Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
Int J Mol Sci ; 25(3)2024 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-38338918

RESUMEN

Due to prolonged forced positioning, the incidence of intraoperative pressure injuries is high. This study aimed to explore the impact of small-molecule antiplatelet drugs on pressure injuries by locally applying them before an injury occurs. In the first part of this study, water-soluble tracers with different molecular weights were applied to normal and early-stage pressure-injured skin. Through digital cameras, spectrophotometers, and histological observations, the penetration of tracers into the epidermis was clarified. In the second part of this study, a water-soluble antiplatelet drug called Trapidil (molecular weight = 205 Da) was applied to the left side of the back of a rat before, during, and after compression, and the contralateral side served as a non-intervention control group. The differences in pressure injuries between the two groups were observed through a digital camera, an ultraviolet camera, and temperature measurement, and skin circulation and perfusion were assessed via an intravenous injection of Evans Blue. The first part of this study found that water-soluble tracers did not easily penetrate normal skin but could more easily penetrate pressure-damaged skin. The smaller the molecular weight of the tracer, the easier it penetrated the skin. Therefore, in the next step of research, water-soluble drugs with smaller molecular weights should be selected. The second part of this study found that, compared with the control group, the occurrence rates and areas of ulcers were lower, the gray value was higher, and the skin temperature was lower in the Trapidil group (p < 0.05). After the intravenous Evans Blue injection, skin circulation and perfusion in the Trapidil group were found to be better. In conclusion, this study found that the topical skin application of a small-molecule antiplatelet agent may have significant effects against pressure injuries by improving post-decompression ischemia, providing new insights into the prevention and treatment of intraoperative pressure injuries.


Asunto(s)
Lesiones por Aplastamiento , Úlcera por Presión , Trapidil , Ratas , Animales , Inhibidores de Agregación Plaquetaria/farmacología , Inhibidores de Agregación Plaquetaria/uso terapéutico , Úlcera por Presión/tratamiento farmacológico , Trapidil/farmacología , Azul de Evans/farmacología , Piel , Agua/farmacología
2.
J Biomol Struct Dyn ; 42(2): 1064-1071, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-37114509

RESUMEN

This study investigated the effects of isosakuranetin (5,7-dihydroxy-4'-methoxyflavanone) on cerebral infarction and blood brain barrier (BBB) damage in cerebral ischemia and reperfusion (I/R) in a rat model. The right middle cerebral artery was occluded for 2 h followed by reperfusion. The experimental rats were divided into five groups: a sham, or control group; vehicle group; and 5 mg/kg, 10 mg/kg, and 20 mg/kg bodyweight isosakuranetin-treated I/R groups. After 24 h of reperfusion, the rats were tested using a six-point neurological function score. The percentage of cerebral infarction was evaluated using 2,3,5-triphenyltetrazolium chloride (TTC) staining. BBB leakage was determined by Evan Blue injection assay and brain morphology changes were observed under light microscopy following staining with hematoxylin and eosin (H&E). The results of neurological function score revealed that isosakuranetin reduced the severity of neurological damage. A dose of 10 and 20 mg/kg bodyweight of isosakuranetin significantly decreased the infarct volume. All three doses of isosakuranetin significantly decreased Evan Blue leakage. The penumbra area of the I/R brains revealed the characteristics of apoptotic cell death. Therefore, isosakuranetin-treated I/R attenuated the brain damage from cerebral I/R injury and further investigation of the mechanisms warrant further investigation to assist in the development of protective strategies against cerebral I/R injury in clinical trials.Communicated by Ramaswamy H. Sarma.


Asunto(s)
Isquemia Encefálica , Flavonoides , Daño por Reperfusión , Ratas , Animales , Barrera Hematoencefálica , Ratas Sprague-Dawley , Azul de Evans/metabolismo , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Infarto Cerebral/tratamiento farmacológico , Infarto Cerebral/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/metabolismo
3.
Food Funct ; 14(22): 10052-10068, 2023 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-37861458

RESUMEN

Background: Myocardial ischemia and reperfusion injury (MIRI) is a severe complication of revascularization therapy in patients with myocardial infarction. Therefore, there is an urgent requirement to find more therapeutic solutions for MIRI. Recently, ferroptosis, which is characterized by lipid peroxidation, was considered a critical contributor to MIRI. Fucoxanthin (FX), a natural antioxidant carotenoid, which is abundant in brown seaweed, exerts protective effects under various pathological conditions. However, whether FX alleviates MIRI is unclear. This study aims to clarify the effects of FX on MIRI. Methods: Mice with left anterior descending artery ligation and reperfusion were used as in vivo models. Neonatal rat cardiomyocytes (NRCs) induced with hypoxia and reperfusion were used as in vitro models. TTC-Evans blue staining was performed to validate the infarction size. Transmission electron microscopy was employed to detect mitochondrial injury in cardiomyocytes. In addition, 4 weeks after MIRI, echocardiography was performed to measure cardiac function; fluorescent probes and western blots were used to detect ferroptosis. Results: TTC-Evans blue staining showed that FX reduced the infarction size induced by MIRI. Transmission electron microscopy showed that FX ameliorated the MIRI-induced myofibril loss and mitochondrion shrinkage. Furthermore, FX improved LVEF and LVFS and inhibited myocardial hypertrophy and fibrosis after 4 weeks in mice with MIRI. In the in vitro study, calcein AM/PI staining and TUNEL staining showed that FX reduced cell death caused by hypoxia and reperfusion treatment. DCFH-DA and MitoSOX probes indicated that FX inhibited cellular and mitochondrial reactive oxygen species (ROS). Moreover, C11-BODIPY 581/591 staining, ferro-orange staining, MDA assay, Fe2+ assay, 4-hydroxynonenal enzyme-linked immunosorbent assay, and western blot were performed and the results revealed that FX ameliorated ferroptosis in vitro and in vivo, as indicated by inhibiting lipid ROS and Fe2+ release, as well as by modulating ferroptosis hallmark FTH, TFRC, and GPX4 expression. Additionally, the protective effects of FX were eliminated by the NRF2 inhibitor brusatol, as observed from western blotting, C11-BODIPY 581/591 staining, and calcein AM/PI staining, indicating that FX exerted cardio-protective effects on MIRI through the NRF2 pathway. Conclusion: Our study showed that FX alleviated MIRI through the inhibition of ferroptosis via the NRF2 signaling pathway.


Asunto(s)
Enfermedad de la Arteria Coronaria , Ferroptosis , Infarto del Miocardio , Isquemia Miocárdica , Daño por Reperfusión Miocárdica , Humanos , Ratas , Ratones , Animales , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Ratas Sprague-Dawley , Transducción de Señal , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Infarto del Miocardio/tratamiento farmacológico , Hipoxia
4.
CNS Neurosci Ther ; 29(11): 3612-3623, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37269080

RESUMEN

AIM: Blood-brain barrier (BBB) dysfunction is one of the hallmarks of ischemic stroke. USP14 has been reported to play a detrimental role in ischemic brain injury. However, the role of USP14 in BBB dysfunction after ischemic stroke is unclear. METHODS: In this study, we tested the role of USP14 in disrupting BBB integrity after ischemic stroke. The USP14-specific inhibitor IU1 was injected into middle cerebral artery occlusion (MCAO) mice once a day. The Evans blue (EB) assay and IgG staining were used to assess BBB leakage 3 days after MCAO. FITC-detran test was slected to examine the BBB leakage in vitro. Behavior tests were conducted to evaluate recovery from ischemic stroke. RESULTS: Middle cerebral artery occlusion increased endothelial cell USP14 expression in the brain. Furthermore, the EB assay and IgG staining showed that USP14 inhibition through IU1 injection protected against BBB leakage after MCAO. Analysis of protein expression revealed a reduction in the inflammatory response and chemokine release after IU1 treatment. In addition, IU1 treatment was found to rescue neuronal loss resulting from ischemic stroke. Behavior tests showed a positive effect of IU1 in attenuating brain injury and improving motor function recovery. In vitro study showed that IU1 treatment could alleviate endothelial cell leakage induced by OGD in cultured bend.3 cells through modulating ZO-1 expression. CONCLUSIONS: Our results demonstrate a role for USP14 in disrupting the integrity of the BBB and promoting neuroinflammation after MCAO.


Asunto(s)
Barrera Hematoencefálica , Lesiones Encefálicas , Isquemia Encefálica , Accidente Cerebrovascular Isquémico , Animales , Ratones , Barrera Hematoencefálica/metabolismo , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Células Endoteliales/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Inmunoglobulina G , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Accidente Cerebrovascular Isquémico/metabolismo , Enfermedades Neuroinflamatorias , Accidente Cerebrovascular/metabolismo
5.
Naunyn Schmiedebergs Arch Pharmacol ; 396(11): 3207-3220, 2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37243759

RESUMEN

Vascular cognitive impairment (VCI) has been one of the major types of cognitive impairment. Blood-brain barrier damage plays an essential part in the pathogenesis of VCI. At present, the treatment of VCI is mainly focused on prevention, with no drug clinically approved for the treatment of VCI. This study aimed to investigate the effects of DL-3-n-butylphthalide (NBP) on VCI rats. A modified bilateral common carotid artery occlusion (mBCCAO) model was applied to mimic VCI. The feasibility of the mBCCAO model was verified by laser Doppler, 13N-Ammonia-Positron Emission Computed Tomography (PET), and Morris Water Maze. Subsequently, the Morris water maze experiment, Evans blue staining, and western blot of tight junction protein were performed to evaluate the effect of different doses of NBP (40 mg/kg, 80 mg/kg) on the improvement of cognitive impairment and BBB disruption induced by mBCCAO. Immunofluorescence was employed to examine the changes in pericyte coverage in the mBCCAO model and the effect of NBP on pericyte coverage was preliminarily explored. mBCCAO surgery led to obvious cognitive impairment and the decrease of whole cerebral blood flow, among which the blood flow in the cortex, hippocampus and thalamus brain regions decreased more significantly. High-dose NBP (80 mg/kg) improved long-term cognitive function in mBCCAO rats, alleviated Evans blue leakage and reduced the loss of tight junction proteins (ZO-1, Claudin-5) in the early course of the disease, thereby exerting a protective effect on the blood-brain barrier. No significant changes in pericyte coverage were observed after mBCCAO. High-dose NBP improved cognitive function in mBCCAO rats. High-dose NBP protected the integrity of BBB by upregulating TJ protein expression, rather than regulating pericyte coverage ratio. NBP could be a potential drug for the treatment of VCI.


Asunto(s)
Benzofuranos , Isquemia Encefálica , Disfunción Cognitiva , Ratas , Animales , Barrera Hematoencefálica/metabolismo , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/metabolismo , Cognición , Disfunción Cognitiva/tratamiento farmacológico , Disfunción Cognitiva/metabolismo
6.
Biomed Chromatogr ; 37(6): e5631, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36987546

RESUMEN

Eucalyptol, a natural epoxide monoterpene, was found in rat brain tissue after oral administration in our previous study, suggesting that the compound may possess the ability to pass the blood-brain barrier (BBB). However, a few studies have demonstrated that eucalyptol does penetrate the BBB. The aims of this study were to determine the opening effect of eucalyptol on the BBB in rats, to establish and validate a method for the determination of eucalyptol in brain tissue, and to reveal its brain pharmacokinetic characteristics. The opening effect of BBB was assessed by dye extravasation and ultrastructural alterations, and the quantitative method of eucalyptol in rat brain tissue was established and confirmed. For pharmacokinetic research, rat brain samples were taken at 0.05, 0.167, 0.5, 1, 2.5, 5, 10, and 15 h after administration. There was a significantly higher extravasation of Evans blue from the brain parenchyma of rats in the medium-dose eucalyptol group (P < 0.01), which was associated with the BBB's altered ultrastructure. It is suggested that eucalyptol increased the permeability of the BBB. After oral administration, eucalyptol was quickly absorbed by the brain. This study provides valuable information on eucalyptol use to treat illnesses of the central nervous system.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Ratas , Animales , Eucaliptol , Ratas Sprague-Dawley , Azul de Evans/farmacología
7.
Diabetes ; 72(5): 638-652, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36821829

