Microbiota and Diabetes Mellitus: Role of Lipid Mediators

datacite.rightshttp://purl.org/coar/access_right/c_abf2eng
dc.contributor.authorSalazar, Juan
dc.contributor.authorAngarita, Lissé
dc.contributor.authorMorillo, Valery
dc.contributor.authorNavarro, Carla
dc.contributor.authorMartínez, María Sofía
dc.contributor.authorChacín, Maricarmen
dc.contributor.authorTorres, Wheeler
dc.contributor.authorRajotia, Arush
dc.contributor.authorRojas, Milagros
dc.contributor.authorCano, Clímaco
dc.contributor.authorAñez, Roberto
dc.contributor.authorRojas, Joselyn
dc.contributor.authorBermúdez, Valmore
dc.date.accessioned2021-01-20T23:07:35Z
dc.date.available2021-01-20T23:07:35Z
dc.date.issued2020
dc.description.abstractDiabetes Mellitus (DM) is an inflammatory clinical entity with different mechanisms involved in its physiopathology. Among these, the dysfunction of the gut microbiota stands out. Currently, it is understood that lipid products derived from the gut microbiota are capable of interacting with cells from the immune system and have an immunomodulatory effect. In the presence of dysbiosis, the concentration of lipopolysaccharides (LPS) increases, favoring damage to the intestinal barrier. Furthermore, a pro-inflammatory environment prevails, and a state of insulin resistance and hyperglycemia is present. Conversely, during eubiosis, the production of short-chain fatty acids (SCFA) is fundamental for the maintenance of the integrity of the intestinal barrier as well as for immunogenic tolerance and appetite/satiety perception, leading to a protective effect. Additionally, it has been demonstrated that alterations or dysregulation of the gut microbiota can be reversed by modifying the eating habits of the patients or with the administration of prebiotics, probiotics, and symbiotics. Similarly, different studies have demonstrated that drugs like Metformin are capable of modifying the composition of the gut microbiota, promoting changes in the biosynthesis of LPS, and the metabolism of SCFA.eng
dc.format.mimetypepdfspa
dc.identifier.doihttps://doi.org/10.3390/nu12103039
dc.identifier.issn20726643
dc.identifier.urihttps://hdl.handle.net/20.500.12442/7003
dc.identifier.urlhttps://www.mdpi.com/2072-6643/12/10/3039
dc.language.isoengeng
dc.publisherMDPI
dc.rightsAttribution-NonCommercial-NoDerivatives 4.0 Internacional*
dc.rights.accessrightsinfo:eu-repo/semantics/openAccesseng
dc.rights.urihttp://creativecommons.org/licenses/by-nc-nd/4.0/*
dc.sourceNutrientseng
dc.sourceVol. 12, No. 10, 2020
dc.subjectDiabetesspa
dc.subjectInflammationeng
dc.subjectMicrobiotaeng
dc.subjectDysbiosiseng
dc.subjectLipopolysaccharideseng
dc.subjectShort-chaineng
dc.subjectFatty acidseng
dc.titleMicrobiota and Diabetes Mellitus: Role of Lipid Mediatorseng
dc.type.driverinfo:eu-repo/semantics/articleeng
dc.type.spaArtículo científicospa
dcterms.referencesInternational Diabetes Federation. IDF Diabetes Atlas. 2015. Available online: https://www.idf.org/elibrary/ epidemiology-research/diabetes-atlas/13-diabetes-atlas-seventh-edition.html (accessed on 20 March 2020).spa
dcterms.referencesMinistery of Health. Bolivarian Republic of Venezuela. Anuary of Morbility. 2011. Available online: https: //www.ovsalud.org/descargas/publicaciones/documentos-oficiales/Anuario-Morbilidad-2011.pdf (accessed on 20 March 2020).eng
dcterms.referencesWorld Health Organization. Global Report on Diabetes; World Health Organization: Geneva, Switzerland, 2016. Available online: http://apps.who.int/iris/bitstream/10665/204871/1/9789241565257_eng.pdf (accessed on 20 March 2020).eng
dcterms.referencesDe La Cruz Vargas, J.A.; Dos Santos, F.; Dyzinger, W.; Herzog, S. Medicina Del Estilo de Vida: Trabajando Juntos Para Revertir La Epidemia de Las Enfermedades Crónicas En Latinoamérica. Cienc. Innov. Salud 2017, 4. [CrossRef]spa
dcterms.referencesBaratieri, T.; Dal Santo Ottoni, J.; Luciana Botti, M.; Serpa Maicel, R.D.C.; Gramazio Soares, L. Risco Cardiovascular Em Usuários de Programa de Atenção a Hipertensos e Diabéticos Em Um Município Do Paraná-Brasil. Cienc. Innov. Salud 2014, 2. [CrossRef]spa
dcterms.referencesMorales, J.; Carcausto, W.; Varillas, Y.; Pérez, J.; Salsavilca, E.; Castro, I.; Rivera, M.; Quispe, M. Actividad Física En Pacientes Con Diabetes Mellitus Del Primer Nivel de Atención de Lima Norte. Rev. Latinoam. Hipertens. 2018, 13, 49–54spa
dcterms.referencesDe Fronzo, R.A. From the Triumvirate to the Ominous Octet: A New Paradigm for the Treatment of Type 2 Diabetes Mellitus. Diabetes 2009, 58, 773–795. [CrossRef] [PubMed]eng
dcterms.referencesSchwartz, S.S.; Epstein, S.; Corkey, B.E.; Grant, S.F.A.; Gavin, J.R.; Aguilar, R.B. The Time Is Right for a New Classification System for Diabetes: Rationale and Implications of the β-Cell–Centric Classification Schema. Diabetes Care 2016, 39, 179–186. [CrossRefeng
dcterms.referencesVatanen, T.; Kostic, A.D.; D’Hennezel, E.; Siljander, H.; Franzosa, E.A.; Yassour, M.; Kolde, R.; Vlamakis, H.; Arthur, T.D.; Hämäläinen, A.-M.; et al. Variation in Microbiome LPS Immunogenicity Contributes to Autoimmunity in Humans. Cell 2016, 165, 842–853. [CrossRef]eng
dcterms.referencesBrown, K.; Godovannyi, A.; Ma, C.; Zhang, Y.; Ahmadi-Vand, Z.; Dai, C.; Gorzelak, M.A.; Chan, Y.; Chan, J.M.; Lochner, A.; et al. Prolonged Antibiotic Treatment Induces a Diabetogenic Intestinal Microbiome That Accelerates Diabetes in NOD Mice. ISME J. 2016, 10, 321–332. [CrossRef]eng
dcterms.referencesPalacios, T.; Vitetta, L.; Coulson, S.; Madigan, C.D.; Denyer, G.S.; Caterson, I.D. The Effect of a Novel Probiotic on Metabolic Biomarkers in Adults with Prediabetes and Recently Diagnosed Type 2 Diabetes Mellitus: Study Protocol for a Randomized Controlled Trial. Trials 2017, 18. [CrossRef]eng
dcterms.referencesPeng, J.; Narasimhan, S.; Marchesi, J.R.; Benson, A.; Wong, F.S.; Wen, L. Long Term Effect of Gut Microbiota Transfer on Diabetes Development. J. Autoimmun. 2014, 53, 85–94. [CrossRef]eng
dcterms.referencesGonzalez, C.M.C.; Quiroz, E.A.N.; Lastre-Amell, G.; Oróstegui-Santander, M.A.; Peña, G.E.G.; Sucerquia, A.; Carrero, L.L.S. Dislipidemia como factor de riesgo cardiovascular: Uso de probióticos en la terapéutica nutricional. Arch. Venez. Farmacol. Ter. 2020, 39, 126–139.spa
dcterms.referencesHerder, C.; Færch, K.; Carstensen-Kirberg, M.; Lowe, G.D.; Haapakoski, R.; Witte, D.R.; Brunner, E.J.; Roden, M.; Tabák, A.G.; Kivimäki, M.; et al. Biomarkers of Subclinical Inflammation and Increases in Glycaemia, Insulin Resistance and Beta-Cell Function in Non-Diabetic Individuals: The Whitehall II Study. Eur. J. Endocrinol. 2016, 175, 367–377. [CrossRef] [PubMed]eng
dcterms.referencesPouvreau, C.; Dayre, A.; Butkowski, E.G.; de Jong, B.; Jelinek, H.F. Inflammation and Oxidative Stress Markers in Diabetes and Hypertension. J. Inflamm. Res. 2018, 11, 61–68. [CrossRef] [PubMed]eng
dcterms.referencesOdegaard, A.O.; Jacobs, D.R.; Sanchez, O.A.; Goff, D.C.; Reiner, A.P.; Gross, M.D. Oxidative Stress, Inflammation, Endothelial Dysfunction and Incidence of Type 2 Diabetes. Cardiovasc. Diabetol. 2016, 15, 51. [CrossRef] [PubMed]eng
dcterms.referencesBermudez, V.; Salazar, J.; Gonzalez, R.; Ortega, A.; Calvo, M.; Olivar, L.C.; Morillo, J.; Miquilena, E.; Chavez-Castillo, M.; Chaparro, R.; et al. Prevalence and Risk Factors Associated with Impaired Fasting Glucose in Adults from Maracaibo City, Venezuela. J. Diabetes Metab. 2019, 7, 100683. [CrossRef]eng
dcterms.referencesRojas, J.; Bermudez, V.; Palmar, J.; Martínez, M.S.; Olivar, L.C.; Nava, M.; Tomey, D.; Rojas, M.; Salazar, J.; Garicano, C.; et al. Pancreatic Beta Cell Death: Novel Potential Mechanisms in Diabetes Therapy. J. Diabetes Res. 2018, 2018, 9601801. [CrossRef]eng
dcterms.referencesMobini, R.; Tremaroli, V.; Ståhlman, M.; Karlsson, F.; Levin, M.; Ljungberg, M.; Sohlin, M.; Bertéus Forslund, H.; Perkins, R.; Bäckhed, F.; et al. Metabolic Effects of Lactobacillus Reuteri DSM 17938 in People with Type 2 Diabetes: A Randomized Controlled Trial. Diabetes Obes. Metab. 2017, 19, 579–589. [CrossRef] [PubMed]eng
dcterms.referencesRodríguez Nieves, R.R.; Torres Ruiz, L.E.; Sarmiento Segarra, K.B.; Narea Illescas, D.I.; Araque Pluas, I.V.; Apolo Montero, A.M.; Ibarra Vélez, L.S.; Alvarado Chiquito, O.L. Prevalencia de Síndrome Metabólico En Trabajadores de Una Empresa de Construcción En Guayaquil, Ecuador. Rev. Latinoam. Hipertens. 2019, 14, 638–643.spa
dcterms.referencesAhmad, R.; Thomas, R.; Kochumon, S.; Sindhu, S. Increased Adipose Tissue Expression of IL-18R and Its Ligand IL-18 Associates with Inflammation and Insulin Resistance in Obesity. Immun. Inflamm. Dis. 2017, 5, 318–335. [CrossRef]eng
dcterms.referencesYan, Y.; Li, S.; Liu, Y.; Bazzano, L.; He, J.; Mi, J.; Chen, W. Temporal Relationship between Inflammation and Insulin Resistance and Their Joint Effect on Hyperglycemia: The Bogalusa Heart Study. Cardiovasc. Diabetol. 2019, 18, 109. [CrossRef]eng
dcterms.referencesEsser, N.; Legrand-Poels, S.; Piette, J.; Scheen, A.J.; Paquot, N. Inflammation as a Link between Obesity, Metabolic Syndrome and Type 2 Diabetes. Diabetes Res. Clin. Pract. 2014, 105, 141–150. [CrossRef]eng
dcterms.referencesZozulinska, D.; Wierusz-Wysocka, B. Type 2 Diabetes Mellitus as Inflammatory Disease. Diabetes Res. Clin. Pract. 2006, 74, S12–S16. [CrossRef]spa
dcterms.referencesRoohi, A.; Tabrizi, M.; Abbasi, F.; Ataie-Jafari, A.; Nikbin, B.; Larijani, B.; Qorbani, M.; Meysamie, A.; Asgarian-Omran, H.; Nikmanesh, B.; et al. Serum IL-17, IL-23, and TGF-β Levels in Type 1 and Type 2 Diabetic Patients and Age-Matched Healthy Controls. Biomed. Res. Int. 2014, 2014, 718946. [CrossRef] [PubMed]eng
dcterms.referencesAbdel-Moneim, A.; Bakery, H.H.; Allam, G. The Potential Pathogenic Role of IL-17/Th17 Cells in Both Type 1 and Type 2 Diabetes Mellitus. Biomed. Pharmacother. 2018, 101, 287–292. [CrossRef] [PubMed]eng
dcterms.referencesVon Scholten, B.J.; Reinhard, H.; Hansen, T.W.; Schalkwijk, C.G.; Stehouwer, C.; Parving, H.-H.; Jacobsen, P.K.; Rossing, P. Markers of Inflammation and Endothelial Dysfunction Are Associated with Incident Cardiovascular Disease, All-Cause Mortality, and Progression of Coronary Calcification in Type 2 Diabetic Patients with Microalbuminuria. J. Diabetes Complicat. 2016, 30, 248–255. [CrossRef]eng
dcterms.referencesSigurdardottir, S.; Zapadka, T.E.; Lindstrom, S.I.; Liu, H.; Taylor, B.E.; Lee, C.A.; Kern, T.S.; Taylor, P.R. Diabetes-Mediated IL-17A Enhances Retinal Inflammation, Oxidative Stress, and Vascular Permeability. Cell. Immunol. 2019, 341, 103921. [CrossRef]eng
dcterms.referencesRomán-Pintos, L.M.; Villegas-Rivera, G.; Rodríguez-Carrizalez, A.D.; Miranda-Díaz, A.G.; Cardona-Muñoz, E.G. Diabetic Polyneuropathy in Type 2 Diabetes Mellitus: Inflammation, Oxidative Stress, and Mitochondrial Function. J. Diabetes Res. 2016, 2016, 3425617. [CrossRef]spa
dcterms.referencesBurcelin, R. Gut Microbiota and Immune Crosstalk in Metabolic Disease. Mol. Metab. 2016, 5, 771–781. [CrossRef]eng
dcterms.referencesHuang, X.; Yan, D.; Xu, M.; Li, F.; Ren, M.; Zhang, J.; Wu, M. Interactive Association of Lipopolysaccharide and Free Fatty Acid with the Prevalence of Type 2 Diabetes: A Community-Based Cross-Sectional Study. J. Diabetes Investig. 2019, 10, 1438–1446. [CrossRef]eng
dcterms.referencesKhondkaryan, L.; Margaryan, S.; Poghosyan, D.; Manukyan, G. Impaired Inflammatory Response to LPS in Type 2 Diabetes Mellitus. Int. J. Inflam. 2018, 2018, 2157434. [CrossRef]eng
dcterms.referencesZhao, L.; Zhang, F.; Ding, X.; Wu, G.; Lam, Y.Y.; Wang, X.; Fu, H.; Xue, X.; Lu, C.; Ma, J.; et al. Gut Bacteria Selectively Promoted by Dietary Fibers Alleviate Type 2 Diabetes. Science 2018, 359, 1151–1156. [CrossRef]eng
dcterms.referencesLi, M.; van Esch, B.C.A.M.; Wagenaar, G.T.M.; Garssen, J.; Folkerts, G.; Henricks, P.A.J. Pro- and Anti-Inflammatory Effects of Short Chain Fatty Acids on Immune and Endothelial Cells. Eur. J. Pharmacol. 2018, 831, 52–59. [CrossRef] [PubMed]eng
dcterms.referencesTurnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The Human Microbiome Project. Nature 2007, 449, 804–810. [CrossRef] [PubMed]eng
dcterms.referencesMartí, J.M.; Martínez-Martínez, D.; Rubio, T.; Gracia, C.; Peña, M.; Latorre, A.; Moya, A.; Garay, C.P. Health and Disease Imprinted in the Time Variability of the Human Microbiome. MSystems 2017, 2. [CrossRef] [PubMed]eng
dcterms.referencesRomero, G. Influencia de La Microbiota Intestinal En La Enfermedad Hepática Crónica. Su Rol En El Hepatocarcinoma. Gen 2016, 70, 64–69spa
dcterms.referencesVargas-Robles, D.D.; Domínguez-Bello, M.G. Microbiota de los indígenas del Amazonas venezolano: Influencia de los estilos de vida. Gac. Med. Caracas 2020, 126, 291–303.spa
dcterms.referencesHill, C.J.; Lynch, D.B.; Murphy, K.; Ulaszewska, M.; Jeffery, I.B.; O’Shea, C.A.; Watkins, C.; Dempsey, E.; Mattivi, F.; Tuohy, K.; et al. Evolution of Gut Microbiota Composition from Birth to 24 Weeks in the INFANTMET Cohort. Microbiome 2017, 5, 4. [CrossRef]eng
dcterms.referencesTorres, Y.; Bermúdez, V.; Garicano, C.; Vilasmil, N.; Bautista, J.; Martínez, M.S.; Rojas-Quintero, J. Desarrollo del sistema inmunológico ¿naturaleza o crianza? Arch. Venez. Farmacol. Ter. 2017, 36, 144–151.spa
dcterms.referencesFaneite Antique, D.P.; Faneite Campos, J. Microbioma perinatal: Nuevos horizontes de la vida. Gac. Med. Caracas 2020, 123, 94–106spa
dcterms.referencesDuranti, S.; Lugli, G.A.; Milani, C.; James, K.; Mancabelli, L.; Turroni, F.; Alessandri, G.; Mangifesta, M.; Mancino, W.; Ossiprandi, M.C.; et al. Bifidobacterium bifidum and the infant gut microbiota: An intriguing case of microbe-host co-evolution. Environ. Microbiol. 2019, 21, 3683–3695. [CrossRef]eng
dcterms.referencesBiagi, E.; Nylund, L.; Candela, M.; Ostan, R.; Bucci, L.; Pini, E.; Nikkïla, J.; Monti, D.; Satokari, R.; Franceschi, C.; et al. Through Ageing, and Beyond: Gut Microbiota and Inflammatory Status in Seniors and Centenarians. PLoS ONE 2010, 5, e10667. [CrossRef]eng
dcterms.referencesJeffery, I.B.; Lynch, D.B.; O’Toole, P.W. Composition and Temporal Stability of the Gut Microbiota in Older Persons. ISME J. 2016, 10, 170–182. [CrossRef] [PubMed]eng
dcterms.referencesAlkanani, A.K.; Hara, N.; Gottlieb, P.A.; Ir, D.; Robertson, C.E.; Wagner, B.D.; Frank, D.N.; Zipris, D. Alterations in Intestinal Microbiota Correlate With Susceptibility to Type 1 Diabetes. Diabetes 2015, 64, 3510–3520. [CrossRef] [PubMed]eng
dcterms.referencesClarke, S.F.; Murphy, E.F.; O’Sullivan, O.; Lucey, A.J.; Humphreys, M.; Hogan, A.; Hayes, P.; O’Reilly, M.; Jeffery, I.B.; Wood-Martin, R.; et al. Exercise and Associated Dietary Extremes Impact on Gut Microbial Diversity. Gut 2014, 63, 1913–1920. [CrossRef] [PubMed]eng
dcterms.referencesCarding, S.; Verbeke, K.; Vipond, D.T.; Corfe, B.M.; Owen, L.J. Dysbiosis of the Gut Microbiota in Disease. Microb. Ecol. Health Dis. 2015, 26. [CrossRef] [PubMed]eng
dcterms.referencesPomié, C.; Blasco-Baque, V.; Klopp, P.; Nicolas, S.; Waget, A.; Loubières, P.; Azalbert, V.; Puel, A.; Lopez, F.; Dray, C.; et al. Triggering the Adaptive Immune System with Commensal Gut Bacteria Protects against Insulin Resistance and Dysglycemia. Mol. Metab. 2016, 5, 392–403. [CrossRef] [PubMedeng
dcterms.referencesMorales, P.; Fujio, S.; Navarrete, P.; Ugalde, J.A.; Magne, F.; Carrasco-Pozo, C.; Tralma, K.; Quezada, M.; Hurtado, C.; Covarrubias, N.; et al. Impact of Dietary Lipids on Colonic Function and Microbiota: An Experimental Approach Involving Orlistat-Induced Fat Malabsorption in Human Volunteers. Clin. Transl. Gastroenterol. 2016, 7, e161. [CrossRef]eng
dcterms.referencesEndesfelder, D.; Zu Castell, W.; Ardissone, A.; Davis-Richardson, A.G.; Achenbach, P.; Hagen, M.; Pflueger, M.; Gano, K.A.; Fagen, J.R.; Drew, J.C.; et al. Compromised Gut Microbiota Networks in Children with Anti-Islet Cell Autoimmunity. Diabetes 2014, 63, 2006–2014. [CrossRef]eng
dcterms.referencesGhosh, S.S.; Wang, J.; Yannie, P.; Ghosh, S. Intestinal Barrier Dysfunction, LPS Translocation, and Disease Development. J. Endocr. Soc. 2020, 4, bvz039. [CrossRef]eng
dcterms.referencesCani, 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. Diabetes 2008, 57, 1470–1481. [CrossRef]eng
dcterms.referencesCani, 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. Diabetes 2008, 57, 1470–1481. [CrossRef]eng
dcterms.referencesTopchiy, E.; Cirstea, M.; Kong, H.; Boyd, J.; Wang, Y.; Russell, J.; Waley, K. Lipopolysaccharide Is Cleared from the Circulation by Hepatocytes via the Low Density Lipoprotein Receptor. PLoS ONE 2016, 11, e0155030. [CrossRef]eng
dcterms.referencesRohr, M.W.; Narasimhulu, C.; Rudeski-Rohr, T.; Parthasarathy, S. Negative Effects of a High-Fat Diet on Intestinal Permeability: A Review. Adv. Nutr. 2020, 11, 77–91. [CrossRef] [PubMed]eng
dcterms.referencesLam, Y.Y.; Ha, C.W.Y.; Hoffmann, J.M.A.; Oscarsson, J.; Dinudom, A.; Mather, T.J.; Cook, D.I.; Hunt, N.H.; Caterson, I.D.; Holmes, A.J.; et al. Effects of Dietary Fat Profile on Gut Permeability and Microbiota and Their Relationships with Metabolic Changes in Mice. Obesity 2015, 23, 1429–1439. [CrossRef] [PubMed]eng
dcterms.referencesGuo, S.; Al-Sadi, R.; Said, H.M.; Ma, T.Y. Lipopolysaccharide Causes an Increase in Intestinal Tight Junction Permeability in Vitro and in Vivo by Inducing Enterocyte Membrane Expression and Localization of TLR-4 and CD14. Am. J. Pathol. 2013, 182, 375–387. [CrossRef] [PubMed]eng
dcterms.referencesGuo, S.; Nighot, M.; Al-Sadi, R.; Alhmoud, T.; Nighot, P.; Ma, T.Y. Lipopolysaccharide Regulation of Intestinal Tight Junction Permeability Is Mediated by TLR4 Signal Transduction Pathway Activation of FAK and MyD88. J. Immunol. 2015, 195, 4999–5010. [CrossRef]eng
dcterms.referencesGaridou, L.; Pomié, C.; Klopp, P.; Waget, A.; Charpentier, J.; Aloulou, M.; Giry, A.; Serino, M.; Stenman, L.; Lahtinen, S.; et al. The Gut Microbiota Regulates Intestinal CD4 T Cells Expressing RORγt and Controls Metabolic Disease. Cell. Metab. 2015, 22, 100–112. [CrossRef]eng
dcterms.referencesCavallari, J.F.; Denou, E.; Foley, K.P.; Khan, W.I.; Schertzer, J.D. Different Th17 Immunity in Gut, Liver, and Adipose Tissues during Obesity: The Role of Diet, Genetics, and Microbes. Gut Microbes 2016, 7, 82–89. [CrossRef]eng
dcterms.referencesGomes, J.; de Assis, J.; Gonçalves, R. Metabolic endotoxemia and diabetes mellitus: A systematic review. Metabolism 2017, 86, 133–144. [CrossRef]eng
dcterms.referencesMatheus, V.A.; Monteiro, L.; Oliveira, R.B.; Maschio, D.A.; Collares-Buzato, C.B. Butyrate Reduces High-Fat Diet-Induced Metabolic Alterations, Hepatic Steatosis and Pancreatic Beta Cell and Intestinal Barrier Dysfunctions in Prediabetic Mice. Exp. Biol. Med. 2017. [CrossRef]eng
dcterms.referencesPedersen, C.; Gallagher, E.; Horton, F.; Ellis, R.J.; Ijaz, U.Z.; Wu, H.; Jaiyeola, E.; Diribe, O.; Duparc, T.; Cani, P.D.; et al. Host–Microbiome Interactions in Human Type 2 Diabetes Following Prebiotic Fibre (Galacto-Oligosaccharide) Intake. Br. J. Nutr. 2016, 116, 1869–1877. [CrossRef]eng
dcterms.referencesSong, M.J.; Kim, K.H.; Yoon, J.M.; Kim, J.B. Activation of Toll-like Receptor 4 Is Associated with Insulin Resistance in Adipocytes. Biochem. Biophys. Res. Commun. 2006, 346, 739–745. [CrossRef]eng
dcterms.referencesAmyot, J.; Semache, M.; Ferdaoussi, M.; Fontés, G.; Poitout, V. Lipopolysaccharides Impair Insulin Gene Expression in Isolated Islets of Langerhans via Toll-Like Receptor-4 and NF-KB Signalling. PLoS ONE 2012, 7, e36200. [CrossRef] [PubMed]eng
dcterms.referencesCao, J.; Peng, J.; An, H.; He, Q.; Boronina, T.; Guo, S.; White, M.F.; Cole, P.A.; He, L. Endotoxemia-Mediated Activation of Acetyltransferase P300 Impairs Insulin Signaling in Obesity. Nat. Commun. 2017, 8, 131. [CrossRef] [PubMed]eng
dcterms.referencesKelder, T.; Stroeve, J.H.M.; Bijlsma, S.; Radonjic, M.; Roeselers, G. Correlation Network Analysis Reveals Relationships between Diet-Induced Changes in Human Gut Microbiota and Metabolic Health. Nutr. Diabetes 2014, 4, e122. [CrossRef] [PubMed]eng
dcterms.referencesMariat, D.; Firmesse, O.; Levenez, F.; Guimarăes, V.; Sokol, H.; Doré, J.; Corthier, G.; Furet, J.P. The Firmicutes/Bacteroidetes ratio of the human microbiota changes with age. BMC Microbiol. 2009, 9, 123. [CrossRef]eng
dcterms.referencesSikalidis, A.; Maykish, A. The Gut Microbiome and Type 2 Diabetes Mellitus: Discussing a Complex Relationship. Biomedicines 2020, 8, 8. [CrossRef]eng
dcterms.referencesMagne, F.; Gotteland, M.; Gauthier, L.; Zazueta, A.; Pesoa, S.; Navarrete, P.; Balamurugan, R. The Firmicutes/Bacteroidetes Ratio: A Relevant Marker of Gut Dysbiosis in Obese Patients. Nutrients 2020, 12, 1474. [CrossRef]eng
dcterms.referencesLarsen, N.; Vogensen, F.; Van den Berg, F.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sørensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut Microbiota in Human Adults with Type 2 Diabetes Differs from Non-Diabetic Adults. PLoS ONE 2010, 5, e9085. [CrossRef]eng
dcterms.referencesEndesfelder, D.; Engel, M.; Davis-Richardson, A.G.; Ardissone, A.N.; Achenbach, P.; Hummel, S.; Winkler, C.; Atkinson, M.; Schatz, D.; Triplett, E.; et al. Towards a Functional Hypothesis Relating Anti-Islet Cell Autoimmunity to the Dietary Impact on Microbial Communities and Butyrate Production. Microbiome 2016, 4, 17. [CrossRef]eng
dcterms.referencesRuiz, L.; Delgado, S.; Ruas-Madiedo, P.; Sánchez, B.; Margolles, A. Bifidobacteria and Their Molecular Communication with the Immune System. Front. Microbiol. 2017, 8, 2345. [CrossRef]spa
dcterms.referencesXu, J.; Lian, F.; Zhao, L.; Zhao, Y.; Chen, X.; Zhang, X.; Guo, Y.; Zhang, C.; Zgou, Q.; Xue, Z.; et al. Structural modulation of gut microbiota during alleviation of type 2 diabetes with a Chinese herbal formula. ISME J. 2015, 9, 552–562. [CrossRef]eng
dcterms.referencesYanagibashi, T.; Hosono, A.; Oyama, A.; Tsuda, M.; Suzuki, A.; Hachimura, S.; Takahashi, Y.; Momose, Y.; Itoh, K.; Hirayama, K.; et al. IgA Production in the Large Intestine Is Modulated by a Different Mechanism than in the Small Intestine: Bacteroides Acidifaciens Promotes IgA Production in the Large Intestine by Inducing Germinal Center Formation and Increasing the Number of IgA+ B Cells. Immunobiology 2013, 218, 645–651. [CrossRef] [PubMed]eng
dcterms.referencesMurri, M.; Leiva, I.; Gomez-Zumaquero, J.M.; Tinahones, F.J.; Cardona, F.; Soriguer, F.; Queipo-Ortuño, M.I. Gut Microbiota in Children with Type 1 Diabetes Differs from That in Healthy Children: A Case-Control Study. BMC Med. 2013, 11, 46. [CrossRef] [PubMed]eng
dcterms.referencesJandhyala, S.M.; Madhulika, A.; Deepika, G.; Rao, G.V.; Reddy, D.N.; Subramanyam, C.; Sasikala, M.; Talukdar, R. Altered Intestinal Microbiota in Patients with Chronic Pancreatitis: Implications in Diabetes and Metabolic Abnormalities. Sci. Rep. 2017, 7, 43640. [CrossRef] [PubMed]eng
dcterms.referencesRemely, M.; Aumueller, E.; Merold, C.; Dworzak, S.; Hippe, B.; Zanner, J.; Pointner, A.; Brath, H.; Haslberger, A.G. Effects of Short Chain Fatty Acid Producing Bacteria on Epigenetic Regulation of FFAR3 in Type 2 Diabetes and Obesity. Gene 2014, 537, 85–92. [CrossRef]eng
dcterms.referencesGurung, M.; Li, Z.; You, H.; Rodrigues, R.; Jump, D.B.; Morgun, A.; Shulzhenko, N. Role of gut microbiota in type 2 diabetes pathophysiology. EBioMedicine 2020, 51, 102590. [CrossRef]eng
dcterms.referencesKim, C.H. Microbiota or Short-Chain Fatty Acids: Which Regulates Diabetes. Cell. Mol. Immunol. 2018, 15, 88–91. [CrossRef]eng
dcterms.referencesGarcía, P.O. La fibra alimentaria y su uso terapéutico en algunas enfermedades crónicas. Gac. Med. Caracas 2020, 120, 107–114.spa
dcterms.referencesPuddu, A.; Sanguineti, R.; Montecucco, F.; Viviani, G.L. Evidence for the Gut Microbiota Short-Chain Fatty Acids as Key Pathophysiological Molecules Improving Diabetes. Mediat. Inflamm. 2014, 2014, 162021. [CrossRef]eng
dcterms.referencesMyhrstad, M.C.W.; Tunsjø, H.; Charnock, C.; Telle-Hansen, V.H. Dietary Fiber, Gut Microbiota, and Metabolic Regulation-Current Status in Human Randomized Trials. Nutrients 2020, 12, 859. [CrossRef]eng
dcterms.referencesRatajczak, W.; Rył, A.; Mizerski, A.; Walczakiewicz, K.; Sipak, O.; Laszczy ´nska, M. Immunomodulatory Potential of Gut Microbiome-Derived Short-Chain Fatty Acids (SCFAs). Acta Biochim. Pol. 2019, 66, 1–12. [CrossRef]eng
dcterms.referencesDávila, L.A.; Pirela, V.B.; Díaz, W.; Villasmil, N.R.; León, S.C.; Contreras, M.C.E.; Bonacich, K.B.; Agüero, S.D.; Vergara, P.C.; Bonacich, R.B.; et al. The Microbiome and the Epigenetics of Diabetes Mellitus. In Diabetes Food Plan; Waisundara, V., Ed.; InTech: London, UK, 2018.spa
dcterms.referencesKimura, I.; Ichimura, A.; Ohue-Kitano, R.; Igarashi, M. Free Fatty Acid Receptors in Health and Disease. Physiol. Rev. 2020, 100, 171–210. [CrossRef] [PubMed]eng
dcterms.referencesLee, S.U.; In, H.J.; Kwon, M.S.; Park, B.; Jo, M.; Kim, M.-O.; Cho, S.; Lee, S.; Lee, H.-J.; Kwak, Y.S.; et al. β-Arrestin 2 Mediates G Protein-Coupled Receptor 43 Signals to Nuclear Factor-KB. Biol. Pharm. Bull. 2013, 36, 1754–1759. [CrossRef] [PubMed]eng
dcterms.referencesCox, M.A.; Jackson, J.; Stanton, M.; Rojas-Triana, A.; Bober, L.; Laverty, M.; Yang, X.; Zhu, F.; Liu, J.; Wang, S.; et al. Short-Chain Fatty Acids Act as Antiinflammatory Mediators by Regulating Prostaglandin E(2) and Cytokines. World J. Gastroenterol. 2009, 15, 5549–5557. [CrossRef] [PubMed]eng
dcterms.referencesHernández, M.A.G.; Canfora, E.E.; Jocken, J.W.E.; Blaak, E.E. The Short-Chain Fatty Acid Acetate in Body Weight Control and Insulin Sensitivity. Nutrients 2019, 11, 1943. [CrossRef]eng
dcterms.referencesLarasati, R.A.; Harbuwono, D.S.; Rahajeng, E.; Pradipta, S.; Nuraeni, H.S.; Susilowati, A.; Wibowo, H. The Role of Butyrate on Monocyte Migration and Inflammation Response in Patient with Type 2 Diabetes Mellitus. Biomedicines 2019, 7, 74. [CrossRef]eng
dcterms.referencesChun, E.; Lavoie, S.; Fonseca-Pereira, D.; Bae, S.; Michaud, M.; Hoveyda, H.R.; Fraser, G.L.; Gallini Comeau, C.A.; Glickman, J.N.; Fuller, M.H.; et al. Metabolite-Sensing Receptor Ffar2 Regulates Colonic Group 3 Innate Lymphoid Cells and Gut Immunity. Immunity 2019, 51, 871–884. [CrossRef]eng
dcterms.referencesWu, W.; Sun, M.; Chen, F.; Cao, A.T.; Liu, H.; Zhao, Y.; Huang, X.; Xiao, Y.; Yao, S.; Zhao, Q.; et al. Microbiota Metabolite Short-Chain Fatty Acid Acetate Promotes Intestinal IgA Response to Microbiota Which Is Mediated by GPR43. Mucosal. Immunol. 2017, 10, 946–956. [CrossRef]eng
dcterms.referencesKim, C.H.; Park, J.; Kim, M. Gut Microbiota-Derived Short-Chain Fatty Acids, T Cells, and Inflammation. Immune Netw. 2014, 14, 277–288. [CrossRef]eng
dcterms.referencesAstakhova, L.; Ngara, M.; Babich, O.; Prosekov, A.; Asyakina, L.; Dyshlyuk, L.; Midtvedt, T.; Zhou, X.; Ernberg, I.; Matskova, L. Short Chain Fatty Acids (SCFA) Reprogram Gene Expression in Human Malignant Epithelial and Lymphoid Cells. PLoS ONE 2016, 11, e0154102. [CrossRef]eng
dcterms.referencesYap, Y.A.; Mariño, E. Dietary SCFAs Immunotherapy: Reshaping the Gut Microbiota in Diabetes. In SpringerLink; Springer: New York, NY, USA, 2020. [CrossRef]eng
dcterms.referencesMariño, E.; Richards, J.L.; McLeod, K.H.; Stanley, D.; Yap, Y.A.; Knight, J.; McKenzie, C.; Kranich, J.; Oliveira, A.C.; Rossello, F.J.; et al. Gut Microbial Metabolites Limit the Frequency of Autoimmune T Cells and Protect against Type 1 Diabetes. Nat. Immunol. 2017, 18, 552–562. [CrossRef]eng
dcterms.referencesCarpio Duran, A.L.; Duran Medina, M.F.; Andrade Valdivieso, M.R.; Espinoza Dunn, M.A.; Rodas Torres, W.P.; Abad Barrera, L.N.; Rodríguez Barzola, C.V.; Yagual Villon, O.A. Terapia Incretinomimética: Evidencia Clínica de La Eficacia de Los Agonistas Del GLP-1R y Sus Efectos Cardio-Protectores. Rev. Latinoam. Hipertens. 2018, 13, 400–415spa
dcterms.referencesRahat-Rozenbloom, S.; Fernandes, J.; Cheng, J.; Wolever, T.M.S. Acute Increases in Serum Colonic Short-Chain Fatty Acids Elicited by Inulin Do Not Increase GLP-1 or PYY Responses but May Reduce Ghrelin in Lean and Overweight Humans. Eur. J. Clin. Nutr. 2017, 71, 953–958. [CrossRef] [PubMed]eng
dcterms.referencesBjerg, A.T.; Kristensen, M.; Ritz, C.; Holst, J.J.; Rasmussen, C.; Leser, T.D.; Wellejus, A.; Astrup, A. Lactobacillus Paracasei Subsp Paracasei L. Casei W8 Suppresses Energy Intake Acutely. Appetite 2014, 82, 111–118. [CrossRef] [PubMed]eng
dcterms.referencesDe Velasco, P.; Ferreira, A.; Crovesy, L.; Marine, T.; Das Graças Tavares do Carmo, M. Fatty Acids, Gut Microbiota, and the Genesis of Obesity. In Biochemistry and Health Benefits of Fatty Acids; Waisundara, V., Ed.; IntechOpen: London, UK, 2018.eng
dcterms.referencesGrasset, E.; Puel, A.; Charpentier, J.; Collet, X.; Christensen, J.E.; Tercé, F.; Burcelin, R. A Specific Gut Microbiota Dysbiosis of Type 2 Diabetic Mice Induces GLP-1 Resistance through an Enteric NO-Dependent and Gut-Brain Axis Mechanism. Cell Metab. 2017, 25, 1075–1090. [CrossRef] [PubMed]eng
dcterms.referencesPerry, R.J.; Peng, L.; Barry, N.A.; Cline, G.W.; Zhang, D.; Cardone, R.L.; Petersen, K.F.; Kibbey, R.G.; Goodman, A.L.; Shulman, G.I. Acetate Mediates a Microbiome–Brain–β-Cell Axis to Promote Metabolic Syndrome. Nature 2016, 534, 213–217. [CrossRef]eng
dcterms.referencesYang, M.; Wang, J.; Wu, S.; Yuan, L.; Zhao, X.; Liu, C.; Xie, J.; Jia, Y.; Lai, Y.; Zhao, A.Z.; et al. Duodenal GLP-1 Signaling Regulates Hepatic Glucose Production through a PKC-δ-Dependent Neurocircuitry. Cell Death Dis. 2017, 8, e2609. [CrossRef]eng
dcterms.referencesVieira, A.T.; Fukumori, C.; Ferreira, C.M. New Insights into Therapeutic Strategies for Gut Microbiota Modulation in Inflammatory Diseases. Clin. Transl. Immunol. 2016, 5, e87. [CrossRef]eng
dcterms.referencesKellow, N.J.; Coughlan, M.T.; Savige, G.S.; Reid, C.M. Effect of Dietary Prebiotic Supplementation on Advanced Glycation, Insulin Resistance and Inflammatory Biomarkers in Adults with Pre-Diabetes: A Study Protocol for a Double-Blind Placebo-Controlled Randomised Crossover Clinical Trial. BMC Endocr. Disord. 2014, 14, 55. [CrossRef]eng
dcterms.referencesKassaian, N.; Aminorroaya, A.; Feizi, A.; Jafari, P.; Amini, M. The Effects of Probiotic and Synbiotic Supplementation on Metabolic Syndrome Indices in Adults at Risk of Type 2 Diabetes: Study Protocol for a Randomized Controlled Trial. Trials 2017, 18, 148. [CrossRef]eng
dcterms.referencesDávila, L.A.; Pirela, V.B.; Villasmil, N.R.; Cisternas, S.; Díaz, W.; Escobar, M.C.; Carrasco, P.; Durán, S.; Buhring, K.; Buhring, R.; et al. New Insights into Alleviating Diabetes Mellitus: Role of Gut Microbiota and a Nutrigenomic Approac. In Diabetes Food Plan; Waisundara, V., Ed.; InTech: London, UK, 2018.spa
dcterms.referencesBolívar González, S.; Talero Barrientos, E.; Motilva Sánchez, V. Efectos de Un Preparado Probiótico En Un Modelo de Colitis Experimental Crónica En Ratones, Inducida Por La Ingesta de Dextrano Sulfato Sódico (DSS). Cienc. Innov. Salud 2015, 3. [CrossRef]spa
dcterms.referencesYao, K.; Zeng, L.; He, Q.; Wang, W.; Lei, J.; Zou, X. Effect of Probiotics on Glucose and Lipid Metabolism in Type 2 Diabetes Mellitus: A Meta-Analysis of 12 Randomized Controlled Trials. Med. Sci. Monit. 2017, 23, 3044–3053. [CrossRef] [PubMed]eng
dcterms.referencesNaito, E.; Yoshida, Y.; Makino, K.; Kounoshi, Y.; Kunihiro, S.; Takahashi, R.; Matsuzaki, T.; Miyazaki, K.; Ishikawa, F. Beneficial Effect of Oral Administration of Lactobacillus Casei Strain Shirota on Insulin Resistance in Diet-Induced Obesity Mice. J. Appl. Microbiol. 2011, 110, 650–657. [CrossRef] [PubMed]eng
dcterms.referencesChen, J.J.; Wang, R.; Li, X.; Wang, R. Bifidobacterium Longum Supplementation Improved High-Fat-Fed-Induced Metabolic Syndrome and Promoted Intestinal Reg I Gene Expression. Exp. Biol. Med. 2011, 236, 823–831. [CrossRef] [PubMed]eng
dcterms.referencesAlfa, M.J.; Strang, D.; Tappia, P.S.; Olson, N.; De Gagne, P.; Bray, D.; Murray, B.-L.; Hiebert, B. A Randomized Placebo Controlled Clinical Trial to Determine the Impact of Digestion Resistant Starch MSPrebiotic® on Glucose, Insulin, and Insulin Resistance in Elderly and Mid-Age Adults. Front. Med. 2017, 4, 260. [CrossRef] [PubMed]eng
dcterms.referencesDe Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-Generated Metabolites Promote Metabolic Benefits via Gut-Brain Neural Circuits. Cell 2014, 156, 84–96. [CrossRef] [PubMed]eng
dcterms.referencesZheng, J.; Yuan, X.; Cheng, G.; Jiao, S.; Feng, C.; Zhao, X.; Yin, H.; Du, Y.; Liu, H. Chitosan Oligosaccharides Improve the Disturbance in Glucose Metabolism and Reverse the Dysbiosis of Gut Microbiota in Diabetic Mice. Carbohydr. Polym. 2018, 190, 77–86. [CrossRef] [PubMed]eng
dcterms.referencesChan, C.; Hyslop, C.M.; Shrivastava, V.; Ochoa, A.; Reimer, R.A.; Huang, C. Oligofructose as an Adjunct in Treatment of Diabetes in NOD Mice. Sci. Rep. 2016, 6, 37627. [CrossRef]eng
dcterms.referencesEverard, A.; Matamoros, S.; Geurts, L.; Delzenne, N.M.; Cani, P.D. Saccharomyces Boulardii Administration Changes Gut Microbiota and Reduces Hepatic Steatosis, Low-Grade Inflammation, and Fat Mass in Obese and Type 2 Diabetic Db/Db Mice. MBio 2014, 5, e01011. [CrossRef]eng
dcterms.referencesKarczewski, J.; Troost, F.J.; Konings, I.; Dekker, J.; Kleerebezem, M.; Brummer, R.-J.M.; Wells, J.M. Regulation of Human Epithelial Tight Junction Proteins by Lactobacillus Plantarum in Vivo and Protective Effects on the Epithelial Barrier. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G851–G859. [CrossRef]eng
dcterms.referencesSharma, P.; Bhardwaj, P.; Singh, R. Administration of Lactobacillus Casei and Bifidobacterium Bifidum Ameliorated Hyperglycemia, Dyslipidemia, and Oxidative Stress in Diabetic Rats. Int. J. Prev. Med. 2016, 7. [CrossRef]eng
dcterms.referencesValladares, R.; Sankar, D.; Li, N.; Williams, E.; Lai, K.-K.; Abdelgeliel, A.S.; Gonzalez, C.F.; Wasserfall, C.H.; Iii, J.L.; Schatz, D.; et al. Lactobacillus Johnsonii N6.2 Mitigates the Development of Type 1 Diabetes in BB-DP Rats. PLoS ONE 2010, 5, e10507. [CrossRef] [PubMed]eng
dcterms.referencesBalakumar, M.; Prabhu, D.; Sathishkumar, C.; Prabu, P.; Rokana, N.; Kumar, R.; Raghavan, S.; Soundarajan, A.; Grover, S.; Batish, V.K.; et al. Improvement in Glucose Tolerance and Insulin Sensitivity by Probiotic Strains of Indian Gut Origin in High-Fat Diet-Fed C57BL/6J Mice. Eur. J. Nutr. 2016, 57, 279–295. [CrossRef] [PubMed]eng
dcterms.referencesYadav, H.; Lee, J.-H.; Lloyd, J.; Walter, P.; Rane, S.G. Beneficial Metabolic Effects of a Probiotic via Butyrate-Induced GLP-1 Hormone Secretion. J. Biol. Chem. 2013, 288, 25088–25097. [CrossRef] [PubMed]eng
dcterms.referencesAsemi, Z.; Khorrami-Rad, A.; Alizadeh, S.-A.; Shakeri, H.; Esmaillzadeh, A. Effects of Synbiotic Food Consumption on Metabolic Status of Diabetic Patients: A Double-Blind Randomized Cross-over Controlled Clinical Trial. Clin. Nutr. 2014, 33, 198–203. [CrossRef] [PubMed]eng
dcterms.referencesEbrahimi, Z.S.; Nasli-Esfahani, E.; Nadjarzade, A.; Mozaffari-Khosravi, H. Effect of Symbiotic Supplementation on Glycemic Control, Lipid Profiles and Microalbuminuria in Patients with Non-Obese Type 2 Diabetes: A Randomized, Double-Blind, Clinical Trial. J. Diabetes Metab. Disord. 2017, 16, 23. [CrossRef] [PubMed]eng
dcterms.referencesShen, T.-C.D. Diet and Gut Microbiota in Health and Disease. In Intestinal Microbiome: Functional Aspects in Health and Disease; Nestlé Nutrition Institute Workshop Series; Karger Publishers: Basel, Switzerland, 2017; Volume 88, pp. 117–126. [CrossRef]eng
dcterms.referencesHeianza, Y.; Sun, D.; Li, X.; DiDonato, J.A.; Bray, G.A.; Sacks, F.M.; Qi, L. Gut Microbiota Metabolites, Amino Acid Metabolites and Improvements in Insulin Sensitivity and Glucose Metabolism: The POUNDS Lost Trial. Gut 2018, 67. [CrossRef]eng
dcterms.referencesMiranda, P.J.P.; Cueva, C. Rol de la metformina en el tratamiento de la diabetes mellitus gestacional: Situación actual. Arch. Venez. Farmacol. Ter. 2019, 38, 234–239.eng
dcterms.referencesNapolitano, A.; Miller, S.; Nicholls, A.W.; Baker, D.; Van Horn, S.; Thomas, E.; Rajpal, D.; Spivak, A.; Brown, J.R.; Nunez, D.J. Novel Gut-Based Pharmacology of Metformin in Patients with Type 2 Diabetes Mellitus. PLoS ONE 2014, 9, e100778. [CrossRef]eng
dcterms.referencesBonora, E.; Cigolini, M.; Bosello, O.; Zancanaro, C.; Capretti, L.; Zavaroni, I.; Coscelli, C.; Butturini, U. Lack of Effect of Intravenous Metformin on Plasma Concentrations of Glucose, Insulin, C-Peptide, Glucagon and Growth Hormone in Non-Diabetic Subjects. Curr. Med. Res. Opin. 1984, 9, 47–51. [CrossRef]spa
dcterms.referencesRidlon, J.M.; Harris, S.C.; Bhowmik, S.; Kang, D.-J.; Hylemon, P.B. Consequences of Bile Salt Biotransformations by Intestinal Bacteria. Gut Microbes 2016, 7, 22–39. [CrossRef]eng
dcterms.referencesForslund, K.; Hildebrand, F.; Nielsen, T.; Falony, G.; Le Chatelier, E.; Sunagawa, S.; Prifti, E.; Vieira-Silva, S.; Gudmundsdottir, V.; Krogh Pedersen, H.; et al. Disentangling Type 2 Diabetes and Metformin Treatment Signatures in the Human Gut Microbiota. Nature 2015, 528, 262–266. [CrossRef] [PubMed]eng
dcterms.referencesDe La Cuesta-Zuluaga, J.; Mueller, N.T.; Corrales-Agudelo, V.; Velásquez-Mejía, E.P.; Carmona, J.A.; Abad, J.M.; Escobar, J.S. Metformin Is Associated With Higher Relative Abundance of Mucin-Degrading Akkermansia Muciniphila and Several Short-Chain Fatty Acid–Producing Microbiota in the Gut. Diabetes Care 2016, dc161324. [CrossRef]eng
dcterms.referencesWu, H.; Esteve, E.; Tremaroli, V.; Khan, M.T.; Caesar, R.; Mannerås-Holm, L.; Ståhlman, M.; Olsson, L.M.; Serino, M.; Planas-Fèlix, M.; et al. Metformin Alters the Gut Microbiome of Individuals with Treatment-Naive Type 2 Diabetes, Contributing to the Therapeutic Effects of the Drug. Nat. Med. 2017, 23, 850–858. [CrossRef] [PubMed]eng
dcterms.referencesLee, H.; Ko, G. Effect of Metformin on Metabolic Improvement and Gut Microbiota. Appl. Environ. Microbiol. 2014, 80, 5935–5943. [CrossRef]eng
dcterms.referencesVallianou, N.; Stratigou, T.; Tsagarakis, S. Metformin and gut microbiota: their interactions and their impact on diabetes. Hormones (Athens) 2019, 2, 141–144. [CrossRef]eng
dcterms.referencesBryrup, T.; Thomsen, C.W.; Kern, T.; Allin, K.H.; Brandslund, I.; Jørgensen, N.R.; Vestergaard, H.; Hansen, T.; Hansen, T.H.; Pedersen, O.; et al. Metformin-Induced Changes of the Gut Microbiota in Healthy Young Men: Results of a Non-Blinded, One-Armed Intervention Study. Diabetologia 2019, 62, 1024–1035. [CrossRef]eng
dcterms.referencesSu, B.; Liu, H.; Li, J.; Sunli, Y.; Liu, B.; Liu, D.; Zhang, P.; Meng, X. Acarbose Treatment Affects the Serum Levels of Inflammatory Cytokines and the Gut Content of Bifidobacteria in Chinese Patients with Type 2 Diabetes Mellitus. J. Diabetes 2015, 7, 729–739. [CrossRef]eng
dcterms.referencesGu, Y.; Wang, X.; Li, J.; Zhang, Y.; Zhong, H.; Liu, R.; Zhang, D.; Feng, Q.; Xie, X.; Hong, J.; et al. Analyses of Gut Microbiota and Plasma Bile Acids Enable Stratification of Patients for Antidiabetic Treatment. Nat. Commun. 2017, 8, 1785. [CrossRef]eng
dcterms.referencesSmith, B.J.; Miller, R.A.; Ericsson, A.C.; Harrison, D.C.; Strong, R.; Schmidt, T.M. Changes in the Gut Microbiome and Fermentation Products Concurrent with Enhanced Longevity in Acarbose-Treated Mice. BMC Microbiol. 2019, 19, 130. [CrossRef]eng
dcterms.referencesBaxter, N.T.; Lesniak, N.A.; Sinani, H.; Schloss, P.D.; Koropatkin, N.M. The Glucoamylase Inhibitor Acarbose Has a Diet-Dependent and Reversible Effect on the Murine Gut Microbiome. MSphere 2019, 4. [CrossRef]eng
dcterms.referencesZhao, L.; Chen, Y.; Xia, F.; Abudukerimu, B.; Zhang, W.; Guo, Y.; Wang, N.; Lu, Y. A Glucagon-Like Peptide-1 Receptor Agonist Lowers Weight by Modulating the Structure of Gut Microbiota. Front. Endocrinol. 2018, 9, 233. [CrossRef] [PubMed]eng
dcterms.referencesWang, L.; Li, P.; Tang, Z.; Yan, X.; Feng, B. Structural Modulation of the Gut Microbiota and the Relationship with Body Weight: Compared Evaluation of Liraglutide and Saxagliptin Treatment. Sci. Rep. 2016, 6, 33251. [CrossRef] [PubMed]eng
dcterms.referencesWang, Z.; Saha, S.; Van Horn, S.; Thomas, E.; Traini, C.; Sathe, G.; Rajpal, D.K.; Brown, J.R. Gut Microbiome Differences between Metformin- and Liraglutide-Treated T2DM Subjects. Endocrinol. Diabetes Metab. 2018, 1, e00009. [CrossRef] [PubMed]eng
dcterms.referencesMoreira, G.V.; Azevedo, F.F.; Ribeiro, L.M.; Santos, A.; Guadagnini, D.; Gama, P.; Liberti, E.A.; Saad, M.; Carvalho, C. Liraglutide Modulates Gut Microbiota and Reduces NAFLD in Obese Mice. J. Nutr. Biochem. 2018, 62, 143–154. [CrossRef]spa
dcterms.referencesSun, L.; Xie, C.; Wang, G.; Wu, Y.; Wu, Q.; Wang, X.; Liu, J.; Deng, Y.; Xia, J.; Chen, B.; et al. Gut Microbiota and Intestinal FXR Mediate the Clinical Benefits of Metformin. Nat. Med. 2018, 24, 1919–1929. [CrossRef]eng
dcterms.referencesFang, S.; Suh, J.M.; Reilly, S.M.; Yu, E.; Osborn, O.; Lackey, D.; Yoshihara, E.; Perino, A.; Jacinto, S.; Lukasheva, Y.; et al. Intestinal FXR Agonism Promotes Adipose Tissue Browning and Reduces Obesity and Insulin Resistance. Nat. Med. 2015, 21, 159–165. [CrossRef]eng
dcterms.referencesPathak, P.; Xie, C.; Nichols, R.G.; Ferrell, J.M.; Boehme, S.; Krausz, K.W.; Patterson, A.D.; Gonzalez, F.J.; Chiang, J.Y.L. Intestine Farnesoid X Receptor Agonist and the Gut Microbiota Activate G-Protein Bile Acid Receptor-1 Signaling to Improve Metabolism. Hepatology 2018, 68, 1574–1588. [CrossRef]eng
dcterms.referencesZhang, X.; Fang, Z.; Zhang, C.; Xia, H.; Jie, Z.; Han, X.; Chen, Y.; Ji, L. Effects of Acarbose on the Gut Microbiota of Prediabetic Patients: A Randomized, Double-Blind, Controlled Crossover Trial. Diabetes Ther. 2017, 8, 293–307. [CrossRef]eng
dcterms.referencesRemely, M.; Hippe, B.; Zanner, J.; Aumueller, E.; Brath, H.; Haslberger, A.G. Gut Microbiota of Obese, Type 2 Diabetic Individuals Is Enriched in Faecalibacterium Prausnitzii, Akkermansia Muciniphila and Peptostreptococcus Anaerobius after Weight Loss. Endocr. Metab. Immune Disord. Drug Targets 2016, 16, 99–106. [CrossRef]eng
dcterms.referencesXu, G.-D.; Cai, L.; Ni, Y.-S.; Tian, S.-Y.; Lu, Y.-Q.; Wang, L.-N.; Chen, L.-L.; Ma, W.-Y.; Deng, S.-P. Comparisons of Effects on Intestinal Short-Chain Fatty Acid Concentration after Exposure of Two Glycosidase Inhibitors in Mice. Biol. Pharm. Bull. 2018, 41, 1024–1033. [CrossRef]eng
dcterms.referencesZhang, Q.; Xiao, X.; Zheng, J.; Li, M.; Yu, M.; Ping, F.; Wang, T.; Wang, X. Featured Article: Structure Moderation of Gut Microbiota in Liraglutide-Treated Diabetic Male Rats. Exp. Biol. Med. 2018, 243, 34–44. [CrossRef] [PubMed]eng
oaire.versioninfo:eu-repo/semantics/publishedVersioneng

Archivos

Bloque original
Mostrando 1 - 1 de 1
Cargando...
Miniatura
Nombre:
Microbiota_and_Diabetes_Mellitus.pdf
Tamaño:
2.08 MB
Formato:
Adobe Portable Document Format
Descripción:
PDF
Bloque de licencias
Mostrando 1 - 1 de 1
No hay miniatura disponible
Nombre:
license.txt
Tamaño:
381 B
Formato:
Item-specific license agreed upon to submission
Descripción:

Colecciones