Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Nat Med ; 78(3): 644-654, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38409483

RESUMO

Atherosclerosis is a cardiovascular disease, accounting for the most common mortality cause worldwide. Notoginsenoside R1 (NGR1) is a characteristic saponin of Radix notoginseng that exhibits anti-inflammatory and antioxidant effects while modulating lipid metabolism. Evidence suggests that NGR1 exerts cardioprotective, neuroprotective, and anti-atherosclerosis effects. However, underlying NGR1 mechanisms alleviating atherosclerosis (AS) have not been examined. This study used a network pharmacology approach to construct the drug-target-disease correlation and protein-protein interaction (PPI) network of NGR1 and AS. Moreover, functional annotation and pathway enrichment analyses deciphered the critical biological processes and signaling pathways potentially regulated by NGR1. The protective effect of NGR1 against AS and the underlying mechanism(s) was assessed in an atherogenic apolipoprotein E-deficient (ApoE-/-) mice in vivo and an oxidized low-density lipoprotein (ox-LDL)-induced macrophage model in vitro. The network pharmacology and molecular docking analyses revealed that NGR1 protects against AS by targeting the NLRP3/caspase-1/IL-1ß pathway. NGR1 reduced foam cell formation in ox-LDL-induced macrophages and decreased atherosclerotic lesion formation, serum lipid metabolism, and inflammatory cytokines in AS mice in vivo. Therefore, NGR1 downregulates the NLRP3 inflammasome complex gene expression of NLRP3, caspase-1, ASC, IL-1ß, and IL-18, in vivo and in vitro.


Assuntos
Aterosclerose , Ginsenosídeos , Inflamassomos , Macrófagos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Farmacologia em Rede , Animais , Ginsenosídeos/farmacologia , Ginsenosídeos/química , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Camundongos , Aterosclerose/tratamento farmacológico , Aterosclerose/metabolismo , Inflamassomos/metabolismo , Inflamassomos/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Masculino , Simulação de Acoplamento Molecular , Lipoproteínas LDL , Camundongos Endogâmicos C57BL , Modelos Animais de Doenças , Apolipoproteínas E/genética , Transdução de Sinais/efeitos dos fármacos
2.
Gene ; 883: 147653, 2023 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-37479096

RESUMO

In response to stress, cells can utilize several processes, such as the activation of the Nrf2/Keap1 pathway as a critical regulator of oxidative stress to protect against oxidative damage. C-Jun N-terminal kinase (JNK), a member of the mitogen-activated protein kinase (MAPK) family, is involved in regulating the NF-E2-related nuclear factor 2 (Nrf2)/antioxidant response element (ARE) signaling pathway. NAD(P)H quinone redox enzyme-1 (NQO1), a downstream target gene of the Nrf2 pathway, plays a vital role in removing peroxide and providing resistance to oxidative injury. We found that microcystins (MCs) stimulated CpNrf2 to express and increase anti-oxidative enzyme activities in a previous experiment. In our current study, the full-length cDNAs of JNK and NQO1 from Cristaria plicata (designated CpJNK and CpNQO1) were cloned. The relative levels of CpJNK and CpNQO1 were high in hepatopancreas. Upon MCs induction, the relative level of CpNQO1 was increased, whereas that of CpJNK was decreased significantly. In contrast, CpNrf2 knockdown upregulated the expression of CpJNK mRNA and phosphorylation of CpJNK protein (Cpp-JNK), but inhibited CpNQO1 expression. Additionally, we found that JNK inhibitor SP600125 stimulated expression of CpNQO1 and CpNrf2 upon exposure to MCs, and we further confirmed that CpNrf2 protein combined with the ARE element in CpNQO1 gene promoter in vitro, and increased CpNQO1-ARE-luciferase activity in a CpNrf2-dependent manner. These findings indicated C. plicata effectively alleviated MC-induced oxidative injury through JNK participated in regulating the Nrf2/NQO1-ARE pathway.


Assuntos
Elementos de Resposta Antioxidante , Unionidae , Animais , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Microcistinas/toxicidade , Microcistinas/genética , Estresse Oxidativo , Proteínas Quinases Ativadas por Mitógeno/genética , Unionidae/genética
3.
Curr Comput Aided Drug Des ; 19(6): 451-464, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36740793

RESUMO

BACKGROUND: Buyang Huanwu Decoction (BHD) is used to regulate blood circulation and clear collaterals and is widely used in coronary heart disease. However, the active compounds and the mechanism of BHD used to treat restenosis are less understood. OBJECTIVE: The study aimed to explore the potential mechanism of Buyang Huanwu decoction BHD for the treatment of restenosis using network pharmacology and molecular docking experiments. METHODS: The bioactive components of BHD and their corresponding targets were retrieved from the Traditional Chinese Medicine Systems Pharmacology (TCMSP) and Encyclopaedia of Traditional Chinese Medicine (ETCM) databases as well as literature. Restenosisassociated therapeutic genes were identified from the OMIM, Drugbank, GEO, and Dis- GeNET databases. Genes related to the vascular smooth muscle cell (VSMC) phenotype were obtained from the gene ontology (GO) database and literature. The core target genes for the drug-disease-VSMC phenotype were identified using the Venn tool and Cytoscape software. Moreover, the "drug-component-target-pathway" network was constructed and analyzed, and pathway enrichment analysis was performed. The connection between the main active components and core targets was analyzed using the AutoDock tool, and PyMOL was used to visualize the results. RESULTS: The "compound-target-disease" network included 80 active ingredients and 599 overlapping targets. Among the bioactive components, quercetin, ligustrazine, ligustilide, hydroxysafflor yellow A, and dihydrocapsaicin had high degree values, and the core targets included TP53, MYC, APP, UBC, JUN, EP300, TGFB1, UBB, SP1, MAPK1, SMAD2, CTNNB1, FOXO3, PIN1, EGR1, TCF4, FOS, SMAD3, and CREBBP. A total of 365 items were obtained from the GO functional enrichment analysis (p < 0.05), whereas the enrichment analysis of the KEGG pathway identified 30 signaling pathways (p < 0.05), which involved the TGF-ß signaling pathway, Wnt signaling pathway, TRAF6-mediated induction of NF-κB and MAPK pathway, TLR7/8 cascade, and others. The molecular docking results revealed quercetin, luteolin, and ligustilide to have good affinity with the core targets MYC and TP53. CONCLUSION: The active ingredients in BHD might act on TP53, MYC, APP, UBC, JUN, and other targets through its active components (such as quercetin, ligustrazine, ligustilide, hydroxysafflor yellow A, and dihydrocapsaicin). This action of BHD may be transmitted via the involvement of multiple signaling pathways, including the TGF-ß signaling pathway, Wnt signaling pathway, TRAF6-mediated induction of NF-κB and MAPK pathway, and TLR7/8 cascade, to treat restenosis by inhibiting the phenotype switching and proliferation of VSMC.


Assuntos
NF-kappa B , Farmacologia em Rede , Simulação de Acoplamento Molecular , Músculo Liso Vascular , Quercetina/farmacologia , Fator 6 Associado a Receptor de TNF , Receptor 7 Toll-Like , Proliferação de Células , Fator de Crescimento Transformador beta
4.
Dev Comp Immunol ; 141: 104629, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36587710

RESUMO

Microcystins (MC) are one of the most abundant and widely distributed cyanotoxins in aquatic systems. MC inhibits the functions of protein phosphatase 1 and 2A (PP1/2A), which can seriously affect ecosystem integrity. The NF-E2-related nuclear factor 2 (Nrf2)/Kelch-like epichlorohydrin-related protein-1 (Keap1) signaling pathway protects against oxidative damage by activating phase II detoxification/antioxidant enzymes. Our previous study revealed that MC upregulates the expression and enhances the activities of the antioxidant enzymes by stimulating the CpNrf2 signaling pathway. In the current study, to further clarify the regulatory role of Keap1 in response to MC-induced oxidative stress in shellfish, we cloned the full-length cDNA of Keap1a and Keap1b from Cristaria plicata (designated CpKeap1a and CpKeap1b), which are 2952 and 3710 bp peptides, respectively. The amino acid sequence of CpKeap1a and CpKeap1b contained Tram-track and Bric-a-brac (BTB), Intervening region (IVR), and Double glycine repeat (DGR) domain. Additionally, CpKeap1a contained two cysteine residues analogous to Cys-273 and -288 in zebrafish, but CpKeap1b did not. Moreover, CpKeap1a and -1b formed a homodimer and heterodimer, respectively, and also formed a heterodimer with CpNrf2. In the hepatopancreas, the expression levels of CpKeap1a and -1b were the highest, but MC treatment down-regulated the expression of these proteins. Moreover, the transcription of antioxidant enzymes with antioxidant response element (ARE-driven enzymes), including CpMnSOD, CpCu/ZnSOD, CpTRX, CpPrx, CpSe-GPx, and Cpsigma-GST was upregulated by CpNrf2 in the hepatopancreas. Compared with the MC-induced group, CpKeap1a-siRNA1117 injection significantly increased the transcription of mRNAs for ARE-driven enzymes and Nrf2. CpKeap1a-siRNA1117 also enhanced the activities of antioxidation enzymes. These findings demonstrated that Keap1a negatively regulated the expression of Nrf2 protein and MC-induced oxidative stress response in C. plicata. Therefore, we speculated that CpKeap1a promoted CpNrf2 by recognizing and binding MC. These events then protected molluscs from MC-induced oxidative damage.


Assuntos
Bivalves , Unionidae , Animais , Antioxidantes/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Epicloroidrina , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Peixe-Zebra/metabolismo , Microcistinas/metabolismo , Ecossistema , Bivalves/genética , Unionidae/genética , Estresse Oxidativo , Proteínas de Transporte/metabolismo , Proteínas de Peixe-Zebra/genética
5.
Aquat Toxicol ; 255: 106398, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36669434

RESUMO

Microcystins (MCs) are the most frequent and widely distributed type of cyanotoxin in aquatic systems, and they cause an imbalance of the body's oxidative system. In a previous experiment, we demonstrated that the mollusk Cristaria plicata can protect against MC-induced oxidative damage through the nuclear factor erythroid 2-related factor 2(Nrf2)/Kelch-like epichlorohydrin-related protein-1 (Keap1) pathway. Here, we evaluated whether selective autophagy affects the Nrf2/Keap1a anti-oxidative stress pathway in C. plicata. Full-length cDNA sequences of p62/SQSTM1 from C. plicata (Cpp62) were divided into 2484 bp fragments. From N-terminal to C-terminal, the amino acid sequence of Cpp62 contained PB1 (Phox and Bem1p domain), ZNF (zinc finger domain) chain, LIR (LC3 interacting region) and UBA (ubiquitin-associated domain) domains, but not the KIR (Keap1 interacting region) domain. We confirmed that Cpp62 did not bind to CpKeap1a in vitro, and the relative level of Cpp62 was the highest in the hepatopancreas. Moreover, MCs significantly upregulated the mRNA and protein levels of Cpp62 in the hepatopancreas after CpKeap1a knockdown, whereas Nrf2 upregulated the transcription levels of Cpp62, suggesting that MCs increased Cpp62 expression via the Nrf2/Keap1a signaling pathway. Moreover, Cpp62 and CpNrf2 proteins have a strong affinity for the NQO1 promoter, but MCs inhibited the ability of CpNrf2 and Cpp62 to upregulate luciferase activity. The results show that Nrf2 and the p62 protein induced p62 expression by binding to ARE (antioxidant response element) sequences in the p62 promoter of C. plicata, thereby promoting p62 to resist MC-induced oxidative stress. Therefore, we speculate that MCs induce p62-dependent autophagy in C. plicata, resulting in the inhibition of Nrf2 transcription and Cpp62 promoter activity. These findings help to reveal the mechanism by which the p62-Nrf2/Keap1 pathway mitigates MC-induced oxidative damage in mussels.


Assuntos
Unionidae , Poluentes Químicos da Água , Animais , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/química , Proteína Sequestossoma-1/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Microcistinas/toxicidade , Microcistinas/metabolismo , Poluentes Químicos da Água/toxicidade , Transdução de Sinais , Estresse Oxidativo
6.
Can J Anaesth ; 70(1): 106-115, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36109453

RESUMO

PURPOSE: Recently, more attention has been given to the costoclavicular space (CCS) as an alternative pathway for ultrasound-guided brachial plexus block (BPB). While 0.5% ropivacaine was used in most related studies, research has shown effective ultrasound-guided supraclavicular BPB using lower local anesthetic concentrations, and our preliminary data have indicated that 0.375% ropivacaine may be effective when given in the CCS. Hence, we hypothesized that the efficacy of 0.375% ropivacaine would be noninferior compared with 0.5% in ultrasound-guided BPB via the CCS. METHODS: We conducted a randomized, double-blind, single-centre, noninferiority clinical trial. Seventy patients undergoing elective forearm or hand surgery were randomly assigned to receive either 20 mL of 0.375% ropivacaine (experimental group) or 0.5% ropivacaine (control group) in the CCS for BPB. We assessed sensory and motor blockade at five, ten, 15, 20, 25, and 30 min after the injection. The primary outcome was the rate of successful BPB. Secondary outcomes included onset time, duration of sensory and motor blockade, and adverse reactions. The depth from the skin to the CCS was also recorded during the procedure. RESULTS: A total of 69 patients were evaluable for block success. There was one failed block in both groups, yielding a BPB block success rate of 97% in both groups. 0.375% Ropivacaine was noninferior to 0.5% ropivacaine (P = 0.98). There was no significant difference in the median [interquartile range (IQR)] onset time of sensory-motor blockade in the experimental group (15 [15-20] min; N = 34) compared with the control group (15 [13-20] min; N = 33; Mann-Whitney test, P = 0.48). The median [IQR] duration of sensory blockade was significantly shorter in the experimental group (455 [398-490] min vs 610 [570-655] min in the control group; Hodges-Lehmann estimator of the difference, 165 min; 95.08% confidence interval (CI), 130 to 195; P < 0.001). Likewise, the median [IQR] duration of motor blockade was significantly shorter in the experimental group (470 [409-500] min vs 625 [578-665] min in the control group; Hodges-Lehmann estimator of the difference, 165 min; 95.08% CI, 130 to 195; P < 0.001). There were no adverse reactions directly related to the technique or the ropivacaine injection in either group. CONCLUSIONS: 0.375% Ropivacainewas noninferior to 0.5% ropivacaine with regard to rate of successful ultrasound-guided costoclavicular BPB. STUDY REGISTRATION: chictr.org.cn (ChiCTR20000306570); registered 8 March 2020.


RéSUMé: OBJECTIF: L'espace costo-claviculaire (ECC) a récemment bénéficié d'un regain d'intérêt comme voie de substitution pour le bloc du plexus brachial (BPB) échoguidé. La ropivacaïne 0,5 % a été utilisée dans la majorité des études sur ce sujet, mais la recherche a montré un BPB supra-claviculaire échoguidé efficace en utilisant de plus faibles concentrations d'anesthésique local et nos données préliminaires ont indiqué que la ropivacaïne à 0,375 % pouvait être efficace en administration dans l'ECC. En conséquence, nous avons émis l'hypothèse selon laquelle l'efficacité de la ropivacaïne 0,375 % serait non inférieure à la ropivacaïne 0,5 % dans le BPB échoguidé via l'ECC. MéTHODES: Nous avons mené un essai clinique monocentrique de non-infériorité, randomisée en double insu. Soixante-dix patients subissant une chirurgie élective de l'avant-bras ou de la main ont été randomisés dans un groupe recevant 20 mL de ropivacaïne 0,375 % (groupe expérimental) ou de ropivacaïne 0,5 % (groupe contrôle) dans l'ECC pour un BPB. Nous avons évalué les blocs sensoriel et moteur à 5, 10, 15, 20, 25 et 30 minutes après l'injection. Le critère d'évaluation principal était le taux de succès du BPB. Les critères d'évaluation secondaires étaient, notamment, le délai d'action, la durée des blocs sensoriel et moteur, et les événements indésirables. La profondeur de la peau à l'ECC a aussi été consignée pendant la procédure. RéSULTATS: Un total de 69 patients était évaluable pour le succès du bloc. Il y a eu un échec du bloc dans chacun des deux groupes, ramenant le taux de succès du BPB à 97 % dans les deux groupes. La ropivacaïne 0,375 % a été non inférieure à la ropivacaïne 0,5 % (P = 0,98). Il n'y a pas eu de différence significative concernant le délai d'action médian (plage interquartile [PIQ]) du bloc sensori-moteur dans le groupe expérimental (15 [15 à 20] minutes; n = 34) comparativement au groupe contrôle (15 [13 à 20] minutes; n = 33; test de Mann­Whitney, P = 0,48). La durée médiane [PIQ] du bloc sensitif a été significativement plus courte dans le groupe expérimental (455 [398 à 490] minutes contre 610 [570 à 655] minutes dans le groupe contrôle; estimateur de la différence de Hodges­Lehmann, 165 minutes; intervalle de confiance [IC] à 95,08 % : 130 à 195; P < 0,001). De même, la durée médiane [PIQ] du bloc moteur a été significativement plus courte dans le groupe expérimental (470 [409 à 500] minutes contre 625 [578 à 665] minutes dans le groupe contrôle; estimateur de la différence de Hodges­Lehmann, 165 minutes; IC à 95,08 %, 130 à 195; P < 0,001). Il n'y a pas eu d'événement indésirable directement lié à la technique ou à l'injection de ropivacaïne dans l'un ou l'autre groupe. CONCLUSIONS: La ropivacaïne 0,375 % a été non inférieure à la ropivacaïne 0,5 % en ce qui concerne le taux de succès du BPB costo-claviculaire échoguidé. ENREGISTREMENT DE L'éTUDE: chictr.org.cn (ChiCTR20000306570); Enregistrée le 8 mars 2020.


Assuntos
Bloqueio do Plexo Braquial , Humanos , Bloqueio do Plexo Braquial/métodos , Ropivacaina , Anestésicos Locais/efeitos adversos , Extremidade Superior , Ultrassonografia
7.
Front Chem ; 10: 1042038, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36300028

RESUMO

ZnO quantum dots (QDs) have received much attention as biomarkers and drug delivery systems in cancer treatment, due to their low cost, ease of preparation, and pH-responsive degradation. However, its applications are limited by the low quantum yield and light absorption. In this work, a lanthanum-doped zinc oxide (La-ZnO) QDs-based drug delivery platform was constructed. The results show that 4% La doping is the most beneficial for improving the fluorescent properties of the ZnO QDs. After loading the drug, the cell activity was 15% at ZnO@DOX and 12% at La-ZnO@DOX. According to in vitro and in vivo experiment results, the La-ZnO QDs show enhancement of the antitumor effect. Dual enhancement of fluorescence and anti-tumor effects make La-ZnO QDs promising as a drug delivery system in cancer treatment.

8.
Int Immunopharmacol ; 101(Pt B): 108331, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34810122

RESUMO

BACKGROUND: Remimazolam is a novel ester-type benzodiazepine with ultrafast onset of sedation effect and fast recovery of consciousness. It has potential advantages in the sedation of sepsis-associated acute liver injury (SALI) patients. However, the effect and mechanism of remimazo lam on inflammation in the liver have not yet been elucidated. This study investigated the anti-inflammatory effects and mechanisms of remimazolam on SALI both in vivo and in vitro. METHODS: Lipopolysaccharide (LPS) plus galactosamine treated rat model and LPS-challenged RAW264.7 cells model were constructed to simulate SALI. Next, the models were used to explore the efficacy of remimazolam treatment on SALI. Benzodiazepine receptor inhibitor, PK11195, was also employed. Hepatic injury was assessed by quantifying levels of transaminases, examining liver pathology, and calculating the number of inflammatory cells in the liver. Inflammatory response was evaluated by determining levels of pro-inflammatory cytokines and chemokines in blood, as well as p38 phosphorylation (p-p38) in the liver. RESULTS: SALIrat models showed significant liver damage as manifested by increased levels of transaminases, proinflammatory cytokines, chemokines, and p-38. Remimazolam treatment reduced the liver injury and pathological changes, suppressed pro-inflammatory reactions, and elevated p-p38. The protective effect of remimazolam on liver injury was significantly blocked by PK11195. In LPS-stimulated RAW264.7 cells, it was found that treatment with remimazolam reduced the inflammatory response in LPS-treated cells in a time-dependent manner and decreased the level of p-p38. These results suggest that PK11195 can block remimazolam-induced inhibition of proinflammatory cytokine release and p-38 phosphorylation. CONCLUSIONS: This study shows that remimazolam can attenuate inflammatory response in SALI, which may be associated with activation of peripheral benzodiazepine receptors and inhibition of p38 phosphorylation in macrophages.


Assuntos
Macrófagos/efeitos dos fármacos , Animais , Benzodiazepinas/farmacologia , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocinas/metabolismo , Galactosamina/farmacologia , Inflamação/patologia , Lipopolissacarídeos/farmacologia , Fígado/patologia , Masculino , Camundongos , NF-kappa B/metabolismo , Células RAW 264.7 , Ratos , Receptores de GABA-A , Sepse/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
9.
Front Immunol ; 12: 625627, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33790896

RESUMO

Melatonin reportedly alleviates sepsis-induced multi-organ injury by inducing autophagy and activating class III deacetylase Sirtuin family members (SIRT1-7). However, whether melatonin attenuates small-intestine injury along with the precise underlying mechanism remain to be elucidated. To investigate this, we employed cecal ligation and puncture (CLP)- or endotoxemia-induced sepsis mouse models and confirmed that melatonin treatment significantly prolonged the survival time of mice and ameliorated multiple-organ injury (lung/liver/kidney/small intestine) following sepsis. Melatonin partially protected the intestinal barrier function and restored SIRT1 and SIRT3 activity/protein expression in the small intestine. Mechanistically, melatonin treatment enhanced NF-κB deacetylation and subsequently reduced the inflammatory response and decreased the TNF-α, IL-6, and IL-10 serum levels; these effects were abolished by SIRT1 inhibition with the selective blocker, Ex527. Correspondingly, melatonin treatment triggered SOD2 deacetylation and increased SOD2 activity and subsequently reduced oxidative stress; this amelioration of oxidative stress by melatonin was blocked by the SIRT3-selective inhibitor, 3-TYP, and was independent of SIRT1. We confirmed this mechanistic effect in a CLP-induced sepsis model of intestinal SIRT3 conditional-knockout mice, and found that melatonin preserved mitochondrial function and induced autophagy of small-intestine epithelial cells; these effects were dependent on SIRT3 activation. This study has shown, to the best of our knowledge, for the first time that melatonin alleviates sepsis-induced small-intestine injury, at least partially, by upregulating SIRT3-mediated oxidative-stress inhibition, mitochondrial-function protection, and autophagy induction.


Assuntos
Antioxidantes/farmacologia , Autofagia/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Intestino Delgado/efeitos dos fármacos , Melatonina/farmacologia , Mitocôndrias/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Sepse/tratamento farmacológico , Sirtuína 3/metabolismo , Animais , Modelos Animais de Doenças , Mediadores da Inflamação/metabolismo , Mucosa Intestinal/enzimologia , Mucosa Intestinal/patologia , Intestino Delgado/enzimologia , Intestino Delgado/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Sepse/enzimologia , Sepse/microbiologia , Sepse/patologia , Sirtuína 1/metabolismo , Sirtuína 3/genética
10.
Dalton Trans ; 50(16): 5607-5616, 2021 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-33908929

RESUMO

New effective antimicrobial agents with novel modes of action are urgently needed due to the continued emergence of drug-resistant bacteria. Here, three ruthenium complexes functionalized with benzothiophene: [Ru(phen)2(BTPIP)](ClO4)2 (Ru(II)-1), [Ru(dmp)2(BTPIP)](ClO4)2 (Ru(II)-2) and [Ru(dmb)2(BTPIP)](ClO4)2 (Ru(II)-3) (dmb = 4,4'-dimethyl-2,2'-bipyridine, phen = 1,10-phenanthroline, dmp = 2,9-dimethyl-1,10-phenanthroline) have been synthesized and their antimicrobial activities in vitro were assessed. Minimum inhibitory concentration (MIC) assays indicated that the three Ru(II)-1, Ru(II)-2 and Ru(II)-3 complexes all showed antibacterial activity against Staphylococcus aureus and Pseudomonas aeruginosa. The most active Ru(II)-3 complex was further tested against biofilms. Furthermore, it was also tested whether complex Ru(II)-3 could serve as an antibacterial adjuvant. Interestingly, the checkerboard data showed that Ru(II)-3 selectively exhibited synergism with aminoglycoside antibiotics. More importantly, the observed synergetic effect might be attributed to the inhibition of the regulatory function of SaCcpA. Finally, in vivo bacterial infection treatment studies through a murine skin infection model and skin irritation test were also conducted. All in all, these results confirmed that ruthenium complexes functionalized with benzothiophene have good antimicrobial activity against Staphylococcus aureus.


Assuntos
Antibacterianos/química , Antibacterianos/farmacologia , Complexos de Coordenação/química , Complexos de Coordenação/farmacologia , Rutênio/química , Staphylococcus aureus/efeitos dos fármacos , Tiofenos/química
11.
Oxid Med Cell Longev ; 2021: 6669075, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33688392

RESUMO

Mitochondria maintain mitochondrial homeostasis through continuous fusion and fission, that is, mitochondrial dynamics, which is precisely mediated by mitochondrial fission and fusion proteins, including dynamin-related protein 1 (Drp1), mitofusin 1 and 2 (Mfn1/2), and optic atrophy 1 (OPA1). When the mitochondrial fission and fusion of cardiomyocytes are out of balance, they will cause their own morphology and function disorders, which damage the structure and function of the heart, are involved in the occurrence and progression of cardiovascular disease such as ischemia-reperfusion injury (IRI), septic cardiomyopathy, and diabetic cardiomyopathy. In this paper, we focus on the latest findings regarding the molecular features and regulatory mechanisms of mitochondrial dynamic disorder in cardiovascular pathologies. Finally, we will address how these findings can be applied to improve the treatment of cardiovascular disease.


Assuntos
Doenças Cardiovasculares/patologia , Doenças Mitocondriais/patologia , Dinâmica Mitocondrial , Animais , Humanos , Modelos Biológicos
12.
Cell Death Dis ; 12(2): 217, 2021 02 26.
Artigo em Inglês | MEDLINE | ID: mdl-33637691

RESUMO

Our previous studies showed that silent mating-type information regulation 2 homologue-1 (SIRT1, a deacetylase) upregulation could attenuate sepsis-induced acute kidney injury (SAKI). Upregulated SIRT1 can deacetylate certain autophagy-related proteins (Beclin1, Atg5, Atg7 and LC3) in vitro. However, it remains unclear whether the beneficial effect of SIRT1 is related to autophagy induction and the underlying mechanism of this effect is also unknown. In the present study, caecal ligation and puncture (CLP)-induced mice, and an LPS-challenged HK-2 cell line were established to mimic a SAKI animal model and a SAKI cell model, respectively. Our results demonstrated that SIRT1 activation promoted autophagy and attenuated SAKI. SIRT1 deacetylated only Beclin1 but not the other autophagy-related proteins in SAKI. SIRT1-induced autophagy and its protective effect against SAKI were mediated by the deacetylation of Beclin1 at K430 and K437. Moreover, two SIRT1 activators, resveratrol and polydatin, attenuated SAKI in CLP-induced septic mice. Our study was the first to demonstrate the important role of SIRT1-induced Beclin1 deacetylation in autophagy and its protective effect against SAKI. These findings suggest that pharmacologic induction of autophagy via SIRT1-mediated Beclin1 deacetylation may be a promising therapeutic approach for future SAKI treatment.


Assuntos
Injúria Renal Aguda/enzimologia , Autofagia , Proteína Beclina-1/metabolismo , Túbulos Renais Proximais/enzimologia , Sepse/complicações , Sirtuína 1/metabolismo , Acetilação , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Injúria Renal Aguda/prevenção & controle , Animais , Autofagia/efeitos dos fármacos , Linhagem Celular , Modelos Animais de Doenças , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Glucosídeos/farmacologia , Humanos , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/ultraestrutura , Masculino , Camundongos Endogâmicos C57BL , Resveratrol/farmacologia , Sepse/microbiologia , Transdução de Sinais , Sirtuína 1/genética , Estilbenos/farmacologia , Fatores de Tempo
13.
Biomed Pharmacother ; 132: 110862, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33069969

RESUMO

Ziyuglycoside II (ZGS II) is a major bioactive ingredient of Sanguisorbae officinalis L., which has been widely used for managing myelosuppression or leukopenia induced by chemotherapy or radiotherapy. In the current study, we investigated the pro-hematopoietic effects and underlying mechanisms of ZGS II in cyclophosphamide-induced leukopenia in mice. The results showed that ZGS II significantly increased the number of total white blood cells and neutrophils in the peripheral blood. Flow cytometry analysis also showed a significant increase in the number of nucleated cells and hematopoietic stem and progenitor cells (HSPCs) including ST-HSCs, MPPs, and GMPs, and enhanced HSPC proliferation in ZGS II treated mice. The RNA-sequencing analysis demonstrated that ZGS II effectively regulated cell differentiation, immune system processes, and hematopoietic system-related pathways related to extracellular matrix (ECM)-receptor interaction, focal adhesion, hematopoietic cell lineage, cytokine-cytokine receptor interaction, the NOD-like receptor signaling pathway, and the osteoclast differentiation pathway. Moreover, ZGS II treatment altered the differentially expressed genes (DEGs) with known functions in HSPC differentiation and mobilization (Cxcl12, Col1a2, and Sparc) and the surface markers of neutrophilic precursors or neutrophils (Ngp and CD177). Collectively, these data suggest that ZGS II protected against chemotherapy-induced leukopenia by regulating HSPC proliferation and differentiation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Ciclofosfamida , Células-Tronco Hematopoéticas/efeitos dos fármacos , Leucopenia/prevenção & controle , Leucopoese/efeitos dos fármacos , Saponinas/farmacologia , Animais , Citoproteção , Modelos Animais de Doenças , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/patologia , Leucopenia/induzido quimicamente , Leucopenia/metabolismo , Leucopenia/patologia , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais
14.
Psychiatry Res ; 293: 113470, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32992097

RESUMO

Olanzapine is an atypical antipsychotic drug that has been increasingly used for treatment in schizophrenia. It has been observed that olanzapine responses in schizophrenia patients vary individually, but the reason has not been elucidated. In the study, we aimed to comprehensively explore the relationships between olanzapine responses and genetic polymorphisms of drug metabolizing enzymes, transporters and target receptors, and so as to interpret the reason of good and poor responses of olanzapine. A total of 241 Chinese Han paranoid schizophrenia who treated with olanzapine alone for 4 weeks were recruited. The positive and negative symptom scale (PANSS) was used to evaluate the efficacy of olanzapine. The genetic polymorphisms were detected by improved multiple ligase detection reaction (iMLDR). Multivariate logistic regression analysis suggested that the genetic polymorphisms of CYP1A2 rs762551, UGT1A4 rs2011425, ABCB1 rs1045642, DRD2 rs1799732 and rs1799978, 5-HTR2A rs6311 were significantly associated with olanzapine response. Multifactor dimensionality reduction (MDR) analysis showed that there was a negative interaction between CYP1A2 rs762551, ABCB1 rs1045642, DRD2 rs1799978, 5-HTR2A rs6311 and the interaction model was the optimal model. Our findings could partially explain the different olanzapine outcome and provided evidence for clarifying the predictive indicators of olanzapine response in further.


Assuntos
Antipsicóticos/uso terapêutico , Povo Asiático/genética , Citocromo P-450 CYP1A2/genética , Olanzapina/uso terapêutico , Esquizofrenia Paranoide/tratamento farmacológico , Adulto , Feminino , Genótipo , Humanos , Masculino , Proteínas de Membrana Transportadoras/genética , Pessoa de Meia-Idade , Polimorfismo Genético , Esquizofrenia Paranoide/etnologia , Esquizofrenia Paranoide/genética , Resultado do Tratamento
15.
Nat Commun ; 11(1): 281, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31941912

RESUMO

Yersinia pestis is transmitted from fleas to rodents when the bacterium develops an extensive biofilm in the foregut of a flea, starving it into a feeding frenzy, or, alternatively, during a brief period directly after feeding on a bacteremic host. These two transmission modes are in a trade-off regulated by the amount of biofilm produced by the bacterium. Here by investigating 446 global isolated Y. pestis genomes, including 78 newly sequenced isolates sampled over 40 years from a plague focus in China, we provide evidence for strong selection pressures on the RNA polymerase ω-subunit encoding gene rpoZ. We demonstrate that rpoZ variants have an increased rate of biofilm production in vitro, and that they evolve in the ecosystem during colder and drier periods. Our results support the notion that the bacterium is constantly adapting-through extended phenotype changes in the fleas-in response to climate-driven changes in the niche.


Assuntos
Proteínas de Bactérias/genética , Peste/microbiologia , Sifonápteros/microbiologia , Yersinia pestis/fisiologia , Animais , Biofilmes , Evolução Biológica , China , Clima , RNA Polimerases Dirigidas por DNA/genética , Reservatórios de Doenças , Ecossistema , Infestações por Pulgas , Variação Genética , Genoma Bacteriano , Interações Hospedeiro-Parasita , Interações Hospedeiro-Patógeno , Marmota/parasitologia , Fenótipo , Filogenia , Sciuridae/parasitologia , Seleção Genética , Sifonápteros/fisiologia , Yersinia pestis/genética
16.
Am J Physiol Renal Physiol ; 316(1): F20-F31, 2019 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-30379096

RESUMO

Sepsis is the leading cause of death in the intensive care unit and continues to lack effective treatment. It is widely accepted that high-mobility group box 1 (HMGB1) is a key inflammatory mediator in the pathogenesis of sepsis. Moreover, some studies indicate that the functions of HMGB1 depend on its molecular localization and posttranslational modifications. Our previous study confirms that sirtuin 1, silent information regulator 2-related enzyme 1 (SIRT1), a type III deacetylase, can ameliorate sepsis-associated acute kidney injury (SA-AKI). We explored the effect and mechanism of SIRT1 on HMGB1 using a mouse model of cecal ligation and puncture-induced sepsis and LPS-treated human kidney (HK-2) cell line. We found that HMGB1 is elevated in the serum but is gradually reduced in kidney cells in the later stages of septic mice. The acetylation modification of HMGB1 is a key process before its nucleus-to-cytoplasm translocation and extracellular secretion in kidney cells, accelerating the development of SA-AKI. Moreover, SIRT1 can physically interact with HMGB1 at the deacetylated lysine sites K28, K29, and K30, subsequently suppressing downstream inflammatory signaling. Thus the SIRT1-HMGB1 signaling pathway is a crucial mechanism in the development of SA-AKI and presents a novel experimental perspective for future SA-AKI research.


Assuntos
Injúria Renal Aguda/prevenção & controle , Proteína HMGB1/metabolismo , Rim/enzimologia , Sepse/complicações , Sirtuína 1/metabolismo , Acetilação , Injúria Renal Aguda/enzimologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Humanos , Rim/patologia , Camundongos Endogâmicos C57BL , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Processamento de Proteína Pós-Traducional , Sepse/enzimologia , Fatores de Tempo
17.
J Surg Res ; 233: 221-230, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30502252

RESUMO

BACKGROUND: The deactivation of SIRT3, a novel deacetylase located in mitochondria, can aggravate multiple organ dysfunction. However, the role of SIRT3 and its downstream targets in ischemia/reperfusion (I/R)-induced acute kidney injury (AKI) remain unknown. MATERIALS AND METHODS: I/R was reproduced in a rat model using a clamp placed on the left and right renal pedicles for 40 min. The rats were intraperitoneally injected with either the vehicle or a selective SIRT3 inhibitor (3-TYP) and scarified at different time points (4, 8, and 24 h after I/R). A portion of the renal tissue was extracted for histological analysis, and another portion was collected for the isolation of renal tubular epithelial cells for Western blotting, SOD2 and SIRT3 activity, cell apoptosis, and the determination of oxidative stress. RESULTS: The I/R-induced AKI model was successfully reproduced and SIRT3 activity was considerably reduced than control (sham operated) group, accompanied by increased acetylation of SOD2 and p53, as well as their elevated physical interaction in extracted mitochondrial protein (all P values < 0.05). Moreover, SIRT3 suppression by 3-TYP treatment (comparing with the vehicle treatment group) aggravated AKI, as evidenced by increased indicators of oxidative stress (increased mitochondrial red fluorescence MitoSOX and decreased reduced glutathione/oxidized glutathione ratio, all P values < 0.01). CONCLUSIONS: The elevation of SOD2 and p53 protein acetylation in the mitochondria of renal tubular epithelial cells is an important signaling event in the pathogenesis of I/R-induced AKI. Thus, deacetylase SIRT3 may be an upstream regulator of both SOD2 and p53, and the SIRT3 deactivation may aggravate AKI.


Assuntos
Injúria Renal Aguda/patologia , Túbulos Renais/patologia , Sirtuínas/metabolismo , Superóxido Dismutase/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Acetilação/efeitos dos fármacos , Injúria Renal Aguda/etiologia , Animais , Apoptose/efeitos dos fármacos , Modelos Animais de Doenças , Feminino , Inibidores de Histona Desacetilases/farmacologia , Humanos , Túbulos Renais/citologia , Masculino , Mitocôndrias/patologia , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Espécies Reativas de Oxigênio/metabolismo , Traumatismo por Reperfusão/complicações , Sirtuínas/antagonistas & inibidores , Organismos Livres de Patógenos Específicos
18.
Curr Microbiol ; 75(12): 1634-1641, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30291406

RESUMO

Yersinia pestis is a dangerous bacterial pathogen that can cause plague. Both RovA and cyclic AMP receptor protein (cAMP-CRP) are required for regulating biofilm- and virulence-related genes in Y. pestis. In this study, the transcriptional regulation between RovA and cAMP-CRP were analyzed by using primer extension, quantitative RT-PCR, LacZ fusion, and electrophoretic mobility shift assay. The results indicated that RovA repressed crp transcription in an indirect manner, while that RovA had no regulatory action on cyaA at the transcriptional level. In addition, cAMP-CRP did not regulate the transcription of rovA. Taken together with our previous results, complex regulatory interactions of RovA, cAMP-CRP, and PhoP/PhoQ in Y. pestis were revealed, which would promote us gain deeper understanding about coordinative modulation of biofilm- and virulence-related regulator genes.


Assuntos
Arvicolinae/microbiologia , Proteínas de Bactérias/genética , Proteína Receptora de AMP Cíclico/genética , Genes Reguladores/genética , Fatores de Transcrição/genética , Transcrição Gênica/genética , Yersinia pestis/genética , Animais , Biofilmes , Regulação Bacteriana da Expressão Gênica/genética , Regulon/genética , Virulência/genética
19.
Artigo em Inglês | MEDLINE | ID: mdl-30333962

RESUMO

Biofilm formation is critical for blocking flea foregut and hence for transmission of Y. pestis by flea biting. In this study, we identified the regulatory role of the AraC-family transcriptional regulator BfvR (YPO1737 in strain CO92) in biofilm formation and virulence of Yersinia pestis biovar Microtus. Crystal violet staining, Caenorhabditis elegans biofilm assay, colony morphology assay, intracellular c-di-GMP concentration determination, and BALB/c mice challenge were employed to reveal that BfvR enhanced Y. pestis biofilm formation while repressed its virulence in mice. Further molecular biological assays demonstrated that BfvR directly stimulated the expression of hmsHFRS, waaAE-coaD, and hmsCDE, which, in turn, affected the production of exopolysaccharide, LPS, and c-di-GMP, respectively. In addition, BfvR directly and indirectly repressed psaABC and psaEF transcription, respectively. We concluded that the modulation of biofilm- and virulence-related genes by BfvR led to increased biofilm formation and reduced virulence of Y. pestis biovar Microtus.


Assuntos
Antígenos de Bactérias/metabolismo , Biofilmes/crescimento & desenvolvimento , Regulação Bacteriana da Expressão Gênica , Fatores de Transcrição/metabolismo , Yersinia pestis/crescimento & desenvolvimento , Yersinia pestis/metabolismo , Animais , Caenorhabditis elegans/microbiologia , GMP Cíclico/análogos & derivados , GMP Cíclico/análise , Modelos Animais de Doenças , Redes Reguladoras de Genes , Genes Reguladores , Camundongos Endogâmicos BALB C , Peste/microbiologia , Peste/patologia , Polissacarídeos Bacterianos/metabolismo , Análise de Sobrevida , Virulência , Yersinia pestis/genética
20.
Sci Rep ; 8(1): 7595, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29765072

RESUMO

Restenosis caused by neointimal hyperplasia significantly decreases long-term efficacy of percutaneous transluminal angioplasty (PTA), stenting, and by-pass surgery for managing coronary and peripheral arterial diseases. A major cause of pathological neointima formation is abnormal vascular smooth muscle cell (VSMC) proliferation and migration. Notoginsenoside R1 (NGR1) is a novel saponin that is derived from Panax notoginseng and has reported cardioprotective, neuroprotective and anti-inflammatory effects. However, its role in modulating VSMC neointima formation remains unexplored. Herein, we report that NGR1 inhibits serum-induced VSMC proliferation and migration by regulating VSMC actin cytoskeleton dynamics. Using a mouse femoral artery endothelium denudation model, we further demonstrate that systemic administration of NGR1 had a potent therapeutic effect in mice, significantly reducing neointimal hyperplasia following acute vessel injury. Mechanistically, we show that NGR1's mode of action is through inhibiting the activation of phosphatidylinositol 3-kinase (PI3K)/Akt signaling. Taken together, this study identified NGR1 as a potential therapeutic agent for combating restenosis after PTA in cardiovascular diseases.


Assuntos
Ginsenosídeos/administração & dosagem , Hiperplasia/tratamento farmacológico , Músculo Liso Vascular/citologia , Neointima/tratamento farmacológico , Animais , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Modelos Animais de Doenças , Ginsenosídeos/farmacologia , Humanos , Hiperplasia/metabolismo , Masculino , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Neointima/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...