Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Neuroinflammation ; 21(1): 70, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38515139

RESUMEN

Myeloperoxidase (MPO) plays critical role in the pathology of cerebral ischemia-reperfusion (I/R) injury via producing hypochlorous acid (HOCl) and inducing oxidative modification of proteins. High-mobility group box 1 (HMGB1) oxidation, particularly disulfide HMGB1 formation, facilitates the secretion and release of HMGB1 and activates neuroinflammation, aggravating cerebral I/R injury. However, the cellular sources of MPO/HOCl in ischemic brain injury are unclear yet. Whether HOCl could promote HMGB1 secretion and release remains unknown. In the present study, we investigated the roles of microglia-derived MPO/HOCl in mediating HMGB1 translocation and secretion, and aggravating the brain damage and blood-brain barrier (BBB) disruption in cerebral I/R injury. In vitro, under the co-culture conditions with microglia BV cells but not the single culture conditions, oxygen-glucose deprivation/reoxygenation (OGD/R) significantly increased MPO/HOCl expression in PC12 cells. After the cells were exposed to OGD/R, MPO-containing exosomes derived from BV2 cells were released and transferred to PC12 cells, increasing MPO/HOCl in the PC12 cells. The HOCl promoted disulfide HMGB1 translocation and secretion and aggravated OGD/R-induced apoptosis. In vivo, SD rats were subjected to 2 h of middle cerebral artery occlusion (MCAO) plus different periods of reperfusion. Increased MPO/HOCl production was observed at the reperfusion stage, accomplished with enlarged infarct volume, aggravated BBB disruption and neurological dysfunctions. Treatment of MPO inhibitor 4-aminobenzoic acid hydrazide (4-ABAH) and HOCl scavenger taurine reversed those changes. HOCl was colocalized with cytoplasm transferred HMGB1, which was blocked by taurine in rat I/R-injured brain. We finally performed a clinical investigation and found that plasma HOCl concentration was positively correlated with infarct volume and neurological deficit scores in ischemic stroke patients. Taken together, we conclude that ischemia/hypoxia could activate microglia to release MPO-containing exosomes that transfer MPO to adjacent cells for HOCl production; Subsequently, the production of HOCl could mediate the translocation and secretion of disulfide HMGB1 that aggravates cerebral I/R injury. Furthermore, plasma HOCl level could be a novel biomarker for indexing brain damage in ischemic stroke patients.


Asunto(s)
Lesiones Encefálicas , Isquemia Encefálica , Proteína HMGB1 , Accidente Cerebrovascular Isquémico , Daño por Reperfusión , Humanos , Ratas , Animales , Ácido Hipocloroso , Microglía/metabolismo , Proteína HMGB1/metabolismo , Ratas Sprague-Dawley , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Barrera Hematoencefálica/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/patología , Neuronas/metabolismo , Daño por Reperfusión/metabolismo , Peroxidasa/metabolismo , Taurina , Disulfuros
2.
CNS Neurosci Ther ; 30(3): e14689, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38516831

RESUMEN

AIMS: Chronic alcohol exposure leads to persistent neurological disorders, which are mainly attributed to neuroinflammation and apoptosis. Stimulator of IFN genes (STING) is essential in the cytosolic DNA sensing pathway and is involved in inflammation and cellular death processes. This study was to examine the expression pattern and biological functions of STING signaling in alcohol use disorder (AUD). METHODS: Cell-free DNA was extracted from human and mouse plasma. C57BL/6J mice were given alcohol by gavage for 28 days, and behavior tests were used to determine their mood and cognition. Cultured cells were treated with ethanol for 24 hours. The STING agonist DMXAA, STING inhibitor C-176, and STING-siRNA were used to intervene the STING. qPCR, western blot, and immunofluorescence staining were used to assess STING signaling, inflammation, and apoptosis. RESULTS: Circulating cell-free mitochondrial DNA (mtDNA) was increased in individuals with AUD and mice chronically exposed to alcohol. Upregulation of STING signaling under alcohol exposure led to inflammatory responses in BV2 cells and mitochondrial apoptosis in PC12 cells. DMXAA exacerbated alcohol-induced cognitive impairment and increased the activation of microglia, neuroinflammation, and apoptosis in the medial prefrontal cortex (mPFC), while C-176 exerted neuroprotection. CONCLUSION: Activation of STING signaling played an essential role in alcohol-induced inflammation and mitochondrial apoptosis in the mPFC. This study identifies STING as a promising therapeutic target for AUD.


Asunto(s)
Disfunción Cognitiva , Enfermedades Neuroinflamatorias , Humanos , Ratones , Animales , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Inflamación/inducido químicamente , Inflamación/metabolismo , Etanol/toxicidad , ADN Mitocondrial/metabolismo , Apoptosis , Disfunción Cognitiva/inducido químicamente
3.
Neurobiol Dis ; 184: 106210, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37352984

RESUMEN

The progressive neurodegenerative disease amyotrophic lateral sclerosis (ALS) is caused by a decline in motor neuron function, resulting in worsened motor impairments, malnutrition, respiratory failure and mortality, and there is a lack of effective clinical treatments. The exact mechanism of motor neuronal degeneration remains unclear. Previously, we reported that ferroptosis, which is characterized by the accumulation of lipid peroxide and glutathione depletion in an iron-dependent manner, contributed to motor neuronal death in ALS cell models with the hSOD1G93A (human Cu/Zn-superoxide dismutase) gene mutation. In this study, we further explored the role of ferroptosis in motor neurons and its regulation in mutant hSOD1G93A cell and mouse models. Our results showed that ferroptosis was activated in hSOD1G93A NSC-34 cells and mouse models, which was accompanied by decreased nuclear retention of nuclear factor erythroid 2-related factor 2 (NRF2) and downregulation of solute carrier family 7 member 11 (SLC7A11) and glutathione peroxidase 4 (GPX4) levels. Moreover, RTA-408, an NRF2 activator, inhibited ferroptosis in hSOD1G93A NSC-34 cells by upregulating the protein expression of SLC7A11 and GPX4. Moreover, hSOD1G93A mice treated with RTA-408 showed obvious improvements in body weight and motor function. Our study demonstrated that ferroptosis contributed to the toxicity of motor neurons and that activating NRF2 could alleviate neuronal degeneration in ALS with the hSOD1G93A mutation.


Asunto(s)
Esclerosis Amiotrófica Lateral , Ferroptosis , Enfermedades Neurodegenerativas , Animales , Humanos , Ratones , Esclerosis Amiotrófica Lateral/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , Neuronas Motoras/metabolismo , Mutación/genética , Enfermedades Neurodegenerativas/metabolismo , Neuroprotección , Factor 2 Relacionado con NF-E2/metabolismo , Superóxido Dismutasa/genética , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo
4.
Brain Behav Immun ; 110: 60-79, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36754245

RESUMEN

BACKGROUND: Chronic alcohol exposure (CAE) during late adolescence increases the risk of anxiety development. Alcohol-induced prefrontal cortex (PFC) microglial activation, characterized by morphological changes and increased associations with neurons, plays a critical role in the pathogenesis of anxiety. Alcohol exposure increases NLRP3 inflammasome expression, increasing cytokine secretion by activated microglia. Cannabinoid type 2 receptor (CB2R), an essential receptor of the endocannabinoid system, regulates microglial activation and neuroinflammatory reactions. We aimed to investigate the role of CB2R activation in ameliorating late adolescent CAE-induced anxiety-like behaviors and microglial activation in C57BL/6J mice. METHODS: Six-week-old C57BL/6J mice were acclimated for 7 days and then were administered alcohol by gavage (4 g/kg, 25 % w/v) for 28 days. The mice were intraperitoneally injected with the specific CB2R agonist AM1241 1 h before alcohol treatment. Anxiety-like behaviors during withdrawal were assessed by open field test and elevated plus maze test 24 h after the last alcohol administration. Microglial activation, microglia-neuron interactions, and CB2R and NLRP3 inflammasome-related molecule expression in the PFC were measured using immunofluorescence, immunohistochemical, qPCR, and Western blotting assays. Microglial morphology was evaluated by Sholl analysis and the cell body-to-total cell size index. Additionally, N9 microglia were activated by LPS in vitro, and the effects of AM1241 on NLRP3 and N9 microglial activation were investigated. RESULTS: After CAE, mice exhibited severe anxiety-like behaviors during withdrawal. CAE induced obvious microglia-neuron associations, and increased expression of microglial activation markers, CB2R, and NLRP3 inflammasome-related molecules in the PFC. Microglia also showed marked filament retraction and reduction and cell body enlargement after CAE. AM1241 treatment ameliorated anxiety-like behaviors in CAE model mice, and it prevented microglial morphological changes, reduced microglial activation marker expression, and suppressed the microglial NLRP3 inflammasome activation and proinflammatory cytokine secretion induced by CAE. AM1241 suppressed the LPS-induced increase in NLRP3 inflammasome-related molecules, IL-1ß release, and M1 phenotype markers (iNOS and CD86) in N9 cell, which was reversed by CB2R antagonist treatment. CONCLUSIONS: CAE caused anxiety-like behaviors in late adolescent mice at least partly by inducing microglial activation and increasing microglia-neuron associations in the PFC. CB2R activation ameliorated these effects by preventing morphological changes and suppressing NLRP3 inflammasome activation in PFC microglia.


Asunto(s)
Ansiedad , Etanol , Inflamasomas , Microglía , Corteza Prefrontal , Receptor Cannabinoide CB2 , Animales , Ratones , Consumo de Bebidas Alcohólicas/efectos adversos , Ansiedad/etiología , Ansiedad/metabolismo , Cannabinoides/farmacología , Citocinas/metabolismo , Etanol/efectos adversos , Inflamasomas/metabolismo , Lipopolisacáridos/farmacología , Ratones Endogámicos C57BL , Microglía/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Corteza Prefrontal/metabolismo , Receptor Cannabinoide CB2/metabolismo , Receptores de Cannabinoides/metabolismo
5.
Mol Neurobiol ; 60(5): 2749-2766, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36717480

RESUMEN

The pyrin domain-containing protein 3 (NLRP3) inflammasome drives the profound cerebral ischemia and reperfusion injury (I/R) and mediates the secretion of IL-1ß (interleukin-1ß), which exerts a subsequent cascade of inflammatory injury. The NLRP3-activated-microglial manipulation in adjacent neuronal and endothelial NLRP3 activation has been confirmed in our previous studies. In the present study, we extended the cognition of how microglia mediated neuronal and endothelial NLRP3-IL-1ß signaling during cerebral ischemia and reperfusion injury. In vitro, Neuro-2a and bEND3 cells were cultured alone or co-cultured with BV2 cells and oxygen-glucose deprivation/reoxygenation (OGD/R) was performed. In vivo, transient middle cerebral artery occlusion (tMCAO) rat models and lentiviral silencing targeting IL-1R1 were performed. The NLRP3 inflammasome activation was evaluated by enzyme-linked immunosorbent assay, western blotting, immunoprecipitation, immunohistochemistry, and immunofluorescence. In the co-culture system after OGD/R treatment, NLRP3 inflammasomes in neurons and endothelial cells were activated by microglial IL-1ß via IL-1ß/IL-1R1/TRAF6 signaling pathway, with the basal protein level of NLRP3. In addition, ruptured lysosomes engulfing ASC specks which were possibly secreted from microglia triggered the enhanced NLRP3 expression. In cortices of tMCAO rats at 24 h of reperfusion, silencing IL-1R1, mainly presented in neurons and endothelial cells, was efficient to block the subsequent inflammatory damage and leukocyte brain infiltration, leading to better neurological outcome. Neuronal and endothelial NLRP3 inflammasomes were activated by microglia in cerebral ischemia and reperfusion injury mainly via IL-1ß/IL-1R1/TRAF6 signaling, which might be therapeutically targetable.


Asunto(s)
Isquemia Encefálica , Daño por Reperfusión , Animales , Ratas , Isquemia Encefálica/metabolismo , Células Endoteliales/metabolismo , Infarto de la Arteria Cerebral Media/complicaciones , Infarto de la Arteria Cerebral Media/metabolismo , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Daño por Reperfusión/metabolismo , Factor 6 Asociado a Receptor de TNF/metabolismo
6.
Ying Yong Sheng Tai Xue Bao ; 34(12): 3347-3356, 2023 Dec.
Artículo en Chino | MEDLINE | ID: mdl-38511374

RESUMEN

Establishing the remote sensing yield estimation model of wheat-maize rotation cultivated land can timely and accurately estimate the comprehensive grain yield. Taking the winter wheat-summer maize rotation cultivated land in Caoxian County, Shandong Province, as test object, using the Sentinel-2 images from 2018 to 2019, we compared the time-series feature classification based on QGIS platform and support vector machine algorithm to select the best method and extract sowing area of wheat-maize rotation cultivated land. Based on the correlation between wheat and maize vegetation index and the statistical yield, we screened the sensitive vegetation indices and their growth period, and obtained the vegetation index integral value of the sensitive spectral period by using the Newton-trapezoid integration method. We constructed the multiple linear regression and three machine learning (random forest, RF; neural network model, BP; support vector machine model, SVM) models based on the integral value combination to get the best and and optimized yield estimation model. The results showed that the accuracy rate of extracting wheat and maize sowing area based on time-series features using QGIS platform reached 94.6%, with the overall accuracy and Kappa coefficient were 5.9% and 0.12 higher than those of the support vector machine algorithm, respectively. The remote sensing yield estimation in sensitive spectral period was better than that in single growth period. The normalized differential vegetation index and ratio vegetation index integral group of wheat and enhanced vegetation index and structure intensify pigment vegetable index integral group of maize could more effectively aggregate spectral information. The optimal combination of vegetation index integral was difference, and the fitting accuracy of machine learning algorithm was higher than that of empirical statistical model. The optimal yield estimation model was the difference value group-random forest (DVG-RF) model of machine learning algorithm (R2=0.843, root mean square error=2.822 kg·hm-2), with a yield estimation accuracy of 93.4%. We explored the use of QGIS platform to extract the sowing area, and carried out a systematical case study on grain yield estimation method of wheat-maize rotation cultivated land. The established multi-vegetation index integral combination model was effective and feasible, which could improve accuracy and efficiency of yield estimation.


Asunto(s)
Triticum , Zea mays , Tecnología de Sensores Remotos/métodos , Grano Comestible , China
7.
Am J Drug Alcohol Abuse ; 48(5): 562-572, 2022 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-35838410

RESUMEN

B a ckground: Circular RNAs (circRNAs) have been crucially implicated in various diseases, however, their involvement in chronic intermittent ethanol (CIE) exposure remains unclear.O bjective: The present study was conducted to evaluate the circular RNA expression alteration in brain samples and to identify the molecular mechanisms underlying chronic intermittent ethanol exposure.M ethods: Male C57BL/6J mice (10 for each group) were given 4 weeks of chronic intermittent ethanol exposure. Whole brain samples were collected for high-throughput sequencing and circRNA bioinformatic analysis. Real-time quantitative PCR (RI-qPCR) and agarose electrophoresis were used to validate the differentially expressed circRNAs. Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes pathway (KEGG) analysis were performed. A p level < 0.05 was considered statistically significant.R esults: Compared with the control group and baseline values, the CIE group showed a significant increase in ethanol intake. High-throughput sequencing revealed 399 significantly different circRNAs in CIE mice, including 150 up-regulated circRNAs and 249 down-regulated circRNAs. GO analysis showed that the most significantly enriched term for biological process, cellular component, and molecular function were GO:0050885, GO:0016020 and GO:0005515, respectively. The most enriched pathways in KEGG analysis were GABAergic synapse (mmu04727), followed by retrograde endocannabinoid (eCB) signaling (mmu04723) and morphine addiction (mmu05032). Among the circRNAs, RT-qPCR confirmed 14 upregulated and 13 downregulated circRNAs in the brain tissues with 9 upregulated and 10 downregulated circRNAs being observed in blood samples.C onclusions: Our study suggests that chronic ethanol exposure upregulates or downregulates circRNAs in the brain, which, in turn, could alter neurotransmitter release and signal transduction.


Asunto(s)
Etanol , ARN Circular , Animales , Regulación hacia Abajo , Endocannabinoides , Perfilación de la Expresión Génica/métodos , Masculino , Ratones , Ratones Endogámicos C57BL , Neurotransmisores , ARN Circular/genética , Sefarosa , Regulación hacia Arriba
8.
J Neuroinflammation ; 19(1): 87, 2022 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-35414088

RESUMEN

BACKGROUND: Chronic morphine exposure induces immunosuppression in the peripheral and central nervous system, resulting in susceptibility of patients to invading pathogens. Mitophagy is a crucial regulator of inflammation, and dysregulated mitophagy may cause immunosuppression, but whether mitophagy is linked with morphine-induced immunosuppression in the brain remains unknown. NLRX1 is the only mitochondrially localized NOD family receptor protein which serves as a critical regulator in immunity and mitophagy activation, but it remains an enigma how NLRX1 functions in the crosstalk between microglial inflammatory defense and mitophagy in the presence of morphine. METHODS: Primary microglia and astrocytes, BV2 and MA cell lines were utilized. Mice were stimulated with repeated morphine treatment to mimic chronic morphine exposure, and activation of mitophagy, lysosomal functions, and inflammation were assayed in specific brain regions and immune organs with or without NLRX1-silencing. RESULTS: Morphine induced microglial mitophagy in a LC3 (microtubule-associated proteins light chain 3)-dependent manner, which was mediated by NLRX1. Contrastingly, morphine impaired lysosomal functions, including generation, acidification and mitophagosome-lysosome fusion, thus leading to insufficient mitophagy activation in microglia. NLRX1-silencing inhibited mitophagy activity and rescued lysosomal functions including generation and acidification in microglia. The NLRX1-mediated incomplete mitophagy in microglial cells contributed to immunosuppression and vulnerability towards pathogenic challenge after morphine treatment. In vivo, NLRX1-mediated microglial mitophagy activation by morphine was mainly located in the murine brain cortex, striatum, and cerebellum, where NLRX1 functioned as a negative immune regulator and facilitated septic shock. Collectively, microglial immune responses to septic shock were amenable to NLRX1 silencing in the brain with morphine treatment. CONCLUSION: Morphine activated insufficient mitophagy in microglia which was regulated by NLRX1, ultimately leading to host immunosuppression and susceptible conditions in the brain.


Asunto(s)
Mitofagia , Choque Séptico , Animales , Humanos , Terapia de Inmunosupresión , Ratones , Ratones Endogámicos NOD , Microglía/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Morfina/toxicidad , Choque Séptico/metabolismo , Choque Séptico/patología
9.
Am J Cancer Res ; 12(2): 893-906, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35261810

RESUMEN

RNA N6-methyladenosine (m6A) modification is gradually thought to be an active participant in the considerable biological processes of glioblastoma (GB), providing us with a novel insight for exploring this disease. However, the role of RNA m6A modification during the epithelial mesenchymal transition (EMT) or vasculogenic mimicry (VM) progression has not been investigated in GB. Here we performed a research to validate the impact exerted by AlkB homolog 5 (ALKBH5), one of "erasers" for RNA m6A and methyltransferase-like 3 (METTL3) which adds m6A modification to the RNAs on the progression of EMT and VM in GB. In this study, we demonstrate that the m6A levels of RNAs were reduced in GB cells and glioma tissues. Patients with high mRNA expression of ALKBH5 acquired relatively shorter median overall survival (OS) time, while patients with relatively high expression of MEETL3 prolonged their disease free survival. ALKBH5 enhanced GB cell proliferation and influenced cell cycle in vitro. Decreased RNA m6A methylation enhanced the progression of the EMT and VM in glioblastoma cells. ALKBH5 strengthened glioblastoma growth and enhanced the EMT and VM process of glioblastoma in vivo. Our study uncovers that RNA m6A methylation suppresses the process of EMT and VM in glioblastoma, providing a new perspective to seek for a potential therapeutic target for GB.

10.
Oxid Med Cell Longev ; 2022: 8217663, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35178161

RESUMEN

BACKGROUND: Oxidative stress and reactive oxygen species (ROS) are important in the pathogenesis of amyotrophic lateral sclerosis (ALS). Hypochlorous acid (HOCl) is a powerful oxidant of the reactive oxygen species (ROS) family. HOCl's role in the progress of ALS remains unclear due to the lack of an effective HOCl detection method. Cumulative evidence supports oxidative damage incurred by mutant hSOD1 contributing to motor neuron death; however, whether HOCl as well as its catalytic enzyme myeloperoxidase (MPO) function in the cell death of SOD1G93A ALS remains elusive. METHODS: The hSOD1WT and hSOD1G93A NSC-34 cell and SOD1G93A ALS mouse models were employed. With a novel fluorescent HOCl probe, HKOCl-3, we detected the expressions of HOCl and its catalytic enzyme, MPO, in the above models in vitro and in vivo. The regulation of MPO/HOCl by hSOD1G93A mutation and cell deaths by MPO/HOCl were also assayed, including apoptosis, ferroptosis, and autophagy. RESULTS: Our results showed that hSOD1G93A mutation promoted the activation of the MPO/HOCl pathway in SOD1G93A ALS cell models. The activation of MPO/HOCl pathways facilitated apoptosis and ferroptosis through increasing the Bax/Bcl-2 ratio and expression of caspase-3 or inhibiting the expressions of GPX4 and NQO1 and thus leading to irreversible lipid peroxidation. Overexpressed FSP1, a glutathione-independent suppressor, could ameliorate ferroptosis. In vivo, we demonstrated that the activation of the MPO/HOCl pathway occurred differently in motor neurons of the motor cortices, brain stems, and spinal cords in male and female SOD1G93A transgenic mice. In addition, inhibiting MPO improved the motor performance of SOD1G93A transgenic mice, as demonstrated by the rotarod test. CONCLUSIONS: We concluded that aggregation of mutant hSOD1 proteins contributed to activation of the MPO/HOCl pathway, triggering apoptosis and ferroptosis in motor neuronal deaths and exerting impaired motor performance.


Asunto(s)
Esclerosis Amiotrófica Lateral/terapia , Apoptosis/fisiología , Ferroptosis/fisiología , Neuronas Motoras/metabolismo , Peroxidasa/metabolismo , Superóxido Dismutasa-1/metabolismo , Esclerosis Amiotrófica Lateral/patología , Animales , Estudios de Casos y Controles , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Estrés Oxidativo , Especies Reactivas de Oxígeno , Transfección
11.
Mol Immunol ; 123: 74-87, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32438202

RESUMEN

BACKGROUND: Idebenone is a well-appreciated mitochondrial protectant while the mechanisms underlying the neuroprotection in cerebral ischemia and reperfusion (I/R) remain elusive. It has been manifested NLRP3 inflammasom activation contributed to I/R induced damage. It raises questions how exactly NLRP3 inflammasom was activated in microglia and neuron and whether idebenone reverses the process in I/R. METHODS: I/R rat model was utilized and BV2, primary microglia and PC12 cells were subjected to oxygen-glucose deprivation (OGD). Then, western-blotting, q-PCR, immunofluorescence staining, ELISA, flow cytometry and immunoprecipitation analysis were performed. RESULTS: We found ROS-NLRP3 singaling was activated in BV2 cells at OGD/R 24 h. Importantly, microglial NLRP3 activation was essential for NLRP3 activation in PC12 cells under microglial-neuronal co-culture circumstance, which has been confirmed to induced neuronal apoptosis. Further, we found mitochondrial dysfunction in OGD/R led to mt-DNA translocation as well as generation of mt-ROS, resulting cytosolic accumulation of oxidized mt-DNA. Ultimately, oxidized mt-DNA binding to NLRP3 contributed to further activation of NLRP3 and dramatically augmented inflammation in BV2 and PC12 cells. Furthermore, idebenone treatment inhibited the process, thus suppressing the NLRP3-mediated inflammatory injury after OGD/R. In vivo, NLRP3 was activated in microglia of I/R rats and inhibition of NLRP3 was observed in idebenone treatment group, which had less neurological deficit and less infarct volume. INTERPRETATION: Our data revealed the anti-inflammatory effects of idebenone via suppressing activation of NLRP3 and ameliorating NLRP3-mediating damage in I/R, which may provide new insight in therapeutic strategy for ischemic stroke.


Asunto(s)
Isquemia Encefálica , Encefalitis/prevención & control , Inflamasomas/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neuroprotección/efectos de los fármacos , Daño por Reperfusión , Ubiquinona/análogos & derivados , Animales , Animales Recién Nacidos , Isquemia Encefálica/complicaciones , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/inmunología , Isquemia Encefálica/metabolismo , Células Cultivadas , Encefalitis/etiología , Inflamasomas/metabolismo , Inflamasomas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Células PC12 , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/complicaciones , Daño por Reperfusión/tratamiento farmacológico , Daño por Reperfusión/inmunología , Daño por Reperfusión/metabolismo , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular/complicaciones , Accidente Cerebrovascular/tratamiento farmacológico , Accidente Cerebrovascular/metabolismo , Accidente Cerebrovascular/patología , Ubiquinona/farmacología , Ubiquinona/uso terapéutico
12.
Int J Neurosci ; 130(9): 857-864, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31928267

RESUMEN

Background and purpose: The optimal treatment for acute ischemic stroke with mild neurologic deficits is unclear. We aimed to compare the efficacy and safety of alteplase versus dual-antiplatelet therapy in acute minor stroke.Methods: We performed a retrospective cohort study of patients with minor ischemic stroke and National Institutes of Health Stroke Scale scores ≤5 presenting within 24 h from last seen normal. Patients were divided into intravenous alteplase or dual-antiplatelet therapy group. The primary outcome was a modified Rankin Scale (mRS) score of 0 or 1 at 90 days. Secondary outcomes included mRS score at 7 days, and composite outcome of vascular events within 90 days. The safety outcome was any intracranial hemorrhage (ICH) according to the ECASS II criteria. Clinical outcomes were compared using a multivariable logistic regression after adjusting for confounding factors. We then performed the propensity score matching as a sensitivity analysis.Results: Two hundred twenty-eight patients met the eligibility criteria were included for analysis between January 2015 and September 2018. In the aspirin-clopidogrel group, 109 patients (91.6%) achieved a favorable functional outcome at 3-month versus 85(78.0%) in the alteplase group (OR 4.463, 95%CI 1.708-11.662, p = .002). The difference of the composite outcome of vascular events were not statistical significance between the two groups (p > .05). Asymptomatic ICH occurred in 0.8% patients who received aspirin-clopidogrel, as compared with 3.7% patients in alteplase group (p = .030).Conclusions: Patients treated with dual-antiplatelet therapy with acute minor ischemic stroke had greater functional outcome at 3 months compared with patients who received alteplase therapy.Classification of evidence: This study provides Class IV evidence that dual-antiplatelet therapy is superior to alteplase for achieving a better functional outcome and does not increase the risk of hemorrhage in acute minor ischemic stroke.


Asunto(s)
Aspirina/farmacología , Clopidogrel/farmacología , Fibrinolíticos/farmacología , Hemorragias Intracraneales/inducido químicamente , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Evaluación de Resultado en la Atención de Salud , Inhibidores de Agregación Plaquetaria/farmacología , Activador de Tejido Plasminógeno/farmacología , Anciano , Aspirina/efectos adversos , Clopidogrel/efectos adversos , Quimioterapia Combinada , Femenino , Fibrinolíticos/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Inhibidores de Agregación Plaquetaria/efectos adversos , Estudios Retrospectivos , Índice de Severidad de la Enfermedad , Activador de Tejido Plasminógeno/efectos adversos
13.
J Cell Physiol ; 235(3): 2609-2618, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31502677

RESUMEN

Most circular RNAs (circRNAs) belong to a novel class of noncoding RNAs that are produced by back-splicing reactions, and they regulate physiological and pathophysiological processes in human disease. Although circRNA expression has been shown to be altered in the ischemic cerebral tissue in animal studies, the expression profile of circRNA in the patients with acute ischemic stroke (AIS) has not been investigated to date. In this investigation, high-throughput sequencing was carried out to compare the circRNA expression of peripheral blood mononuclear cells (PBMCs) from five patients with AIS and five healthy subjects. A total of 521 circRNAs were expressed differentially between the patients with AIS and healthy controls (p < .05, fold difference ≥2) including 373 upregulated circRNAs and 148 downregulated circRNAs in patients with AIS compared to controls. Thirteen candidate circRNAs were verified by quantitative real-time polymerase chain reaction (qRT-PCR). Bioinformatics analyses showed that these differentially expressed circRNAs were highly conserved, as well as eight circRNAs that were confirmed by qRT-PCR containing binding sites to multiple microRNAs. Kyoto Encyclopedia of Genes and Genomes pathway enrichment and gene ontology analyses indicated that the aberrantly expressed circRNAs participated in many pathophysiological processes of AIS, especially regarding inflammation and immunity. In conclusion, patients with AIS differentially express certain circRNAs in PBMCs, which may be diagnostic biomarkers or potential therapeutic targets.


Asunto(s)
Accidente Cerebrovascular Isquémico/patología , Leucocitos Mononucleares/citología , ARN Circular/análisis , ARN Circular/biosíntesis , Biomarcadores/análisis , Estudios de Casos y Controles , Biología Computacional , Femenino , Perfilación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Accidente Cerebrovascular Isquémico/mortalidad , Masculino , Persona de Mediana Edad , Estudios Prospectivos , ARN Circular/genética , Reacción en Cadena en Tiempo Real de la Polimerasa
14.
Brain Behav ; 9(10): e01398, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31532082

RESUMEN

OBJECTIVES: Approximately, half of the acute stroke patients with minor symptoms were excluded from thrombolysis in some randomized controlled trials (RCTs). There is little evidence on treating minor strokes with rt-PA. Here, we performed a systematic review and meta-analysis to assess the safety and efficacy of thrombolysis in these patients. METHODS: PubMed, Embase, Web of Science, and Cochrane Library were searched in July 2018. All available RCTs and retrospective comparative studies that compared thrombolysis with nonthrombolysis' for acute minor stroke (NIHSS ≤ 5) with quantitative outcomes were included. RESULTS: Ten studies, including a total of 4,333 patients, were identified. The risk of intracranial hemorrhage (ICH) was higher in the rt-PA group as compared with that in the non-rt-PA group (3.8% vs. 0.6%; p = .0001). However, there is no significant difference in the rate of mortality between the two groups (p = .96). The pooled rate of a good outcome in 90 days was 67.8% in those with rt-PA and 63.3% in those without rt-PA (p = .07). Heterogeneity was 43% between the studies (p = .08). After adjusting for the heterogeneity, thrombolysis was associated with good outcome (68.3% vs. 63.0%, OR 1.47; 95% CI 1.14-1.89; p = .003). In post hoc analyses, including only RCTs, the pooled rate of good outcome had no significant differences between the two groups (86.6% vs. 85.7%, 95% CI 0.44-3.17, p = .74; 87.4% vs. 91.9%, 95% CI 0.35-1.41, p = .32; before and after adjusting separately). CONCLUSIONS: Although thrombolysis might increase the risk of ICH based on existing studies, patients with acute minor ischemic stroke could still benefit from thrombolysis at 3 months from the onset.


Asunto(s)
Fibrinolíticos/uso terapéutico , Reperfusión/métodos , Accidente Cerebrovascular/tratamiento farmacológico , Terapia Trombolítica/métodos , Activador de Tejido Plasminógeno/uso terapéutico , Humanos , Masculino , Estudios Retrospectivos , Resultado del Tratamiento
15.
Brain Behav Immun ; 82: 63-75, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31376498

RESUMEN

Chronic excessive drinking leads to a wide spectrum of neurological disorders, including cognitive deficits, such as learning and memory impairment. However, the neurobiological mechanisms underlying these deleterious changes are still poorly understood. We conducted a comprehensive study to investigate the role and mechanism of autophagy in alcohol-induced memory impairment. To establish an ethanol-induced memory impairment mouse model, we allowed C57BL/6J mice intermittent access to 20% ethanol (four-bottle choice) to escalate ethanol drinking levels. Memory impairment was confirmed by a Morris water maze test. We found that mice exposed to EtOH (ethanol) and EtOH combined with the autophagy inhibitor 3-methyladenine (3-MA) showed high alcohol intake and blood alcohol concentration. We confirmed that the EtOH group exhibited notable memory impairment. Inhibition of autophagy by 3-MA worsened ethanol-induced memory impairment. Ethanol induced autophagy in the hippocampus of mice as indicated by western blotting, electron microscopy, RT-qPCR, and fluorescence confocal microscopy. We determined that the mTOR/BECN1 (S14) pathway is involved in ethanol-induced autophagy in vivo. Further, ethanol-induced autophagy suppressed the NLRP3 inflammatory and apoptosis pathways in the hippocampus in mice and in vitro. These findings suggest that autophagy activation in hippocampal cells alleviates ethanol-induced memory impairment in association with anti-apoptotic and anti-inflammatory pathways.


Asunto(s)
Autofagia/efectos de los fármacos , Etanol/efectos adversos , Memoria/efectos de los fármacos , Memoria/fisiología , Animales , Apoptosis/efectos de los fármacos , Autofagia/fisiología , Nivel de Alcohol en Sangre , Muerte Celular/efectos de los fármacos , Disfunción Cognitiva/metabolismo , Etanol/farmacología , Hipocampo/metabolismo , Masculino , Trastornos de la Memoria/metabolismo , Ratones , Ratones Endogámicos C57BL , Microglía , Neuroinmunomodulación , Serina-Treonina Quinasas TOR/metabolismo
16.
Stem Cells Int ; 2019: 8421281, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31428161

RESUMEN

Mesenchymal stromal cells (MSCs) have become the most commonly used adult stem cells in regenerative medicine. Preclinical studies have shown that MSCs-based therapy is a potential new treatment approach for neurological diseases. Intrathecal injection has unique feature which allows stem cells to directly migrate to the lesion site in patients with central nervous system (CNS) diseases. In this study, we evaluate the safety and feasibility of intrathecal allogeneic bone marrow-derived MSCs (BM-MSCs) in patients with neurological diseases. This open-label clinical study included 37 patients (14 diseases). Eligible patients underwent a baseline assessment and were intrathecally injected with allogeneic BM-MSCs (1 × 106 cells/kg, 4 consecutive treatments at 1-week intervals). After four infusions, the patients were followed up for at least 6 months. Adverse events, cerebrospinal fluid (CSF) test results, clinical symptoms, physical examination, and haematological and imaging examinations were used to assess the safety and feasibility of the treatment. Also, we performed a systematic review of the safety of all types of intrathecal stem cells and compared our result to previous studies. In our study, the highest adverse event was a slight ache at the injection site (4.11%), followed by fever (3.42%) and mild headache (2.05%). No severe adverse events were reported. After the intrathecal injections, the white blood cell (WBC) counts in the CSF increased in 30 patients and the protein concentration in the CSF exceeded the normal range in 26 patients, while other CSF indicators remained normal. Moreover, these patients had no suspected manifestations of CNS infection. Haematological and imaging examinations showed no abnormal changes after BM-MSCs infusion. Compared with previous studies, the incidence of adverse events was nearly consistent or even lower for headache, fever, nausea, and neck pain. In conclusion, repeated intrathecal allogeneic BM-MSCs are safe, feasible, and promising for the treatment of patients with neurological diseases.

17.
Transl Stroke Res ; 10(2): 170-177, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29796934

RESUMEN

Mesenchymal stromal cells (MSCs) can differentiate into multiple tissues. Preclinical studies have shown that MSC-based therapy is a potential new treatment approach for ischemic stroke. These results support the urgent need for further studies of MSC transplantation in the treatment of ischemic stroke in humans. Here, we develop a prospective, randomized, controlled, observer-blinded phase II trial to assess the clinical safety, feasibility, and therapeutic mechanisms of allogenic bone marrow-derived MSCs (BM-MSCs) by intrathecal infusion in the treatment of patients with cerebral infarction within the middle cerebral artery and with a National Institutes of Health Stroke Scale (NIHSS) score from 15 to 25. Sample size calculation has determined that a patient population of 118, with ischemic stroke between 30 and 90 days following onset, will be randomly divided into experimental (n = 59) and control (n = 59) groups. Then eligible patients will receive four intrathecal infusions of allogenic BM-MSCs (1 × 106 cells/kg body weight) once a week. All patients have detailed functional assessments and magnetic resonance imaging prior to cell infusion and at intervals up to 1 year after. The primary outcome is the score on the modified Rankin Scale at 90 days after treatment, and the second outcomes include multiple indicators of safety and feasibility. And this trial has been registered as ChiCTR-INR-16008908 (25 July 2016).


Asunto(s)
Isquemia Encefálica/complicaciones , Trasplante de Células Madre Mesenquimatosas/métodos , Accidente Cerebrovascular/etiología , Accidente Cerebrovascular/cirugía , Adolescente , Adulto , Anciano , Isquemia Encefálica/cirugía , Femenino , Estudios de Seguimiento , Humanos , Imagen por Resonancia Magnética , Masculino , Células Madre Mesenquimatosas/fisiología , Persona de Mediana Edad , Método Simple Ciego , Resultado del Tratamiento , Adulto Joven
19.
J Neuroinflammation ; 15(1): 242, 2018 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-30153825

RESUMEN

BACKGROUND: Nod-like receptor protein 3 (NLRP3) inflammasome is a crucial factor in mediating inflammatory responses after cerebral ischemia/reperfusion (I/R), but the cellular location of NLRP3 inflammasome in cerebral I/R has yet come to a conclusion, and there is still no specific evidence to state the relationship between mitochondria and the NLRP3 inflammasome in cerebral I/R. METHODS: In the present study, we detected the cellular localization of NLRP3 inflammasomes in a transient middle cerebral artery occlusion (tMCAO) rat model and a transwell co-culture cell system under oxygen-glucose deprivation/reoxygenation (OGD/R) conditions. Then, we investigated the relationship between mitochondrial dysfunction and the activation of NLRP3 inflammasomes in different cell types after OGD/R and cerebral I/R injury. RESULTS: Our results showed that NLRP3 inflammasomes were first activated in microglia soon after cerebral I/R injury onset and then were expressed in neurons and microvascular endothelial cells later, but they were mainly in neurons. Furthermore, mitochondrial dysfunction played an important role in activating NLRP3 inflammasomes in microglia after OGD/R, and mitochondrial protector could inhibit the activation of NLRP3 inflammasomes in cerebral I/R rats. CONCLUSION: Our findings may provide novel insights into the cell type-dependent activation of NLRP3 inflammasomes at different stages of cerebral I/R injury and the role of mitochondrial dysfunction in activating the NLRP3 inflammasome pathway.


Asunto(s)
Infarto de la Arteria Cerebral Media/complicaciones , Enfermedades Mitocondriales/etiología , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Daño por Reperfusión/fisiopatología , Animales , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Masculino , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Potencial de la Membrana Mitocondrial/genética , Ratones , Ratones Endogámicos C57BL , Microglía/efectos de los fármacos , Microglía/metabolismo , Enfermedades Mitocondriales/fisiopatología , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteínas del Tejido Nervioso/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Oxígeno/administración & dosificación , Ratas , Ratas Sprague-Dawley
20.
CNS Neurol Disord Drug Targets ; 17(5): 370-382, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29732997

RESUMEN

BACKGROUND AND OBJECTIVE: Hydroxysafflor yellow A (HSYA) was reported to suppress inflammation in ischaemic microglia. However, the mechanism through which HSYA inhibits inflammation caused by cerebral ischaemia and reperfusion injury remains unknown. Here, we have mimicked acute cerebral ischaemia and reperfusion injury by subjecting male Sprague-Dawley rats to transient middle cerebral artery occlusion for 90 minutes and have demonstrated that toll-like receptor 9 (TLR9) was upregulated from day 3 after reperfusion, accompanied by the persistent activation of the pro-inflammatory nuclear factor-κB (NF-κB) pathway from 6 hours to day 7. HSYA was injected intraperitoneally at a dose of 6 mg/kg per day, which activated TLR9 in microglia of ischaemic cortex at 6 hours after reperfusion and then obviously suppressed the NF-κB pathway from day 1 to day 7. Meanwhile, HSYA also activated the anti-inflammatory pathway through interferon regulatory factor 3 from day 1 to day 3. The anti-inflammatory effect of HSYA was partially reversed by TLR9-siRNA interference in primary microglia, which was stimulated by oxygen-glucose deprivation and reoxygenation treatment. The regulation of TLR9-mediated inflammation by HSYA was consistent with the recovery of neurological deficits in rats. CONCLUSION: Therefore, our findings support that HSYA exerts anti-inflammatory effects by reprogramming the TLR9 signalling pathway during treatment of acute cerebral ischaemia and reperfusion injury.


Asunto(s)
Isquemia Encefálica/tratamiento farmacológico , Encéfalo/efectos de los fármacos , Chalcona/análogos & derivados , Quinonas/uso terapéutico , Reperfusión , Transducción de Señal/efectos de los fármacos , Receptor Toll-Like 9/metabolismo , Animales , Animales Recién Nacidos , Encéfalo/metabolismo , Infarto Encefálico/tratamiento farmacológico , Infarto Encefálico/etiología , Isquemia Encefálica/complicaciones , Hipoxia de la Célula/efectos de los fármacos , Células Cultivadas , Chalcona/química , Chalcona/farmacología , Chalcona/uso terapéutico , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Lateralidad Funcional/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Glucosa/deficiencia , Etiquetado Corte-Fin in Situ , Inflamación/tratamiento farmacológico , Inflamación/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Confocal , Examen Neurológico , Quinonas/química , Quinonas/farmacología , Interferencia de ARN/fisiología , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Factores de Tiempo , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...