Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
J Neurosci ; 37(11): 3072-3084, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28188219

RESUMEN

Neurogenesis is essential to brain development and plays a central role in the response to brain injury. Stroke and head trauma stimulate proliferation of endogenous neural stem cells (NSCs); however, the survival of young neurons is sharply reduced by postinjury inflammation. Cellular mitochondria are critical to successful neurogenesis and are a major target of inflammatory injury. Mitochondrial protection was shown to improve survival of young neurons. This study tested whether reducing cellular microRNA-210 (miR-210) would enhance mitochondrial function and improve survival of young murine neurons under inflammatory conditions. Several studies have demonstrated the potential of miR-210 inhibition to enhance and protect mitochondrial function through upregulation of mitochondrial proteins. Here, miR-210 inhibition significantly increased neuronal survival and protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase in differentiating NSC cultures exposed to inflammatory mediators. Unexpectedly, we found that reducing miR-210 significantly attenuated NSC proliferation upon induction of differentiation. Further investigation revealed that increased mitochondrial function suppressed the shift to primarily glycolytic metabolism and reduced mitochondrial length characteristic of dividing cells. Activation of AMP-regulated protein kinase-retinoblastoma signaling is important in NSC proliferation and the reduction of this activation observed by miR-210 inhibition is one mechanism contributing to the reduced proliferation. Postinjury neurogenesis occurs as a burst of proliferation that peaks in days, followed by migration and differentiation over weeks. Our studies suggest that mitochondrial protective miR-210 inhibition should be delayed until after the initial burst of proliferation, but could be beneficial during the prolonged differentiation stage.SIGNIFICANCE STATEMENT Increasing the success of endogenous neurogenesis after brain injury holds therapeutic promise. Postinjury inflammation markedly reduces newborn neuron survival. This study found that enhancement of mitochondrial function by reducing microRNA-210 (miR-210) levels could improve survival of young neurons under inflammatory conditions. miR-210 inhibition protected the activity of mitochondrial enzymes cytochrome c oxidase and aconitase. Conversely, we observed decreased precursor cell proliferation likely due to suppression of the AMP-regulated protein kinase-retinoblastoma axis with miR-210 inhibition. Therefore, mitochondrial protection is a double-edged sword: early inhibition reduces proliferation, but inhibition later significantly increases neuroblast survival. This explains in part the contradictory published reports of the effects of miR-210 on neurogenesis.


Asunto(s)
Proliferación Celular , Supervivencia Celular/inmunología , Encefalitis/inmunología , MicroARNs/inmunología , Mitocondrias/inmunología , Neurogénesis/inmunología , Neuronas/inmunología , Animales , Citocinas/inmunología , Encefalitis/patología , Femenino , Inflamación/inmunología , Inflamación/patología , Masculino , Ratones , Mitocondrias/patología , Neuronas/patología
2.
Mol Cell Neurosci ; 82: 118-125, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28522364

RESUMEN

Whether the effect of miR-181a is sexually dimorphic in stroke is unknown. Prior work showed protection of male mice with miR-181a inhibition. Estrogen receptor-α (ERα) is an identified target of miR181 in endometrium. Therefore we investigated the separate and joint effects of miR-181a inhibition and 17ß-estradiol (E2) replacement after ovariectomy. Adult female mice were ovariectomized and implanted with an E2- or vehicle-containing capsule for 14d prior to 1h middle cerebral artery occlusion (MCAO). Each group received either miR-181a antagomir or mismatch control by intracerebroventricular injection 24h before MCAO. After MCAO neurologic deficit and infarct volume were assessed. Primary male and female astrocyte cultures were subjected to glucose deprivation with miR-181a inhibitor or transfection control, and E2 or vehicle control, with/without ESRα knockdown with small interfering RNA. Cell death was assessed by propidium iodide staining, and lactate dehydrogenase assay. A miR-181a/ERα target site blocker (TSB), with/without miR-181a mimic, was used to confirm targeting of ERα by miR-181a in astrocytes. Individually, miR-181a inhibition or E2 decreased infarct volume and improved neurologic score in female mice, and protected male and female astrocyte cultures. Combined miR-181a inhibition plus E2 afforded greater protection of female mice and female astrocyte cultures, but not in male astrocyte cultures. MiR-181a inhibition only increased ERα levels in vivo and in female cultures, while ERα knockdown with siRNA increased cell death in both sexes. Treatment with ERα TSB was strongly protective in both sexes. In conclusion, the results of the present study suggest miR-181a inhibition enhances E2-mediated stroke protection in females in part by augmenting ERα production, a mechanism detected in female mice and female astrocytes. Sex differences were observed with combined miR-181a inhibition/E2 treatment, and miR-181a targeting of ERα.


