Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
1.
Redox Rep ; 29(1): 2332038, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38563333

RESUMO

OBJECTIVES: Gentamicin is one of the most common ototoxic drugs that can lower patients' quality of life. Oxidative stress is a key factors inducing sensory hair cell death during gentamicin administration. So far, there are no effective drugs to prevent or treat gentamicin- induced hearing loss. A recent study found cystic fibrosis transmembrane conductance regulator (CFTR) as a new target to modulate cellular oxidative balance. The objective of this study was to estimate the effect of the CFTR activator ivacaftor on gentamicin-induced ototoxicity and determine its mechanism. METHODS: The hair cell count was analyzed by Myosin 7a staining. Apoptosis was analyzed by TUNEL Apoptosis Kit. Cellular reactive oxygen species (ROS) level was detected by DCFH-DA probes. The Nrf2 related proteins expression levels were analyzed by western blot. RESULTS: An in vitro cochlear explant model showed that gentamicin caused ROS accumulation in sensory hair cells and induced apoptosis, and this effect was alleviated by pretreatment with ivacaftor. Western blotting showed that ivacaftor administration markedly increased the protein expression of nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO1), and NAD(P)H:quinone oxidoreductase 1 (NQO1). The protective effect of ivacaftor was abolished by the Nrf2 inhibitor ML385. DISCUSSION: Our results indicate the protective role of the CFTR-Nrf2-HO1/NQO1 pathway in gentamicin-induced ototoxicity. Ivacaftor may be repositioned or repurposed towards aminoglycosides-induced hearing loss.


Assuntos
Aminofenóis , Perda Auditiva , Ototoxicidade , Quinolonas , Humanos , Gentamicinas/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Qualidade de Vida , Estresse Oxidativo , Apoptose , NAD(P)H Desidrogenase (Quinona)/metabolismo , NAD(P)H Desidrogenase (Quinona)/farmacologia
2.
Redox Rep ; 29(1): 2341537, 2024 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38629506

RESUMO

BACKGROUND: Acrylamide is a toxic substance formed in some foods that require high-temperature cooking processes and has been implicated as a gonadotoxic agent. Zinc, on the other hand, is a known antioxidant with fertility-enhancing properties. Hence, this study was designed to explore the possible ameliorative effect of zinc in acrylamide-induced gonadotoxicity. METHODS: Twenty-four male Wistar rats were randomized into control, acrylamide (10 mg/kg of acrylamide), acrylamide + 1 mg/kg of zinc, and acrylamide + 3 mg/kg of zinc. The administration was via the oral route and lasted for 56 days. RESULTS: Zinc treatment ameliorated acrylamide-impaired sperm quality, normal testicular histoarchitecture, and hormonal balance, which was accompanied by increased testicular malondialdehyde and interleukin-1ß and decreased testicular superoxide dismutase (SOD) and catalase (CAT). Furthermore, zinc prevented acrylamide-induced downregulation of testicular nuclear factor erythroid 2-related factor 2 (Nrf2), heme oxygenase-1 (HO-1), and B-cell lymphoma 2 (BCl2) expression and upregulation of testicular nuclear factor kappa B (NF-κB) and bcl-2-like protein 4 (bax) expression. CONCLUSION: In conclusion, zinc may protect against acrylamide-induced testicular toxicity, mediated by its antioxidant, anti-inflammatory, and antiapoptotic effects.


Assuntos
Antioxidantes , NF-kappa B , Ratos , Animais , Masculino , NF-kappa B/metabolismo , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Proteína X Associada a bcl-2/metabolismo , Proteína X Associada a bcl-2/farmacologia , Zinco/farmacologia , Acrilamida/toxicidade , Ratos Wistar , Sêmen/metabolismo , Estresse Oxidativo , Transdução de Sinais , Apoptose , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia
3.
Microbiol Spectr ; 12(4): e0322023, 2024 Apr 02.
Artigo em Inglês | MEDLINE | ID: mdl-38441979

RESUMO

Equid alphaherpesvirus 8 (EqHV-8) is one of the most economically important viruses that is known to cause severe respiratory disease, abortion, and neurological syndromes in equines. However, no effective vaccines or therapeutic agents are available to control EqHV-8 infection. Heme oxygenase-1 (HO-1) is an antioxidant defense enzyme that displays significant cytoprotective effects against different viral infections. However, the literature on the function of HO-1 during EqHV-8 infection is little. We explored the effects of HO-1 on EqHV-8 infection and revealed its potential mechanisms. Our results demonstrated that HO-1 induced by cobalt-protoporphyrin (CoPP) or HO-1 overexpression inhibited EqHV-8 replication in susceptible cells. In contrast, HO-1 inhibitor (zinc protoporphyria) or siRNA targeting HO-1 reversed the anti-EqHV-8 activity. Furthermore, biliverdin, a metabolic product of HO-1, mediated the anti-EqHV-8 effect of HO-1 via both the protein kinase C (PKC)ß/extracellular signal-regulated kinase (ERK)1/ERK2 and nitric oxide (NO)-dependent cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) signaling pathways. In addition, CoPP protected the mice by reducing the EqHV-8 infection in the lungs. Altogether, these results indicated that HO-1 can be developed as a promising therapeutic strategy to control EqHV-8 infection.IMPORTANCEEqHV-8 infections have threatened continuously donkey and horse industry worldwide, which induces huge economic losses every year. However, no effective vaccination strategies or drug against EqHV-8 infection until now. Our present study found that one host protien HO-1 restrict EqHV-8 replication in vitro and in vivo. Furthermore, we demonstrate that HO-1 and its metabolite biliverdin suppress EqHV-8 relication via the PKCß/ERK1/ERK2 and NO/cGMP/PKG pathways. Hence, we believe that HO-1 can be developed as a promising therapeutic strategy to control EqHV-8 infection.


Assuntos
Proteínas Quinases Dependentes de GMP Cíclico , Heme Oxigenase-1 , Cavalos , Animais , Camundongos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Proteínas Quinases Dependentes de GMP Cíclico/metabolismo , Proteínas Quinases Dependentes de GMP Cíclico/farmacologia , Biliverdina/farmacologia , Transdução de Sinais , Replicação Viral
4.
Am J Physiol Cell Physiol ; 326(2): C589-C605, 2024 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-38189132

RESUMO

The pathophysiology of muscle damage in peripheral artery disease (PAD) includes increased oxidant production and impaired antioxidant defenses. Epicatechin (EPI), a naturally occurring flavanol, has antioxidant properties that may mediate the beneficial effects of natural products such as cocoa. In a phase II randomized trial, a cocoa-flavanol-rich beverage significantly improved walking performance compared with a placebo in people with PAD. In the present work, the molecular mechanisms underlying the therapeutic effect of cocoa flavanols were investigated by analyzing baseline and follow-up muscle biopsies from participants. Increases in nuclear factor erythroid 2-related factor 2 (Nrf2) target antioxidants heme oxygenase-1 (HO-1) and NAD(P)H dehydrogenase [quinone] 1 (NQO1) in the cocoa group were significantly associated with reduced accumulation of central nuclei, a myopathy indicator, in type II muscle fibers (P = 0.017 and P = 0.023, respectively). Protein levels of the mitochondrial respiratory complex III subunit, cytochrome b-c1 complex subunit 2 (UQCRC2), were significantly higher in the cocoa group than in the placebo group (P = 0.032), and increases in UQCRC2 were significantly associated with increased levels of Nrf2 target antioxidants HO-1 and NQO1 (P = 0.001 and P = 0.035, respectively). Exposure of non-PAD human myotubes to ex vivo serum from patients with PAD reduced Nrf2 phosphorylation, an indicator of activation, increased hydrogen peroxide production and oxidative stress, and reduced mitochondrial respiration. Treatment of myotubes with EPI in the presence of serum from patients with PAD increased Nrf2 phosphorylation and protected against PAD serum-induced oxidative stress and mitochondrial dysfunction. Overall, these findings suggest that cocoa flavanols may enhance antioxidant capacity in PAD via Nrf2 activation.NEW & NOTEWORTHY The current study supports the hypothesis that in people with PAD, cocoa flavanols activate Nrf2, thereby increasing antioxidant protein levels, protecting against skeletal muscle damage, and increasing mitochondrial protein abundance. These results suggest that Nrf2 activation may be an important therapeutic target for improving walking performance in people with PAD.


Assuntos
Cacau , Catequina , Doença Arterial Periférica , Humanos , Antioxidantes/metabolismo , Antioxidantes/farmacologia , Cacau/química , Catequina/metabolismo , Catequina/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Músculos , Fator 2 Relacionado a NF-E2/metabolismo , Estresse Oxidativo , Doença Arterial Periférica/tratamento farmacológico , Doença Arterial Periférica/metabolismo , Polifenóis/metabolismo , Polifenóis/farmacologia
5.
J Cardiovasc Transl Res ; 17(1): 183-196, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37603208

RESUMO

Ferroptosis plays a critical role in pulmonary arterial hypertension (PAH)-induced right ventricular (RV) dysfunction, but key genes remain largely unclear. We here identified HMOX1 as an essential ferroptosis-related differentially expressed gene in PAH by bioinformatic analysis using FerrDb, GSE119754, and GSE3675 datasets, respectively. Notably, there were marked increases in HMOX1 and iron levels in RV of monocrotaline-induced PAH rats with reduced TAPSE levels. More importantly, treatment with ferrostatin-1 effectively attenuated RV hypertrophy, remodeling, myocardial fibrosis, and dysfunction in PAH rats. In cultured H9C2 cells and primary neonatal rat cardiomyocytes, pretreatment with ferrostatin-1 and knockdown HMOX1 by siRNA strikingly blunted hypoxia-induced promotion of lipid peroxidation, ferroptosis, and cardiomyocyte injury by potentiating glutathione (GSH) and nitric oxide signaling, respectively. In summary, ferrostatin-1 attenuates RV hypertrophy, fibrosis, and dysfunction in PAH by suppressing the HMOX1/GSH signaling. Targeting HMOX1 ferroptosis signaling functions as a potential therapeutic strategy for patients with PAH.


Assuntos
Cicloexilaminas , Hipertensão Pulmonar , Fenilenodiaminas , Hipertensão Arterial Pulmonar , Disfunção Ventricular Direita , Humanos , Ratos , Animais , Hipertrofia Ventricular Direita/etiologia , Hipertrofia Ventricular Direita/prevenção & controle , Hipertensão Arterial Pulmonar/tratamento farmacológico , Hipertensão Arterial Pulmonar/genética , Miócitos Cardíacos , Remodelação Ventricular , Modelos Animais de Doenças , Heme Oxigenase-1/genética , Heme Oxigenase-1/farmacologia , Heme Oxigenase-1/uso terapêutico
6.
Int J Immunopathol Pharmacol ; 37: 3946320231219348, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38031977

RESUMO

OBJECTIVE: Our research aims to assess the influence of erastin, a ferroptosis-inducing agent, on cervical cancer cells. INTRODUCTION: Cervical cancer is a prevalent malignancy in females. Dysregulation of ferroptosis, a form of cell demise reliant on iron, is implicated in several cancers. METHODS: The effect of erastin on HeLa and SiHa was detected by transwell assay, scratch test, and colony formation assay, while cell apoptosis was detected using flow cytometry. Cellular reactive oxygen species (ROS) generation was detected using the dichloro-dihydro-fluorescein diacetate assay. Sequencing analysis identified differentially expressed genes (DEGs), and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Enrichment analyses were employed to identify the target gene. Subsequently, the utilization of small interfering RNA (siRNA) was employed to suppress the targeted gene expression in HeLa cells, thereby effectively mitigating the impact of erastin on various cellular processes including invasion, colony formation, migration, and ROS generation. RESULTS: The findings indicate that erastin attenuates the viability of both HeLa cells (IC50 = 30.88 µM) and SiHa cells (IC50 = 29.40 µM). Treatment with erastin at 10 µM inhibits the invasion, colony formation, and migration of both HeLa and SiHa cells within 24 h. Ferrostatin-1 (1 µM) notably alleviates the inhibitory effects of erastin of HeLa and SiHa cells. Upregulation of nuclear factor erythroid 2-related factor 2 (Nrf2) and its downstream target, heme oxygenase-1 (HO-1), was found in erastin-treated cells compared to the control group. When knocked down HO-1 in HeLa cells, effectively counteracting the effects of erastin on the invasion, colony formation, migration, and ROS production in HeLa cells. CONCLUSION: Our research demonstrates that erastin induces ferroptosis and the accumulation of ROS in cervical cancer cells by activating the Nrf2/HO-1 pathway, significantly reducing cell proliferation and motility. These findings propose a potential molecular mechanism of erastin-mediated cervical cancer development.


Assuntos
Ferroptose , Neoplasias do Colo do Útero , Feminino , Humanos , Fator 2 Relacionado a NF-E2 , Neoplasias do Colo do Útero/tratamento farmacológico , Células HeLa , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Espécies Reativas de Oxigênio/metabolismo , RNA Interferente Pequeno , Transdução de Sinais
7.
Anticancer Agents Med Chem ; 23(20): 2209-2216, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37711016

RESUMO

BACKGROUND: Osteosarcoma is the most common malignant bone tumor, with highly proliferative and metastatic properties. Previous studies have reported that arctigenin (Arc), a bioactive lignin compound, showed excellent anti-tumor activities in a variety of human cancers. However, its role in osteosarcoma has not been studied. OBJECTIVE: We aimed to investigate the anti-tumor effects of Arc on osteosarcoma cell proliferation, migration, invasion, apoptosis, and cell cycle. METHODS: Effects of Arc on osteosarcoma cell proliferation were detected by MTT and colony formation assay. Flow cytometry analysis was performed to assess the cell apoptosis and cycle arrest. Transwell assay was used to evaluate the capability of migration and invasion. qRT-PCR and Western blot were employed to determine the changes in mRNA and protein levels. RESULTS: Arc could significantly suppress the proliferation, colony formation, and induce cell apoptosis and S phase cycle arrest of MG63 and U-2 OS cells in a dose-dependent manner. In addition, we also observed an inhibitory effect of Arc treatment on osteosarcoma cell invasion, migration, and epithelial-mesenchymal transition (EMT). HMOX1, encoding enzyme heme oxygenase-1, was predicted to be a candidate target of Arc using STITCH. Arc treatment significantly reduced the mRNA and protein levels of HMOX1. Furthermore, overexpression of HMOX1 could partly reverse the inhibitory effects of Arc on osteosarcoma cell malignant phenotypes. CONCLUSION: Our results suggest that Arc inhibits the proliferation, metastasis and promotes cell apoptosis and cycle arrest of osteosarcoma cells by downregulating HMOX1 expression.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Humanos , Heme Oxigenase-1/genética , Heme Oxigenase-1/farmacologia , Linhagem Celular Tumoral , Apoptose , Proliferação de Células , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Osteossarcoma/metabolismo , RNA Mensageiro , Movimento Celular
8.
Turk J Gastroenterol ; 34(11): 1171-1179, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37768306

RESUMO

BACKGROUND/AIMS: The major complication of liver resection is hepatic ischemia/reperfusion injury. Propofol appears to have organprotective effects. Our study aimed to study the protective role of propofol against hepatic ischemia/reperfusion injury and the potential mechanisms. MATERIALS AND METHODS: Mice and human hepatocytes (LO2) were used to establish 2 models: the ischemia/reperfusion injury model in vivo and the hypoxia/reoxygenation model in vitro, respectively. Alanine and aspartate aminotransferase serum levels were detected to evaluate the extent of hepatic cellular injury. Malondialdehyde, superoxide dismutase, glutathione, and catalase expression levels were measured to evaluate the oxidative damage in mice liver. Lactate dehydrogenase levels were detected for hepatocyte cytotoxicity severity. Nuclear factor, erythroid-like 2 and heme oxygenase 1 expression levels were detected. RESULTS: In the ischemia/reperfusion model, propofol pretreatment significantly reduced the alanine aminotransferase and aspartate aminotransferase expression levels, alleviating the hepatic cellular injury. Propofol also protected the mice liver from oxidative damage. In the hypoxia/reoxygenation model, propofol pretreatment reduced lactate dehydrogenase expression levels, suggesting its protective effects in LO2 cells. Furthermore, propofol increased the nuclear factor, erythroid-like 2 and heme oxygenase 1 expression levels both in vivo and in vitro. CONCLUSION: Propofol acts through the nuclear factor, erythroid-like 2, and heme oxygenase 1 pathway to protect the mice liver against ischemia/reperfusion injury and hepatocytes against hypoxia/reoxygenation injury. Propofol should be used as an effective therapeutic drug for hepatic ischemia/reperfusion injury.


Assuntos
Hepatopatias , Propofol , Traumatismo por Reperfusão , Humanos , Camundongos , Animais , Propofol/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Heme Oxigenase-1/uso terapêutico , Hepatócitos/metabolismo , Hepatopatias/etiologia , Hepatopatias/prevenção & controle , Hepatopatias/tratamento farmacológico , Traumatismo por Reperfusão/prevenção & controle , Traumatismo por Reperfusão/tratamento farmacológico , Isquemia/metabolismo , Hipóxia/tratamento farmacológico , Hipóxia/metabolismo , Aspartato Aminotransferases , Lactato Desidrogenases/metabolismo
9.
Planta Med ; 89(15): 1468-1482, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-37541437

RESUMO

Boswellia is a traditional medicine for bruises and injuries. Its main active ingredient, acetyl-11-keto-beta-boswellic acid, has antioxidant and antiapoptotic effects. In this experiment, we used Sprague-Dawley rats to make a sciatic nerve injury model to detect the transcription factor NF-E2-related factor 2/heme oxygenase 1 signaling pathway and apoptosis, combined with clinical indicators, for testing whether acetyl-11-keto-beta-boswellic acid can reduce oxidative stress and promote sciatic nerve repair. Our results showed that acetyl-11-keto-beta-boswellic acid administration promoted myelin regeneration and functional recovery in the rat sciatic nerve, reduced lipid peroxidation levels, upregulated the expression of various antioxidant enzymes and enhanced enzyme activity, decreased the expression levels of apoptosis-related proteins, and promoted nuclear translocation of the transcription factor NF-E2-related factor 2 protein. In vitro studies revealed that acetyl-11-keto-beta-boswellic acid reduced H2O2-induced reactive oxygen species production, restored mitochondrial membrane potential, upregulated the expression of various antioxidant enzymes, and downregulated apoptosis-related indicators in Schwann cells, and these therapeutic effects of acetyl-11-keto-beta-boswellic acid were reversed after ML385 treatment in Schwann cells. In summary, acetyl-11-keto-beta-boswellic acid alleviates oxidative stress and apoptosis caused by sciatic nerve injury in rats by activating the transcription factor NF-E2-related factor 2/heme oxygenase 1 signaling pathway, promotes the recovery of sciatic nerve function in rats, and is a promising therapeutic agent to promote sciatic nerve repair by alleviating excessive oxidative stress.


Assuntos
Heme Oxigenase-1 , Triterpenos , Ratos , Animais , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Ratos Sprague-Dawley , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Peróxido de Hidrogênio/metabolismo , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo , Transdução de Sinais , Triterpenos/farmacologia , Triterpenos/uso terapêutico , Triterpenos/metabolismo , Nervo Isquiático/metabolismo , Células de Schwann/metabolismo
10.
Allergol Immunopathol (Madr) ; 51(4): 10-18, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37422775

RESUMO

BACKGROUND: Asthma is a severe chronic respiratory disease affecting all age groups with increasing prevalence. Anti-inflammatory strategies are promising options for the treatment of asthma. Although the inhibitory effect of aloin on inflammation has been demonstrated in various diseases, its effect on asthma remains unknown. METHODS: A mice asthma model was established by treating with ovalbumin (OVA). The effects and mechanism of aloin on the OVA-treated mice were determined by enzyme-linked--immunosorbent serologic assay, biochemical examination, hematoxylin and eosin and Masson's staining, and Western blot assay. RESULTS: OVA treatment in mice significantly increased the number of total cells, neutrophils, eosinophils, and macrophages and the concentration of interleukin (IL)-4, IL-5, and IL-13, which were attenuated with the administration of aloin. The content of malondialdehyde was enhanced in OVA-treated mice, with the decreased levels of superoxide dismutase and glutathione, which were reversed with aloin treatment. Aloin treatment reduced the airway resistance of OVA-induced mice. The inflammatory cell infiltration around small airways was accompanied by the thickening and contraction of bronchial walls and pulmonary collagen deposition in OVA-treated mice; however, these conditions were ameliorated with aloin treatment. Mechanically, aloin upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2)-heme oxygenase 1 (HO-1) pathway but inhibited the level of transforming growth factor beta-SMAD2/3 genes (TGF-ß/Smad2/3) axis in OVA-induced mice. CONCLUSION: Aloin treatment lessened airway hyperresponsiveness, airway remodeling, inflammation, and oxidative stress in OVA-treated mice, and was closely related to the activation of Nrf2/HO-1 pathway and the weakening of TGF-ß/Smad2/3 pathway.


