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

Robin Christensen, Berit L Heitmann, Karina Winther Andersen, Ole Haagen Nielsen, Signe Bek Sørensen, Mohamad Jawhara, Anette Bygum, Lone Hvid, Jakob Grauslund, Jimmi Wied, Henning Glerup, Ulrich Fredberg, Jan Alexander Villadsen, Søren Geill Kjær, Jan Fallingborg, Seyed A G R Moghadd, Torben Knudsen, Jacob Brodersen, Jesper Frøjk, Jens Frederik Dahlerup, Anders Bo Bojesen, Grith Lykke Sorensen, Steffen Thiel, Nils J Færgeman, Ivan Brandslund, Tue Bjerg Bennike, Allan Stensballe, Erik Berg Schmidt, Andre Franke, David Ellinghaus, Philip Rosenstiel, Jeroen Raes, Mette Boye, Lars Werner, Charlotte Lindgaard Nielsen, Heidi Lausten Munk, Anders Bathum Nexøe, Torkell Ellingsen, Uffe Holmskov, Jens Kjeldsen, Vibeke Andersen, Robin Christensen, Berit L Heitmann, Karina Winther Andersen, Ole Haagen Nielsen, Signe Bek Sørensen, Mohamad Jawhara, Anette Bygum, Lone Hvid, Jakob Grauslund, Jimmi Wied, Henning Glerup, Ulrich Fredberg, Jan Alexander Villadsen, Søren Geill Kjær, Jan Fallingborg, Seyed A G R Moghadd, Torben Knudsen, Jacob Brodersen, Jesper Frøjk, Jens Frederik Dahlerup, Anders Bo Bojesen, Grith Lykke Sorensen, Steffen Thiel, Nils J Færgeman, Ivan Brandslund, Tue Bjerg Bennike, Allan Stensballe, Erik Berg Schmidt, Andre Franke, David Ellinghaus, Philip Rosenstiel, Jeroen Raes, Mette Boye, Lars Werner, Charlotte Lindgaard Nielsen, Heidi Lausten Munk, Anders Bathum Nexøe, Torkell Ellingsen, Uffe Holmskov, Jens Kjeldsen, Vibeke Andersen

Abstract

Introduction: Chronic inflammatory diseases (CIDs) are frequently treated with biological medications, specifically tumour necrosis factor inhibitors (TNFi)). These medications inhibit the pro-inflammatory molecule TNF alpha, which has been strongly implicated in the aetiology of these diseases. Up to one-third of patients do not, however, respond to biologics, and lifestyle factors are assumed to affect treatment outcomes. Little is known about the effects of dietary lifestyle as a prognostic factor that may enable personalised medicine. The primary outcome of this multidisciplinary collaborative study will be to identify dietary lifestyle factors that support optimal treatment outcomes.

Methods and analysis: This prospective cohort study will enrol 320 patients with CID who are prescribed a TNFi between June 2017 and March 2019. Included among the patients with CID will be patients with inflammatory bowel disease (Crohn's disease and ulcerative colitis), rheumatic disorders (rheumatoid arthritis, axial spondyloarthritis, psoriatic arthritis), inflammatory skin diseases (psoriasis, hidradenitis suppurativa) and non-infectious uveitis. At baseline (pretreatment), patient characteristics will be assessed using patient-reported outcome measures, clinical assessments of disease activity, quality of life and lifestyle, in addition to registry data on comorbidity and concomitant medication(s). In accordance with current Danish standards, follow-up will be conducted 14-16 weeks after treatment initiation. For each disease, evaluation of successful treatment response will be based on established primary and secondary endpoints, including disease-specific core outcome sets. The major outcome of the analyses will be to detect variability in treatment effectiveness between patients with different lifestyle characteristics.

Ethics and dissemination: The principle goal of this project is to improve the quality of life of patients suffering from CID by providing evidence to support dietary and other lifestyle recommendations that may improve clinical outcomes. The study is approved by the Ethics Committee (S-20160124) and the Danish Data Protecting Agency (2008-58-035). Study findings will be disseminated through peer-reviewed journals, patient associations and presentations at international conferences.

Trial registration number: NCT03173144; Pre-results.

Keywords: biomarker and lifestyle; lifestyle and chronic inflammatory disease; patient related outcome measures; personalized medicine; treatment outcome; western style diet.

Conflict of interest statement

Competing interests: All authors declare no conflict of interest. However, the following authors declare: B. Heitmann has received funding from ‘MatPrat’, the information office for Norwegian egg and meat; L. Hvid is on the advisory board for Abbvie A/S; J. Fallingborg is on the advisory boards for AbbVie A/S, MSD Denmark, Takeda Pharma A/S, and Ferring Pharmaceuticals A/S; V. Andersen receives compensation for consultancy and for being a member of the advisory board for MSD Denmark (Merck) and Janssen A/S. The funding sponsors had no role in the design of the study; in the collection, analysis, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

© Article author(s) (or their employer(s) unless otherwise stated in the text of the article) 2018. All rights reserved. No commercial use is permitted unless otherwise expressly granted.

Figures

Figure 1
Figure 1
Hypothesis for effects of diet in relation to treatment effect. (Left) Low levels of fibre intake may promote microbial metabolism of mucus as the main energy source. This will lead to decrease of the mucus layer. Further, degradation of mucus releases free sulfate, which would then become available for use by sulfate-reducing bacteria (eg, Bilophila wadsworthia) for microbial produced hydrogen sulfide. In addition, high intake of food containing organic sulfur and sulfate additives, such as meat and processed meat, may increase the amount of sulfate for microbial produced hydrogen sulfide. The resultant hydrogen sulfide from low intake of fibre and high intake of meat may reduce the disulfide bonds in the mucus network rendering the mucus layer penetrable to, for example, bacteria. Then, MAMPs from microbes or contained in the diet may reach the epithelium and activate the pattern recognition receptors such as TLRs on the enterocytes (IEC) and next activate NFkB, type I interferon and other inflammatory pathways. This leads to production of pro-inflammatory (TNF, IL-1β, IL-6, IFN, IL-17, etc), and anti-inflammatory (primarily IL-10) cytokines and chemokines that will next activate innate lymphocytic cells and other immune cells and the immune system in general. There is some support for such a mechanism in chronic inflammatory disease, including findings of high amounts of sulfate-reducing bacteria in patients with UC; an association between the highest tertile of carbohydrate-restricted diet and RA, in a nested case–control study among 386 individuals who developed RA and 1886 matched controls from the Swedish Västerbotten Intervention Program cohort with prospectively sampled dietary survey; association of high-fibre intake with low risk of Crohn’s disease among 170 776 participants from the prospective Nurses’ Health Study I; 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. Finally, a prospective study of 191 patients with UC in remission found that high consumption of meat, particularly red and processed meat, protein and alcohol was associated with risk of relapse, and that high sulfur or sulfate intakes may offer an explanation for the observed findings. Additionally, support of the notation that diet may affect systemic immune response is provided by the finding that intake of low glycaemic index diet was found to lower secretion of TNF and IL-6 from stimulated peripheral blood mononuclear cells from obese humans. (Right) Intake of high fibre and low meat may promote an effective mucosal barrier and support the effects of outcome after drug targeting the pro-inflammatory molecule TNF (TNF inhibitors). Intake of soluble plant fibre has been found to block bacterial adhesion to gut enterocytes in animal and cell studies. The genetic architecture of the individual may also impact the influence of lifestyle factors. Hence, to provide lifestyle recommendations, we need to understand the effects of lifestyle on the immune system and how lifestyle may improve the therapeutic outcome and reduce the need of medical treatment in the individual person. Information on diet and non-diet lifestyle exposures may be collected by using, for example, questionnaires and lifestyle-associated biomarkers or a combination of these methods. Evidence-based biomarkers for lifestyle assessment are scarce and mostly used for studies on healthy individuals. IEC, intestinal epithelial cells; IL, interleukin; MAMPS, microbial-associated molecular patterns; NFkB, nuclear factor kappa B; RA, rheumatoid arthritis; TLR, Toll-like receptors; TNF, tumour necrosis factor; UC, ulcerative colitis.
Figure 2
Figure 2
Organisation and patient research partners. IBD, inflammatory bowel disease.

References

    1. Nielsen OH, Ainsworth MA. Tumor necrosis factor inhibitors for inflammatory bowel disease. N Engl J Med 2013;369:754–62. 10.1056/NEJMct1209614
    1. Baumgart DC, Carding SR. Inflammatory bowel disease: cause and immunobiology. Lancet 2007;369:1627–40. 10.1016/S0140-6736(07)60750-8
    1. Baumgart DC, Sandborn WJ. Inflammatory bowel disease: clinical aspects and established and evolving therapies. 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. 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. 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. Cross M, Smith E, Hoy D, et al. . The global burden of rheumatoid arthritis: estimates from the global burden of disease 2010 study. Ann Rheum Dis 2014;73:1316–22. 10.1136/annrheumdis-2013-204627
    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. Kaplan GG. The global burden of IBD: from 2015 to 2025. Nat Rev Gastroenterol Hepatol 2015;12:720–7. 10.1038/nrgastro.2015.150
    1. Scott DL, Wolfe F, Huizinga TW. Rheumatoid arthritis. Lancet 2010;376:1094–108. 10.1016/S0140-6736(10)60826-4
    1. Lettre G, Rioux JD. Autoimmune diseases: insights from genome-wide association studies. Hum Mol Genet 2008;17:R116–R121. 10.1093/hmg/ddn246
    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. Yadav P, Ellinghaus D, Rémy G, et al. . Genetic factors interact with tobacco smoke to modify risk for inflammatory bowel disease in humans and mice. Gastroenterology 2017;153:550–65. 10.1053/j.gastro.2017.05.010
    1. Liu JZ, van Sommeren S, Huang H, et al. . Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations. Nat Genet 2015;47:979–86. 10.1038/ng.3359
    1. Cortes A, Hadler J, Pointon JP, et al. . Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci. Nat Genet 2013;45:730–8. 10.1038/ng.2667
    1. Bowes J, Budu-Aggrey A, Huffmeier U, et al. . Dense genotyping of immune-related susceptibility loci reveals new insights into the genetics of psoriatic arthritis. Nat Commun 2015;6:6046 10.1038/ncomms7046
    1. Okada Y, Wu D, Trynka G, et al. . Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature 2014;506:376–81. 10.1038/nature12873
    1. Tsoi LC, Spain SL, Knight J, et al. . Identification of 15 new psoriasis susceptibility locihighlights the role of innate immunity.Nat Genet 2012;44:1341–8. 10.1038/ng.2467
    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;108:575–82. 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:S462 10.1097/MIB.0000000000000798
    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. Lund H, Brunnhuber K, Juhl C, et al. . Towards evidence based research. BMJ 2016;355:i5440 10.1136/bmj.i5440
    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. Heatley RV. Nutritional replenishment as an alternative to TNF-alpha in Crohn’s disease. Lancet 1997;349:1702 10.1016/S0140-6736(05)62678-5
    1. Lee D, Baldassano RN, Otley AR, et al. . Comparative effectiveness of nutritional and biological therapy in north american children with active crohn’s isease. Inflamm Bowel Dis 2015;21:1786–93. 10.1097/MIB.0000000000000426
    1. Andersen V, Holmskov U, Sørensen SB, et al. . A proposal for a study on treatment selection and lifestyle recommendations in chronic inflammatory diseases: a Danish multidisciplinary collaboration on prognostic factors and personalised medicine. Nutrients 2017;9:499 10.3390/nu9050499
    1. RADS. Rådet for Anvendelse af Dyr Sygehusmedicin. 2017. (accessed 20.2.2017).
    1. Ali FM, Johns N, Finlay AY, et al. . Comparison of the paper-based and electronic versions of the Dermatology Life Quality Index: evidence of equivalence. Br J Dermatol 2017. (published Online First: 2017/01/24). 10.1111/bjd.15314
    1. Campbell N, Ali F, Finlay AY, et al. . Equivalence of electronic and paper-based patient-reported outcome measures. Qual Life Res 2015;24:1949–61. 10.1007/s11136-015-0937-3
    1. Khanna R, Bressler B, Levesque BG, et al. . Early combined immunosuppression for the management of Crohn’s disease (REACT): a cluster randomised controlled trial. Lancet 2015;386:1825–34. 10.1016/S0140-6736(15)00068-9
    1. Vermeire S, O’Byrne S, Keir M, et al. . Etrolizumab as induction therapy for ulcerative colitis: a randomised, controlled, phase 2 trial. Lancet 2014;384:309–18. 10.1016/S0140-6736(14)60661-9
    1. Taylor PC, Keystone EC, van der Heijde D, et al. . Baricitinib versus placebo or adalimumab in rheumatoid arthritis. N Engl J Med 2017;376:652–62. 10.1056/NEJMoa1608345
    1. Baeten D, Sieper J, Braun J, et al. . Secukinumab, an interleukin-17A inhibitor, in ankylosing spondylitis. N Engl J Med 2015;373:2534–48. 10.1056/NEJMoa1505066
    1. Sieper J, Rudwaleit M, Baraliakos X, et al. . The assessment of spondyloarthritis international society (ASAS) handbook: a guide to assess spondyloarthritis. Ann Rheum Dis 2009;68(Suppl 2):ii1–ii44. 10.1136/ard.2008.104018
    1. Mease PJ, McInnes IB, Kirkham B, et al. . Secukinumab Inhibition of Interleukin-17A in Patients with Psoriatic Arthritis. N Engl J Med 2015;373:1329–39. 10.1056/NEJMoa1412679
    1. Lebwohl M, Strober B, Menter A, et al. . Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med 2015;373:1318–28. 10.1056/NEJMoa1503824
    1. Kimball AB, Okun MM, Williams DA, et al. . Two Phase 3 Trials of Adalimumab for Hidradenitis Suppurativa. N Engl J Med 2016;375:422–34. 10.1056/NEJMoa1504370
    1. Jaffe GJ, Dick AD, Brézin AP, et al. . Adalimumab in patients with active noninfectious uveitis. N Engl J Med 2016;375:932–43. 10.1056/NEJMoa1509852
    1. Peyrin-Biroulet L, Sandborn W, Sands BE, et al. . Selecting therapeutic targets in inflammatory bowel disease (stride): determining therapeutic goals for treat-to-target. Am J Gastroenterol 2015;110:1324–38. 10.1038/ajg.2015.233
    1. OPEN. 2017. (accessed 20 Febraury 2017).
    1. Drosos GI, Pozo JL. The causes and mechanisms of meniscal injuries in the sporting and non-sporting environment in an unselected population. Knee 2004;11:143–9. 10.1016/S0968-0160(03)00105-4
    1. Hemingway H, Croft P, Perel P, et al. . Prognosis research strategy (PROGRESS) 1: a framework for researching clinical outcomes. BMJ 2013;346:e5595 10.1136/bmj.e5595
    1. Altman DG, Royston P. What do we mean by validating a prognostic model? Stat Med 2000;19:453–73. 10.1002/(SICI)1097-0258(20000229)19:4<453::AID-SIM350>;2-5
    1. White IR, Horton NJ, Carpenter J, et al. . Strategy for intention to treat analysis in randomised trials with missing outcome data. BMJ 2011;342:d40 10.1136/bmj.d40
    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. European prospective investigastion into cancer and nutrition EPIC. 2014.
    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. 10.1093/ije/20.4.906
    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. Heitmann BL, Lissner L. Dietary underreporting by obese individuals: is it specific or non-specific? BMJ 1995;311:986–9. 10.1136/bmj.311.7011.986
    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. de Wit MP, Berlo SE, Aanerud GJ, et al. . European league against rheumatism recommendations for the inclusion of patient representatives in scientific projects. Ann Rheum Dis 2011;70:722–6. 10.1136/ard.2010.135129
    1. Kappelman MD, Long MD, Martin C, et al. . Evaluation of the patient-reported outcomes measurement information system in a large cohort of patients with inflammatory bowel diseases. Clin Gastroenterol Hepatol 2014;12:1315–23. 10.1016/j.cgh.2013.10.019
    1. Cheung PP, de Wit M, Bingham CO, et al. . Recommendations for the involvement of patient research partners (PRP) in OMERACT working groups. A report from the OMERACT 2014 working group on PRP. J Rheumatol 2016;43:187–93. 10.3899/jrheum.141011
    1. Steyerberg EW, Moons KG, van der Windt DA, et al. . Prognosis research strategy (PROGRESS) 3: Prognostic model research. PLoS Med 2013;10:e1001381 10.1371/journal.pmed.1001381
    1. Riley RD, Hayden JA, Steyerberg EW, et al. . Prognosis research strategy (PROGRESS) 2: Prognostic factor research. PLoS Med 2013;10:e1001380 10.1371/journal.pmed.1001380
    1. Hingorani AD, Windt DA, Riley RD, et al. . Prognosis research strategy (PROGRESS) 4: stratified medicine research. BMJ 2013;346:e5793 10.1136/bmj.e5793
    1. Osler M, Linneberg A, Glümer C, et al. . The cohorts at the Research Centre for Prevention and Health, formerly ’The Glostrup Population Studies'. Int J Epidemiol 2011;40:602–10. 10.1093/ije/dyq041
    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. 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. 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. 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. 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. Ijssennagger N, van der Meer R, van Mil SW. 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. 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. Sundström B, Johansson I, Rantapää-Dahlqvist S. Diet and alcohol as risk factors for rheumatoid arthritis: a nested case-control study. Rheumatol Int 2015;35:533–9. 10.1007/s00296-014-3185-x
    1. Jowett SL, Seal CJ, Pearce MS, et al. . Influence of dietary factors on the clinical course of ulcerative colitis: a prospective cohort study. Gut 2004;53:1479–84. 10.1136/gut.2003.024828
    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;141:1089–94. 10.3945/jn.111.139964
    1. Merga Y, Campbell BJ, Rhodes JM. Mucosal barrier, bacteria and inflammatory bowel disease: possibilities for therapy. Dig Dis 2014;32:475–83. 10.1159/000358156
    1. Hedrick VE, Dietrich AM, Estabrooks PA, et al. . Dietary biomarkers: advances, limitations and future directions. Nutr J 2012;11:109 10.1186/1475-2891-11-109
    1. Corella D, Ordovás JM. Biomarkers: background, classification and guidelines for applications in nutritional epidemiology. Nutr Hosp 2015;31(Suppl 3):177–88. 10.3305/nh.2015.31.sup3.8765
    1. Holen T, Norheim F, Gundersen TE, et al. . Biomarkers for nutrient intake with focus on alternative sampling techniques. Genes Nutr 2016;11:12 10.1186/s12263-016-0527-1
    1. Zhang Y, Schöttker B, Florath I, et al. . Smoking-associated DNA methylation biomarkers and their predictive value for all-cause and cardiovascular mortality. Environ Health Perspect 2016;124:67–74. 10.1289/ehp.1409020
    1. Garneau V, Rudkowska I, Paradis AM, et al. . Omega-3 fatty acids status in human subjects estimated using a food frequency questionnaire and plasma phospholipids levels. Nutr J 2012;11:46 10.1186/1475-2891-11-46
    1. Warensjö Lemming E, Nälsén C, Becker W, et al. . Relative validation of the dietary intake of fatty acids among adults in the swedish national dietary survey using plasma phospholipid fatty acid composition. J Nutr Sci 2015;4:e25 10.1017/jns.2015.1
    1. Wallingford SC, Pilkington SM, Massey KA, et al. . Three-way assessment of long-chain n-3 PUFA nutrition: by questionnaire and matched blood and skin samples. Br J Nutr 2013;109:701–8. 10.1017/S0007114512001997
    1. Dragsted LO. Biomarkers of meat intake and the application of nutrigenomics. Meat Sci 2010;84:301–7. 10.1016/j.meatsci.2009.08.028
    1. Risérus U, Marklund M. Milk fat biomarkers and cardiometabolic disease. Curr Opin Lipidol 2017;28:46–51. 10.1097/MOL.0000000000000381
    1. Johnsen NF, Hausner H, Olsen A, et al. . Intake of whole grains and vegetables determines the plasma enterolactone concentration of Danish women. J Nutr 2004;134:2691–7.
    1. Nørskov NP, Kyrø C, Olsen A, et al. . High-throughput lc-ms/ms method for direct quantification of glucuronidated, sulfated, and free enterolactone in human plasma. J Proteome Res 2016;15:1051–8. 10.1021/acs.jproteome.5b01117
    1. Moreno ML, Cebolla Á, Muñoz-Suano A, et al. . Detection of gluten immunogenic peptides in the urine of patients with coeliac disease reveals transgressions in the gluten-free diet and incomplete mucosal healing. Gut 2017;66:250–7. 10.1136/gutjnl-2015-310148
    1. Zamora-Ros R, Touillaud M, Rothwell JA, et al. . Measuring exposure to the polyphenol metabolome in observational epidemiologic studies: current tools and applications and their limits. Am J Clin Nutr 2014;100:11–26. 10.3945/ajcn.113.077743
    1. Krogholm KS, Bredsdorff L, Alinia S, et al. . Free fruit at workplace intervention increases total fruit intake: a validation study using 24 h dietary recall and urinary flavonoid excretion. Eur J Clin Nutr 2010;64:1222–8. 10.1038/ejcn.2010.130
    1. Linseisen J, Rohrmann S. Biomarkers of dietary intake of flavonoids and phenolic acids for studying diet-cancer relationship in humans. Eur J Nutr 2008;47(Suppl 2):60–8. 10.1007/s00394-008-2007-x
    1. Brantsaeter AL, Haugen M, Rasmussen SE, et al. . Urine flavonoids and plasma carotenoids in the validation of fruit, vegetable and tea intake during pregnancy in the Norwegian Mother and Child Cohort Study (MoBa). Public Health Nutr 2007;10:838–47. 10.1017/S1368980007339037
    1. Neale EP, Batterham MJ, Tapsell LC. Consumption of a healthy dietary pattern results in significant reductions in C-reactive protein levels in adults: a meta-analysis. Nutr Res 2016;36:391–401. 10.1016/j.nutres.2016.02.009
    1. Stedile N, Canuto R, Col CD, et al. . Dietary total antioxidant capacity is associated with plasmatic antioxidant capacity, nutrient intake and lipid and DNA damage in healthy women. Int J Food Sci Nutr 2016;67:479–88. 10.3109/09637486.2016.1164670
    1. Shah RV, Murthy VL, Allison MA, et al. . Diet and adipose tissue distributions: the multi-ethnic study of atherosclerosis. Nutr Metab Cardiovasc Dis 2016;26:185–93. 10.1016/j.numecd.2015.12.012
    1. Sotos-Prieto M, Bhupathiraju SN, Falcon LM, et al. . Association between a healthy lifestyle score and inflammatory markers among puerto rican adults. Nutr Metab Cardiovasc Dis 2016;26:178–84. 10.1016/j.numecd.2015.12.004
    1. Ko BJ, Park KH, Shin S, et al. . Diet quality and diet patterns in relation to circulating cardiometabolic biomarkers. Clin Nutr 2016;35:484–90. 10.1016/j.clnu.2015.03.022
    1. Marques-Rocha JL, Milagro FI, Mansego ML, et al. . LINE-1 methylation is positively associated with healthier lifestyle but inversely related to body fat mass in healthy young individuals. Epigenetics 2016;11:49–60. 10.1080/15592294.2015.1135286
    1. Umoh FI, Kato I, Ren J, et al. . Markers of systemic exposures to products of intestinal bacteria in a dietary intervention study. Eur J Nutr 2016;55:793–8. 10.1007/s00394-015-0900-7
    1. Loprinzi PD, Lee IM, Andersen RE, et al. . Association of concurrent healthy eating and regular physical activity with cardiovascular disease risk factors in u.S. Youth. Am J Health Promot 2015;30:2–8. 10.4278/ajhp.140213-QUAN-71
    1. Yu E, Rimm E, Qi L, et al. . Diet, lifestyle, biomarkers, genetic factors, and risk of cardiovascular disease in the nurses' health studies. Am J Public Health 2016;106:1616–23. 10.2105/AJPH.2016.303316
    1. Satija A, Bhupathiraju SN, Rimm EB, et al. . Plant-based dietary patterns and incidence of type 2 diabetes in US men and women: results from three prospective cohort studies. PLoS Med 2016;13:e1002039 10.1371/journal.pmed.1002039
    1. Ardisson Korat AV, Willett WC, Hu FB. Diet, lifestyle, and genetic risk factors for type 2 diabetes: a review from the Nurses' Health Study, Nurses' Health Study 2, and Health Professionals' Follow-up Study. Curr Nutr Rep 2014;3:345–54. 10.1007/s13668-014-0103-5
    1. Nettleton JA, Steffen LM, Palmas W, et al. . Associations between microalbuminuria and animal foods, plant foods, and dietary patterns in the multiethnic study of atherosclerosis. Am J Clin Nutr 2008;87:1825–36.
    1. Armstrong AW, Harskamp CT, Dhillon JS, et al. . Psoriasis and smoking: a systematic review and meta-analysis. Br J Dermatol 2014;170:304–14. 10.1111/bjd.12670
    1. Ibfelt EH, Jensen DV, Hetland ML. The Danish nationwide clinical register for patients with rheumatoid arthritis: DANBIO. Clin Epidemiol 2016;8:737–42. 10.2147/CLEP.S99490
    1. Gniadecki R, Kragballe K, Dam TN, et al. . Comparison of drug survival rates for adalimumab, etanercept and infliximab in patients with psoriasis vulgaris. Br J Dermatol 2011;164:1091–6. 10.1111/j.1365-2133.2011.10213.x
    1. Bek S, Nielsen JV, Bojesen AB, et al. . Systematic review: genetic biomarkers associated with anti-TNF treatment response in inflammatory bowel diseases. Aliment Pharmacol Ther 2016;44:554–67. 10.1111/apt.13736
    1. Moore HM, Kelly AB, Jewell SD, et al. . Biospecimen reporting for improved study quality (BRISQ). JProteomeRes 2011;10:3429–38.
    1. Betsou F, Lehmann S, Ashton G, et al. . Standard preanalytical coding for biospecimens: defining the sample PREanalytical code. Cancer Epidemiol Biomarkers Prev 2010;19:1004–11. 10.1158/1055-9965.EPI-09-1268
    1. Armyra K, Kouris A, Markantoni V, et al. . Hidradenitis suppurativa treated with tetracycline in combination with colchicine: a prospective series of 20 patients. Int J Dermatol 2017;56:346–50. 10.1111/ijd.13428
    1. Skoie IM, Dalen I, Ternowitz T, et al. . Fatigue in psoriasis: a controlled study. Br J Dermatol 2017;177:505–12. 10.1111/bjd.15375
    1. Stjernman H, Grännö C, Järnerot G, et al. . Short health scale: a valid, reliable, and responsive instrument for subjective health assessment in Crohn’s disease. Inflamm Bowel Dis 2008;14:47–52. 10.1002/ibd.20255
    1. Hjortswang H, Järnerot G, Curman B, et al. . The Short Health Scale: a valid measure of subjective health in ulcerative colitis. Scand J Gastroenterol 2006;41:1196–203. 10.1080/00365520600610618

Source: PubMed

3
구독하다