1
|
Pascual G, Sabater A, Bizzotto J, Seniuk R, Sanchis P, Ledesma-Bazan S, Labanca E, Scorticati C, Mazza O, Vazquez E, Toro A, Prada F, Gueron G, Cotignola J. AR (CAG) n Microsatellite and APEX1 c.444T>G (p.Asp148Glu) Polymorphisms as Independent Prognostic Biomarkers in Prostate Cancer: Insights from an Argentinian Cohort. Cancers (Basel) 2024; 16:3815. [PMID: 39594771 PMCID: PMC11592882 DOI: 10.3390/cancers16223815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/05/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND/OBJECTIVES Prostate cancer (PCa) is the leading malignancy and the third most common cause of cancer-related death in Argentinian men. Predicting outcomes in localized PCa remains difficult due to tumor heterogeneity. In this study, we assessed the impact of AR (CAG)n and APEX1 c.444T>G polymorphisms on biochemical relapse in Argentine patients with localized PCa. METHODS We genotyped blood samples from 123 PCa patients for AR (CAG)n and APEX1 p.Asp148Glu (c.444T>G) polymorphisms. Associations with clinicopathological parameters and biochemical relapse-free survival (BRFS) were assessed. RESULTS AR (CAG)20-23 was associated with a family history of breast/ovarian cancer (p = 0.0469). The combination of AR (CAG)20-23 and APEX1 c.444TT/GG correlated with a 2.89 times higher risk of biochemical relapse (log-rank p = 0.006). Multivariable analysis confirmed AR and APEX1 polymorphisms as independent predictors of biochemical relapse (HR = 3.95, p = 0.002). In patients with PSA levels <10 ng/mL, combined AR (CAG)20-23 and APEX1 c.444TT/GG genotypes were significantly associated with an increased risk of biochemical relapse (HR = 2.61, p = 0.044). Multivariable analysis confirmed the prognostic significance of these genotypes (HR = 3.44, p = 0.02). CONCLUSIONS This study has identified AR (CAG)n and APEX1 c.444T>G polymorphisms as independent predictors of PCa relapse in Argentinian patients, suggesting their potential use in improving prognostic models.
Collapse
Affiliation(s)
- Gaston Pascual
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Agustina Sabater
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Juan Bizzotto
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Rocio Seniuk
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Pablo Sanchis
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Sabrina Ledesma-Bazan
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Estefania Labanca
- Department of Genitourinary Medical Oncology and The David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Carlos Scorticati
- Departamento de Urología, Ciudad Autónoma de Buenos Aires (CABA), Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires C1120AAF, Argentina
| | - Osvaldo Mazza
- Departamento de Urología, Ciudad Autónoma de Buenos Aires (CABA), Hospital de Clínicas José de San Martín, Universidad de Buenos Aires, Buenos Aires C1120AAF, Argentina
| | - Elba Vazquez
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Ayelen Toro
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Federico Prada
- Instituto de Tecnología (INTEC), Universidad Argentina de la Empresa (UADE), Buenos Aires C1073AAO, Argentina
| | - Geraldine Gueron
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| | - Javier Cotignola
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina; (G.P.); (A.S.)
- Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN), CONICET-Universidad de Buenos Aires, Buenos Aires C1428EGA, Argentina
| |
Collapse
|
2
|
Sun Z, Chen G, Wang L, Sang Q, Xu G, Zhang N. APEX1 promotes the oncogenicity of hepatocellular carcinoma via regulation of MAP2K6. Aging (Albany NY) 2022; 14:7959-7971. [PMID: 36205565 PMCID: PMC9596212 DOI: 10.18632/aging.204325] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/17/2022] [Indexed: 12/24/2022]
Abstract
Objective: Apurinic/apyrimidinic endonuclease 1 (APEX1), a key enzyme responsible for DNA base excision repair, has been linked to development and progression of cancers. In this work, we aimed to explore the role of APEX1 in hepatocellular carcinoma (HCC) and elucidate its molecular mechanism. Methods: The expression of APEX1 in HCC tissues and matched adjacent normal tissues (n = 80 cases) was evaluated by immunohistochemistry. Web-based tools UALCAN and the Kaplan-Meier plotter were used to analyze the Cancer Genome Atlas database to compare expression of APEX1 mRNA to 5-year overall survival. APEX1 was stably silenced in two HCC cell lines, Hep 3B and Bel-7402, with shRNA technology. An in vivo tumorigenesis model was established by subcutaneously injecting sh-APEX1-transfected Bel-7402 cells into mice, and tumor growth was determined. We performed high-throughput transcriptome sequencing in sh-APEX1-treated HCC cells to identify the key KEGG signaling pathways induced by silencing of APEX1. Results: APEX1 was significantly upregulated and predicted poor clinical overall survival in HCC patients. Silencing APEX1 inhibited the proliferation of HCC cells in vivo and in vitro, and it repressed invasion and migration and increased apoptosis and the percentage of cells in G1. Differentially expressed genes upon APEX1 silencing included genes involved in TNF signaling. A positive correlation between the expression of APEX1 and MAP2K6 was noted, and overexpressing MAP2K6 overcame cancer-related phenotypes associated with APEX1 silencing. Conclusion: APEX1 enhances the malignant properties of HCC via MAP2K6. APEX1 may represent a valuable prognostic biomarker and therapeutic target in HCC.
Collapse
Affiliation(s)
- Zhipeng Sun
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Institute for Precision Medicine, Tsinghua University, Beijing, China
| | - Guangyang Chen
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Liang Wang
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Qing Sang
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Guangzhong Xu
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing, China
| | - Nengwei Zhang
- Oncology Surgery Department, Beijing Shijitan Hospital, Capital Medical University, Peking University Ninth School of Clinical Medicine, Beijing, China
| |
Collapse
|
3
|
Gong Z, Platek ME, Till C, Goodman PJ, Tangen CM, Platz EA, Neuhouser ML, Thompson IM, Santella RM, Ambrosone CB. Associations Between Polymorphisms in Genes Related to Oxidative Stress and DNA Repair, Interactions With Serum Antioxidants, and Prostate Cancer Risk: Results From the Prostate Cancer Prevention Trial. Front Oncol 2022; 11:808715. [PMID: 35096612 PMCID: PMC8795906 DOI: 10.3389/fonc.2021.808715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/21/2021] [Indexed: 02/03/2023] Open
Abstract
Study of polymorphisms in genes related to the generation and removal of oxidative stress and repair of oxidative DNA damage will lead to new insights into the genetic basis of prostate cancer. In the Prostate Cancer Prevention Trial (PCPT), a double-blind, randomized controlled trial testing finasteride versus placebo for prostate cancer prevention, we intend to investigate the role of oxidative stress/DNA repair mechanisms in prostate cancer etiology and whether these polymorphisms modify prostate cancer risk by interacting with antioxidant status in both placebo and finasteride arms. We evaluated associations of selected candidate polymorphisms in genes in these pathways, and interactions with pre-diagnostic serum antioxidants, and the risk of prostate cancer among 1,598 cases and 1,706 frequency-matched controls enrolled in the PCPT. Odds ratios (ORs) and 95% confidence intervals (CIs) were estimated using multivariable-adjusted logistic regression models. While there were no statistically significant associations observed in the placebo arm, several SNPs were associated with prostate cancer in the finasteride arm. Specifically, APEX1-rs1760944 was associated with increased risk of total prostate cancer (per minor allele: p-trend=0.04). OGG1-rs1052133 was positively (CG/GG vs. CC: OR=1.32, 95% CI: 1.01-1.73) and NOS3-rs1799983 was inversely (per minor allele: p-trend=0.04) associated with risk of low-grade prostate cancer. LIG3-rs1052536 and XRCC1-rs25489 were suggestively associated with reduced risk of high-grade prostate cancer (per minor allele: both p-trend=0.04). In the placebo arm, significant associations were observed among men with higher serum lycopene for APEX1-rs1760944 and NQO1-rs1800566, or higher serum β-cryptoxanthin for ERCC4-rs1800067. In the finasteride arm, stronger associations were observed among men with lower serum lycopene for NOS3-rs1799983, higher serum α-carotene, β-carotene, and β-cryptoxanthin for LIG3-rs1052536, or lower serum retinol for SOD2-rs1799725. These results suggest that germline variations in oxidative stress and DNA repair pathways may contribute to prostate carcinogenesis and that these associations may differ by intraprostatic sex steroid hormone status and be further modified by antioxidant status. Findings provide insights into the complex role of gene, gene-antioxidant and -finasteride interactions in prostate cancer etiology, and thus may lead to the development of preventative strategies.
Collapse
Affiliation(s)
- Zhihong Gong
- Department of Cancer Prevention & Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Mary E Platek
- Department of Cancer Prevention & Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Cathee Till
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Phyllis J Goodman
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Catherine M Tangen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Elizabeth A Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States.,Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, United States
| | - Marian L Neuhouser
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Ian M Thompson
- Department of Urology, University of Texas Health Science Center, San Antonio, TX, United States
| | - Regina M Santella
- Department of Environmental Health Sciences and Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, United States
| | - Christine B Ambrosone
- Department of Cancer Prevention & Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
4
|
Saad AM, Abdel-Megied AES, Elbaz RA, Hassab El-Nabi SE, Elshazli RM. Genetic variants of APEX1 p.Asp148Glu and XRCC1 p.Gln399Arg with the susceptibility of hepatocellular carcinoma. J Med Virol 2021; 93:6278-6291. [PMID: 34289138 DOI: 10.1002/jmv.27217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Accepted: 07/17/2021] [Indexed: 12/30/2022]
Abstract
The DNA repair genes have a crucial function in the base excision repair (BER) mechanism among different cancerous disorders, particularly hepatocellular carcinoma (HCC). The foremost objective of this study is to explore the association of genetic variants of the APEX1 p.Asp148Glu and the XRCC1 p.Gln399Arg with the susceptibility of HCC and to identify the computational bioinformatics frameworks of these missense variants. A total of 250 participants were enrolled in this study, including 150 HCC patients and 100 cancer-free controls. The genomic DNA was characterized and genotyped by applying the PCR-CTPP method. The frequency of the APEX1 (rs1130409*Glu) allele was statistically significant with increased risk of HCC (OR = 1.66, 95% CI = 1.12-2.45), while the XRCC1 (rs25487*Gln) allele conferred a protection against the progression of HCC (OR = 0.64, 95% CI = 0.42-0.96). Furthermore, HCC patients carrying the APEX1 p.Asp148Glu and the XRCC1 p.Gln399Arg variants indicated no significant difference with the clinical, and laboratory parameters (p > .05). Our findings confirmed that the APEX1 p.Asp148Glu variant was associated with increased risk of HCC, while the XRCC1 p.Gln399Arg variant revealed protection against the development of HCC.
Collapse
Affiliation(s)
- Ahmad M Saad
- Biochemistry Section, Department of Chemistry, Faculty of Science, Menoufia University, Menoufia, Egypt
| | | | - Rizk A Elbaz
- Genetic Unit, Children Hospital, Mansoura University, Mansoura, Egypt
| | | | - Rami M Elshazli
- Biochemistry and Molecular Genetics Unit, Department of Basic Sciences, Faculty of Physical Therapy, Horus University-Egypt, New Damietta, Egypt
| |
Collapse
|
5
|
Tan A, Ruan P, Sun P. APEX1/miR-24 axis: a promising therapeutic target in endometriosis. Arch Gynecol Obstet 2021; 304:131-141. [PMID: 33502561 DOI: 10.1007/s00404-021-05963-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 11/17/2020] [Indexed: 11/25/2022]
Abstract
PURPOSE The present work aimed to explore the aberrant expression of APEX1 in endometrial stromal cells (ESC) and the underlying mechanisms. METHODS The levels of APEX1 and miR-24 in endometriosis tissues were tested by qRT-PCR and Western blot. After cell transfection, cells were correspondingly classified into pcDNA3.1-NC, sh-NC, mimic NC, inhibitor NC, pcDNA3.1-APEX1, sh-APEX1, miR-24 mimic, miR-24 inhibitor, sh-NC + inhibitor NC, inhibitor-NC + sh-APEX1, sh-NC + miR-24 inhibitor, pcDNA3.1-NC + mimic NC, mimic NC + pcDNA3.1-APEX1 and pcDNA3.1-NC + miR-24 mimic group. Besides, cell proliferation, apoptosis in addition to apoptosis-related proteins Bax, Bcl-2 and cleaved-casase-3 were analyzed by BrdU assay, flow cytometry (FCM) and Western blot assays, respectively. Additionally, RIP assay was conducted to determine the interaction between pri-miR-24 and miR-24. RESULTS APEX1 and miR-24 were highly expressed in endometriosis tissues. Overexpression of APEX1 and miR-24 potentiates ESC proliferation and inhibits apoptosis, while those effects could be reversed by APEX1 and miR-24 silencing. Meanwhile, APEX1 and miR-24 could elevate ESC apoptosis-related proteins Bax and cleaved-caspase-3 and decrease Bcl-2 expression. Importantly, APEX1 was positively correlated with miR-24 expression. CONCLUSION APEX1 promotes ESC proliferation and inhibits apoptosis by upregulating miR-24 expression.
Collapse
Affiliation(s)
- Aili Tan
- Department of Obstetrics and Gynecology, Wuhan University Renmin Hospital, Wuhan, 430060, Hubei, China
| | - Peng Ruan
- Department of Oncology, Wuhan University Renmin Hospital, No. 99, Zhangzhidong Road, Wuchang District, Wuhan, 430060, Hubei, China.
| | - Pengxing Sun
- Department of Obstetrics and Gynecology, Wuhan University Renmin Hospital, Wuhan, 430060, Hubei, China
| |
Collapse
|
6
|
AP endonuclease 1 (Apex1) influences brain development linking oxidative stress and DNA repair. Cell Death Dis 2019; 10:348. [PMID: 31024003 PMCID: PMC6484078 DOI: 10.1038/s41419-019-1578-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 03/27/2019] [Accepted: 04/04/2019] [Indexed: 12/13/2022]
Abstract
Brain and neurons are particularly sensitive to reactive oxygen species (ROS). Oxidative damage from ROS results in increased 8-oxoguanine in DNA followed by repair through the base excision repair (BER) pathway. We reported earlier that AP endonuclease 1 (Apex1) not only participates directly in BER but also regulates transcription factor Creb1. Here, we investigated how Apex1 affects brain to respond effectively to oxidative damage during zebrafish development. Loss of Apex1 resulted in increased ROS, 8-oxoguanine, and abasic sites as well as loss of Ogg1, which recognizes 8-oxoguanine and is required for its repair. Moreover, knock-down of Apex1 not only resulted in reduction of expression of several major proteins in the BER pathway (Polb and Ogg1), and it also resulted in maldistribution and loss of four key brain transcription factors (fezf2, otx2, egr2a, and pax2a), leading to abnormal brain development. These results were independent of p53 protein level. In contrast, exposure to exogenous H2O2 resulted in increased transcription and protein of Apex1 along with other BER components, as well as Creb1. Taken together, these results indicate that oxidative stress increased when the level of Apex1 was reduced, revealing a novel pathway of how Apex1 manages oxidative stress in developing brain.
Collapse
|
7
|
Cypriano AS, Alves G, Ornellas AA, Scheinkman J, Almeida R, Scherrer L, Lage C. Relationship between XPD, RAD51, and APEX1 DNA repair genotypes and prostate cancer risk in the male population of Rio de Janeiro, Brazil. Genet Mol Biol 2017; 40:751-758. [PMID: 29111564 PMCID: PMC5738611 DOI: 10.1590/1678-4685-gmb-2017-0039] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 07/24/2017] [Indexed: 12/26/2022] Open
Abstract
Susceptibility to cancer ensues in individuals carrying malfunctioning DNA repair
mechanisms. The impact of Single Nucleotide Polymorphisms (SNPs) in key DNA
repair mechanisms on risk for prostate cancer was investigated in this
case-control study. Samples consisted of 110 patients with confirmed prostate
cancer and 200 unaffected men, from Rio de Janeiro, Brazil.
XPD/Lys751Gln (rs13181), APEX1/Asp148Glu
(rs1130409), and RAD51/G135C (rs1801320) SNPs were analyzed by
PCR-RFLP. Allelic and genotypic frequencies were calculated and compared by
Chi-Square test. The association between SNPs and clinical/epidemiological data
was considered significant by Odds Ratio analysis, with IC95% and a
p-value≤0.05. Only the XPD/Lys751Gln SNP significantly
increased susceptibility to disease in southeastern Brazilian men, with
p≤0.001 [OR=2.36 (1.46-3.84)], with no association with
APEX1 or RAD51 SNPs. Combined
XPD+RAD51 SNPs were highly associated with
the disease, p≤0.005 [OR=3.40 (1.32-9.20)]. A Chi-Square
significant association between XPD/Lys751Gln and Gleason score
was also observed (OR=9.31; IC95%=1.19–428.0; p=0.022).
Epidemiological inquiries revealed that exposure to pesticides significantly
impacted the risk for prostate cancer in this population. DNA repair
dysfunctions seem to prevail among workers exposed to chemical byproducts to
cancer in this specific tissue. Non-invasive genotyping SNPs may help assessment
of prostate cancer risk in environmentally exposed populations.
Collapse
Affiliation(s)
- Ana Sheila Cypriano
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal de Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Gilda Alves
- Instituto Nacional de Câncer, Hospital do Câncer I, Rio de Janeiro, RJ, Brazil.,Laboratório de Marcadores Circulantes, Departamento de Patologia, Faculdade de Ciências Médicas, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Antonio Augusto Ornellas
- Instituto Nacional de Câncer, Hospital do Câncer I, Rio de Janeiro, RJ, Brazil.,Hospital Mário Kroeff, Rio de Janeiro, RJ, Brazil
| | | | | | | | - Claudia Lage
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal de Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
8
|
Li G, Yang M, Li X, Deng S. Comprehensive assessment of the association between estrogen receptor of alpha polymorphisms and the risk of prostate cancer: evidence from a meta-analysis. Oncotarget 2017; 8:102310-102320. [PMID: 29254246 PMCID: PMC5731956 DOI: 10.18632/oncotarget.21117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Accepted: 08/27/2017] [Indexed: 11/25/2022] Open
Abstract
We performed a meta analysis to access the relationship of estrogen receptor of alpha (ESRα) polymorphisms with the risk of prostate cancer (PC). Twenty-four case-control studies (including 5477 cases and 10708 controls) were recruited for meta-analysis. The strongest association with the risk of PC was observed between ESRα rs9340799 and rs2234693 under the two genotypic models of allele and codominance in the overall population (p < 0.05). Under the subgroup analysis of ethnicity, we observed that ESRα rs9340799 was significantly associated with the susceptibility to PC in European population (AvsG, p = 0.000; AAvsGG, p = 0.002), while there was no difference in Asian (AvsG, p = 0.493; AAvsGG, p = 0.736) or African population (AvsG, p = 0.800; AAvsGG, p = 0.788). The results also showed that significant association between rs2234693 and the susceptibility to PC in European (CvsT, p = 0.004; CCvsTT, p = 0.001) and Asian population (CvsT, p = 0.004; CCvsTT, p = 0.003), but not in African population (CvsT, p = 0.636; CCvsTT, p = 0.669). The meta-analysis indicated that ESRα rs9340799 and rs2234693 might contribute to susceptibility and development of PC in European population.
Collapse
Affiliation(s)
- Guang Li
- Laboratory of Forensic Medicine and Bioinformatics, Chongqing Medical University, Chongqing, China
| | - Meng Yang
- Research Department, Children Hospital of Chongqing Medical University, Chongqing, China
| | - Xian Li
- Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing, China
| | - Shixiong Deng
- Laboratory of Forensic Medicine and Bioinformatics, Chongqing Medical University, Chongqing, China
| |
Collapse
|
9
|
Xiao X, Yang Y, Ren Y, Zou D, Zhang K, Wu Y. rs1760944 Polymorphism in the APE1 Region is Associated with Risk and Prognosis of Osteosarcoma in the Chinese Han Population. Sci Rep 2017; 7:9331. [PMID: 28839218 PMCID: PMC5570937 DOI: 10.1038/s41598-017-09750-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 06/16/2017] [Indexed: 01/16/2023] Open
Abstract
The effects of single nucleotide polymorphisms (SNPs) at APE1 have been investigated in several types of cancer. However, no reports of the association of APE1 polymorphisms with osteosarcoma (OS) have been published. The present study was designed to determine whether APE1 polymorphisms (rs1130409, rs1760944, rs1760941, rs2275008, rs17111750) are associated with OS. A 2-stage case-control study was performed in a total of 378 OS patients and 616 normal controls. Individuals carrying TG and GG genotypes had significantly lower risk of developing OS than those with the WT genotype TT at rs1760944 (OR = 0.65, 95%CI 0.49–0.86; OR = 0.50, 95%CI 0.34–0.74, respectively). OS patients with allele G at rs1760944 were less susceptible to low differentiation tumor and metastasis (OR = 0.73, 95%CI 0.54–0.98; OR = 0.63, 95%CI 0.43–0.92, respectively). Kaplan-Meier curves and log-rank results revealed that OS patients harboring genotype GG and G allele at rs1760944 had better survival (P < 0.001 for both). In addition, the APE1 protein was underexpressed in individuals who carried G allele at rs1760944. This study suggested that APE1 rs1760944 polymorphism is associated with decreased risk of developing OS and better survival of OS patients.
Collapse
Affiliation(s)
- Xing Xiao
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yun Yang
- School of Medicine, Shandong University, Jinan, Shandong, China
| | - Yanjun Ren
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Debo Zou
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Kaining Zhang
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yingguang Wu
- Department of Spine Surgery, Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China.
| |
Collapse
|