2, 4, 8, 12 or 24H post-insult. Brain tissue from the frontal cortex and basal ganglia was harvested for assessment of glial cell activation profiles and transcription levels of inflammatory markers in HI piglets with comparison to a control group of newborn piglets. Fluorescence microscopy was used to observe microglia, astrocytes, neurons, degenerating neurons and possibly apoptotic cells, and quantitative polymerase chain reaction was used to measure gene expression of several cytokines and chemokines. HI injury was associated with microglial activation and morphological changes to astrocytes at all time points examined. Gene expression analyses of inflammation-related markers revealed significantly higher expression of pro-inflammatory cytokines tumor necrosis factor-α (TNFα) and interleukin 1 beta (IL-1ß), chemokines cxc-chemokine motif ligand (CXCL)8 and CXCL10, and anti-inflammatory cytokine transforming growth factor (TGF)ß in every HI group, with some region-specific differences noted. No significant difference was observed in the level of C-X-C chemokine receptor (CCR)5 over time. This high degree of neuroinflammation was associated with a reduction in the number of neurons in piglets at 12H and 24H in the frontal cortex, and the putamen at 12H. This reduction of neurons was not associated with increased numbers of degenerating neurons or potentially apoptotic cells. HI injury triggered a robust early neuroinflammatory response associated with a reduction in neurons in cortical and subcortical regions in our piglet model of HIE. This neuroinflammatory response may be targeted using novel therapeutics to reduce neuropathology in our piglet model of neonatal HIE.


Assuntos
Citocinas , Hipóxia-Isquemia Encefálica , Animais , Suínos , Citocinas/metabolismo , Animais Recém-Nascidos , Fator de Necrose Tumoral alfa/metabolismo , Doenças Neuroinflamatórias , Neuroglia/metabolismo , Encéfalo/metabolismo , Hipóxia/metabolismo , Microglia/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Fator de Crescimento Transformador beta/metabolismo , Inflamação/patologia
2.
Pediatr Res ; 92(1): 25-31, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34482377

RESUMO

BACKGROUND: The objective of this study was to systematically review the literature to determine the effect of combined hypothermia (HTH) and mesenchymal stem cell (MSC) therapy (administered during or immediately before or after HTH) compared with HTH alone on brain injury and neurobehavioural outcomes in animal models of neonatal hypoxic-ischaemic encephalopathy. METHODS: Primary outcomes assessed were neuropathological measures and neurobehavioural measures of brain outcome. Secondary outcomes were brain protein proinflammatory cytokine status. Risk of bias (ROB) was assessed with the Systematic Review Centre for Laboratory Animal Experimentation (SYRCLE) ROB assessment tool. RESULTS: Of 393 studies identified, 3 studies in postnatal day 7 (P7) male Sprague-Dawley rats met the inclusion criteria. Meta-analyses were undertaken for neuropathological measures (apoptotic cells, astrocytes, microglia), neurobehavioral measures (rotarod test and negative geotaxis), and proinflammatory cytokine levels. Two of the three studies scored low or unclear ROB across all measures. Treatment with HTH-MSCs together significantly improved astrocyte optical density by standardised mean difference (SMD) of 0.71 [95% confidence interval (CI) -1.14, -0.28]. No other measures showed significant differences. CONCLUSIONS: There is insufficient preclinical data to confirm the efficacy of combined HTH-MSC therapy over HTH alone. Future studies should utilise a reporting checklist such as in SYRCLE or Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines to improve reporting standards. IMPACT: Very few articles investigating the use of MSCs for the treatment of hypoxic-ischaemic encephalopathy are clinically relevant. Continuing to publish studies in models of hypoxic-ischaemic encephalopathy without the inclusion of HTH therapy does not progress the field towards improved clinical outcomes. This study shows that HTH and MSC therapy improves measures of astrogliosis. More studies are required to establish the efficacy of HTH and MSCs on measures of neuropathology and neurobehavior. The reporting of preclinical data in this space could be improved by using reporting checklists such as the SYRCLE or ARRIVE tools.


