Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 21(1): 433, 2023 07 04.
Artigo em Inglês | MEDLINE | ID: mdl-37403143

RESUMO

BACKGROUND: Cervical cancer (CC) has poor prognosis and high mortality rate for its metastasis during the disease progression. Epithelial-mesenchymal transition (EMT) and anoikis are initial and pivotal steps during the metastatic process. Although higher levels of Nrf2 are associated with aggressive tumor behaviors in cervical cancer, the detailed mechanism of Nrf2 in cervical cancer metastasis, especially EMT and anoikis, remains unclear. METHODS: Immunohistochemistry (IHC) was used to examine Nrf2 expression in CC. Wound healing assay and transwell analysis were used to evaluate the migration ability of CC cells. Western blot, qTR-PCR and immunofluorescent staining were used to verify the expression level of Nrf2, the EMT associated markers and anoikis associated proteins. Flow cytometry assays and cell counting were used to detect the apoptosis of cervical cancer cells. The lung and lymph node metastatic mouse model were established for studies in vivo. The interaction between Nrf2 and Snail1 was confirmed by rescue-of-function assay. RESULTS: When compared with cervical cancer patients without lymph node metastasis, Nrf2 was highly expressed in patients with lymph node metastasis. And Nrf2 was proved to enhance the migration ability of HeLa and SiHa cells. In addition, Nrf2 was positively correlated with EMT processes and negatively associated with anoikis in cervical cancer. In vivo, a xenograft assay also showed that Nrf2 facilitated both pulmonary and lymphatic distant metastasis of cervical cancer. Rescue-of-function assay further revealed the mechanism that Nrf2 impacted the metastasis of CC through Snail1. CONCLUSION: Our fundings established Nrf2 plays a crucial role in the metastasis of cervical cancer by enhancing EMT and resistance to anoikis by promoting the expression of Snail1, with potential value as a therapeutic candidate.


Assuntos
Fator 2 Relacionado a NF-E2 , Neoplasias do Colo do Útero , Feminino , Animais , Camundongos , Humanos , Linhagem Celular Tumoral , Metástase Linfática/patologia , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias do Colo do Útero/patologia , Células HeLa , Transição Epitelial-Mesenquimal , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Metástase Neoplásica
2.
Biomed Pharmacother ; 165: 115097, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37406514

RESUMO

OBJECTIVE: Acetaminophen (APAP) is one of the world's popular and safe painkillers, and overdose can cause severe liver damage and even acute liver failure. The effect and mechanism of the xanthohumol on acetaminophen-induced hepatotoxicity remains unclear. METHODS: The hepatoprotective effects of xanthohumol were studied using APAP-induced HepG2 cells and acute liver injury of mouse, seperately. RESULTS: In vitro, xanthohumol inhibited H2O2- and acetaminophen-induced cytotoxicity and oxidative stress. Xanthohumol up-regulated the expression of Nrf2. Further mechanistic studies showed that xanthohumol triggered Nrf2 activation via the AMPK/Akt/GSK3ß pathway to exert a cytoprotective effect. In vivo, xanthohumol significantly ameliorated acetaminophen-induced mortality, the elevation of ALT and AST, GSH depletion, MDA formation and histopathological changes. Xanthohumol effectively suppressed the phosphorylation and mitochondrial translocation of JNK, mitochondrial translocation of Bax, the activation o cytochrome c, AIF secretion and Caspase-3. In vivo, xanthohumol increased Nrf2 nuclear transcription and AMPK, Akt and GSK3ß phosphorylation in vivo. In addition, whether xanthohumol protected against acetaminophen-induced liver injury in Nrf2 knockout mice has not been illustated. CONCLUSION: Thus, xanthohumol exerted a hepatoprotective effect by inhibiting oxidative stress and mitochondrial dysfunction through the AMPK/Akt/GSK3ß/Nrf2 antioxidant pathway.


Assuntos
Acetaminofen , Doença Hepática Induzida por Substâncias e Drogas , Animais , Camundongos , Acetaminofen/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Transdução de Sinais , Glicogênio Sintase Quinase 3 beta/metabolismo , Peróxido de Hidrogênio/farmacologia , Estresse Oxidativo , Fígado , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Doença Hepática Induzida por Substâncias e Drogas/metabolismo
3.
Biomed Pharmacother ; 165: 115047, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37390709

RESUMO

Epidemiologic studies have shown that fine particulate matter 2.5 (PM2.5) exaggerates airway inflammation associated with acute exacerbation of chronic obstructive pulmonary disease (AECOPD). Daphnetin (Daph) is a natural compound with a variety of biological activities. At present, there are limited data on whether Daph can protect against cigarette smoke (CS)-induced chronic obstructive pulmonary disease (COPD) and PM2.5-CS-induced AECOPD. Therefore, this study systematically evaluated the effects of Daph on CS-induced COPD and PM2.5-CS-induced AECOPD and determined its mechanism of action. First, in vitro studies showed that PM2.5 exacerbated cytotoxicity and NLRP3 inflammasome-mediated pyroptosis induced by low-dose cigarette smoke extracts (CSE). However, the effect was reversed by si-NLRP3 and MCC950. Similar results were obtained in PM2.5-CS-induced AECOPD mice. The results of the mechanistic studies suggested that blocking NLRP3 abolished PM2.5 combined with cigarette induced cytotoxicity, lung damage, NLRP3 inflammasome activation and pyroptosis in vitro and in vivo. Second, Daph suppressed the expression of NLRP3 inflammasome and pyroptosis in BEAS-2B cells. Third, Daph significantly protected against CS-induced COPD and PM2.5-CS-induced AECOPD by inhibiting the NLRP3 inflammasome and pyroptosis in mice. Our findings identified the NLRP3 inflammasome as a critical contributor to PM2.5-CS-induced airway inflammation, and Daph as a negative regulator of NLRP3-mediated pyroptosis, which has implications for the pathophysiology of AECOPD.


Assuntos
Inflamassomos , Doença Pulmonar Obstrutiva Crônica , Camundongos , Animais , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Material Particulado/toxicidade , Piroptose , Inflamação/tratamento farmacológico , Doença Pulmonar Obstrutiva Crônica/tratamento farmacológico
4.
Br J Pharmacol ; 180(19): 2532-2549, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37005797

RESUMO

BACKGROUND AND PURPOSE: Our previous research showed that ferroptosis plays a crucial role in the pathophysiology of PM2.5-induced lung injury. The present study aimed to investigate the protective role of the Nrf2 signalling pathway and its bioactive molecule tectoridin in PM2.5-induced lung injury by regulating ferroptosis. EXPERIMENTAL APPROACH: We examined the regulatory effect of Nrf2 on ferroptosis in PM2.5-induced lung injury and Beas-2b cells using Nrf2-knockout (KO) mice and Nrf2 siRNA transfection. The effects and underlying mechanisms of tectoridin on PM2.5-induced lung injury were evaluated in vitro and in vivo. KEY RESULTS: Nrf2 deletion increased iron accumulation and ferroptosis-related protein expression in vivo and vitro, further exacerbating lung injury and cell death in response to PM2.5 exposure. Tectoridin activated Nrf2 target genes and ameliorated cell death caused by PM2.5. In addition, tectoridin prevented lipid peroxidation, iron accumulation and ferroptosis in vitro, but in siNrf2-treated cells, these effects almost disappeared. In addition, tectoridin effectively mitigated PM2.5-induced respiratory system damage, as evaluated by HE, PAS, and inflammatory factors. Tectoridin also augmented the antioxidative Nrf2 signalling pathway and prevented changes in ferroptosis-related morphological and biochemical indicators, including MDA levels, GSH depletion and GPX4 and xCT downregulation, in PM2.5-induced lung injury. However, the effects of tectoridin on ferroptosis and respiratory injury were almost abolished in Nrf2-KO mice. CONCLUSION AND IMPLICATIONS: Our data proposed the protective effect of Nrf2 activation on PM2.5-induced lung injury by inhibiting ferroptosis-mediated lipid peroxidation and highlight the potential of tectoridin as a PM2.5-induced lung injury treatment.


Assuntos
Ferroptose , Lesão Pulmonar , Animais , Camundongos , Lesão Pulmonar/induzido quimicamente , Lesão Pulmonar/prevenção & controle , Fator 2 Relacionado a NF-E2 , Camundongos Knockout , Material Particulado/toxicidade , Ferro
5.
Cell Death Dis ; 14(2): 100, 2023 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-36765041

RESUMO

Dissemination of ovarian cancer (OvCa) cells can lead to inoperable metastatic lesions in the bowel and omentum, which have a poor prognosis despite surgical and chemotherapeutical options. A better understanding of the mechanisms underlying metastasis is urgently needed. In this study, bioinformatics analyses revealed that UBE2E2, a less-studied ubiquitin (Ub)-conjugating enzyme (E2), was upregulated in OvCa and was associated with poor prognosis. Subsequently, we performed western blot analysis and IHC staining with 88 OvCa and 26 normal ovarian tissue samples, which further confirmed that UBE2E2 protein is highly expressed in OvCa tissue but weakly expressed in normal tissue. Furthermore, the silencing of UBE2E2 blocked OvCa cell migration, epithelial-mesenchymal transition (EMT) and metastasis in vitro, whereas UBE2E2 overexpression exerted the opposite effects. Mechanistically, UBE2E2 promoted p62 accumulation and increased the activity of the Nrf2-antioxidant response element (ARE) system, which ultimately activated the Snail signaling pathway by inhibiting the ubiquitin-mediated degradation of Snail. Additionally, co-IP and immunofluorescence demonstrated that a direct interaction exists between UBE2E2 and Nrf2, and the N-terminal of UBE2E2 (residues 1-52) is required and sufficient for its interaction with Nrf2 protein. Mutations in the active site cysteine (Cys139) impaired both the function and cellular distribution of UBE2E2. More importantly, the deletion of UBE2E2 reduced tumorigenicity and metastasis in xenograft OvCa mouse models. Taken together, our findings reveal the role of the UBE2E2-Nrf2-p62-Snail signaling axis in OvCa and thus provides novel therapeutic targets for the prevention of OvCa metastasis.


Assuntos
Antioxidantes , Neoplasias Ovarianas , Feminino , Humanos , Animais , Camundongos , Antioxidantes/uso terapêutico , Fator 2 Relacionado a NF-E2/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal/genética , Neoplasias Ovarianas/patologia , Ubiquitinas , Enzimas de Conjugação de Ubiquitina/genética
6.
Phytother Res ; 36(5): 2161-2172, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35285100

RESUMO

Oxidative stress and inflammation play important roles in pleurisy. Leonurine (Leo) has been confirmed to exert antioxidative and antiinflammatory effects in many preclinical experiments, but these effects have not been studied in pleurisy. The aim of this study was to explore the therapeutic effect and mechanism of Leo in a carrageenan (CAR)-induced pleurisy model. In this study, we found that the increase of reactive oxygen species (ROS), myeloperoxidase (MPO), and malondialdehyde (MDA) and decrease of glutathione (GSH) induced by CAR could be reversed by the treatment of Leo. Leo effectively reduced the levels of proinflammatory cytokines interleukin-1ß (IL-1ß), tumor necrosis factor-α (TNF-α), and the percentages of mature macrophages and increased the levels of antiinflammatory cytokines (IL-10). Furthermore, Western blotting revealed that Leo significantly activated the Nrf2 pathway to restrain the thioredoxin-interacting protein/NOD-like receptor protein 3 (TXNIP/NLRP3) and nuclear factor kappa-B (NF-κB) pathways. However, the protective effect of Leo was significantly weakened in Nrf2-deficient mice. These results indicate that Leo confers potent protection against CAR-induced pleurisy by inhibiting the TXNIP/NLRP3 and NF-κB pathways dependent on Nrf2, which may serve as a promising agent for attenuating pleurisy.


Assuntos
NF-kappa B , Pleurisia , Animais , Carragenina/toxicidade , Proteínas de Transporte , Ácido Gálico/análogos & derivados , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pleurisia/induzido quimicamente , Pleurisia/tratamento farmacológico , Tiorredoxinas/metabolismo
8.
Med Oncol ; 38(5): 54, 2021 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-33811596

RESUMO

The fatality rate of ovarian cancer ranks first among gynecological tumors, and the prognosis is poor. Diosmetin (Dio), a natural flavonoid obtained from citrus fruits, has been shown to have anti-tumor effects in lung, liver, and skin cancers. We aimed to investigate the effects of Dio on ovarian cancer A2780 and SKOV3 cells along with the underlying mechanisms. Our data showed that Dio inhibited the proliferation, migration, and invasion of these cells and induced their apoptosis. Moreover, Dio upregulated the levels of Bax and cleaved Caspase-3 and PARP while downregulating the level of Bcl2. Mechanistically, our results revealed that Dio inhibited Nrf2 and induced the production of reactive oxygen species (ROS). The ROS scavenger N-acetyl-L-cysteine (NAC) suppressed the inhibitory effect of Dio on the proliferation of the ovarian cancer cells. Additionally, overexpression of Nrf2 partially suppressed the Dio-induced apoptosis and proliferation inhibition in these cells. These findings indicate that Dio exerts an anti-tumor activity by upregulating ROS levels and inhibiting Nrf2, indicating that Dio is a promising chemotherapeutic candidate for the treatment of ovarian cancer.


Assuntos
Apoptose/efeitos dos fármacos , Flavonoides/farmacologia , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias Ovarianas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Apoptose/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Relação Dose-Resposta a Droga , Feminino , Flavonoides/uso terapêutico , Humanos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/patologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
9.
Int Immunopharmacol ; 96: 107601, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33812255

RESUMO

Gentamicin (GM), an aminoglycoside antibiotic, is one of the most effective drugs used in the treatment of various types of bacterial infections, but the major adverse effect and drug-induced nephrotoxicity of GM limit its clinical applications. Daphnetin (Daph) is a natural coumarin derivative that is clinically used to treat rheumatoid arthritis and coagulopathy and exhibits antioxidant effects. However, the effect of Daph on GM-induced nephrotoxicity has not yet been elucidated. This study investigated Daph-mediated protection against GM-induced nephrotoxicity in mice and explored the underlying mechanisms of GM-induced renal dysfunction in mice. We found that Daph treatment significantly reduced GM-induced nephrotoxicity mainly by ameliorating renal injury in mice and attenuating cell damage in vitro. Mechanistically, we found that Daph upregulated the expression level of Nrf2 and its regulated antioxidant enzymes HO-1, NQO1, GCLC and GCLM in vivo and in vitro. GM upregulated the expression levels of NOX4, cleaved Caspase-3 and p53 and the BAX/BCL2 ratio in vivo to stimulate oxidative stress and apoptosis. However, Daph treatment significantly improved the oxidative stress and apoptosis caused by GM, thereby exerting antioxidative and antiapoptotic effects. Our study was the first to suggest that the natural product Daph protects against GM-induced nephrotoxicity through the activation of Nrf2 which regulates oxidative stress and apoptosis. The pharmacological activation of Nrf2 may be useful as a novel therapy to prevent renal injury.


Assuntos
Injúria Renal Aguda/prevenção & controle , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Gentamicinas/efeitos adversos , Estresse Oxidativo/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Umbeliferonas/farmacologia , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/patologia , Animais , Antioxidantes/administração & dosagem , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/prevenção & controle , Injeções Intraperitoneais , Camundongos Endogâmicos ICR , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Fator 2 Relacionado a NF-E2/metabolismo , Substâncias Protetoras/administração & dosagem , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Umbeliferonas/administração & dosagem
10.
Pharmacol Res ; 167: 105561, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33737241

RESUMO

Autophagy-mediated cell death plays a critical role in the pathogenesis of PMs-induced lung injury. Hyperoside (Hyp), a flavonoid glycosides, is known to exert protective effects on many diseases by inhibiting autophagic activity. The current study aimed to explore the protective effect and mechanism of Hyp against PMs-induced lung injury in PM2.5 challenged Beas-2b cells in vitro and BALB/C mice in vivo. In vitro, we found that the organic solvent-extractable fraction of SRM1649b (O-PMs) caused more severe cytotoxicity in Beas-2b cells than the water solvent-extractable fraction of SRM1649b (W-PMs). O-PMs treatment dose-dependently upregulated the expression of autophagy markers (beclin-1, p62, atg3 and LC3II) and apoptotic proteins. This cytotoxicity of O-PMs was attenuated by Hyp pretreatment in parallel with downregulation of the expression of autophagy markers, apoptotic proteins, and p-AMPK and upregulation of p-mTOR expression. Notably, the therapeutic effect of Hyp was attenuated by pretreated with AICAR (an AMPK inducer), but enhanced by CC and 3-MA treatment. In vivo, Hyp reduced pathological lung injury and decreased the levels of PMs-induced inflammatory cytokines (TNF-α and IL-6), and the number of total cells in the BALF by inhibiting AMPK/mTOR signaling. Furthermore, cotreatment with AICAR (500 mg/kg) reduced but did not abrogate the pulmonary protective effect of Hyp. These findings indicate that Hyp protects against PMs-induced lung injury by suppressing autophagy deregulation and apoptosis through regulation of the AMPK/mTOR pathway.


Assuntos
Autofagia/efeitos dos fármacos , Lesão Pulmonar/tratamento farmacológico , Lesão Pulmonar/etiologia , Material Particulado/efeitos adversos , Substâncias Protetoras/uso terapêutico , Quercetina/análogos & derivados , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Linhagem Celular , Humanos , Lesão Pulmonar/metabolismo , Lesão Pulmonar/patologia , Masculino , Camundongos Endogâmicos BALB C , Quercetina/uso terapêutico , Serina-Treonina Quinases TOR/metabolismo
11.
Cancer Cell Int ; 21(1): 116, 2021 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-33596893

RESUMO

BACKGROUND: Acquired resistance to therapeutic drugs has become an important issue in treating ovarian cancer. Studies have shown that the prevalent chemotherapy resistance (cisplatin, paclitaxel etc.) for ovarian cancer occurs partly because of decreased production of reactive oxygen species within the mitochondria of ovarian cancer cells. MAIN BODY: Nuclear erythroid-related factor-2 (Nrf2) mainly controls the regulation of transcription of genes through the Keap1-Nrf2-ARE signaling pathway and protects cells by fighting oxidative stress and defending against harmful substances. This protective effect is reflected in the promotion of tumor cell growth and their resistance to chemotherapy drugs. Therefore, inhibition of the Nrf2 pathway may reverse drug resistance. In this review, we describe the functions of Nrf2 in drug resistance based on Nrf2-associated signaling pathways determined in previous studies. CONCLUSIONS: Further studies on the relevant mechanisms of Nrf2 may help improve the outcomes of ovarian cancer therapy.

12.
Phytomedicine ; 82: 153465, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33486268

RESUMO

BACKGROUND: Ovarian cancer is one of the most common gynecological malignancies in the world. Daphnetin (Daph) was previously reported to possess antitumor potential, but its potential and molecular mechanisms in ovarian cancer remain poorly understood. PURPOSE: In the current study, we aimed to explore the antitumor effect and detailed mechanisms of Daph in ovarian cancer cells. METHODS: The cytotoxic effect of Daph on ovarian cells was determined in vitro and in vivo. Cell growth, proliferation, apoptosis and ROS generation were measured by CCK8 assays, colony formation assays and flow cytometry. Western blotting was used to evaluate the related signal proteins. Immunofluorescence and transmission electron microscopy were used to evaluate markers of autophagy and autophagic flux. The antitumor effects were observed in the A2780 xenograft model. Moreover, Daph-induced autophagy was observed by enhanced LC3-II accumulation and endogenous LC3 puncta, and an autophagy inhibitor further enhanced the antitumor efficacy of Daph, which indicated that the cytoprotective role of autophagy in ovarian cancer. RESULTS: We found that Daph exhibited antitumor effects by inducing ROS-dependent apoptosis in ovarian cancer, which could be reversed by N-acetyl cysteine (NAC). The AMPK/Akt/mTOR pathway was involved in Daph-mediated cytoprotective autophagy, and when Daph-mediated the expression level of AMPK and autophagy were blocked, there was robust inhibition of cell proliferation and induction of apoptosis. In addition, in the A2780 xenograft model, combined treatment with Daph and an autophagy inhibitor showed obvious synergetic effects on the inhibition of cell viability and promotion of apoptosis, without any side effects. CONCLUSION: Our results suggest that Daph triggers ROS-induced cell apoptosis and induces cytoprotective autophagy by modulating the AMPK/Akt/mTOR pathway. Moreover, the combination of Daph and autophagy inhibitor may be a potential therapeutic strategy for ovarian cancer.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Autofagia/efeitos dos fármacos , Neoplasias Ovarianas/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Umbeliferonas/farmacologia , Apoptose/efeitos dos fármacos , Morte Celular , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Front Pharmacol ; 11: 579178, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33363464

RESUMO

Cisplatin (CDDP) is a widely used drug for cancer treatment that exhibits major side effects in normal tissues, such as nephrotoxicity in kidneys. The Nrf2 signaling pathway, a regulator of mitochondrial dysfunction, oxidative stress and inflammation, is a potential therapeutic target in CDDP-induced nephrotoxicity. We explored the underlying mechanisms in wild-type (WT) and Nrf2-/- mice on CDDP-induced renal dysfunction in vivo. We found that Nrf2 deficiency aggravated CDDP-induced nephrotoxicity, and Daph treatment significantly ameliorated the renal injury characterized by biochemical markers in WT mice and reduced the CDDP-induced cell damage. In terms of the mechanism, Daph upregulated the SIRT1 and SIRT6 expression in vivo and in vitro. Furthermore, Daph inhibited the expression level of NOX4, whereas it activated Nrf2 translocation and antioxidant enzymes HO-1 and NQO1, and alleviated oxidative stress and mitochondrial dysfunction. Moreover, Daph suppressed CDDP-induced NF-κB and MAPK inflammation pathways, as well as p53 and cleaved caspase-3 apoptosis pathways. Notably, the protective effects of Daph in WT mice were completely abrogated in Nrf2-/- mice. Moreover, Daph enhanced, rather than attenuated, the tumoricidal effect of CDDP.

14.
Food Funct ; 11(5): 4471-4484, 2020 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-32377661

RESUMO

Early pulmonary fibrosis after acute lung injury leads to poor prognosis and high mortality. Pterostilbene (Pts), a bioactive component in blueberries, possesses anti-inflammatory, antioxidative and antifibrotic properties. However, the effects of Pts on lipopolysaccharide (LPS)-induced pulmonary fibrosis are still unknown. In our study, the Pts group showed lower lung injury and fibrosis scores, and lower levels of hydroxyproline and protein (collagen I and transforming growth factor-ß) than the scores and levels in mice treated with LPS. MMP-1 was the degrading enzyme of collagen I and LPS caused the inhibition of MMP-1, disturbing the degradation of collagen. Additionally, Pts remarkably reversed the LPS-induced inhibition of interleukin-10 and the release of tumor necrosis factor-α, interleukin-6 and interleukin-1ß. In terms of cellular pathways, Pts treatment ameliorated LPS-activated nuclear factor kappa B (NF-κB) and NOD-like receptor NLRP3 signaling. Besides, LPS-induced low levels of A20 could be activated by Pts. In addition, Pts treatment reversed the high levels of Caspase-3, poly ADP-ribose polymerase (PARP) and Bcl2-associated X protein (Bax) expression and the low levels of B cell lymphoma/lewkmia-2 (Bcl2) that had been induced by LPS. Moreover, oxidative stress is also involved in the pathogenesis of fibrosis. Our findings indicate that LPS injection triggered the production of myeloperoxidase (MPO) and malondialdehyde (MDA) and the depletion of superoxide dismutase (SOD) and glutathione (GSH), and that these effects were notably reversed by treatment with Pts. In addition, Pts induced the dissociation of Kelch-like epichlorohydrin-associated protein-1 (Keap-1) and NF-E2 related factor-2 (Nrf2) and the activation of downstream genes (heme oxygenase-1, NAD(P)H:quinine oxidoreductase, glutamate-cysteine ligase catalytic subunit and glutamate-cysteine ligase modifier). In conclusion, oxidative stress, apoptosis and inflammation are involved in early pulmonary fibrosis and Pts exerts a protective effect by activating Keap-1/Nrf2, inhibiting caspase-dependent A20/NF-κB and NLRP3 signaling pathways.


Assuntos
Antioxidantes/uso terapêutico , Mirtilos Azuis (Planta) , Estresse Oxidativo/efeitos dos fármacos , Fibrose Pulmonar/prevenção & controle , Estilbenos/uso terapêutico , Animais , Antioxidantes/administração & dosagem , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Inflamação , Lipopolissacarídeos , Camundongos , Camundongos Endogâmicos , Fitoterapia , Fibrose Pulmonar/induzido quimicamente , Coelhos , Distribuição Aleatória , Estilbenos/administração & dosagem , Estilbenos/farmacologia
15.
Phytomedicine ; 71: 153241, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32454347

RESUMO

BACKGROUND: Oxidative stress-triggered fatal hepatotoxicity is an essential pathogenic factor in acute liver failure (ALF). AIMS: To investigate the protective effect of daphnetin (Daph) on tert-butyl hydroperoxide (t-BHP) and acetaminophen (APAP)-induced hepatotoxicity through altering Nrf2/Trx-1 pathway activation. MATERIALS AND METHODS: In vivo, male C57BL/6 mice with Wild-type (WT) and Nrf2-/- were divided into five groups and acute liver injury model were established by APAP or LPS/GalN after injection with Daph (20, 40, or 80 mg/kg), seperately. Then, liver tissue and serum were collected for biochemical determination, TUNEL and H & E staining, and western blot analysis. In vitro, HepG2 cells were used to investigate the protective effect and mechanism of daphnetin against ROS and apoptosis induced by t-BHP via apoptosis detection, western blot, immunofluorescence analysis, and sgRNA transfection. RESULTS: Our results indicated that Daph efficiently inhibited t-BHP-stimulated hepatotoxicity, and modulated Trx-1 expression and Nrf2 activation which decreased Keap1-overexpression in HepG2 cells. Moreover, Daph inhibited t-BHP-excited hepatotoxicity and enhanced Trx-1 expression, which was reversed in Nrf2-/- HepG2 cells. In vivo, a survival rate analysis first suggested that Daph significantly reduced the lethality induced by APAP or GalN/LPS in a Nrf2-dependent or -independent manner by using Nrf2-/- mice, respectively. Next, further results implicated that Daph not only effectively alleviated APAP-induced an increase of ALT and AST levels, histopathological changes, ROS overproduction, malondialdehyde (MDA) formation and GSH/GSSG reduction, but it also relieved hepatic apoptosis by strengthening the suppression of cleaved-caspase-3 and expression of P53 protein. Additionally, Daph attenuated mitochondrial dysfunction by suppressing ASK1/JNK activation and decreasing apoptosis-inducing factor (AIF) and Cytochrome c release and Bax mitochondrial translocation. Daph inhibited inflammatory responses by inactivating the thioredoxin-interacting protein (Txnip)/NLRP3 inflammasome. Furthermore, Daph efficiently enhanced Nrf2 nuclear translocation and Trx-1 expression. However, these effects in WT mice were eliminated in Nrf2-/- mice. CONCLUSIONS: These investigations demonstrated that Daph treatment has protective potential against oxidative stress-driven hepatotoxicity by inhibition of ASK1/JNK and Txnip/NLRP3 activation, which may be strongly related to the Nrf2/Trx-1 upregulation.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Inflamassomos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Umbeliferonas/farmacologia , Acetaminofen/efeitos adversos , Animais , Proteínas de Transporte/metabolismo , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Células Hep G2 , Humanos , Inflamassomos/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , MAP Quinase Quinase 4/metabolismo , MAP Quinase Quinase Quinase 5/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/fisiologia , Substâncias Protetoras/farmacologia
16.
Front Pharmacol ; 11: 264, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32256355

RESUMO

Cisplatin (CDDP) is a widely used chemotherapeutic agent for various solid tumors, but its severe side effects, particularly nephrotoxicity, limit its clinical application. Isoorientin (Iso) is a flavonoid-like compound known to have antioxidant effects. As oxidative injury plays a vital role in CDDP-induced acute kidney injury (AKI), the effect of Iso on CDDP-induced nephrotoxicity has not yet been researched. We assessed the effects of Iso against CDDP-induced nephrotoxicity in vitro using mTEC cells and further explored the mechanisms underlying CDDP-induced renal dysfunction in vivo in WT and Nrf2-/- mice. The results showed that Iso treatment significantly reduced CDDP-induced nephrotoxicity via attenuating cell damage in vitro and via ameliorating renal injury, as determined by biochemical markers, in mice. The molecular mechanism underlying this protection was also investigated. Iso up-regulated the expression levels of SIRT1 and SIRT6 in vivo and in vitro. In addition, Iso activated Nrf2 translocation and the expression levels of its downstream antioxidant enzymes, such as HO-1 and NQO1, whereas it inhibited the expression level of NOX4, thus decreasing oxidative stress. Notably, the protective effects of Iso observed in WT mice were completely abolished in Nrf2-/- mice. Collectively, these data indicate that the protective effect of Iso on CDDP-induced nephrotoxicity by SIRT1- and SIRT6-mediated Nrf2 activation regulates oxidative stress, inflammation and apoptosis. The absence of Nrf2 exacerbates CDDP-induced renal damage, and the pharmacological activation of Nrf2 may represent a novel therapy to prevent kidney injury.

17.
Front Oncol ; 9: 769, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31475112

RESUMO

AMPK-mediated autophagy and Akt/mTOR pathways play important roles in current cancer treatments. Oridonin (Ori), an ent-kaurane diterpenoid isolated from Isodon rubescens, exerts extensive anti-tumor potential and controversial effects on autophagy. In this study, we investigated the effect of Ori on the autophagy, apoptosis, and AMPK/Akt/mTOR pathways and determined whether Ori was related to the increased cisplatin sensitivity observed in A549 cells. First, we found that Ori suppressed Akt/mTOR, Bcl2 and autophagy flux with enhanced levels of Atg3, P62, and LC3II, which was also shown as the accumulation of autophagosomes. AMPK and pro-apoptotic proteins (caspase3, Bax, and PARP) were activated in Ori-treated cells. With the pretreatment of compound c (AMPK inhibitor), the activation of autophagosomes, apoptosis and the inhibition of Akt/mTOR pathways induced by Ori were all reversed. The Ori-activated apoptosis-related markers mentioned previously and the cell-killing effect were restrained by 3-MA (inhibitor of autophagosomes) treatment. Therefore, we hypothesized that the Ori-induced pro-apoptotic effect was mediated by AMPK/Akt/mTOR-dependent accumulation of impaired autophagosomes. Furthermore, Ori could increase the sensitivity of cisplatin through its increased cell-killing, autophagy-suppressing and apoptosis-inducing activities. In addition to sensitizing cisplatin, Ori also alleviated cisplatin-induced acute renal injury in vivo, manifested as depleted BUN, CRE, kidney index, and weight loss compared to the cisplatin group. In summary, apart from its protective effect on cisplatin-induced nephrotoxicity, Ori enhanced cisplatin sensitivity via its pro-apoptotic activity mediated by AMPK/Akt/mTOR-dependent autophagosome activation, which may be a potential therapeutic target for non-small cell lung cancer.

18.
Cell Commun Signal ; 17(1): 62, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31186013

RESUMO

BACKGROUND: Oxidative stress and the resulting inflammation are essential pathological processes in acute lung injury (ALI). Nuclear factor erythroid 2-related factor 2 (Nrf2), a vital transcriptional factor, possesses antioxidative potential and has become a primary target to treat many diseases. Oridonin (Ori), isolated from the plant Rabdosia Rrubescens, is a natural substance that possesses antioxidative and anti-inflammatory effects. Our aim was to study whether the anti-inflammatory and antioxidant effects of Ori on LPS-induced ALI were mediated by Nrf2. METHODS: MTT assays, Western blotting analysis, a mouse model, and hematoxylin-eosin (H & E) staining were employed to explore the mechanisms by which Ori exerts a protective effect on LPS-induced lung injury in RAW264.7 cells and in a mouse model. RESULTS: Our results indicated that Ori increased the expression of Nrf2 and its downstream genes (HO-1, GCLM), which was mediated by the activation of Akt and MAPK. Additionally, Ori inhibited LPS-induced activation of the pro-inflammatory pathways NLRP3 inflammasome and NF-κB pathways. These two pathways were also proven to be Nrf2-independent by the use of a Nrf2 inhibitor. In keeping with these findings, Ori alleviated LPS-induced histopathological changes, the enhanced production of myeloperoxidase and malondialdehyde, and the depleted expression of GSH and superoxide dismutase in the lung tissue of mice. Furthermore, the expression of LPS-induced NLRP3 inflammasome and NF-κB pathways was more evident in Nrf2-deficient mice but could still be reversed by Ori. CONCLUSIONS: Our results demonstrated that Ori exerted protective effects on LPS-induced ALI via Nrf2-independent anti-inflammatory and Nrf2-dependent antioxidative activities.


Assuntos
Lesão Pulmonar Aguda/metabolismo , Lesão Pulmonar Aguda/prevenção & controle , Diterpenos do Tipo Caurano/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Lesão Pulmonar Aguda/patologia , Animais , Relação Dose-Resposta a Droga , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Inflamassomos/metabolismo , Lipopolissacarídeos/efeitos adversos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
19.
Chem Biol Interact ; 308: 269-278, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-31153982

RESUMO

Although cisplatin is an effective anticancer drug, its clinical application is limited due to various side effects, especially nephrotoxicity. In this study, we investigated the protective effects and possible mechanisms of hesperetin on cisplatin-induced kidney damage. In vitro, hesperetin significantly attenuated oxidative stress-induced apoptosis by reducing ROS levels in cisplatin-treated HK-2 cells. Simultaneously, hesperetin activated the Nrf2 signaling pathway and regulated its downstream genes, including NQO1 and HO-1. In vivo, hesperetin could significantly attenuate cisplatin-induced nephrotoxicity, blood urea nitrogen (BUN) and serum creatinine (SCr). Furthermore, hesperetin clarifies cisplatin-induced oxidative stress by reducing MDA/MPO levels and increasing SOD/GSH levels. In addition, from the histopathological analysis of the kidney, hesperetin significantly reduced the nephrotoxicity caused by cisplatin compared with the cisplatin group. Moreover, western blotting of renal tissue revealed that hesperetin activates Nrf2 in a dose-dependent manner, attenuates the MAPK signaling pathway against inflammation, and inhibits the expression of apoptotic proteins to protect kidneys from AKI caused by cisplatin. Altogether, these findings suggest that hesperetin may be a potential protectant against cisplatin-induced nephrotoxicity.


Assuntos
Injúria Renal Aguda/patologia , Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Hesperidina/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Injúria Renal Aguda/tratamento farmacológico , Injúria Renal Aguda/etiologia , Animais , Antioxidantes/metabolismo , Nitrogênio da Ureia Sanguínea , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/uso terapêutico , Creatinina/sangue , Hesperidina/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Rim/efeitos dos fármacos , Rim/patologia , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
20.
Int J Biol Sci ; 15(4): 788-799, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30906210

RESUMO

Farrerol has been shown to have antioxidative potential via Nrf2 activation, which in turn is involved in the prevention of hepatotoxicity. The purpose of the current study was to explore the protective effect of farrerol against acetaminophen-induced hepatotoxicity and its underlying mechanisms. Mice were used to evaluate the hepatoprotective effect of farrerol on liver injury induced by acetaminophen in vivo. HepG2 cells were utilized to further determine the functional role and mechanisms by which Nrf2 and autophagy are involved in the hepatoprotective effect of farrerol in vitro. We found that treatment with farrerol leads to a significant reduction in acetaminophen-induced hepatotoxicity by decreasing mortality, histopathological liver changes, and ALT and AST levels. Furthermore, farrerol effectively suppressed mitochondrial dysfunction by reducing JNK phosphorylation, Bax mitochondrial translocation, AIF and cytochrome c release. Further investigations revealed that the activation of Nrf2 and the induction of autophagy via the AMPK/AKT pathway by farrerol contributed to its hepatoprotective activity in vitro. In addition, farrerol inhibited acetaminophen-induced the mortality and histopathological changes in WT mice were evidently alleviated but not abrogated in Nrf2 -/- mice, which attributed to the induction of autophagy. Together, farrerol has protective potential against acetaminophen-induced hepatotoxicity which may be associated with activation of Nrf2 and autophagy.


Assuntos
Autofagia/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Doença Hepática Induzida por Substâncias e Drogas/metabolismo , Cromonas/uso terapêutico , Fator 2 Relacionado a NF-E2/metabolismo , Acetaminofen/farmacologia , Animais , Sobrevivência Celular/efeitos dos fármacos , Feminino , Células Hep G2 , Humanos , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA