Adiposity, metabolites, and colorectal cancer risk: Mendelian randomization study

Caroline J Bull, Joshua A Bell, Neil Murphy, Eleanor Sanderson, George Davey Smith, Nicholas J Timpson, Barbara L Banbury, Demetrius Albanes, Sonja I Berndt, Stéphane Bézieau, D Timothy Bishop, Hermann Brenner, Daniel D Buchanan, Andrea Burnett-Hartman, Graham Casey, Sergi Castellví-Bel, Andrew T Chan, Jenny Chang-Claude, Amanda J Cross, Albert de la Chapelle, Jane C Figueiredo, Steven J Gallinger, Susan M Gapstur, Graham G Giles, Stephen B Gruber, Andrea Gsur, Jochen Hampe, Heather Hampel, Tabitha A Harrison, Michael Hoffmeister, Li Hsu, Wen-Yi Huang, Jeroen R Huyghe, Mark A Jenkins, Corinne E Joshu, Temitope O Keku, Tilman Kühn, Sun-Seog Kweon, Loic Le Marchand, Christopher I Li, Li Li, Annika Lindblom, Vicente Martín, Anne M May, Roger L Milne, Victor Moreno, Polly A Newcomb, Kenneth Offit, Shuji Ogino, Amanda I Phipps, Elizabeth A Platz, John D Potter, Conghui Qu, J Ramón Quirós, Gad Rennert, Elio Riboli, Lori C Sakoda, Clemens Schafmayer, Robert E Schoen, Martha L Slattery, Catherine M Tangen, Kostas K Tsilidis, Cornelia M Ulrich, Fränzel J B van Duijnhoven, Bethany van Guelpen, Kala Visvanathan, Pavel Vodicka, Ludmila Vodickova, Hansong Wang, Emily White, Alicja Wolk, Michael O Woods, Anna H Wu, Peter T Campbell, Wei Zheng, Ulrike Peters, Emma E Vincent, Marc J Gunter, Caroline J Bull, Joshua A Bell, Neil Murphy, Eleanor Sanderson, George Davey Smith, Nicholas J Timpson, Barbara L Banbury, Demetrius Albanes, Sonja I Berndt, Stéphane Bézieau, D Timothy Bishop, Hermann Brenner, Daniel D Buchanan, Andrea Burnett-Hartman, Graham Casey, Sergi Castellví-Bel, Andrew T Chan, Jenny Chang-Claude, Amanda J Cross, Albert de la Chapelle, Jane C Figueiredo, Steven J Gallinger, Susan M Gapstur, Graham G Giles, Stephen B Gruber, Andrea Gsur, Jochen Hampe, Heather Hampel, Tabitha A Harrison, Michael Hoffmeister, Li Hsu, Wen-Yi Huang, Jeroen R Huyghe, Mark A Jenkins, Corinne E Joshu, Temitope O Keku, Tilman Kühn, Sun-Seog Kweon, Loic Le Marchand, Christopher I Li, Li Li, Annika Lindblom, Vicente Martín, Anne M May, Roger L Milne, Victor Moreno, Polly A Newcomb, Kenneth Offit, Shuji Ogino, Amanda I Phipps, Elizabeth A Platz, John D Potter, Conghui Qu, J Ramón Quirós, Gad Rennert, Elio Riboli, Lori C Sakoda, Clemens Schafmayer, Robert E Schoen, Martha L Slattery, Catherine M Tangen, Kostas K Tsilidis, Cornelia M Ulrich, Fränzel J B van Duijnhoven, Bethany van Guelpen, Kala Visvanathan, Pavel Vodicka, Ludmila Vodickova, Hansong Wang, Emily White, Alicja Wolk, Michael O Woods, Anna H Wu, Peter T Campbell, Wei Zheng, Ulrike Peters, Emma E Vincent, Marc J Gunter

Abstract

Background: Higher adiposity increases the risk of colorectal cancer (CRC), but whether this relationship varies by anatomical sub-site or by sex is unclear. Further, the metabolic alterations mediating the effects of adiposity on CRC are not fully understood.

Methods: We examined sex- and site-specific associations of adiposity with CRC risk and whether adiposity-associated metabolites explain the associations of adiposity with CRC. Genetic variants from genome-wide association studies of body mass index (BMI) and waist-to-hip ratio (WHR, unadjusted for BMI; N = 806,810), and 123 metabolites from targeted nuclear magnetic resonance metabolomics (N = 24,925), were used as instruments. Sex-combined and sex-specific Mendelian randomization (MR) was conducted for BMI and WHR with CRC risk (58,221 cases and 67,694 controls in the Genetics and Epidemiology of Colorectal Cancer Consortium, Colorectal Cancer Transdisciplinary Study, and Colon Cancer Family Registry). Sex-combined MR was conducted for BMI and WHR with metabolites, for metabolites with CRC, and for BMI and WHR with CRC adjusted for metabolite classes in multivariable models.

Results: In sex-specific MR analyses, higher BMI (per 4.2 kg/m2) was associated with 1.23 (95% confidence interval (CI) = 1.08, 1.38) times higher CRC odds among men (inverse-variance-weighted (IVW) model); among women, higher BMI (per 5.2 kg/m2) was associated with 1.09 (95% CI = 0.97, 1.22) times higher CRC odds. WHR (per 0.07 higher) was more strongly associated with CRC risk among women (IVW OR = 1.25, 95% CI = 1.08, 1.43) than men (IVW OR = 1.05, 95% CI = 0.81, 1.36). BMI or WHR was associated with 104/123 metabolites at false discovery rate-corrected P ≤ 0.05; several metabolites were associated with CRC, but not in directions that were consistent with the mediation of positive adiposity-CRC relations. In multivariable MR analyses, associations of BMI and WHR with CRC were not attenuated following adjustment for representative metabolite classes, e.g., the univariable IVW OR for BMI with CRC was 1.12 (95% CI = 1.00, 1.26), and this became 1.11 (95% CI = 0.99, 1.26) when adjusting for cholesterol in low-density lipoprotein particles.

Conclusions: Our results suggest that higher BMI more greatly raises CRC risk among men, whereas higher WHR more greatly raises CRC risk among women. Adiposity was associated with numerous metabolic alterations, but none of these explained associations between adiposity and CRC. More detailed metabolomic measures are likely needed to clarify the mechanistic pathways.

Keywords: Body mass index; CCFR; CORECT; Colorectal cancer; Epidemiology; GECCO; Mendelian randomization; Metabolism; NMR; Waist-to-hip ratio.

Conflict of interest statement

None to declare.

