Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Biol Sex Differ ; 15(1): 30, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38566248

RESUMO

BACKGROUND: Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of learning disabilities and memory deficits in children. In both human and animal studies, female neonate brains are less susceptible to HI than male brains. Phosphorylation of the nerve growth factor receptor TrkB has been shown to provide sex-specific neuroprotection following in vivo HI in female mice in an estrogen receptor alpha (ERα)-dependent manner. However, the molecular and cellular mechanisms conferring sex-specific neonatal neuroprotection remain incompletely understood. Here, we test whether female neonatal hippocampal neurons express autonomous neuroprotective properties and assess the ability of testosterone (T) to alter this phenotype. METHODS: We cultured sexed hippocampal neurons from ERα+/+ and ERα-/- mice and subjected them to 4 h oxygen glucose deprivation and 24 h reoxygenation (4-OGD/24-REOX). Sexed hippocampal neurons were treated either with vehicle control (VC) or the TrkB agonist 7,8-dihydroxyflavone (7,8-DHF) following in vitro ischemia. End points at 24 h REOX were TrkB phosphorylation (p-TrkB) and neuronal survival assessed by immunohistochemistry. In addition, in vitro ischemia-mediated ERα gene expression in hippocampal neurons were investigated following testosterone (T) pre-treatment and TrkB antagonist therapy via q-RTPCR. Multifactorial analysis of variance was conducted to test for significant differences between experimental conditions. RESULTS: Under normoxic conditions, administration of 3 µM 7,8-DHF resulted an ERα-dependent increase in p-TrkB immunoexpression that was higher in female, as compared to male neurons. Following 4-OGD/24-REOX, p-TrkB expression increased 20% in both male and female ERα+/+ neurons. However, with 3 µM 7,8-DHF treatment p-TrkB expression increased further in female neurons by 2.81 ± 0.79-fold and was ERα dependent. 4-OGD/24-REOX resulted in a 56% increase in cell death, but only female cells were rescued with 3 µM 7,8-DHF, again in an ERα dependent manner. Following 4-OGD/3-REOX, ERα mRNA increased ~ 3 fold in female neurons. This increase was blocked with either the TrkB antagonist ANA-12 or pre-treatment with T. Pre-treatment with T also blocked the 7,8-DHF- dependent sex-specific neuronal survival in female neurons following 4-OGD/24-REOX. CONCLUSIONS: OGD/REOX results in sex-dependent TrkB phosphorylation in female neurons that increases further with 7,8-DHF treatment. TrkB phosphorylation by 7,8-DHF increased ERα mRNA expression and promoted cell survival preferentially in female hippocampal neurons. The sex-dependent neuroprotective actions of 7,8-DHF were blocked by either ANA-12 or by T pre-treatment. These results are consistent with a model for a female-specific neuroprotective pathway in hippocampal neurons in response to hypoxia. The pathway is activated by 7,8-DHF, mediated by TrkB phosphorylation, dependent on ERα and blocked by pre-exposure to T.


Assuntos
Receptor alfa de Estrogênio , Fármacos Neuroprotetores , Criança , Feminino , Animais , Masculino , Camundongos , Humanos , Receptor alfa de Estrogênio/metabolismo , Neuroproteção , Caracteres Sexuais , Testosterona/farmacologia , Testosterona/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/metabolismo , Neurônios/metabolismo , Hipocampo/metabolismo , Isquemia , Hipóxia/metabolismo , RNA Mensageiro/metabolismo
2.
Biol Sex Differ ; 15(1): 1, 2024 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-38178264

RESUMO

BACKGROUND: Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of life-long neurological morbidities that result in learning and memory impairments. Evidence suggests that male neonates are more susceptible to the detrimental effects of HI, yet the mechanisms mediating these sex-specific responses to neural injury in neonates remain poorly understood. We previously tested the effects of treatment with a small molecule agonist of the tyrosine kinase B receptor (TrkB), 7,8-dihydroxyflavone (DHF) following neonatal HI and determined that females, but not males exhibit increased phosphorylation of TrkB and reduced apoptosis in their hippocampi. Moreover, these female-specific effects of the TrkB agonist were found to be dependent upon the expression of Erα. These findings demonstrated that TrkB activation in the presence of Erα comprises one pathway by which neuroprotection may be conferred in a female-specific manner. The goal of this study was to determine the role of Erα-dependent TrkB-mediated neuroprotection in memory and anxiety in young adult mice exposed to HI during the neonatal period. METHODS: In this study, we used a unilateral hypoxic ischemic (HI) mouse model. Erα+/+ or Erα-/- mice were subjected to HI on postnatal day (P) 9 and mice were treated with either vehicle control or the TrkB agonist, DHF, for 7 days following HI. When mice reached young adulthood, we used the novel object recognition, novel object location and open field tests to assess long-term memory and anxiety-like behavior. The brains were then assessed for tissue damage using immunohistochemistry. RESULTS: Neonatal DHF treatment prevented HI-induced decrements in recognition and location memory in adulthood in females, but not in males. This protective effect was absent in female mice lacking Erα. The female-specific improved recognition and location memory outcomes in adulthood conferred by DHF therapy after neonatal HI tended to be or were Erα-dependent, respectively. Interestingly, DHF triggered anxiety-like behavior in both sexes only in the mice that lacked Erα. When we assessed the severity of injury, we found that DHF therapy did not decrease the percent tissue loss in proportion to functional recovery. We additionally observed that the presence of Erα significantly reduced overall HI-associated mortality in both sexes. CONCLUSIONS: These observations provide evidence for a therapeutic role for DHF in which TrkB-mediated sustained recovery of recognition and location memories in females are Erα-associated and dependent, respectively. However, the beneficial effects of DHF therapy did not include reduction of gross tissue loss but may be derived from the enhanced functioning of residual tissues in a cell-specific manner.


