Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 361
Filtrar
Más filtros

País/Región como asunto
Intervalo de año de publicación
1.
Nature ; 582(7812): 395-398, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32494010

RESUMEN

Neuroprotectant strategies that have worked in rodent models of stroke have failed to provide protection in clinical trials. Here we show that the opposite circadian cycles in nocturnal rodents versus diurnal humans1,2 may contribute to this failure in translation. We tested three independent neuroprotective approaches-normobaric hyperoxia, the free radical scavenger α-phenyl-butyl-tert-nitrone (αPBN), and the N-methyl-D-aspartic acid (NMDA) antagonist MK801-in mouse and rat models of focal cerebral ischaemia. All three treatments reduced infarction in day-time (inactive phase) rodent models of stroke, but not in night-time (active phase) rodent models of stroke, which match the phase (active, day-time) during which most strokes occur in clinical trials. Laser-speckle imaging showed that the penumbra of cerebral ischaemia was narrower in the active-phase mouse model than in the inactive-phase model. The smaller penumbra was associated with a lower density of terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL)-positive dying cells and reduced infarct growth from 12 to 72 h. When we induced circadian-like cycles in primary mouse neurons, deprivation of oxygen and glucose triggered a smaller release of glutamate and reactive oxygen species, as well as lower activation of apoptotic and necroptotic mediators, in 'active-phase' than in 'inactive-phase' rodent neurons. αPBN and MK801 reduced neuronal death only in 'inactive-phase' neurons. These findings suggest that the influence of circadian rhythm on neuroprotection must be considered for translational studies in stroke and central nervous system diseases.


Asunto(s)
Relojes Circadianos/fisiología , Ritmo Circadiano/fisiología , Modelos Animales de Enfermedad , Neuronas/patología , Neuroprotección , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/prevención & control , Animales , Isquemia Encefálica/patología , Isquemia Encefálica/fisiopatología , Isquemia Encefálica/prevención & control , Glucosa/deficiencia , Humanos , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Accidente Cerebrovascular/fisiopatología , Investigación Biomédica Traslacional , Insuficiencia del Tratamiento
2.
Circ Res ; 128(3): 363-382, 2021 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-33301355

RESUMEN

RATIONALE: Cerebrovascular function is critical for brain health, and endogenous vascular protective pathways may provide therapeutic targets for neurological disorders. S1P (Sphingosine 1-phosphate) signaling coordinates vascular functions in other organs, and S1P1 (S1P receptor-1) modulators including fingolimod show promise for the treatment of ischemic and hemorrhagic stroke. However, S1P1 also coordinates lymphocyte trafficking, and lymphocytes are currently viewed as the principal therapeutic target for S1P1 modulation in stroke. OBJECTIVE: To address roles and mechanisms of engagement of endothelial cell S1P1 in the naive and ischemic brain and its potential as a target for cerebrovascular therapy. METHODS AND RESULTS: Using spatial modulation of S1P provision and signaling, we demonstrate a critical vascular protective role for endothelial S1P1 in the mouse brain. With an S1P1 signaling reporter, we reveal that abluminal polarization shields S1P1 from circulating endogenous and synthetic ligands after maturation of the blood-neural barrier, restricting homeostatic signaling to a subset of arteriolar endothelial cells. S1P1 signaling sustains hallmark endothelial functions in the naive brain and expands during ischemia by engagement of cell-autonomous S1P provision. Disrupting this pathway by endothelial cell-selective deficiency in S1P production, export, or the S1P1 receptor substantially exacerbates brain injury in permanent and transient models of ischemic stroke. By contrast, profound lymphopenia induced by loss of lymphocyte S1P1 provides modest protection only in the context of reperfusion. In the ischemic brain, endothelial cell S1P1 supports blood-brain barrier function, microvascular patency, and the rerouting of blood to hypoperfused brain tissue through collateral anastomoses. Boosting these functions by supplemental pharmacological engagement of the endothelial receptor pool with a blood-brain barrier penetrating S1P1-selective agonist can further reduce cortical infarct expansion in a therapeutically relevant time frame and independent of reperfusion. CONCLUSIONS: This study provides genetic evidence to support a pivotal role for the endothelium in maintaining perfusion and microvascular patency in the ischemic penumbra that is coordinated by S1P signaling and can be harnessed for neuroprotection with blood-brain barrier-penetrating S1P1 agonists.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Arterias Cerebrales/metabolismo , Células Endoteliales/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Ataque Isquémico Transitorio/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Esfingosina/análogos & derivados , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/patología , Barrera Hematoencefálica/fisiopatología , Arterias Cerebrales/efectos de los fármacos , Arterias Cerebrales/patología , Arterias Cerebrales/fisiopatología , Circulación Cerebrovascular , Modelos Animales de Enfermedad , Células Endoteliales/patología , Femenino , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/fisiopatología , Infarto de la Arteria Cerebral Media/prevención & control , Ataque Isquémico Transitorio/patología , Ataque Isquémico Transitorio/fisiopatología , Ataque Isquémico Transitorio/prevención & control , Accidente Cerebrovascular Isquémico/patología , Accidente Cerebrovascular Isquémico/fisiopatología , Accidente Cerebrovascular Isquémico/prevención & control , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microcirculación , Fármacos Neuroprotectores/farmacología , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato/agonistas , Receptores de Esfingosina-1-Fosfato/genética , Grado de Desobstrucción Vascular
3.
Pharm Biol ; 60(1): 2189-2199, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36307999

RESUMEN

CONTEXT: Effective treatment of ischaemic stroke is required to combat its high prevalence and incidence. Although the combination of Astragalus membranaeus (Fisch.) Bge. (Fabaceae) and Carthamus tinctorius L. (Asteraceae) is used in traditional Chinese medicine for the treatment of stroke, its underlying mechanism remains unclear. OBJECTIVE: The objective of this study is to elucidate the mechanism underlying Huangqi-Honghua (HQ-HH) for the treatment of ischaemic stroke by gut microbiota analysis and metabonomics. MATERIALS AND METHODS: Sprague-Dawley rats were randomly assigned to the sham, model, HQ-HH, and Naoxintong (NXT) groups. The middle cerebral artery occlusion-reperfusion model was established after 7 days of intragastric administration in the HQ-HH (4.5 g/kg, qd) and NXT (1.0 g/kg, qd) groups. The neurological examination, infarct volume, gut microbiota, bile acids, and inflammation markers were assessed after 72 h of reperfusion. RESULTS: Compared with the model group, HQ-HH significantly reduced the neurological deficit scores of the model rats (2.0 ± 0.2 vs. 3.16 ± 0.56), and reduced the cerebral infarct volume (27.83 ± 3.95 vs. 45.17 ± 2.75), and reduced the rate of necrotic neurons (26.35 ± 4.37 vs. 53.50 ± 9.61). HQ-HH regulating gut microbiota, activating the bile acid receptor FXR, maintaining the homeostasis of bile acid, reducing Th17 cells and increasing Treg cells in the rat brain, reducing the inflammatory response, and improving cerebral ischaemia-reperfusion injury. CONCLUSIONS: These data indicate that HQ-HH combination can improve ischaemic stroke by regulating the gut microbiota to affect bile acid metabolism, providing experimental evidence for the wide application of HQ-HH in clinical practice of ischaemic stroke.


Asunto(s)
Isquemia Encefálica , Carthamus tinctorius , Microbioma Gastrointestinal , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Accidente Cerebrovascular , Animales , Ratas , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/prevención & control , Ratas Sprague-Dawley , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/prevención & control , Daño por Reperfusión/tratamiento farmacológico , Reperfusión , Ácidos y Sales Biliares/uso terapéutico
4.
Biochem Biophys Res Commun ; 577: 52-57, 2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34507065

RESUMEN

Focal ischemia causes irreversible brain damage if cerebral blood flow is not restored promptly. Acute phase excitotoxicity and pro-oxidant and inflammatory events in the sub-chronic phase elicit coagulative necrosis, vascular injury, cerebral oedema, and neurobehavioral deficits. Earlier, in pre-clinical studies arbutin protected behavioral functions and improved therapeutic outcomes in different models of brain and metabolic disorders. Arbutin is natural hydroquinone that might protect against ischemia-reperfusion (I/R) injury. In this study, cerebro-protective effects of arbutin were evaluated in the middle cerebral artery occlusion-reperfusion (MCAo/R) mouse model. Mice were administered arbutin (50, 100 mg/kg, i.p.) for 21 days, and subjected to MCAo/R or sham surgery on day 14. Results showed brain infarction, blood-brain barrier dysfunction, oedema, and neurological deficits 24 h post-MCAo/R injury that were prevented by arbutin. Behavioral evaluations over the sub-chronic phase revealed MCAo/R triggered spatial and working memory deficits. Arbutin protected the memory against MCAo/R injury and decreased hydroxy-2'-deoxyguanosine, protein carbonyls, inflammatory cytokines (tumor necrosis factor-α, myeloperoxidase, matrix metalloproteinase-9, inducible nitric oxide synthase), and enhanced glutathione levels in the ischemia ipsilateral hemisphere. Arbutin decreased brain acetylcholinesterase activity, glutamate, and enhanced GABA levels against MCAo/R. Arbutin can alleviate I/R pathogenesis and protects neurobehavioral functions in the MCAo/R mouse model.


Asunto(s)
Arbutina/farmacología , Encéfalo/efectos de los fármacos , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/prevención & control , Daño por Reperfusión/prevención & control , Animales , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/fisiología , Encéfalo/fisiopatología , Cromatografía Líquida de Alta Presión , Ácido Glutámico/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Memoria a Corto Plazo/efectos de los fármacos , Memoria a Corto Plazo/fisiología , Ratones , Fármacos Neuroprotectores/farmacología , Neurotransmisores/metabolismo , Permeabilidad/efectos de los fármacos , Daño por Reperfusión/fisiopatología , Ácido gamma-Aminobutírico/metabolismo
5.
J Neuroinflammation ; 18(1): 265, 2021 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-34772416

RESUMEN

BACKGROUND: Lymphocytes have dichotomous functions in ischemic stroke. Regulatory T cells are protective, while IL-17A from innate lymphocytes promotes the infarct growth. With recent advances of T cell-subtype specific transgenic mouse models it now has become possible to study the complex interplay of T cell subpopulations in ischemic stroke. METHODS: In a murine model of experimental stroke we analyzed the effects of IL-10 on the functional outcome for up to 14 days post-ischemia and defined the source of IL-10 in ischemic brains based on immunohistochemistry, flow cytometry, and bone-marrow chimeric mice. We used neutralizing IL-17A antibodies, intrathecal IL-10 injections, and transgenic mouse models which harbor a deletion of the IL-10R on distinct T cell subpopulations to further explore the interplay between IL-10 and IL-17A pathways in the ischemic brain. RESULTS: We demonstrate that IL-10 deficient mice exhibit significantly increased infarct sizes on days 3 and 7 and enlarged brain atrophy and impaired neurological outcome on day 14 following tMCAO. In ischemic brains IL-10 producing immune cells included regulatory T cells, macrophages, and microglia. Neutralization of IL-17A following stroke reversed the worse outcome in IL-10 deficient mice and intracerebral treatment with recombinant IL-10 revealed that IL-10 controlled IL-17A positive lymphocytes in ischemic brains. Importantly, IL-10 acted differentially on αß and γδ T cells. IL-17A producing CD4+ αß T cells were directly controlled via their IL-10-receptor (IL-10R), whereas IL-10 by itself had no direct effect on the IL-17A production in γδ T cells. The control of the IL-17A production in γδ T cells depended on an intact IL10R signaling in regulatory T cells (Tregs). CONCLUSIONS: Taken together, our data indicate a key function of IL-10 in restricting the detrimental IL-17A-signaling in stroke and further supports that IL-17A is a therapeutic opportunity for stroke treatment.


