Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 25
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Am J Chin Med ; 51(6): 1595-1611, 2023.
Article de Anglais | MEDLINE | ID: mdl-37489112

RÉSUMÉ

Cinobufagin, a cardiotonic steroid derived from toad venom extracts, exhibits significant anticancer properties by inhibiting Na[Formula: see text]/K[Formula: see text]-ATPase in cancer cells. It is frequently used in clinical settings to treat advanced-stage cancer patients, improving their quality of life and survival time. However, its long-term use can result in multidrug resistance to other chemotherapy drugs, and the exact mechanism underlying this effect remains unknown. Therefore, this study explores the molecular mechanism underlying the anticancer effects of cinobufagin in hepatocellular carcinomas (HCCs), specifically in HepG2 and Huh-7 cells. As determined using transcriptome analysis, cinobufagin-triggered protective autophagy suppressed cell apoptosis in liver cancer HepG2 and Huh-7 cells by inhibiting the phosphoinositide-3-Kinase (PI3K)-AKT serine/threonine kinase (AKT)-mammalian target of rapamycin (mTOR) pathway. Cinobufagin-inhibited cell proliferation, induced apoptosis, and generated cell autophagy by upregulating the expression of MAP1 light chain 3 protein II, Beclin1, and autophagy-related protein 12-5. In addition, the autophagy inhibitor MRT68921 improved the antiproliferative and proapoptotic effects of cinobufagin in the studied cell lines. Overall, this study suggests that combining cinobufagin with an autophagy inhibitor can effectively treat HCC, providing a potential strategy for cancer therapy.


Sujet(s)
Venins d'amphibien , Carcinome hépatocellulaire , Tumeurs du foie , Humains , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Lignée cellulaire tumorale , Qualité de vie , Apoptose/génétique , Prolifération cellulaire , Autophagie/génétique , Venins d'amphibien/pharmacologie , Venins d'amphibien/usage thérapeutique
2.
ACS Biomater Sci Eng ; 9(6): 3402-3413, 2023 06 12.
Article de Anglais | MEDLINE | ID: mdl-37140447

RÉSUMÉ

Current anticancer research shows that a combination of multiple treatment methods can greatly improve the killing of tumor cells. Using the latest microfluidic swirl mixer technology, combined with chemotherapy and photothermal-ablation therapy, we developed multiresponsive targeted antitumor nanoparticles (NPs) made of folate-functionalized gelatin NPs under 200 nm in size and with encapsulated CuS NPs, Fe3O4 NPs, and curcumin (Cur). By exploring gelatin's structure, adjusting its concentration and pH, and fine-tuning the fluid dynamics in the microfluidic device, the best preparation conditions were obtained for gelatin NPs with an average particle size of 90 ± 7 nm. The comparative targeting of the drug delivery system (DDS) was demonstrated on lung adenocarcinoma A549 cells (low level of folate receptors) and breast adenocarcinoma MCF-7 cells (high level of folate receptors). Folic acid helps achieve targeting and accurate delivery of NPs to the MCF-7 tumor cells. The synergistic photothermal ablation and curcumin's anticancer activity are achieved through infrared light irradiation (980 nm), while Fe3O4 is guided with an external magnetic field to target gelatin NPs and accelerate the uptake of drugs, thus efficiently killing tumor cells. The method described in this work is simple, easy to repeat, and has great potential to be scaled up for industrial production and subsequent clinical use.


Sujet(s)
Antinéoplasiques , Curcumine , Nanoparticules , Humains , Curcumine/pharmacologie , Curcumine/composition chimique , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Gélatine , Microfluidique , Nanoparticules/composition chimique , Acide folique
3.
Biomater Sci ; 11(8): 2845-2859, 2023 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-36857655

RÉSUMÉ

Antimicrobial resistance (AMR) has become a major global health concern prompting the quest for new antibiotics with higher efficiency and less proneness to drug resistance. Antimicrobial peptides (AMPs) offer such properties and have therefore gained increasing attention as a new generation of antibiotics to overcome AMR. In an attempt to develop new highly selective and highly efficient antifungal peptides, a sequence (named At1) originating from the natural AMP Ponericin-W1 was used as a lead sequence for rational design of a series of short cationic antifungal peptides named At2-At12. The charge, hydrophobicity, and terminal amino acids of the peptides were modified in a systematic way to investigate the effect of such structural changes on the biological activity of the peptides. Among all the designed peptides, three peptides (coded as At3, At5 and At10) exhibited high antifungal activity without any significant hemolytic activity in human red blood cells. The higher selectivity of these peptides for fungal cells over human cells was further confirmed in cocultures of Candida albicans and human foreskin fibroblasts. These three peptides lacked any hydrophilic residues in their hydrophobic domain, contained lysine residues in their hydrophilic region and had an overall charge of 7+. They also had a higher helical content in microbial membrane mimicking DPPG SUVs than the rest of the peptides. The fungi did not develop any resistance to the designed antifungal peptides even after 25 generations indicating low AMR. At5 was also used in vivo for the treatment of wounds infected with Candida albicans in mice and showed superiority over fluconazole for treating infection and accelerating wound healing. There was an interplay between the hydrophobicity and positive charge density to determine the antifungal activity of the peptides. The results from this study suggest this class of antifungal peptides as promising candidates for antifungal drugs with high efficiency, high biocompatibility and low propensity for drug resistance.


Sujet(s)
Antifongiques , Peptides antimicrobiens , Humains , Souris , Animaux , Peptides antimicrobiens cationiques/composition chimique , Candida albicans , Antibactériens/composition chimique
4.
Int J Pharm ; 622: 121857, 2022 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-35623489

RÉSUMÉ

Liposomes have been widely used in nanomedicine for the delivery of hydrophobic and hydrophilic anticancer agents. The most common applications of these formulations are vaccines and anticancer formulations (e.g., mRNA, small molecule drugs). However, large-scale production with precise control of size and size distribution of the lipid-based drug delivery systems (DDSs) is one of the major challenges in the pharmaceutical industry. In this study, we used newly designed microfluidic swirl mixers with simple 3D mixing chamber structures to prepare liposomes at a larger scale (up to 320 mL/min or 20 L/h) than the commercially available devices. This design demonstrated high productivity and better control of liposome size and polydispersity index (PDI) than conventional liposome preparation methods. The microfluidic swirl mixer devices were used to produce curcumin-loaded liposomes under different processing conditions which were later characterized and studied in vitro to evaluate their efficiency as DDSs. The obtained results demonstrated that the liposomes can effectively deliver curcumin into cancer cells. Therefore, the microfluidic swirl mixers are promising devices for reproducible and scalable manufacturing of DDSs.


Sujet(s)
Curcumine , Tumeurs , Systèmes de délivrance de médicaments , Liposomes/composition chimique , Microfluidique/méthodes , Nanomédecine , Taille de particule
5.
Mol Med Rep ; 23(3)2021 03.
Article de Anglais | MEDLINE | ID: mdl-33495805

RÉSUMÉ

Subsequently to the publication of the above paper, an interested reader drew to the authors' attention that several pairings of panels in Fig. 5, as shown on p. 5599, were strikingly similar. After having examined their original data, the authors realized that they uploaded some images incorrectly during the process of compiling this figure, and that there were duplicated data panels in this figure. However, the authors were able to consult their original data, and had access to the correct images. The revised version of Fig. 5, showing the correct data for the Akt/Control, p­Akt/Control, mTOR/0.05 µM Ouabain, HIF­1α/0.05 µM Ouabain and Akt/0.5 µM Ouabain experiments, is shown opposite. Note that the replacement of the erroneous data does not affect either the results or the conclusions reported in this paper, and all the authors agree to this Corrigendum. The authors are grateful to the Editor of Molecular Medicine Reports for granting them this opportunity to publish a Corrigendum, and apologize to the readership for any inconvenience caused. [the original article was published in Molecular Medicine Reports 17: 5595­5600, 2018; DOI: 10.3892/mmr.2018.8587].

6.
Am J Chin Med ; 48(3): 651-678, 2020.
Article de Anglais | MEDLINE | ID: mdl-32349518

RÉSUMÉ

Cinobufagin is a Na+/K+-ATPase (NKA) inhibitor with excellent anticancer effects to prolong the survival of patients. The purpose of the present study was to clarify the underlying mechanism of the anticancer effects of cinobufagin using overexpression or inhibition of aurora kinase A (AURKA) signaling. First, high expression of Na+/K+-ATPase alpha 1 subunit (ATP1A1) and AURAK resulted in increased malignant transformation in hepatocellular carcinoma (HCC) patients using the cancer genome atlas (TCGA) data and tissue samples. After treatment with cinobufagin, we successfully screened 202, 249, and 335 changing expression proteins in Huh-7 cells under normal, overexpression, and inhibition of AURKA using tandem mass tags (TMT)-labeled quantitative proteomics coupled to 2D liquid chromatography-tandem mass spectrometry (LC-MS/MS). Bioinformatics analysis revealed that these molecules were closely associated with chromosome segregation, DNA damage, and regulation of translation processes. We further confirmed that cinobufagin induced DNA damage and chromosome segregation disorders and suppresses translational processing in oncogenes by decreasing the expression of AURKA, mechanistic target of rapamycin kinase (mTOR), p-mTOR, p-extracellular regulated protein kinases (ERK), eukaryotic translation initiation factor 4E (eIF4E), and p-eIF4E, while increasing the expression of p-eukaryotic translation initiation factor 4E binding protein 1 (4E-BP1) (S65, T37, T46, T45) and increasing the interaction between eIF4 and 4E-BP1. Our results suggested that cinobufagin performed an antitumor effects in liver cancer cells by inhibiting the AURKA-mTOR-eIF4E axis.


Sujet(s)
Antinéoplasiques d'origine végétale , Aurora kinase A/métabolisme , Bufanolide/pharmacologie , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Protéines de liaison à l'ADN/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Facteurs de transcription/métabolisme , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , Altération de l'ADN/effets des médicaments et des substances chimiques , Expression des gènes/effets des médicaments et des substances chimiques , Oncogènes/génétique , Sodium-Potassium-Exchanging ATPase/métabolisme , Cellules cancéreuses en culture
7.
Mol Med Rep ; 19(5): 4119-4128, 2019 May.
Article de Anglais | MEDLINE | ID: mdl-30942456

RÉSUMÉ

The Na+/K+­ATPase inhibitor cinobufagin exhibits numerous anticancer effects on hepatocellular carcinoma (HCC) cells expressing wild­type p53 via inhibition of aurora kinase A (AURKA) and activation of p53 signaling. However, the effects of cinobufagin on HCC cells expressing mutant p53 remain unclear. In the present study, the anticancer effects of cinobufagin were investigated on HCC Huh­7 cells with mutant p53, and the effects of AURKA overexpression or inhibition on the anticancer effects of cinobufagin were analyzed. Viability, cell cycle progression and apoptosis of cells were determined using an MTT assay, flow cytometry and Hoechst 33342 staining, respectively. The expression levels of p53 and p73 signaling­associated proteins were investigated via western blot analysis. The results demonstrated that the expression levels of AURKA, B­cell lymphoma 2 (Bcl­2), cyclin­dependent kinase 1, cyclin B1, proliferating cell nuclear antigen and heterogeneous nuclear ribonucleoprotein K, as well as the phosphorylation of p53 and mouse double minute 2 homolog, were significantly decreased in Huh­7 cells treated with 5 µmol/l cinobufagin for 24 h. Conversely, the expression levels of Bcl­2­associated X protein, p21, p53 upregulated modulator of apoptosis and phorbol­12­myristate­13­acetate­induced protein 1, were significantly increased by cinobufagin treatment. Overexpression or inhibition of AURKA suppressed or promoted the anticancer effects of cinobufagin on Huh­7 cells, respectively. These results indicated that cinobufagin may induce anticancer effects on Huh­7 cells via the inhibition of AURKA and p53 signaling, and via the activation of p73 signaling, in an AURKA­dependent manner.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Bufanolide/pharmacologie , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Protéine tumorale p73/métabolisme , Apoptose/effets des médicaments et des substances chimiques , Aurora kinase A/métabolisme , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Humains , Protéine p53 suppresseur de tumeur/métabolisme
8.
Mol Med Rep ; 17(4): 5595-5600, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29436645

RÉSUMÉ

Glioma is one of the most malignant forms of brain tumor, and has been of persistent concern due to its high recurrence and mortality rates, and limited therapeutic options. As a cardiac glycoside, ouabain has widespread applications in congestive heart diseases due to its positive cardiac inotropic effect by inhibiting Na+/K+­ATPase. Previous studies have demonstrated that ouabain has antitumor activity in several types of human tumor, including glioma. However, the exact underlying mechanism remains to be elucidated. The purpose of present study was to elucidate the effect of ouabain on human glioma cell apoptosis and investigate the exact mechanism. U­87MG cells were treated with various concentrations of ouabain for 24 h, following which cell viability and survival rate were assessed using a 3­(4,5-dimethylthiazol-2­yl)­2,5­diphenyltetrazolium bromide assay. The dynamic changes and cell motility were observed using digital holographic microscopy. Additionally, western blot analysis and high­content screening assays were used to detect the protein expression levels of phosphorylated (p­)Akt, mammalian target of rapamycin (mTOR), p­mTOR and hypoxia­inducible factor (HIF)­1α, respectively. Compared with the control group, ouabain suppressed U­87MG cell survival, and attenuated cell motility in a dose­dependent manner (P<0.01). The downregulation of p­Akt, mTOR, p­mTOR and HIF­1α were observed following treatment with 2.5 and 25 µmol/l of ouabain. These results suggested that ouabain exerted suppressive effects on tumor cell growth and motility, leading to cell death via regulating the intracellular Akt/mTOR signaling pathway and inhibiting the expression of HIF­1α in glioma cells. The present study examined the mechanism underlying the antitumor property of ouabain, providing a novel potential therapeutic agent for glioma treatment.


Sujet(s)
Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Gliome/génétique , Gliome/métabolisme , Sous-unité alpha du facteur-1 induit par l'hypoxie/génétique , Ouabaïne/pharmacologie , Protéines proto-oncogènes c-akt/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Sérine-thréonine kinases TOR/métabolisme , Lignée cellulaire tumorale , Mouvement cellulaire/effets des médicaments et des substances chimiques , Survie cellulaire/effets des médicaments et des substances chimiques , Gliome/anatomopathologie , Humains , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme
9.
Mol Med Rep ; 17(4): 5213-5221, 2018 04.
Article de Anglais | MEDLINE | ID: mdl-29393410

RÉSUMÉ

An increasing amount of evidence indicates that the inhibition of ß adrenergic signaling can result in the inhibition of tumor growth. However, the role of propranolol in liver cancer and the underlying mechanism remain to be elucidated. The present study aimed to investigate the role of propranolol in liver cancer cell lines and provide evidence for further clinical study. Propranolol was added at different concentrations to HepG2 and HepG2.2.15 liver cancer cells and HL­7702 normal human liver cells. The proliferation of the cell lines was monitored by live­cell imaging at a range of time intervals. Immunofluorescence using DAPI and Hoechst 33342/propidium iodide (PI) staining, Annexin V­FITC/PI double­staining flow cytometry, western blotting and reverse transcription­quantitative polymerase chain reaction were used to investigate the effect of propranolol on liver cancer cell apoptosis. The proliferation of HepG2 and HepG2.2.15 cells was inhibited by 40 and 80 µmol/l propranolol. However, the proliferation of HL­7702 cells was not affected by <160 µmol/l propranolol. Propranolol treatment decreased the expression of adrenergic receptor ß­2 to a greater extent than adrenergic receptor ß­1, and induced apoptosis in the liver cancer cells. The apoptotic rates of HepG2 and HepG2.2.15 cells increased following treatment with propranolol, while the apoptotic rate of HL­7702 cells was not affected. Propranolol promoted poly (ADP­ribose) polymerase cleavage and decreased the expression of full­length caspase­3 in liver cancer cell lines; it induced S­phase arrest in HepG2 and HepG2.2.15 cell lines, while HL­7702 cells were arrested at the G0/G1 phase of the cell cycle. Thus, it was demonstrated that propranolol inhibited proliferation, promoted apoptosis and induced S-phase arrest in HepG2 and HepG2.2.15 cells.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Propranolol/pharmacologie , Antagonistes bêta-adrénergiques/pharmacologie , Apoptose/génétique , Marqueurs biologiques , Carcinome hépatocellulaire , Cycle cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cytométrie en flux , Humains , Tumeurs du foie , Récepteurs bêta-1 adrénergiques/génétique , Récepteurs bêta-1 adrénergiques/métabolisme , Récepteurs bêta-2 adrénergiques/génétique , Récepteurs bêta-2 adrénergiques/métabolisme
10.
Int J Mol Med ; 39(6): 1409-1420, 2017 Jun.
Article de Anglais | MEDLINE | ID: mdl-28487965

RÉSUMÉ

Heme oxygenase-1 (HO-1) is an inducible and cytoprotective enzyme that provides a defense against oxidant damage. The present study screened 137 HO-1/interacting proteins using a profound co-immunoprecipitation (Co-IP) coupled with proteomics, and profiled the global HO-1 interactome network, including oxidative phosphorylation, endoplasmic reticulum and transport vesicle functions. Among these molecules, we observed that a novel interactor, emopamil-binding protein (EBP), is closely related to the cholesterol metabolism process. This study demonstrated that cholesterol promotes excessive oxidative stress and alters the energy metabolism in cardiomyocytes, further triggering numerous cardiovascular diseases. We observed that cholesterol caused the overexpression of EBP and HO-1 by the activation of AKT and Nrf2/mTOR pathways. In addition, HO-1 and EBP performed a myocardial protective function. The overexpression of HO-1 alleviated the cholesterol-induced excessive oxidative stress status by inhibition of the carbohydrate metabolism. Notably, we also confirmed that the loss of partial HO-1 activity aggravated the oxidative damage and cardiac systolic function induced by a high-fat diet in HO-1 heterozygous (HO-1+/-) mice. These findings indicate that the HO-1/EBP interaction plays a protective role in alleviating the dysfunction of oxidative stress and cardiac systolic function induced by cholesterol stimulation.


Sujet(s)
Protéines de transport/métabolisme , Cholestérol/métabolisme , Heme oxygenase-1/métabolisme , Hypoxie/métabolisme , Myocytes cardiaques/métabolisme , Transduction du signal , Steroid isomerases/métabolisme , Animaux , Métabolisme glucidique , Lignée cellulaire , Hypoxie/anatomopathologie , Mâle , Souris de lignée C57BL , Myocytes cardiaques/anatomopathologie , Facteur-2 apparenté à NF-E2/métabolisme , Stress oxydatif , Cartes d'interactions protéiques , Protéines proto-oncogènes c-akt/métabolisme , Sérine-thréonine kinases TOR/métabolisme
11.
Mol Med Rep ; 15(4): 2154-2162, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28259939

RÉSUMÉ

Zing finger protein 580 (ZFP580) is a novel Cys2-His2 zinc-finger transcription factor that has an anti-apoptotic role in myocardial cells. It is involved in the endothelial transforming growth factor­ß1 (TGF­ß1) signal transduction pathway as a mothers against decapentaplegic homolog (Smad)2 binding partner. The aim of the present study was to determine the involvement of ZFP580 in TGF­ß1­mediated cytoprotection against chemical hypoxia­induced apoptosis, using H9c2 cardiac myocytes. Hypoxia was chemically induced in H9c2 myocardial cells by exposure to cobalt chloride (CoCl2). In response to hypoxia, cell viability was decreased, whereas the expression levels of hypoxia inducible factor-1α and ZFP580 were increased. Pretreatment with TGF­ß1 attenuated CoCl2­induced cell apoptosis and upregulated ZFP580 protein expression; however, these effects could be suppressed by SB431542, an inhibitor of TGF­ß type I receptor and Smad2/3 phosphorylation. Furthermore, suppression of ZFP580 expression by RNA interference reduced the anti­apoptotic effects of TGF­ß1 and thus increased CoCl2­induced apoptosis. B­cell lymphoma (Bcl)­2­associated X protein/Bcl­2 ratio, reactive oxygen species generation and caspase­3 activation were also increased following ZFP580 inactivation. In conclusion, these results indicate that ZFP580 is a component of the TGF-ß1/Smad signaling pathway, and is involved in the protective effects of TGF­ß1 against chemical hypoxia­induced cell apoptosis, through inhibition of the mitochondrial apoptotic pathway.


Sujet(s)
Apoptose/effets des médicaments et des substances chimiques , Hypoxie cellulaire/effets des médicaments et des substances chimiques , Cobalt/toxicité , Myocytes cardiaques/cytologie , Myocytes cardiaques/effets des médicaments et des substances chimiques , Facteurs de transcription/métabolisme , Facteur de croissance transformant bêta-1/métabolisme , Animaux , Lignée cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Cytoprotection , Sous-unité alpha du facteur-1 induit par l'hypoxie/métabolisme , Myocytes cardiaques/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Rats , Espèces réactives de l'oxygène/métabolisme , Récepteur de type I du facteur de croissance transformant bêta , Récepteurs TGF-bêta/métabolisme , Protéine Smad2/métabolisme , Protéine Smad-3/métabolisme
12.
J Proteomics ; 130: 211-20, 2016 Jan 01.
Article de Anglais | MEDLINE | ID: mdl-26435418

RÉSUMÉ

Hypoxic status alters the energy metabolism and induces cell injury in cardiomyocytes, and it further triggers the occurrence and development of cardiovascular diseases. Our previous studies have shown that salidroside (SAL) exhibits anti-hypoxic activity. However, the mechanisms remain obscure. In the present study, we successfully screened 92 different expression proteins in CoCl2-induced hypoxic conditions, 106 different expression proteins in the SAL-mediated anti-hypoxic group were compared with the hypoxic group using quantitative proteomics strategy, respectively. We confirmed that SAL showed a positive protective function involving the acetyl-CoA metabolic, tricarboxylic acid (TCA) cycle using bioinformatics analysis. We also demonstrated that SAL plays a critical role in restoring the TCA cycle and in protecting cardiomyocytes from oxidative injury via up-regulation expressions of PDHE1-B, ACO2, SUCLG1, SUCLG2 and down-regulation of MDH2. SAL also inhibited H9c2 cell apoptosis by inhibiting the activation of pro-apoptotic molecules caspase 3 and caspase 9 as well as activation of the anti-apoptotic molecular Bcl-2. Additionally, SAL also improved mitochondrial membrane potential (ΔΨm), reduced reactive oxygen species (ROS) and intercellular Ca(2+) concentration ([Ca(2+)]i) accumulation and inhibited the excessive consumption of ATP in H9c2 cells.


Sujet(s)
Cobalt/composition chimique , Glucosides/composition chimique , Myocytes cardiaques/métabolisme , Phénols/composition chimique , Protéomique/méthodes , Triacides carboxyliques/composition chimique , Adénosine triphosphate/composition chimique , Apoptose , Calcium/composition chimique , Caspase-3/métabolisme , Caspase-9/métabolisme , Lignée cellulaire , Chromatographie en phase liquide , Cycle citrique , Biologie informatique , Hypoxie/anatomopathologie , Potentiels de membrane , Oxygène/composition chimique , Extraits de plantes/composition chimique , Protéome , Protéines proto-oncogènes c-bcl-2/métabolisme , Espèces réactives de l'oxygène/métabolisme , Rhodiola/composition chimique , Spectrométrie de masse en tandem
13.
J Proteome Res ; 14(11): 4594-602, 2015 Nov 06.
Article de Anglais | MEDLINE | ID: mdl-26491887

RÉSUMÉ

Many studies have shown the Na(+)/K(+)-ATPase (NKA) might be a potential target for anticancer therapy. Cardiac glycosides (CGs), as a family of naturally compounds, inhibited the NKA activity. The present study investigates the antitumor effect of ouabain and elucidates the pharmacological mechanisms of CG activity in liver cancer HepG2 cell using SILAC coupled to LC-MS/MS method. Bioinformatics analysis of 330 proteins that were changed in cells under treatment with 0.5 µmol/L ouabain showed that the biological processes are associated with an acute inflammatory response, cell cycle, oxidation reduction, chromosome segregation, and DNA metabolism. We confirmed that ouabain induced chromosome segregation disorder and S-cell cycle block by decreasing the expression of AURKA, SMC2, Cyclin D, and p-CDK1 as well as increasing the expression of p53. We found that the overexpression or inhibition of AURKA significantly reduced or enhanced the ouabain-mediated the anticancer effects. Our findings suggest that AURKA is involved in the anticancer mechanisms of ouabain in HepG2 cells.


Sujet(s)
Antinéoplasiques/pharmacologie , Aurora kinase A/antagonistes et inhibiteurs , Antienzymes/pharmacologie , Régulation de l'expression des gènes tumoraux , Ouabaïne/pharmacologie , Phase S/effets des médicaments et des substances chimiques , Sodium-Potassium-Exchanging ATPase/antagonistes et inhibiteurs , Animaux , Aurora kinase A/génétique , Aurora kinase A/métabolisme , Protéine-kinase CDC2 , Carcinome hépatocellulaire/traitement médicamenteux , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Protéines de transport/antagonistes et inhibiteurs , Protéines de transport/génétique , Protéines de transport/métabolisme , Protéines du cycle cellulaire , Chromatographie en phase liquide , Ségrégation des chromosomes/effets des médicaments et des substances chimiques , Cycline D/antagonistes et inhibiteurs , Cycline D/génétique , Cycline D/métabolisme , Kinases cyclines-dépendantes/antagonistes et inhibiteurs , Kinases cyclines-dépendantes/génétique , Kinases cyclines-dépendantes/métabolisme , Femelle , Réseaux de régulation génique/effets des médicaments et des substances chimiques , Cellules HepG2 , Humains , Tumeurs du foie/traitement médicamenteux , Tumeurs du foie/génétique , Tumeurs du foie/métabolisme , Tumeurs du foie/anatomopathologie , Souris , Souris nude , Protéines nucléaires/antagonistes et inhibiteurs , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Phase S/génétique , Transduction du signal , Sodium-Potassium-Exchanging ATPase/génétique , Sodium-Potassium-Exchanging ATPase/métabolisme , Spectrométrie de masse en tandem , Protéine p53 suppresseur de tumeur/agonistes , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme , Tests d'activité antitumorale sur modèle de xénogreffe
14.
PLoS One ; 9(4): e94635, 2014.
Article de Anglais | MEDLINE | ID: mdl-24722354

RÉSUMÉ

BACKGROUND: ZFP580 is a novel C2H2 type zinc-finger transcription factor recently identified by our laboratory. We previously showed that ZFP580 may be involved in cell survival and growth. The aim of this study was to elucidate whether ZFP580 is involved in the cardioprotective effects of intermittent high-altitude (IHA) hypoxia against myocardial ischemia-reperfusion (I/R) injury. METHODS AND RESULTS: After rats were subjected to myocardial ischemia for 30 min followed by reperfusion, ZFP580 expression in the left ventricle was measured. ZFP580 protein expression was found to be up-regulated within 1 h and decreased at 2 h after reperfusion. Comparing normoxic and IHA hypoxia-adapted rats (5000 m, 6 h day-1, 6 weeks) following I/R injury (30 min ischemia and 2 h reperfusion), we found that adaptation to IHA hypoxia attenuated infarct size and plasma leakage of lactate dehydrogenase and creatine kinase-MB. In addition, ZFP580 expression in the myocardium was up-regulated by IHA hypoxia. Consistent with this result, ZFP580 expression was found to be significantly increased in cultured H9c2 myocardial cells in the hypoxic preconditioning group compared with those in the control group following simulated I/R injury (3 h simulated ischemic hypoxia and 2 h reoxygenation). To determine the role of ZFP580 in apoptosis, lentivirus-mediated gene transfection was performed in H9c2 cells 72 h prior to simulated I/R exposure. The results showed that ZFP580 overexpression significantly inhibited I/R-induced apoptosis and caspase-3 activation. H9c2 cells were pretreated with or without PD98059, an inhibitor of ERK1/2 phosphorylation, and Western blot results showed that PD98059 (10 µM) markedly suppressed I/R-induced up-regulation of ZFP580 expression. CONCLUSIONS: Our findings demonstrate that the cardioprotective effect of IHA hypoxia against I/R injury is mediated via ZFP580, a downstream target of ERK1/2 signaling with anti-apoptotic roles in myocardial cells.


Sujet(s)
Adaptation physiologique/physiologie , Hypoxie/métabolisme , Ischémie myocardique/métabolisme , Lésion de reperfusion myocardique/métabolisme , Facteurs de transcription/métabolisme , Doigts de zinc/physiologie , Altitude , Animaux , Apoptose/physiologie , MB Creatine kinase/métabolisme , Ventricules cardiaques/métabolisme , L-Lactate dehydrogenase/métabolisme , Mâle , Myocarde/métabolisme , Phosphorylation , Rats , Rat Wistar , Régulation positive
15.
Zhongguo Ying Yong Sheng Li Xue Za Zhi ; 30(5): 396-400, 2014 Sep.
Article de Chinois | MEDLINE | ID: mdl-25581930

RÉSUMÉ

OBJECTIVE: To elucidate whether ZFP580 is involved in the cardioprotective effects of intermittent hypobaric hypoxia (IHH) against myocardial ischemia/reperfusion (I/R) injury. METHODS: Thirty two male Wistar rats were randomly divided into 2 groups (n = 16): normoxia control group and IHH preconditioning group. Rats in IHH group were exposed in a hypobaric chamber (equivalent to an altitude of 5 000 m) for a 6 h period each day for 42 d. Plasma was collected and lactate dehydrogenase (LDH) and creatine kinase-MB (CK-MB) were measured after 2 h of myocardial I/R injury. ZFP580 protein expression in myocardial tissue was assayed by Western blot. Other 8 rats in each group were used to evaluate I/R-induced cardiac infarction by TTC staining. Lentivirus-mediated gene transfection was performed in H9c2 cells 72 h prior to simulated ischemia/reperfusion (SI/R) exposure. The degree of cell apoptosis was determined by annexin V/7-AAD staining and flow cytometry analysis. RESULTS: Compared with normoxia control group, adaptation to IHH attenuated infarct size and plasma leakage of LDH and CK-MB. In addition, ZFP580 expression in the myocardium was up-regulated by IHH. The results of gene transfection showed that ZFP580 overexpression significantly inhibited cells apoptosis induced by SI/R. CONCLUSION: Our findings demonstrate that the cardioprotective effect of IHH against I/R injury is mediated via ZFP580, a novel transcription factor, with anti-apoptotic roles in myocardial cells.


Sujet(s)
Hypoxie/physiopathologie , Lésion de reperfusion myocardique/physiopathologie , Facteurs de transcription/métabolisme , Animaux , Apoptose , Lignée cellulaire , MB Creatine kinase/métabolisme , L-Lactate dehydrogenase/métabolisme , Mâle , Myocarde/métabolisme , Myocytes cardiaques/cytologie , Rats , Rat Wistar
16.
J Gastroenterol Hepatol ; 29(4): 851-9, 2014 Apr.
Article de Anglais | MEDLINE | ID: mdl-24303990

RÉSUMÉ

BACKGROUND AND AIM: Recent studies have shown that imbalance between tumor-infiltrating interleukin (IL)-17(+) T cells and regulatory T cells (Tregs) is an important regulator of progression in various cancers, but little is known regarding this imbalance in hepatocellular carcinoma (HCC). This study explored the role of imbalance between IL-17(+) T cells and Tregs in the immunopathogenesis of HCC in patients with chronic hepatitis B (CHB) infection. METHODS: Fifty-six of patient-matched tumors and peritumoral surgical specimens from 56 patient with HCC and 136 liver biopsies specimens from 46 patients with CHB, 37 with atypical hyperplasia (AH), and 53 with HCC were enrolled. The expressions of IL-17, FoxP3, CD4, and CD8 in liver tissue were measured by immunochemistry for the evaluation of liver-infiltrating lymphocytes. RESULTS: The density of liver infiltrated FoxP3(+) Tregs was increased in a stepwise manner from CHB to AH then HCC, while there was a decreasing trend for the density of IL-17(+) T cells and CD8(+) T cells. In surgical specimens of less differentiated HCC, the quantity of tumor-infiltrating FoxP3(+) Tregs was significantly lower and IL-17(+) T cells and CD8(+) T cells were significantly higher. Additionally, peritumoral IL-17(+) T cells were increased in poorly differentiated HCC. High intratumoral FoxP3(+) Tregs with high intratumoral IL-17(+) T cells showed a significantly lower overall survival (OS) and disease-free survival (DFS) compared with other groups (OS, P = 0.033; DFS, P = 0.004). High intratumoral FoxP3(+) Tregs with high peritumoral IL-17(+) T cells showed a significantly lower survival rate compared with other groups (OS, P < 0.001 and DFS, P < 0.001). CONCLUSION: Our findings suggest that intrahepatic IL-17(+) T cells and FoxP3(+) Tregs may cooperate to promote the progression of HCC.


Sujet(s)
Carcinome hépatocellulaire/immunologie , Facteurs de transcription Forkhead/métabolisme , Interleukine-17/métabolisme , Tumeurs du foie/immunologie , Lymphocytes T régulateurs/immunologie , Adulte , Sujet âgé , Carcinome hépatocellulaire/génétique , Carcinome hépatocellulaire/mortalité , Carcinome hépatocellulaire/anatomopathologie , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/immunologie , Évolution de la maladie , Femelle , Expression des gènes , Humains , Foie/métabolisme , Tumeurs du foie/génétique , Tumeurs du foie/mortalité , Tumeurs du foie/anatomopathologie , Mâle , Adulte d'âge moyen , Pronostic , Taux de survie
17.
Zhongguo Zhong Yao Za Zhi ; 37(22): 3467-71, 2012 Nov.
Article de Chinois | MEDLINE | ID: mdl-23373223

RÉSUMÉ

OBJECTIVE: To study the effect of pseudolaric acid B (PLAB) on cell proliferation and cycle of human prostate carcinoma DU-145 cells. method: Its inhibitory effect on the cell growth was measured by MTT method. Characteristics of cell death were determined by Hoechest 33342 staining. The cell cycle was detected by flow cytometry. The expressions of cyclin B1, cyclin D1 and CDK1 were detected by Real time-PCR and Western blot, respectively. RESULT: PLAB notably inhibited DU-145 cell growth in a dose- and time dependent manner (P < 0.05). Its IC50 values of PLAB for DU-145 cells for 24, 48 and 72 h were 4.53, 2.39 and 2.08 micromol x L(-1), respectively. Having been treated with 5 micromol x L(-1) PLAB for 24 h, the cells showed such apoptosis characteristics as nuclei chromatin condensation and apoptotic body. With the increase in PLAB concentration, the proportion of G2/M phase cells strikingly increased in a dose- and time dependent manner (P < 0.05), meanwhile cyclin B1 and CDK1 showed over-expressions (P < 0.05), and the cyclin D1 showed under-expression (P < 0.05). CONCLUSION: PLAB can inhibit the growth of DU-145 cells and induce the cell cycle G2/M arrest, accompanied with the over-expression of cyclin B1 and CDK1, which may be related with its regulation cycle-associated protein degradation.


Sujet(s)
Points de contrôle du cycle cellulaire/effets des médicaments et des substances chimiques , Diterpènes/pharmacologie , Médicaments issus de plantes chinoises/pharmacologie , Tumeurs de la prostate/physiopathologie , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Humains , Mâle , Tumeurs de la prostate/traitement médicamenteux
18.
Cell Biol Int ; 35(11): 1153-7, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21599657

RÉSUMÉ

ZNF580 (gene ID 51157), a novel gene encoding a C2H2 (Cys2-His2) zinc-finger transcription factor, may be involved in the maintenance of vascular endothelium homoeostasis. To investigate the physiological role of the transcription factor ZNF580, we screened human foetal brain cDNA library with a yeast two-hybrid system and identified 14 proteins that interact with ZNF580. The interaction between ZNF580 and Smad2 was confirmed by co-immunoprecipitation. Co-localization between endogenous ZNF580 and Smad2 was mainly found in the nuclei of EA.hy926 endothelial cells with immunofluorescence and confocal microscopy. Our results suggest that ZNF580 is a binding partner of Smad2 and is involved in the signal transduction of the TGF-ß (transforming growth factor-ß) signalling pathway, which provides a basis for additional research to investigate the role of ZNF580 in the maintenance of vascular endothelium homoeostasis and the onset of atherosclerotic diseases.


Sujet(s)
Cartographie d'interactions entre protéines , Protéine Smad2/métabolisme , Facteurs de transcription/métabolisme , Technique de Western , Noyau de la cellule/génétique , Noyau de la cellule/métabolisme , Cellules endothéliales/cytologie , Cellules endothéliales/métabolisme , Technique d'immunofluorescence , Banque de gènes , Cellules HEK293 , Homéostasie , Humains , Immunoprécipitation , Plasmides/génétique , Plasmides/métabolisme , Transduction du signal , Protéine Smad2/génétique , Facteurs de transcription/génétique , Transfection , Facteur de croissance transformant bêta/métabolisme , Techniques de double hybride
19.
J Steroid Biochem Mol Biol ; 125(3-5): 181-91, 2011 Jul.
Article de Anglais | MEDLINE | ID: mdl-21215801

RÉSUMÉ

Recent studies revealed the potential of Na(+)/K(+)-ATPase as a target for anticancer therapy and showed additional modes of action of cardiotonic steroids (CSs), a diverse family of naturally derived compounds, as inhibitors of Na(+)/K(+)-ATPase. The results from epidemiological studies showed significantly lower mortality rates in cancer patients receiving CSs, which sparked interest in the anticancer properties of these drugs. The present study was designed to investigate the anticancer effect of CSs (ouabain or cinobufagin) and to elucidate the molecular mechanisms of CS activity in hepatoma cell lines (HepG2 and SMMC-7721). Ouabain and cinobufagin significantly inhibited cell proliferation by attenuating the phosphorylation of extracellular regulated kinase (ERK) and down-regulating the expression of C-myc. These CSs also induced cell apoptosis by increasing the concentration of intracellular free calcium ([Ca(2+)](i)) and induced S phase cell cycle arrest by down-regulating the expression of Cyclin A, cyclin dependent kinase 2 (CDK2) and proliferating cell nuclear antigen (PCNA) as well as up-regulating the expression of cyclin dependent kinase inhibitor 1A (p21(CIP1)). Overexpression of ERK reversed the antiproliferation effect of ouabain or cinobufagin in HepG2 and SMMC-7721 cells. Currently, the first generation of CS-based anticancer drugs (UNBS1450 and Anvirzel) are in Phase I clinical trials. These data clearly support their potential use as cancer therapies.


Sujet(s)
Bufanolide/pharmacologie , Cycle cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Extracellular Signal-Regulated MAP Kinases/métabolisme , Ouabaïne/pharmacologie , Phosphorylation/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Technique de Western , Calcium/métabolisme , Carcinome hépatocellulaire/métabolisme , Carcinome hépatocellulaire/anatomopathologie , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Cycline A/génétique , Cycline A/métabolisme , Kinase-2 cycline-dépendante/génétique , Kinase-2 cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/génétique , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Cellules HepG2 , Humains , Microscopie confocale , Antigène nucléaire de prolifération cellulaire/génétique , Antigène nucléaire de prolifération cellulaire/métabolisme , RT-PCR
20.
Biol Pharm Bull ; 33(5): 743-51, 2010.
Article de Anglais | MEDLINE | ID: mdl-20460749

RÉSUMÉ

Recent research has shown that the Na(+)/K(+)-ATPase alpha1 subunit is a novel anti-cancer target, which plays pivotal roles in malignant cell ion transport, metabolism, migration and signal transduction. The purpose of the present study was to investigate the anti-cancer effects of ouabain and Na(+)/K(+)-ATPase alpha1 small interfering ribonucleic acid (siRNA) on HepG2 cell proliferation, apoptosis and cell cycle, and to explore the molecular mechanisms. The expression of Na(+)/K(+)-ATPase alpha1 subunit in human hepatocellular carcinoma (HCC), normal liver tissues and human HCC line (HepG2, SMMC-7721 and Bel-7402) has been investigated. Using the ouabain and Na(+)/K(+)-ATPase alpha1 subunit siRNA, which target the Na(+)/K(+)-ATPase, we have evaluated the effects of inhibiting Na(+)/K(+)-ATPase alpha1 in human HepG2 cells with respect to cell proliferation, morphology, cell cycle, impact on intracellular Ca2++, reactive oxygen species (ROS) concentration, and correlated gene expression level on messenger ribonucleic acid (mRNA) and protein. Our data showed that the expression Na(+)/K(+)-ATPase alpha1 subunit in HCC tissues is higher than that in normal liver tissues. Ouabain and Na(+)/K(+)-ATPase alpha1 siRNA could inhibit HepG2 cell proliferation. Ouabain could induce HepG2 cell apoptosis and generate S phase arrest, and siRNA could enhance the anti-cancer effect of ouabain that induced HepG2 cells apoptosis via an intracellular Ca(2+) and ROS increase-mediated, and generated cell cycle S phase arresting by decreasing the CyclinA1/cyclin-dependent kinase 2 (CDK2)/proliferating cell nuclear antigen (PCNA) complex product and increasing the expression of cyclin-dependent kinase inhibitor 1A (P21(CIP1)). We believe that targeting of the Na(+)/K(+)-ATPase alpha1 subunit in human HCC cells could provide new sight into the treatment of HCC.


Sujet(s)
Antinéoplasiques d'origine végétale/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Carcinome hépatocellulaire/métabolisme , Tumeurs du foie/métabolisme , Foie/effets des médicaments et des substances chimiques , Ouabaïne/pharmacologie , Sodium-Potassium-Exchanging ATPase/antagonistes et inhibiteurs , Antinéoplasiques d'origine végétale/usage thérapeutique , Calcium/métabolisme , Carcinome hépatocellulaire/traitement médicamenteux , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cycline A1/métabolisme , Kinase-2 cycline-dépendante/métabolisme , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Cellules HepG2 , Humains , Foie/métabolisme , Tumeurs du foie/traitement médicamenteux , Phytothérapie , Extraits de plantes/pharmacologie , Antigène nucléaire de prolifération cellulaire/métabolisme , Structure tertiaire des protéines , Petit ARN interférent/métabolisme , Espèces réactives de l'oxygène/métabolisme , Phase S/effets des médicaments et des substances chimiques
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE