User menu

Accès à distance ? S'identifier sur le proxy UCLouvain

Microbial regulation of organismal energy homeostasis

  • Open access
  • PDF
  • 3.49 M
  1. Cani Patrice D, Human gut microbiome: hopes, threats and promises, 10.1136/gutjnl-2018-316723
  2. Barr Jeremy J., A bacteriophages journey through the human body, 10.1111/imr.12565
  3. Forde Amanda, Hill Colin, Phages of life - the path to pharma : Phages of life, 10.1111/bph.14106
  4. Sender Ron, Fuchs Shai, Milo Ron, Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans, 10.1016/j.cell.2016.01.013
  5. Li Junhua, , Jia Huijue, Cai Xianghang, Zhong Huanzi, Feng Qiang, Sunagawa Shinichi, Arumugam Manimozhiyan, Kultima Jens Roat, Prifti Edi, Nielsen Trine, Juncker Agnieszka Sierakowska, Manichanh Chaysavanh, Chen Bing, Zhang Wenwei, Levenez Florence, Wang Juan, Xu Xun, Xiao Liang, Liang Suisha, Zhang Dongya, Zhang Zhaoxi, Chen Weineng, Zhao Hailong, Al-Aama Jumana Yousuf, Edris Sherif, Yang Huanming, Wang Jian, Hansen Torben, Nielsen Henrik Bjørn, Brunak Søren, Kristiansen Karsten, Guarner Francisco, Pedersen Oluf, Doré Joel, Ehrlich S Dusko, Bork Peer, Wang Jun, An integrated catalog of reference genes in the human gut microbiome, 10.1038/nbt.2942
  6. Qin Junjie, , Li Ruiqiang, Raes Jeroen, Arumugam Manimozhiyan, Burgdorf Kristoffer Solvsten, Manichanh Chaysavanh, Nielsen Trine, Pons Nicolas, Levenez Florence, Yamada Takuji, Mende Daniel R., Li Junhua, Xu Junming, Li Shaochuan, Li Dongfang, Cao Jianjun, Wang Bo, Liang Huiqing, Zheng Huisong, Xie Yinlong, Tap Julien, Lepage Patricia, Bertalan Marcelo, Batto Jean-Michel, Hansen Torben, Le Paslier Denis, Linneberg Allan, Nielsen H. Bjørn, Pelletier Eric, Renault Pierre, Sicheritz-Ponten Thomas, Turner Keith, Zhu Hongmei, Yu Chang, Li Shengting, Jian Min, Zhou Yan, Li Yingrui, Zhang Xiuqing, Li Songgang, Qin Nan, Yang Huanming, Wang Jian, Brunak Søren, Doré Joel, Guarner Francisco, Kristiansen Karsten, Pedersen Oluf, Parkhill Julian, Weissenbach Jean, Bork Peer, Ehrlich S. Dusko, Wang Jun, A human gut microbial gene catalogue established by metagenomic sequencing, 10.1038/nature08821
  7. Le Chatelier Emmanuelle, , Nielsen Trine, Qin Junjie, Prifti Edi, Hildebrand Falk, Falony Gwen, Almeida Mathieu, Arumugam Manimozhiyan, Batto Jean-Michel, Kennedy Sean, Leonard Pierre, Li Junhua, Burgdorf Kristoffer, Grarup Niels, Jørgensen Torben, Brandslund Ivan, Nielsen Henrik Bjørn, Juncker Agnieszka S., Bertalan Marcelo, Levenez Florence, Pons Nicolas, Rasmussen Simon, Sunagawa Shinichi, Tap Julien, Tims Sebastian, Zoetendal Erwin G., Brunak Søren, Clément Karine, Doré Joël, Kleerebezem Michiel, Kristiansen Karsten, Renault Pierre, Sicheritz-Ponten Thomas, de Vos Willem M., Zucker Jean-Daniel, Raes Jeroen, Hansen Torben, Bork Peer, Wang Jun, Ehrlich S. Dusko, Pedersen Oluf, Richness of human gut microbiome correlates with metabolic markers, 10.1038/nature12506
  8. Cotillard Aurélie, , Kennedy Sean P., Kong Ling Chun, Prifti Edi, Pons Nicolas, Le Chatelier Emmanuelle, Almeida Mathieu, Quinquis Benoit, Levenez Florence, Galleron Nathalie, Gougis Sophie, Rizkalla Salwa, Batto Jean-Michel, Renault Pierre, Doré Joel, Zucker Jean-Daniel, Clément Karine, Ehrlich Stanislav Dusko, , Dietary intervention impact on gut microbial gene richness, 10.1038/nature12480
  9. Dao Maria Carlota, Everard Amandine, Aron-Wisnewsky Judith, Sokolovska Nataliya, Prifti Edi, Verger Eric O, Kayser Brandon D, Levenez Florence, Chilloux Julien, Hoyles Lesley, Dumas Marc-Emmanuel, Rizkalla Salwa W, Doré Joel, Cani Patrice D, Clément Karine, , Akkermansia muciniphilaand improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology, 10.1136/gutjnl-2014-308778
  10. Vandeputte, D. et al. Quantitative microbiome profiling links gut community variation to microbial load. Nature 551, 507–511 (2017).
  11. Turnbaugh Peter J., Ley Ruth E., Mahowald Michael A., Magrini Vincent, Mardis Elaine R., Gordon Jeffrey I., An obesity-associated gut microbiome with increased capacity for energy harvest, 10.1038/nature05414
  12. Backhed F., Ding H., Wang T., Hooper L. V., Koh G. Y., Nagy A., Semenkovich C. F., Gordon J. I., The gut microbiota as an environmental factor that regulates fat storage, 10.1073/pnas.0407076101
  13. Wichmann Anita, Allahyar Ava, Greiner Thomas U., Plovier Hubert, Lundén Gunnel Östergren, Larsson Thomas, Drucker Daniel J., Delzenne Nathalie M., Cani Patrice D., Bäckhed Fredrik, Microbial Modulation of Energy Availability in the Colon Regulates Intestinal Transit, 10.1016/j.chom.2013.09.012
  14. Turnbaugh Peter J., Hamady Micah, Yatsunenko Tanya, Cantarel Brandi L., Duncan Alexis, Ley Ruth E., Sogin Mitchell L., Jones William J., Roe Bruce A., Affourtit Jason P., Egholm Michael, Henrissat Bernard, Heath Andrew C., Knight Rob, Gordon Jeffrey I., A core gut microbiome in obese and lean twins, 10.1038/nature07540
  15. Turnbaugh P. J., Ridaura V. K., Faith J. J., Rey F. E., Knight R., Gordon J. I., The Effect of Diet on the Human Gut Microbiome: A Metagenomic Analysis in Humanized Gnotobiotic Mice, 10.1126/scitranslmed.3000322
  16. Dewulf Evelyne M, Cani Patrice D, Claus Sandrine P, Fuentes Susana, Puylaert Philippe GB, Neyrinck Audrey M, Bindels Laure B, de Vos Willem M, Gibson Glenn R, Thissen Jean-Paul, Delzenne Nathalie M, Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women, 10.1136/gutjnl-2012-303304
  17. Salazar Nuria, Dewulf Evelyne M., Neyrinck Audrey M., Bindels Laure B., Cani Patrice D., Mahillon Jacques, de Vos Willem M., Thissen Jean-Paul, Gueimonde Miguel, de los Reyes-Gavilán Clara G., Delzenne Nathalie M., Inulin-type fructans modulate intestinal Bifidobacterium species populations and decrease fecal short-chain fatty acids in obese women, 10.1016/j.clnu.2014.06.001
  18. Ridaura V. K., Faith J. J., Rey F. E., Cheng J., Duncan A. E., Kau A. L., Griffin N. W., Lombard V., Henrissat B., Bain J. R., Muehlbauer M. J., Ilkayeva O., Semenkovich C. F., Funai K., Hayashi D. K., Lyle B. J., Martini M. C., Ursell L. K., Clemente J. C., Van Treuren W., Walters W. A., Knight R., Newgard C. B., Heath A. C., Gordon J. I., Gut Microbiota from Twins Discordant for Obesity Modulate Metabolism in Mice, 10.1126/science.1241214
  19. Brahe L. K., Astrup A., Larsen L. H., Is butyrate the link between diet, intestinal microbiota and obesity-related metabolic diseases? : Butyrate and obesity-related diseases, 10.1111/obr.12068
  20. Neis Evelien PJG, van Eijk Hans MH, Lenaerts Kaatje, Olde Damink Steven WM, Blaak Ellen E, Dejong Cornelis HC, Rensen Sander S, Distal versus proximal intestinal short-chain fatty acid release in man, 10.1136/gutjnl-2018-316161
  21. De Vadder Filipe, Kovatcheva-Datchary Petia, Goncalves Daisy, Vinera Jennifer, Zitoun Carine, Duchampt Adeline, Bäckhed Fredrik, Mithieux Gilles, Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits, 10.1016/j.cell.2013.12.016
  22. RÉMÉSY C., DEMIGNÉ C., CHARTIER F., Origin and utilization of volatile fatty acids in the rat, 10.1051/rnd:19800725
  23. Singh Vishal, Chassaing Benoit, Zhang Limin, San Yeoh Beng, Xiao Xia, Kumar Manish, Baker Mark T., Cai Jingwei, Walker Rachel, Borkowski Kamil, Harvatine Kevin J., Singh Nagendra, Shearer Gregory C., Ntambi James M., Joe Bina, Patterson Andrew D., Gewirtz Andrew T., Vijay-Kumar Matam, Microbiota-Dependent Hepatic Lipogenesis Mediated by Stearoyl CoA Desaturase 1 (SCD1) Promotes Metabolic Syndrome in TLR5-Deficient Mice, 10.1016/j.cmet.2015.09.028
  24. Schwarzer M., Makki K., Storelli G., Machuca-Gayet I., Srutkova D., Hermanova P., Martino M. E., Balmand S., Hudcovic T., Heddi A., Rieusset J., Kozakova H., Vidal H., Leulier F., Lactobacillus plantarum strain maintains growth of infant mice during chronic undernutrition, 10.1126/science.aad8588
  25. Smythe P.M., CHANGES IN INTESTINAL BACTERIAL FLORA AND ROLE OF INFECTION IN KWASHIORKOR, 10.1016/s0140-6736(58)91336-9
  26. Gupta Sourav, Mohammed Monzoorul, Ghosh Tarini, Kanungo Suman, Nair Gopinath, Mande Sharmila S, Metagenome of the gut of a malnourished child, 10.1186/1757-4749-3-7
