Metabolomics Approach: Linking Enterotypes to Personalized Diets: Comparison
Please note this is a comparison between Version 2 by Madeline Bartsch and Version 3 by Rita Xu.

Recent advances in high-throughput DNA sequencing have catalyzed a deeper exploration of the human microbiome and its intricate relationship with metabolic health. Researchers examine the influence of dietary choices on the production of microbial metabolites and provide a comprehensive review of studies investigating the interplay between enterotypes and diet. The goal is to contribute to the refinement of personalized dietary recommendations and provide valuable insights to advance the understanding of metabolic health.

Recent advances in high-throughput DNA sequencing have catalyzed a deeper exploration of the human microbiome and its intricate relationship with metabolic health. Our review examines different methods of microbiome analysis, with a particular focus on metabolomics. We examine the influence of dietary choices on the production of microbial metabolites and provide a comprehensive review of studies investigating the interplay between enterotypes and diet. The goal is to contribute to the refinement of personalized dietary recommendations and provide valuable insights to advance our understanding of metabolic health.

  • metabolomics
  • enterotypes
  • personalized dietary recommendations
  • Prevotella
  • Bacteroides

1. Introduction

Over the past two decades, research into the human microbiome and its link to metabolic health has been driven by advances in high-throughput DNA sequencing. Studies have revealed associations between changes in gut microbiota composition and metabolic disorders such as obesity [1][2], Type 2 Diabetes Mellitus (T2DM) [3], and cardiovascular diseases [4][5]. Influencing factors include age [6], gender [7], geography [8], genetics [9], and environmental elements [10][11][12], with dietary choices [13] playing a significant role.

To comprehend the intricate relationship between dietary patterns, the gut microbiome, and their effect on host metabolism, researchers have introduced enterotypes as a classification framework [14]. These categorize individuals based on their gut microbiota composition. To unravel the functional implications of enterotypes on host metabolism, it is necessary to integrate metabolomics, a discipline that studies small molecular compounds. Metabolomics provides a comprehensive overview of the metabolites produced by the gut microbiota in response to diet.

2.

This review focuses on methods analyzing the microbiome, particularly metabolomics, and explores how dietary choices influence microbial metabolites. It provides an overview of studies on the link between enterotypes and diet. The review concludes by evaluating how metabolomics can improve personalized dietary recommendations. It draws insights from the complex relationship between enterotypes and diet.

Methods in Microbiome Research: An Overview

The Human Microbiome Project initiative (HMP) has been instrumental in establishing fundamental frameworks for microbiota analysis [15]. Specifically, amplicon-based sequencing of 16S ribosomal ribonucleic acid (rRNA) has been widely used to analyze microbiome composition.

Key figures, such as those involved in the HMP initiative, have made significant contributions. The shift from amplicon-based sequencing to metagenomic analyses represents a milestone, as demonstrated by influential projects such as MetaHIT [16]. Metagenomic methods not only reveal taxonomic composition but also offer insights into functional metabolic capabilities, such as identifying genes that encode enzymes responsible for breaking down food components.

Metaproteomics in microbiome research was introduced in 2008 by Verberkmoes et al [17]. This technique enables a detailed investigation of microbial proteins and links them to specific microorganisms. Furthermore, Booijink et al. made a significant advancement by initiating a metatranscriptome analysis of the human fecal microbiome, which provides tractable links between microbial genetic potential and molecular activity [18].

The seamless progression from amplicon-based sequencing through metagenomics to metaproteomics and metatranscriptomics has established a comprehensive foundation for exploring the functional aspects of the microbiome. To gain a truly holistic understanding of its role in human health, the focus shifts further into the realm of metabolomics. Metabolomic techniques offer insight into the overall metabolic status and interactions within the microbiome. Despite the diverse characteristics of metabolites, metabolomics employs various technologies, including mass spectrometry (MS) and nuclear magnetic resonance (NMR) spectroscopy, to measure defined sets of known metabolites or perform a comprehensive analysis of the metabolome [19].

As part of this progression, the use of mass spectrometry (MS), particularly gas chromatography-mass spectrometry (GC-MS) and liquid chromatography-mass spectrometry (LC-MS), has become more prevalent in scientific studies. MS has the ability to detect metabolites with high sensitivity, impartiality, and efficiency [20]. Metabolomics, when combined with artificial intelligence (AI) and machine learning (ML) techniques, has the potential to enhance theour understanding of the microbiome's significance for personalized biomarker discovery, and nutritional recommendations [21].

Ethical considerations in metabolomics research are of utmost importance, emphasizing the need for privacy, informed consent, and transparency in handling participant data. Collaboration among researchers, ethicists, policymakers, and healthcare professionals is essential to address ethical concerns, foster responsible advancement, and maximize the benefits of metabolomics, microbial research, and personalized nutrition for society [22][23].

3. The Tight Interaction between Diet, the Gut Microbiome, and Its Metabolites

The investigation of the human microbiome has experienced significant expansion, with a specific focus on the intricate relationship between diet, the gut microbiome, and microbial metabolites. This interplay substantially influences theour overall health, as nutrients impact microbial growth, leading to the production of diverse metabolites.

3.1. Carbohydrates and Dietary Fiber

Carbohydrates and Dietary Fiber

The utilization of carbohydrates and dietary fiber holds particular importance. Microbiota-accessible carbohydrates (MACs), primarily obtained from plant-based sources, are indigestible carbohydrates that play a pivotal role in the interaction between the human body and the microbiome [24][25]. The microbiome possesses carbohydrate-active enzymes (CAZymes) that break down complex dietary carbohydrates, resulting in the production of short-chain fatty acids (SCFAs) as the primary end products. SCFAs, comprising acetate, propionate, and butyrate, exert various effects on human metabolism, influencing gut barrier integrity [26], gut motility [27], hormone regulation [28][29][30][31], chromatin dynamics [32][33], gut-brain connections [34], and immune responses [35]

3.2. Proteins and Amino Acids

Proteins and Amino Acids

In the absence of fermentable fibers, gut microbes adapt by utilizing amino acids from dietary proteins [36][37][38]. Proteolytic bacteria, including Bacteroides and Clostridium species, contribute to amino acid fermentation, yielding SCFAs and branched-chain fatty acids  [38][39]. The breakdown of branched-chain amino acids (BCAAs) like valine, leucine, and isoleucine, associated with an impaired insulin resistance, highlights the intricate relationship between protein fermentation, microbial metabolites, and metabolic health  [20][40][41].

3.3. Dietary Fat and Bile Acids

Dietary Fat and Bile Acids

Contrary to previous beliefs, dietary fat significantly influences the composition of the gut microbiota, subsequently impacting the host's health [42]. A diet rich in saturated fats can induce metabolic endotoxemia [43], a pro-inflammatory condition mediated through toll-like receptor 4 (TLR4) and CD14, leading to outcomes like weight gain, increased adiposity, and insulin resistance [44]. Bile acids, crucial for fat digestion and absorption, also engage with the gut microbiota [45]. An imbalance in the gut microbiota can perturb the bile acid receptor farnesoid X receptor (FXR), potentially contributing to conditions such as colorectal cancer [46].

3.4. Plant- and Animal-Derived Bioactive Compounds

Plant- and Animal-Derived Bioactive Compounds

Plant-derived polyphenols, abundant in various dietary sources, undergo significant modifications by the gut microbiota, influencing their absorption and bioavailability [47]. Despite limited bioavailability, polyphenols exhibit anti-inflammatory, antioxidant, and antimicrobial properties, impacting conditions such as cardiovascular disease [48], cancer [49], metabolic disorders [50], Alzheimer’s disease [51], and inflammatory bowel disease [52]. Additionally, nutrients from animal-derived foods, like choline, betaine, and l-carnitine, interact with the gut microbiome, contributing to the formation of trimethylamine-N-oxide (TMAO) and influencing health outcomes [53][54].

This overview highlights the importance of microbial metabolites in the relationship between dietary components and human health when exploring diet-microbiome interactions.

4. Interindividual Differences in Microbial Responses to Diet According to Enterotypes

Understanding the intricate relationship between an individual's microbiota and their response to dietary elements highlights the impracticality of a universal dietary approach. Recognizing the significance of precision nutrition becomes imperative in designing tailored dietary plans, mitigating the challenges of variability inherent in nutrition research. This chapter explores the concept of enterotypes, a categorization based on gut microbiota composition, shedding light on their applications, current status, and future directions in personalized dietary recommendations.

The concept of enterotypes was introduced in 2011 by Arumgam et al., who identified three distinct clusters based on microbiota composition across diverse populations [14]. Subsequent studies, including those by Liang et al., reinforced the presence of enterotypes and revealed variations, particularly in Asian populations [55]. However, challenges arose regarding the definition and identification of enterotypes, prompting further investigation.

Enterotypes, initially characterized by dominant genera like Bacteroides and Prevotella, have shown consistent patterns across various populations [56][57][58]. Although regional dietary distinctions likely contribute to subgroup variations within enterotypes, both Bacteroides and Prevotella enterotypes exhibit equal associations with diverse health conditions [59][60]. Notably, the role of Bifidobacterium in the Bacteroides enterotype, particularly in Japan, underscores the influence of regional dietary habits [61].

Applications and Current Status

The prominence of enterotypes lies in simplifying the complex gut microbiome landscape, offering avenues for microbiota-based diagnostics, therapeutic interventions, disease prevention strategies, and personalized dietary recommendations. However, challenges in precisely defining enterotypes led to a modified concept, emphasizing the need for standardization in enterotyping methods.

Enterotypes, particularly the Prevotella-dominated (P-type) and Bacteroides-dominated (B-type) types play a crucial role in reflecting individuals' long-term dietary patterns. The P-type, associated with vegetarians, high-fiber and carbohydrate-rich diets, and traditional diets, showcases efficient hydrolytic enzymes for plant fiber degradation but has limited lipid and protein fermentation capacity. In contrast, the B-type features specialized enzymes tailored for the degradation of animal-derived carbohydrates and proteins, exhibiting an enhanced saccharolytic and proteolytic potential [62][63][64].

Studies conducted by a research group from Denmark highlight the functional differences between Prevotella and Bacteroides in various human dietary intervention studies. The Prevotella to Bacteroides ratio (P/B ratio) was closely related to alterations in body fat and weight, with a caloric deficit of 500 kcal for 24 weeks leading to more significant weight and body fat loss in individuals with a high P/B ratio. This group also observed that a high-fiber diet, characteristic of traditional dietary patterns in P-type individuals, likely results in more efficient and substantial weight loss [65][66][67][68][69][70]. These functional distinctions underscore the relevance of considering individualized dietary plans based on enterotypes.

Current research offers promising insights into personalized dietary recommendations grounded in enterotypes and metabolomics methods. Despite the challenges stemming from microbiome intricacies, advancements in AI and machine learning tools exhibit potential in predicting metabolic responses to specific diets. The future trajectory involves surmounting challenges in comprehending microbiome functions and harnessing these advanced tools for more precise predictions. This ongoing effort holds the promise of enhanced dietary guidance tailored to individual microbiome compositions.

In conclusion, the integration of enterotypes and metabolomics provides valuable insights into the potential for personalized dietary recommendations based on an individual's unique microbiome composition. Despite the challenges inherent in understanding the intricacies of the microbiome and its multifaceted functions, the application of metabolomics emerges as a promising avenue for deciphering these complexities. These findings lay the foundation for more nuanced and tailored dietary guidance, offering the potential for improved metabolic responses based on an individual's distinct microbiome composition.

References

  1. Maggie A. Stanislawski; Dana Dabelea; Leslie A. Lange; Brandie D. Wagner; Catherine A. Lozupone; Gut microbiota phenotypes of obesity. npj Biofilms Microbiomes. 2019, 5, 1-9.
  2. Mariona Pinart; Andreas Dötsch; Kristina Schlicht; Matthias Laudes; Jildau Bouwman; Sofia K. Forslund; Tobias Pischon; Katharina Nimptsch; Gut Microbiome Composition in Obese and Non-Obese Persons: A Systematic Review and Meta-Analysis. Nutr.. 2021, 14, 12.
  3. Ge Yang; Jinlong Wei; Pinyi Liu; Qihe Zhang; Yuan Tian; Guowen Hou; Lingbin Meng; Ying Xin; Xin Jiang; Role of the gut microbiota in type 2 diabetes and related diseases. Metab.. 2021, 117, 154712.
  4. Hui Xu; Xiang Wang; Wenke Feng; Qi Liu; Shanshan Zhou; Quan Liu; Lu Cai; The gut microbiota and its interactions with cardiovascular disease. Microb. Biotechnol.. 2020, 13, 637-656.
  5. Shadi Ahmadmehrabi; W.H. Wilson Tang; Gut microbiome and its role in cardiovascular diseases. Curr. Opin. Cardiol.. 2017, 32, 761-766.
  6. Tanya Yatsunenko; Federico E. Rey; Mark J. Manary; Indi Trehan; Maria Gloria Dominguez-Bello; Monica Contreras; Magda Magris; Glida Hidalgo; Robert N. Baldassano; Andrey P. Anokhin; Andrew C. Heath; Barbara Warner; Jens Reeder; Justin Kuczynski; J. Gregory Caporaso; Catherine A. Lozupone; Christian Lauber; Jose Carlos Clemente; Dan Knights; Rob Knight; Jeffrey I. Gordon; Human gut microbiome viewed across age and geography. Nat.. 2012, 486, 222-227.
  7. The Human Microbiome Project Consortium; Structure, function and diversity of the healthy human microbiome. Nat.. 2012, 486, 207-214.
  8. Juan S Escobar; Bernadette Klotz; Beatriz E Valdes; Gloria M Agudelo; The gut microbiota of Colombians differs from that of Americans, Europeans and Asians. BMC Microbiol.. 2014, 14, 1-14.
  9. Omer Weissbrod; Daphna Rothschild; Elad Barkan; Eran Segal; Host genetics and microbiome associations through the lens of genome wide association studies. Curr. Opin. Microbiol.. 2018, 44, 9-19.
  10. Gwen Falony; Marie Joossens; Sara Vieira-Silva; Jun Wang; Youssef Darzi; Karoline Faust; Alexander Kurilshikov; Marc Jan Bonder; Mireia Valles-Colomer; Doris Vandeputte; Raul Y. Tito; Samuel Chaffron; Leen Rymenans; Chloë Verspecht; Lise De Sutter; Gipsi Lima-Mendez; Kevin D’hoe; Karl Jonckheere; Daniel Homola; Roberto Garcia; Ettje F. Tigchelaar; Linda Eeckhaudt; Jingyuan Fu; Liesbet Henckaerts; Alexandra Zhernakova; Cisca Wijmenga; Jeroen Raes; Population-level analysis of gut microbiome variation. Sci.. 2016, 352, 560-564.
  11. Henrik M. Roager; Lea B. S. Hansen; Martin I. Bahl; Henrik L. Frandsen; Vera Carvalho; Rikke J. Gøbel; Marlene D. Dalgaard; Damian R. Plichta; Morten H. Sparholt; Henrik Vestergaard; Torben Hansen; Thomas Sicheritz-Pontén; H. Bjørn Nielsen; Oluf Pedersen; Lotte Lauritzen; Mette Kristensen; Ramneek Gupta; Tine R. Licht; Colonic transit time is related to bacterial metabolism and mucosal turnover in the gut. Nat. Microbiol.. 2016, 1, 16093.
  12. Francesco Asnicar; Emily R Leeming; Eirini Dimidi; Mohsen Mazidi; Paul W Franks; Haya Al Khatib; Ana M Valdes; Richard Davies; Elco Bakker; Lucy Francis; Andrew Chan; Rachel Gibson; George Hadjigeorgiou; Jonathan Wolf; Timothy D Spector; Nicola Segata; Sarah E Berry; Blue poo: impact of gut transit time on the gut microbiome using a novel marker. Gut. 2021, 70, 1665-1674.
  13. Niv Zmora; Jotham Suez; Eran Elinav; You are what you eat: diet, health and the gut microbiota. Nat. Rev. Gastroenterol. Hepatol.. 2018, 16, 35-56.
  14. Manimozhiyan Arumugam; MetaHIT Consortium (additional members); Jeroen Raes; Eric Pelletier; Denis Le Paslier; Takuji Yamada; Daniel R. Mende; Gabriel R. Fernandes; Julien Tap; Thomas Bruls; Jean-Michel Batto; Marcelo Bertalan; Natalia Borruel; Francesc Casellas; Leyden Fernandez; Laurent Gautier; Torben Hansen; Masahira Hattori; Tetsuya Hayashi; Michiel Kleerebezem; Ken Kurokawa; Marion Leclerc; Florence Levenez; Chaysavanh Manichanh; H. Bjørn Nielsen; Trine Nielsen; Nicolas Pons; Julie Poulain; Junjie Qin; Thomas Sicheritz-Ponten; Sebastian Tims; David Torrents; Edgardo Ugarte; Erwin G. Zoetendal; Jun Wang; Francisco Guarner; Oluf Pedersen; Willem M. de Vos; Søren Brunak; Joel Doré; Jean Weissenbach; S. Dusko Ehrlich; Peer Bork; Enterotypes of the human gut microbiome. Nat.. 2011, 473, 174-180.
  15. Peter J. Turnbaugh; Ruth E. Ley; Micah Hamady; Claire M. Fraser-Liggett; Rob Knight; Jeffrey I. Gordon; The Human Microbiome Project. Nat.. 2007, 449, 804-810.
  16. S. Dusko Ehrlich; Metagenomics of the intestinal microbiota: potential applications. null. 2010, 34, S23-S28.
  17. Nathan C Verberkmoes; Alison L Russell; Manesh Shah; Adam Godzik; Magnus Rosenquist; Jonas Halfvarson; Mark G Lefsrud; Juha Apajalahti; Curt Tysk; Robert L Hettich; Janet K Jansson; Shotgun metaproteomics of the human distal gut microbiota. ISME J.. 2008, 3, 179-189.
  18. Carien C. G. M. Booijink; Jos Boekhorst; Erwin G. Zoetendal; Hauke Smidt; Michiel Kleerebezem; Willem M. de Vos; Metatranscriptome Analysis of the Human Fecal Microbiota Reveals Subject-Specific Expression Profiles, with Genes Encoding Proteins Involved in Carbohydrate Metabolism Being Dominantly Expressed. Appl. Environ. Microbiol.. 2010, 76, 5533-5540.
  19. Mireia Valles-Colomer; Cristina Menni; Sarah E. Berry; Ana M. Valdes; Tim D. Spector; Nicola Segata; Cardiometabolic health, diet and the gut microbiome: a meta-omics perspective. Nat. Med.. 2023, 29, 551-561.
  20. Kimberly A. Krautkramer; Jing Fan; Fredrik Bäckhed; Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol.. 2020, 19, 77-94.
  21. Vidya Niranjan; Akshay Uttarkar; Aakaanksha Kaul; Maryanne Varghese. A Machine Learning-Based Approach Using Multi-omics Data to Predict Metabolic Pathways; Springer Science and Business Media LLC: Dordrecht, GX, Netherlands, 2022; pp. 441-452.
  22. Alessandro Di Minno; Monica Gelzo; Marianna Caterino; Michele Costanzo; Margherita Ruoppolo; Giuseppe Castaldo; Challenges in Metabolomics-Based Tests, Biomarkers Revealed by Metabolomic Analysis, and the Promise of the Application of Metabolomics in Precision Medicine. Int. J. Mol. Sci.. 2022, 23, 5213.
  23. Cecil M. Lewis; Laura‐Isobel McCall; Richard R. Sharp; Paul G. Spicer; Ethical priority of the most actionable system of biomolecules: the metabolome. Am. J. Phys. Anthr.. 2019, 171, 177-181.
  24. Jacob F. Wardman; Rajneesh K. Bains; Peter Rahfeld; Stephen G. Withers; Carbohydrate-active enzymes (CAZymes) in the gut microbiome. Nat. Rev. Microbiol.. 2022, 20, 542-556.
  25. Lawrence A. David; Corinne F. Maurice; Rachel N. Carmody; David B. Gootenberg; Julie E. Button; Benjamin E. Wolfe; Alisha V. Ling; A. Sloan Devlin; Yug Varma; Michael A. Fischbach; Sudha B. Biddinger; Rachel J. Dutton; Peter J. Turnbaugh; Diet rapidly and reproducibly alters the human gut microbiome. Nat.. 2013, 505, 559-563.
  26. Naschla Gasaly; Paul de Vos; Marcela A. Hermoso; Impact of Bacterial Metabolites on Gut Barrier Function and Host Immunity: A Focus on Bacterial Metabolism and Its Relevance for Intestinal Inflammation. Front. Immunol.. 2021, 12, 658354.
  27. Ara Koh; Filipe De Vadder; Petia Kovatcheva-Datchary; Fredrik Bäckhed; From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell. 2016, 165, 1332-1345.
  28. P. Larraufie; C. Martin-Gallausiaux; N. Lapaque; J. Dore; F. M. Gribble; F. Reimann; H. M. Blottiere; SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci. Rep.. 2018, 8, 1-9.
  29. Christopher S. Reigstad; Charles E. Salmonson; John F. Rainey Iii; Joseph H. Szurszewski; David R. Linden; Justin L. Sonnenburg; Gianrico Farrugia; Purna C. Kashyap; Gut microbes promote colonic serotonin production through an effect of short‐chain fatty acids on enterochromaffin cells. FASEB J.. 2014, 29, 1395-1403.
  30. Eun-Young Lee; Xilin Zhang; Junki Miyamoto; Ikuo Kimura; Tomoaki Taknaka; Kenichi Furusawa; Takahito Jomori; Kosuke Fujimoto; Satoshi Uematsu; Takashi Miki; Gut carbohydrate inhibits GIP secretion via a microbiota/SCFA/FFAR3 pathway. J. Endocrinol.. 2018, 239, 267-276.
  31. Kento Mio; Reina Ogawa; Natsuki Tadenuma; Seiichiro Aoe; Arabinoxylan as well as β-glucan in barley promotes GLP-1 secretion by increasing short-chain fatty acids production. Biochem. Biophys. Rep.. 2022, 32, 101343.
  32. Markus Waldecker; Tanja Kautenburger; Heike Daumann; Cordula Busch; Dieter Schrenk; Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. J. Nutr. Biochem.. 2008, 19, 587-593.
  33. Mahmoud L. Soliman; Thad A. Rosenberger; Acetate supplementation increases brain histone acetylation and inhibits histone deacetylase activity and expression. Mol. Cell. Biochem.. 2011, 352, 173-180.
  34. Boushra Dalile; Lukas Van Oudenhove; Bram Vervliet; Kristin Verbeke; The role of short-chain fatty acids in microbiota–gut–brain communication. Nat. Rev. Gastroenterol. Hepatol.. 2019, 16, 461-478.
  35. Daniela Parada Venegas; Marjorie K. De la Fuente; Glauben Landskron; María Julieta González; Rodrigo Quera; Gerard Dijkstra; Hermie J. M. Harmsen; Klaas Nico Faber; Marcela A. Hermoso; Short Chain Fatty Acids (SCFAs)-Mediated Gut Epithelial and Immune Regulation and Its Relevance for Inflammatory Bowel Diseases. Front. Immunol.. 2019, 10, 277.
  36. Rebecca Wall; R Paul Ross; Fergus Shanahan; Liam O’mahony; Caitlin O’mahony; Mairead Coakley; Orla Hart; Peadar Lawlor; Eamonn M Quigley; Barry Kiely; Gerald F Fitzgerald; Catherine Stanton; Metabolic activity of the enteric microbiota influences the fatty acid composition of murine and porcine liver and adipose tissues. Am. J. Clin. Nutr.. 2009, 89, 1393-1401.
  37. Pieter Evenepoel; Dirk Claus; Benny Geypens; Martin Hiele; Karen Geboes; Paul Rutgeerts; Yvo Ghoos; Xinxin Liu; Jean-Marc Blouin; Arlette Santacruz; Annaïg Lan; Mireille Andriamihaja; Sabina Wilkanowicz; Pierre-Henri Benetti; Daniel Tomé; Yolanda Sanz; François Blachier; Anne-Marie Davila; Henrike M. Hamer; Vicky De Preter; Karen Windey; Kristin Verbeke; Mamy Eklou-Lawson; Nathalie Petit; Serge Delpal; Fadhila Allek; Anne Blais; Corine Delteil; Amount and fate of egg protein escaping assimilation in the small intestine of humans. Am. J. Physiol. Liver Physiol.. 1999, 277, G935-G943.
  38. Alexandria Bartlett; Manuel Kleiner; Dietary protein and the intestinal microbiota: An understudied relationship. iScience. 2022, 25, 105313.
  39. G. T. Macfarlane; J. H. Cummings; C. Allison; Protein Degradation by Human Intestinal Bacteria. Microbiol.. 1986, 132, 1647-1656.
  40. Christopher B. Newgard; Interplay between Lipids and Branched-Chain Amino Acids in Development of Insulin Resistance. Cell Metab.. 2012, 15, 606-614.
  41. Christopher B. Newgard; Jie An; James R. Bain; Michael J. Muehlbauer; Robert D. Stevens; Lillian F. Lien; Andrea M. Haqq; Svati H. Shah; Michelle Arlotto; Cris A. Slentz; James Rochon; Dianne Gallup; Olga Ilkayeva; Brett R. Wenner; William S. Yancy; Howard Eisenson; Gerald Musante; Richard S. Surwit; David S. Millington; Mark D. Butler; Laura P. Svetkey; A Branched-Chain Amino Acid-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance. Cell Metab.. 2009, 9, 311-326.
  42. Laure Gabert; Cécile Vors; Corinne Louche‐Pélissier; Valérie Sauvinet; Stéphanie Lambert‐Porcheron; Jocelyne Drai; Martine Laville; Michel Désage; Marie‐Caroline Michalski; 13C tracer recovery in human stools after digestion of a fat‐rich meal labelled with [1,1,1‐13C3]tripalmitin and [1,1,1‐13C3]triolein. Rapid Commun. Mass Spectrom.. 2011, 25, 2697-2703.
  43. Jacques Amar; Rémy Burcelin; Jean Bernard Ruidavets; Patrice D Cani; Josette Fauvel; Marie Christine Alessi; Bernard Chamontin; Jean Ferriéres; Energy intake is associated with endotoxemia in apparently healthy men. Am. J. Clin. Nutr.. 2008, 87, 1219-1223.
  44. Patrice D. Cani; Rodrigo Bibiloni; Claude Knauf; Aurélie Waget; Audrey M. Neyrinck; Nathalie M. Delzenne; Rémy Burcelin; Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet–Induced Obesity and Diabetes in Mice. Diabetes. 2008, 57, 1470-1481.
  45. Stephanie L. Collins; Jonathan G. Stine; Jordan E. Bisanz; C. Denise Okafor; Andrew D. Patterson; Bile acids and the gut microbiota: metabolic interactions and impacts on disease. Nat. Rev. Microbiol.. 2022, 21, 236-247.
  46. Soeren Ocvirk; Stephen J.D. O’keefe; Dietary fat, bile acid metabolism and colorectal cancer. Semin. Cancer Biol.. 2020, 73, 347-355.
  47. Laura Marín; Elisa M. Miguélez; Claudio J. Villar; Felipe Lombó; Bioavailability of Dietary Polyphenols and Gut Microbiota Metabolism: Antimicrobial Properties. BioMed Res. Int.. 2015, 2015, 1-18.
  48. Lee Hooper; Colin Kay; Asmaa Abdelhamid; Paul A Kroon; Jeffrey S Cohn; Eric B Rimm; Aedín Cassidy; Effects of chocolate, cocoa, and flavan-3-ols on cardiovascular health: a systematic review and meta-analysis of randomized trials. Am. J. Clin. Nutr.. 2012, 95, 740-751.
  49. Rahul K. Lall; Deeba N. Syed; Vaqar M. Adhami; Mohammad Imran Khan; Hasan Mukhtar; Dietary Polyphenols in Prevention and Treatment of Prostate Cancer. Int. J. Mol. Sci.. 2015, 16, 3350-3376.
  50. Mateus de Lima Macena; Lara Fernanda da Silva Nunes; Andreza Ferreira da Silva; Isabele Rejane Oliveira Maranhão Pureza; Dafiny Rodrigues Silva Praxedes; Juliana Célia de Farias Santos; Nassib Bezerra Bueno; Effects of dietary polyphenols in the glycemic, renal, inflammatory, and oxidative stress biomarkers in diabetic nephropathy: a systematic review with meta-analysis of randomized controlled trials. Nutr. Rev.. 2022, 80, 2237-2259.
  51. Chiara Colizzi; The protective effects of polyphenols on Alzheimer's disease: A systematic review. Alzheimer's Dementia: Transl. Res. Clin. Interv.. 2018, 5, 184-196.
  52. Fang Liu; Deming Li; Xinjing Wang; Yuan Cui; Xinli Li; Polyphenols intervention is an effective strategy to ameliorate inflammatory bowel disease: a systematic review and meta-analysis. Int. J. Food Sci. Nutr.. 2020, 72, 14-25.
  53. Siyu He; Hong Jiang; Caili Zhuo; Wei Jiang; Trimethylamine/Trimethylamine-N-Oxide as a Key Between Diet and Cardiovascular Diseases. Cardiovasc. Toxicol.. 2021, 21, 593-604.
  54. Doudou Li; Ying Lu; Shuai Yuan; Xiaxia Cai; Yuan He; Jie Chen; Qiong Wu; Di He; Aiping Fang; Yacong Bo; Peige Song; Debby Bogaert; Kostas Tsilidis; Susanna C Larsson; Huanling Yu; Huilian Zhu; Evropi Theodoratou; Yimin Zhu; Xue Li; Gut microbiota–derived metabolite trimethylamine-N-oxide and multiple health outcomes: an umbrella review and updated meta-analysis. Am. J. Clin. Nutr.. 2022, 116, 230-243.
  55. Chao Liang; Han-Chi Tseng; Hui-Mei Chen; Wei-Chi Wang; Chih-Min Chiu; Jen-Yun Chang; Kuan-Yi Lu; Shun-Long Weng; Tzu-Hao Chang; Chao-Hsiang Chang; Chen-Tsung Weng; Hwei-Ming Wang; Hsien-Da Huang; Diversity and enterotype in gut bacterial community of adults in Taiwan. BMC Genom.. 2017, 18, 1-11.
  56. Chao Kang; Yong Zhang; Xiaohui Zhu; Kai Liu; Xiaolan Wang; Mengting Chen; Jian Wang; Hua Chen; Suocheng Hui; Li Huang; Qianyong Zhang; Jundong Zhu; Bin Wang; Mantian Mi; Healthy Subjects Differentially Respond to Dietary Capsaicin Correlating with Specific Gut Enterotypes. J. Clin. Endocrinol. Metab.. 2016, 101, 4681-4689.
  57. Mi Young Lim; Mina Rho; Yun-Mi Song; Kayoung Lee; Joohon Sung; GwangPyo Ko; Stability of Gut Enterotypes in Korean Monozygotic Twins and Their Association with Biomarkers and Diet. Sci. Rep.. 2014, 4, 7348.
  58. Tingting Chen; Wenmin Long; Chenhong Zhang; Shuang Liu; Liping Zhao; Bruce R. Hamaker; Fiber-utilizing capacity varies in Prevotella- versus Bacteroides-dominated gut microbiota. Sci. Rep.. 2017, 7, 1-7.
  59. Nehal Adel Abdelsalam; Shaimaa M. Hegazy; Ramy K. Aziz; The curious case of Prevotella copri. Gut Microbes. 2023, 15, 2249152.
  60. Hassan Zafar; Milton H. Saier Jr; Gut Bacteroides species in health and disease. Gut Microbes. 2021, 13, 1-20.
  61. Suguru Nishijima; Wataru Suda; Kenshiro Oshima; Seok-Won Kim; Yuu Hirose; Hidetoshi Morita; Masahira Hattori; The gut microbiome of healthy Japanese and its microbial and functional uniqueness. DNA Res.. 2016, 23, 125-133.
  62. Paul I. Costea; Falk Hildebrand; Manimozhiyan Arumugam; Fredrik Bäckhed; Martin J. Blaser; Frederic D. Bushman; Willem M. de Vos; S. Dusko Ehrlich; Claire M. Fraser; Masahira Hattori; Curtis Huttenhower; Ian B. Jeffery; Dan Knights; James D. Lewis; Ruth E. Ley; Howard Ochman; Paul W. O’toole; Christopher Quince; David A. Relman; Fergus Shanahan; Shinichi Sunagawa; Jun Wang; George M. Weinstock; Gary D. Wu; Georg Zeller; Liping Zhao; Jeroen Raes; Rob Knight; Peer Bork; Enterotypes in the landscape of gut microbial community composition. Nat. Microbiol.. 2017, 3, 8-16.
  63. Sara Vieira-Silva; Gwen Falony; Youssef Darzi; Gipsi Lima-Mendez; Roberto Garcia Yunta; Shujiro Okuda; Doris Vandeputte; Mireia Valles-Colomer; Falk Hildebrand; Samuel Chaffron; Jeroen Raes; Species–function relationships shape ecological properties of the human gut microbiome. Nat. Microbiol.. 2016, 1, 16088-16088.
  64. Jing Li; Ruiqing Fu; Yajun Yang; Hans-Peter Horz; Yaqun Guan; Yan Lu; Haiyi Lou; Lei Tian; Shijie Zheng; Hongjiao Liu; Meng Shi; Kun Tang; Sijia Wang; Shuhua Xu; A metagenomic approach to dissect the genetic composition of enterotypes in Han Chinese and two Muslim groups. Syst. Appl. Microbiol.. 2017, 41, 1-12.
  65. M F Hjorth; H M Roager; T M Larsen; S K Poulsen; T R Licht; M I Bahl; Y Zohar; A Astrup; Pre-treatment microbial Prevotella-to-Bacteroides ratio, determines body fat loss success during a 6-month randomized controlled diet intervention. Int. J. Obes.. 2017, 42, 580-583.
  66. Mads F. Hjorth; Trine Blædel; Line Q. Bendtsen; Janne K. Lorenzen; Jacob B. Holm; Pia Kiilerich; Henrik M. Roager; Karsten Kristiansen; Lesli H. Larsen; Arne Astrup; Prevotella-to-Bacteroides ratio predicts body weight and fat loss success on 24-week diets varying in macronutrient composition and dietary fiber: results from a post-hoc analysis. Int. J. Obes.. 2018, 43, 149-157.
  67. Lars Christensen; Mads F. Hjorth; Lukasz Krych; Tine Rask Licht; Lotte Lauritzen; Faidon Magkos; Henrik M. Roager; Prevotella abundance and salivary amylase gene copy number predict fat loss in response to wholegrain diets. Front. Nutr.. 2022, 9, 947349.
  68. Mads F Hjorth; Lars Christensen; Thomas M Larsen; Henrik M Roager; Lukasz Krych; Witold Kot; Dennis S Nielsen; Christian Ritz; Arne Astrup; Pretreatment Prevotella-to-Bacteroides ratio and salivary amylase gene copy number as prognostic markers for dietary weight loss. Am. J. Clin. Nutr.. 2020, 111, 1079-1086.
  69. Lars Christensen; Stine Vuholm; Henrik M Roager; Dennis S Nielsen; Lukasz Krych; Mette Kristensen; Arne Astrup; Mads F Hjorth; Prevotella Abundance Predicts Weight Loss Success in Healthy, Overweight Adults Consuming a Whole-Grain Diet Ad Libitum: A Post Hoc Analysis of a 6-Wk Randomized Controlled Trial. J. Nutr.. 2019, 149, 2174-2181.
  70. Lars Christensen; Claudia V. Sørensen; Frederikke U. Wøhlk; Louise Kjølbæk; Arne Astrup; Yolanda Sanz; Mads F. Hjorth; Alfonso Benítez-Páez; Microbial enterotypes beyond genus level: Bacteroides species as a predictive biomarker for weight change upon controlled intervention with arabinoxylan oligosaccharides in overweight subjects. Gut Microbes. 2020, 12, 1847627.
More
Video Production Service