Assuntos
Hipotermia , Hipóxia-Isquemia Encefálica , Células-Tronco Mesenquimais , Animais , Citocinas , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/terapia , Masculino , Modelos Animais , Ratos , Ratos Sprague-Dawley
4.
Front Physiol ; 10: 541, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31133875

RESUMO

Intrauterine growth restriction (IUGR) is a condition where the fetus does not achieve optimal growth, commonly caused by placental insufficiency. The chronic decrease in blood flow restricts oxygen and nutrient supply to the fetus, which can damage numerous organ systems, with the fetal brain being particularly vulnerable. Although white matter and neuronal injury are evident in IUGR infants, the specific mechanisms underlying these changes are poorly understood. Inflammation is considered to be a main driver in exacerbating brain injury. Using a spontaneous piglet model of IUGR, we aim to determine whether administration of the anti-inflammatory drug ibuprofen will decrease inflammation at postnatal day 4 (P4). The treatment group received ibuprofen (20 mg/kg/day on day 1 and 10 mg/kg/day on days 2 and 3) in piglet formula during the morning feed each day and brains examined on P4. Markers of inflammation, apoptosis, cell proliferation, neuronal injury, and white matter injury were examined. Ibuprofen treatment ameliorated the increase in numbers of microglia and astrocytes in the parietal cortex and white matter tracts of the IUGR piglet brain on P4 as well as decreasing proinflammatory cytokines. Ibuprofen treatment prevented the reduction in apoptosis, neuronal cell counts, and myelin index in the IUGR piglets. Our findings demonstrate ibuprofen reduces the inflammatory response in the IUGR neonatal brain and concurrently reduces neuronal and white matter impairment.

5.
J Physiol ; 596(23): 5675-5686, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29700828

RESUMO

Brain injury in intrauterine growth restricted (IUGR) infants is a major contributing factor to morbidity and mortality worldwide. Adverse outcomes range from mild learning difficulties, to attention difficulties, neurobehavioral issues, cerebral palsy, epilepsy, and other cognitive and psychiatric disorders. While the use of medication to ameliorate neurological deficits in IUGR neonates has been identified as warranting urgent research for several years, few trials have been reported. This review summarises clinical trials focusing on brain protection in the IUGR newborn as well as therapeutic interventions trialled in animal models of IUGR. Therapeutically targeting mechanisms of brain injury in the IUGR neonate is fundamental to improving long-term neurodevelopmental outcomes. Inflammation is a key mechanism in neonatal brain injury; and therefore an appealing target. Ibuprofen, an anti-inflammatory drug currently used in the preterm neonate, may be a potential therapeutic candidate to treat brain injury in the IUGR neonate. To better understand the potential of ibuprofen and other therapeutic agents to be neuroprotective in the IUGR neonate, long-term follow-up information of neurodevelopmental outcomes must be studied. Where agents such as ibuprofen are shown to be effective, have a good safety profile and are relatively inexpensive, they can be widely adopted and lead to improved outcomes.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Retardo do Crescimento Fetal/tratamento farmacológico , Animais , Humanos , Recém-Nascido
6.
Brain Struct Funct ; 223(2): 1025-1033, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29282556

RESUMO

The GABAA receptor provides the majority of inhibitory neurotransmission in the adult central nervous system but in immature brain is responsible for much of the excitatory drive, a requirement for normal brain development. It is well established that GABAA receptor subunit expression changes across the course of brain development. In the present study, we have identified a splice variant of the GABAA receptor α3 subunit which appears unique to the developing brain, referred to here as the GABAA receptor α3 subunit neonatal variant (GABAA receptor α3N). RT-PCR and sequence analysis revealed splicing of exon 8 of the α3 subunit. Western blot analysis showed expression of GABAA receptor α3N in the cortex of several neonatal species and significantly reduced expression of this splice variant in the corresponding adult brains. Expression was evident in multiple brain regions and decreased across development in the pig. Fractionation revealed differential cellular localisation in the parietal cortex, hippocampus and thalamus of the full-length GABAA receptor α3 and GABAA receptor α3N. Immunoprecipitation showed direct interaction with the GABAA receptor subunits α1 and γ2 but not with gephyrin.