Periods of low oxygen delivery and blood flow to the brains of newborns are known to cause life-long impairments to their cognitive ability as adults. Interestingly, male newborns are more susceptible to this injury than females. The mechanisms causing this sex difference are poorly understood. Here we test the role of the nerve growth factor receptor tyrosine kinase B (TrkB) in providing long-term neuroprotection following neonatal hypoxia­ischemia (HI) in mice. We have previously shown that when mice are treated with the TrkB agonist 7,8-dihydroxyflavone (DHF) in the days following neonatal HI, the result is short-term neuroprotection only in females and this protection is dependent on the presence of the estrogen receptor alpha receptor ([Formula: see text]). In this study, we extend these observations by subjecting mice either with or without [Formula: see text] to HI. Some of the mice were then treated with DHF immediately after HI. As adults, we performed tests to assess the mice's memory and anxiety-like behavior. At the end of these tests, we assessed the brains for tissue loss. Our results show that as adults the DHF treatment following HI in neonatal mice preserved memory only in females and this effect was dependent on the presence of [Formula: see text]. In addition, DHF therapy triggered anxiety-like behavior in mice lacking [Formula: see text]. We also show that this neuroprotection is not dependent on preservation of brain tissue following the injury. These results provide insight into the mechanisms behind the female resistance to hypoxic ischemic episodes as newborns.


Assuntos
Hipóxia-Isquemia Encefálica , Receptores Proteína Tirosina Quinases , Animais , Camundongos , Masculino , Feminino , Receptores Proteína Tirosina Quinases/uso terapêutico , Neuroproteção , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Isquemia , Hipóxia
3.
Res Sq ; 2023 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-37720039

RESUMO

Background: Neonatal hypoxia ischemia (HI) related brain injury is one of the major causes of life-long neurological morbidities that result in learning and memory impairments. Evidence suggests that male neonates are more susceptible to the detrimental effects of HI, yet the mechanisms mediating these sex-specific responses to neural injury in neonates remain poorly understood. We previously tested the effects of treatment with a small molecule agonist of the tyrosine kinase B receptor (TrkB), 7,8-dihydroxyflavone (DHF) following neonatal HI and determined that females, but not males exhibit increased phosphorylation of TrkB and reduced apoptosis in their hippocampi. Moreover, these female-specific effects of the TrkB agonist were found to be dependent upon the expression of ERα. These findings demonstrated that TrkB activation in the presence of ERα comprises one pathway by which neuroprotection may be conferred in a female-specific manner. The goal of this study was to determine the role of ERα-dependent TrkB-mediated neuroprotection in memory and anxiety in young adult mice exposed to HI during the neonatal period. Methods: In this study we used a unilateral hypoxic ischemic (HI) mouse model. ERα+/+ or ERα-/- mice were subjected to HI on postnatal day (P) 9 and mice were treated with either vehicle control or the TrkB agonist, DHF, for seven days following HI. When mice reached young adulthood, we used the novel object recognition, novel object location and open field tests to assess long-term memory and anxiety like behavior. The brains were then assessed for tissue damage using immunohistochemistry. Results: Neonatal DHF treatment prevented HI-induced decrements in recognition and location memory in adulthood in females, but not in males. This protective effect was absent in female mice lacking ERα. Thus, the female-specific and ERα-dependent neuroprotection conferred by DHF therapy after neonatal HI was associated with improved learning and memory outcomes in adulthood. Interestingly, DHF triggered anxiety like behavior in both sexes only in the mice that lacked ERα. When we assessed the severity of injury, we found that DHF therapy did not decrease the percent tissue loss in proportion to functional recovery. We additionally observed that the presence of ERα significantly reduced overall HI-associated mortality in both sexes. Conclusions: These observations provide evidence for a therapeutic role for DHF in which sustained recovery of memory in females is TrkB-mediated and ERα-dependent. However, the beneficial effects of DHF therapy did not include reduction of gross tissue loss but may be derived from the enhanced functioning of residual tissues in a cell-specific manner.

4.
Neurochem Int ; 127: 137-147, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30639264

RESUMO

BACKGROUND: Neuroinflammation plays an important role in ischemic brain injury and recovery, however the interplay between brain development and the neuroinflammatory response is poorly understood. We previously described age-dependent differences in the microglial response and the effect of microglial inhibition. Here we investigate whether age-dependent microglial responses may be related to pre-injury developmental differences in microglial phenotype. METHODS: Measures of microglia morphology were quantified using semi-automated software analysis of immunostained sections from postnatal day 2 (P2), P9, P30 and P60 mice using IMARIS. Microglia were isolated from P2, P9, P30 and P60 mice, and expression of markers of classical and alternative microglial activation was assessed, as well as transforming growth factor beta (TGF-ß) receptor, Serpine1, Mer Tyrosine Kinase (MerTK), and the suppressor of cytokine signaling (SOCS3). Hypoxia-ischemia (HI) was induced in P9 and P30 mice using unilateral carotid artery ligation and exposure to 10% oxygen for 50 min. Microglia morphology and microglial expression of genes in the TGF-ß and MerTK pathways were determined in ipsilateral and contralateral hippocampus. RESULTS: A progressive and significant increase in microglia branching morphology was seen in all brain regions from P2 to P30. No consistent classical or alternative activation profile was seen in isolated microglia. A clear transition to increased expression of TGF-ß and its downstream effector serpine1 was seen between P9 and P30. A similar increase in expression was seen in MerTK and its downstream effector SOCS3. HI resulted in a significant decrease in branching morphology only in the P9 mice, and expression of TGF-ß receptor, Serpine1, MerTK, and SOCS3 were elevated in P30 mice compared to P9 post-HI. CONCLUSION: Microglia maturation is associated with changes in morphology and gene expression, and microglial responses to ischemia in the developing brain differ based on the age at which injury occurs.