Figures

Fig. 1
Fig. 1
Study aims and assumptions. Study aims are to (1) estimate the total effect of adiposity on CRC risk using genetic instruments for BMI and WHR ((i) unadjusted for BMI) and (2) estimate the mediated effect of adiposity on CRC risk by metabolites from targeted NMR metabolomics. Aim 2 is addressed using two approaches: (1) two-step MR wherein effects are examined of adiposity on metabolites (ii) and of adiposity-related metabolites on CRC risk (iii) and (2) multivariable MR wherein effects of adiposity on CRC (i) are examined with adjustment for the effect of representative metabolite classes on CRC (iii). Sex-specific analyses were performed when sex-specific GWAS estimates for exposure and outcome were both available. When ≥ 2 SNP instruments were available, up to 4 MR models were applied: the inverse-variance-weighted (IVW) model which assumes that none of the SNPs are pleiotropic [28], the weighted median (WM) model which allows up to half of the included SNPs to be pleiotropic and is less influenced by outliers [28], the weighted mode model which assumes that the most common effect is consistent with the true causal effect [29], and the MR-Egger model which provides an estimate of association magnitude allowing all SNPs to be pleiotropic [30]. Analyses with metabolites as outcomes were conducted within discovery aims wherein P value thresholds are applied to prioritize traits with the strongest evidence of association to be taken forward into further stages of analysis (with CRC risk). Analyses with CRC as outcomes were conducted within estimation aims wherein P values are interpreted as continuous indicators of evidence strength and focus is on effect size and precision [31, 32]
Fig. 2
Fig. 2
Associations of BMI and WHR with CRC risk based on two-sample MR. Sex-combined estimates are based on GWAS done among women and men together (for both exposure and outcome). Sex-specific estimates are based on GWAS done separately among women and men (for exposure as well as outcome)
Fig. 3
Fig. 3
Associations of BMI- or WHR-related lipid metabolites with CRC risk based on two-sample MR (IVW method). Estimates reflect the OR (95% CI) for CRC per SD higher metabolite that is associated (as an outcome) with BMI or WHR. +/− symbols indicate the direction of association of BMI or WHR with that metabolite
Fig. 4
Fig. 4
Associations of BMI- or WHR-related non-lipid metabolites with CRC risk based on two-sample MR (IVW method). Estimates reflect the OR (95% CI) for CRC per SD higher metabolite that is associated (as an outcome) with BMI or WHR. +/− symbols indicate the direction of association of BMI or WHR with that metabolite
Fig. 5
Fig. 5
Associations of BMI and WHR with CRC risk independent of various metabolite classes based on multivariable MR. Metabolite classes are based on a single representative metabolite from a previous network analysis [43], as follows: VLDL (triglycerides in small VLDL); IDL and LDL (total cholesterol in medium LDL), HDL (triglycerides in very large HDL), Omega-3 and PUFA (other polyunsaturated fatty acids than 18:2), Omega-6 (18:2, linoleic acid), MUFA and other fatty acids (Omega-9 and saturated fatty acids), glycemia (glucose), substrates (citrate), branched-chain amino acids (leucine), and other amino acids (glutamine). Adipose adjustments include the alternative adiposity trait (WHR or BMI) as a positive control

References

    1. Sung H, Siegel RL, Rosenberg PS, Jemal A. Emerging cancer trends among young adults in the USA: analysis of a population-based cancer registry. Lancet Public Health. 2019;4(3):E137-E147. 10.1016/S2468-2667(18)30267-6.
    1. Mauri G, Sartore-Bianchi A, Russo AG, Marsoni S, Bardelli A, Siena S. Early-onset colorectal cancer in young individuals. Mol Oncol. 2019;13(2):109–131. doi: 10.1002/1878-0261.12417.
    1. World Cancer Research Fund/American Institute for Cancer Research. Continuous Update Project Expert Report. Diet, nutrition, physical activity and colorectal cancer. 2018.
    1. Lauby-Secretan B, Scoccianti C, Loomis D, Grosse Y, Bianchini F, Straif K. Body fatness and cancer—viewpoint of the IARC Working Group. N Engl J Med. 2016;375(8):794–798. doi: 10.1056/NEJMsr1606602.
    1. Luo J, Hendryx M, Manson JE, Figueiredo JC, LeBlanc ES, Barrington W, et al. Intentional weight loss and obesity-related cancer risk. JNCI Cancer Spectrum. 2019;3(4):pkz054. doi: 10.1093/jncics/pkz054.
    1. Davey Smith G, Ebrahim S. ‘Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease? Int J Epidemiol. 2003;32(1):1–22. doi: 10.1093/ije/dyg070.
    1. Thrift AP, Gong J, Peters U, Chang-Claude J, Rudolph A, Slattery ML, et al. Mendelian randomization study of body mass index and colorectal cancer risk. Cancer Epidemiol Biomark Prev. 2015;24(7):1024–1031. doi: 10.1158/1055-9965.EPI-14-1309.
    1. Jarvis D, Mitchell JS, Law PJ, Palin K, Tuupanen S, Gylfe A, et al. Mendelian randomisation analysis strongly implicates adiposity with risk of developing colorectal cancer. Br J Cancer. 2016;115(2):266–272. doi: 10.1038/bjc.2016.188.
    1. Gao C, Patel CJ, Michailidou K, Peters U, Gong J, Schildkraut J, et al. Mendelian randomization study of adiposity-related traits and risk of breast, ovarian, prostate, lung and colorectal cancer. Int J Epidemiol. 2016;45(3):896–908. doi: 10.1093/ije/dyw129.
    1. Gunter MJ, Riboli E. Obesity and gastrointestinal cancers—where do we go from here? Nature Rev Gastroenterol Hepatol. 2018;15(11):651. doi: 10.1038/s41575-018-0073-y.
    1. Dombrowski SU, Knittle K, Avenell A, Araujo-Soares V, Sniehotta FF. Long term maintenance of weight loss with non-surgical interventions in obese adults: systematic review and meta-analyses of randomised controlled trials. BMJ. 2014;348:g2646. doi: 10.1136/bmj.g2646.
    1. World Cancer Research Fund/American Institute for Cancer Research. Diet, nutrition, physical activity and colorectal cancer: continuous update project. 2017.
    1. Lawlor DA. Commentary: two-sample Mendelian randomization: opportunities and challenges. Int J Epidemiol. 2016;45(3):908. doi: 10.1093/ije/dyw127.
    1. Aschard H, Vilhjálmsson BJ, Joshi AD, Price AL, Kraft P. Adjusting for heritable covariates can bias effect estimates in genome-wide association studies. Am J Hum Genet. 2015;96(2):329–339. doi: 10.1016/j.ajhg.2014.12.021.
    1. Holmes MV, Ala-Korpela M, Davey SG. Mendelian randomization in cardiometabolic disease: challenges in evaluating causality. Nat Rev Cardiol. 2017;14:577–590. doi: 10.1038/nrcardio.2017.78.
    1. Hartwig FP, Tilling K, Davey-Smith G, Lawlor DA, Borges M-CJB. Bias in two-sample Mendelian randomization by using covariable-adjusted summary associations. bioRxiv. 2019. p. 816363.
    1. Holmes MV, Davey SG. Problems in interpreting and using GWAS of conditional phenotypes illustrated by ‘alcohol GWAS’. Mol Psych. 2019;24(2):167. doi: 10.1038/s41380-018-0037-1.
    1. Rosen ED, Spiegelman BM. What we talk about when we talk about fat. Cell. 2014;156(1):20–44. doi: 10.1016/j.cell.2013.12.012.
    1. Kahn SE, Hull RL, Utzschneider KM. Mechanisms linking obesity to insulin resistance and type 2 diabetes. Nature. 2006;444(7121):840–846. doi: 10.1038/nature05482.
    1. Würtz P, Wang Q, Kangas AJ, Richmond RC, Skarp J, Tiainen M, et al. Metabolic signatures of adiposity in young adults: Mendelian randomization analysis and effects of weight change. PLoS Med. 2014;11(12):e1001765. doi: 10.1371/journal.pmed.1001765.
    1. Rodriguez-Broadbent H, Law PJ, Sud A, Palin K, Tuupanen S, Gylfe A, et al. Mendelian randomisation implicates hyperlipidaemia as a risk factor for colorectal cancer. Int J Cancer. 2017;140(12):2701–2708. doi: 10.1002/ijc.30709.
    1. Song M, Lu Y, Gunter M, Murphy N, Banbury BL, Ma W, et al. Type 2 diabetes and glycemic traits in relation to colorectal cancer risk: a Mendelian randomization study. 2018.
    1. May-Wilson S, Sud A, Law PJ, Palin K, Tuupanen S, Gylfe A, et al. Pro-inflammatory fatty acid profile and colorectal cancer risk: a Mendelian randomisation analysis. Eur J Cancer. 2017;84:228–238. doi: 10.1016/j.ejca.2017.07.034.
    1. Würtz P, Kangas AJ, Soininen P, Lawlor DA, Davey Smith G, Ala-Korpela M. Quantitative serum NMR metabolomics in large-scale epidemiology: a primer on-omic technology. Am J Epidemiol. 2017:kwx016.
    1. Kettunen J, Demirkan A, Würtz P, Draisma HH, Haller T, Rawal R, et al. Genome-wide study for circulating metabolites identifies 62 loci and reveals novel systemic effects of LPA. Nat Commun. 2016;7:11122. doi: 10.1038/ncomms11122.
    1. Huyghe JR, Bien SA, Harrison TA, Kang HM, Chen S, Schmit SL, et al. Discovery of common and rare genetic risk variants for colorectal cancer. Nature Genet. 2019;51(1):76. doi: 10.1038/s41588-018-0286-6.
    1. Davey Smith G, Hemani G. Mendelian randomization: genetic anchors for causal inference in epidemiological studies. Hum Mol Gen. 2014;23(R1):R89–R98. doi: 10.1093/hmg/ddu328.
    1. Bowden J, Davey Smith G, Haycock PC, Burgess S. Consistent estimation in Mendelian randomization with some invalid instruments using a weighted median estimator. Genet Epidemiol. 2016;40(4):304–314. doi: 10.1002/gepi.21965.
    1. Hartwig FP, Davey Smith G, Bowden J. Robust inference in summary data Mendelian randomization via the zero modal pleiotropy assumption. Int J Epidemiol. 2017;46(6):1985–1998. doi: 10.1093/ije/dyx102.
    1. Bowden J, Davey Smith G, Burgess S. Mendelian randomization with invalid instruments: effect estimation and bias detection through Egger regression. Int J Epidemiol. 2015;44(2):512–525. doi: 10.1093/ije/dyv080.
    1. Sterne JA, Davey SG. Sifting the evidence—what’s wrong with significance tests? BMJ. 2001;322(7280):226–231. doi: 10.1136/bmj.322.7280.226.
    1. Wasserstein RL, Lazar NA. The ASA’s statement on p-values: context, process, and purpose. Am Statistician. 2016;70(2):129–133. doi: 10.1080/00031305.2016.1154108.
    1. Pulit SL, Stoneman C, Morris AP, Wood AR, Glastonbury CA, Tyrrell J, et al. Metaanalysis of genome-wide association studies for body fat distribution in 694 649 individuals of European ancestry. Hum Mol Gen. 2019;28(1):166–74.
    1. Shim H, Chasman DI, Smith JD, Mora S, Ridker PM, Nickerson DA, et al. A multivariate genome-wide association analysis of 10 LDL subfractions, and their response to statin treatment, in 1868 Caucasians. PLoS One. 2015;10(4):e0120758. doi: 10.1371/journal.pone.0120758.
    1. Haycock PC, Burgess S, Wade KH, Bowden J, Relton C, Davey SG. Best (but oft-forgotten) practices: the design, analysis, and interpretation of Mendelian randomization studies. Am J Clin Nutr. 2016;103(4):965–978. doi: 10.3945/ajcn.115.118216.
    1. Hemani G, Zheng J, Elsworth B, Wade KH, Haberland V, Baird D, et al. The MR-base platform supports systematic causal inference across the human phenome. eLife. 2018;7:e34408. doi: 10.7554/eLife.34408.
    1. Burgess S, Bowden J, Fall T, Ingelsson E, Thompson SG. Sensitivity analyses for robust causal inference from Mendelian randomization analyses with multiple genetic variants. Epidemiology. 2017;28(1):30. doi: 10.1097/EDE.0000000000000559.
    1. Wald A. The fitting of straight lines if both variables are subject to error. Ann Mathematical Statistics. 1940;11(3):284–300. doi: 10.1214/aoms/1177731868.
    1. Bowden J, Hemani G, Davey Smith GJAjoe. Invited commentary: Detecting individual and global horizontal pleiotropy in Mendelian randomization—a job for the humble heterogeneity statistic? 2018;187(12):2681–5.
    1. Zheng J, Baird D, Borges M-C, Bowden J, Hemani G, Haycock P, et al. Recent developments in Mendelian randomization studies. Curr Epidemiol Rep. 2017;4(4):330–345. doi: 10.1007/s40471-017-0128-6.
    1. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J Royal Statistic Soc: Series B (Methodological) 1995;57(1):289–300.
    1. Sanderson E, Davey Smith G, Windmeijer F, Bowden J. An examination of multivariable Mendelian randomization in the single-sample and two-sample summary data settings. Int J Epidemiol. 2018;dyy262:1–15.
    1. Kujala UM, Mäkinen V-P, Heinonen I, Soininen P, Kangas AJ, Leskinen TH, et al. Long-term leisure-time physical activity and serum metabolome. Circulation. 2012:CIRCULATIONAHA. 112.105551.
    1. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic studies of body mass index yield new insights for obesity biology. Nature. 2015;518(7538):197–206. doi: 10.1038/nature14177.
    1. Shungin D, Winkler TW, Croteau-Chonka DC, Ferreira T, Locke AE, Mägi R, et al. New genetic loci link adipose and insulin biology to body fat distribution. Nature. 2015;518(7538):187–196. doi: 10.1038/nature14132.
    1. Sanderson E, Spiller W, Bowden J. Testing and correcting for weak and pleiotropic instruments in two-sample multivariable Mendelian randomisation. BioRxiv. 2020. 10.1101/2020.04.02.021980.
    1. Murphy N, Jenab M, Gunter MJ. Adiposity and gastrointestinal cancers: epidemiology, mechanisms and future directions. Nat Rev Gastroenterol Hepatol. 2018;15:659–670. doi: 10.1038/s41575-018-0038-1.
    1. Wells JC. Sexual dimorphism of body composition. Best Pract Res Clin Endocrinol Metab. 2007;21(3):415–430. doi: 10.1016/j.beem.2007.04.007.
    1. Lovejoy J, Champagne C, De Jonge L, Xie H, Smith S. Increased visceral fat and decreased energy expenditure during the menopausal transition. Int J Obes. 2008;32(6):949–958. doi: 10.1038/ijo.2008.25.
    1. Bell JA, Carslake D, O’Keeffe LM, Frysz M, Howe LD, Hamer M, et al. Associations of body mass and fat indexes with cardiometabolic traits. J Am Coll Cardiol. 2018;72(24):3142–3154. doi: 10.1016/j.jacc.2018.09.066.
    1. Flegal KM, Shepherd JA, Looker AC, Graubard BI, Borrud LG, Ogden CL, et al. Comparisons of percentage body fat, body mass index, waist circumference, and waist-stature ratio in adults. Am J Clin Nutr. 2009;89(2):500–508. doi: 10.3945/ajcn.2008.26847.
    1. Wei H-J, Zeng R, Lu J-H, Lai W-FT, Chen W-H, Liu H-Y, et al. Adipose-derived stem cells promote tumor initiation and accelerate tumor growth by interleukin-6 production. Oncotarget. 2015;6:7713–7726. doi: 10.18632/oncotarget.3481.
    1. Hotamisligil G. Inflammation and metabolic disorders. Nature. 2006;444(7121):860–867. doi: 10.1038/nature05485.
    1. Rinaldi S, Cleveland R, Norat T, Biessy C, Rohrmann S, Linseisen J, et al. Serum levels of IGF-I, IGFBP-3 and colorectal cancer risk: results from the EPIC cohort, plus a meta-analysis of prospective studies. Int J Cancer. 2010;126:NA-NA.
    1. Tran TT, Naigamwalla D, Oprescu AI, Lam L, McKeown-Eyssen G, Bruce WR, et al. Hyperinsulinemia, but not other factors associated with insulin resistance, acutely enhances colorectal epithelial proliferation in vivo. Endocrinol. 2006;147:1830–1837. doi: 10.1210/en.2005-1012.
    1. Kiunga GA, Raju J, Sabljic N, Bajaj G, Good CK, Bird RP. Elevated insulin receptor protein expression in experimentally induced colonic tumors. Cancer Lett. 2004;211:145–153. doi: 10.1016/j.canlet.2004.02.015.
    1. Kaaks R, Toniolo P, Akhmedkhanov A, Lukanova A, Biessy C, Dechaud H, et al. Serum C-peptide, insulin-like growth factor (IGF)-I, IGF-binding proteins, and colorectal cancer risk in women. J Natl Cancer Inst. 2000;92:1592–1600. doi: 10.1093/jnci/92.19.1592.
    1. Murphy N, Carreras-Torres R, Song M, Chan AT, Martin RM, Papadimitriou N, et al. Circulating levels of insulin-like growth factor 1 and insulin-like growth factor binding protein 3 associate with risk of colorectal cancer based on serologic and Mendelian randomization analyses. Gastroenterology. 2019;158(5):1300–312.e20. 10.1053/j.gastro.2019.12.020.
    1. Burgess S, Thompson SG, CRP CHD Genetics Collaboration. Avoiding bias from weak instruments in Mendelian randomization studies. International Journal of Epidemiology 2011;40(3):755–64.
    1. Gonzalez EC, Roetzheim RG, Ferrante JM, Campbell R. Predictors of proximal vs. distal colorectal cancers. Dis Colon Rectum. 2001;44:251–258. doi: 10.1007/BF02234301.
    1. Jacobs ET, Thompson PA, Martínez MaE. Diet, gender, and colorectal neoplasia. J Clin Gastroenterol 2007;41:731–746.
    1. Okubo R, Masuda H, Nemoto N. p53 mutation found to be a significant prognostic indicator in distal colorectal cancer. Oncol Rep. 2001;8(3):509-14.
    1. Pekow J, Meckel K, Dougherty U, Butun F, Mustafi R, Lim J, et al. Tumor suppressors miR-143 and miR-145 and predicted target proteins API5, ERK5, K-RAS, and IRS-1 are differentially expressed in proximal and distal colon. Am J Physiol-Gastrointestinal Liver Physiol. 2015;308:G179–GG87. doi: 10.1152/ajpgi.00208.2014.
    1. Missiaglia E, Jacobs B, D’Ario G, Di Narzo AF, Soneson C, Budinska E, et al. Distal and proximal colon cancers differ in terms of molecular, pathological, and clinical features. Ann Oncol. 2014;25:1995–2001. doi: 10.1093/annonc/mdu275.
    1. Dale KM, Coleman CI, Henyan NN, Kluger J, White CM. Statins and cancer risk: a meta-analysis. JAMA. 2006;295(1):74–80. doi: 10.1001/jama.295.1.74.
    1. Liu Y, Tang W, Wang J, Xie L, Li T, He Y, et al. Association between statin use and colorectal cancer risk: a meta-analysis of 42 studies. Cancer Causes Control. 2014;25(2):237–49.
    1. Lytras T, Nikolopoulos G, Bonovas S. Statins and the risk of colorectal cancer: An updated systematic review and meta-analysis of 40 studies. World J Gastroenterol. 2014;20(7):1858–70.
    1. Yao X, Tian Z. Dyslipidemia and colorectal cancer risk: a meta-analysis of prospective studies. Cancer Causes Control. 2015;26(2):257–268. doi: 10.1007/s10552-014-0507-y.
    1. Lee S, Zhang C, Kilicarslan M, Piening BD, Bjornson E, Hallström BM, et al. Integrated network analysis reveals an association between plasma mannose levels and insulin resistance. Cell Metab. 2016;24(1):172–184. doi: 10.1016/j.cmet.2016.05.026.
    1. Lawlor DA, Tilling K, Davey SG. Triangulation in aetiological epidemiology. Int J Epidemiol. 2016;45(6):1866–1886.
    1. Burgess S, Davies NM, Thompson SG. Bias due to participant overlap in two-sample Mendelian randomization. Genet Epidemiol. 2016;40(7):597–608. doi: 10.1002/gepi.21998.

Source: PubMed

3
Subscribe