Cani, Patrice D.
[UCL]
Everard, Amandine
[UCL]
Obesity and diabetes have reached epidemic proportions. Evidence suggests that besides dietary habits and physical activity, other environmental factors, such as gut microbes, are recognized as additional partners implicated in the control of energy homeostasis. Studies on the human gut microbiota have shown that the general population can be stratified on the sole basis of three dominant bacteria (i.e., the concept of enterotypes), while some others have suggested categorizing the population according to their microbiome gene richness. Both aspects have been strengthened by recent studies investigating the impact of nutrients (e.g., dietary fibers, fat feeding) and dietary habits (i.e., vegans versus omnivores) of different populations. Using preclinical models, quite a few novel mechanisms have been proposed in these gut microbiota-host interactions, including the role of novel bacteria, the regulation of antimicrobial peptide production, the maintenance of the gut barrier function and intestinal innate immunity. In this review, we discuss several of the aforementioned aspects. Nonetheless, determining the overall mechanisms by which microbes dialogue with host cells will require further investigations before anticipating the development of next-generation nutritional interventions using prebiotics, probiotics, synbiotics, or even specific nutrients for promoting health benefits.
- Savage D C, Microbial Ecology of the Gastrointestinal Tract, 10.1146/annurev.mi.31.100177.000543
- Blottière Hervé M, de Vos Willem M, Ehrlich S Dusko, Doré Joël, Human intestinal metagenomics: state of the art and future, 10.1016/j.mib.2013.06.006
- 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, Antolin Maria, Artiguenave François, Blottiere Hervé, Borruel Natalia, Bruls Thomas, Casellas Francesc, Chervaux Christian, Cultrone Antonella, Delorme Christine, Denariaz Gérard, Dervyn Rozenn, Forte Miguel, Friss Carsten, van de Guchte Maarten, Guedon Eric, Haimet Florence, Jamet Alexandre, Juste Catherine, Kaci Ghalia, Kleerebezem Michiel, Knol Jan, Kristensen Michel, Layec Severine, Le Roux Karine, Leclerc Marion, Maguin Emmanuelle, Melo Minardi Raquel, Oozeer Raish, Rescigno Maria, Sanchez Nicolas, Tims Sebastian, Torrejon Toni, Varela Encarna, de Vos Willem, Winogradsky Yohanan, Zoetendal Erwin, Bork Peer, Ehrlich S. Dusko, Wang Jun, A human gut microbial gene catalogue established by metagenomic sequencing, 10.1038/nature08821
- 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, Pons Nicolas, Le Chatelier Emmanuelle, Batto Jean-Michel, Kennedy Sean, Haimet Florence, Winogradski Yohanan, Pelletier Eric, LePaslier Denis, Artiguenave François, Bruls Thomas, Weissenbach Jean, Turner Keith, Parkhill Julian, Antolin Maria, Casellas Francesc, Borruel Natalia, Varela Encarna, Torrejon Antonio, Denariaz Gérard, Derrien Muriel, van Hylckama Vlieg Johan E T, Viega Patrick, Oozeer Raish, Knoll Jan, Rescigno Maria, Brechot Christian, M'Rini Christine, Mérieux Alexandre, Yamada Takuji, Tims Sebastian, Zoetendal Erwin G, Kleerebezem Michiel, de Vos Willem M, Cultrone Antonella, Leclerc Marion, Juste Catherine, Guedon Eric, Delorme Christine, Layec Séverine, Khaci Ghalia, van de Guchte Maarten, Vandemeulebrouck Gaetana, Jamet Alexandre, Dervyn Rozenn, Sanchez Nicolas, Blottière Hervé, Maguin Emmanuelle, Renault Pierre, Tap Julien, Mende Daniel R, Bork Peer, Wang Jun, An integrated catalog of reference genes in the human gut microbiome, 10.1038/nbt.2942
- Doré Joël, Blottière Hervé, The influence of diet on the gut microbiota and its consequences for health, 10.1016/j.copbio.2015.01.002
- Hartstra Annick V., Bouter Kristien E.C., Bäckhed Fredrik, Nieuwdorp Max, Insights Into the Role of the Microbiome in Obesity and Type 2 Diabetes, 10.2337/dc14-0769
- Villanueva-Millán M. J., Pérez-Matute P., Oteo J. A., Gut microbiota: a key player in health and disease. A review focused on obesity, 10.1007/s13105-015-0390-3
- Scott, Microb. Ecol. Health Dis., 26, 25877 (2015)
- Maranduba Carlos Magno da Costa, De Castro Sandra Bertelli Ribeiro, Souza Gustavo Torres de, Rossato Cristiano, da Guia Francisco Carlos, Valente Maria Anete Santana, Rettore João Vitor Paes, Maranduba Claudinéia Pereira, Souza Camila Maurmann de, Carmo Antônio Márcio Resende do, Macedo Gilson Costa, Silva Fernando de Sá, Intestinal Microbiota as Modulators of the Immune System and Neuroimmune System: Impact on the Host Health and Homeostasis, 10.1155/2015/931574
- Festi Davide, Schiumerini Ramona, Eusebi Leonardo Henry, Marasco Giovanni, Taddia Martina, Colecchia Antonio, Gut microbiota and metabolic syndrome, 10.3748/wjg.v20.i43.16079
- Ettinger Grace, MacDonald Kyle, Reid Gregor, Burton Jeremy P, The influence of the human microbiome and probiotics on cardiovascular health, 10.4161/19490976.2014.983775
- Brandsma Eelke, Houben Tom, Fu Jingyuan, Shiri-Sverdlov Ronit, Hofker Marten H., The immunity–diet–microbiota axis in the development of metabolic syndrome : , 10.1097/mol.0000000000000154
- Devkota Suzanne, Chang Eugene B., Interactions between Diet, Bile Acid Metabolism, Gut Microbiota, and Inflammatory Bowel Diseases, 10.1159/000371687
- Devkota, Nature, 487, 104 (2012)
- Druart C., Alligier M., Salazar N., Neyrinck A. M., Delzenne N. M., Modulation of the Gut Microbiota by Nutrients with Prebiotic and Probiotic Properties, 10.3945/an.114.005835
- Cani Patrice D, Van Hul Matthias, Novel opportunities for next-generation probiotics targeting metabolic syndrome, 10.1016/j.copbio.2014.10.006
- Graber, J. Bacteriol., 89, 47 (1965)
- Cummings J H, Wiggins H S, Jenkins D J, Houston H, Jivraj T, Drasar B S, Hill M J, Influence of diets high and low in animal fat on bowel habit, gastrointestinal transit time, fecal microflora, bile acid, and fat excretion., 10.1172/jci109020
- SUGAWARA Masayoshi, SUZUKI Kunio, ENDO Kimiko, TASHIRO Yasuhito, NAKAMURA Katsuki, SUZUKI Kunihiko, FUJISAWA Tomohiko, SHIRAGAMI Nobue, MITSUOKA Tomotari, Effect of Dietary Fat and Fiber on Fecal Flora, Bacterial Metabolites, and Fecal Properties in Japanese Volunteers., 10.3177/jnsv.38.317
- Cani P. D., Amar J., Iglesias M. A., Poggi M., Knauf C., Bastelica D., Neyrinck A. M., Fava F., Tuohy K. M., Chabo C., Waget A., Delmee E., Cousin B., Sulpice T., Chamontin B., Ferrieres J., Tanti J.-F., Gibson G. R., Casteilla L., Delzenne N. M., Alessi M. C., Burcelin R., Metabolic Endotoxemia Initiates Obesity and Insulin Resistance, 10.2337/db06-1491
- Cani P. D., Neyrinck A. M., Fava F., Knauf C., Burcelin R. G., Tuohy K. M., Gibson G. R., Delzenne N. M., Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia, 10.1007/s00125-007-0791-0
- 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
- Turnbaugh Peter J., Bäckhed Fredrik, Fulton Lucinda, Gordon Jeffrey I., Diet-Induced Obesity Is Linked to Marked but Reversible Alterations in the Mouse Distal Gut Microbiome, 10.1016/j.chom.2008.02.015
- Fleissner Christine K., Huebel Nora, Abd El-Bary Mohamed Mostafa, Loh Gunnar, Klaus Susanne, Blaut Michael, Absence of intestinal microbiota does not protect mice from diet-induced obesity, 10.1017/s0007114510001303
- Hosoi Toru, Yokoyama Shota, Matsuo Suguru, Akira Shizuo, Ozawa Koichiro, Myeloid Differentiation Factor 88 (MyD88)-Deficiency Increases Risk of Diabetes in Mice, 10.1371/journal.pone.0012537
- Clavel Thomas, Desmarchelier Charles, Haller Dirk, Gérard Philippe, Rohn Sascha, Lepage Patricia, Daniel Hannelore, Intestinal microbiota in metabolic diseases : From bacterial community structure and functions to species of pathophysiological relevance, 10.4161/gmic.29331
- Harley Isaac T.W., Giles Daniel A., Pfluger Paul T., Burgess Stacey L., Walters Stephanie, Hembree Jazzminn, Raver Christine, Rewerts Cheryl L., Downey Jordan, Flick Leah M., Stankiewicz Traci E., McAlees Jaclyn W., Wills-Karp Marsha, Balfour Sartor R., Divanovic Senad, Tschöp Matthias H., Karp Christopher L., Differential colonization with segmented filamentous bacteria and Lactobacillus murinus do not drive divergent development of diet-induced obesity in C57BL/6 mice, 10.1016/j.molmet.2013.04.004
- Hildebrandt Marie A., Hoffmann Christian, Sherrill–Mix Scott A., Keilbaugh Sue A., Hamady Micah, Chen Ying–Yu, Knight Rob, Ahima Rexford S., Bushman Frederic, Wu Gary D., High-Fat Diet Determines the Composition of the Murine Gut Microbiome Independently of Obesity, 10.1053/j.gastro.2009.08.042
- Serino Matteo, Luche Elodie, Gres Sandra, Baylac Audrey, Bergé Mathieu, Cenac Claire, Waget Aurelie, Klopp Pascale, Iacovoni Jason, Klopp Christophe, Mariette Jerome, Bouchez Olivier, Lluch Jerome, Ouarné Francoise, Monsan Pierre, Valet Philippe, Roques Christine, Amar Jacques, Bouloumié Anne, Théodorou Vassilia, Burcelin Remy, Metabolic adaptation to a high-fat diet is associated with a change in the gut microbiota, 10.1136/gutjnl-2011-301012
- de La Serre C. B., Ellis C. L., Lee J., Hartman A. L., Rutledge J. C., Raybould H. E., Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation, 10.1152/ajpgi.00098.2010
- Ravussin Yann, Koren Omry, Spor Ayme, LeDuc Charles, Gutman Roee, Stombaugh Jesse, Knight Rob, Ley Ruth E., Leibel Rudolph L., Responses of Gut Microbiota to Diet Composition and Weight Loss in Lean and Obese Mice, 10.1038/oby.2011.111
- Zhang Xu, Zhao Yufeng, Zhang Menghui, Pang Xiaoyan, Xu Jia, Kang Chaoying, Li Meng, Zhang Chenhong, Zhang Zhiguo, Zhang Yifei, Li Xiaoying, Ning Guang, Zhao Liping, Structural Changes of Gut Microbiota during Berberine-Mediated Prevention of Obesity and Insulin Resistance in High-Fat Diet-Fed Rats, 10.1371/journal.pone.0042529
- Neyrinck Audrey M., Possemiers Sam, Druart Céline, Van de Wiele Tom, De Backer Fabienne, Cani Patrice D., Larondelle Yvan, Delzenne Nathalie M., Prebiotic Effects of Wheat Arabinoxylan Related to the Increase in Bifidobacteria, Roseburia and Bacteroides/Prevotella in Diet-Induced Obese Mice, 10.1371/journal.pone.0020944
- Qiao Yi, Sun Jin, Ding Yinyi, Le Guowei, Shi Yonghui, Alterations of the gut microbiota in high-fat diet mice is strongly linked to oxidative stress, 10.1007/s00253-012-4323-6
- Everard Amandine, Lazarevic Vladimir, Gaïa Nadia, Johansson Maria, Ståhlman Marcus, Backhed Fredrik, Delzenne Nathalie M, Schrenzel Jacques, François Patrice, Cani Patrice D, Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity, 10.1038/ismej.2014.45
- Everard Amandine, Geurts Lucie, Caesar Robert, Van Hul Matthias, Matamoros Sébastien, Duparc Thibaut, Denis Raphael G. P., Cochez Perrine, Pierard Florian, Castel Julien, Bindels Laure B., Plovier Hubert, Robine Sylvie, Muccioli Giulio G., Renauld Jean-Christophe, Dumoutier Laure, Delzenne Nathalie M., Luquet Serge, Bäckhed Fredrik, Cani Patrice D., Intestinal epithelial MyD88 is a sensor switching host metabolism towards obesity according to nutritional status, 10.1038/ncomms6648
- Carmody Rachel N., Gerber Georg K., Luevano Jesus M., Gatti Daniel M., Somes Lisa, Svenson Karen L., Turnbaugh Peter J., Diet Dominates Host Genotype in Shaping the Murine Gut Microbiota, 10.1016/j.chom.2014.11.010
- Chaplin Alice, Parra Pilar, Serra Francisca, Palou Andreu, Conjugated Linoleic Acid Supplementation under a High-Fat Diet Modulates Stomach Protein Expression and Intestinal Microbiota in Adult Mice, 10.1371/journal.pone.0125091
- Daniel Hannelore, Gholami Amin Moghaddas, Berry David, Desmarchelier Charles, Hahne Hannes, Loh Gunnar, Mondot Stanislas, Lepage Patricia, Rothballer Michael, Walker Alesia, Böhm Christoph, Wenning Mareike, Wagner Michael, Blaut Michael, Schmitt-Kopplin Philippe, Kuster Bernhard, Haller Dirk, Clavel Thomas, High-fat diet alters gut microbiota physiology in mice, 10.1038/ismej.2013.155
- 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, Guedon Eric, Delorme Christine, Layec Séverine, Khaci Ghalia, van de Guchte Maarten, Vandemeulebrouck Gaetana, Jamet Alexandre, Dervyn Rozenn, Sanchez Nicolas, Maguin Emmanuelle, Haimet Florence, Winogradski Yohanan, Cultrone Antonella, Leclerc Marion, Juste Catherine, Blottière Hervé, Pelletier Eric, LePaslier Denis, Artiguenave François, Bruls Thomas, Weissenbach Jean, Turner Keith, Parkhill Julian, Antolin Maria, Manichanh Chaysavanh, Casellas Francesc, Boruel Natalia, Varela Encarna, Torrejon Antonio, Guarner Francisco, Denariaz Gérard, Derrien Muriel, van Hylckama Vlieg Johan E. T., Veiga Patrick, Oozeer Raish, Knol Jan, Rescigno Maria, Brechot Christian, M’Rini Christine, Mérieux Alexandre, Yamada Takuji, Richness of human gut microbiome correlates with metabolic markers, 10.1038/nature12506
- Arumugam Manimozhiyan, Raes Jeroen, Pelletier Eric, Le Paslier Denis, Yamada Takuji, Mende Daniel R., Fernandes Gabriel R., Tap Julien, Bruls Thomas, Batto Jean-Michel, Bertalan Marcelo, Borruel Natalia, Casellas Francesc, Fernandez Leyden, Gautier Laurent, Hansen Torben, Hattori Masahira, Hayashi Tetsuya, Kleerebezem Michiel, Kurokawa Ken, Leclerc Marion, Levenez Florence, Manichanh Chaysavanh, Nielsen H. Bjørn, Nielsen Trine, Pons Nicolas, Poulain Julie, Qin Junjie, Sicheritz-Ponten Thomas, Tims Sebastian, Torrents David, Ugarte Edgardo, Zoetendal Erwin G., Wang Jun, Guarner Francisco, Pedersen Oluf, de Vos Willem M., Brunak Søren, Doré Joel, Antolín María, Artiguenave François, Blottiere Hervé M., Almeida Mathieu, Brechot Christian, Cara Carlos, Chervaux Christian, Cultrone Antonella, Delorme Christine, Denariaz Gérard, Dervyn Rozenn, Foerstner Konrad U., Friss Carsten, van de Guchte Maarten, Guedon Eric, Haimet Florence, Huber Wolfgang, van Hylckama-Vlieg Johan, Jamet Alexandre, Juste Catherine, Kaci Ghalia, Knol Jan, Lakhdari Omar, Layec Severine, Le Roux Karine, Maguin Emmanuelle, Mérieux Alexandre, Melo Minardi Raquel, M'rini Christine, Muller Jean, Oozeer Raish, Parkhill Julian, Renault Pierre, Rescigno Maria, Sanchez Nicolas, Sunagawa Shinichi, Torrejon Antonio, Turner Keith, Vandemeulebrouck Gaetana, Varela Encarna, Winogradsky Yohanan, Zeller Georg, Weissenbach Jean, Ehrlich S. Dusko, Bork Peer, Enterotypes of the human gut microbiome, 10.1038/nature09944
- Wu G. D., Chen J., Hoffmann C., Bittinger K., Chen Y.-Y., Keilbaugh S. A., Bewtra M., Knights D., Walters W. A., Knight R., Sinha R., Gilroy E., Gupta K., Baldassano R., Nessel L., Li H., Bushman F. D., Lewis J. D., Linking Long-Term Dietary Patterns with Gut Microbial Enterotypes, 10.1126/science.1208344
- David Lawrence A., Maurice Corinne F., Carmody Rachel N., Gootenberg David B., Button Julie E., Wolfe Benjamin E., Ling Alisha V., Devlin A. Sloan, Varma Yug, Fischbach Michael A., Biddinger Sudha B., Dutton Rachel J., Turnbaugh Peter J., Diet rapidly and reproducibly alters the human gut microbiome, 10.1038/nature12820
- Huse Susan M., Ye Yuzhen, Zhou Yanjiao, Fodor Anthony A., A Core Human Microbiome as Viewed through 16S rRNA Sequence Clusters, 10.1371/journal.pone.0034242
- Claesson, Nature, 488, 178 (2012)
- Nakayama Jiro, Watanabe Koichi, Jiang Jiahui, Matsuda Kazunori, Chao Shiou-Huei, Haryono Pri, La-ongkham Orawan, Sarwoko Martinus-Agus, Sujaya I. Nengah, Zhao Liang, Chen Kang-Ting, Chen Yen-Po, Chiu Hsueh-Hui, Hidaka Tomoko, Huang Ning- Xin, Kiyohara Chikako, Kurakawa Takashi, Sakamoto Naoshige, Sonomoto Kenji, Tashiro Kousuke, Tsuji Hirokazu, Chen Ming-Ju, Leelavatcharamas Vichai, Liao Chii-Cherng, Nitisinprasert Sunee, Rahayu Endang S., Ren Fa-Zheng, Tsai Ying-Chieh, Lee Yuan-Kun, Diversity in gut bacterial community of school-age children in Asia, 10.1038/srep08397
- Lim Mi Young, Rho Mina, Song Yun-Mi, Lee Kayoung, Sung Joohon, Ko GwangPyo, Stability of Gut Enterotypes in Korean Monozygotic Twins and Their Association with Biomarkers and Diet, 10.1038/srep07348
- Roager H. M., Licht T. R., Poulsen S. K., Larsen T. M., Bahl M. I., Microbial Enterotypes, Inferred by the Prevotella-to-Bacteroides Ratio, Remained Stable during a 6-Month Randomized Controlled Diet Intervention with the New Nordic Diet, 10.1128/aem.03549-13
- Wu Gary D, Compher Charlene, Chen Eric Z, Smith Sarah A, Shah Rachana D, Bittinger Kyle, Chehoud Christel, Albenberg Lindsey G, Nessel Lisa, Gilroy Erin, Star Julie, Weljie Aalim M, Flint Harry J, Metz David C, Bennett Michael J, Li Hongzhe, Bushman Frederic D, Lewis James D, Comparative metabolomics in vegans and omnivores reveal constraints on diet-dependent gut microbiota metabolite production, 10.1136/gutjnl-2014-308209
- 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, consortium ANR MicroObes, Doré Joel, Zucker Jean-Daniel, Clément Karine, Ehrlich Stanislav Dusko, Blottière Hervé, Leclerc Marion, Juste Catherine, de Wouters Tomas, Lepage Patricia, Fouqueray Charlene, Basdevant Arnaud, Henegar Cornelieu, Godard Cindy, Fondacci Marine, Rohia Alili, Hajduch Froogh, Weissenbach Jean, Pelletier Eric, Le Paslier Denis, Gauchi Jean-Pierre, Gibrat Jean-François, Loux Valentin, Carré Wilfrid, Maguin Emmanuelle, van de Guchte Maarten, Jamet Alexandre, Boumezbeur Fouad, Layec Séverine, Dietary intervention impact on gut microbial gene richness, 10.1038/nature12480
- Lagkouvardos Ilias, Kläring Karoline, Heinzmann Silke S., Platz Stefanie, Scholz Birgit, Engel Karl-Heinz, Schmitt-Kopplin Philippe, Haller Dirk, Rohn Sascha, Skurk Thomas, Clavel Thomas, Gut metabolites and bacterial community networks during a pilot intervention study with flaxseeds in healthy adult men, 10.1002/mnfr.201500125
- Schaubeck Monika, Clavel Thomas, Calasan Jelena, Lagkouvardos Ilias, Haange Sven Bastiaan, Jehmlich Nico, Basic Marijana, Dupont Aline, Hornef Mathias, Bergen Martin von, Bleich André, Haller Dirk, Dysbiotic gut microbiota causes transmissible Crohn's disease-like ileitis independent of failure in antimicrobial defence, 10.1136/gutjnl-2015-309333
- Erridge, Am. J. Clin. Nutr., 86, 1286 (2007)
- Amar, Am. J. Clin. Nutr., 87, 1219 (2008)
- Pussinen P. J., Havulinna A. S., Lehto M., Sundvall J., Salomaa V., Endotoxemia Is Associated With an Increased Risk of Incident Diabetes, 10.2337/dc10-1676
- Lassenius Mariann I., Pietiläinen Kirsi H., Kaartinen Kati, Pussinen Pirkko J., Syrjänen Jaana, Forsblom Carol, Pörsti Ilkka, Rissanen Aila, Kaprio Jaakko, Mustonen Jukka, Groop Per-Henrik, Lehto Markku, , Bacterial Endotoxin Activity in Human Serum Is Associated With Dyslipidemia, Insulin Resistance, Obesity, and Chronic Inflammation, 10.2337/dc10-2197
- Laugerette Fabienne, Vors Cécile, Géloën Alain, Chauvin Marie-Agnès, Soulage Christophe, Lambert-Porcheron Stéphanie, Peretti Noël, Alligier Maud, Burcelin Rémy, Laville Martine, Vidal Hubert, Michalski Marie-Caroline, Emulsified lipids increase endotoxemia: possible role in early postprandial low-grade inflammation, 10.1016/j.jnutbio.2009.11.011
- Moreno-Navarrete José María, Escoté Xavier, Ortega Francisco, Serino Matteo, Campbell Mark, Michalski Marie-Caroline, Laville Martine, Xifra Gemma, Luche Elodie, Domingo Pere, Sabater Mónica, Pardo Gerard, Waget Aurelie, Salvador Javier, Giralt Marta, Rodriguez-Hermosa Jose I., Camps Marta, Kolditz Catherine I., Viguerie Nathalie, Galitzky Jean, Decaunes Pauline, Ricart Wifredo, Frühbeck Gema, Villarroya Francesc, Mingrone Geltrude, Langin Dominique, Zorzano Antonio, Vidal Hubert, Vendrell Joan, Burcelin Remy, Vidal-Puig Antonio, Fernández-Real José Manuel, A role for adipocyte-derived lipopolysaccharide-binding protein in inflammation- and obesity-associated adipose tissue dysfunction, 10.1007/s00125-013-3015-9
- Gu Yeyi, Yu Shan, Park Jong Yung, Harvatine Kevin, Lambert Joshua D., Dietary cocoa reduces metabolic endotoxemia and adipose tissue inflammation in high-fat fed mice, 10.1016/j.jnutbio.2013.12.004
- Wang Jing-Hua, Bose Shambhunath, Kim Gi-Cheol, Hong Seung-Ug, Kim Ji-Hun, Kim Jai-eun, Kim Hojun, Flos Lonicera Ameliorates Obesity and Associated Endotoxemia in Rats through Modulation of Gut Permeability and Intestinal Microbiota, 10.1371/journal.pone.0086117
- Kaliannan K., Hamarneh S. R., Economopoulos K. P., Nasrin Alam S., Moaven O., Patel P., Malo N. S., Ray M., Abtahi S. M., Muhammad N., Raychowdhury A., Teshager A., Mohamed M. M. R., Moss A. K., Ahmed R., Hakimian S., Narisawa S., Millan J. L., Hohmann E., Warren H. S., Bhan A. K., Malo M. S., Hodin R. A., Intestinal alkaline phosphatase prevents metabolic syndrome in mice, 10.1073/pnas.1220180110
- Peng Xiaohui, Nie Yao, Wu Jianjun, Huang Qiang, Cheng Yuqiang, Juglone prevents metabolic endotoxemia-induced hepatitis and neuroinflammation via suppressing TLR4/NF-κB signaling pathway in high-fat diet rats, 10.1016/j.bbrc.2015.04.124
- Luck Helen, Tsai Sue, Chung Jason, Clemente-Casares Xavier, Ghazarian Magar, Revelo Xavier S., Lei Helena, Luk Cynthia T., Shi Sally Yu, Surendra Anuradha, Copeland Julia K., Ahn Jennifer, Prescott David, Rasmussen Brittany A., Chng Melissa Hui Yen, Engleman Edgar G., Girardin Stephen E., Lam Tony K.T., Croitoru Kenneth, Dunn Shannon, Philpott Dana J., Guttman David S., Woo Minna, Winer Shawn, Winer Daniel A., Regulation of Obesity-Related Insulin Resistance with Gut Anti-inflammatory Agents, 10.1016/j.cmet.2015.03.001
- Kless Caroline, Müller Veronika Maria, Schüppel Valentina Luise, Lichtenegger Martina, Rychlik Michael, Daniel Hannelore, Klingenspor Martin, Haller Dirk, Diet-induced obesity causes metabolic impairment independent of alterations in gut barrier integrity, 10.1002/mnfr.201400840
- Cani P. D., Possemiers S., Van de Wiele T., Guiot Y., Everard A., Rottier O., Geurts L., Naslain D., Neyrinck A., Lambert D. M., Muccioli G. G., Delzenne N. M., Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability, 10.1136/gut.2008.165886
- Robertson M. Denise, Dietary-resistant starch and glucose metabolism : , 10.1097/mco.0b013e3283536931
- Slavin Joanne, Fiber and Prebiotics: Mechanisms and Health Benefits, 10.3390/nu5041417
- Birt D. F., Boylston T., Hendrich S., Jane J.-L., Hollis J., Li L., McClelland J., Moore S., Phillips G. J., Rowling M., Schalinske K., Scott M. P., Whitley E. M., Resistant Starch: Promise for Improving Human Health, 10.3945/an.113.004325
- Geurts L., Neyrinck A.M., Delzenne N.M., Knauf C., Cani P.D., Gut microbiota controls adipose tissue expansion, gut barrier and glucose metabolism: novel insights into molecular targets and interventions using prebiotics, 10.3920/bm2012.0065
- Apajalahti J. H. A., Kettunen H., Kettunen A., Holben W. E., Nurminen P. H., Rautonen N., Mutanen M., Culture-Independent Microbial Community Analysis Reveals that Inulin in the Diet Primarily Affects Previously Unknown Bacteria in the Mouse Cecum, 10.1128/aem.68.10.4986-4995.2002
- Everard, Diabetes, 60, 2775 (2011)
- Derrien M., Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium, 10.1099/ijs.0.02873-0
- Everard Amandine, Lazarevic Vladimir, Derrien Muriel, Girard Myriam, Muccioli Giulio G., Neyrinck Audrey M., Possemiers Sam, Van Holle Ann, François Patrice, de Vos Willem M., Delzenne Nathalie M., Schrenzel Jacques, Cani Patrice D., Responses of Gut Microbiota and Glucose and Lipid Metabolism to Prebiotics in Genetic Obese and Diet-Induced Leptin-Resistant Mice, 10.2337/db11-0227
- 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
- Shin Na-Ri, Lee June-Chul, Lee Hae-Youn, Kim Min-Soo, Whon Tae Woong, Lee Myung-Shik, Bae Jin-Woo, An increase in theAkkermansiaspp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice, 10.1136/gutjnl-2012-303839
- Tachon Sybille, Zhou June, Keenan Michael, Martin Roy, Marco Maria L., The intestinal microbiota in aged mice is modulated by dietary resistant starch and correlated with improvements in host responses, 10.1111/j.1574-6941.2012.01475.x
- Anhê Fernando F, Roy Denis, Pilon Geneviève, Dudonné Stéphanie, Matamoros Sébastien, Varin Thibault V, Garofalo Carole, Moine Quentin, Desjardins Yves, Levy Emile, Marette André, A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increasedAkkermansiaspp. population in the gut microbiota of mice, 10.1136/gutjnl-2014-307142
- Lukovac S., Belzer C., Pellis L., Keijser B. J., de Vos W. M., Montijn R. C., Roeselers G., Differential Modulation by Akkermansia muciniphila and Faecalibacterium prausnitzii of Host Peripheral Lipid Metabolism and Histone Acetylation in Mouse Gut Organoids, 10.1128/mbio.01438-14
- Reunanen Justus, Kainulainen Veera, Huuskonen Laura, Ottman Noora, Belzer Clara, Huhtinen Heikki, de Vos Willem M., Satokari Reetta, Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer, 10.1128/aem.04050-14
- Zackular J. P., Baxter N. T., Iverson K. D., Sadler W. D., Petrosino J. F., Chen G. Y., Schloss P. D., The Gut Microbiome Modulates Colon Tumorigenesis, 10.1128/mbio.00692-13
- 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
- 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
- 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
- Poggi M., Bastelica D., Gual P., Iglesias M. A., Gremeaux T., Knauf C., Peiretti F., Verdier M., Juhan-Vague I., Tanti J. F., Burcelin R., Alessi M. C., C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet, 10.1007/s00125-007-0654-8
- Caricilli Andréa M., Picardi Paty K., de Abreu Lélia L., Ueno Mirian, Prada Patrícia O., Ropelle Eduardo R., Hirabara Sandro Massao, Castoldi Ângela, Vieira Pedro, Camara Niels O. S., Curi Rui, Carvalheira José B., Saad Mário J. A., Gut Microbiota Is a Key Modulator of Insulin Resistance in TLR 2 Knockout Mice, 10.1371/journal.pbio.1001212
- Davis Jeremy E., Braucher Douglas R., Walker-Daniels Jennifer, Spurlock Michael E., Absence of Tlr2 protects against high-fat diet-induced inflammation and results in greater insulin-stimulated glucose transport in cultured adipocytes, 10.1016/j.jnutbio.2009.12.008
- Kim Kyung-Ah, Gu Wan, Lee In-Ah, Joh Eun-Ha, Kim Dong-Hyun, High Fat Diet-Induced Gut Microbiota Exacerbates Inflammation and Obesity in Mice via the TLR4 Signaling Pathway, 10.1371/journal.pone.0047713
- Michelsen K. S., Wong M. H., Shah P. K., Zhang W., Yano J., Doherty T. M., Akira S., Rajavashisth T. B., Arditi M., Lack of Toll-like receptor 4 or myeloid differentiation factor 88 reduces atherosclerosis and alters plaque phenotype in mice deficient in apolipoprotein E, 10.1073/pnas.0403249101
- Pierre Nicolas, Deldicque Louise, Barbé Caroline, Naslain Damien, Cani Patrice D., Francaux Marc, Toll-Like Receptor 4 Knockout Mice Are Protected against Endoplasmic Reticulum Stress Induced by a High-Fat Diet, 10.1371/journal.pone.0065061
- Shi Hang, Kokoeva Maia V., Inouye Karen, Tzameli Iphigenia, Yin Huali, Flier Jeffrey S., TLR4 links innate immunity and fatty acid–induced insulin resistance, 10.1172/jci28898
- Chassaing Benoit, Koren Omry, Goodrich Julia K., Poole Angela C., Srinivasan Shanthi, Ley Ruth E., Gewirtz Andrew T., Dietary emulsifiers impact the mouse gut microbiota promoting colitis and metabolic syndrome, 10.1038/nature14232
Bibliographic reference |
Cani, Patrice D. ; Everard, Amandine. Talking microbes: When gut bacteria interact with diet and host organs.. In: Molecular Nutrition & Food Research, Vol. 60, no. 1, p. 58-66 (2016) |
Permanent URL |
http://hdl.handle.net/2078.1/170441 |