User menu

Accès à distance ? S'identifier sur le proxy UCLouvain

Resistant starch can improve insulin sensitivity independently of the gut microbiota

  • Open access
  • PDF
  • 1.94 M
  1. World Health Organization. Obesity and overweight. Fact sheet N°311, available at http://www.who.int/mediacentre/factsheets/fs311/en/ . 2015. Accessed 28 June 2015.
  2. Alberti K. G. M. M., Zimmet P., Shaw J., Metabolic syndrome-a new world-wide definition. A Consensus Statement from the International Diabetes Federation, 10.1111/j.1464-5491.2006.01858.x
  3. DeFronzo Ralph A., Ferrannini Ele, Groop Leif, Henry Robert R., Herman William H., Holst Jens Juul, Hu Frank B., Kahn C. Ronald, Raz Itamar, Shulman Gerald I., Simonson Donald C., Testa Marcia A., Weiss Ram, Type 2 diabetes mellitus, 10.1038/nrdp.2015.19
  4. Patsouris David, Li Ping-Ping, Thapar Divya, Chapman Justin, Olefsky Jerrold M., Neels Jaap G., Ablation of CD11c-Positive Cells Normalizes Insulin Sensitivity in Obese Insulin Resistant Animals, 10.1016/j.cmet.2008.08.015
  5. Kaczmarczyk Melissa M., Miller Michael J., Freund Gregory G., The health benefits of dietary fiber: Beyond the usual suspects of type 2 diabetes mellitus, cardiovascular disease and colon cancer, 10.1016/j.metabol.2012.01.017
  6. Jakobsdottir Greta, Nyman Margareta, Fåk Frida, Designing future prebiotic fiber to target metabolic syndrome, 10.1016/j.nut.2013.08.013
  7. Slavin Joanne, Fiber and Prebiotics: Mechanisms and Health Benefits, 10.3390/nu5041417
  8. Robertson MD, Bickerton AS, Dennis AL, Vidal H, Frayn KN. Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am J Clin Nutr. 2005;82:559–67.
  9. Robertson M. Denise, Wright John W., Loizon Emmanuelle, Debard Cyrille, Vidal Hubert, Shojaee-Moradie Fariba, Russell-Jones David, Umpleby A. Margot, Insulin-Sensitizing Effects on Muscle and Adipose Tissue after Dietary Fiber Intake in Men and Women with Metabolic Syndrome, 10.1210/jc.2012-1513
  10. Johnston K. L., Thomas E. L., Bell J. D., Frost G. S., Robertson M. D., Resistant starch improves insulin sensitivity in metabolic syndrome, 10.1111/j.1464-5491.2010.02923.x
  11. Maki K. C., Pelkman C. L., Finocchiaro E. T., Kelley K. M., Lawless A. L., Schild A. L., Rains T. M., Resistant Starch from High-Amylose Maize Increases Insulin Sensitivity in Overweight and Obese Men, 10.3945/jn.111.152975
  12. Higgins Janine A., Resistant Starch and Energy Balance: Impact on Weight Loss and Maintenance, 10.1080/10408398.2011.629352
  13. 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
  14. Keenan M. J., Zhou J., Hegsted M., Pelkman C., Durham H. A., Coulon D. B., Martin R. J., Role of Resistant Starch in Improving Gut Health, Adiposity, and Insulin Resistance, 10.3945/an.114.007419
  15. Bindels Laure B., Walter Jens, Ramer-Tait Amanda E., Resistant starches for the management of metabolic diseases : , 10.1097/mco.0000000000000223
  16. 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
  17. 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
  18. Bindels Laure B., Dewulf Evelyne M., Delzenne Nathalie M., GPR43/FFA2: physiopathological relevance and therapeutic prospects, 10.1016/j.tips.2013.02.002
  19. Smith P. M., Howitt M. R., Panikov N., Michaud M., Gallini C. A., Bohlooly-Y M., Glickman J. N., Garrett W. S., The Microbial Metabolites, Short-Chain Fatty Acids, Regulate Colonic Treg Cell Homeostasis, 10.1126/science.1241165
  20. Arpaia Nicholas, Campbell Clarissa, Fan Xiying, Dikiy Stanislav, van der Veeken Joris, deRoos Paul, Liu Hui, Cross Justin R., Pfeffer Klaus, Coffer Paul J., Rudensky Alexander Y., Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation, 10.1038/nature12726
  21. Trompette Aurélien, Gollwitzer Eva S, Yadava Koshika, Sichelstiel Anke K, Sprenger Norbert, Ngom-Bru Catherine, Blanchard Carine, Junt Tobias, Nicod Laurent P, Harris Nicola L, Marsland Benjamin J, Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, 10.1038/nm.3444
  22. Bindels Laure B., Delzenne Nathalie M., Cani Patrice D., Walter Jens, Towards a more comprehensive concept for prebiotics, 10.1038/nrgastro.2015.47
  23. Delzenne Nathalie M., Bindels Laure B., Gut microbiota: Ganoderma lucidum, a new prebiotic agent to treat obesity?, 10.1038/nrgastro.2015.137
  24. Chang Chih-Jung, Lin Chuan-Sheng, Lu Chia-Chen, Martel Jan, Ko Yun-Fei, Ojcius David M., Tseng Shun-Fu, Wu Tsung-Ru, Chen Yi-Yuan Margaret, Young John D., Lai Hsin-Chih, Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota, 10.1038/ncomms8489
  25. Chassaing Benoit, Miles-Brown Jennifer, Pellizzon Michael, Ulman Edward, Ricci Matthew, Zhang Limin, Patterson Andrew D., Vijay-Kumar Matam, Gewirtz Andrew T., Lack of soluble fiber drives diet-induced adiposity in mice, 10.1152/ajpgi.00172.2015
  26. Martinez I., Wallace G., Zhang C., Legge R., Benson A. K., Carr T. P., Moriyama E. N., Walter J., Diet-Induced Metabolic Improvements in a Hamster Model of Hypercholesterolemia Are Strongly Linked to Alterations of the Gut Microbiota, 10.1128/aem.00380-09
  27. Martinez I., Perdicaro D. J., Brown A. W., Hammons S., Carden T. J., Carr T. P., Eskridge K. M., Walter J., Diet-Induced Alterations of Host Cholesterol Metabolism Are Likely To Affect the Gut Microbiota Composition in Hamsters, 10.1128/aem.03046-12
  28. Bindels Laure B., Beck Raphaël, Schakman Olivier, Martin Jennifer C., De Backer Fabienne, Sohet Florence M., Dewulf Evelyne M., Pachikian Barbara D., Neyrinck Audrey M., Thissen Jean-Paul, Verrax Julien, Calderon Pedro Buc, Pot Bruno, Grangette Corinne, Cani Patrice D., Scott Karen P., Delzenne Nathalie M., Restoring Specific Lactobacilli Levels Decreases Inflammation and Muscle Atrophy Markers in an Acute Leukemia Mouse Model, 10.1371/journal.pone.0037971
  29. Yang Junyi, Bindels Laure B., Segura Munoz Rafael R., Martínez Inés, Walter Jens, Ramer-Tait Amanda E., Rose Devin J., Disparate Metabolic Responses in Mice Fed a High-Fat Diet Supplemented with Maize-Derived Non-Digestible Feruloylated Oligo- and Polysaccharides Are Linked to Changes in the Gut Microbiota, 10.1371/journal.pone.0146144
  30. 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
  31. Nichenametla Sailendra N., Weidauer Lee A., Wey Howard E., Beare Tianna M., Specker Bonny L., Dey Moul, Resistant starch type 4-enriched diet lowered blood cholesterols and improved body composition in a double blind controlled cross-over intervention, 10.1002/mnfr.201300829
  32. Zhou June, Martin Roy J., Raggio Anne M., Shen Li, McCutcheon Kathleen, Keenan Michael J., The importance of GLP-1 and PYY in resistant starch's effect on body fat in mice, 10.1002/mnfr.201400904
  33. Martínez Inés, Kim Jaehyoung, Duffy Patrick R., Schlegel Vicki L., Walter Jens, Resistant Starches Types 2 and 4 Have Differential Effects on the Composition of the Fecal Microbiota in Human Subjects, 10.1371/journal.pone.0015046
  34. Ding Shengli, Chi Michael M., Scull Brooks P., Rigby Rachael, Schwerbrock Nicole M. J., Magness Scott, Jobin Christian, Lund Pauline K., High-Fat Diet: Bacteria Interactions Promote Intestinal Inflammation Which Precedes and Correlates with Obesity and Insulin Resistance in Mouse, 10.1371/journal.pone.0012191
  35. Bäckhed Fredrik, Manchester Jill K., Semenkovich Clay F., Gordon Jeffrey I., Mechanisms underlying the resistance to diet-induced obesity in germ-free mice, 10.1073/pnas.0605374104
  36. Rabot S., Membrez M., Bruneau A., Gerard P., Harach T., Moser M., Raymond F., Mansourian R., Chou C. J., Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism, 10.1096/fj.10-164921
  37. 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
  38. Dodevska Margarita S., Sobajic Sladjana S., Djordjevic Predrag B., Dimitrijevic-Sreckovic Vesna S., Spasojevic-Kalimanovska Vesna V., Djordjevic Brizita I., Effects of total fibre or resistant starch-rich diets within lifestyle intervention in obese prediabetic adults, 10.1007/s00394-015-0831-3
  39. Gentile Christopher L., Ward Emery, Holst Jens Juul, Astrup Arne, Ormsbee Michael J., Connelly Scott, Arciero Paul J., Resistant starch and protein intake enhances fat oxidation and feelings of fullness in lean and overweight/obese women, 10.1186/s12937-015-0104-2
  40. Bodinham C. L., Smith L., Thomas E. L., Bell J. D., Swann J. R., Costabile A., Russell-Jones D., Umpleby A. M., Robertson M. D., Efficacy of increased resistant starch consumption in human type 2 diabetes, 10.1530/ec-14-0036
  41. Bodinham Caroline L., Smith Leanne, Wright John, Frost Gary S., Robertson M. Denise, Dietary Fibre Improves First-phase Insulin Secretion in Overweight Individuals, 10.1371/journal.pone.0040834
  42. 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
  43. Martínez Inés, Lattimer James M, Hubach Kelcie L, Case Jennifer A, Yang Junyi, Weber Casey G, Louk Julie A, Rose Devin J, Kyureghian Gayaneh, Peterson Daniel A, Haub Mark D, Walter Jens, Gut microbiome composition is linked to whole grain-induced immunological improvements, 10.1038/ismej.2012.104
  44. 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
  45. Prawitt J., Abdelkarim M., Stroeve J. H. M., Popescu I., Duez H., Velagapudi V. R., Dumont J., Bouchaert E., van Dijk T. H., Lucas A., Dorchies E., Daoudi M., Lestavel S., Gonzalez F. J., Oresic M., Cariou B., Kuipers F., Caron S., Staels B., Farnesoid X Receptor Deficiency Improves Glucose Homeostasis in Mouse Models of Obesity, 10.2337/db11-0030
  46. Samuel B. S., Shaito A., Motoike T., Rey F. E., Backhed F., Manchester J. K., Hammer R. E., Williams S. C., Crowley J., Yanagisawa M., Gordon J. I., Effects of the gut microbiota on host adiposity are modulated by the short-chain fatty-acid binding G protein-coupled receptor, Gpr41, 10.1073/pnas.0808567105
  47. Lumeng Carey N., Bodzin Jennifer L., Saltiel Alan R., Obesity induces a phenotypic switch in adipose tissue macrophage polarization, 10.1172/jci29881
  48. Weisberg Stuart P., Hunter Deborah, Huber Reid, Lemieux Jacob, Slaymaker Sarah, Vaddi Kris, Charo Israel, Leibel Rudolph L., Jr. Anthony W. Ferrante, CCR2 modulates inflammatory and metabolic effects of high-fat feeding, 10.1172/jci24335
  49. Kanda H., MCP-1 contributes to macrophage infiltration into adipose tissue, insulin resistance, and hepatic steatosis in obesity, 10.1172/jci26498
  50. 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
  51. Muccioli Giulio G, Naslain Damien, Bäckhed Fredrik, Reigstad Christopher S, Lambert Didier M, Delzenne Nathalie M, Cani Patrice D, The endocannabinoid system links gut microbiota to adipogenesis, 10.1038/msb.2010.46
  52. 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
  53. Suhre Karsten, Meisinger Christa, Döring Angela, Altmaier Elisabeth, Belcredi Petra, Gieger Christian, Chang David, Milburn Michael V., Gall Walter E., Weinberger Klaus M., Mewes Hans-Werner, Hrabé de Angelis Martin, Wichmann H.-Erich, Kronenberg Florian, Adamski Jerzy, Illig Thomas, Metabolic Footprint of Diabetes: A Multiplatform Metabolomics Study in an Epidemiological Setting, 10.1371/journal.pone.0013953
  54. Zhao Xinjie, Fritsche Jens, Wang Jiangshan, Chen Jing, Rittig Kilian, Schmitt-Kopplin Philippe, Fritsche Andreas, Häring Hans-Ulrich, Schleicher Erwin D., Xu Guowang, Lehmann Rainer, Metabonomic fingerprints of fasting plasma and spot urine reveal human pre-diabetic metabolic traits, 10.1007/s11306-010-0203-1
  55. Vincent R. P., Omar S., Ghozlan S., Taylor D. R., Cross G., Sherwood R. A., Fandriks L., Olbers T., Werling M., Alaghband-Zadeh J., Roux C. W. l., Higher circulating bile acid concentrations in obese patients with type 2 diabetes, 10.1177/0004563212473450
  56. Wewalka Marlene, Patti Mary-Elizabeth, Barbato Corinne, Houten Sander M., Goldfine Allison B., Fasting Serum Taurine-Conjugated Bile Acids Are Elevated in Type 2 Diabetes and Do Not Change With Intensification of Insulin, 10.1210/jc.2013-3367
  57. Haeusler R. A., Astiarraga B., Camastra S., Accili D., Ferrannini E., Human Insulin Resistance Is Associated With Increased Plasma Levels of 12 -Hydroxylated Bile Acids, 10.2337/db13-0639
  58. Thomas Charles, Gioiello Antimo, Noriega Lilia, Strehle Axelle, Oury Julien, Rizzo Giovanni, Macchiarulo Antonio, Yamamoto Hiroyasu, Mataki Chikage, Pruzanski Mark, Pellicciari Roberto, Auwerx Johan, Schoonjans Kristina, TGR5-Mediated Bile Acid Sensing Controls Glucose Homeostasis, 10.1016/j.cmet.2009.08.001
  59. Perino Alessia, Pols Thijs Willem Hendrik, Nomura Mitsunori, Stein Sokrates, Pellicciari Roberto, Schoonjans Kristina, TGR5 reduces macrophage migration through mTOR-induced C/EBPβ differential translation, 10.1172/jci76289
  60. Ebihara K, Shiraishi R, Okuma K. Hydroxypropyl-modified potato starch increases fecal bile acid excretion in rats. J Nutr. 1998;128:848–54.
  61. Trautwein Elke A, Forgbert Karin, Rieckhoff Dörte, Erbersdobler Helmut F, Impact of β-cyclodextrin and resistant starch on bile acid metabolism and fecal steroid excretion in regard to their hypolipidemic action in hamsters1Presented in part at Experimental Biology 98, 18–22 April, 1998, San Francisco, CA, USA.1, 10.1016/s0005-2760(98)00174-x
  62. van Munster Ivo P., Tangerman Albert, Nagengast Fokko M., Effect of resistant starch on colonic fermentation, bile acid metabolism, and mucosal proliferation, 10.1007/bf02087431
  63. Sayin Sama I., Wahlström Annika, Felin Jenny, Jäntti Sirkku, Marschall Hanns-Ulrich, Bamberg Krister, Angelin Bo, Hyötyläinen Tuulia, Orešič Matej, Bäckhed Fredrik, Gut Microbiota Regulates Bile Acid Metabolism by Reducing the Levels of Tauro-beta-muricholic Acid, a Naturally Occurring FXR Antagonist, 10.1016/j.cmet.2013.01.003
  64. 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
  65. Salonen Anne, Lahti Leo, Salojärvi Jarkko, Holtrop Grietje, Korpela Katri, Duncan Sylvia H, Date Priya, Farquharson Freda, Johnstone Alexandra M, Lobley Gerald E, Louis Petra, Flint Harry J, de Vos Willem M, Impact of diet and individual variation on intestinal microbiota composition and fermentation products in obese men, 10.1038/ismej.2014.63
  66. 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
  67. Bodinham Caroline L., Al-Mana Najlaa M., Smith Leanne, Robertson M. Denise, Endogenous plasma glucagon-like peptide-1 following acute dietary fibre consumption, 10.1017/s0007114513000731
  68. Zhou J., Martin R. J., Tulley R. T., Raggio A. M., McCutcheon K. L., Shen L., Danna S. C., Tripathy S., Hegsted M., Keenan M. J., Dietary resistant starch upregulates total GLP-1 and PYY in a sustained day-long manner through fermentation in rodents, 10.1152/ajpendo.90637.2008
  69. Belobrajdic Damien P, King Roger A, Christophersen Claus T, Bird Anthony R, Dietary resistant starch dose-dependently reduces adiposity in obesity-prone and obesity-resistant male rats, 10.1186/1743-7075-9-93
  70. Harazaki Tomomi, Inoue Seiya, Imai Chihiro, Mochizuki Kazuki, Goda Toshinao, Resistant starch improves insulin resistance and reduces adipose tissue weight and CD11c expression in rat OLETF adipose tissue, 10.1016/j.nut.2013.10.020
  71. Keenan Michael J., Janes Marlene, Robert Julina, Martin Roy J., Raggio Anne M., McCutcheon Kathleen L., Pelkman Christine, Tulley Richard, Goita M'Famara, Durham Holiday A., Zhou June, Senevirathne Reshani N., Resistant starch from high amylose maize (HAM-RS2) reduces body fat and increases gut bacteria in ovariectomized (OVX) rats, 10.1002/oby.20109
  72. Lu Hongjia, Gui Yu, Guo Ting, Wang Qianqian, Liu Xiong, Effect of the particle size of cellulose from sweet potato residues on lipid metabolism and cecal conditions in ovariectomized rats, 10.1039/c4fo00799a
  73. Chen Hsiao-Ling, Lin You-Mei, Wang Yi-Chun, Comparative Effects of Cellulose and Soluble Fibers (Pectin, Konjac Glucomannan, Inulin) on Fecal Water Toxicity toward Caco-2 Cells, Fecal Bacteria Enzymes, Bile Acid, and Short-Chain Fatty Acids, 10.1021/jf102127k
  74. Claus S. P., Ellero S. L., Berger B., Krause L., Bruttin A., Molina J., Paris A., Want E. J., de Waziers I., Cloarec O., Richards S. E., Wang Y., Dumas M.-E., Ross A., Rezzi S., Kochhar S., Van Bladeren P., Lindon J. C., Holmes E., Nicholson J. K., Colonization-Induced Host-Gut Microbial Metabolic Interaction, 10.1128/mbio.00271-10
  75. Swann J. R., Want E. J., Geier F. M., Spagou K., Wilson I. D., Sidaway J. E., Nicholson J. K., Holmes E., Systemic gut microbial modulation of bile acid metabolism in host tissue compartments, 10.1073/pnas.1006734107
  76. Belkaid Yasmine, Hand Timothy W., Role of the Microbiota in Immunity and Inflammation, 10.1016/j.cell.2014.03.011
  77. Lane DJ. 16S/23S rRNA sequencing. In: Stackebrandt E, Goodfellow M, editors. Nucleic acid techniques in bacterial systematics. New York: John Wiley & Sons, Inc; 1991.
  78. Woo K. S., Seib P. A., Cross-Linked Resistant Starch: Preparation and Properties, 10.1094/cchem.2002.79.6.819
  79. Muniyappa R., Lee S., Chen H., Quon M. J., Current approaches for assessing insulin sensitivity and resistance in vivo: advantages, limitations, and appropriate usage, 10.1152/ajpendo.00645.2007
  80. Matthews D. R., Hosker J. P., Rudenski A. S., Naylor B. A., Treacher D. F., Turner R. C., Homeostasis model assessment: insulin resistance and ?-cell function from fasting plasma glucose and insulin concentrations in man, 10.1007/bf00280883
  81. Wallace T. M., Levy J. C., Matthews D. R., Use and Abuse of HOMA Modeling, 10.2337/diacare.27.6.1487
  82. Lee S., Muniyappa R., Yan X., Chen H., Yue L. Q., Hong E.-G., Kim J. K., Quon M. J., Comparison between surrogate indexes of insulin sensitivity and resistance and hyperinsulinemic euglycemic clamp estimates in mice, 10.1152/ajpendo.00676.2007
  83. Eren A. Murat, Vineis Joseph H., Morrison Hilary G., Sogin Mitchell L., A Filtering Method to Generate High Quality Short Reads Using Illumina Paired-End Technology, 10.1371/journal.pone.0066643
  84. Schloss P. D., Westcott S. L., Ryabin T., Hall J. R., Hartmann M., Hollister E. B., Lesniewski R. A., Oakley B. B., Parks D. H., Robinson C. J., Sahl J. W., Stres B., Thallinger G. G., Van Horn D. J., Weber C. F., Introducing mothur: Open-Source, Platform-Independent, Community-Supported Software for Describing and Comparing Microbial Communities, 10.1128/aem.01541-09
  85. Edgar Robert C, UPARSE: highly accurate OTU sequences from microbial amplicon reads, 10.1038/nmeth.2604
  86. Cole James R., Wang Qiong, Fish Jordan A., Chai Benli, McGarrell Donna M., Sun Yanni, Brown C. Titus, Porras-Alfaro Andrea, Kuske Cheryl R., Tiedje James M., Ribosomal Database Project: data and tools for high throughput rRNA analysis, 10.1093/nar/gkt1244
  87. Caporaso J Gregory, Kuczynski Justin, Stombaugh Jesse, Bittinger Kyle, Bushman Frederic D, Costello Elizabeth K, Fierer Noah, Peña Antonio Gonzalez, Goodrich Julia K, Gordon Jeffrey I, Huttley Gavin A, Kelley Scott T, Knights Dan, Koenig Jeremy E, Ley Ruth E, Lozupone Catherine A, McDonald Daniel, Muegge Brian D, Pirrung Meg, Reeder Jens, Sevinsky Joel R, Turnbaugh Peter J, Walters William A, Widmann Jeremy, Yatsunenko Tanya, Zaneveld Jesse, Knight Rob, QIIME allows analysis of high-throughput community sequencing data, 10.1038/nmeth.f.303
  88. Vázquez-Baeza Yoshiki, Pirrung Meg, Gonzalez Antonio, Knight Rob, EMPeror: a tool for visualizing high-throughput microbial community data, 10.1186/2047-217x-2-16
  89. Segata Nicola, Izard Jacques, Waldron Levi, Gevers Dirk, Miropolsky Larisa, Garrett Wendy S, Huttenhower Curtis, Metagenomic biomarker discovery and explanation, 10.1186/gb-2011-12-6-r60
  90. Han Jun, Liu Yang, Wang Renxue, Yang Juncong, Ling Victor, Borchers Christoph H., Metabolic Profiling of Bile Acids in Human and Mouse Blood by LC–MS/MS in Combination with Phospholipid-Depletion Solid-Phase Extraction, 10.1021/ac503816u
  91. Faul Franz, Erdfelder Edgar, Lang Albert-Georg, Buchner Axel, G*Power 3: A flexible statistical power analysis program for the social, behavioral, and biomedical sciences, 10.3758/bf03193146
  92. Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc B. 1995;57:289–300.
  93. R Development Core Team. R: a language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2012. http://www.R-project.org/ . Accessed July 2015.
Bibliographic reference Bindels, Laure B. ; Segura Munoz, Rafael R ; Gomes-Neto, João Carlos ; Mutemberezi, Valentin ; Martínez, Inés ; et. al. Resistant starch can improve insulin sensitivity independently of the gut microbiota. In: Microbiome, Vol. 5, no.1, p. 12 [1-16] (2017)
Permanent URL http://hdl.handle.net/2078.1/182300