Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Life Sci ; 333: 122172, 2023 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-37832632

RESUMO

AIMS: Cachexia, a metabolic syndrome, affects 21 % of patients suffering from ischemic encephalopathy. However, the specific mechanism and prevention measures are still unclear. Omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been proven to reduce inflammatory cytokine levels during ischemic events, but whether they have a protective effect against cachexia after hypoxic-ischemic brain damage (HIBD) remains unclear. MAIN METHODS: C57BL/6J wild-type and mfat-1 transgenic male mice were treated with and without HIBD. One day after HIBD, the epididymal white fat, gastrocnemius muscle and hypothalamus were weighed and analyzed the phenotypic changes. RNA sequencing was applied to gastrocnemius muscle to identify differential genes and pathways in HIBD groups. The effect of HPA axis on cachexia post-HIBD was examined via adrenalectomy, dexamethasone (0.1 mg/kg), and corticosterone injection (100 mg/kg). KEY FINDINGS: The results showed that the incidence of cachexia in mfat-1 mice, which produce high proportion of n-3 PUFAs, was significantly lower than that in wild-type mice post-HIBD. Cachexia-related factors, such as inflammation, muscle atrophy and lipid metabolism were significantly improved in mfat-1 HIBD. RNA sequencing revealed that catabolic and proteasome pathways were significantly downregulated. In hypothalamus, inflammatory cytokines, lipid peroxidation levels were reduced. Corticosterone, glucocorticoid receptor, and dexamethasone suppression test all showed that mfat-1 improved the dysfunction of the HPA axis post-HIBD. The present study elucidated for the first time that mfat-1 reduced HIBD-induced hyperactivation of the HPA axis in mice by reducing inflammation and oxidative stress and contributed to the reduction of metabolic imbalance in peripheral tissues. SIGNIFICANCE: Our study provides mechanistic information for the development of intervention strategies to prevent cachexia.


Assuntos
Sistema Hipotálamo-Hipofisário , Hipóxia-Isquemia Encefálica , Humanos , Camundongos , Animais , Masculino , Sistema Hipotálamo-Hipofisário/metabolismo , Caquexia/etiologia , Caquexia/prevenção & controle , Caquexia/metabolismo , Corticosterona/metabolismo , Camundongos Endogâmicos C57BL , Sistema Hipófise-Suprarrenal/metabolismo , Camundongos Transgênicos , Hipotálamo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Inflamação/metabolismo , Dexametasona/metabolismo , Animais Recém-Nascidos , Encéfalo/metabolismo
2.
Food Funct ; 14(14): 6610-6623, 2023 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-37395364

RESUMO

Hypoxic-ischemic encephalopathy (HIE) is one of the leading causes of acute neonatal death and chronic neurological damage, and severe HIE can have secondary sequelae such as cognitive impairment and cerebral palsy, for which effective interventions are lacking. In this study, we found that continuous 30-day intake of Acer truncatum Bunge seed oil (ASO) reduced brain damage and improved cognitive ability in HIE rats. Using lipidomic strategies, we observed that HIE rats had decreased unsaturated fatty acids and increased lysophospholipids in the brain. However, after 30 days of ASO treatment, phospholipids, plasmalogens, and unsaturated fatty acids increased, while lysophospholipids and oxidized glycerophospholipids decreased in both serum and the brain. Enrichment analysis showed that ASO intake mainly affected sphingolipid metabolism, fat digestion and absorption, glycerolipid metabolism and glycerophospholipid metabolic pathways in serum and the brain. Cluster, correlation, and confirmatory factor analyses showed that cognitive improvement after ASO administration was attributed to increased essential phospholipids and ω3/6/9 fatty acids, coupled with decreased oxidized glycerophospholipids in HIE rats. Our findings indicate that ASO has the potential to be developed as an effective food supplement for ischemic hypoxic newborns.


Assuntos
Acer , Hipóxia-Isquemia Encefálica , Ratos , Animais , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Lipidômica , Cognição , Glicerofosfolipídeos , Óleos de Plantas/farmacologia
3.
Mol Neurobiol ; 58(10): 5383-5395, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34319540

RESUMO

Brain photobiomodulation (PBM) is an innovative treatment for a variety of neurological conditions, including cerebral ischemia. However, the capability of PBM for ischemic stroke needs to be further explored and its mechanisms of action remain currently unclear. The aim of the present research was to identify a treatment protocol capable of inducing neuroprotection and to investigate the molecular mechanisms activated by a dual-wavelength near infrared (NIR) laser source in an organotypic hippocampal slice model of hypoxia/ischemia. Hippocampal slices were exposed to oxygen and glucose deprivation (OGD) for 30 min followed by NIR laser light (fluence 3.71, 7.42, or 14.84 J/cm2; wavelengths 808 nm and 905 nm) delivered immediately or 30 min or 60 min after OGD, in order to establish a therapeutic window. Neuronal injury was assessed by propidium iodide fluorescence 24 h later. Our results show that NIR laser irradiation attenuates OGD neurotoxicity once applied immediately or 30 min after OGD. Western blot analysis of proteins involved in neuroinflammation (iNOS, COX-2, NFkB subunit p65, and Bcl-2) and in glutamatergic-mediated synaptic activity (vGluT1, EAAT2, GluN1, and PSD95) showed that the protein modifications induced by OGD were reverted by NIR laser application. Moreover, CA1 confocal microscopy revealed that the profound morphological changes induced by OGD were reverted by NIR laser radiation. In conclusion, NIR laser radiation attenuates OGD neurotoxicity in organotypic hippocampal slices through attenuation of inflammatory mechanisms. These findings shed light on molecular definition of NIR neuroprotective mechanisms, thus underlining the potential benefit of this technique for the treatment of cerebral ischemia.


Assuntos
Hipocampo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/terapia , Terapia a Laser/métodos , Terapia com Luz de Baixa Intensidade/métodos , Neuroproteção/fisiologia , Animais , Feminino , Hipocampo/patologia , Hipóxia-Isquemia Encefálica/patologia , Masculino , Microscopia de Fluorescência/métodos , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar
4.
Int J Dev Neurosci ; 81(6): 510-519, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34021639

RESUMO

Perinatal asphyxia is a peripartum event that can cause permanent sequelae to the newborns, affecting the brain development. Recently, it has been demonstrated that epigenetics mechanisms play an important role in this injury and that folic acid (FA) supplementation during pregnancy can affect these epigenetics modifications as well as gene expression. We have identified both positive and negative effects of FA treatment in rats submitted to a model of neonatal hypoxia-ischemia (HI). Considering that FA supplementation is already used in pregnant women and that HI occurs in the peripartum period, this study was designated to evaluate how gestational FA supplementation and neonatal HI affect: apoptosis (caspase-3) and expression of synaptic proteins (synapsin and PSD-95) and the methylation of histone H3 lysine (K) 4 and 27 in the rat hippocampus. Pregnant Wistar rats were divided according to the diets: standard (SD), supplemented with 2 mg/kg of FA or with 20 mg/kg of FA. HI procedure was performed at the 7th PND. Protein expression and H3 methylation were evaluated at the 60th PND in the rats' hippocampus. Neonatal HI increased caspase-3 expression decreased synapsin expression and reduced H3K4me2, -me3 and H3K27me2, -me3 in the ipsilateral hippocampus. FA only prevented the augment in caspase-3 expression. In conclusion, neonatal HI caused lasting effects on caspase-3-mediated cell death (prevented by the FA) and synaptic proteins in the rats' hippocampus. This is the first study to show that histone modifications may contribute to these pathological findings in the hippocampus of HI animals.


Assuntos
Caspase 3/metabolismo , Ácido Fólico/administração & dosagem , Hipocampo/efeitos dos fármacos , Histonas/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Sinapsinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Metilação de DNA , Feminino , Hipocampo/metabolismo , Masculino , Gravidez , Ratos , Ratos Wistar
5.
Nutrients ; 14(1)2021 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-35011052

RESUMO

Perinatal hypoxia-ischemia (HI) is a major cause of neonatal brain injury, leading to long-term neurological impairments. Medical nutrition can be rapidly implemented in the clinic, making it a viable intervention to improve neurodevelopment after injury. The omega-3 (n-3) fatty acids docosahexaenoic acid (DHA, 22:6n-3) and eicosapentaenoic acid (EPA, 20:5n-3), uridine monophosphate (UMP) and choline have previously been shown in rodents to synergistically enhance brain phospholipids, synaptic components and cognitive performance. The objective of this study was to test the efficacy of an experimental diet containing DHA, EPA, UMP, choline, iodide, zinc, and vitamin B12 in a mouse model of perinatal HI. Male and female C57Bl/6 mice received the experimental diet or an isocaloric control diet from birth. Hypoxic ischemic encephalopathy was induced on postnatal day 9 by ligation of the right common carotid artery and systemic hypoxia. To assess the effects of the experimental diet on long-term motor and cognitive outcome, mice were subjected to a behavioral test battery. Lesion size, neuroinflammation, brain fatty acids and phospholipids were analyzed at 15 weeks after HI. The experimental diet reduced lesion size and neuroinflammation specifically in males. In both sexes, brain n-3 fatty acids were increased after receiving the experimental diet. The experimental diet also improved novel object recognition, but no significant effects on motor performance were observed. Current data indicates that early life nutritional supplementation with a combination of DHA, EPA, UMP, choline, iodide, zinc, and vitamin B12 may provide neuroprotection after perinatal HI.


Assuntos
Colina/administração & dosagem , Suplementos Nutricionais , Ácidos Docosa-Hexaenoicos/administração & dosagem , Ácido Eicosapentaenoico/administração & dosagem , Hipóxia-Isquemia Encefálica/dietoterapia , Doenças Neuroinflamatórias/dietoterapia , Uridina Monofosfato/administração & dosagem , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Ácidos Graxos Ômega-3/metabolismo , Feminino , Humanos , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Masculino , Camundongos Endogâmicos C57BL , Doenças Neuroinflamatórias/metabolismo , Doenças Neuroinflamatórias/patologia , Caracteres Sexuais
6.
Can J Physiol Pharmacol ; 98(10): 733-740, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32551885

RESUMO

This study aims to investigate the mechanisms through which fructose diphosphate (FDP) causes anti-hypoxia and anti-fatigue effects and improves learning and memory. Mice were divided into three groups: low-dose FDP (FDP-L), high-dose FDP (FDP-H), and a control group. Acute toxic hypoxia induced by carbon monoxide, sodium nitrite, and potassium cyanide and acute cerebral ischemic hypoxia were used to investigate the anti-hypoxia ability of FDP. The tests of rod-rotating, mouse tail suspension, and swimming endurance were used to explore the anti-fatigue effects of FDP. The Morris water maze experiment was used to determine the impact of FDP on learning and memory ability. Poisoning-induced hypoxic tests showed that mouse survival time was significantly prolonged in the FDP-L and FDP-H groups compared with the control group (p < 0.05). In the exhaustive swimming test, FDP significantly shortened struggling time and prolonged the time of mass-loaded swimming; the rod-rotating test showed that endurance time was significantly prolonged by using FDP (p < 0.05). FDP significantly decreased lactate and urea nitrogen levels and increased hepatic and muscle glycogen and glucose transporter-4 and Na+-K+-ATPase (p < 0.05). To conclude, FDP enhances hypoxia tolerance and fatigue resistance and improves learning and memory ability through regulating glucose and energy metabolism.


Assuntos
Comportamento Animal/efeitos dos fármacos , Metabolismo Energético/efeitos dos fármacos , Fadiga/prevenção & controle , Frutosedifosfatos/farmacologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Hipóxia/prevenção & controle , Aprendizagem/efeitos dos fármacos , Memória/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Fadiga/metabolismo , Fadiga/fisiopatologia , Fadiga/psicologia , Hipóxia/metabolismo , Hipóxia/fisiopatologia , Hipóxia/psicologia , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Hipóxia-Isquemia Encefálica/psicologia , Locomoção/efeitos dos fármacos , Camundongos , Teste do Labirinto Aquático de Morris/efeitos dos fármacos , Teste de Desempenho do Rota-Rod , Natação
7.
Mol Neurobiol ; 57(5): 2194-2205, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31974940

RESUMO

Hypoxic-ischemic (HI) brain injury remains an important cause of brain damage in neonates with potential life-long consequences. Caffeine, which is a competitive inhibitor of adenosine receptors, is commonly used as treatment for preterm apnoea in clinical settings. In the current study, we investigated the effects of caffeine given at 0 h, 6 h, 12 h or 24 h after HI in P10 mouse pups. Open field and rotarod behavioural tests were performed 2 weeks after injury, and brain morphology was then evaluated. Gene expression and immunohistological analyses were assessed in mice 1- and 5-day post-HI. A single dose of caffeine directly after HI resulted in a reduction of the lesion in the grey and white matter, judged by immunostaining of MAP2 and MBP, respectively, compared to PBS-treated controls. In addition, the number of amoeboid microglia and apoptotic cells, the area covered by astrogliosis, and the expression of pro-inflammatory cytokines were significantly decreased. Behavioural assessment after 2 weeks showed increased open-field activity after HI, and this was normalised if caffeine was administered immediately after the injury. Later administrations of caffeine did not change the outcomes when compared to the vehicle group. In conclusion, caffeine only yielded neuroprotection and immunomodulation in a neonatal model of brain hypoxia ischaemia if administered immediately after injury.


Assuntos
Cafeína/administração & dosagem , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Imunomodulação/efeitos dos fármacos , Neuroglia/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Antagonistas de Receptores Purinérgicos P1/administração & dosagem , Animais , Lesões Encefálicas/patologia , Cafeína/farmacologia , Cafeína/uso terapêutico , Fragmentação do DNA/efeitos dos fármacos , Doenças Desmielinizantes/prevenção & controle , Esquema de Medicação , Avaliação Pré-Clínica de Medicamentos , Comportamento Exploratório , Feminino , Gliose/etiologia , Gliose/prevenção & controle , Hipóxia-Isquemia Encefálica/metabolismo , Inflamação/genética , Inflamação/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/efeitos dos fármacos , Microglia/metabolismo , Neuroglia/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Antagonistas de Receptores Purinérgicos P1/farmacologia , Antagonistas de Receptores Purinérgicos P1/uso terapêutico , Distribuição Aleatória , Teste de Desempenho do Rota-Rod , Método Simples-Cego , Organismos Livres de Patógenos Específicos
8.
Neural Plast ; 2020: 8828826, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33488693

RESUMO

Aim. Neonatal hypoxic-ischemic encephalopathy (HIE) is a significant cause of perinatal morbidity and mortality. Chinese Tuina is an effective treatment for HIE, but the molecular mechanisms are yet unknown. This study investigated the effect and mechanisms of Chinese Tuina on the inflammatory response in neonatal HIE rats. Main Methods. 30 male neonatal rats were divided randomly into 3 groups: sham, HIE, and HIE with Chinese Tuina (CHT) groups. The HIE and CHT groups were subjected to left common carotid occlusion and hypoxia at 3 days postnatal (P3). The pups in the CHT group received Chinese Tuina treatment on the next day for 28 days. The weight was measured at P4, P9, P13, P21, and P31. The behavioral functions were determined at P21. The protein expression and the methylation level in promoter regions of TNF-α and IL-10 were determined by Western blotting, immunohistochemistry, and pyrosequencing, respectively, at P33. Key Findings. The weight gain in the HIE group was slow compared with that of the CHT group. The rats in the CHT group performed better both in the balance beam and hang plate experiment. Chinese Tuina inhibited the expression of TNF-α and upregulated the expression of IL-10. Neonatal hypoxic-ischemic injury downregulated the methylation level in promoter regions of TNF-α at all CpG points but not IL-10. However, Chinese Tuina did not change the methylation level in promoter regions of TNF-α and IL-10. Significance. Chinese Tuina protected against HIE through inhibiting the neuroinflammatory reaction. While HIE markedly downregulated the methylation level of TNF-α, the protective effects of Chinese Tuina were independent of the regulation of the methylation level of TNF-α and IL-10.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/terapia , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/metabolismo , Massagem/métodos , Animais , Animais Recém-Nascidos , Feminino , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley
9.
Kaohsiung J Med Sci ; 36(1): 43-53, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31642199

RESUMO

Accumulating evidences have proved the protective role of traditional Chinese medicine in improving neurological damage induced by cerebral hypoxia-ischemia. Herein, we hypothesized that Dendrobium officinale aqueous extract exerted neuroprotection against brain damage. Initially, a model of hypoxic-ischemic brain damage (HIBD) was induced in neonatal rats, which were subsequently intragastrically administered with different doses of Dendrobium officinale aqueous extract. Next, the antioxidant capacity was examined by enzyme-linked immunosorbent assay. 2,3,5-Triphenyltetrazolium chloride and terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling staining assays were adopted to determine neuronal apoptosis in brain tissues. Furthermore, neurotrophic factors and hypoxia-inducible factor-1α (HIF-1α) expression was identified by Western blot analysis. The neonatal rat models of HIBD presented impaired neurobehaviors and antioxidant capacity, increased neuronal apoptosis and expression of HIF-1α and histone deacetylase 1 (HDAC1), as well as diminished expression of neurotrophic factors and K+ -Cl- -cotransporter 2 (KCC2). Notably, in response to different doses of Dendrobium officinale aqueous extract, the impairment on neurobehaviors and antioxidant capacity was alleviated, accompanied by reduced levels of nitric oxide synthase, nitric oxide, and malondialdehyde, and increased superoxide dismutase activity. Besides, the neuronal apoptosis was inhibited as reflected by down-regulated cleaved caspase-3 and Bax and up-regulated Bcl-2. Moreover, we also found accelerated expression of neurotrophic factors and KCC2 and diminished expression of HIF-1α and HDAC1. Altogether, this present study highlights that the aqueous extract of Dendrobium officinale can suppress the neuronal apoptosis and enhance the expression of neurotrophic factors to protect neonatal rats against HIBD.


Assuntos
Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Dendrobium/química , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Extratos Vegetais/uso terapêutico , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Histona Desacetilase 1/metabolismo , Marcação In Situ das Extremidades Cortadas , Malondialdeído/metabolismo , Neuroproteção/efeitos dos fármacos , Óxido Nítrico/metabolismo , Óxido Nítrico Sintase/metabolismo , Ratos , Superóxido Dismutase/metabolismo , Simportadores/metabolismo
10.
Int J Dev Neurosci ; 79: 86-95, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31693927

RESUMO

INTRODUCTION: Neonatal Hypoxia-Ischemia (HI) is a major cause of morbidity and mortality, and is frequently associated with short and long-term neurologic and cognitive impairments. The HI injury causes mitochondrial damage leading to increased production of reactive oxygen species (ROS). Phytoestrogens are non-steroidal plant substances structurally and functionally similar to estrogen. Coumestrol is a potent isoflavonoid with a protective effect against ischemic brain damage in adult rats. Our aim was to determine if coumestrol treatment following neonatal HI attenuates the long-term cognitive deficits induced by neonatal HI, as well as to investigate one possible mechanism underlying its potential effect. METHODS: On the 7th postnatal day, male Wistar rats were submitted to the Levine-Rice HI model. Intraperitoneal injections of 20 mg/kg of coumestrol, or vehicle, were administered immediately pre-hypoxia or 3 h post-hypoxia. At 12 h after HI the mitochondrial status and ROS levels were determined. At 60th postnatal day the cognitive deficits were revealed in the Morris water maze reference and working spatial memories. Following behavioral analysis, histological assessment was performed and reactive astrogliosis was measured by GFAP expression. RESULTS: Results demonstrate that both pre- and post-HI administration of coumestrol were able to counteract the long-term cognitive and morphological impairments caused by HI, as well as to block the late reactive astrogliosis. The pre-HI administration of coumestrol was able to prevent the early mitochondrial dysfunction in the hippocampus of injured rat pups. CONCLUSION: Present data suggest that coumestrol exerts protection against experimental neonatal brain hypoxia-ischemia through, at least in part, early modulation of mitochondrial function.


Assuntos
Encéfalo/efeitos dos fármacos , Cognição/efeitos dos fármacos , Disfunção Cognitiva/tratamento farmacológico , Cumestrol/farmacologia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Animais , Encéfalo/metabolismo , Disfunção Cognitiva/metabolismo , Cumestrol/uso terapêutico , Hipóxia-Isquemia Encefálica/metabolismo , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Mitocôndrias/metabolismo , Fármacos Neuroprotetores/uso terapêutico , Fitoestrógenos/farmacologia , Fitoestrógenos/uso terapêutico , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo
11.
Cell Transplant ; 28(12): 1686-1699, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31694396

RESUMO

Hyperbaric oxygen (HBO) therapy may promote neurological recovery from hypoxic-ischemic encephalopathy (HIE). However, the therapeutic effects of HBO and its associated mechanisms remain unknown. The canonical Wnt/ß-catenin signaling pathways and bone morphogenetic protein (BMP) play important roles in mammalian nervous system development. The present study examined whether HBO stimulates the differentiation of neural stem cells (NSCs) and its effect on Wnt3/ß-catenin and BMP2 signaling pathways. We showed HBO treatment (2 ATA, 60 min) promoted differentiation of NSCs into neurons and oligodendrocytes in vitro. In addition, rat hypoxic-ischemic brain damage (HIBD) tissue extracts also promoted the differentiation of NSCs into neurons and oligodendrocytes, with the advantage of reducing the number of astrocytes. These effects were most pronounced when these two were combined together. In addition, the expression of Wnt3a, BMP2, and ß-catenin nuclear proteins were increased after HBO treatment. However, blockade of Wnt/ß-catenin or BMP signaling inhibited NSC differentiation and reduced the expression of Wnt3a, BMP2, and ß-catenin nuclear proteins. In conclusion, HBO promotes differentiation of NSCs into neurons and oligodendrocytes and reduced the number of astrocytes in vitro possibly through regulation of Wnt3/ß-catenin and BMP2 signaling pathways. HBO may serve as a potential therapeutic strategy for treating HIE.


Assuntos
Proteína Morfogenética Óssea 2/metabolismo , Diferenciação Celular , Oxigenoterapia Hiperbárica , Hipóxia-Isquemia Encefálica , Células-Tronco Neurais/metabolismo , Via de Sinalização Wnt , Proteína Wnt3/metabolismo , beta Catenina/metabolismo , Animais , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Hipóxia-Isquemia Encefálica/terapia , Células-Tronco Neurais/patologia , Ratos , Ratos Sprague-Dawley
12.
Neuromolecular Med ; 21(4): 505-516, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31515728

RESUMO

Brain endothelial cells play an important role in maintaining blood flow homeostasis in the brain. Cerebral ischemia is a major cause of endothelial dysfunction which can disrupt the blood-brain barrier (BBB). Oxygen-glucose deprivation (OGD)/reperfusion promote cell death and BBB breakdown in brain endothelial cells. Acetyl-11-keto-ß-boswellic acid (AKBA), a biologically active phytoconstituent of the medicinal plant Boswellia serrata, has been shown to be protective against various inflammatory diseases as well as ischemic brain injury. The molecular mechanisms underlying these beneficial characteristics of AKBA are poorly understood. We subjected bEND.3 cells to OGD/reperfusion to investigate the protective role of AKBA in this model. We found that AKBA treatment attenuated endothelial cell death and oxidative stress assessed by means of TUNEL assay, cleaved-caspase-3, and dihydroethidium (DHE) staining. Furthermore, OGD downregulated tight junction proteins ZO-1 and Occludin levels, and increased the expressions of inflammatory cytokines TNF-α, ICAM-1, and complement C3a receptor (C3aR). We also noticed the increased phosphorylation of ERK 1/2 in bEND.3 cells in OGD group. AKBA treatment significantly attenuated expression levels of these inflammatory proteins and prevented the degradation of ZO-1 and Occludin following OGD. In conclusion, AKBA treatment provides protection against endothelial cell dysfunction following OGD by attenuating oxidative stress and inflammation.


Assuntos
Anti-Inflamatórios/farmacologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Boswellia/química , Ativação do Complemento/efeitos dos fármacos , Inativadores do Complemento/farmacologia , Células Endoteliais/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Plantas Medicinais/química , Triterpenos/farmacologia , Animais , Hipóxia Celular , Dano ao DNA , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Glucose/farmacologia , Inflamação , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Oxigênio/farmacologia , Extratos Vegetais/química , Espécies Reativas de Oxigênio/análise
13.
Nutr Res ; 64: 93-108, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30802728

RESUMO

Pilose antler polypeptides (PAP) have recently been found to be effective in the treatment of brain damage in hypoxic-ischemic encephalopathy (HIE). However, the impacts of hypoxic-ischemic (HI)-induced injury on oxidative stress and inflammation in peripheral tissues remain unclear. In the present study, we hypothesized that the administration of PAP might exert a protective effect on HI-induced peripheral tissue dysfunction. To that end, HI-injured rats were administered PAP for 3 weeks, and then the metabolic phenotypes and gut microbiota were evaluated by qPCR and 16S rRNA sequencing analysis. Hepatic lipid accumulation, systemic oxidative stress and inflammation, as well as impaired gut barrier function and altered gut microbiota were found in HI-injured rats, which were reversed by the treatment of PAP. PAP treatment modulated the abundance and composition of gut microbiota, and PICRUSt analyses revealed that PAP treatment also led to a functional change in the microbial communities. These protective effects of PAP were associated with attenuated susceptibility to bacterial infections, decreased antibiotic synthesis and changed cellular processes and signaling, which may cause inflammation, barrier dysfunction, oxidative stress and mitochondria dysfunction in HI rats. In conclusion, these results suggested that PAP protected against HI-induced peripheral tissue damage in rats and therefore might be a potential candidate for the treatment of HIE and its complications.


Assuntos
Chifres de Veado/química , Cervos , Microbioma Gastrointestinal/efeitos dos fármacos , Hipóxia-Isquemia Encefálica , Inflamação/prevenção & controle , Estresse Oxidativo/efeitos dos fármacos , Peptídeos/farmacologia , Animais , Infecções Bacterianas/prevenção & controle , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/fisiopatologia , Hipóxia , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Isquemia , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia , Masculino , Mitocôndrias/efeitos dos fármacos , Peptídeos/uso terapêutico , Reação em Cadeia da Polimerase , RNA Ribossômico 16S , Ratos Sprague-Dawley , Transdução de Sinais
14.
J Cereb Blood Flow Metab ; 39(1): 118-130, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-28949271

RESUMO

Hypoxic ischemic encephalopathy (HIE) leads to significant morbidity and mortality. Impaired autoregulation after hypoxia-ischaemia has been suggested to contribute further to injury. Thalamic lactate/N-Acetylasperate (Lac/NAA) peak area ratio of > 0.3 on proton (1H) magnetic resonance spectroscopy (MRS) is associated with poor neurodevelopment outcome following HIE. Cytochrome-c-oxidase (CCO) plays a central role in mitochondrial oxidative metabolism and ATP synthesis. Using a novel broadband NIRS system, we investigated the impact of pressure passivity of cerebral metabolism (CCO), oxygenation (haemoglobin difference (HbD)) and cerebral blood volume (total haemoglobin (HbT)) in 23 term infants following HIE during therapeutic hypothermia (HT). Sixty-minute epochs of data from each infant were studied using wavelet analysis at a mean age of 48 h. Wavelet semblance (a measure of phase difference) was calculated to compare reactivity between mean arterial blood pressure (MABP) with oxCCO, HbD and HbT. OxCCO-MABP semblance correlated with thalamic Lac/NAA ( r = 0.48, p = 0.02). OxCCO-MABP semblance also differed between groups of infants with mild to moderate and severe injury measured using brain MRI score ( p = 0.04), thalamic Lac/NAA ( p = 0.04) and neurodevelopmental outcome at one year ( p = 0.04). Pressure passive changes in cerebral metabolism were associated with injury severity indicated by thalamic Lac/NAA, MRI scores and neurodevelopmental assessment at one year of age.


Assuntos
Encéfalo/metabolismo , Hipóxia-Isquemia Encefálica/metabolismo , Mitocôndrias/metabolismo , Trifosfato de Adenosina/biossíntese , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Pressão Sanguínea , Circulação Cerebrovascular , Deficiências do Desenvolvimento/diagnóstico por imagem , Deficiências do Desenvolvimento/etiologia , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Feminino , Homeostase , Humanos , Hipóxia-Isquemia Encefálica/complicações , Hipóxia-Isquemia Encefálica/diagnóstico por imagem , Recém-Nascido , Ácido Láctico/metabolismo , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Masculino , Tálamo/metabolismo
15.
Free Radic Biol Med ; 131: 98-114, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30458277

RESUMO

The transcriptional factor Nrf2, a master regulator of oxidative stress and inflammation that are tightly linked to the development and progression of cerebral ischemia pathology, plays a vital role in inducing the endogenous neuroprotective process. Here, hypoxic-ischemia (HI) was performed in adult Nrf2 knockout and wildtype mice that were orally pretreated either with standardized Korean red ginseng extract (Ginseng) or dimethyl fumarate (DMF), two candidate Nrf2 inducers, to determine whether the putative protection was through an Nrf2-dependent mechanism involving the attenuation of reactive gliosis. Results show that Nrf2 target cytoprotective genes were distinctly elevated following HI. Pretreatment with Ginseng or DMF elicited robust neuroprotection against the deterioration of acute cerebral ischemia damage in an Nrf2-dependent manner as revealed by the reductions of neurological deficits score, infarct volume and brain edema, as well as enhanced expression levels of Nrf2 target antioxidant proteins and anti-inflammation mediators. In both ischemic striatum and cortex, the dynamic pattern of attenuated reactive gliosis in astrocytes and microglia, including affected astrocytic dysfunction in glutamate metabolism and water homeostasis, correlated well with the Nrf2-dependent neuroprotection by Ginseng or DMF. Furthermore, such neuroprotective benefits extended to the late phase of ischemic brain damage after HI, as evidenced by improvements in neurobehavioral outcomes, infarct volume and brain edema. Overall, pretreatment with Ginseng or DMF identically attenuates reactive gliosis and confers long-lasting neuroprotective efficacy against ischemic brain damage through an Nrf2-dependent mechanism. This study also provides new insight into the profitable contribution of reactive gliosis in the Nrf2-dependent neuroprotection in acute brain injury.


Assuntos
Fumarato de Dimetilo/farmacologia , Gliose/tratamento farmacológico , Hipóxia-Isquemia Encefálica/tratamento farmacológico , Fator 2 Relacionado a NF-E2/genética , Fármacos Neuroprotetores/farmacologia , Panax/química , Animais , Aquaporina 4/genética , Aquaporina 4/metabolismo , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Artérias Carótidas/cirurgia , Córtex Cerebral/irrigação sanguínea , Córtex Cerebral/metabolismo , Córtex Cerebral/fisiopatologia , Transtornos Cerebrovasculares/cirurgia , Corpo Estriado/irrigação sanguínea , Corpo Estriado/metabolismo , Corpo Estriado/fisiopatologia , Modelos Animais de Doenças , Regulação da Expressão Gênica , Proteína Glial Fibrilar Ácida/genética , Proteína Glial Fibrilar Ácida/metabolismo , Gliose/genética , Gliose/metabolismo , Gliose/fisiopatologia , Humanos , Hipóxia-Isquemia Encefálica/genética , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Microglia/efeitos dos fármacos , Microglia/metabolismo , Microglia/patologia , NAD(P)H Desidrogenase (Quinona)/genética , NAD(P)H Desidrogenase (Quinona)/metabolismo , Fator 2 Relacionado a NF-E2/deficiência , Extratos Vegetais/farmacologia
16.
Lancet Neurol ; 18(1): 35-45, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30447969

RESUMO

BACKGROUND: In neonatal encephalopathy, the clinical manifestations of injury can only be reliably assessed several years after an intervention, complicating early prognostication and rendering trials of promising neuroprotectants slow and expensive. We aimed to determine the accuracy of thalamic proton magnetic resonance (MR) spectroscopy (MRS) biomarkers as early predictors of the neurodevelopmental abnormalities observed years after neonatal encephalopathy. METHODS: We did a prospective multicentre cohort study across eight neonatal intensive care units in the UK and USA, recruiting term and near-term neonates who received therapeutic hypothermia for neonatal encephalopathy. We excluded infants with life-threatening congenital malformations, syndromic disorders, neurometabolic diseases, or any alternative diagnoses for encephalopathy that were apparent within 6 h of birth. We obtained T1-weighted, T2-weighted, and diffusion-weighted MRI and thalamic proton MRS 4-14 days after birth. Clinical neurodevelopmental tests were done 18-24 months later. The primary outcome was the association between MR biomarkers and an adverse neurodevelopmental outcome, defined as death or moderate or severe disability, measured using a multivariable prognostic model. We used receiver operating characteristic (ROC) curves to examine the prognostic accuracy of the individual biomarkers. This trial is registered with ClinicalTrials.gov, number NCT01309711. FINDINGS: Between Jan 29, 2013, and June 25, 2016, we recruited 223 infants who all underwent MRI and MRS at a median age of 7 days (IQR 5-10), with 190 (85%) followed up for neurological examination at a median age of 23 months (20-25). Of those followed up, 31 (16%) had moderate or severe disability, including one death. Multiple logistic regression analysis could not be done because thalamic N-acetylaspartate (NAA) concentration alone accurately predicted an adverse neurodevelopmental outcome (area under the curve [AUC] of 0·99 [95% CI 0·94-1·00]; sensitivity 100% [74-100]; specificity 97% [90-100]; n=82); the models would not converge when any additional variable was examined. The AUC (95% CI) of clinical examination at 6 h (n=190) and at discharge (n=167) were 0·72 (0·65-0·78) and 0·60 (0·53-0·68), respectively, and the AUC of abnormal amplitude integrated EEG at 6 h (n=169) was 0·73 (0·65-0·79). On conventional MRI (n=190), cortical injury had an AUC of 0·67 (0·60-0·73), basal ganglia or thalamic injury had an AUC of 0·81 (0·75-0·87), and abnormal signal in the posterior limb of internal capsule (PLIC) had an AUC of 0·82 (0·76-0·87). Fractional anisotropy of PLIC (n=65) had an AUC of 0·82 (0·76-0·87). MRS metabolite peak-area ratios (n=160) of NAA-creatine (<1·29) had an AUC of 0·79 (0·72-0·85), of NAA-choline had an AUC of 0·74 (0·66-0·80), and of lactate-NAA (>0·22) had an AUC of 0·94 (0·89-0·97). INTERPRETATION: Thalamic proton MRS measures acquired soon after birth in neonatal encephalopathy had the highest accuracy to predict neurdevelopment 2 years later. These methods could be applied to increase the power of neuroprotection trials while reducing their duration. FUNDING: National Institute for Health Research UK.


Assuntos
Encéfalo/diagnóstico por imagem , Hipotermia Induzida , Hipóxia-Isquemia Encefálica/terapia , Espectroscopia de Ressonância Magnética , Ácido Aspártico/análogos & derivados , Ácido Aspártico/metabolismo , Encéfalo/metabolismo , Feminino , Humanos , Hipóxia-Isquemia Encefálica/diagnóstico por imagem , Hipóxia-Isquemia Encefálica/metabolismo , Lactente , Recém-Nascido , Masculino , Estudos Prospectivos , Tálamo , Resultado do Tratamento
17.
Oxid Med Cell Longev ; 2018: 9416432, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30258527

RESUMO

In the cerebral hypoxia-ischemia rat model, the prophylactic administration of zinc can cause either cytotoxicity or preconditioning effect, whereas the therapeutic administration of selenium decreases the ischemic damage. Herein, we aimed to explore whether supplementation of low doses of prophylactic zinc and therapeutic selenium could protect from a transient hypoxic-ischemic event. We administrated zinc (0.2 mg/kg of body weight; ip) daily for 14 days before a 10 min common carotid artery occlusion (CCAO). After CCAO, we administrated sodium selenite (6 µg/kg of body weight; ip) daily for 7 days. In the temporoparietal cerebral cortex, we determined nitrites by the Griess method and lipid peroxidation by the Gerard-Monnier assay. qPCR was used to measure mRNA of nitric oxide synthases, antioxidant enzymes, chemokines, and their receptors. We measured the enzymatic activity of SOD and GPx and protein levels of chemokines and their receptors by ELISA. We evaluated long-term memory using the Morris-Water maze test. Our results showed that prophylactic administration of zinc caused a preconditioning effect, decreasing nitrosative/oxidative stress and increasing GPx and SOD expression and activity, as well as eNOS expression. The therapeutic administration of selenium maintained this preconditioning effect up to the late phase of hypoxia-ischemia. Ccl2, Ccr2, Cxcl12, and Cxcr4 were upregulated, and long-term memory was improved. Pyknotic cells were decreased suggesting prevention of neuronal cell death. Our results show that the prophylactic zinc and therapeutic selenium administration induces effective neuroprotection in the early and late phases after CCAO.


Assuntos
Antioxidantes/metabolismo , Córtex Cerebral/efeitos dos fármacos , Hipóxia-Isquemia Encefálica/metabolismo , Memória/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Selenito de Sódio/administração & dosagem , Zinco/administração & dosagem , Animais , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Wistar
18.
Neurochem Res ; 43(6): 1210-1226, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29696512

RESUMO

Notoginsenoside R1 (NGR1) is a predominant phytoestrogen extracted from Panax notoginseng that has recently been reported to play important roles in the treatment of cardiac dysfunction, diabetic kidney disease, and acute liver failure. Studies have suggested that NGR1 may be a viable treatment of hypoxic-ischemic brain damage (HIBD) in neonates by reducing endoplasmic reticulum stress via estrogen receptors (ERs). However, whether NGR1 has other neuroprotective mechanisms or long-term neuroprotective effects is unclear. In this study, oxygen-glucose deprivation/reoxygenation (OGD/R) in primary cortical neurons and unilateral ligation of the common carotid artery (CCL) in 7-day-old postnatal Sprague Dawley (SD) rats followed by exposure to a hypoxic environment were used to mimic an HIBD episode. We assessed the efficacy of NGR1 by measuring neuronal damage with MTT assay and assessed brain injury by TTC staining and brain water content detection 24-48 h after OGD/HIE. Simultaneously, we measured the long-term neurophysiological effects using the beam walking test (5 weeks after HI) and Morris water maze test 5-6 weeks after HI. Expression of PI3K-Akt-mTOR/JNK (24 h after HI or OGD/R) proteins was detected by Western blotting after stimulation with HI, NGR1, LY294002 (PI3K inhibitor), 740Y-P (PI3K agonist), or ICI 182780(estrogen receptors inhibitor). The results indicated that NGR1 exerted neuroprotective effects by inhibiting neuronal apoptosis and promoting cell survival via the PI3K-Akt-mTOR/JNK signaling pathways by targeting ER in neonatal hypoxic-ischemic injury.


Assuntos
Ginsenosídeos/uso terapêutico , Hipóxia-Isquemia Encefálica/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fármacos Neuroprotetores/uso terapêutico , Fosfatidilinositol 3-Quinases/biossíntese , Proteínas Proto-Oncogênicas c-akt/biossíntese , Serina-Treonina Quinases TOR/biossíntese , Animais , Animais Recém-Nascidos , Células Cultivadas , Ginsenosídeos/farmacologia , Hipóxia-Isquemia Encefálica/prevenção & controle , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Sprague-Dawley
19.
Exp Neurol ; 300: 222-231, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29199131

RESUMO

We investigated whether electroacupuncture (EA) and treadmill (TM) exercise improve behaviors related to motor and memory dysfunction in a cerebral palsy-like rat model via activation of oligodendrogenesis. A neonatal hypoxia-ischemia model was created using Sprague-Dawley rats (P7), and these underwent EA stimulation and treadmill training from 3 to 5weeks after hypoxia-ischemia induction. EA treatment was delivered via electrical stimulation (2Hz, 1mA) at two acupoints, Baihui (GV20) and Zusanli (ST36). Behavioral tests showed that EA alleviated motor dysfunction caused by hypoxia-ischemia on a rotarod test, and TM exercise alleviated motor and memory dysfunction seen on cylinder and passive avoidance tests. Combined therapy with EA and TM exercise showed synergistic effects on the cylinder, rotarod, and catwalk tests. TM exercise significantly restored corpus callosum thickness, and combined therapy with EA and TM restored myelin basic protein (MBP) levels in this region. While EA stimulation only increased activation of cAMP-response element binging protein (CREB) in oligodendrocytes of the corpus callosum, TM exercise increased newly generated oligodendrocyte progenitor cells or oligodendrocytes via activation of CREB. Synergistic effects on oligodendrogenesis were also observed by the combined therapy. Furthermore, the combined therapy induced mature brain-derived neurotrophic factor (BDNF) expression in the cerebral cortex. These results demonstrate that combined therapy with EA and TM exercise may restore myelin components following neonatal hypoxia-ischemia via upregulation of oligodendrogenesis involving CREB/BDNF signaling, which subsequently improves motor and memory function. Therefore, combined therapy with EA and TM exercise offers another treatment option for functional recovery from injuries caused by neonatal hypoxia-ischemia, such as cerebral palsy.


Assuntos
Terapia Combinada/métodos , Doenças Desmielinizantes/terapia , Eletroacupuntura/métodos , Teste de Esforço/métodos , Hipóxia-Isquemia Encefálica/terapia , Oligodendroglia/fisiologia , Animais , Animais Recém-Nascidos , Proliferação de Células/fisiologia , Corpo Caloso/citologia , Corpo Caloso/metabolismo , Corpo Caloso/patologia , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Feminino , Hipóxia-Isquemia Encefálica/metabolismo , Hipóxia-Isquemia Encefálica/patologia , Masculino , Gravidez , Ratos , Ratos Sprague-Dawley
20.
PLoS One ; 12(9): e0185202, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28934366

RESUMO

Melatonin is a promising neuroprotective agent after perinatal hypoxic-ischemic (HI) brain injury. We used in-vivo 1H magnetic resonance spectroscopy to investigate effects of melatonin treatment on brain metabolism after HI. Postnatal day 7 Sprague-Dawley rats with unilateral HI brain injury were treated with either melatonin 10 mg/kg dissolved in phosphate-buffered saline (PBS) with 5% dimethyl sulfoxide (DMSO) or vehicle (5% DMSO and/or PBS) directly and at 6 hours after HI. 1H MR spectra from the thalamus in the ipsilateral and contralateral hemisphere were acquired 1 day after HI. Our results showed that injured animals had a distinct metabolic profile in the ipsilateral thalamus compared to sham with low concentrations of total creatine, choline, N-acetyl aspartate (NAA), and high concentrations of lipids. A majority of the melatonin-treated animals had a metabolic profile characterized by higher total creatine, choline, NAA and lower lipid levels than other HI animals. When comparing absolute concentrations, melatonin treatment resulted in higher glutamine levels and lower lipid concentrations compared to DMSO treatment as well as higher macromolecule levels compared to PBS treatment day 1 after HI. DMSO treated animals had lower concentrations of glucose, creatine, phosphocholine and macromolecules compared to sham animals. In conclusion, the neuroprotective effects of melatonin were reflected in a more favorable metabolic profile including reduced lipid levels that likely represents reduced cell injury. Neuroprotective effects may also be related to the influence of melatonin on glutamate/glutamine metabolism. The modulatory effects of the solvent DMSO on cerebral energy metabolism might have masked additional beneficial effects of melatonin.


Assuntos
Hipóxia-Isquemia Encefálica/metabolismo , Melatonina/farmacologia , Fármacos Neuroprotetores/farmacologia , Animais , Animais Recém-Nascidos , Relação Dose-Resposta a Droga , Espectroscopia de Ressonância Magnética , Ratos , Ratos Sprague-Dawley , Tálamo/efeitos dos fármacos , Tálamo/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA