Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Neurotox Res ; 39(4): 1274-1284, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33939098

ABSTRACT

Thimerosal (THIM) induces neurotoxic changes including neuronal death and releases apoptosis inducing factors from mitochondria to cytosol. THIM alters the expression level of factors involved in apoptosis. On the other hand, the anti-apoptotic effects of exercise have been reported. In this study, we aimed to discover the effect of three protocols of treadmill exercise on the expression level of mitochondrial transcription factor A (TFAM), peroxisome proliferator-activated receptor γ coactivator-1α (PGC-1α), BCL-2-associated death (BAD), BCL-2-associated X (BAX), BCL-XL, and BCL-2 (a pro-survival BCL-2 protein) in the hippocampus of control and THIM-exposed rats. Male Wistar rats were used in this research. Real-time PCR was applied to assess genes expression. The results showed that THIM increased the expression of pro-apoptotic factors (BAD and BAX), decreased the expression of anti-apoptotic factors (BCL-2 and BCL-XL), and decreased the expression of factors involved in mitochondrial biogenesis (TFAM and PGC-1α). Treadmill exercise protocols reversed the effect of THIM on all genes. In addition, treadmill exercise protocols decreased the expression of BAD and BAX, increased the expression of BCL-2, and increased the expression of TFAM and PGC-1α in control rats. In conclusion, THIM induced a pro-apoptotic effect and disturbed mitochondrial biogenesis and stability, whereas treadmill exercise reversed these effects.


Subject(s)
Exercise Test/methods , Hippocampus/drug effects , Physical Conditioning, Animal/physiology , Thimerosal/toxicity , Animals , Gene Expression , Hippocampus/metabolism , Male , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/biosynthesis , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Physical Conditioning, Animal/methods , Preservatives, Pharmaceutical/toxicity , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Rats, Wistar , Transcription Factors/biosynthesis , Transcription Factors/genetics , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/genetics , bcl-X Protein/biosynthesis , bcl-X Protein/genetics
2.
Protein Expr Purif ; 175: 105707, 2020 11.
Article in English | MEDLINE | ID: mdl-32682909

ABSTRACT

14-3-3 protein isoforms regulate multiple processes in eukaryotes, including apoptosis and cell division. 14-3-3 proteins preferentially recognize phosphorylated unstructured motifs, justifying the protein-peptide binding approach to study 14-3-3/phosphotarget complexes. Tethering of human 14-3-3σ with partner phosphopeptides via a short linker has provided structural information equivalent to the use of synthetic phosphopeptides, simultaneously facilitating purification and crystallization. Nevertheless, the broader applicability to other 14-3-3 isoforms and phosphopeptides was unclear. Here, we designed a novel 14-3-3ζ chimera with a conserved phosphopeptide from BAD, whose complex with 14-3-3 is a gatekeeper of apoptosis regulation. The chimera could be bacterially expressed and purified without affinity tags. Co-expressed PKA efficiently phosphorylates BAD within the chimera and blocks its interaction with a known 14-3-3 phosphotarget, suggesting occupation of the 14-3-3 grooves by the tethered BAD phosphopeptide. Efficient crystallization of the engineered protein suggests suitability of the "chimeric" approach for studies of other relevant 14-3-3 complexes.


Subject(s)
14-3-3 Proteins , Protein Engineering , Recombinant Fusion Proteins , bcl-Associated Death Protein , 14-3-3 Proteins/biosynthesis , 14-3-3 Proteins/chemistry , 14-3-3 Proteins/genetics , 14-3-3 Proteins/isolation & purification , Crystallography, X-Ray , Humans , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/isolation & purification , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/chemistry , bcl-Associated Death Protein/genetics , bcl-Associated Death Protein/isolation & purification
3.
Bull Exp Biol Med ; 168(2): 205-209, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31784847

ABSTRACT

The expression of molecular and cellular regulators of apoptosis (proapoptotic protein Bad and antiapoptotic protein Bcl-2) was measured in the follicular apparatus of rat ovaries during the recovery period (days 7 and 14) after hyperthermia (up to rectal temperature 43.5°C). The Bcl-2/Bad index was calculated. The expression of Bcl-2 in the follicular apparatus of rat ovaries increased on day 7 after the exposure. The Bcl-2/Bad index also increased, which suggests that the development of apoptosis by the mitochondrial pathway in follicles was limited at this term after hyperthermia. On day 14 after hyperthermia, the area of immunohistochemical staining for the antiapoptotic protein Bcl-2 significantly decreased in cells of the ovarian follicular epithelium, but the expression of the proapoptotic protein Bad significantly increased; these changes led to a decrease in Bcl-2/Bad index, which attested to weakening of the antiapoptotic defense and activation of oocyte apoptosis by the mitochondrial pathway.


Subject(s)
Hypothermia/pathology , Ovarian Follicle/metabolism , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Recovery of Function/physiology , bcl-Associated Death Protein/biosynthesis , Animals , Apoptosis/physiology , Female , Mitochondria/metabolism , Rats , Rats, Wistar
4.
Int J Mol Med ; 43(2): 693-700, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30483729

ABSTRACT

Oxidative stress serves an important role in hypertensive brain damage. Peroxisome proliferator­activated receptor γ (PPAR­Î³) agonists possess antioxidative and anti­apoptotic effects. The present study verified the possibility that rosiglitazone serves a neuroprotective role by alleviating oxidative stress and cell apoptosis in the hippocampi of spontaneously hypertensive rats (SHRs). SHRs and age­matched Wistar­Kyoto (WKY; both 56 weeks old) rats received gavage administration of vehicle or rosiglitazone (5 mg/kg/day) for eight weeks. Systolic blood pressure (SBP) was measured by the indirect tail­cuff method. The expression ratio of activated astrocytes was analyzed by glial fibrillary acidic protein immunohistochemistry. PPAR­Î³, inducible nitric oxide synthase (iNOS), gp47phox, B­cell lymphoma 2 (Bcl­2), Bcl­2­associated X protein (Bax) and caspase­3 expression were investigated by quantitative polymerase chain reaction and western blot analysis. The number of apoptotic cells in the hippocampus of four groups was detected using the terminal deoxynucleotidyl transferase­mediated dUTP end­labeling (TUNEL) method. Compared with the WKY group, the SHR group exhibited decreased Bcl­2 and PPAR­Î³ expression, increased SBP, increased ratio of activated astrocytes and TUNEL­positive cells, increased expression of iNOS, gp47phox, caspase­3 and Bax. Rosiglitazone administration increased Bcl­2 and PPAR­Î³ expression, decreased the ratio of activated astrocytes and TUNEL­positive cells, decreased iNOS, gp47phox, caspase­3 and Bax expression in the hippocampi of SHRs. However, rosiglitazone did not significantly decreased SBP in the SHR group. Therefore, rosiglitazone exerts neuroprotective effect through antioxidative and anti­apoptotic pathways, which was independent of blood pressure control.


Subject(s)
Apoptosis/drug effects , Hippocampus/drug effects , Hypertension/physiopathology , Hypoglycemic Agents/administration & dosage , Rosiglitazone/administration & dosage , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/pathology , Blood Pressure/drug effects , Caspase 3/biosynthesis , Hippocampus/metabolism , Hippocampus/physiopathology , Hypertension/metabolism , Hypertension/pathology , Male , Oxidative Stress/drug effects , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Reactive Oxygen Species/metabolism , bcl-2-Associated X Protein/biosynthesis , bcl-Associated Death Protein/biosynthesis
5.
Parasite Immunol ; 40(6): e12531, 2018 06.
Article in English | MEDLINE | ID: mdl-29633291

ABSTRACT

Benign prostatic hyperplasia (BPH) is characterized by the proliferation of stromal and epithelial cell types in the prostate, and interactions between the two types of cells. We demonstrated previously that proliferation of prostate stromal cells was induced by BPH epithelial cells in response to Trichomonas vaginalis (Tv) infection via crosstalk with mast cells. In this study, we investigated whether IL-6 released by the proliferating stromal cells in turn induce the BPH epithelial cells to multiply. When culture supernatants of the proliferating prostate stromal cells were added to BPH epithelial cells, the latter multiplied, and expression of cyclin D1, FGF2 and Bcl-2 increased. Blocking the IL-6 signalling pathway with anti-IL-6R antibody or JAK1/2 inhibitor inhibited the proliferation of the BPH epithelial cells and reduced the expression of IL-6, IL-6R and STAT3. Also, epithelial-mesenchymal transition was detected in the proliferating BPH epithelial cells. In conclusion, IL-6 released from proliferating prostate stromal cells induced by BPH epithelial cells infected with Tv in turn induces multiplication of the BPH epithelial cells. This result provides first evidence that the inflammatory microenvironment of prostate stromal cells resulting from Tv infection induces the proliferation of prostate epithelial cells by stromal-epithelial interaction.


Subject(s)
Cell Proliferation/physiology , Epithelial Cells/metabolism , Interleukin-6/metabolism , Prostatic Hyperplasia/pathology , Stromal Cells/metabolism , Trichomonas Infections/pathology , Cyclin D1/biosynthesis , Epithelial-Mesenchymal Transition/physiology , Fibroblast Growth Factor 2/biosynthesis , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/biosynthesis , Male , Mast Cells/metabolism , Prostate/cytology , STAT3 Transcription Factor/biosynthesis , Signal Transduction , Trichomonas vaginalis/immunology , bcl-Associated Death Protein/biosynthesis
6.
PLoS Genet ; 13(7): e1006913, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28753606

ABSTRACT

Genes and pathways that allow cells to cope with oncogene-induced stress represent selective cancer therapeutic targets that remain largely undiscovered. In this study, we identify a RhoJ signaling pathway that is a selective therapeutic target for BRAF mutant cells. RhoJ deletion in BRAF mutant melanocytes modulates the expression of the pro-apoptotic protein BAD as well as genes involved in cellular metabolism, impairing nevus formation, cellular transformation, and metastasis. Short-term treatment of nascent melanoma tumors with PAK inhibitors that block RhoJ signaling halts the growth of BRAF mutant melanoma tumors in vivo and induces apoptosis in melanoma cells in vitro via a BAD-dependent mechanism. As up to 50% of BRAF mutant human melanomas express high levels of RhoJ, these studies nominate the RhoJ-BAD signaling network as a therapeutic vulnerability for fledgling BRAF mutant human tumors.


Subject(s)
Melanoma/genetics , Proto-Oncogene Proteins B-raf/genetics , bcl-Associated Death Protein/biosynthesis , p21-Activated Kinases/genetics , rho GTP-Binding Proteins/genetics , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Enzyme Inhibitors/administration & dosage , Gene Expression Regulation, Neoplastic/drug effects , Humans , Melanocytes/drug effects , Melanocytes/pathology , Melanoma/drug therapy , Melanoma/pathology , Mutation , Neoplasm Metastasis , Nevus/genetics , Nevus/pathology , Signal Transduction/drug effects , bcl-Associated Death Protein/genetics , p21-Activated Kinases/antagonists & inhibitors
7.
Biochem Biophys Res Commun ; 489(2): 179-186, 2017 07 22.
Article in English | MEDLINE | ID: mdl-28549584

ABSTRACT

OBJECTIVE: Previous studies found bone resorption and chondrocytes loss in mouse models of mid-palatal suture when given continuous compressive force, although chondrocytes response remained unknown. Herein, we design this study to determine how continuous compression force induces chondrocytes apoptosis. METHODS: Thirty C57BL/6 male mice (aged 6 weeks) were randomly assigned into controls (not ligated to a spring), blank controls (ligated with no compression) and the compression group (ligated with 20-g compression). After 4 d, palatal tissues were sampled and stained by TB and safranin-O. Tunel staining measured the percentage of apoptotic chondrocytes, and immunohistochemistry was performed to label apoptosis-associated proteins (e.g., Bcl-2, Bcl-xl, Bax, Bak, Bid, Bad, caspase-3, caspase-8 and caspase-9). Intergroup comparison was made by the rank sum test, and P < 0.05 was defined as statistical significance. RESULTS: After 7d of induction, TB and safranin-O staining revealed that the cartilage area in the compression group was significantly decreased, while the control group remained largely unaltered. Tunel staining showed that apoptotic cell numbers in the mid-palatal suture were significantly higher than the control group. Immunohistochemistry showed that mice in the compression group had significantly increased expression of caspase-3, caspase-9, Bad, Bak, Bax and Bid; However, caspase-8 remained unaltered. No expression of Bcl-2 and Bcl-xl was detected. CONCLUSIONS: Continuous compression force induces chondrocytes apoptosis in the mid-palatal suture. This process might be associated with the mitochondrial pathway.


Subject(s)
Apoptosis , Chondrocytes/metabolism , Chondrocytes/pathology , Pressure/adverse effects , Sutures/adverse effects , Up-Regulation , Animals , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Caspase 3/biosynthesis , Caspase 9/biosynthesis , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Phenazines , Tolonium Chloride , bcl-2 Homologous Antagonist-Killer Protein/biosynthesis , bcl-2-Associated X Protein/biosynthesis , bcl-Associated Death Protein/biosynthesis
8.
Oncol Rep ; 36(5): 2793-2799, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666310

ABSTRACT

Cyclin Y (CCNY) is a newly identified PFTK1 interacting protein and has been found to be associated with the proliferation and tumorigenesis of human non-small cell lung cancer. In the present study, we analyzed the expression levels of CCNY in 65 cases of breast cancer (BC) tissues and in four BC cell lines, BT-474, MDA-MB-231, T-47D and MCF-7. Lentivirus-mediated short hairpin RNA (shRNA) was employed to knock down CCNY expression in MCF-7 and MDA-MB-231 cells. The effects of CCNY depletion on cell growth were examined by MTT, colony formation and flow cytometry assays. The results showed that immunohistochemical expression of CCNY in tumor tissues is stronger than that in normal tissues. CCNY was also expressed in all four BC cells. The knockdown of CCNY resulted in a significant reduction in cell proliferation and colony formation ability. Cell cycle analysis showed that CCNY knockdown arrested MDA-MB­231 cells in the G0/G1 phase. Furthermore, depletion of CCNY inhibited BC cell growth via the activation of Bad and GSK3ß, as well as cleavages of PARP and caspase-3 in a p53-dependent manner. Therefore, we believe that CCNY has biological effect in BC development, and its inhibition via an RNA interference lentiviral system may provide a therapeutic option for BC.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Cell Proliferation/genetics , Cyclins/genetics , Apoptosis/genetics , Biomarkers, Tumor/therapeutic use , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Caspase 3/biosynthesis , Caspase 3/genetics , Cyclins/therapeutic use , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/biosynthesis , Glycogen Synthase Kinase 3 beta/genetics , Humans , Lentivirus/genetics , MCF-7 Cells , Poly (ADP-Ribose) Polymerase-1/biosynthesis , Poly (ADP-Ribose) Polymerase-1/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Tumor Suppressor Protein p53/genetics , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/genetics
9.
Int J Oncol ; 49(2): 793-802, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27279602

ABSTRACT

It has been reported that lower doses of the opioid antagonist naltrexone are able to reduce tumour growth by interfering with cell signalling as well as by modifying the immune system. We have evaluated the gene expression profile of a cancer cell line after treatment with low-dose naltrexone (LDN), and assessed the effect that adapting treatment schedules with LDN may have on enhancing efficacy. LDN had a selective impact on genes involved with cell cycle regulation and immune modulation. Similarly, the pro-apoptotic genes BAD and BIK1 were increased only after LDN. Continuous treatment with LDN had little effect on growth in different cell lines; however, altering the treatment schedule to include a phase of culture in the absence of drug following an initial round of LDN treatment, resulted in enhanced cell killing. Furthermore, cells pre-treated with LDN were more sensitive to the cytotoxic effects of a number of common chemotherapy agents. For example, priming HCT116 with LDN before treatment with oxaliplatin significantly increased cell killing to 49±7.0 vs. 14±2.4% in cultures where priming was not used. Interestingly, priming with NTX before oxaliplatin resulted in just 32±1.8% cell killing. Our data support further the idea that LDN possesses anticancer activity, which can be improved by modifying the treatment schedule.


Subject(s)
Colorectal Neoplasms/drug therapy , Dose-Response Relationship, Drug , Naltrexone/administration & dosage , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , HCT116 Cells , Humans , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Mitochondrial Proteins , Organoplatinum Compounds/administration & dosage , Oxaliplatin , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/genetics
10.
Oncol Rep ; 36(1): 99-107, 2016 Jul.
Article in English | MEDLINE | ID: mdl-27122127

ABSTRACT

The Korean prostrate spurge Euphorbia supina (Euphorbiaceae family) has been used as a folk medicine in Korea against a variety of ailments such as bronchitis, hemorrhage, jaundice and multiple gastrointestinal diseases. Polyphenols from Korean E. supina (PES) which include quercetin and kaempferol derivatives have anticancer properties. Hence, we investigated the anticancer effects of PES on U937 human leukemic cells. Firstly, PES significantly inhibited the proliferation of U937 cells in a dose-dependent manner. PES induced accumulation of the sub-G1 DNA content (apoptotic cell population), apoptotic bodies and chromatin condensation and DNA fragmentation in the U937 cells. PES also induced activation of caspase-3, -8 and -9, subsequent cleavage of PARP, and significantly suppressed XIAP, cIAP-1 and cIAP-2 in a dose-dependent manner. Furthermore, PES activated Bid, and induced the loss of mitochondrial membrane potential (MMP, ΔΨm) along with upregulation of pro-apoptotic proteins (Bax and Bad), and downregulation of anti-apoptotic proteins (Bcl-2 and Bcl-xL) and cytochrome c release. The Fas receptor was upregulated by PES in a dose-dependent manner, suggesting that the extrinsic pathway was also involved in the PES-induced apoptosis. Moreover, the PES-induced apoptosis was at least in part associated with extracellular signal-regulated kinase (ERK) activation in the U937 human leukemic cells. This study provides evidence that PES may be useful in the treatment of leukemia.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Euphorbia/chemistry , Extracellular Signal-Regulated MAP Kinases/metabolism , Leukemia/drug therapy , Phytochemicals/pharmacology , Polyphenols/pharmacology , Antineoplastic Agents/isolation & purification , BH3 Interacting Domain Death Agonist Protein/metabolism , Baculoviral IAP Repeat-Containing 3 Protein , Caspase 3/metabolism , Caspase 8/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cytochromes c/metabolism , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Inhibitor of Apoptosis Proteins/metabolism , Leukemia/pathology , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Phosphoinositide-3 Kinase Inhibitors , Poly(ADP-ribose) Polymerases/metabolism , Polyphenols/isolation & purification , Republic of Korea , U937 Cells , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , bcl-2-Associated X Protein/biosynthesis , bcl-Associated Death Protein/biosynthesis
11.
Oncotarget ; 7(14): 17290-300, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26981780

ABSTRACT

We determined whether the myelofibrosis drug ruxolitinib, an inhibitor of Janus kinases 1/2 (JAK1 and JAK2), could interact with the multiple sclerosis drug dimethyl-fumarate (DMF) to kill tumor cells; studies used the in vivo active form of the drug, mono-methyl fumarate (MMF). Ruxolitinib interacted with MMF to kill brain, breast, lung and ovarian cancer cells, and enhanced the lethality of standard of care therapies such as paclitaxel and temozolomide. MMF also interacted with other FDA approved drugs to kill tumor cells including Celebrex® and Gilenya®. The combination of [ruxolitinib + MMF] inactivated ERK1/2, AKT, STAT3 and STAT5; reduced expression of MCL-1, BCL-XL, SOD2 and TRX; increased BIM expression; decreased BAD S112 S136 phosphorylation; and enhanced pro-caspase 3 cleavage. Expression of activated forms of STAT3, MEK1 or AKT each significantly reduced drug combination lethality; prevented BAD S112 S136 dephosphorylation and decreased BIM expression; and preserved TRX, SOD2, MCL-1 and BCL-XL expression. The drug combination increased the levels of reactive oxygen species in cells, and over-expression of TRX or SOD2 prevented drug combination tumor cell killing. Over-expression of BCL-XL or knock down of BAX, BIM, BAD or apoptosis inducing factor (AIF) protected tumor cells. The drug combination increased AIF : HSP70 co-localization in the cytosol but this event did not prevent AIF : eIF3A association in the nucleus.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Dimethyl Fumarate/pharmacology , Drug Synergism , Mitochondria/drug effects , Neoplasms/drug therapy , Pyrazoles/pharmacology , Animals , Bcl-2-Like Protein 11/biosynthesis , Cell Line, Tumor , Dimethyl Fumarate/administration & dosage , Humans , Janus Kinase 1/antagonists & inhibitors , Mice , Mitochondria/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Nitriles , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrazoles/administration & dosage , Pyrimidines , Rats , Reactive Oxygen Species/metabolism , Signal Transduction , bcl-Associated Death Protein/biosynthesis
12.
Mol Med Rep ; 13(2): 1593-601, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26717963

ABSTRACT

Cardiomyocyte apoptosis is a major event in the pathogenesis of diabetic cardiomyopathy. Currently, no single effective treatment for diabetic cardiomyopathy exists. The present study investigated whether advanced oxidative protein products (AOPPs) have a detrimental role in the survival of cardiomyocytes and if glucagon-like peptide-1 (GLP-1) exerts a cardioprotective effect under these circumstances. The present study also aimed to determine the underlying mechanisms. H9c2 cells were exposed to increasing concentrations of AOPPs in the presence or absence of GLP-1, and the viability and apoptotic rate were detected using a cell counting kit-8 assay and flow cytometry, respectively. In addition, a phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) inhibitor, LY294002, was employed to illustrate the mechanism of the antiapoptotic effect of GLP-1. The expression levels of the apoptotic-associated proteins, Akt, B-cell lymphoma (Bcl)-2, Bcl-2-associated death promoter (Bad), Bcl-2-associated X protein (Bax) and caspase-3 were measured by western blotting. It was revealed that GLP-1 significantly attenuated AOPP-induced cell toxicity and apoptosis. AOPPs inactivated the phosphorylation of Akt, reduced the phosphorylation of Bad, decreased the expression of Bcl-2, increased the expression of Bax and the activation of caspase-3 in H9c2 cells. GLP-1 reversed the above changes induced by AOPPs and the protective effects of GLP-1 were abolished by the PI3K inhibitor, LY294002. In conclusion, the present data suggested that GLP-1 protected cardiomyocytes against AOPP-induced apoptosis, predominantly via the PI3K/Akt/Bad pathway. These results provided a conceivable mechanism for the development of diabetic cardiomyopathy and rendered a novel application of GLP-1 exerting favorable cardiac effects for the treatment of diabetic cardiomyopathy.


Subject(s)
Advanced Oxidation Protein Products/metabolism , Diabetic Cardiomyopathies/genetics , Glucagon-Like Peptide 1/metabolism , Myocytes, Cardiac/metabolism , Advanced Oxidation Protein Products/genetics , Animals , Apoptosis/drug effects , Chromones/administration & dosage , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/pathology , Glucagon-Like Peptide 1/genetics , Humans , Morpholines/administration & dosage , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/physiology , Oncogene Protein v-akt/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphoinositide-3 Kinase Inhibitors , Rats , bcl-Associated Death Protein/biosynthesis
13.
Growth Factors ; 33(5-6): 319-25, 2015.
Article in English | MEDLINE | ID: mdl-26567452

ABSTRACT

Mesenchymal stem cells (MSC) secrete a great variety of cytokines that have beneficial paracrine actions. Hepatocyte growth factor (HGF) promotes proliferation in several cell types. The aim of the present study was to investigate the protective effect of HGF gene-transfected MSC (HGF-MSC) in acetaminophen (AAP)-treated hepatocytes. We transfected the HGF gene into MSCs and confirmed HGF expression by RT-PCR and western blot. The concentration of HGF in HGF-MSC conditioned media (HGFCM) was upregulated compared with that in control MSCCM samples. Cell viability was increased in HGFCM-treated hepatocytes. Expression of Mcl-1, an anti-apoptosis protein, was increased and expression of pro-apoptosis proteins (Bad, Bik and Bid) was decreased in HGFCM-treated hepatocytes. HGF-MSC had protective effects on AAP-induced hepatocyte damage by enhancing proliferation. These results suggest that HGF-expressing MSCs may provide regenerative potential for liver cell damage.


Subject(s)
Acetaminophen/adverse effects , Analgesics, Non-Narcotic/adverse effects , Culture Media, Conditioned/pharmacology , Hepatocyte Growth Factor/metabolism , Hepatocytes/metabolism , Mesenchymal Stem Cells/metabolism , Acetaminophen/pharmacology , Analgesics, Non-Narcotic/pharmacology , Animals , Apoptosis Regulatory Proteins/biosynthesis , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , DNA Fragmentation/drug effects , Gene Expression , Hepatocyte Growth Factor/biosynthesis , Hepatocyte Growth Factor/genetics , Humans , L-Lactate Dehydrogenase/metabolism , Liver/injuries , Membrane Proteins/biosynthesis , Mice , Mitochondrial Proteins , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Transfection , bcl-Associated Death Protein/biosynthesis
14.
Tumour Biol ; 36(6): 4367-76, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25618598

ABSTRACT

Glucose-regulated protein 78 (GRP78) is expressed as part of the molecular response to endoplasmic reticulum (ER) stress and mediates protein folding within the cell. GRP78 is also an important biomarker of cancer progression and the therapeutic response of patients with different cancer types. However, the role of GRP78 in the cytotoxic effect of 17-DMAG in colon cancer cells remains unclear. GRP78 expression was knocked down by small interfering RNA (siRNA). The anticancer effects of 17-DMAG were assessed by an 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay, a flow cytometric cell-cycle analysis, and an Annexin V-propidium iodide (PI) apoptotic assay. We found that HT-29 cells expressed a lower level of GRP78 compared with DLD-1 cells. The MTT assay revealed that HT-29 cells were more sensitive to 17-DMAG treatment than DLD-1 cells. GRP78 knock down (GRP78KD) cells demonstrated an increased sensitivity to 17-DMAG treatment compared with the scrambled control cells. Based on the cell-cycle analysis and Annexin V-PI apoptotic assay, apoptosis dramatically increased in GRP78KD cells compared with scrambled control DLD-1 cells after these cells were treated with 17-DMAG. Finally, we observed a decrease in the level of Bcl-2 and an increase in the levels of Bad and Bax in GRP78KD cells treated with 17-DMAG. These results are consistent with an increased sensitivity to 17-DMAG after knock down of GRP78. The level of GRP78 expression may determine the therapeutic efficacy of 17-DMAG against colon cancer cells.


Subject(s)
Benzoquinones/administration & dosage , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Heat-Shock Proteins/genetics , Lactams, Macrocyclic/administration & dosage , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Endoplasmic Reticulum Chaperone BiP , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Gene Silencing , HSP90 Heat-Shock Proteins/biosynthesis , HT29 Cells , Heat-Shock Proteins/metabolism , Humans , Proto-Oncogene Proteins c-bcl-2/biosynthesis , bcl-2-Associated X Protein/biosynthesis , bcl-Associated Death Protein/biosynthesis
15.
Oncol Rep ; 33(3): 1358-64, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25592110

ABSTRACT

Recombinant hirudin (rH) is a highly potent and specific inhibitor of thrombin, and has been shown to inhibit the growth and metastasis of several types of cancers in experimental tumor models. The objective of this study was to evaluate the antitumor effects and explore the underlying mechanisms of rH in Hep-2 human laryngeal carcinoma (LC) cells. Hep-2 cells were treated with various concentrations of rH for 24 h. The cell viability was evaluated by a water-soluble tetrazolium salt (WST) assay. The adhesion ability of the cells was evaluated by cell adhesion to fibronectin. Cell migration and invasion were measured with the Boyden chamber assay. Cell apoptosis was detected by Hoechst 33324 fluorescence staining. A chicken chorioallantoic membrane (CAM) assay was used to assess the effects of rH on angiogenesis in vivo. Western blotting was used to detect the expression levels of vascular endothelial growth factor receptor (VEGF-R), focal adhesion kinase (FAK), Bcl-2-associated agonist of cell death (Bad) and B-cell CLL/lymphoma 2 (Bcl-2) proteins. rH significantly inhibited the cell viability and induced apoptosis in LC Hep-2 cells in a dose-dependent manner, as compared with phosphate-buffered saline (PBS) as control. These results were accompanied by a decrease in the anti-apoptotic protein Bcl-2 and an increase in the pro-apoptotic protein Bad. Moreover, rH dose-dependently inhibited the adhesion, migration and invasion of the Hep-2 cells, compared to the vehicle PBS. In addition, rH robustly suppressed angiogenesis in the CAM assay. Importantly, the expression of adhesion and angiogenesis-associated proteins FAK and VEGF-R was significantly downregulated by rH in a dose-dependent manner. The present findings demonstrate that rH exerts antitumor effects in Hep-2 human laryngeal cancer cells via multiple mechanisms and suggests that targeting thrombin by rH is a potential strategy for the treatment of LC.


Subject(s)
Antithrombins/pharmacology , Hirudins/pharmacology , Laryngeal Neoplasms/drug therapy , Laryngeal Neoplasms/pathology , Recombinant Proteins/pharmacology , Animals , Apoptosis/drug effects , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Chick Embryo , Fibronectins/metabolism , Focal Adhesion Kinase 1/biosynthesis , Humans , Neoplasm Invasiveness/pathology , Neovascularization, Pathologic/chemically induced , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, Vascular Endothelial Growth Factor/biosynthesis , Thrombin/antagonists & inhibitors , bcl-Associated Death Protein/biosynthesis
16.
Arch Bronconeumol ; 51(7): 328-37, 2015 Jul.
Article in English, Spanish | MEDLINE | ID: mdl-25017817

ABSTRACT

INTRODUCTION: Emphysema has been associated with decreased VEGF and VEGFR-2 expression and the presence of high numbers of apoptotic alveolar cells. Keratinocyte growth factor stimulates VEGF synthesis which in turn confers normal lung structure maintenance via the Akt pathway. In this study the potential role of rHuKGF in the improvement of deregulated Akt mediated cell survival pathway in emphysematous mice was investigated. METHODS: Three experimental groups, i.e., emphysema, treatment and control groups, were prepared. Lungs of mice were treated on 3 occasions by oropharyngeal instillation of 10mg rHuKGF per kg body weight after induction of emphysema with porcine pancreatic elastase. Subsequently, lung tissues from mice were collected for histopathology and molecular biology studies. RESULTS AND DISCUSSION: Histopathology photomicrographs and destructive index analysis have shown that elastase-induced airspace enlargement and loss of alveoli recovered in the treatment group. rHuKGF stimulates VEGF production which in turn induces the Akt mediated cell survival pathway in emphysematous lungs. mRNA expression of VEGF, VEGFR, PI3K and Akt was significantly increased while Pten, Caspase-9 and Bad was notably decreased in treatment group when compared with emphysema group, being comparable with the control group. Moreover, VEGF protein expression was in accordance with that found for mRNA. CONCLUSION: Therapeutic rHuKGF supplementation improves the deregulated Akt pathway in emphysema, resulting in alveolar cell survival through activation of the endogenous VEGF-dependent cell survival pathway. Hence rHuKGF may prove to be a potential drug in the treatment of emphysema.


Subject(s)
Fibroblast Growth Factor 7/therapeutic use , Proto-Oncogene Proteins c-akt/physiology , Pulmonary Emphysema/drug therapy , Animals , Apoptosis/drug effects , Caspase 9/biosynthesis , Caspase 9/genetics , Cell Survival , Disease Models, Animal , Drug Evaluation, Preclinical , Fibroblast Growth Factor 7/pharmacology , Gene Expression Regulation/drug effects , Humans , Instillation, Drug , Lung/drug effects , Lung/pathology , Male , Mice , Mice, Inbred C57BL , PTEN Phosphohydrolase/biosynthesis , PTEN Phosphohydrolase/genetics , Pancreatic Elastase/toxicity , Phosphatidylinositol 3-Kinases/biosynthesis , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/enzymology , Pulmonary Emphysema/pathology , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Random Allocation , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor Receptor-2/biosynthesis , Vascular Endothelial Growth Factor Receptor-2/genetics , bcl-Associated Death Protein/biosynthesis , bcl-Associated Death Protein/genetics
17.
Int J Clin Exp Pathol ; 8(10): 13075-82, 2015.
Article in English | MEDLINE | ID: mdl-26722503

ABSTRACT

It is well known that lung cancer is the 1st leading cause of death worldwide. Many reports have demonstrated that Bad, the Bcl-xL/Bcl-2-associated death promoter plays a crucial role in the intrinsic apoptosis pathway. The aim of this study was to explore the expression of Bad and its clinical significance in small cell lung carcinoma (SCLC) By analyzing the expression of Bad in 147 SCLC patient specimen, we found that Bad expression was remarkably decreased in 55.8% (82/147) cases, compared with the neighboring non-tumor tissues. Further study showed that Bad expression was correlated with adverse clinical characters such as clinical stage (P = 0.001), tumor size (P = 0.036) and tumor recurrence (P = 0.030). Furthermore, the results of Kaplan-Meier analysis showed that low Bad expression was significantly correlated to overall survival (P < 0.0001) and disease-free survival (P = 0.017) of patients with SCLC. Moreover, multivariate analyses revealed that Bad was an independent indicator of overall survival in SCLC (hazard ration = 0.620, 95% confidence interval: 0.389-0.987, P < 0.001). In summary, we can conclude that patients with SCLC represent downregulation of Bad and the latter could be served as a useful biomarker for the outcomes of SCLC.


Subject(s)
Biomarkers, Tumor/analysis , Lung Neoplasms/pathology , Small Cell Lung Carcinoma/pathology , bcl-Associated Death Protein/biosynthesis , Adult , Aged , Aged, 80 and over , Blotting, Western , Disease-Free Survival , Down-Regulation , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Male , Middle Aged , Prognosis , Proportional Hazards Models , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/mortality , Young Adult , bcl-Associated Death Protein/analysis
19.
Neurosci Lett ; 579: 134-9, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-25043191

ABSTRACT

BAD-deficient mice and fasting have several common functional roles in seizures, beta-hydroxybutyrate (BHB) uptake in brain and alteration in counterregulatory hormonal regulation during hypoglycemia. Neuronal specific insulin receptor knockout (NIRKO) mice display impaired counterregulatory hormonal responses during hypoglycemia. In this study we investigated the fasting mediated expression of p-BAD(ser155) and p-AKT(ser473) in different regions of brain (prefrontal cortex, hippocampus, midbrain and hypothalamus). Fasting specifically increases p-BAD(ser155) and p-AKT(ser473) in prefrontal cortex and decreases in other regions of brain. Our results suggest that fasting may increase the uptake BHB by decreasing p-BAD(ser155) in the brain during hypoglycemia except prefrontal cortex and it uncovers specific functional area of p-BAD(ser155) and p-AKT(ser473) that may regulates counter regulatory hormonal response. Overall in support with previous findings, fasting mediated hypoglycemia activates prefrontal cortex insulin signaling which influences the hypothalamic paraventricular nucleus mediated activation of sympathoadrenal hormonal responses.


Subject(s)
Fasting/metabolism , Oncogene Protein v-akt/biosynthesis , Prefrontal Cortex/metabolism , bcl-Associated Death Protein/biosynthesis , Animals , Blood Glucose/metabolism , Brain Chemistry/genetics , Insulin/physiology , Liver/metabolism , Mice , Mice, Knockout , Oncogene Protein v-akt/genetics , Receptor, Insulin/genetics , Signal Transduction/genetics , bcl-Associated Death Protein/genetics
20.
PLoS One ; 9(5): e96843, 2014.
Article in English | MEDLINE | ID: mdl-24806443

ABSTRACT

Early embryonic lethality is common, particularly in dairy cattle. We made cattle embryos more sensitive to environmental stressors by raising the threshold of embryo survival signaling required to overcome the deleterious effects of overexpressing the proapoptotic protein BAD. Two primary fibroblast cell lines expressing BAD and exhibiting increased sensitivity to stress-induced apoptosis were used to generate transgenic Day 13/14 BAD embryos. Transgenic embryos were normal in terms of retrieval rates, average embryo length or expression levels of the trophectoderm marker ASCL2. However both lines of BAD-tg embryos lost the embryonic disc and thus the entire epiblast lineage at significantly greater frequencies than either co-transferrred IVP controls or LacZ-tg embryos. Embryos without epiblast still contained the second ICM-derived lineage, the hypopblast, albeit frequently in an impaired state, as shown by reduced expression of the hypoblast markers GATA4 and FIBRONECTIN. This indicates a gradient of sensitivity (epiblast > hypoblast > TE) to BAD overexpression. We postulate that the greater sensitivity of specifically the epiblast lineage that we have seen in our transgenic model, reflects an inherent greater susceptibility of this lineage to environmental stress and may underlie the epiblast-specific death seen in phantom pregnancies.


Subject(s)
Embryonic Development/genetics , Germ Layers/metabolism , bcl-Associated Death Protein/biosynthesis , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cattle , Embryo Culture Techniques , Embryo, Mammalian , Fibronectins/metabolism , GATA4 Transcription Factor/metabolism , Gene Expression Regulation, Developmental , bcl-Associated Death Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL