1
|
AHCYL1 Is a Novel Biomarker for Predicting Prognosis and Immunotherapy Response in Colorectal Cancer. JOURNAL OF ONCOLOGY 2022; 2022:5054324. [PMID: 35578598 PMCID: PMC9107370 DOI: 10.1155/2022/5054324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 04/18/2022] [Indexed: 11/18/2022]
Abstract
Background Colorectal cancer (CRC) is the third most frequent cancer worldwide. The AHCYL1 gene is required for CNV and has a close association with the tumor immune microenvironment. However, the predictive value of the AHCYL1 gene in patients with CRC remains unknown. Methods AHCYL1 gene with prognostic potential was comprehensively analyzed. Next, using LASSO Cox regression, we fully examined and integrated the AHCYL1 and AHCYL1-related genes from TCGA database. Meanwhile, TCGA database was used to study the connection between AHCYL1 and the tumor immune microenvironment and tumor mutation burden (TMB) in CRC. The influence of AHCYL1 in tumor growth and the recruiting ability of CD8+ T cells were verified, respectively, in vivo and in tissues. To ascertain the connection between AHCYL1 and AHCYL1-related genes and the prognosis of CRC, a prognostic model was created and validated. Result We demonstrated that AHCYL1 has a differential expression and patients with AHCYL1 deletion get shorter survival in CRC. Additionally, the tissues without AHCYL1 have a weaker ability to recruit the natural killer (NK) cell, CD8+ T cells, and tumor-infiltrating lymphocytes (TILs) and response to immunotherapy. Additionally, knockdown of AHCYL1 promoted tumor growth in the CRC mouse model and recruited lower CD8+ T cells in CRC tissues. TCGA database was used to classify patients into low- and high-risk categories based on the expression of four genes. Meanwhile, we discovered an association between the low-risk group and a lower TMB and a higher response to immunotherapy. Finally, a predictive nomogram based on these genes was developed and verified, yielding a C-index of 0.74. Conclusion For CRC patients, the prognostic model based on AHCYL1 and AHCYL1-related genes showed a high predictive performance in terms of prognosis and immunotherapy response.
Collapse
|
2
|
Xu X, Gong C, Wang Y, Hu Y, Liu H, Fang Z. Multi-omics analysis to identify driving factors in colorectal cancer. Epigenomics 2020; 12:1633-1650. [PMID: 32573269 DOI: 10.2217/epi-2020-0073] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Aim: We aim to identify driving genes of colorectal cancer (CRC) through multi-omics analysis. Materials & methods: We downloaded multi-omics data of CRC from The Cancer Genome Atlas dataset. Integrative analysis of single-nucleotide variants, copy number variations, DNA methylation and differentially expressed genes identified candidate genes that carry CRC risk. Kernal genes were extracted from the weighted gene co-expression network analysis. A competing endogenous RNA network composed of CRC-related genes was constructed. Biological roles of genes were further investigated in vitro. Results: We identified LRRC26 and REP15 as novel prognosis-related driving genes for CRC. LRRC26 hindered tumorigenesis of CRC in vitro. Conclusion: Our study identified novel driving genes and may provide new insights into the molecular mechanisms of CRC.
Collapse
Affiliation(s)
- Xi Xu
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Chaoju Gong
- Central Laboratory, The Municipal Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221106, PR China
| | - Yunfeng Wang
- Institute for Integrative Biology of the Cell, UMR 9198, CNRS, Commissariat à l'Energie Atomique et aux Énergies Alternatives (CEA), Université Paris-Sud, 91198 Gif-sur-Yvette, Palaiseau, 91120, France
| | - Yanyan Hu
- Central Laboratory, Sanmen People's Hospital of Zhejiang Province, Sanmen, 317100, PR China
| | - Hong Liu
- Zhejiang Normal University - Jinhua People's Hospital Joint Center for Biomedical Research, Jinhua, 321004, PR China.,The Affiliated Hospital of Jinhua Polytechnic College, Jinhua, 321000, PR China
| | - Zejun Fang
- Central Laboratory, Sanmen People's Hospital of Zhejiang Province, Sanmen, 317100, PR China.,Central Laboratory, Sanmenwan Branch, The First Affiliated Hospital, College of Medicine, Zhejiang University, Sanmen, 317100, PR China
| |
Collapse
|
3
|
Zhang J, Fu Y, Chen J, Li Q, Guo H, Yang B. Genetic variant of TMBIM1 is associated with the susceptibility of colorectal cancer in the Chinese population. Clin Res Hepatol Gastroenterol 2019; 43:324-329. [PMID: 30447906 DOI: 10.1016/j.clinre.2018.10.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 02/04/2023]
Abstract
BACKGROUND AND AIMS Recent meta-analysis of genome-wide association studies (GWASs) identified a novel variant rs992157 at 2q35 that was associated with colorectal cancer (CRC) in the population of European ancestry. We aimed to replicate the association of rs992157 with CRC in the Chinese population and to further determine the real susceptible gene of CRC as indicated by this variant. METHODS 824 CRC patients and 1063 healthy controls were included. The frequency of the genotype and the allele of rs992157 were compared between the patients and the controls and between different subgroups of patients classified by status of metastasis. Expression level of TMBIM1 was compared between the tumor tissue and the adjacent normal tissues collected from 43 patients during surgery. Besides, the relationship between genotypes of rs992157 and the tissue expression of TMBIM1 was analyzed. RESULTS Patients were found to have significantly higher frequency of allele G than the controls (44.2% vs. 40.0%, P = 0.009; OR = 1.18). Moreover, allele G was associated with an increased risk of lymph node metastasis (P = 0.02) and distant metastasis of CRC (P = 0.04). The mean expression level of TMBIM1 was significantly higher in tumor tissue than in the adjacent normal tissues (0.0019 ± 0.00068 vs. 0.00041 ± 0.00024, P < 0.001). In addition, patients with genotype GG were found to have remarkably higher TMBIM1 expression in the tumors than those with genotype AA (0.0024 ± 0.00052 vs. 0.0015 ± 0.00078, P = 0.005). CONCLUSION Variant rs992157 is significantly associated with the susceptibility and progression of CRC. It can increase the risk of CRC possibly via up-regulation of TMBIM1.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, PR China
| | - Yiwei Fu
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, PR China
| | - Jiebin Chen
- Department of Paediatrics, Jiangsu Taizhou People's Hospital, Taizhou, PR China
| | - Qianjun Li
- Department of Gastroenterology, Huai'an First People's Hospital of Nanjing Medical University, Huai'an, PR China
| | - Huimin Guo
- Department of Gastroenterology, The Drum Tower Hospital of Nanjing University Medical School, Nanjing, PR China
| | - Bin Yang
- Department of Gastroenterology, Jiangsu Taizhou People's Hospital, Taizhou, PR China.
