1
|
DeBenedictis JN, Baars E, Ochoteco-Asensio J, van Breda SG, de Kok TM. Genetic Variability Impacts Genotoxic and Transcriptome Responses in the Human Colon after the Consumption of Processed Red Meat Products and Those with Added Phytochemical Extracts. Nutrients 2024; 16:425. [PMID: 38337709 PMCID: PMC10857093 DOI: 10.3390/nu16030425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/22/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
The PHYTOME study investigated the effect of consuming processed meat products on outcomes related to colorectal cancer risk without testing the impact of genetic variability on these responses. This research aims to elucidate the genetic impact on apparent total N-nitroso compound (ATNC) excretion, colonic DNA adduct formation, ex vivo-induced DNA damage, and gene expression changes in colon biopsies of healthy participants. Through a systematic literature review, candidate polymorphisms were selected and then detected using TaqMan and PCR analysis. The effect of genotype on study outcomes was determined via a linear mixed model and analysis of variance. Machine learning was used to evaluate relative allele importance concerning genotoxic responses, which established a ranking of the most protective alleles and a combination of genotypes (gene scores). Participants were grouped by GSTM1 genotype and differentially expressed genes (DEGs), and overrepresented biological pathways were compared between groups. Stratifying participants by ten relevant genes revealed significant variations in outcome responses. After consumption of processed red meat, variations in NQO1 and COMT impacted responses in ATNC levels (µmol/L) (+9.56 for wildtype vs. heterozygous) and DNA adduct levels (pg/µg DNA) (+1.26 for variant vs. wildtype and +0.43 for variant vs. heterozygous), respectively. After phytochemicals were added to the meat, GSTM1 variation impacted changes in DNA adduct levels (-6.12 for deletion vs. wildtype). The gene scores correlated with these responses and DEGs were identified by GSTM1 genotype. The altered pathways specific to the GSTM1 wildtype group included 'metabolism', 'cell cycle', 'vitamin D receptor', and 'metabolism of water-soluble vitamins and co-factors'. Genotype impacted both the potential genotoxicity of processed red meat and the efficacy of protective phytochemical extracts.
Collapse
Affiliation(s)
| | | | | | - Simone G. van Breda
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands (J.O.-A.)
| | | |
Collapse
|
2
|
Sivasubramanian BP, Dave M, Panchal V, Saifa-Bonsu J, Konka S, Noei F, Nagaraj S, Terpari U, Savani P, Vekaria PH, Samala Venkata V, Manjani L. Comprehensive Review of Red Meat Consumption and the Risk of Cancer. Cureus 2023; 15:e45324. [PMID: 37849565 PMCID: PMC10577092 DOI: 10.7759/cureus.45324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/15/2023] [Indexed: 10/19/2023] Open
Abstract
Red and processed meat consumption rates are increasing in the United States. In this review, we present the current evidence that links red meat consumption and cancer development. A literature search was conducted in the PubMed and Google Scholar databases to review red meat consumption and its association with breast cancer and gastrointestinal cancer. Due to the presence of heme iron, which triggers oxidative reactions that eventually result in tumor formation, red meat consumption is strongly associated with the development of breast cancer. Ingestion of red meat increases Helicobacter pylori infections, resulting in enhanced expression of the CagA gene and the secretion of pro-inflammatory cytokines. This is the leading cause of gastric cancer. There is a strong correlation between heterocyclic amines and polycyclic aromatic hydrocarbons in red meat and the development of pancreatic cancer. However, additional research is necessary to confirm this finding. Adult colorectal cancer is caused by the formation of heterocyclic amines and DNA adducts due to the intake of red and processed meats cooked at higher temperatures. The consumption of poultry is associated with a reduced risk of breast and gastrointestinal cancers, but the results are inconsistent. The evidence is strong for the association between red meat and breast cancer and most gastric cancers. The presence of aromatic hydrocarbons, heterocyclic amines, and heme iron in red meat has been found to be behind tumorigenesis. Poultry has been shown to have a low association with cancer, but additional research is needed.
Collapse
Affiliation(s)
- Barath Prashanth Sivasubramanian
- Infectious Diseases, University of Texas Health Science Center at San Antonio, San Antonio, USA
- Internal Medicine, Employees State Insurance Corporation Medical College (ESIC-MC) & Post Graduate Institute of Medical Sciences and Research (PGIMSR), Chennai, IND
| | - Mihir Dave
- Internal Medicine, Medical College Baroda, Vadodara, IND
| | - Viraj Panchal
- Internal Medicine, Smt. Nathiba Hargovandas Lakhmichand Municipal Medical College, Ahmedabad, IND
| | | | - Srujana Konka
- Internal Medicine, Prathima Institute of Medical Sciences, Nagunur, IND
| | - Farahnaz Noei
- Internal Medicine, University of Ottawa, Ottawa, CAN
| | | | | | - Priya Savani
- Internal Medicine, Surat Municipal Institute of Medical Education & Research, Surat, IND
| | | | | | - Lokesh Manjani
- Internal Medicine, MedStar Washington Hospital Center, Washington, USA
| |
Collapse
|
3
|
Miao YD, Quan WX, Dong X, Gan J, Ji CF, Wang JT, Zhang F. Prognosis-related metabolic genes in the development of colorectal cancer progress and perspective. Gene 2023; 862:147263. [PMID: 36758843 DOI: 10.1016/j.gene.2023.147263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/10/2023]
Abstract
Colorectal cancer (CRC) is one of the most commonplace malignant tumors in the world. The occurrence and development of CRC are involved in numerous events. Metabolic reprogramming is one of the hallmarks of cancer and is convoluted and associated with carcinogenesis. Lots of metabolic genes are involved in the occurrence and progression of CRC. Study methods combining tumor genomics and metabolomics are more likely to explore this field in depth. In this mini-review, we make the latest progress and future prospects into the different molecular mechanisms of seven prognosis-related metabolic genes, we screened out in previous research, involved in the occurrence and development of CRC.
Collapse
Affiliation(s)
- Yan-Dong Miao
- The Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China
| | - Wu-Xia Quan
- Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China
| | - Xin Dong
- The Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China
| | - Jian Gan
- Department of Gastroenterology, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China
| | - Cui-Feng Ji
- Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China
| | - Jiang-Tao Wang
- Department of Thyroid and Breast Surgery, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China
| | - Fang Zhang
- The Cancer Center, Yantai Affiliated Hospital of Binzhou Medical University, The 2nd Medical College of Binzhou Medical University, Yantai 264100, China.
| |
Collapse
|
4
|
Zhu C, Wang Z, Cai J, Pan C, Lin S, Zhang Y, Chen Y, Leng M, He C, Zhou P, Wu C, Fang Y, Li Q, Li A, Liu S, Lai Q. VDR Signaling via the Enzyme NAT2 Inhibits Colorectal Cancer Progression. Front Pharmacol 2021; 12:727704. [PMID: 34867333 PMCID: PMC8635240 DOI: 10.3389/fphar.2021.727704] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 10/04/2021] [Indexed: 12/31/2022] Open
Abstract
Recent epidemiological and preclinical evidence indicates that vitamin D3 inhibits colorectal cancer (CRC) progression, but the mechanism has not been completely elucidated. This study was designed to determine the protective effects of vitamin D3 and identify crucial targets and regulatory mechanisms in CRC. First, we confirmed that 1,25(OH)2D3, the active form of vitamin D3, suppressed the aggressive phenotype of CRC in vitro and in vivo. Based on a network pharmacological analysis, N-acetyltransferase 2 (NAT2) was identified as a potential target of vitamin D3 against CRC. Clinical data of CRC patients from our hospital and bioinformatics analysis by online databases indicated that NAT2 was downregulated in CRC specimens and that the lower expression of NAT2 was correlated with a higher metastasis risk and lower survival rate of CRC patients. Furthermore, we found that NAT2 suppressed the proliferation and migration capacity of CRC cells, and the JAK1/STAT3 signaling pathway might be the underlying mechanism. Moreover, Western blot and immunofluorescence staining assays demonstrated that 1,25(OH)2D3 promoted NAT2 expression, and the chromatin immunoprecipitation assay indicated that the vitamin D receptor (VDR) transcriptionally regulated NAT2. These findings expand the potential uses of vitamin D3 against CRC and introduce VDR signaling via the enzyme NAT2 as a potential diagnostic and therapeutic target for CRC.
Collapse
Affiliation(s)
- Chaojun Zhu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zihuan Wang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianqun Cai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chunqiu Pan
- Department of Emergency Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Simin Lin
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Zhang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuting Chen
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengxin Leng
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chengcheng He
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peirong Zhou
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China.,Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Changjie Wu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yuxin Fang
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qingyuan Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Aimin Li
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Side Liu
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiuhua Lai
- Guangdong Provincial Key Laboratory of Gastroenterology, Department of Gastroenterology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
5
|
Abstract
Nowadays multiple heterogeneous chemicals affect the human body. They include drugs, household chemicals, dyes, food supplements and others. The human organism can modify, inactivate, and eliminate the chemicals by biotransformation enzymes. But it is well known that biotransformation can lead to toxification phenomenon. Individuals differ from each other by the rate of chemical modification that promotes accumulation of toxins and carcinogens in some patients. An N-acetyltransferase 2 enzyme participates in the aromatic amines second phase metabolism. This work reviews the acetyltransferase gene polymorphism possible role in diseases development including drug-induced organs damage.Gene of acetyltransferase has polymorphisms associated with two haplotypes of fast and slow substrate acetylation. Gene alleles combine in three genotypes: fast, intermediate, and slow acetylators. Acetylation rate plays a significant role in side effects development during tuberculosis treatment and cancer pathogenesis. Recently, new data described the role of enzyme in development of non-infectious diseases in the human. Scientists consider that slow acetylation genotype in combination with high xenobiotic load result in accumulation of toxic substances able to damage cells.Therefore, acetyltransferase genotyping helps to reveal risk groups of cancer and non-infectious disease development and to prescribe more effective and safe doses of drugs.
Collapse
|
6
|
Kossenas K, Constantinou C. Epidemiology, Molecular Mechanisms, and Clinical Trials: an Update on Research on the Association Between Red Meat Consumption and Colorectal Cancer. Curr Nutr Rep 2021; 10:435-467. [PMID: 34665439 DOI: 10.1007/s13668-021-00377-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2021] [Indexed: 11/28/2022]
Abstract
PURPOSE OF THE REVIEW Colorectal cancer is the second most common cause of cancer death in the world. The aim of this review is to provide an update on recent epidemiological studies, the molecular mechanisms involved, and ongoing clinical trials investigating the relationship between red meat consumption and colorectal cancer. RECENT FINDINGS Evidence in the literature proposes an association between red meat consumption and development of colorectal cancer, and there is some insight with regard to the mechanisms involved. Twenty studies of the IARC report (1990-2015) showed that red meat is positively associated with colorectal cancer whereas 14 studies either supported no positive association or no statistically significant association between red meat consumption and risk for CRC. More recent epidemiological studies conducted from 2016 and onwards provided further evidence that adherence to diets low in red and/or processed meat reduces the risk of colorectal cancer. Evidence from recent studies supports that quantity, doneness, and preparation of red meat play a role in colorectal carcinogenesis. Red meat's degradation products allow for the creation of a pro-inflammatory colonic microenvironment, and the gut microbiome plays a role in colorectal carcinogenesis. Heme, hydrogen sulfide, lipid peroxidation, nitroso compounds, and the bacterium Fusobacterium Nucleatum (as well as possibly other bacteria such as Akkermansia muciniphila, Eubacterium cylindroides, Eubacterium eligens 1 and 2, and Eubacterium rectale 1 and 2) also partake in the process of colorectal carcinogenesis. Several clinical trials are underway investigating the effects of different diets and red meat substitution products on colorectal cancer incidence as well as the underlying molecular mechanisms involved in the process.
Collapse
Affiliation(s)
- Konstantinos Kossenas
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, P.O. Box 24005, 21 Ilia Papakyriakou, 2414 Engomi, CY-1700, Nicosia, Cyprus
| | - Constantina Constantinou
- Department of Basic and Clinical Sciences, University of Nicosia Medical School, P.O. Box 24005, 21 Ilia Papakyriakou, 2414 Engomi, CY-1700, Nicosia, Cyprus.
| |
Collapse
|
7
|
Le Marchand L. The role of heterocyclic aromatic amines in colorectal cancer: the evidence from epidemiologic studies. Genes Environ 2021; 43:20. [PMID: 34099058 PMCID: PMC8183058 DOI: 10.1186/s41021-021-00197-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 05/31/2021] [Indexed: 11/29/2022] Open
Abstract
Since Dr. Sugimura’s discovery of heterocyclic aromatic amines (HAA) in broiled fish, many epidemiological studies have been conducted to investigate their role in human cancers, often focusing on colorectal cancer. The difficulty in measuring HAA exposure from meat and fish intake in these studies has resulted in inconsistent findings. Because studying individuals who may be particularly susceptible to the carcinogenic effects of HAA might facilitate the demonstration of a link with cancer, multiple studies have focused on individuals with the high activity phenotype for CYP1A2 and/or NAT2, the two main metabolic enzymes involved in the bioactivation of HAA. These investigations have also yielded inconsistent results. Two recent large pooled analyses of colorectal cancer studies have helped clarify the overall evidence. One was conducted in whites and reported no interaction of red meat intake and NAT2 genotype on risk in Whites. The other was conducted in Japanese and African Americans, two populations with high rates of the disease and a prevalence of the at-risk rapid NAT2 phenotype 10- and 2-fold greater than in whites, respectively. In those groups, a significant interaction was found, with the association of red meat with colorectal cancer being strongest among individuals with the rapid NAT2 phenotype, intermediate among those with the intermediate phenotype and not significant among those with the slow NAT2 phenotype. Recent research on biomarkers has focused on PhIP hair content, as a marker of exposure to HAA, and on DNA adducts using new sensitive quantitative methods, as markers of early biological effects. These advances, when brought to bear, may contribute greatly to the further elucidation of the carcinogenicity of HAA in humans.
Collapse
Affiliation(s)
- Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, 701 Ilalo Street, Honolulu, HI, 9681, USA.
| |
Collapse
|
8
|
Huang BZ, Wang S, Bogumil D, Wilkens LR, Wu L, Blot WJ, Zheng W, Shu XO, Pandol SJ, Le Marchand L, Setiawan VW. Red meat consumption, cooking mutagens, NAT1/2 genotypes and pancreatic cancer risk in two ethnically diverse prospective cohorts. Int J Cancer 2021; 149:10.1002/ijc.33598. [PMID: 33844845 PMCID: PMC8594451 DOI: 10.1002/ijc.33598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/17/2021] [Accepted: 03/19/2021] [Indexed: 12/17/2022]
Abstract
There is limited evidence on the association between red meat consumption and pancreatic cancer among ethnic minorities. We assessed this relationship in two large prospective cohorts: the Multiethnic Cohort Study (MEC) and the Southern Community Cohort Study (SCCS). Demographic, dietary and other risk factor data were collected at cohort entry. Red meat intake was assessed using cohort-specific validated food frequency questionnaires. Incident pancreatic cancer cases were identified via linkages to state cancer registries. Cox regression was used to calculate relative risks (RRs) and 95% confidence intervals (CIs) for the association of red meat intake with pancreatic cancer risk in each cohort. We performed additional analyses to evaluate cooking methods, mutagens and effect modification by NAT1/2 genotypes. From a total of 184 542 (MEC) and 66 793 (SCCS) at-risk participants, we identified 1618 (MEC) and 266 (SCCS) incident pancreatic cancer cases. Red meat consumption was associated with pancreatic cancer risk in the MEC (RRQ4vsQ1 1.18, 95% CI 1.02-1.37) and with borderline statistical significance in the SCCS (RRQ4vsQ1 1.31, 95% CI 0.93-1.86). This association was significant in African Americans (RRQ4vsQ1 1.49, 95% CI 1.06-2.11) and Latinos (RRQ4vsQ1 1.44, 95% CI 1.02-2.04) in the MEC, and among African Americans (RRQ4vsQ1 1.55, 95% CI 1.03-2.33) in the SCCS. NAT2 genotypes appeared to modify the relationship between red meat and pancreatic cancer in the MEC (pinteraction = 0.03). Our findings suggest that the associations for red meat may be strongest in African Americans and Latinos. The mechanisms underlying the increased risk for these populations should be further investigated.
Collapse
Affiliation(s)
- Brian Z. Huang
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Department of Research & Evaluation, Kaiser Permanente Southern California, Pasadena, CA
| | - Songren Wang
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - David Bogumil
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Lynne R. Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Lang Wu
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - William J. Blot
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Stephen J. Pandol
- Division of Gastroenterology, Department of Medicine, Cedars-Sinai Medical Center and Department of Veterans Affairs, Los Angeles, CA
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI
| | - Veronica Wendy Setiawan
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
- Norris Comprehensive Cancer Center, Los Angeles, CA
| |
Collapse
|
9
|
Medina-Sanson A, Núñez-Enríquez JC, Hurtado-Cordova E, Pérez-Saldivar ML, Martínez-García A, Jiménez-Hernández E, Fernández-López JC, Martín-Trejo JA, Pérez-Lorenzana H, Flores-Lujano J, Amador-Sánchez R, Mora-Ríos FG, Peñaloza-González JG, Duarte-Rodríguez DA, Torres-Nava JR, Flores-Bautista JE, Espinosa-Elizondo RM, Román-Zepeda PF, Flores-Villegas LV, González-Ulivarri JE, Martínez-Silva SI, Espinoza-Anrubio G, Almeida-Hernández C, Ramírez-Colorado R, Hernández-Mora L, García-López LR, Cruz-Ojeda GA, Godoy-Esquivel AE, Contreras-Hernández I, Medina-Hernández A, López-Caballero MG, Hernández-Pineda NA, Granados-Kraulles J, Rodríguez-Vázquez MA, Torres-Valle D, Cortés-Reyes C, Medrano-López F, Pérez-Gómez JA, Martínez-Ríos A, Aguilar-De Los Santos A, Serafin-Díaz B, Bekker-Méndez VC, Mata-Rocha M, Morales-Castillo BA, Sepúlveda-Robles OA, Ramírez-Bello J, Rosas-Vargas H, Hidalgo-Miranda A, Mejía-Aranguré JM, Jiménez-Morales S. Genotype-Environment Interaction Analysis of NQO1, CYP2E1, and NAT2 Polymorphisms and the Risk of Childhood Acute Lymphoblastic Leukemia: A Report From the Mexican Interinstitutional Group for the Identification of the Causes of Childhood Leukemia. Front Oncol 2020; 10:571869. [PMID: 33072605 PMCID: PMC7537417 DOI: 10.3389/fonc.2020.571869] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 08/17/2020] [Indexed: 11/26/2022] Open
Abstract
Background: Acute lymphoblastic leukemia (ALL) is the main type of cancer in children. In Mexico and other Hispanic populations, the incidence of this neoplasm is one of the highest reported worldwide. Functional polymorphisms of various enzymes involved in the metabolism of xenobiotics have been associated with an increased risk of developing ALL, and the risk is different by ethnicity. The aims of the present study were to identify whether NQO1, CYP2E1, and NAT2 polymorphisms or some genotype-environmental interactions were associated with ALL risk in Mexican children. Methods: We conducted a case-control study including 478 pediatric patients diagnosed with ALL and 284 controls (children without leukemia). Ancestry composition of a subset of cases and controls was assessed using 32 ancestry informative markers. Genetic-environmental interactions for the exposure to hydrocarbons were assessed by logistic regression analysis. Results: The polymorphisms rs1801280 (OR 1.54, 95% CI 1.21–1.93), rs1799929 (OR 1.96, 95% CI 1.55–2.49), and rs1208 (OR 1.44, 95% CI 1.14–1.81) were found to increase the risk of ALL; being the risks higher under a recessive model (OR 2.20, 95% CI 1.30–1.71, OR 3.87, 95% CI 2.20–6.80, and OR 2.26, 95% CI 1.32–3.87, respectively). Gene-environment interaction analysis showed that NAT2 rs1799929 TT genotype confers high risk to ALL under exposure to fertilizers, insecticides, hydrocarbon derivatives, and parental tobacco smoking. No associations among NQO1, CYP2E1, and ALL were observed. Conclusion: Our study provides evidence for the association between NAT2 polymorphisms/gene-environment interactions, and the risk of childhood ALL in Mexican children.
Collapse
Affiliation(s)
- Aurora Medina-Sanson
- Departamento de Hemato-Oncología, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Mexico City, Mexico.,Programa de Maestría y Doctorado en Ciencias Médicas de la Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM)Mexico City, Mexico
| | - Juan Carlos Núñez-Enríquez
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Eduardo Hurtado-Cordova
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico.,Universidad Xochicalco, Campos Tijuana, Tijuana, Mexico
| | - María Luisa Pérez-Saldivar
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Anayeli Martínez-García
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico.,Facultad de Estudios Superiores Zaragoza, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Elva Jiménez-Hernández
- Servicio de Hematología Pediátrica, Centro Médico Nacional "La Raza", Hospital General "Gaudencio González Garza", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Jorge Alfonso Martín-Trejo
- Servicio de Hematología Pediátrica, Centro Médico Nacional "Siglo XXI", UMAE Hospital de Pediatría "Dr. Silvestre Frenk Freund", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Héctor Pérez-Lorenzana
- Servicio de Cirugía Pediátrica, Hospital General "Gaudencio González Garza", Centro Médico Nacional Siglo XXI (CMN) "La Raza", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Janet Flores-Lujano
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Raquel Amador-Sánchez
- Servicio de Hematología Pediátrica, Hospital General Regional "Carlos McGregor Sánchez Navarro", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Felix Gustavo Mora-Ríos
- Cirugía Pediátrica del Hospital Regional "General Ignacio Zaragoza", Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | | | - David Aldebarán Duarte-Rodríguez
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - José Refugio Torres-Nava
- Servicio de Oncología, Hospital Pediátrico de Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | | | | | - Pedro Francisco Román-Zepeda
- Coordinación Clínica y Servicio de Cirugía pediátrica, Hospital General Regional (HGR) No. 1 "Dr. Carlos Mac Gregor Sánchez Navarro", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Luz Victoria Flores-Villegas
- Servicio de Hematología Pediátrica, Centro Médico Nacional "20 de Noviembre", Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Juana Esther González-Ulivarri
- Jefatura de Enseñanza, Hospital Pediátrico de Iztacalco, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Sofía Irene Martínez-Silva
- Jefatura de Enseñanza, Hospital Pediátrico de Iztapalapa, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Gilberto Espinoza-Anrubio
- Servicio de Pediatría, Hospital General Zona (HGZ) No. 8 "Dr. Gilberto Flores Izquierdo", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Carolina Almeida-Hernández
- Jefatura de Enseñanza, Hospital General de Ecatepec "Las Américas", Instituto de Salud del Estado de México (ISEM), Mexico City, Mexico
| | - Rosario Ramírez-Colorado
- Jefatura de Enseñanza, Hospital Pediátrico La Villa, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Luis Hernández-Mora
- Jefatura de Enseñanza, Hospital Pediátrico San Juan de Aragón, Secretaría de Salud (SS), Mexico City, Mexico
| | - Luis Ramiro García-López
- Servicio de Pediatría, Hospital Pediátrico de Tacubaya, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Gabriela Adriana Cruz-Ojeda
- Coordinación Clínica de Educación e Investigación en Salud, Hospital General de Zona (HGZ) No. 47, IMSS, Mexico City, Mexico
| | - Arturo Emilio Godoy-Esquivel
- Servicio de Cirugía Pediátrica, Hospital Pediátrico de Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Iris Contreras-Hernández
- Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Abraham Medina-Hernández
- Pediatría, Hospital Materno-Pediátrico de Xochimilco, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - María Guadalupe López-Caballero
- Jefatura de Enseñanza, Hospital Pediátrico de Coyoacán, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Norma Angélica Hernández-Pineda
- Coordinación Clínica y Pediatría del Hospital General de Zona 76, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Jorge Granados-Kraulles
- Coordinación Clínica y Pediatría del Hospital General de Zona 76, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - María Adriana Rodríguez-Vázquez
- Coordinación Clínica y Pediatría del Hospital General de Zona 68, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Delfino Torres-Valle
- Coordinación Clínica y Pediatría del Hospital General de Zona 71, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Carlos Cortés-Reyes
- Pediatría, Hospital General Dr. Darío Fernández Fierro, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Francisco Medrano-López
- Coordinación Clínica y Servicio de Pediatría, Hospital General Regional (HGR) No. 72 "Dr. Vicente Santos Guajardo", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Jessica Arleet Pérez-Gómez
- Coordinación Clínica y Servicio de Pediatría, Hospital General Regional (HGR) No. 72 "Dr. Vicente Santos Guajardo", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Annel Martínez-Ríos
- Cirugía Pediátrica del Hospital Regional "General Ignacio Zaragoza", Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - Antonio Aguilar-De Los Santos
- Coordinación Clínica y Pediatría del Hospital General de Zona 98, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Berenice Serafin-Díaz
- Coordinación Clínica y Pediatría del Hospital General de Zona 57, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Vilma Carolina Bekker-Méndez
- Hospital de Infectología "Dr. Daniel Méndez Hernández", "La Raza", Instituto Mexicano del Seguro Social (IMSS), Unidad de Investigación Médica en Inmunología e Infectología, Mexico City, Mexico
| | - Minerva Mata-Rocha
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Blanca Angélica Morales-Castillo
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Omar Alejandro Sepúlveda-Robles
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Haydeé Rosas-Vargas
- Unidad de Investigación Médica en Genética Humana, UMAE Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional "Siglo XXI", Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Alfredo Hidalgo-Miranda
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Juan Manuel Mejía-Aranguré
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Médica de Alta Especialidad Hospital de Pediatría "Dr. Silvestre Frenk Freund", Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico.,Coordinación de Investigación en Salud, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Silvia Jiménez-Morales
- Laboratorio de Genómica del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
10
|
Wang ST, Cui WQ, Pan D, Jiang M, Chang B, Sang LX. Tea polyphenols and their chemopreventive and therapeutic effects on colorectal cancer. World J Gastroenterol 2020; 26:562-597. [PMID: 32103869 PMCID: PMC7029350 DOI: 10.3748/wjg.v26.i6.562] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 12/30/2019] [Accepted: 01/11/2020] [Indexed: 02/06/2023] Open
Abstract
Colorectal cancer (CRC), a multifactorial disease, is usually induced and developed through complex mechanisms, including impact of diet and lifestyle, genomic abnormalities, change of signaling pathways, inflammatory response, oxidation stress, dysbiosis, and so on. As natural polyphenolic phytochemicals that exist primarily in tea, tea polyphenols (TPs) have been shown to have many clinical applications, especially as anticancer agents. Most animal studies and epidemiological studies have demonstrated that TPs can prevent and treat CRC. TPs can inhibit the growth and metastasis of CRC by exerting the anti-inflammatory, anti-oxidative or pro-oxidative, and pro-apoptotic effects, which are achieved by modulations at multiple levels. Many experiments have demonstrated that TPs can modulate several signaling pathways in cancer cells, including the mitogen-activated protein kinase pathway, phosphatidylinositol-3 kinase/Akt pathway, Wnt/β-catenin pathway, and 67 kDa laminin receptor pathway, to inhibit proliferation and promote cell apoptosis. In addition, novel studies have also suggested that TPs can prevent the growth and metastasis of CRC by modulating the composition of gut microbiota to improve immune system and decrease inflammatory responses. Molecular pathological epidemiology, a novel multidisciplinary investigation, has made great progress on CRC, and the further molecular pathological epidemiology research should be developed in the field of TPs and CRC. This review summarizes the existing in vitro and in vivo animal and human studies and potential mechanisms to examine the effects of tea polyphenols on CRC.
Collapse
Affiliation(s)
- Shi-Tong Wang
- Department of Cardiovascular Ultrasound, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Wen-Qi Cui
- Department of Neurology, Shengjing Hospital, Affiliated Hospital of China Medical University, Shenyang 110004, Liaoning Province, China
| | - Dan Pan
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Min Jiang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| | - Li-Xuan Sang
- Department of Geriatrics, First Affiliated Hospital of China Medical University, Shenyang 110001, Liaoning Province, China
| |
Collapse
|
11
|
Mint Sidi Ould Deoula M, Huybrechts I, El Kinany K, Boudouaya H, Hatime Z, El Asri A, Benslimane A, Nejjari C, Sidi Adil I, El Rhazi K. Behavioral, Nutritional, and Genetic Risk Factors of Colorectal Cancers in Morocco: Protocol for a Multicenter Case-Control Study. JMIR Res Protoc 2020; 9:e13998. [PMID: 31929106 PMCID: PMC7006499 DOI: 10.2196/13998] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 06/27/2019] [Accepted: 07/19/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Colorectal cancer (CRC) has been reported as the third most commonly diagnosed cancer worldwide and is currently considered as a major public health concern. A peak increase in incidence has been noted in economically transitioning countries like Morocco where industrialization started shifting from a traditional lifestyle and diet toward a more westernized diet and lifestyle. OBJECTIVE This paper aims to present the protocol of a large-scale Moroccan case-control study that aims at investigating associations of diet, other lifestyle factors, and genetic traits with CRC risk in Morocco. METHODS A case-control study was conducted between 2009 and 2017, including 3032 case-control pairs (1516 cases and 1516 controls) matched on sex, age, and center in 5 major public health hospitals in Morocco. Questionnaires on sociodemographic data, lifestyle, family history of CRC, and nonsteroidal anti-inflammatory drugs (NSAIDs) were completed by trained investigators during face-to-face interviews. In addition, participants completed a semiquantitative food-frequency questionnaire, developed to assess food intake in the Moroccan population. Information regarding genetic factors was recorded for cases, and paraffin blocks (with embedded tumor tissues) are available in 3 collaborating hospitals. Conditional logistic regression analysis is planned to assess associations between diet and CRC risk. Binary logistic regression is considered to predict associations between mutations and nutritional risk factors including only CRC case series. RESULTS Altogether, 2966 cases-control pairs (1483 cases and 1483 controls) were considered eligible and included in this study. Both cases and controls did not differ significantly with respect to age (P=.36), sex (P=.51), center (P>.99), marital status (P=.30), and NSAID use (P=.08). However, participants in the control group were significantly more likely to have a high income level and live in urban areas and to have a high level of education than cases. CONCLUSIONS This is the first study investigating potential risk factors of CRC such as lifestyle, diet, and genetic factors, originating from a southern Mediterranean country with low but increasing CRC prevalence. Identified risk factors allow the establishment of evidence-based preventive actions regarding nutrition and other lifestyle habits adapted to the Moroccan context. In brief, this study will promote cancer research and prevention in Morocco. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) RR1-10.2196/13998.
Collapse
Affiliation(s)
| | | | | | | | - Zineb Hatime
- Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | | | | | | | | | | |
Collapse
|
12
|
Yang T, Li X, Montazeri Z, Little J, Farrington SM, Ioannidis JP, Dunlop MG, Campbell H, Timofeeva M, Theodoratou E. Gene-environment interactions and colorectal cancer risk: An umbrella review of systematic reviews and meta-analyses of observational studies. Int J Cancer 2019; 145:2315-2329. [PMID: 30536881 PMCID: PMC6767750 DOI: 10.1002/ijc.32057] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 11/06/2018] [Accepted: 11/15/2018] [Indexed: 12/14/2022]
Abstract
The cause of colorectal cancer (CRC) is multifactorial, involving both genetic variants and environmental risk factors. We systematically searched the MEDLINE, EMBASE, China National Knowledge Infrastructure (CNKI) and Wanfang databases from inception to December 2016, to identify systematic reviews and meta-analyses of observational studies that investigated gene-environment (G×E) interactions in CRC risk. Then, we critically evaluated the cumulative evidence for the G×E interactions using an extension of the Human Genome Epidemiology Network's Venice criteria. Overall, 15 articles reporting systematic reviews of observational studies on 89 G×E interactions, 20 articles reporting meta-analyses of candidate gene- or single-nucleotide polymorphism-based studies on 521 G×E interactions, and 8 articles reporting 33 genome-wide G×E interaction analyses were identified. On the basis of prior and observed scores, only the interaction between rs6983267 (8q24) and aspirin use was found to have a moderate overall credibility score as well as main genetic and environmental effects. Though 5 other interactions were also found to have moderate evidence, these interaction effects were tenuous due to the lack of main genetic effects and/or environmental effects. We did not find highly convincing evidence for any interactions, but several associations were found to have moderate strength of evidence. Our conclusions are based on application of the Venice criteria which were designed to provide a conservative assessment of G×E interactions and thus do not include an evaluation of biological plausibility of an observed joint effect.
Collapse
Affiliation(s)
- Tian Yang
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
| | - Xue Li
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
| | - Zahra Montazeri
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Julian Little
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Susan M. Farrington
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| | - John P.A. Ioannidis
- Stanford Prevention Research Center, Departments of Medicine, of Health Research and Policy, and of Biomedical Data Science, Stanford University School of Medicine, and Department of StatisticsStanford University School of Humanities and SciencesStanfordCaliforniaUSA
- Meta‐Research Innovation Center at Stanford (METRICS)Stanford UniversityStanfordCaliforniaUSA
| | - Malcolm G. Dunlop
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
| | - Maria Timofeeva
- Colon Cancer Genetics Group, Medical Research Council Human Genetics Unit, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| | - Evropi Theodoratou
- Centre for Global Health Research, Usher Institute of Population Health Sciences and InformaticsThe University of EdinburghEdinburghUnited Kingdom
- Cancer Research UK Edinburgh Centre, Medical Research Council Institute of Genetics & Molecular MedicineWestern General Hospital, The University of EdinburghEdinburghUnited Kingdom
| |
Collapse
|
13
|
Klusek J, Nasierowska-Guttmejer A, Kowalik A, Wawrzycka I, Chrapek M, Lewitowicz P, Radowicz-Chil A, Klusek J, Głuszek S. The Influence of Red Meat on Colorectal Cancer Occurrence Is Dependent on the Genetic Polymorphisms of S-Glutathione Transferase Genes. Nutrients 2019; 11:E1682. [PMID: 31336627 PMCID: PMC6683097 DOI: 10.3390/nu11071682] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/17/2019] [Accepted: 07/19/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND It is postulated that both individual genotype and environmental factors such as diet may modify the risk of developing colorectal cancer (CRC). The influences of GST gene polymorphism and red meat intake on CRC occurrence in the Polish population were analyzed in this study. METHODS Genotyping was performed with the qPCR method. RESULTS A high frequency of meat consumption was associated with an over 2-fold increase in the risk of colorectal cancer odds ratio (OR) adjusted for sex and age = 2.4, 95% confidence interval (CI); 1.3-4.4). However, after analyzing the genetic profiles, in the absence of polymorphisms of all three analyzed genes, there was no association between a high frequency of meat consumption and the occurrence of CRC. In the case of GSTM1 gene polymorphism, the high frequency of meat consumption increased the risk of CRC by almost more than 4 times (OR adjusted for sex and age = 3.8, 95% CI: 1.6-9.1). For GSTP1 gene polymorphism, a 3-fold increase in CRC risk was observed with a high frequency of meat consumption (OR adjusted for sex and age = 3.4, 95% CI: 1.4-8.1). In the case of GSTT1 gene polymorphism, the increase in risk of CRC was not statistically significant (OR adjusted for sex and age = 1.9, 95% CI: 0.4-8.5). CONCLUSIONS The frequency of red meat intake in non-smokers increases the risk of colon cancer in the case of GST gene polymorphisms.
Collapse
Affiliation(s)
- Justyna Klusek
- Department of Surgery and Surgical Nursery with a Research Laboratory and Genetic Laboratory, Faculty of Medicine and Health Sciences, Jan Kochanowski University, 19, 5-317 Kielce, Poland.
| | - Anna Nasierowska-Guttmejer
- Department of Pathology, Faculty of Medicine and Health Sciences, Jan Kochanowski University, 19, 25-317 Kielce, Poland
| | - Artur Kowalik
- Department of Molecular Diagnostic, Holy Cross Cancer Centre, 19, 25-734 Kielce, Poland
| | - Iwona Wawrzycka
- Department of Surgery and Surgical Nursery with a Research Laboratory and Genetic Laboratory, Faculty of Medicine and Health Sciences, Jan Kochanowski University, 19, 5-317 Kielce, Poland
- Department of General, Oncological and Endocrinological Surgery, Voivodeship Hospital, 19, 25-736 Kielce, Poland
| | - Magdalena Chrapek
- Department of Probability Calculus and Statistics, Institute of Mathematics, Jan Kochanowski University, 19, 25-406 Kielce, Poland
| | - Piotr Lewitowicz
- Department of Pathology, Faculty of Medicine and Health Sciences, Jan Kochanowski University, 19, 25-317 Kielce, Poland
| | - Agnieszka Radowicz-Chil
- Department of Pathology, Faculty of Medicine and Health Sciences, Jan Kochanowski University, 19, 25-317 Kielce, Poland
| | - Jolanta Klusek
- Department of Physiology, Institute of Biology, Jan Kochanowski University, 19, 25-406 Kielce, Poland
| | - Stanisław Głuszek
- Department of Surgery and Surgical Nursery with a Research Laboratory and Genetic Laboratory, Faculty of Medicine and Health Sciences, Jan Kochanowski University, 19, 5-317 Kielce, Poland
- Department of General, Oncological and Endocrinological Surgery, Voivodeship Hospital, 19, 25-736 Kielce, Poland
| |
Collapse
|
14
|
The Impact of Coffee and Its Selected Bioactive Compounds on the Development and Progression of Colorectal Cancer In Vivo and In Vitro. Molecules 2018; 23:molecules23123309. [PMID: 30551667 PMCID: PMC6321559 DOI: 10.3390/molecules23123309] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022] Open
Abstract
Coffee is one of the most popular beverages worldwide. Coffee contains bioactive compounds that affect the human body such as caffeine, caffeic acid, chlorogenic acids, trigonelline, diterpenes, and melanoidins. Some of them have demonstrated potential anticarcinogenic effects in animal models and in human cell cultures, and may play a protective role against colorectal cancer. Colorectal cancer (CRC) is the third leading cause of cancer-related mortality in the USA and other countries. Dietary patterns, as well as the consumption of beverages, may reduce the risk of CRC incidence. In this review, we focus on published epidemiological studies concerning the association of coffee consumption and the risk of development of colorectal cancer, and provide a description of selected biologically active compounds in coffee that have been investigated as potential cancer-combating compounds: Caffeine, caffeic acid (CA), chlorogenic acids (CGAs), and kahweol in relation to colorectal cancer progression in in vitro settings. We review the impact of these substances on proliferation, viability, invasiveness, and metastasis, as well as on susceptibility to chemo- and radiotherapy of colorectal cancer cell lines cultured in vitro.
Collapse
|
15
|
Effects of gene polymorphisms of metabolic enzymes on the association between red and processed meat consumption and the development of colon cancer; a literature review. J Nutr Sci 2018; 7:e26. [PMID: 30305892 PMCID: PMC6176493 DOI: 10.1017/jns.2018.17] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 06/08/2018] [Accepted: 08/17/2018] [Indexed: 12/15/2022] Open
Abstract
The role of environmental factors and genetic susceptibility in the development of colon cancer (CC) has been already proven, but the role of gene polymorphisms in modifying the risk of environmental factors such as nutritional factors is still unknown. This study aimed to investigate the effect of polymorphisms of involved genes in the association between red meat consumption and the development of CC. The present review was carried out using keywords such as polymorphism and/or protein and/or red meat and/or processed meat and/or colon cancer. PubMed and Science Direct databases were used to collect all related articles published from 2001 to 2017. The presence of SNP in the coding genes of proteins involved in metabolism of nutrients could play significant roles in the extent of the effects of nutrition in the development of CC. The effect of dietary proteins greatly depends on the polymorphisms in the metabolising genes of these substances. Gene polymorphisms may have a role in colorectal cancer risk, especially in people with high meat intake, and this leads to a difference in the effects of meat consumption in different individuals. To conclude, dietary recommendations for the prevention and control of CC should be modified based on the genotype of different individuals. Increasing our knowledge on this field of nutritional genomics can lead to personalised preventive and therapeutic recommendations for CC patients.
Collapse
|
16
|
Jeon J, Du M, Schoen RE, Hoffmeister M, Newcomb PA, Berndt SI, Caan B, Campbell PT, Chan AT, Chang-Claude J, Giles GG, Gong J, Harrison TA, Huyghe JR, Jacobs EJ, Li L, Lin Y, Le Marchand L, Potter JD, Qu C, Bien SA, Zubair N, Macinnis RJ, Buchanan DD, Hopper JL, Cao Y, Nishihara R, Rennert G, Slattery ML, Thomas DC, Woods MO, Prentice RL, Gruber SB, Zheng Y, Brenner H, Hayes RB, White E, Peters U, Hsu L. Determining Risk of Colorectal Cancer and Starting Age of Screening Based on Lifestyle, Environmental, and Genetic Factors. Gastroenterology 2018; 154:2152-2164.e19. [PMID: 29458155 PMCID: PMC5985207 DOI: 10.1053/j.gastro.2018.02.021] [Citation(s) in RCA: 193] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Revised: 01/22/2018] [Accepted: 02/06/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND & AIMS Guidelines for initiating colorectal cancer (CRC) screening are based on family history but do not consider lifestyle, environmental, or genetic risk factors. We developed models to determine risk of CRC, based on lifestyle and environmental factors and genetic variants, and to identify an optimal age to begin screening. METHODS We collected data from 9748 CRC cases and 10,590 controls in the Genetics and Epidemiology of Colorectal Cancer Consortium and the Colorectal Transdisciplinary study, from 1992 through 2005. Half of the participants were used to develop the risk determination model and the other half were used to evaluate the discriminatory accuracy (validation set). Models of CRC risk were created based on family history, 19 lifestyle and environmental factors (E-score), and 63 CRC-associated single-nucleotide polymorphisms identified in genome-wide association studies (G-score). We evaluated the discriminatory accuracy of the models by calculating area under the receiver operating characteristic curve values, adjusting for study, age, and endoscopy history for the validation set. We used the models to project the 10-year absolute risk of CRC for a given risk profile and recommend ages to begin screening in comparison to CRC risk for an average individual at 50 years of age, using external population incidence rates for non-Hispanic whites from the Surveillance, Epidemiology, and End Results program registry. RESULTS In our models, E-score and G-score each determined risk of CRC with greater accuracy than family history. A model that combined both scores and family history estimated CRC risk with an area under the receiver operating characteristic curve value of 0.63 (95% confidence interval, 0.62-0.64) for men and 0.62 (95% confidence interval, 0.61-0.63) for women; area under the receiver operating characteristic curve values based on only family history ranged from 0.53 to 0.54 and those based only E-score or G-score ranged from 0.59 to 0.60. Although screening is recommended to begin at age 50 years for individuals with no family history of CRC, starting ages calculated based on combined E-score and G-score differed by 12 years for men and 14 for women, for individuals with the highest vs the lowest 10% of risk. CONCLUSIONS We used data from 2 large international consortia to develop CRC risk calculation models that included genetic and environmental factors along with family history. These determine risk of CRC and starting ages for screening with greater accuracy than the family history only model, which is based on the current screening guideline. These scoring systems might serve as a first step toward developing individualized CRC prevention strategies.
Collapse
Affiliation(s)
- Jihyoun Jeon
- Department of Epidemiology, University of Michigan, Ann Arbor, Michigan.
| | - Mengmeng Du
- Memorial Sloan Kettering, New York, New York
| | - Robert E Schoen
- Department of Medicine and Epidemiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Polly A Newcomb
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland
| | - Bette Caan
- Division of Research, Kaiser Permanente Medical Care Program, Oakland, California
| | - Peter T Campbell
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Andrew T Chan
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jenny Chang-Claude
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Graham G Giles
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, and Centre for Epidemiology and Biostatistics, School of Global and Population Health, University of Melbourne, Melbourne, Australia
| | - Jian Gong
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Eric J Jacobs
- Epidemiology Research Program, American Cancer Society, Atlanta, Georgia
| | - Li Li
- Case Western Reserve University, Cleveland, Ohio
| | - Yi Lin
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Stephanie A Bien
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Niha Zubair
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Robert J Macinnis
- Cancer Epidemiology and Intelligence Division, Cancer Council Victoria, and Centre for Epidemiology and Biostatistics, School of Global and Population Health, University of Melbourne, Melbourne, Australia
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Genetic Epidemiology Laboratory, Department of Pathology, The University of Melbourne, Parkville, Victoria, Australia; Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Genetic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - John L Hopper
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Victoria, Australia; Department of Epidemiology, School of Public Health and Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Yin Cao
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Reiko Nishihara
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah
| | - Duncan C Thomas
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Michael O Woods
- Memorial University of Newfoundland, St John's, Newfoundland, Canada
| | - Ross L Prentice
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
| | - Yingye Zheng
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Hermann Brenner
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center, Heidelberg, Germany; Division of Preventive Oncology, German Cancer Research Center and National Center for Tumor Diseases, Heidelberg, Germany; German Cancer Consortium, German Cancer Research Center, Heidelberg, Germany
| | - Richard B Hayes
- Division of Epidemiology, Department of Population Health, New York University School of Medicine, New York, New York
| | - Emily White
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington.
| |
Collapse
|
17
|
Augustus GJ, Ellis NA. Colorectal Cancer Disparity in African Americans: Risk Factors and Carcinogenic Mechanisms. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 188:291-303. [PMID: 29128568 DOI: 10.1016/j.ajpath.2017.07.023] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/01/2017] [Accepted: 07/20/2017] [Indexed: 12/13/2022]
Abstract
African Americans have the highest incidence and mortality rates of colorectal cancer (CRC) of any ethnic group in the United States. Although some of these disparities can be explained by differences in access to care, cancer screening, and other socioeconomic factors, disparities remain after adjustment for these factors. Consequently, an examination of recent advances in the understanding of ethnicity-specific factors, including genetic and environmental factors relating to risk of CRC, the biology of CRC progression, and the changes in screening and mortality, is important for evaluating our progress toward eliminating the disparities. An overarching limitation in this field is the number and sample size of studies performed to characterize the etiological bases of CRC incidence and mortality in African Americans. Despite this limitation, significant differences in etiology are manifest in many studies. These differences need validation, and their impacts on disparities need more detailed investigation. Perhaps most heartening, improvements in CRC screening can be attributed to the smallest difference in CRC incidence between African Americans and whites since the late 1980s. Cancer mortality, however, remains a persistent difference.
Collapse
Affiliation(s)
- Gaius J Augustus
- Cancer Biology Graduate Interdisciplinary Program, University of Arizona, Tucson, Arizona.
| | - Nathan A Ellis
- University of Arizona Cancer Center, University of Arizona, Tucson, Arizona; Department of Cellular and Molecular Medicine, University of Arizona, Tucson, Arizona.
| |
Collapse
|
18
|
Troeung L, Sodhi-Berry N, Martini A, Malacova E, Ee H, O'Leary P, Lansdorp-Vogelaar I, Preen DB. Increasing Incidence of Colorectal Cancer in Adolescents and Young Adults Aged 15-39 Years in Western Australia 1982-2007: Examination of Colonoscopy History. Front Public Health 2017; 5:179. [PMID: 28791283 PMCID: PMC5522835 DOI: 10.3389/fpubh.2017.00179] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 07/03/2017] [Indexed: 12/31/2022] Open
Abstract
Aims To examine trends in colorectal cancer (CRC) incidence and colonoscopy history in adolescents and young adults (AYAs) aged 15–39 years in Western Australia (WA) from 1982 to 2007. Design Descriptive cohort study using population-based linked hospital and cancer registry data. Method Five-year age-standardized and age-specific incidence rates of CRC were calculated for all AYAs and by sex. Temporal trends in CRC incidence were investigated using Joinpoint regression analysis. The annual percentage change (APC) in CRC incidence was calculated to identify significant time trends. Colonoscopy history relative to incident CRC diagnosis was examined and age and tumor grade at diagnosis compared for AYAs with and without pre-diagnosis colonoscopy. CRC-related mortality within 5 and 10 years of incident diagnosis were compared for AYAs with and without pre-diagnosis colonoscopy using mortality rate ratios (MRRs) derived from negative binomial regression. Results Age-standardized CRC incidence among AYAs significantly increased in WA between 1982 and 2007, APC = 3.0 (95% CI 0.7–5.5). Pre-diagnosis colonoscopy was uncommon among AYAs (6.0%, 33/483) and 71% of AYAs were diagnosed after index (first ever) colonoscopy. AYAs with pre-diagnosis colonoscopy were older at CRC diagnosis (mean 36.7 ± 0.7 years) compared to those with no prior colonoscopy (32.6 ± 0.2 years), p < 0.001. At CRC diagnosis, a significantly greater proportion of AYAs with pre-diagnosis colonoscopy had well-differentiated tumors (21.2%) compared to those without (5.6%), p = 0.001. CRC-related mortality was significantly lower for AYAs with pre-diagnosis colonoscopy compared to those without, for both 5-year [MRR = 0.44 (95% CI 0.27–0.75), p = 0.045] and 10-year morality [MRR = 0.43 (95% CI 0.24–0.83), p = 0.043]. Conclusion CRC incidence among AYAs in WA has significantly increased over the 25-year study period. Pre-diagnosis colonoscopy is associated with lower tumor grade at CRC diagnosis as well as significant reduction in both 5- and 10-year CRC-related mortality rates. These findings warrant further research into the balance in benefits and harms of targeted screening for AYA at highest risk.
Collapse
Affiliation(s)
- Lakkhina Troeung
- Centre for Health Services Research, School of Population Health, The University of Western Australia, Perth, WA, Australia
| | - Nita Sodhi-Berry
- Centre for Health Services Research, School of Population Health, The University of Western Australia, Perth, WA, Australia.,Occupational Respiratory Epidemiology, School of Population Health, The University of Western Australia, Perth, WA, Australia
| | - Angelita Martini
- Centre for Health Services Research, School of Population Health, The University of Western Australia, Perth, WA, Australia
| | - Eva Malacova
- Centre for Health Services Research, School of Population Health, The University of Western Australia, Perth, WA, Australia.,Department of Health, Safety and Environment, School of Public Health, Curtin University, Perth, WA, Australia
| | - Hooi Ee
- Department of Gastroenterology, Sir Charles Gairdner Hospital, Queen Elizabeth II Medical Centre, Nedlands, WA, Australia
| | - Peter O'Leary
- Health Policy and Management, Faculty of Health Sciences, School of Public Health, Curtin University, Perth, WA, Australia.,School of Women's and Infants' Health, The University of Western Australia, Perth, WA, Australia
| | - Iris Lansdorp-Vogelaar
- Department of Public Health, Erasmus MC University Medical Centre, Rotterdam, Netherlands
| | - David B Preen
- Centre for Health Services Research, School of Population Health, The University of Western Australia, Perth, WA, Australia
| |
Collapse
|
19
|
Etemadi A, Sinha R, Ward MH, Graubard BI, Inoue-Choi M, Dawsey SM, Abnet CC. Mortality from different causes associated with meat, heme iron, nitrates, and nitrites in the NIH-AARP Diet and Health Study: population based cohort study. BMJ 2017; 357:j1957. [PMID: 28487287 PMCID: PMC5423547 DOI: 10.1136/bmj.j1957] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Objective To determine the association of different types of meat intake and meat associated compounds with overall and cause specific mortality.Design Population based cohort study.Setting Baseline dietary data of the NIH-AARP Diet and Health Study (prospective cohort of the general population from six states and two metropolitan areas in the US) and 16 year follow-up data until 31 December 2011.Participants 536 969 AARP members aged 50-71 at baseline.Exposures Intake of total meat, processed and unprocessed red meat (beef, lamb, and pork) and white meat (poultry and fish), heme iron, and nitrate/nitrite from processed meat based on dietary questionnaire. Adjusted Cox proportional hazards regression models were used with the lowest fifth of calorie adjusted intakes as reference categories.Main outcome measure Mortality from any cause during follow-up.Results An increased risk of all cause mortality (hazard ratio for highest versus lowest fifth 1.26, 95% confidence interval 1.23 to 1.29) and death due to nine different causes associated with red meat intake was observed. Both processed and unprocessed red meat intakes were associated with all cause and cause specific mortality. Heme iron and processed meat nitrate/nitrite were independently associated with increased risk of all cause and cause specific mortality. Mediation models estimated that the increased mortality associated with processed red meat was influenced by nitrate intake (37.0-72.0%) and to a lesser degree by heme iron (20.9-24.1%). When the total meat intake was constant, the highest fifth of white meat intake was associated with a 25% reduction in risk of all cause mortality compared with the lowest intake level. Almost all causes of death showed an inverse association with white meat intake.Conclusions The results show increased risks of all cause mortality and death due to nine different causes associated with both processed and unprocessed red meat, accounted for, in part, by heme iron and nitrate/nitrite from processed meat. They also show reduced risks associated with substituting white meat, particularly unprocessed white meat.
Collapse
Affiliation(s)
- Arash Etemadi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Rashmi Sinha
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Mary H Ward
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Barry I Graubard
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Maki Inoue-Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Sanford M Dawsey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| | - Christian C Abnet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20850, USA
| |
Collapse
|
20
|
Procopciuc LM, Osian G, Iancu M. N-acetyl transferase 2/environmental factors and their association as a modulating risk factor for sporadic colon and rectal cancer. J Clin Lab Anal 2016; 31. [PMID: 27883249 DOI: 10.1002/jcla.22098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/26/2016] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVES The aim of this study was to evaluate the association between environmental factors and colon or rectal cancer after adjusting for N-acetyl transferase 2 (NAT2) phenotypes. METHODS Ninety-six patients with sporadic colon cancer, 54 with sporadic rectal cancer and 162 control subjects were genotyped for NAT2-T341C, G590A, G857A, A845C, and C481T using sequencing and PCR-RFLP analysis. RESULTS The risk for colon cancer was increased in carriers of the homozygous negative genotypes for NAT2*5C-T341C, NAT2*6B-G590A, NAT2*7B-G857A, NAT2*18-A845C, and NAT2*5A-C481T. The risk for rectal cancer was increased in carriers of the homozygous negative genotypes for NAT2*5C-T341C, NAT2*7B-G857A, and NAT2*5A-C481T. High fried red meat intake associated with NAT2-T341C, G590A, G857A, A845C, and C481T rapid acetylator allele determines a risk of 2.39 (P=.002), 2.39 (P=.002), 2.37 (P=.002), 2.28 (P=.004), and 2.51 (P=.001), respectively, for colon cancer, whereas in the case of rectal cancer, the risk increased to 7.55 (P<.001), 7.7 (P<.001), 7.83 (P<.001), 7.51 (P<.001), and 8.62 (P<.001), respectively. Alcohol consumption associated with the NAT2 -T341C, G590A, G857A, A845C, and C481T rapid acetylator allele induces a risk of 10.63 (P<.001), 12.04 (P<.001), 9.76 (P<.001), 10.25 (P<.001), and 9.54 (P<.001), respectively, for colon cancer, whereas the risk for rectal cancer is 9.72 (P<.001), 11.24 (P<.001), 13.07 (P<.001), 10.04 (P<.001), and 9.43 (P<.001), respectively. Smokers with NAT2-T341C, G590A, G857A, A845C, and C481T rapid acetylator allele have a risk of 4.87, 4.25, 4.18, 3.81, and 3.82, respectively, to develop colon cancer. CONCLUSIONS Fried red meat, alcohol, and smoking increase the risk of sporadic CRC, especially of colon cancer, in the case of rapid acetylators for the NAT2 variants.
Collapse
Affiliation(s)
- Lucia M Procopciuc
- Department of Medical Biochemistry, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Gelu Osian
- Multi Organ Transplant Center, King Fahad Specialist Hospital, Dammam, Saudi Arabia
| | - Mihaela Iancu
- Department of Medical Informatics and Biostatistics, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| |
Collapse
|
21
|
McCartney DMA, Byrne DG, Cantwell MM, Turner MJ. Cancer incidence in Ireland—the possible role of diet, nutrition and lifestyle. J Public Health (Oxf) 2016. [DOI: 10.1007/s10389-016-0769-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
22
|
Ma C, Gu L, Yang M, Zhang Z, Zeng S, Song R, Xu C, Sun Y. rs1495741 as a tag single nucleotide polymorphism of N-acetyltransferase 2 acetylator phenotype associates bladder cancer risk and interacts with smoking: A systematic review and meta-analysis. Medicine (Baltimore) 2016; 95:e4417. [PMID: 27495060 PMCID: PMC4979814 DOI: 10.1097/md.0000000000004417] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Rs1495741 has been identified to infer N-acetyltransferase 2 (NAT2) acetylator phenotype, and to decrease the risk of bladder cancer. However, a number of studies conducted in various regions showed controversial results. To quantify the association between rs1495741 and the risk of bladder cancer and to estimate the interaction effect of this genetic variant with smoking, we performed a systematic literature review and meta-analysis involving 14,815 cases and 58,282 controls from 29 studies. Our results indicates rs1495741 significantly associated with bladder cancer risk (OR = 0.85, 95% CI = 0.82-0.89, test for heterogeneity P = 0.36, I = 7.0%). And we verified this association in populations from Europe, America, and Asia. Further, our stratified meta-analysis showed rs1495741's role is typically evident only in ever smokers, which suggests its interaction with smoking. This study may provide new insight into gene-environment study on bladder cancer.
Collapse
Affiliation(s)
| | | | - Mingyuan Yang
- Department of Spine Surgery, Changhai Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | - Chuanliang Xu
- Department of Urology
- Correspondence: Chuanliang Xu, Department of Urology, Changhai Hospital No.168 Changhai Road, Shanghai, 200433 China (e-mail: )
| | | |
Collapse
|
23
|
Using national dietary intake data to evaluate and adapt the US Diet History Questionnaire: the stepwise tailoring of an FFQ for Canadian use. Public Health Nutr 2016; 19:3247-3255. [PMID: 27349130 DOI: 10.1017/s1368980016001506] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
OBJECTIVE To evaluate the Canadian Diet History Questionnaire I (C-DHQ I) food list and to adapt the US DHQ II for Canada using Canadian dietary survey data. DESIGN Twenty-four-hour dietary recalls reported by adults in a national Canadian survey were analysed to create a food list corresponding to C-DHQ I food questions. The percentage contribution of the food list to the total survey intake of seventeen nutrients was used as the criterion to evaluate the suitability of the C-DHQ I to capture food intake in Canadian populations. The data were also analysed to identify foods and to modify portion sizes for the C-DHQ II. SETTING The Canadian Community Health Survey (CCHS) - Cycle 2.2 Nutrition (2004). SUBJECTS Adults (n 20 159) who completed 24 h dietary recalls during in-person interviews. RESULTS Four thousand five hundred and thirty-three foods and recipes were grouped into 268 Food Groups, of which 212 corresponded to questions on the C-DHQ I. Nutrient intakes captured by the C-DHQ I ranged from 79 % for fat to 100 % for alcohol. For the new C-DHQ II, some food questions were retained from the original US DHQ II while others were added based on foods reported in CCHS and foods available on the Canadian market since 2004. Of 153 questions, 143 were associated with portion sizes of which fifty-three were modified from US values. Sex-specific nutrient profiles for the C-DHQ II nutrient database were derived using CCHS data. CONCLUSIONS The C-DHQ I and II are designed to optimize the capture of foods consumed by Canadian populations.
Collapse
|
24
|
Wang H, Iwasaki M, Haiman CA, Kono S, Wilkens LR, Keku TO, Berndt SI, Tsugane S, Le Marchand L. Interaction between Red Meat Intake and NAT2 Genotype in Increasing the Risk of Colorectal Cancer in Japanese and African Americans. PLoS One 2015; 10:e0144955. [PMID: 26683305 PMCID: PMC4684304 DOI: 10.1371/journal.pone.0144955] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 11/25/2015] [Indexed: 01/28/2023] Open
Abstract
Heterocyclic aromatic amines formed in cooked meat may be an underlying mechanism for the red meat-colorectal cancer (CRC) association. These compounds require bioactivaction by N-acetyltransferase 2 (NAT2). An interaction effect between red meat consumption and NAT2 in increasing CRC risk has been inconsistently reported in whites. We investigated this interaction in two populations in which the high-activity rapid NAT2 phenotype is 10- and 2-fold more common than in whites. We meta-analyzed four studies of Japanese (2,217 cases, 3,788 controls) and three studies of African Americans (527 cases, 4,527 controls). NAT2 phenotype was inferred from an optimized seven-SNP genotyping panel. Processed and total red meat intakes were associated with an increased CRC risk in Japanese and in both ethnic groups combined (P’s ≤ 0.002). We observed an interaction between processed meat intake and NAT2 in Japanese (P = 0.04), African Americans (P = 0.02), and in both groups combined (P = 0.006). The association of processed meat with CRC was strongest among individuals with the rapid NAT2 phenotype (combined analysis, OR for highest vs. lowest quartile: 1.62, 95% CI: 1.28–2.05; Ptrend = 8.0×10−5), intermediate among those with the intermediate NAT2 phenotype (1.29, 95% CI: 1.05–1.59; Ptrend = 0.05) and null among those with the slow phenotype (Ptrend = 0.45). A similar interaction was found for NAT2 and total red meat (Pinteraction = 0.03). Our findings support a role for NAT2 in modifying the association between red meat consumption and CRC in Japanese and African Americans.
Collapse
Affiliation(s)
- Hansong Wang
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Motoki Iwasaki
- Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| | - Christopher A. Haiman
- Department of Preventive Medicine, Keck School of Medicine and Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Suminori Kono
- Department of Preventive Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Lynne R. Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
| | - Temitope O. Keku
- Center for Gastrointestinal Biology and Disease, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Sonja I. Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Shoichiro Tsugane
- Research Center for Cancer Prevention and Screening, National Cancer Center, Tokyo, Japan
| | - Loïc Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, Hawaii, United States of America
- * E-mail:
| |
Collapse
|
25
|
Food Processing and the Mediterranean Diet. Nutrients 2015; 7:7925-64. [PMID: 26393643 PMCID: PMC4586566 DOI: 10.3390/nu7095371] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 09/03/2015] [Accepted: 09/09/2015] [Indexed: 02/06/2023] Open
Abstract
The benefits of the Mediterranean diet (MD) for protecting against chronic disorders such as cardiovascular disease are usually attributed to high consumption of certain food groups such as vegetables, and low consumption of other food groups such as meat. The influence of food processing techniques such as food preparation and cooking on the nutrient composition and nutritional value of these foods is not generally taken into consideration. In this narrative review, we consider the mechanistic and epidemiological evidence that food processing influences phytochemicals in selected food groups in the MD (olives, olive oil, vegetables and nuts), and that this influences the protective effects of these foods against chronic diseases associated with inflammation. We also examine how the pro-inflammatory properties of meat consumption can be modified by Mediterranean cuisine. We conclude by discussing whether food processing should be given greater consideration, both when recommending a MD to the consumer and when evaluating its health properties.
Collapse
|
26
|
Abstract
Diet and inflammation have been suggested to be important risk factors for colorectal cancer (CRC). In the present study, we examined the association between the dietary inflammatory index (DII) and the risk of CRC in a multi-centre case-control study conducted between 1992 and 1996 in Italy. The study included 1225 incident colon cancer cases, 728 incident rectal cancer cases and 4154 controls hospitalised for acute non-neoplastic diseases. The DII was computed based on dietary intake assessed using a validated seventy-eight-item FFQ that included assessment of alcohol intake. Logistic regression models were used to estimate the OR adjusted for age, sex, study centre, education, BMI, alcohol drinking, physical activity and family history of CRC. Energy intake was adjusted using the residual method. Subjects with higher DII scores (i.e. with a more pro-inflammatory diet) had a higher risk of CRC, with the DII being used both as a continuous variable (OR(continuous) 1.13, 95 % CI 1.09, 1.18) and as a categorical variable (OR(quintile 5 v. 1) 1.55, 95 % CI 1.29, 1.85; P for trend < 0.0001). Similar results were observed when the analyses were carried out separately for colon and rectal cancer cases. These results indicate that a pro-inflammatory diet is associated with an increased risk of CRC.
Collapse
|
27
|
Song M, Garrett WS, Chan AT. Nutrients, foods, and colorectal cancer prevention. Gastroenterology 2015; 148:1244-60.e16. [PMID: 25575572 PMCID: PMC4409470 DOI: 10.1053/j.gastro.2014.12.035] [Citation(s) in RCA: 435] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 11/26/2014] [Accepted: 12/01/2014] [Indexed: 02/07/2023]
Abstract
Diet has an important role in the development of colorectal cancer. In the past few decades, findings from extensive epidemiologic and experimental investigations have linked consumption of several foods and nutrients to the risk of colorectal neoplasia. Calcium, fiber, milk, and whole grains have been associated with a lower risk of colorectal cancer, and red meat and processed meat have been associated with an increased risk. There is substantial evidence for the potential chemopreventive effects of vitamin D, folate, fruits, and vegetables. Nutrients and foods also may interact, as a dietary pattern, to influence colorectal cancer risk. Diet likely influences colorectal carcinogenesis through several interacting mechanisms. These include the direct effects on immune responsiveness and inflammation, and the indirect effects of overnutrition and obesity-risk factors for colorectal cancer. Emerging evidence also implicates the gut microbiota as an important effector in the relationship between diet and cancer. Dietary modification therefore has the promise of reducing colorectal cancer incidence.
Collapse
Affiliation(s)
- Mingyang Song
- Department of Nutrition, Harvard School of Public Health, Boston, MA,Department of Epidemiology, Harvard School of Public Health, Boston, MA
| | - Wendy S. Garrett
- Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA,Department of Genetics and Complex Diseases, Harvard School of Public Health, Boston, MA,Department of Medicine, Harvard Medical School, Boston, MA,Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Andrew T. Chan
- Department of Medicine, Harvard Medical School, Boston, MA,Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Brigham and Women's Hospital, Boston, MA,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
Beyerle J, Frei E, Stiborova M, Habermann N, Ulrich CM. Biotransformation of xenobiotics in the human colon and rectum and its association with colorectal cancer. Drug Metab Rev 2015; 47:199-221. [PMID: 25686853 DOI: 10.3109/03602532.2014.996649] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In humans, the liver is generally considered to be the major organ contributing to drug metabolism, but studies during the last years have suggested an important role of the extra-hepatic drug metabolism. The gastrointestinal tract (GI-tract) is the major path of entry for a wide variety of compounds including food, and orally administered drugs, but also compounds - with neither nutrient nor other functional value - such as carcinogens. These compounds are metabolized by a large number of enzymes, including the cytochrome P450 (CYP), the glutathione S-transferase (GST) family, the uridine 5'-diphospho- glucuronosyltransferase (UDP-glucuronosyltransferase - UGT) superfamily, alcohol-metabolizing enzymes, sulfotransferases, etc. These enzymes can either inactivate carcinogens or, in some cases, generate reactive species with higher reactivity compared to the original compound. Most data in this field of research originate from animal or in vitro studies, wherein human studies are limited. Here, we review the human studies, in particular the studies on the phenotypic expression of these enzymes in the colon and rectum to get an impression of the actual enzyme levels in this primary organ of exposure. The aim of this review is to give a summary of currently available data on the relation between the CYP, the GST and the UGT biotransformation system and colorectal cancer obtained from clinical and epidemiological studies in humans.
Collapse
Affiliation(s)
- Jolantha Beyerle
- Division of Preventive Oncology, German Cancer Research Center (DKFZ) and National Center for Tumor Diseases (NCT) , Heidelberg , Germany
| | | | | | | | | |
Collapse
|