Impact of red meat, processed meat and fibre intake on risk of late-onset chronic inflammatory diseases: prospective cohort study on lifestyle factors using the Danish 'Diet, Cancer and Health' cohort (PROCID-DCH): protocol

Nathalie Fogh Rasmussen, Katrine Hass Rubin, Maria Stougaard, Anne Tjønneland, Egon Stenager, Merete Lund Hetland, Bente Glintborg, Anette Bygum, Vibeke Andersen, Nathalie Fogh Rasmussen, Katrine Hass Rubin, Maria Stougaard, Anne Tjønneland, Egon Stenager, Merete Lund Hetland, Bente Glintborg, Anette Bygum, Vibeke Andersen

Abstract

Introduction: Chronic inflammatory diseases (CIDs) (Crohn's disease, ulcerative colitis, psoriasis, psoriatic arthritis, rheumatoid arthritis and multiple sclerosis) are diseases of the immune system that have some shared genetic and environmental predisposing factors, but still few studies have investigated the effects of lifestyle on disease risk of several CIDs. The primary aim of this prospective cohort study is to investigate the impact of fibre, red meat and processed meat on risk of late-onset CID, with the perspective that results of this study can contribute in supporting future diet recommendations for effective personalised prevention.

Methods and analysis: The study will use data from 57 053 persons from the prospective Danish cohort study 'Diet, Cancer and Health' together with National Health Registry data. The follow-up period is from December 1993 to December 2018. Questionnaire data on diet and lifestyle were collected at entry to the Diet, Cancer and Health study. The outcome CID is defined as having a diagnosis of one of the CIDs registered in the National Patient Registry or, for multiple sclerosis, in the Danish Multiple Sclerosis Registry during follow-up and being treated with a drug used for the specific disease. The major outcome of the analyses will be to detect variability in risk of late onset of any CID and, if power allows, disease risk of late onset of each CID diagnosis between persons with different fibre and red meat, and processed meat intake. The outcome will be adjusted for age, sex, body mass index, physical activity, energy, alcohol, fermented dairy products, education, smoking status, hormone replacement therapy and comorbidity.

Ethics and dissemination: The study is approved by the Danish Data Protection Agency (2012-58-0018). The core study is an open register-based cohort study. The study does not need approval from the Ethics committee or Institutional Review Board by Danish law. Study findings will be disseminated through peer-reviewed journals, patient associations and presentations at international conferences.

Trial registration number: NCT03456206; Post-results.

Keywords: chronic inflammatory diseases; diet; disease risk; fibre; lifestyle; red meat.

Conflict of interest statement

Competing interests: BG declares to have received research funding from Abbvie, Biogen, Pfizer. MLH declares to have received research funding from BMS, MSD, Pfizer, Biogen, Samsung, CellTrion, Lilly and Novartis. AB has participated in the development of educational material for Biogen. All other authors declare no conflict of interest.

© Author(s) (or their employer(s)) 2019. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.

Figures

Figure 1
Figure 1
Hypothesis for effects of diet in relation to the development of chronic inflammatory diseases (CIDs). Diet (meat, fibre, animal fat, n-3 and n-6 polyunsaturated fatty acids, vitamins A and D, carotenoids, smoking, gluten102) may affect the immune system either directly or indirectly via, for example, the activity and composition of the gut microbiome. The effect of low intake of fibre/high intake of red and processed meat is shown at left: in short, low intake of fibre (which could otherwise serve as a nutrient for the microbes) may lead to the microbial metabolism of mucus and to decrease of the intestinal mucus layer. A high intake of red and processed meat may render the mucus layer penetrable to, for example, bacteria by reducing the disulphide bonds in the mucus network. Thus, microbes may reach the epithelium and activate the immune system. There is some support for such a mechanism in CIDs, including findings of; high amounts of sulphate-reducing bacteria in patients with inflammatory bowel disease (IBD); association of high-fibre intake with low risk of IBD among 170 776 participants from the prospective Nurses’ Health Study I; and association of high intake of red meat and total protein and risk of developing inflammatory polyarthritis in the population-based prospective cohort of 25 630 participants from the European Prospective Investigation of Cancer in Norfolk. IEC, intestinal epithelial cell; MAMPs, microbe-associated molecular patterns; NFkB, nuclear factor kappa-light-chain-enhancer of activated B cells; TLRs, toll-like receptors; TNF, tumor necrosis factor. Figure from Christensen et al (copyright 2018 by Vibeke Andersen).

References

    1. Amato MP, Derfuss T, Hemmer B, et al. . Environmental modifiable risk factors for multiple sclerosis: Report from the 2016 ECTRIMS focused workshop. Mult Scler 2017:1–14. 10.1177/1352458516686847
    1. Parisi R, Symmons DP, Griffiths CE, et al. . Global epidemiology of psoriasis: a systematic review of incidence and prevalence. J Invest Dermatol 2013;133:377–85. 10.1038/jid.2012.339
    1. Baumgart DC, Carding SR. Gastroenterology 1: Inflammatory bowel disease: cause and immunobiology. The Lancet 2007;369:1627.
    1. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. The Lancet 2007;369:1641–57. 10.1016/S0140-6736(07)60751-X
    1. Strober W, Fuss I, Mannon P. The fundamental basis of inflammatory bowel disease. J Clin Invest 2007;117:514–21. 10.1172/JCI30587
    1. Taurog JD, Chhabra A, Colbert RA, et al. . Ankylosing spondylitis and axial spondyloarthritis. N Engl J Med 2016;374:2563–74. 10.1056/NEJMra1406182
    1. Löfvendahl S, Theander E, Svensson Å, et al. . Validity of diagnostic codes and prevalence of physician-diagnosed psoriasis and psoriatic arthritis in southern Sweden–a population-based register study. PLoS One 2014;9:e98024 10.1371/journal.pone.0098024
    1. Christensen R, Heitmann BL, Andersen KW, et al. . Impact of red and processed meat and fibre intake on treatment outcomes among patients with chronic inflammatory diseases: protocol for a prospective cohort study of prognostic factors and personalised medicine. BMJ Open 2018;8:e018166 10.1136/bmjopen-2017-018166
    1. Molodecky NA, Soon IS, Rabi DM, et al. . Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 2012;142:46–54. 10.1053/j.gastro.2011.10.001
    1. Leray E, Moreau T, Fromont A, et al. . Epidemiology of multiple sclerosis. Rev Neurol 2016;172:3–13. 10.1016/j.neurol.2015.10.006
    1. Browne P, Chandraratna D, Angood C, et al. . Atlas of Multiple Sclerosis 2013: A growing global problem with widespread inequity. Neurology 2014;83:1022–4. 10.1212/WNL.0000000000000768
    1. Ng SC, Shi HY, Hamidi N, et al. . Worldwide incidence and prevalence of inflammatory bowel disease in the 21st century: a systematic review of population-based studies. Lancet 2018;390:2769–78. 10.1016/S0140-6736(17)32448-0
    1. WHO. Chronic Diseases and Health Promotion. 2017.
    1. Poddubnyy D, Rudwaleit M. Efficacy and safety of adalimumab treatment in patients with rheumatoid arthritis, ankylosing spondylitis and psoriatic arthritis. Expert Opin Drug Saf 2011;10:655–73. 10.1517/14740338.2011.581661
    1. Egeberg A, Skov L, Gislason GH, et al. . Incidence and Prevalence of Psoriasis in Denmark. Acta Derm Venereol 2017;97:808–12. 10.2340/00015555-2672
    1. Egeberg A, Kristensen LE, Thyssen JP, et al. . Incidence and prevalence of psoriatic arthritis in Denmark: a nationwide register linkage study. Ann Rheum Dis 2017;76:1591–7. 10.1136/annrheumdis-2016-210963
    1. Olivieri I, Palazzi C, Peruz G, et al. . Management issues with elderly-onset rheumatoid arthritis: an update. Drugs Aging 2005;22:809–22. 10.2165/00002512-200522100-00002
    1. Taleban S, Colombel JF, Mohler MJ, et al. . Inflammatory bowel disease and the elderly: a review. J Crohns Colitis 2015;9:507–15. 10.1093/ecco-jcc/jjv059
    1. Koch-Henriksen N, Thygesen LC, Stenager E, et al. . Incidence of MS has increased markedly over six decades in Denmark particularly with late onset and in women. Neurology 2018;90:e1954–e1963. 10.1212/WNL.0000000000005612
    1. Beecham AH, Patsopoulos NA, Xifara DK, et al. . Analysis of immune-related loci identifies 48 new susceptibility variants for multiple sclerosis. Nat Genet 2013;45:1353–60. 10.1038/ng.2770
    1. Ellinghaus D, Jostins L, Spain SL, et al. . Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nat Genet 2016;48:510–8. 10.1038/ng.3528
    1. Anderson CA, Boucher G, Lees CW, et al. . Meta-analysis identifies 29 additional ulcerative colitis risk loci, increasing the number of confirmed associations to 47. Nat Genet 2011;43:246–52. 10.1038/ng.764
    1. Ellinghaus D, Zhang H, Zeissig S, et al. . Association between variants of PRDM1 and NDP52 and Crohn’s disease, based on exome sequencing and functional studies. Gastroenterology 2013;145:339–47. 10.1053/j.gastro.2013.04.040
    1. Goyette P, Boucher G, Mallon D, et al. . High-density mapping of the MHC identifies a shared role for HLA-DRB1*01:03 in inflammatory bowel diseases and heterozygous advantage in ulcerative colitis. Nat Genet 2015;47:172–9. 10.1038/ng.3176
    1. Ananthakrishnan AN, Khalili H, Higuchi LM, et al. . Higher predicted vitamin D status is associated with reduced risk of Crohn’s disease. Gastroenterology 2012;142:482–9. 10.1053/j.gastro.2011.11.040
    1. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. . Long-term intake of dietary fat and risk of ulcerative colitis and Crohn’s disease. Gut 2014;63:776–84. 10.1136/gutjnl-2013-305304
    1. Ananthakrishnan AN, Khalili H, Konijeti GG, et al. . A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology 2013;145:970–7. 10.1053/j.gastro.2013.07.050
    1. Ananthakrishnan AN, Khalili H, Song M, et al. . Zinc intake and risk of Crohn’s disease and ulcerative colitis: a prospective cohort study. Int J Epidemiol 2015;44:1995–2005. 10.1093/ije/dyv301
    1. Ananthakrishnan AN, Khalili H, Song M, et al. . High School Diet and Risk of Crohn’s Disease and Ulcerative Colitis. Inflamm Bowel Dis 2015;21:2311–9. 10.1097/MIB.0000000000000501
    1. Chan SS, Luben R, Olsen A, et al. . Association between high dietary intake of the n-3 polyunsaturated fatty acid docosahexaenoic acid and reduced risk of Crohn’s disease. Aliment Pharmacol Ther 2014;39:834–42. 10.1111/apt.12670
    1. Chan SS, Luben R, Olsen A, et al. . Body mass index and the risk for Crohn’s disease and ulcerative colitis: data from a European Prospective Cohort Study (The IBD in EPIC Study). Am J Gastroenterol 2013;108:575–582. 10.1038/ajg.2012.453
    1. Chan SS, Luben R, van Schaik F, et al. . Carbohydrate intake in the etiology of Crohn’s disease and ulcerative colitis. Inflamm Bowel Dis 2014;20:2013–21. 10.1097/MIB.0000000000000168
    1. John S, Luben R, Shrestha SS, et al. . Dietary n-3 polyunsaturated fatty acids and the aetiology of ulcerative colitis: a UK prospective cohort study. Eur J Gastroenterol Hepatol 2010;22:602–6. 10.1097/MEG.0b013e3283352d05
    1. de Silva PS, Luben R, Shrestha SS, et al. . Dietary arachidonic and oleic acid intake in ulcerative colitis etiology: a prospective cohort study using 7-day food diaries. Eur J Gastroenterol Hepatol 2014;26:11–18. 10.1097/MEG.0b013e328365c372
    1. de Silva PS, Olsen A, Christensen J, et al. . An association between dietary arachidonic acid, measured in adipose tissue, and ulcerative colitis. Gastroenterology 2010;139:1912–7. 10.1053/j.gastro.2010.07.065
    1. Hart AR, Luben R, Olsen A, et al. . Diet in the aetiology of ulcerative colitis: a European prospective cohort study. Digestion 2008;77:57–64. 10.1159/000121412
    1. Opstelten JL, Leenders M, Dik VK, et al. . Dairy Products, Dietary Calcium, and Risk of Inflammatory Bowel Disease: Results From a European Prospective Cohort Investigation. Inflamm Bowel Dis 2016;22:1403–11. 10.1097/MIB.0000000000000798
    1. Bordoni A, Danesi F, Dardevet D, et al. . Dairy products and inflammation: a review of the clinical evidence. Crit Rev Food Sci Nutr 2017;57:2497–525. 10.1080/10408398.2014.967385
    1. Lahiri M, Luben RN, Morgan C, et al. . Using lifestyle factors to identify individuals at higher risk of inflammatory polyarthritis (results from the European Prospective Investigation of Cancer-Norfolk and the Norfolk Arthritis Register–the EPIC-2-NOAR Study). Ann Rheum Dis 2014;73:219–26. 10.1136/annrheumdis-2012-202481
    1. Pattison DJ, Symmons DP, Lunt M, et al. . Dietary risk factors for the development of inflammatory polyarthritis: evidence for a role of high level of red meat consumption. Arthritis Rheum 2004;50:3804–12. 10.1002/art.20731
    1. Lauer K. Multiple sclerosis in relation to meat preservation in France and Switzerland. Neuroepidemiology 1989;8:308–15. 10.1159/000110199
    1. Lauer K. The food pattern in geographical relation to the risk of multiple sclerosis in the Mediterranean and Near East region. J Epidemiol Community Health 1991;45:251–2. 10.1136/jech.45.3.251
    1. Lauer K. Notes on the epidemiology of multiple sclerosis, with special reference to dietary habits. Int J Mol Sci 2014;15:3533–45. 10.3390/ijms15033533
    1. Lauer K. The history of nitrite in human nutrition: a contribution from German cookery books. J Clin Epidemiol 1991;44:261–4. 10.1016/0895-4356(91)90037-A
    1. Sharif K, Watad A, Bragazzi NL, et al. . Physical activity and autoimmune diseases: Get moving and manage the disease. Autoimmun Rev 2018;17:53–72. 10.1016/j.autrev.2017.11.010
    1. Jones PD, Kappelman MD, Martin CF, et al. . Exercise decreases risk of future active disease in patients with inflammatory bowel disease in remission. Inflamm Bowel Dis 2015;21:1063–71. 10.1097/MIB.0000000000000333
    1. Bilski J, Mazur-Bialy AI, Wierdak M, et al. . The impact of physical activity and nutrition on inflammatory bowel disease: the potential role of cross talk between adipose tissue and skeletal muscle. J Physiol Pharmacol 2013;64:143–55.
    1. Wolk K, Mallbris L, Larsson P, et al. . Excessive body weight and smoking associates with a high risk of onset of plaque psoriasis. Acta Derm Venereol 2009;89:492–7. 10.2340/00015555-0711
    1. Oliver JE, Silman AJ. Risk factors for the development of rheumatoid arthritis. Scand J Rheumatol 2006;35:169–74. 10.1080/03009740600718080
    1. Mahid SS, Minor KS, Soto RE, et al. . Smoking and inflammatory bowel disease: a meta-analysis. Mayo Clin Proc 2006;81:1462–71. 10.4065/81.11.1462
    1. Klareskog L, Malmström V, Lundberg K, et al. . Smoking, citrullination and genetic variability in the immunopathogenesis of rheumatoid arthritis. Semin Immunol 2011;23:92–8. 10.1016/j.smim.2011.01.014
    1. Li W, Han J, Choi HK, et al. . Smoking and risk of incident psoriasis among women and men in the United States: a combined analysis. Am J Epidemiol 2012;175:402–13. 10.1093/aje/kwr325
    1. Costenbader KH, Feskanich D, Mandl LA, et al. . Smoking intensity, duration, and cessation, and the risk of rheumatoid arthritis in women. Am J Med 2006;119:503.e1–503.e9. 10.1016/j.amjmed.2005.09.053
    1. Setty AR, Curhan G, Choi HK. Smoking and the risk of psoriasis in women: Nurses' Health Study II. Am J Med 2007;120:953–9. 10.1016/j.amjmed.2007.06.020
    1. Alfredsson L, Olsson T. Lifestyle and Environmental Factors in Multiple Sclerosis. Cold Spring Harb Perspect Med 2018:a028944 (published Online First: 2018/05/08). 10.1101/cshperspect.a028944
    1. Palacios N, Alonso A, Brønnum-Hansen H, et al. . Smoking and increased risk of multiple sclerosis: parallel trends in the sex ratio reinforce the evidence. Ann Epidemiol 2011;21:536–42. 10.1016/j.annepidem.2011.03.001
    1. Swanson GR, Sedghi S, Farhadi A, et al. . Pattern of alcohol consumption and its effect on gastrointestinal symptoms in inflammatory bowel disease. Alcohol 2010;44:223–8. 10.1016/j.alcohol.2009.10.019
    1. Khasawneh M, Spence AD, Addley J, et al. . The role of smoking and alcohol behaviour in the management of inflammatory bowel disease. Best Pract Res Clin Gastroenterol 2017;31:553–9. 10.1016/j.bpg.2017.10.004
    1. Abdollahpour I, Nedjat S, Mansournia MA, et al. . Lifestyle factors and multiple sclerosis: A population-based incident case-control study. Mult Scler Relat Disord 2018;22:128–33. 10.1016/j.msard.2018.03.022
    1. Kelly KR, Haus JM, Solomon TP, et al. . A low-glycemic index diet and exercise intervention reduces TNF(alpha) in isolated mononuclear cells of older, obese adults. J Nutr 2011;141:1089–1094. 10.3945/jn.111.139964
    1. Andersen V, Hansen AK, Heitmann BL. Potential Impact of Diet on Treatment Effect from Anti-TNF Drugs in Inflammatory Bowel Disease. Nutrients 2017;9:286 10.3390/nu9030286
    1. Vieira AT, Castelo PM, Ribeiro DA, et al. . Influence of Oral and Gut Microbiota in the Health of Menopausal Women. Front Microbiol 2017;8 10.3389/fmicb.2017.01884
    1. Tjønneland A, Olsen A, Boll K, et al. . Study design, exposure variables, and socioeconomic determinants of participation in Diet, Cancer and Health: a population-based prospective cohort study of 57,053 men and women in Denmark. Scand J Public Health 2007;35:432–41. 10.1080/14034940601047986
    1. Würtz AM, Hansen MD, Tjønneland A, et al. . Substitution of meat and fish with vegetables or potatoes and risk of myocardial infarction. Br J Nutr 2016;116:1602–10. 10.1017/S0007114516003500
    1. Sørensen M, Autrup H, Olsen A, et al. . Prospective study of NAT1 and NAT2 polymorphisms, tobacco smoking and meat consumption and risk of colorectal cancer. Cancer Lett 2008;266:186–93. 10.1016/j.canlet.2008.02.046
    1. Egeberg R, Olsen A, Christensen J, et al. . Associations between red meat and risks for colon and rectal cancer depend on the type of red meat consumed. J Nutr 2013;143:464–72. 10.3945/jn.112.168799
    1. Charlson ME, Pompei P, Ales KL, et al. . A new method of classifying prognostic comorbidity in longitudinal studies: development and validation. J Chronic Dis 1987;40:373–83. 10.1016/0021-9681(87)90171-8
    1. Quan H, Li B, Couris CM, et al. . Updating and validating the Charlson comorbidity index and score for risk adjustment in hospital discharge abstracts using data from 6 countries. Am J Epidemiol 2011;173:676–82. 10.1093/aje/kwq433
    1. Stata Statistical Software [program]. 15 version, 2017.
    1. Mason K, Thygesen LC, Stenager E, et al. . Evaluating the use and limitations of the Danish National Patient Register in register-based research using an example of multiple sclerosis. Acta Neurol Scand 2012;125:213–7. 10.1111/j.1600-0404.2011.01558.x
    1. Lophaven SN, Lynge E, Burisch J. The incidence of inflammatory bowel disease in Denmark 1980-2013: a nationwide cohort study. Aliment Pharmacol Ther 2017;45:961–72. 10.1111/apt.13971
    1. Kingwell E, Marriott JJ, Jetté N, et al. . Incidence and prevalence of multiple sclerosis in Europe: a systematic review. BMC Neurol 2013;13:128 10.1186/1471-2377-13-128
    1. Bentzen J, Flachs EM, Stenager E, et al. . Prevalence of multiple sclerosis in Denmark 1950–2005. Mult Scler 2010;16:520–5. 10.1177/1352458510364197
    1. Koch-Henriksen N, Magyari M, Laursen B. Registers of multiple sclerosis in Denmark. Acta Neurol Scand 2015;132:4–10. 10.1111/ane.12424
    1. Society TDMS. Fakta om sclerose 2013. (accessed May 2018).
    1. Kiadaliri AA, Kristensen LE, Englund M. Burden of rheumatoid arthritis in the Nordic region, 1990-2015: a comparative analysis using the Global Burden of Disease Study 2015. Scand J Rheumatol 2018;47:1–101. 10.1080/03009742.2017.1314002
    1. Pedersen JK, Svendsen AJ, Hørslev-Petersen K. Incidence of Rheumatoid Arthritis in the Southern part of Denmark from 1995 to 2001. Open Rheumatol J 2007;1:18–23. 10.2174/1874312900701010018
    1. Pedersen CB. The Danish Civil Registration System. Scand J Public Health 2011;39:22–5. 10.1177/1403494810387965
    1. Schmidt M, Schmidt SA, Sandegaard JL, et al. . The Danish National Patient Registry: a review of content, data quality, and research potential. Clin Epidemiol 2015;7:449–90. 10.2147/CLEP.S91125
    1. Pottegård A, Schmidt SAJ, Wallach-Kildemoes H, et al. . Data Resource Profile: The Danish National Prescription Registry. Int J Epidemiol 2017;46:dyw213 10.1093/ije/dyw213
    1. Helweg-Larsen K. The Danish Register of Causes of Death. Scand J Public Health 2011;39:26–9. 10.1177/1403494811399958
    1. Ibfelt EH, Sørensen J, Jensen DV, et al. . Validity and completeness of rheumatoid arthritis diagnoses in the nationwide DANBIO clinical register and the Danish National Patient Registry. Clin Epidemiol 2017;9:627–32. 10.2147/CLEP.S141438
    1. Lindhardsen J, Gislason GH, Jacobsen S, et al. . Non-steroidal anti-inflammatory drugs and risk of cardiovascular disease in patients with rheumatoid arthritis: a nationwide cohort study. Ann Rheum Dis 2014;73:1515–21. 10.1136/annrheumdis-2012-203137
    1. European prospective investigation into cancer and nutrition E. EPIC Study 2014. (accessed March 2018).
    1. Racine A, Carbonnel F, Chan SS, et al. . Dietary Patterns and Risk of Inflammatory Bowel Disease in Europe: Results from the EPIC Study. Inflamm Bowel Dis 2016;22:345–54. 10.1097/MIB.0000000000000638
    1. Togo P, Heitmann BL, Sørensen TI, et al. . Consistency of food intake factors by different dietary assessment methods and population groups. Br J Nutr 2003;90:667–78. 10.1079/BJN2003943
    1. Tjønneland A, Overvad K, Haraldsdóttir J, et al. . Validation of a semiquantitative food frequency questionnaire developed in Denmark. Int J Epidemiol 1991;20:906–12.
    1. Osler M, Heitmann BL. The validity of a short food frequency questionnaire and its ability to measure changes in food intake: a longitudinal study. Int J Epidemiol 1996;25:1023–9. 10.1093/ije/25.5.1023
    1. Køster-Rasmussen R, Siersma V, Halldorsson TI, et al. . Missing portion sizes in FFQ–alternatives to use of standard portions. Public Health Nutr 2015;18:1914–21. 10.1017/S1368980014002389
    1. Tjonneland A, Haraldsdóttir J, Overvad K, et al. . Influence of individually estimated portion size data on the validity of a semiquantitative food frequency questionnaire. Int J Epidemiol 1992;21:770–7. 10.1093/ije/21.4.770
    1. Feskanich D, Rimm EB, Giovannucci EL, et al. . Reproducibility and validity of food intake measurements from a semiquantitative food frequency questionnaire. J Am Diet Assoc 1993;93:790–6. 10.1016/0002-8223(93)91754-E
    1. Hunter DJ, Sampson L, Stampfer MJ, et al. . Variability in portion sizes of commonly consumed foods among a population of women in the United States. Am J Epidemiol 1988;127:1240–9. 10.1093/oxfordjournals.aje.a114916
    1. Andersen V, Chan S, Luben R, et al. . Fibre intake and the development of inflammatory bowel disease: A European prospective multi-centre cohort study (EPIC-IBD). J Crohns Colitis 2018;12:129–36. 10.1093/ecco-jcc/jjx136
    1. Goldbohm RA, van ’t Veer P, van den Brandt PA, et al. . Reproducibility of a food frequency questionnaire and stability of dietary habits determined from five annually repeated measurements. Eur J Clin Nutr 1995;49:420–9.
    1. Johnsen NF, Christensen J, Thomsen BL, et al. . Physical activity and risk of colon cancer in a cohort of Danish middle-aged men and women. Eur J Epidemiol 2006;21:877–84. 10.1007/s10654-006-9076-z
    1. uk biobank. Improving the health of future generations (accessed May 2018).
    1. Andersen V, Vogel U. Dietary fibres and meat in relation to Colorectal Cancer. Norske Gastroenterologisk Forening-nytt 2014;21:34–6.
    1. Devkota S, Wang Y, Musch MW, et al. . Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature 2012;487:104–8.
    1. Strober W. Vitamin A rewrites the ABCs of oral tolerance. Mucosal immunology 2008;1:92–5.
    1. Agmon-Levin N, Theodor E, Segal RM, et al. . Vitamin D in systemic and organ-specific autoimmune diseases. Clinical reviews in allergy & immunology 2013;45:256–66.
    1. Kaulmann A, Bohn T. Carotenoids, inflammation, and oxidative stress–implications of cellular signaling pathways and relation to chronic disease prevention. Nutr Res 2014;34:907–29. 10.1016/j.nutres.2014.07.010
    1. Ejsing-Duun M, Josephsen J, Aasted B, et al. . Dietary gluten reduces the number of intestinal regulatory T cells in mice. Scandinavian journal of immunology 2008;67:553–9.
    1. Campbell DJ, Koch MA. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat Rev Immunol 2011;11:119–30. 10.1038/nri2916
    1. Pedros C, Duguet F, Saoudi A, et al. . Disrupted regulatory T cell homeostasis in inflammatory bowel diseases. World J Gastroenterol 2016;22:974–95. 10.3748/wjg.v22.i3.974
    1. Yao CK, Muir JG, Gibson PR. Review article: insights into colonic protein fermentation, its modulation and potential health implications. Aliment Pharmacol Ther 2016;43:181–96. 10.1111/apt.13456
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol 2016;16:341–52. 10.1038/nri.2016.42
    1. Desai MS, Seekatz AM, Koropatkin NM, et al. . A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 2016;167:1339–53. 10.1016/j.cell.2016.10.043
    1. Png CW, Lindén SK, Gilshenan KS, et al. . Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol 2010;105:2420–8. 10.1038/ajg.2010.281
    1. Windey K, De Preter V, Louat T, et al. . Modulation of protein fermentation does not affect fecal water toxicity: a randomized cross-over study in healthy subjects. PLoS One 2012;7:e52387 10.1371/journal.pone.0052387
    1. Ijssennagger N, van der Meer R, van Mil SWC. Sulfide as a Mucus Barrier-Breaker in Inflammatory Bowel Disease? Trends Mol Med 2016;22:190–9. 10.1016/j.molmed.2016.01.002
    1. Gibson GR, Macfarlane GT, Cummings JH. Sulphate reducing bacteria and hydrogen metabolism in the human large intestine. Gut 1993;34:437–9. 10.1136/gut.34.4.437
    1. Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 2011;474:298–306. 10.1038/nature10208
    1. Cui J, Chen Y, Wang HY, et al. . Mechanisms and pathways of innate immune activation and regulation in health and cancer. Hum Vaccin Immunother 2014;10:3270–85. 10.4161/21645515.2014.979640
    1. Jia W, Whitehead RN, Griffiths L, et al. . Diversity and distribution of sulphate-reducing bacteria in human faeces from healthy subjects and patients with inflammatory bowel disease. FEMS Immunol Med Microbiol 2012;65:55–68. 10.1111/j.1574-695X.2012.00935.x
    1. Larsen L, Jensen MD, Larsen MD, et al. . The Danish National Registry for Biological Therapy in Inflammatory Bowel Disease. Clin Epidemiol 2016;8:607–12. 10.2147/CLEP.S99478

Source: PubMed

Подписаться