RESUMEN

NADPH oxidases (NOXs) are major players in generating reactive oxygen species (ROS) and are implicated in various neurodegenerative ocular pathologies. The aim of this study was to investigate the role of a NOX4 inhibitor (GLX7013114) in two in vivo, experimental streptozotocin (STZ) paradigms depicting the early events of diabetic retinopathy (DR). Animals in the diabetic treated group received GLX7013114 topically (20 µL/eye, 10 mg/mL, once daily) for 14 days (paradigm A: preventive) and 7 days (paradigm B: treated) at 48 h and 4 weeks after STZ injection, respectively. Several methodologies were used (immunohistochemistry, Western blot, real-time PCR, ELISA, pattern electroretinography [PERG]) to assess the diabetes-induced early events of DR, namely oxidative stress, neurodegeneration, and neuroinflammation, and the effect of GLX7013114 on the diabetic insults. GLX7013114, administered as eye drops (paradigms A and B), was beneficial in treating the oxidative nitrative stress, activation of caspase-3 and micro- and macroglia, and attenuation of neuronal markers. It also attenuated the diabetes-induced increase in vascular endothelial growth factor, Evans blue dye leakage, and proinflammatory cytokine (TNF-α protein, IL-1ß/IL-6 mRNA) levels. PERG amplitude values suggested that GLX7013114 protected retinal ganglion cell function (paradigm B). This study provides new findings regarding the pharmacological profile of the novel NOX4 inhibitor GLX7013114 as a promising therapeutic candidate for the treatment of the early stage of DR. ARTICLE HIGHLIGHTS: NADPH oxidases (NOXs) are implicated in the early pathological events of diabetic retinopathy (DR). The NOX4 inhibitor GLX7013114, topically administered, reduced oxidative damage and apoptosis in the rat streptozotocin model of DR. GLX7013114 protected retinal neurons and retinal ganglion cell function and reduced the expression of pro-inflammatory cytokines in the diabetic retina. GLX7013114 diminished the diabetes-induced increase in vascular endothelial growth factor levels and Evans blue dye leakage in retinal tissue. GLX7013114 exhibits neuroprotective, anti-inflammatory, and vasculoprotective properties that suggest it may have a role as a putative therapeutic for the early events of DR.


Asunto(s)
Diabetes Mellitus , Retinopatía Diabética , Ratas , Animales , Retinopatía Diabética/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/metabolismo , Estreptozocina/farmacología , Retina/metabolismo , NADPH Oxidasas/metabolismo , NADPH Oxidasas/farmacología , NADPH Oxidasas/uso terapéutico , Citocinas/metabolismo , Diabetes Mellitus/metabolismo , NADPH Oxidasa 4/genética , NADPH Oxidasa 4/metabolismo
8.
Mol Neurobiol ; 60(1): 235-246, 2023 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-36258136

RESUMEN

We have previously shown that selective inhibition of histone deacetylase 3 (HDAC3) decreases infarct volume and improves long-term functional outcomes after stroke. In this study, we examined the effects of HDAC3 inhibition on cerebral edema and blood-brain barrier (BBB) leakage and explored its underlying mechanisms. Adult male Wistar rats were subjected to 2-h middle cerebral artery occlusion (MCAO) and randomly treated i.p. with either vehicle or a selective HDAC3 inhibitor (RGFP966) at 2 and 24 h after stroke. Modified neurological severity scores (mNSS) were calculated at 2 h, 1 day, and 3 days. H&E, Evans blue dye (EBD) assay, and fluorescein isothiocyanate (FITC)-dextran were employed to assess cerebral edema and BBB leakage. Western blot for matrix metalloproteinase-9 (MMP9), MMP-9 zymography, and immunostaining for HDAC3, GFAP, Iba-1, albumin, aquaporin-4, claudin-5, ZO-1, and NF-kB were performed. Early RGFP966 administration decreased cerebral edema (p = 0.002) and BBB leakage, as measured by EBD assay, FITC-dextran, and albumin extravasation (p < 0.01). RGFP966 significantly increased tight junction proteins (claudin-5 and ZO-1) in the peri-infarct area. RGFP966 also significantly decreased HDAC3 in GFAP + astrocytes, which correlated with better mNSS (r = 0.67, p = 0.03) and decreased cerebral edema (r = 0.64, p = 0.04). RGFP966 decreased aquaporin-4 in GFAP + astrocytes (p = 0.002), as well as, the inflammatory markers Iba-1, NF-kB, and MMP9 in the ischemic brain (p < 0.05). Early HDAC3 inhibition decreases cerebral edema and BBB leakage. BBB protection by RGFP966 is mediated in part by the upregulation of tight junction proteins, downregulation of aquaporin-4 and HDAC3 in astrocytes, and decreased neuroinflammation.


Asunto(s)
Acuaporinas , Edema Encefálico , Accidente Cerebrovascular , Ratas , Animales , Masculino , Barrera Hematoencefálica/metabolismo , Edema Encefálico/complicaciones , Edema Encefálico/tratamiento farmacológico , Edema Encefálico/metabolismo , Claudina-5/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , FN-kappa B/metabolismo , Ratas Wistar , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Albúminas/metabolismo , Acuaporinas/metabolismo
9.
Microvasc Res ; 145: 104453, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36356686

RESUMEN

Increased microvascular permeability at the level of the blood-brain barrier (BBB) often leads to vasogenic brain edema following traumatic brain injury (TBI). These pathologic conditions compromise the integrity of the neurovascular unit resulting in severe brain dysfunction. To quantify this permeability and assess ionic equillibrium, preclinical researchers have relied on the use of various molecular weight permeable dyes such as Evans Blue that normally cannot enter the brain parenchyma under homeostatic conditions. Evans Blue, the most cited of the molecular weight dyes, has reported reproducibility issues because of harsh extraction processes, suboptimal detection via absorbance, and wide excitation fluorescence spectra associated with the dye. Our laboratory group transitioned to Alexa Fluor 680, a far-red dye with improved sensitivity compared to Evans Blue and thus improved reproducibility to alleviate this issue. To evaluate our reproducibility and increase the rigor of our experimental design, we retrospectively analyzed our controlled cortical impact (CCI) experiments over the past 10 years to evaluate effect size with larger samples and potential sources of variability. All of our BBB permeability experiments were performed with Male, Sprague Dawley rats weighing between 225 and 300 g. Historically, Sprague Dawleys were randomly divided into treatment groups: SHAM, CCI, and a stem cell-based treatment from years 2007-2020. The assessment of microvascular hyperpermeability were evaluated by comparing the mean at minimum threshold, area at 1 k-2 k, and intensity density obtained from Alexa Fluor 680 permeability data. Studies utilizing Evans Blue were further compared by tip depth, diameter size, and the hemisphere of injury. Statistical evaluation utilizing the G Power software analysis did not yield a significant difference in sample size comparing experimental groups for Evans Blue and Alexa Fluor 680 analyzed brain tissue. Our analysis also demonstrated a trend in that recent studies (years 2018-2020) have yielded more compact sample sizes between experimental groups in Alexa Fluor 680 analyzed rats. This retrospective study further revealed that Alexa Fluor 680 image analysis provides greater sensitivity to BBB permeability following TBI in comparison to Evans Blue. Significant differences in sample size were not detected between Evans Blue and Alexa Fluor 680; there were significant differences found throughout year to year analysis at the lower range of thresholds. SUMMARY STATEMENT: This work provides a comparative analysis of BBB permeability assay techniques after CCI model of injury in rats.


Asunto(s)
Barrera Hematoencefálica , Lesiones Traumáticas del Encéfalo , Ratas , Animales , Masculino , Estudios Retrospectivos , Ratas Sprague-Dawley , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Proyectos de Investigación , Reproducibilidad de los Resultados , Lesiones Traumáticas del Encéfalo/diagnóstico , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Encéfalo , Permeabilidad , Colorantes/farmacología , Colorantes/uso terapéutico
10.
Eur J Drug Metab Pharmacokinet ; 47(6): 869-877, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36197653

RESUMEN

BACKGROUND AND OBJECTIVES: No treatment modalities have been identified to prevent neuron damage induced by traumatic brain injury (TBI). The objective of this study was to investigate whether ginsenoside Rb1 (GS-Rb1) could be utilized to exert neuroprotective effects in TBI. METHODS: Lateral fluid percussion injury (LFPI) was used to induce an experimental TBI model. Lewis rats were divided into a GS-Rb1 group (5, 10, 20 mg/kg, intraperitoneally injected daily), a sham group, and a vehicle group. Neurological impairments were assessed with brain water content, Evans blue extravasation, neurological deficit scores, and Morris water maze test. TUNEL and NeuN staining were utilized to detect neuron apoptosis. The relative expression of apoptosis- and autophagy-relevant molecules were assayed with real-time PCR and western blot. RESULTS: GS-Rb1 inhibited TBI-induced brain edema and Evans blue extravasation in a dose-dependent manner. Furthermore, GS-Rb1 improved neurological impairments with diminished neurological deficit scores, decreased escape latencies, increased time in the target quadrant, and increased number of platform site crossings. GS-Rb1 protected against neuron apoptosis with downregulated Bax expression and upregulated Bcl-2 expression. It was worth noting that TBI increased the LC3II/LC3I ratio and upregulated the relative expression of Beclin-1, Atg-7, and Atg-3; moreover, TBI downregulated the relative expression of P62. The administration of GS-Rb1 further strengthened the relative expression of autophagy-related molecules. CONCLUSIONS: GS-Rb1 alleviates neurological impairments induced by TBI with upregulated autophagy.


Asunto(s)
Autofagia , Lesiones Traumáticas del Encéfalo , Ratas , Animales , Azul de Evans/farmacología , Ratas Endogámicas Lew , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo
11.
J Pharmacol Sci ; 150(3): 135-145, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36184118

RESUMEN

Histamine is a major neurotransmitter and alleviates neuronal damage after ischemic injury via H2 receptors. Herein, we investigated the effects of H2 receptor agonists on the blood-brain barrier (BBB) disruption after traumatic brain injury (TBI). Male ddY mice were used to generate the TBI model, in which a fluid percussion injury (FPI) was induced by a hydraulic impact. The BBB disruption was evaluated using Evans blue extravasation. H2 receptor agonists, amthamine and dimaprit, were administered into the lateral cerebroventricle (i.c.v.) or tail vein (i.v.) from 3 hours to 3 days after FPI. The i.c.v. or i.v. administration of amthamine and dimaprit reduced FPI-induced Evans blue extravasation and promoted mRNA expression of vascular protective factors, including angiopoietin-1 and sonic hedgehog. The co-administration of ranitidine, a H2 receptor antagonist, inhibited these effects. Expression of the H2 receptor was observed in astrocytes and brain microvascular endothelial cells (BMECs) in the injured cortex. Treatment with amthamine and dimaprit promoted mRNA expression of vascular protective factors in astrocytes and BMECs. These results suggest that H2 receptor agonists alleviate TBI-induced BBB disruption by increasing the expression of vascular protective factors in astrocytes and BMECs.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Agonistas de los Receptores Histamínicos , Angiopoyetina 1/metabolismo , Angiopoyetina 1/farmacología , Animales , Barrera Hematoencefálica/metabolismo , Lesiones Traumáticas del Encéfalo/tratamiento farmacológico , Lesiones Traumáticas del Encéfalo/metabolismo , Dimaprit/metabolismo , Dimaprit/farmacología , Células Endoteliales/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Proteínas Hedgehog , Histamina/farmacología , Agonistas de los Receptores Histamínicos/metabolismo , Agonistas de los Receptores Histamínicos/farmacología , Masculino , Ratones , Factores Protectores , ARN Mensajero/metabolismo , Ranitidina/metabolismo , Ranitidina/farmacología , Receptores Histamínicos H2/genética , Receptores Histamínicos H2/metabolismo , Tiazoles
12.
Brain Behav Immun ; 106: 247-261, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36089218

RESUMEN

The α-synuclein (α-syn) is involved in methamphetamine (METH)-induced neurotoxicity. Neurons can transfer excessive α-syn to neighboring neurons and glial cells. The effects of α-syn aggregation in astrocytes after METH exposure on the blood-brain barrier (BBB) remains unclear. Our previous study demonstrated that nuclear receptor-related protein 1 (Nurr1), a member of the nuclear receptor family widely expressed in the brain, was involved in the process of METH-induced α-syn accumulated in astrocytes to activate neuroinflammation. The role Nurr1 plays in astrocyte-mediated neuroinflammation, which results in BBB injury induced by METH, remains uncertain. This study found that METH up-regulated α-syn expression in neurons extended to astrocytes, thereby eliciting astrocyte activation, increasing and decreasing IL-1ß, IL-6, TNF-α, and GDNF levels by down-regulating Nurr1 expression, and ultimately damaging the BBB. Specifically, the permeability of BBB to Evans blue and sodium fluorescein (NaF) increased; IgG deposits in the brain parenchyma increased; the Claudin5, Occludin, and PDGFRß levels decreased. Several ultrastructural pathological changes occurred in the BBB, such as abnormal cerebral microvascular diameter, astrocyte end-foot swelling, decreased pericyte coverage, and loss of tight junctions. However, knockout or inhibition of α-syn or astrocyte-specific overexpression of Nurr1 partially alleviated these symptoms and BBB injury. Moreover, the in vitro experiments confirmed that METH increased α-syn level in the primary cultured neurons, which could be further transferred to primary cultured astrocytes, resulting in decreased Nurr1 levels. The decreased Nurr1 levels mediated the increase of IL-1ß, IL-6, and TNF-α, and the decrease of GDNF, thereby changing the permeability to NaF, transendothelial electrical resistance, and Claudin5 and Occludin levels of primary cultured brain microvascular endothelial cells. Based on our findings, we proposed a new mechanism to elucidate METH-induced BBB injury and presented α-syn and Nurr1 as promising drug intervention targets to reduce BBB injury and resulting neurotoxicity in METH abusers.


Asunto(s)
Estimulantes del Sistema Nervioso Central , Metanfetamina , Síndromes de Neurotoxicidad , Astrocitos/metabolismo , Barrera Hematoencefálica/metabolismo , Estimulantes del Sistema Nervioso Central/farmacología , Células Endoteliales/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Fluoresceína/metabolismo , Fluoresceína/farmacología , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Factor Neurotrófico Derivado de la Línea Celular Glial/farmacología , Humanos , Inmunoglobulina G , Interleucina-6/metabolismo , Metanfetamina/metabolismo , Enfermedades Neuroinflamatorias , Neuronas/metabolismo , Síndromes de Neurotoxicidad/metabolismo , Ocludina/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , alfa-Sinucleína/metabolismo
13.
Acta Parasitol ; 67(4): 1514-1520, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35951222

RESUMEN

PURPOSE: Malarial parasites are susceptible to oxidative stress. The effects of α-tocopheryloxy acetic acid (α-TEA), a vitamin E analog, on infection by Plasmodium berghei ANKA and P. falciparum in mice and human red blood cells (RBCs), respectively, were examined in this study. METHODS: For in vivo studies in mice, RBCs infected with P. berghei ANKA were inoculated via intraperitoneal injection and α-TEA was administered to C57BL/6 J male mice after infection. The blood-brain barrier (BBB) permeability was examined by Evans blue staining in experimental cerebral malaria at 7 days after infection. The in vitro inhibitory effect of α-TEA on P. falciparum 3D7 (chloroquine-sensitive strain) and K1 (multidrug-resistant strain) was tested using a SYBR Green I-based assay. RESULTS: When 1.5% α-TEA was administered for 14 days after infection, 88% of P. berghei ANKA-infected mice survived during the experimental period. Nevertheless, all the control mice died within 12 days of infection. Furthermore, the Evans blue intensity in α-TEA-treated mice brains was less than that in untreated mice, indicating that α-TEA might inhibit the destruction of the BBB and progression of cerebral malaria. The in vitro experiment revealed that α-TEA inhibited the proliferation of both the 3D7 and K1 strains. CONCLUSION: This study showed that α-TEA is effective against murine and human malaria in vivo and in vitro, respectively. Although α-TEA alone has a sufficient antimalarial effect, future research could focus on the structure-activity relationship to achieve better pharmacokinetics and decrease the cytotoxicity and/or the combined effect of α-TEA with existing drugs. In addition, the prophylactic antimalarial activity of premedication with α-TEA may also be an interesting perspective in the future.


Asunto(s)
Antimaláricos , Malaria Cerebral , Malaria Falciparum , Humanos , Ratones , Masculino , Animales , Plasmodium berghei , Antimaláricos/farmacología , Antimaláricos/uso terapéutico , Malaria Cerebral/tratamiento farmacológico , Malaria Cerebral/parasitología , Ácido Acético/farmacología , Ácido Acético/uso terapéutico , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Ratones Endogámicos C57BL , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum
14.
Brain Behav ; 12(9): e2687, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35957622

RESUMEN

BACKGROUND: Subarachnoid hemorrhage (SAH) is a severe acute neurological disorder. SAH causes neuroinflammation and leads to early brain injury (EBI) and secondary injury. MicroRNAs are crucial regulators in a variety of neurological diseases. This study was performed to decipher how miR-340-5p functions in SAH. METHODS: An experimental mouse model with SAH was established by the intravascular perforation, and the in vitro SAH model was constructed by exposing cocultured primary neurons and microglia to oxyhemoglobin. After overexpression of miR-340-5p in mice, the neurobehavioral disorders were evaluated by Garcia test; brain edema was evaluated by wet-dry method; blood-brain barrier (BBB) damage was detected with Evan's blue staining; levels of inflammatory cytokines were detected with enzyme-linked immunosorbent assay. After miR-340-5p was transfected in to microglia, Iba-1 expression was detected by Western blot, and neuronal apoptosis were detected with flow cytometry. The targeting relationship between miR-340-5p and STING was verified by dual-luciferase reporter gene assay and RNA immunoprecipitation assay. RESULTS: MiR-340-5p was significantly inhibited in the brain tissues of mice with SAH and microglia of SAH model, and neurological impairment, brain edema, BBB injury, and neuroinflammation were significantly alleviated in mice after overexpressing miR-340-5p. STING was identified as a target of miR-340-5p, and STING overexpression could counteract the effects of miR-340-5p overexpression on neurons. CONCLUSION: MiR-340-5p can attenuate EBI caused by SAH-induced neuroinflammation by inhibiting STING.


Asunto(s)
Edema Encefálico , Lesiones Encefálicas , MicroARNs , Hemorragia Subaracnoidea , Animales , Edema Encefálico/metabolismo , Lesiones Encefálicas/complicaciones , Citocinas/metabolismo , Azul de Evans/farmacología , Proteínas de la Membrana , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Enfermedades Neuroinflamatorias , Neuronas/metabolismo , Oxihemoglobinas/metabolismo , Transducción de Señal , Hemorragia Subaracnoidea/complicaciones , Hemorragia Subaracnoidea/genética , Hemorragia Subaracnoidea/metabolismo
15.
J Neuroinflammation ; 19(1): 189, 2022 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-35842640

RESUMEN

BACKGROUND: Nafamostat mesylate (nafamostat, NM) is an FDA-approved serine protease inhibitor that exerts anti-neuroinflammation and neuroprotective effects following rat spinal cord injury (SCI). However, clinical translation of nafamostat has been limited by an unclear administration time window and mechanism of action. METHODS: Time to first dose of nafamostat administration was tested on rats after contusive SCI. The optimal time window of nafamostat was screened by evaluating hindlimb locomotion and electrophysiology. As nafamostat is a serine protease inhibitor known to target thrombin, we used argatroban (Arg), a thrombin-specific inhibitor, as a positive control in the time window experiments. Western blot and immunofluorescence of thrombin expression level and its enzymatic activity were assayed at different time points, as well its receptor, the protease activated receptor 1 (PAR1) and downstream protein matrix metalloproteinase-9 (MMP9). Blood-spinal cord barrier (BSCB) permeability leakage indicator Evans Blue and fibrinogen were analyzed along these time points. The infiltration of peripheral inflammatory cell was observed by immunofluorescence. RESULTS: The optimal administration time window of nafamostat was 2-12 h post-injury. Argatroban, the thrombin-specific inhibitor, had a similar pattern. Thrombin expression peaked at 12 h and returned to normal level at 7 days post-SCI. PAR1, the thrombin receptor, and MMP9 were significantly upregulated after SCI. The most significant increase of thrombin expression was detected in vascular endothelial cells (ECs). Nafamostat and argatroban significantly downregulated thrombin and MMP9 expression as well as thrombin activity in the spinal cord. Nafamostat inhibited thrombin enrichment in endothelial cells. Nafamostat administration at 2-12 h after SCI inhibited the leakage of Evans Blue in the epicenter and upregulated tight junction proteins (TJPs) expression. Nafamostat administration 8 h post-SCI effectively inhibited the infiltration of peripheral macrophages and neutrophils to the injury site. CONCLUSIONS: Our study provides preclinical information of nafamostat about the administration time window of 2-12 h post-injury in contusive SCI. We revealed that nafamostat functions through inhibiting the thrombin-mediated BSCB breakdown and subsequent peripheral immune cells infiltration.


Asunto(s)
Metaloproteinasa 9 de la Matriz , Traumatismos de la Médula Espinal , Animales , Benzamidinas , Barrera Hematoencefálica/metabolismo , Células Endoteliales/metabolismo , Azul de Evans/metabolismo , Azul de Evans/farmacología , Guanidinas , Metaloproteinasa 9 de la Matriz/metabolismo , Ratas , Ratas Sprague-Dawley , Receptor PAR-1/metabolismo , Inhibidores de Serina Proteinasa/farmacología , Inhibidores de Serina Proteinasa/uso terapéutico , Médula Espinal , Traumatismos de la Médula Espinal/tratamiento farmacológico , Traumatismos de la Médula Espinal/metabolismo , Trombina/metabolismo
16.
Exp Eye Res ; 217: 108977, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35139333

RESUMEN

Loss-of-function mutations in the Wnt co-receptor, low-density lipoprotein receptor-related protein 5 (LRP5), result in familial exudative vitreoretinopathy (FEVR), osteoporosis-pseudoglioma syndrome (OPPG), and Norrie disease. CRISPR/Cas9 gene editing was used to produce rat strains deficient in Lrp5. The purpose of this study was to validate this rat model for studies of hypovascular, exudative retinopathies. The retinal vasculature of wildtype and Lrp5 knockout rats was stained with Giffonia simplifolia isolectin B4 and imaged by fluorescence microscopy. Effects on retinal structure were investigated by histology. The integrity of the blood-retina barrier was analyzed by measurement of permeability to Evans blue dye and staining for claudin-5. Retinas were imaged by fundus photography and SD-OCT, and electroretinograms were recorded. Lrp5 gene deletion led to sparse superficial retinal capillaries and loss of the deep and intermediate plexuses. Autofluorescent exudates were observed and are correlated with increased Evans blue permeability and absence of claudin-5 expression in superficial vessels. OCT images show pathology similar to OCT of humans with FEVR, and retinal thickness is reduced by 50% compared to wild-type rats. Histology and OCT reveal that photoreceptor and outer plexiform layers are absent. The retina failed to demonstrate an ERG response. CRISPR/Cas9 gene-editing produced a predictable rat Lrp5 knockout model with extensive defects in the retinal vascular and neural structure and function. This rat model should be useful for studies of exudative retinal vascular diseases involving the Wnt and norrin pathways.


Asunto(s)
Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad , Retina , Animales , Claudina-5/biosíntesis , Claudina-5/genética , Azul de Evans/farmacología , Vitreorretinopatías Exudativas Familiares/genética , Vitreorretinopatías Exudativas Familiares/metabolismo , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/genética , Proteína-5 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Mutación , Ratas , Retina/metabolismo , Relación Estructura-Actividad
17.
Int J Neurosci ; 132(11): 1061-1071, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33287606

RESUMEN

Backgroud: Hyperhomocysteinemia (HHcy) is implicated in various neurovascular disorders including vascular dementia, subarachnoid hemorrhage and stroke. Elevated homocysteine (Hcy) levels are associated with increased oxidative stress and compromised blood-brain barrier (BBB) integrity. Hydrogen sulfide (H2S) has recently emerged as potent neuroprotective molecule in various neurological conditions including those associated with HHcy. The present study evaluates the protective effect of sodium hydrogen sulfide (NaHS; a source of H2S) on HHcy-induced BBB dysfunction and underpin molecular mechanisms.Materials and methods: Supplementation of NaHS restored the increased BBB permeability in the cortex and hippocampus of HHcy animals assessed in terms of diffused sodium fluorescein and Evans blue tracer dyes in the brain. Activity of matrix metalloproteinases (MMPs) assessed by gelatinase activity and in situ gelatinase assay was restored to the normal in the cortex and hippocampus of HHcy animals supplemented with NaHS.Results: Application of gelatin zymography revealed that specifically MMP-9 activity was increased in the cortex and hippocampus of HHcy animals, which was inhibited by NaHS supplementation. Real-time RT-PCR analysis showed that NaHS administration also decreased mRNA expression of MMP-9 in the hippocampus of HHcy animals. NaHS supplementation was further observed to reduce water retention in the brain regions of Hcy treated animals.Conclusion: Taken together, these findings suggest that NaHS supplementation ameliorates HHcy-induced BBB permeability and brain edema by inhibiting the mRNA expression and activity of MMP-9. Therefore, H2S and H2S releasing drugs may be used as a novel therapeutic approach to treat HHcy-associated neurovascular disorders.


Asunto(s)
Sulfuro de Hidrógeno , Hiperhomocisteinemia , Animales , Sulfuro de Hidrógeno/farmacología , Sulfuro de Hidrógeno/uso terapéutico , Sulfuro de Hidrógeno/metabolismo , Hiperhomocisteinemia/complicaciones , Hiperhomocisteinemia/tratamiento farmacológico , Barrera Hematoencefálica , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/farmacología , Metaloproteinasa 9 de la Matriz/uso terapéutico , Azul de Evans/metabolismo , Azul de Evans/farmacología , Azul de Evans/uso terapéutico , Fluoresceína/metabolismo , Fluoresceína/farmacología , Fluoresceína/uso terapéutico , Gelatina/metabolismo , Gelatina/farmacología , Gelatina/uso terapéutico , Permeabilidad , ARN Mensajero/metabolismo , Sodio , Colorantes/metabolismo , Colorantes/farmacología , Colorantes/uso terapéutico , Homocisteína , Agua/metabolismo , Agua/farmacología
18.
Sci Rep ; 11(1): 6521, 2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33753753

RESUMEN

Drug delivery in diffuse intrinsic pontine glioma is significantly limited by the blood-brain barrier (BBB). Focused ultrasound (FUS), when combined with the administration of microbubbles can effectively open the BBB permitting the entry of drugs across the cerebrovasculature into the brainstem. Given that the utility of FUS in brainstem malignancies remains unknown, the purpose of our study was to determine the safety and feasibility of this technique in a murine pontine glioma model. A syngeneic orthotopic model was developed by stereotactic injection of PDGF-B+PTEN-/-p53-/- murine glioma cells into the pons of B6 mice. A single-element, spherical-segment 1.5 MHz ultrasound transducer driven by a function generator through a power amplifier was used with concurrent intravenous microbubble injection for tumor sonication. Mice were randomly assigned to control, FUS and double-FUS groups. Pulse and respiratory rates were continuously monitored during treatment. BBB opening was confirmed with gadolinium-enhanced MRI and Evans blue. Kondziela inverted screen testing and sequential weight lifting measured motor function before and after sonication. A subset of animals were treated with etoposide following ultrasound. Mice were either sacrificed for tissue analysis or serially monitored for survival with daily weights. FUS successfully caused BBB opening while preserving normal cardiorespiratory and motor function. Furthermore, the degree of intra-tumoral hemorrhage and inflammation on H&E in control and treated mice was similar. There was also no difference in weight loss and survival between the groups (p > 0.05). Lastly, FUS increased intra-tumoral etoposide concentration by more than fivefold. FUS is a safe and feasible technique for repeated BBB opening and etoposide delivery in a preclinical pontine glioma model.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Neoplasias del Tronco Encefálico/tratamiento farmacológico , Sistemas de Liberación de Medicamentos , Glioma/tratamiento farmacológico , Animales , Transporte Biológico/efectos de los fármacos , Tronco Encefálico/diagnóstico por imagen , Tronco Encefálico/efectos de los fármacos , Neoplasias del Tronco Encefálico/diagnóstico por imagen , Neoplasias del Tronco Encefálico/genética , Neoplasias del Tronco Encefálico/patología , Modelos Animales de Enfermedad , Etopósido/farmacología , Azul de Evans/farmacología , Gadolinio/farmacología , Glioma/diagnóstico por imagen , Glioma/genética , Glioma/patología , Humanos , Imagen por Resonancia Magnética , Ratones , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/farmacología , Puente/diagnóstico por imagen , Puente/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/genética , Proteínas Proto-Oncogénicas c-sis/farmacología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/farmacología , Ultrasonografía
19.
mBio ; 12(2)2021 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-33785634

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a recently emerged virus that causes coronavirus infectious disease 2019 (COVID-19). SARS-CoV-2 spike protein, like SARS-CoV-1, uses the angiotensin converting enzyme 2 (ACE2) as a cellular receptor to initiate infection. Compounds that interfere with the SARS-CoV-2 spike protein receptor binding domain protein (RBD)-ACE2 receptor interaction may function as entry inhibitors. Here, we used a dual strategy of molecular docking and surface plasmon resonance (SPR) screening of compound libraries to identify those that bind to human ACE2 or the SARS-CoV-2 spike protein receptor binding domain (RBD). Molecular modeling screening interrogated 57,641 compounds and focused on the region of ACE2 that is engaged by RBD of the SARS-CoV-2 spike glycoprotein and vice versa. SPR screening used immobilized human ACE2 and SARS-CoV-2 Spike protein to evaluate the binding of these proteins to a library of 3,141 compounds. These combined screens identified compounds from these libraries that bind at KD (equilibrium dissociation constant) <3 µM affinity to their respective targets, 17 for ACE2 and 6 for SARS-CoV-2 RBD. Twelve ACE2 binders and six of the RBD binders compete with the RBD-ACE2 interaction in an SPR-based competition assay. These compounds included registered drugs and dyes used in biomedical applications. A Vero-E6 cell-based SARS-CoV-2 infection assay was used to evaluate infection blockade by candidate entry inhibitors. Three compounds demonstrated dose-dependent antiviral in vitro potency-Evans blue, sodium lifitegrast, and lumacaftor. This study has identified potential drugs for repurposing as SARS-CoV-2 entry inhibitors or as chemical scaffolds for drug development.IMPORTANCE SARS-CoV-2, the causative agent of COVID-19, has caused more than 60 million cases worldwide with almost 1.5 million deaths as of November 2020. Repurposing existing drugs is the most rapid path to clinical intervention for emerging diseases. Using an in silico screen of 57,641 compounds and a biophysical screen of 3,141 compounds, we identified 22 compounds that bound to either the angiotensin converting enzyme 2 (ACE2) and/or the SARS-CoV-2 spike protein receptor binding domain (SARS-CoV-2 spike protein RBD). Nine of these drugs were identified by both screening methods. Three of the identified compounds, Evans blue, sodium lifitegrast, and lumacaftor, were found to inhibit viral replication in a Vero-E6 cell-based SARS-CoV-2 infection assay and may have utility as repurposed therapeutics. All 22 identified compounds provide scaffolds for the development of new chemical entities for the treatment of COVID-19.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Antivirales/farmacología , Tratamiento Farmacológico de COVID-19 , Glicoproteína de la Espiga del Coronavirus/metabolismo , Acoplamiento Viral/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Aminopiridinas/farmacología , Animales , Benzodioxoles/farmacología , Línea Celular , Chlorocebus aethiops , Evaluación Preclínica de Medicamentos , Reposicionamiento de Medicamentos , Azul de Evans/farmacología , Humanos , Simulación del Acoplamiento Molecular , Fenilalanina/análogos & derivados , Fenilalanina/farmacología , Unión Proteica/efectos de los fármacos , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Sulfonas/farmacología , Resonancia por Plasmón de Superficie , Células Vero
20.
Cell Mol Neurobiol ; 41(2): 293-307, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-32382851

RESUMEN

The main pathological features of ischemic stroke include neuronal damage and blood-brain barrier (BBB) dysfunction. Previous studies have shown that Evans Blue, a dye used to probe BBB integrity, could enter the brain only during the pathological status of ischemic stroke, indicating the potential pathologically activated therapeutic use of this chemical to treat ischemic stroke. In this study, we have reported that Evans Blue could produce in vitro neuroprotective effects against iodoacetic acid (IAA)-induced hypoxia neuronal death in HT22 cells. We further found that P2X purinoreceptor 4 (P2X4R), a subtype of ATP-gated cation channel, was expressed in HT22 cells. Evans Blue could prevent IAA-induced increase of P2X4R mRNA and protein expression. Interestingly, shRNA of P2X4R could protect against IAA-induced activation of p38, and SB203580, a specific inhibitor of p38, could reverse IAA-induced neurotoxicity, indicating that p38 is a downstream signaling molecule of P2X4R. Molecular docking analysis further demonstrated the possible interaction between Evans Blue and the ATP binding site of P2X4R. Most importantly, pre-treatment of Evans Blue could largely reduce neurological and behavioral abnormity, and decrease brain infarct volume in middle cerebral artery occlusion/reperfusion (MCAO) rats. All these results strongly suggested that Evans Blue could exert neuroprotective effects via inhibiting the P2X4R/p38 pathway, possibly by acting on the ATP binding site of P2X4R, indicating that Evans Blue might be further developed as a pathologically activated therapeutic drug against ischemic stroke.


Asunto(s)
Azul de Evans/farmacología , Fármacos Neuroprotectores/farmacología , Receptores Purinérgicos P2X4/metabolismo , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Conducta Animal/efectos de los fármacos , Infarto Encefálico/complicaciones , Infarto Encefálico/patología , Muerte Celular/efectos de los fármacos , Línea Celular , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Ácido Yodoacético , Masculino , Ratones , Modelos Biológicos , Simulación del Acoplamiento Molecular , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...