Part of Referencing Articles For The Sample Intelligence
[1] Portincasa P, et al. Curcumin and Fennel Essential Oil Improve Symptoms and Quality of Life in Patients with Irritable Bowel Syndrome. J Gastrointestin Liver Dis;25(2):151-7. (2016).
[2] Markus V, et al. Anti-Quorum Sensing Activity of Stevia Extract, Stevioside, Rebaudioside A and Their Aglycon Steviol. Molecules. 25(22):5480. (2020).
[3] Lever E, et al. Systematic review: the effect of prunes on gastrointestinal function. Aliment Pharmacol Ther. (2014).
[4] Jalanka J, et al. The Effect of Psyllium Husk on Intestinal Microbiota in Constipated Patients and Healthy Controls. International Journal of Molecular Sciences. (2019).
[5] Maier TV, et al. Impact of Dietary Resistant Starch on the Human Gut Microbiome, Metaproteome, and Metabolome. American Soc. Microbiology mBio 8:e01343-17. (2017).
[6] Pallister T, et al. Food: a new form of personalised (gut microbiome) medicine for chronic diseases? J R Soc Med. 109(9):331-6. (2016).
[7] Tillisch K, et al. Consumption of Fermented Milk Product With Probiotic Modulates Brain Activity. Gastroenterology. 144(7):10.1053/j.gastro.2013.02.043. (2013).
[8] Schmidt K, et al. Prebiotic intake reduces the waking cortisol response and alters emotional bias in healthy volunteers. Psychopharmacology 232(10): 1793–1801. (2015).
[9] Clarke SF, et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 63, 1913. (2014).
[10] Allen JM, et al. Exercise alters gut microbiota composition and function in lean and obese humans. Med Sci Sports Exerc. 50(4):747– 57. (2018).
[11] Mitchell CM, et al. Does Exercise Alter Gut Microbial Composition? A Systematic Review. Med Sci Sports Exerc. 51(1):160-167. (2019).
[12] Othaim A, et al. Amounts and Botanical Diversity of Dietary Fruits and Vegetables Affect Distinctly the Human Gut Microbiome, Current Developments in Nutrition, Volume 4, Issue
Supplement_2 Page 1545. (2020).
[13] Cappello G, et al. Peppermint oil (Mintoil) in the treatment of irritable bowel syndrome: a prospective double blind placebo-controlled randomized trial. Dig Liver Dis. 39(6):530-6.
(2007).
[14] Khanna R, et al. Peppermint oil for the treatment of irritable bowel syndrome: a systematic review and meta-analysis. J Clin Gastroenterol. 48(6):505-12. (2014).
[15] Beißner F, et al. Therapeutische Empfehlungen, Akupunkt 61: 2. (2018).
[16] Schmid CM, et al. Characterization of agonistic (aroma-active and physiologically active) compounds in thyme, oregano and marjoram, Techn Univ München. (2018).
[17] Martín-Peláez S, et al. Effect of virgin olive oil and thyme phenolic compounds on blood lipid prole: implications of human gut microbiota. Eur J Nutr 56: 119. (2017).
[18] Europäisches Arzneibuch (http://www.edqm.eu)
[19] Ried K, et al. Potential of garlic (Allium sativum) in lowering high blood pressure: mechanisms of action and clinical relevance. Integr Blood Press Control. 7:71-82. (2014).
[20] Sahebkar A, et al. Effect of garlic on plasma lipoprotein(a) concentrations: A systematic review and meta-analysis of randomized controlled clinical trials. Nutrition. 32(1):33-40.
(2016).
[21] Capili B, et al. Addressing the Role of Food in Irritable Bowel Syndrome Symptom Management. J Nurse Pract. 12(5):324-329. (2016).
[22] Rowland I, et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr 57: 1. (2018).
[23] Tianthong W, et al. A randomized, double-blind, placebo-controlled trial on the efcacy of ginger in the prevention of abdominal distention in post cesarean section patients. Sci Rep.
8(1):6835. (2018).
[24] Singh RP, et al. Cuminum cyminum – A Popular Spice: An Updated Review. Pharmacogn J. 9(3):292-301. (2017).
[25] Gentile L, et al. Oleuropein: Molecular Dynamics and Computation. 24(39):4315-4328. (2017).
[26] Gavahian M, et al. Health benets of olive oil and its components: Impacts on gut microbiota antioxidant activities, and prevention of noncommunicable diseases Trends in Food Sc. &
Tech. 88:220-227. (2019).
[27] Pacheco C, et al. Retention and pre-colon bioaccessibility of oleuropein in starchy food matrices, and the effect of microencapsulation by using inulin. J. Funct. Foods 41:112-117.
(2018).
[28] López de las Hazas MC, et al. Differential absorption and metabolism of hydroxytyrosol and its precursors oleuropein and secoiridoids. J Funct Foods 22: 52-63 .(2016).
[29] Zielińska A, et al. Abundance of active ingredients in sea-buckthorn oil. Lipids Health Dis. 16: 95. (2017).
[30] Olas B, et al. The benecial health aspects of sea buckthorn (Elaeagnus rhamnoides (L.) A.Nelson) oil. J Ethnopharmacol.;213:183-190. (2018).
[31] Yang B, et al. Clinical evidence on potential health benets of berries. Curr Op Food Science 2:36-42. (2015).
[32] Dreher ML, et al. Starch digestibility of foods: a nutritional perspective. Crit Rev Food Sci Nutr. 20(1):47-71.(1984)
[33] Lin AH, et al. Structure and Digestion of Common Complementary Food Starches. J Pediatr Gastroenterol Nutr. 66 Suppl 3:S35-S38. (2018).
[34] Stargrove M, et al. Herb, Nutrient and Drug Interactions: Clinical Implications and Therapeutic Strategies, 1. Auage. St. Louis, Missouri: Elsevier Health Sciences. (2008).
[35] Markus V, et al. Inhibitory Effects of Artificial Sweeteners on Bacterial Quorum Sensing. Int. J. Mol. Sci. (2021).
[36] Donaldson GP, et al. Gut biogeography of the bacterial microbiota. Nat Rev Microbiol 14, 20–32 (2015).
[37] Li J, et al. An integrated catalog of reference genes in the human gut microbiome. Nat Biotechnol 32, 834–841 (2014).
[38] De Angelis M, et al. Diet influences the functions of the human intestinal microbiome. Sci Rep 10, 4247 (2020).
[39] Chatelier E, et al. Richness of human gut microbiome correlates with metabolic markers. Nature 500, 541 (2013).
[40] Cotillard A, et al. Dietary intervention impact on gut microbial gene richness. Nature 500, 585 (2013).
[41] Human Microbiome Project Consortium, A framework for human microbiome research, Nature 486(7402):215-221. (2012)
[42] Lozupone CA, et al. Diversity, stability and resilience of the human gut microbiota. Nature 489, 220 (2012).
[43] Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature 486, 222 (2012).
[44] Human Microbiome Project Consortium, Structure, function and diversity of the healthy human microbiome, Nature 486:207-214 (2012).
[45] Martinez-Guryn M, et al. Regional diversity of the gastroinestinal microbiome, Cell Host & Microbe 26:314-324 (2019).
[46] Masco L, et al. Polyphasic taxonomic analysis of Bidobacterium animalis and Bidobacterium lactis reveals relatedness at the subspecies level: reclassication of Bidobacterium animalis
as Bidobacterium animalis subsp. animalis subsp. nov. and Bidobacterium lactis as Bidobacterium animalis subsp. lactis subsp. nov., Int J Syst Evol Microbiol. 54(4):1137-43 (2004).
[47] O'Callaghan A, et al. Bifidobacteria and Their Role as Members of the Human Gut Microbiota. Front Microbiol. 7: 925 (2016).
[48] Rivière A, et al. Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategies for Their Stimulation in the Human Gut. Front Microbiol. 7: 979 (2016).
[49] Naumova N, et al. Human Gut Microbiome Response to Short-Term Bifidobacterium-Based Probiotic Treatment. Indian J Microbiol 60, 451–457 (2020).
[50] Malinen E, et al. Association of symptoms with gastrointestinal microbiota in irritable bowel syndrome. World J Gastroentero 16, 4532–4540 (2010).
[51] Markowiak-Kopeć, et al. The Effect of Probiotics on the Production of Short-Chain Fatty Acids by Human Intestinal Microbiome. Nutrients 12:1107 (2020).
[52] Mayengbam S, et al. Impact of dietary ber supplementation on modulating microbiota-host-metabolic axes in obesity. J Nutritional Biochem (2018).
[53] Heeney DD, et al. Intestinal Lactobacillus in health and disease, a driver or just along for the ride? Curr Opin Biotechnol. 49:140-147 (2018).
[54] Marco ML, et al. Health benets of fermented foods: microbiota and beyond, Curr Opin Biotechnol. 44:94-102. (2017).
[55] Holzapfel WH, et al. Taxonomy and important features of probiotic microorganisms in food and nutrition , American J Clinical Nutrition,73(2):365S–373 (2001).
[56] Tingirikari JMR, Microbiota-accessible pectic poly- and oligosaccharides in gut health. Food Funct. 9(10):5059-5073 (2018).
[57] Ndeh D, et al. Complex pectin metabolism by gut bacteria reveals novel catalytic functions. Nature 544(7648):65-70 (2017).
[58] Tomas-Barberan FA, et al. Advances in Health-Promoting Food Ingredients. J. Agric. Food Chem., 67, 33, 9121-9123 (2019) .
[59] Duda‐Chodak A, et al. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: a review, Eur J Nutr, 54(3):325-41 (2015) .
[60] Pasolli E, et al., Extensive Unexplored Human Microbiome Diversity Revealed by Over 150,000 Genomes from Metagenomes Spanning Age, Geography, and Lifestyle, Call,
176(3):649-664 (2019).
[61] King CH, et al. Baseline human gut microbiota profile in healthy people and standard reporting template. PLoS ONE 14(9): e0206484. (2019).
[62] Ranjan R, et al.Multiomic Strategies Reveal Diversity and Important Functional Aspects of Human Gut Microbiome. Biomed Res Int.;6074918. (2018).
[63] Massot-Cladera, et al.Gut Health-Promoting Benefits of a Dietary Supplement of Vitamins with Inulin and Acacia Fibers in Rats. Nutrients.12(8):2196.(2020).
[64] Cronin P, et al. Dietary Fibre Modulates the Gut Microbiota. Nutrients.13, 1655.(2021).
[65] Yang, et al.The effects of psyllium husk on gut microbiota composition and function in chronically constipated women of reproductive age using 16S rRNA gene sequencing analysis.
Aging (Albany NY).;13(11):15366-15383.(2021).
[66] Fu, et al. Associations among Dietary Omega-3 Polyunsaturated Fatty Acids, the Gut Microbiota, and Intestinal Immunity.Mediators of Inflammation (2021).
[67] Wawrzyniak P, et al. Nutritional Lipids and Mucosal Inflammation. Mol. Nutr. Food Res, 65, 1901269.(2021).
[68] Tu, et al. Characterization of the Functional Changes in Mouse Gut Microbiome Associated with Increased Akkermansia muciniphila Population Modulated by Dietary Black
Raspberries.American Chemical Society (2018).
[69] Jin, et al. Effects of green tea consumption on human fecal microbiota with special reference to Bifidobacterium species. Microbiology and Immunology, 56: 729-739 (2012).
[70] Arumugam M, et al. Enterotypes of the human gut microbiome. Nature 473, 174 (2011).
[71] Wu GD, et al. Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes. Science 334, 105–108 (2011).
[72] Costea PI, et al. Enterotypes in the landscape of gut microbial community composition. Nat Microbiol 3, 8–16 (2018).
[73] Ley RE, et al. Gut microbiota in 2015: Prevotella in the gut: choose carefully. Nat Rev Gastroenterology Hepatology 13, 69–70 (2016).
[74] Ley RE, et al. Microbial ecology: Human gut microbes associated with obesity. Nature 444, 1022 (2006).
[75] Schwiertz A, et al. Microbiota and SCFA in Lean and Overweight Healthy Subjects. Obesity 18, 190–195 (2010).
[76] Whisner CM, et al. Diet, physical activity and screen time but not body mass index are associated with the gut microbiome of a diverse cohort of college students living in university
housing: a cross-sectional study. BMC Microbiol.;18(1):210 (2018).
[77] Langille MG, et al. Predictive functional proling of microbial communities using 16S rRNA marker gene sequences. Nature Biotechnology 31:814–821 (2013).
[78] Gao J, et al. Predictive functional proling using marker gene sequences and community diversity analyses of microbes in full-scale anaerobic sludge digesters. Bioprocess Biosyst Eng.
39(7):1115-27 (2016).
[79] Chen L, et al. Assessment of Bacterial Communities and Predictive Functional Proling in Soils Subjected to Short-Term Fumigation- Incubation. Microb Ecol. 72(1):240-251 (2016).
[80] Hjorth MR, et al. Pre-treatment microbial Prevotella-to-Bacteroides ratio, determines body fat loss success during a 6-month randomized controlled diet intervention. Int J Obes
(Lond). 42(3):580-583 (2018).
[81] Vital M, et al. Metagenomic Insights into the Degradation of Resistant Starch by Human Gut Microbiota. Appl Environ Microbiol. 84(23) (2018).
[82] Alfa MJ, et al. A randomized trial to determine the impact of a digestion resistant starch composition on the gut microbiome in older and mid-age adults, Clinical Nutrition 37, 797e807
(2018) .
[83] Braune A, et al, Bacterial species involved in the conversion of dietary flavonoids in the human gut, Gut Microbes. 7(3):216-34 (2016).
[84] Aherne SA, et al. Dietary avonols: chemistry, food content, and metabolism, Nutrition;18(1):75-81 (2002) .
[85] Xu Y, et al, Coenzyme Q10 Improves Lipid Metabolism and Ameliorates Obesity by Regulating CaMKII-Mediated PDE4 Inhibition, Sci Rep.;7(1):8253 (2017).
[86] Samiento A, et al. Coenzyme Q10 Supplementation and Exercise in Healthy Humans: A Systematic Review, Curr Drug Metab.;17(4):345-58 (2016).
[87] Merra G, et al. Influence of Mediterranean Diet on Human Gut Microbiota. Nutrients. (2020).
[88] Machate DJ, et al. Fatty Acid Diets: Regulation of Gut Microbiota Composition and Obesity and Its Related Metabolic Dysbiosis. International Journal of Molecular Sciences.
21(11):4093 (2020).
[89] Boschmann M, et al. Water Drinking Induces Thermogenesis through Osmosensitive Mechanisms, The Journal of Clinical Endocrinology & Metabolism, Volume 92, Issue 8, Pages
3334–3337 (2007).
[90] Vij VA, et al. Effect of 'water induced thermogenesis' on body weight, body mass index and body composition of overweight subjects. J Clin Diagn Res.;7(9):1894-6. (2013).
[91] Dinh, et al. The effects of green tea on lipid metabolism and its potential applications for obesity and related metabolic disorders - An existing update.Diabetes & Metabolic Syndrome:
Clinical Research & Reviews 13. (2019).
[92] Moon, et al. Clinical Evidence and Mechanisms of High-Protein Diet-Induced Weight Loss. JOMES;29:166-173. (2020).
[93] Dietger M, et al. Energieverbrauch I – Grundumsatz im Fit und gesund von 1 bis Hundert: Ernährung und Bewegung - Aktuelles medizinisches Wissen zur Gesundheit.Springer Berlin
Heidelberg. (2018).
[94] Elmadfa. Ernährungslehre. (2015).
[95] Lassen P, et al. Protein supplementation during an energy-restricted diet induces visceral fat loss and gut microbiota amino acid metabolism activation: a randomized trial. Sci Rep 11,
15620 (2021).
[96] Hjorth MF, et al. Pretreatment Prevotella-to-Bacteroides ratio and markers of glucose metabolism as prognostic markers for dietary weight loss maintenance. Eur J Clin Nutr 74, 338–
347 (2020).
[97] Covasa M, et al. Intestinal Sensing by Gut Microbiota: Targeting Gut Peptides. Front. Endocrinol. (2019).
[98] Fachgesellschaft für Ernährungstherapie und Prävention (FET) eV (Zugriff 12.08.2021).
[99] Forslund K, et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 528, 262 (2015).
[100] Everard A, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc National Acad Sci 110, 9066–9071 (2013).
[101] Dao M, et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 65, 426
(2016).
[102] Schneeberger M, et al. Akkermansia muciniphila inversely correlates with the onset of inammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice.
Sci Rep-uk 5, srep16643 (2015).
[103] Raman M, et al. Fecal Microbiome and Volatile Organic Compound Metabolome in Obese Humans With Nonalcoholic Fatty Liver Disease. Clin Gastroenterol H 11, 868-875.e3 (2013).
[104] Del Chierico F, et al. Gut Microbiota Markers in Obese Adolescent and Adult Patients: Age-Dependent Differential Patterns. Front Microbiol. 9:1210 (2018).
[105] Haro C, et al. Intestinal Microbiota Is Inuenced by Gender and Body Mass Index. Plos ONe 11, e0154090 (2016).
[106] Zhernakova A, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352, 565–569 (2016).
[107] Haro C, et al. The gut microbial community in metabolic syndrome patients is modied by diet. J Nutr Biochem. 27:27-31 (2016).
[108] Verdam FJ, et al. Human intestinal microbiota composition is associated with local and systemic inammation in obesity. Obesity 21, E607–E615 (2013).
[109] Murri M, et al. Gut microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. Bmc Med 11, 46 (2013).
[110] Xu J, et al. Evolution of symbiotic bacteria in the distal human intestine. PLoS Biol. 5(7):e156 (2007).
[111] Medina-Vera I, et al. A dietary intervention with functional foods reduces metabolic endotoxaemia and attenuates biochemical abnormalities by modifying faecal microbiota in people
with type 2 diabetes. Diabetes Metab.: S1262-3636(18)30175-7 (2018).
[112] Zupancic ML, et al. Analysis of the Gut Microbiota in the Old Order Amish and Its Relation to the Metabolic Syndrome. Plos One 7, e43052 (2012).
[113] Maslowski KM, et al. Regulation of inammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 461, 1282 (2009).
[114] Wang K, et al. Parabacteroides distasonis Alleviates Obesity and Metabolic Dysfunctions via Production of Succinate and Secondary Bile Acids. Cell Rep.26(1):222-235 (2019).
[115] Clarke SF, et al. The gut microbiota and its relationship to diet and obesity. Gut Microbes 3, 186–202 (2012).
[116] Santacruz A, et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Brit J Nutr 104, 83–92 (2010).
[117] Niccolai E, et al. The Gut–Brain Axis in the Neuropsychological Disease Model of Obesity: A Classical Movie Revised by the Emerging Director “Microbiome”. Nutrients 11:156 (2019).
[118] Heianza Y, et al. Changes in Gut Microbiota-Related Metabolites and Long-term Successful Weight Loss in Response to Weight- Loss Diets: The POUNDS Lost Trial. Diabetes Care.
41(3):413-419 (2018).
[119] Bortolin RC, et al. Guarana supplementation attenuated obesity, insulin resistance, and adipokines dysregulation induced by a standardized human Western diet via brown adipose
tissue activation. Phytother Res. 33(5):1394-1403 (2019).
[120] De Filippo C, et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europeand rural Africa.Proc. Natl. Acad. Sci. USA, 107, 14691–14696
(2010).
[121] Yeoh YK, et al. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut.;70(4):698 (2021).
[122] Gu S, et al. Alterations of the Gut Microbiota in Patients With Coronavirus Disease 2019 or H1N1 Influenza. Clinical Infectious Diseases.;71(10):2669-78 (2020).
[123] Atarashi K, et al. Induction of colonic regulatory T cells by indigenous Clostridium species.Science (2011).
[124] Zuo T, et al. Alterations in Gut Microbiota of Patients With COVID-19 During Time of Hospitalization. Gastroenterology.;159(3):944-55.e8 (2020).
[125] Tao W, et al. Analysis of the intestinal microbiota in COVID-19 patients and its correlation with the inflammatory factor IL-18. Medicine in Microecology.;5:100023 (2020).
[126] Alameddine J, et a. Faecalibacterium prausnitzii Skews Human DC to Prime IL10-Producing T Cells Through TLR2/6/JNK Signaling and IL-10, IL-27, CD39, and IDO-1 Induction.
Frontiers in immunology.;10:143 (2019).
[127] Chattopadhyay I, et al. SARS-CoV-2-Indigenous Microbiota Nexus: Does Gut Microbiota Contribute to Inflammation and Disease Severity in COVID-19? Front Cell Infect
Microbiol.;11:590874 (2021).
[128] Hiippala K, et al. Isolation of Anti-Inflammatory and Epithelium Reinforcing Bacteroides and Parabacteroides Spp. from A Healthy Fecal Donor. Nutrients.;12(4):935 (2020).
[129] Sánchez-Alcoholado L, et al. Gut Microbiota-Mediated Inflammation and Gut Permeability in Patients with Obesity and Colorectal Cancer. International journal of molecular
sciences.;21(18) (2020).
[130] Kaakoush NO, et al. Sutterella Species, IgA-degrading Bacteria in Ulcerative Colitis. Trends in Microbiology.;28(7):519-22 (2020).
[131] Wang J, et al. The Relationship Between Gut Microbiota and Inflammatory Diseases: The Role of Macrophages.;11(1065) (2020).
[132] Zhai R, et al. Strain-Specific Anti-inflammatory Properties of Two Akkermansia muciniphila Strains on Chronic Colitis in Mice. Front Cell Infect Microbiol.;9:239 (2019).
[133] Liu JH, et al. Akkermansia muciniphila promotes type H vessel formation and bone fracture healing by reducing gut permeability and inflammation. Disease Models &
Mechanisms.;13(11) (2020).
[134] Charlet R, et al. Bacteroides thetaiotaomicron and Lactobacillus johnsonii modulate intestinal inflammation and eliminate fungi via enzymatic hydrolysis of the fungal cell wall.
Scientific Reports.;10(1):11510 (2020).
[135] Sun J, et al. Anti-inflammatory properties and gut microbiota modulation of an alkali-soluble polysaccharide from purple sweet potato in DSS-induced colitis mice. International Journal
of Biological Macromolecules.;153:708-22 (2020).
[136] Baldelli V, et al. The Role of Enterobacteriaceae in Gut Microbiota Dysbiosis in Inflammatory Bowel Diseases. Microorganisms.;9(4) (2021).
[137] Derrien M, et al. Akkermansia muciniphila and its role in regulating host functions. Microbial Pathogenesis.;106:171-81 (2017).
[138] Hiippala K, et al. The Potential of Gut Commensals in Reinforcing Intestinal Barrier Function and Alleviating Inflammation. Nutrients.;10(8) (2018).
[139] Zhang L, et al. Akkermansia muciniphila can reduce the damage of gluco/lipotoxicity, oxidative stress and inflammation, and normalize intestine microbiota in streptozotocin-induced
diabetic rats. Pathogens and disease.;76(4) (2018).
[140] Delday M, et al. Bacteroides thetaiotaomicron Ameliorates Colon Inflammation in Preclinical Models of Crohn's Disease. Inflammatory bowel diseases.;25(1):85-9 (2019).
[141] Zhaoyan L, et al. Antioxidant and Anti-Inflammatory Properties of Recombinant Bifidobacterium bifidum BGN4 Expressing Antioxidant Enzymes.Microorganisms.Volume 9 (2021).
[142] Tang L, et al. Clinical Significance of the Correlation between Changes in the Major Intestinal Bacteria Species and COVID-19 Severity. Engineering.;6(10):1178-84 (2020).
[143] Menezes-Garcia Z, et al. Colonization by Enterobacteriaceae is crucial for acute inflammatory responses in murine small intestine via regulation of corticosterone production, Gut
Microbes.(2020) 11:6, 1531-1546 (2020).
[144] Henke, et al.Capsular polysaccharide correlates with immune response to the human gut microbe Ruminococcus gnavus. Proceedings of the National Academy of Sciences May
(2021).
[145] Körner U, et al. Nahrungsmittelallergien und -unverträglichkeiten: Diagnostik, Therapie und Beratung: Georg Thieme Verlag KG (2020).
[146] Mullin GE, et al. Integrative gastroenterology. New York: Oxford University Press (2011).
[147] Mitchell AE, et al. Ten-year comparison of the influence of organic and conventional crop management practices on the content of flavonoids in tomatoes. J Agric Food
Chem.;55(15):6154-9 (2007).
[148] Schmiedel, et al. Europäisches Arzneibuch 8. Ausgabe, 8. Nachtrag (Ph. Eur. 8.8): Amtliche deutsche Ausgabe: Deutscher Apotheker Verlag;(2016).
[149] Wong C, et al. Potential Benefits of Dietary Fibre Intervention in Inflammatory Bowel Disease. International journal of molecular sciences.;17(6) (2016).
[150] Felson DT, et al. Dietary fatty acids for the treatment of OA, including fish oil. Ann Rheum Dis.;75(1):1-2 (2016).
[151] Biesalski HK,et al. Ernährungsmedizin: nach dem neuen Curriculum Ernährungsmedizin der Bundesärztekammer;276 Tabellen: Thieme;(2010).
[152] Zhao Y, et al. Intestinal microbiota and chronic constipation. SpringerPlus.;5(1):1130 (2016).
[153] Höfler E, et al. Praktische Diätetik: Grundlagen, Ziele und Umsetzung der Ernährungstherapie;mit 205 Tabellen sowie 141 Übungsaufgaben: Wiss. Verlag-Ges.;(2012).
[154] Österreichische Gesellschaft für Ernährung. Vitamine, Mineralstoffe, Spurenelemente. (2019).
[155] Puchner R, et al. Rheumatologie aus der Praxis: Entzündliche Gelenkerkrankungen – mit Fallbeispielen: Springer Berlin Heidelberg;(2017).
[156] Innes JK, et al. Omega-6 fatty acids and inflammation. Prostaglandins, leukotrienes, and essential fatty acids.;132:41-8 (2018).
[157] Valacchi G, et al. The dual action of ozone on the skin. The British journal of dermatology.;153(6):1096-100 (2005).
[158] Magnani ND, et al. Skin Damage Mechanisms Related to Airborne Particulate Matter Exposure. Toxicological Sciences.;149(1):227-36 (2015).
[159] Riccio P, et al. Undigested Food and Gut Microbiota May Cooperate in the Pathogenesis of Neuroinflammatory Diseases: A Matter of Barriers and a Proposal on the Origin of Organ
Specificity. Nutrients 11, 2714 (2019).
[160] Calder PC, et al. Feeding the immune system. The Proceedings of the Nutrition Society.;72(3):299-309.(2013).
[161] Dosch SF, et al. SARS coronavirus spike protein-induced innate immune response occurs via activation of the NF-kappaB pathway in human monocyte macrophages in vitro. Virus
Res;142(1-2):19-27. (2009).
[162] Wang Y, et al.The Membrane Protein of Severe Acute Respiratory Syndrome Coronavirus Functions as a Novel Cytosolic Pathogen-Associated Molecular Pattern To Promote Beta
Interferon Induction via a Toll-Like-Receptor-Related TRAF3-Independent Mechanism. mBio.;7(1):e01872-15.(2016).
[163] Al-Qahtani AA, et al. Middle east respiratory syndrome corona virus spike glycoprotein suppresses macrophage responses via DPP4-mediated induction of IRAK-M and PPARγ.
Oncotarget.;8(6):9053-66.(2017).
[164] Hsieh, et al. Association between dietary flavonoid intakes and C-reactive protein levels: A cross-sectional study in Taiwan. Journal of Nutritional Science, 10, E15. (2021).
[165] Asgharpour, et al. Efficacy of Oral Administration of Allium sativum Powder “Garlic Extract” on Lipid Profile, Inflammation, and Cardiovascular Indices among Hemodialysis
Patients.Evidence-Based Complementary and Alternative Medicine (2021).
[166] Azeez, et al. 6 - Antiinflammatory effects of turmeric (Curcuma longa) and ginger (Zingiber officinale).Inflammation and Natural Products,Academic Press,Pages 127-146 (2021).
[167] Zhang, et al. Butyrate in Energy Metabolism: There Is Still More to Learn.Trends in Endocrinology & Metabolism,Volume 32, Issue 3. (2021).
[168] Mattiuzzo E, et al. In Vitro Effects of Low Doses of β-Caryophyllene, Ascorbic Acid and d-Glucosamine on Human Chondrocyte Viability and Inflammation. Pharmaceuticals. (2021).
[169] Demirel-Yalciner, et al. alpha-Tocopherol supplementation reduces inflammation and apoptosis in high cholesterol mediated nonalcoholic steatohepatitis. BioFactors.). 47: 403– 413.
(2021).
[170] Fu, et al. Low Vitamin D Status Is Associated with Inflammation in Patients with Chronic Obstructive Pulmonary Disease. The Journal of Immunology February 1, 206 (3) 515-523
(2021).
[171] Serrano D, et al. Microbiome as Mediator of Diet on Colorectal Cancer Risk: The Role of Vitamin D, Markers of Inflammation and Adipokines. Nutrients. 13(2):363. (2021).
[172] Shibabaw T, et al. Omega-3 polyunsaturated fatty acids: anti-inflammatory and anti-hypertriglyceridemia mechanisms in cardiovascular disease. Mol Cell Biochem 476, 993–1003
(2021).
[173] Mariamenatu, et al. Overconsumption of Omega-6 Polyunsaturated Fatty Acids (PUFAs) versus Deficiency of Omega-3 PUFAs in Modern-Day Diets: The Disturbing Factor for Their
“Balanced Antagonistic Metabolic Functions” in the Human Body. Hindawi. (2021).
[174] Cox S, et al. Food Matters Food-additive emulsifiers: the worst thing since sliced bread? The Lancet Gastroenterology & Hepatology, Volume 6, Issue 7, 532 (2021).
[175] Li K, et al. Bacteroides thetaiotaomicron relieves colon inflammation by activating aryl hydrocarbon receptor and modulating CD4+T cell homeostasis, International
Immunopharmacology, Volume 90, (2021).
[176] Pang W, et al. Bacteroides thetaiotaomicron Ameliorates Experimental Allergic Airway Inflammation via Activation of ICOS+Tregs and Inhibition of Th2 Response .Frontiers in
Immunology .VOLUME 12. (2021).
[177] Quévrain E, et al. Isentification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn’s diseaseGut;65:415-(2016).
[178] Ghosh S, et al. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cellular and Molecular Gastroenterology and Hepatology.;11(5):1463-82 (2021).
[179] Lenoir M, et al. Butyrate mediates anti-inflammatory effects of Faecalibacterium prausnitzii in intestinal epithelial cells through Dact3. Gut microbes.;12(1):1826748 (2020).
[180] Deutsche Gesellschaft für Ernährung. Referenzwerte für die Nährstoffzufuhr D-A-CH: Deutsche Gesellschaft für Ernährung;(2018).
[181] Groschwitz KR, et al. Intestinal barrier function: molecular regulation and disease pathogenesis. J Allergy Clin Immunol 124(1):3–22. https://doi.org/10.1016/j.jaci (2009).
[182] Mu Q, et al. Leaky Gut As a Danger Signal for Autoimmune Diseases. Front Immunol;8:598 (2017).
[183] Zielińska A, et al. Nowak I. Abundance of active ingredients in sea-buckthorn oil. Lipids in Health and Disease.;16 (2017).
[184] Macia L, et al. Microbial influences on epithelial integrity and immune function as a basis for inflammatory diseases. Immunological reviews.;245(1):164-76. (2012).
[185] Kelly CJ, et al. Crosstalk between Microbiota-Derived Short-Chain Fatty Acids and Intestinal Epithelial HIF Augments Tissue Barrier Function. Cell host & microbe.;17(5):662-71
(2015).
[186] Viennois E, et al. First victim, later aggressor: How the intestinal microbiota drives the pro-inflammatory effects of dietary emulsifiers? Gut microbes.9(3):1-4 (2018).
[187] Sicard JF, et al. Interactions of Intestinal Bacteria with Components of the Intestinal Mucus. Front Cell Infect Microbiol;7:387 (2017).
[188] Mullin GE, et al. Integrative gastroenterology. New York: Oxford University Press;(2011).
[189] Patrick RP, et al. Vitamin D and the omega-3 fatty acids control serotonin synthesis and action, part 2: relevance for ADHD, bipolar disorder, schizophrenia, and impulsive behavior.
FASEB journal : official publication of the Federation of American Societies for Experimental Biology,;29(6):2207-22 (2015).
[190] Barreau F, et al. Hugot J, Intestinal barrier dysfunction triggered by invasive bacteria. Curr Opin Microbiol 17, 91–98 (2014).
[191] Wang L, et al. Intestinal REG3 lectins protect against alcoholic steatohepatitis by reducingmucosa-associated microbiota and preventing bacterial translocation. Cell Host Microbe, 19,
227–239 (2016).
[192] Chen, et al. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Medicine 8:43 (2016).
[193] Von Ossowski I, et al. Mucosal Adhesion Properties of the Probiotic Lactobacillus rhamnosus GG SpaCBA and SpaFED Pilin Subunits. Appl Environ Microb 76, 2049–2057 (2010).
[194] Kinashi, et al .Partners in Leaky Gut Syndrome: Intestinal Dysbiosis and Autoimmunity. Front. Immunol. 12:673708 (2021).
[195] Kishino S, et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proc Natl Acad Sci USA;110(44):17808-13 (2013).
[196] He C, et al. Vitamin A inhibits the action of LPS on the intestinal epithelial barrier function and tight junction proteins. Food Funct.10, 1235–1242 (2019).
[197] Riccio P, et al. Diet, gutmicrobiota, and vitamins A+D, in multiple sclerosis. Review. Neurotherapeutics 15, 75-91 (2018).
[198] Zhu W, et al. 1,25(OH)2D3 deficiency-induced gut microbial dysbiosis degrades the colonic mucus barrier in Cyp27b1 knockout mouse modl. Gut Pathog. 11 8 (2019).
[199] Cantorna MT, et al. Vitamin A and vitamin D regulate the microbial complexity, barrier function, and the mucosal immune responses to ensure intestinal homeostasis. Crit. Rev.
Biochem. Mol. Biol, 54, 184–192 (2019).
[200] Madeo, et al. Spermidine in health and disease. Science (2018).
[201] Ma, et al. Spermidine improves gut barrier integrity and gut microbiota function in diet-induced obese mice, Gut Microbes, 12:1, 1832857 (2020).
[202] Balakrishnan, et al. Autoimmunity-Associated Gut Commensals Modulate Gut Permeability and Immunity in Humanized Mice, Military Medicine, Volume 184, Issue Supplement_1,
Pages 529–536 (2019).
[203] Yamada T, et al. Mucin O-glycans facilitate symbiosynthesis to maintain gut immune homeostasis. EBioMedicine. 48:513-525. (2019).
[204] Hiippala K, et al. Mucosal Prevalence and Interactions with the Epithelium Indicate Commensalism of Sutterella spp. Front Microbiol 7, 1706 (2016).
[205] Tana C, et al.Altered profiles of intestinal microbiota and organic acids may be the origin of symptoms in irritable bowel syndrome.Neurogastroenterol Motil. May;22(5):512-9, e114-5
(2010).
[206] Zhang et al.Primary Human Colonic Mucosal Barrier Crosstalk with Super Oxygen-Sensitive Faecalibacterium prausnitzii in Continuous Culture (2021).
[207] Crost EH, et al. Utilisation of mucin glycans by the human gut symbiont Ruminococcus gnavus is strain-dependent. PLoS ONE 8:e76341. (2013)
[208] Roberts JL, et al.Bifidobacterium adolescentis supplementation attenuates fracture-induced systemic sequelae. Biomed Pharmacother (2020).
[209] Van der Lugt, et al. Akkermansia muciniphila ameliorates the age-related decline in colonic mucus thickness and attenuates immune activation in accelerated aging Ercc1−/Δ7 mice.
Immun Ageing 16, 6 (2019).
[210] Ouyang J, et al. The Bacterium Akkermansia muciniphila: A Sentinel for Gut Permeability and Its Relevance to HIV-Related Inflammation. Front. Immunol. 11:645 (2020).
[211] Huck O, et al. Akkermansia muciniphila reduces Porphyromonas gingivalis-induced inflammation and periodontal bone destruction. J Clin Periodontol.;47: 202– 212 (2020).
[212] Cruz-Aguliar RM, et al. An Open-Labeled Study on Fecal Microbiota Transfer in Irritable Bowel Syndrome Patients Reveals Improvement in Abdominal Pain Associated with the
Relative Abundance of Akkermansia Muciniphila. Digestion 1–12 (2018).
[213] Png CW, et al. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am J Gastroenterol., 105(11):2420-8 (2010).
[214] Liu Y, et al. Dietary quality and the colonic mucosa-associated gut microbiome in humans. Am J Clin Nutr.;110(3):701-712 (2019)
[215] Fanning S. et al. Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection. Proc Natl Acad Sci U S A.
(2012).
[216] Coyne MJ, et al. Human Symbionts Use a Host-Like Pathway for Surface Fucosylation. Science 307, 1778–1781 (2005).
[217] Grander C, et al.. Recovery of ethanol-induced Akkermansia muciniphila depletion ameliorates alcoholic liver disease. Gut. 2018 May;67(5):891-901 (2017).
[218] Lapiere, et al. Prophylactic Faecalibacterium prausnitzii treatment prevents the acute breakdown of colonic epithelial barrier in a preclinical model of pelvic radiation disease, Gut
Microbes, 12:1 (2020).
[219] Xu J, et al. Faecalibacterium prausnitzii-derived microbial anti-inflammatory molecule regulates intestinal integrity in diabetes mellitus mice via modulating tight junction protein
expression. Journal of Diabetes (2020).
[220] Duncan SH, et al. Proposal of Roseburia faecis sp. nov., Roseburia hominis sp. nov. and Roseburia inulinivorans sp. nov., based on isolates from human faeces. Int. J. Syst. Evol.
Microbiol. 56, 2437–2441 (2006).
[221] Louis P, et al. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 19, 29–41 (2017).
[222] Lopez-Siles M, et al. Mucosa-associated Faecalibacterium prausnitzii phylotype richness is reduced in patients with inflammatory bowel disease. Appl Environ Microbiol.(2015).
[223] Miquel S, et al. Identification of Metabolic Signatures Linked to Anti-Inflammatory Effects of Faecalibacterium prausnitzii (2015).
[224] Graziani F, et al. Ruminococcus gnavus E1 modulates mucin expression and intestinal glycosylation. J Appl Microbiol, 120: 1403-1417. (2016).
[225] Qin P, et al. Characterization a Novel Butyric Acid-Producing Bacterium Collinsella aerofaciens Subsp. Shenzhenensis Subsp. Nov. Microorganisms (2019).
[226] Marietta EV, et al. Suppression of inflammatory arthritis by human gut-derived Prevotella histicola in humanized mice Arthritis Rheumatol., 68 (2016)
[227] Astbury, et al. Lower gut microbiome diversity and higher abundance of proinflammatory genus Collinsella are associated with biopsy-proven nonalcoholic steatohepatitis, Gut
Microbes, 11:3, 569-580 (2020).
[228] Achamrah N, et al. Glutamine and the regulation of intestinal permeability: from bench to bedside. Curr Opin Clin Nutr Metab Care. 20(1):86-91. (2017).
[229] Burrin DG, et al. Metabolic fate and function of dietary glutamate in the gut. Am J Clin Nutr. 90(3):850S-856S (2009).
[230] David LA, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505, 559 (2014).
[231] Huang JY,et al. The human commensal Bacteroides fragilis binds intestinal mucin. Anaerobe.17(4):137-41 (2011).
[232] Swidsinski A, et al. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J Clin Microbiol. 43(7):3380-9. (2005).
[233] Brunkwall L, et al. Self-reported bowel symptoms are associated with differences in overall gut microbiota composition and enrichment of Blautia in a population-based cohort.
Journal of gastroenterology and hepatology.;36(1):174-80 (2021).
[234] Xu C, et al. Multiomics Study of Gut Bacteria and Host Metabolism in Irritable Bowel Syndrome and Depression Patients.;10(660) (2020).
[235] Matsumoto H, et al. Mucosa-Associated Microbiota in Patients with Irritable Bowel Syndrome: A Comparison of Subtypes. Digestion.;102(suppl 1)(1):49-56 (2021).
[236] Lopetuso LR, et al. Towards a disease-associated common trait of gut microbiota dysbiosis: The pivotal role of Akkermansia muciniphila. Digestive and Liver Disease.;52(9):1002-10
(2020).
[237] Paulina P, et al. Qualitative Identification of Roseburia hominis in Faeces Samples Obtained from Patients with Irritable Bowel Syndrome and Healthy Individuals. Proceedings.;66(1)
(2020).
[238] O’Toole PW, et al. Next-generation probiotics: The spectrum from probiotics to live biotherapeutics. Nat. Microbiol.2:17057 (2017).
[239] Masoodi I, et al. Microbial dysbiosis in irritable bowel syndrome: A single-center metagenomic study in Saudi Arabia. JGH Open.;4(4):649-55 (2020).
[240] Agnello M, et al. Gut microbiome composition and risk factors in a large cross-sectional IBS cohort. BMJ Open Gastroenterology.;7(1):e000345 (2020).
[241] Sciavilla P, et al. Gut microbiota profiles and characterization of cultivable fungal isolates in IBS patients. Applied Microbiology and Biotechnology.;105(8):3277-88 (2021).
[242] Rajilić-Stojanović M, et al. Intestinal Microbiota And Diet in IBS: Causes, Consequences, or Epiphenomena? Am J Gastroenterology 110, 278 (2015).
[243] Yang M, et al. Mucosal-Associated Microbiota Other Than Luminal Microbiota Has a Close Relationship With Diarrhea-Predominant Irritable Bowel Syndrome.;10(606) (2020).
[244] Enck P, et al. Dysbiosis in Functional Bowel Disorders. Ann Nutr Metab.72(4):296-306 (2018).
[245] Pittayanon R, et al. Gut Microbiota in Patients With Irritable Bowel Syndrome—A Systematic Review.Gastroenterology, Volume 157, Issue 1, 97 - 108 (2019).
[246] Barandouzi Z, et al. Altered Gut Microbiota in Irritable Bowel Syndrome and Its Association with Food Components. J. Pers. Med, 11, 35 (2021).
[247] Mei L, et al. Gut microbiota composition and functional prediction in diarrhea-predominant irritable bowel syndrome. BMC Gastroenterol 21,.105 (2021).
[248] Fodor A, et al. Rifaximin is associated with modest, transient decreases in multiple taxa in the gut microbiota of patients with diarrhoea-predominant irritable bowel syndrome, Gut
Microbes 10:1, 22-33 (2019).
[249] Vich Vila A, et al. Gut microbiota composition and functional changes in inflammatory bowel disease and irritable bowel syndrome.Sci. Transl. Med. 10, eaap8914 (2018).
[250] Peter J, et al. A Microbial Signature of Psychological Distress in Irritable Bowel Syndrome, Psychosom Med., 80(8): 698–709 (2018).
[251] Labus JS, et al. Evidence for an association of gut microbial Clostridia with brain functional connectivity and gastrointestinal sensorimotor function in patients with irritable bowel
syndrome, based on tripartite network analysis. Microbiome, 7:45 (2019).
[252] Lopetuso LR, et al. Gut Microbiota in Health, Diverticular Disease, Irritable Bowel Syndrome, and Inflammatory Bowel Diseases: Time for Microbial Marker of Gastrointestinal
Disorders.Dig Dis. 36(1):56-65 (2018).
[253] Frank DN, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases Jan;17(1):179-84 (2011).
[254] Kassinen A, et al.The fecal microbiota of irritable bowel syndrome patients differs significantly from that of healthy subjects. Gastroenterology. Jul;133(1):24-33 (2007).
[255] Lyra A, et al. Diarrhoea-predominant irritable bowel syndrome distinguishable by 16S rRNA gene phylotype quantification. World J Gastroenterol. Dec 21;15(47):5936-45 (2009).
[256] Hynönen U, et al. Isolation and whole genome sequencing of a Ruminococcus-like bacterium, associated with irritable bowel syndrome. Anaerobe, 39, 60-67 (2016).
[257] Salonen A, et al. Gastrointestinal microbiota in irritable bowel syndrome: present state and perspectives. Microbiology, 156(11), 3205–3215 (2010).
[258] Malinen E, et al. Analysis of the Fecal Microbiota of Irritable Bowel Syndrome Patients and Healthy Controls with Real-Time PCR. Am J Gastroenterology 100, ajg200561 (2005).
[259] Rajilić–Stojanović M, et al. Global and Deep Molecular Analysis of Microbiota Signatures in Fecal Samples From Patients With Irritable Bowel Syndrome. Gastroenterology 141, 1792–
1801 (2011).
[260] Kerckhoffs AP, Samsom M, van der Rest ME, de Vogel J, Knol J, Ben-Amor K, Akkermans LM. Lower Bifidobacteria counts in both duodenal mucosa-associated and fecal microbiota in
irritable bowel syndrome patients. World J Gastroenterol. Jun 21;15(23):2887-92 (2009).
[261] Mazzawi T, et al. The kinetics of gut microbial community composition in patients with irritable bowel syndrome following fecal microbiota transplantation. PLoS ONE 13(11):
e0194904 (2018).
[262] Jalanka-Tuovinen J, et al. Faecal microbiota composition and host–microbe cross-talk following gastroenteritis and in postinfectious irritable bowel syndrome. Gut 63, 1737 (2014).
[263] Rizzello F, Dietary geraniol ameliorates intestinal dysbiosis and relieves symptoms in irritable bowel syndrome patients: a pilot study. BMC Complement Altern Med. 18: 338 (2018).
[264] Sokol H, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. Oct
28;105(43):16731-6 (2008).
[265] Hod K, et al. The effect of a multispecies probiotic on microbiota composition in a clinical trial of patients with diarrhea predominant irritable bowel syndrome. Neurogastroenterol
Motil. 30(12):e13456 (2018).
[266] Lopes-Siles M, et al. lterations in the Abundance and Co-occurrence of Akkermansia muciniphila and Faecalibacterium prausnitzii in the Colonic Mucosa of Inflammatory Bowel
Disease Subjects. Frontiers in Cellular and Infection Microbiology.;8:281 (2018).
[267] Carroll IM, et al. Alterations in composition and diversity of the intestinal microbiota in patients with diarrhea-predominant irritable bowel syndrome. Neurogastroenterol Motil.
24(6):521-30, e248 (2012).
[268] Costabile A, et al. Effect of Breadmaking Process on In Vitro Gut Microbiota Parameters in Irritable Bowel Syndrome. PLOS ONE.;9(10):e111225 (2014).
[269] Ziegler JU, et al.Wheat and the irritable bowel syndrome – FODMAP levels of modern and ancient species and their retention during bread making. Journal of Functional
Foods;25:257-66 (2016).
[270] Williams CE, et al.Vitamin D status in irritable bowel syndrome and the impact of supplementation on symptoms: what do we know and what do we need to know? European journal
of clinical nutrition.;72(10):1358-63 (2008).
[271] Jonsson AL, et al. Role of gut microbiota in atherosclerosis. Nat Rev Cardiol. 14(2):79-87 (2017).
[272] Luedde M, et al. Heart failure is associated with depletion of core intestinal microbiota. ESC Heart Fail. 4, 282–290 (2017).
[273] Pedersen N, et al. Ehealth: Low FODMAP diet vs Lactobacillus rhamnosus GG in irritable bowel syndrome, Gastroenterology 20:43 16215 (2014).
[274] Reddel S, et al. The Impact of Low-FODMAPs, Gluten-Free, and Ketogenic Diets on Gut Microbiota Modulation in Pathological Conditions. Nutrients. 11(2) (2019).
[275] Altobelli E, et al. Low-FODMAP Diet Improves Irritable Bowel Syndrome Symptoms: A Meta-Analysis. Nutrients 9(9) (2017).
[276] Schmidt T, et al. The Human Gut Microbiome: From Association to Modulation. Cell 172, 1198–1215 (2018).
[277] Jeffery IB,. et al. An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota. Gut 61, 997 (2012).
[278] Donner, et al. Serotonin: Zum Glück gibt´s was zum Essen.UGB-FORUM 5/05.S. 245-8.(2005).
[279] Wilkins CH, et al. Vitamin D deficiency is associated with low mood and worse cognitive performance in older adults. Am J Geriatr Psychiatry.;14(12):1032-40. (2006).
[280] Glabska, et al. The Influence of Vitamin D Intake and Status on Mental Health in Children: A Systematic Review. Nutrients , 13, 952. (2021).
[281] Tillisch K, et al. Consumption of fermented milk product with probiotic modulates brain activity. Gastroenterology. (2013).
[282] Van de Wouw, et al. Distinct actions of the fermented beverage kefir on host behaviour, immunity and microbiome gut-brain modules in the mouse Microbiome (2020)
[283] Mocking, et al.Meta-analysis and meta-regression of omega-3 polyunsaturated fatty acid supplementation for major depressive disorder.Translational psychiatry (2016).
[284] Trebatická J, et al. Omega-3 fatty-acids modulate symptoms of depressive disorder, serum levels of omega-3 fatty acids and omega-6/omega-3 ratio in children. A randomized,
double-blind and controlled trial. Psychiatry Research, Volume 287 (2020).
[285] Stengler M, et al. The Role of Folate and MTHFR Polymorphisms in the Treatment of Depression. Altern Ther Health Med.;27(2):53-57. PMID: 32827402. (2021).
[286] Mansoori A, e tal.. The Effects of Bariatric Surgery on Vitamin B Status and Mental Health. Nutrients 20;13(4):1383. (2021).
[287] Zheng P, et al.. Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism. Mol. Psychiatr.21, 786 (2016).
[288] Vacca M, et al. The Controversial Role of Human Gut Lachnospiraceae. Microorganisms.;8(4):573. (2020).
[289] Chen Z, et al. Comparative metaproteomics analysis shows altered fecal microbiota signatures in patients with major depressive disorder. Neuroreport.(2018).
[290] Cheng, et al. Identifying psychiatric disorder-associated gutmicrobiota using microbiota-related gene setenrichment analysis (2019).
[291] Yang, et al. Landscapes ofbacterial andmetabolic signatures andtheir interaction inmajor depressive disorders.SCIENCE ADVANCES. (2020).
[292] Barandouzi, et al. Altered Composition of Gut Microbiota in Depression: A Systematic Review.Frontiers in Psychiatry.(2020).
[293] Jianguo L,et al. Altered gut metabolome contributes to depression-like behaviors in rats exposed to chronic unpredictable mild stress. Transl Psychiatry 9, 40 (2019).
[294] Martin del Campo, et al. P1144 ODORIBACTER AND ANAEROTRUNCUS: GUT MICROBIOME SIGNATURE MIGHT BE RELATED TO COGNITIVE IMPAIRMENT IN PATIENTS ON
PERITONEAL DIALYSIS, Nephrology Dialysis Transplantation, Volume 35, Issue Supplement_3 (2020).
[295] Rong H,et al. Similarly in depression, nuances of gut microbiota: Evidences from a shotgun metagenomics sequencing study on major depressive disorder versus bipolar disorder with
current major depressive episode patients. Journal of Psychiatric Research (2019).
[296] Ma W, et al. Chronic paradoxical sleep deprivation-induced depression-like behavior, energy metabolism and microbial changes in rats. Life Sci. (2019).
[297] Liu, et al. Reductions in anti-inflammatory gut bacteria are associated with depression in a sample of young adults,Brain, Behavior, and Immunity (2020).
[298] Madan, et al. The gut microbiota is associated with psychiatric symptom severity and treatment outcome among individuals with serious mental illness. J. Affect. Disord. 264, 98–106.
(2020).
[299] Chen JJ, et al. Age-specific differential changes on gut microbiota composition in patients withmajor depressive disorder. Aging 12, 2764–2776. (2020).
[300] Liśkiewicz P, et al. Analysis of gut microbiota and intestinal integrity markers of inpatients with major depressive disorder. Prog Neuropsychopharmacol Biol Psychiatry (2021).
[301] Tian, et al.Towards a psychobiotic therapy for depression: Bifidobacterium breve CCFM1025 reverses chronic stress-induced depressive symptoms and gut microbial abnormalities in
mice,Neurobiology of Stress,Volume 12 (2020).
[302] Xie, et al.Oral treatment with Lactobacillus reuteri attenuates depressive-like behaviors and serotonin metabolism alterations induced by chronic social defeat stress,Journal of
Psychiatric Research,Volume 122, (2019).
[303] Kosuge, et al.Heat-sterilized Bifidobacterium breve prevents depression-like behavior and interleukin-1β expression in mice exposed to chronic social defeat stress,Brain, Behavior,
and Immunity,Volume 96, (2021).
[304] Han SK, et al. Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98 Alleviate Escherichia coli-Induced depression and Gut Dysbiosis in Mice. J Microbiol Biotechnol.
PMID: 32347078. (2020).
[305] Tian, et al. Unraveling the Microbial Mechanisms Underlying the Psychobiotic Potential of a Bifidobacterium breve Strain. Molecular Nutrition & Food Research. (2021).
[306] Simpson, et al.Bugs and Brains: The Gut and Mental Health Study Characterising the gut microbiota in anxiety, depression, irritable bowel syndrome, and their comorbidity. (2021).
[307] Valles-Colomer M, et al., The neuroactive potential of the human gut microbiota in quality of life and depression. Nature Microbiology, 4(4): p. 623-632. (2019).
[308] Stevens BR, et al.. Depressive hypertension: A proposed human endotype of brain/gut microbiome dysbiosis. Am Heart J. 39:27-37. (2021).
[309] Zhang Q, et al Gut Microbiome Composition Associated With Major Depressive Disorder and Sleep Quality. Frontiers in psychiatry, 12, 645045 (2021).
[310] Khlevner J, et al. Brain-Gut Axis: Clinical Implications. Gastroenterol Clin North Am. 47(4):727-739 (2018).
[311] Cheung SG, et al. Systematic Review of Gut Microbiota and Major Depression. Front Psychiatry. 10:34 (2019).
[312] Andrew M, et al. Associations among diet, the gastrointestinal microbiota, and negative emotional states in adults, Nutr Neurosci. 22:1-10 (2019).
[313] McGaughey KD, et al. Relative abundance of Akkermansia spp. and other bacterial phylotypes correlates with anxiety- and depressive-like behavior following social defeat in mice.
Nature Sci Rep. 9(1):3281 (2019).
[314] Mangiola F, et al. Gut microbiota in autism and mood disorders. World J Gastroentero 22, 361–368 (2016).
[315] Jiang H, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav. Immun. 48, 186–194 (2015).
[316] Aizawa E, et al. Possible association of Bifidobacterium an dLactobacillus in the gut microbiota of patients with major depressiv disorder, Jounral of Affectice Disorders (2016).
[317] Kelly J R, et al. Transferring the blues: depression-associated gut microbiota induces neurobehavioural changes in the rat. J. Psychiatr. Res. 82, 109–118. (2016).
[318] Messaoudi M, et al. Assessment of psychotropic-like properties of a probiotic formulation (Lactobacillus helveticus R0052 and Bifidobacterium longum R0175) in rats and human
subjects. (2010).
[319] Allen AP, et al. Bifidobacterium lingum 1714 as a trasnaltion psycholobiotic: Modulation of stress electrophysiology and neurocognition in healt volunteers. Translation Psychiarty
6:111 e393 (2016).
[320] Hayek N, et al. Chocolate, gut microbiota, and human health. Front Pharmacol. 4:11 (2013).
[321] Brickman AM, et al. Enhancing dentate gyrus function with dietary avanols improves cognition in older adults. Nat Neurosci. 17(12):1798–1803 (2014).
[322] Fraga CG, et al.The effects of polyphenols and other bioactives on human health. Food Funct. 10(2):514-528 (2019).
[323] Waclawiková B, et al. Role of Microbiota and Tryptophan Metabolites in the Remote Effect of Intestinal Inammation on Brain and Depression. Pharmaceuticals (Basel). 11(3): 63
(2018).
[324] O'Mahony SM, et al. Serotonin, tryptophan metabolism and the brain-gut-microbiome axis. Behav Brain Res. 277:32-48 (2015).
[325] Lindseth G, et al. The Effects of Dietary Tryptophan on Affective Disorders. Arch Psychiatr Nurs. 29(2):102–107 (2015).
[326] Liu S, et al. Metagenomic analysis of the gut microbiome in atherosclerosis patients identify cross-cohort microbial signatures and potential therapeutic target. The FASEB Journal.
(2020).
[327] Xu, et al. Compositional and Functional Alterations of Gut Microbiota in Patients with Stroke,Nutrition, Metabolism and Cardiovascular Diseases (2021).
[328] Companys J, et al. Gut Microbiota Profile and Its Association with Clinical Variables and Dietary Intake in Overweight/Obese and Lean Subjects: A Cross-Sectional Study.
Nutrients.;13(6):2032.(2021).
[329] Ozato N, et al. Blautia genus associated with visceral fat accumulation in adults 20–76 years of age. npj Biofilms Microbiomes 5, 28 (2019).
[330] Zhu, et al. Dysbiosis signatures of gut microbiota in coronary artery disease.hysiological Genomics50:10, 893-903 (2018).
[331] Chen, et al. Oolong tea extract and citrus peel polymethoxyflavones reduce transformation of L-Carnitine to trimethylamine-N-oxide and decrease vascular inflammation in L-Carnitine
feeding mice. Journal of Agricultural and Food Chemistry, vol. 67, no. 28, pp. 7869–7879, (2019).
[332] Shi J, et al. Berberine treatment reduces atherosclerosis by mediating gut microbiota in ApoE-/- mice. Biomedicine & Pharmacotherapy, vol. 107, pp. 1556–1563, (2018).
[333] Anwar U, et al. Trigonelline inhibits intestinal microbial metabolism of choline and its associated cardiovascular risk Journal of Pharmaceutical and Biomedical Analysis, vol. 159, pp.
100–112 (2018).
[334] Li X, et al. Berberine attenuates choline-induced atherosclerosis by inhibiting trimethylamine and trimethylamine-N-oxide production via manipulating the gut microbiome. npj Biofilms
Microbiomes 7, 36 (2021).
[335] Iglesias-Carres, et al. Use of dietary phytochemicals for inhibition of trimethylamine N-oxide formation. The Journal of Nutritional Biochemistry, 91, 108600. (2021).
[336] Roohbakhsh E, et al. The Effect of Interval Training and Consuming Fenugreek Seed Extract on FGF-21 and VEGF Gene Expression in Patients With Coronary Artery Diseases.
Horizon Med Sci. (2021).
[337] Dalla Via A, et al. Urinary TMAO Levels Are Associated with the Taxonomic Composition of the Gut Microbiota and with the Choline TMA-Lyase Gene (cutC) Harbored by
Enterobacteriaceae. Nutrients. (2020).
[338] Dan, et al. Differential Analysis of Hypertension-Associated Intestinal Microbiota. Int. J. Med. Sci. 16, 872–881 (2019).
[339] Huart J, et al. Gut Microbiota and Fecal Levels of Short-Chain Fatty Acids Differ Upon 24-Hour Blood Pressure Levels in Men. Hypertension (2019).
[340] Verhaar, et al. Associations between gut microbiota, faecal short-chain fatty acids, and blood pressure across ethnic groups: The HELIUS study.Eur. Heart J.(2020).
[341] Genoni, et al. Long‐term Paleolithic diet is associated with lower resistant starch intake, diferent gut microbiota composition and increased serum TMAO concentrations.European
Journal of Nutrition (2020).
[342] Macpherson, et al. Gut Microbiota-Dependent Trimethylamine N-Oxide Associates With Inflammation in Common Variable Immunodeficiency.Frontiers in Immunology .(2020).
[343] Skye, et al. Microbial Transplantation With Human Gut Commensals Containing CutC Is Sufficient to Transmit Enhanced Platelet Reactivity and Thrombosis Potential (2018).
[344] Griffin, et al. A mediterranean diet does not alter plasma trimethylamine N-oxide concentrations in healthy adults at risk for colon cancer. Food Funct.10:2138–47.(2019).
[345] Yoshida N, et al. A possible beneficial effect of Bacteroides on faecal lipopolysaccharide activity and cardiovascular diseases. Sci Rep 10, 13009 (2020).
[346] Depommier C, et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: a proof-of-concept exploratory study. Nat Med 25, 1096–1103
(2019).
[347] Ferguson JF, et al. Nutrigenomics, the Microbiome, and Gene-Environment Interactions: New Directions in Cardiovascular Disease Research, Prevention, and Treatment. A Scientic
Statement from the American Heart Association. Circ Cardiovasc Genet (2016).
[348] Tang WHW, et al. Dietary metabolism, the gut microbiome, and heart failure. Nat Rev Cardiol.;16(3):137-154 (2019).
[349] Burri BJ, et al. Absorption, metabolism, and functions of β-cryptoxanthin, Nutr Rev. 74(2): 69–82 (2016).
[350] Goszcz K, et al. Bioactive polyphenols and cardiovascular disease: Chemical antagonists, pharmacological agents or xenobiotics that drive an adaptive response? Br. J. Pharmacol. 174,
1209–1225 (2017).
[351] De Bruyne T, et al. Dietary Polyphenols Targeting Arterial Stiffness: Interplay of Contributing Mechanisms and Gut Microbiome- Related Metabolism. Nutrients. 11:3 (2019).
[352] Jie Z, et al. The gut microbiome in atherosclerotic cardiovascular disease. Nat Commun. 8(1):845 (2017).
[353] Kummen M, et al. The gut microbial profile in patients with primary sclerosing cholangitis is distinct from patients with ulcerative colitis without biliary disease and healthy controls.
Gut 66, 611–619 (2017).
[354] Karlsson FH, et al. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat Commun 3, 1245 (2012).
[355] Kasahara K, et al. Interactions between Roseburia intestinalis and diet modulate atherogenesis in a murine model. Nat. Microbiol. 3:1461–1471 (2018).
[356] Watson H, et al. A randomised trial of the effect of omega-3 polyunsaturated fatty acid supplements on the human intestinal microbiotaGut..(2018).
[357] Do, et al. High-Glucose or -Fructose Diet Cause Changes of the Gut Microbiota and Metabolic Disorders in Mice without Body Weight Change. Nutrients. (2018).
[358] Emamat H, et al. A. Artificial sweeteners are related to non-alcoholic fatty liver disease: Microbiota dysbiosis as a novel potential mechanismEXCU (2020).
[359] Friedman S, et al. Mechanisms of NAFLD development and therapeutic strategies. Nature Medicine (2018).
[360] Tian, et al. Perilla Oil Has Similar Protective Effects of Fish Oil on High-Fat Diet-Induced Nonalcoholic Fatty Liver Disease and Gut Dysbiosis (2016).
[361] Vadarlis, et al. Systematic review with meta-analysis: The effect of vitamin E supplementation in adult patients with non-alcoholic fatty liver disease.J Gastroenterol Hepatol (2020).
[362] Gjurašin, et al. Non-Alcoholic Fatty Liver Disease is Associated with an Increased Mortality in Adult Patients with Group B Streptococcus Invasive Disease (2020).
[363] Iwaki M, et al. Gut microbiota composition associated with hepatic fibrosis in non-obese patients with non-alcoholic fatty liver disease. Journal of Gastroenterology and Hepatology,
36: 2275– 2284. (2021).
[364] Shen, et al. Gut microbiota dysbiosis in patients with non-alcoholic fatty liver disease.Hepatobiliary & pancreatic diseases international: HBPD INT (2017).
[365] Demir, et al. Phenotyping non-alcoholic fatty liver disease by the gutmicrobiota: Ready for prime time? Journal of Gastroenterology and Hepatology (2020).
[366] Da Silva, et al.Nonalcoholic fatty livedisease is associated with dysbiosis independent of body mass index and insulin resistance. 8: 1466. (2018).
[367] Li F, et al. Characteristics of fecal microbiota in non-alcoholic fatty liver disease patients. Sci China Life Sci;61:770–8 (2018).
[368] Qiao, et al.Activation of a Specific Gut Bacteroides-Folate-Liver Axis Benefits for the Alleviation of Nonalcoholic Hepatic Steatosis,Cell Reports,Volume 32, Issue 6,108005 (2020).
[369] Tsai, et al. Gut microbiota dysbiosis in patients with biopsy-proven nonalcoholic fatty liver disease: a cross-sectional study in Taiwan. Nutrients. 12:820 (2020).
[370] Schwimmer JB, et al. Microbiome signatures associated with steatohepatitis and moderate to severe fibrosis in children with nonalcoholic fatty liver disease. Gastroenterology. (2019).
[371] Oh TG, et al. A Universal Gut-Microbiome-Derived Signature Predicts Cirrhosis. Cell metabolism (2020).
[372] Rau, et al. Fecal SCFAs and SCFA-producing bacteria in gut microbiome of human NAFLD as a putative link to systemic T-cell activation and advanced disease. United European
Gastroenterol J (2018).
[373] Ponziani, et al. Hepatocellular Carcinoma Is Associated With Gut Microbiota Profile and Inflammation in Nonalcoholic Fatty Liver Disease. Hepatology. (2019).
[374] Ren, et al. Correlation analysis of gut microbiota and biochemical indexes in patients with non-alcoholic fatty liver disease.Chinese Journal of Hepatology. (2019).
[375] Wu, et al. Identification of the keystone species in non-alcoholic fatty liver disease by causal inference and dynamic intervention modeling.bioRxiv. (2020).
[376] Kravetz, et al. Effect of Gut Microbiota and PNPLA3 rs738409 Variant on Nonalcoholic Fatty Liver Disease (NAFLD) in Obese Youth, The Journal of Clinical Endocrinology &
Metabolism, Volume 105, Issue 10. (2020).
[377] Zhao, et al. Metagenome of gutmicrobiota of children with nonalcoholic fatty liver disease. Front Pediatr.(2019).
[378] Rao, et al. Gut Akkermansia muciniphila ameliorates metabolic dysfunction-associated fatty liver disease by regulating the metabolism of L-aspartate via gut-liver axis. Gut Microbes.
(2021).
[379] Mokhtari Z, et al. Nonalcoholic Fatty Liver Disease, the Gut Microbiome, and Diet. Adv Nutr. 8(2):240-252 (2017).
[380] Safari Z, et al. The links between the gut microbiome and non‐alcoholic fatty liver disease (NAFLD), Cellular and Molecular Life Sciences 76(8):1541-1558, (2019).
[381] Pierantonelli I, et al. Nonalcoholic Fatty Liver Disease: Basic Pathogenetic Mechanisms in the Progression From NAFLD to NASH. Transplantation. 103(1):e1-e13 (2019).
[382] Bajaj JS, et al. Linkage of gut microbiome with cognition in hepatic encephalopathy. Am J Physiol-gastr L 302, G168–G175 (2012).
[383] Bajaj JS, et al. Colonic mucosal microbiome differs from stool microbiome in cirrhosis and hepatic encephalopathy and is linked to cognition and inammation. Am J Physiol-gastro 303,
G675–G685 (2012).
[384] Qin N, et al. Alterations of the human gut microbiome in liver cirrhosis. Nature 513:59–64 (2014).
[385] Jiang, W, et al. Dysbiosis gut microbiota associated with inammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci Rep-uk
5, 8096 (2015).
[386] Nistal E, et al. An altered fecal microbiota prole in patients with non- alcoholic fatty liver disease (NAFLD) associated with obesity. Rev Esp Enferm Dig. 111(4):275-282 (2019).
[387] Zhu L, et al. Characterization of gut microbiomes in nonalcoholic steatohepatitis (NASH) patients: a connection between endogenous alcohol and NASH. Hepatology 57(2):601-9
(2013).
[388] Torres J, et al. The features of mucosa-associated microbiota in primary sclerosing cholangitis Aliment Pharmacol Ther. 43(7):790- 801 (2016).
[389] Del Chierico F, et al. Gut microbiota proling of pediatric nonalcoholic fatty liver disease and obese patients unveiled by an integrated meta-omics-based approach. Hepatology,
65(2):451-464 (2017).
[390] Caussy C, et al. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat Commun. 10(1):1406 (2019).
[391] Mouzaki M, et al. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 58, 120–127 (2013).
[392] Sohail MU, et al. Role of the Gastrointestinal Tract Microbiome in the Pathophysiology of Diabetes Mellitus. Journal of Diabetes Research 2017:9631435, 9 (2017).
[393] Jørgensen PB, et al. A possible link between food and mood: dietary impact on gut microbiota and behavior in BALB/c mice. PLoS One 9(8):e103398 (2014).
[394] Owen L, et al. The role of diet and nutrition on mental health and wellbeing. Proc Nutr Soc. 76(4):425-426 (2017).
[395] Noble EE, et al. Gut to Brain Dysbiosis: Mechanisms Linking Western Diet Consumption, the Microbiome, and Cognitive Impairment. Front Behav Neurosci. 11:9 (2017).
[396] Oriachad CS, et al. Food for thought: The role of nutrition in the microbiota-gut–brain axis. Clin Nut Experimental 6:25-38 (2016).
[397] Franco R, et al. The central role of glutathione in the pathophysiology of human diseases. Medycyna pracy. 113(4-5):234-58 (2007).
[398] Menni C, et al. Omega-3 fatty acids correlate with gut microbiome diversity and production of N-carbamylglutamate in middle aged and elderly women Sci Rep. 7: 11079 (2017).
[399] Robertson RC, et al. Omega-3 polyunsaturated fatty acids critically regulate behaviour and gut microbiota development in adolescence and adulthood. Brain Behav Immun. 59:21-37
(2017).
[400] Calder PC, et al. Omega-3 polyunsaturated fatty acids and inammatory processes: nutrition or pharmacology? Br J Clin Pharmacol. 75(3):645-62 (2013).
[401] Huang L, et al. Dysbiosis of gut microbiota was closely associated with psoriasis. Sci China Life Sci.62(6):807-815 (2019).
[402] Madden, et al. How lifestyle factors and their associated pathogenetic mechanisms impact psoriasis.Clinical Nutrition, Volume 39, Issue 4, 1026 - 1040. (2020).
[403] Kashani, et al. Priyankar Dey. Dietary Inflammatory Index in Relation to Psoriasis Risk, Cardiovascular Risk Factors and Clinical Outcomes;A Result from Case-Control Study in
Psoriasis Patients. Applied Physiology, Nutrition, and Metabolism. (2021).
[404] Kanda N, et al.Nutrition and Psoriasis. International Journal of Molecular Sciences. (2020).
[405] Myers B, et al. The gut microbiome in psoriasis and psoriatic arthritis. Best Pract. Res. Clin. Rheumatol. 2019, 33, 101494. (2019).
[406] Visser M, et al. Bacterial Dysbiosis and Translocation in Psoriasis Vulgaris. Front. Cell Infect. Microbiol. 9,(2019).
[407] Hsu D, et al. Role of skin and gut microbiota in the pathogenesis of psoriasis, an inflammatory skin disease. Microecol.Med.(2020).
[408] Chen L, et al.Skin and gut microbiome in psoriasis: Gaining insight into the pathophysiology of it and finding novel therapeutic strategies. Front. Microbiol. (2020).
[409] Chen YJ, et al. Intestinal microbiota profiling and predicted metabolic dysregulation in psoriasis patients. Exp Dermatol27: 1336– 1343. (2018).
[410] Tan L, et al. The Akkermansia muciniphila is a gut microbiota signature in psoriasis. Exp Dermatol.Feb;27(2):144-149(2018).
[411] Hidalgo-Cantabrana C, et al. Gut microbiota dysbiosis in a cohort of patients with psoriasis. Br J Dermatol, 181: 1287-1295. (2019).
[412] Shapiro, et al. Psoriatic patients have a distinct structural and functional fecal microbiota compared with controls. J Dermatol. (2019).
[413] Dei-Cas I, et al. Metagenomic analysis of gut microbiota in non-treated plaque psoriasis patients stratified by disease severity: development of a new Psoriasis-Microbiome Index. Sci
Repul 29;10(1):12754. (2020).
[414] Zhang X, et al. Dysbiosis of gut microbiota and its correlation with dysregulation of cytokines in psoriasis patients. BMC Microbiol 21, 78 (2021).
[415] Xiao, et al. Deciphering Gut Microbiota Dysbiosis and Corresponding Genetic and Metabolic Dysregulation in Psoriasis Patients Using Metagenomics Sequencing .Frontiers in Cellular
and Infection Microbiology. (2021).
[416] Sun C, et al. Involvement of Gut Microbiota in the Development of Psoriasis Vulgaris.medRxiv . (2020).
[417] Ye S, et al. Diversity analysis of gut microbiota between healthy controls and those with atopic dermatitis in a Chinese population. J. Dermatol., 48: 158-167. (2021).
[418] Millsop JW, et al. Diet and psoriasis, part III: role of nutritional supplements, J Am Acad Dermatol. 71(3):561-9 (2014).
[419] Del Duca E, et al. Superiority of a vitamin B12-containing emollient compared to a standard emollient in the maintenance treatment of mild-to-moderate plaque psoriasis. Int J
Immunopathol Pharmacol. 30(4):439-444 (2017).
[420] Simopoulos AP, et al. The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed Pharmacother;56(8):365-79. (2002).
[421] Vaughn AR, et al. Skin-gut axis: The relationship between intestinal bacteria and skin health. World J Dermatol. 6(4): 52-58 (2017).
[422] Gianchecchi E, et al. Recent Advances on Microbiota Involvement in the Pathogenesis of Autoimmunity. Int. Journal of Mol Science 20(283) (2019).
[423] Salem I, et al. The Gut Microbiome as a Major Regulator of the Gut-Skin Axis. Front. Microbiol. 9:1459 (2019).
[424] Codoñer FM, et al. Gut microbial composition in patients with psoriasis. Sci. Rep. 8:3812 (2018).
[425] Wang, et al. Dietary Supplementation with Inulin Modulates the Gut Microbiota and Improves Insulin Sensitivity in Prediabetes.International Journal of Endocrinology (2021).
[426] Drouin-Chartier, et al. Changes in dairy product consumption and risk of type 2 diabetes: results from 3 large prospective cohorts of US men and women. The American journal of
clinical nutrition. (2019).
[427] Celli GB, et al. Gastroretentive systems - a proposed strategy to modulate anthocyanin release and absorption for the management of diabetes. Drug delivery.;23(6):1892-901.
(2016).
[428] Wan,g et al. Enterotype Bacteroides Is Associated with a High Risk in Patients with Diabetes: A Pilot Study . Journal of Diabetes Research. (2020).
[429] Del Chierico, et al. Fecal microbiota signatures of insulin resistance, inflammation, and metabolic syndrome in youth with obesity: a pilot study. Acta Diabetol 58, 1009–1022 (2021).
[430] Frost F, et al. A structured weight loss program increases gut microbiota phylogenetic diversity and reduces levels of Collinsella in obese type 2 diabetics: A pilot study. PLoS ONE
14(7): e0219489 (2019).
[431] Nirmalkar K, et al. Gut Microbiota and Endothelial Dysfunction Markers in Obese Mexican Children and Adolescents. Nutrients. (2018).
[432] Zhu Y, et al. Effects of oat β-glucan, oat resistant starch, and the whole oat flour on insulin resistance, inflammation, and gut microbiota in high-fat-diet-induced type 2 diabetic
rats,Journal of Functional Foods. (2020).
[433] Sánchez-Tapia M, et al.Consumption of Cooked Black Beans Stimulates a Cluster of Some Clostridia Class Bacteria Decreasing Inflammatory Response and Improving Insulin
Sensitivity. Nutrients.;12(4):1182 (2020).
[434] Shih CT, et al. Akkermansia muciniphila is Negatively Correlated with Hemoglobin A1c in Refractory Diabetes. Microorganisms. (2020).
[435] Kulkarni P, et al. Could dysbiosis of inflammatory and anti-inflammatory gut bacteria have an implications in the development of type 2 diabetes? A pilot investigation. BMC Res Notes
14, 52 (2021).
[436] Almugadam, et al. Alterations of Gut Microbiota in Type 2 Diabetes Individuals and the Confounding Effect of Antidiabetic Agents.Journal of Diabetes Research (2020).
[437] Cai, et al. Ethanol extract of propolis prevents high-fat diet-induced insulin resistance and obesity in association with modulation of gut microbiota in mice. Food Res Int.;130:108939.
(2020).
[438] Zhang, et al. Decreased Abundance of Akkermansia muciniphila Leads to the Impairment of Insulin Secretion and Glucose Homeostasis in Lean Type 2 Diabetes. 8, 2100536. (2021).
[439] Maskarinec G, et al.The gut microbiome and type 2 diabetes status in the Multiethnic Cohort. PLoS ONE 16(6): e0250855. (2021).
[440] Cinek O, et al.The bacteriome at the onset of type 1 diabetes: a study from four geographically distant African and Asian countries. Diabetes Research and Clinical Practice, vol. 144,
pp. 51–62. (2018).
[441] Higuchi, et al.Intestinal dysbiosis in autoimmune diabetes is correlated with poor glycemic control and increased interleukin-6: a pilot study.Frontiers in Immunology, vol. 9. (2018).
[442] Huang, et al.Gut microbiota profiling in Han Chinese with type 1 diabetes.Diabetes Research and Clinical Practice, vol. 141, pp. 256–263 (2018).
[443] Leiva-Gea, et al.Gut microbiota differs in composition and functionality between children with type 1 diabetes and MODY2 and healthy control subjects: a case-control study.Diabetes
Care, vol. 41, no. 11, pp. 2385–2395 (2018).
[444] Karlsson FH, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 498, 99 (2013).
[445] Wang N, et al. Proteomics, metabolomics and metagenomics for type 2 diabetes and its complications. Life Science 1;212:194-202 (2018).
[446] Kim YA, et al. Probiotics, prebiotics, synbiotics and insulin sensitivity. Nutrition Research Reviews : 1-17 (2017).
[447] Lambeth SM, et al. Composition, Diversity and Abundance of Gut Microbiome in Prediabetes and Type 2 Diabetes, J Diabetes Obes, 2(3): 1–7, (2015).
[448] Qin J, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 55 (2012).
[449] Wu X, et al. Molecular Characterisation of the Faecal Microbiota in Patients with Type II Diabetes. Curr Microbiol 61, 69–78 (2010).
[450] Hwang N, et al. Genes and Gut Bacteria Involved in Luminal Butyrate Reduction Caused by Diet and Loperamide. Genes (Basel) 28;8(12) (2017).
[451] Al-Otaibi FE, et al. Non-vertebral Veillonella species septicemia and osteomyelitis in a patient with diabetes: a case report and review of the literature, J Med Case Rep, 8:365 (2014).
[452] Lippert K, et al. Gut microbiota dysbiosis associated with glucose metabolism disorders and the metabolic syndrome in older adults. Benef Microbes. 8(4):545-556 (2017).
[453] Zhang X, et al. Human Gut Microbiota Changes Reveal the Progression of Glucose Intolerance. Plos One 8, e71108 (2013).
[454] Moreno-Indias I, et al. Insulin resistance is associated with specic gut microbiota in appendix samples from morbidly obese patients. Am J Transl Res 8, 5672–5684 (2016).
[455] Chen W, et al. Review of Ginseng Anti-Diabetic Studies. Molecules.;24(24) (2019).
[456] Satija A, 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;13(6):e1002039
(2016).
[457] Różańska D, et al. The signicance of anthocyanins in the prevention and treatment of type 2 diabetes. Adv Clin Exp Med.;27(1):135-142 (2018).
[458] Alpizar-Rodriguez D, et al. Prevotella copE in individuals at risk for rheumatoid arthritis. Ann Rheum Dis.;78(5):590.(2019).
[459] Boer CG, et al. Intestinal microbiome composition and its relation to joint pain and inflammation. Nat Commun.;10(1):4881.(2019).
[460] Volkmann ER, et al. Systemic sclerosis is associated with specific alterations in gastrointestinal microbiota in two independent cohorts. BMJ Open Gastroenterol.Apr 1;4(1):e000134.
(2017).
[461] Wells PM, et al. Associations between gut microbiota and genetic risk for rheumatoid arthritis in the absence of disease: a cross-sectional study. The Lancet Rheumatology;2(7):e418-
e27 (2020).
[462] Gupta VK, et al.Gut Microbiome Predicts Clinically Important Improvement in Patients with Rheumatoid Arthritis. medRxiv;(2020).
[463] Natalello G, et al. Gut microbiota analysis in systemic sclerosis according to disease characteristics and nutritional status. Clinical and experimental rheumatology;38 Suppl 125(3):73-
84 (2020).
[464] Mena-Vázquez N, et al. Expansion of Rare and Harmful Lineages is Associated with Established Rheumatoid Arthritis. Journal of Clinical Medicine.;9(4). (2020).
[465] Moen K, et al. Immunoglobulin G and A Antibody Responses to Bacteroides forsythus and Prevotella intermedia in Sera and Synovial Fluids of Arthritis Patients. Clin Diagn Lab
Immun 10, 1043–1050 (2003).
[466] Kishikawa T, et al. Metagenome-wide association study of gut microbiome revealed novel aetiology of rheumatoid arthritis in the Japanese population. Ann Rheum Dis;79(1):103.
(2020).
[467] Mclean MH, et al. Does the microbiota play a role in the pathogenesis of autoimmune diseases?. Gut, 64(2):332-41 (2015).
[468] Chen J, et al. Dysbiosis of the gut microbiome is a risk factor for osteoarthritis in older female adults: a case control study. BMC Bioinformatics 22;299 (2021).
[469] Henson, et al. Interrogation of the perturbed gut microbiota in gouty arthritis patients through in silico metabolic modeling. Eng. Life Sci. (2021).
[470] Rooney C, et al. Investigating the role of the gut microbiome as a trigger for arthritis development in individuals at risk of rheumatoid arthritis.Microbiology Society (2020).
[471] Yong W, et al. Associations of Changes in Serum Inflammatory Factors, MMP-3, 25(OH)D and Intestinal Flora with Osteoporosis and Disease Activity in Rheumatoid Arthritis Patients.
Clinical Laboratory. Dec;66(12) (2020).
[472] Boulygina, et al. Gut Microbiota in Rheumatoid Arthritis: Potential Bacterial Drivers and Protectors. medRxiv (2019).
[473] Balekuduru, et al. Alterations in fecal microbiota in patients with inflammatory bowel disease and enteropathic arthropathy.103-110 (2021).
[474] Chiang, et al. An Association of Gut Microbiota with Different Phenotypes in Chinese Patients with Rheumatoid Arthritis.Journal of Clinical Medicine (2019).
[475] Wang, et al. AB0824 ANALYSIS OF GUT MICROBIOTA IN RHEUMATOID ARTHRITIS PATIENTS .Annals of the Rheumatic Diseases 2021;80:1436.(2021).
[476] Wei M, et al. High-Throughput Absolute Quantification Sequencing Revealed Osteoporosis-Related Gut Microbiota Alterations in Han Chinese Elderly. Front. Cell. Infect. Microbiol.
2021. 11:63037 (2021).
[477] Picchianti-Diamanti A, et al. Analysis of Gut Microbiota in Rheumatoid Arthritis Patients: Disease-Related Dysbiosis and Modifications Induced by Etanercept. Int J Mol
Sci.;19(10):2938 (2018).
[478] Scher JU, et al. Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. Elife 2, e01202 (2013).
[479] Pianta A, et al. Evidence of the Immune Relevance of Prevotella copri, a Gut Microbe, in Patients With Rheumatoid Arthritis. Arthritis Rheumatol 69, 964–975 (2017).
[480] Horta-Baas G, et al. Intestinal dysbiosis and rheumatoid arthritis: a link between gut microbiota and the pathogenesis of rheumatoid arthritis. J Immunol Res. 2017:1–13 (2017).
[481] Forbes JD, et al. A comparative study of the gut microbiota in immune-mediated inflammatory diseases—does a common dysbiosis exist?. Microbiome 6, 221 (2018).
[482] Kimura Y, et al. Periodontal pathogens participate in synovitis in patients with rheumatoid arthritis in clinical remission: a retrospective case–control study. Rheumatology 54, 2257–
2263 (2015).
[483] Yeoh N, et al. The role of the microbiome in rheumatic diseases. Curr. Rheumatol. Rep. 15 (2013).
[484] Liu Z, et al. Self-Balance of Intestinal Flora in Spouses of Patients With Rheumatoid Arthritis. Front Med (Lausanne).;7:538 (2020).
[485] Hemshekhar M, et al. A bioavailable form of curcumin, in combination with vitamin-D- and omega-3-enriched diet, modifies disease onset and outcomes in a murine model of
collagen-induced arthritis. Arthritis Res Ther 23, 39 (2021).
[486] Aparicio-Soto M, et al. The phenolic fraction of extra virgin olive oil modulates the activation and the inflammatory response of T cells from patients with systemic lupus
erythematosus and healthy donors. Mol Nutr Food Res.(2017).
[487] Hong, et al. Tunisian Olea europaea L. leaf extract suppresses Freund's complete adjuvant-induced rheumatoid arthritis and lipopolysaccharide-induced inflammatory responses,
Journal of Ethnopharmacology, Volume 268, (2021).
[488] Pianta, et al. Identification of Novel, Immunogenic HLA-DR-Presented Prevotella copri Peptides in Patients with Rheumatoid Arthritis Patients with Rheumatoid Arthritis. (2021).
[489] Bustamante MF, et al. Design of an anti-inflammatory diet (ITIS diet) for patients with rheumatoid arthritis. Contemp Clin Trials Commun. (2020).
[490] Campbell AW, et al. Autoimmunity and the Gut, Autoimmune Diseases (2014).
[491] Fernandez-Moreno M, et al. Mitochondrial DNA (mtDNA) haplogroups and serum levels of anti-oxidant enzymes in patients with osteoarthritis. BMC Musculoskelet Disord.;12: 264.
(2011).
[492] Felson DT, et al. Dietary fatty acids for the treatment of OA, including sh oil. Ann Rheum Dis 75:1–2 (2016).
[493] Höfer S, et al. Praktische Diätetik. Wissenschaftl. Verlagsgesellschaft Stuttgart, XII (2018).