  27. Monira Shirajum, Nakamura Shota, Gotoh Kazuyoshi, Izutsu Kaori, Watanabe Haruo, Alam Nur Haque, Endtz Hubert Ph., Cravioto Alejandro, Ali Sk. Imran, Nakaya Takaaki, Horii Toshihiro, Iida Tetsuya, Alam Munirul, Gut Microbiota of Healthy and Malnourished Children in Bangladesh, 10.3389/fmicb.2011.00228
  28. Ghosh Tarini Shankar, Sen Gupta Sourav, Bhattacharya Tanudeep, Yadav Deepak, Barik Anamitra, Chowdhury Abhijit, Das Bhabatosh, Mande Sharmila S., Nair G. Balakrish, Gut Microbiomes of Indian Children of Varying Nutritional Status, 10.1371/journal.pone.0095547
  29. Subramanian Sathish, Huq Sayeeda, Yatsunenko Tanya, Haque Rashidul, Mahfuz Mustafa, Alam Mohammed A., Benezra Amber, DeStefano Joseph, Meier Martin F., Muegge Brian D., Barratt Michael J., VanArendonk Laura G., Zhang Qunyuan, Province Michael A., Petri Jr William A., Ahmed Tahmeed, Gordon Jeffrey I., Persistent gut microbiota immaturity in malnourished Bangladeshi children, 10.1038/nature13421
  30. De Filippo C., Cavalieri D., Di Paola M., Ramazzotti M., Poullet J. B., Massart S., Collini S., Pieraccini G., Lionetti P., Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa, 10.1073/pnas.1005963107
  31. Byndloss Mariana X., Olsan Erin E., Rivera-Chávez Fabian, Tiffany Connor R., Cevallos Stephanie A., Lokken Kristen L., Torres Teresa P., Byndloss Austin J., Faber Franziska, Gao Yandong, Litvak Yael, Lopez Christopher A., Xu Gege, Napoli Eleonora, Giulivi Cecilia, Tsolis Renée M., Revzin Alexander, Lebrilla Carlito B., Bäumler Andreas J., Microbiota-activated PPAR-γ signaling inhibits dysbiotic Enterobacteriaceae expansion, 10.1126/science.aam9949
  32. Cani Patrice D., Gut cell metabolism shapes the microbiome, 10.1126/science.aao2202
  33. Waterson Michael J., Horvath Tamas L., Neuronal Regulation of Energy Homeostasis: Beyond the Hypothalamus and Feeding, 10.1016/j.cmet.2015.09.026
  34. Williams Kevin W, Elmquist Joel K, From neuroanatomy to behavior: central integration of peripheral signals regulating feeding behavior, 10.1038/nn.3217
  35. Suzuki Keisuke, Jayasena Channa N., Bloom Stephen R., Obesity and Appetite Control, 10.1155/2012/824305
  36. Cani Patrice D., Dewever Cédric, Delzenne Nathalie M., Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats, 10.1079/bjn20041225
  37. Delzenne, N. M., Cani, P. D., Daubioul, C. & Neyrinck, A. M. Impact of inulin and oligofructose on gastrointestinal peptides. Br. J. Nutr. 93 (Suppl 1), S157–S161 (2005).
  38. Cani Patrice D., Neyrinck Audrey M., Maton Nicole, Delzenne Nathalie M., Oligofructose Promotes Satiety in Rats Fed a High-Fat Diet: Involvement of Glucagon-Like Peptide-1, 10.1038/oby.2005.117
  39. Cani P D, Joly E, Horsmans Y, Delzenne N M, Oligofructose promotes satiety in healthy human: a pilot study, 10.1038/sj.ejcn.1602350
  40. Cani Patrice D., Hoste Sophie, Guiot Yves, Delzenne Nathalie M., Dietary non-digestible carbohydrates promote L-cell differentiation in the proximal colon of rats, 10.1017/s0007114507691648
  41. Karaki Shin-ichiro, Mitsui Retsu, Hayashi Hisayoshi, Kato Ikuo, Sugiya Hiroshi, Iwanaga Toshihiko, Furness John B., Kuwahara Atsukazu, Short-chain fatty acid receptor, GPR43, is expressed by enteroendocrine cells and mucosal mast cells in rat intestine, 10.1007/s00441-005-0140-x
  42. Karaki Shin-ichiro, Tazoe Hideaki, Hayashi Hisayoshi, Kashiwabara Hidefumi, Tooyama Kazunari, Suzuki Yuichi, Kuwahara Atsukazu, Expression of the short-chain fatty acid receptor, GPR43, in the human colon, 10.1007/s10735-007-9145-y
  43. Tazoe Hideaki, Otomo Yasuko, Karaki Shin-ichiro, Kato Ikuo, Fukami Yasuyuki, Terasaki Masaki, Kuwahara Atsukazu, Expression of short-chain fatty acid receptor GPR41 in the human colon, 10.2220/biomedres.30.149
  44. Tolhurst G., Heffron H., Lam Y. S., Parker H. E., Habib A. M., Diakogiannaki E., Cameron J., Grosse J., Reimann F., Gribble F. M., Short-Chain Fatty Acids Stimulate Glucagon-Like Peptide-1 Secretion via the G-Protein-Coupled Receptor FFAR2, 10.2337/db11-1019
  45. Psichas A, Sleeth M L, Murphy K G, Brooks L, Bewick G A, Hanyaloglu A C, Ghatei M A, Bloom S R, Frost G, The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents, 10.1038/ijo.2014.153
  46. Ronveaux Charlotte C, Tomé Daniel, Raybould Helen E, Glucagon-Like Peptide 1 Interacts with Ghrelin and Leptin to Regulate Glucose Metabolism and Food Intake through Vagal Afferent Neuron Signaling, 10.3945/jn.114.206029
  47. Hagemann Dirk, Holst Jens J., Gethmann Arnica, Banasch Matthias, Schmidt Wolfgang E., Meier Juris J., Glucagon-like peptide 1 (GLP-1) suppresses ghrelin levels in humans via increased insulin secretion, 10.1016/j.regpep.2007.03.002
  48. Murphy Kevin G., Bloom Stephen R., Gut hormones and the regulation of energy homeostasis, 10.1038/nature05484
  49. Batterham Rachel L., Cowley Michael A., Small Caroline J., Herzog Herbert, Cohen Mark A., Dakin Catherine L., Wren Alison M., Brynes Audrey E., Low Malcolm J., Ghatei Mohammad A., Cone Roger D., Bloom Stephen R., Gut hormone PYY3-36 physiologically inhibits food intake, 10.1038/nature00887
  50. Kanoski Scott E., Hayes Matthew R., Skibicka Karolina P., GLP-1 and weight loss: unraveling the diverse neural circuitry, 10.1152/ajpregu.00520.2015
  51. Cani Patrice D., Knauf Claude, How gut microbes talk to organs: The role of endocrine and nervous routes, 10.1016/j.molmet.2016.05.011
  52. Rastelli Marialetizia, Knauf Claude, Cani Patrice D., Gut Microbes and Health: A Focus on the Mechanisms Linking Microbes, Obesity, and Related Disorders : Mechanisms Linking Microbes and Obesity, 10.1002/oby.22175
  53. Lin Hua V., Frassetto Andrea, Kowalik Jr Edward J., Nawrocki Andrea R., Lu Mofei M., Kosinski Jennifer R., Hubert James A., Szeto Daphne, Yao Xiaorui, Forrest Gail, Marsh Donald J., Butyrate and Propionate Protect against Diet-Induced Obesity and Regulate Gut Hormones via Free Fatty Acid Receptor 3-Independent Mechanisms, 10.1371/journal.pone.0035240
  54. Chambers Edward S, Viardot Alexander, Psichas Arianna, Morrison Douglas J, Murphy Kevin G, Zac-Varghese Sagen E K, MacDougall Kenneth, Preston Tom, Tedford Catriona, Finlayson Graham S, Blundell John E, Bell Jimmy D, Thomas E Louise, Mt-Isa Shahrul, Ashby Deborah, Gibson Glen R, Kolida Sofia, Dhillo Waljit S, Bloom Stephen R, Morley Wayne, Clegg Stuart, Frost Gary, Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults, 10.1136/gutjnl-2014-307913
  55. Li Zhuang, Yi Chun-Xia, Katiraei Saeed, Kooijman Sander, Zhou Enchen, Chung Chih Kit, Gao Yuanqing, van den Heuvel José K, Meijer Onno C, Berbée Jimmy F P, Heijink Marieke, Giera Martin, Willems van Dijk Ko, Groen Albert K, Rensen Patrick C N, Wang Yanan, Butyrate reduces appetite and activates brown adipose tissue via the gut-brain neural circuit, 10.1136/gutjnl-2017-314050
  56. Goswami Chayon, Iwasaki Yusaku, Yada Toshihiko, Short-chain fatty acids suppress food intake by activating vagal afferent neurons, 10.1016/j.jnutbio.2018.03.009
  57. Frost Gary, Sleeth Michelle L., Sahuri-Arisoylu Meliz, Lizarbe Blanca, Cerdan Sebastian, Brody Leigh, Anastasovska Jelena, Ghourab Samar, Hankir Mohammed, Zhang Shuai, Carling David, Swann Jonathan R., Gibson Glenn, Viardot Alexander, Morrison Douglas, Louise Thomas E, Bell Jimmy D., The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism, 10.1038/ncomms4611
  58. Tennoune N, Chan P, Breton J, Legrand R, Chabane Y N, Akkermann K, Järv A, Ouelaa W, Takagi K, Ghouzali I, Francois M, Lucas N, Bole-Feysot C, Pestel-Caron M, do Rego J-C, Vaudry D, Harro J, Dé E, Déchelotte P, Fetissov S O, Bacterial ClpB heat-shock protein, an antigen-mimetic of the anorexigenic peptide α-MSH, at the origin of eating disorders, 10.1038/tp.2014.98
  59. Breton Jonathan, Tennoune Naouel, Lucas Nicolas, Francois Marie, Legrand Romain, Jacquemot Justine, Goichon Alexis, Guérin Charlène, Peltier Johann, Pestel-Caron Martine, Chan Philippe, Vaudry David, do Rego Jean-Claude, Liénard Fabienne, Pénicaud Luc, Fioramonti Xavier, Ebenezer Ivor S., Hökfelt Tomas, Déchelotte Pierre, Fetissov Sergueï O., Gut Commensal E. coli Proteins Activate Host Satiety Pathways following Nutrient-Induced Bacterial Growth, 10.1016/j.cmet.2015.10.017
  60. Strandwitz, P. Neurotransmitter modulation by the gut microbiota. Brain Res. 1693 (Pt B), 128–133 (2018).
  61. Yano Jessica M., Yu Kristie, Donaldson Gregory P., Shastri Gauri G., Ann Phoebe, Ma Liang, Nagler Cathryn R., Ismagilov Rustem F., Mazmanian Sarkis K., Hsiao Elaine Y., Indigenous Bacteria from the Gut Microbiota Regulate Host Serotonin Biosynthesis, 10.1016/j.cell.2015.02.047
  62. Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney R D, Shanahan F, Dinan T G, Cryan J F, The microbiome-gut-brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner, 10.1038/mp.2012.77
  63. Kelly John R., Borre Yuliya, O' Brien Ciaran, Patterson Elaine, El Aidy Sahar, Deane Jennifer, Kennedy Paul J., Beers Sasja, Scott Karen, Moloney Gerard, Hoban Alan E., Scott Lucinda, Fitzgerald Patrick, Ross Paul, Stanton Catherine, Clarke Gerard, Cryan John F., Dinan Timothy G., Transferring the blues: Depression-associated gut microbiota induces neurobehavioural changes in the rat, 10.1016/j.jpsychires.2016.07.019
  64. Zheng P, Zeng B, Zhou C, Liu M, Fang Z, Xu X, Zeng L, Chen J, Fan S, Du X, Zhang X, Yang D, Yang Y, Meng H, Li W, Melgiri N D, Licinio J, Wei H, Xie P, Gut microbiome remodeling induces depressive-like behaviors through a pathway mediated by the host’s metabolism, 10.1038/mp.2016.44
  65. De Palma Giada, Lynch Michael D. J., Lu Jun, Dang Vi T., Deng Yikang, Jury Jennifer, Umeh Genevieve, Miranda Pedro M., Pigrau Pastor Marc, Sidani Sacha, Pinto-Sanchez Maria Ines, Philip Vivek, McLean Peter G., Hagelsieb Moreno-Gabriel, Surette Michael G., Bergonzelli Gabriela E., Verdu Elena F., Britz-McKibbin Philip, Neufeld Josh D., Collins Stephen M., Bercik Premysl, Transplantation of fecal microbiota from patients with irritable bowel syndrome alters gut function and behavior in recipient mice, 10.1126/scitranslmed.aaf6397
  66. Sampson Timothy R., Debelius Justine W., Thron Taren, Janssen Stefan, Shastri Gauri G., Ilhan Zehra Esra, Challis Collin, Schretter Catherine E., Rocha Sandra, Gradinaru Viviana, Chesselet Marie-Francoise, Keshavarzian Ali, Shannon Kathleen M., Krajmalnik-Brown Rosa, Wittung-Stafshede Pernilla, Knight Rob, Mazmanian Sarkis K., Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease, 10.1016/j.cell.2016.11.018
  67. Minter Myles R., Zhang Can, Leone Vanessa, Ringus Daina L., Zhang Xiaoqiong, Oyler-Castrillo Paul, Musch Mark W., Liao Fan, Ward Joseph F., Holtzman David M., Chang Eugene B., Tanzi Rudolph E., Sisodia Sangram S., Antibiotic-induced perturbations in gut microbial diversity influences neuro-inflammation and amyloidosis in a murine model of Alzheimer’s disease, 10.1038/srep30028
  68. Berer Kerstin, Gerdes Lisa Ann, Cekanaviciute Egle, Jia Xiaoming, Xiao Liang, Xia Zhongkui, Liu Chuan, Klotz Luisa, Stauffer Uta, Baranzini Sergio E., Kümpfel Tania, Hohlfeld Reinhard, Krishnamoorthy Gurumoorthy, Wekerle Hartmut, Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice, 10.1073/pnas.1711233114
  69. Hsiao Elaine Y., McBride Sara W., Hsien Sophia, Sharon Gil, Hyde Embriette R., McCue Tyler, Codelli Julian A., Chow Janet, Reisman Sarah E., Petrosino Joseph F., Patterson Paul H., Mazmanian Sarkis K., Microbiota Modulate Behavioral and Physiological Abnormalities Associated with Neurodevelopmental Disorders, 10.1016/j.cell.2013.11.024
  70. Castro Daniel C., Cole Shannon L., Berridge Kent C., Lateral hypothalamus, nucleus accumbens, and ventral pallidum roles in eating and hunger: interactions between homeostatic and reward circuitry, 10.3389/fnsys.2015.00090
  71. Ding Li, Zhang Jin, Glucagon-like peptide-1 activates endothelial nitric oxide synthase in human umbilical vein endothelial cells, 10.1038/aps.2011.149
  72. Rotondo Alessandra, Amato Antonella, Lentini Laura, Baldassano Sara, Mulè Flavia, Glucagon-like peptide-1 relaxes gastric antrum through nitric oxide in mice, 10.1016/j.peptides.2010.09.028
  73. Grasset Estelle, Puel Anthony, Charpentier Julie, Collet Xavier, Christensen Jeffrey E., Tercé François, Burcelin Rémy, A Specific Gut Microbiota Dysbiosis of Type 2 Diabetic Mice Induces GLP-1 Resistance through an Enteric NO-Dependent and Gut-Brain Axis Mechanism, 10.1016/j.cmet.2017.04.013
  74. Catry Emilie, Bindels Laure B, Tailleux Anne, Lestavel Sophie, Neyrinck Audrey M, Goossens Jean-François, Lobysheva Irina, Plovier Hubert, Essaghir Ahmed, Demoulin Jean-Baptiste, Bouzin Caroline, Pachikian Barbara D, Cani Patrice D, Staels Bart, Dessy Chantal, Delzenne Nathalie M, Targeting the gut microbiota with inulin-type fructans: preclinical demonstration of a novel approach in the management of endothelial dysfunction, 10.1136/gutjnl-2016-313316
  75. Brandl Katharina, Kumar Vipin, Eckmann Lars, Gut-liver axis at the frontier of host-microbial interactions, 10.1152/ajpgi.00361.2016
  76. Tilg Herbert, Cani Patrice D, Mayer Emeran A, Gut microbiome and liver diseases, 10.1136/gutjnl-2016-312729
  77. Schramm Christoph, Bile Acids, the Microbiome, Immunity, and Liver Tumors, 10.1056/nejmcibr1807106
  78. Chevre Raphael, Silvestre-Roig Carlos, Soehnlein Oliver, Nutritional Modulation of Innate Immunity: The Fat–Bile–Gut Connection, 10.1016/j.tem.2018.08.002
  79. Pathak Preeti, Xie Cen, Nichols Robert G., Ferrell Jessica M., Boehme Shannon, Krausz Kristopher W., Patterson Andrew D., Gonzalez Frank J., Chiang John Y.L., Intestine farnesoid X receptor agonist and the gut microbiota activate G-protein bile acid receptor-1 signaling to improve metabolism, 10.1002/hep.29857
  80. Ma Chi, Han Miaojun, Heinrich Bernd, Fu Qiong, Zhang Qianfei, Sandhu Milan, Agdashian David, Terabe Masaki, Berzofsky Jay A., Fako Valerie, Ritz Thomas, Longerich Thomas, Theriot Casey M., McCulloch John A., Roy Soumen, Yuan Wuxing, Thovarai Vishal, Sen Shurjo K., Ruchirawat Mathuros, Korangy Firouzeh, Wang Xin Wei, Trinchieri Giorgio, Greten Tim F., Gut microbiome–mediated bile acid metabolism regulates liver cancer via NKT cells, 10.1126/science.aan5931
  81. Fiorucci Stefano, Mencarelli Andrea, Palladino Giuseppe, Cipriani Sabrina, Bile-acid-activated receptors: targeting TGR5 and farnesoid-X-receptor in lipid and glucose disorders, 10.1016/j.tips.2009.08.001
  82. Watanabe Mitsuhiro, Houten Sander M., Mataki Chikage, Christoffolete Marcelo A., Kim Brian W., Sato Hiroyuki, Messaddeq Nadia, Harney John W., Ezaki Osamu, Kodama Tatsuhiko, Schoonjans Kristina, Bianco Antonio C., Auwerx Johan, Bile acids induce energy expenditure by promoting intracellular thyroid hormone activation, 10.1038/nature04330
  83. Anhê Fernando F, Nachbar Renato T, Varin Thibault V, Trottier Jocelyn, Dudonné Stéphanie, Le Barz Mélanie, Feutry Perrine, Pilon Geneviève, Barbier Olivier, Desjardins Yves, Roy Denis, Marette André, Treatment with camu camu (Myrciaria dubia) prevents obesity by altering the gut microbiota and increasing energy expenditure in diet-induced obese mice, 10.1136/gutjnl-2017-315565
  84. Ziętak Marika, Chabowska-Kita Agnieszka, Kozak Leslie Paul, Brown fat thermogenesis: Stability of developmental programming and transient effects of temperature and gut microbiota in adults, 10.1016/j.biochi.2016.12.006
  85. Chevalier Claire, Stojanović Ozren, Colin Didier J., Suarez-Zamorano Nicolas, Tarallo Valentina, Veyrat-Durebex Christelle, Rigo Dorothée, Fabbiano Salvatore, Stevanović Ana, Hagemann Stefanie, Montet Xavier, Seimbille Yann, Zamboni Nicola, Hapfelmeier Siegfried, Trajkovski Mirko, Gut Microbiota Orchestrates Energy Homeostasis during Cold, 10.1016/j.cell.2015.11.004
  86. Ziętak Marika, Kovatcheva-Datchary Petia, Markiewicz Lidia H., Ståhlman Marcus, Kozak Leslie P., Bäckhed Fredrik, Altered Microbiota Contributes to Reduced Diet-Induced Obesity upon Cold Exposure, 10.1016/j.cmet.2016.05.001
  87. Zhang, X. Y. et al. Huddling remodels gut microbiota to reduce energy requirements in a small mammal species during cold exposure. Microbiome 6, 103 (2018).
  88. Derrien M., Collado M. C., Ben-Amor K., Salminen S., de Vos W. M., The Mucin Degrader Akkermansia muciniphila Is an Abundant Resident of the Human Intestinal Tract, 10.1128/aem.01226-07
  89. Everard A., Belzer C., Geurts L., Ouwerkerk J. P., Druart C., Bindels L. B., Guiot Y., Derrien M., Muccioli G. G., Delzenne N. M., de Vos W. M., Cani P. D., Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity, 10.1073/pnas.1219451110
  90. Schneeberger Marc, Everard Amandine, Gómez-Valadés Alicia G., Matamoros Sébastien, Ramírez Sara, Delzenne Nathalie M., Gomis Ramon, Claret Marc, Cani Patrice D., Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice, 10.1038/srep16643
  91. Cani Patrice D., de Vos Willem M., Next-Generation Beneficial Microbes: The Case of Akkermansia muciniphila, 10.3389/fmicb.2017.01765
  92. Plovier Hubert, Everard Amandine, Druart Céline, Depommier Clara, Van Hul Matthias, Geurts Lucie, Chilloux Julien, Ottman Noora, Duparc Thibaut, Lichtenstein Laeticia, Myridakis Antonis, Delzenne Nathalie M, Klievink Judith, Bhattacharjee Arnab, van der Ark Kees C H, Aalvink Steven, Martinez Laurent O, Dumas Marc-Emmanuel, Maiter Dominique, Loumaye Audrey, Hermans Michel P, Thissen Jean-Paul, Belzer Clara, de Vos Willem M, Cani Patrice D, A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice, 10.1038/nm.4236
  93. Pierre Joseph F., Martinez Kristina B., Ye Honggang, Nadimpalli Anuradha, Morton Timothy C., Yang Jinghui, Wang Qiang, Patno Noelle, Chang Eugene B., Yin Deng Ping, Activation of bile acid signaling improves metabolic phenotypes in high-fat diet-induced obese mice, 10.1152/ajpgi.00202.2016
  94. Gao, X. et al. Polyphenol- and caffeine-rich postfermented pu-erh tea improves diet-induced metabolic syndrome by remodeling intestinal homeostasis in mice. Infect. Immun. 86, e00601–e00617 (2017).
  95. Liu Jie, Li Yanfang, Yang Puyu, Wan Jianchun, Chang Qimeng, Wang Thomas T. Y., Lu Weiying, Zhang Yaqiong, Wang Qin, Yu Liangli Lucy, Gypenosides Reduced the Risk of Overweight and Insulin Resistance in C57BL/6J Mice through Modulating Adipose Thermogenesis and Gut Microbiota, 10.1021/acs.jafc.7b03382
  96. Worthmann Anna, John Clara, Rühlemann Malte C, Baguhl Miriam, Heinsen Femke-Anouska, Schaltenberg Nicola, Heine Markus, Schlein Christian, Evangelakos Ioannis, Mineo Chieko, Fischer Markus, Dandri Maura, Kremoser Claus, Scheja Ludger, Franke Andre, Shaul Philip W, Heeren Joerg, Cold-induced conversion of cholesterol to bile acids in mice shapes the gut microbiome and promotes adaptive thermogenesis, 10.1038/nm.4357
  97. Kim Kyoung-Han, Kim Yun Hye, Son Joe Eun, Lee Ju Hee, Kim Sarah, Choe Min Seon, Moon Joon Ho, Zhong Jian, Fu Kiya, Lenglin Florine, Yoo Jeong-Ah, Bilan Philip J, Klip Amira, Nagy Andras, Kim Jae-Ryong, Park Jin Gyoon, Hussein Samer MI, Doh Kyung-Oh, Hui Chi-chung, Sung Hoon-Ki, Intermittent fasting promotes adipose thermogenesis and metabolic homeostasis via VEGF-mediated alternative activation of macrophage, 10.1038/cr.2017.126
  98. Mattson Mark P., Longo Valter D., Harvie Michelle, Impact of intermittent fasting on health and disease processes, 10.1016/j.arr.2016.10.005
  99. Anton Stephen D., Moehl Keelin, Donahoo William T., Marosi Krisztina, Lee Stephanie A., Mainous Arch G., Leeuwenburgh Christiaan, Mattson Mark P., Flipping the Metabolic Switch: Understanding and Applying the Health Benefits of Fasting : Flipping the Metabolic Switch, 10.1002/oby.22065
  100. Li Guolin, Xie Cen, Lu Siyu, Nichols Robert G., Tian Yuan, Li Licen, Patel Daxeshkumar, Ma Yinyan, Brocker Chad N., Yan Tingting, Krausz Kristopher W., Xiang Rong, Gavrilova Oksana, Patterson Andrew D., Gonzalez Frank J., Intermittent Fasting Promotes White Adipose Browning and Decreases Obesity by Shaping the Gut Microbiota, 10.1016/j.cmet.2017.08.019
  101. Sonoyama K., Fujiwara R., Takemura N., Ogasawara T., Watanabe J., Ito H., Morita T., Response of Gut Microbiota to Fasting and Hibernation in Syrian Hamsters, 10.1128/aem.00692-09
  102. Remely Marlene, Hippe Berit, Geretschlaeger Isabella, Stegmayer Sonja, Hoefinger Ingrid, Haslberger Alexander, Increased gut microbiota diversity and abundance of Faecalibacterium prausnitzii and Akkermansia after fasting: a pilot study, 10.1007/s00508-015-0755-1
  103. Costello Elizabeth K, Gordon Jeffrey I, Secor Stephen M, Knight Rob, Postprandial remodeling of the gut microbiota in Burmese pythons, 10.1038/ismej.2010.71
  104. Belzer Clara, de Vos Willem M, Microbes inside—from diversity to function: the case of Akkermansia, 10.1038/ismej.2012.6
  105. Weitkunat Karolin, Stuhlmann Christin, Postel Anna, Rumberger Sandra, Fankhänel Maria, Woting Anni, Petzke Klaus Jürgen, Gohlke Sabrina, Schulz Tim J., Blaut Michael, Klaus Susanne, Schumann Sara, Short-chain fatty acids and inulin, but not guar gum, prevent diet-induced obesity and insulin resistance through differential mechanisms in mice, 10.1038/s41598-017-06447-x
  106. Gao Z., Yin J., Zhang J., Ward R. E., Martin R. J., Lefevre M., Cefalu W. T., Ye J., Butyrate Improves Insulin Sensitivity and Increases Energy Expenditure in Mice, 10.2337/db08-1637
  107. Lu, Y. et al. Short chain fatty acids prevent high-fat-diet-induced obesity in mice by regulating G protein–coupled receptors and gut microbiota. Sci. Rep. 6, 37589 (2016).
  108. Canfora Emanuel E., Jocken Johan W., Blaak Ellen E., Short-chain fatty acids in control of body weight and insulin sensitivity, 10.1038/nrendo.2015.128
  109. Hanatani Satoko, Motoshima Hiroyuki, Takaki Yuki, Kawasaki Shuji, Igata Motoyuki, Matsumura Takeshi, Kondo Tatsuya, Senokuchi Takafumi, Ishii Norio, Kawashima Junji, Kukidome Daisuke, Shimoda Seiya, Nishikawa Takeshi, Araki Eiichi, Acetate alters expression of genes involved in beige adipogenesis in 3T3-L1 cells and obese KK-Ay mice, 10.3164/jcbn.16-23
  110. Sahuri-Arisoylu M, Brody L P, Parkinson J R, Parkes H, Navaratnam N, Miller A D, Thomas E L, Frost G, Bell J D, Reprogramming of hepatic fat accumulation and 'browning' of adipose tissue by the short-chain fatty acid acetate, 10.1038/ijo.2016.23
  111. Bouter K EC, Bakker G J, Levin E., Hartstra A V, Kootte R S, Udayappan S D, Katiraei S., Bahler L., Gilijamse P. W., Tremaroli V., Stahlman M., Holleman F., van Riel N. A. W., Verberne H J, Romijn J A, Dallinga-Thie G M, Serlie M J, Ackermans M T, Kemper E M, van Dijk Willems K., Backhed F., Groen A K, Nieuwdorp M., Differential metabolic effects of oral butyrate treatment in lean versus metabolic syndrome subjects : , 10.1038/s41424-018-0025-4
  112. Canfora, E. E. et al. Colonic infusions of short-chain fatty acid mixtures promote energy metabolism in overweight/obese men: a randomized crossover trial. Sci. Rep. 7, 2360 (2017).
  113. Chambers Edward S., Byrne Claire S., Aspey Karen, Chen Yanjie, Khan Saadiyah, Morrison Douglas J., Frost Gary, Acute oral sodium propionate supplementation raises resting energy expenditure and lipid oxidation in fasted humans, 10.1111/dom.13159
  114. Rahat-Rozenbloom S, Fernandes J, Gloor G B, Wolever T M S, Evidence for greater production of colonic short-chain fatty acids in overweight than lean humans, 10.1038/ijo.2014.46
  115. Schwiertz Andreas, Taras David, Schäfer Klaus, Beijer Silvia, Bos Nicolaas A., Donus Christiane, Hardt Philip D., Microbiota and SCFA in Lean and Overweight Healthy Subjects, 10.1038/oby.2009.167
  116. Pertwee R G, The pharmacology of cannabinoid receptors and their ligands: an overview, 10.1038/sj.ijo.0803272
  117. Geurts Lucie, Everard Amandine, Van Hul Matthias, Essaghir Ahmed, Duparc Thibaut, Matamoros Sébastien, Plovier Hubert, Castel Julien, Denis Raphael G. P., Bergiers Marie, Druart Céline, Alhouayek Mireille, Delzenne Nathalie M., Muccioli Giulio G., Demoulin Jean-Baptiste, Luquet Serge, Cani Patrice D., Adipose tissue NAPE-PLD controls fat mass development by altering the browning process and gut microbiota, 10.1038/ncomms7495
  118. Pacher P., The Endocannabinoid System as an Emerging Target of Pharmacotherapy, 10.1124/pr.58.3.2
  119. Silvestri Cristoforo, Di Marzo Vincenzo, The Endocannabinoid System in Energy Homeostasis and the Etiopathology of Metabolic Disorders, 10.1016/j.cmet.2013.03.001
  120. Mazier Wilfrid, Saucisse Nicolas, Gatta-Cherifi Blandine, Cota Daniela, The Endocannabinoid System: Pivotal Orchestrator of Obesity and Metabolic Disease, 10.1016/j.tem.2015.07.007
  121. Muccioli, G. et al. The endocannabinoid system links gut microbiota to adipogenesis. Mo. Syst. Biol. 6, 392 (2010).
  122. Cluny Nina L., Keenan Catherine M., Reimer Raylene A., Le Foll Bernard, Sharkey Keith A., Prevention of Diet-Induced Obesity Effects on Body Weight and Gut Microbiota in Mice Treated Chronically with Δ9-Tetrahydrocannabinol, 10.1371/journal.pone.0144270
  123. Rousseaux Christel, Thuru Xavier, Gelot Agathe, Barnich Nicolas, Neut Christel, Dubuquoy Laurent, Dubuquoy Caroline, Merour Emilie, Geboes Karen, Chamaillard Mathias, Ouwehand Arthur, Leyer Greg, Carcano Didier, Colombel Jean-Frédéric, Ardid Denis, Desreumaux Pierre, Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors, 10.1038/nm1521
  124. Cani Patrice D., Plovier Hubert, Van Hul Matthias, Geurts Lucie, Delzenne Nathalie M., Druart Céline, Everard Amandine, Endocannabinoids — at the crossroads between the gut microbiota and host metabolism, 10.1038/nrendo.2015.211
  125. Mikkelsen K. H., Allin K. H., Knop F. K., Effect of antibiotics on gut microbiota, glucose metabolism and body weight regulation: a review of the literature, 10.1111/dom.12637
  126. Cox Laura M., Blaser Martin J., Antibiotics in early life and obesity, 10.1038/nrendo.2014.210
  127. Rodrigues Richard R., Greer Renee L., Dong Xiaoxi, DSouza Karen N., Gurung Manoj, Wu Jia Y., Morgun Andrey, Shulzhenko Natalia, Antibiotic-Induced Alterations in Gut Microbiota Are Associated with Changes in Glucose Metabolism in Healthy Mice, 10.3389/fmicb.2017.02306
  128. Cani P. D., Bibiloni R., Knauf C., Waget A., Neyrinck A. M., Delzenne N. M., Burcelin R., Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet-Induced Obesity and Diabetes in Mice, 10.2337/db07-1403
  129. Cho Ilseung, Yamanishi Shingo, Cox Laura, Methé Barbara A., Zavadil Jiri, Li Kelvin, Gao Zhan, Mahana Douglas, Raju Kartik, Teitler Isabel, Li Huilin, Alekseyenko Alexander V., Blaser Martin J., Antibiotics in early life alter the murine colonic microbiome and adiposity, 10.1038/nature11400
  130. Cox Laura M., Blaser Martin J., Pathways in Microbe-Induced Obesity, 10.1016/j.cmet.2013.05.004
  131. Francino M. P., Antibiotics and the Human Gut Microbiome: Dysbioses and Accumulation of Resistances, 10.3389/fmicb.2015.01543
  132. Reijnders Dorien, Goossens Gijs H., Hermes Gerben D.A., Neis Evelien P.J.G., van der Beek Christina M., Most Jasper, Holst Jens J., Lenaerts Kaatje, Kootte Ruud S., Nieuwdorp Max, Groen Albert K., Olde Damink Steven W.M., Boekschoten Mark V., Smidt Hauke, Zoetendal Erwin G., Dejong Cornelis H.C., Blaak Ellen E., Effects of Gut Microbiota Manipulation by Antibiotics on Host Metabolism in Obese Humans: A Randomized Double-Blind Placebo-Controlled Trial, 10.1016/j.cmet.2016.06.016
  133. Nguyen T. L. A., Vieira-Silva S., Liston A., Raes J., How informative is the mouse for human gut microbiota research?, 10.1242/dmm.017400
  134. Gough E. K., Moodie E. E. M., Prendergast A. J., Johnson S. M. A., Humphrey J. H., Stoltzfus R. J., Walker A. S., Trehan I., Gibb D. M., Goto R., Tahan S., de Morais M. B., Manges A. R., The impact of antibiotics on growth in children in low and middle income countries: systematic review and meta-analysis of randomised controlled trials, 10.1136/bmj.g2267
  135. Million Matthieu, Diallo Aldiouma, Raoult Didier, Gut microbiota and malnutrition, 10.1016/j.micpath.2016.02.003
  136. Alcoba Gabriel, Kerac Marko, Breysse Serge, Salpeteur Cécile, Galetto-Lacour Annick, Briend André, Gervaix Alain, Do Children with Uncomplicated Severe Acute Malnutrition Need Antibiotics? A Systematic Review and Meta-Analysis, 10.1371/journal.pone.0053184
  137. Jacobson Eugene D., Chodos Robert B., Faloon William W., An experimental malabsorption syndrome induced by neomycin, 10.1016/0002-9343(60)90146-7
  138. Munukka Eveliina, Rintala Anniina, Toivonen Raine, Nylund Matts, Yang Baoru, Takanen Anna, Hänninen Arno, Vuopio Jaana, Huovinen Pentti, Jalkanen Sirpa, Pekkala Satu, Faecalibacterium prausnitzii treatment improves hepatic health and reduces adipose tissue inflammation in high-fat fed mice, 10.1038/ismej.2017.24
  139. Maurice Corinne Ferrier, Haiser Henry Joseph, Turnbaugh Peter James, Xenobiotics Shape the Physiology and Gene Expression of the Active Human Gut Microbiome, 10.1016/j.cell.2012.10.052
  140. Saad Rama, Rizkallah Mariam R, Aziz Ramy K, Gut Pharmacomicrobiomics: the tip of an iceberg of complex interactions between drugs and gut-associated microbes, 10.1186/1757-4749-4-16
  141. Imhann Floris, Vich Vila Arnau, Bonder Marc Jan, Lopez Manosalva Ailine G., Koonen Debby P.Y., Fu Jingyuan, Wijmenga Cisca, Zhernakova Alexandra, Weersma Rinse K., The influence of proton pump inhibitors and other commonly used medication on the gut microbiota, 10.1080/19490976.2017.1284732
  142. Le Bastard Q., Al-Ghalith G. A., Grégoire M., Chapelet G., Javaudin F., Dailly E., Batard E., Knights D., Montassier E., Systematic review: human gut dysbiosis induced by non-antibiotic prescription medications, 10.1111/apt.14451
  143. Falony Gwen, Joossens Marie, Vieira-Silva Sara, Wang Jun, Darzi Youssef, Faust Karoline, Kurilshikov Alexander, Bonder Marc Jan, Valles-Colomer Mireia, Vandeputte Doris, Tito Raul Y., Chaffron Samuel, Rymenans Leen, Verspecht Chloë, De Sutter Lise, Lima-Mendez Gipsi, D’hoe Kevin, Jonckheere Karl, Homola Daniel, Garcia Roberto, Tigchelaar Ettje F., Eeckhaudt Linda, Fu Jingyuan, Henckaerts Liesbet, Zhernakova Alexandra, Wijmenga Cisca, Raes Jeroen, Population-level analysis of gut microbiome variation, 10.1126/science.aad3503
  144. Wang Zeneng, Klipfell Elizabeth, Bennett Brian J., Koeth Robert, Levison Bruce S., DuGar Brandon, Feldstein Ariel E., Britt Earl B., Fu Xiaoming, Chung Yoon-Mi, Wu Yuping, Schauer Phil, Smith Jonathan D., Allayee Hooman, Tang W. H. Wilson, DiDonato Joseph A., Lusis Aldons J., Hazen Stanley L., Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease, 10.1038/nature09922
  145. Lee Duk-Hee, Porta Miquel, Jacobs David R., Vandenberg Laura N., Chlorinated Persistent Organic Pollutants, Obesity, and Type 2 Diabetes, 10.1210/er.2013-1084
  146. Lu Kun, Mahbub Ridwan, Fox James G., Xenobiotics: Interaction with the Intestinal Microflora, 10.1093/ilar/ilv018
  147. Caparrós-Martín Jose A., Lareu Ricky R., Ramsay Joshua P., Peplies Jörg, Reen F. Jerry, Headlam Henrietta A., Ward Natalie C., Croft Kevin D., Newsholme Philip, Hughes Jeffery D., O’Gara Fergal, Statin therapy causes gut dysbiosis in mice through a PXR-dependent mechanism, 10.1186/s40168-017-0312-4
  148. de la Cuesta-Zuluaga Jacobo, Mueller Noel T., Corrales-Agudelo Vanessa, Velásquez-Mejía Eliana P., Carmona Jenny A., Abad José M., Escobar Juan S., Metformin Is Associated With Higher Relative Abundance of Mucin-DegradingAkkermansia muciniphilaand Several Short-Chain Fatty Acid–Producing Microbiota in the Gut, 10.2337/dc16-1324
  149. Ma Wei, Chen Ji, Meng Yuhong, Yang Jichun, Cui Qinghua, Zhou Yuan, Metformin Alters Gut Microbiota of Healthy Mice: Implication for Its Potential Role in Gut Microbiota Homeostasis, 10.3389/fmicb.2018.01336
  150. Ursell Luke K., Knight Rob, Xenobiotics and the Human Gut Microbiome: Metatranscriptomics Reveal the Active Players, 10.1016/j.cmet.2013.02.013
  151. Zhao Xia, Zhang Zhujun, Hu Bin, Huang Wei, Yuan Chao, Zou Lingyun, Response of Gut Microbiota to Metabolite Changes Induced by Endurance Exercise, 10.3389/fmicb.2018.00765
  152. Monda Vincenzo, Villano Ines, Messina Antonietta, Valenzano Anna, Esposito Teresa, Moscatelli Fiorenzo, Viggiano Andrea, Cibelli Giuseppe, Chieffi Sergio, Monda Marcellino, Messina Giovanni, Exercise Modifies the Gut Microbiota with Positive Health Effects, 10.1155/2017/3831972
  153. Chen Jingyuan, Guo Yuan, Gui Yajun, Xu Danyan, Physical exercise, gut, gut microbiota, and atherosclerotic cardiovascular diseases, 10.1186/s12944-017-0653-9
  154. Cerdá Begoña, Pérez Margarita, Pérez-Santiago Jennifer D., Tornero-Aguilera Jose F., González-Soltero Rocío, Larrosa Mar, Gut Microbiota Modification: Another Piece in the Puzzle of the Benefits of Physical Exercise in Health?, 10.3389/fphys.2016.00051
  155. ALLEN JACOB M., MAILING LUCY J., NIEMIRO GRACE M., MOORE RACHEL, COOK MARC D., WHITE BRYAN A., HOLSCHER HANNAH D., WOODS JEFFREY A., Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans : , 10.1249/mss.0000000000001495
  156. Hsu Yi Ju, Chiu Chien Chao, Li Yen Peng, Huang Wen Ching, Huang Yen Te, Huang Chi Chang, Chuang Hsiao Li, Effect of Intestinal Microbiota on Exercise Performance in Mice : , 10.1519/jsc.0000000000000644
  157. MATSUMOTO Megumi, INOUE Ryo, TSUKAHARA Takamitsu, USHIDA Kazunari, CHIJI Hideyuki, MATSUBARA Noritaka, HARA Hiroshi, Voluntary Running Exercise Alters Microbiota Composition and Increases n-Butyrate Concentration in the Rat Cecum, 10.1271/bbb.70474
  158. den Besten Gijs, van Eunen Karen, Groen Albert K., Venema Koen, Reijngoud Dirk-Jan, Bakker Barbara M., The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, 10.1194/jlr.r036012
  159. Clark Allison, Mach Núria, The Crosstalk between the Gut Microbiota and Mitochondria during Exercise, 10.3389/fphys.2017.00319
  160. Newgard Christopher B., An Jie, Bain James R., Muehlbauer Michael J., Stevens Robert D., Lien Lillian F., Haqq Andrea M., Shah Svati H., Arlotto Michelle, Slentz Cris A., Rochon James, Gallup Dianne, Ilkayeva Olga, Wenner Brett R., Yancy William S., Eisenson Howard, Musante Gerald, Surwit Richard S., Millington David S., Butler Mark D., Svetkey Laura P., A Branched-Chain Amino Acid-Related Metabolic Signature that Differentiates Obese and Lean Humans and Contributes to Insulin Resistance, 10.1016/j.cmet.2009.02.002
  161. Pedersen Helle Krogh, Gudmundsdottir Valborg, Nielsen Henrik Bjørn, Hyotylainen Tuulia, Nielsen Trine, Jensen Benjamin A. H., Forslund Kristoffer, Hildebrand Falk, Prifti Edi, Falony Gwen, Le Chatelier Emmanuelle, Levenez Florence, Doré Joel, Mattila Ismo, Plichta Damian R., Pöhö Päivi, Hellgren Lars I., Arumugam Manimozhiyan, Sunagawa Shinichi, Vieira-Silva Sara, Jørgensen Torben, Holm Jacob Bak, Trošt Kajetan, Consortium MetaHIT, Kristiansen Karsten, Brix Susanne, Raes Jeroen, Wang Jun, Hansen Torben, Bork Peer, Brunak Søren, Oresic Matej, Ehrlich S. Dusko, Pedersen Oluf, Human gut microbes impact host serum metabolome and insulin sensitivity, 10.1038/nature18646
  162. Org, E. et al. Relationships between gut microbiota, plasma metabolites, and metabolic syndrome traits in the METSIM cohort. Genome Biol. 18, 70 (2017).
  163. Liu Ruixin, Hong Jie, Xu Xiaoqiang, Feng Qiang, Zhang Dongya, Gu Yanyun, Shi Juan, Zhao Shaoqian, Liu Wen, Wang Xiaokai, Xia Huihua, Liu Zhipeng, Cui Bin, Liang Peiwen, Xi Liuqing, Jin Jiabin, Ying Xiayang, Wang Xiaolin, Zhao Xinjie, Li Wanyu, Jia Huijue, Lan Zhou, Li Fengyu, Wang Rui, Sun Yingkai, Yang Minglan, Shen Yuxin, Jie Zhuye, Li Junhua, Chen Xiaomin, Zhong Huanzi, Xie Hailiang, Zhang Yifei, Gu Weiqiong, Deng Xiaxing, Shen Baiyong, Xu Xun, Yang Huanming, Xu Guowang, Bi Yufang, Lai Shenghan, Wang Jian, Qi Lu, Madsen Lise, Wang Jiqiu, Ning Guang, Kristiansen Karsten, Wang Weiqing, Gut microbiome and serum metabolome alterations in obesity and after weight-loss intervention, 10.1038/nm.4358
  164. Ben-Othman Nouha, Vieira Andhira, Courtney Monica, Record Fabien, Gjernes Elisabet, Avolio Fabio, Hadzic Biljana, Druelle Noémie, Napolitano Tiziana, Navarro-Sanz Sergi, Silvano Serena, Al-Hasani Keith, Pfeifer Anja, Lacas-Gervais Sandra, Leuckx Gunter, Marroquí Laura, Thévenet Julien, Madsen Ole Dragsbaek, Eizirik Decio Laks, Heimberg Harry, Kerr-Conte Julie, Pattou François, Mansouri Ahmed, Collombat Patrick, Long-Term GABA Administration Induces Alpha Cell-Mediated Beta-like Cell Neogenesis, 10.1016/j.cell.2016.11.002
  165. de Vadder Filipe, Mithieux Gilles, Gut-brain signaling in energy homeostasis: the unexpected role of microbiota-derived succinate, 10.1530/joe-17-0542
  166. De Vadder Filipe, Kovatcheva-Datchary Petia, Zitoun Carine, Duchampt Adeline, Bäckhed Fredrik, Mithieux Gilles, Microbiota-Produced Succinate Improves Glucose Homeostasis via Intestinal Gluconeogenesis, 10.1016/j.cmet.2016.06.013
  167. Ley Ruth E., Prevotella in the gut: choose carefully : Gut microbiota in 2015, 10.1038/nrgastro.2016.4
  168. Scher Jose U, Sczesnak Andrew, Longman Randy S, Segata Nicola, Ubeda Carles, Bielski Craig, Rostron Tim, Cerundolo Vincenzo, Pamer Eric G, Abramson Steven B, Huttenhower Curtis, Littman Dan R, Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis, 10.7554/elife.01202
  169. Heianza Yoriko, Sun Dianjianyi, Li Xiang, DiDonato Joseph A, Bray George A, Sacks Frank M, Qi Lu, Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism: the POUNDS Lost trial, 10.1136/gutjnl-2018-316155
  170. Zhu Weifei, Gregory Jill C., Org Elin, Buffa Jennifer A., Gupta Nilaksh, Wang Zeneng, Li Lin, Fu Xiaoming, Wu Yuping, Mehrabian Margarete, Sartor R. Balfour, McIntyre Thomas M., Silverstein Roy L., Tang W.H. Wilson, DiDonato Joseph A., Brown J. Mark, Lusis Aldons J., Hazen Stanley L., Gut Microbial Metabolite TMAO Enhances Platelet Hyperreactivity and Thrombosis Risk, 10.1016/j.cell.2016.02.011
  171. Gao Xiang, Liu Xiaofang, Xu Jie, Xue Changhu, Xue Yong, Wang Yuming, Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet, 10.1016/j.jbiosc.2014.03.001
  172. Koeth Robert A., Levison Bruce S., Culley Miranda K., Buffa Jennifer A., Wang Zeneng, Gregory Jill C., Org Elin, Wu Yuping, Li Lin, Smith Jonathan D., Tang W.H. Wilson, DiDonato Joseph A., Lusis Aldons J., Hazen Stanley L., γ-Butyrobetaine Is a Proatherogenic Intermediate in Gut Microbial Metabolism of L -Carnitine to TMAO, 10.1016/j.cmet.2014.10.006
  173. Agus Allison, Planchais Julien, Sokol Harry, Gut Microbiota Regulation of Tryptophan Metabolism in Health and Disease, 10.1016/j.chom.2018.05.003
  174. Zhang, L. S. & Davies, S. S. Microbial metabolism of dietary components to bioactive metabolites: opportunities for new therapeutic interventions. Genome Med. 8, 46 (2016).
  175. Dodd, D. et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 551, 648–652 (2017).
  176. Tuomainen Marjo, Lindström Jaana, Lehtonen Marko, Auriola Seppo, Pihlajamäki Jussi, Peltonen Markku, Tuomilehto Jaakko, Uusitupa Matti, de Mello Vanessa D., Hanhineva Kati, Associations of serum indolepropionic acid, a gut microbiota metabolite, with type 2 diabetes and low-grade inflammation in high-risk individuals, 10.1038/s41387-018-0046-9
  177. de Mello, V. D. et al. Indolepropionic acid and novel lipid metabolites are associated with a lower risk of type 2 diabetes in the Finnish Diabetes Prevention Study. Sci. Rep. 7, 46337 (2017).
  178. Krishnan Smitha, Ding Yufang, Saedi Nima, Choi Maria, Sridharan Gautham V., Sherr David H., Yarmush Martin L., Alaniz Robert C., Jayaraman Arul, Lee Kyongbum, Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages, 10.1016/j.celrep.2018.03.109
  179. Chimerel Catalin, Emery Edward, Summers David K., Keyser Ulrich, Gribble Fiona M., Reimann Frank, Bacterial Metabolite Indole Modulates Incretin Secretion from Intestinal Enteroendocrine L Cells, 10.1016/j.celrep.2014.10.032
  180. Beaumont Martin, Neyrinck Audrey M., Olivares Marta, Rodriguez Julie, de Rocca Serra Audrey, Roumain Martin, Bindels Laure B., Cani Patrice D., Evenepoel Pieter, Muccioli Giulio G., Demoulin Jean-Baptiste, Delzenne Nathalie M., The gut microbiota metabolite indole alleviates liver inflammation in mice, 10.1096/fj.201800544
  181. Koh Ara, Molinaro Antonio, Ståhlman Marcus, Khan Muhammad Tanweer, Schmidt Caroline, Mannerås-Holm Louise, Wu Hao, Carreras Alba, Jeong Heeyoon, Olofsson Louise E., Bergh Per-Olof, Gerdes Victor, Hartstra Annick, de Brauw Maurits, Perkins Rosie, Nieuwdorp Max, Bergström Göran, Bäckhed Fredrik, Microbially Produced Imidazole Propionate Impairs Insulin Signaling through mTORC1, 10.1016/j.cell.2018.09.055
  182. Klurfeld David M., Davis Cindy D., Karp Robert W., Allen-Vercoe Emma, Chang Eugene B., Chassaing Benoit, Fahey George C., Hamaker Bruce R., Holscher Hannah D., Lampe Johanna W., Marette Andre, Martens Eric, O’Keefe Stephen J., Rose Devin J., Saarela Maria, Schneeman Barbara O., Slavin Joanne L., Sonnenburg Justin L., Swanson Kelly S., Wu Gary D., Lynch Christopher J., Considerations for best practices in studies of fiber or other dietary components and the intestinal microbiome, 10.1152/ajpendo.00058.2018
  183. Routy Bertrand, Gopalakrishnan Vancheswaran, Daillère Romain, Zitvogel Laurence, Wargo Jennifer A., Kroemer Guido, The gut microbiota influences anticancer immunosurveillance and general health, 10.1038/s41571-018-0006-2
  184. Newberry Sydne J., Probiotics for the Prevention and Treatment of Antibiotic-Associated Diarrhea : A Systematic Review and Meta-analysis, 10.1001/jama.2012.3507
  185. Wei, D. et al. Probiotics for the prevention or treatment of chemotherapy- or radiotherapy-related diarrhoea in people with cancer. Cochrane Database Syst. Rev. 8, CD008831 (2018).
  186. Goldenberg, J. Z. et al. Probiotics for the prevention of Clostridium difficile–associated diarrhea in adults and children. Cochrane Database Syst. Rev. 12, CD006095 (2017).
  187. Zmora Niv, Zilberman-Schapira Gili, Suez Jotham, Mor Uria, Dori-Bachash Mally, Bashiardes Stavros, Kotler Eran, Zur Maya, Regev-Lehavi Dana, Brik Rotem Ben-Zeev, Federici Sara, Cohen Yotam, Linevsky Raquel, Rothschild Daphna, Moor Andreas E., Ben-Moshe Shani, Harmelin Alon, Itzkovitz Shalev, Maharshak Nitsan, Shibolet Oren, Shapiro Hagit, Pevsner-Fischer Meirav, Sharon Itai, Halpern Zamir, Segal Eran, Elinav Eran, Personalized Gut Mucosal Colonization Resistance to Empiric Probiotics Is Associated with Unique Host and Microbiome Features, 10.1016/j.cell.2018.08.041
  188. Suez Jotham, Zmora Niv, Zilberman-Schapira Gili, Mor Uria, Dori-Bachash Mally, Bashiardes Stavros, Zur Maya, Regev-Lehavi Dana, Ben-Zeev Brik Rotem, Federici Sara, Horn Max, Cohen Yotam, Moor Andreas E., Zeevi David, Korem Tal, Kotler Eran, Harmelin Alon, Itzkovitz Shalev, Maharshak Nitsan, Shibolet Oren, Pevsner-Fischer Meirav, Shapiro Hagit, Sharon Itai, Halpern Zamir, Segal Eran, Elinav Eran, Post-Antibiotic Gut Mucosal Microbiome Reconstitution Is Impaired by Probiotics and Improved by Autologous FMT, 10.1016/j.cell.2018.08.047
  189. Kootte Ruud S., Levin Evgeni, Salojärvi Jarkko, Smits Loek P., Hartstra Annick V., Udayappan Shanti D., Hermes Gerben, Bouter Kristien E., Koopen Annefleur M., Holst Jens J., Knop Filip K., Blaak Ellen E., Zhao Jing, Smidt Hauke, Harms Amy C., Hankemeijer Thomas, Bergman Jacques J.G.H.M., Romijn Hans A., Schaap Frank G., Olde Damink Steven W.M., Ackermans Mariette T., Dallinga-Thie Geesje M., Zoetendal Erwin, de Vos Willem M., Serlie Mireille J., Stroes Erik S.G., Groen Albert K., Nieuwdorp Max, Improvement of Insulin Sensitivity after Lean Donor Feces in Metabolic Syndrome Is Driven by Baseline Intestinal Microbiota Composition, 10.1016/j.cmet.2017.09.008
  190. Kovatcheva-Datchary Petia, Nilsson Anne, Akrami Rozita, Lee Ying Shiuan, De Vadder Filipe, Arora Tulika, Hallen Anna, Martens Eric, Björck Inger, Bäckhed Fredrik, Dietary Fiber-Induced Improvement in Glucose Metabolism Is Associated with Increased Abundance of Prevotella, 10.1016/j.cmet.2015.10.001
  191. Zeevi David, Korem Tal, Zmora Niv, Israeli David, Rothschild Daphna, Weinberger Adina, Ben-Yacov Orly, Lador Dar, Avnit-Sagi Tali, Lotan-Pompan Maya, Suez Jotham, Mahdi Jemal Ali, Matot Elad, Malka Gal, Kosower Noa, Rein Michal, Zilberman-Schapira Gili, Dohnalová Lenka, Pevsner-Fischer Meirav, Bikovsky Rony, Halpern Zamir, Elinav Eran, Segal Eran, Personalized Nutrition by Prediction of Glycemic Responses, 10.1016/j.cell.2015.11.001
  192. Kondo Tomoo, Kishi Mikiya, Fushimi Takashi, Kaga Takayuki, Acetic Acid Upregulates the Expression of Genes for Fatty Acid Oxidation Enzymes in Liver To Suppress Body Fat Accumulation, 10.1021/jf900470c
  193. Hong Yeon-Hee, Nishimura Yukihiko, Hishikawa Daisuke, Tsuzuki Hiroaki, Miyahara Hisae, Gotoh Chizu, Choi Ki-Choon, Feng Dan Dan, Chen Chen, Lee Hong-Gu, Katoh Kazuo, Roh Sang-Gun, Sasaki Shinichi, Acetate and Propionate Short Chain Fatty Acids Stimulate Adipogenesis via GPCR43, 10.1210/en.2005-0545
  194. Ge Hongfei, Li Xiaofan, Weiszmann Jennifer, Wang Ping, Baribault Helene, Chen Jin-Long, Tian Hui, Li Yang, Activation of G Protein-Coupled Receptor 43 in Adipocytes Leads to Inhibition of Lipolysis and Suppression of Plasma Free Fatty Acids, 10.1210/en.2008-0059
  195. Jocken Johan W. E., González Hernández Manuel A., Hoebers Nicole T. H., van der Beek Christina M., Essers Yvonne P. G., Blaak Ellen E., Canfora Emanuel E., Short-Chain Fatty Acids Differentially Affect Intracellular Lipolysis in a Human White Adipocyte Model, 10.3389/fendo.2017.00372
  196. Jia Yimin, Hong Jian, Li Huifang, Hu Yun, Jia Longfei, Cai Demin, Zhao Ruqian, Butyrate stimulates adipose lipolysis and mitochondrial oxidative phosphorylation through histone hyperacetylation-associated β3 -adrenergic receptor activation in high-fat diet-induced obese mice : Butyrate stimulates adipose lipolysis and oxidative phosphorylation, 10.1113/ep086114
  197. van Nood Els, Vrieze Anne, Nieuwdorp Max, Fuentes Susana, Zoetendal Erwin G., de Vos Willem M., Visser Caroline E., Kuijper Ed J., Bartelsman Joep F.W.M., Tijssen Jan G.P., Speelman Peter, Dijkgraaf Marcel G.W., Keller Josbert J., Duodenal Infusion of Donor Feces for Recurrent Clostridium difficile, 10.1056/nejmoa1205037
  198. Chapman Brandon C., Moore Hunter B., Overbey Douglas M., Morton Alex P., Harnke Ben, Gerich Mark E., Vogel Jon D., Fecal microbiota transplant in patients with Clostridium difficile infection : A systematic review, 10.1097/ta.0000000000001195
  199. Carlucci Christian, Petrof Elaine O., Allen-Vercoe Emma, Fecal Microbiota-based Therapeutics for Recurrent Clostridium difficile Infection, Ulcerative Colitis and Obesity, 10.1016/j.ebiom.2016.09.029
  200. Vrieze Anne, Van Nood Els, Holleman Frits, Salojärvi Jarkko, Kootte Ruud S., Bartelsman Joep F.W.M., Dallinga–Thie Geesje M., Ackermans Mariette T., Serlie Mireille J., Oozeer Raish, Derrien Muriel, Druesne Anne, Van Hylckama Vlieg Johan E.T., Bloks Vincent W., Groen Albert K., Heilig Hans G.H.J., Zoetendal Erwin G., Stroes Erik S., de Vos Willem M., Hoekstra Joost B.L., Nieuwdorp Max, Transfer of Intestinal Microbiota From Lean Donors Increases Insulin Sensitivity in Individuals With Metabolic Syndrome, 10.1053/j.gastro.2012.06.031
  201. Cani Patrice D, Van Hul Matthias, Novel opportunities for next-generation probiotics targeting metabolic syndrome, 10.1016/j.copbio.2014.10.006
  202. Patel Rachna, DuPont Herbert L., New Approaches for Bacteriotherapy: Prebiotics, New-Generation Probiotics, and Synbiotics, 10.1093/cid/civ177
  203. Garruti Gabriella, Di Ciaula Agostino, Wang Helen H., Wang David Q.-H., Portincasa Piero, Cross-Talk Between Bile Acids and Gastro-Intestinal and Thermogenic: Clues from Bariatric Surgery, 10.5604/01.3001.0010.5499
  204. Liu Haijun, Hu Cheng, Zhang Xueli, Jia Weiping, Role of gut microbiota, bile acids and their cross-talk in the effects of bariatric surgery on obesity and type 2 diabetes, 10.1111/jdi.12687
  205. Cani Patrice D., Gut microbiota — at the intersection of everything?, 10.1038/nrgastro.2017.54
  206. Aron-Wisnewsky, J. et al. Major microbiota dysbiosis in severe obesity: fate after bariatric surgery. Gut 68, 70–82 (2018).
  207. Cani, P. D. Severe obesity and gut microbiota: does bariatric surgery really reset the system? Gut 68, 5–6 (2018).
  208. Wang Zeneng, Roberts Adam B., Buffa Jennifer A., Levison Bruce S., Zhu Weifei, Org Elin, Gu Xiaodong, Huang Ying, Zamanian-Daryoush Maryam, Culley Miranda K., DiDonato Anthony J., Fu Xiaoming, Hazen Jennie E., Krajcik Daniel, DiDonato Joseph A., Lusis Aldons J., Hazen Stanley L., Non-lethal Inhibition of Gut Microbial Trimethylamine Production for the Treatment of Atherosclerosis, 10.1016/j.cell.2015.11.055
  209. Laurans Ludivine, Venteclef Nicolas, Haddad Yacine, Chajadine Mouna, Alzaid Fawaz, Metghalchi Sarvenaz, Sovran Bruno, Denis Raphael G. P., Dairou Julien, Cardellini Marina, Moreno-Navarrete Jose-Maria, Straub Marjolene, Jegou Sarah, McQuitty Claire, Viel Thomas, Esposito Bruno, Tavitian Bertrand, Callebert Jacques, Luquet Serge H., Federici Massimo, Fernandez-Real José Manuel, Burcelin Remy, Launay Jean-Marie, Tedgui Alain, Mallat Ziad, Sokol Harry, Taleb Soraya, Genetic deficiency of indoleamine 2,3-dioxygenase promotes gut microbiota-mediated metabolic health, 10.1038/s41591-018-0060-4
  210. Chen Zhongyi, Guo Lilu, Zhang Yongqin, L. Walzem Rosemary, Pendergast Julie S., Printz Richard L., Morris Lindsey C., Matafonova Elena, Stien Xavier, Kang Li, Coulon Denis, McGuinness Owen P., Niswender Kevin D., Davies Sean S., Incorporation of therapeutically modified bacteria into gut microbiota inhibits obesity, 10.1172/jci72517
Bibliographic reference Cani, Patrice D. ; Van Hul, Matthias ; Lefort, Charlotte ; Depommier, Clara ; Rastelli, Marialetizia ; et. al. Microbial regulation of organismal energy homeostasis. In: Nature Metabolism, Vol. 1, no. 1, p. 34-46 (2019)
Permanent URL http://hdl.handle.net/2078.1/212417