Assuntos
Encéfalo/citologia , Neurônios/metabolismo , Receptores de GABA-A/metabolismo , Fatores Etários , Animais , Encéfalo/anatomia & histologia , Fracionamento Celular , Humanos , Imunoprecipitação , Modelos Moleculares , Suínos
8.
Placenta ; 54: 111-116, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27939102

RESUMO

While placental function is fundamental to normal fetal development, the blood-brain barrier provides a second checkpoint critical to protecting the fetal brain and ensuring healthy brain development. The placenta is considered the key barrier between the mother and fetus, regulating delivery of essential nutrients, removing waste as well as protecting the fetus from potentially noxious substances. However, disturbances to the maternal environment and subsequent adaptations to placental function may render the placenta ineffective for providing a suitable environment for the developing fetus and to providing sufficient protection from harmful substances. The developing brain is particularly vulnerable to changes in the maternal/fetal environment. Development of the blood-brain barrier and maturation of barrier transporter systems work to protect the fetal brain from exposure to drugs, excluding them from the fetal CNS. This review will focus on the role of the 'other' key barrier during gestation - the blood-brain barrier - which has been shown to be functional as early as 8 weeks' gestation.


Assuntos
Barreira Hematoencefálica/crescimento & desenvolvimento , Desenvolvimento Fetal , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Feminino , Humanos , Placenta/metabolismo , Gravidez , Proteínas Carreadoras de Solutos/metabolismo
9.
Placenta ; 54: 117-124, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-27916232

RESUMO

Disruption to the maternal environment during pregnancy from events such as hypoxia, stress, toxins, inflammation, and reduced placental blood flow can affect fetal development. Intrauterine growth restriction (IUGR) is commonly caused by chronic placental insufficiency, interrupting supply of oxygen and nutrients to the fetus resulting in abnormal fetal growth. IUGR is a major cause of perinatal morbidity and mortality, occurring in approximately 5-10% of pregnancies. The fetal brain is particularly vulnerable in IUGR and there is an increased risk of long-term neurological disorders including cerebral palsy, epilepsy, learning difficulties, behavioural difficulties and psychiatric diagnoses. Few studies have focused on how growth restriction interferes with normal brain development in the IUGR neonate but recent studies in growth restricted animal models demonstrate increased neuroinflammation. This review describes the role of neuroinflammation in the progression of brain injury in growth restricted neonates. Identifying the mediators responsible for alterations in brain development in the IUGR infant is key to prevention and treatment of brain injury in these infants.


Assuntos
Lesões Encefálicas/etiologia , Retardo do Crescimento Fetal/imunologia , Animais , Astrócitos/fisiologia , Lesões Encefálicas/metabolismo , Citocinas/metabolismo , Feminino , Retardo do Crescimento Fetal/metabolismo , Humanos , Inflamação , Microglia/fisiologia , Gravidez
10.
J Neurochem ; 139(3): 471-484, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27456541

RESUMO

Seizures are a common manifestation of hypoxic-ischaemic brain injury in the neonate. In status epilepticus models alterations to GABAA R subunit expression have been suggested to contribute to (i) abnormal development of the GABAergic system, (ii) why seizures become self-sustaining and (iii) the development of pharmacoresistance. Detailed investigation of GABAA R subunit protein expression after neonatal hypoxia-ischaemia (HI) is currently insufficient. Using our pig model of HI and subsequent spontaneous neonatal seizures, we investigated changes in protein expression of the three predominant α-subunits of the GABAA R; α1 , α2 and α3 . Anaesthetized, ventilated newborn pigs (< 24 h old) were subjected to 30 min HI and subsequently recovered to 24 or 72 h. Amplitude-integrated electroencephalography was used to monitor brain activity and identify seizure activity. Brain tissue was collected post-mortem and GABAA R α-subunit protein expression was analysed using western blot and immunohistochemistry. GABAA R α1 and α3 protein expression was significantly reduced in animals that developed seizures after HI; HI animals that did not develop seizures did not exhibit the same reductions. Immunohistochemistry revealed decreased α1 and α3 expression, and α1 redistribution from the cell membrane to the cytosol, in the hippocampus of seizure animals. Multivariate analyses, controlling for HI severity and neuronal injury, revealed that seizures were independently associated with significant GABAA R α3 reduction. This is the first study to show loss and redistribution of GABAA R α-subunits in a neonatal brain experiencing seizures. Our findings are similar to those reported in models of SE and in chronic epilepsy.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Receptores de GABA-A/metabolismo , Convulsões/metabolismo , Animais , Animais Recém-Nascidos , Comportamento Animal , Química Encefálica , Eletroencefalografia/efeitos dos fármacos , Feminino , Hipocampo/metabolismo , Imuno-Histoquímica , Masculino , Respiração Artificial , Convulsões/psicologia , Suínos
11.
Stroke ; 44(3): 809-11, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23362078

RESUMO

BACKGROUND AND PURPOSE: To determine the optimal dose of 2-iminobiotin (2-IB) for the treatment of moderate to severe asphyxia in a neonatal piglet model of hypoxia-ischemia. METHODS: Newborn piglets were subjected to a 30-minute hypoxia-ischemia insult and randomly treated with vehicle or 2-IB (0.1 mg/kg, 0.2 mg/kg, or 1.0 mg/kg). aEEG background and seizure activity were scored after hypoxia-ischemia every 4 h until 24 h and at 48 h and neurobehavioral scores were obtained. Brain tissue was collected and processed for analysis of caspase-3 activity, histology, and tyrosine nitration. RESULTS: A dose range of 0.1 to 1.0 mg/kg/dose of 2-IB improved short-term outcome as demonstrated by an increased survival with a normal aEEG and decreased nitrotyrosine staining in the 2-IB-treated animals, indicating decreased cellular damage. Neurobehavior, caspase-3 activity in thalamus, and histology scores were not significantly different. CONCLUSIONS: Based on survival with a normal aEEG, 0.2 mg/kg 2-IB is likely to be the most appropriate dose for use in future clinical trials in neonates with perinatal hypoxia-ischemia.


Assuntos
Asfixia/tratamento farmacológico , Asfixia/etiologia , Biotina/análogos & derivados , Hipóxia-Isquemia Encefálica/complicações , Óxido Nítrico Sintase/antagonistas & inibidores , Animais , Biotina/uso terapêutico , Encéfalo/metabolismo , Encéfalo/patologia , Caspase 3/metabolismo , Relação Dose-Resposta a Droga , Eletroencefalografia , Modelos Animais , Suínos , Fatores de Tempo , Resultado do Tratamento
12.
Neurochem Res ; 37(11): 2364-78, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22528834

RESUMO

Glial fibrillary acidic protein (GFAP) is an intermediate filament protein expressed in the astrocyte cytoskeleton that plays an important role in the structure and function of the cell. GFAP can be phosphorylated at six serine (Ser) or threonine (Thr) residues but little is known about the role of GFAP phosphorylation in physiological and pathophysiological states. We have generated antibodies against two phosphorylated GFAP (pGFAP) proteins: p8GFAP, where GFAP is phosphorylated at Ser-8 and p13GFAP, where GFAP is phosphorylated at Ser-13. We examined p8GFAP and p13GFAP expression in the control neonatal pig brain and at 24 and 72 h after an hypoxic-ischemic (HI) insult. Immunohistochemistry demonstrated pGFAP expression in astrocytes with an atypical cytoskeletal morphology, even in control brains. Semi-quantitative western blotting revealed that p8GFAP expression was significantly increased at 24 h post-insult in HI animals with seizures in frontal, parietal, temporal and occipital cortices. At 72 h post-insult, p8GFAP and p13GFAP expression were significantly increased in HI animals with seizures in brain regions that are vulnerable to cellular damage (cortex and basal ganglia), but no changes were observed in brain regions that are relatively spared following an HI insult (brain stem and cerebellum). Increased pGFAP expression was associated with poor neurological outcomes such as abnormal encephalography and neurobehaviour, and increased histological brain damage. Phosphorylation of GFAP may play an important role in astrocyte remodelling during development and disease and could potentially contribute to the plasticity of the central nervous system.


Assuntos
Animais Recém-Nascidos , Proteína Glial Fibrilar Ácida/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Animais , Western Blotting , Eletroencefalografia , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/fisiopatologia , Imuno-Histoquímica , Fosforilação , Suínos
13.
Brain Res ; 1319: 164-74, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20079338

RESUMO

White matter damage is a significant problem in the human pre-term baby. Damage to white matter is usually associated with injury or insults to babies born prematurely, typically before 32weeks' gestation, however there is increasing evidence of both grey and white matter damage occurring after 32weeks' gestation. Astrocytes play a vital role in white matter, regulating molecules such as glutamate in the extracellular space and preventing excitotoxic damage to neighbouring oligodendrocytes and axons. We have previously described dramatic changes in grey matter astrocytes in response to a hypoxic/ischemic (H/I) insult around the time of birth. In this study, we have used GFAP immunohistochemistry and Golgi-Kopsch staining to examine the morphology of white matter astrocytes in control neonatal pig brains, and in the brains of animals exposed to the same (perinatal) H/I insult. We demonstrate that the areal percentage of the section occupied by GFAP-immunoreactive processes and cell bodies is significantly decreased (by 46%, P<0.0001) in subcortical white matter from H/I brains. This loss of GFAP was accompanied by alterations in astrocyte morphology and an overall decrease in the size (field of section occupied by an individual astrocyte) of white matter astrocytes from 649microm(2) to 426microm(2), as revealed by Golgi-Kopsch staining and image analysis. These data suggest that astrocytes may contribute to the pathology of white matter damage following an H/I insult around the time of birth, and suggest that astrocytes may offer a novel target for therapies to improve outcomes after H/I.


Assuntos
Astrócitos/metabolismo , Astrócitos/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Animais , Animais Recém-Nascidos , Tamanho Celular , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Imuno-Histoquímica , Masculino , Microscopia Confocal , Bainha de Mielina , Suínos
14.
Glia ; 58(2): 181-94, 2010 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-19606499

RESUMO

Astrocytes play a vital role in the brain; their structural integrity and sustained function are essential for neuronal viability, especially after injury or insult. In this study, we have examined the response of astrocytes to hypoxia/ischemia (H/I), employing multiple methods (immunohistochemistry, iontophoretic cell injection, Golgi-Kopsch staining, and D-aspartate uptake) in a neonatal pig model of H/I. We have identified morphological changes in cortical gray matter astrocytes in response to H/I. Initial astrocytic changes were evident as early as 8 h post-insult, before histological evidence for neuronal damage. By 72 h post-insult, astrocytes exhibited significantly fewer processes that were shorter, thicker, and had abnormal terminal swellings, compared with astrocytes from control brains that exhibited a complex structure with multiple fine branching processes. Quantification and image analysis of astrocytes at 72 h post-insult revealed significant decreases in the average astrocyte size, from 686 microm(2) in controls to 401 microm(2) in H/I brains. Sholl analysis revealed a significant decrease (>60%) in the complexity of astrocyte branching between 5 and 20 microm from the cell body. D-Aspartate uptake studies revealed that the H/I insult resulted in impaired astrocyte function, with significantly reduced clearance of the glutamate analog, D-aspartate. These results suggest that astrocytes may be involved in the pathophysiological events of H/I brain damage at a far earlier time point than first thought. Developing therapies that prevent or reverse these astrocytic changes may potentially improve neuronal survival and thus might be a useful strategy to minimize brain damage after an H/I insult.


Assuntos
Astrócitos/patologia , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/patologia , Fibras Nervosas Amielínicas , Animais , Animais Recém-Nascidos , Astrócitos/fisiologia , Encéfalo/fisiopatologia , Tamanho Celular , Ácido D-Aspártico/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Ácido Glutâmico/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Imuno-Histoquímica , Isoquinolinas , Masculino , Suínos , Fatores de Tempo
15.
J Chem Neuroanat ; 39(3): 211-20, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20034557

RESUMO

We have examined brains of neonatal pigs that were rendered hypoxic. Glutamine synthetase (GS), a key enzyme in the detoxification of glutamate and ammonia, was rapidly lost from astrocytes in regions susceptible to damage, including the CA1 of hippocampus and various cortical regions. Conversely, resilient areas such as the dentate gyrus exhibited little or no loss of GS. Onset of loss was rapid, patches of loss being evident by 1h post-insult, and loss was extensive by 24h and did not recover by 72 h. Examination of counterstained sections revealed that GS losses preceded any overt neuronal damage. Loss of GS from astrocytes would plausibly lead to a rise in intracellular glutamate, and could explain why reversal of astrocytic glutamate transport during hypoxia/ischaemia is conceptually possible.


Assuntos
Astrócitos/enzimologia , Encéfalo/enzimologia , Glutamato-Amônia Ligase/metabolismo , Hipóxia/fisiopatologia , Animais , Animais Recém-Nascidos , Encéfalo/patologia , Imuno-Histoquímica , Suínos
16.
J Child Neurol ; 24(2): 183-93, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19182156

RESUMO

Dopamine D(1) and D(2) receptor protein expression was examined by Western blotting in newborn infants dying from cerebral asphyxia between 31 and 42 weeks' gestation, and matched controls. Frontal, occipital, temporal, and motor cortex tissue samples were obtained at autopsy (median postmortem interval 35 hours) and frozen for storage at -80 degrees C. A total of 2 immunoreactive bands were detected with each primary antibody in infant brain, whereas a single band was present in adult human and rat tissue. Immunoreactivity varied between cortical areas for both receptors, but their regional patterns differed significantly. D(1) protein levels were higher in motor and temporal cortex than in frontal or occipital cortex. D(2) protein showed graded expression frontal > motor > occipital > temporal cortex. Asphyxia cases showed lower expression of the upper D(2) immunoreactive band, but no difference in regional pattern. Lower D(2) receptor expression may attenuate stress responses and underlie increased vulnerability to hypoxia at birth.


Assuntos
Asfixia Neonatal/metabolismo , Córtex Cerebral/metabolismo , Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , Western Blotting , Humanos , Imuno-Histoquímica , Recém-Nascido , Recém-Nascido Prematuro , Análise Multivariada , Análise de Regressão
17.
J Biol Chem ; 282(40): 29414-23, 2007 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-17684014

RESUMO

Glial fibrillary acidic protein (GFAP) is an enigmatic protein; it currently has no unambiguously defined role. It is expressed in the cytoskeleton of astrocytes in the mammalian brain. We have used co-immunoprecipitation to identify in vivo binding partners for GFAP in the rat and pig brain. We demonstrate interactions between GFAP, the glutamate transporter GLAST, the PDZ-binding protein NHERF1, and ezrin. These interactions are physiologically relevant; we demonstrate in vitro that transport of D-aspartate (a glutamate analogue) is significantly increased in the presence of GFAP and NHERF1. Moreover, we demonstrate in vivo that expression of GFAP is essential in retaining GLAST in the plasma membranes of astrocytes after an hypoxic insult. These data indicate that the cytoskeleton of the astrocyte plays an important role in protecting the brain against glutamate-mediated excitotoxicity.


Assuntos
Encéfalo/metabolismo , Proteínas do Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Transportador 1 de Aminoácido Excitatório/fisiologia , Proteína Glial Fibrilar Ácida/metabolismo , Animais , Transporte Biológico , Ácido D-Aspártico/metabolismo , Transportador 1 de Aminoácido Excitatório/metabolismo , Humanos , Camundongos , Microscopia de Fluorescência , Modelos Biológicos , Ratos , Suínos , Transfecção
18.
Brain Res ; 1100(1): 110-7, 2006 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-16765329

RESUMO

A comparison of a constant (continuous delivery of 4% FiO2) and a variable (initial 5% FiO2 with adjustments to induce low amplitude EEG (LAEEG) and hypotension) hypoxic/ischemic insult was performed to determine which insult was more effective in producing a consistent degree of survivable neuropathological damage in a newborn piglet model of perinatal asphyxia. We also examined which physiological responses contributed to this outcome. Thirty-nine 1-day-old piglets were subjected to either a constant hypoxic/ischemic insult of 30- to 37-min duration or a variable hypoxic/ischemic insult of 30-min low peak amplitude EEG (LAEEG <5 microV) including 10 min of low mean arterial blood pressure (MABP <70% of baseline). Control animals (n = 6) received 21% FiO2 for the duration of the experiment. At 72 h, the piglets were euthanased, their brains removed and fixed in 4% paraformaldehyde and assessed for hypoxic/ischemic injury by histological analysis. Based on neuropathology scores, piglets were grouped as undamaged or damaged; piglets that did not survive to 72 h were grouped separately as dead. The variable insult resulted in a greater number of piglets with neuropathological damage (undamaged = 12.5%, damaged = 68.75%, dead = 18.75%) while the constant insult resulted in a large proportion of undamaged piglets (undamaged = 50%, damaged = 22.2%, dead = 27.8%). A hypoxic insult varied to maintain peak amplitude EEG <5 microV results in a greater number of survivors with a consistent degree of neuropathological damage than a constant hypoxic insult. Physiological variables MABP, LAEEG, pH and arterial base excess were found to be significantly associated with neuropathological outcome.


Assuntos
Hipóxia-Isquemia Encefálica/patologia , Consumo de Oxigênio/fisiologia , Oxigênio/administração & dosagem , Envelhecimento/fisiologia , Anestesia , Animais , Animais Recém-Nascidos , Gasometria , Pressão Sanguínea/fisiologia , Peso Corporal/fisiologia , Encéfalo/patologia , Interpretação Estatística de Dados , Eletroencefalografia/efeitos dos fármacos , Respiração Artificial , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...