Asunto(s)
Interleucina-10/uso terapéutico , Interleucina-17/antagonistas & inhibidores , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Animales , Anticuerpos Neutralizantes/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Inmunohistoquímica , Infarto de la Arteria Cerebral Media/prevención & control , Inyecciones Espinales , Interleucina-10/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores de Interleucina-10/antagonistas & inhibidores , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Resultado del Tratamiento
6.
J Neuroinflammation ; 18(1): 40, 2021 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531049

RESUMEN

BACKGROUND: Nucleotide-binding oligomerization domain-like receptor pyrin domain-containing protein 3 (NLRP3) plays an important role in mediating inflammatory responses during ischemic stroke. Bile acid receptor Takeda-G-protein-receptor-5 (TGR5) has been identified as an important component in regulating brain inflammatory responses. In this study, we investigated the mechanism of TGR5 in alleviating neuroinflammation after middle cerebral artery occlusion (MCAO). METHODS: Sprague-Dawley rats were subjected to MCAO and TGR5 agonist INT777 was administered intranasally 1 h after MCAO. Small interfering RNAs (siRNA) targeting TGR5 and Pellino3 were administered through intracerebroventricular injection 48 h before MCAO. Infarct volumes and neurologic scores were evaluated, and ELISA, flow cytometry, immunofluorescence staining, immunoblotting, and co-immunoprecipitation were used for the evaluations. RESULTS: Endogenous TGR5 and Pellino3 levels increased after MCAO. TGR5 activation by INT777 significantly decreased pro-inflammatory cytokine, cleaved caspase-8, and NLRP3 levels, thereby reducing brain infarctions; both short- and long-term neurobehavioral assessments showed improvements. Ischemic damage induced the interaction of TGR5 with Pellino3. Knockdown of either TGR5 or Pellino3 increased the accumulation of cleaved caspase-8 and NLRP3, aggravated cerebral impairments, and abolished the anti-inflammatory effects of INT777 after MCAO. CONCLUSIONS: TGR5 activation attenuated brain injury by inhibiting neuroinflammation after MCAO, which could be mediated by Pellino3 inhibition of caspase-8/NLRP3.


Asunto(s)
Caspasa 8/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Mediadores de Inflamación/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Administración Intranasal , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Ácidos Cólicos/administración & dosificación , Infarto de la Arteria Cerebral Media/prevención & control , Mediadores de Inflamación/antagonistas & inhibidores , Inyecciones Intraventriculares , Masculino , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , ARN Interferente Pequeño/administración & dosificación , Ratas , Ratas Sprague-Dawley , Receptores Acoplados a Proteínas G/agonistas , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores
7.
J Neuroinflammation ; 18(1): 119, 2021 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-34022892

RESUMEN

BACKGROUND: Cerebral ischemia-reperfusion (I/R) injury is a major cause of early complications and unfavorable outcomes after endovascular thrombectomy (EVT) therapy in patients with acute ischemic stroke (AIS). Recent studies indicate that modulating microglia/macrophage polarization and subsequent inflammatory response may be a potential adjunct therapy to recanalization. Annexin A1 (ANXA1) exerts potent anti-inflammatory and pro-resolving properties in models of cerebral I/R injury. However, whether ANXA1 modulates post-I/R-induced microglia/macrophage polarization has not yet been fully elucidated. METHODS: We retrospectively collected blood samples from AIS patients who underwent successful recanalization by EVT and analyzed ANXA1 levels longitudinally before and after EVT and correlation between ANXA1 levels and 3-month clinical outcomes. We also established a C57BL/6J mouse model of transient middle cerebral artery occlusion/reperfusion (tMCAO/R) and an in vitro model of oxygen-glucose deprivation and reoxygenation (OGD/R) in BV2 microglia and HT22 neurons to explore the role of Ac2-26, a pharmacophore N-terminal peptide of ANXA1, in regulating the I/R-induced microglia/macrophage activation and polarization. RESULTS: The baseline levels of ANXA1 pre-EVT were significantly lower in 23 AIS patients, as compared with those of healthy controls. They were significantly increased to the levels found in controls 2-3 days post-EVT. The increased post-EVT levels of ANXA1 were positively correlated with 3-month clinical outcomes. In the mouse model, we then found that Ac2-26 administered at the start of reperfusion shifted microglia/macrophage polarization toward anti-inflammatory M2-phenotype in ischemic penumbra, thus alleviating blood-brain barrier leakage and neuronal apoptosis and improving outcomes at 3 days post-tMCAO/R. The protection was abrogated when mice received Ac2-26 together with WRW4, which is a specific antagonist of formyl peptide receptor type 2/lipoxin A4 receptor (FPR2/ALX). Furthermore, the interaction between Ac2-26 and FPR2/ALX receptor activated the 5' adenosine monophosphate-activated protein kinase (AMPK) and inhibited the downstream mammalian target of rapamycin (mTOR). These in vivo findings were validated through in vitro experiments. CONCLUSIONS: Ac2-26 modulates microglial/macrophage polarization and alleviates subsequent cerebral inflammation by regulating the FPR2/ALX-dependent AMPK-mTOR pathway. It may be investigated as an adjunct strategy for clinical prevention and treatment of cerebral I/R injury after recanalization. Plasma ANXA1 may be a potential biomarker for outcomes of AIS patients receiving EVT.


Asunto(s)
Anexina A1/metabolismo , Diferenciación Celular , Infarto de la Arteria Cerebral Media/prevención & control , Macrófagos , Microglía/metabolismo , Daño por Reperfusión/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Anciano , Animales , Anexina A1/farmacología , Anexina A1/uso terapéutico , Diferenciación Celular/efectos de los fármacos , Línea Celular , Modelos Animales de Enfermedad , Femenino , Proteínas de Homeodominio/metabolismo , Humanos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Persona de Mediana Edad , Péptidos/uso terapéutico , Receptores de Formil Péptido/metabolismo , Daño por Reperfusión/inmunología , Estudios Retrospectivos
8.
Basic Res Cardiol ; 116(1): 32, 2021 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-33942194

RESUMEN

Stroke remains one of the most common causes of death and disability worldwide. Several preclinical studies demonstrated that the brain can be effectively protected against ischaemic stroke by two seemingly distinct treatments: remote ischaemic conditioning (RIC), involving cycles of ischaemia/reperfusion applied to a peripheral organ or tissue, or by systemic administration of glucagon-like-peptide-1 (GLP-1) receptor (GLP-1R) agonists. The mechanisms underlying RIC- and GLP-1-induced neuroprotection are not completely understood. In this study, we tested the hypothesis that GLP-1 mediates neuroprotection induced by RIC and investigated the effect of GLP-1R activation on cerebral blood vessels, as a potential mechanism of GLP-1-induced protection against ischaemic stroke. A rat model of ischaemic stroke (90 min of middle cerebral artery occlusion followed by 24-h reperfusion) was used. RIC was induced by 4 cycles of 5 min left hind limb ischaemia interleaved with 5-min reperfusion periods. RIC markedly (by ~ 80%) reduced the cerebral infarct size and improved the neurological score. The neuroprotection established by RIC was abolished by systemic blockade of GLP-1R with a specific antagonist Exendin(9-39). In the cerebral cortex of GLP-1R reporter mice, ~ 70% of cortical arterioles displayed GLP-1R expression. In acute brain slices of the rat cerebral cortex, activation of GLP-1R with an agonist Exendin-4 had a strong dilatory effect on cortical arterioles and effectively reversed arteriolar constrictions induced by metabolite lactate or oxygen and glucose deprivation, as an ex vivo model of ischaemic stroke. In anaesthetised rats, Exendin-4 induced lasting increases in brain tissue PO2, indicative of increased cerebral blood flow. These results demonstrate that neuroprotection against ischaemic stroke established by remote ischaemic conditioning is mediated by a mechanism involving GLP-1R signalling. Potent dilatory effect of GLP-1R activation on cortical arterioles suggests that the neuroprotection in this model is mediated via modulation of cerebral blood flow and improved brain perfusion.


Asunto(s)
Arteriolas/efectos de los fármacos , Circulación Cerebrovascular/efectos de los fármacos , Miembro Posterior/irrigación sanguínea , Incretinas/farmacología , Infarto de la Arteria Cerebral Media/prevención & control , Precondicionamiento Isquémico , Accidente Cerebrovascular Isquémico/prevención & control , Fármacos Neuroprotectores/farmacología , Fragmentos de Péptidos/farmacología , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología , Animales , Arteriolas/metabolismo , Arteriolas/fisiopatología , Modelos Animales de Enfermedad , Receptor del Péptido 1 Similar al Glucagón/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/fisiopatología , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/fisiopatología , Masculino , Ratas Sprague-Dawley , Flujo Sanguíneo Regional
9.
Chem Res Toxicol ; 34(4): 1016-1023, 2021 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-33728903

RESUMEN

Stroke is a significant cardiovascular disease that influences the health of human beings all over the world, especially the elderly population. It is reported that the blood-brain barrier (BBB) can be easily destroyed by stroke, which is one of the main factors responsible for macrophage infiltration and central nervous inflammation. Here, we report the protective effects of Trelagliptin against BBB injury and macrophage infiltration. Our results indicate that the infraction volume, the neurological score, and macrophage infiltration staining with CD68 were increased in middle cerebral artery occlusion (MCAO) mice but significantly reversed by treatment with Trelagliptin. Additionally, Trelagliptin reduced the permeability of the BBB by increasing the expression of the tight junction zonula occludens protein-1 (ZO-1) in the cerebral cortex. In an in vitro hypoxia model of endothelial cells, the increased migration of macrophages, enlarged permeability of endothelial monolayer, downregulation of ZO-1, and elevated expression level of CXCL1 by hypoxic conditions were all reversed by treatment with Trelagliptin in a dose-dependent manner. Our results demonstrate that Trelagliptin might mitigate macrophage infiltration by preventing the breakdown of the blood-brain barrier in the brains of MCAO mice.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/efectos de los fármacos , Infarto de la Arteria Cerebral Media/prevención & control , Macrófagos/efectos de los fármacos , Uracilo/análogos & derivados , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Uracilo/química , Uracilo/farmacología
10.
Neurochem Res ; 46(12): 3301-3313, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34431027

RESUMEN

The present study was conducted to investigate the effects of different doses of recombinant human Chemerin (rhChemerin) on brain damage, spatial memory, blood-brain barrier (BBB) disruption and cellular and molecular mechanisms in a mouse stroke model. The mouse stroke model was developed by blocking the middle cerebral artery for 1 h and performing reperfusion for 23 h. Immediately, one and three hours after the stroke, 200, 400 and 800 ng/mouse of intranasal rhChemerin was administered. Neuronal and BBB damage, spatial memory and neurological performance were examined 24 h after the stroke. Western blotting and immunofluorescence were utilized to determine the effects of rhChemerin on the expressions of nuclear factor kappa B (NF-κB), pro-inflammatory cytokines such as TNF-α and IL-1ß, anti-inflammatory cytokines such as IL-10 and TGF-ß and vascular endothelial growth factor (VEGF). Administering 400 and 800 ng/mouse of rhChemerin in the mice immediately and one hour after ischemia minimized the infarct size, BBB opening, spatial memory and neurological impairment (P < 0.001). Furthermore, 800 ng/mouse of rhChemerin significantly diminished terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling (TUNEL)-positive (apoptotic) cells, suppressed the expressions of NF-kB, TNF-α and IL-1ß and upregulated IL-10 and VEGF in the cortex and hippocampus of the mice. The present findings showed that rhChemerin administered immediately and one hour after stroke alleviates neuronal and BBB injures and improves spatial memory. These effects of rhChemerin may be mediated by inhibiting inflammatory pathways and apoptotic machinery.


Asunto(s)
Edema Encefálico/prevención & control , Lesiones Encefálicas/prevención & control , Quimiocinas/farmacología , Infarto de la Arteria Cerebral Media/prevención & control , Péptidos y Proteínas de Señalización Intercelular/farmacología , Fármacos Neuroprotectores/farmacología , Proteínas Recombinantes/farmacología , Accidente Cerebrovascular/complicaciones , Animales , Apoptosis , Barrera Hematoencefálica/metabolismo , Edema Encefálico/etiología , Edema Encefálico/metabolismo , Edema Encefálico/patología , Lesiones Encefálicas/etiología , Lesiones Encefálicas/metabolismo , Lesiones Encefálicas/patología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/etiología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Ratones , Transducción de Señal
11.
Metab Brain Dis ; 36(6): 1341-1351, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33842985

RESUMEN

MicroRNAs (miRNAs) are known as important regulators of gene expression and play important roles in diverse biological activities. However, the involvement of miRNAs in cerebral ischemia remains elusive. In the present study, using the middle cerebral artery occlusion (MCAO) model and oxygen-glucose deprivation/reperfusion (OGD/RP)-induced cell injury model, we found that the expression levels of miR-34c-5p were significantly reduced in MCAO rats and OGD/RP cells. Overexpression of miR-34c-5p could improve the increased brain infarction, brain water content and neurological scores in MCAO rats, as well as the abnormal expression of inflammatory cytokines (TNF-α, IL-6, COX-2, iNOS, IL-10) in OGD/RP cells. Moreover, overexpression of miR-34c-5p was found to inhibit the activity of nuclear factor-kappa B (NF-κB) by regulating the expression of nuclear receptor coactivator 1 (NCOA1), and increase the apoptotic rate of cortical neurons by inhibiting the expression of Caspase-3 and Bax and upregulating the expression of Bcl-2. Taken together, our findings demonstrated that miR-34c-5p plays an important role in cerebral ischemia/reperfusion injury, which may be mediated through inflammatory and apoptotic signaling pathways.


Asunto(s)
Antiinflamatorios/uso terapéutico , Apoptosis/efectos de los fármacos , Isquemia Encefálica/prevención & control , MicroARNs/genética , Daño por Reperfusión/prevención & control , Animales , Conducta Animal/efectos de los fármacos , Agua Corporal , Química Encefálica , Isquemia Encefálica/genética , Isquemia Encefálica/psicología , Citocinas/genética , Hipoxia/complicaciones , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/prevención & control , Masculino , FN-kappa B/genética , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/genética , Daño por Reperfusión/psicología , Transducción de Señal/genética
12.
Int J Neurosci ; 131(3): 264-278, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32125198

RESUMEN

Purpose: Cerebral ischemic stroke, caused by obstruction of the blood flow to the brain, initiates a complex cascade of pathophysiological changes. The aim of the present study was to assess the protective role and the underlying mechanism of troxerutin and cerebroprotein hydrolysate (TCH) injections for five days in rats subjected to middle cerebral artery occlusion (MCAO).Materials and Methods: Male Sprague-Dawley rats treated with either TCH or a vehicle (0.9% saline) via intraperitoneal injection were examined one or three days after MCAO.Results: TCH alleviated neurological deficits and reduced infarct volume, innate immune response, blood-brain barrier destruction, and suppressed cell apoptosis. The therapeutic effects of TCH were achieved by diminished neuronal nitric oxide synthase (nNOS) and inducible nitric oxide synthase (iNOS), and increased endothelial nitric oxide synthase (eNOS). Furthermore, L-NAME showed an inhibitory effect against TCH after MCAO on eNOS expression, NO and peroxynitrite production, neurobehavioral score, and infarct volume.Conclusions: The results indicate that injection of TCH has multifaceted neuroprotective effects against MCAO via regulation of the various NOS isoforms.


Asunto(s)
Anticoagulantes/administración & dosificación , Modelos Animales de Enfermedad , Hidroxietilrutósido/análogos & derivados , Infarto de la Arteria Cerebral Media/prevención & control , Fármacos Neuroprotectores/administración & dosificación , Acoplamiento Neurovascular/efectos de los fármacos , Animales , Hidroxietilrutósido/administración & dosificación , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Masculino , Acoplamiento Neurovascular/fisiología , Ratas , Ratas Sprague-Dawley
13.
J Stroke Cerebrovasc Dis ; 30(3): 105485, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33360253

RESUMEN

PURPOSE: Cerebral ischemic injury contributes to severe dysfunction of the brain, which triggers extremely high mortality and disability. The role of microRNA (miR)-181a-5p is documented in cerebral ischemic injury. Therefore, this study intended to further figure out the mechanism of miR-181a-5p in cerebral ischemic injury. METHODS: miR-181a-5p expression in middle cerebral artery occlusion (MCAO) mouse model, oxygen-glucose-deprivation/reoxygenation (OGD/R) N2a cell model, and serum from acute ischemic injury (ACI) patients was evaluated using reverse transcription quantitative polymerase chain reaction (RT-qPCR). Gain- and loss-of-function assays were implemented in MCAO mice and OGD/R-induced N2a cells. In mice, the cerebral infarction area was assessed with 2,3,5-triphenyltetrazolium chloride staining, the number of damaged neurons by Nissl staining, and apoptosis by TdT-mediated dUTP-biotin nick end-labeling staining. Moreover, N2a cell apoptosis and proliferation were determined with flow cytometry or 5-ethynyl-2'-deoxyuridine staining, respectively. The expression of En2 and Wnt/ß-catenin pathway-related factors was determined with RT-qPCR and Western blot analysis. The targeting relationship between miR-181a-5p and En2 was evaluated by dual luciferase reporter gene assay. RESULTS: miR-181a-5p was highly expressed in serum of ACI patients, MCAO mice, and OGD/R-induced N2a cells. En2, lowly expressed in MCAO mice, was targeted by miR-181a-5p, and miR-181a-5p down-regulation activated the Wnt/ß-catenin pathway. Furthermore, miR-181a-5p inhibition or En2 overexpression reduced cerebral infarction area, the number of damaged neurons, and apoptosis in MCAO mice, and also diminished apoptosis and accelerated proliferation of OGD/R-induced N2a cells. CONCLUSION: miR-181a-5p suppression activated Wnt/ß-catenin pathway and sequentially attenuated cerebral ischemic injury by targeting En2.


Asunto(s)
Encéfalo/metabolismo , Proteínas de Homeodominio/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Vía de Señalización Wnt , Animales , Apoptosis , Encéfalo/patología , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Regulación hacia Abajo , Proteínas de Homeodominio/genética , Infarto de la Arteria Cerebral Media/genética , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/prevención & control , Masculino , Ratones Endogámicos C57BL , MicroARNs/sangre , MicroARNs/genética , Proteínas del Tejido Nervioso/genética , Neuronas/patología
14.
Glia ; 68(1): 76-94, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31420975

RESUMEN

Ischemic preconditioning (IPC) is an experimental phenomenon in which a subthreshold ischemic insult applied to the brain reduces damage caused by a subsequent more severe ischemic episode. Identifying key molecular and cellular mediators of IPC will provide critical information needed to develop novel therapies for stroke. Here we report that the transcriptomic response of acutely isolated preconditioned cortical microglia is dominated by marked upregulation of genes involved in cell cycle activation and cellular proliferation. Notably, this transcriptional response occurs in the absence of cortical infarction. We employed ex vivo flow cytometry, immunofluorescent microscopy, and quantitative stereology methods on brain tissue to evaluate microglia proliferation following IPC. Using cellular colocalization of microglial (Iba1) and proliferation (Ki67 and BrdU) markers, we observed a localized increase in the number of microglia and proliferating microglia within the preconditioned hemicortex at 72, but not 24, hours post-IPC. Our quantification demonstrated that the IPC-induced increase in total microglia was due entirely to proliferation. Furthermore, microglia in the preconditioned hemisphere had altered morphology and increased soma volumes, indicative of an activated phenotype. Using transgenic mouse models with either fractalkine receptor (CX3CR1)-haploinsufficiency or systemic type I interferon signaling loss, we determined that microglial proliferation after IPC is dependent on fractalkine signaling but independent of type I interferon signaling. These findings suggest there are multiple distinct targetable signaling pathways in microglia, including CX3CR1-dependent proliferation that may be involved in IPC-mediated protection.


Asunto(s)
Ciclo Celular/fisiología , Corteza Cerebral/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Precondicionamiento Isquémico/métodos , Microglía/metabolismo , Transcriptoma/fisiología , Animales , Proliferación Celular/fisiología , Corteza Cerebral/patología , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL
15.
Stroke ; 51(6): 1886-1890, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32404037

RESUMEN

Background and Purpose- Previous experimental studies found that the infusion of human purified nascent HDL (high-density lipoprotein) significantly reduced infarct volume and hemorrhagic transformation rate by decreasing neutrophil recruitment. ApoA1-M (apolipoprotein A1-Milano) is a natural variant of human ApoA1 that confers protection against atherosclerosis. Recombinant ApoA1-M has been formulated as a complex with phospholipids to mimic the properties of nascent HDL. The aim of this study was to assess the impact of intravenous ApoA1-M in a transient middle cerebral artery occlusion stroke model in rats. Methods- In a first experiment, rats were subjected to 120-minute transient middle cerebral artery occlusion and intravenous ApoA1-M was infused immediately or 4 hours after occlusion. In a second experiment, rats were subjected to 240-minute transient middle cerebral artery occlusion and intravenous ApoA1-M was infused with or without recombinant tPA (tissue-type plasminogen activator) immediately after recanalization. Primary outcome criteria were the infarct volume and hemorrhagic transformation rate measured at 24 hours. Platelets, coagulation, and neutrophil activation biomarkers were measured in brain homogenates and plasma. Additional in vitro experiments studied the effects of ApoA1-M on platelet aggregation and platelet-neutrophil interactions. Results- The infusion of ApoA1-M immediately or 4 hours after 120-minute transient middle cerebral artery occlusion significantly reduced the infarct volume compared with saline (P=0.034 and P=0.036, respectively). Compared with tPA alone, co-administration of ApoA1-M and tPA showed similar rates of hemorrhagic transformation. ApoA1-M had no significant inhibition effect on neutrophil activation biomarkers. Platelet activation was slightly decreased in rats treated with ApoA1-M compared with saline. In vitro, the incubation of human and rat platelet-rich plasma with ApoA1-M significantly reduced ADP-induced platelet aggregation (P=0.001 and P=0.02, respectively). Conclusions- ApoA1-Milano significantly decreased the infarct volume through an inhibition of platelet aggregation but did not reduce hemorrhagic transformation and neutrophils activation as expected after previous experimental studies with nascent HDL. Visual Overview- An online visual overview is available for this article.


Asunto(s)
Apolipoproteína A-I/farmacología , Aterosclerosis/prevención & control , Infarto de la Arteria Cerebral Media/prevención & control , Animales , Aterosclerosis/sangre , Aterosclerosis/patología , Biomarcadores/sangre , Plaquetas/metabolismo , Plaquetas/patología , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media/sangre , Infarto de la Arteria Cerebral Media/patología , Masculino , Activación Neutrófila/efectos de los fármacos , Neutrófilos/metabolismo , Neutrófilos/patología , Agregación Plaquetaria/efectos de los fármacos , Ratas , Proteínas Recombinantes/farmacología
16.
Stroke ; 51(9): 2844-2853, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32772683

RESUMEN

BACKGROUND AND PURPOSE: Although VEGF165 (vascular endothelial growth factor-165) is able to enhance both angiogenesis and neurogenesis, it also increases vascular permeability through the blood-brain barrier. Heparan sulfate (HS) sugars play important roles in regulating VEGF bioactivity in the pericellular compartment. Here we asked whether an affinity-purified VEGF165-binding HS (HS7) could augment endogenous VEGF activity during stroke recovery without affecting blood-brain barrier function. METHODS: Both rat brain endothelial cell line 4 and primary rat neural progenitor cells were used to evaluate the potential angiogenic and neurogenic effects of HS7 in vitro. For in vivo experiments, male Sprague-Dawley rats were subjected to 100 minutes of transient focal cerebral ischemia, then treated after 4 days with either PBS or HS7. One week later, infarct volume, behavioral sequelae, immunohistochemical markers of angiogenesis and neural stem cell proliferation were assessed. RESULTS: HS7 significantly enhanced VEGF165-mediated angiogenesis in rat brain endothelial cell line 4 brain endothelial cells, and increased the proliferation and differentiation of primary neural progenitor cells, both via the VEGFR2 (vascular endothelial growth factor receptor 2) pathway. Intracerebroventricular injection of HS7 improved neurological outcome in ischemic rats without changing infarct volumes. Immunostaining of the compromised cerebrum demonstrated increases in collagen IV/Ki67 and nestin/Ki67 after HS7 exposure, consistent with its ability to promote angiogenesis and neurogenesis, without compromising blood-brain barrier integrity. CONCLUSIONS: A VEGF-activating glycosaminoglycan sugar, by itself, is able to enhance endogenous VEGF165 activity during the post-ischemic recovery phase of stroke.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Heparitina Sulfato/uso terapéutico , Accidente Cerebrovascular/tratamiento farmacológico , Factor A de Crecimiento Endotelial Vascular/uso terapéutico , Animales , Barrera Hematoencefálica/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Heparitina Sulfato/administración & dosificación , Infarto de la Arteria Cerebral Media/prevención & control , Inyecciones Intraventriculares , Ataque Isquémico Transitorio/tratamiento farmacológico , Ataque Isquémico Transitorio/fisiopatología , Masculino , Neovascularización Fisiológica/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Recuperación de la Función , Factor A de Crecimiento Endotelial Vascular/administración & dosificación , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
17.
Neurochem Res ; 45(8): 1888-1901, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32447509

RESUMEN

Selenium has been shown to possess antioxidant and neuroprotective effects by modulating mitochondrial function and activating mitochondrial biogenesis. Our previous study has also suggested that selenium protected neurons against glutamate toxicity and hyperglycemia-induced damage by regulating mitochondrial fission and fusion. However, it is still not known whether the mitochondrial biogenesis is involved in selenium alleviating hyperglycemia-aggravated cerebral ischemia reperfusion (I/R) injury. The object of this study is to define whether selenium protects neurons against hyperglycemia-aggravated cerebral I/R injury by promoting mitochondrial biogenesis. In vitro oxygen deprivation plus high glucose model decreased cell viability, enhanced reactive oxygen species production, and meanwhile stimulated mitochondrial biogenesis signaling. Pretreated with selenium significantly decreased cell death and further activated the mitochondrial biogenesis signaling. In vivo 30 min of middle cerebral artery occlusion in the rats under hyperglycemic condition enhanced neurological deficits, enlarged infarct volume, exacerbated neuronal damage and oxidative stress compared with normoglycemic ischemic rats after 24 h reperfusion. Consistent to the in vitro results, selenium treatment alleviated ischemic damage in hyperglycemic ischemic animals. Furthermore, selenium reduced the structural changes of mitochondria caused by hyperglycemic ischemia and further promoted the mitochondrial biogenesis signaling. Selenium activates mitochondrial biogenesis signaling, protects mitochondrial structure integrity and ameliorates cerebral I/R injury in hyperglycemic rats.


Asunto(s)
Infarto de la Arteria Cerebral Media/prevención & control , Mitocondrias/efectos de los fármacos , Fármacos Neuroprotectores/uso terapéutico , Biogénesis de Organelos , Selenito de Sodio/uso terapéutico , Animales , Muerte Celular/efectos de los fármacos , Línea Celular , Hiperglucemia/fisiopatología , Infarto de la Arteria Cerebral Media/fisiopatología , Masculino , Ratones , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Daño por Reperfusión/prevención & control , Transducción de Señal/efectos de los fármacos , Superóxido Dismutasa/metabolismo , Proteína Desacopladora 2/metabolismo
18.
Neurochem Res ; 45(2): 437-454, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31865520

RESUMEN

Pyroptosis is a newly identified lytic form of programmed cell death which is characterized by plasma membrane blebbing and rupture. Pyroptosis occurs in cerebral ischemia injury, and contributes to the activation and secretion of the inflammatory cytokines interleukin (IL)-1ß, IL-18, and IL-6. Previous reports have found that Dendrobium alkaloids (DNLA) can exert neuroprotective effects against oxygen-glucose deprivation/reperfusion (OGD/R) damage in vitro, but the mechanisms underlying these effects remain elusive. In this study, we investigated whether DNLA exerted therapeutic benefits against cerebral ischemia-reperfusion (CIR) damage via ameliorating pyroptosis and inflammation. OGD/R damage was induced in HT22 cells pretreated with DNLA (0.03, 0.3, or 3 mg/ml, 24 h prior to OGD/R), MCC950 (10 ng/ml, 1 h prior), and VX765 (10 ng/ml, 1 h prior). Neuronal apoptosis, necrosis, pyroptosis, and pathological changes were analyzed 24 h following OGD/R. Further to this, male C57/BL mice pretreated with different concentrations of DNLA (0.5 or 5 mg/kg, ip.) for 24 h and VX765 (50 mg/kg, ip., 1 h before CIR) underwent transient middle cerebral artery occlusion and reperfusion. We found that DNLA pretreatment resulted in a lower neurologic deficit score, a reduced infarct volume, fewer pyroptotic cells, and reduced levels of inflammatory factors 24 h after CIR. Furthermore, DNLA administration also reduced the levels of the pyroptosis-associated proteins Caspase-1 and gasdermin-D, particularly in the hippocampal CA1 region. Similar decreases were observed in the levels of the inflammatory factors IL-1ß, IL-6, and IL-18. OGD/R-associated ultrastructural damage was seen to improve following DNLA administration, likely due to the regulation of the tight junction protein Pannexin-1 by DNLA. Overall, these findings demonstrate that DNLA can protect against CIR damage through inhibiting pyroptosis-induced neuronal death, providing new therapeutic insights for CIR injury.


Asunto(s)
Alcaloides/uso terapéutico , Infarto de la Arteria Cerebral Media/prevención & control , Necrosis/prevención & control , Fármacos Neuroprotectores/uso terapéutico , Piroptosis/efectos de los fármacos , Daño por Reperfusión/prevención & control , Animales , Caspasa 1/metabolismo , Inhibidores de Caspasas/farmacología , Línea Celular , Dipéptidos/farmacología , Hipocampo/metabolismo , Hipocampo/patología , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Endogámicos C57BL , Necrosis/metabolismo , Necrosis/patología , Proteínas de Unión a Fosfato/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología , para-Aminobenzoatos/farmacología
19.
J Cardiovasc Pharmacol ; 77(2): 208-216, 2020 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-33351536

RESUMEN

ABSTRACT: Ischemic stroke is the leading cause of globe death and permanent disability, but its therapeutic strategies are limited. Over the past decades, multiprotein complexes called inflammasomes have been shown as promising targets in ischemic stroke. Here, we examined vinpocetine (Vinp), a synthetic drug, playing a neuroprotective role against ischemic stroke in mice through regulating NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome activation. Middle cerebral artery occlusion/reperfusion (MCAO/R) was applied to mimic ischemic stroke in vivo. Vinp was administrated by intraperitoneal injection with different dose (5 or 10 mg/kg) 1 hour after reperfusion. Then, neurological assessment and infarct size were performed, and interleukin-1ß (IL-1ß) and IL-18 levels were evaluated using ELISA. The levels of NLRP3 inflammasome components and its upstream nuclear factor-κB (NF-κB) were determined using real-time PCR or Western blot. The experimental results indicated that posttreatment with Vinp decreased cerebral infarct size, improved behavior recover, reduced NLRP3 inflammasome expression, and suppressed the transfer of NF-κB to nucleus and proinflammatory cytokine release in middle cerebral artery occlusion/reperfusion mice. In conclusion, this study demonstrates that Vinp alleviates ischemic stroke by regulating levels of NLRP3 inflammasome, NF-κB, and proinflammatory cytokines in vivo, offering an alternative medication for ischemic stroke associated with inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Encéfalo/efectos de los fármacos , Infarto de la Arteria Cerebral Media/prevención & control , Inflamasomas/metabolismo , Accidente Cerebrovascular Isquémico/prevención & control , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Fármacos Neuroprotectores/farmacología , Alcaloides de la Vinca/farmacología , Animales , Encéfalo/metabolismo , Encéfalo/patología , Modelos Animales de Enfermedad , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Inflamasomas/genética , Mediadores de Inflamación/metabolismo , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Masculino , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Transducción de Señal
20.
J Stroke Cerebrovasc Dis ; 29(11): 105286, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33066914

RESUMEN

BACKGROUND: Stroke is one of the leading causes of death and disability worldwide. Scalp acupuncture and exercise therapy have been proven as two effective methods for the treatment of stroke. However, their combined action and mechanisms have not been fully elucidated. The present study aimed to investigate the protective effect of scalp acupuncture combined with exercise therapy on neurons in rats with ischemic brain injury. METHODS: 100 rats were randomly divided into 5 groups including sham group, model group, acupuncture group, rehabilitation group, and experimental group (scalp acupuncture combined with exercise therapy). Middle cerebral artery occlusion (MCAO) model in rats was established according to Longa modified suture method to mimic ischemic stroke. The modified Bedexer's neurological function score was used to evaluate the neurological deficits of rats and the brain infarct volume was measured using 2, 3, 5-triphenyl tetrazolium chloride monohydrate (TTC) staining. Moreover, the apoptosis in the hippocampus was detected by western blotting and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The pro-inflammatory cytokines such as interleukin-1 beta (IL-1ß) and tumor necrosis factor-alpha (TNF-α), reactive oxygen species (ROS) and superoxide dismutase (SOD) were determined by corresponding kits. Immunohistochemistry or immunofluorescence was performed to detect the expression of brain-derived neurotrophic factor (BDNF), S100ß and glial fibrillary acidic protein (GFAP) in the hippocampi of rats. RESULTS: The neurological deficit score, the expression levels of apoptotic factors such as cleaved caspase-3 and Bax, and the TUNEL-positive cell rate of the experimental group were significantly lower than those of the acupuncture group and the rehabilitation group. However, apoptosis inhibitor Bcl-2 showed downregulated expression in the MCAO model rats but this trend was reverted by single and combinatorial treatments. In addition, the contents of TNF-α, IL-1ß and ROS in the acupuncture group and the rehabilitation group were significantly lower than those in the model group, but higher than the experimental group. While the opposite results were obtained in SOD activity. Furthermore, compared with the model group, the ratios of BDNF, S100ß, and GFAP-positive cells in the acupuncture, rehabilitation and experimental groups were significantly increased, and the highest ratios were recorded in the experimental group. CONCLUSIONS: This study demonstrated that scalp acupuncture combined with exercise therapy effectively counteracts ischemic brain injury via the downregulation of pro-inflammatory mediators and ROS, the increased production of the antioxidant enzyme SOD, neurotrophic factor BDNF and astrocyte activities.


Asunto(s)
Puntos de Acupuntura , Terapia por Acupuntura , Apoptosis , Encéfalo/patología , Terapia por Ejercicio , Infarto de la Arteria Cerebral Media/prevención & control , Cuero Cabelludo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Conducta Animal , Encéfalo/metabolismo , Encéfalo/fisiopatología , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Citocinas/metabolismo , Modelos Animales de Enfermedad , Proteína Ácida Fibrilar de la Glía/metabolismo , Infarto de la Arteria Cerebral Media/metabolismo , Infarto de la Arteria Cerebral Media/patología , Infarto de la Arteria Cerebral Media/psicología , Mediadores de Inflamación/metabolismo , Masculino , Necrosis , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Subunidad beta de la Proteína de Unión al Calcio S100/metabolismo , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA