Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 49
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
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
2.
Neuromolecular Med ; 21(2): 170-181, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30900118

RESUMEN

Mild traumatic brain injury (mTBI) can result in permanent impairment in memory and learning and may be a precursor to other neurological sequelae. Clinical treatments to ameliorate the effects of mTBI are lacking. Inhibition of microRNA-181a (miR-181a) is protective in several models of cerebral injury, but its role in mTBI has not been investigated. In the present study, miR-181a-5p antagomir was injected intracerebroventricularly 24 h prior to closed-skull cortical impact in young adult male mice. Paw withdrawal, open field, zero maze, Y maze, object location and novel object recognition tests were performed to assess neurocognitive dysfunction. Brains were assessed immunohistologically for the neuronal marker NeuN, the perineuronal net marker wisteria floribunda lectin (WFA), cFos, and the interneuron marker parvalbumin. Protein quantification was performed with immunoblots for synaptophysin and postsynaptic density 95 (PSD95). Fluorescent in situ hybridization was utilized to localize hippocampal miR-181a expression. MiR-181a antagomir treatment reduced neuronal miR-181a expression after mTBI, restored deficits in novel object recognition and increased hippocampal parvalbumin expression in the dentate gyrus. These changes were associated with decreased dentate gyrus hyperactivity indicated by a relative reduction in PSD95 and cFos expression. These results suggest that miR-181a inhibition may be a therapeutic approach to reduce hippocampal excitotoxicity and prevent cognitive dysfunction following mTBI.


Asunto(s)
Antagomirs/uso terapéutico , Lesiones Traumáticas del Encéfalo/terapia , Conducta Exploratoria/efectos de los fármacos , Traumatismos Cerrados de la Cabeza/terapia , MicroARNs/antagonistas & inhibidores , Parvalbúminas/biosíntesis , Reconocimiento en Psicología/efectos de los fármacos , Animales , Antagomirs/administración & dosificación , Antagomirs/farmacología , Lesiones Traumáticas del Encéfalo/genética , Lesiones Traumáticas del Encéfalo/metabolismo , Corteza Cerebral/química , Corteza Cerebral/lesiones , Corteza Cerebral/patología , Simulación por Computador , Traumatismos Cerrados de la Cabeza/genética , Traumatismos Cerrados de la Cabeza/metabolismo , Hipocampo/química , Hipocampo/lesiones , Hipocampo/patología , Hiperalgesia/etiología , Hiperalgesia/genética , Hiperalgesia/prevención & control , Masculino , Aprendizaje por Laberinto , Trastornos de la Memoria/etiología , Trastornos de la Memoria/genética , Trastornos de la Memoria/prevención & control , Ratones , Ratones Endogámicos C57BL , MicroARNs/biosíntesis , MicroARNs/genética , Prueba de Campo Abierto , Parvalbúminas/genética , Premedicación , Distribución Aleatoria , Método Simple Ciego , Sinapsis/química
3.
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
4.
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.

5.
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
6.
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.

7.
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
8.
J Neurochem ; 140(5): 799-813, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-27987215

RESUMEN

Cortical spreading depression (CSD), based on its similarities with peri-infarct depolarization, is an ideal model for investigating transformation from the ischemic penumbra to infarct core. However, the underlying mechanisms remain unclear. To our knowledge, this is the first study to use a middle cerebral artery occlusion ischemic-reperfusion (I/R) injury model to determine whether AMP-activated protein kinase (AMPK)-dependent autophagy contributes to the neuroprotection of CSD preconditioning in rat cortex. In this study, we topically applied a pledget soaked in 1 mol/L KCl solution on rat cortex for 2 h to elicite CSD or 1 mol/L NaCl solution as a control. The results demonstrated that CSD preconditioning significantly decreased the infarct volume, neurological deficits and neuronal apoptosis in the cortical penumbra of middle cerebral artery occlusion rats, which was inhibited by the autophagy inhibitor 3-methyladenine (3-MA, 200 nmol). Furthermore, CSD increased the protein levels of the autophagy markers LC3-II, Beclin-1 and the p-AMPK (Thr172 )/AMPK ratio at 12 h and decreased P62 and p-P70S6K (Thr389 ). Moreover, the AMPK inhibitor Compound C (20 mg/kg) down-regulated the LC3-II, p-AMPK (Thr172 )/AMPK and ULK1 levels, up-regulated the P62 and p-P70S6K (Thr389 ) levels induced by CSD. The neuroprotection of CSD is likely a result of AMPK-mediated autophagy activity and autophagy-induced neuronal cells apoptosis inhibition. These novel findings support a central role for AMPK and autophagy in CSD-induced ischemic tolerance. AMPK-mediated autophagy may represent a new target for stroke.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Autofagia , Isquemia Encefálica/patología , Depresión de Propagación Cortical/fisiología , Precondicionamiento Isquémico , Neuroprotección , Daño por Reperfusión/prevención & control , Accidente Cerebrovascular/patología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Adenina/análogos & derivados , Adenina/farmacología , Animales , Conducta Animal , Isquemia Encefálica/metabolismo , Isquemia Encefálica/psicología , Infarto Cerebral/patología , Depresión de Propagación Cortical/efectos de los fármacos , Infarto de la Arteria Cerebral Media/patología , Masculino , Enfermedades del Sistema Nervioso/etiología , Enfermedades del Sistema Nervioso/patología , Enfermedades del Sistema Nervioso/psicología , Cloruro de Potasio/farmacología , Ratas , Ratas Wistar , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , Accidente Cerebrovascular/metabolismo
9.
Mitochondrion ; 30: 248-54, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27553862

RESUMEN

Neurons in the cornu ammonis 1 (CA1) region of the hippocampus are vulnerable to cerebral ischemia, while dentate gyrus (DG) neurons are more resistant. This effect is mediated by local astrocytes, and may reflect differences in subregional hippocampal expression of miR-29a. We investigated the role of miR-29a on survival of hippocampal astrocytes cultured selectively from CA1 and DG in response to glucose deprivation (GD). CA1 astrocytes exhibited more cell death and a greater decrease in miR-29a than DG astrocytes. A reciprocal change was observed in the mitochondrial voltage dependent cation channel-1 (VDAC1), a regulator of mitochondria and target of miR-29a. In CA1 astrocytes, increasing miR-29a decreased VDAC1 and improved cell survival, while knockdown of VDAC1 improved survival. Finally, the protective effect of miR-29a was eliminated by inhibition of miR-29a/VDAC1 binding. These findings suggest that the selective vulnerability of the CA1 to injury may be due in part to a limited miR-29a response in CA1 astrocytes, allowing a greater increase in VDAC1-mediated cellular dysfunction in CA1 astrocytes.


Asunto(s)
Astrocitos/fisiología , Giro Dentado/citología , Regulación de la Expresión Génica , Hipocampo/citología , MicroARNs/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , Animales , Supervivencia Celular , Células Cultivadas , Ratones
10.
J Cereb Blood Flow Metab ; 35(12): 1977-84, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26126866

RESUMEN

MicroRNA-29b (miR-29b) is involved in regulating ischemia process, but the molecular mechanism is unclear. In this work, we explored the function of miR-29b in cerebral ischemia. The level of miR-29b in white blood cells was evaluated in patients and mice after ischemic stroke. Brain infarct volume and National Institute of Health stroke scale (NIHSS) scores were analyzed to determine the relationship between miR-29b expression and the severity of stroke. The relationship of miR-29b and aquaporin-4 (AQP4) was further studied in mice. We found that miR-29b was significantly downregulated in stroke patients (P<0.05). MiR-29b level negatively associated with NIHSS scores (r=-0.349, P<0.01) and brain infarct volume (r=-0.321, P<0.05). In ischemic mice, miR-29b in the brain and blood were both downregulated (r=0.723, P<0.05). MiR-29b overexpression reduced infarct volume (49.50±6.55 versus 35.48±2.28 mm(3), P<0.05), edema (164±4% versus 108±4%, P<0.05), and blood-brain barrier (BBB) disruption compared with controls (15±9% versus 7±3%, P<0.05). Aquaporin-4 expression greatly decreased after miR-29b overexpression (28±7% versus 11±3%, P<0.05). Dual-luciferase reporter system showed that AQP-4 was the direct target of miR-29b (P<0.05). We concluded that miR-29b could potentially predict stroke outcomes as a novel circulating biomarker, and miR-29b overexpression reduced BBB disruption after ischemic stroke via downregulating AQP-4.


Asunto(s)
Acuaporina 4/genética , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/genética , MicroARNs/genética , Animales , Biomarcadores , Barrera Hematoencefálica/patología , Edema Encefálico/patología , Infarto Cerebral/genética , Infarto Cerebral/patología , Regulación hacia Abajo/efectos de los fármacos , Femenino , Vectores Genéticos , Humanos , Infarto de la Arteria Cerebral Media/patología , Lentivirus/genética , Lentivirus/metabolismo , Masculino , Ratones , MicroARNs/sangre , MicroARNs/efectos de los fármacos , Valor Predictivo de las Pruebas , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/genética
11.
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
12.
Chin Med J (Engl) ; 128(11): 1510-5, 2015 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-26021509

RESUMEN

BACKGROUND: Sevoflurane and propofol are widely used anesthetics for surgery. Studies on the mechanisms of general anesthesia have focused on changes in protein expression properties and membrane lipid. MicroRNAs (miRNAs) regulate neural function by altering protein expression. We hypothesize that sevoflurane and propofol affect miRNA expression profiles in the brain, expect to understand the mechanism of anesthetic agents. METHODS: Rats were randomly assigned to a 2% sevoflurane group, 600 µg·kg - 1·min - 1 propofol group, and a control group without anesthesia (n = 4, respectively). Treatment group was under anesthesia for 6 h, and all rats breathed spontaneously with continuous monitoring of respiration and blood gases. Changes in rat cortex miRNA expression profiles were analyzed by miRNA microarrays and validated by quantitative real-time polymerase chain reaction (qRT-PCR). Differential expression of miRNA using qRT-PCR among the control, sevoflurane, and propofol groups were compared using one-way analysis of variance (ANOVA). RESULTS: Of 677 preloaded rat miRNAs, the microarray detected the expression of 277 miRNAs in rat cortex (40.9%), of which 9 were regulated by propofol and (or) sevoflurane. Expression levels of three miRNAs (rno-miR-339-3p, rno-miR-448, rno-miR-466b-1FNx01) were significantly increased following sevoflurane and six (rno-miR-339-3p, rno-miR-347, rno-miR-378FNx01, rno-miR-412FNx01, rno-miR-702-3p, and rno-miR-7a-2FNx01) following propofol. Three miRNAs (rno-miR-466b-1FNx01, rno-miR-3584-5p and rno-miR-702-3p) were differentially expressed by the two anesthetic treatment groups. CONCLUSIONS: Sevoflurane and propofol anesthesia induced distinct changes in brain miRNA expression patterns, suggesting differential regulation of protein expression. Determining the targets of these differentially expressed miRNAs may help reveal both the common and agent-specific actions of anesthetics on neurological and physiological function.


Asunto(s)
Éteres Metílicos/farmacología , MicroARNs/genética , Propofol/farmacología , Anestesia General , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Reacción en Cadena en Tiempo Real de la Polimerasa , Sevoflurano
13.
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
14.
Exp Neurol ; 264: 1-7, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25433215

RESUMEN

miR-181 has deleterious effects on stroke outcome, and reducing miR-181a levels prior to middle cerebral artery occlusion (MCAO) was shown previously to be protective. Here we tested the effect of post-ischemic treatment with miR-181a antagomir by intracerebroventricular and intravenous routes of administration on infarct size, neurological outcome, inflammatory response and long term behavioral outcome. Post-treatment with miR-181a antagomir significantly reduced infarction size, improved neurological deficits and reduced NF-κB activation, numbers of infiltrating leukocytes and levels of Iba1. Targets affected by miR-181a antagomir administered after stroke onset include BCL2 and X-linked inhibitor of apoptosis protein (XIAP). Post-treatment with miR-181a antagomir significantly improved behavioral outcome assessed by rotarod at one month. These findings indicate that post-treatment with miR-181a antagomir has neuroprotective effects against ischemic neuronal damage and neurological impairment in mice, and the protection is long lasting including recovery of motor function and coordination over one month. The ability to protect the brain with post-treatment with miR-181a antagomir with long lasting effect makes this a promising therapeutic target and may be an innovative and effective new approach for stroke therapy.


Asunto(s)
Infarto Encefálico/tratamiento farmacológico , Isquemia Encefálica/tratamiento farmacológico , Regulación de la Expresión Génica/efectos de los fármacos , MicroARNs/antagonistas & inhibidores , Oligonucleótidos/administración & dosificación , Recuperación de la Función/efectos de los fármacos , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Infarto Encefálico/etiología , Isquemia Encefálica/complicaciones , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patología , Proteínas de Unión al Calcio/metabolismo , Modelos Animales de Enfermedad , Inyecciones Intravenosas , Inyecciones Intraventriculares , Masculino , Ratones , Ratones Endogámicos C57BL , MicroARNs/química , MicroARNs/metabolismo , Proteínas de Microfilamentos/metabolismo , Actividad Motora/efectos de los fármacos , Enfermedades del Sistema Nervioso/tratamiento farmacológico , Enfermedades del Sistema Nervioso/etiología , Peroxidasa/metabolismo , Desempeño Psicomotor/efectos de los fármacos , Factores de Tiempo
15.
Antioxid Redox Signal ; 22(2): 187-202, 2015 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24359188

RESUMEN

SIGNIFICANCE: Cerebral ischemia is a major cause of death and disability throughout the world, yet therapeutic options remain limited. The interplay between the cellular redox state and the immune response plays a critical role in determining the extent of neural cell injury after ischemia and reperfusion. Excessive amounts of reactive oxygen species (ROS) generated by mitochondria and other sources act both as triggers and effectors of inflammation. This review will focus on the interplay between these two mechanisms. RECENT ADVANCES: MicroRNAs (miRNAs) are important post-transcriptional regulators that interact with multiple target messenger RNAs coordinately regulating target genes, including those involved in controlling mitochondrial function, redox state, and inflammatory pathways. This review will focus on the regulation of mitochondria, ROS, and inflammation by miRNAs in the chain of deleterious intra- and intercellular events that lead to brain cell death after cerebral ischemia. CRITICAL ISSUES: Although pretreatment using miRNAs was effective in cerebral ischemia in rodents, testing treatment after the onset of ischemia is an essential next step in the development of acute stroke treatment. In addition, miRNA formulation and delivery into the CNS remain a challenge in the clinical translation of miRNA therapy. FUTURE DIRECTIONS: Future research should focus on post-treatment and potential clinical use of miRNAs.


Asunto(s)
MicroARNs/genética , Especies Reactivas de Oxígeno/metabolismo , Accidente Cerebrovascular/genética , Animales , Humanos , Mitocondrias/metabolismo , Oxidación-Reducción
16.
Adv Neurobiol ; 11: 171-88, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25236729

RESUMEN

Astrocytes have been shown to protect neurons from delayed neuronal death and increase their survival in cerebral ischemia. One of the main mechanisms of astrocyte protection is rapid removal of excess glutamate from synaptic sites by astrocytic plasma membrane glutamate transporters such as GLT-1/EAAT-2, reducing excitotoxicity. Astrocytic mitochondrial function is essential for normal GLT-1 function. Manipulating astrocytic mitochondrial and GLT-1 function is thus an important strategy to enhance neuronal survival and improve outcome following cerebral ischemia. Increasing evidence supports the involvement of microRNAs (miRNA), some of them being astrocyte-enriched, in the regulation of cerebral ischemia. This chapter will first update the information about astrocytes, GLT-1, astrocytic mitochondria, and delayed neuronal death. Then we will focus on two recently reported astrocyte-enriched miRNAs (miR-181 and miR-29 families), their effects on astrocytic mitochondria and GLT-1 as well as on outcome after cerebral ischemia.

17.
Neurosci Lett ; 565: 53-8, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24269978

RESUMEN

Astrocytes have been shown to protect neurons and increase their survival in many pathological settings. Manipulating astrocyte functions is thus an important strategy to enhance neuronal survival and improve outcome following cerebral ischemia. Increasing evidence supports the involvement of microRNAs (miRNA), some of them being astrocyte-enriched, in the regulation of cerebral ischemia. This mini review will focus on several recently reported astrocyte-enriched miRNAs (miR-181 and miR-29 families and miR-146a), their validated targets, regional expression and effects on outcome after cerebral ischemia.


Asunto(s)
Astrocitos/metabolismo , Isquemia Encefálica/metabolismo , MicroARNs/metabolismo , Animales , Astrocitos/patología , Isquemia Encefálica/patología , Isquemia Encefálica/terapia , Supervivencia Celular , Terapia Genética , Humanos , MicroARNs/genética , Neuronas/patología
18.
Neurochem Int ; 77: 2-8, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24373752

RESUMEN

The BCL-2 family is centrally involved in the mechanism of cell death after cerebral ischemia. It is well known that the proteins of the BCL-2 family are key regulators of apoptosis through controlling mitochondrial outer membrane permeabilization. Recent findings suggest that many BCL-2 family members are also directly involved in controlling transmission of Ca(2+) from the endoplasmic reticulum (ER) to mitochondria through a specialization called the mitochondria-associated ER membrane (MAM). Increasing evidence supports the involvement of microRNAs (miRNAs), some of them targeting BCL-2 family proteins, in the regulation of cerebral ischemia. In this mini-review, after highlighting current knowledge about the multiple functions of BCL-2 family proteins and summarizing their relationship to outcome from cerebral ischemia, we focus on the regulation of BCL-2 family proteins by miRNAs, especially miR-29 which targets multiple BCL-2 family proteins.


Asunto(s)
Isquemia Encefálica/genética , Genes bcl-2/genética , MicroARNs/genética , Proteínas Proto-Oncogénicas c-bcl-2/genética , Secuencia de Aminoácidos , Animales , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Datos de Secuencia Molecular
19.
Transl Stroke Res ; 4(6): 693-703, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24323423

RESUMEN

The highly evolutionarily conserved 70 kDa heat shock protein (HSP70) family was first understood for its role in protein folding and response to stress. Subsequently, additional functions have been identified for it in regulation of organelle interaction, of the inflammatory response, and of cell death and survival. Overexpression of HSP70 family members is associated with increased resistance to and improved recovery from cerebral ischemia. MicroRNAs (miRNAs) are important posttranscriptional regulators that interact with multiple target messenger RNAs (mRNA) coordinately regulating target genes, including chaperones. The members of the HSP70 family are now appreciated to work together as networks to facilitate organelle communication and regulate inflammatory signaling and cell survival after cerebral ischemia. This review will focus on the new concept of the role of the chaperone network in the organelle network and its novel regulation by miRNA.


Asunto(s)
Isquemia Encefálica/metabolismo , MicroARNs/metabolismo , Chaperonas Moleculares/metabolismo , Orgánulos/metabolismo , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , MicroARNs/genética , Mitocondrias/metabolismo , Chaperonas Moleculares/genética , Orgánulos/genética , Pliegue de Proteína , ARN Mensajero
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...