Assuntos
Expressão Gênica/fisiologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia/metabolismo , Microglia/patologia , Animais , Animais Recém-Nascidos , Encéfalo/metabolismo , Forma Celular , Modelos Animais de Doenças , Hipocampo/metabolismo , Inflamação/metabolismo , Camundongos Endogâmicos C57BL , Microglia/citologia , Microglia/metabolismo
5.
J Vis Exp ; (116)2016 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-27805577

RESUMO

Astrogliosis following hypoxia/ischemia (HI)-related brain injury plays a role in increased morbidity and mortality in neonates. Recent clinical studies indicate that the severity of brain injury appear to be sex dependent, and that the male neonates are more susceptible to the effects of HI-related brain injury, resulting in more severe neurological outcomes as compared to females with comparable brain injuries. The development of reliable methods to isolate and maintain highly enriched populations of sexed hippocampal astrocytes is essential to understand the cellular basis of sex differences in the pathological consequences of neonatal HI. In this study, we describe a method for creating sex specific hippocampal astrocyte cultures that are subjected to a model of in-vitro ischemia, oxygen-glucose deprivation, followed by reoxygenation. Subsequent reactive astrogliosis was examined by immunostaining for the Glial Fibrillary Acidic Protein (GFAP) and S100B. This method provides a useful tool to study the role of male and female hippocampal astrocytes following neonatal HI, separately.


Assuntos
Astrócitos , Hipocampo , Caracteres Sexuais , Animais , Animais Recém-Nascidos , Técnicas de Cultura de Células , Modelos Animais de Doenças , Feminino , Humanos , Hipóxia-Isquemia Encefálica , Masculino , Camundongos
6.
J Neuroimmunol ; 291: 18-27, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26857490

RESUMO

We previously found increased microglial proliferation and pro-inflammatory cytokine release in infant mice compared to juvenile mice after hypoxia-ischemia (HI). The aim of the current study was to assess for differences in the effect of microglial suppression on HI-induced brain injury in infant and juvenile mice. HI was induced in neonatal (P9) and juvenile (P30) mice and minocycline or vehicle was administered at 2h and 24h post-HI. P9 minocycline-treated mice demonstrated early but transient improvements in neurologic injury, while P30 minocycline-treated mice demonstrated sustained improvements in cerebral atrophy and Morris Water Maze performance at 60days post-HI.


Assuntos
Envelhecimento , Lesões Encefálicas/etiologia , Lesões Encefálicas/patologia , Encéfalo/patologia , Hipóxia-Isquemia Encefálica/complicações , Microglia/metabolismo , Animais , Animais Recém-Nascidos , Lesões Encefálicas/tratamento farmacológico , Antígeno CD11b/metabolismo , Modelos Animais de Doenças , Citometria de Fluxo , Lateralidade Funcional , Hipóxia-Isquemia Encefálica/patologia , Deficiências da Aprendizagem/tratamento farmacológico , Deficiências da Aprendizagem/etiologia , Antígenos Comuns de Leucócito/metabolismo , Imageamento por Ressonância Magnética , Aprendizagem em Labirinto , Camundongos , Microglia/efeitos dos fármacos , Microglia/patologia , Minociclina/uso terapêutico , Exame Neurológico , Estatísticas não Paramétricas , Fatores de Tempo
7.
eNeuro ; 3(1)2016.
Artigo em Inglês | MEDLINE | ID: mdl-26839918

RESUMO

Male neonate brains are more susceptible to the effects of perinatal asphyxia resulting in hypoxia and ischemia (HI)-related brain injury. The relative resistance of female neonatal brains to adverse consequences of HI suggests that there are sex-specific mechanisms that afford females greater neuroprotection and/or facilitates recovery post-HI. We hypothesized that HI preferentially induces estrogen receptor α (ERα) expression in female neonatal hippocampi and that ERα is coupled to Src family kinase (SFK) activation that in turn augments phosphorylation of the TrkB and thereby results in decreased apoptosis. After inducing the Vannucci's HI model on P9 (C57BL/6J) mice, female and male ERα wild-type (ERα(+/+)) or ERα null mutant (ERα(-/-)) mice received vehicle control or the selective TrkB agonist 7,8-dihydroxyflavone (7,8-DHF). Hippocampi were collected for analysis of mRNA of ERα and BDNF, protein levels of ERα, p-TrkB, p-src, and cleaved caspase 3 (c-caspase-3) post-HI. Our results demonstrate that: (1) HI differentially induces ERα expression in the hippocampus of the female versus male neonate, (2) src and TrkB phosphorylation post-HI is greater in females than in males after 7,8-DHF therapy, (3) src and TrkB phosphorylation post-HI depend on the presence of ERα, and (4) TrkB agonist therapy decreases the c-caspase-3 only in ERα(+/+) female mice hippocampus. Together, these observations provide evidence that female-specific induction of ERα expression confers neuroprotection with TrkB agonist therapy via SFK activation and account for improved functional outcomes in female neonates post-HI.


Assuntos
Receptor alfa de Estrogênio/metabolismo , Hipocampo/fisiologia , Hipóxia-Isquemia Encefálica/metabolismo , Receptor trkB/metabolismo , Animais , Animais Recém-Nascidos , Apoptose , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Receptor alfa de Estrogênio/genética , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/metabolismo , Receptor trkB/agonistas , Quinases da Família src/metabolismo
8.
Behav Brain Res ; 286: 236-240, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25746451

RESUMO

5-Hydroxymethylcytosine (5-hmC) is a novel environmentally sensitive DNA modification that is highly enriched in post-mitotic neurons and is associated with active transcription of neuronal genes. Recently, 5-hmC was functionally linked to learning and cognition and these studies revealed an accumulation of 5-hmC in the prefrontal cortex of mice undergoing fear extinction. These studies led us to hypothesize a role for 5-hmC in response to stress. To test this hypothesis, we combined immunohistochemistry, tandem mass spectrometry, and tet-assisted sodium bisulfite sequencing (TAB-seq) analyses on tissue and DNA from the hippocampus of 7-week old male mice exposed to a single 30-min restraint stress. After first identifying that the broad neuronal distribution of 5-hmC is not disrupted by acute stress, we used TAB-seq to find a stress-induced increase of 5-hmC in the 3'UTR of the glucocorticoid receptor gene (Nr3c1). Nr3c1 has a well-defined role in the stress pathway and these data suggest that 5-hmC contributes to these processes. Together, these data indicate that a deeper investigation of stress-related 5-hmC levels may reveal an environmental impact on this newly discovered epigenetic mark in the brain.


Assuntos
Citosina/análogos & derivados , Hipocampo/metabolismo , Receptores de Glucocorticoides/metabolismo , Estresse Psicológico/metabolismo , Regiões 3' não Traduzidas , 5-Metilcitosina/análogos & derivados , Doença Aguda , Animais , Citosina/metabolismo , Modelos Animais de Doenças , Hipocampo/patologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Distribuição Aleatória , Receptores de Glucocorticoides/genética , Restrição Física , Estresse Psicológico/patologia
9.
PLoS One ; 9(1): e84294, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24392123

RESUMO

Hypoxia ischemia (HI)-related brain injury is the major cause of long-term morbidity in neonates. One characteristic hallmark of neonatal HI is the development of reactive astrogliosis in the hippocampus. However, the impact of reactive astrogliosis in hippocampal damage after neonatal HI is not fully understood. In the current study, we investigated the role of Na(+)/H(+) exchanger isoform 1 (NHE1) protein in mouse reactive hippocampal astrocyte function in an in vitro ischemia model (oxygen/glucose deprivation and reoxygenation, OGD/REOX). 2 h OGD significantly increased NHE1 protein expression and NHE1-mediated H(+) efflux in hippocampal astrocytes. NHE1 activity remained stimulated during 1-5 h REOX and returned to the basal level at 24 h REOX. NHE1 activation in hippocampal astrocytes resulted in intracellular Na(+) and Ca(2+) overload. The latter was mediated by reversal of Na(+)/Ca(2+) exchange. Hippocampal astrocytes also exhibited a robust release of gliotransmitters (glutamate and pro-inflammatory cytokines IL-6 and TNFα) during 1-24 h REOX. Interestingly, inhibition of NHE1 activity with its potent inhibitor HOE 642 not only reduced Na(+) overload but also gliotransmitter release from hippocampal astrocytes. The noncompetitive excitatory amino acid transporter inhibitor TBOA showed a similar effect on blocking the glutamate release. Taken together, we concluded that NHE1 plays an essential role in maintaining H(+) homeostasis in hippocampal astrocytes. Over-stimulation of NHE1 activity following in vitro ischemia disrupts Na(+) and Ca(2+) homeostasis, which reduces Na(+)-dependent glutamate uptake and promotes release of glutamate and cytokines from reactive astrocytes. Therefore, blocking sustained NHE1 activation in reactive astrocytes may provide neuroprotection following HI.


Assuntos
Astrócitos/metabolismo , Glucose/metabolismo , Hipocampo/metabolismo , Oxigênio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Transporte Biológico , Isquemia Encefálica/genética , Isquemia Encefálica/metabolismo , Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Morte Celular , Células Cultivadas , Citocinas/metabolismo , Modelos Animais de Doenças , Ácido Glutâmico/metabolismo , Camundongos , Neurotransmissores/metabolismo , Cultura Primária de Células , Sódio/metabolismo , Trocador de Sódio e Cálcio/metabolismo , Trocador 1 de Sódio-Hidrogênio , Regulação para Cima
10.
CNS Neurol Disord Drug Targets ; 12(3): 360-70, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23469848

RESUMO

In this study, we investigated the effects of a bioactive high-affinity TrkB receptor agonist 7,8- dihydroxyflavone (7,8 DHF) on neonatal brain injury in female and male mice after hypoxia ischemia (HI). HI was induced by exposure of postnatal day 9 (P9) mice to 10% O2 for 50 minutes at 37°C after unilateral ligation of the left common carotid artery. Animals were randomly assigned to HI-vehicle control group [phosphate buffered saline (PBS), intraperitoneally (i.p.)] or HI + 7,8 DHF-treated groups (5 mg/kg in PBS, i.p at 10 min, 24 h, or with subsequent daily injections up to 7 days after HI). The HI-vehicle control mice exhibited neuronal degeneration in the ipsilateral hippocampus and cortex with increased Fluoro-Jade C positive staining and loss of microtubule associated protein 2 expression. In contrast, the 7,8 DHF-treated mice showed less hippocampal neurodegeneration and astrogliosis, with more profound effects in female than in male mice. Moreover, 7,8 DHF-treated mice improved motor learning and spatial learning at P30-60 compared to the HI-vehicle control mice. Diffusion tensor imaging of ex vivo brain tissues at P90 after HI revealed less reduction of fractional anisotropy values in the ipsilateral corpus callosum of 7,8 DHF-treated brains, which was accompanied with better preserved myelin basic protein expression and CA1 hippocampal structure. Taken together, these findings strongly suggest that TrkB agonist 7,8 DHF is protective against HI-mediated hippocampal neuronal death, white matter injury, and improves neurological function, with a more profound response in female than in male mice.


Assuntos
Flavonas/farmacologia , Degeneração Neural/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Receptor trkB/agonistas , Caracteres Sexuais , Envelhecimento , Animais , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Corpo Caloso/patologia , Feminino , Flavonas/uso terapêutico , Gliose/complicações , Gliose/tratamento farmacológico , Hipocampo/metabolismo , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Aprendizagem/efeitos dos fármacos , Masculino , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Proteína Básica da Mielina/metabolismo , Fibras Nervosas Mielinizadas/patologia , Neuroimagem , Fármacos Neuroprotetores/uso terapêutico , Recuperação de Função Fisiológica/efeitos dos fármacos
11.
CNS Neurol Disord Drug Targets ; 12(3): 338-49, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23469850

RESUMO

In the present study, we tested whether the ongoing differentiation of microglia in the immature brain results in more robust microglial activation and pro-inflammatory responses than juvenile brains following hypoxia-ischemia (HI). Under normoxic conditions, microglial activation profiles were assessed in postnatal day 9 and postnatal day 30 mice (P9 and P30) by analyzing relative expression levels of CD45 in CD11b+/CD45+ microglia/macrophages. Flow cytometry analysis revealed that the hippocampi of P9 and P30 brains exhibited higher levels of CD45 expression in CD11b+/CD45+ cells than in the cortex and striatum. In response to HI, there was an early increase in number of CD11b+/CD45+ microglia/macrophages in the ipsilateral hippocampus of P9 mice. These cells transformed from a "ramified" to an "amoeboid" morphology in the CA1 region, which was accompanied by a loss of microtubule-associated protein 2 immunostaining in this brain region. The peak response of microglial activation in the ipsilateral hippocampus of P9 mice occurred on day 2 post-HI, which was in contrast to a delayed and persistent microglial activation in the cortex and striatum (peak on day 9 post-HI). P9 brains demonstrated a 2-3 fold greater increase in microglia counts than P30 brains in each region (hippocampus, cortex, and striatum) during day 1-17 post-HI. P9 brains also showed more robust expression of pro-inflammatory cytokines (tumor necrosis factor-alpha, interleukin-1ß) than P30 brains. Taken together, compared to P30 mice, P9 mice demonstrated differences in microglial activation and pro-inflammatory responses after HI, which may be important in brain damage and tissue repair.


Assuntos
Envelhecimento/fisiologia , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Hipocampo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Antígenos Comuns de Leucócito/biossíntese , Microglia/metabolismo , Animais , Encéfalo/fisiopatologia , Caspase 3/metabolismo , Proliferação de Células , Córtex Cerebral/metabolismo , Corpo Estriado/metabolismo , Interleucina-1beta/biossíntese , Camundongos , Microglia/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Fator de Necrose Tumoral alfa/biossíntese
12.
Cell Physiol Biochem ; 30(1): 33-48, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22759954

RESUMO

The hallmark of apoptosis is a significant reduction in cell volume (AVD) resulting from loss of K(+)(i) and Cl(-)(i). Loss of cell volume and lowering of ionic strength of intracellular K(+) and Cl(-) occur before any other detectable characteristics of apoptosis. In the present study, temozolomide (TMZ) triggered loss of K(+)(i) and Cl(-)(i) and AVD in primary glioblastoma multiforme (GBM) cancer cells (GC) and GC cancer stem cells (GSC). We hypothesize that Na(+)-K(+)-2Cl(-) cotransporter isoform 1 (NKCC1) counteracts AVD during apoptosis in GBM cancer cells by regulating cell volume and Cl(-) homeostasis. NKCC1 protein was expressed in both GC and GSC and played an essential role in regulatory volume increase (RVI) in response to hypertonic cell shrinkage and isotonic cell shrinkage. Blocking NKCC1 activity with its potent inhibitor bumetanide abolished RVI. These cells maintained a basal [Cl(-)](i) (~ 68 mM) above the electrochemical equilibrium for Cl(-)(i). NKCC1 also functioned to replenish Cl(-)(i) levels following the loss of Cl(-)(i). TMZ-treated cells exhibited increased phosphorylation of NKCC1 and its up-stream novel Cl(-)/volume-sensitive regulatory kinase WNK1. Inhibition of NKCC1 activity with bumetanide accelerated AVD, early apoptosis, as well as activation of caspase-3 and caspase-8. Taken together, this study strongly suggests that NKCC1 is an essential mechanism in GBM cells to maintain K(+), Cl(-), and volume homeostasis to counteract TMZ-induced loss of K(+), Cl(-) and AVD. Therefore, blocking NKCC1 function augments TMZ-induced apoptosis in glioma cells.


Assuntos
Apoptose/efeitos dos fármacos , Bumetanida/farmacologia , Dacarbazina/análogos & derivados , Inibidores de Simportadores de Cloreto de Sódio e Potássio/farmacologia , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Caspase 3/metabolismo , Caspase 8/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Tamanho Celular/efeitos dos fármacos , Cloretos/metabolismo , Dacarbazina/farmacologia , Sinergismo Farmacológico , Glioblastoma , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Antígenos de Histocompatibilidade Menor , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/fisiologia , Potássio/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Membro 2 da Família 12 de Carreador de Soluto , Temozolomida , Imagem com Lapso de Tempo , Proteína Quinase 1 Deficiente de Lisina WNK
13.
J Neurochem ; 120(4): 622-30, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22129278

RESUMO

Docosahexaenoic acid (DHA) has neuroprotective effects in several neurodegenerative disease conditions. However, the underlying mechanisms are not well understood. In the present study, we investigated the effects of DHA on astrocyte Ca(2+) signaling under in vitro ischemic conditions (oxygen/glucose deprivation and reoxygenation, OGD/REOX). OGD (2h) triggered a Ca(2+) (ER) store overload (∼1.9-fold). Ca(2+) uptake by the Ca(2+) (ER) stores was further augmented during REOX and Ca(2+) (ER) was elevated by ∼4.7-fold at 90min REOX. Interestingly, Ca(2+) (ER) stores abruptly released Ca(2+) at ∼120min REOX and emptied at 160min REOX. Depletion of Ca(2+) (ER) stores led to delayed elevation of intracellular Ca(2+) concentration (Ca(2+) (cyt) ) and cell death. Activation of the purinergic receptor P2Y1 was responsible for the release of Ca(2+) (ER) . Most importantly, DHA blocked the initial Ca(2+) (ER) store overload, the delayed depletion of Ca(2+) (ER) , and rise in Ca(2+) (cyt) , which was in part via inhibiting d-myo-inositol 1,4,5-triphosphate receptors. The DHA metabolite DiHDoHE exhibited similar effects. DHA also attenuated expression of phosphorylated eukaryotic initiation factor 2α and activating transcription factor-4, two ER stress markers, following in vitro ischemia. Taken together, these findings suggest that DHA has protective effects in astrocytes following in vitro ischemia, in part, by inhibiting Ca(2+) dysregulation and ER stress.


Assuntos
Astrócitos/efeitos dos fármacos , Sinalização do Cálcio/efeitos dos fármacos , Cálcio/antagonistas & inibidores , Ácidos Docosa-Hexaenoicos/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Isquemia/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Hipóxia Celular/efeitos dos fármacos , Hipóxia Celular/fisiologia , Células Cultivadas , Retículo Endoplasmático/metabolismo , Retículo Endoplasmático/patologia , Estresse do Retículo Endoplasmático/fisiologia , Isquemia/metabolismo , Camundongos , Oxirredução/efeitos dos fármacos
14.
J Neurosci ; 30(45): 15210-20, 2010 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-21068326

RESUMO

H(+) extrusion is important for sustained NADPH oxidase activation after "respiratory" burst in macrophage/microglia activation. In this study, we investigated the role of Na(+)/H(+) exchanger isoform 1 (NHE-1) in activation of microglia after lipopolysaccharide (LPS) or oxygen and glucose deprivation and reoxygenation (OGD/REOX) exposure. NHE-1 functioned in maintaining basal pH(i) of immortalized M4T.4 microglia or mouse primary microglia. Pharmacological inhibition of NHE-1 activity with the potent inhibitor cariporide [HOE 642 (4-isopropyl-3-methylsulfonyl-benzoyl-guanidine-methanesulfonate)] abolished pH(i) regulation in microglia under basal conditions. Activation of microglia either by LPS, phorbol myristate acetate, or OGD/REOX accelerated pH(i) regulation and caused pH(i) elevation, which was accompanied with an increase in [Na(+)](i) and [Ca(2+)](i) as well as production of superoxide anion and cytokines. Interestingly, inhibition of NHE-1 not only abolished pH(i) regulation but also reduced production of superoxide anion as well as expression of cytokines and inducible nitric oxide synthase. Together, these results reveal that there was a concurrent activation of NHE-1 in microglia in response to proinflammatory stimuli. The study suggests that NHE-1 functions to maintain microglial pH(i) homeostasis allowing for sustained NADPH oxidase function and "respiratory" burst.


Assuntos
Encéfalo/metabolismo , Homeostase/fisiologia , Microglia/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Animais , Western Blotting , Encéfalo/citologia , Encéfalo/efeitos dos fármacos , Cálcio/metabolismo , Linhagem Celular , Células Cultivadas , Imunofluorescência , Glucose/deficiência , Guanidinas/farmacologia , Hipóxia/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Microglia/citologia , Microglia/efeitos dos fármacos , NADPH Oxidases/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Explosão Respiratória/fisiologia , Sulfonas/farmacologia
15.
J Biol Chem ; 285(45): 35155-68, 2010 Nov 05.
Artigo em Inglês | MEDLINE | ID: mdl-20817726

RESUMO

Neuronal dendrites are vulnerable to injury under diverse pathological conditions. However, the underlying mechanisms for dendritic Na(+) overload and the selective dendritic injury remain poorly understood. Our current study demonstrates that activation of NHE-1 (Na(+)/H(+) exchanger isoform 1) in dendrites presents a major pathway for Na(+) overload. Neuronal dendrites exhibited higher pH(i) regulation rates than soma as a result of a larger surface area/volume ratio. Following a 2-h oxygen glucose deprivation and a 1-h reoxygenation, NHE-1 activity was increased by ∼70-200% in dendrites. This elevation depended on activation of p90 ribosomal S6 kinase. Moreover, stimulation of NHE-1 caused dendritic Na(+)(i) accumulation, swelling, and a concurrent loss of Ca(2+)(i) homeostasis. The Ca(2+)(i) overload in dendrites preceded the changes in soma. Inhibition of NHE-1 or the reverse mode of Na(+)/Ca(2+) exchange prevented these changes. Mitochondrial membrane potential in dendrites depolarized 40 min earlier than soma following oxygen glucose deprivation/reoxygenation. Blocking NHE-1 activity not only attenuated loss of dendritic mitochondrial membrane potential and mitochondrial Ca(2+) homeostasis but also preserved dendritic membrane integrity. Taken together, our study demonstrates that NHE-1-mediated Na(+) entry and subsequent Na(+)/Ca(2+) exchange activation contribute to the selective dendritic vulnerability to in vitro ischemia.


Assuntos
Cálcio/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Dendritos/metabolismo , Homeostase , Hidrogênio/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Sódio/metabolismo , Animais , Hipóxia Celular , Células Cultivadas , Dendritos/patologia , Concentração de Íons de Hidrogênio , Transporte de Íons , Camundongos , Proteínas Quinases S6 Ribossômicas 90-kDa/metabolismo , Trocador 1 de Sódio-Hidrogênio
16.
J Neurochem ; 114(5): 1436-46, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20557423

RESUMO

In the present study, we investigated changes of cytosolic Ca2+([Ca2+](cyt)), endoplasmic reticulum Ca2+([Ca2+](ER)) and mitochondrial Ca2+(Ca2+(m)) in astrocytes following oxygen/glucose deprivation and reoxygenation (OGD/REOX). Two hours OGD did not cause changes in [Ca2+](cyt), but led to a significant increase in [Ca2+](ER). The elevation in [Ca2+](ER) continued and reached a peak level (130 +/- 2 microM) by 90 min REOX. An abrupt release of Ca2+(ER) occurred during 1.5-2.5 h REOX, which was accompanied with a delayed and sustained rise in [Ca2+](cyt). Moreover, Ca2+(m) content was increased significantly within 15 min REOX followed by a secondary rise (approximately 4.5-fold) and a release of mitochondrial cytochrome c (Cyt c). Astrocytes exhibited translocation of Cyt c from mitochondria to endoplasmic reticulum (ER) and up regulation of ER stress protein p-eIF2alpha. Blocking Na+-K+-Cl(-) cotransporter isoform 1 activity, either by its potent inhibitor bumetanide or genetic ablation, abolished release of ER Ca2+, delayed rise in [Ca2+](cyt) and Ca2+(m). Inhibition of the reverse mode operation of the Na+/Ca2+ exchanger significantly attenuated OGD/REOX-mediated Cyt c release. In summary, this study illustrates that OGD/REOX triggers a time-dependent loss of Ca2+ homeostasis in cytosol and organelles (ER and mitochondria) in astrocytes. Collective stimulation of Na+-K+-Cl(-) cotransporter isoform 1 and reverse mode function of Na+/Ca2+ exchanger contributes to these changes.


Assuntos
Astrócitos/metabolismo , Sinalização do Cálcio/fisiologia , Citocromos c/metabolismo , Retículo Endoplasmático/metabolismo , Glucose/deficiência , Mitocôndrias/metabolismo , Oxigênio/metabolismo , Animais , Astrócitos/enzimologia , Hipóxia Celular/fisiologia , Células Cultivadas , Retículo Endoplasmático/enzimologia , Camundongos , Camundongos Mutantes , Mitocôndrias/enzimologia , Consumo de Oxigênio/fisiologia
17.
Integr Biol (Camb) ; 2(1): 58-64, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20473413

RESUMO

Regulation of intracellular pH (pH(i)) in neurons is crucial to maintain their physiological function. In the current study, newly-developed polydimethylsiloxane (PDMS) microfluidic devices were used to independently investigate pH(i) regulation in neuronal soma and neurites. Embryonic cortical neurons were cultured in PDMS microfluidic devices with soma growing in one chamber (seeded) and neurites extending through a set of perpendicular microchannels into the opposite parallel chamber (non-seeded). Neurons in the microchambers were characterized by the vital dye calcein-red, polarized mitochondria, and expression of neuronal specific beta-tubulin (type-III), axonal Tau-1 protein, dendritic microtubule associated protein (MAP-2), and Na(+)/H(+) exchanger isoform 1 (NHE-1). Neurites exhibited higher resting pH(i) than soma (7.16 +/- 0.09 vs. 6.90 +/- 0.15). The neurites had a proton extrusion rate 3.7-fold faster than in soma following NH(4)Cl prepulse-mediated acidification (p < 0.05). The difference in the pH(i) regulation rates between neurites and soma can be accounted for by the larger surface area to volume ratio in the neurites. Interestingly, pharmacological inhibition of NHE-1 activity blocked the pH(i) regulation in soma and in neurites by approximately 70% (p < 0.05). Taken together, our study demonstrated that the microfluidic devices provide a useful tool to study neuronal pH(i) regulation in soma and their neurites. We conclude that NHE-1 plays an important role in regulation of pH(i) in both compartments.


Assuntos
DNA Glicosilases/metabolismo , Técnicas Analíticas Microfluídicas/métodos , Neuritos/metabolismo , Neurônios/metabolismo , ATPase Trocadora de Sódio-Potássio/fisiologia , Animais , Técnicas de Cultura de Células/métodos , Células Cultivadas , Concentração de Íons de Hidrogênio , Ratos
18.
J Neurosci ; 30(10): 3555-66, 2010 Mar 10.
Artigo em Inglês | MEDLINE | ID: mdl-20219989

RESUMO

The node of Ranvier is a tiny segment of a myelinated fiber with various types of specializations adapted for generation of high-speed nerve impulses. It is ionically specialized with respect to ion channel segregation and ionic fluxes, and metabolically specialized in ionic pump expression and mitochondrial density augmentation. This report examines the interplay of three important parameters (calcium fluxes, Na pumps, mitochondrial motility) at nodes of Ranvier in frog during normal nerve activity. First, we used calcium dyes to resolve a highly localized elevation in axonal calcium at a node of Ranvier during action potentials, and showed that this calcium elevation retards mitochondrial motility during nerve impulses. Second, we found, surprisingly, that physiologic activation of the Na pumps retards mitochondrial motility. Blocking Na pumps alone greatly prevents action potentials from retarding mitochondrial motility, which reveals that mitochondrial motility is coupled to Na/K-ATPase. In conclusion, we suggest that during normal nerve activity, Ca elevation and activation of Na/K-ATPase act, possibly in a synergistic manner, to recruit mitochondria to a node of Ranvier to match metabolic needs.


Assuntos
Potenciais de Ação/fisiologia , Cálcio/fisiologia , Mitocôndrias/enzimologia , Fibras Nervosas Mielinizadas/enzimologia , Nós Neurofibrosos/enzimologia , ATPase Trocadora de Sódio-Potássio/fisiologia , Potenciais de Ação/efeitos dos fármacos , Animais , Bloqueadores dos Canais de Cálcio/farmacologia , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Fibras Nervosas Mielinizadas/efeitos dos fármacos , Fibras Nervosas Mielinizadas/metabolismo , Nós Neurofibrosos/efeitos dos fármacos , Nós Neurofibrosos/metabolismo , Xenopus laevis
19.
J Neurochem ; 112(1): 173-82, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19840218

RESUMO

In this study, we investigated whether disruption of Na(+) and Ca(2+) homeostasis via activation of Na(+)-K(+)-Cl(-) cotransporter isoform 1 (NKCC1) and reversal of Na(+)/Ca(2+) exchange (NCX(rev)) affects protein aggregation and degradation following oxygen-glucose deprivation (OGD). Cultured cortical neurons were subjected to 2 h OGD and 1-24 h reoxygenation (REOX). Redistribution of ubiquitin and formation of ubiquitin-conjugated protein aggregates occurred in neurons as early as 2 h REOX. The protein aggregation progressed further by 8 h REOX. There was no significant recovery at 24 h REOX. Moreover, the proteasome activity in neurons was inhibited by 80-90% during 2-8 h REOX and recovered partially at 24 h REOX. Interestingly, pharmacological inhibition or genetic ablation of NKCC1 activity significantly decreased accumulation of ubiquitin-conjugated protein aggregates and improved proteasome activity. A similar protective effect was obtained by blocking NCX(rev) activity. Inhibition of NKCC1 activity also preserved intracellular ATP and Na(+) homeostasis during 0-24 h REOX. In a positive control study, disruption of endoplasmic reticulum Ca(2+) with thapsigargin triggered redistribution of free ubiquitin and protein aggregation. We conclude that overstimulation of NKCC1 and NCX(rev) following OGD/REOX partially contributes to protein aggregation and proteasome dysfunction as a result of ionic dysregulation.


Assuntos
Cálcio/fisiologia , Proteínas de Membrana Transportadoras/metabolismo , Neurônios/metabolismo , Sódio/fisiologia , Animais , Cátions Bivalentes/antagonistas & inibidores , Cátions Bivalentes/metabolismo , Cátions Monovalentes/antagonistas & inibidores , Cátions Monovalentes/metabolismo , Hipóxia Celular/fisiologia , Células Cultivadas , Feminino , Glucose/deficiência , Homeostase/genética , Homeostase/fisiologia , Hipóxia/metabolismo , Proteínas de Membrana Transportadoras/fisiologia , Camundongos , Neurônios/fisiologia , Gravidez , Complexo de Endopeptidases do Proteassoma/fisiologia , Inibidores de Proteassoma , Dobramento de Proteína , Multimerização Proteica , Simportadores de Cloreto de Sódio-Potássio/deficiência , Simportadores de Cloreto de Sódio-Potássio/genética , Simportadores de Cloreto de Sódio-Potássio/metabolismo , Membro 2 da Família 12 de Carreador de Soluto
20.
Exp Neurol ; 218(2): 213-20, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19427306

RESUMO

Recent discoveries show that caspase-independent cell death pathways are a pervasive mechanism in neurodegenerative diseases, and apoptosis-inducing factor (AIF) is an important effector of this mode of neuronal death. There are currently two known mechanisms underlying AIF release following excitotoxic stress, PARP-1 and calpain. To test whether there is an interaction between PARP-1 and calpain in triggering AIF release, we used the NMDA toxicity model in rat primary cortical neurons. Exposure to NMDA resulted in AIF truncation and nuclear translocation, and shRNA-mediated knockdown of AIF resulted in neuroprotection. Both calpain and PARP-1 are involved with AIF processing as AIF truncation, nuclear translocation and neuronal death were attenuated by calpain inhibition using adeno-associated virus-mediated overexpression of the endogenous calpain inhibitor, calpastatin, or treatment with the PARP-1 inhibitor 3-ABA. Activation of PARP-1 is necessary for calpain activation as PARP-1 inhibition blocked mitochondrial calpain activation. Finally, NMDA toxicity induces mitochondrial Ca(2+) dysregulation in a PARP-1 dependent manner. Thus, PARP-1 and mitochondrial calpain activation are linked via PARP-1-induced alterations in mitochondrial Ca(2+) homeostasis. Collectively, these findings link the two seemingly independent mechanisms triggering AIF-induced neuronal death.


Assuntos
Fator de Indução de Apoptose/biossíntese , Cálcio/metabolismo , Calpaína/metabolismo , Mitocôndrias/metabolismo , Neurônios/metabolismo , Poli(ADP-Ribose) Polimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Células Cultivadas , Córtex Cerebral/citologia , DNA Complementar , Agonistas de Aminoácidos Excitatórios/farmacologia , Vetores Genéticos , Proteínas de Fluorescência Verde/metabolismo , Mitocôndrias/efeitos dos fármacos , N-Metilaspartato/farmacologia , Neurônios/efeitos dos fármacos , Poli(ADP-Ribose) Polimerase-1 , RNA Interferente Pequeno , Ratos , Ratos Sprague-Dawley , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...