Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
J Exp Clin Cancer Res ; 40(1): 220, 2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34210327

ABSTRACT

BACKGROUND: Metastasis is a major challenge in cervical cancer treatment. Previous studies have shown that the dual functional protein apurinic/apyrimidinic endonuclease 1 (APE1) promotes tumor metastasis and is overexpressed in cervical cancer. However, the biological role and mechanism of APE1 in cervical cancer metastasis have rarely been studied. METHODS: We used gene set enrichment analysis (GSEA) to determine the APE1-related signaling pathways in cervical cancer. To investigate the role and mechanism of APE1 in cervical cancer metastasis and invasion, immunohistochemistry, immunofluorescence, western blotting, secondary structure prediction, coimmunoprecipitation, luciferase reporter, and electrophoretic mobility shift assays were performed. The inhibitory effects of the APE1 redox function inhibitor APX3330 on cervical cancer metastasis were evaluated using animal models. RESULTS: Clinical data showed that high expression of APE1 was associated with lymph node metastasis in cervical cancer patients. GSEA results showed that APE1 was associated with epithelial to mesenchymal transition (EMT) in cervical cancer. Ectopic expression of APE1 promoted EMT and invasion of cervical cancer cells, whereas inhibition of APE1 suppressed EMT and invasion of cervical cancer cells in a redox function-dependent manner. Notably, APE1 redox function inhibitor APX3330 treatment dramatically suppressed cervical cancer cell lymph node and distant metastasis in vivo. Furthermore, we found that APE1 enhanced the interaction between ZEB1 and the E-cadherin promoter by binding to ZEB1, thereby suppressing the expression of E-cadherin, a negative regulator of EMT. CONCLUSION: Our findings help to elucidate the role played by APE1 in cervical cancer metastasis and targeting APE1 redox function may be a novel strategy for inhibiting cervical cancer metastasis.


Subject(s)
Antigens, CD/metabolism , Cadherins/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Uterine Cervical Neoplasms/metabolism , Zinc Finger E-box-Binding Homeobox 1/metabolism , Animals , Antigens, CD/genetics , Cadherins/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Epithelial-Mesenchymal Transition , Female , HeLa Cells , Heterografts , Humans , Lymphatic Metastasis , Mice , Middle Aged , Neoplasm Metastasis , Oxidation-Reduction , Transfection , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology , Zinc Finger E-box-Binding Homeobox 1/genetics
2.
Sci Rep ; 10(1): 3758, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111912

ABSTRACT

Although APE2 plays essential roles in base excision repair and ATR-Chk1 DNA damage response (DDR) pathways, it remains unknown how the APE2 gene is altered in the human genome and whether APE2 is differentially expressed in cancer patients. Here, we report multiple-cancer analyses of APE2 genomic alterations and mRNA expression from cancer patients using available data from The Cancer Genome Atlas (TCGA). We observe that APE2 genomic alterations occur at ~17% frequency in 14 cancer types (n = 21,769). Most frequent somatic mutations of APE2 appear in uterus (2.89%) and skin (2.47%) tumor samples. Furthermore, APE2 expression is upregulated in tumor tissue compared with matched non-malignant tissue across 5 cancer types including kidney, breast, lung, liver, and uterine cancers, but not in prostate cancer. We also examine the mRNA expression of 13 other DNA repair and DDR genes from matched samples for 6 cancer types. We show that APE2 mRNA expression is positively correlated with PCNA, APE1, XRCC1, PARP1, Chk1, and Chk2 across these 6 tumor tissue types; however, groupings of other DNA repair and DDR genes are correlated with APE2 with different patterns in different cancer types. Taken together, this study demonstrates alterations and abnormal expression of APE2 from multiple cancers.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Endonucleases/biosynthesis , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Multifunctional Enzymes/biosynthesis , Mutation , Neoplasm Proteins/biosynthesis , Neoplasms/enzymology , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Endonucleases/genetics , Humans , Multifunctional Enzymes/genetics , Neoplasm Proteins/genetics , Neoplasms/genetics
3.
Exp Neurol ; 307: 1-11, 2018 09.
Article in English | MEDLINE | ID: mdl-29772245

ABSTRACT

Inflammatory pain is a complex and multifactorial disorder. Apurinic/apyrimidinic endonuclease 1 (APE1), also called Redox Factor-1 (Ref-1), is constitutively expressed in the central nervous system and regulates various cellular functions including oxidative stress. In the present study, we investigated APE1 modulation and associated pain behavior changes in the complete Freund's adjuvant (CFA) model of inflammatory pain in rats. In addition we tested the anti-inflammatory effects of E3330, a selective inhibitor of APE1-redox activity, in CFA pain condition. We demonstrate that APE1 expression and subcellular distribution are significantly altered in rats at 4 days post CFA injection. We observed around 30% reduction in the overall APE1 mRNA and protein levels. Interestingly, our data point to an increased nuclear accumulation in the inflamed group as compared to the sham group. E3330 inhibitor injection in CFA rats normalized APE1 mRNA expression and changed its distribution toward cytosolic accumulation. Furthermore, intrathecal injection of E3330 decreased inflammation (i.e. reduced IL-6 expression) and alleviated pain, as assessed by measuring the paw withdrawal threshold with the von Frey test. In conclusion, our data indicate that changes in APE1 expression and sub-cellular distribution are implicated in inflammatory pain mechanisms mediated by APE1 redox functions. Further studies are required to elucidate the exact function of APE1 in inflammatory pain processes.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Oxidative Stress/physiology , Pain/metabolism , Animals , Benzoquinones/pharmacology , Benzoquinones/therapeutic use , Freund's Adjuvant/toxicity , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Pain/chemically induced , Pain/drug therapy , Propionates/pharmacology , Propionates/therapeutic use , Rats , Rats, Sprague-Dawley
4.
Environ Sci Pollut Res Int ; 25(12): 12150-12158, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29455351

ABSTRACT

Several possible mechanisms have been examined to gain an understanding on the carcinogenic properties of lead, which include among others, mitogenesis, alteration of gene expression, oxidative damage, and inhibition of DNA repair. The aim of the present study was to explore if low concentrations of lead, relevant for human exposure, interfere with Ape1 function, a base excision repair enzyme, and its role in cell transformation in Balb/c-3T3. Lead acetate 5 and 30 µM induced APE1 mRNA and upregulation of protein expression. This increase in mRNA expression is consistent throughout the chronic exposure. Additionally, we also found an impaired function of Ape1 through molecular beacon-based assay. To evaluate the impact of lead on foci formation, a Balb/c-3T3 two-step transformation model was used. Balb/c-3T3 cells were pretreated 1 week with low concentrations of lead before induction of transformation with n-methyl-n-nitrosoguanidine (MNNG) (0.5 µg/mL) and 12-O-tetradecanoylphorbol-13-acetate (TPA) (0.1 µg/mL) (a classical two-step protocol). Morphological cell transformation increased in response to lead pretreatment that was paralleled with an increase in Ape1 mRNA and protein overexpression and an impairment of Ape1 activity and correlating with foci number. In addition, we found that lead pretreatment and MNNG (transformation initiator) increased DNA damage, determined by comet assay. Our data suggest that low lead concentrations (5, 30 µM) could play a facilitating role in cellular transformation, probably through the impaired function of housekeeping genes such as Ape1, leading to DNA damage accumulation and chromosomal instability, one of the most important hallmarks of cancer induced by chronic exposures.


Subject(s)
Carcinogens, Environmental/toxicity , Cell Transformation, Neoplastic/drug effects , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Lead/toxicity , Models, Biological , Animals , BALB 3T3 Cells , Cell Proliferation/drug effects , Cell Survival/drug effects , Comet Assay , Gene Expression/drug effects , Humans , Methylnitronitrosoguanidine/pharmacology , Mice , Tetradecanoylphorbol Acetate/pharmacology
5.
Metab Brain Dis ; 32(5): 1705-1716, 2017 10.
Article in English | MEDLINE | ID: mdl-28676971

ABSTRACT

Glial cells protect themselves from the elevated reactive oxygen species (ROS) via developing unusual mechanisms to maintain the genomic stability, and reprogramming of the cellular antioxidant system to cope with the adverse effects. In the present study non-cytotoxic dose of oxidants, H2O2 (100 µM) and GO (10 µU/ml) was used to induce moderate oxidative stress via generating ROS in human glioblastoma cell line U-87 MG cells, which showed a marked increase in the antioxidant capacity as studied by measuring the modulation in expression levels and activities of superoxide dismutase (SOD1 and SOD2) and catalase (CAT) enzymes, and the GSH content. However, pretreatment (3 h) of Curcumin and Quercetin (10 µM) followed by the treatment of oxidants enhanced the cell survival, and the levels/activities of the antioxidants studied. Oxidative stress also resulted in an increase in the nitrite levels in the culture supernatants, and further analysis by immunocytochemistry showed an increase in iNOS expression. In addition, phytochemical pretreatment decreased the nitrite level in the culture supernatants of oxidatively stressed U-87 MG cells. Elevated ROS also increased the expression of COX-2 and APE1 enzymes and pretreatment of Curcumin and Quercetin decreased COX-2 expression and increased APE1 expression in the oxidatively stressed U-87 MG cells. The immunocytochemistry also indicates for APE1 enhanced stress-dependent subcellular localization to the nuclear compartment, which advocates for enhanced DNA repair and redox functions of APE1 towards survival of U-87 MG cells. It can be concluded that intracellular oxidants activate the key enzymes involved in antioxidant mechanisms, NO-dependent survival mechanisms, and also in the DNA repair pathways for glial cell survival in oxidative-stress micro-environment.


Subject(s)
Antioxidants/metabolism , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Glucose Oxidase/metabolism , Hydrogen Peroxide/metabolism , Antioxidants/pharmacology , Brain Neoplasms/pathology , Catalase/metabolism , Cell Line, Tumor , Cell Survival , Cyclooxygenase 2/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Glioblastoma/pathology , Glutathione/metabolism , Humans , Nitric Oxide Synthase Type II/biosynthesis , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism
6.
Mech Ageing Dev ; 161(Pt A): 149-162, 2017 01.
Article in English | MEDLINE | ID: mdl-27364693

ABSTRACT

It is unknown whether DNA damage accumulation is an upstream instigator or secondary effect of the cell death process in different populations of adult postmitotic neurons in the central nervous system. In two different mouse models of injury-induced neurodegeneration characterized by relatively synchronous accumulation of mitochondria, oxidative stress, and DNA damage prior to neuronal apoptosis, we enforced the expression of human 8-oxoguanine DNA glycosylase (hOGG1) and human apurinic-apyrimidinic endonuclease-1/Ref1 (hAPE) using recombinant adenoviruses (Ad). Thalamic lateral geniculate neurons and lumbar spinal cord motor neurons were transduced by Ad-hOGG1 and Ad-hAPE injections into the occipital cortex and skeletal muscle, respectively, prior to their target deprivation- and axotomy-induced retrograde apoptosis. Enforced expression of hOGG1 and hAPE in thalamus and spinal cord was confirmed by western blotting and immunohistochemistry. In injured populations of neurons in thalamus and spinal cord, a DNA damage response (DDR) was registered, as shown by localization of phospho-activated p53, Rad17, and replication protein A-32 immunoreactivities, and this DDR was attenuated more effectively by enforced hAPE expression than by hOGG1 expression. Enforced expression of hOGG1 and hAPE significantly protected thalamic neurons and motor neurons from retrograde apoptosis induced by target deprivation and axotomy. We conclude that a DDR response is engaged pre-apoptotically in different types of injured mature CNS neurons and that DNA repair enzymes can regulate the survival of retrogradely dying neurons, suggesting that DNA damage and activation of DDR are upstream mechanisms for this form of adult neurodegeneration in vivo, thus identifying DNA repair as a therapeutic target for neuroprotection.


Subject(s)
Adenoviridae , Apoptosis , DNA Glycosylases/biosynthesis , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Motor Neurons/metabolism , Neurodegenerative Diseases/metabolism , Transduction, Genetic , Animals , Axotomy , DNA Glycosylases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Models, Animal , Humans , Male , Mice , Mice, Transgenic , Motor Neurons/pathology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , Neurodegenerative Diseases/therapy , Replication Protein A/genetics , Replication Protein A/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
7.
Antioxid Redox Signal ; 26(12): 616-629, 2017 04 20.
Article in English | MEDLINE | ID: mdl-27835927

ABSTRACT

The APEX nuclease (multifunctional DNA repair enzyme) 1 (APEX1) has a disordered N-terminus, a redox, and a DNA repair domain. APEX1 has anti-apoptotic properties, which have been linked to both domains depending on cell type and experimental conditions. AIMS: As protection against apoptosis is a hallmark of vessel integrity, we wanted to elucidate whether APEX1 acts anti-apoptotic in primary human endothelial cells and, if so, what the underlying mechanisms are. RESULTS: APEX1 inhibits apoptosis in endothelial cells by reducing Cathepsin D (CatD) cleavage, potentially by binding to the unprocessed form. Diminished CatD activation results in increased Thioredoxin-1 protein levels leading to reduced Caspase 3 activation. Consequently, apoptosis rates are decreased. This depends on the first twenty amino acids in APEX1, because APEX1 (21-318) induces CatD activity, decreases Thioredoxin-1 protein levels, and, thus, increases Caspase 3 activity and apoptosis. Along the same lines, APEX1 (1-20) inhibits Caspase 3 cleavage and apoptosis. Furthermore, re-expression of Thioredoxin-1 via lentiviral transduction rescues endothelial cells from APEX1 (21-318)-induced apoptosis. In an in vivo model of restenosis, which is characterized by oxidative stress, endothelial activation, and smooth muscle cell proliferation, Thioredoxin-1 protein levels are reduced in the endothelium of the carotids. INNOVATION: APEX1 acts anti-apoptotic in endothelial cells. This anti-apoptotic effect depends on the first 20 amino acids of APEX1. CONCLUSION: As proper function of the endothelium during life span is a hallmark for individual health span, a detailed characterization of the functions of the APEX1N-terminus is required to understand all its cellular properties. Antioxid. Redox Signal. 26, 616-629.


Subject(s)
Apoptosis/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Graft Occlusion, Vascular/genetics , Thioredoxins/biosynthesis , Amino Acids/genetics , Amino Acids/metabolism , Blood Vessels/metabolism , Blood Vessels/pathology , Carotid Arteries/metabolism , Carotid Arteries/pathology , Caspase 3/genetics , Caspase 3/metabolism , Cathepsin D/genetics , Cell Proliferation/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Endothelial Cells/metabolism , Gene Expression Regulation , Graft Occlusion, Vascular/pathology , Humans , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Oxidative Stress/genetics , Thioredoxins/genetics
8.
Stem Cells Transl Med ; 5(8): 1067-78, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27334489

ABSTRACT

UNLABELLED: : Overcoming the insufficient survival of cell grafts is an essential objective in cell-based therapy. Apurinic/apyrimidinic endonuclease/redox factor 1 (APE1) promotes cell survival and may enhance the therapeutic effect of engrafted cells. The aim of this study is to determine whether APE1 overexpression in cardiac progenitor cells (CPCs) could ameliorate the efficiency of cell-based therapy. CPCs isolated from 8- to 10-week-old C57BL/6 mouse hearts were infected with retrovirus harboring APE1-DsRed (APE1-CPC) or a DsRed control (control-CPC). Oxidative stress-induced apoptosis was then assessed in APE1-CPCs, control-CPCs, and neonatal rat ventricular myocytes (NRVMs) cocultured with these CPCs. This analysis revealed that APE1 overexpression inhibited CPC apoptosis with activation of transforming growth factor ß-activated kinase 1 (TAK1) and nuclear factor (NF)-κB. In the coculture model, NRVM apoptosis was inhibited to a greater extent in the presence of APE1-CPCs compared with control-CPCs. Moreover, the number of surviving DsRed-positive CPC grafts was significantly higher 7 days after the transplant of APE1-CPCs into a mouse myocardial infarction model, and the left ventricular ejection fraction showed greater improvement with attenuation of fibrosis 28 days after the transplant of APE1-CPCs compared with control-CPCs. Additionally, fewer inflammatory macrophages and a higher percentage of cardiac α-sarcomeric actinin-positive CPC-grafts were observed in mice injected with APE1-CPCs compared with control-CPCs after 7 days. In conclusion, antiapoptotic APE1-CPC graft, which increased TAK1-NF-κB pathway activation, survived effectively in the ischemic heart, restored cardiac function, and reduced cardiac inflammation and fibrosis. APE1 overexpression in CPCs may serve as a novel strategy to improve cardiac cell therapy. SIGNIFICANCE: Improving the survival of cell grafts is essential to maximize the efficacy of cell therapy. The authors investigated the role of APE1 in CPCs under ischemic conditions and evaluated the therapeutic efficacy of transplanted APE1-overexpressing CPCs in a mouse model of myocardial infarction. APE1 hindered apoptosis in CPC grafts subjected to oxidative stress caused in part by increased TAK1-NF-κB pathway activation. Furthermore, APE1-CPC grafts that effectively survived in the ischemic heart restored cardiac function and attenuated fibrosis through pleiotropic mechanisms that remain to be characterized. These findings suggest that APE1 overexpression in CPCs may be a novel strategy to reinforce cardiac cell therapy.


Subject(s)
Apoptosis , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Genetic Therapy/methods , Myocardial Infarction/surgery , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/transplantation , Regeneration , Stem Cell Transplantation/methods , Stem Cells/enzymology , Animals , Cell Differentiation , Cell Survival , Cells, Cultured , Coculture Techniques , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Disease Models, Animal , Enzyme Induction , Fibrosis , Graft Survival , Inflammation Mediators/metabolism , Male , Mice, Inbred C57BL , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Neovascularization, Physiologic , Oxidative Stress , Phenotype , Recovery of Function , Signal Transduction , Stem Cells/pathology , Stroke Volume , Time Factors , Transfection , Ventricular Function, Left , Ventricular Remodeling
9.
Genes Genet Syst ; 91(3): 183-188, 2016 Nov 26.
Article in English | MEDLINE | ID: mdl-27150717

ABSTRACT

Reduced-genome Escherichia coli strains lacking up to 38.9% of the parental chromosome have been constructed by combining large-scale chromosome deletion mutations. Functionally redundant genes involved in essential processes can be systematically identified using these reduced-genome strains. One large-scale chromosome deletion mutation could be introduced into the wild-type strain but not into the largest reduced-genome strain, suggesting a synthetic lethal interaction between genes removed by the deletion and those already absent in the reduced-genome strain. Thus, introduction of the deletion mutation into a series of reduced-genome mutants could allow the identification of other chromosome deletion mutations responsible for the synthetic lethal phenotype. We identified a synthetic lethality caused by disruption of nfo and xthA, two genes encoding apurinic/apyrimidinic (AP) endonucleases involved in the DNA base excision repair pathway, and two other large-scale chromosome deletions. We constructed temperature-sensitive mutants harboring quadruple-deletion mutations in the affected genes/chromosome regions. Using these mutants, we identified two multi-copy suppressors: holC, encoding the chi subunit of DNA polymerase III, and yoaA, encoding a putative DNA helicase. Addition of the yoaA disruption increased the methyl methanesulfonate (MMS) sensitivity of xthA single-deletion or xthA nfo double-deletion mutants. This increased MMS sensitivity was not suppressed by the presence of multi-copy holC. These results indicate that yoaA is involved in MMS sensitivity and suggest that YoaA functions together with HolC.


Subject(s)
DNA Helicases/genetics , DNA Polymerase III/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Escherichia coli Proteins/genetics , Synthetic Lethal Mutations/genetics , Chromosomes, Bacterial/genetics , DNA Helicases/biosynthesis , DNA Polymerase III/biosynthesis , DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Deoxyribonuclease IV (Phage T4-Induced)/biosynthesis , Deoxyribonuclease IV (Phage T4-Induced)/genetics , Escherichia coli/enzymology , Escherichia coli/genetics , Escherichia coli Proteins/biosynthesis , Genome, Bacterial/drug effects , Genotype , Methyl Methanesulfonate/pharmacology , Mutant Proteins/biosynthesis , Mutant Proteins/genetics , Mutation , Signal Transduction/drug effects , Synthetic Lethal Mutations/drug effects
10.
Nucleic Acids Res ; 44(7): 3165-75, 2016 Apr 20.
Article in English | MEDLINE | ID: mdl-26773055

ABSTRACT

DNA constantly undergoes chemical modification due to endogenous and exogenous mutagens. The DNA base excision repair (BER) pathway is the frontline mechanism handling the majority of these lesions, and primarily involves a DNA incision and subsequent resealing step. It is imperative that these processes are extremely well-coordinated as unrepaired DNA single strand breaks (SSBs) can be converted to DNA double strand breaks during replication thus triggering genomic instability. However, the mechanism(s) governing the BER process are poorly understood. Here we show that accumulation of unrepaired SSBs triggers a p53/Sp1-dependent downregulation of APE1, the endonuclease responsible for the DNA incision during BER. Importantly, we demonstrate that impaired p53 function, a characteristic of many cancers, leads to a failure of the BER coordination mechanism, overexpression of APE1, accumulation of DNA strand breaks and results in genomic instability. Our data provide evidence for a previously unrecognized mechanism for coordination of BER by p53, and its dysfunction in p53-inactivated cells.


Subject(s)
DNA Repair , Genomic Instability , Tumor Suppressor Protein p53/metabolism , Cells, Cultured , DNA Breaks, Single-Stranded , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Down-Regulation , Humans , Sp1 Transcription Factor/metabolism
12.
Eur Rev Med Pharmacol Sci ; 19(16): 3063-71, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26367730

ABSTRACT

OBJECTIVE: To study the expression of programmed death-ligand1 (PD-L1) in hepatocellular carcinoma and its relationship with clinicopathological characteristics and, prognosis of hepatocellular carcinoma and APE1, P53 protein expression levels. PATIENTS AND METHODS: A total of 128 patients with hepatocellular carcinoma were enrolled in this study. The expression of PD-L1, APE1 and P53 were detected by immunohistochemistry.Use immunohistochemical ABC staining method to detect the expression levels of PD-L1, APE1 and P53 protein in the hepatocellular carcinoma of 128 cases. RESULTS: Positive The positive expression rates levels of PD-L1, APE1, and P53 protein in hepatocellular carcinoma tissues are were 82.03%, 92.19%, and 60.94%. PD-L1 positive expression were significantly associated with clinical stage, The PD-L1 protein has a high expression in patients with I ~ II stage liver cancerHBV infection positive and nonportal vein thrombosis (p=0.041; p=0.030; p=0.014). It is inversely correlated with P53 and PD-L1 expression (correlation coefficient -0.227, p=0.010), and positively correlated with APE1 expression (correlation coefficient 0.189, p=0.032). The expression of PD-L1 is associated with the survival time of patients with hepatocellular carcinoma, and the median survival time of patients with high expression of PD-L1 is ten months. The median survival time of patients with low expression is five months (p=0.001). The relationship between the expression of APE1 and P53 protein and overall survival time of patients with hepatocellular carcinoma has not been found. CONCLUSIONS: The PD-L1 and APE1 expression in hepatocellular carcinoma are related to the level of the expression of P53 protein. The expression state of PD-L1 may be a prognostic factor in hepatocellular carcinoma.


Subject(s)
B7-H1 Antigen/biosynthesis , Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Liver Neoplasms/metabolism , Tumor Suppressor Protein p53/biosynthesis , Adolescent , Adult , Aged , B7-H1 Antigen/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Female , Humans , Immunohistochemistry , Liver Neoplasms/pathology , Male , Middle Aged , Prognosis , Tumor Suppressor Protein p53/genetics , Young Adult
13.
J Nanosci Nanotechnol ; 15(7): 4815-28, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26373042

ABSTRACT

China is one of the countries with the highest incidence of gastric cancer, and accounts for over 40% of all new gastric cancer cases in the world. Genetic factors as well as environmental factors play a role in development of gastric cancer. To investigate the independent roles of single nucleotide polymorphisms (SNPs) in base excision repair (BER) genes (APE1 and NEIL2), carcinogen metabolism gene (CYP2E1) and tumor suppressor pathway gene (MDM2) for gastric cancer susceptibility in a Chinese population, we conducted a hospital based case-control study to evaluate the potential association between these polymorphisms and susceptibility to gastric cancer in a Northern Jiangsu population. We also associated the NEIL-2 mRNA expression with the studied NEIL2 SNP genotypes to assess whether the genotypes have influence on the NEIL2 mRNA (hence protein) expression. Five SNPs, APE 1 (rs2275008), NEIL 2 (rs804270), MDM2 (rs2279744), and CYP 2E1 (rs2480256 and rs2031920), were genotyped by TaqMan assays in 105 gastric cancer cases and 118 controls. Genotype frequency distribution showed that the APE 1 SNP (rs2275008), NEIL 2 SNP (rs804270), MDM2 SNP (rs2279744), and CYP 2E1 SNP (rs2031920) had more mutant alleles in gastric cancer cases than controls (76.19, 68.57, 54.29, and 43.81%, respectively), while CYP 2E1 SNP (rs2480256) had large percentage of both alleles (43.81%). Risk analysis revealed that there was increased risk for gastric cancer in subjects with mutant alleles in APE 1 (rs2275008: OR 5.49, 95% CI = 2.6-5.7, p <.0001), NEIL 2 (rs804270: OR 2.3, 95% CI = 1.22-4.3, p=0.01), MDM2 (rs2279744: OR 14.65, 95% CI = 5.63-8.15, p < .0001), and CYP 2E1 (rs2031920: OR 8.385, 95% CI = 3.2-5.3, p < .0001) SNPs. Moreover, the NEIL2 mRNA expression analysis showed that there was significant differential expression of NEIL2 mRNA among the randomly tested NEIL2 genotypes (p = 0.005), with low expression seen in variant genotypes than in other genotypes. In conclusion, variant alleles in the NEIL2 (rs804270), APE1 (rs2275008), CYP2E1 (rs2031920) and MDM2 (rs2279744) SNPs may independently influence susceptibility to gastric cancer in a Northern Jiangsu Chinese population. The genotypes may also independently influence their respective gene mRNA expression, as seen in our study, where there was differential expression of the NEIL2 mRNA among the genotypes, with low NEIL2 mRNA expression seen in the variant genotype.


Subject(s)
Cytochrome P-450 CYP2E1/genetics , DNA Glycosylases/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Proto-Oncogene Proteins c-mdm2/genetics , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Alleles , Asian People , Case-Control Studies , China/epidemiology , Cytochrome P-450 CYP2E1/biosynthesis , DNA Glycosylases/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Female , Gene Expression Regulation , Gene Frequency , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-mdm2/biosynthesis , Stomach Neoplasms/epidemiology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology
14.
Oncol Rep ; 34(4): 1667-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26238022

ABSTRACT

Molecular evidence indicates that alterations in genes involved in the maintenance of genome stability may be related to susceptibility to bladder carcinoma. Our goal was to evaluate the prognostic role of base excision repair (BER) genes in a cohort of patients diagnosed with primary urothelial carcinoma of the bladder (UCB). The levels of all APE1, XRCC1 and POLB transcripts were detected by quantitative real-time PCR (qPCR) technique in tumor samples from 52 patients undergoing transurethral resection (TUR) for primary UCB at the Department of Urology, Brazilian National Cancer Institute, Rio de Janeiro. Increased levels of APE1, XRCC1 and POLB transcripts were significantly associated with high-grade tumors when compared to these levels in low-grade tumors (p<0.01) and could be attributed to different mechanisms of transcriptional regulation as a response to tumorigenesis and oxidative stress. By analyzing the collected data in the present study, regardless of pathological grade or stage, univariate analysis revealed that the reduced levels of APE1 transcripts were significantly associated with cancer-specific mortality (p=0.032). Furthermore, the variant genotype (TG/GG) of the APE1 T1349G polymorphism was observed in 75% of a subset of patients who concomitantly experienced reduced levels of the APE1 transcript and death and/or recurrence events. Taken together, our data reinforce the idea that human DNA repair mechanisms must be finely regulated in order to avoid instability leading to tumorigenesis and poor clinical outcomes in UCB patients.


Subject(s)
DNA Polymerase beta/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , Neoplasm Recurrence, Local/genetics , Urinary Bladder Neoplasms/genetics , Aged , Aged, 80 and over , Brazil , DNA Polymerase beta/biosynthesis , DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-Binding Proteins/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Neoplasm Grading , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Polymorphism, Single Nucleotide , Prognosis , Survival Analysis , Urinary Bladder Neoplasms/mortality , Urinary Bladder Neoplasms/pathology , X-ray Repair Cross Complementing Protein 1
15.
Mutat Res ; 779: 96-104, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26164266

ABSTRACT

Peripheral neuropathy is one of the major side effects of treatment with the anticancer drug, cisplatin. One proposed mechanism for this neurotoxicity is the formation of platinum adducts in sensory neurons that could contribute to DNA damage. Although this damage is largely repaired by nuclear excision repair (NER), our previous findings suggest that augmenting the base excision repair pathway (BER) by overexpressing the repair protein APE1 protects sensory neurons from cisplatin-induced neurotoxicity. The question remains whether APE1 contributes to the ability of the NER pathway to repair platinum-damage in neuronal cells. To examine this, we manipulated APE1 expression in sensory neuronal cultures and measured Pt-removal after exposure to cisplatin. When neuronal cultures were treated with increasing concentrations of cisplatin for two or three hours, there was a concentration-dependent increase in Pt-damage that peaked at four hours and returned to near baseline levels after 24h. In cultures where APE1 expression was reduced by ∼ 80% using siRNA directed at APE1, there was a significant inhibition of Pt-removal over eight hours which was reversed by overexpressing APE1 using a lentiviral construct for human wtAPE1. Overexpressing a mutant APE1 (C65 APE1), which only has DNA repair activity, but not its other significant redox-signaling function, mimicked the effects of wtAPE1. Overexpressing DNA repair activity mutant APE1 (226 + 177APE1), with only redox activity was ineffective suggesting it is the DNA repair function of APE1 and not its redox-signaling, that restores the Pt-damage removal. Together, these data provide the first evidence that a critical BER enzyme, APE1, helps regulate the NER pathway in the repair of cisplatin damage in sensory neurons.


Subject(s)
DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Peripheral Nervous System Diseases/genetics , Sensory Receptor Cells/drug effects , Animals , Cisplatin/adverse effects , DNA Damage/drug effects , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Gene Expression Regulation/drug effects , Humans , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Primary Cell Culture , Rats , Sensory Receptor Cells/metabolism
16.
Mutat Res ; 779: 124-33, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26201249

ABSTRACT

Increased paternal age is associated with a greater risk of producing children with genetic disorders originating from de novo germline mutations. Mice mimic the human condition by displaying an age-associated increase in spontaneous mutant frequency in spermatogenic cells. The observed increase in mutant frequency appears to be associated with a decrease in the DNA repair protein, AP endonuclease 1 (APEX1) and Apex1 heterozygous mice display an accelerated paternal age effect as young adults. In this study, we directly tested if APEX1 over-expression in cell lines and transgenic mice could prevent increases in mutagenesis. Cell lines with ectopic expression of APEX1 had increased APEX1 activity and lower spontaneous and induced mutations in the lacI reporter gene relative to the control. Spermatogenic cells obtained from mice transgenic for human APEX1 displayed increased APEX1 activity, were protected from the age-dependent increase in spontaneous germline mutagenesis, and exhibited increased apoptosis in the spermatogonial cell population. These results directly indicate that increases in APEX1 level confer protection against the murine paternal age effect, thus highlighting the role of APEX1 in preserving reproductive health with increasing age and in protection against genotoxin-induced mutagenesis in somatic cells.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Mutagenesis/genetics , Paternal Age , Spermatogenesis/genetics , Animals , Apoptosis/genetics , DNA Repair/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Gene Expression Regulation, Developmental , Germ-Line Mutation , Humans , Male , Mice , Mice, Transgenic , Spermatozoa/metabolism , Spermatozoa/pathology
17.
Am J Physiol Cell Physiol ; 309(5): C296-307, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26108661

ABSTRACT

Tumor-forming endothelial cells have highly elevated levels of Nox-4 that release H2O2 into the nucleus, which is generally not compatible with cell survival. We sought to identify compensatory mechanisms that enable tumor-forming endothelial cells to survive and proliferate under these conditions. Ape-1/ref-1 (Apex-1) is a multifunctional protein that promotes DNA binding of redox-sensitive transcription factors, such as AP-1, and repairs oxidative DNA damage. A validated mouse endothelial cell (EOMA) tumor model was used to demonstrate that Nox-4-derived H2O2 causes DNA oxidation that induces Apex-1 expression. Apex-1 functions as a chaperone to keep transcription factors in a reduced state. In EOMA cells Apex-1 enables AP-1 binding to the monocyte chemoattractant protein-1 (mcp-1) promoter and expression of that protein is required for endothelial cell tumor formation. Intraperitoneal injection of the small molecule inhibitor E3330, which specifically targets Apex-1 redox-sensitive functions, resulted in a 50% decrease in tumor volume compared with mice injected with vehicle control (n = 6 per group), indicating that endothelial cell tumor proliferation is dependent on Apex-1 expression. These are the first reported results to establish Nox-4 induction of Apex-1 as a mechanism promoting endothelial cell tumor formation.


Subject(s)
Cell Proliferation , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Endothelial Cells/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Animals , Benzoquinones/administration & dosage , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Endothelial Cells/drug effects , Endothelial Cells/pathology , Female , Mice , Mice, 129 Strain , Neoplasms/drug therapy , Propionates/administration & dosage
18.
Hum Mol Genet ; 24(16): 4516-29, 2015 Aug 15.
Article in English | MEDLINE | ID: mdl-25976310

ABSTRACT

Ataxia oculomotor apraxia type 1 (AOA1) is an autosomal recessive disease caused by mutations in APTX, which encodes the DNA strand-break repair protein aprataxin (APTX). CoQ10 deficiency has been identified in fibroblasts and muscle of AOA1 patients carrying the common W279X mutation, and aprataxin has been localized to mitochondria in neuroblastoma cells, where it enhances preservation of mitochondrial function. In this study, we show that aprataxin deficiency impairs mitochondrial function, independent of its role in mitochondrial DNA repair. The bioenergetics defect in AOA1-mutant fibroblasts and APTX-depleted Hela cells is caused by decreased expression of SDHA and genes encoding CoQ biosynthetic enzymes, in association with reductions of APE1, NRF1 and NRF2. The biochemical and molecular abnormalities in APTX-depleted cells are recapitulated by knockdown of APE1 in Hela cells and are rescued by overexpression of NRF1/2. Importantly, pharmacological upregulation of NRF1 alone by 5-aminoimidazone-4-carboxamide ribonucleotide does not rescue the phenotype, which, in contrast, is reversed by the upregulation of NRF2 by rosiglitazone. Accordingly, we propose that the lack of aprataxin causes reduction of the pathway APE1/NRF1/NRF2 and their target genes. Our findings demonstrate a critical role of APTX in transcription regulation of mitochondrial function and the pathogenesis of AOA1 via a novel pathomechanistic pathway, which may be relevant to other neurodegenerative diseases.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , DNA-Binding Proteins/deficiency , Down-Regulation , Fibroblasts/metabolism , Mitochondria/metabolism , NF-E2-Related Factor 2/biosynthesis , Nuclear Proteins/deficiency , Nuclear Respiratory Factor 1/biosynthesis , Signal Transduction , Ataxia/genetics , Ataxia/metabolism , Ataxia/pathology , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-Binding Proteins/genetics , Female , Fibroblasts/pathology , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , Humans , Male , Mitochondria/pathology , NF-E2-Related Factor 2/genetics , Nuclear Proteins/genetics , Nuclear Respiratory Factor 1/genetics
19.
Drug Des Devel Ther ; 9: 901-9, 2015.
Article in English | MEDLINE | ID: mdl-25733810

ABSTRACT

INTRODUCTION: Gastric cancer is a fatal malignancy with a rising incidence rate. Effective methods for early diagnosis, monitoring metastasis, and prognosis are currently unavailable for gastric cancer. In this study, we examined the association of programmed death ligand-1 (PD-L1) and apurinic/apyrimidinic endonuclease 1 (APE1) expression with the prognosis of gastric cancer. METHODS: The expressions of PD-L1 and APE1 were detected by immunohistochemistry in 107 cases of human gastric carcinoma. The correlation of PD-L1 and APE1 expression with the clinicopathologic features of gastric carcinoma was analyzed by SPSS version 19.0. RESULTS: The positive expression rates of PD-L1 and APE1 in gastric cancer tissues were 50.5% (54/107) and 86.9% (93/107), respectively. PD-L1 and APE1 positive expressions were significantly associated with depth of invasion, lymph node metastasis, pathological type, overall survival, and higher T stage. Furthermore, the expression of PD-L1 in highly differentiated gastric cancers was higher than that in poorly differentiated cancers (P=0.008). Moreover, the expression of APE1 and PD-L1 in gastric cancers was positively correlated (r=0.336, P<0.01). Multivariate analysis showed that the depth of invasion was a significant prognostic factor (risk ratio 19.91; P=0.000), but there was no significant relationship with PD-L1, APE1, prognosis, and other characteristics. CONCLUSION: The deregulation of PD-L1 and APE1 might contribute to the development and the poor prognosis of gastric cancer. Our findings suggest that high expression of PD-L1 and APE1 is a risk factor of gastric cancer and a new biomarker to predict the prognosis of gastric cancer. Furthermore, our findings suggest that targeting the PD-L1 and APE1 signaling pathways may be a new strategy for cancer immune therapy and targeted therapy for gastric cancer, especially in patients with deep invasion and lymph node metastasis.


Subject(s)
B7-H1 Antigen/metabolism , Carcinogenesis/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Stomach Neoplasms/diagnosis , Stomach Neoplasms/metabolism , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/biosynthesis , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Female , Humans , Immunohistochemistry , Male , Middle Aged , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Up-Regulation
20.
Mol Oncol ; 9(6): 1071-80, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25681012

ABSTRACT

Medulloblastoma (MB) and ependymoma (EP) are the most common pediatric brain tumors, afflicting 3000 children annually. Radiotherapy (RT) is an integral component in the treatment of these tumors; however, the improvement in survival is often accompanied by radiation-induced adverse developmental and psychosocial sequelae. Therefore, there is an urgent need to develop strategies that can increase the sensitivity of brain tumors cells to RT while sparing adjacent healthy brain tissue. Apurinic endonuclease 1 (Ape1), an enzyme in the base excision repair pathway, has been implicated in radiation resistance in cancer. Pharmacological and specificity limitations inherent to small molecule inhibitors of Ape1 have hindered their clinical development. Here we report on a nanoparticle (NP) based siRNA delivery vehicle for knocking down Ape1 expression and sensitizing pediatric brain tumor cells to RT. The NP comprises a superparamagnetic iron oxide core coated with a biocompatible, biodegradable coating of chitosan, polyethylene glycol (PEG), and polyethyleneimine (PEI) that is able to bind and protect siRNA from degradation and to deliver siRNA to the perinuclear region of target cells. NPs loaded with siRNA against Ape1 (NP:siApe1) knocked down Ape1 expression over 75% in MB and EP cells, and reduced Ape1 activity by 80%. This reduction in Ape1 activity correlated with increased DNA damage post-irradiation, which resulted in decreased cell survival in clonogenic assays. The sensitization was specific to therapies generating abasic lesions as evidenced by NP:siRNA not increasing sensitivity to paclitaxel, a microtubule disrupting agent. Our results indicate NP-mediated delivery of siApe1 is a promising strategy for circumventing pediatric brain tumor resistance to RT.


Subject(s)
Brain Neoplasms , DNA Repair , Ependymoma , Gamma Rays , Medulloblastoma , Nanoparticles , Radiation-Sensitizing Agents/pharmacology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Brain Neoplasms/radiotherapy , Cell Line, Tumor , DNA Repair/drug effects , DNA Repair/radiation effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/biosynthesis , Ependymoma/metabolism , Ependymoma/pathology , Ependymoma/radiotherapy , Humans , Medulloblastoma/metabolism , Medulloblastoma/pathology , Medulloblastoma/radiotherapy , Neoplasm Proteins/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...