1
|
Wang R, Lin L, Han Y, Li Z, Zhen J, Zhang Y, Sun F, Lu Y. Exosome-delivered miR-153 from Trichinella spiralis promotes apoptosis of intestinal epithelial cells by downregulating Bcl2. Vet Res 2023; 54:52. [PMID: 37381058 DOI: 10.1186/s13567-023-01186-6] [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: 04/25/2023] [Accepted: 05/22/2023] [Indexed: 06/30/2023] Open
Abstract
Trichinellosis, a helminthic zoonosis, exhibits a cosmopolitan distribution and is a public health concern. In previous studies, it was reported that the exosomes secreted by Trichinella spiralis larvae (TsExos) largely affected cell biological activities. miRNAs, as exosome-delivered cargoes, affect the biological activities of the host by targeting genes. The present study aimed to elucidate the mechanisms by which miRNAs interact with intestinal epithelial cells. First, a miRNA library of TsExos was constructed; then, based on high-throughput miRNA sequencing results, miR-153 and its predicted target genes, namely, Agap2, Bcl2 and Pten, were selected for follow-up studies. The dual-luciferase reporter assays revealed that miR-153 directly targeted Bcl2 and Pten. Furthermore, real-time qPCR and Western blotting revealed that only Bcl2 was downregulated by TsExo-delivered miR-153 in porcine intestinal epithelial cells (IPEC-J2). Bcl2, an important antiapoptotic protein, plays an essential role in cell apoptosis as a common intersecting molecule of various signal transduction pathways. Therefore, we hypothesized that miR-153 derived from TsExos causes cell apoptosis by targeting Bcl2. The results suggested that miR-153 could induce apoptosis, reduce mitochondrial membrane potential, affect cell proliferation, and cause damage and substantial oxidative stress. Furthermore, miR-153 coincubated with IPEC-J2 cells stimulated the accumulation of the proapoptotic proteins Bax and Bad, which belong to the Bcl2 family of proteins, and the apoptosis-implementing proteins Caspase 9 and Caspase 3. Moreover, studies have suggested that miR-153 can promote apoptosis by regulating the MAPK and p53 signalling pathways involved in apoptosis. Thus, exosome-mediated miR-153 delivery secreted by T. spiralis could induce apoptosis and affect the MAPK and p53 signalling pathways by downregulating Bcl2 in IPEC-J2 cells. The study highlights the mechanisms underlying the invasion of T. spiralis larva.
Collapse
Affiliation(s)
- Ruibiao Wang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Lihao Lin
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yang Han
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Zhixin Li
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jingbo Zhen
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yuheng Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Feng Sun
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yixin Lu
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China.
| |
Collapse
|
2
|
Liao CY, Yang SF, Wu TJ, Chang H, Huang CYF, Liu YF, Wang CH, Liou JC, Hsu SL, Lee H, Sheu GT, Chang JT. Novel function of PERP-428 variants impacts lung cancer risk through the differential regulation of PTEN/MDM2/p53-mediated antioxidant activity. Free Radic Biol Med 2021; 167:307-320. [PMID: 33731308 DOI: 10.1016/j.freeradbiomed.2021.02.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 02/04/2021] [Accepted: 02/10/2021] [Indexed: 01/11/2023]
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Identifying genetic risk factors and understanding their mechanisms will help reduce lung cancer incidence. The p53 apoptosis effect is related to PMP-22 (PERP), a tetraspan membrane protein, and an apoptotic effector protein downstream of p53. Although historically considered a tumor suppressor, PERP is highly expressed in lung cancers. Stable knockdown of PERP expression induces CL1-5 and A549 lung cancer cell death, but transient knockdown has no effect. Interestingly, relative to the PERP-428GG genotype, PERP-428CC was associated with the highest lung cancer risk (OR = 5.38; 95% CI = 2.12-13.65, p < 0.001), followed by the PERP-428CG genotype (OR = 2.34; 95% CI = 1.55-3.55, p < 0.001). Ectopic expression of PERP-428G, but not PERP-428C, protects lung cancer cells against ROS-induced DNA damage. Mechanistically, PERP-428 SNPs differentially regulate p53 protein stability. p53 negatively regulates the expression of the antioxidant enzymes catalase (CAT) and glutathione reductase (GR), thereby modulating redox status. p53 protein stability is higher in PERP-428C-expressing cells than in PERP-428G-expressing cells because MDM2 expression is decreased and p53 Ser20 phosphorylation is enhanced in PERP-428C-expressing cells. The MDM2 mRNA level is decreased in PERP-428C-expressing cells via PTEN-mediated downregulation of the MDM2 constitutive p1 promoter. This study reveals that in individuals with PERP-428CC, CAT/GR expression is decreased via the PTEN/MDM2/p53 pathway. These individuals have an increased lung cancer risk. Preventive antioxidants and avoidance of ROS stressors are recommended to prevent lung cancer or other ROS-related chronic diseases.