| |
Collapse
|
4
|
Clinical significance of germline copy number variation in susceptibility of human diseases. J Genet Genomics 2018; 45:3-12. [PMID: 29396143 DOI: 10.1016/j.jgg.2018.01.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 12/27/2017] [Accepted: 01/02/2018] [Indexed: 02/06/2023]
Abstract
Germline copy number variation (CNV) is considered to be an important form of human genetic polymorphisms. Previous studies have identified amounts of CNVs in human genome by advanced technologies, such as comparative genomic hybridization, single nucleotide genotyping, and high-throughput sequencing. CNV is speculated to be derived from multiple mechanisms, such as nonallelic homologous recombination (NAHR) and nonhomologous end-joining (NHEJ). CNVs cover a much larger genome scale than single nucleotide polymorphisms (SNPs), and may alter gene expression levels by means of gene dosage, gene fusion, gene disruption, and long-range regulation effects, thus affecting individual phenotypes and playing crucial roles in human pathogenesis. The number of studies linking CNVs with common complex diseases has increased dramatically in recent years. Here, we provide a comprehensive review of the current understanding of germline CNVs, and summarize the association of germline CNVs with the susceptibility to a wide variety of human diseases that were identified in recent years. We also propose potential issues that should be addressed in future studies.
Collapse
|
5
|
Zhang D, Li Z, Xu X, Zhou D, Tang S, Yin X, Xu F, Li H, Zhou Y, Zhu T, Deng H, Zhang S, Huang Q, Wang J, Yin W, Zhu Y, Lai M. Deletions at SLC18A1 increased the risk of CRC and lower SLC18A1 expression associated with poor CRC outcome. Carcinogenesis 2017; 38:1057-1062. [PMID: 28968818 DOI: 10.1093/carcin/bgx088] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 07/24/2017] [Indexed: 12/26/2022] Open
Abstract
Copy number variations (CNVs) contribute to the development of colorectal cancer (CRC). We conducted a two-stage association study to identify CNV risk loci for CRC. We performed a gene-based rare CNV study on 694 sporadic CRC and 1641 controls using Illumina Human-OmniExpress-12v1.0 BeadChips, and further replicated in 934 CRC cases and 2680 controls for risk CNVs by using TaqMan Copy Number Assay. Tumor buddings, cancer cells in the center of primary tumor and normal intestinal epithelial cells were captured using laser capture microdissection (LCM) and were assayed using AffymetrixGeneChip® Human Genome U133 Plus 2.0 Array. In addition, The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus data were assessed for the effects of risk CNVs. We found that germline deletions affecting the last six exons of SLC18A1 significantly associated with CRC with a combined P value of 6.4 × 10-5 by a two-stage analysis. Both in TCGA CRC RNA seq dataset and GDS4382, SLC18A1 was significantly down regulated in CRC tissues than in paired normal tissues (N = 32 and 17 pairs, P = 0.004 and 0.009, respectively). In LCM samples, similar observations were obtained that the expression levels of SLC18A1 in the tumor buddings, cancer cells in the center of primary tumor, and stroma of both tumor budding and cancer cells were lower than normal intestinal epithelial and stromal cells (fold change = 0.17-0.62, 0.12-0.57 and 0.37-0.68, respectively). In summary, the germline deletions at SLC18A1 contributed to the development of CRC. The role of SLC18A1 required further exploration.
Collapse
Affiliation(s)
- Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Zhenli Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Xiaohong Xu
- Clinical laboratory, The Affiliated Zhejiang Cancer Hospital of Zhejiang Chinese Medical University, Hangzhou 310022, China
| | - Dan Zhou
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Shunli Tang
- The Second Department of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiaoyang Yin
- The Second Department of Clinical Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Fangying Xu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Hui Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Yuan Zhou
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Tao Zhu
- Departments of Pathology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou 310016, China
| | - Hong Deng
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Shuai Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Qiong Huang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Jing Wang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| | - Wei Yin
- The Core Facilities, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yimin Zhu
- Department of Epidemiology and Biostatistics, Zhejiang University School of Public Health, Hangzhou 310058, China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou 310058, China
| |
Collapse
|
6
|
Thean LF, Low YS, Lo M, Teo YY, Koh WP, Yuan JM, Chew MH, Tang CL, Cheah PY. Genome-wide association study identified copy number variants associated with sporadic colorectal cancer risk. J Med Genet 2017; 55:181-188. [DOI: 10.1136/jmedgenet-2017-104913] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/19/2017] [Accepted: 10/11/2017] [Indexed: 12/18/2022]
Abstract
BackgroundMultiple single nucleotide polymorphisms (SNPs) have been associated with colorectal cancer (CRC) risk. The role of structural or copy number variants (CNV) in CRC, however, remained unclear. We investigated the role of CNVs in patients with sporadic CRC.MethodsA genome-wide association study (GWAS) was performed on 1000 Singapore Chinese patients aged 50 years or more with no family history of CRC and 1000 ethnicity-matched, age-matched and gender-matched healthy controls using the Affymetrix SNP 6 platform. After 16 principal component corrections, univariate and multivariate segmentations followed by association testing were performed on 1830 samples that passed quality assurance tests.ResultsA rare CNV region (CNVR) at chromosome 14q11 (OR=1.92 (95% CI 1.59 to 2.32), p=2.7e-12) encompassing CHD8, and common CNVR at chromosomes 3q13.12 (OR=1.54 (95% CI 1.33 to 1.77), p=2.9e-9) and 12p12.3 (OR=1.69 (95% CI 1.41 to 2.01), p=2.8e-9) encompassing CD47 and RERG/ARHGDIB, respectively, were significantly associated with CRC risk. CNV loci were validated in an independent replication panel using an optimised copy number assay. Whole-genome expression data in matched tumours of a subset of cases demonstrated that copy number loss at CHD8 was significantly associated with dysregulation of several genes that perturb the Wnt, TP53 and inflammatory pathways.ConclusionsA rare CNVR at 14q11 encompassing the chromatin modifier CHD8 was significantly associated with sporadic CRC risk. Copy number loss at CHD8 altered expressions of genes implicated in colorectal tumourigenesis.
Collapse
|
7
|
Li M, Carey J, Cristiano S, Susztak K, Coresh J, Boerwinkle E, Kao WHL, Beaty TH, Köttgen A, Scharpf RB. Genome-Wide Association of Copy Number Polymorphisms and Kidney Function. PLoS One 2017; 12:e0170815. [PMID: 28135296 PMCID: PMC5279752 DOI: 10.1371/journal.pone.0170815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 01/11/2017] [Indexed: 01/08/2023] Open
Abstract
Genome-wide association studies (GWAS) using single nucleotide polymorphisms (SNPs) have identified more than 50 loci associated with estimated glomerular filtration rate (eGFR), a measure of kidney function. However, significant SNPs account for a small proportion of eGFR variability. Other forms of genetic variation have not been comprehensively evaluated for association with eGFR. In this study, we assess whether changes in germline DNA copy number are associated with GFR estimated from serum creatinine, eGFRcrea. We used hidden Markov models (HMMs) to identify copy number polymorphic regions (CNPs) from high-throughput SNP arrays for 2,514 African (AA) and 8,645 European ancestry (EA) participants in the Atherosclerosis Risk in Communities (ARIC) study. Separately for the EA and AA cohorts, we used Bayesian Gaussian mixture models to estimate copy number at regions identified by the HMM or previously reported in the HapMap Project. We identified 312 and 464 autosomal CNPs among individuals of EA and AA, respectively. Multivariate models adjusted for SNP-derived covariates of population structure identified one CNP in the EA cohort near genome-wide statistical significance (Bonferroni-adjusted p = 0.067) located on chromosome 5 (876-880kb). Overall, our findings suggest a limited role of CNPs in explaining eGFR variability.
Collapse
Affiliation(s)
- Man Li
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Division of Nephrology and Hypertension, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah, United States of America
| | - Jacob Carey
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Stephen Cristiano
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Katalin Susztak
- Renal Electrolyte and Hypertension Division, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, United States of America
| | - Eric Boerwinkle
- Human Genetics Center, University of Texas Health Science Center at Houston, Houston, Texas, United States of America
| | - Wen Hong L. Kao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Welch Center for Prevention, Epidemiology and Clinical Research, Baltimore, Maryland, United States of America
| | - Terri H. Beaty
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Anna Köttgen
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
- Division of Genetic Epidemiology, Medical Center–University of Freiburg, Faculty of Medicine, Freiburg, Germany
| | - Robert B. Scharpf
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
8
|
Arriba M, García JL, Rueda D, Pérez J, Brandariz L, Nutu OA, Alonso L, Rodríguez Y, Urioste M, González-Sarmiento R, Perea J. Unsupervised Analysis of Array Comparative Genomic Hybridization Data from Early-Onset Colorectal Cancer Reveals Equivalence with Molecular Classification and Phenotypes. Neoplasia 2017; 19:28-34. [PMID: 27987438 PMCID: PMC5166699 DOI: 10.1016/j.neo.2016.11.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 11/02/2016] [Accepted: 11/07/2016] [Indexed: 02/07/2023]
Abstract
AIM To investigate whether chromosomal instability (CIN) is associated with tumor phenotypes and/or with global genomic status based on MSI (microsatellite instability) and CIMP (CpG island methylator phenotype) in early-onset colorectal cancer (EOCRC). METHODS Taking as a starting point our previous work in which tumors from 60 EOCRC cases (≤45 years at the time of diagnosis) were analyzed by array comparative genomic hybridization (aCGH), in the present study we performed an unsupervised hierarchical clustering analysis of those aCGH data in order to unveil possible associations between the CIN profile and the clinical features of the tumors. In addition, we evaluated the MSI and the CIMP statuses of the samples with the aim of investigating a possible relationship between copy number alterations (CNAs) and the MSI/CIMP condition in EOCRC. RESULTS Based on the similarity of the CNAs detected, the unsupervised analysis stratified samples into two main clusters (A, B) and four secondary clusters (A1, A2, B3, B4). The different subgroups showed a certain correspondence with the molecular classification of colorectal cancer (CRC), which enabled us to outline an algorithm to categorize tumors according to their CIMP status. Interestingly, each subcluster showed some distinctive clinicopathological features. But more interestingly, the CIN of each subcluster mainly affected particular chromosomes, allowing us to define chromosomal regions more specifically affected depending on the CIMP/MSI status of the samples. CONCLUSIONS Our findings may provide a basis for a new form of classifying EOCRC according to the genomic status of the tumors.
Collapse
Key Words
- acgh, array comparative genomic hybridization
- cimp, cpg island methylator phenotype
- cin, chromosomal instability
- cna, copy number alteration
- crc, colorectal cancer
- dfs, disease-free survival
- eocrc, early-onset colorectal cancer
- gii, genomic instability index
- ls, lynch syndrome
- macs, microsatellite and chromosome stable tumors
- mmr, mismatch repair
- msi, microsatellite instability
- mss, microsatellite stability
- os, overall survival
Collapse
Affiliation(s)
- María Arriba
- Centre for Biomedical Research of the 12 de Octubre University Hospital, Avda. de Córdoba, S/N, 28041, Madrid, Spain
| | - Juan L García
- Biomedical Research Institute of Salamanca (IBSAL). University Hospital of Salamanca-USAL-CSIC, P° de San Vicente, 58-182, 37007, Salamanca, Spain; Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Daniel Rueda
- Molecular Biology Laboratory, 12 de Octubre University Hospital, Avda. de Córdoba, S/N, 28041, Madrid, Spain
| | - Jessica Pérez
- Biomedical Research Institute of Salamanca (IBSAL). University Hospital of Salamanca-USAL-CSIC, P° de San Vicente, 58-182, 37007, Salamanca, Spain; Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - Lorena Brandariz
- Department of Surgery, 12 de Octubre University Hospital. Avda. de Córdoba, S/N, 28041, Madrid, Spain
| | - Oana A Nutu
- Department of Surgery, 12 de Octubre University Hospital. Avda. de Córdoba, S/N, 28041, Madrid, Spain
| | - Laura Alonso
- Department of Surgery, 12 de Octubre University Hospital. Avda. de Córdoba, S/N, 28041, Madrid, Spain
| | - Yolanda Rodríguez
- Department of Pathology, 12 de Octubre University Hospital, Avda. de Córdoba, S/N, 28041, Madrid, Spain
| | - Miguel Urioste
- Familial Cancer Clinical Unit, Human Cancer Genetics Program. Spanish National Cancer Research Centre (CNIO), C/Melchor Fernández Almagro, 3, 28029, Madrid, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER), Institute of Health Carlos III, C/Monforte de Lemos 3-5, Pabellón 11, 28029, Madrid, Spain
| | - Rogelio González-Sarmiento
- Biomedical Research Institute of Salamanca (IBSAL). University Hospital of Salamanca-USAL-CSIC, P° de San Vicente, 58-182, 37007, Salamanca, Spain; Institute of Molecular and Cellular Biology of Cancer (IBMCC), University of Salamanca-CSIC, Campus Miguel de Unamuno, 37007, Salamanca, Spain
| | - José Perea
- Centre for Biomedical Research of the 12 de Octubre University Hospital, Avda. de Córdoba, S/N, 28041, Madrid, Spain; Department of Surgery, 12 de Octubre University Hospital. Avda. de Córdoba, S/N, 28041, Madrid, Spain.
| |
Collapse
|
9
|
Candidate predisposing germline copy number variants in early onset colorectal cancer patients. Clin Transl Oncol 2016; 19:625-632. [PMID: 27888432 DOI: 10.1007/s12094-016-1576-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/10/2016] [Indexed: 01/04/2023]
Abstract
PURPOSE A great proportion of the heritability of colorectal cancer (CRC) still remains unexplained, and rare variants, as well as copy number changes, have been proposed as potential candidates to explain the so-called 'missing heritability'. We aimed to identify rare high-to-moderately penetrant copy number variants (CNVs) in patients suspected of having hereditary CRC due to an early onset. METHODS/PATIENTS We have selected for genome-wide copy number analysis, 27 MMR-proficient early onset CRC patients (<50 years) without identifiable germline mutations in Mendelian genes related to this phenotype. Rare CNVs were selected by removing all CNVs detected at MAF >1% in the in-house control CNV database (n = 629 healthy controls). Copy number assignment was checked by duplex real-time quantitative PCR or multiplex ligation probe amplification. Somatic mutation analysis in candidate genes included: loss of heterozygosity studies, point mutation screening, and methylation status of the promoter. RESULTS We have identified two rare germline deletions involving the AK3 and SLIT2 genes in two patients. The search for a second somatic mutational event in the corresponding CRC tumors showed loss of heterozygosity in AK3, and promoter hypermethylation in SLIT2. Both genes have been previously related to colorectal carcinogenesis. CONCLUSIONS These findings suggest that AK3 and SLIT2 may be potential candidates involved in genetic susceptibility to CRC.
Collapse
|
10
|
Park C, Kim JI, Hong SN, Jung HM, Kim TJ, Lee S, Kim SJ, Kim HC, Kim DH, Cho B, Park JH, Sung J, Lee DS, Kang M, Son HJ, Kim YH. A copy number variation in PKD1L2 is associated with colorectal cancer predisposition in korean population. Int J Cancer 2016; 140:86-94. [PMID: 27605020 DOI: 10.1002/ijc.30421] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Accepted: 08/15/2016] [Indexed: 12/30/2022]
Abstract
Recently reported genome-wide association studies have identified more than 20 common low-penetrance colorectal cancer (CRC) susceptibility loci. Recent studies have reported that copy number variations (CNVs) are considered important human genomic variants related to cancer, while the contribution of CNVs remains unclear. We performed array comparative genomic hybridization (aCGH) in 36 CRC patients and 47 controls. Using breakpoint PCR, we confirmed the breakpoint of the PKD1L2 deletion region. High frequency of PKD1L2 CNV was observed in CRC cases. We validated the association between PKD1L2 variation and CRC risk in 1,874 cases and 2,088 controls (OR = 1.44, 95% CI = 1.04-1.98, p = 0.028). Additionally, PKD1L2 CNV is associated with increased CRC risk in patients younger than 50 years (OR = 2.14, 95% CI 1.39-3.30, p = 5.8 × 10-4 ). In subgroup analysis according to body mass index (BMI), we found that the CN loss of PKD1L2 with BMI above or equal to 25 exhibited a significant increase in CRC risk (OR = 2.29, 95% CI 1.29-4.05, p = 0.005). PKD1L2 CNV with BMI above or equal to 25 and age below 50 is associated with a remarkably increased risk of colorectal cancer (OR = 5.24, 95% CI 2.36-11.64, p= 4.8 × 10-5 ). Moreover, we found that PKD1L2 variation in obese patients (BMI ≥ 25) was associated with poor survival rate (p = 0.026). Our results suggest that the common PKD1L2 CNV is associated with CRC, and PKD1L2 CNV with high BMI and/or age below 50 exhibited a significant increased risk of CRC. In obese patients, PKD1L2 variation was associated with poor survival.
Collapse
Affiliation(s)
- Changho Park
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jong-Il Kim
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea.,Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hey Mi Jung
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Tae Jun Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seungbok Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Seong Jin Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Duk-Hwan Kim
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon, Korea
| | - Belong Cho
- Department of Family Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Jin-Ho Park
- Department of Family Medicine, Seoul National University College of Medicine, Seoul, Korea
| | - Joohon Sung
- Complex Disease and Genome Epidemiology Branch, Department of Epidemiology, School of Public Health, Seoul National University, Seoul, Korea
| | - Dong-Sung Lee
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul, Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea.,Medical Research Center, Genomic Medicine Institute (GMI), Seoul National University, Seoul, Korea
| | - Mingon Kang
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Jung Son
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Ho Kim
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Xicola RM, Bontu S, Doyle BJ, Rawson J, Garre P, Lee E, de la Hoya M, Bessa X, Clofent J, Bujanda L, Balaguer F, Castellví-Bel S, Alenda C, Jover R, Ruiz-Ponte C, Syngal S, Andreu M, Carracedo A, Castells A, Newcomb PA, Lindor N, Potter JD, Baron JA, Ellis NA, Caldes T, LLor X. Association of a let-7 miRNA binding region of TGFBR1 with hereditary mismatch repair proficient colorectal cancer (MSS HNPCC). Carcinogenesis 2016; 37:751-758. [PMID: 27234654 PMCID: PMC4967215 DOI: 10.1093/carcin/bgw064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 04/24/2016] [Accepted: 05/15/2016] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to identify novel colorectal cancer (CRC)-causing alleles in unexplained familial CRC cases. In order to do so, coding regions in five candidate genes (MGMT, AXIN2, CTNNB1, TGFBR1 and TGFBR2) were sequenced in 11 unrelated microsatellite-stable hereditary non-polyposis CRC (MSS HNPCC) cases. Selected genetic variants were genotyped in a discovery set of 27 MSS HNPCC cases and 85 controls. One genetic variant, rs67687202, in TGFBR1 emerged as significant (P = 0.002), and it was genotyped in a replication set of 87 additional MSS HNPCC-like cases and 338 controls where it was also significantly associated with MSS HNPCC cases (P = 0.041). In the combined genotype data, rs67687202 was associated with a moderate increase in CRC risk (OR = 1.68; 95% CI = 1.13-2.50; P = 0.010). We tested a highly correlated SNP rs868 in 723 non-familial CRC cases compared with 629 controls, and it was not significantly associated with CRC risk (P = 0.370). rs868 is contained in a let-7 miRNA binding site in the 3'UTR of TGFBR1, which might provide a functional basis for the association in MSS HNPCC. In luciferase assays, the risk-associated allele for rs868 was associated with half the luciferase expression in the presence of miRNA let-7b-5p compared with protective allele, suggesting more binding of let-7b-5p and less TGFBR1 expression. Thus, rs868 potentially is a CRC risk-causing allele. Our results support the concept that rs868 is associated with lower TGFBR1 expression thereby increasing CRC risk.
Collapse
Affiliation(s)
- Rosa M Xicola
- Department of Medicine and Cancer Center, Yale University, New Haven, CT, USA
| | - Sneha Bontu
- Department of Medicine and Cancer Center, Yale University, New Haven, CT, USA
| | - Brian J Doyle
- Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Jamie Rawson
- Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Pilar Garre
- Laboratorio de Oncología Molecular, Hospital Clinico San Carlos, IdISSC , Madrid, Spain
| | - Esther Lee
- Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Miguel de la Hoya
- Laboratorio de Oncología Molecular, Hospital Clinico San Carlos, IdISSC , Madrid, Spain
| | - Xavier Bessa
- Department of Gastroenterology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Joan Clofent
- Department of Gastroenterology, Hospital de Sagunto, Sagunto, Valencia, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, CIBERehd, Universidad del País Vasco (UPV/EHU), San Sebastian, Spain
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clinic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - Sergi Castellví-Bel
- Department of Gastroenterology, Hospital Clinic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - Cristina Alenda
- Department of Gastroenterology and Department of Pathology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Rodrigo Jover
- Department of Gastroenterology and Department of Pathology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Clara Ruiz-Ponte
- Fundación Pública Galega de Medicina Xenómica (FPGMX)-SERGAS, Grupo de Medicina Xenómica, IDIS, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Santiago de Compostela, Galiza, Spain
| | - Sapna Syngal
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Montserrat Andreu
- Department of Gastroenterology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Angel Carracedo
- Fundación Pública Galega de Medicina Xenómica (FPGMX)-SERGAS, Grupo de Medicina Xenómica, IDIS, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Santiago de Compostela, Galiza, Spain
| | - Antoni Castells
- Department of Gastroenterology, Hospital Clinic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | | | - Noralane Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, USA
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Department of Epidemiology, University of Washington, Seattle, WA, USA Centre for Public Health Research, Wellington, New Zealand
| | - John A Baron
- Department of Biostatistics and Epidemiology, Dartmouth College, Lebanon, NH, USA
| | - Nathan A Ellis
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Trinidad Caldes
- Laboratorio de Oncología Molecular, Hospital Clinico San Carlos, IdISSC , Madrid, Spain
| | - Xavier LLor
- Department of Medicine and Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
12
|
Li Z, Yu D, Gan M, Shan Q, Yin X, Tang S, Zhang S, Shi Y, Zhu Y, Lai M, Zhang D. A genome-wide assessment of rare copy number variants in colorectal cancer. Oncotarget 2016; 6:26411-23. [PMID: 26315111 PMCID: PMC4694911 DOI: 10.18632/oncotarget.4621] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 07/06/2015] [Indexed: 12/30/2022] Open
Abstract
Colorectal cancer (CRC) is a complex disease with an estimated heritability of approximately 35%. However, known CRC-related common single nucleotide polymorphisms (SNPs) can only explain ~0.65% of the heritability. This “missing heritability” may be explained partially by rare copy number variants (CNVs). In this study, we performed a genome-wide scan using Illumina Human-Omni Express BeadChip, 694 sporadic CRC cases and 1641 controls were eventually included in our analysis after quality control. The global burden analysis revealed a 1.53-fold excess of rare CNVs in CRC cases compared with controls (P < 1 × 10−6), and the difference being more pronounced for genic rare CNVs and CNVs overlapped with coding regions (1.65-fold and 1.84-fold, respectively, both P < 1 × 10−6). Interestingly, both the cases in the lowest and middle tertile of age carried a higher burden of rare CNVs comparing to the highest tertile. Furthermore, 639 CNV-disrupted genes exclusive to CRC cases were found to be significantly enriched in gene ontology (GO) terms concerning nucleosome assembly and olfactory receptor activity. Our study was the first to evaluate the burden of rare CNVs in sporadic CRC and suggested that rare CNVs contributed to the missing heritability of CRC.
Collapse
Affiliation(s)
- Zhenli Li
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Dan Yu
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Meifu Gan
- Department of Pathology, Taizhou Hospital, Linhai, Zhejiang, 317000, China
| | - Qiaonan Shan
- Zhejiang University School of Clinical Medicine, Hangzhou, Zhejiang, 310058, China
| | - Xiaoyang Yin
- Zhejiang University School of Clinical Medicine, Hangzhou, Zhejiang, 310058, China
| | - Shunli Tang
- Zhejiang University School of Clinical Medicine, Hangzhou, Zhejiang, 310058, China
| | - Shuai Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Yongyong Shi
- Bio-X Institutes, Key Laboratory for The Genetics of Developmental and Neuropsychiatric Disorders, Ministry of Education, Shanghai Jiao Tong University, Shanghai, 200000, China
| | - Yimin Zhu
- Department of Epidemiology & Biostatistics, Zhejiang University School of Public Health, Hangzhou, Zhejiang, 310058, China
| | - Maode Lai
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| | - Dandan Zhang
- Department of Pathology, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.,Key Laboratory of Disease Proteomics of Zhejiang Province, Hangzhou, Zhejiang, 310058, China
| |
Collapse
|
13
|
Gong J, Tian J, Lou J, Ke J, Li L, Li J, Yang Y, Gong Y, Zhu Y, Zhang Y, Zhong R, Chang J, Miao X. A functional polymorphism inlnc-LAMC2-1:1confers risk of colorectal cancer by affecting miRNA binding. Carcinogenesis 2016; 37:443-51. [DOI: 10.1093/carcin/bgw024] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 02/12/2016] [Indexed: 02/07/2023] Open
|
14
|
Ananthakrishnan AN, Du M, Berndt SI, Brenner H, Caan BJ, Casey G, Chang-Claude J, Duggan D, Fuchs CS, Gallinger S, Giovannucci EL, Harrison TA, Hayes RB, Hoffmeister M, Hopper JL, Hou L, Hsu L, Jenkins MA, Kraft P, Ma J, Nan H, Newcomb PA, Ogino S, Potter JD, Seminara D, Slattery ML, Thornquist M, White E, Wu K, Peters U, Chan AT. Red meat intake, NAT2, and risk of colorectal cancer: a pooled analysis of 11 studies. Cancer Epidemiol Biomarkers Prev 2015; 24:198-205. [PMID: 25342387 PMCID: PMC4294960 DOI: 10.1158/1055-9965.epi-14-0897] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Red meat intake has been associated with risk of colorectal cancer, potentially mediated through heterocyclic amines. The metabolic efficiency of N-acetyltransferase 2 (NAT2) required for the metabolic activation of such amines is influenced by genetic variation. The interaction between red meat intake, NAT2 genotype, and colorectal cancer has been inconsistently reported. METHODS We used pooled individual-level data from the Colon Cancer Family Registry and the Genetics and Epidemiology of Colorectal Cancer Consortium. Red meat intake was collected by each study. We inferred NAT2 phenotype based on polymorphism at rs1495741, highly predictive of enzyme activity. Interaction was assessed using multiplicative interaction terms in multivariate-adjusted models. RESULTS From 11 studies, 8,290 colorectal cancer cases and 9,115 controls were included. The highest quartile of red meat intake was associated with increased risk of colorectal cancer compared with the lowest quartile [OR, 1.41; 95% confidence interval (CI), 1.29-1.55]. However, a significant association was observed only for studies with retrospective diet data, not for studies with diet prospectively assessed before cancer diagnosis. Combining all studies, high red meat intake was similarly associated with colorectal cancer in those with a rapid/intermediate NAT2 genotype (OR, 1.38; 95% CI, 1.20-1.59) as with a slow genotype (OR, 1.43; 95% CI, 1.28-1.61; P interaction = 0.9). CONCLUSION We found that high red meat intake was associated with increased risk of colorectal cancer only from retrospective case-control studies and not modified by NAT2 enzyme activity. IMPACT Our results suggest no interaction between NAT2 genotype and red meat intake in mediating risk of colorectal cancer.
Collapse
Affiliation(s)
| | - Mengmeng Du
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sonja I Berndt
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany. German Cancer Consortium, Heidelberg, Germany
| | | | - Graham Casey
- University of Southern California, Los Angeles, California
| | - Jenny Chang-Claude
- German Cancer Consortium, Heidelberg, Germany. Division of Cancer Epidemiology, German Cancer Research Center (DFKZ), Heidelberg, Germany
| | - David Duggan
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Charles S Fuchs
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts. Channing Division of Network Medicine, Boston, Massachusetts
| | - Steven Gallinger
- Mount Sinai Hospital and University Health Network Toronto General Hospital, Toronto, Canada
| | - Edward L Giovannucci
- Channing Division of Network Medicine, Boston, Massachusetts. Harvard School of Public Health, Boston, Massachusetts
| | | | | | | | - John L Hopper
- Melbourne School of Population Health, The University of Melbourne, Australia
| | - Lifang Hou
- Department of Preventive Medicine and The Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Evanston, Illinois
| | - Li Hsu
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Mark A Jenkins
- Melbourne School of Population Health, The University of Melbourne, Australia
| | - Peter Kraft
- Harvard School of Public Health, Boston, Massachusetts
| | - Jing Ma
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, Massachusetts. Channing Division of Network Medicine, Boston, Massachusetts
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Simon Cancer Center, Indiana University, Indianapolis, Indiana
| | | | - Shuji Ogino
- Channing Division of Network Medicine, Boston, Massachusetts. Harvard School of Public Health, Boston, Massachusetts. Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - John D Potter
- Fred Hutchinson Cancer Research Center, Seattle, Washington. University of Washington School of Public Health, Seattle, Washington. Centre for Public Health Research, Massey University, Wellington, New Zealand
| | - Daniela Seminara
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | | | | | - Emily White
- Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Kana Wu
- Channing Division of Network Medicine, Boston, Massachusetts. Harvard School of Public Health, Boston, Massachusetts
| | - Ulrike Peters
- Fred Hutchinson Cancer Research Center, Seattle, Washington. University of Washington School of Public Health, Seattle, Washington.
| | - Andrew T Chan
- Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
15
|
Abstract
A number of studies have evaluated the role of gene-diet interaction in the etiology of colorectal cancer (CRC). Historically, these studies focused on established dietary risk factors and genes involved in their metabolism. However, results from these candidate gene studies were inconsistent, possibly due to multiple testing and publication bias. In recent years, genome-wide association studies have identified a number of CRC susceptibility loci, and subsequent meta-analyses have observed limited evidence that diet may modify the risk associated with these susceptibility loci. Statistical techniques have been recently developed to evaluate the presence of interaction across the entire genome; results from these genome-wide studies have demonstrated limited evidence of interaction and have failed to replicate results from candidate gene studies and those using established susceptibility loci. However, larger sample sizes are likely needed to elucidate modest or weak interaction in genome-wide studies of gene-diet interaction.
Collapse
|
16
|
Copy number variation analysis on a non-Hodgkin lymphoma case-control study identifies an 11q25 duplication associated with diffuse large B-cell lymphoma. PLoS One 2014; 9:e105382. [PMID: 25133503 PMCID: PMC4136881 DOI: 10.1371/journal.pone.0105382] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 07/23/2014] [Indexed: 11/19/2022] Open
Abstract
Recent GWAS have identified several susceptibility loci for NHL. Despite these successes, much of the heritable variation in NHL risk remains to be explained. Common copy-number variants are important genomic sources of variability, and hence a potential source to explain part of this missing heritability. In this study, we carried out a CNV analysis using GWAS data from 681 NHL cases and 749 controls to explore the relationship between common structural variation and lymphoma susceptibility. Here we found a novel association with diffuse large B-cell lymphoma (DLBCL) risk involving a partial duplication of the C-terminus region of the LOC283177 long non-coding RNA that was further confirmed by quantitative PCR. For chronic lymphocytic leukemia/small lymphocytic lymphoma (CLL/SLL), known somatic deletions were identified on chromosomes 13q14, 11q22-23, 14q32 and 22q11.22. Our study shows that GWAS data can be used to identify germline CNVs associated with disease risk for DLBCL and somatic CNVs for CLL/SLL.
Collapse
|
17
|
Demichelis F, Stanford JL. Genetic predisposition to prostate cancer: Update and future perspectives. Urol Oncol 2014; 33:75-84. [PMID: 24996773 DOI: 10.1016/j.urolonc.2014.04.021] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 04/27/2014] [Accepted: 04/28/2014] [Indexed: 02/08/2023]
Abstract
OBJECTIVE Prostate cancer is the second most frequent cancer in men worldwide and kills over 250,000 men worldwide every year. Prostate cancer is a heterogeneous disease at the clinical and the molecular level. The Scandinavian Twin Registry Study demonstrated that in contrast to most malignancies where environment was the overriding influence, heritable factors account for more than fifty percent of prostate cancers. METHODS AND MATERIALS We review the literature on prostate cancer risk variants (rare and common) including SNPs and Copy Number Variants (CNVs) and discuss the potential implications of significant variants for prostate cancer patient care. RESULTS The search for prostate cancer susceptibility genes has included both family-based studies and case-control studies utilizing a variety of approaches from array-based to sequencing-based studies. A major challenge is to identify genetic variants associated with more aggressive, potentially lethal prostate cancer and to understand their role in the progression of the disease. CONCLUSION Future risk models useful in the clinical setting will likely incorporate several risk loci rather than single variants and may be dependent on an individual patient's ethnic background.
Collapse
Affiliation(s)
- Francesca Demichelis
- Centre for Integrative Biology, University of Trento, Trento, Italy; Institute for Computational Biomedicine, Weill Medical College of Cornell University, New York, NY; Institute for Precision Medicine, Weill Medical College of Cornell University and New York Presbyterian Hospital, New York, NY.
| | - Janet L Stanford
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA; Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA
| |
Collapse
|
18
|
Rasool S, Rasool V, Naqvi T, Ganai BA, Shah BA. Genetic unraveling of colorectal cancer. Tumour Biol 2014; 35:5067-82. [PMID: 24573608 DOI: 10.1007/s13277-014-1713-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 01/29/2014] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer is a common disease in both men and women (being the third most common cancer in men and the second most common among women) and thus represents an important and serious public health issue, especially in the western world. Although it is a well-established fact that cancers of the large intestine produce symptoms relatively earlier at a stage that can be easily cured by resection, a large number of people lose their lives to this deadly disease each year. Recent times have seen an important change in the incidence of colorectal cancer in different parts of the world. The etiology of colorectal cancer is multifactorial and is likely to involve the actions of genes at multiple levels along the multistage carcinogenesis process. Exhaustive efforts have been made out in the direction of unraveling the role of various environmental factors, gene mutations, and polymorphisms worldwide (as well as in Kashmir-"a valley of gastrointestinal cancers") that have got a role to play in the development of this disease so that antitumor drugs could be developed against this cancer, first, and, finally, the responsiveness or resistance to these agents could be understood for combating this global issue.
Collapse
Affiliation(s)
- Sabha Rasool
- Department of Biochemistry, University of Kashmir, Hazratbal, Srinagar, 190006, Kashmir, India
| | | | | | | | | |
Collapse
|