Asunto(s)
Astrocitos/metabolismo , Isquemia Encefálica/genética , Receptor alfa de Estrógeno/genética , Ataque Isquémico Transitorio/metabolismo , MicroARNs/genética , Animales , Astrocitos/efectos de los fármacos , Isquemia Encefálica/metabolismo , Modelos Animales de Enfermedad , Femenino , Ataque Isquémico Transitorio/genética , Masculino , Ratones Endogámicos C57BL , Neuronas/metabolismo , Fármacos Neuroprotectores/farmacología , Daño por Reperfusión/genética , Daño por Reperfusión/metabolismo , Factores Sexuales
3.
Stroke ; 46(8): 2271-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26130091

RESUMEN

BACKGROUND AND PURPOSE: Interleukin (IL)-4 protects from middle cerebral artery occlusion in male mice. Females generally show less injury in response to the same ischemic challenge, but the underlying mechanisms are not fully understood. We tested the importance of IL-4 in female protection using IL-4 knockout (KO) mice. METHODS: IL-4 KO and wild-type (WT) mice of both sexes were subjected to middle cerebral artery occlusion. Infarct volume was assessed by triphenyltetrazolium chloride staining and neurobehavioral outcome by neuroscore. T cell proliferation was assessed after Concanavalin A exposure. Ischemic brain immune cell populations were analyzed by fluorescence-activated cell sorting and immunostaining. RESULTS: Infarction in WT females during estrus and proestrus phases was significantly smaller than in males; neurological score was better. Infarction volume was larger and neurological score worse in IL-4 KO compared with WT in both sexes, with no sex difference. Proliferation of T cells was inhibited in WT females with higher proliferation and no sex difference in IL-4 KO. Macrophage numbers and total T cells in the ischemic hemisphere were lower in WT females, and monocytes increased markedly in IL-4 KOs with no sex difference. The reduced macrophage infiltration in WT-females was predominantly M2. Loss of IL-4 increased CD68+ and iNOS+ cells and reduced YM1+ and Arg1+ cells in both sexes. CONCLUSIONS: IL-4 is required for female neuroprotection during the estrus phase of the estrus cycle. Protected WT females show a predominance of M2-activated microglia/macrophages and reduced inflammatory infiltration. Increasing macrophage M2 polarization, with or without added inhibition of infiltration, may be a new approach to stroke treatment.


Asunto(s)
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevención & control , Interleucina-4/deficiencia , Caracteres Sexuales , Animales , Isquemia Encefálica/patología , Femenino , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Distribución Aleatoria
4.
Stroke ; 46(2): 551-6, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25604249

RESUMEN

BACKGROUND AND PURPOSE: MicroRNA (miR)-200c increases rapidly in the brain after transient cerebral ischemia but its role in poststroke brain injury is unclear. Reelin, a regulator of neuronal migration and synaptogenesis, is a predicted target of miR-200c. We hypothesized that miR-200c contributes to injury from transient cerebral ischemia by targeting reelin. METHODS: Brain infarct volume, neurological score and levels of miR-200c, reelin mRNA, and reelin protein were assessed in mice subjected to 1 hour of middle cerebral artery occlusion with or without intracerebroventricular infusion of miR-200c antagomir, mimic, or mismatch control. Direct targeting of reelin by miR-200c was assessed in vitro by dual luciferase assay and immunoblot. RESULTS: Pretreatment with miR-200c antagomir decreased post-middle cerebral artery occlusion brain levels of miR-200c, resulting in a significant reduction in infarct volume and neurological deficit. Changes in brain levels of miR-200c inversely correlated with reelin protein expression. Direct targeting of the Reln 3' untranslated region by miR-200c was verified with dual luciferase assay. Inhibition of miR-200c resulted in an increase in cell survival subsequent to in vitro oxidative injury. This effect was blocked by knockdown of reelin mRNA, whereas application of reelin protein afforded protection. CONCLUSIONS: These findings suggest that the poststroke increase in miR-200c contributes to brain cell death by inhibiting reelin expression, and that reducing poststroke miR-200c is a potential target to mitigate stroke-induced brain injury.


Asunto(s)
Isquemia Encefálica/metabolismo , Moléculas de Adhesión Celular Neuronal/biosíntesis , Proteínas de la Matriz Extracelular/biosíntesis , MicroARNs/administración & dosificación , MicroARNs/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Serina Endopeptidasas/biosíntesis , Animales , Isquemia Encefálica/patología , Moléculas de Adhesión Celular Neuronal/antagonistas & inhibidores , Células Cultivadas , Proteínas de la Matriz Extracelular/antagonistas & inhibidores , Marcación de Gen , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Proteína Reelina
5.
Glia ; 61(11): 1784-94, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24038396

RESUMEN

Following transient forebrain ischemia, astrocytes play a key role in determining whether or not neurons in the hippocampal CA1 sector go on to die in a delayed fashion. MicroRNAs (miRNAs) are a novel class of RNAs that control gene expression at the post-transcriptional level and the miR-29 family is highly expressed in astrocytes. In this study we assessed levels of miR-29 in hippocampus following forebrain ischemia and found that after transient forebrain ischemia and short periods of reperfusion, miR-29a significantly increased in the resistant dentate gyrus, but decreased in the vulnerable CA1 region of the hippocampus. We demonstrate that miR-29a targets BH3-only proapoptotic BCL2 family member PUMA by luciferase reporter assay and by Western blot. Comparing primary neuron and astrocyte cultures, and postnatal brain, we verified the strongly astrocytic expression of miR-29a. We further found that miR-29a mimic protects and miR-29a inhibitor aggravates cell injury and mitochondrial function after ischemia-like stresses in vitro. Lastly, by overexpressing and reducing miR-29a we demonstrate the protective effect of miR-29a on CA1 delayed neuronal death after forebrain ischemia. Our data suggest that by targeting a pro-apoptotic BCL2 family member, increasing levels of miR-29a might emerge as a strategy for protection against ischemia-reperfusion injury.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Astrocitos/metabolismo , Ataque Isquémico Transitorio/metabolismo , MicroARNs/metabolismo , Neuronas/metabolismo , Prosencéfalo/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Muerte Celular/fisiología , Mitocondrias/genética , Mitocondrias/metabolismo , Ratas , Daño por Reperfusión/metabolismo
6.
Neurobiol Dis ; 45(1): 555-63, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21983159

RESUMEN

MicroRNAs (miRNA) are short (~22nt) single stranded RNAs that downregulate gene expression. Although recent studies indicate extensive miRNA changes in response to ischemic brain injury, there is currently little information on the roles of specific miRNAs in this setting. Heat shock proteins (HSP) of the HSP70 family have been extensively studied for their multiple roles in cellular protection, but there is little information on their regulation by miRNAs. We used bioinformatics to identify miR-181 as a possible regulator of several HSP70 family members. We validated GRP78/BIP as a target by dual luciferase assay. In response to stroke in the mouse we find that miR-181 increases in the core, where cells die, but decreases in the penumbra, where cells survive. Increased levels of miR-181a are associated with decreased GRP78 protein levels, but increased levels of mRNA, implicating translational arrest. We manipulated levels of miR-181a using plasmid overexpression of pri-miR-181ab or mimic to increase, and antagomir or inhibitor to reduce levels. Increased miR-181a exacerbated injury both in vitro and in the mouse stroke model. Conversely, reduced levels were associated with reduced injury and increased GRP78 protein levels. Studies in C6 cells show that if GRP78 levels are maintained miR-181a no longer exerts a toxic effect. These data demonstrate that miR-181 levels change in response to stroke and inversely correlate with levels of GRP78. Importantly, reducing or blocking miR-181a protects the brain from stroke.


Asunto(s)
Isquemia Encefálica/genética , Encéfalo/metabolismo , Proteínas de Choque Térmico/genética , MicroARNs/genética , Accidente Cerebrovascular/genética , Animales , Isquemia Encefálica/metabolismo , Chaperón BiP del Retículo Endoplásmico , Expresión Génica , Proteínas de Choque Térmico/metabolismo , Ratones , MicroARNs/metabolismo , Neuronas/metabolismo , Accidente Cerebrovascular/metabolismo
7.
Stroke ; 42(7): 2026-32, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21597016

RESUMEN

BACKGROUND AND PURPOSE: Stroke causes brain injury with activation of an inflammatory response that can contribute to injury. We tested the hypothesis that the anti-inflammatory cytokine interleukin-4 (IL-4) reduces injury after stroke using IL-4 knockout (KO) adult male mice. METHODS: IL-4 KO and wild-type mice were subjected to transient middle cerebral artery occlusion. Outcome was assessed by triphenyltetrazolium chloride staining for infarct volume, neuroscore and spontaneous activity for behavioral outcome, and immunostaining and stereological counting for cellular response. RESULTS: Infarction volume at 24 hours was significantly larger in IL-4 KO mice, neurological score was significantly worse, and spontaneous activity was reduced compared with wild-type mice. Increased macrophage/microglial infiltration, increased numbers of myeloperoxidase-positive cells, and increased Th1/Th2 ratio were observed in the infarct core in IL-4 KO mice. Reduced astrocyte activation was observed in the cortical penumbra in IL-4 KO mice. Recombinant IL-4 administered intracerebroventricularly before middle cerebral artery occlusion significantly reduced infarct volume, improved neurological score, reduced macrophages/microglia, and lowered the Th1/Th2 ratio in IL-4 KO mice, but not in wild-type. CONCLUSIONS: Loss of IL-4 signaling in KO mice was associated with worse outcome, and this was reversed by giving exogenous IL-4. Worsened outcome was associated with increased inflammation in the core, which was reversed in IL-4 KO but not significantly changed in wild-type mice by exogenous IL-4. This is consistent with IL-4 signaling leading to reduced inflammation in the core and a possible beneficial role for activated astrocytes in the penumbra.


Asunto(s)
Lesiones Encefálicas/etiología , Lesiones Encefálicas/genética , Interleucina-4/genética , Ataque Isquémico Transitorio/complicaciones , Animales , Astrocitos/metabolismo , Modelos Animales de Enfermedad , Inflamación , Ataque Isquémico Transitorio/genética , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Recombinantes/uso terapéutico , Transducción de Señal , Resultado del Tratamiento
8.
Glia ; 58(9): 1042-9, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20235222

RESUMEN

Brief forebrain ischemia is a model of the delayed hippocampal neuronal loss seen in patients following cardiac arrest and resuscitation. Previous studies demonstrated that selective dysfunction of hippocampal CA1 subregion astrocytes occurs hours to days before delayed neuronal death. In this study we tested the strategy of directing protection to astrocytes to protect neighboring neurons from forebrain ischemia. Two well-studied protective proteins, heat shock protein 72 (Hsp72) or superoxide dismutase 2 (SOD2), were genetically targeted for expression in astrocytes using the astrocyte-specific human glial fibrillary acidic protein (GFAP) promoter. The expression constructs were injected stereotacticly immediately above the hippocampal CA1 region on one side of the rat brain two days prior to forebrain ischemia. Cell type specific expression was confirmed by double label immunohistochemistry. When the expression constructs were injected two days before transient forebrain ischemia, the loss of CA1 hippocampal neurons observed seven days later was significantly reduced on the injected side compared with controls. This neuroprotection was associated with significantly better preservation of astrocyte glutamate transporter-1 immunoreactivity at 5-h reperfusion and reduced oxidative stress. Improving the resistance of astrocytes to ischemic stress by targeting either the cytosolic or mitochondrial compartment was thus associated with preservation of CA1 neurons following forebrain ischemia. Targeting astrocytes is a promising strategy for neuronal preservation following cardiac arrest and resuscitation.


Asunto(s)
Astrocitos/fisiología , Isquemia Encefálica/fisiopatología , Región CA1 Hipocampal/fisiopatología , Proteínas del Choque Térmico HSP72/metabolismo , Neuronas/fisiología , Superóxido Dismutasa/metabolismo , Animales , Región CA1 Hipocampal/patología , Hipoxia de la Célula/fisiología , Células Cultivadas , Transportador 2 de Aminoácidos Excitadores/metabolismo , Proteína Ácida Fibrilar de la Glía/genética , Proteínas del Choque Térmico HSP72/genética , Humanos , Masculino , Ratones , Neuronas/patología , Estrés Oxidativo/fisiología , Regiones Promotoras Genéticas , Prosencéfalo/patología , Prosencéfalo/fisiopatología , Ratas , Ratas Sprague-Dawley , Superóxido Dismutasa/genética , Transfección
9.
Anesth Analg ; 108(1): 280-7, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19095863

RESUMEN

BACKGROUND: Several beta-adrenergic receptor (betaAR) antagonists have been shown to have neuroprotective effects against cerebral ischemia. However, clenbuterol, a beta(2)AR agonist, was shown to have neuroprotective activity by increasing nerve growth factor expression. We used beta(2)AR knockout mice and a beta(2) selective antagonist to test the effect of loss of beta(2)ARs on outcome from transient focal cerebral ischemia. METHODS: Ischemia was induced by the intraluminal suture method, for 60 min of middle cerebral artery occlusion (MCAO) followed by 24 h reperfusion. Neurological score was determined at 24 h reperfusion and infarct size was determined by cresyl violet or 2,3,5-triphenyltetrazolium chloride staining. beta(2)AR knockout mice and wild-type congenic FVB/N controls were studied, as well as 2 groups of wild type mice given either ICI 118,551 (0.2 mg/kg) or 0.9% saline intraperitoneally 30 min before MCAO (n = 10 per group). Changes in expression of heat shock protein (Hsp)72 after ischemia were examined by immunohistochemistry and western blots. RESULTS: Compared with wild type littermates, infarct volume was decreased by 22.3% in beta(2)AR knockout mice (39.7 +/- 10.7 mm(3) vs 51.0 +/- 11.4 mm(3), n = 10/group, P = 0.034) after 60 min of MCAO followed by 24 h reperfusion. Pretreatment with a beta(2)AR selective antagonist, ICI 118,551, also decreased infarct size significantly, by 25.1%, compared with the saline control (32.8 +/- 11.9 mm(3) vs 43.8 +/- 10.3 mm(3), n = 10/group, P = 0.041). Neurological scores were also significantly improved in mice lacking the beta(2)AR or pretreated with ICI 118,551. After cerebral ischemia, total levels of Hsp72 and the number of Hsp72 immunopositive cells were greater in mice lacking beta(2) AR. CONCLUSION: Brain injury is reduced and neurological outcome improved after MCAO in mice lacking the beta(2)AR, or in wild type mice pretreated with a selective beta(2)AR antagonist. This is consistent with a shift away from prosurvival signaling to prodeath signaling in the presence of beta(2)AR activation in cerebral ischemia. Protection is associated with higher levels of Hsp72, a known antideath protein. The effect of beta(2)AR signaling in the setting of cerebral ischemia is complex and warrants further study.


Asunto(s)
Antagonistas Adrenérgicos beta/farmacología , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Ataque Isquémico Transitorio/prevención & control , Fármacos Neuroprotectores/farmacología , Propanolaminas/farmacología , Receptores Adrenérgicos beta 2/deficiencia , Antagonistas de Receptores Adrenérgicos beta 2 , Animales , Modelos Animales de Enfermedad , Proteínas del Choque Térmico HSP72/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ataque Isquémico Transitorio/etiología , Ataque Isquémico Transitorio/metabolismo , Ataque Isquémico Transitorio/patología , Masculino , Ratones , Ratones Noqueados , Actividad Motora/efectos de los fármacos , Receptores Adrenérgicos beta 2/genética , Regulación hacia Arriba
10.
Prog Neurobiol ; 176: 73-85, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30121237

RESUMEN

Cerebral ischemia remains a major cause of death and disability worldwide, yet therapeutic options remain limited. Differences in sex and age play an important role in the final outcome in response to cerebral ischemia in both experimental and clinical studies: males have a higher risk and worse outcome than females at younger ages and this trend reverses in older ages. Although the molecular mechanisms underlying sex dimorphism are complex and are still not well understood, studies suggest steroid hormones, sex chromosomes, differential cell death and immune pathways, and sex-specific microRNAs may contribute to the outcome following cerebral ischemia. This review focuses on differential effects between males and females on cell death and immunological pathways in response to cerebral ischemia, the central role of innate sex differences in steroid hormone signaling, and upstreamregulation of sexually dimorphic gene expression by microRNAs.


Asunto(s)
Isquemia Encefálica , MicroARNs , Caracteres Sexuales , Inmunidad Adaptativa/fisiología , Animales , Femenino , Humanos , Inmunidad Innata/fisiología , Masculino , Accidente Cerebrovascular/genética , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/fisiopatología , Transcriptoma
11.
eNeuro ; 6(4)2019.
Artículo en Inglés | MEDLINE | ID: mdl-31427401

RESUMEN

The cellular and molecular mechanisms regulating postinjury neurogenesis in the adult hippocampus remain undefined. We have previously demonstrated that preinjury treatment with anti-microRNA (miR)-181a preserved neurons and prevented astrocyte dysfunction in the hippocampal cornu ammonis-1 (CA1) following transient forebrain ischemia. In the present study, we assessed postinjury treatment with anti-miR-181a on recovery of CA1 neurons following transient forebrain ischemia in rats. Stereotactic CA1 injection of miR-181a antagomir at either 2 h or 7 d postinjury resulted in improved restoration of CA1 measured at 28 d postinjury. Treatment with antagomir was associated with overexpression of the mir-181a target cell adhesion-associated, oncogene-related protein and enhanced expression of the neuroprogenitor cell marker doublecortin (DCX) in the CA1. Assessment of GFAP+ cell fate by Cre/Lox-mediated deletion demonstrated that some GFAP+ cells in CA1 exhibited de novo DCX expression in response to injury. In vitro experiments using primary neuronal stem cells confirmed that miR-181a inhibition augmented the expression of DCX and directed cellular differentiation toward a neuronal fate. These results suggest that miR-181a inhibition plays a central role in the restoration of CA1 neurons via augmentation of early latent neurogenic gene activation in neural progenitor cells, including some reactive astrocytes. Therapeutic interventions targeting this restorative process may represent a novel postinjury approach to improve clinical outcomes in survivors of forebrain ischemia.


Asunto(s)
Antagomirs/administración & dosificación , Isquemia Encefálica/metabolismo , Región CA1 Hipocampal/metabolismo , MicroARNs/antagonistas & inhibidores , Neuronas/metabolismo , Animales , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Región CA1 Hipocampal/efectos de los fármacos , Proteína Doblecortina , Masculino , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Prosencéfalo/efectos de los fármacos , Prosencéfalo/fisiopatología , Ratas Sprague-Dawley
12.
J Neurosci ; 27(16): 4253-60, 2007 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-17442809

RESUMEN

Transient global ischemia, as with cardiac arrest, causes loss of CA1 hippocampal neurons 2-4 d later, whereas nearby dentate gyrus (DG) neurons are relatively resistant. Whether differential astrocyte vulnerability in ischemic injury contributes to CA1 neuronal death is uncertain. Here, we find that CA1 astrocytes are more sensitive to ischemia than DG astrocytes. In rats subjected to transient forebrain ischemia, CA1 astrocytes lose glutamate transport activity and immunoreactivity for GFAP, S100beta, and glutamate transporter GLT-1 within a few hours of reperfusion, but without astrocyte cell death. Oxidative stress may contribute to the observed selective CA1 changes, because CA1 astrocytes show early increases in mitochondrial free radicals and reduced mitochondrial membrane potential. Similar changes were not observed in DG astrocytes. Upregulation of GLT-1 expression in astrocytes with ceftriaxone protected CA1 neurons from forebrain ischemia. We suggest that greater oxidative stress and loss of GLT-1 function selectively in CA1 astrocytes is central to the well known delayed death of CA1 neurons.


Asunto(s)
Astrocitos/metabolismo , Transportador 2 de Aminoácidos Excitadores/metabolismo , Hipocampo/fisiopatología , Ataque Isquémico Transitorio/fisiopatología , Animales , Astrocitos/patología , Ceftriaxona/farmacología , Supervivencia Celular , Proteína Ácida Fibrilar de la Glía/metabolismo , Ácido Glutámico/farmacocinética , Inmunohistoquímica , Masculino , Potenciales de la Membrana , Mitocondrias/metabolismo , Estrés Oxidativo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba
13.
Cureus ; 9(6): e1414, 2017 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-28861331

RESUMEN

The effects of high dose gamma radiation on brain tissue are poorly understood, with both limited and major changes reported. The present study compared the effects of gamma irradiation on the expression of interneuron markers within the hippocampal cornu ammonis 1 (CA1) region with expression in control matched rats. This area was chosen for study because of its well-characterized circuitry. Male Sprague-Dawley rats were exposed to 60 Gy of whole brain gamma radiation and after 24 or 48 hours, the brains were removed, fixed and sectioned to quantitate expression of parvalbumin (PV), calbindin-D28K (CB), reelin, neuropeptide-Y (NPY), and somatostatin. All of these markers increased in expression over the first 48 hours, except NPY, which decreased. This provides novel information on changes in gene expression in the hippocampal interneurons following radiation. Staining for Beclin 1, a marker of autophagy, increased most strongly in the subgranular zone (SGZ) of the dentate gyrus (DG). Overall, the results are consistent with the hypothesis that increased intracellular calcium follows irradiation, leading to an increased expression of calcium binding proteins. Increased autophagy occurs in the neurogenic zone of the dentate hilus, consistent with reduced effective neurogenesis after irradiation.

14.
Cureus ; 9(3): e1076, 2017 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-28401026

RESUMEN

Studies on the effects of gamma radiation on brain tissue have produced markedly differing results, ranging from little effect to major pathology, following irradiation. The present study used control-matched animals to compare effects on a well characterized brain region following gamma irradiation. Male Sprague-Dawley rats were exposed to 60 Gy of whole brain gamma radiation and, after 24-hours, 48-hours, and one-week periods, hippocampal brain slices were isolated and measured for anatomical and physiological differences. There were no major changes observed in tissue appearance or evoked synaptic responses at any post-irradiation time point. However, exposure to 60 Gy of irradiation resulted in a small, but statistically significant (14% change; ANOVA p < 0.005; n = 9) reduction in synaptic inhibition seen at 100 ms, indicating a selective depression of the gamma-aminobutyric acid (GABAA) slow form of inhibition. Population spike (PS) amplitudes also transiently declined by ~ 10% (p < 0.005; n = 9) when comparing the 24-hour group to sham group. Effects on PS amplitude recovered to baseline 48 hour and one week later. There were no obvious negative pathological effects; however, a subtle depression in circuit level inhibition was observed and provides evidence for 'radiomodulation' of brain circuits.

15.
J Cereb Blood Flow Metab ; 26(7): 937-50, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16292251

RESUMEN

Heat shock protein (Hsp)70 can suppress both necrosis and apoptosis induced by various injuries in vivo and in vitro. However, the relative importance of different functions and binding partners of Hsp70 in ischemic protection is unknown. To explore this question, we tested the ability of Hsp70-K71E, an adenosine triphosphate (ATP)ase-deficient point mutant, and Hsp70-381-640, a deletion mutant lacking the ATPase domain and encoding the carboxyl-terminal portion, to protect against ischemia-like injury in vivo and in vitro. Heat shock protein 70-wild type (-WT), -K71E, -381-640, and control vector plasmid LXSN were expressed in primary murine astrocyte cultures. Astrocytes overexpressing Hsp70-WT, -K71E, or -381-640 were all significantly protected from 4 h combined oxygen-glucose deprivation and 24 h reperfusion when assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay or propidium iodide staining and cell counting (P < 0.05). Brains of rats were transfected with plasmids encoding Hsp70-WT, -K71E, -381-640, or LXSN 24 h before 2 h middle cerebral artery occlusion followed by 24 h reperfusion. Animals that overexpressed either of the mutant proteins or Hsp70-WT had significantly better neurological scores and smaller infarcts than control animals. Protection by both mutants was associated with reduced protein aggregation, as assessed by ubiquitin immunohistochemistry and reduced nuclear translocation of apoptosis-inducing factor. The results show that the carboxyl-terminal portion of Hsp70 is sufficient for neuroprotection. This indicates that neither the ability to fold denatured proteins nor interactions with cochaperones or other proteins that bind the amino-terminal half of Hsp70 are essential to ischemic protection.


Asunto(s)
Isquemia Encefálica/prevención & control , Terapia Genética/métodos , Proteínas HSP70 de Choque Térmico/uso terapéutico , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/fisiopatología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Regulación de la Expresión Génica , Glucosa/farmacología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Ratones , Neuronas/metabolismo , Oxígeno/farmacología , Mutación Puntual , Estructura Terciaria de Proteína/genética , Ubiquitina/antagonistas & inhibidores , Ubiquitina/metabolismo
16.
Neurobiol Aging ; 27(4): 562-9, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16257478

RESUMEN

Protein aggregation and misfolding are central mechanisms of both acute and chronic neurodegeneration. Overexpression of chaperone Hsp70 protects from stroke in animal and cell culture models. Although it is accepted that chaperones protect cells, the mechanism of protection by chaperones in ischemic injury is poorly understood. In particular, the relative importance of preventing protein aggregation compared to facilitating correct protein folding during ischemia and recovery is not known. To test the importance of protein folding and minimize interaction with co-chaperones we studied the bacterial chaperonin GroEL (HSPD1) and a folding-deficient mutant D87K. Both molecules protected cells from ischemia-like injury, and reduced infarct volume and improved neurological outcome after middle cerebral artery occlusion in rats. Protection was associated with reduced protein aggregation, assessed by ubiquitin immunohistochemistry. Marked neuroprotection by the folding-deficient chaperonin demonstrates that inhibition of aggregation is sufficient to protect the brain from ischemia. This suggests that strategies to maintain protein solubility and inhibit aggregation in the face of acute insults such as stroke may be a useful protective strategy.


Asunto(s)
Ácido Aspártico/genética , Chaperonina 60/genética , Isquemia/prevención & control , Lisina/genética , Mutación/fisiología , Análisis de Varianza , Animales , Animales Recién Nacidos , Astrocitos/metabolismo , Western Blotting/métodos , Encéfalo/citología , Encéfalo/patología , Agregación Celular/genética , Agregación Celular/fisiología , Recuento de Células/métodos , Muerte Celular/fisiología , Células Cultivadas , Chaperonina 60/administración & dosificación , Cricetinae , Cricetulus , Técnica del Anticuerpo Fluorescente/métodos , Proteína Ácida Fibrilar de la Glía/metabolismo , Glucosa/deficiencia , Calor/efectos adversos , Hipoxia/complicaciones , Hipoxia/etiología , Hipoxia/terapia , Técnicas In Vitro , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/terapia , Isquemia/etiología , Isquemia/patología , Ratones , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/metabolismo , Sales de Tetrazolio , Factores de Tiempo , Transfección/métodos
17.
FEBS Lett ; 580(20): 4865-71, 2006 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-16916513

RESUMEN

The Na(+)-driven Cl(-)/HCO(3)(-) exchanger (NCBE) plays an important role in the regulation of intracellular pH (pH(i)). We previously identified two variants of NCBE from rat brain of which the variant with a carboxyterminal PSD-95/Dlg/ZO-1 (PDZ) motif (rb2NCBE) colocalized with the actin cytoskeleton. Increased rb2NCBE activity by PKA inhibition and reduction by forskolin and cAMP agonist suggest PKA regulation of NCBE. Disruption of actin filaments also decreased rb2NCBE activity. EBP50 and FLAG-rb2NCBE were reciprocally co-immunoprecipitated from rb2NCBE transfected cells. It is concluded that NCBE activity is inhibited by PKA and depends on the integrity of the actin cytoskeleton within a multiprotein complex at the plasma membrane.


Asunto(s)
Encéfalo/metabolismo , Antiportadores de Cloruro-Bicarbonato/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Transducción de Señal/fisiología , Sodio/metabolismo , Actinas/metabolismo , Adenilil Ciclasas/metabolismo , Animales , Astrocitos/citología , Astrocitos/metabolismo , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Antiportadores de Cloruro-Bicarbonato/genética , Proteínas Quinasas Dependientes de AMP Cíclico/antagonistas & inhibidores , Citocalasina B/metabolismo , Proteínas del Citoesqueleto/metabolismo , Citoesqueleto/metabolismo , Activación Enzimática , Concentración de Iones de Hidrógeno , Ratones , Complejos Multiproteicos , Células 3T3 NIH , Fosfoproteínas/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Ratas , Intercambiadores de Sodio-Hidrógeno
18.
Cell Stress Chaperones ; 11(2): 180-6, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-16817324

RESUMEN

Wild-type inducible Hsp70 (WT) and 2 folding deficient mutants protect the brain against focal cerebral ischemia in vivo and brain cells from oxygen-glucose deprivation (OGD) in vitro, but the protective mechanisms remain unclear. Mitochondria are central to both normal physiological function and the regulation of cell death. We tested the effect of overexpressing Hsp70 and 2 mutants, Hsp70-K71 E, an adenosine triphosphatase (ATPase)-deficient point mutant, and Hsp70-381-640, a deletion mutant lacking the ATPase domain on mitochondrial physiology under glucose deprivation (GD) stress in primary cultured astrocytes. Mitochondrial membrane potential was assessed using a potentiometric fluorescent dye tetramethylrhodamine ethyl ester (TMRE). By 5 hours of GD, the mitochondria in the LXSN control transfected astrocytes had markedly reduced membrane potential. However, in the Hsp70-WT, -K71E, and -381-640 groups, there was no apparent change in TMRE signal during 5 hours of GD. Oxygen consumption was measured to assess oxidative respiration. Overexpression of Hsp70-K71 E and -381-640 prevented the decrease in state III respiration observed at 5 hours, and all 3 prevented the increase in state IV respiration found in LXSN controls after 5 hours of GD. Reactive oxygen species (ROS) production was assessed with hydroethidine. Hsp70 and its mutants all significantly reduced the increases in ROS accumulation during 5 hours of GD. The results demonstrate that the protective effect of the carboxyl-terminal half of Hsp70 and of the full-length molecule is associated with better maintained mitochondrial membrane potential, better maintained state IV respiration, and reduced ROS generation during GD.


Asunto(s)
Astrocitos/metabolismo , Glucosa/metabolismo , Proteínas HSP70 de Choque Térmico/biosíntesis , Mitocondrias/fisiología , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/efectos de los fármacos , Respiración de la Célula/efectos de los fármacos , Respiración de la Célula/fisiología , Células Cultivadas , Glucosa/deficiencia , Glucosa/farmacología , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/fisiología , Humanos , Ratones , Mitocondrias/efectos de los fármacos , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/biosíntesis , Proteínas Mutantes/genética , Proteínas Mutantes/fisiología , Especies Reactivas de Oxígeno/metabolismo
20.
Neurosci Lett ; 380(3): 229-33, 2005 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-15862891

RESUMEN

Our prior work demonstrated that geldanamycin (GA) reduced injury due to oxygen-glucose deprivation (OGD) in primary astrocyte cultures. Using medium with an ionic composition similar to that observed during in vivo global ischemia, the selectivity and temporal profile of CA1 neuronal damage seen in vivo was mimicked with OGD in mouse hippocampal organotypic slice cultures. The present study tested the ability of GA to reduce delayed neuronal death in such cultures. Treating organotypic cultures with 100 nM GA for 24 h prior to OGD induced Hsp70 and significantly reduced CA1 neuronal damage. Staining with ubiquitin to identify protein aggregates revealed reduced redistribution of ubiquitin, consistent with reduced protein aggregation likely due at least in part to induction of Hsp70 by GA.


Asunto(s)
Hipocampo/efectos de los fármacos , Hipoxia-Isquemia Encefálica/metabolismo , Degeneración Nerviosa/prevención & control , Fármacos Neuroprotectores/farmacología , Quinonas/farmacología , Animales , Benzoquinonas , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Inhibidores Enzimáticos/farmacología , Glucosa/deficiencia , Proteínas HSP70 de Choque Térmico/agonistas , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipoxia-Isquemia Encefálica/patología , Lactamas Macrocíclicas , Ratones , Modelos Biológicos , Degeneración Nerviosa/metabolismo , Degeneración Nerviosa/patología , Técnicas de Cultivo de Órganos , Complejo de la Endopetidasa Proteasomal/efectos de los fármacos , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Factores de Tiempo , Resultado del Tratamiento , Ubiquitina/agonistas , Ubiquitina/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA