Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 651: 121-126, 2023 04 09.
Article in English | MEDLINE | ID: mdl-36822125

ABSTRACT

Hydroxyl radical (·OH) generated by the Fenton reaction between transition metal ions and hydrogen peroxide (H2O2) can induce significant cellular damage. However, the specific mechanism of ·OH-induced cell death has not been systematically studied. In this study, we reacted FeSO4 and Fe3O4 magnetic nanoparticles with H2O2 and found that ·OH generated from the intracellular Fenton reaction can lead to significant cell death. The Fenton reaction between Fe2+ with H2O2 resulted in a shift in lipid peroxidation and cell cycle arrest. It is noteworthy that the ·OH generated from the Fenton reaction triggered severe apoptosis but did not lead to DNA double-strand breakage. Our results suggest that the Fenton reaction had acute cytotoxicity, which was primarily due to ·OH produced from the Fenton reaction inducing lipid peroxidation and apoptosis and modulating the cell cycle, but not by inducing DNA damage.


Subject(s)
Hydrogen Peroxide , Iron , Lipid Peroxidation , Hydrogen Peroxide/pharmacology , Hydrogen Peroxide/metabolism , Hydroxyl Radical , DNA/metabolism , Cell Cycle , Apoptosis
2.
BMC Cancer ; 22(1): 628, 2022 Jun 08.
Article in English | MEDLINE | ID: mdl-35672728

ABSTRACT

BACKGROUND: Eukaryotic translation elongation factors 1 δ (EEF1D), has garnered much attention with regards to their role in the drug resistance of cancers. In this paper, we investigated the effects and mechanisms of increasing the sensitivity of ovarian cancer cells to cisplatin or cis-dichlorodiammine platinum (DDP) by knockdown and knockout of EEF1D gene in cellular and animal models. METHODS: The EEF1D gene was knocked-down or -out by siRNA or CRISPR/Cas9 respectively in human ovarian cancer cell SKOV3, DDP-resistant subline SKOV3/DDP, and EEF1D gene in human primary ovarian cancer cell from 5 ovarian cancer patients with progressive disease/stable disease (PD/SD) was transiently knocked down by siRNA interference. The mice model bearing xenografted tumor was established with subcutaneous inoculation of SKOV3/DDP. RESULTS: The results show that reducing or removing EEF1D gene expression significantly increased the sensitivity of human ovarian cancer cells to DDP in inhibiting viability and inducing apoptosis in vitro and in vivo, and also boosted DDP to inhibit xenografted tumor growth. Interfering with EEF1D gene expression in mice xenografted tumor significantly affected the levels of OPTN, p-Akt, Bcl-2, Bax, cleaved caspase-3 and ERCC1 compared to DDP treated mice alone, and had less effect on PI3K, Akt and caspase-3. CONCLUSIONS: The knocking down or out EEF1D gene expression could enhance the sensitivity of ovarian cancer cells to DDP partially, which may be achieved via inactivating the PI3K/AKT signaling pathway, thus inducing cell apoptosis and decreasing repairment of DNA damage. Our study provides a novel therapeutic strategy for the treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis , Carcinoma, Ovarian Epithelial/drug therapy , Caspase 3/metabolism , Cell Line, Tumor , Cell Proliferation , Cisplatin/pharmacology , Cisplatin/therapeutic use , Drug Resistance, Neoplasm/genetics , Female , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peptide Elongation Factor 1/genetics , Peptide Elongation Factor 1/metabolism , Peptide Elongation Factor 1/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/genetics
3.
Transl Psychiatry ; 12(1): 230, 2022 06 04.
Article in English | MEDLINE | ID: mdl-35662233

ABSTRACT

OPTN is an autophagy receptor involved in autophagic degradation. Here we studied the role of OPTN in attenuating the neurotoxicity induced by mutated Tau protein. We constructed recombinant adeno-associated viruses with OPTN and Tau-P301L genes, respectively. Through virus coinfection on neuronal cell line HT22 in vitro and Kunming mice in vivo, we found that autophagy- and apoptosis-associated genes are altered by Tau-P301L at both mRNA and protein levels, which are restored by OPTN expression. Functionally, OPTN suppresses apoptosis and enhances cellular viability in Tau-P301L expressing HT22 cells, and increases learning and memory in Tau-P301L expressing mice, respectively. Last, we found that OPTN reduces the p-Tau levels in vitro and in vivo. Our results reveal the function of OPTN in lowering the p-Tau level and the expressions of apoptosis genes, and increasing the expressions of autophagic genes, indicating a beneficial role of OPTN in Tau pathology.


Subject(s)
Autophagy , tau Proteins , Animals , Autophagy/genetics , Carrier Proteins/metabolism , Cell Cycle Proteins , Cells, Cultured , Membrane Transport Proteins/metabolism , Mice , Neurons/metabolism , tau Proteins/metabolism
4.
J Cancer ; 12(20): 6081-6093, 2021.
Article in English | MEDLINE | ID: mdl-34539881

ABSTRACT

Ovarian cancer is the leading cause of death in gynecologic malignancies. Ovarian cancer as a metastatic malignant tumor is highly recurrent and prone to drug resistance. Bioactive peptides are an emerging area of biomedical research in reducing resistance of tumor cell to drugs. In this paper, we investigated the effects and mechanisms of bioactive hexapeptide (PGPIPN) derived in milk protein on the sensitivity of ovarian cancer cells to cis-dichlorodiammine platinum (DDP). Human ovarian cancer cell lines (SKOV3 and COC1), their DDP-resistant sublines (SKOV3/DDP and COC1/DDP) and human primary ovarian cancer cells were cultured in vitro under the combined treatment of DDP (close to IC50) and different concentrations of PGPIPN. The viabilities, apoptosis and cell cycle changes were respectively measured by WST-8 and flow cytometry. The mRNA and protein expression levels of HSF1, HSP70, MDR1, ERCC1 and ß-actin gene were respectively assayed by RT-qPCR and western blotting. The results showed that PGPIPN significantly increased the sensitivity of human ovarian cancer cells to DDP in inhibiting viability and inducing apoptosis in vitro. But the effects in sensitive cells were lower than DDP-resistant cells. PGPIPN significantly changed the cell cycles in all human ovarian cancer cells, which leaded to a significant increase in the percentage of cells blocked at G2/M phase and decrease the percentage of cells at G1 phases in a dose-dependent manner. PGPIPN affected the expression levels of HSF1, HSP70, MDR1 and ERCC1 genes. Compared with cells in DDP treatment alone, the expression levels of HSF1 and HSP70 in human ovarian cancer cells treated with DDP and PGPIPN together significantly decreased in dose-dependent manner. PGPIPN significantly decreased MDR1 and ERCC1 of drug-resistant ovarian cancer cell lines and human primary ovarian cancer cell in a dose-dependent manner. Pifithrin-µ (PFTµ, HSP70 inhibitor) decreased or removed the effects of peptide in increasing the sensitivity of ovarian cancer cells to DDP. This suggests that PGPIPN enhanced the sensitivity of ovarian cancer cells to DDP partially via reducing the activity of HSF1/HSP70 signaling pathway, thus inducing cell apoptosis and decreasing repairment of DNA damage.

5.
ACS Omega ; 6(19): 12598-12604, 2021 May 18.
Article in English | MEDLINE | ID: mdl-34056410

ABSTRACT

Using density functional theory and a cluster approach, we study the reaction potential surface and compute Gibbs free energies for the acylate reaction of ß-lactamase with penicillin G, where the solvent effect is important and taken into consideration. Two reaction paths are investigated: one is a multi-step process with a rate-limit energy barrier of 19.1 kcal/mol, which is relatively small, and the reaction can easily occur; the other is a one-step process with a barrier of 45.0 kcal/mol, which is large and thus makes the reaction hard to occur. The reason why the two paths have different barriers is explained.

6.
Immunotherapy ; 13(2): 113-123, 2021 02.
Article in English | MEDLINE | ID: mdl-33076728

ABSTRACT

Aim: To explore the expression of programmed death-1 (PD-1) or programmed death ligand 1 (PD-L1), natural killer T (NKT) and hepatoma cells in coculture system, and the influence of abolishing PD-1 on antitumor efficiency. Materials & methods: CRISPR/Cas9 technology, flow cytometry, ELISA, CCK-8 assay and mouse models were performed to investigate the interactions between PD-1/PD-L1 expression on NKT and hepatoma cells, respectively. Results: The NKT and hepatoma cells mutually affected the expression of PD-1/PD-L1. The killing effect was positively correlated with NKT-mediated PD-L1 expression on hepatoma cells. Conclusion: Hepatoma cells in different genetic background responded differently to NKT-induced PD-L1 stimulation, and those cells with lower PD-L1 expression fail to PD-1 blocking intervention. Additionally, the killing effect was more time-efficient with PD-1 knockout than with monoclonal antibody blockade.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Natural Killer T-Cells/immunology , Programmed Cell Death 1 Receptor/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/immunology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Gene Knockout Techniques , Humans , Interferon-gamma/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , Mice , Mice, Nude , Natural Killer T-Cells/metabolism , Natural Killer T-Cells/transplantation , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology
7.
Onco Targets Ther ; 13: 7387-7398, 2020.
Article in English | MEDLINE | ID: mdl-32801762

ABSTRACT

BACKGROUND/AIMS: Long noncoding RNAs (lncRNAs) play a critical role in tumorigenesis and progression of ovarian cancer (OC). This study focused on the function and potential mechanism toward LEMD1-AS1 (LEMD1 antisense RNA 1) in the progression of ovarian cancer. MATERIALS AND METHODS: The expression of LEMD1-AS1 in OC tissues was evaluated in TCGA and Gene Expression Omnibus datasets (GSE119056) and confirmed in OC cell lines via qRT-PCR (quantitative real-time polymerase chain reaction). Then, the location of LEMD1-AS1 in the cytoplasmic and nuclear RNAs extracted from OV cells was detected by qRT-PCR. Cell Counting Kit-8 (CCK-8), colony formation, wound-healing and transwell assays were applied to examine cell viability, proliferation, migration and invasion, respectively. Further, the effect of LEMD1-AS1 on OC tumor growth was determined via subcutaneous xenotransplanted tumor model. The potential target for LEMD1-AS1 was validated via dual-luciferase activity assay, RNA pull-down and RNA immunoprecipitation. RESULTS: The expression of LEMD1-AS1 was decreased in OC tissues and cell lines. Forced overexpression of LEMD1-AS1 inhibited the proliferation, migration and invasion of ovarian cancer cells and transplanted tumor growth in nude mice. We found that LEMD1-AS1 was mainly located in the cytoplasm of OC cells and contained complementary sites of miR-183-5p. Mechanistically, our results showed that LEMD1-AS1 could directly interact with miR-183-5p and tumor protein p53 (TP53). The anti-tumor role of LEMD1-AS1 on OC progression depended on miR-183-5p-mediated TP53 expression. CONCLUSION: LEMD1-AS1 suppresses OC progression through sponging miR-183-5p and regulation of TP53, suggesting a novel biomarker and target for OC.

8.
Int J Mol Med ; 46(3): 1107-1117, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32705158

ABSTRACT

Bioactive peptides are an emerging area of biomedical research in the study of numerous human diseases, including acute alcoholic liver injury (AALI). To study the role and mechanism of the milk­derived hexapeptide Pro­Gly­Pro­Ile­Pro­Asn (PGPIPN) in preventing and reducing AALI, the present study established a mouse model of AALI. PGPIPN was used as a therapeutic drug, and glutathione (GSH) was used as a positive control. The body and liver weights of mice were measured, and the liver indexes were calculated to observe mice health. The pathological morphology of liver tissues stained with hematoxylin and eosin were examined to analyze hepatic injury, and hepatocyte apoptosis was measured with a TUNEL assay. The concentrations or activities of alanine aminotransferase (ALT), aspartate aminotransferase, tumor necrosis factor­α, interleukin (IL)­1ß, IL­6, triglyceride, total cholesterol, malondialdehyde, superoxide dismutase and GSH peroxidase (GSH­PX) were detected in serum and/or liver homogenates. The 78 kDa glucose­regulated protein (GRP78), protein kinase R­like (PKR) endoplasmic reticulum kinase (PERK), phosphorylated (p)­PERK, eukaryotic initiation factor 2α (eIF­2α), p-eIF-2α, inositol­requiring enzyme 1α (IRE­1α), spliced X­box binding protein 1 (XBP­1s), C/EBP homologous protein (CHOP), caspase­3 and cleaved caspase­3 proteins associated with endoplasmic reticulum stress in hepatocytes were assessed by western blotting, and RNA levels of XBP­1s, CHOP and caspase­3 genes were assessed by reverse transcription­quantitative PCR. The results suggested that PGPIPN attenuated alcoholic hepatocyte damage in animal models and reduced hepatocyte oxidative stress in a dose­dependent manner. Moreover, PGPIPN reduced endoplasmic reticulum stress by regulating the expression levels of p­PERK, p­eIF­2α, XBP­1s, CHOP, caspase­3 and cleaved caspase­3. Collectively, the present results indicated that PGPIPN, as a potential therapeutic drug for AALI, exerted a protective effect on the liver and could reduce liver damage.


Subject(s)
Endoplasmic Reticulum Stress/drug effects , Liver Diseases, Alcoholic/drug therapy , Oligopeptides/therapeutic use , Protective Agents/therapeutic use , Animals , Apoptosis/drug effects , Endoplasmic Reticulum Chaperone BiP , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Diseases, Alcoholic/metabolism , Liver Diseases, Alcoholic/pathology , Liver Diseases, Alcoholic/prevention & control , Male , Mice , Milk/chemistry , Oligopeptides/chemistry , Oxidative Stress/drug effects , Protective Agents/chemistry
9.
Oncotarget ; 8(50): 88079-88093, 2017 Oct 20.
Article in English | MEDLINE | ID: mdl-29152143

ABSTRACT

PGPIPN is a therapeutic hexapeptide derived from bovine ß-casein. Here we investigated the role and mechanism of this peptide on alcoholic fatty liver disease (AFLD). We took human hepatic cell line LO2 and hepatocellular carcinoma cell line HepG2 to establish the models of steatosis hepatocyte induced by alcohol, taken PGPIPN as pharmacological intervention. And we also established the model of AFLD mice, taken PGPIPN as therapeutic drug and glutathione (GSH) as positive control. We assayed the biochemical materials related to liver injury, lipid metabolism and oxidation, and observed morphology change and fat accumulation of hepatocyte. The gene expressions and/or activities related to liver injury, lipid metabolism and oxidation, such as ACC, PPAR-γ, CHOP and Caspase-3, were assessed by real time PCR and western blot. Our results showed PGPIPN alleviated hepatic steatosis in both model cells and AFLD model mice. PGPIPN can effectively reduce the lipid accumulation and oxidative stress of hepatocyte in a dose-dependent manner. PGPIPN alleviated alcohol-induced cell steatosis and injuries by regulating the gene expressions and/or activities of ACC, PPAR-γ, CHOP and Caspase-3. Our results demonstrated PGPIPN had the protective and therapeutic effect on AFLD, which may serve as a potential therapeutic agent for AFLD.

10.
Anticancer Agents Med Chem ; 17(4): 635-643, 2017.
Article in English | MEDLINE | ID: mdl-27357543

ABSTRACT

BACKGROUND: Some bioactive peptides derived from natural resources or synthesized by rational design have been shown to have very good anticancer effects. We designed an anticancer fusion peptide (ACFP) based on the structure of bovine lactoferricin (LfcinB) and hexapeptide (PGPIPN) derived from bovine milk protein. OBJECTIVE: To prepare ACFP through genetic engineering and study its antiovarian cancer activity. METHOD: ACFP gene was produced by a flexible link arm connecting LfcinB and PGPIPN. ACFP was inductively expressed in Escherichia coli by the recombinant plasmid pGEX-KG-ACFP. ACFP was prepared and purified by affinity chromatography, and identified by polyacrylamide gel electrophoresis (PAGE), high-performance liquid chromatography (HPLC) and mass spectrometry (MS). Recombinant lentivirus vectors were produced by cotransfecting 293T cells with constructed plasmid pLJM1-ACFP, envelope plasmid Δ8.91 and pVSVG using Lipofectamine. ACFP gene was transfected into ovarian cancer cells by pLJM1-ACFP lentivirus. Cell Viability was assayed by the methyl thiazolyl tetrazolium (MTT). The apoptosis of ovarian cancer SKOV3 cells was measured by flow cytometry and observed by Hoechst33258 staining. RESULTS: ACFP was successfully prepared and purified by genetic engineering. ACFP more effectively inhibited the viability of human ovarian cancer SKOV3 cells than the single parent peptides in vitro. ACFP was found to have no cytotoxicity towards untransformed cells. The ACFP gene in cancer cells infected with pLJM1-ACFP lentivirus could significantly inhibit the viability of SKOV3 cells and induce their apoptosis. CONCLUSION: ACFP is a potential therapeutic agent for the treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Milk Proteins/chemistry , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Peptides/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/isolation & purification , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Flow Cytometry , Genetic Engineering , HEK293 Cells , Humans , Peptides/chemistry , Peptides/genetics , Structure-Activity Relationship
11.
Oncotarget ; 7(19): 28435-47, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27078846

ABSTRACT

Tuberous sclerosis complex (TSC), caused by loss-of-function mutations in the TSC1 or TSC2 gene, is characterized by benign tumor formation in multiple organs. Hyperactivation of mammalian target of rapamycin complex 1 (mTORC1) is the primary alteration underlying TSC tumors. By analyzing Tsc2-null mouse embryonic fibroblasts (MEFs) and rat uterine leiomyoma-derived Tsc2-null ELT3 cells, we detected evidence for the involvement of cyclooxygenase 2 (COX2) as a downstream target of mTORC1 in the development of TSC tumors. We showed that loss of TSC2 led to decreased COX2 expression through activation of an mTORC1/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Overexpression of COX2 promoted proliferation and tumoral growth of Tsc2-null cells. COX2 knockdown inhibited the proliferation of the control cells. COX2 enhanced Tsc2-null cell growth through upregulation of interleukin-6 (IL-6). In addition, rapamycin in combination with celecoxib, a COX2 inhibitor, strongly inhibited Tsc2-deficient cell growth. We conclude that downregulation of COX2 exerts a protective effect against hyperactivated mTORC1-mediated tumorigenesis caused by the loss of TSC2, and the combination of rapamycin and celecoxib may be an effective new approach to treating TSC.


Subject(s)
Cyclooxygenase 2/biosynthesis , Mechanistic Target of Rapamycin Complex 1/metabolism , Neoplasms/genetics , Tuberous Sclerosis/complications , Tumor Suppressor Proteins/deficiency , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carcinogenesis/metabolism , Celecoxib/pharmacology , Cell Proliferation/drug effects , Down-Regulation , Mice , Neoplasms/metabolism , Rats , Sirolimus/pharmacology , Tuberous Sclerosis/metabolism , Tuberous Sclerosis/pathology , Tuberous Sclerosis Complex 2 Protein
12.
BMC Cancer ; 16: 246, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-27012847

ABSTRACT

BACKGROUND: ACFP is an anti-cancer fusion peptide derived from bovine milk protein. This study was to investigate the anti-cancer function and underlying mechanisms of ACFP in ovarian cancer. METHODS: Fresh ovarian tumor tissues were collected from 53 patients who underwent initial debulking surgery, and primary cancer cells were cultured. Normal ovarian surface epithelium cells (NOSECs), isolated from 7 patients who underwent surgery for uterine fibromas, were used as normal control tissue. Anti-viabilities of ACFP were assessed by WST-1 (water-soluble tetrazolium 1), and apoptosis was measured using a flow cytometry-based assay. Gene expression profiles of ovarian cancer cells treated with ACFP were generated by cDNA microarray, and the expression of apoptotic-specific genes, such as bcl-xl, bax, akt, caspase-3, CDC25C and cyclinB1, was assessed by real time PCR and western blot analysis. RESULTS: Treatment with ACFP inhibited the viability and promoted apoptosis of primary ovarian cancer cells but exhibited little or no cytotoxicity toward normal primary ovarian cells. Mechanistically, the anti-cancer effects of ACFP in ovarian cells were shown to occur partially via changes in gene expression and related signal pathways. Gene expression profiling highlighted that ACFP treatment in ovarian cancer cells repressed the expression of bcl-xl, akt, CDC25C and cyclinB1 and promoted the expression of bax and caspase-3 in a time- and dose-dependent manner. CONCLUSIONS: Our results suggest that ACFP may represent a potential therapeutic agent for ovarian cancer that functions by altering the expression and signaling of cancer-related pathways in ovarian cancer cells.


Subject(s)
Milk Proteins/administration & dosage , Oligonucleotide Array Sequence Analysis/methods , Ovarian Neoplasms/drug therapy , Peptides/administration & dosage , Animals , Apoptosis/drug effects , Caspase 3/biosynthesis , Cattle , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclin B1/biosynthesis , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Signal Transduction/drug effects , bcl-2-Associated X Protein/biosynthesis , cdc25 Phosphatases/biosynthesis
13.
Int J Oncol ; 48(4): 1721-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26893013

ABSTRACT

Some bioactive peptides derived from natural resources or synthesized by rational design have been proved to have very good anticancer effect. We studied the inhibition of PGPIPN, a hexapeptide derived from bovine ß-casein, on the invasion and metastasis of human ovarian cancer cells in vitro and its molecular mechanism. The human ovarian cancer cells studied include the cell line SKOV3 as well as the primary ovarian cancer cells from ovarian tumor tissues of 37 patients at initial debulking surgery, diagnosed as serous ovarian adenocarcinoma. We showed that PGPIPN inhibited the invasion of ovarian cancer cells with Transwell chamber assay, the migration of ovarian cancer cells with cell scratch assay and colony formation of ovarian cancer cells. The expression (mRNAs and proteins) of genes relevant to invasion and metastasis, MTA1, and NM23H1 were analyzed by real-time PCR and western blotting. PGPIPN repressed the expression of MTA1, and promoted NM23H1. The effects of PGPIPN were dose-dependent. Thus, our study suggests that PGPIPN is a potential therapeutic agent for adjuvant therapy of human malignant ovarian tumors.


Subject(s)
Histone Deacetylases/metabolism , NM23 Nucleoside Diphosphate Kinases/metabolism , Oligopeptides/pharmacology , Ovarian Neoplasms/metabolism , Repressor Proteins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Dose-Response Relationship, Drug , Female , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylases/genetics , Humans , NM23 Nucleoside Diphosphate Kinases/genetics , Neoplasm Invasiveness , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Repressor Proteins/genetics , Trans-Activators
14.
PLoS One ; 11(1): e0146094, 2016.
Article in English | MEDLINE | ID: mdl-26771557

ABSTRACT

AIM: SB-710411 is a rat selective urotensin-II (U-II) receptor antagonist, which can block U-II-induced contraction of the aorta and inhibit U-II-induced myocardial fibrosis in rats. However, the effect of SB-710411 on myocardial ischemia-reperfusion (I/R) injury is unclear. The present study was designed to investigate whether SB-710411 has a protective effect on myocardial I/R injury in rats and the possible mechanisms. METHODS AND RESULTS: Myocardial I/R injury was induced by occluding the left anterior descending coronary artery in adult male Sprague-Dawley rats. Hemodynamic parameters, electrocardiogram (ECG), infarct size, histological alteration, lactate dehydrogenase (LDH), creatine phosphokinase-MB (CK-MB), cardiac troponin I (cTnI), RhoA, and the protein expressions of U-II receptor (UTR), ROCK1 and ROCK2 were evaluated. Cardiac I/R injury significantly up-regulated the expressions of UTR, ROCK1 and ROCK2 proteins in rat myocardium. SB-710411 1.0 and 2.0 µg/kg significantly reduced cardiac I/R-induced the infarct size and histological damage in rat myocardium, markedly inhibited the changes of hemodynamic parameters and the increases of ST-segment in ECG, the serum LDH and CK-MB activities and cTnI level in rats subjected to myocardial I/R injury. Furthermore, SB-710411 obviously prevented myocardial I/R-increased RhoA activity and UTR, ROCK1 and ROCK2 protein expressions. CONCLUSIONS: Our results indicate that cardiac I/R injury increases myocardial UTR expression, and SB-710411 has a potent protective effect on myocardial I/R injury in rats. The cardioprotection may be associated with the inhibition of UTR-RhoA/ROCK pathway.


Subject(s)
Myocardial Reperfusion Injury/prevention & control , Peptides, Cyclic/therapeutic use , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cardiotonic Agents/therapeutic use , Heart/drug effects , Male , Rats , Rats, Sprague-Dawley , Urotensins/antagonists & inhibitors
15.
PLoS One ; 8(4): e60701, 2013.
Article in English | MEDLINE | ID: mdl-23593287

ABSTRACT

Bioactive peptides, either derived from nature resources or synthesized by rational design, have been demonstrated potential for therapeutic agents against numerous human diseases, including cancer. However, the mechanism of therapeutic peptides against cancer has not been well elucidated. Here we show that PGPIPN, a hexapeptide derived from bovine ß-casein, inhibited the proliferation of human ovarian cancer cells line SKOV(3) as well as the primary ovarian cancer cells in vitro. Consistently, PGPIPIN also decreased tumor growth rate in xenograft ovarian cancer model mice in a dose-dependent manner. Further study demonstrated that the anti-tumor effect of PGPIPN is partially through promoting cell apoptosis by inhibiting BCL2 pathway. Thus, our study suggests that PGPIPN is a potential therapeutic agent for the treatment of ovarian cancer or other types of cancer.


Subject(s)
Molecular Targeted Therapy/methods , Oligopeptides/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Caseins/chemistry , Cattle , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Mice , Oligopeptides/chemistry , Oligopeptides/therapeutic use , Xenograft Model Antitumor Assays
16.
BMC Cancer ; 10: 354, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20602797

ABSTRACT

BACKGROUND: In recent years, some miRNAs have been reported to be connected closely with the development of human hepatocellular carcinoma. In our previous studies, a set of miRNAs were revealed to be dysregulated in HCC tissues. However, the functions of these miRNAs in HCC remain largely undefined. METHODS: The expression profiles of miR-183 were compared between HCC tissues and adjacent normal liver tissues using qRT-PCR method. This method was used to screen the potential target genes of miR-183. A luciferase reporter assay was conducted to confirm target association. Finally, the functional effect of miR-183 in hepatoma cells was examined. RESULTS: Among the 25 HCC samples analyzed, microRNA-183 was significantly up-regulated (twofold to 367-fold) in 17 samples compared with the matching nontumoral liver tissues. Programmed cell death 4 (PDCD4) was identified as the target gene of miR-183. Moreover, PDCD4 is a proapoptotic molecule involved in TGF-beta1-induced apoptosis in human HCC cells, we found that miR-183 transfectants were resistant to apoptosis induced by TGF-beta1. CONCLUSIONS: We conclude that miR-183 can inhibit apoptosis in human HCC cells by repressing the PDCD4 expression, and miR-183 may play an important role in HCC development.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , MicroRNAs/physiology , RNA-Binding Proteins/metabolism , Transforming Growth Factor beta1/metabolism , Adult , Aged , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/genetics , Blotting, Western , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , DNA Primers/chemistry , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Liver/metabolism , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Luciferases/metabolism , Male , Middle Aged , Prognosis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/genetics , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , Transforming Growth Factor beta1/genetics , Tumor Cells, Cultured
17.
Bioinformatics ; 26(7): 976-8, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20179076

ABSTRACT

SUMMARY: The semantic comparisons of Gene Ontology (GO) annotations provide quantitative ways to compute similarities between genes and gene groups, and have became important basis for many bioinformatics analysis approaches. GOSemSim is an R package for semantic similarity computation among GO terms, sets of GO terms, gene products and gene clusters. Four information content (IC)- and a graph-based methods are implemented in the GOSemSim package, multiple species including human, rat, mouse, fly and yeast are also supported. The functions provided by the GOSemSim offer flexibility for applications, and can be easily integrated into high-throughput analysis pipelines. AVAILABILITY: GOSemSim is released under the GNU General Public License within Bioconductor project, and freely available at http://bioconductor.org/packages/2.6/bioc/html/GOSemSim.html.


Subject(s)
Computational Biology/methods , Genes , Software , Algorithms , Animals , Humans , Natural Language Processing , Proteins/genetics , Semantics , Terminology as Topic , Vocabulary, Controlled
18.
Thromb Haemost ; 102(6): 1194-203, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19967151

ABSTRACT

To resolve the therapeutic dilemma between efficacy of thrombolysis and bleeding risk associated with the use of a combination of thrombolytic and anticoagulant treatments, we created a fusion protein. Staphylokinase was fused to the N-terminus of hirudin using thrombin recognition sequence as linker peptide, resulting in a fusion protein STH. We hypothesised that STH would be cleaved by thrombin at the thrombus site, releasing staphylokinase and hirudin to perform bifunctionally, and attenuating bleeding risk. SDS-PAGE and Western blot analyses indicated that the linker peptide could be specially recognised and cleaved by thrombin. Amidolytic and thromboelastogram assays showed that the N-terminus of hirudin in STH was blocked by staphylokinase and linker peptide, impeding hirudin's anticoagulant activity. Once cleaved, STH displayed 35.7% of the anticoagulant activity of equimolar hirudin and exhibited anticoagulant effects in the fibrin clot lysis assay. Thrombin-binding and fibrin clot lysis assays showed that the C-terminus of hirudin retained its high affinity for thrombin. Moreover, STH showed improved thrombolytic effects and a lower bleeding risk in animals. Thus, STH may have the capacity to perform bifunctionally and release anticoagulant activity in a thrombus-targeted manner in vivo, which may reduce the bleeding risk that often accompanies high thrombolytic efficacy in the treatment of thromboembolic diseases.


Subject(s)
Fibrinolytic Agents/pharmacology , Animals , Base Sequence , Cattle , DNA Primers/genetics , Disease Models, Animal , Fibrinolytic Agents/chemistry , Hemorrhage/prevention & control , Hirudins/chemistry , Hirudins/genetics , Hirudins/pharmacology , Humans , In Vitro Techniques , Metalloendopeptidases/chemistry , Metalloendopeptidases/genetics , Metalloendopeptidases/pharmacology , Mice , Plasminogen/metabolism , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/pharmacology , Thrombelastography , Thrombin/metabolism , Thromboembolism/drug therapy , Thrombolytic Therapy/adverse effects , Thrombolytic Therapy/methods , Vena Cava, Inferior
19.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 17(4): 986-9, 2009 Aug.
Article in Chinese | MEDLINE | ID: mdl-19698244

ABSTRACT

This study was purposed to investigate the angiogenesis-promoting activities of human mesenchymal stem cells (hMSCs) modified by hepatocyte growth factor (HGF) and the underlying mechanisms. The hMSCs were transfected by recombinant adenoviral vector carrying human HGF gene and seeded onto the chicken chorioallantoic membrane. Three days later, the number of blood vessels was counted and their angiogenic response was compared with those of hMSCs of same generation, recombinant basic fibroblast growth factor (bFGF) and alpha-MEM as control. The expression levels of bFGF, VEGF, angiopoietin-1 and angiopoietin-2 were evaluated by RT-PCR assay. The results showed that gene-modified hMSCs exhibited greatest activity to promote angiogenesis while the angiogenic response was nearly same between groups treated by hMSCs and bFGF, all of which were significantly higher than that observed in control (p < 0.01). RT-PCR analysis revealed that hMSCs constitutively expressed multiple angiogenesis-associated growth factors and their levels seemed up-regulated by HGF gene transfer. It is concluded that HGF gene-modified hMSCs show a potent angiogenesis-promoting function and may be useful in the treatment of ischemic disorders.


Subject(s)
Hepatocyte Growth Factor/genetics , Mesenchymal Stem Cells , Neovascularization, Physiologic/genetics , Animals , Cells, Cultured , Chick Embryo , Chickens , Humans , Transfection
20.
Acta Pharmacol Sin ; 30(5): 617-27, 2009 May.
Article in English | MEDLINE | ID: mdl-19363518

ABSTRACT

AIM: Oncolytic adenovirus, also called conditionally replicating adenovirus (CRAD), can selectively propagate in tumor cells and cause cell lysis. The released viral progeny can infect neighboring cancer cells, initiating a cascade that can lead to the ultimate destruction of the tumor. Suicide gene therapy using herpes simplex virus thymidine kinase (HSV-TK) and ganciclovir (GCV) offers a potential treatment strategy for cancer and is undergoing preclinical trials for a variety of tumors. We hypothesized that HSV-TK gene therapy combined with oncolytic adenoviral therapy would have an enhanced effect compared with the individual effects of the therapies and is a potential novel therapeutic strategy to treat liver cancer. METHODS: To address our hypothesis, a novel CRAD was created, which consisted of a telomerase-dependent oncolytic adenovirus engineered to express E1A and HSV-TK genes (Ad-ETK). The combined effect of Ad-ETK and GCV was assessed both in vitro and in vivo in nude mice bearing HepG2 cell-derived tumors. Expression of the therapeutic genes by the transduced tumor cells was analyzed by RT-PCR and Western blotting. RESULTS: We confirmed that Ad-ETK had antitumorigenic effects on human hepatocellular carcinoma (HCC) both in vitro and in vivo, and the TK/GCV system enhanced oncolytic adenoviral therapy. We confirmed that both E1A and HSV-TK genes were expressed in vivo. CONCLUSION: The Ad-ETK construct should provide a relatively safe and selective approach to killing cancer cells and should be investigated as an adjuvant therapy for hepatocellular carcinoma.


Subject(s)
Adenoviridae/genetics , Carcinoma, Hepatocellular/therapy , Ganciclovir/administration & dosage , Genetic Therapy/methods , Liver Neoplasms, Experimental/therapy , Oncolytic Virotherapy/methods , Thymidine Kinase/genetics , Animals , Cell Line, Tumor , Combined Modality Therapy , Genes, Transgenic, Suicide , Humans , Mice , Mice, Nude , Neoplasm Transplantation , Simplexvirus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...