Assuntos
Asma , Fator 2 Relacionado a NF-E2 , Animais , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Ovalbumina , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Pulmão , Inflamação/metabolismo , Camundongos Endogâmicos BALB C , Modelos Animais de Doenças
11.
J Neuropathol Exp Neurol ; 82(8): 722-733, 2023 07 20.
Artigo em Inglês | MEDLINE | ID: mdl-37403613

RESUMO

Vitamin D supplementation is reported to have anti-inflammatory and neuroprotective effects during cerebral ischemia-reperfusion injury (CIRI), but the protective mechanism has not been fully elucidated. In this study, rats were given prior administrations of 1,25-vitamin D3 (1,25-VitD3) for a week and subjected to 2 hours of middle cerebral artery occlusion (MCAO) followed by 24 hours of reperfusion. Supplementation with 1,25-VitD3 significantly reduced neurological deficit scores and cerebral infarction areas, and increased surviving neurons. Oxygen-glucose deprivation/reoxygenation (OGD/R)-induced rat cortical neuron cells (RN-C) were subjected to 1,25-VitD3 treatment. Administration of 1,25-VitD3 improved cell viability and inhibited lactate dehydrogenase (LDH) activity and cell apoptosis in OGD/R-stimulated RN-C, as assessed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-h-tetrazolium bromide (MTT) assay, LDH activity assays and TdT-mediated dUTP nick end labeling (TUNEL) staining, respectively. Notably, western blot assay showed that 1,25-VitD3 upregulated nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) to alleviate oxidative stress, but reduced proteins and inflammatory cytokines related to NLR pyrin domain containing 3 (NLRP3)-mediated pyroptosis, resulting in decreased pyroptosis and neuroinflammation in vivo and in vitro. Transfection of pcDNA-Nrf2 in RN-C also inhibited pyroptosis and OGD/R-induced cell death whereas breakdown of Nrf2 signals destroyed the protective effect of 1,25-VitD3 on OGD/R-stimulated RN-C. In conclusion, 1,25-VitD3 protects neurons against CIRI through activating the antioxidant Nrf2/HO-1 pathway to restrain NLRP3-mediated pyroptosis.


Assuntos
Isquemia Encefálica , Traumatismo por Reperfusão , Ratos , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Antioxidantes/metabolismo , Piroptose , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Vitamina D/metabolismo , Vitamina D/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Transdução de Sinais , Isquemia Encefálica/metabolismo , Estresse Oxidativo , Infarto da Artéria Cerebral Média/metabolismo , Traumatismo por Reperfusão/tratamento farmacológico , Traumatismo por Reperfusão/metabolismo , Oxigênio , Reperfusão , Neurônios/metabolismo
12.
Acta Biomater ; 166: 604-614, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37156432

RESUMO

Ferroptosis-based nanoplatforms have shown great potential in cancer therapy. However, they also face issues such as degradation and metabolism. Carrier-free nanoplatforms consisting of active drugs can effectively avoid the security issues associated with additional carrier ingredients. Herein, a biomimetic carrier-free nanoplatform (HESN@CM) was designed to treat cancer by modulating cascade metabolic pathways of ferroptosis. CCR2-overexpressing macrophage membrane-modified HESN can target cancer cells via the CCR2-CCL2 axis. The acidic tumor microenvironment (TME) can disrupt the supramolecular interaction of HESN, releasing hemin and erastin. Then, erastin could induce cancer cells ferroptosis by inhibiting system XC- pathways, while hemin, a vital component of blood to transport oxygen, could be broken down by heme oxygenase-1 (HO-1), increasing the intracellular Fe2+ concentration to induce cancer cells' ferroptosis further. Meanwhile, erastin could enhance the activity of HO-1, further promoting the release of Fe2+ from hemin. As a result, HESN@CM demonstrated superior therapeutic efficacy in both primary and metastatic tumors in vitro and in vivo. The carrier-free HESN@CM provided cascade ferroptosis tumor therapy strategies for potential clinical application. STATEMENT OF SIGNIFICANCE: CCR2-overexpressing biomimetic carrier-free nanoplatform (HESN@CM) was designed for cancer treatment by modulating metabolic pathways of ferroptosis. HESN modified with CCR2-overexpressing macrophage membrane can target tumor cells via the CCR2-CCL2 axis. HESN was composed of hemin and erastin without additional vectors. Erastin could directly induce ferroptosis, while hemin could be broken down by heme oxygenase-1 (HO-1), increasing the intracellular Fe2+ concentration to enhance ferroptosis further. Meanwhile, erastin could improve the activity of HO-1, promoting the release of Fe2+ from hemin. Therefore, HESN@CM with good bioavailability, stability, and simple preparation can realize cascade ferroptosis tumor therapy and have the potential prospect of clinical translation.


Assuntos
Ferroptose , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Hemina/farmacologia , Biomimética , Linhagem Celular Tumoral
13.
Braz. J. Anesth. (Impr.) ; 73(2): 177-185, March-Apr. 2023. graf
Artigo em Inglês | LILACS | ID: biblio-1439592

RESUMO

Abstract Background The precise underlying mechanism of antioxidant effects of dexmedetomidine-induced neuroprotection against cerebral ischemia has not yet been fully elucidated. Activation of Nuclear factor erythroid 2-related factor (Nrf2) and Heme Oxygenase-1 (HO-1) represents a major antioxidant-defense mechanism. Therefore, we determined whether dexmedetomidine increases Nrf2/HO-1 expression after global transient cerebral ischemia and assessed the involvement of Protein Kinase C (PKC) in the dexmedetomidine-related antioxidant mechanism. Methods Thirty-eight rats were randomly assigned to five groups: sham (n = 6), ischemic (n = 8), chelerythrine (a PKC inhibitor; 5 mg.kg-1 IV administered 30 min before cerebral ischemia) (n = 8), dexmedetomidine (100 µg.kg-1 IP administered 30 min before cerebral ischemia (n = 8), and dexmedetomidine + chelerythrine (n = 8). Global transient cerebral ischemia (10 min) was applied in all groups, except the sham group; histopathologic changes and levels of nuclear Nrf2 and cytoplasmic HO-1 were examined 24 hours after ischemia insult. Results We found fewer necrotic and apoptotic cells in the dexmedetomidine group relative to the ischemic group (p< 0.01) and significantly higher Nrf2 and HO-1 levels in the dexmedetomidine group than in the ischemic group (p< 0.01). Additionally, chelerythrine co-administration with dexmedetomidine attenuated the dexmedetomidine-induced increases in Nrf2 and HO-1 levels (p< 0.05 and p< 0.01, respectively) and diminished its beneficial neuroprotective effects. Conclusion Preischemic dexmedetomidine administration elicited neuroprotection against global transient cerebral ischemia in rats by increasing Nrf2/HO-1 expression partly via PKC signaling, suggesting that this is the antioxidant mechanism underlying dexmedetomidine-mediated neuroprotection.


Assuntos
Animais , Ratos , Traumatismo por Reperfusão/prevenção & controle , Isquemia Encefálica , Proteína Quinase C/metabolismo , Proteína Quinase C/farmacologia , Ataque Isquêmico Transitório , Estresse Oxidativo , Fármacos Neuroprotetores/farmacologia , Dexmedetomidina/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Heme Oxigenase (Desciclizante)/farmacologia , Antioxidantes/metabolismo , Antioxidantes/farmacologia
14.
Biomater Adv ; 148: 213363, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36881963

RESUMO

Silver nanoparticles (AgNPs) have inconsistent findings against inflammation. Although a wealth of literature on the beneficial effects of green-synthesized AgNPs has been published, a detailed mechanistic study of green AgNPs on the protective effects against lipopolysaccharide (LPS)-induced neuroinflammation using human microglial cells (HMC3) has not yet been reported. For the first time, we studied the inhibitory effect of biogenic AgNPs on inflammation and oxidative stress induced by LPS in HMC3 cells. X-ray photoelectron spectroscopy, Fourier-transform infrared spectroscopy, and transmission electron microscopy were used to characterize AgNPs produced from honeyberry. Co-treatment with AgNPs significantly reduced mRNA expressions of inflammatory molecules such as interleukin (IL)-6 and tumor necrosis factor-α, while increasing the expressions of anti-inflammatory markers such as IL-10 and transforming growth factor (TGF)-ß. HMC3 cells were also switched from M1 to M2, as shown by lower expression of M1 markers such as cluster of differentiation (CD)80, CD86, and CD68 and higher expression of M2 markers such as CD206, CD163, and triggering receptors expressed on myeloid cells (TREM2). Furthermore, AgNPs inhibited LPS-induced toll-like receptor (TLR)4 signaling, as evidenced by decreased expression of myeloid differentiation factor 88 (MyD88) and TLR4. In addition, AgNPs reduced the production of reactive oxygen species (ROS) and enhanced the expression of nuclear factor-E2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1), while decreasing the expression of inducible nitric oxide synthase. The docking score of the honeyberry phytoconstituents ranged from -14.93 to - 4.28 KJ/mol. In conclusion, biogenic AgNPs protect against neuroinflammation and oxidative stress by targeting TLR4/MyD88 and Nrf2/HO-1 signaling pathways in a LPS-induced in vitro model. Biogenic AgNPs could be utilized as potential nanomedicine against LPS-induced inflammatory disorders.


Assuntos
Lipopolissacarídeos , Nanopartículas Metálicas , Humanos , Lipopolissacarídeos/toxicidade , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Fator 88 de Diferenciação Mieloide/farmacologia , Simulação de Acoplamento Molecular , Prata/farmacologia , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Doenças Neuroinflamatórias , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo , Nanopartículas Metálicas/uso terapêutico , Transdução de Sinais , Fenótipo , Linhagem Celular , Inflamação/induzido quimicamente , Inflamação/metabolismo , Encéfalo/metabolismo
15.
J Appl Microbiol ; 134(1)2023 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-36724245

RESUMO

AIMS: The occurrence of alcoholic liver injury is related to the oxidative stress. Bacteria for alleviating alcoholic related liver injury have received widespread attention. Study aims to investigate the alleviated efficacy of Lactiplantibacillus plantarum (L. plantarum) P101 on alcohol-induced liver injury and its potential mechanism. METHODS AND RESULTS: The model of alcoholic liver injury was obtained according to the NIAAA method and the mice were treated with L. plantarum P101 (108 CFU.mice-1). Results showed that treatment of L. plantarum P101 could significantly improve liver function and antioxidant capacity. Furthermore, L. plantarum P101 significantly up-regulated Nuclear factor erythroid 2-related factor (Nrf2) and its target molecule, Hemeoxygenase 1 (HO-1), by promoting nuclear translocation of Nrf2. Moreover, inflammatory factors and pro-apoptotic protein (Caspase3) levels were significantly decreased in mice treated with L. plantarum P101. CONCLUSIONS: This study confirmed that the beneficial effect of L. plantarum P101 supplement was achieved via regulating Nrf2/HO-1 antioxidant pathway, and alleviated alcoholic liver injury.


Assuntos
Antioxidantes , Fator 2 Relacionado a NF-E2 , Animais , Camundongos , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Fígado , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Estresse Oxidativo , Lactobacillaceae/química
16.
Vascular ; 31(4): 818-827, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35321597

RESUMO

OBJECTIVE: As one of the independent risk factors for atherosclerosis (AS), oxidized low-density lipoprotein (ox-LDL) can trigger damage to the vascular intima and induce the expression of various adhesion molecules. This study aimed to explore the effects of galangin, an extract of galangal, on ox-LDL-induced vascular endothelial cells. METHODS: The effects of different concentrations of galangin or ox-LDL on the metabolic activity of vascular endothelial cells were determined using the CCK8 assay. Afterward, the role of galangin in the expression levels of inflammatory factors was assessed using RT-qPCR and Western blotting. In addition, the influences of galangin on apoptosis and endothelial-mesenchymal transition (EndMT) were also evaluated. Through molecular docking, the Heme oxygenase-1 (HO-1) signaling pathway was proposed, and then the effects of the HO-1 signaling pathway on the regulatory roles of galangin were evaluated. RESULTS: In this study, galangin was found to effectively increase the metabolic activity of ox-LDL-induced cells in a concentration-dependent manner. In addition, galangin was found to reduce ox-LDL-induced cell inflammation, apoptosis, and EndMT. Moreover, galangin could combine with HO-1 and regulate the HO-1 signaling pathway. The effects of galangin on cells were shown to be through the HO-1 signaling pathway. CONCLUSION: To sum up, galangin reduced ox-LDL-induced inflammation, apoptosis, and EndMT of vascular endothelial cells via regulating the HO-1 signaling pathway.


Assuntos
Células Endoteliais , Heme Oxigenase-1 , Humanos , Células Endoteliais/metabolismo , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Simulação de Acoplamento Molecular , Transdução de Sinais , Lipoproteínas LDL , Apoptose , Inflamação/metabolismo
17.
Braz J Anesthesiol ; 73(2): 177-185, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-34560114

RESUMO

BACKGROUND: The precise underlying mechanism of antioxidant effects of dexmedetomidine-induced neuroprotection against cerebral ischemia has not yet been fully elucidated. Activation of Nuclear factor erythroid 2-related factor (Nrf2) and Heme Oxygenase-1 (HO-1) represents a major antioxidant-defense mechanism. Therefore, we determined whether dexmedetomidine increases Nrf2/HO-1 expression after global transient cerebral ischemia and assessed the involvement of Protein Kinase C (PKC) in the dexmedetomidine-related antioxidant mechanism. METHODS: Thirty-eight rats were randomly assigned to five groups: sham (n...=...6), ischemic (n...=...8), chelerythrine (a PKC inhibitor; 5...mg.kg-1 IV administered 30...min before cerebral ischemia) (n...=...8), dexmedetomidine (100.....g.kg-1 IP administered 30...min before cerebral ischemia (n...=...8), and dexmedetomidine...+...chelerythrine (n...=...8). Global transient cerebral ischemia (10...min) was applied in all groups, except the sham group; histopathologic changes and levels of nuclear Nrf2 and cytoplasmic HO-1 were examined 24...hours after ischemia insult. RESULTS: We found fewer necrotic and apoptotic cells in the dexmedetomidine group relative to the ischemic group (p...<...0.01) and significantly higher Nrf2 and HO-1 levels in the dexmedetomidine group than in the ischemic group (p...<...0.01). Additionally, chelerythrine co-administration with dexmedetomidine attenuated the dexmedetomidine-induced increases in Nrf2 and HO-1 levels (p...<...0.05 and p...<...0.01, respectively) and diminished its beneficial neuroprotective effects. CONCLUSION: Preischemic dexmedetomidine administration elicited neuroprotection against global transient cerebral ischemia in rats by increasing Nrf2/HO-1 expression partly via PKC signaling, suggesting that this is the antioxidant mechanism underlying dexmedetomidine-mediated neuroprotection.


Assuntos
Isquemia Encefálica , Dexmedetomidina , Ataque Isquêmico Transitório , Fármacos Neuroprotetores , Traumatismo por Reperfusão , Ratos , Animais , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Antioxidantes/farmacologia , Antioxidantes/metabolismo , Proteína Quinase C/metabolismo , Proteína Quinase C/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Fator 2 Relacionado a NF-E2/farmacologia , Dexmedetomidina/farmacologia , Ratos Sprague-Dawley , Estresse Oxidativo , Fármacos Neuroprotetores/farmacologia , Traumatismo por Reperfusão/prevenção & controle
18.
Planta Med ; 89(1): 30-45, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35764305

RESUMO

Various age-related chronic diseases have been linked to oxidative stress. The cellular antioxidant response pathway is regulated by the transcription factor nuclear erythroid factor 2. Therefore, plant-derived nuclear erythroid factor 2 activators might be useful therapeutics to stimulate the body's defense mechanisms. Our study focused on the discovery of potent nuclear erythroid factor 2 activators from medicinal plants. Initially, a variety of medicinal plant extracts were screened for nuclear erythroid factor 2 activity using a nuclear erythroid factor 2 luciferase reporter cell line. Among these, Valerian (Valeriana officinalis) root was identified as a potent candidate. Sequential extraction and bioassay-guided fractionation led to the isolation of four nuclear erythroid factor 2-active compounds, which were structurally identified by NMR and LC/HRMS as the known compounds isovaltrate, valtrate, jatamanvaltrate-P, and valerenic acid. These four compounds were then tested in relevant biological assays. Firstly, their effects on the expression of glutathione S-transferase, glutamate-cysteine ligase catalytic subunit, glutathione peroxidase, and heme oxygenase 1 were determined in HepG2 cells. Glutathione S-transferase P1 and glutamate-cysteine ligase catalytic subunit were upregulated by isovaltrate, valtrate, and jatamanvaltrate-P, while heme oxygenase 1 was upregulated by isovaltrate, jatamanvaltrate-P, and valerenic acid. The four compounds also increased the levels of glutathione and its metabolite, CysGly. As glutathione aids in the detoxification of hydrogen peroxide, cytoprotective effects of these four nuclear erythroid factor 2 activators against hydrogen peroxide toxicity were investigated, and indeed, the compounds significantly improved cell survival. This study provides evidence that four valepotriates from the roots of V. officinalis are activators of nuclear erythroid factor 2-mediated antioxidant and detoxification pathways. Our data might expand the medical use of this plant beyond its current application as a sleep aid.


Assuntos
Antioxidantes , Valeriana , Antioxidantes/química , Fator 2 Relacionado a NF-E2/metabolismo , Valeriana/química , Glutamato-Cisteína Ligase/metabolismo , Glutamato-Cisteína Ligase/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo , Glutationa/metabolismo
19.
Gen Thorac Cardiovasc Surg ; 71(5): 280-290, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36008747

RESUMO

OBJECTIVES: Doxorubicin is a type of effective antitumor drug but can contribute to cardiomyocyte injuries. We aimed to dissect the mechanism of the HMOX1/CTGF axis in DOX-induced cardiomyocyte injury, mitochondrial dysfunction, and ferroptosis. METHODS: Bioinformatics analysis was conducted to retrieve differentially expressed genes in a DOX-induced mouse model. Mouse cardiomyocytes, HL-1 cells, were induced with l µM DOX, after which gain- or loss-of-function assays were applied. CCK-8, fluorescent probe assay, flow cytometry, and corresponding kits were employed to detect cell viability, ROS levels, mitochondrial membrane potential and cell apoptosis, and GSH and Fe2+ contents, respectively. qRT-PCR or Western blot assay was adopted to test HMOX1, CTGF, BCL-2, Caspase3, Cleaved-Caspase3, and GPX4 expression. RESULTS: Bioinformatics analysis showed that HMOX1 and CTGF were highly expressed in DOX-induced mice and correlated with each other. Also, HMOX1 and CTGF expression was high in HL-1 cells after DOX treatment, along with an obvious decrease in cell viability and GSH and GPX4 expression, an increase in ROS levels, apoptosis, and Fe2+ contents, and mitochondrial membrane potential dysfunction or loss. HMOX1 or CTGF silencing diminished cell apoptosis, Cleaved-Caspase3 expression, Fe2+ contents, and ROS levels, enhanced cell viability and the expression of GSH, GPX4, and BCL-2, and recovered mitochondrial membrane potential in DOX-induced HL-1 cells. Nevertheless, the effects of HMOX1 silencing on the viability, apoptosis, ferroptosis, and mitochondrial dysfunction of DOX-induced HL-1 cells were counteracted by CTGF overexpression. CONCLUSIONS: In conclusion, HMOX1 silencing decreased CTGF expression to alleviate DOX-induced injury, mitochondrial dysfunction, and ferroptosis of mouse cardiomyocytes.


Assuntos
Ferroptose , Camundongos , Animais , Ferroptose/genética , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/farmacologia , Doxorrubicina/toxicidade , Doxorrubicina/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Mitocôndrias/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Membrana/farmacologia , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia
20.
J Appl Oral Sci ; 30: e20220158, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36350873

RESUMO

OBJECTIVE: Carcinoembryonic antigen-related cell adhesion molecule 1 (CEACAM1) is a member of the carcinoembryonic antigen family. Although its expression has been found in chronic oral inflammatory epithelium, this study aimed to know whether CEACAM1 in oral keratinocytes participates in host immune response against Candida albicans . METHODOLOGY: We investigated CEACAM1 expression in oral keratinocytes induced by C. albicans as well as by Candida cell wall component ß-glucan particles (ß-GPs). Furthermore, the effects of CEACAM1 on ß-GPs-induced heme oxygenase-1 (HO-1) expression and its related signals were examined. RESULTS: Fluorescence staining showed CEACAM1 expression in oral keratinocytes (RT7) cells, whereas quantitative reverse transcription (RT)-PCR indicated that both live and heat-killed C. albicans increased CEACAM1 mRNA expression in RT7 cells. Examinations using quantitative RT-PCR and western blotting indicated that CEACAM1 expression was also increased by ß-GPs derived from C. albicans . Specific siRNA for CEACAM1 decreased HO-1 expression induced by ß-GPs from C. albicans as well as the budding yeast microorganism Saccharomyces cerevisiae . Moreover, knockdown of CEACAM1 decreased ß-GPs-induced ROS activity in the early phase and translocation of Nrf2 into the nucleus. CONCLUSION: CEACAM1 in oral keratinocytes may have a critical role in regulation of HO-1 for host immune defense during Candida infection.


Assuntos
Heme Oxigenase-1 , beta-Glucanas , Heme Oxigenase-1/metabolismo , Heme Oxigenase-1/farmacologia , beta-Glucanas/farmacologia , beta-Glucanas/metabolismo , Antígeno Carcinoembrionário/metabolismo , Antígeno Carcinoembrionário/farmacologia , Molécula 1 de Adesão Celular/metabolismo , Glucanos/metabolismo , Glucanos/farmacologia , Candida , Queratinócitos , Candida albicans/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...