Collapse
Affiliation(s)
- Chen-Yi Liao
- Institute of Medicine, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan; CSMU Lung Cancer Research Center, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Ting-Jian Wu
- Institute of Medicine, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Han Chang
- Department of Pathology, China Medical University Hospital, No. 91, Hsueh-Shih Road, Taichung, 40402 Taiwan.
| | - Chi-Ying F Huang
- Institute of Biopharmaceutical Sciences, National Yang Ming University, No. 155, Sec. 2, Linong Street, Taipei, 11221, Taiwan.
| | - Yu-Fan Liu
- Department of Biomedical Sciences, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Chi-Hsiang Wang
- Institute of Medical and Molecular Toxicology, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Jhong-Chio Liou
- Institute of Medical and Molecular Toxicology, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Shih-Lan Hsu
- Department of Education & Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Sec. 4, Taichung 407204, Taiwan.
| | - Huei Lee
- Graduate Institute of Cancer Biology and Drug Discovery, Taipei Medical University, 250 Wu-Hsing Street, Taipei, 11031, Taiwan.
| | - Gwo-Tarng Sheu
- Institute of Medicine, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan; CSMU Lung Cancer Research Center, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan; Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| | - Jinghua Tsai Chang
- Institute of Medicine, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan; CSMU Lung Cancer Research Center, Chung Shan Medical University, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan; Divisions of Medical Oncology and Pulmonary Medicine, Chung Shan Medical University Hospital, 110 Sec. 1, Chien-Kuo N. Road, Taichung, 40203, Taiwan.
| |
Collapse
|
3
|
Roberts O, Paraoan L. PERP-ing into diverse mechanisms of cancer pathogenesis: Regulation and role of the p53/p63 effector PERP. Biochim Biophys Acta Rev Cancer 2020; 1874:188393. [PMID: 32679166 DOI: 10.1016/j.bbcan.2020.188393] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/11/2020] [Accepted: 07/12/2020] [Indexed: 12/20/2022]
Abstract
The tetraspan plasma membrane protein PERP (p53 apoptosis effector related to PMP22) is a lesser-known transcriptional target of p53 and p63. A member of the PMP22/GAS3/EMP membrane protein family, PERP was originally identified as a p53 target specifically trans-activated during apoptosis, but not during cell-cycle arrest. Several studies have since shown downregulation of PERP expression in numerous cancers, suggesting that PERP is a tumour suppressor protein. This review focusses on the important advances made in elucidating the mechanisms regulating PERP expression and its function as a tumour suppressor in diverse human cancers, including breast cancer and squamous cell carcinoma. Investigating PERP's role in clinically-aggressive uveal melanoma has revealed that PERP engages a positive-feedback loop with p53 to regulate its own expression, and that p63 is required beside p53 to achieve pro-apoptotic levels of PERP in this cancer. Furthermore, the recent discovery of the apoptosis-mediating interaction of PERP with SERCA2b at the plasma membrane-endoplasmic reticulum interface demonstrates a novel mechanism of PERP stabilisation, and how PERP can mediate Ca2+ signalling to facilitate apoptosis. The multi-faceted role of PERP in cancer, involving well-documented functions in mediating apoptosis and cell-cell adhesion is discussed, alongside PERP's emerging roles in epithelial-mesenchymal transition, and PERP crosstalk with inflammation signalling pathways, and other signalling pathways. The potential for restoring PERP expression as a means of cancer therapy is also considered.
Collapse
Affiliation(s)
- Owain Roberts
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Luminita Paraoan
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| |
Collapse
|
4
|
Wu W, Liu D, Jiang S, Zhang K, Zhou H, Lu Q. Polymorphisms in gene MMP-2 modify the association of cadmium exposure with hypertension risk. ENVIRONMENT INTERNATIONAL 2019; 124:441-447. [PMID: 30684802 DOI: 10.1016/j.envint.2019.01.041] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 12/22/2018] [Accepted: 01/15/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND Cadmium exposure has been inconsistently related with hypertension. However, epidemiologic data on the genetic susceptibility to the hypertensive effect of cadmium exposure are limited. OBJECTIVES We investigated whether the associations between cadmium exposure and hypertension risk differed by genetic polymorphisms in MMPs genes. METHODS The present study of 497 hypertension cases and 497 healthy controls was conducted in a Chinese population. Urinary cadmium levels were measured with inductively coupled plasma-mass spectrometer (ICP-MS). Multivariable logistic regression models were analyzed after controlling major confounders. RESULTS Within the multivariable logistic regression models, compared with the lowest tertile, the highest tertile of urinary cadmium had a 1.33-fold (95% CI: 1.01, 1.93) increased risk of hypertension. Carriers of rs243865 T allele and rs243866 A allele in MMP-2 were suggested to have increased risks of hypertension. The associations of urinary cadmium with hypertension risk were modified by rs14070 (P-value for interaction = 0.022) and rs7201 (P-value for interaction = 0.009) in gene MMP-2. Positively significant trends for increasing odds of hypertension with cadmium levels were observed among the wild types of rs14070 and rs7201, respectively. CONCLUSIONS Increasing urinary cadmium concentrations were positively associated with hypertension risk in a Chinese population, and the associations were modified by polymorphism of rs14070 and rs7201 in gene MMP-2.
Collapse
Affiliation(s)
- Weixiang Wu
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China; Department of Clinical Laboratory, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Dayang Liu
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Shunli Jiang
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Ke Zhang
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China; Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, #1277 Jiefang Road, Wuhan, Hubei 430022, China
| | - Hao Zhou
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | - Qing Lu
- State Key Laboratory of Environment Health (Incubation), Key Laboratory of Environment and Health, Ministry of Education, Key Laboratory of Environment and Health (Wuhan), Ministry of Environmental Protection, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China.
| |
Collapse
|
5
|
TNFR1 single nucleotide polymorphisms are not associated with cervical HPV-induced pre-malignant lesion but regulate in situ cervical TNFR1 expression. Oncotarget 2019; 10:953-965. [PMID: 30847024 PMCID: PMC6398171 DOI: 10.18632/oncotarget.26627] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/14/2019] [Indexed: 11/25/2022] Open
Abstract
TNF-α is involved in HPV infection control by triggering cell signaling through binding in specific receptors TNFR1 and TNFR2. Genetic polymorphisms in these receptors may influence TNF-α signaling. Herein, we investigated TNFR1 rs767455 and rs2234649 single nucleotide polymorphisms, and TNFR1 protein expression in cervical squamous intraepithelial lesions (SIL) to identify their role in cervical pre-malignant development. SIL patients (n = 179) and healthy volunteers (n = 227) were enrolled for TNFR1 genotyping analysis by PCR-RFLP in blood samples and TNFR1 protein expression in cervical tissue by immunohistochemistry. No statistical differences regard genotypes and allelic frequencies for both polymorphisms were observed. Cervical TNFR1-expressing cells were rare in epithelium and basal layer regardless the groups. However, a progressive increase in infiltrating cells was observed in the stromal area, mainly in high SIL (HSIL) group compared to low SIL (LSIL, p < 0.001) and control (p < 0.001) groups. TNFR1-expressing cells frequency was higher in TNFR1 rs767455AG/GG (p < 0.001), and in rs2234649AA (p < 0.001) genotypes carries in HSIL subgroup. These data indicated that TNFR1-expression is abrogated in cervical epithelium, where HPV-induced pre-malignant lesion occurs, increasing its frequency in inflammatory cells in stroma, and is genetically controlled by TNFR1 rs767455AG/GG and rs234649AA genotypes. These biomarkers may be useful to identify cervical precancerous lesions progression.
Collapse
|
6
|
Dong J, Levine DM, Buas MF, Zhang R, Onstad L, Fitzgerald RC, Corley DA, Shaheen NJ, Lagergren J, Hardie LJ, Reid BJ, Iyer PG, Risch HA, Caldas C, Caldas I, Pharoah PD, Liu G, Gammon MD, Chow WH, Bernstein L, Bird NC, Ye W, Wu AH, Anderson LA, MacGregor S, Whiteman DC, Vaughan TL, Thrift AP. Interactions Between Genetic Variants and Environmental Factors Affect Risk of Esophageal Adenocarcinoma and Barrett's Esophagus. Clin Gastroenterol Hepatol 2018; 16:1598-1606.e4. [PMID: 29551738 PMCID: PMC6162842 DOI: 10.1016/j.cgh.2018.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 02/23/2018] [Accepted: 03/09/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Genome-wide association studies (GWAS) have identified more than 20 susceptibility loci for esophageal adenocarcinoma (EA) and Barrett's esophagus (BE). However, variants in these loci account for a small fraction of cases of EA and BE. Genetic factors might interact with environmental factors to affect risk of EA and BE. We aimed to identify single nucleotide polymorphisms (SNPs) that may modify the associations of body mass index (BMI), smoking, and gastroesophageal reflux disease (GERD), with risks of EA and BE. METHODS We collected data on single BMI measurements, smoking status, and symptoms of GERD from 2284 patients with EA, 3104 patients with BE, and 2182 healthy individuals (controls) participating in the Barrett's and Esophageal Adenocarcinoma Consortium GWAS, the UK Barrett's Esophagus Gene Study, and the UK Stomach and Oesophageal Cancer Study. We analyzed 993,501 SNPs in DNA samples of all study subjects. We used standard case-control logistic regression to test for gene-environment interactions. RESULTS For EA, rs13429103 at chromosome 2p25.1, near the RNF144A-LOC339788 gene, showed a borderline significant interaction with smoking status (P = 2.18×10-7). Ever smoking was associated with an almost 12-fold increase in risk of EA among individuals with rs13429103-AA genotype (odds ratio=11.82; 95% CI, 4.03-34.67). Three SNPs (rs12465911, rs2341926, rs13396805) at chromosome 2q23.3, near the RND3-RBM43 gene, interacted with GERD symptoms (P = 1.70×10-7, P = 1.83×10-7, and P = 3.58×10-7, respectively) to affect risk of EA. For BE, rs491603 at chromosome 1p34.3, near the EIF2C3 gene, and rs11631094 at chromosome 15q14, at the SLC12A6 gene, interacted with BMI (P = 4.44×10-7) and pack-years of smoking history (P = 2.82×10-7), respectively. CONCLUSION The associations of BMI, smoking, and GERD symptoms with risks of EA and BE appear to vary with SNPs at chromosomes 1, 2, and 15. Validation of these suggestive interactions is warranted.
Collapse
Affiliation(s)
- Jing Dong
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas
| | - David M Levine
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington
| | - Matthew F Buas
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington; Department of Cancer Prevention and Control, Roswell Park Cancer Institute, Buffalo, New York
| | - Rui Zhang
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, Washington
| | - Lynn Onstad
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Rebecca C Fitzgerald
- Medical Research Council Cancer Unit, Hutchison-MRC Research Centre, University of Cambridge, Cambridge, United Kingdom
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, California; San Francisco Medical Center, Kaiser Permanente Northern California, San Francisco, California
| | - Nicholas J Shaheen
- Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Jesper Lagergren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; School of Cancer Sciences, King's College London, London, United Kingdom
| | - Laura J Hardie
- Division of Epidemiology, LICAMM, School of Medicine, University of Leeds, Leeds, United Kingdom
| | - Brian J Reid
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Prasad G Iyer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Carlos Caldas
- Cancer Research UK, Cambridge Institute, Cambridge, United Kingdom; Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Isabel Caldas
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom
| | - Paul D Pharoah
- Department of Oncology, University of Cambridge, Cambridge, United Kingdom; Department of Public Health and Primary Care, University of Cambridge, Cambridge, United Kingdom
| | - Geoffrey Liu
- Pharmacogenomic Epidemiology, Ontario Cancer Institute, Toronto, Canada
| | - Marilie D Gammon
- Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina
| | - Wong-Ho Chow
- Department of Epidemiology, MD Anderson Cancer Center, Houston, Texas
| | - Leslie Bernstein
- Department of Population Sciences, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, California
| | - Nigel C Bird
- Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Anna H Wu
- Department of Preventive Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Lesley A Anderson
- Centre for Public Health, Queen's University Belfast, Belfast, United Kingdom
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - David C Whiteman
- Cancer Control, QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Thomas L Vaughan
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Aaron P Thrift
- Section of Epidemiology and Population Sciences, Department of Medicine, Baylor College of Medicine, Houston, Texas; Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
7
|
Granata S, Dalla Gassa A, Carraro A, Brunelli M, Stallone G, Lupo A, Zaza G. Sirolimus and Everolimus Pathway: Reviewing Candidate Genes Influencing Their Intracellular Effects. Int J Mol Sci 2016; 17:ijms17050735. [PMID: 27187382 PMCID: PMC4881557 DOI: 10.3390/ijms17050735] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/21/2016] [Accepted: 05/06/2016] [Indexed: 02/07/2023] Open
Abstract
Sirolimus (SRL) and everolimus (EVR) are mammalian targets of rapamycin inhibitors (mTOR-I) largely employed in renal transplantation and oncology as immunosuppressive/antiproliferative agents. SRL was the first mTOR-I produced by the bacterium Streptomyces hygroscopicus and approved for several medical purposes. EVR, derived from SRL, contains a 2-hydroxy-ethyl chain in the 40th position that makes the drug more hydrophilic than SRL and increases oral bioavailability. Their main mechanism of action is the inhibition of the mTOR complex 1 and the regulation of factors involved in a several crucial cellular functions including: protein synthesis, regulation of angiogenesis, lipid biosynthesis, mitochondrial biogenesis and function, cell cycle, and autophagy. Most of the proteins/enzymes belonging to the aforementioned biological processes are encoded by numerous and tightly regulated genes. However, at the moment, the polygenic influence on SRL/EVR cellular effects is still not completely defined, and its comprehension represents a key challenge for researchers. Therefore, to obtain a complete picture of the cellular network connected to SRL/EVR, we decided to review major evidences available in the literature regarding the genetic influence on mTOR-I biology/pharmacology and to build, for the first time, a useful and specific “SRL/EVR genes-focused pathway”, possibly employable as a starting point for future in-depth research projects.
Collapse
Affiliation(s)
- Simona Granata
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| | | | - Amedeo Carraro
- Liver Transplant Unit, Department of General Surgery and Odontoiatrics, University/Hospital of Verona, 37126 Verona, Italy.
| | - Matteo Brunelli
- Department of Pathology and Diagnostics, University of Verona, Azienda Ospedaliera Universitaria Integrata, 37126 Verona, Italy.
| | - Giovanni Stallone
- Nephrology, Dialysis and Transplantation Unit, University of Foggia, 71122 Foggia, Italy.
| | - Antonio Lupo
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| | - Gianluigi Zaza
- Renal Unit, Department of Medicine, University/Hospital of Verona, 37126 Verona, Italy.
| |
Collapse
|
8
|
Datta S, Ray A, Singh R, Mondal P, Basu A, De Sarkar N, Majumder M, Maiti G, Baral A, Jha GN, Mukhopadhyay I, Panda C, Chowdhury S, Ghosh S, Roychoudhury S, Roy B. Sequence and expression variations in 23 genes involved in mitochondrial and non-mitochondrial apoptotic pathways and risk of oral leukoplakia and cancer. Mitochondrion 2015; 25:28-33. [DOI: 10.1016/j.mito.2015.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Revised: 08/15/2015] [Accepted: 09/18/2015] [Indexed: 12/25/2022]
|
9
|
Wan X, Li X, Yang J, Lv W, Wang Q, Chen Y, Li Y. Genetic association between PIK3CA gene and oral squamous cell carcinoma: a case control study conducted in Chongqing, China. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:13360-13366. [PMID: 26722541 PMCID: PMC4680486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/23/2015] [Indexed: 06/05/2023]
Abstract
PIK3CA has been shown to be involved in many malignant tumors. This study was designed to determine the expression level of PIK3CA in oral squamous cell carcinoma (OSCC) and the association of gene polymorphisms of PIK3CA with OSCC in Chinese population. The expression of PIK3CA was detected by real-time PCR in tumor and pericarcinomatous tissues of 10 OSCC patients. Nine single-nucleotide polymorphisms (SNPs) of PIK3CA (rs1607237, rs17849079, rs2677764, rs2699887, rs4855094, rs4975596, rs6443624, rs7651265 and rs7736074) in blood of 113 OSCC patients and 184 normal controls were genotyped using matrix-assisted laser desorption/ionization time of flight mass spectrometry (MALDI-TOF MS) assay. The gene expression of PIK3CA was significantly higher in tumor tissues of OSCC patients than that in pericarcinomatous tissues (P = 0.012). An increased frequency of the C allele of PIK3CA rs1607237 was observed in OSCC patients as compared with controls; However, the significance was lost after Bonferroni correction (P = 0.048, pc = 0.576). In further stratification analysis, although the frequencies of PIK3CA rs4975596 A allele in male patients and rs1607237 C allele in female patients were increased (P = 0.032, P = 0.020, respectively), the significance was also missing when Bonferroni correction was performed (P c = 0.384, (P c = 0.24, respectively). The prevalence of other SNPs of PIK3CA did not differ between OSCC patients and controls. The expression of PIK3CA was increased in OSCC tumors; however, none of the nine tested SNPs of PIK3CA was associated with susceptibility to OSCC in the studied population.
Collapse
Affiliation(s)
- Xiaoxiao Wan
- Departments of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| | - Xian Li
- Departments of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| | - Junyan Yang
- Departments of Laboratory Medicine, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| | - Wei Lv
- Departments of Laboratory Medicine, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| | - Qiming Wang
- Departments of Laboratory Medicine, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| | - Ying Chen
- Departments of Laboratory Medicine, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| | - Yong Li
- Departments of Oral and Maxillofacial Surgery, Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical UniversityChongqing 401147, China
| |
Collapse
|
10
|
Dai JY, de Dieu Tapsoba J, Buas MF, Onstad LE, Levine DM, Risch HA, Chow WH, Bernstein L, Ye W, Lagergren J, Bird NC, Corley DA, Shaheen NJ, Wu AH, Reid BJ, Hardie LJ, Whiteman DC, Vaughan TL. A newly identified susceptibility locus near FOXP1 modifies the association of gastroesophageal reflux with Barrett's esophagus. Cancer Epidemiol Biomarkers Prev 2015; 24:1739-47. [PMID: 26377193 DOI: 10.1158/1055-9965.epi-15-0507] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/19/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Important risk factors for esophageal adenocarcinoma and its precursor, Barrett's esophagus, include gastroesophageal reflux disease, obesity, and cigarette smoking. Recently, genome-wide association studies have identified seven germline single-nucleotide polymorphisms (SNP) that are associated with risk of Barrett's esophagus and esophageal adenocarcinoma. Whether these genetic susceptibility loci modify previously identified exposure-disease associations is unclear. METHODS We analyzed exposure and genotype data from the BEACON Consortium discovery phase GWAS, which included 1,516 esophageal adenocarcinoma case patients, 2,416 Barrett's esophagus case patients, and 2,187 control participants. We examined the seven newly identified susceptibility SNPs for interactions with body mass index, smoking status, and report of weekly heartburn or reflux. Logistic regression models were used to estimate ORs for these risk factors stratified by SNP genotype, separately for Barrett's esophagus and esophageal adenocarcinoma. RESULTS The odds ratio for Barrett's esophagus associated with at least weekly heartburn or reflux varied significantly with the presence of at least one minor allele of rs2687201 (nominal P = 0.0005, FDR = 0.042). ORs (95% CIs) for weekly heartburn or reflux among participants with 0, 1, or 2 minor alleles of rs2687201 were 6.17 (4.91-7.56), 3.56 (2.85-4.44), and 3.97 (2.47-6.37), respectively. No statistically significant interactions were observed for smoking status and body mass index. CONCLUSION Reflux symptoms are more strongly associated with Barrett's esophagus risk among persons homozygous for the major allele of rs2687201, which lies approximately 75 kb downstream of the transcription factor gene FOXP1. IMPACT The novel gene-exposure interaction discovered in this study provides new insights into the etiology of esophageal adenocarcinoma.
Collapse
Affiliation(s)
- James Y Dai
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Biostatistics, University of Washington, School of Public Health, Seattle, Washington.
| | - Jean de Dieu Tapsoba
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Matthew F Buas
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Lynn E Onstad
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - David M Levine
- Department of Biostatistics, University of Washington, School of Public Health, Seattle, Washington
| | - Harvey A Risch
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
| | - Wong-Ho Chow
- Department of Epidemiology, MD Anderson Cancer Center, Houston, Texas
| | - Leslie Bernstein
- Department of Population Sciences, Beckman Research Institute and City of Hope Comprehensive Cancer Center, Duarte, California
| | - Weimin Ye
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Jesper Lagergren
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden. Division of Cancer Studies, King's College London, London, United Kingdom
| | - Nigel C Bird
- Department of Oncology, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Douglas A Corley
- Division of Research, Kaiser Permanente Northern California, Oakland, California. San Francisco Medical Center, Kaiser Permanente Northern California, San Francisco, California
| | - Nicholas J Shaheen
- Division of Gastroenterology and Hepatology, University of North Carolina School of Medicine, University of North Carolina, Chapel Hill, North Carolina
| | - Anna H Wu
- Department of Preventive Medicine, University of Southern California/Norris Comprehensive Cancer Center, Los Angeles, California
| | - Brian J Reid
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Laura J Hardie
- Division of Epidemiology, University of Leeds, Leeds, United Kingdom
| | - David C Whiteman
- Cancer Control, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Thomas L Vaughan
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, University of Washington, School of Public Health, Seattle, Washington.
| |
Collapse
|
11
|
Wu F, Jasmine F, Kibriya MG, Liu M, Cheng X, Parvez F, Islam T, Ahmed A, Rakibuz-Zaman M, Jiang J, Roy S, Paul-Brutus R, Slavkovich V, Islam T, Levy D, VanderWeele TJ, Pierce BL, Graziano JH, Ahsan H, Chen Y. Interaction between arsenic exposure from drinking water and genetic polymorphisms on cardiovascular disease in Bangladesh: a prospective case-cohort study. ENVIRONMENTAL HEALTH PERSPECTIVES 2015; 123:451-7. [PMID: 25575156 PMCID: PMC4421763 DOI: 10.1289/ehp.1307883] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 01/07/2015] [Indexed: 05/20/2023]
Abstract
BACKGROUND Epidemiologic data on genetic susceptibility to cardiovascular effects of arsenic exposure from drinking water are limited. OBJECTIVE We investigated whether the association between well-water arsenic and cardiovascular disease (CVD) differed by 170 single nucleotide polymorphisms (SNPs) in 17 genes related to arsenic metabolism, oxidative stress, inflammation, and endothelial dysfunction. METHOD We conducted a prospective case-cohort study nested in the Health Effects of Arsenic Longitudinal Study, with a random subcohort of 1,375 subjects and 447 incident fatal and nonfatal cases of CVD. Well-water arsenic was measured in 2000 at baseline. The CVD cases, 56 of which occurred in the subcohort, included 238 coronary heart disease cases, 165 stroke cases, and 44 deaths due to other CVD identified during follow-up from 2000 to 2012. RESULTS Of the 170 SNPs tested, multiplicative interactions between well-water arsenic and two SNPs, rs281432 in ICAM1 (padj = 0.0002) and rs3176867 in VCAM1 (padj = 0.035), were significant for CVD after adjustment for multiple testing. Compared with those with GC or CC genotype in rs281432 and lower well-water arsenic, the adjusted hazard ratio (aHR) for CVD was 1.82 (95% CI: 1.31, 2.54) for a 1-SD increase in well-water arsenic combined with the GG genotype, which was greater than expected given aHRs of 1.08 and 0.96 for separate effects of arsenic and the genotype alone, respectively. Similarly, the joint aHR for arsenic and the rs3176867 CC genotype was 1.34 (95% CI: 0.95, 1.87), greater than expected given aHRs for their separate effects of 1.02 and 0.84, respectively. CONCLUSIONS Associations between CVD and arsenic exposure may be modified by genetic variants related to endothelial dysfunction.
Collapse
Affiliation(s)
- Fen Wu
- Department of Population Health, New York University School of Medicine, New York, New York, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hazelton WD, Curtius K, Inadomi JM, Vaughan TL, Meza R, Rubenstein JH, Hur C, Luebeck EG. The Role of Gastroesophageal Reflux and Other Factors during Progression to Esophageal Adenocarcinoma. Cancer Epidemiol Biomarkers Prev 2015; 24:1012-23. [PMID: 25931440 DOI: 10.1158/1055-9965.epi-15-0323-t] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 04/10/2015] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND U.S. esophageal adenocarcinoma (EAC) incidence increased over 5-fold between 1975 and 2009. Symptomatic gastroesophageal reflux disease (sGERD) elevates the risk for EAC. However, a simple calculation suggests that changes in sGERD prevalence can explain at most approximately 16% of this trend. Importantly, a mechanistic understanding of the influence of sGERD and other factors (OF) on EAC is lacking. METHODS A multiscale model was developed to estimate temporal trends for sGERD and OF, and their mechanistic role during carcinogenesis. Model calibration was to Surveillance, Epidemiology, and End Results (SEER) incidence and age-dependent sGERD data using maximum likelihood and Markov chain Monte Carlo (MCMC) methods. RESULTS Among men, 77.8% [95% credibility interval (CI), 64.9%-85.6%] of the incidence trend is attributable to OF, 13.4% (95% CI, 11.4%-17.3%) to sGERD, and 8.8% (95% CI, 4.2%-13.7%) to sGERD-OF interactions. Among women, 32.6% (95% CI, 27.0%-39.9%) of the trend is attributable to OF, 13.6% (95% CI, 12.5%-15.9%) to sGERD, and 47.4% (95% CI, 30.7%-64.6%) to interactions. The predicted trends were compared with historical trends for obesity, smoking, and proton pump inhibitor use. Interestingly, predicted OF cohort trends correlated most highly with median body mass index (BMI) at age 50 (r = 0.988 for men; r = 0.998 for women). CONCLUSIONS sGERD and OF mechanistically increase premalignant cell promotion, which increases EAC risk exponentially with exposure duration. IMPACT Surveillance should target individuals with long-duration sGERD and OF exposures.
Collapse
Affiliation(s)
- William D Hazelton
- Program in Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington. Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Kit Curtius
- Department of Applied Mathematics, University of Washington, Seattle, Washington
| | - John M Inadomi
- Division of Gastroenterology, University of Washington, Seattle, Washington
| | - Thomas L Vaughan
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington. Department of Epidemiology, School of Medicine, University of Washington, Seattle, Washington
| | - Rafael Meza
- Department of Epidemiology, School of Public Health, University of Michigan Medical School, Ann Arbor, Michigan
| | - Joel H Rubenstein
- Division of Gastroenterology, University of Michigan Medical School, Ann Arbor, Michigan. Veterans Affairs Center for Clinical Management Research, Ann Arbor, Michigan
| | - Chin Hur
- Institute for Technology Assessment, Massachusetts General Hospital, Boston, Massachusetts. Gastrointestinal Unit, Massachusetts General Hospital, Boston, Massachusetts. Harvard Medical School, Boston, Massachusetts
| | - E Georg Luebeck
- Program in Computational Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington. Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, Washington
| |
Collapse
|
13
|
Le Morvan V, Litière S, Laroche-Clary A, Ait-Ouferoukh S, Bellott R, Messina C, Cameron D, Bonnefoi H, Robert J. Identification of SNPs associated with response of breast cancer patients to neoadjuvant chemotherapy in the EORTC-10994 randomized phase III trial. THE PHARMACOGENOMICS JOURNAL 2014; 15:63-8. [PMID: 24958282 DOI: 10.1038/tpj.2014.24] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Revised: 04/14/2014] [Accepted: 04/22/2014] [Indexed: 11/09/2022]
Abstract
Using cell line panels we identified associations between single-nucleotide polymorphisms (SNPs) and chemosensitivity. To validate these findings in clinics, we genotyped a subset of patients included in a neoadjuvant breast cancer trial to explore the relationship between genotypes and clinical outcome according to treatment received and p53 status. We genotyped 384 selected SNPs in the germline DNA extracted from formalin-fixed paraffin-embedded non-invaded lymph nodes of 243 patients. The polymorphisms of five selected genes were first studied, and then all 384 SNPs were considered. Correction for multiple testing was applied. CYP1B1 polymorphism was significantly associated with pathological complete response (pCR) in patients who had received DNA-damaging agents. MDM2, MDM4 and TP53BP1 polymorphisms were significantly associated with pCR in patients harboring a p53-positive tumor. In the complete SNP panel, there was a significant association between overall survival (OS) and a SNP of ADH1C, R272Q (P=0.0023). By multivariate analysis, only ADH1C genotype and p53 status were significantly associated with OS.
Collapse
Affiliation(s)
- V Le Morvan
- INSERM U916, Institut Bergonié, Université Bordeaux Segalen, Bordeaux, France
| | - S Litière
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | - A Laroche-Clary
- INSERM U916, Institut Bergonié, Université Bordeaux Segalen, Bordeaux, France
| | - S Ait-Ouferoukh
- INSERM U916, Institut Bergonié, Université Bordeaux Segalen, Bordeaux, France
| | - R Bellott
- INSERM U916, Institut Bergonié, Université Bordeaux Segalen, Bordeaux, France
| | - C Messina
- European Organisation for Research and Treatment of Cancer (EORTC), Brussels, Belgium
| | | | - H Bonnefoi
- INSERM U916, Institut Bergonié, Université Bordeaux Segalen, Bordeaux, France
| | - J Robert
- INSERM U916, Institut Bergonié, Université Bordeaux Segalen, Bordeaux, France
| |
Collapse
|
14
|
Yu Y, Zheng S, Zhang S, Jin W, Liu H, Jin M, Chen Z, Ding Z, Wang L, Chen K. Polymorphisms of inflammation-related genes and colorectal cancer risk: a population-based case-control study in China. Int J Immunogenet 2014; 41:289-97. [PMID: 24762198 DOI: 10.1111/iji.12119] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 02/04/2014] [Accepted: 03/13/2014] [Indexed: 01/01/2023]
Abstract
The previous studies found that chronic inflammation related to an increased risk of colorectal cancer (CRC). This study aims to explore the associations of polymorphisms in inflammation-related genes (IL10, IL10RA, IL6R, TNFRSF1A, TNFRSF1B, LTA and IL4) and their interactions with the risk of colorectal cancer among Chinese population. A population-based case-control study including 299 cases and 296 controls was conducted from January 2001 to December 2009. Multivariate unconditional logistic regression was used to analyse the association of nine SNPs in inflammation-related genes with the risk of CRC, colon cancer and rectal cancer, respectively. Generalized multifactor dimensionality reduction (GMDR) was implemented to explore the gene-gene interactions among all SNPs on CRC. A decreased risk of colorectal cancer in subjects with rs1800872 AC genotype of IL10 (OR = 0.643, 95%CI = 0.453, 0.912) or AC/CC genotype (OR = 0.636, 95%CI = 0.457, 0.885) was observed, compared with those with AA genotype. Meanwhile, similar associations were observed between rs1800872 and rectal cancer. Additionally, in rs1061624 of TNFRSF1B gene, AG genotype (OR=0.566; 95% CI= 0.362, 0.885) and AG/GG genotype (OR=0.638; 95% CI=0.420, 0.971) were significantly associated with a decreased risk of rectal cancer, respectively. Our findings indicated that mutants in IL10 and TNFRSF1B genes may change the CRC risk. However, there is no interaction between inflammation-related genes on CRC risk.
Collapse
Affiliation(s)
- Y Yu
- Department of Epidemiology & Health Statistics, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China; Chronic Disease Research Institute, School of Public Health, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Yin J, Tang W, Shao A, Wang L, Wang X, Ding G, Liu C, Chen Y, Chen S, Gu H. Caspase8 rs1035142 G>T polymorphism was associated with an increased risk of esophageal cancer in a Chinese population. Mol Biol Rep 2014; 41:2037-43. [DOI: 10.1007/s11033-014-3052-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 01/04/2014] [Indexed: 11/29/2022]
|
16
|
Gene–environment interactions in heavy metal and pesticide carcinogenesis. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2014; 760:1-9. [DOI: 10.1016/j.mrgentox.2013.11.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 11/19/2013] [Accepted: 11/25/2013] [Indexed: 01/05/2023]
|
17
|
Clemons NJ, Phillips WA, Lord RV. Signaling pathways in the molecular pathogenesis of adenocarcinomas of the esophagus and gastroesophageal junction. Cancer Biol Ther 2013; 14:782-95. [PMID: 23792587 PMCID: PMC3909547 DOI: 10.4161/cbt.25362] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Esophageal adenocarcinoma develops in response to severe gastroesophageal reflux disease through the precursor lesion Barrett esophagus, in which the normal squamous epithelium is replaced by a columnar lining. The incidence of esophageal adenocarcinoma in the United States has increased by over 600% in the past 40 years and the overall survival rate remains less than 20% in the community. This review highlights some of the signaling pathways for which there is some evidence of a role in the development of esophageal adenocarcinoma. An increasingly detailed understanding of the biology of this cancer has emerged recently, revealing that in addition to the well-recognized alterations in single genes such as p53, p16, APC, and telomerase, there are interactions between the components of the reflux fluid, the homeobox gene Cdx2, and the Wnt, Notch, and Hedgehog signaling pathways.
Collapse
Affiliation(s)
- Nicholas J Clemons
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne, Australia; Sir Peter MacCallum Department of Oncology; University of Melbourne, Melbourne, Australia; Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne, Australia
| | - Wayne A Phillips
- Surgical Oncology Research Laboratory; Peter MacCallum Cancer Centre; East Melbourne, Australia; Sir Peter MacCallum Department of Oncology; University of Melbourne, Melbourne, Australia; Department of Surgery (St. Vincent's Hospital); University of Melbourne; Melbourne, Australia
| | - Reginald V Lord
- St. Vincent's Centre for Applied Medical Research; Sydney, Australia; Notre Dame University School of Medicine; Sydney, Australia
| |
Collapse
|