Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Cell Physiol ; 233(6): 5070-5077, 2018 06.
Article in English | MEDLINE | ID: mdl-29231977

ABSTRACT

Bcl-2 homologous antagonist/killer (BAK1) is a critical regulator of mitochondrial apoptosis. Although upregulation of BAK1 induces apoptosis has been established, the underlying molecular mechanism is far from clear. 4,4'-diisothiocyanostilbene-2,2'-disulfonic acid (DIDS), an organic anion used as a blocker of anion exchangers and chloride channels, has been proved to rescue cell apoptosis both in vitro and in vivo. However, whether DIDS can inhibit BAK1-induced mitochondrial apoptosis remains undefined. Thus, this study aimed to explore whether DIDS could protect BAK1-induced apoptosis through GSK3ß/ß-catenin signaling pathway. The results showed overexpression BAK1 in 293T cells induced mitochondrial apoptosis accompanied by increasing the expression levels of cleaved caspase-9, -3, poly (ADP-ribose) polymerase (PARP) and reducing the MMP. Furthermore, overexpression BAK1 decreased the expression levels of Ser9-GSK3ß and ß-catenin. In addition, lithium chloride (LiCl), an activator of Wnt/ß-catenin signaling pathway, markedly attenuated overexpression BAK1-induced mitochondrial apoptosis by restoring the expression levels of Ser9-GSK3ß and ß-catenin. Finally, DIDS absolutely abolished overexpression BAK1-mediated mitochondrial apoptosis through recovering the expression levels of Ser9-GSK3ß and ß-catenin. Taken together, our results reveal that DIDS blocks overexpression BAK1-induced mitochondrial apoptosis through GSK3ß/ß-catenin pathway.


Subject(s)
4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Apoptosis/drug effects , Glycogen Synthase Kinase 3 beta/metabolism , Mitochondria/drug effects , Wnt Signaling Pathway/drug effects , bcl-2 Homologous Antagonist-Killer Protein/metabolism , HEK293 Cells , Humans , Mitochondria/enzymology , Mitochondria/pathology , Phosphorylation , Up-Regulation , bcl-2 Homologous Antagonist-Killer Protein/genetics
2.
Molecules ; 22(4)2017 Apr 07.
Article in English | MEDLINE | ID: mdl-28387735

ABSTRACT

Olaquindox, a feed additive, has drawn public attention due to its potential mutagenicity, genotoxicity, hepatoxicity and nephrotoxicity. The purpose of this study was to investigate the role of tuberous sclerosis complex (TSC2) pathways in olaquindox-induced autophagy in human embryonic kidney 293 (HEK293) cells. The results revealed that olaquindox treatment reduced the cell viability of HEK293 cells and downregulated the expression of TSC2 in a dose- and time-dependent manner. Meanwhile, olaquindox treatment markedly induced the production of reactive oxygen species (ROS), cascaded to autophagy, oxidative stress, and apoptotic cell death, which was effectively eliminated by the antioxidant N-acetylcysteine (NAC). Furthermore, overexpression of TSC2 attenuated olaquindox-induced autophagy in contrast to inducing the production of ROS, oxidative stress and apoptosis. Consistently, knockdown of TSC2 upregulated autophagy, and decreased olaquindox-induced cell apoptosis. In conclusion, our findings indicate that TSC2 partly participates in olaquindox-induced autophagy, oxidative stress and apoptosis, and demonstrate that TSC2 has a negative regulation role in olaquindox-induced autophagy in HEK293 cells.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Quinoxalines/pharmacology , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/metabolism , Cell Survival/drug effects , Down-Regulation , HEK293 Cells , Humans , Oxidative Stress/drug effects , Tuberous Sclerosis Complex 2 Protein
3.
Molecules ; 22(1)2017 Jan 13.
Article in English | MEDLINE | ID: mdl-28098804

ABSTRACT

Olaquindox, a quinoxaline 1,4-dioxide derivative, is widely used as a feed additive in many countries. The potential genotoxicity of olaquindox, hence, is of concern. However, the proper mechanism of toxicity was unclear. The aim of the present study was to investigate the effect of growth arrest and DNA damage 45 alpha (GADD45a) on olaquindox-induced DNA damage and cell cycle arrest in HepG2 cells. The results showed that olaquindox could induce reactive oxygen species (ROS)-mediated DNA damage and S-phase arrest, where increases of GADD45a, cyclin A, Cdk 2, p21 and p53 protein expression, decrease of cyclin D1 and the activation of phosphorylation-c-Jun N-terminal kinases (p-JNK), phosphorylation-p38 (p-p38) and phosphorylation-extracellular signal-regulated kinases (p-ERK) were involved. However, GADD45a knockdown cells treated with olaquindox could significantly decrease cell viability, exacerbate DNA damage and increase S-phase arrest, associated with the marked activation of p-JNK, p-p38, but not p-ERK. Furthermore, SP600125 and SB203580 aggravated olaquindox-induced DNA damage and S-phase arrest, suppressed the expression of GADD45a. Taken together, these findings revealed that GADD45a played a protective role in olaquindox treatment and JNK/p38 pathways may partly contribute to GADD45a regulated olaquindox-induced DNA damage and S-phase arrest. Our findings increase the understanding on the molecular mechanisms of olaquindox.


Subject(s)
Anti-Bacterial Agents/pharmacology , Cell Cycle Proteins/genetics , Food Additives/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , MAP Kinase Kinase 4/genetics , Nuclear Proteins/genetics , Quinoxalines/pharmacology , p38 Mitogen-Activated Protein Kinases/genetics , Animal Feed/analysis , Anthracenes/pharmacology , Apoptosis/drug effects , Cell Cycle Proteins/agonists , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Comet Assay , DNA Fragmentation/drug effects , Hep G2 Cells , Humans , Imidazoles/pharmacology , MAP Kinase Kinase 4/metabolism , Nuclear Proteins/agonists , Nuclear Proteins/metabolism , Phosphorylation/drug effects , Pyridines/pharmacology , Reactive Oxygen Species/agonists , Reactive Oxygen Species/metabolism , S Phase/drug effects , Signal Transduction , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Cell Biol Toxicol ; 32(2): 141-52, 2016 04.
Article in English | MEDLINE | ID: mdl-27085326

ABSTRACT

The present study is undertaken to explore quinocetone-induced autophagy and its possible mechanism. Western blotting and green fluorescence protein (GFP)-LC3 vector transfection were performed to determine the ratio of LC3 conversion and its subcellular localization. Results revealed that the quinocetone induced autophagy in time- and dose-dependent manners. Besides, we tested the expressions of immunoglobulin heavy chain binding protein (BiP) and C/EBP homologous protein (CHOP) and the transcription of BiP, HerpUD, and sec24D by western blotting and RT-PCR, respectively. Results showed that quinocetone also induced endoplasmic reticulum (ER) stress during quinocetone-induced autophagy. Furthermore, we observed the cleavage of ATF6, the phosphorylation of MRLC, and the expression of death-associated protein kinase (DAPK1) by western blotting; the transcription of DAPK1 by RT-PCR; and the subcellular localization of ATF6 and mAtg9 by immunofluorescence. These results suggest that quinocetone stimulates the MRLC-mediated mAtg9 trafficking, which is critical for autophagosome formation, via the ATF6 upregulated expression of DAPK1. Last, we generated ATF6 and DAPK1 stable knockdown HepG2 cell lines and found that the conversion ratios of LC3 were decreased upon the treatment of quinocetone. Together, we propose that quinocetone induces autophagy through ER stress signaling pathway-induced cytoskeleton activation.


Subject(s)
Activating Transcription Factor 6/metabolism , Autophagy-Related Proteins/metabolism , Autophagy/drug effects , Death-Associated Protein Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Membrane Proteins/metabolism , Quinoxalines/pharmacology , Vesicular Transport Proteins/metabolism , Activating Transcription Factor 6/genetics , Apoptosis/drug effects , Autophagy-Related Proteins/genetics , Cell Movement/drug effects , Death-Associated Protein Kinases/genetics , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , HEK293 Cells , Heat-Shock Proteins/metabolism , Hep G2 Cells , Humans , Membrane Proteins/genetics , Phosphorylation , Signal Transduction , Transcriptional Activation/drug effects , Vesicular Transport Proteins/genetics
5.
Toxicol Mech Methods ; 26(1): 11-21, 2016.
Article in English | MEDLINE | ID: mdl-26446980

ABSTRACT

The study aims at evaluating the combination of the quinocetone and the ML-7 in preclinical hepatocellular carcinoma models. To this end, the effect of quinocetone and ML-7 on apoptosis induction and signaling pathways was analyzed on HepG2 cell lines. Here, we report that ML-7, in a nontoxic concentration, sensitized the HepG2 cells to quinocetone-induced cytotoxicity. Also, ML-7 profoundly enhances quinocetone-induced apoptosis in HepG2 cell line. Mechanistic investigations revealed that ML-7 and quinocetone act in concert to trigger the cleavage of caspase-8 as well as Bax/Bcl-2 ratio up-regulation and subsequent cleavage of Bid, capsases-9 and -3. Importantly, ML-7 weakened the quinocetone-induced Akt pathway activation, but strengthened the phosphorylation of p-38, ERK and JNK. Further treatment of Akt activator and p-38 inhibitor almost completely abolished the ML-7/quinocetone-induced apoptosis. In contrast, the ERK and JNK inhibitor aggravated the ML-7/quinocetone-induced apoptosis, indicating that the synergism critically depended on p-38 phosphorylation and HepG2 cells provoke Akt, ERK and JNK signaling pathways to against apoptosis. In conclusion, the rational combination of quinocetone and ML-7 presents a promising approach to trigger apoptosis in hepatocellular carcinoma, which warrants further investigation.


Subject(s)
Apoptosis/drug effects , Azepines/toxicity , Cell Survival/drug effects , Mitogen-Activated Protein Kinase Kinases/metabolism , Naphthalenes/toxicity , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/toxicity , Azepines/administration & dosage , Azepines/chemistry , Drug Therapy, Combination , Gene Expression Regulation/drug effects , Hep G2 Cells , Humans , Mitogen-Activated Protein Kinase Kinases/genetics , Naphthalenes/administration & dosage , Naphthalenes/chemistry , Proto-Oncogene Proteins c-akt/genetics , Quinoxalines/administration & dosage , Quinoxalines/chemistry
6.
Toxicol Mech Methods ; 26(4): 301-10, 2016 May.
Article in English | MEDLINE | ID: mdl-27098396

ABSTRACT

Quinocetone (QCT, 3-methyl-2-quinoxalin benzenevinylketo-1, 4-dioxide) is widely used as a veterinary drug and animal feed additive in China. Although it promotes growth and improves feed efficiency, QCT's in vitro and in vivo toxicities remain uncertain. This study was conducted to explore the mechanism of QCT-induced autophagy in HepG2 cells. By the results obtained from monodansylcadaverine (MDC) staining, ultrastructural observation by transmission electron microscopy (TEM), as well as Western blotting analysis for LC3, p62, and Beclin-1, it was demonstrated that QCT induced autophagy in HepG2 cells. Furthermore, PI3K/AKT inhibitor significantly enhanced QCT-induced autophagy, while TSC2 knockdown attenuated this process. In addition, inhibition of autophagy by pharmacological approach remarkably increased the viability of QCT-treated cells detected by MTT assay, suggesting that QCT-triggered autophagy may play as a promotion mechanism for cell death. Meanwhile, apoptosis was markedly downregulated after autophagy blockage, and evaluated by flow cytometry and Western blotting analysis for caspase-3 cleavage. Consequently, these results suggested that QCT-induced autophagy was mediated by AKT/TSC2/p70S6K signaling pathway, and inhibition of autophagy promoted QCT-treated cell survival by attenuating apoptosis.


Subject(s)
Anti-Bacterial Agents/toxicity , Autophagy/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/toxicity , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Tumor Suppressor Proteins/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Survival/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Flow Cytometry , Gene Knockdown Techniques , Hep G2 Cells , Hepatocytes , Humans , Microscopy, Electron, Transmission , Signal Transduction , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/genetics
7.
Toxicol Mech Methods ; 24(9): 654-65, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25162335

ABSTRACT

Tunicamycin (TM) causes accumulation of unfolded protein in endoplasmic reticulum (ER) lumen and introduces from elsewhere ER stress. This study was to assess the apoptosis and autophagy effect induced by TM on HepG2 cells and the role of autophagy in the system. The viability of HepG2 cells was significantly inhibited by TM in a dose-dependent manner detected by MTT assay. Then, the apoptotic morphology change, increasing apoptotic cell rate suggested that apoptosis was induced by TM in a time- and dose-dependent manner. To further determine the involvement of caspase-dependent pathway in TM-induced apoptosis, we discover that the activity of caspase-3/7, 8, 9 and cleavage of PARP markedly increased after TM treatment and the apoptosis was effectively attenuated by using caspase-9 and pan caspase inhibitor. Moreover, provided the rising stained acidic vacuoles and an increased level of LC3II and activation of Beclin1, we concluded that autophagy could be triggered by TM in a time- and dose-dependent manner. In addition, the inhibition of autophagy efficiently promoted TM-induced cell death identified by MTT assay. Meanwhile, the apoptotic cell rate and caspase-3 activation increased significantly after autophagy blockage. In conclusion, we found that TM initiated apoptosis and autophagy both in a time- and dose-dependent manner in HepG2 cells; and inhibition of autophagy may promote TM-induced cell death through enhancing apoptosis.


Subject(s)
Autophagy , Caspases/metabolism , Tunicamycin/pharmacology , Apoptosis , Enzyme Activation , Flow Cytometry , Hep G2 Cells , Humans
8.
Res Sq ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38645058

ABSTRACT

Genome wide association studies (GWASs) have identified numerous risk loci associated with prostate cancer, yet unraveling their functional significance remains elusive. Leveraging our high-throughput SNPs-seq method, we pinpointed rs4519489 within the multi-ancestry GWAS-discovered 2p25 locus as a potential functional SNP due to its significant allelic differences in protein binding. Here, we conduct a comprehensive analysis of rs4519489 and its associated gene, NOL10, employing diverse cohort data and experimental models. Clinical findings reveal a synergistic effect between rs4519489 genotype and NOL10 expression on prostate cancer prognosis and severity. Through unbiased proteomics screening, we reveal that the risk allele A of rs4519489 exhibits enhanced binding to USF1, a novel oncogenic transcription factor (TF) implicated in prostate cancer progression and prognosis, resulting in elevated NOL10 expression. Furthermore, we elucidate that NOL10 regulates cell cycle pathways, fostering prostate cancer progression. The concurrent expression of NOL10 and USF1 correlates with aggressive prostate cancer characteristics and poorer prognosis. Collectively, our study offers a robust strategy for functional SNP screening and TF identification through high-throughput SNPs-seq and unbiased proteomics, highlighting the rs4519489-USF1-NOL10 regulatory axis as a promising biomarker or therapeutic target for clinical diagnosis and treatment of prostate cancer.

9.
J Exp Clin Cancer Res ; 42(1): 198, 2023 Aug 08.
Article in English | MEDLINE | ID: mdl-37550764

ABSTRACT

BACKGROUND: Aberrant somatic genomic alteration including copy number amplification is a hallmark of cancer genomes. We previously profiled genomic landscapes of prostate cancer (PCa), yet the underlying causal genes with prognostic potential has not been defined. It remains unclear how a somatic genomic event cooperates with inherited germline variants contribute to cancer predisposition and progression. METHODS: We applied integrated genomic and clinical data, experimental models and bioinformatic analysis to identify GATA2 as a highly prevalent metastasis-associated genomic amplification in PCa. Biological roles of GATA2 in PCa metastasis was determined in vitro and in vivo. Global chromatin co-occupancy and co-regulation of GATA2 and SMAD4 was investigated by coimmunoprecipitation, ChIP-seq and RNA-seq assays. Tumor cellular assays, qRT-PCR, western blot, ChIP, luciferase assays and CRISPR-Cas9 editing methods were performed to mechanistically understand the cooperation of GATA2 with SMAD4 in promoting TGFß1 and AR signaling and mediating inherited PCa risk and progression. RESULTS: In this study, by integrated genomics and experimental analysis, we identified GATA2 as a prevalent metastasis-associated genomic amplification to transcriptionally augment its own expression in PCa. Functional experiments demonstrated that GATA2 physically interacted and cooperated with SMAD4 for genome-wide chromatin co-occupancy and co-regulation of PCa genes and metastasis pathways like TGFß signaling. Mechanistically, GATA2 was cooperative with SMAD4 to enhance TGFß and AR signaling pathways, and activated the expression of TGFß1 via directly binding to a distal enhancer of TGFß1. Strinkingly, GATA2 and SMAD4 globally mediated inherited PCa risk and formed a transcriptional complex with HOXB13 at the PCa risk-associated rs339331/6q22 enhancer, leading to increased expression of the PCa susceptibility gene RFX6. CONCLUSIONS: Our study prioritizes causal genomic amplification genes with prognostic values in PCa and reveals the pivotal roles of GATA2 in transcriptionally activating the expression of its own and TGFß1, thereby co-opting to TGFß1/SMAD4 signaling and RFX6 at 6q22 to modulate PCa predisposition and progression.


Subject(s)
Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/pathology , Prostate/metabolism , Signal Transduction , Transforming Growth Factor beta/metabolism , Chromatin , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Smad4 Protein/genetics , Smad4 Protein/metabolism , GATA2 Transcription Factor/genetics , GATA2 Transcription Factor/metabolism
10.
Oncogene ; 41(30): 3804-3820, 2022 07.
Article in English | MEDLINE | ID: mdl-35773413

ABSTRACT

Loss of α6ß4-dependent hemidesmosomal adhesions has been observed during prostate cancer progression. However, the significance and underlying mechanisms by which aberrant hemidesmosome assembly may modulate tumorigenesis remain elusive. Using an extensive CRISPR/Cas9-mediated genetic engineering approaches in different prostate cancer cell lines combined with in vivo tumorigenesis studies in mice, bone marrow-on-chip assays and bioinformatics, as well as histological analysis of prostate cancer patient cohorts, we demonstrated that simultaneous loss of PTEN and hemidesmosomal adhesions induced several tumorigenic properties including proliferation, migration, resistance to anoikis, apoptosis, and drug treatment in vitro, and increased metastatic capacity in vivo. These effects were plectin-depended and plectin was associated with actin-rich adhesions upon hemidesmosome disruption in PTEN-negative prostate cancer cells leading to activation of EGFR/PI3K/Akt- and FAK/Src-pathways. These results suggest that analysis of PTEN and hemidesmosomal proteins may have diagnostic value helping to stratify prostate cancer patients with high risk for development of aggressive disease and highlight actin-associated plectin as a potential therapeutic target specifically in PTEN/hemidesmosome dual-negative prostate cancer.


Subject(s)
Plectin , Prostatic Neoplasms , Actins , Animals , Anoikis , Carcinogenesis , Focal Adhesions/metabolism , Humans , Male , Mice , PTEN Phosphohydrolase/genetics , Phosphatidylinositol 3-Kinases , Plectin/genetics , Prostatic Neoplasms/pathology
11.
Nat Commun ; 13(1): 7320, 2022 11 28.
Article in English | MEDLINE | ID: mdl-36443337

ABSTRACT

Genome-wide association studies have identified 270 loci conferring risk for prostate cancer (PCa), yet the underlying biology and clinical impact remain to be investigated. Here we observe an enrichment of transcription factor genes including HNF1B within PCa risk-associated regions. While focused on the 17q12/HNF1B locus, we find a strong eQTL for HNF1B and multiple potential causal variants involved in the regulation of HNF1B expression in PCa. An unbiased genome-wide co-expression analysis reveals PCa-specific somatic TMPRSS2-ERG fusion as a transcriptional mediator of this locus and the HNF1B eQTL signal is ERG fusion status dependent. We investigate the role of HNF1B and find its involvement in several pathways related to cell cycle progression and PCa severity. Furthermore, HNF1B interacts with TMPRSS2-ERG to co-occupy large proportion of genomic regions with a remarkable enrichment of additional PCa risk alleles. We finally show that HNF1B co-opts ERG fusion to mediate mechanistic and biological effects of the PCa risk-associated locus 17p13.3/VPS53/FAM57A/GEMIN4. Taken together, we report an extensive germline-somatic interaction between TMPRSS2-ERG fusion and genetic variations underpinning PCa risk association and progression.


Subject(s)
Genome-Wide Association Study , Prostatic Neoplasms , Male , Humans , Prostate , Prostatic Neoplasms/genetics , Pelvis , Germ Cells , Transcriptional Regulator ERG/genetics , Hepatocyte Nuclear Factor 1-beta/genetics , Serine Endopeptidases/genetics , Oncogene Proteins, Fusion/genetics
12.
Toxicol In Vitro ; 47: 195-206, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29229420

ABSTRACT

Quinocetone (QCT) has been approved and widely used as an animal feed additive in China since 2003. However, investigations indicate that QCT shows potential toxicity both in vitro and in vivo. Although voltage dependent anion channel 1 (VDAC1) involved in regulating QCT-induced apoptotic cell death has been established, the role of voltage dependent anion channel 2 (VDAC2) in QCT-induced toxicity remains unclear. In this study, we showed that QCT-induced cell death was coupled to VDAC2 oligomerization. Moreover, VDAC inhibitor 4, 4'-diisothiocyano stilbene-2, 2'-disulfonic acid (DIDS) alleviated QCT-induced cell death and VDAC2 oligomerization. Meanwhile, overexpression VDAC2 aggravated QCT-induced VDAC2 oligomerization. In addition, caspase inhibitor Z-VAD-FMK and reactive oxidative species (ROS) scavenger N-acetyl-l-cysteine (NAC) apparently blocked QCT-induced cell death and VDAC2 oligomerization. Finally, overexpression N-terminal truncated VDAC2 attenuated QCT-induced VDAC2 oligomerization but had no influence on its localization to mitochondria when comparing to the full length of VDAC2. Taken together, our results reveal that ROS-mediated VDAC2 oligomerization is associated with QCT-induced apoptotic cell death. The N-terminal region of VDAC2 is required for QCT-induced VDAC2 oligomerization.


Subject(s)
Hepatocytes/drug effects , Mitochondria, Liver/drug effects , Oxidants/toxicity , Quinoxalines/toxicity , Reactive Oxygen Species/agonists , Voltage-Dependent Anion Channel 2/metabolism , 4,4'-Diisothiocyanostilbene-2,2'-Disulfonic Acid/pharmacology , Caspase Inhibitors/pharmacology , Dimerization , Free Radical Scavengers , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hep G2 Cells , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Membrane Transport Modulators/pharmacology , Microscopy, Fluorescence , Mitochondria, Liver/metabolism , Osmolar Concentration , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Multimerization/drug effects , Protein Transport/drug effects , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Voltage-Dependent Anion Channel 2/antagonists & inhibitors , Voltage-Dependent Anion Channel 2/chemistry
13.
Food Chem Toxicol ; 108(Pt A): 148-160, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28757460

ABSTRACT

Olaquindox, a quinoxaline 1,4-di-N-oxide, is known as an antibacterial agent and feed additive to treat bacterial infections and promote animal growth. However, the potential mechanism of toxicity is still unknown. The present study aims to explore the molecular mechanism of p21 on olaquindox-induced mitochondrial apoptosis and S-phase arrest in human hepatoma G2 cells (HepG2). As a result, olaquindox promoted production of ROS, suppressed the protein expression p21 in p53-independent way and phosphorylated p21. Meanwhile, olaquindox activated AKT and Nrf2/HO-1 pathway, up-regulated Bax/Bcl-2 ratio, disrupted mitochondrial membrane potential (MMP) and subsequently caused cytochrome c release and a cascade activation of caspase, eventually induced apoptosis. Olaquindox could induce S-phase arrest in HepG2 cells involved with the increase of Cyclin A, Cyclin E and CDK 2. Furthermore, knockdown of p21 decreased cell viability, enhanced oxidative stress, aggravated olaquindox-induced mitochondrial apoptosis and S-phase arrest involvement of activating PI3K/AKT and inhibiting Nrf2/HO-1 pathway. PI3K/AKT inhibitor (LY294002) and HO-1inhibitor (ZnPP-IX) both increased olaquindox-induced apoptosis and S-phase arrest. In conclusion, knockdown of p21 increased olaquindox-induced mitochondrial apoptosis and S-phase arrest through further activating PI3K/AKT and inhibiting Nrf2/HO-1pathway. Our study provided new insights into the molecular mechanism of olaquindox and shed light on the role of p21.


Subject(s)
Apoptosis/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Mitochondria/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinoxalines/pharmacology , Gene Expression Regulation/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hep G2 Cells , Humans , Mitochondria/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics
14.
Food Chem Toxicol ; 105: 161-176, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28343033

ABSTRACT

Quinocetone (QCT) has been used as an animal feed additive in China since 2003. However, investigations indicate that QCT has potential toxicity due to the fact that it shows cytotoxicity, genotoxicity, hepatotoxicity, nephrotoxicity and immunotoxicity in vitro and animal models. Although QCT-induced mitochondrial apoptosis has been established, the molecular mechanism remains unclear. This study was aimed to investigate the role of voltage-dependent anion channel 1 (VDAC1) oligomerization and Wnt/ß-catenin pathway in QCT-induced mitochondrial apoptosis. The results showed VDAC inhibitor 4, 4-diisothiocyano stilbene-2, 2-disulfonic acid (DIDS) partly compromised QCT-induced cell viability decrease (from 34.1% to 68.5%) and mitochondrial apoptosis accompanied by abating VDAC1 oligomerization, cytochrome c (Cyt c) release and the expression levels of cleaved caspase-9, -3 and poly (ADP-ribose) polymerase (PARP). Meanwhile, overexpression VDAC1 exacerbated QCT-induced VDAC1 oligomerization and Cyt c release. In addition, lithium chloride (LiCl), an activator of Wnt/ß-catenin pathway, markedly attenuated QCT-induced mitochondrial apoptosis by partly restoring the expression levels of Wnt1 and ß-catenin. Finally, reactive oxygen species (ROS) scavenger N-acetyl-l-cysteine (NAC) obviously blocked QCT-induced VDAC1 oligomerization and the inhibition of Wnt1/ß-catenin pathway. Taken together, our results reveal that QCT induces mitochondrial apoptosis by ROS-dependent promotion of VDAC1 oligomerization and suppression of Wnt1/ß-catenin pathway.


Subject(s)
Apoptosis/drug effects , Mitochondria/metabolism , Quinoxalines/toxicity , Reactive Oxygen Species/metabolism , Voltage-Dependent Anion Channel 1/metabolism , Wnt1 Protein/metabolism , beta Catenin/metabolism , Cell Line, Tumor , Hep G2 Cells , Humans , Mitochondria/drug effects , Mitochondria/genetics , Signal Transduction/drug effects , Voltage-Dependent Anion Channel 1/chemistry , Voltage-Dependent Anion Channel 1/genetics , Wnt1 Protein/genetics , beta Catenin/genetics
15.
Food Chem Toxicol ; 88: 1-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26687534

ABSTRACT

Furazolidone (FZD), a synthetic nitrofuran with a broad spectrum of antimicrobial activities, has been shown to exhibit marked genotoxity and cytotoxicity in vitro, but the proper mechanism was unclear. P21(Waf1/Cip1) (p21), a cyclin-dependent kinase, is critically involved in cell cycle arrest and apoptosis in response to DNA injury. This study was aimed to explore the role of p21 in FZD-induced apoptosis in HepG2 cells and uncover its possible mechanism. Firstly, we demonstrated that FZD (50 µg/mL) treatment increased the mRNA level of p21 but reduced the protein level of p21 by shortening its half-life. Moreover, the degradation of p21 was associated with the inhibition of PI3K/Akt pathway by FZD. Then, the change of p21 protein expression modulated FZD-induced apoptosis. Overexpression of p21 attenuated FZD-induced caspase-3 activation and ROS generation, eventually reduced apoptosis. Conversely, knockdown of p21 by siRNA enhanced FZD-induced those phenomenon. In addition, the influence of p21 on FZD-induced ROS generation might be associated with Nrf2/HO-1 pathway which was a key regulator in defense response against oxidative stress. In conclusion, these findings demonstrated that p21 plays a critical role in FZD-induced apoptosis in HepG2 cells through influencing the caspase-3 activation and ROS generation.


Subject(s)
Apoptosis/drug effects , Caspase 3/metabolism , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Furazolidone/toxicity , Reactive Oxygen Species/metabolism , Caspase 3/genetics , Cell Survival , Cyclin-Dependent Kinase Inhibitor p21/genetics , Down-Regulation , Gene Expression Regulation/physiology , Hep G2 Cells , Humans , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism
16.
Environ Toxicol Pharmacol ; 46: 140-146, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27458702

ABSTRACT

Olaquindox, a quinoxaline 1, 4-dioxide derivative, has been widely used as a feed additive for promoting animal growth in China. The aim of present study was to investigate the effect of grow arrest and DNA damage 45 alpha (GADD45a) on olaquindox-induced apoptosis in HepG2 cells. The result showed that olaquindox induced the decrease of cell viability in a dose dependent manner. Compared to the control group, olaquindox treatment at 400 and 800µg/mL increased the expression level of GADD45a protein and reactive oxygen species (ROS) production, decreased mitochondrial membrane potential (MMP), and subsequently increased the expression of Bax while decreased the expression of Bcl-2, leading to the release of cytochrome c (Cyt c). However, knockdown of GADD45a enhanced olaquindox-induced ROS production, disrupted MMP and subsequently caused Cyt c release, then further increased olaquindox- induced cell apoptosis by increasing the activities of caspase-9, caspase-3, and poly (ADP-ribose) polymerase (PARP). In conclusion, the results revealed that GADD45a played a critical role in olaquindox-induced apoptosis in HepG2 cells, which may embrace the regulatory ability on the mitochondrial apoptosis pathway.


Subject(s)
Apoptosis/drug effects , Cell Cycle Proteins/metabolism , Mitochondria/drug effects , Nuclear Proteins/metabolism , Quinoxalines/toxicity , Apoptosis/genetics , Caspase 3/metabolism , Caspase 9/metabolism , Cell Cycle Proteins/genetics , Cell Death/drug effects , Cell Death/genetics , Gene Knockdown Techniques , Hep G2 Cells/drug effects , Hep G2 Cells/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Nuclear Proteins/genetics , Poly(ADP-ribose) Polymerases/metabolism , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL