Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 92
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Mitochondrion ; 59: 105-112, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33933660

RESUMO

Brain-enriched microRNA-338 (miR-338) is known to play a central role in brain mitochondrial function, however the role of miR-338 in stroke injury remains unknown. This study investigated the role of miR-338 in injury from transient focal cerebral ischemia in mice, and in cell survival and mitochondrial function after in vitro ischemia in astrocyte and neuronal cultures. Pre-treatment of mice with intracerebroventricular injection of miR-338 antagomir 24 h prior to 1 h of middle cerebral artery occlusion (MCAO) significantly reduced infarct size and improved neurological score at both 24 h and 7d after injury. Levels of the miR-338 target cytochrome-c oxidase subunit 4I1 (COX4I1), which plays an essential role in maintaining brain mitochondrial ATP production, were increased in miR-338 antagomir-treated mice. Mouse primary astrocyte cell cultures subjected to glucose deprivation exhibited increased cell survival when pre-treated with miR-338 inhibitor, and greater cell death with miR-338 mimic. Decreased miR-338 levels were associated with increased ATP production, augmented cytochrome c oxidative (CcO) activity and preservation of COX4I1. In vitro protection with miR-338 inhibitor was blocked by concurrent knockdown of COX4I1 with small interfering RNA. Parallel studies in mouse neuronal N2a cultures resulted in preserved ATP content and CcO activity with miR-338 inhibition, indicating a shared miR-338-dependent response to ischemic stress between brain cell types. These results suggest that miR-338 inhibition and/or COX4I1-targeted therapies may be novel clinical strategies to protect against stroke injury via preservation of mitochondrial function in multiple cell types.


Assuntos
Astrócitos/citologia , Isquemia Encefálica/genética , Complexo IV da Cadeia de Transporte de Elétrons/genética , MicroRNAs/genética , Mitocôndrias/metabolismo , Neurônios/citologia , Animais , Astrócitos/química , Isquemia Encefálica/etiologia , Isquemia Encefálica/metabolismo , Sobrevivência Celular , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/química , Cultura Primária de Células
2.
Front Cell Neurosci ; 13: 394, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31551712

RESUMO

In the present study, we assessed efficacy of exosomes harvested from human and mouse stem cell cultures in protection of mouse primary astrocyte and neuronal cell cultures following in vitro ischemia, and against ischemic stroke in vivo. Cell media was collected from primary mouse neural stem cell (NSC) cultures or from human induced pluripotent stem cell-derived cardiomyocyte (iCM) cultures. Exosomes were extracted and purified by polyethylene glycol complexing and centrifugation, and exosome size and concentration were determined with a NanoSiteTM particle analyzer. Exosomes were applied to primary mouse cortical astrocyte or neuronal cultures prior to, and/or during, combined oxygen-glucose deprivation (OGD) injury. Cell death was assessed via lactate dehydrogenase (LHD) and propidium iodide staining 24 h after injury. NSC-derived exosomes afforded marked protection to astrocytes following OGD. A more modest (but significant) level of protection was observed with human iCM-derived exosomes applied to astrocytes, and with NSC-derived exosomes applied to primary neuronal cultures. In subsequent experiments, NSC-derived exosomes were injected intravenously into adult male mice 2 h after transient (1 h) middle cerebral artery occlusion (MCAO). Gross motor function was assessed 1 day after reperfusion and infarct volume was assessed 4 days after reperfusion. Mice treated post-stroke with intravenous NSC-derived exosomes exhibited significantly reduced infarct volumes. Together, these results suggest that exosomes isolated from mouse NSCs provide neuroprotection against experimental stroke possibly via preservation of astrocyte function. Intravenous NSC-derived exosome treatment may therefore provide a novel clinical adjuvant for stroke in the immediate post-injury period.

3.
eNeuro ; 6(4)2019.
Artigo em Inglês | MEDLINE | ID: mdl-31427401

RESUMO

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.


Assuntos
Antagomirs/administração & dosagem , Isquemia Encefálica/metabolismo , Região CA1 Hipocampal/metabolismo , MicroRNAs/antagonistas & inibidores , Neurônios/metabolismo , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Região CA1 Hipocampal/efeitos dos fármacos , Proteína Duplacortina , Masculino , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Prosencéfalo/efeitos dos fármacos , Prosencéfalo/fisiopatologia , Ratos Sprague-Dawley
4.
Front Neurosci ; 13: 609, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31354401

RESUMO

Recent studies demonstrate significant neuroimmune changes in postpartum females, a period that also carries an increased risk of stroke. Oxytocin, a major hormone upregulated in the brains of nursing mothers, has been shown to both modulate neuroinflammation and protect against stroke. In the present study we assessed whether and how nursing modulates the neuroimmune response and injury after stroke. We observed that postpartum nursing mice were markedly protected from 1 h of transient middle cerebral artery occlusion (MCAO) relative to either non-pregnant/non-postpartum or non-nursing (pups removed) postpartum females. Nursing mice also expressed reduced levels of pro-inflammatory cytokines, had decreased migration of blood leukocytes into the brain following MCAO, and displayed peripheral neuroimmune changes characterized by increased spleen weight and increased fraction of spleen monocytes. Intranasal oxytocin treatment in non-pregnant females in part recapitulated the protective and anti-inflammatory effects associated with nursing. In summary, the results of the present study demonstrate that nursing in the postpartum period provides relative protection against transient ischemic stroke associated with decreased brain leukocytes and increased splenic monocytes. These effects appear to be regulated, at least in part, by oxytocin.

5.
Neuromolecular Med ; 21(2): 170-181, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30900118

RESUMO

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.


Assuntos
Antagomirs/uso terapêutico , Lesões Encefálicas Traumáticas/terapia , Comportamento Exploratório/efeitos dos fármacos , Traumatismos Cranianos Fechados/terapia , MicroRNAs/antagonistas & inibidores , Parvalbuminas/biossíntese , Reconhecimento Psicológico/efeitos dos fármacos , Animais , Antagomirs/administração & dosagem , Antagomirs/farmacologia , Lesões Encefálicas Traumáticas/genética , Lesões Encefálicas Traumáticas/metabolismo , Córtex Cerebral/química , Córtex Cerebral/lesões , Córtex Cerebral/patologia , Simulação por Computador , Traumatismos Cranianos Fechados/genética , Traumatismos Cranianos Fechados/metabolismo , Hipocampo/química , Hipocampo/lesões , Hipocampo/patologia , Hiperalgesia/etiologia , Hiperalgesia/genética , Hiperalgesia/prevenção & controle , Masculino , Aprendizagem em Labirinto , Transtornos da Memória/etiologia , Transtornos da Memória/genética , Transtornos da Memória/prevenção & controle , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/biossíntese , MicroRNAs/genética , Teste de Campo Aberto , Parvalbuminas/genética , Pré-Medicação , Distribuição Aleatória , Método Simples-Cego , Sinapses/química
6.
Stroke ; 49(6): 1488-1495, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29748423

RESUMO

BACKGROUND AND PURPOSE: Inflammatory cells play a significant role in secondary injury after ischemic stroke. Recent studies have suggested that a lack of autophagy in myeloid cells causes augmented proinflammatory cytokine release and prolonged inflammation after tissue injury. In this study, we investigated the roles of myeloid cell autophagy in ischemic brain injury. METHODS: Focal cerebral ischemia was induced via transient middle cerebral artery occlusion in mice with autophagy-deficient myeloid lineage cells (Atg5flox/flox LysMCre+) and in their littermate controls (Atg5flox/flox). Infarct volume, neurological function, inflammatory cell infiltration, and proinflammatory cytokine expression levels were evaluated. RESULTS: Mice lacking autophagy in myeloid lineage cells had a lower survival rate for 14 days than control mice (20% versus 70%; P<0.05). Although there was no difference in infarct volume at 12 hours between the 2 groups, mice lacking autophagy in myeloid lineage cells had larger infarct volumes at later time points (3 and 7 days after reperfusion) with worse neurological deficit scores and lower grip test scores. There were a higher number of ionized calcium binding adaptor molecule 1-positive cells and cells expressing M1 marker CD16/32 in mice lacking autophagy in myeloid cells at the later time points. Moreover, these mice had higher expression levels of proinflammatory cytokines at later time points; however, there was no difference in ionized calcium binding adaptor molecule 1-positive cells or mRNA levels of proinflammatory cytokines at the earlier time point (12 hours after reperfusion). CONCLUSIONS: These data suggest that the lack of myeloid cell autophagy aggravates secondary injury by augmenting and prolonging inflammation after ischemic stroke without affecting the initial injury.


Assuntos
Autofagia/fisiologia , Lesões Encefálicas/metabolismo , Linhagem da Célula/fisiologia , Células Mieloides/metabolismo , Acidente Vascular Cerebral/metabolismo , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Citocinas/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/metabolismo , Inflamação/metabolismo , Camundongos Transgênicos
7.
Transl Stroke Res ; 8(6): 585-596, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28646450

RESUMO

Preconditioning is a paradigm in which sublethal stress-prior to a more injurious insult-induces protection against injury. In the central nervous system (CNS), preconditioning against ischemic stroke is induced by short durations of ischemia, brief seizures, exposure to anesthetics, and other stresses. Increasing evidence supports the contribution of microRNAs (miRNAs) to the pathogenesis of cerebral ischemia and ischemic tolerance induced by preconditioning. Studies investigating miRNA changes induced by preconditioning have to date identified 562 miRNAs that change expression levels after preconditioning, and 15% of these changes were reproduced in at least one additional study. Of miRNAs assessed as changed by preconditioning in more than one study, about 40% changed in the same direction in more than one study. Most of the studies to assess the role of specific miRNAs in the neuroprotective mechanism of preconditioning were performed in vitro, with fewer studies manipulating individual miRNAs in vivo. Thus, while many miRNAs change in response to preconditioning stimuli, the mechanisms underlying their effects are not well understood. The data does suggest that miRNAs may play significant roles in preconditioning-induced neuroprotection. This review focuses on the current state of knowledge of the possible role of miRNAs in preconditioning-induced cerebral protection.


Assuntos
Isquemia Encefálica , Precondicionamento Isquêmico , MicroRNAs , Neuroproteção/genética , Animais , Humanos , Acidente Vascular Cerebral
8.
Mol Cell Neurosci ; 82: 118-125, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28522364

RESUMO

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.


Assuntos
Astrócitos/metabolismo , Isquemia Encefálica/genética , Receptor alfa de Estrogênio/genética , Ataque Isquêmico Transitório/metabolismo , MicroRNAs/genética , Animais , Astrócitos/efeitos dos fármacos , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Feminino , Ataque Isquêmico Transitório/genética , Masculino , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/genética , Traumatismo por Reperfusão/metabolismo , Fatores Sexuais
9.
Cureus ; 9(3): e1076, 2017 Mar 04.
Artigo em Inglês | MEDLINE | ID: mdl-28401026

RESUMO

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.

10.
Nat Med ; 23(4): 450-460, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28288111

RESUMO

Although blood-brain barrier (BBB) compromise is central to the etiology of diverse central nervous system (CNS) disorders, endothelial receptor proteins that control BBB function are poorly defined. The endothelial G-protein-coupled receptor (GPCR) Gpr124 has been reported to be required for normal forebrain angiogenesis and BBB function in mouse embryos, but the role of this receptor in adult animals is unknown. Here Gpr124 conditional knockout (CKO) in the endothelia of adult mice did not affect homeostatic BBB integrity, but resulted in BBB disruption and microvascular hemorrhage in mouse models of both ischemic stroke and glioblastoma, accompanied by reduced cerebrovascular canonical Wnt-ß-catenin signaling. Constitutive activation of Wnt-ß-catenin signaling fully corrected the BBB disruption and hemorrhage defects of Gpr124-CKO mice, with rescue of the endothelial gene tight junction, pericyte coverage and extracellular-matrix deficits. We thus identify Gpr124 as an endothelial GPCR specifically required for endothelial Wnt signaling and BBB integrity under pathological conditions in adult mice. This finding implicates Gpr124 as a potential therapeutic target for human CNS disorders characterized by BBB disruption.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Glioblastoma/genética , Infarto da Artéria Cerebral Média/genética , Hemorragias Intracranianas/genética , Receptores Acoplados a Proteínas G/genética , Junções Íntimas/metabolismo , Animais , Barreira Hematoencefálica/ultraestrutura , Modelos Animais de Doenças , Células Endoteliais/ultraestrutura , Matriz Extracelular/metabolismo , Citometria de Fluxo , Imunofluorescência , Glioblastoma/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Hemorragias Intracranianas/metabolismo , Camundongos , Camundongos Knockout , Microscopia Eletrônica , Microvasos , Pericitos/ultraestrutura , Reação em Cadeia da Polimerase em Tempo Real , Junções Íntimas/ultraestrutura , Via de Sinalização Wnt
11.
J Neurosci ; 37(11): 3072-3084, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28188219

RESUMO

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.


Assuntos
Proliferação de Células , Sobrevivência Celular/imunologia , Encefalite/imunologia , MicroRNAs/imunologia , Mitocôndrias/imunologia , Neurogênese/imunologia , Neurônios/imunologia , Animais , Citocinas/imunologia , Encefalite/patologia , Feminino , Inflamação/imunologia , Inflamação/patologia , Masculino , Camundongos , Mitocôndrias/patologia , Neurônios/patologia
12.
Mitochondrion ; 30: 248-54, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27553862

RESUMO

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.


Assuntos
Astrócitos/fisiologia , Giro Denteado/citologia , Regulação da Expressão Gênica , Hipocampo/citologia , MicroRNAs/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Animais , Sobrevivência Celular , Células Cultivadas , Camundongos
13.
Anesthesiology ; 123(4): 810-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26270940

RESUMO

BACKGROUND: Isoflurane induces cell death in neurons undergoing synaptogenesis via increased production of pro-brain-derived neurotrophic factor (proBDNF) and activation of postsynaptic p75 neurotrophin receptor (p75). Astrocytes express p75, but their role in neuronal p75-mediated cell death remains unclear. The authors investigated whether astrocytes have the capacity to buffer increases in proBDNF and protect against isoflurane/p75 neurotoxicity. METHODS: Cell death was assessed in day in vitro (DIV) 7 mouse primary neuronal cultures alone or in co-culture with age-matched or DIV 21 astrocytes with propidium iodide 24 h after 1 h exposure to 2% isoflurane or recombinant proBDNF. Astrocyte-targeted knockdown of p75 in co-culture was achieved with small-interfering RNA and astrocyte-specific transfection reagent and verified with immunofluorescence microscopy. proBDNF levels were assessed by enzyme-linked immunosorbent assay. Each experiment used six to eight replicate cultures/condition and was repeated at least three times. RESULTS: Exposure to isoflurane significantly (P < 0.05) increased neuronal cell death in primary neuronal cultures (1.5 ± 0.7 fold, mean ± SD) but not in co-culture with DIV 7 (1.0 ± 0.5 fold) or DIV 21 astrocytes (1.2 ± 1.2 fold). Exogenous proBDNF dose dependently induced neuronal cell death in both primary neuronal and co-cultures, an effect enhanced by astrocyte p75 inhibition. Astrocyte-targeted p75 knockdown in co-cultures increased media proBDNF (1.2 ± 0.1 fold) and augmented isoflurane-induced neuronal cell death (3.8 ± 3.1 fold). CONCLUSIONS: The presence of astrocytes provides protection to growing neurons by buffering increased levels of proBDNF induced by isoflurane. These findings may hold clinical significance for the neonatal and injured brain where increased levels of proBDNF impair neurogenesis.


Assuntos
Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Fator Neurotrófico Derivado do Encéfalo/biossíntese , Isoflurano/toxicidade , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Precursores de Proteínas/biossíntese , Animais , Astrócitos/patologia , Fator Neurotrófico Derivado do Encéfalo/antagonistas & inibidores , Células Cultivadas , Técnicas de Cocultura , Camundongos , Neurônios/patologia , Precursores de Proteínas/antagonistas & inibidores
14.
J Neurosci Res ; 93(11): 1703-12, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26147710

RESUMO

Recent studies have demonstrated that neural stem cell (NSC) culture at physiologically normoxic conditions (2-5% O2) is advantageous in terms of neuronal differentiation and survival. Neuronal differentiation is accompanied by a remarkable shift to mitochondrial oxidative metabolism compared with preferentially glycolytic metabolism of proliferating cells. However, metabolic changes induced by growth in a normoxic (5%) O2 culture environment in NSCs have been minimally explored. This study demonstrates that culturing under 5% O2 conditions results in higher levels of mitochondrial oxidative metabolism, decreased glycolysis, and reduced levels of reactive oxygen species in NSC cultures. Inflammation is one of the major environmental factors limiting postinjury NSC neuronal differentiation and survival. Our results show that NSCs differentiated under 5% O2 conditions possess better resistance to in vitro inflammatory injury compared with those exposed to 20% O2. The present work demonstrates that lower, more physiologically normal O2 levels support metabolic changes induced during NSC neuronal differentiation and provide increased resistance to inflammatory injury, thus highlighting O2 tension as an important determinant of cell fate and survival in various stem cell therapies.


Assuntos
Técnicas de Cultura de Células/métodos , Diferenciação Celular/fisiologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Oxigênio/farmacologia , Animais , Apoptose/fisiologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Inflamação/metabolismo , Camundongos
15.
Stroke ; 46(8): 2271-6, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26130091

RESUMO

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.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/prevenção & controle , Interleucina-4/deficiência , Caracteres Sexuais , Animais , Isquemia Encefálica/patologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Distribuição Aleatória
16.
Stroke ; 46(2): 551-6, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25604249

RESUMO

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.


Assuntos
Isquemia Encefálica/metabolismo , Moléculas de Adesão Celular Neuronais/biossíntese , Proteínas da Matriz Extracelular/biossíntese , MicroRNAs/administração & dosagem , MicroRNAs/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Serina Endopeptidases/biossíntese , Animais , Isquemia Encefálica/patologia , Moléculas de Adesão Celular Neuronais/antagonistas & inibidores , Células Cultivadas , Proteínas da Matriz Extracelular/antagonistas & inibidores , Marcação de Genes , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteína Reelina
17.
Exp Neurol ; 264: 1-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25433215

RESUMO

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.


Assuntos
Infarto Encefálico/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Regulação da Expressão Gênica/efeitos dos fármacos , MicroRNAs/antagonistas & inibidores , Oligonucleotídeos/administração & dosagem , Recuperação de Função Fisiológica/efeitos dos fármacos , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Infarto Encefálico/etiologia , Isquemia Encefálica/complicações , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Injeções Intravenosas , Injeções Intraventriculares , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/química , MicroRNAs/metabolismo , Proteínas dos Microfilamentos/metabolismo , Atividade Motora/efeitos dos fármacos , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Peroxidase/metabolismo , Desempenho Psicomotor/efeitos dos fármacos , Fatores de Tempo
18.
Antioxid Redox Signal ; 22(2): 187-202, 2015 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24359188

RESUMO

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.


Assuntos
MicroRNAs/genética , Espécies Reativas de Oxigênio/metabolismo , Acidente Vascular Cerebral/genética , Animais , Humanos , Mitocôndrias/metabolismo , Oxirredução
19.
Neurochem Res ; 40(2): 301-7, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24993363

RESUMO

Astrocytes are critical regulators of neuronal function and an effective target for stroke therapy in animal models. Identifying individual targets with the potential for simultaneous activation of multiple downstream pathways that regulate astrocyte homeostasis may be a necessary element for successful clinical translation. Mitochondria and microRNAs each represent individual targets with multi-modal therapeutic potential. Mitochondria regulate metabolism and apoptosis, while microRNAs have the capacity to bind and inhibit numerous mRNAs. By combining strategies targeted at maintaining astrocyte function during and following cerebral ischemia, a synergistic therapeutic effect may be achieved.


Assuntos
Astrócitos/fisiologia , MicroRNAs/fisiologia , Mitocôndrias/fisiologia , Acidente Vascular Cerebral/terapia , Animais , Modelos Animais de Doenças , Humanos , Acidente Vascular Cerebral/patologia
20.
Adv Neurobiol ; 11: 171-88, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25236729

RESUMO

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.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...