Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
J Neuroinflammation ; 20(1): 50, 2023 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-36829205

RESUMO

BACKGROUND: The repair of white matter injury is of significant importance for functional recovery after ischemic stroke, and the up-regulation of triggering receptors expressed on myeloid cells 2 (TREM2) after ischemic stroke is neuroprotective and implicated in remyelination. However, the lack of effective therapies calls for the need to investigate the regenerative process of remyelination and the role of rehabilitation therapy. This study sought to investigate whether and how moderate physical exercise (PE) promotes oligodendrogenesis and remyelination in rats with transient middle cerebral artery occlusion (tMCAO). METHODS: Male Sprague-Dawley rats (weighing 250-280 g) were subjected to tMCAO. AAV-shRNA was injected into the lateral ventricle to silence the Trem2 gene before the operation. The rats in the physical exercise group started electric running cage training at 48 h after the operation. The Morris water maze and novel object recognition test were used to evaluate cognitive function. Luxol fast blue staining, diffusion tensor imaging, and electron microscopy were used to observe myelin injury and repair. Immunofluorescence staining was applied to observe the proliferation and differentiation of oligodendrocyte precursor cells (OPCs). Expression of key molecules were detected using immunofluorescence staining, quantitative real-time polymerase chain reaction, Western blotting, and Enzyme-linked immunosorbent assay, respectively. RESULTS: PE exerted neuroprotective efects by modulating microglial state, promoting remyelination and recovery of neurological function of rats over 35 d after stroke, while silencing Trem2 expression in rats suppressed the aforementioned effects promoted by PE. In addition, by leveraging the activin-A neutralizing antibody, we found a direct beneficial effect of PE on microglia-derived activin-A and its subsequent role on oligodendrocyte differentiation and remyelination mediated by the activin-A/Acvr axis. CONCLUSIONS: The present study reveals a novel regenerative role of PE in white matter injury after stroke, which is mediated by upregulation of TREM2 and microglia-derived factor for oligodendrocytes regeneration. PE is an effective therapeutic approach for improving white matter integrity and alleviating neurological function deficits after ischemic stroke.


Assuntos
Lesões Encefálicas , Isquemia Encefálica , AVC Isquêmico , Acidente Vascular Cerebral , Substância Branca , Ratos , Masculino , Animais , Microglia/metabolismo , Substância Branca/metabolismo , AVC Isquêmico/metabolismo , Isquemia Encefálica/metabolismo , Imagem de Tensor de Difusão , Ratos Sprague-Dawley , Acidente Vascular Cerebral/metabolismo , Infarto da Artéria Cerebral Média/metabolismo , Lesões Encefálicas/metabolismo
2.
Arch Phys Med Rehabil ; 99(5): 1011-1022.e1, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29357280

RESUMO

OBJECTIVE: To investigate the effectiveness of neuromuscular electrical stimulation (NMES) with or without other interventions in improving lower limb activity after chronic stroke. DATA SOURCES: Electronic databases, including PubMed, EMBase, Cochrane Library, PEDro (Physiotherapy Evidence Database), and PsycINFO, were searched from the inception to January 2017. STUDY SELECTION: We selected the randomized controlled trials (RCTs) involving chronic stroke survivors with lower limb dysfunction and comparing NMES or combined with other interventions with a control group of no electrical stimulation treatment. DATA EXTRACTION: The primary outcome was defined as lower limb motor function, and the secondary outcomes included gait speed, Berg Balance Scale, timed Up and Go, 6-minute walk test, Modified Ashworth Scale, and range of motion. DATA SYNTHESIS: Twenty-one RCTs involving 1481 participants were identified from 5759 retrieved articles. Pooled analysis showed that NMES had a moderate but statistically significant benefit on lower limb motor function (standard mean difference 0.42, 95% confidence interval 0.26-0.58), especially when NMES was combined with other interventions or treatment time within either 6 or 12 weeks. NMES also had significant benefits on gait speed, balance, spasticity, and range of motion but had no significant difference in walking endurance after NMES. CONCLUSIONS: NMES combined with or without other interventions has beneficial effects in lower limb motor function in chronic stroke survivors. These data suggest that NMES should be a promising therapy to apply in chronic stroke rehabilitation to improve the capability of lower extremity in performing activities.


Assuntos
Terapia por Estimulação Elétrica/métodos , Hemiplegia/reabilitação , Reabilitação do Acidente Vascular Cerebral/métodos , Acidente Vascular Cerebral/complicações , Doença Crônica , Hemiplegia/etiologia , Hemiplegia/fisiopatologia , Humanos , Extremidade Inferior/fisiopatologia , Modalidades de Fisioterapia , Amplitude de Movimento Articular , Recuperação de Função Fisiológica , Acidente Vascular Cerebral/fisiopatologia , Resultado do Tratamento , Teste de Caminhada , Caminhada/fisiologia , Velocidade de Caminhada
3.
Med Sci Monit ; 23: 6072-6081, 2017 Dec 23.
Artigo em Inglês | MEDLINE | ID: mdl-29274273

RESUMO

BACKGROUND The role of nicotinic acetylcholine receptor alpha7 subunit (a7nAchR) in the treatment of acute cerebral ischemia by VNS has not been thoroughly clarified to date. Therefore, this study aimed to investigate the specific role of a7nAchR and explore whether this process is involved in the mechanisms of VNS-induced neuroprotection in rats undergoing permanent middle cerebral artery occlusion (PMCAO) surgery. MATERIAL AND METHODS Rats received a7nAChR antagonist (A) or antagonist placebo injection for control (AC), followed by PMCAO and VNS treatment, whereas the a7nAChR agonist (P) was utilized singly without VNS treatment but only with PMCAO pretreatment. The rats were randomly divided into 6 groups: sham PMCAO, PMCAO, PMCAO+VNS, PMCAO+VNS+A, PMCAO+VNS+AC, and PMCAO+P. Neurological function and cerebral infarct volume were measured to evaluate the level of brain injury at 24 h after PMCAO or PMCAO-sham. Moreover, the related proteins levels of a7nAChR, p-JAK2, and p-STAT3 in the ischemic penumbra were assessed by Western blot analysis. RESULTS Rats pretreated with VNS had significantly improved neurological function and reduced cerebral infarct volume after PMCAO injury (p<0.05). In addition, VNS enhanced the levels of a7nAchR, p-JAK2, and p-STAT3 in the ischemic penumbra (p<0.05). However, inhibition of a7nAchR not only attenuated the beneficial neuroprotective effects induced by VNS, but also decreased levels of p-JAK2 and p-STAT3. Strikingly, pharmacological activation of a7nAchR can partially substitute for VNS-induced beneficial neurological protection. CONCLUSIONS These results suggest that a7nAchR is a pivotal mediator of VNS-induced neuroprotective effects on PMCAO injury, which may be related to suppressed inflammation via activation of the a7nAchR/JAK2 anti-inflammatory pathway.


Assuntos
Isquemia Encefálica/terapia , Janus Quinase 2/metabolismo , Estimulação do Nervo Vago/métodos , Receptor Nicotínico de Acetilcolina alfa7/metabolismo , Animais , Lesões Encefálicas/tratamento farmacológico , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Modelos Animais de Doenças , Infarto da Artéria Cerebral Média/metabolismo , Infarto da Artéria Cerebral Média/cirurgia , Inflamação/tratamento farmacológico , Masculino , Fármacos Neuroprotetores/uso terapêutico , Ratos , Ratos Sprague-Dawley , Fator de Transcrição STAT3/metabolismo , Nervo Vago/metabolismo , Receptor Nicotínico de Acetilcolina alfa7/agonistas , Receptor Nicotínico de Acetilcolina alfa7/antagonistas & inibidores
4.
Neural Regen Res ; 19(8): 1772-1780, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-38103244

RESUMO

JOURNAL/nrgr/04.03/01300535-202408000-00031/figure1/v/2023-12-16T180322Z/r/image-tiff Proliferation of neural stem cells is crucial for promoting neuronal regeneration and repairing cerebral infarction damage. Transcranial magnetic stimulation (TMS) has recently emerged as a tool for inducing endogenous neural stem cell regeneration, but its underlying mechanisms remain unclear. In this study, we found that repetitive TMS effectively promotes the proliferation of oxygen-glucose deprived neural stem cells. Additionally, repetitive TMS reduced the volume of cerebral infarction in a rat model of ischemic stroke caused by middle cerebral artery occlusion, improved rat cognitive function, and promoted the proliferation of neural stem cells in the ischemic penumbra. RNA-sequencing found that repetitive TMS activated the Wnt signaling pathway in the ischemic penumbra of rats with cerebral ischemia. Furthermore, PCR analysis revealed that repetitive TMS promoted AKT phosphorylation, leading to an increase in mRNA levels of cell cycle-related proteins such as Cdk2 and Cdk4. This effect was also associated with activation of the glycogen synthase kinase 3ß/ß-catenin signaling pathway, which ultimately promotes the proliferation of neural stem cells. Subsequently, we validated the effect of repetitive TMS on AKT phosphorylation. We found that repetitive TMS promoted Ca2+ influx into neural stem cells by activating the P2 calcium channel/calmodulin pathway, thereby promoting AKT phosphorylation and activating the glycogen synthase kinase 3ß/ß-catenin pathway. These findings indicate that repetitive TMS can promote the proliferation of endogenous neural stem cells through a Ca2+ influx-dependent phosphorylated AKT/glycogen synthase kinase 3ß/ß-catenin signaling pathway. This study has produced pioneering results on the intrinsic mechanism of repetitive TMS to promote neural function recovery after ischemic stroke. These results provide a strong scientific foundation for the clinical application of repetitive TMS. Moreover, repetitive TMS treatment may not only be an efficient and potential approach to support neurogenesis for further therapeutic applications, but also provide an effective platform for the expansion of neural stem cells.

5.
Exp Neurol ; 364: 114380, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-36914085

RESUMO

BACKGROUND: Neuroinflammation and oxidative stress are important pathological mechanisms underlying cerebral ischemic stroke. Increasing evidence suggests that regulation autophagy in ischemic stroke may improve neurological functions. In this study, we aimed to explore whether exercise pretreatment attenuates neuroinflammation and oxidative stress in ischemic stroke by improving autophagic flux. METHODS: 2,3,5-Triphenyltetrazolium chloride staining was used to determine the infarction volume, and modified Neurological Severity Scores and rotarod test were used to evaluate neurological functions after ischemic stroke. The levels of oxidative stress, neuroinflammation, neuronal apoptosis and degradation, autophagic flux, and signaling pathway proteins were determined using immunofluorescence, dihydroethidium, TUNEL, and Fluoro-Jade B staining, western blotting, and co-immunoprecipitation. RESULTS: Our results showed that, in middle cerebral artery occlusion (MCAO) mice, exercise pretreatment improved neurological functions and defective autophagy, and reduced neuroinflammation and oxidative stress. Mechanistically, after using chloroquine, impaired autophagy abolished the neuroprotection of exercise pretreatment. And transcription factor EB (TFEB) activation mediated by exercise pretreatment contributes to improving autophagic flux after MCAO. Furthermore, we showed that TFEB activation mediated by exercise pretreatment in MCAO was regulated by the AMPK-mTOR and AMPK-FOXO3a-SKP2-CARM1 signaling pathways. CONCLUSIONS: Exercise pretreatment has the potential to improve the prognosis of ischemic stroke patients, and it can exert neuroprotective effects in ischemic stroke by inhibiting neuroinflammation and oxidative stress, which might be due to the TFEB-mediated autophagic flux. And targeting autophagic flux may be promising strategies for the treatment of ischemic stroke.


Assuntos
AVC Isquêmico , Acidente Vascular Cerebral , Camundongos , Animais , Doenças Neuroinflamatórias , Proteínas Quinases Ativadas por AMP , Autofagia/fisiologia , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/patologia , Estresse Oxidativo
6.
Front Psychiatry ; 13: 864481, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35573384

RESUMO

Increasing evidence indicates that inflammatory responses may influence brain neurochemical pathways, inducing depressive-like behaviors. Ultrasound stimulation (US) is a promising non-invasive treatment for neuropsychiatric diseases. We investigated whether US can suppress inflammation and improve depressive-like behaviors. Mice were intraperitoneally injected with lipopolysaccharide to induce depressive-like behaviors. Ultrasound wave was delivered into the prefrontal cortex (PFC) for 30 min. Depressive- and anxiety-like behaviors were evaluated through the forced swimming test (FST), tail suspension test (TST), and elevated plus maze (EPM). Biochemical analyses were performed to assess the expression of inflammatory cytokines in the PFC and serum. The results indicated that US of the PFC significantly improved depressive-like behaviors in the TST (p < 0.05) and FST (p < 0.05). Anxiety-like behaviors also improved in the EPM (p < 0.05). Furthermore, the lipopolysaccharide-mediated upregulation of IL-6, IL-1ß, and TNF-α in the PFC was significantly reduced (p < 0.05) by US. In addition, no tissue damage was observed. Overall, US of PFC can effectively improve lipopolysaccharide-induced depressive-like behaviors, possibly through the downregulation of inflammatory cytokines in the PFC. US may be a safe and promising tool for improvement of depression.

7.
Neurorehabil Neural Repair ; 35(8): 680-691, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34032160

RESUMO

The effects of low-frequency repetitive transcranial magnetic stimulation (LF-rTMS) on treating poststroke aphasia (PSA) remain inconclusive. We aimed to evaluate the efficacy and safety of LF-rTMS on language function poststroke and determine potential factors that may affect treatment effects. Electronic databases, including MEDLINE, EMBASE, and Cochrane Library were searched to identify relevant randomized controlled trials (RCTs) concerning the effects of LF-rTMS on language performance poststroke. We adopted fixed- and random-effects models to estimate intervention effects, which were represented by the Hedges' g and 95% CIs. Subgroup analyses regarding several factors potentially influencing the effects of LF-rTMS on language recovery were also conducted. A total of 14 RCTs involving 374 participants were included in the meta-analysis. The pooled analysis showed the positive and significant effects of LF-rTMS on language function, both short-term (Hedges' g = 0.65; P < .05) and long-term (Hedges' g = 0.46; P < .05). Subgroup analyses demonstrated that LF-rTMS for 20 minutes per day over 10 days yielded the largest effect size (Hedges' g = 1.02; P < .05) and that LF-rTMS significantly improved language performance in the chronic stage after stroke (Hedges' g = 0.55; P < .05). Patients with different native languages might have diverse responses to LF-rTMS treatment efficacy. Additionally, there were significant improvements in language subtests, including naming, repetition, comprehension, and writing. Overall, this updated meta-analysis demonstrated that LF-rTMS has significant positive effects on PSA, with moderate treatment effects. It provides additional evidence to support LF-rTMS as a promising complementary therapy to promote language recovery in PSA.


Assuntos
Afasia/terapia , Idioma , Reabilitação do Acidente Vascular Cerebral/métodos , Acidente Vascular Cerebral/complicações , Estimulação Magnética Transcraniana/métodos , Afasia/etiologia , Afasia/fisiopatologia , Humanos , Acidente Vascular Cerebral/fisiopatologia , Resultado do Tratamento
8.
Front Cell Neurosci ; 14: 593130, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33324166

RESUMO

Background: Exosomes, especially stem cell-derived exosomes, have been widely studied in pre-clinical research of ischemic stroke. However, their pooled effects remain inconclusive. Methods: Relevant literature concerning the effects of exosomes on neurological performance in a rodent model of ischemic stroke was identified via searching electronic databases, including PubMed, Embase, and Web of Science. The primary outcomes included neurological function scores (NFS) and infarct volume (IV), and the secondary outcomes were several pro-inflammatory factors and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling-positive cells. Subgroup analyses regarding several factors potentially influencing the effects of exosomes on NFS and IV were also conducted. Results: We identified 21 experiments from 18 studies in the meta-analysis. Pooled analyses showed the positive and significant effects of exosomes on NFS (standardized mean difference -2.79; 95% confidence interval -3.81 to -1.76) and IV (standardized mean difference -3.16; 95% confidence interval -4.18 to -2.15). Our data revealed that the effects of exosomes on neurological outcomes in rodent stroke models might be related to routes of administration and exosomes sources. In addition, there was significant attenuation in pro-inflammatory factors, including interleukin-6, tumor necrosis factor-α and interleukin-1ß, and terminal deoxynucleotidyl transferase deoxyuridine triphosphate nick end labeling-positive cells when undergoing exosomes treatment. Conclusion: Cell-derived exosomes treatment demonstrated statistically significant improvements in structural and neurological function recovery in animal models of ischemic stroke. Our results also provide relatively robust evidence supporting cell-derived exosomes as a promising therapy to promote neurological recovery in stroke individuals.

9.
J Hum Hypertens ; 32(2): 94-104, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29317741

RESUMO

Uncertainties still remain in terms of the efficacy of anti-hypertensive treatment on the risk of major cardiovascular (CV) events within prehypertensive levels. This review aims to assess the efficacy and safety of anti-hypertensives on the CV risks in populations within prehypertensive levels. Randomized controlled trials (RCTs) concerning active treatment vs placebo in populations within prehypertensive levels were identified through electronic database and manual search. Outcomes included the first co-primary outcomes, stroke, heart failure (HF), myocardial infarction (MI), all-cause mortality, and cardiovascular mortality. The first co-primary outcomes were defined as composite cardiovascular disease (CVD) events in the included studies. A total of 29 RCTs involving 127,641 participants were identified. Pooled analysis showed active treatment was associated with a significant 7% reduction in risk of the first co-primary outcomes, 14% in stroke, and 10% in HF as compared to placebo (0.86, 0.77-0.96; 0.93, 0.89-0.98; and 0.90, 0.83-0.97). However, there were no significant reductions in risk of MI, all-cause mortality, and cardiovascular mortality. A significant reduction in risk of the first co-primary outcomes was observed in subpopulations with systolic blood pressure (SBP) 130-139 mmHg (0.94, 0.89-0.99) or prior CVDs (0.88, 0.82-0.94). Meta-regression analyses showed no significant relative risk reductions proportional to the magnitude of the mean baseline BP, mean on-treatment BP, the mean absolute change in BP, the proportion of patients with hypertension, and mean age. In summary, anti-hypertensive treatment has beneficial cardiovascular effects in populations within prehypertensive levels, especially in subpopulations with SBP 130-139 mmHg or prior CVDs.


Assuntos
Anti-Hipertensivos/uso terapêutico , Insuficiência Cardíaca/prevenção & controle , Infarto do Miocárdio/prevenção & controle , Pré-Hipertensão/tratamento farmacológico , Acidente Vascular Cerebral/prevenção & controle , Humanos , Pré-Hipertensão/complicações
10.
Neural Regen Res ; 10(6): 925-31, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26199609

RESUMO

Curcumin has been shown to significantly improve spatial memory impairment induced by HIV-1 gp120 V3 in rats, but the electrophysiological mechanism remains unknown. Using extracellular microelectrode recording techniques, this study confirmed that the gp120 V3 loop could suppress long-term potentiation in the rat hippocampal CA1 region and synaptic plasticity, and that curcumin could antagonize these inhibitory effects. Using a Fura-2/AM calcium ion probe, we found that curcumin resisted the effects of the gp120 V3 loop on hippocampal synaptosomes and decreased Ca(2+) concentration in synaptosomes. This effect of curcumin was identical to nimodipine, suggesting that curcumin improved the inhibitory effects of gp120 on synaptic plasticity, ameliorated damage caused to the central nervous system, and might be a potential neuroprotective drug.

SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa