Gut Microbiome: Profound Implications for Diet and Disease

Nutrients - Tập 11 Số 7 - Trang 1613
Ronald D. Hills1, Benjamin Pontefract2,3, Hillary R. Mishcon1, Cody A. Black4,1, Steven C. Sutton1, Cory R. Theberge1
1Department of Pharmaceutical Sciences, College of Pharmacy, University of New England, Portland, ME 04103, USA
2College of Pharmacy, Ferris State University, Big Rapids, MI 49307, USA
3Pharmacy Service, Boise Veterans Affairs Medical Center, Boise, ID 83702, USA
4College of Pharmacy, University of Texas at Austin, San Antonio, TX 78229, USA

Tóm tắt

The gut microbiome plays an important role in human health and influences the development of chronic diseases ranging from metabolic disease to gastrointestinal disorders and colorectal cancer. Of increasing prevalence in Western societies, these conditions carry a high burden of care. Dietary patterns and environmental factors have a profound effect on shaping gut microbiota in real time. Diverse populations of intestinal bacteria mediate their beneficial effects through the fermentation of dietary fiber to produce short-chain fatty acids, endogenous signals with important roles in lipid homeostasis and reducing inflammation. Recent progress shows that an individual’s starting microbial profile is a key determinant in predicting their response to intervention with live probiotics. The gut microbiota is complex and challenging to characterize. Enterotypes have been proposed using metrics such as alpha species diversity, the ratio of Firmicutes to Bacteroidetes phyla, and the relative abundance of beneficial genera (e.g., Bifidobacterium, Akkermansia) versus facultative anaerobes (E. coli), pro-inflammatory Ruminococcus, or nonbacterial microbes. Microbiota composition and relative populations of bacterial species are linked to physiologic health along different axes. We review the role of diet quality, carbohydrate intake, fermentable FODMAPs, and prebiotic fiber in maintaining healthy gut flora. The implications are discussed for various conditions including obesity, diabetes, irritable bowel syndrome, inflammatory bowel disease, depression, and cardiovascular disease.

Từ khóa


Tài liệu tham khảo

Conlon, 2015, The impact of diet and lifestyle on gut microbiota and human health, Nutrients, 7, 17, 10.3390/nu7010017

Singh, 2017, Influence of diet on the gut microbiome and implications for human health, J. Transl. Med., 15, 73, 10.1186/s12967-017-1175-y

Brennan, 2016, Gut microbiota, inflammation, and colorectal cancer, Annu. Rev. Microbiol., 70, 395, 10.1146/annurev-micro-102215-095513

Valdes, 2018, Role of the gut microbiota in nutrition and health, BMJ, 361, k2179, 10.1136/bmj.k2179

Qin, 2010, A human gut microbial gene catalogue established by metagenomic sequencing, Nature, 464, 59, 10.1038/nature08821

Almeida, 2019, A new genomic blueprint of the human gut microbiota, Nature, 568, 499, 10.1038/s41586-019-0965-1

Rothschild, 2018, Environment dominates over host genetics in shaping human gut microbiota, Nature, 555, 210, 10.1038/nature25973

Noble, 2001, Waist-to-hip ratio versus BMI as predictors of cardiac risk in obese adult women, West. J. Med., 174, 240, 10.1136/ewjm.174.4.240-a

Hamer, 2017, Normal-Weight central obesity and risk for mortality, Ann. Intern. Med., 166, 917, 10.7326/L17-0022

Gentile, 2018, The gut microbiota at the intersection of diet and human health, Science, 362, 776, 10.1126/science.aau5812

Lauderdale, 2000, Body mass index in a US national sample of Asian Americans: Effects of nativity, years since immigration and socioeconomic status, Int. J. Obes., 24, 1188, 10.1038/sj.ijo.0801365

Vangay, 2018, US immigration westernizes the human gut microbiome, Cell, 175, 962, 10.1016/j.cell.2018.10.029

Arumugam, 2011, Enterotypes of the human gut microbiome, Nature, 473, 174, 10.1038/nature09944

Wu, 2011, Linking long-term dietary patterns with gut microbial enterotypes, Science, 334, 105, 10.1126/science.1208344

Jonkers, 2015, Intestinal microbiota and diet in IBS: Causes, consequences, or epiphenomena?, Am. J. Gastroenterol., 110, 278, 10.1038/ajg.2014.427

Hall, 2017, A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients, Genome Med., 9, 103, 10.1186/s13073-017-0490-5

Henke, 2019, Ruminococcus gnavus, a member of the human gut microbiome associated with Crohn’s disease, produces an inflammatory polysaccharide, Proc. Natl. Acad. Sci. USA, 116, 12672, 10.1073/pnas.1904099116

Kim, 2014, Colorectal cancer in inflammatory bowel disease: The risk, pathogenesis, prevention and diagnosis, World J. Gastroenterol., 20, 9872, 10.3748/wjg.v20.i29.9872

Jandhyala, 2015, Role of the normal gut microbiota, World J. Gastroenterol., 21, 8787, 10.3748/wjg.v21.i29.8787

Winter, 2014, Dysbiosis in the inflamed intestine: Chance favors the prepared microbe, Gut Microbes, 5, 71, 10.4161/gmic.27129

2014, Escherichia coli in chronic inflammatory bowel diseases: An update on adherent invasive Escherichia coli pathogenicity, World J. Gastrointest. Pathophysiol., 5, 213, 10.4291/wjgp.v5.i3.213

Lewis, 2015, Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn’s disease, Cell Host Microbe, 18, 489, 10.1016/j.chom.2015.09.008

Margolis, 2010, Potential association between the oral tetracycline class of antimicrobials used to treat acne and inflammatory bowel disease, Am. J. Gastroenterol., 105, 2610, 10.1038/ajg.2010.303

Lloyd, 2004, Artificial nutrition: Principles and practice of enteral feeding, Clin. Colon Rectal Surg., 17, 107, 10.1055/s-2004-828657

Llewellyn, 2018, Interactions between diet and the intestinal microbiota alter intestinal permeability and colitis severity in mice, Gastroenterology, 154, 1037, 10.1053/j.gastro.2017.11.030

Chiba, 2010, Lifestyle-related disease in Crohn’s disease: Relapse prevention by a semi-vegetarian diet, World J. Gastroenterol., 16, 2484, 10.3748/wjg.v16.i20.2484

Lewis, 2017, Diet as a trigger or therapy for inflammatory bowel diseases, Gastroenterology, 152, 398, 10.1053/j.gastro.2016.10.019

Roy, 2006, Short-chain fatty acids: Ready for prime time?, Nutr. Clin. Pract., 21, 351, 10.1177/0115426506021004351

Sims, 2014, In vitro fermentation of prebiotic oligosaccharides by Bifidobacterium lactis HN019 and Lactobacillus spp., Anaerobe, 25, 11, 10.1016/j.anaerobe.2013.11.001

Andoh, 2016, Physiological role of gut microbiota for maintaining human health, Digestion, 93, 176, 10.1159/000444066

Ohira, 2017, Are short chain fatty acids in gut microbiota defensive players for inflammation and atherosclerosis?, J. Atheroscler. Thromb., 24, 660, 10.5551/jat.RV17006

Sivaprakasam, 2016, Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis, Pharmacol. Ther., 164, 144, 10.1016/j.pharmthera.2016.04.007

Morrison, 2016, Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism, Gut Microbes, 7, 189, 10.1080/19490976.2015.1134082

Bergman, 1990, Energy contributions of volatile fatty acids from the gastrointestinal tract in various species, Physiol. Rev., 70, 567, 10.1152/physrev.1990.70.2.567

Kim, 2014, Perspectives on the therapeutic potential of short-chain fatty acid receptors, BMB Rep., 47, 173, 10.5483/BMBRep.2014.47.3.272

Singh, 2014, Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis, Immunity, 40, 128, 10.1016/j.immuni.2013.12.007

Ang, 2016, GPR41 and GPR43 in obesity and inflammation—Protective or causative?, Front. Immunol., 7, 28, 10.3389/fimmu.2016.00028

Litvak, 2018, Colonocyte metabolism shapes the gut microbiota, Science, 362, eaat9076, 10.1126/science.aat9076

Haslam, 2018, Antibiotic exposure and reduced short chain fatty acid production after hematopoietic stem cell transplant, Biol. Blood Marrow Transpl., 24, 2418, 10.1016/j.bbmt.2018.07.030

Wassenaar, 2016, Insights from 100 years of research with probiotic E. coli, Eur. J. Microbiol. Immunol., 6, 147, 10.1556/1886.2016.00029

Byndloss, 2017, Microbiota-activated PPAR-gamma signaling inhibits dysbiotic Enterobacteriaceae expansion, Science, 357, 570, 10.1126/science.aam9949

Winter, 2013, Host-derived nitrate boosts growth of E. coli in the inflamed gut, Science, 339, 708, 10.1126/science.1232467

Xu, 2018, 5-Aminosalicylic acid alters the gut bacterial microbiota in patients with ulcerative colitis, Front. Microbiol., 9, 1274, 10.3389/fmicb.2018.01274

Pang, 2014, Fecal biomarkers of intestinal health and disease in children, Front. Pediatr., 2, 6, 10.3389/fped.2014.00006

Abdulamir, 2010, The impact of the level of the intestinal short chain fatty acids in inflammatory bowel disease patients versus healthy subjects, Open Biochem. J., 4, 53, 10.2174/1874091X01004010053

Sanchez, 2019, Faecal bacterial and short-chain fatty acids signature in hypercholesterolemia, Sci. Rep., 9, 1772, 10.1038/s41598-019-38874-3

Imperiale, 2019, Performance characteristics of fecal immunochemical tests for colorectal cancer and advanced adenomatous polyps: A systematic review and meta-analysis, Ann. Intern. Med., 170, 319, 10.7326/M18-2390

Lazaridis, 2018, Current insights into the innate immune system dysfunction in irritable bowel syndrome, Ann. Gastroenterol., 31, 171

Yao, 2012, Subtypes of irritable bowel syndrome on Rome III criteria: A multicenter study, J. Gastroenterol. Hepatol., 27, 760, 10.1111/j.1440-1746.2011.06930.x

Lacy, 2016, Bowel Disorders, Gastroenterology, 150, 1393, 10.1053/j.gastro.2016.02.031

Hughes, 2013, Immune activation in irritable bowel syndrome: Can neuroimmune interactions explain symptoms?, Am. J. Gastroenterol., 108, 1066, 10.1038/ajg.2013.120

Langhorst, 2009, Elevated human beta-defensin-2 levels indicate an activation of the innate immune system in patients with irritable bowel syndrome, Am. J. Gastroenterol., 104, 404, 10.1038/ajg.2008.86

Farup, P.G., Rudi, K., and Hestad, K. (2016). Faecal short-chain fatty acids - a diagnostic biomarker for irritable bowel syndrome?. BMC Gastroenterol., 16.

Linsalata, M., Riezzo, G., D’Attoma, B., Clemente, C., Orlando, A., and Russo, F. (2018). Noninvasive biomarkers of gut barrier function identify two subtypes of patients suffering from diarrhoea predominant-IBS: A case-control study. BMC Gastroenterol., 18.

Mu, 2017, Leaky gut as a danger signal for autoimmune diseases, Front. Immunol., 8, 598, 10.3389/fimmu.2017.00598

Briskey, 2016, Probiotics modify tight-junction proteins in an animal model of nonalcoholic fatty liver disease, Ther. Adv. Gastroenterol., 9, 463, 10.1177/1756283X16645055

Wang, 2016, Intestinal REG3 lectins protect against alcoholic steatohepatitis by reducing mucosa-associated microbiota and preventing bacterial translocation, Cell Host Microbe, 19, 227, 10.1016/j.chom.2016.01.003

Carabotti, 2015, The gut-brain axis: Interactions between enteric microbiota, central and enteric nervous systems, Ann. Gastroenterol., 28, 203

Quigley, E.M.M. (2018). The gut-brain axis and the microbiome: Clues to pathophysiology and opportunities for novel management strategies in irritable bowel syndrome (IBS). J. Clin. Med., 7.

Strati, 2017, New evidences on the altered gut microbiota in autism spectrum disorders, Microbiome, 5, 24, 10.1186/s40168-017-0242-1

Fowlie, G., Cohen, N., and Ming, X. (2018). The perturbance of microbiome and gut-brain axis in autism spectrum disorders. Int. J. Mol. Sci., 19.

Francavilla, 2015, Autism spectrum disorders and intestinal microbiota, Gut Microbes, 6, 207, 10.1080/19490976.2015.1035855

Sinaiscalco, 2018, Autism and neuro-immune-gut link, AIMS Mol. Sci., 5, 166, 10.3934/molsci.2018.2.166

Wheless, 2008, History of the ketogenic diet, Epilepsia, 49, 3, 10.1111/j.1528-1167.2008.01821.x

Reddel, S., Putignani, L., and Del Chierico, F. (2019). The impact of low-FODMAPs, gluten-free, and ketogenic diets on gut microbiota modulation in pathological conditions. Nutrients, 11.

Stafstrom, 2012, The ketogenic diet as a treatment paradigm for diverse neurological disorders, Front. Pharmacol., 3, 59, 10.3389/fphar.2012.00059

Olson, 2018, The gut microbiota mediates the anti-seizure effects of the ketogenic diet, Cell, 173, 1728, 10.1016/j.cell.2018.04.027

Wong, 2003, GABA, gamma-hydroxybutyric acid, and neurological disease, Ann. Neurol., 54, S3, 10.1002/ana.10696

Doenyas, 2018, Dietary interventions for autism spectrum disorder: New perspectives from the gut-brain axis, Physiol. Behav., 194, 577, 10.1016/j.physbeh.2018.07.014

Sampson, 2016, Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson’s disease, Cell, 167, 1469, 10.1016/j.cell.2016.11.018

Sarkar, 2016, Psychobiotics and the manipulation of bacteria-gut-brain signals, Trends Neurosci., 39, 763, 10.1016/j.tins.2016.09.002

Skonieczna-Zydecka, K., Marlicz, W., Misera, A., Koulaouzidis, A., and Loniewski, I. (2018). Microbiome-the missing link in the gut-brain axis: Focus on its role in gastrointestinal and mental health. J. Clin. Med., 7.

Cammarota, 2017, European consensus conference on faecal microbiota transplantation in clinical practice, Gut, 66, 569, 10.1136/gutjnl-2016-313017

Kang, 2017, Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: An open-label study, Microbiome, 5, 10, 10.1186/s40168-016-0225-7

Choi, 2016, Fecal microbiota transplantation: Current applications, effectiveness, and future perspectives, Clin. Endosc., 49, 257, 10.5946/ce.2015.117

Frissen, 2017, Fecal microbiota transplantation in metabolic syndrome: History, present and future, Gut Microbes, 8, 253, 10.1080/19490976.2017.1293224

Kootte, 2017, Improvement of insulin sensitivity after lean donor feces in metabolic syndrome Is driven by baseline intestinal microbiota composition, Cell Metab., 26, 611, 10.1016/j.cmet.2017.09.008

Foster, 2013, Gut-brain axis: How the microbiome influences anxiety and depression, Trends Neurosci., 36, 305, 10.1016/j.tins.2013.01.005

Falony, 2019, The neuroactive potential of the human gut microbiota in quality of life and depression, Nat. Microbiol., 4, 623, 10.1038/s41564-018-0337-x

Wallace, 2017, The effects of probiotics on depressive symptoms in humans: A systematic review, Ann. Gen. Psychiatry, 16, 14, 10.1186/s12991-017-0138-2

Boehme, M., Van De Wouw, M., Bastiaanssen, T.F.S., Olavarria-Ramirez, L., Lyons, K., Fouhy, F., Golubeva, A.V., Moloney, G.M., Minuto, C., and Sandhu, K.V. (2019). Mid-life microbiota crises: Middle age is associated with pervasive neuroimmune alterations that are reversed by targeting the gut microbiome. Mol. Psychiatry, in press.

Banta, 2019, Mental health status and dietary intake among California adults: A population-based survey, Int. J. Food Sci. Nutr., 70, 759, 10.1080/09637486.2019.1570085

Sonnenburg, 2016, Diet-microbiota interactions as moderators of human metabolism, Nature, 535, 56, 10.1038/nature18846

Ridaura, 2013, Gut microbiota from twins discordant for obesity modulate metabolism in mice, Science, 341, 1241214, 10.1126/science.1241214

Turnbaugh, 2006, An obesity-associated gut microbiome with increased capacity for energy harvest, Nature, 444, 1027, 10.1038/nature05414

Turnbaugh, 2009, A core gut microbiome in obese and lean twins, Nature, 457, 480, 10.1038/nature07540

Nielsen, 2013, Richness of human gut microbiome correlates with metabolic markers, Nature, 500, 541, 10.1038/nature12506

Cotillard, 2013, Dietary intervention impact on gut microbial gene richness, Nature, 500, 585, 10.1038/nature12480

Everard, 2013, Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity, Proc. Natl. Acad. Sci. USA, 110, 9066, 10.1073/pnas.1219451110

Anhe, 2015, A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice, Gut, 64, 872, 10.1136/gutjnl-2014-307142

Dao, 2016, Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology, Gut, 65, 426, 10.1136/gutjnl-2014-308778

Depommier, 2019, Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study, Nat. Med., 25, 1096, 10.1038/s41591-019-0495-2

Zhai, Q., Feng, S., Arjan, N., and Chen, W. (2018). A next generation probiotic, Akkermansia muciniphila. Crit. Rev. Food Sci. Nutr., in press.

Liou, 2013, Conserved shifts in the gut microbiota due to gastric bypass reduce host weight and adiposity, Sci. Transl. Med., 5, 178ra41, 10.1126/scitranslmed.3005687

Kasubuchi, 2015, Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation, Nutrients, 7, 2839, 10.3390/nu7042839

Zhang, 2009, Human gut microbiota in obesity and after gastric bypass, Proc. Natl. Acad. Sci. USA, 106, 2365, 10.1073/pnas.0812600106

Furet, 2010, Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: Links with metabolic and low-grade inflammation markers, Diabetes, 59, 3049, 10.2337/db10-0253

Jiao, A.R., Diao, H., Yu, B., He, J., Yu, J., Zheng, P., Huang, Z.Q., Luo, Y.H., Luo, J.Q., and Mao, X.B. (2018). Oral administration of short chain fatty acids could attenuate fat deposition of pigs. PLoS ONE, 13.

Canfora, 2017, Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: A randomized crossover trial, Sci. Rep., 7, 2360, 10.1038/s41598-017-02546-x

Chambers, 2018, Role of gut microbiota-generated short-chain fatty acids in metabolic and cardiovascular health, Curr. Nutr. Rep., 7, 198, 10.1007/s13668-018-0248-8

Fernandes, 2014, Adiposity, gut microbiota and faecal short chain fatty acids are linked in adult humans, Nutr. Diabetes, 4, e121, 10.1038/nutd.2014.23

Menni, 2017, Gut microbiome diversity and high-fibre intake are related to lower long-term weight gain, Int. J. Obes., 41, 1099, 10.1038/ijo.2017.66

Li, 2018, Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit, Gut, 67, 1269, 10.1136/gutjnl-2017-314050

Roshanravan, 2017, Effect of butyrate and inulin supplementation on glycemic status, lipid profile and glucagon-like peptide 1 level in patients with type 2 diabetes: A randomized double-blind, placebo-controlled trial, Horm. Metab. Res., 49, 886, 10.1055/s-0043-119089

Sen, 2017, Diet-driven microbiota dysbiosis is associated with vagal remodeling and obesity, Physiol. Behav., 173, 305, 10.1016/j.physbeh.2017.02.027

Cani, 2007, Metabolic endotoxemia initiates obesity and insulin resistance, Diabetes, 56, 1761, 10.2337/db06-1491

Boutagy, 2016, Metabolic endotoxemia with obesity: Is it real and is it relevant?, Biochimie, 124, 11, 10.1016/j.biochi.2015.06.020

Pedersen, 2016, Host-microbiome interactions in human type 2 diabetes following prebiotic fibre (galacto-oligosaccharide) intake, Br. J. Nutr., 116, 1869, 10.1017/S0007114516004086

Cox, 2017, Increased intestinal permeability as a risk factor for type 2 diabetes, Diabetes Metab., 43, 163, 10.1016/j.diabet.2016.09.004

Colantonio, A.G., Werner, S.L., and Brown, M. (2019). The effects of prebiotics and substances with prebiotic properties on metabolic and inflammatory biomarkers in individuals with type 2 diabetes mellitus: A systematic review. J. Acad. Nutr. Diet., in press.

Dahl, 2017, Health benefits of fiber fermentation, J. Am. Coll. Nutr., 36, 127, 10.1080/07315724.2016.1188737

Weickert, 2018, Impact of dietary fiber consumption on insulin resistance and the prevention of type 2 diabetes, J. Nutr., 148, 7, 10.1093/jn/nxx008

Reynolds, 2019, Carbohydrate quality and human health: A series of systematic reviews and meta-analyses, Lancet, 393, 434, 10.1016/S0140-6736(18)31809-9

McKenzie, 2016, British Dietetic Association systematic review and evidence-based practice guidelines for the dietary management of irritable bowel syndrome in adults (2016 update), J. Hum. Nutr. Diet., 29, 549, 10.1111/jhn.12385

McRorie, 2015, Evidence-based approach to fiber supplements and clinically meaningful health benefits, Part 1: What to look for and how to recommend an effective fiber therapy, Nutr. Today, 50, 82, 10.1097/NT.0000000000000082

Lambeau, 2017, Fiber supplements and clinically proven health benefits: How to recognize and recommend an effective fiber therapy, J. Am. Assoc. Nurse Pract., 29, 216, 10.1002/2327-6924.12447

Jakobsdottir, G., Xu, J., Molin, G., Ahrne, S., and Nyman, M. (2013). High-fat diet reduces the formation of butyrate, but increases succinate, inflammation, liver fat and cholesterol in rats, while dietary fibre counteracts these effects. PLoS ONE, 8.

Arora, 2019, Microbial fermentation of flaxseed fibers modulates the transcriptome of GPR41-expressing enteroendocrine cells and protects mice against diet-induced obesity, Am. J. Physiol. Endocrinol. Metab., 316, E453, 10.1152/ajpendo.00391.2018

Simpson, 2015, Review article: Dietary fibre-microbiota interactions, Aliment. Pharmacol. Ther., 42, 158, 10.1111/apt.13248

Zhernakova, 2016, Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity, Science, 352, 565, 10.1126/science.aad3369

Trompette, 2014, Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, Nat. Med., 20, 159, 10.1038/nm.3444

Suez, 2014, Artificial sweeteners induce glucose intolerance by altering the gut microbiota, Nature, 514, 181, 10.1038/nature13793

Zeevi, 2015, Personalized nutrition by prediction of glycemic responses, Cell, 163, 1079, 10.1016/j.cell.2015.11.001

Azulay, 2019, Model of personalized postprandial glycemic response to food developed for an Israeli cohort predicts responses in Midwestern American individuals, Am. J. Clin. Nutr., 110, 63, 10.1093/ajcn/nqz028

Bashiardes, 2018, Towards utilization of the human genome and microbiome for personalized nutrition, Curr. Opin. Biotechnol., 51, 57, 10.1016/j.copbio.2017.11.013

Qin, 2012, A metagenome-wide association study of gut microbiota in type 2 diabetes, Nature, 490, 55, 10.1038/nature11450

Wu, 2017, Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug, Nat. Med., 23, 850, 10.1038/nm.4345

Bauer, 2018, Metformin alters upper small intestinal microbiota that impact a glucose-SGLT1-sensing glucoregulatory pathway, Cell Metab., 27, 101, 10.1016/j.cmet.2017.09.019

Mueller, 2017, Metformin Is associated with higher relative abundance of mucin-degrading Akkermansia muciniphila and several short-chain fatty acid-producing microbiota in the gut, Diabetes Care, 40, 54, 10.2337/dc16-1324

Bryrup, 2019, Metformin-induced changes of the gut microbiota in healthy young men: Results of a non-blinded, one-armed intervention study, Diabetologia, 62, 1024, 10.1007/s00125-019-4848-7

Baxter, 2019, The glucoamylase inhibitor acarbose has a diet-dependent and reversible effect on the murine gut microbiome, mSphere, 4, e00528-18, 10.1128/mSphere.00528-18

Wang, 2011, Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, Nature, 472, 57, 10.1038/nature09922

Gregory, 2015, Transmission of atherosclerosis susceptibility with gut microbial transplantation, J. Biol. Chem., 290, 5647, 10.1074/jbc.M114.618249

Tang, 2013, Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk, N. Engl. J. Med., 368, 1575, 10.1056/NEJMoa1109400

Senthong, 2016, Intestinal microbiota-generated metabolite trimethylamine-N-oxide and 5-year mortality risk in stable coronary artery disease: The contributory role of intestinal microbiota in a COURAGE-like patient cohort, J. Am. Heart Assoc., 5, e002816, 10.1161/JAHA.115.002816

Petersen, 2017, Healthy dietary patterns for preventing cardiometabolic disease: The role of plant-based foods and animal products, Curr. Dev. Nutr., 1, e001289, 10.3945/cdn.117.001289

Soliman, G.A. (2018). Dietary cholesterol and the lack of evidence in cardiovascular disease. Nutrients, 10.

Zhong, 2019, Associations of dietary cholesterol or egg consumption with incident cardiovascular disease and mortality, JAMA, 321, 1081, 10.1001/jama.2019.1572

Wang, 2015, Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis, Cell, 163, 1585, 10.1016/j.cell.2015.11.055

Chen, 2017, Gut microbiota-dependent metabolite trimethylamine N-oxide contributes to cardiac dysfunction in Western diet-induced obese mice, Front. Physiol., 8, 139, 10.3389/fphys.2017.00139

Rath, 2017, Uncovering the trimethylamine-producing bacteria of the human gut microbiota, Microbiome, 5, 54, 10.1186/s40168-017-0271-9

Koeth, 2013, Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis, Nat. Med., 19, 576, 10.1038/nm.3145

Koeth, 2019, L-Carnitine in omnivorous diets induces an atherogenic gut microbial pathway in humans, J. Clin. Investig., 129, 373, 10.1172/JCI94601

Alshahrani, S.M., Fraser, G.E., Sabate, J., Knutsen, R., Shavlik, D., Mashchak, A., Lloren, J.I., and Orlich, M.J. (2019). Red and processed meat and mortality in a low meat intake population. Nutrients, 11.

Bellavia, 2016, High red meat intake and all-cause cardiovascular and cancer mortality: Is the risk modified by fruit and vegetable intake?, Am. J. Clin. Nutr., 104, 1137, 10.3945/ajcn.116.135335

Micha, 2017, Association between dietary factors and mortality from heart disease, stroke, and type 2 diabetes in the United States, JAMA, 317, 912, 10.1001/jama.2017.0947

Micha, 2010, Red and processed meat consumption and risk of incident coronary heart disease, stroke, and diabetes mellitus: A systematic review and meta-analysis, Circulation, 121, 2271, 10.1161/CIRCULATIONAHA.109.924977

Satija, 2019, Meta-analysis of randomized controlled trials of red meat consumption in comparison with various comparison diets on cardiovascular risk factors, Circulation, 139, 1828, 10.1161/CIRCULATIONAHA.118.035225

Hills, 2018, Guide to popular diets, food choices, and their health outcome, Health Care Curr. Rev., 6, 223

Jin, Q., Black, A., Kales, S.N., Vattem, D., Ruiz-Canela, M., and Sotos-Prieto, M. (2019). Metabolomics and microbiomes as potential tools to evaluate the effects of the Mediterranean diet. Nutrients, 11.

Lara, 2019, Dietary patterns and incident heart failure in U.S. adults without known coronary disease, J. Am. Coll. Cardiol., 73, 2036, 10.1016/j.jacc.2019.01.067

Grosso, 2017, A comprehensive meta-analysis on evidence of Mediterranean diet and cardiovascular disease: Are individual components equal?, Crit. Rev. Food Sci. Nutr., 57, 3218, 10.1080/10408398.2015.1107021

Pellegrini, 2016, High-level adherence to a Mediterranean diet beneficially impacts the gut microbiota and associated metabolome, Gut, 65, 1812, 10.1136/gutjnl-2015-309957

Alexander, 2015, Red meat and colorectal cancer: A quantitative update on the state of the epidemiologic science, J. Am. Coll. Nutr., 34, 521, 10.1080/07315724.2014.992553

Schwingshackl, L., Schwedhelm, C., Galbete, C., and Hoffmann, G. (2017). Adherence to Mediterranean Diet and risk of cancer: An updated systematic review and meta-analysis. Nutrients, 9.

Thomas, 2019, Metagenomic analysis of colorectal cancer datasets identifies cross-cohort microbial diagnostic signatures and a link with choline degradation, Nat. Med., 25, 667, 10.1038/s41591-019-0405-7

Wirbel, 2019, Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer, Nat. Med., 25, 679, 10.1038/s41591-019-0406-6

Youn, 2019, Association of choline and betaine levels with cancer incidence and survival: A meta-analysis, Clin. Nutr., 38, 100, 10.1016/j.clnu.2018.01.042

Bishehsari, F., Engen, P.A., Preite, N.Z., Tuncil, Y.E., Naqib, A., Shaikh, M., Rossi, M., Wilber, S., Green, S.J., and Hamaker, B.R. (2018). Dietary fiber treatment corrects the composition of gut microbiota, promotes SCFA production, and suppresses colon carcinogenesis. Genes, 9.

Song, 2015, Dietary fibre and the risk of colorectal cancer: A case- control study, Asian Pac. J. Cancer Prev., 16, 3747, 10.7314/APJCP.2015.16.9.3747

Zhang, 2019, Preventable cancer burden associated with poor diet in the United States, JNCI Cancer Spectr., 3, pkz034, 10.1093/jncics/pkz034

Vonaesch, 2018, Pathogens, microbiome and the host: Emergence of the ecological Koch’s postulates, FEMS Microbiol. Rev., 42, 273, 10.1093/femsre/fuy003

Tomkovich, 2019, Human colon mucosal biofilms from healthy or colon cancer hosts are carcinogenic, J. Clin. Investig., 130, 1699, 10.1172/JCI124196

Hill, 2014, Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic, Nat. Rev. Gastroenterol. Hepatol., 11, 506, 10.1038/nrgastro.2014.66

Shen, 2014, Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn’s disease, and pouchitis: Meta-analysis of randomized controlled trials, Inflamm. Bowel Dis., 20, 21, 10.1097/01.MIB.0000437495.30052.be

Wilkins, 2017, Probiotics for gastrointestinal conditions: A summary of the evidence, Am. Fam. Physician, 96, 170

Tojo, 2014, Intestinal microbiota in health and disease: Role of bifidobacteria in gut homeostasis, World J. Gastroenterol., 20, 15163, 10.3748/wjg.v20.i41.15163

Sanchez, 2017, Probiotics, gut microbiota, and their influence on host health and disease, Mol. Nutr. Food Res., 61, 1600240, 10.1002/mnfr.201600240

Allen, 2010, Probiotics for treating acute infectious diarrhoea, Cochrane Database Syst. Rev., 11, CD003048

Goldenberg, 2017, Probiotics for the prevention of Clostridium difficile-associated diarrhea in adults and children, Cochrane Database Syst. Rev., 12, CD006095

Goldenberg, 2018, Probiotics to prevent Clostridium difficile infection in patients receiving antibiotics, JAMA, 320, 499, 10.1001/jama.2018.9064

Fijan, 2014, Microorganisms with claimed probiotic properties: An overview of recent literature, Int. J. Env. Res. Public Health, 11, 4745, 10.3390/ijerph110504745

Dimidi, 2017, Mechanisms of action of probiotics and the gastrointestinal microbiota on gut motility and constipation, Adv. Nutr., 8, 484, 10.3945/an.116.014407

Gil, 2019, Mechanisms of action of probiotics, Adv. Nutr., 10, S49, 10.1093/advances/nmy063

LeBlanc, 2017, Beneficial effects on host energy metabolism of short-chain fatty acids and vitamins produced by commensal and probiotic bacteria, Microb. Cell Fact., 16, 79, 10.1186/s12934-017-0691-z

Ford, 2014, Efficacy of prebiotics, probiotics, and synbiotics in irritable bowel syndrome and chronic idiopathic constipation: Systematic review and meta-analysis, Am. J. Gastroenterol., 109, 1547, 10.1038/ajg.2014.202

Biagi, 2011, Global and deep molecular analysis of microbiota signatures in fecal samples from patients with irritable bowel syndrome, Gastroenterology, 141, 1792, 10.1053/j.gastro.2011.07.043

2016, Bifidobacteria and their role as members of the human gut microbiota, Front. Microbiol., 7, 925

Giamarellos-Bourboulis, E.J., Pyleris, E., Barbatzas, C., Pistiki, A., and Pimentel, M. (2016). Small intestinal bacterial overgrowth is associated with irritable bowel syndrome and is independent of proton pump inhibitor usage. BMC Gastroenterol., 16.

Saffouri, 2019, Small intestinal microbial dysbiosis underlies symptoms associated with functional gastrointestinal disorders, Nat. Commun., 10, 2012, 10.1038/s41467-019-09964-7

2018, The unregulated probiotic market, Clin. Gastroenterol. Hepatol., 17, 809

Campana, 2017, Strain-specific probiotic properties of lactic acid bacteria and their interference with human intestinal pathogens invasion, Gut Pathog., 9, 12, 10.1186/s13099-017-0162-4

Wang, L., Hu, L., Xu, Q., Yin, B., Fang, D., Wang, G., Zhao, J., Zhang, H., and Chen, W. (2017). Bifidobacterium adolescentis exerts strain-specific effects on constipation induced by loperamide in BALB/c mice. Int. J. Mol. Sci., 18.

Conway, T., and Cohen, P.S. (2015). Commensal and pathogenic Escherichia coli metabolism in the gut. Microbiol. Spectr., 3.

Goswami, 2015, Coculture of Escherichia coli O157:H7 with a nonpathogenic E. coli strain increases toxin production and virulence in a germfree mouse model, Infect. Immun., 83, 4185, 10.1128/IAI.00663-15

Crook, 2019, Adaptive strategies of the candidate probiotic E. coli Nissle in the mammalian gut, Cell Host Microbe, 25, 499, 10.1016/j.chom.2019.02.005

Lerner, 2017, Potential effects of horizontal gene exchange in the human gut, Front. Immunol., 8, 1630, 10.3389/fimmu.2017.01630

Trinchieri, 2017, Efficacy and safety of a multistrain probiotic formulation depends from manufacturing, Front. Immunol., 8, 1474, 10.3389/fimmu.2017.01474

Hod, 2018, The effect of a multispecies probiotic on microbiota composition in a clinical trial of patients with diarrhea-predominant irritable bowel syndrome, Neurogastroenterol. Motil., 30, e13456, 10.1111/nmo.13456

Lee, 2018, The therapeutic effect of a multistrain probiotic on diarrhea-predominant irritable bowel syndrome: A pilot study, Gastroenterol. Res. Pract., 2018, 8791916, 10.1155/2018/8791916

Plaza-Diaz, J., Ruiz-Ojeda, F.J., Vilchez-Padial, L.M., and Gil, A. (2017). Evidence of the anti-inflammatory effects of probiotics and synbiotics in intestinal chronic diseases. Nutrients, 9.

Floch, 2015, Recommendations for probiotic use--2015 Update: Proceedings and consensus opinion, J. Clin. Gastroenterol., 49, S69, 10.1097/MCG.0000000000000420

Markowiak, P., and Slizewska, K. (2017). Effects of probiotics, prebiotics, and synbiotics on human health. Nutrients, 9.

Hughes, 2017, Bifidobacterium breve reduces apoptotic epithelial cell shedding in an exopolysaccharide and MyD88-dependent manner, Open Biol., 7, 160155, 10.1098/rsob.160155

Lam, Y.Y., Ha, C.W., Campbell, C.R., Mitchell, A.J., Dinudom, A., Oscarsson, J., Cook, D.I., Hunt, N.H., Caterson, I.D., and Holmes, A.J. (2012). Increased gut permeability and microbiota change associate with mesenteric fat inflammation and metabolic dysfunction in diet-induced obese mice. PLoS ONE, 7.

Wang, 2011, Lactobacillus rhamnosus GG treatment potentiates intestinal hypoxia-inducible factor, promotes intestinal integrity and ameliorates alcohol-induced liver injury, Am. J. Pathol., 179, 2866, 10.1016/j.ajpath.2011.08.039

Reid, 2006, Probiotics to prevent the need for, and augment the use of, antibiotics, Can. J. Infect. Dis. Med. Microbiol., 17, 219, 10.1155/2006/934626

Sherman, 2005, Probiotics reduce enterohemorrhagic Escherichia coli O157:H7- and enteropathogenic E. coli O127:H6-induced changes in polarized T84 epithelial cell monolayers by reducing bacterial adhesion and cytoskeletal rearrangements, Infect. Immun., 73, 5183, 10.1128/IAI.73.8.5183-5188.2005

Ferrario, 2014, Modulation of fecal Clostridiales bacteria and butyrate by probiotic intervention with Lactobacillus paracasei DG varies among healthy adults, J. Nutr., 144, 1787, 10.3945/jn.114.197723

Barbara, 2018, Probiotics in irritable bowel syndrome: Where are we?, Neurogastroenterol. Motil., 30, e13513, 10.1111/nmo.13513

Baxter, 2019, Dynamics of human gut microbiota and short-chain fatty acids in response to dietary interventions with three fermentable fibers, mBio, 10, e02566-18, 10.1128/mBio.02566-18

Ribeiro, 2017, Gut microbiota and probiotics: Focus on diabetes mellitus, Crit. Rev. Food Sci. Nutr., 57, 2296, 10.1080/10408398.2014.934438

Sanchez, 2014, Effect of Lactobacillus rhamnosus CGMCC1.3724 supplementation on weight loss and maintenance in obese men and women, Br. J. Nutr., 111, 1507, 10.1017/S0007114513003875

Sabico, 2018, Effects of a 6-month multi-strain probiotics supplementation in endotoxemic, inflammatory and cardiometabolic status of T2DM patients: A randomized, double-blind, placebo-controlled trial, Clin. Nutr., 38, 1561, 10.1016/j.clnu.2018.08.009

Abildgaard, 2017, Probiotic treatment reduces depressive-like behaviour in rats independently of diet, Psychoneuroendocrinology, 79, 40, 10.1016/j.psyneuen.2017.02.014

Kadooka, 2013, Effect of Lactobacillus gasseri SBT2055 in fermented milk on abdominal adiposity in adults in a randomised controlled trial, Br. J. Nutr., 110, 1696, 10.1017/S0007114513001037

Kim, 2018, Lactobacillus gasseri BNR17 supplementation reduces the visceral fat accumulation and waist circumference in obese adults: A randomized, double-blind, placebo-controlled trial, J. Med. Food, 21, 454, 10.1089/jmf.2017.3937

Valcheva, 2016, Prebiotics: Definition and protective mechanisms, Best Pract. Res. Clin. Gastroenterol., 30, 27, 10.1016/j.bpg.2016.02.008

Nie, Y., Lin, Q., and Luo, F. (2017). Effects of non-starch polysaccharides on inflammatory bowel disease. Int. J. Mol. Sci., 18.

Gil, 2019, Effects of sweeteners on the gut microbiota: A review of experimental studies and clinical trials, Adv. Nutr., 10, S31, 10.1093/advances/nmy037

Leenen, 2007, Inulin and oligofructose in chronic inflammatory bowel disease, J. Nutr., 137, 2572S, 10.1093/jn/137.11.2572S

Musilova, 2015, Prebiotic effects of a novel combination of galactooligosaccharides and maltodextrins, J. Med. Food, 18, 685, 10.1089/jmf.2013.0187

Nicolucci, 2017, Prebiotics reduce body fat and alter intestinal microbiota in children who are overweight or with obesity, Gastroenterology, 153, 711, 10.1053/j.gastro.2017.05.055

Rios, 2019, Protective effect of prebiotic and exercise intervention on knee health in a rat model of diet-induced obesity, Sci. Rep., 9, 3893, 10.1038/s41598-019-40601-x

Chen, 2017, Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota, Sci. Rep., 7, 2594, 10.1038/s41598-017-02995-4

Thomas, 2018, What is the evidence for a role for diet and nutrition in osteoarthritis?, Rheumatology, 57, iv61, 10.1093/rheumatology/key011

Watson, 2018, A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiota, Gut, 67, 1974, 10.1136/gutjnl-2017-314968

Costantini, L., Molinari, R., Farinon, B., and Merendino, N. (2017). Impact of omega-3 fatty acids on the gut microbiota. Int. J. Mol. Sci., 18.

Menni, 2017, Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women, Sci. Rep., 7, 11079, 10.1038/s41598-017-10382-2

Kim, 2019, Coffee consumption and all-cause and cause-specific mortality: A meta-analysis by potential modifiers, Eur. J. Epidemiol., 34, 731, 10.1007/s10654-019-00524-3

Falony, 2016, Population-level analysis of gut microbiome variation, Science, 352, 560, 10.1126/science.aad3503

Vandeputte, 2016, Stool consistency is strongly associated with gut microbiota richness and composition, enterotypes and bacterial growth rates, Gut, 65, 57, 10.1136/gutjnl-2015-309618

Tropini, 2018, Transient osmotic perturbation causes long-term alteration to the gut microbiota, Cell, 173, 1742, 10.1016/j.cell.2018.05.008

Lee, S.H., Yun, Y., Kim, S.J., Lee, E.J., Chang, Y., Ryu, S., Shin, H., Kim, H.L., Kim, H.N., and Lee, J.H. (2018). Association between cigarette smoking status and composition of gut microbiota: Population-based cross-sectional study. J. Clin. Med., 7.

Song, 2013, Cohabiting family members share microbiota with one another and with their dogs, eLife, 2, e00458, 10.7554/eLife.00458

Parajuli, 2018, Urbanization reduces transfer of diverse environmental microbiota indoors, Front. Microbiol., 9, 84, 10.3389/fmicb.2018.00084

Mahnert, 2019, Man-made microbial resistances in built environments, Nat. Commun., 10, 968, 10.1038/s41467-019-08864-0

Fragiadakis, 2019, Links between environment, diet, and the hunter-gatherer microbiome, Gut Microbes, 10, 216, 10.1080/19490976.2018.1494103

Fu, 2015, The gut microbiome contributes to a substantial proportion of the variation in blood lipids, Circ. Res., 117, 817, 10.1161/CIRCRESAHA.115.306807

Elshazly, 2013, Non-high-density lipoprotein cholesterol, guideline targets, and population percentiles for secondary prevention in 1.3 million adults: The VLDL-2 study (very large database of lipids), J. Am. Coll. Cardiol., 62, 1960, 10.1016/j.jacc.2013.07.045

Ravnskov, 2018, LDL-C does not cause cardiovascular disease: A comprehensive review of the current literature, Expert Rev. Clin. Pharmacol., 11, 959, 10.1080/17512433.2018.1519391

Wu, 2019, Age, sex, and cardiovascular risk attributable to lipoprotein cholesterol among chinese individuals with coronary artery disease: A case-control study, Metab. Syndr. Relat. Disord., 17, 223, 10.1089/met.2018.0067

Ference, 2017, Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European Atherosclerosis Society Consensus Panel, Eur. Heart J., 38, 2459, 10.1093/eurheartj/ehx144

Kastelein, 2008, Lipids, apolipoproteins, and their ratios in relation to cardiovascular events with statin treatment, Circulation, 117, 3002, 10.1161/CIRCULATIONAHA.107.713438

Meeusen, 2016, Should apolipoprotein B replace LDL cholesterol as therapeutic targets are lowered?, Curr. Opin. Lipidol., 27, 359, 10.1097/MOL.0000000000000313

Greene, 2006, Plasma LDL and HDL characteristics and carotenoid content are positively influenced by egg consumption in an elderly population, Nutr. Metab., 3, 6, 10.1186/1743-7075-3-6

Willeit, 2018, Baseline and on-statin treatment lipoprotein(a) levels for prediction of cardiovascular events: Individual patient-data meta-analysis of statin outcome trials, Lancet, 392, 1311, 10.1016/S0140-6736(18)31652-0

Tsimikas, S., Gordts, P., Nora, C., Yeang, C., and Witztum, J.L. (2019). Statin therapy increases lipoprotein(a) levels. Eur. Heart J., in press.

Choi, 2010, Statins do not decrease small, dense low-density lipoprotein, Tex. Heart Inst. J., 37, 421

Magge, 2012, Low-FODMAP diet for treatment of irritable bowel syndrome, Gastroenterol. Hepatol., 8, 739

Skodje, 2018, Fructan, rather than gluten, induces symptoms in patients with self-reported non-Celiac gluten sensitivity, Gastroenterology, 154, 529, 10.1053/j.gastro.2017.10.040

Biesiekierski, 2013, No effects of gluten in patients with self-reported non-celiac gluten sensitivity after dietary reduction of fermentable, poorly absorbed, short-chain carbohydrates, Gastroenterology, 145, 320, 10.1053/j.gastro.2013.04.051

Fasano, 2015, Nonceliac gluten sensitivity, Gastroenterology, 148, 1195, 10.1053/j.gastro.2014.12.049

Halmos, E.P., and Gibson, P.R. (2019). Controversies and reality of the FODMAP diet for patients with irritable bowel syndrome. J. Gastroenterol. Hepatol., in press.

Staudacher, H.M., Ralph, F.S.E., Irving, P.M., Whelan, K., and Lomer, M.C.E. (2019). Nutrient intake, diet quality, and diet diversity in irritable bowel syndrome and the impact of the low FODMAP diet. J. Acad. Nutr. Diet., in press.

Claesson, 2012, Gut microbiota composition correlates with diet and health in the elderly, Nature, 488, 178, 10.1038/nature11319

Halmos, 2015, Diets that differ in their FODMAP content alter the colonic luminal microenvironment, Gut, 64, 93, 10.1136/gutjnl-2014-307264

Dieterich, 2019, Influence of low FODMAP and gluten-free diets on disease activity and intestinal microbiota in patients with non-celiac gluten sensitivity, Clin. Nutr., 38, 697, 10.1016/j.clnu.2018.03.017

Kosinski, C., and Jornayvaz, F.R. (2017). Effects of ketogenic diets on cardiovascular risk factors: Evidence from animal and human studies. Nutrients, 9.

Ebbeling, 2018, Effects of a low carbohydrate diet on energy expenditure during weight loss maintenance: Randomized trial, BMJ, 363, k4583, 10.1136/bmj.k4583

Fung, 2011, Low-carbohydrate diet scores and risk of type 2 diabetes in men, Am. J. Clin. Nutr., 93, 844, 10.3945/ajcn.110.004333

Seidelmann, 2018, Dietary carbohydrate intake and mortality: A prospective cohort study and meta-analysis, Lancet Public Health, 3, e419, 10.1016/S2468-2667(18)30135-X

Swidsinski, 2017, Reduced mass and diversity of the colonic microbiome in patients with multiple sclerosis and their improvement with ketogenic diet, Front. Microbiol., 8, 1141, 10.3389/fmicb.2017.01141

Zhang, 2018, Altered gut microbiome composition in children with refractory epilepsy after ketogenic diet, Epilepsy Res., 145, 163, 10.1016/j.eplepsyres.2018.06.015

Kearns, C.E., Apollonio, D., and Glantz, S.A. (2017). Sugar industry sponsorship of germ-free rodent studies linking sucrose to hyperlipidemia and cancer: An historical analysis of internal documents. PLoS Biol., 15.

Hoffmann, C., Dollive, S., Grunberg, S., Chen, J., Li, H., Wu, G.D., Lewis, J.D., and Bushman, F.D. (2013). Archaea and fungi of the human gut microbiome: Correlations with diet and bacterial residents. PLoS ONE, 8.

Samuel, 2006, A humanized gnotobiotic mouse model of host-archaeal-bacterial mutualism, Proc. Natl. Acad. Sci. USA, 103, 10011, 10.1073/pnas.0602187103

McNeil, 1984, The contribution of the large intestine to energy supplies in man, Am. J. Clin. Nutr., 39, 338, 10.1093/ajcn/39.2.338

Ghoshal, 2016, Irritable bowel syndrome, particularly the constipation-predominant form, involves an increase in Methanobrevibacter smithii, which is associated with higher methane production, Gut Liver, 10, 932, 10.5009/gnl15588

Suhr, 2017, Fungi in the healthy human gastrointestinal tract, Virulence, 8, 352, 10.1080/21505594.2016.1247140

Otasevic, 2018, The dietary modification and treatment of intestinal Candida overgrowth - a pilot study, J. Mycol. Med., 28, 623, 10.1016/j.mycmed.2018.08.002

Bertolini, M., Ranjan, A., Thompson, A., Diaz, P.I., Sobue, T., Maas, K., and Dongari-Bagtzoglou, A. (2019). Candida albicans induces mucosal bacterial dysbiosis that promotes invasive infection. PLoS Pathog., 15.

Cottier, 2015, MIG1 regulates resistance of Candida albicans against the fungistatic effect of weak organic acids, Eukaryot. Cell, 14, 1054, 10.1128/EC.00129-15

Allonsius, 2017, Interplay between Lactobacillus rhamnosus GG and Candida and the involvement of exopolysaccharides, Microb. Biotechnol., 10, 1753, 10.1111/1751-7915.12799

Haas, 1955, The treatment of celiac disease with the specific carbohydrate diet; report on 191 additional cases, Am. J. Gastroenterol., 23, 344

(2019, July 14). Breaking the Vicious Cycle and the Specific Carbohydrate Diet. Available online: www.breakingtheviciouscycle.info.

Gottschall, E. (1994). Breaking the Vicious Cycle: Intestinal Health Through Diet, Kirkton Press.

(2019, June 26). Nutrition in Immune Balance (NiMBAL)—Food Table. Available online: www.nimbal.org/legalillegal-food-list.

Kakodkar, 2015, The specific carbohydrate diet for inflammatory bowel disease: A case series, J. Acad. Nutr. Diet., 115, 1226, 10.1016/j.jand.2015.04.016

Chassaing, 2015, Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome, Nature, 519, 92, 10.1038/nature14232

Chassaing, 2017, Colonic microbiota encroachment correlates with dysglycemia in humans, Cell. Mol. Gastroenterol. Hepatol., 4, 205, 10.1016/j.jcmgh.2017.04.001

Miclotte, L., and Van De Wiele, T. (2019). Food processing, gut microbiota and the globesity problem. Crit. Rev. Food Sci. Nutr., 1–14. in press.

Li, 2017, Intermittent fasting promotes white adipose browning and decreases obesity by shaping the gut microbiota, Cell Metab., 26, 672, 10.1016/j.cmet.2017.08.019

Chevalier, 2015, Gut microbiota orchestrates energy homeostasis during cold, Cell, 163, 1360, 10.1016/j.cell.2015.11.004

Zarrinpar, 2014, Diet and feeding pattern affect the diurnal dynamics of the gut microbiome, Cell Metab., 20, 1006, 10.1016/j.cmet.2014.11.008

Cignarella, 2018, Intermittent fasting confers protection in CNS autoimmunity by altering the gut microbiota, Cell Metab., 27, 1222, 10.1016/j.cmet.2018.05.006

Rangan, 2019, Fasting-mimicking diet modulates microbiota and promotes intestinal regeneration to reduce inflammatory bowel disease pathology, Cell Rep., 26, 2704, 10.1016/j.celrep.2019.02.019

Catterson, 2018, Short-term, intermittent fasting induces long-lasting gut health and TOR-independent lifespan extension, Curr. Biol., 28, 1714, 10.1016/j.cub.2018.04.015

Muccioli, 2010, The endocannabinoid system links gut microbiota to adipogenesis, Mol. Syst. Biol., 6, 392, 10.1038/msb.2010.46

Pesce, 2018, Endocannabinoids in the treatment of gasytrointestinal inflammation and symptoms, Curr. Opin. Pharmacol., 43, 81, 10.1016/j.coph.2018.08.009

De Filippis, D., Esposito, G., Cirillo, C., Cipriano, M., De Winter, B.Y., Scuderi, C., Sarnelli, G., Cuomo, R., Steardo, L., and De Man, J.G. (2011). Cannabidiol reduces intestinal inflammation through the control of neuroimmune axis. PLoS ONE, 6.

Uranga, 2018, Cannabinoid pharmacology and therapy in gut disorders, Biochem. Pharmacol., 157, 134, 10.1016/j.bcp.2018.07.048

2011, Obesity and cannabis use: Results from 2 representative national surveys, Am. J. Epidemiol., 174, 929, 10.1093/aje/kwr200

Cluny, N.L., Keenan, C.M., Reimer, R.A., Le Foll, B., and Sharkey, K.A. (2015). Prevention of diet-induced obesity effects on body weight and gut microbiota in mice treated chronically with delta9-tetrahydrocannabinol. PLoS ONE, 10.

Zhang, 2013, Antibiotic administration routes significantly influence the levels of antibiotic resistance in gut microbiota, Antimicrob. Agents Chemother., 57, 3659, 10.1128/AAC.00670-13

Solomon, 2016, Effect of proton pump inhibitors and antibiotics on the gut microbiome of hospitalised older persons, J. Infect., 72, 498, 10.1016/j.jinf.2016.01.015

Blaser, 2016, Antibiotic use and its consequences for the normal microbiome, Science, 352, 544, 10.1126/science.aad9358

Su, 2018, Meta-analysis: Proton pump inhibitors moderately increase the risk of small intestinal bacterial overgrowth, J. Gastroenterol., 53, 27, 10.1007/s00535-017-1371-9

Trifan, 2017, Proton pump inhibitors therapy and risk of Clostridium difficile infection: Systematic review and meta-analysis, World J. Gastroenterol., 23, 6500, 10.3748/wjg.v23.i35.6500

Gregoire, 2018, Systematic review: Human gut dysbiosis induced by non-antibiotic prescription medications, Aliment. Pharmacol. Ther., 47, 332, 10.1111/apt.14451

Imhann, 2016, Proton pump inhibitors affect the gut microbiome, Gut, 65, 740, 10.1136/gutjnl-2015-310376

Bahr, 2015, Use of the second-generation antipsychotic, risperidone, and secondary weight gain are associated with an altered gut microbiota in children, Transl. Psychiatry, 5, e652, 10.1038/tp.2015.135

Flowers, 2017, Interaction between atypical antipsychotics and the gut microbiome in a bipolar disease cohort, Pharmacotherapy, 37, 261, 10.1002/phar.1890

Rogers, 2016, The influence of non-steroidal anti-inflammatory drugs on the gut microbiome, Clin. Microbiol. Infect., 22, 178, 10.1016/j.cmi.2015.10.003

Meng, J., Yu, H., Ma, J., Wang, J., Banerjee, S., Charboneau, R., Barke, R.A., and Roy, S. (2013). Morphine induces bacterial translocation in mice by compromising intestinal barrier function in a TLR-dependent manner. PLoS ONE, 8.

Sharon, 2019, Human gut microbiota from autism spectrum disorder promote behavioral symptoms in mice, Cell, 177, 1600, 10.1016/j.cell.2019.05.004

Martinez, 2014, Understanding gut microbiota in elderly’s health will enable intervention through probiotics, Benef. Microbes, 5, 235, 10.3920/BM2013.0079

Davenport, E.R., Mizrahi-Man, O., Michelini, K., Barreiro, L.B., Ober, C., and Gilad, Y. (2014). Seasonal variation in human gut microbiome composition. PLoS ONE, 9.

Jeffery, 2015, Gut microbiota and aging, Science, 350, 1214, 10.1126/science.aac8469

Armour, 2019, A metagenomic meta-analysis reveals functional signatures of health and disease in the human gut microbiome, mSystems, 4, e00332-18, 10.1128/mSystems.00332-18

Brunt, 2019, Suppression of the gut microbiome ameliorates age-related arterial dysfunction and oxidative stress in mice, J. Physiol., 597, 2361, 10.1113/JP277336

Smith, 2017, Regulation of life span by the gut microbiota in the short-lived African turquoise killifish, eLife, 6, e27014, 10.7554/eLife.27014

Tirosh, 2019, The short-chain fatty acid propionate increases glucagon and FABP4 production, impairing insulin action in mice and humans, Sci. Transl. Med., 11, eaav0120, 10.1126/scitranslmed.aav0120

Perry, 2016, Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic syndrome, Nature, 534, 213, 10.1038/nature18309

Si, 2018, Gut microbiome-induced shift of acetate to butyrate positively manages dysbiosis in high fat diet, Mol. Nutr. Food Res., 62, 1700670, 10.1002/mnfr.201700670

Arrieta, 2018, Microbial eukaryotes: A missing link in gut microbiome studies, mSystems, 3, e00201-17, 10.1128/mSystems.00201-17

Rampelli, 2017, Characterization of the human DNA gut virome across populations with different subsistence strategies and geographical origin, Environ. Microbiol., 19, 4728, 10.1111/1462-2920.13938

Hsu, 2019, Dynamic modulation of the gut microbiota and metabolome by bacteriophages in a mouse model, Cell Host Microbe, 25, 803, 10.1016/j.chom.2019.05.001

Norman, 2015, Disease-specific alterations in the enteric virome in inflammatory bowel disease, Cell, 160, 447, 10.1016/j.cell.2015.01.002

Audebert, 2016, The Blastocystis Investigation Group; Loywick, A.; Merlin, S.; Viscogliosi, E.; Chabe, M. Colonization with the enteric protozoa Blastocystis is associated with increased diversity of human gut bacterial microbiota, Sci. Rep., 6, 25255, 10.1038/srep25255

Poirier, P., Wawrzyniak, I., Vivares, C.P., Delbac, F., and El Alaoui, H. (2012). New insights into Blastocystis spp.: A potential link with irritable bowel syndrome. PLoS Pathog., 8.

Yason, 2019, Interactions between a pathogenic Blastocystis subtype and gut microbiota: In vitro and in vivo studies, Microbiome, 7, 30, 10.1186/s40168-019-0644-3

Reynolds, 2018, Asymptomatic intestinal colonization with protist Blastocystis is strongly associated with distinct microbiome ecological patterns, mSystems, 3, e00007-18, 10.1128/mSystems.00007-18

Gentekaki, E., Curtis, B.A., Stairs, C.W., Klimes, V., Elias, M., Salas-Leiva, D.E., Herman, E.K., Eme, L., Arias, M.C., and Henrissat, B. (2017). Extreme genome diversity in the hyper-prevalent parasitic eukaryote Blastocystis. PLoS Biol., 15.

Zmora, 2019, You are what you eat: Diet, health and the gut microbiota, Nat. Rev. Gastroenterol. Hepatol., 16, 35, 10.1038/s41575-018-0061-2

Chen, 2019, Association among dietary supplement use, nutrient intake, and mortality among U.S. adults: A cohort study, Ann. Intern. Med., 170, 604, 10.7326/M18-2478

Fetissov, 2017, Role of the gut microbiota in host appetite control: Bacterial growth to animal feeding behaviour, Nat. Rev. Endocrinol., 13, 11, 10.1038/nrendo.2016.150

Thorburn, 2014, Diet, metabolites, and “Western-lifestyle” inflammatory diseases, Immunity, 40, 833, 10.1016/j.immuni.2014.05.014

Vieira-Silva, S., Sabino, J., Valles-Colomer, M., Falony, G., Kathagen, G., Caenepeel, C., Cleynen, I., Van Der Merwe, S., Vermeire, S., and Raes, J. (2019). Quantitative microbiome profiling disentangles inflammation- and bile duct obstruction-associated microbiota alterations across PSC/IBD diagnoses. Nat. Microbiol., in press.

Hillman, 2017, Microbial ecology along the gastrointestinal tract, Microbes Environ., 32, 300, 10.1264/jsme2.ME17017

Estelle, 2018, Characterization of bacterial microbiota compositions along the intestinal tract in pigs and their interactions and functions, Sci. Rep., 8, 12727, 10.1038/s41598-018-30932-6

Johnson, 2019, Daily sampling reveals personalized diet-microbiome associations in humans, Cell Host Microbe, 25, 789, 10.1016/j.chom.2019.05.005

David, 2014, Diet rapidly and reproducibly alters the human gut microbiome, Nature, 505, 559, 10.1038/nature12820

Zimmermann, 2019, Mapping human microbiome drug metabolism by gut bacteria and their genes, Nature, 570, 462, 10.1038/s41586-019-1291-3

De Toro-Martin, J., Arsenault, B.J., Despres, J.P., and Vohl, M.C. (2017). Precision nutrition: A review of personalized nutritional approaches for the prevention and management of metabolic syndrome. Nutrients, 9.

Zhang, 2018, Time for food: The impact of diet on gut microbiota and human health, Nutrition, 51–52, 80, 10.1016/j.nut.2017.12.005