Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 566
Filtrar
1.
JMIR Serious Games ; 12: e57304, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39302638

RESUMEN

Background: The video game industry has introduced a new form of monetization through microtransactions. A controversial example has been the so-called "loot boxes" (LBs) as virtual objects, which are randomized and bought with legal money. In recent years, LBs have come to connect 2 distinct problem behaviors, namely internet gaming disorder (IGD) and online gambling disorder (OGD). Many association studies have been conducted on the 3 constructs, but few have delved into the relationship of problematic use of LBs (PU-LB) with IGD and OGD. Objective: This study aims to explore the mediating role of the PU-LB between IGD and OGD. Methods: This cross-sectional and analytical study used incidental sampling in 24 Spanish schools. The final sample consisted of 542 participants (male: n=523, 96.5%; age: range 11-30 y) who played video games, bought LBs, and had gambled online in the last 12 months. Participants then completed the Spanish versions of the Internet Gaming Disorder Scale-Short Form, Online Gambling Disorder Questionnaire, and PU-LB scale. Results: IGD scores were found to be significantly associated with both PU-LB (r=0.473, P<.001) and OGD (r=0.209, P<.001). Moreover, PU-LB was significantly associated with OGD (r=0.351, P<.001). The structural equation model results indicated that IGD had no significant direct effect on OGD (P=.903). However, the indirect effect of IGD on OGD through PU-LB was significant (P<.001). Therefore, PU-LB fully mediated the relationship between IGD and OGD. Furthermore, these results were found in the subsamples of both minors (<18 y) and young adults (≥18 y). Conclusions: It is suggested that there is a mediation effect of problematic LB use between internet gambling and online gambling problems in both minors and young adults. This has potential practical implications by providing more evidence on how LBs have become a hinge feature between 2 clinically relevant and independent issues. In this regard, adequate industry self-regulation is needed, and effective legislation for the protection of minors is necessary.

2.
Int J Biol Macromol ; : 135731, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39299420

RESUMEN

Phytohormones play crucial roles in regulation of plant growth and tolerance to abiotic stresses. The 2-oxoglutarate-dependent dioxygenase (2OGD) superfamily responds to hormone biosynthesis and metabolism in plants. However, the Nt2OGD family in tobacco has not been fully explored. In this study, we identify 126 members of the Nt2OGD family, and 60 of them are involved in hormone biosynthesis and metabolism process (Nt2OGD-Hs), including 1-aminocyclopropane-1-carboxylic acid oxidases (ACO), dioxygenases for auxin oxidation (DAO), gibberellin (GA) 20-oxidases and 3-oxidases (GA20ox and GA3ox), carbon-19 and carbon-20 GA 2-oxidases (C19-GA2ox and C20-GA2ox), lateral branching oxidoreductases (LBO), jasmonate-induced oxygenases (JOX), downy mildew resistant 6, and DMR6-like oxygenases (DMR6/DLO). Gene duplication analysis suggests the segmental duplication and whole genome duplication (WGD) might be a potential mechanism for the expansion of this family. Expression analysis reveals that most of Nt2OGD-Hs show tissue-specific expression patterns, and some of them respond to environmental conditions. Of Nt2OGD-Hs, the expression of NtJOX3 and NtJOX5, which are involved in JA metabolism, exhibits remarkable changes during drought treatments. Silencing of NtJOX3 or NtJOX5 increases tobacco tolerance to drought stress. Furthermore, knocking out OsJOX3 and OsJOX4, respectively in rice, result in high tolerance to drought. Taken together, our work comprehensively identifies the Nt2OGD family in tobacco and provides new insights into roles of the JA pathway in drought tolerance in plants.

3.
Folia Med Cracov ; 64(1): 39-52, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-39254580

RESUMEN

BACKGROUND: Kidney diseases are a major global health problem affecting millions of people. Despite this, there is as yet no effective drug therapy improving outcome in patients with renal disease. The aim of this study was to examine the nephroprotective effect of α-lipoic acid (ALA) in vitro and to examine the effect of ALA administered in vivo on the production of reactive sulfur species (RSS), including hydrogen sulfide (H2S) and compounds containing sulfane sulfur. METHODS: The effect of ALA was studied in vitro by determining the viability of human embryonic kidney cells (HEK293) in normoxic and hypoxic conditions as well as in vivo in two groups of chronic kidney disease (CKD) patients: non-dialyzed (ND) and undergoing continuous ambulatory peritoneal dialysis (PD) after 30 days of ALA supplementation. RESULTS: The results revealed that the viability of HEK293 cells was significantly decreased by hypoxic conditions, while ALA administered during hypoxia increased the viability to the level observed in normoxic conditions. Studies performed in plasma of CKD patients after ALA supplementation suggested that ALA did not affect the parameters of oxidative stress, while significantly increased the level of reactive sulfane sulfur in both ND and PD patients suffering from CKD. The results suggest that ALA can exert nephroprotective effects which are related to sulfane sulfur production.


Asunto(s)
Insuficiencia Renal Crónica , Ácido Tióctico , Humanos , Ácido Tióctico/farmacología , Insuficiencia Renal Crónica/sangre , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/tratamiento farmacológico , Células HEK293 , Masculino , Femenino , Supervivencia Celular/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Persona de Mediana Edad , Antioxidantes/farmacología , Sulfuro de Hidrógeno/farmacología
4.
Mater Today Bio ; 28: 101184, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39221214

RESUMEN

Currently, the construction of novel biomimetic reduced graphene oxide (RGO)-based nanocomposites to induce neurite sprouting and repair the injured neurons represents a promising strategy in promoting neuronal development or treatment of cerebral anoxia or ischemia. Here, we present an effective method for constructing palladium-reduced graphene oxide (Pd-RGO) nanocomposites by covalently bonding Pd onto RGO surfaces to enhance neurite sprouting of cultured neurons. As described, the Pd-RGO nanocomposites exhibit the required physicochemical features for better biocompatibility without impacting cell viability. Primary neurons cultured on Pd-RGO nanocomposites had significantly increased number and length of neuronal processes, including both axons and dendrites, compared with the control. Western blotting showed that Pd-RGO nanocomposites improved the expression levels of growth associate protein-43 (GAP-43), as well as ß-III tubulin, Tau-1, microtubule-associated protein-2 (MAP2), four proteins that are involved in regulating neurite sprouting and outgrowth. Importantly, Pd-RGO significantly promoted neurite length and complexity under oxygen-glucose deprivation/re-oxygenation (OGD/R) conditions, an in vitro cellular model of ischemic brain damage, that closely relates to neuronal GAP-43 expression. Furthermore, using the middle cerebral artery occlusion (MCAO) model in rats, we found Pd-RGO effectively reduced the infarct area, decreased neuronal apoptosis in the brain, and improved the rats' behavioral outcomes after MCAO. Together, these results indicate the great potential of Pd-RGO nanocomposites as a novel excellent biomimetic material for neural interfacing that shed light on its applications in brain injuries.

5.
Cell Biochem Biophys ; 2024 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-39240442

RESUMEN

Cerebral ischemia/reperfusion injury (IRI) is a primary pathophysiological basis of ischemic stroke, a dreadful cerebrovascular event carrying substantial disability and lethality. Triggering receptor expressed on myeloid cells 2 (TREM2) is a membrane glycoprotein that has been notified as a protective factor for cerebral ischemic stroke. On this basis, the paper is thereby goaled to interpret the probable activity and downstream mechanism of TREM2 against cerebral IRI. Cerebral IRI was simulated in murine microglial BV2 cells under oxygen-glucose deprivation and reperfusion (OGD/R) conditions. Western blotting ascertained the expressions of TREM2 and janus kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) axis-associated proteins. ELISA and RT-qPCR assayed the secretion of inflammatory cytokines. Immunofluorescence and western blotting estimated macrophage polarization. Glycolysis activation was measured through evaluating lactic acid and extracellular acidification rate (ECAR). RT-qPCR and western blotting examined the expressions of glycolytic genes. TREM2 was abnormally expressed and JAK2/STAT3 axis was aberrantly activated in BV2 cells in response to OGD/R. Elevation of TREM2 repressed the inflammatory reaction and glycolysis, inhibited the JAK2/STAT3 axis, whereas promoted M1-to-M2 polarization in OGD/R-injured BV2 cells. Upregulated TREM2 inactivated the glycolytic pathway to relieve OGD/R-induced inflammatory injury and M1 macrophage polarization. Besides, STAT3 activator, colivelin, aggravated the glycolysis, inflammatory injury and drove M1-like macrophage polarization in TREM2-overexpressing BV2 cells exposed to OGD/R. Collectively, TREM2 might produce anti-inflammatory potential in cerebral IRI, which might dependent on the inactivation of glycolytic pathway via intermediating the JAK2/STAT3 axis.

6.
Brain Res ; 1845: 149218, 2024 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-39218334

RESUMEN

Ischemic stroke, caused by diminished or interrupted cerebral blood flow, triggers the activation of microglial cells and subsequent inflammatory responses. Formononetin (FMN) has been observed to inhibit BV2 microglial cell activation and alleviate ensuing neuroinflammatory reactions. Despite extensive research, the precise underlying mechanism remains unclear. To investigate the neuroinflammatory response following FMN-mediated inhibition of BV2 microglial activation, we employed an in vitro oxygen-glucose deprivation/reperfusion (OGD/R) model. BV2 microglial cells were categorized into four groups: control, FMN, OGD/R, and OGD/R+FMN. Cell viability was assessed using the CCK-8 assay, while flow cytometry assessed M1 and M2 cell populations within BV2 cells. Immunofluorescence was utilized to detect the expression levels of apoptosis-inducing factor (AIF), p53, Toll-like receptor 4 (TLR4), and NF-κB p65. Western blotting (WB) was conducted to quantify p65/p-p65, IκB-α/p-IκB-α, and TLR4 protein levels in each group. Additionally, ELISA was employed to measure IL-1ß and TNF-α levels in cell supernatants from each group. The results revealed a significant increase in the proportion of iNOS/CD206-positive M1/M2 cells in the OGD/R group compared to the control group (p < 0.05). There was also a notable increase in nuclear translocation of NF-κB p65 and elevated expression of inflammatory factors IL-1ß and TNF-α in cell supernatants. Moreover, levels of p-p65, p-IκB-α, and TLR4 proteins were significantly elevated in the OGD/R group (p < 0.05). However, the addition of FMN reversed these effects. Specifically, FMN administration notably attenuated cell death and inflammation in BV2 microglia induced by OGD/R through modulation of the TLR4/NF-κB signaling pathway.These findings suggest that FMN may serve as a potential therapeutic agent against neuroinflammation associated with ischemic stroke by targeting microglial activation pathways.

7.
Acta Biomater ; 186: 424-438, 2024 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-39122135

RESUMEN

Mitophagy influences the progression and prognosis of ischemic stroke (IS). However, whether DNA methylation in the brain is associated with altered mitophagy in hypoxia-injured neurons remains unclear. Here, miR-138-5p was found to be highly expressed in exosomes secreted by astrocytes stimulated with oxygen and glucose deprivation/re-oxygenation (OGD/R), which could influence the recovery of OGD/R-injured neurons through autophagy. Mechanistically, miR-138-5p promotes the stable expression of Ras homolog enriched in brain like 1(Rhebl1) through DNA-methyltransferase-3a (DNMT3A), thereby enhancing ubiquitin-dependent mitophagy to maintain mitochondrial homeostasis. Furthermore, we employed glycosylation engineering and bioorthogonal click reactions to load mirna onto the surface of microglia and deliver them to injured region utilising the inflammatory chemotactic properties of microglia to achieve drug-targeted delivery to the central nervous system (CNS). Our findings demonstrate miR-138-5p improves mitochondrial function in neurons through the miR-138-5p/DNMT3A/Rhebl1 axis. Additionally, our engineered cell vector-targeted delivery system could be promising for treating IS. STATEMENT OF SIGNIFICANCE: In this study, we demonstrated that miR-138-5p in exosomes secreted by astrocytes under hypoxia plays a critical role in the treatment of hypoxia-injured neurons. And we find a new target of miR-138-5p, DNMT3A, which affects neuronal mitophagy and thus exerts a protective effect by regulating the methylation of Rbebl1. Furthermore, we have developed a carrier delivery system by combining miR-138-5p with the cell membrane of microglia and utilized the inflammatory chemotactic properties of microglia to deliver this system to the brain via intravenous injection. This groundbreaking study not only provides a novel therapeutic approach for ischemia-reperfusion treatment but also establishes a solid theoretical foundation for further research on targeted drug delivery for central nervous system diseases with promising clinical applications.


Asunto(s)
ADN Metiltransferasa 3A , MicroARNs , Mitofagia , Neuronas , Mitofagia/efectos de los fármacos , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Animales , Hipoxia de la Célula/efectos de los fármacos , Humanos , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Exosomas/metabolismo , Ratones , Astrocitos/metabolismo , Astrocitos/efectos de los fármacos
8.
Exp Neurol ; 380: 114910, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39098715

RESUMEN

Structural and functional alterations in brain microvascular endothelial cells (BMECs) caused by oxygen-glucose deprivation (OGD) are involved in the pathogenesis of various brain disorders. AlkB homolog 5 (ALKBH5) is a primary m6A demethylase that regulates various cell processes, but its distinct roles in BMEC function remain to be clarified. In the present study, in mouse middle cerebral artery occlusion (MCAO) model, knockout of ALKBH5 reduced neurological deficits, infarct volumes and tissue apoptosis caused by ischemia/reperfusion injury. Evans blue leakage and decreased expression of the tight junction protein ZO-1 and Occludin were also attenuated by ALKBH5 knockout. During the exploration of the underlying mechanisms of the role of ALKBH5 in BMECs, we found that the expression of ALKBH5 was induced at both the mRNA and protein levels by hypoxia; however, its protein stability was impaired by OGD treatment. Knockdown of ALKBH5 expression increased total m6A levels and alleviated OGD-induced BMEC injury. At the same time, the selective ALKBH5 inhibitor Cpd 20m also exhibited a protective effect on cell injury. In contrast, overexpression of ALKBH5 increased the sensitivity of BMECs to OGD. Interestingly, the m6A sequencing data revealed that knockdown of ALKBH5altered the expression of many genes via m6A upregulation. The gene expression alterations were verified by real-time PCR. Taken together, our results suggest that ALKBH5, as well as its target genes, plays important roles in the regulation of brain microvascular endothelial cell function through its RNA demethylase activity.


Asunto(s)
Desmetilasa de ARN, Homólogo 5 de AlkB , Células Endoteliales , Glucosa , Ratones Noqueados , Animales , Ratones , Desmetilasa de ARN, Homólogo 5 de AlkB/metabolismo , Desmetilasa de ARN, Homólogo 5 de AlkB/genética , Células Endoteliales/metabolismo , Glucosa/deficiencia , Encéfalo/metabolismo , Encéfalo/patología , Masculino , Microvasos/patología , Microvasos/metabolismo , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Infarto de la Arteria Cerebral Media/patología , Adenosina/análogos & derivados , Adenosina/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/patología
9.
Int J Mol Sci ; 25(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39201562

RESUMEN

Neonatal hypoxic-ischemic encephalopathy (HIE) is a critical condition characterized by significant brain damage due to insufficient blood flow and oxygen delivery at birth, leading to high rates of neonatal mortality and long-term neurological deficits worldwide. 2,3-Diphosphoglyceric acid (2,3-DPG), a small molecule metabolite prevalent in erythrocytes, plays an important role in regulating oxygen delivery, but its potential neuroprotective role in hypoxic-ischemic brain damage (HIBD) has yet to be fully elucidated. Our research reveals that the administration of 2,3-DPG effectively reduces neuron damage caused by hypoxia-ischemia (HI) both in vitro and in vivo. We observed a notable decrease in HI-induced neuronal cell apoptosis, attributed to the downregulation of Bax and cleaved-caspase 3, alongside an upregulation of Bcl-2 expression. Furthermore, 2,3-DPG significantly alleviates oxidative stress and mitochondrial damage induced by oxygen-glucose deprivation/reperfusion (OGD/R). The administration of 2,3-DPG in rats subjected to HIBD resulted in a marked reduction in brain edema and infarct volume, achieved through the suppression of neuronal apoptosis and neuroinflammation. Using RNA-seq analysis, we validated that 2,3-DPG offers protection against neuronal apoptosis under HI conditions by modulating the p38 MAPK pathway. These insights indicated that 2,3-DPG might act as a promising novel therapeutic candidate for HIE.


Asunto(s)
Apoptosis , Hipoxia-Isquemia Encefálica , Proteínas Quinasas p38 Activadas por Mitógenos , Animales , Hipoxia-Isquemia Encefálica/metabolismo , Hipoxia-Isquemia Encefálica/tratamiento farmacológico , Hipoxia-Isquemia Encefálica/patología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ratas , Apoptosis/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Fármacos Neuroprotectores/uso terapéutico , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Neuronas/patología , Ratas Sprague-Dawley , Masculino , Mitocondrias/metabolismo , Mitocondrias/efectos de los fármacos
10.
J Biochem Mol Toxicol ; 38(9): e23827, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39193856

RESUMEN

Ischemic stroke is one main type of cerebrovascular disorders with leading cause of death and disability worldwide. Astrocytes are the only nerve cell type storing glycogen in the brain, which regulate the glucose metabolism and handle the energy supply and survive of neurons. Astrocyte ferroptosis contributes to neuron injury in brain disorders. N-myc downstream-regulated gene 2 (NDRG2) has been implicated in the progression of brain diseases, including ischemic stroke. However, whether NDRG2 could affect the glucose metabolism and ferroptosis of astrocytes during ischemic stroke remains largely unknown. Mouse astrocytes were treated with oxygen-glucose deprivation/reoxygenation (OGD/R) to establish the in vitro model. Glial fibrillary acidic protein, NDRG2, Wnt3a and ß-catenin expression levels were detected by immunofluorescence staining and western blot analyses. Glucose metabolism was investigated by glucose uptake, lactate production, nicotinamide adenine dinucleotide phosphate hydrogen/nicotinamide adenine dinucleotide phosphate (NADPH/NADP+), ATP and glycolysis enzymes (HK2, PKM2 and lactate dehydrogenase A [LDHA]) levels. Ferroptosis was assessed via reactive oxygen species (ROS), glutathione (GSH), iron and ferroptosis-related markers (GPX4 and PTGS2) contents. Glycolysis enzymes and ferroptosis-related markers levels were measured via western blot. NDRG2 expression was elevated in OGD/R-induced astrocytes. NDRG2 overexpression aggravated OGD/R-induced loss of glucose metabolism through reducing glucose uptake, lactate production, NADPH/NADP+ and ATP levels. NDRG2 upregulation exacerbated OGD/R-caused reduction of glycolysis enzymes (HK2, PKM2 and LDHA) levels. NDRG2 promoted OGD/R-induced ferroptosis of astrocytes by increasing ROS, iron and PTGS2 levels and decreasing GSH and GPX4 levels. NDRG2 overexpression enhanced OGD/R-induced decrease of Wnt/ß-catenin signaling activation by reducing Wnt3a and ß-catenin expression. NDRG2 silencing played an opposite effect. Inhibition of Wnt/ß-catenin signaling activation by IWR-1 attenuated the influences of NDRG2 knockdown on glucose metabolism, glycolysis enzymes levels and ferroptosis. These findings demonstrated that NDRG2 contributes to OGD/R-induced inhibition of glucose metabolism and promotion of ferroptosis in astrocytes through inhibiting Wnt/ß-catenin signaling activation, which might be associated with ischemic stroke progression.


Asunto(s)
Astrocitos , Ferroptosis , Glucosa , Vía de Señalización Wnt , beta Catenina , Astrocitos/metabolismo , Animales , Glucosa/metabolismo , Ratones , beta Catenina/metabolismo , Glucólisis , Especies Reactivas de Oxígeno/metabolismo , Células Cultivadas , Oxígeno/metabolismo , Proteínas Adaptadoras Transductoras de Señales
11.
Biochem Biophys Res Commun ; 733: 150452, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39067246

RESUMEN

Due to the complex pathogenesis of acute ischemic stroke (AIS), further investigation into its underlying mechanisms is necessary. Presently, existing literature indicates a close association between ferroptosis and AIS injury; however, the precise mechanism and molecular target of ferroptosis in AIS injury remain elusive. By RNA sequencing, we found a significant increase in LCN2 expression in the ischemic cortex. In order to investigate the potential role of LCN2 in modulating AIS injury through the regulation of ferroptosis, we utilized RNA interference (RNAi) knockdown and gene overexpression experiments. The findings from experiments conducted both in vitro and in vivo revealed a marked increase in ferroptosis levels within the AIS model group. Suppression of the LCN2 gene resulted in a significant reduction in ferroptosis levels in OGD/R cells. Conversely, upregulation of LCN2 exacerbated ferroptosis levels in OGD/R cells. The results suggest that elevated levels of ferroptosis may result from heightened expression of LCN2, thereby exacerbating ischemia/reperfusion injury. This study indicates the involvement of ferroptosis in the pathogenesis of AIS and highlights LCN2 as a regulator of ferroptosis in AIS-induced injury, suggesting a potential therapeutic target for ischemic stroke.

12.
J Cardiothorac Surg ; 19(1): 435, 2024 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-38997740

RESUMEN

BACKGROUND: Cerebral ischemia-reperfusion injury (I/R) can affect patient outcomes and can even be life-threatening. This study aimed to explore the role of Shionone in cerebral I/R and reveal its mechanism of action through the cerebral I/R in vitro model. METHODS: SH-SY5Y cells were subjected to oxygen-glucose deprivation/reoxygenation (OGD/R) to induce cerebral I/R in vitro model. SH-SY5Y cells were treated with different concentrations of Shionone. Cell counting kit-8 and flow cytometry assays were used to detect cell viability and apoptosis levels. The levels of superoxide dismutase, catalase, and malondialdehyde were determined using their corresponding kits to examine the level of oxidative stress. The inflammation response was detected by IL-6, IL-1ß, and TNF-α levels, using enzyme-linked-immunosorbent-assay. RT-qPCR was performed to measure the mRNA levels of p38 and NF-κB. Western blotting was used to quantify the apoptosis-related proteins and p38MAPK/NF-κB signaling pathway proteins. RESULTS: Shionone exhibited no toxic effects on SH-SY5Y cells. Shionone inhibited OGD/R-induced cell apoptosis, improved the inflammatory response caused by OGD/R, and reduced the level of oxidative stress in cells. Western blot assay results showed that Shionone alleviated OGD/R-induced injury by inhibiting the activity of the p38 MAPK/NF-κB signaling pathway. The p38/MAPK agonist P79350 reversed the beneficial effects of Shionone. CONCLUSION: Shionone alleviates cerebral I/R and may thus be a novel therapeutic strategy for treating cerebral I/R.


Asunto(s)
Apoptosis , Glucosa , FN-kappa B , Oxígeno , Daño por Reperfusión , Proteínas Quinasas p38 Activadas por Mitógenos , Humanos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Glucosa/deficiencia , FN-kappa B/metabolismo , Daño por Reperfusión/metabolismo , Daño por Reperfusión/prevención & control , Apoptosis/efectos de los fármacos , Oxígeno/metabolismo , Estrés Oxidativo/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Línea Celular Tumoral
13.
Brain Res Bull ; 215: 111032, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39029715

RESUMEN

BACKGROUND: Buyang Huanwu Decoction (BHD) is widely used in Chinese clinical practice for the treatment and prevention of ischemic cerebral vascular diseases. This study was designed to investigate the effects of BHD on ischemic stroke (IS) and its underlying mechanism. METHODS: The middle cerebral artery occlusion (MCAO) rat model and oxygen-glucose deprivation and reoxygenation (OGD/R) rat brain microvascular endothelial cell (RBMVEC) models were established. Brain infarction size and neurological score were calculated following MCAO surgery. Evans blue was used to measure blood brain barrier (BBB) permeability. Cell counting kit-8 (CCK-8) and TUNEL assays were performed to evaluate the cell viability and apoptosis of RBMVECs. Dual-luciferase reporter assay was used to analyze the transcriptional activities of apoptosis-related genes. RESULTS: Results showed that higher infarction volume, neurological scores, and BBB permeability in the MCAO group rats were reduced after BHD treatment. Drug serum (DS) treatment had no impact on the normal RBMVECs' cell viability and cell apoptosis. Besides, DS treatment decreased the lactate production, glucose uptake, and extracellular acidification rate in normal and OGD/R-induced RBMVECs. DS treatment downregulated the protein levels of pan-lysine lactylation (kla), histone H3 lysine 18 lactylation (H3K18la), and the transcriptional of apoptotic protease activating factor-1 (Apaf-1) in OGD/R-treated RBMVECs. In addition, Apaf-1 overexpression decreased cell viability and increased apoptosis and glycolysis activity of OGD/R-treated RBMVECs. CONCLUSION: In summary, BHD inhibited glycolysis and apoptosis via suppressing the pan-kla and H3K18la protein levels and the Apaf-1 transcriptional activity, thus restraining the progression of IS.


Asunto(s)
Apoptosis , Medicamentos Herbarios Chinos , Células Endoteliales , Glucólisis , Accidente Cerebrovascular Isquémico , Ratas Sprague-Dawley , Animales , Medicamentos Herbarios Chinos/farmacología , Apoptosis/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Glucólisis/efectos de los fármacos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Ratas , Masculino , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Supervivencia Celular/efectos de los fármacos , Infarto de la Arteria Cerebral Media/tratamiento farmacológico , Infarto de la Arteria Cerebral Media/metabolismo , Glucosa/metabolismo , Microvasos/efectos de los fármacos , Microvasos/metabolismo , Modelos Animales de Enfermedad
14.
Int Immunopharmacol ; 138: 112588, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38955031

RESUMEN

Dexmedetomidine (Dex) is widely used in the sedation in intensive care units and as an anesthetic adjunct. Considering the anti-inflammatory and antioxidant properties of Dex, we applied in vivo rat model as well as in vitro cardiomyocyte models (embryonic rat cardiomyocytes H9c2 cells and neonatal rat cardiomyocytes, NRCMs) to evaluate the effects of Dex against myocardial ischemia reperfusion (I/R) injury. Transcriptomic sequencing for gene expression in heart tissues from control rats and Dex-treated rats identified that genes related to fatty acid metabolism were significantly regulated by Dex. Among these genes, the elongation of long-chain fatty acids (ELOVL) family member 6 (Elovl6) was most increased upon Dex-treatment. By comparing the effects of Dex on both wild type and Elovl6-knockdown H9c2 cells and NRCMs under oxygen-glucose deprivation/reoxygenation (OGD/R) challenge, we found that Elovl6 knockdown attenuated the protection efficiency of Dex, which was supported by the cytotoxicity endpoints (cell viability and lactate dehydrogenase release) and apoptosis as well as key gene expressions. These results indicate that Dex exhibited the protective function against myocardial I/R injury via fatty acid metabolism pathways and Elovl6 plays a key role in the process, which was further confirmed using palmitate exposure in both cells, as well as in an in vivo rat model. Overall, this study systematically evaluates the protective effects of Dex on the myocardial I/R injury and provides better understanding on the fatty acid metabolism underlying the beneficial effects of Dex.


Asunto(s)
Apoptosis , Dexmedetomidina , Elongasas de Ácidos Grasos , Ácidos Grasos , Daño por Reperfusión Miocárdica , Miocitos Cardíacos , Animales , Dexmedetomidina/farmacología , Dexmedetomidina/uso terapéutico , Daño por Reperfusión Miocárdica/tratamiento farmacológico , Daño por Reperfusión Miocárdica/metabolismo , Elongasas de Ácidos Grasos/genética , Elongasas de Ácidos Grasos/metabolismo , Ratas , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ácidos Grasos/metabolismo , Masculino , Línea Celular , Apoptosis/efectos de los fármacos , Ratas Sprague-Dawley , Acetiltransferasas/metabolismo , Acetiltransferasas/genética , Supervivencia Celular/efectos de los fármacos
15.
Biomolecules ; 14(7)2024 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-39062570

RESUMEN

Background: The regulation of divalent metal transporter-1 (DMT1) by insulin has been previously described in Langerhans cells and significant neuroprotection was found by insulin and insulin-like growth factor 1 treatment during experimental cerebral ischemia in acute ischemic stroke patients and in a rat 6-OHDA model of Parkinson's disease, where DMT1 involvement is described. According to the regulation of DMT1, previously described as a target gene of NF-kB in the early phase of post-ischemic neurodegeneration, both in vitro and in vivo, and because insulin controls the NFkB signaling with protection from ischemic cell death in rat cardiomyocytes, we evaluated the role of insulin in relation to DMT1 expression and function during ischemic neurodegeneration. Methods: Insulin neuroprotection is evaluated in differentiated human neuroblastoma cells, SK-N-SH, and in primary mouse cortical neurons exposed to oxygen glucose deprivation (OGD) for 8 h or 3 h, respectively, with or without 300 nM insulin. The insulin neuroprotection during OGD was evaluated in both cellular models in terms of cell death, and in SK-N-SH for DMT1 protein expression and acute ferrous iron treatment, performed in acidic conditions, known to promote the maximum DMT1 uptake as a proton co-transporter; and the transactivation of 1B/DMT1 mouse promoter, already known to be responsive to NF-kB, was analyzed in primary mouse cortical neurons. Results: Insulin neuroprotection during OGD was concomitant to the down-regulation of both DMT1 protein expression and 1B/DMT1 mouse promoter transactivation. We also showed the insulin-dependent protection from cell death after acute ferrous iron treatment. In conclusion, although preliminary, this evaluation highlights the peculiar role of DMT1 as a possible pharmacological target, involved in neuroprotection by insulin during in vitro neuronal ischemia and acute ferrous iron uptake.


Asunto(s)
Proteínas de Transporte de Catión , Muerte Celular , Regulación hacia Abajo , Insulina , Neuronas , Animales , Insulina/metabolismo , Insulina/farmacología , Humanos , Proteínas de Transporte de Catión/metabolismo , Proteínas de Transporte de Catión/genética , Ratones , Muerte Celular/efectos de los fármacos , Neuronas/metabolismo , Neuronas/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Neuroprotección/efectos de los fármacos , Línea Celular Tumoral , Fármacos Neuroprotectores/farmacología , Hierro/metabolismo , Isquemia Encefálica/metabolismo , Isquemia Encefálica/tratamiento farmacológico , Isquemia Encefálica/patología , Glucosa/metabolismo , Compuestos Ferrosos/farmacología
16.
Fitoterapia ; 177: 106076, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38897247

RESUMEN

Two new triterpenoids, ilexsaponin U (1) and ilexsaponin V (2), and three new phenylpropanoids, pubescenoside S (3), pubescenoside T (38), and pubescenoside U (39), along with thirty-four existing compounds were isolated from the roots of Ilex pubescens. The elucidation of their structures involved comprehensive spectroscopic techniques, including IR, UV, HR-ESI-MS, and NMR experiments. The anti-inflammatory effects of almost all the compounds were evaluated in LPS-induced RAW264.7 cells. Among these, compounds 1, 4, 8, 11, 12, 26, 27, 29 and 33 exhibited varying degrees of inhibition of inflammatory factors. Notably, compounds 1, 4 and 8 significantly inhibited the mRNA levels of iNOS, IL-6, IL-1ß and TNFα, comparable to or exceeding the effect of the positive control (dexamethasone, DEX). We also evaluated the cardioprotective effects of these compounds in OGD/R-induced H9c2 cells. The results revealed that compounds 2, 3, 7, 8, 26, 35, 36 and 37 at 20 µM significantly increased cell viability by 24.9 ± 3.4%, 28.0 ± 0.3%, 37.6 ± 0.2%, 44.86 ± 0.5%, 9.47 ± 2.1%, 23.9 ± 0.4%, 39.5 ± 3.1% and 28.2 ± 0.1%, respectively. Some of them exhibited effects equal to or greater than that of the positive control (diazoxide, DZ) at 100 µM, showing a 21.9 ± 3.0% increase.


Asunto(s)
Antiinflamatorios , Ilex , Fitoquímicos , Raíces de Plantas , Triterpenos , Ilex/química , Ratones , Animales , Raíces de Plantas/química , Células RAW 264.7 , Triterpenos/farmacología , Triterpenos/aislamiento & purificación , Triterpenos/química , Antiinflamatorios/farmacología , Antiinflamatorios/aislamiento & purificación , Estructura Molecular , Fitoquímicos/farmacología , Fitoquímicos/aislamiento & purificación , Ratas , Cardiotónicos/farmacología , Cardiotónicos/aislamiento & purificación , China , Óxido Nítrico Sintasa de Tipo II/metabolismo
17.
Biomed Pharmacother ; 177: 116894, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38878634

RESUMEN

BACKGROUND: To explore the neuroprotective mechanism of artemisinin against ischemic stroke from the perspective of NLRP3-mediated pyroptosis. METHODS: Serum metabolomics technology was used to analyze the serum samples of mice, and KEGG metabolic pathway was analyzed for the different metabolites in the samples. PIT model and OGD/R model were used to simulate ischemic stroke damage in vivo and in vitro. Hoechst 33342 staining, Annexin V-FITC/PI staining and TUNEL staining were used to detect the pyroptosis rate of cells. The contents of IL-1ß and IL-18 in PC12 cells and serum of mice were detected by ELISA. The expressions of NLRP3, ASC-1, Caspase-1 and TXNIP in PC12 cells and mouse brain tissue were detected by Western Blot. RESULTS: Serum metabolic profiles of animal models identified 234 different metabolites and 91 metabolic pathways. Compared with the Sham group and the Stroke+ART group, the KEGG pathway in the Stroke group was concentrated in the Necroptosis pathway associated with cell growth and death, and the NLRP3 inflammasome-mediated pyroptosis pathway was activated in the Necroptosis pathway after ischemic stroke. The results of in vivo and in vitro experiments showed that pretreatment with 10 µM artemisinin reduced ROS production, decreased Δψm, reduced pyroptosis, maintained neuronal cell morphology, and down-regulated the contents of IL-1ß and IL-18 as well as the expression of key proteins of NLRP3, ASC-1, Caspase-1 and TXNIP(p<0.01). CONCLUSION: Artemisinin can reduce neuronal pyroptosis induced by ischemic stroke by inhibiting ROS/TXNIP/NLRP3/Caspase-1 signaling pathway.


Asunto(s)
Artemisininas , Proteínas Portadoras , Caspasa 1 , Accidente Cerebrovascular Isquémico , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Piroptosis , Especies Reactivas de Oxígeno , Transducción de Señal , Animales , Piroptosis/efectos de los fármacos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Caspasa 1/metabolismo , Transducción de Señal/efectos de los fármacos , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/patología , Artemisininas/farmacología , Ratones , Proteínas Portadoras/metabolismo , Células PC12 , Especies Reactivas de Oxígeno/metabolismo , Masculino , Ratas , Proteínas de Ciclo Celular/metabolismo , Fármacos Neuroprotectores/farmacología , Inflamasomas/metabolismo , Inflamasomas/efectos de los fármacos , Modelos Animales de Enfermedad , Tiorredoxinas
18.
Exp Brain Res ; 242(8): 1841-1850, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38842755

RESUMEN

Vascular dementia (VaD) is the most common cause of dementia in older adults. Due to the lack of effective treatment options, there is an urgent need to find an effective pharmaceutical compound to combat VaD. Piracetam has been reported to improve impaired cognitive function in a variety of conditions in both human and animal models. However, the role and mechanism of Piracetam in VaD remain unclear. Therefore this study aimed to elucidate the effect of Piracetam on a cellular model of VaD in vitro. We found that Piracetam enhanced the growth of OGD-stimulated SH-SY5Y cells. In addition, Piracetam inhibited the oxidative stress of OGD-stimulated SH-SY5Y cells. Further, Piracetam improved mitochondrial function of OGD-stimulated SH-SY5Y cells. Mechanistically, Piracetam inhibited the PI3K/Akt/mTOR pathway in OGD-stimulated SH-SY5Y cells. Collectively, Piracetam improved oxidative stress and mitochondrial dysfunction of OGD-stimulated SH-SY5Y cells through PI3K/Akt/mTOR axis. Hence, Piracetam has the potential to serve as a promising drug of VaD.


Asunto(s)
Demencia Vascular , Mitocondrias , Estrés Oxidativo , Piracetam , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Humanos , Demencia Vascular/tratamiento farmacológico , Demencia Vascular/metabolismo , Piracetam/farmacología , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Línea Celular Tumoral , Fármacos Neuroprotectores/farmacología , Glucosa/metabolismo , Relación Dosis-Respuesta a Droga
19.
Sci Rep ; 14(1): 11240, 2024 05 16.
Artículo en Inglés | MEDLINE | ID: mdl-38755191

RESUMEN

Nao-an Dropping Pill (NADP) is a Chinese patent medicine which commonly used in clinic for ischemic stroke (IS). However, the material basis and mechanism of its prevention or treatment of IS are unclear, then we carried out this study. 52 incoming blood components were resolved by UHPLC-MS/MS from rat serum, including 45 prototype components. The potential active prototype components hydroxysafflor yellow A, ginsenoside F1, quercetin, ferulic acid and caffeic acid screened by network pharmacology showed strongly binding ability with PIK3CA, AKT1, NOS3, NFE2L2 and HMOX1 by molecular docking. In vitro oxygen-glucose deprivation/reperfusion (OGD/R) experimental results showed that NADP protected HA1800 cells from OGD/R-induced apoptosis by affecting the release of LDH, production of NO, and content of SOD and MDA. Meanwhile, NADP could improve behavioral of middle cerebral artery occlusion/reperfusion (MCAO/R) rats, reduce ischemic area of cerebral cortex, decrease brain water and glutamate (Glu) content, and improve oxidative stress response. Immunohistochemical results showed that NADP significantly regulated the expression of PI3K, Akt, p-Akt, eNOS, p-eNOS, Nrf2 and HO-1 in cerebral ischemic tissues. The results suggested that NADP protects brain tissues and ameliorates oxidative stress damage to brain tissues from IS by regulating PI3K/Akt/eNOS and Nrf2/HO-1 signaling pathways.


Asunto(s)
Medicamentos Herbarios Chinos , Accidente Cerebrovascular Isquémico , Factor 2 Relacionado con NF-E2 , Óxido Nítrico Sintasa de Tipo III , Animales , Ratas , Apoptosis/efectos de los fármacos , Medicamentos Herbarios Chinos/farmacología , Medicamentos Herbarios Chinos/química , Medicamentos Herbarios Chinos/uso terapéutico , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1/metabolismo , Accidente Cerebrovascular Isquémico/tratamiento farmacológico , Accidente Cerebrovascular Isquémico/metabolismo , Accidente Cerebrovascular Isquémico/prevención & control , Simulación del Acoplamiento Molecular , Factor 2 Relacionado con NF-E2/efectos de los fármacos , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico Sintasa de Tipo III/efectos de los fármacos , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
20.
Zhongguo Zhong Yao Za Zhi ; 49(10): 2745-2753, 2024 May.
Artículo en Chino | MEDLINE | ID: mdl-38812175

RESUMEN

This study investigated the protective effect of ginsenoside Rg_1(GRg_1) on oxygen and glucose deprivation/reoxygenation(OGD/R)-injured rat adrenal pheochromocytoma(PC12) cells and whether the underlying mechanism was related to the regulation of inositol-requiring enzyme 1(IRE1)-c-Jun N-terminal kinase(JNK)-C/EBP homologous protein(CHOP) signaling pathway. An OGD/R model was established in PC12 cells, and PC12 cells were randomly classified into control, model, OGD/R+GRg_1(0.1, 1, 10 µmol·L~(-1)), OGD/R+GRg_1+rapamycin(autophagy agonist), OGD/R+GRg_1+3-methyladenine(3-MA,autophagy inhibitor), OGD/R+GRg_1+tunicamycin(endoplasmic reticulum stress agonist), OGD/R+GRg_1+4-phenylbutyric acid(4-PBA, endoplasmic reticulum stress inhibitor), and OGD/R+GRg_1+3,5-dibromosalicylaldehyde(DBSA, IRE1 inhibitor) groups. Except the control group, the other groups were subjected to OGD/R treatment, i.e., oxygen and glucose deprivation for 6 h followed by reoxygenation for 6 h. Cell viability was detected by the 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium bromide(MTT) assay. Apoptosis was detected by Hoechst 33342 staining, and the fluorescence intensity of autophagosomes by the monodansylcadaverine(MDC) assay. Western blot was employed to determine the expression of autophagy-related proteins(Beclin1, LC3-Ⅱ, and p62) and the pathway-related proteins [IRE1, p-IRE1, JNK, p-JNK, glucose-regulated protein 78(GRP78), and CHOP]. The results showed that GRg_1 dose-dependently increased the viability of PC12 cells and down-regulated the expression of Beclin1, LC3-Ⅱ, p-IRE1, p-JNK, GRP78, and CHOP, compared with the model group. Furthermore, GRg_1 decreased the apoptosis rate and MDC fluorescence intensity and up-regulated the expression of p62 protein. Compared with the OGD/R+GRg_1(10 µmol·L~(-1)) group, OGD/R+GRg_1+rapamycin and OGD/R+GRg_1+tunicamycin groups showed increased apoptosis rate and MDC fluorescence intensity, up-regulated protein levels of Beclin1, LC3-Ⅱ, p-IRE1, p-JNK, GRP78, and CHOP, decreased relative cell survival rate, and down-regulated protein level of p62. The 3-MA, 4-PBA, and DBSA groups exerted the opposite effects. Taken together, GRg_1 may ameliorate OGD/R-induced PC12 cell injury by inhibiting autophagy via the IRE1-JNK-CHOP pathway.


Asunto(s)
Apoptosis , Ginsenósidos , Glucosa , Proteínas Serina-Treonina Quinasas , Factor de Transcripción CHOP , Animales , Ratas , Células PC12 , Factor de Transcripción CHOP/metabolismo , Factor de Transcripción CHOP/genética , Glucosa/metabolismo , Ginsenósidos/farmacología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Apoptosis/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Autofagia/efectos de los fármacos , Endorribonucleasas/metabolismo , Endorribonucleasas/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/genética , Oxígeno/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Complejos Multienzimáticos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA