Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 204
Filtrar
Mais filtros

Medicinas Complementares
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Mol Neurobiol ; 58(10): 5338-5355, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34302281

RESUMO

Evidence for the clinical use of neuroprotective drugs for the treatment of cerebral ischemia (CI) is still greatly limited. Spatial/temporal disorientation and cognitive dysfunction are among the most prominent long-term sequelae of CI. Cannabidiol (CBD) is a non-psychotomimetic constituent of Cannabis sativa that exerts neuroprotective effects against experimental CI. The present study investigated possible neuroprotective mechanisms of action of CBD on spatial memory impairments that are caused by transient global cerebral ischemia (TGCI) in rats. Hippocampal synaptic plasticity is a fundamental mechanism of learning and memory. Thus, we also evaluated the impact of CBD on neuroplastic changes in the hippocampus after TGCI. Wistar rats were trained to learn an eight-arm aversive radial maze (AvRM) task and underwent either sham or TGCI surgery. The animals received vehicle or 10 mg/kg CBD (i.p.) 30 min before surgery, 3 h after surgery, and then once daily for 14 days. On days 7 and 14, we performed a retention memory test. Another group of rats that received the same pharmacological treatment was tested in the object location test (OLT). Brains were removed and processed to assess neuronal degeneration, synaptic protein levels, and dendritic remodeling in the hippocampus. Cannabidiol treatment attenuated ischemia-induced memory deficits. In rats that were subjected to TGCI, CBD attenuated hippocampal CA1 neurodegeneration and increased brain-derived neurotrophic factor levels. Additionally, CBD protected neurons against the deleterious effects of TGCI on dendritic spine number and the length of dendritic arborization. These results suggest that the neuroprotective effects of CBD against TGCI-induced memory impairments involve changes in synaptic plasticity in the hippocampus.


Assuntos
Canabidiol/uso terapêutico , Hipocampo/efeitos dos fármacos , Ataque Isquêmico Transitório/prevenção & controle , Plasticidade Neuronal/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Canabidiol/farmacologia , Modelos Animais de Doenças , Hipocampo/metabolismo , Hipocampo/patologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Plasticidade Neuronal/fisiologia , Neuroproteção/fisiologia , Técnicas de Cultura de Órgãos , Ratos , Ratos Wistar , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia , Sinapses/metabolismo , Sinapses/patologia
2.
Molecules ; 26(8)2021 Apr 09.
Artigo em Inglês | MEDLINE | ID: mdl-33918660

RESUMO

Angelica gigas Nakai root contains decursin which exerts beneficial properties such as anti-amnesic and anti-inflammatory activities. Until now, however, the neuroprotective effects of decursin against transient ischemic injury in the forebrain have been insufficiently investigated. Here, we revealed that post-treatment with decursin and the root extract saved pyramidal neurons in the hippocampus following transient ischemia for 5 min in gerbil forebrain. Through high-performance liquid chromatography, we defined that decursin was contained in the extract as 7.3 ± 0.2%. Based on this, we post-treated with 350 mg/kg of extract, which is the corresponding dosage of 25 mg/kg of decursin that exerted neuroprotection in gerbil hippocampus against the ischemia. In addition, behavioral tests were conducted to evaluate ischemia-induced dysfunctions via tests of spatial memory (by the 8-arm radial maze test) and learning memory (by the passive avoidance test), and post-treatment with the extract and decursin attenuated ischemia-induced memory impairments. Furthermore, we carried out histochemistry, immunohistochemistry, and double immunohistofluorescence. Pyramidal neurons located in the subfield cornu ammonis 1 (CA1) among the hippocampal subfields were dead at 5 days after the ischemia; however, treatment with the extract and decursin saved the pyramidal neurons after ischemia. Immunoglobulin G (IgG, an indicator of extravasation), which is not found in the parenchyma in normal brain tissue, was apparently shown in CA1 parenchyma from 2 days after the ischemia, but IgG leakage was dramatically attenuated in the CA1 parenchyma treated with the extract and decursin. Furthermore, astrocyte endfeet, which are a component of the blood-brain barrier (BBB), were severely damaged at 5 days after the ischemia; however, post-treatment with the extract and decursin dramatically attenuated the damage of the endfeet. In brief, therapeutic treatment of the extract of Angelica gigas Nakai root and decursin after 5 min transient forebrain ischemia protected hippocampal neurons from the ischemia, showing that ischemia-induced BBB leakage and damage of astrocyte endfeet was significantly attenuated by the extract and decursin. Based on these findings, we suggest that Angelica gigas Nakai root containing decursin can be employed as a pharmaceutical composition to develop a therapeutic strategy for brain ischemic injury.


Assuntos
Angelica/química , Astrócitos/patologia , Benzopiranos/uso terapêutico , Barreira Hematoencefálica/patologia , Butiratos/uso terapêutico , Ataque Isquêmico Transitório/patologia , Extratos Vegetais/uso terapêutico , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Benzopiranos/química , Benzopiranos/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Butiratos/química , Butiratos/farmacologia , Gerbillinae , Proteína Glial Fibrilar Ácida/metabolismo , Transportador de Glucose Tipo 1/metabolismo , Imunoglobulina G/metabolismo , Masculino , Neuraminidase/metabolismo , Fármacos Neuroprotetores/farmacologia , Fármacos Neuroprotetores/uso terapêutico , Extratos Vegetais/farmacologia , Padrões de Referência , Memória Espacial/efeitos dos fármacos
3.
Nutrients ; 12(8)2020 Aug 17.
Artigo em Inglês | MEDLINE | ID: mdl-32824513

RESUMO

Pycnogenol® (an extract of the bark of French maritime pine tree) is used for dietary supplement and known to have excellent antioxidative efficacy. However, there are few reports on neuroprotective effect of Pycnogenol® supplementation and its mechanisms against ischemic injury following transient forebrain ischemia (TFI) in gerbils. Now, we examined neuroprotective effect and its mechanisms of Pycnogenol® in the gerbils with 5-min TFI, which evokes a significant death (loss) of pyramidal cells located in the cornu ammonis (CA1) region of gerbil hippocampus from 4-5 days post-TFI. Gerbils were pretreated with 30, 40, and 50 mg/kg of Pycnogenol® once a day for 7 days before TFI surgery. Treatment with 50 mg/kg, not 30 or 40 mg/kg, of Pycnogenol® potently protected learning and memory, as well as CA1 pyramidal cells, from ischemic injury. Treatment with 50 mg/kg Pycnogenol® significantly enhanced immunoreactivity of antioxidant enzymes (superoxide dismutases and catalase) in the pyramidal cells before and after TFI induction. Furthermore, the treatment significantly reduced the generation of superoxide anion, ribonucleic acid oxidation and lipid peroxidation in the pyramidal cells. Moreover, interestingly, its neuroprotective effect was abolished by administration of sodium azide (a potent inhibitor of SODs and catalase activities). Taken together, current results clearly indicate that Pycnogenol® supplementation can prevent neurons from ischemic stroke through its potent antioxidative role.


Assuntos
Antioxidantes , Região CA1 Hipocampal/citologia , Suplementos Nutricionais , Flavonoides/administração & dosagem , Flavonoides/farmacologia , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Transtornos da Memória/etiologia , Transtornos da Memória/prevenção & controle , Fármacos Neuroprotetores , Extratos Vegetais/administração & dosagem , Extratos Vegetais/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/patologia , Animais , Catalase/metabolismo , Modelos Animais de Doenças , Gerbillinae , Peroxidação de Lipídeos/efeitos dos fármacos , Masculino , Células Piramidais/enzimologia , Superóxido Dismutase/metabolismo
4.
Mol Neurobiol ; 57(10): 4305-4321, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32700252

RESUMO

Buyang Huanwu Decoction (BHD), a classic traditional Chinese medicine (TCM) formula, has been used for recovering neurological dysfunctions and treating post-stroke disability in China for 200 years. In the present study, we investigated the effects of BHD on inhibiting neuronal apoptosis, promoting proliferation and differentiation of neural stem cells (NSCs) and neurite formation and enhancing learning and memory functional recovery in an experimental rat ischemic stroke model. BHD significantly reduced infarct volume and decreased cell apoptosis in the ischemic brain. BHD enhanced neuronal cell viability in vitro. BHD dose-dependently promoted the proliferation of NSCs in ischemic rat brains in vivo. Moreover, BHD promoted neuronal and astrocyte differentiation in primary cultured NSCs in vitro. Water maze test revealed that BHD promoted the recovery of learning function but not memory functions in the transient ischemic rats. We then investigated the changes of the cellular signaling molecules by using two-dimension (2D) gel electrophoresis and focused on the PI3K/Akt/Bad and Jak2/Stat3/cyclin D1signaling pathway to uncover its underlying mechanisms for its neuroprotective and neurogenetic effects. BHD significantly upregulated the expression of p-PI3K, p-Akt, and p-Bad as well as the expression of p-Jak, p-Stat3, and cyclin D1 in vitro and in vivo. In addition, BHD upregulated Hes1 and downregulated cav-1 in vitro and in vivo. Taken together, these results suggest that BHD has neuroprotective effects and neurogenesis-promoting effects via activating PI3K/Akt/Bad and Jak2/Stat3/Cyclin D1 signaling pathways. Graphical Abstract Buyang Huanwu Decoction (BHD) activates the PI3K-AKT-BAD pathway in the ischemic brain for neuroprotection. BHD also activates JAK2/STAT3/Cyclin D1 signaling cascades for promoting neurogenesis in the hippocampus of post-ischemic brains. Moreover, BHD inhibits the expression of caveolin-1 and increases the expression of HES1 for promoting neuronal differentiation. The neuroprotective and neurogenesis-promoting effects in the hippocampus of post-ischemic brains promote learning ability.


Assuntos
Medicamentos de Ervas Chinesas/uso terapêutico , Ataque Isquêmico Transitório/tratamento farmacológico , AVC Isquêmico/tratamento farmacológico , Neurogênese , Fármacos Neuroprotetores/uso terapêutico , Proteômica , Transdução de Sinais , Proteínas 14-3-3/metabolismo , Animais , Apoptose/efeitos dos fármacos , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Astrócitos/patologia , Axônios/patologia , Caveolina 1/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Ciclina D1/metabolismo , Regulação para Baixo/efeitos dos fármacos , Medicamentos de Ervas Chinesas/farmacologia , Receptores ErbB/metabolismo , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/fisiopatologia , AVC Isquêmico/complicações , AVC Isquêmico/patologia , AVC Isquêmico/fisiopatologia , Janus Quinase 2/metabolismo , Masculino , Memória/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/patologia , Neurite (Inflamação)/patologia , Neurogênese/efeitos dos fármacos , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Células PC12 , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Recuperação de Função Fisiológica/efeitos dos fármacos , Traumatismo por Reperfusão/complicações , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/patologia , Fator de Transcrição STAT3/metabolismo , Fatores de Transcrição HES-1/metabolismo , Regulação para Cima/efeitos dos fármacos , Xantenos/farmacologia , Proteína de Morte Celular Associada a bcl/metabolismo
5.
J Chem Neuroanat ; 101: 101683, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31499173

RESUMO

We previously found that fish oil (FO) facilitated memory recovery in the absence of pyramidal neuron rescue after transient, global cerebral ischemia (TGCI). Fish oil preserved the expression of microtubule-associated protein 2 (MAP-2), suggesting a relationship between dendritic plasticity and memory recovery that is mediated by FO after TGCI. The present study examined whether postischemic treatment with FO prevents ischemia-induced loss of dendritic processes in remaining pyramidal neurons. The effects of FO on neuroplasticity-related proteins were also examined after TGCI. Rats were subjected to TGCI (15 min, four-vessel occlusion model) and then received vehicle or FO (300 mg/kg docosahexaenoic acid) once daily for 7 days. The first dose was administered 4 h postischemia. Golgi-Cox staining was used to evaluate dentrict morphology in the pyramidal neurons of hippocampus (CA1 and CA3 subfields) and prefrontal cortex (PFC). Neuronal nuclei protein (NeuN), brain-derived neurotrophic factor (BDNF), growth-associated protein 43 (GAP-43), synaptophysin (SYP), and postsynaptic density protein 95 (PSD-95) levels were measured by Western blot in both structures. Fifteen minutes of TGCI reduced consistently the length of dendrites, number of dendritic branches and dendritic spine density (average of 25%, 43%, 32%, respectively) 7, 14, and 21 days postischemia, indicating that they did not recover spontaneously. This outcome of TGCI was reversed by FO treatment, an effect that was sustained even after treatment cessation. The NeuN and BDNF protein levels were reduced in both the hippocampus and PFC, which were recovered by FO treatment. GAP-43 protein levels decreased after ischemia in the PFC only, and this effect was also mitigated by FO. Neither SYP nor PSD-95 levels were altered by ischemia, but PDS-95 levels almost doubled after FO treatment in the ischemic group. These data support our hypothesis that synaptic plasticity at the level of dendrites may at least partially underlie the memory-protective effect of FO after TGCI and strengthen the possibility that FO has therapeutic potential for treating the sequelae of brain ischemia/reperfusion injury.


Assuntos
Dendritos/efeitos dos fármacos , Óleos de Peixe/farmacologia , Ataque Isquêmico Transitório/patologia , Plasticidade Neuronal/efeitos dos fármacos , Sinapses/efeitos dos fármacos , Animais , Dendritos/patologia , Ácidos Docosa-Hexaenoicos/farmacologia , Hipocampo/efeitos dos fármacos , Hipocampo/patologia , Masculino , Fármacos Neuroprotetores/farmacologia , Ratos , Ratos Wistar , Sinapses/patologia
6.
Toxicol Appl Pharmacol ; 377: 114613, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31207256

RESUMO

Realgar and cinnabar are commonly used mineral medicine containing arsenic and mercury in Traditional Chinese Medicine (TCM). Angong Niuhuang Wan (AGNHW) is a representative realgar- and cinnabar-containing TCM formula for treating acute ischemic stroke, but its toxicology and neuropharmacological effects are not well addressed. In this study, we compared the neuropharmacological effects of AGNHW and modified AGNHW in an experimental ischemic stroke rat model. Male SD rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) plus 22 h of reperfusion. Although oral administration of AGNHW for 7 days in the rats increased arsenic level in the blood and liver tissue, there were no significant changes in the arsenic level in kidney, mercury level in the blood, liver and kidney as well as hepatic and renal functions in MCAO rats. AGNHW revealed neuroprotective properties by reducing infarction volume, preserving blood-brain barrier integrity and improving neurological functions against cerebral ischemia-reperfusion injury. Interestingly, removing realgar and/or cinnabar from AGNHW abolished the neuroprotective effects. Meanwhile, AGNHW could scavenge peroxynitrite, down-regulate the expression of p47phox, 3-NT and MMP-9 and up-regulate the expression of ZO-1 and claudin-5 in the ischemic brains, which were abolished by removing realgar and/or cinnabar from AGNHW. Notably, realgar or cinnabar had no neuroprotection when used alone. Taken together, oral administration of AGNHW for one week should be safe for treating ischemic stroke with neuroprotective effects. Realgar and cinnabar are necessary elements with synergetic actions with other herbal materials for the neuroprotective effects of AGNHW against cerebral ischemia-reperfusion injury.


Assuntos
Arsenicais/química , Arsenicais/farmacologia , Produtos Biológicos/química , Produtos Biológicos/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Ataque Isquêmico Transitório/tratamento farmacológico , Nefropatias/induzido quimicamente , Nefropatias/patologia , Compostos de Mercúrio/química , Compostos de Mercúrio/farmacologia , Fármacos Neuroprotetores/farmacologia , Sulfetos/química , Sulfetos/farmacologia , Animais , Arsênio/sangue , Arsênio/metabolismo , Sequestradores de Radicais Livres/farmacologia , Infarto da Artéria Cerebral Média/patologia , Infarto da Artéria Cerebral Média/prevenção & controle , Ataque Isquêmico Transitório/patologia , Masculino , Medicina Tradicional Chinesa , Mercúrio/sangue , Mercúrio/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/prevenção & controle
7.
Neuroscience ; 410: 128-139, 2019 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-31095985

RESUMO

Transient ischemic attack (TIA) represents brief neurological dysfunction of vascular origin without detectable infarction. Despite major clinical relevance characterization of post-TIA molecular changes using appropriate experimental model is lacking and no therapeutic agent has been established yet. Neurosteroid dehydroepiandrosterone (DHEA) arose as one of the candidates for cerebral ischemia treatment but its effects on TIA-like condition remain unknown. Seeking an animal model applicable for investigation of molecular alterations in mild ischemic conditions such as TIA, 15-min bilateral common carotid artery occlusion with 24-h reperfusion was performed to induce ischemia/ reperfusion (I/R) injury in adult male Wistar rats. Additionally, effects of 4-h post-operative DHEA treatment (20 mg/kg) were investigated in physiological and I/R conditions in hippocampus (HIP) and prefrontal cortex (PFC). The study revealed absence of sensorimotor deficits, cerebral infarcts and neurodegeneration along with preserved HIP and PFC overall neuronal morphology and unaltered malondialdehyde and reduced glutathione level following I/R and/or DHEA treatment. I/R induced nitric oxide burst in HIP and PFC was accompanied with increased neuronal nitric oxide synthase protein level exclusively in HIP. DHEA had no effects in physiological conditions, while increase of Bax/Bcl2 ratio and dissipation of mitochondrial membrane potential in treated I/R group suggested DHEA-mediated exacerbation of post-ischemic changes that might lead to pro-apoptotic events in HIP. Interestingly, DHEA restored I/R-induced NO to the control level in PFC. Obtained results indicated that I/R may serve as an appropriate model for investigation of molecular changes and treatment outcome following mild ischemic conditions such as TIA.


Assuntos
Doenças das Artérias Carótidas/metabolismo , Artéria Carótida Primitiva/metabolismo , Desidroepiandrosterona/administração & dosagem , Mediadores da Inflamação/metabolismo , Ataque Isquêmico Transitório/metabolismo , Adjuvantes Imunológicos/administração & dosagem , Animais , Doenças das Artérias Carótidas/tratamento farmacológico , Doenças das Artérias Carótidas/patologia , Artéria Carótida Primitiva/efeitos dos fármacos , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Hipocampo/patologia , Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/patologia , Masculino , Córtex Pré-Frontal/efeitos dos fármacos , Córtex Pré-Frontal/metabolismo , Córtex Pré-Frontal/patologia , Ratos , Ratos Wistar , Resultado do Tratamento
8.
J Biol Regul Homeost Agents ; 32(5): 1143-1149, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30334406

RESUMO

Prior studies have demonstrated that ulinastatin (UTI) plays a beneficial role in regulating cerebral ischemic injury evoked by cardiac arrest (CA). It is noteworthy to find interventions that can enhance effects of this drug and thereby increase its clinical application. Xuebijing (XBJ) is comprised of extracts from Chinese herbs and has been widely used in China as an anti-endotoxicity drug for the treatment of sepsis and ischemic disorders associated with multiple organ dysfunction syndrome. Thus, in this study we examined the effects of a combination of UTI and XBJ to improve neural injury in the process of neurological functions after transient cerebral ischemia. Our results show that CA impaired Nrf2- antioxidant response element (Nrf2-ARE) and superoxide dismutase (SOD) in the hippocampus CA1 region. This process further amplified products of oxidative stress, namely 8-isoprostaglandin F2α (8-iso PGF2α) and 8-hydroxy-2'-deoxyguanosine (8-OHdG). A lower dose of UTI failed to restore Nrf2-ARE and attenuate 8-iso PGF2α and 8-OHdG SOD following CA; however, systemic administration of XBJ amplified the effects of this dose of UTI on antioxidative signal pathway of the hippocampus. Overall, the results of this study have implications for the enhanced neuroprotective role played by a combination of XBJ and UTI in improving neural injury observed in transient cerebral ischemia; and Nrf2-ARE signal is a part of key mechanisms that are involved in neuroprotective effects of XBJ and UTI.


Assuntos
Medicamentos de Ervas Chinesas/farmacologia , Glicoproteínas/farmacologia , Hipocampo/efeitos dos fármacos , Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fármacos Neuroprotetores/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sinergismo Farmacológico , Hipocampo/metabolismo , Ataque Isquêmico Transitório/patologia , Estresse Oxidativo/efeitos dos fármacos
9.
Chin J Nat Med ; 16(6): 428-435, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30047464

RESUMO

Artemisia capillaris Thunberg is a medicinal plant used as a traditional medicine in many cultures. It is an effective remedy for liver problems including hepatitis. Recent pharmacological reports have indicated that Artemisia species can exert various neurological effects. Previously, we reported a memory-enhancing effect of Artemisia species. However, the mechanisms underlying the neuroprotective effect of A. capillaris (AC) are still unknown. In the present study, we investigated the effect of an ethanol extract of AC on ischemic brain injury in a mouse model of transient forebrain ischemia. The mice were treated with AC for seven days, beginning one day before induction of transient forebrain ischemia. Behavioral deficits were investigated using the Y-maze. Nissl and Fluoro-jade B staining were used to indicate the site of injury. To determine the underlying mechanisms for the drug, we measured acetylcholinesterase activity. AC (200 mg·kg-1) treatment reduced transient forebrain ischemia-induced neuronal cell death in the hippocampal CA1 region. The AC-treated group also showed significant amelioration in the spontaneous alternation of the Y-maze test performance, compared to that in the untreated transient forebrain ischemia group. Moreover, AC treatment showed a concentration-dependent inhibitory effect on acetylcholinesterase activity in vitro. Finally, the effect of AC on forebrain ischemia was blocked by mecamylamine, a nonselective nicotinic acetylcholine receptor antagonist. Our results suggested that in a model of forebrain ischemia, AC protected against neuronal death through the activation of nicotinic acetylcholine receptors.


Assuntos
Artemisia , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/fisiopatologia , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Receptores Colinérgicos/metabolismo , Acetilcolinesterase/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Antagonistas Colinérgicos/farmacologia , Modelos Animais de Doenças , Etanol/química , Hipocampo/patologia , Hipocampo/fisiopatologia , Ataque Isquêmico Transitório/tratamento farmacológico , Masculino , Mecamilamina/farmacologia , Memória/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Modelos Neurológicos , Fármacos Neuroprotetores/administração & dosagem , Fitoterapia , Componentes Aéreos da Planta/química , Extratos Vegetais/administração & dosagem
10.
J Neurol ; 264(10): 2158-2161, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28894947

RESUMO

Spreading somatosensory symptoms appearing as Jacksonian sensory march are usually considered to be due to an epileptic seizure. We report on three cases in which these symptoms were caused by thalamic ischemia. Two patients presented with stereotypically recurring hemiparesthesias lasting 2-5 min that gradually spread from the face to the arm and leg on one side. A first cerebral magnetic resonance imaging including DWI was negative in both cases, whereas new thalamic infarctions appeared on repeated imaging when clinical symptoms remained. A third case with a thalamic ischemia did not show recurring events, but also presented with purely sensory spreading symptoms. In all three cases EEG and cardiovascular diagnostics revealed normal results. Pure sensory stroke has previously been described as a result of ischemia of the thalamus or the internal capsule presenting as a sudden onset hemisensory deficit, but spreading symptoms have rarely been reported. According to our observations, thalamic TIAs are an important differential diagnosis of somatosensory epileptic auras presenting with Jacksonian sensory march which require a different clinical management.


Assuntos
Ataque Isquêmico Transitório/patologia , Tálamo/diagnóstico por imagem , Idoso , Imagem de Difusão por Ressonância Magnética , Humanos , Processamento de Imagem Assistida por Computador , Masculino , Pessoa de Meia-Idade
11.
Physiol Behav ; 177: 196-207, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28483394

RESUMO

A series of our previous studies demonstrated that fish oil (FO), equivalent to 300mg/kg docosahexahenoic acid (DHA), facilitates memory recovery after transient, global cerebral ischemia (TGCI) in the aversive radial maze (AvRM). The present study sought to address two main issues: (i) whether the memory-protective effect of FO that has been observed in the AvRM can be replicated in the passive avoidance test (PAT) and object location test (OLT) and (ii) whether FO at doses that are lower than those used previously can also prevent TGCI-induced memory loss. In Experiment 1, naive rats were trained in the PAT, subjected to TGCI (4-vessel occlusion model), and tested for retrograde memory performance 8 and 15days after ischemia. Fish oil (300mg/kg/day DHA) was given orally for 8days. The first dose was delivered 4h postischemia. In Experiment 2, the rats were subjected to TGCI, treated with the same FO regimen, and then trained and tested in the OLT. In Experiment 3, the rats were trained in the AvRM, subjected to TGCI, administered FO (100, 200, and 300mg/kg DHA), and tested for memory performance up to 3weeks after TGCI. At the end of the behavioral tests, the brains were examined for neurodegeneration and neuroblast proliferation. All of the behavioral tests (PAT, OLT, and AvRM) were sensitive to ischemia, but only the AvRM was able to detect the memory-protective effect of FO. Ischemia-induced neurodegeneration and neuroblast proliferation were unaffected by FO treatment. These results suggest that (i) the beneficial effect of FO on memory recovery after TGCI is task-dependent, (ii) doses of FO<300mg/kg DHA can protect memory function in the radial maze, and (iii) cognitive recovery occurs in the absence of neuronal rescue and/or hippocampal neurogenesis.


Assuntos
Óleos de Peixe/farmacologia , Hipocampo/efeitos dos fármacos , Ataque Isquêmico Transitório/tratamento farmacológico , Transtornos da Memória/tratamento farmacológico , Fármacos Neuroprotetores/farmacologia , Recuperação de Função Fisiológica/efeitos dos fármacos , Animais , Antioxidantes/farmacologia , Aprendizagem da Esquiva/efeitos dos fármacos , Aprendizagem da Esquiva/fisiologia , Complexo Nuclear Basolateral da Amígdala/efeitos dos fármacos , Complexo Nuclear Basolateral da Amígdala/patologia , Modelos Animais de Doenças , Hipocampo/patologia , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/psicologia , Masculino , Transtornos da Memória/etiologia , Transtornos da Memória/patologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/etiologia , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/psicologia , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Neurônios/patologia , Ratos Wistar , Recuperação de Função Fisiológica/fisiologia , Memória Espacial/efeitos dos fármacos , Memória Espacial/fisiologia
12.
Mol Med Rep ; 16(1): 133-142, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28534982

RESUMO

Chrysanthemum indicum Linné extract (CIL) is used in herbal medicine in East Asia. In the present study, gerbils were orally pre­treated with CIL, and changes of antioxidant enzymes including superoxide dismutase (SOD) 1 and SOD2, catalase (CAT) and glutathione peroxidase (GPX) in the hippocampal CA1 region following 5 min of transient cerebral ischemia were investigated and the neuroprotective effect of CIL in the ischemic CA1 region was examined. SOD1, SOD2, CAT and GPX immunoreactivities were observed in the pyramidal cells of the CA1 region and their immunoreactivities were gradually decreased following ischemia­reperfusion and barely detectable at 5 days post­ischemia. CIL pre­treatment significantly increased immunoreactivities of SOD1, CAT and GPX, but not SOD2, in the CA1 pyramidal cells of the sham­operated animals. In addition, SOD1, SOD2, CAT and GPX immunoreactivities in the CA1 pyramidal cells were significantly higher compared with the ischemia­operated animals. Furthermore, it was identified that pre­treatment with CIL protected the CA1 pyramidal cells in the CA1 region using neuronal nuclei immunohistochemistry and Fluoro­Jade B histofluorescence staining; the protected CA1 pyramidal cells were 67.5% compared with the sham­operated animals. In conclusion, oral CIL pre­treatment increased endogenous antioxidant enzymes in CA1 pyramidal cells in the gerbil hippocampus and protected the cells from transient cerebral ischemic insult. This finding suggested that CIL is promising for the prevention of ischemia­induced neuronal damage.


Assuntos
Antioxidantes/metabolismo , Região CA1 Hipocampal/metabolismo , Chrysanthemum/química , Ataque Isquêmico Transitório/metabolismo , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Células Piramidais/efeitos dos fármacos , Células Piramidais/metabolismo , Animais , Biomarcadores , Catalase/metabolismo , Modelos Animais de Doenças , Gerbillinae , Glutationa Peroxidase/metabolismo , Imuno-Histoquímica , Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/patologia , Masculino , Superóxido Dismutase-1/metabolismo
13.
Transl Stroke Res ; 7(6): 548-561, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27714669

RESUMO

White matter injury induced by ischemic stroke elicits sensorimotor impairments, which can be further deteriorated by persistent proinflammatory responses. We previously reported that delayed and repeated treatments with omega-3 polyunsaturated fatty acids (n-3 PUFAs) improve spatial cognitive functions and hippocampal integrity after ischemic stroke. In the present study, we report a post-stroke n-3 PUFA therapeutic regimen that not only confers protection against neuronal loss in the gray matter but also promotes white matter integrity. Beginning 2 h after 60 min of middle cerebral artery occlusion (MCAO), mice were randomly assigned to receive intraperitoneal docosahexaenoic acid (DHA) injections (10 mg/kg, daily for 14 days), alone or in combination with dietary fish oil (FO) supplements starting 5 days after MCAO. Sensorimotor functions, gray and white matter injury, and microglial responses were examined up to 28 days after MCAO. Our results showed that DHA and FO combined treatment-facilitated long-term sensorimotor recovery and demonstrated greater beneficial effect than DHA injections alone. Mechanistically, n-3 PUFAs not only offered direct protection on white matter components, such as oligodendrocytes, but also potentiated microglial M2 polarization, which may be important for white matter repair. Notably, the improved white matter integrity and increased M2 microglia were strongly linked to the mitigation of sensorimotor deficits after stroke upon n-3 PUFA treatments. Together, our results suggest that post-stroke DHA injections in combination with FO dietary supplement benefit white matter restoration and microglial responses, thereby dictating long-term functional improvements.


Assuntos
Ácido Eicosapentaenoico/farmacologia , Ácido Eicosapentaenoico/uso terapêutico , Ataque Isquêmico Transitório/tratamento farmacológico , Leucoencefalopatias/etiologia , Microglia/efeitos dos fármacos , Animais , Antígenos CD/metabolismo , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Circulação Cerebrovascular/efeitos dos fármacos , Corpo Caloso/patologia , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/uso terapêutico , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas dos Microfilamentos/metabolismo , Proteína Básica da Mielina/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Doenças do Sistema Nervoso/tratamento farmacológico , Doenças do Sistema Nervoso/etiologia , Acidente Vascular Cerebral/complicações , Fatores de Tempo
14.
Brain Res ; 1648(Pt A): 257-265, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27431936

RESUMO

Ischemic preconditioning with sublethal stress triggers defensive mechanisms against ischemic brain damage; however, such manipulations are potentially dangerous and, therefore, safe stimuli have been sought. Hyperoxia preconditioning by administration of hyperbaric (HBO) or normobaric oxygen (NBO) may have neuroprotective potential. The aim of this study was to determine whether preconditioning with HBO and air (HBA) applied at 2.5 absolute pressure (ATA) or NBO preconditioning induces ischemic tolerance in the brain of gerbils subjected to 3min transient cerebral ischemia. Neuronal cell survival, changes in brain temperature, the generation of factors involved in neurodegeneration and basic behavior in nest building were all tested. Hyperoxic preconditioning prevented ischemia-induced neuronal cell loss, reduced the number of TUNEL positive cells in the CA1 region of the hippocampus and improved the nest building process compared to untreated ischemic animals. Preconditioning also suppressed the production of reactive oxygen species and increased Bax expression normally observed after an ischemic episode. Only HBO preconditioning inhibited ischemia-evoked increases in brain temperature. Our results show that hyperoxic preconditioning results in induction of ischemic tolerance and prevents ischemia-induced neuronal damage in the gerbil brain. Pressurized air preconditioning was as effective as HBO or NBO preconditioning in providing neuroprotection. The observed neuroprotection probably results from mild oxidative stress evoked by increased brain tissue oxidation and activation of antioxidant and antiapoptotic defenses.


Assuntos
Oxigenoterapia Hiperbárica/métodos , Ataque Isquêmico Transitório/prevenção & controle , Precondicionamento Isquêmico/métodos , Animais , Apoptose , Temperatura Corporal , Encéfalo/metabolismo , Sobrevivência Celular , Gerbillinae , Hipocampo/patologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Comportamento de Nidação , Prosencéfalo/fisiopatologia , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Espécies Reativas de Oxigênio , Proteína X Associada a bcl-2/metabolismo
15.
Cell Physiol Biochem ; 38(5): 1952-62, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27161043

RESUMO

BACKGROUND/AIMS: Glycine is a strychnine-sensitive inhibitory neurotransmitter in the central nervous system (CNS), especially in the spinal cord, brainstem, and retina. The objective of the present study was to investigate the potential neuroprotective effects of GlyT1 inhibitor N [3-(4'-fluorophenyl)-3-(4'-phenylphenoxy) propyl] sarcosine (NFPS) in the rat model of experimental stroke. METHODS: In vivo ischaemia was induced by transient middle cerebral artery occlusion (tMCAO). The methods of Western Blotting, Nissl Staining and Morris water maze methods were applied to analyze the anti-ischaemia mechanism. RESULTS: The results showed that high dose of NFPS (H-NFPS) significantly reduced infarct volume, neuronal injury and the expression of cleaved caspase-3, enhanced Bcl-2/Bax, and improved spatial learning deficits which were administered three hours after transient middle cerebral artery occlusion (tMCAO) induction in rats, while, low dose of NFPS (L-NFPS) exacerbated the injury of ischaemia. These findings suggested that low and high dose of NFPS produced opposite effects. Importantly, it was demonstrated that H-NFPS-dependent neuronal protection was inverted by salicylate (Sal), a specific GlyR x0251;1 antagonist. Such effects could probably be attributed to the enhanced glycine level in both synaptic and extrasynaptic clefts and the subsequently altered extrasynaptic GlyRs and their subtypes. CONCLUSIONS: These data imply that GlyT1 inhibitor NFPS may be a novel target for clinical treatment of transient focal cerebral ischaemia and reperfusion which are associated with altered GlyR alpha 1 subunits.


Assuntos
Proteínas da Membrana Plasmática de Transporte de Glicina/antagonistas & inibidores , Ataque Isquêmico Transitório/patologia , Fármacos Neuroprotetores/farmacologia , Receptores de Glicina/metabolismo , Sarcosina/análogos & derivados , Animais , Western Blotting , Encéfalo/patologia , Caspase 3/metabolismo , Modelos Animais de Doenças , Glicina/metabolismo , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Imuno-Histoquímica , Infarto da Artéria Cerebral Média/complicações , Ataque Isquêmico Transitório/etiologia , Ataque Isquêmico Transitório/metabolismo , Masculino , Aprendizagem em Labirinto , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Receptores de Glicina/antagonistas & inibidores , Salicilatos/farmacologia , Sarcosina/farmacologia , Proteína X Associada a bcl-2/metabolismo
16.
Behav Brain Res ; 311: 425-439, 2016 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-27235715

RESUMO

We reported that fish oil (FO) prevented the loss of spatial memory caused by transient, global cerebral ischemia (TGCI), provided the treatment covered the first days prior to and after ischemia. Continuing these studies, trained rats were subjected to TGCI, and FO was administered for 10days, with a time window of efficacy (TWE) of 4, 8 or 12h post-ischemia. Retrograde memory was assessed up to 43days after TGCI. In another experiment, ischemic rats received FO with a 4- or 12-h TWE, and dendritic density was assessed in the hippocampus and cerebral cortex. The brain lipid profile was evaluated in sham-operated and ischemic rats that were treated with FO or vehicle with a 4-h TWE. Ischemia-induced retrograde amnesia was prevented by FO administration that was initiated with either a 4- or 8-h TWE. Fish oil was ineffective after a 12-h TWE. Independent of the TWE, FO did not prevent ischemic neuronal death. In the hippocampus, but not cerebral cortex, TGCI-induced dendritic loss was prevented by FO with a 4-h TWE but not 12-h TWE. The level of docosahexaenoic acid almost doubled in the hippocampus in ischemic, FO-treated rats (4-h TWE). The data indicate that (i) the anti-amnesic effect of FO can be observed with a TWE of up to 8h, (ii) the stimulation of dendritic neuroplasticity may have contributed to this effect, and (iii) DHA in FO may be the main active constituent in FO that mediates the cognitive and neuroplasticity effects on TGCI.


Assuntos
Dendritos/efeitos dos fármacos , Óleos de Peixe/administração & dosagem , Hipocampo/efeitos dos fármacos , Ataque Isquêmico Transitório/tratamento farmacológico , Memória de Longo Prazo/efeitos dos fármacos , Fármacos Neuroprotetores/administração & dosagem , Amnésia Retrógrada/tratamento farmacológico , Amnésia Retrógrada/etiologia , Amnésia Retrógrada/metabolismo , Amnésia Retrógrada/patologia , Animais , Córtex Cerebral/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Dendritos/metabolismo , Dendritos/patologia , Modelos Animais de Doenças , Ácidos Docosa-Hexaenoicos/metabolismo , Hipocampo/metabolismo , Hipocampo/patologia , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Ataque Isquêmico Transitório/psicologia , Masculino , Memória de Longo Prazo/fisiologia , Doenças Neurodegenerativas/tratamento farmacológico , Doenças Neurodegenerativas/metabolismo , Doenças Neurodegenerativas/patologia , Doenças Neurodegenerativas/psicologia , Plasticidade Neuronal/efeitos dos fármacos , Plasticidade Neuronal/fisiologia , Nootrópicos/administração & dosagem , Ratos Wistar , Fatores de Tempo
17.
Mol Med Rep ; 13(4): 2957-66, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26936330

RESUMO

Ilexonin A is a compound isolated from the root of a plant commonly used in traditional Chinese medicine. The aim of the present study was to investigate the possible protective mechanism of Ilexonin A in rats subjected to occlusion of the middle cerebral artery (MCAO). Transient focal cerebral ischemia was induced by 2 h of MCAO, followed by reperfusion. Ilexonin A at doses of 20, 40 and 80 mg/kg were administered via intraperitoneal injection immediately following ischemia/reperfusion. The expression levels of glial fibrillary acidic protein (GFAP), ionized calcium­binding adapter molecule­1 (Iba­1), vascular endothelial growth factor (VEGF), fetal liver kinase­1 (Flk­1) and Nestin were examined using immunostaining and Western blot analysis of the peri­infarct region following ischemia/reperfusion. Ilexonin A significantly decreased the infarct volume and improved neurological deficits in a dose­dependent manner. The expression levels of VEGF, Flk­1 and Nestin were significantly increased in the rats treated with Ilexonin A, compared with the rats administered with saline. Following treatment with Ilexonin A, a higher number of GFAP­positive astrocytes were found in the Ilexonin A­treated rats at 1, 3 and 7 days, compared with the rats exposed to ischemia only, however, there were fewer astrocytes at 14 days, compared with the ischemia group. Ilexonin A significantly decreased the protein expression of Iba­1. The results of the present study suggested that the protective effects of Ilexonin A were associated with revascularization, neuronal regeneration, and the regulation of astrocyte and microglia cell activation.


Assuntos
Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Fármacos Neuroprotetores/farmacologia , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Infarto Encefálico/metabolismo , Infarto Encefálico/patologia , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Proteína Glial Fibrilar Ácida/metabolismo , Ataque Isquêmico Transitório/tratamento farmacológico , Ataque Isquêmico Transitório/fisiopatologia , Masculino , Microglia/metabolismo , Microglia/patologia , Fármacos Neuroprotetores/química , Compostos Orgânicos/química , Compostos Orgânicos/farmacologia , Ratos , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Traumatismo por Reperfusão/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Neuroscience ; 322: 66-77, 2016 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-26873001

RESUMO

BACKGROUND AND PURPOSE: The initial steps in the cascade leading to cell death are still unknown because of the limitations of the existing methodology, strategy, and modalities used. METHODS: Imaging mass spectrometry (IMS) was used to measure dynamic molecular changes of phosphatidylcholine (PC) species in the rat hippocampus after transient global ischemia (TGI) for 6min. Fresh frozen sections were obtained after euthanizing the rats on Days 1, 2, 4, 7, 10, 14, and 21. Histopathology and IMS of adjacent sections compared morphological and molecular changes, respectively. RESULTS: Histopathological changes were absent immediately after TGI (at Day 1, superacute phase). At Days 2-21 after TGI (from subacute to chronic phases), histopathology revealed neuronal death associated with gliosis, inflammation, and accumulation of activated microglia in CA1. IMS detected significant molecular changes after TGI in the same CA1 domain: increase of PC (diacyl-16:0/22:6) in the superacute phase and increase of PC (diacyl-16:0/18:1) in the subacute to chronic phases. CONCLUSIONS: Histopathology and IMS can provide comprehensive and complementary information on cell death mechanisms in the hippocampal CA1 after global ischemia. IMS provided novel data on molecular changes in phospholipids immediately after TGI. Increased level of PC (diacyl-16:0/22:6) in the pyramidal cell layer of hippocampal CA1 prior to the histopathological change may represent an early step in delayed neuronal death mechanisms.


Assuntos
Região CA1 Hipocampal/metabolismo , Ataque Isquêmico Transitório/metabolismo , Espectrometria de Massas/métodos , Fosfatidilcolinas/metabolismo , Doença Aguda , Animais , Astrócitos/metabolismo , Astrócitos/patologia , Região CA1 Hipocampal/patologia , Morte Celular/fisiologia , Doença Crônica , Modelos Animais de Doenças , Progressão da Doença , Gliose/metabolismo , Gliose/patologia , Imuno-Histoquímica , Ataque Isquêmico Transitório/patologia , Masculino , Microglia/metabolismo , Microglia/patologia , Ratos Sprague-Dawley , Fatores de Tempo
19.
Neuroreport ; 27(1): 23-32, 2016 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-26555034

RESUMO

Glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonists have been shown to be neuroprotective in previous studies in animal models of Alzheimer's or Parkinson's disease. Recently, novel dual-GLP-1/GIP receptor agonists that activate both receptors (DA) were developed to treat diabetes. We tested the protective effects of a novel potent DA against middle cerebral artery occlusion injury in rats and compared it with a potent GLP-1 analog, Val(8)-GLP-1(glu-PAL). Animals were evaluated for neurologic deficit score, infarct volume, and immunohistochemical analyses of the brain at several time points after ischemia. The Val(8)-GLP-1(glu-PAL)-treated and DA-treated groups showed significantly reduced scores of neurological dysfunction, cerebral infarction size, and percentage of TUNEL-positive apoptotic neurons. Furthermore, the expression of the apoptosis marker Bax, the inflammation marker iNOS, and the survival marker Bcl-2 was significantly increased. The DA-treated group was better protected against neurodegeneration than the Val(8)-GLP-1(glu-PAL) group, and the scores of neurological dysfunction, cerebral infarction size, and expression of Bcl-2 were higher, whereas the percentage of TUNEL-positive neurons and the levels of Bax and iNOS were lower in the DA group. DA treatment reduced the infarct volume and improved the functional deficit. It also suppressed the inflammatory response and cell apoptosis after reperfusion. In conclusion, the novel GIP and GLP-1 dual-receptor agonist is more neuroprotective than a GLP-1 receptor agonist in key biomarkers of neuronal degeneration.


Assuntos
Encéfalo/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/análogos & derivados , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Ataque Isquêmico Transitório/tratamento farmacológico , Lipopeptídeos/farmacologia , Receptores dos Hormônios Gastrointestinais/agonistas , Animais , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Infarto da Artéria Cerebral Média , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/metabolismo , Ataque Isquêmico Transitório/patologia , Masculino , Atividade Motora/efeitos dos fármacos , Degeneração Neural/tratamento farmacológico , Degeneração Neural/etiologia , Degeneração Neural/metabolismo , Degeneração Neural/patologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Neurônios/patologia , Fármacos Neuroprotetores/farmacologia , Óxido Nítrico Sintase Tipo II/metabolismo , Distribuição Aleatória , Ratos Sprague-Dawley , Receptores dos Hormônios Gastrointestinais/metabolismo , Proteína X Associada a bcl-2/metabolismo
20.
Nutrition ; 31(7-8): 1008-17, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26059376

RESUMO

OBJECTIVE: The aim of this study was to evaluate the possible beneficial effects of Mentat against transient global ischemia and reperfusion-induced brain injury in rats. METHODS: The neuroprotective effects of Mentat were evaluated against transient global ischemia and reperfusion (I/R)-induced brain injury in rats. Various neurobehavioral and biochemical parameters were assessed, followed by morphologic and histopathologic evaluation of brain tissue to conclude the protective effect of Mentat. Additionally, in vitro antioxidant assays were performed to explore the antioxidant capacity of Mentat and detailed liquid chromatography-mass spectrometry (LC-MS/MS) profiling was carried out to identify the active phytoconstituents responsible for the protective effects of Mentat. RESULTS: Sixty minutes of transient global ischemia followed by 24 h reperfusion (I/R) caused significant alterations in the cognitive and neurologic functions in the ischemia control group (P < 0.01) compared with the sham control. Furthermore, 2,3,5-triphenyltetrazolium chloride staining of the ischemia control group showed 20.85% ± 0.39% of cerebral infarct area (P < 0.01), increased brain volume (% edema 17.81% ± 1.576%; P < 0.01), and increased lipid peroxidation (P < 0.01) in the brain homogenate. Additionally, the histopathology of the ischemia control group showed severe brain injury compared with the sham control group. Interestingly, pretreatment with Mentat (250 and 500 mg/kg, p.o.) and quercetin (20 mg/kg, p.o.) for 7 d has alleviated all pathological changes observed due to I/R injury. Mentat also showed very good antioxidant activity in in vitro assays (2,2-diphenyl-l-picrylhydrazyl, ferric-reducing antioxidant power, and oxygen radical absorbance capacity assays). Furthermore, the detailed LC-MS/MS analysis of Mentat was performed and enclosed for identifying the actives responsible for its protective effects. CONCLUSIONS: These findings suggest that Mentat is a neuroprotective agent that may be a useful adjunct in the management of ischemic stroke and its rehabilitation especially with respect to associated memory impairment and other related neurologic conditions.


Assuntos
Infarto Cerebral/prevenção & controle , Ataque Isquêmico Transitório/tratamento farmacológico , Peroxidação de Lipídeos/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Extratos Vegetais/farmacologia , Traumatismo por Reperfusão/prevenção & controle , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Edema Encefálico/tratamento farmacológico , Edema Encefálico/etiologia , Infarto Cerebral/etiologia , Infarto Cerebral/patologia , Cromatografia Líquida/métodos , Modelos Animais de Doenças , Ataque Isquêmico Transitório/complicações , Ataque Isquêmico Transitório/patologia , Masculino , Espectrometria de Massas/métodos , Fármacos Neuroprotetores/administração & dosagem , Extratos Vegetais/administração & dosagem , Quercetina/administração & dosagem , Quercetina/farmacologia , Ratos , Ratos Wistar , Traumatismo por Reperfusão/etiologia , Traumatismo por Reperfusão/patologia , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA