Letter to the Editor Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Diabetes. Jan 15, 2022; 13(1): 65-69
Published online Jan 15, 2022. doi: 10.4239/wjd.v13.i1.65
Gut microbiota-derived metabolites are novel targets for improving insulin resistance
Rosana MC Bastos, Érika B Rangel, Hospital Israelita Albert Einstein, São Paulo 05652-001, SP, Brazil
Érika B Rangel, Nephrology Division, Federal University of São Paulo, São Paulo 04023-900, SP, Brazil
ORCID number: Rosana MC Bastos (0000-0002-4348-1487); Érika B Rangel (0000-0003-0982-2484).
Author contributions: Bastos RMC and Rangel ÉB wrote the letter; Rangel ÉB revised the letter and gave the final approval.
Supported by São Paulo Research Foundation, No. 2013/19560-6 and No. 2017/23195-2; and EFSD (European Foundation for the Study of Diabetes)/Sanofi (to Rangel ÉB).
Conflict-of-interest statement: The authors declare no conflicts of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Érika B Rangel, MD, PhD, Assistant Professor, Senior Scientist, Hospital Israelita Albert Einstein, Av Albert Einstein 627, Building A, 2SS, São Paulo 05652-001, SP, Brazil. erikabr@uol.com.br
Received: July 3, 2021
Peer-review started: July 3, 2021
First decision: July 28, 2021
Revised: August 1, 2021
Accepted: December 31, 2021
Article in press: December 31, 2021
Published online: January 15, 2022

Abstract

The gut microbiota plays a key role in metabolic diseases. Gut-microbiota-derived metabolites are found in different dietary sources, including: Carbohydrate (acetate, propionate, butyrate, also known as short-chain fatty acids, as well as succinate); protein (hydrogen sulfide, indole, and phenylacetic acid); and lipids (resveratrol-, ferulic acid-, linoleic acid-, catechin- and berry-derived metabolites). Insulin resistance, which is a global pandemic metabolic disease that progresses to type 2 diabetes mellitus, can be directly targeted by these metabolites. Gut-microbiota-derived metabolites have broad effects locally and in distinct organs, in particular skeletal muscle, adipose tissue, and liver. These metabolites can modulate glucose metabolism, including the increase in glucose uptake and lipid oxidation in skeletal muscle, and decrease in lipogenesis and gluconeogenesis associated with lipid oxidation in the liver through activation of phosphatidylinositol 3-kinase - serine/threonine-protein kinase B and AMP-activated protein kinase. In adipose tissue, gut-microbiota-derived metabolites stimulate adipogenesis and thermogenesis, inhibit lipolysis, and attenuate inflammation. Importantly, an increase in energy expenditure and fat oxidation occurs in the whole body. Therefore, the therapeutic potential of current pharmacological and non-pharmacological approaches used to treat diabetes mellitus can be tested to target specific metabolites derived from intestinal bacteria, which may ultimately ameliorate the hyperglycemic burden.

Key Words: Insulin resistance, Gut microbiota, Metabolites, Host metabolism, Metabolic organs, Novel targets

Core Tip: The gut-microbiota-derived metabolites play a key role in metabolic diseases. Insulin signaling pathways are directly targeted by these metabolites, as they promote an increase in glucose uptake and lipid oxidation in skeletal muscle; a decrease in lipogenesis and gluconeogenesis associated with an increase in lipid oxidation in the liver; and an improvement in thermogenesis and inflammation in the adipose tissue. Collectively, these findings pave the way for the development of novel drugs or for investigation of the therapeutic potential of drugs currently used to treat insulin resistance, targeting the gut-microbiota-derived metabolites.



TO THE EDITOR

We read with interest the recent publication by Jang and Lee[1] on the relationship of mechanisms linking the gut microbiota-derived metabolites to insulin resistance published in this journal.

The gut microbiota plays a key role in metabolic diseases. Gut-microbiota-derived metabolites are found in different dietary sources, including: Carbohydrate (acetate, propionate, butyrate, and succinate); protein (hydrogen sulfide, indole, and phenylacetic acid); and lipids (resveratrol-, ferulic acid-, linoleic acid-, cathecin- and berry-derived metabolites). Insulin signaling pathways are directly targeted by these metabolites. Therefore, gut-microbiota-derived metabolites, in particular, the short-chain fatty acids (SCFAs), increase glucose uptake and lipid oxidation in skeletal muscle, whereas in the liver, SCFAs decrease lipogenesis and gluconeogenesis, increasing the lipid oxidation through activation of phosphatidylinositol 3-kinase - serine/threonine-protein kinase B (PI3K-AKT-PKB) and AMP-activated protein kinase. In adipose tissue, SCFAs stimulate adipogenesis and thermogenesis, inhibit lipolysis, and attenuate inflammation. Therefore, an increase in energy expenditure and fat oxidation occurs in the whole body. Collectively, these findings pave the way for the development of novel drugs or for investigation of the therapeutic potential of drugs currently used to treat insulin resistance, targeting the gut-microbiota-derived metabolites.

Notably, preclinical models and clinical studies substantiate the interaction between intestinal microbiota and the pathophysiology of insulin resistance in type 2 diabetes mellitus (DM)[2-4].

Therefore, this current article provides an overview of the important role of the specific microbiota-derived compounds in insulin-responsive tissues, acting as risk factors or protectors for the development of insulin resistance, and highlights the biologic implications of the muscle–liver–adipose tissue axis interaction.

Even though the authors documented the potential role of some bacterial metabolites as regulators of metabolic functions in the body, such as SCFAs derived from carbohydrates (propionate, butyrate and acetate), and the protein- and lipid-derived metabolites, in modulating pathways of insulin signaling, the impact of these bacterial metabolites on host metabolism warrants further investigation.

Importantly, succinate is a metabolite of the tricarboxylic acid cycle and is produced equally by microbiota and the host[5]. Although this metabolite contributes to improving glucose homeostasis through the activation of intestinal gluconeogenesis[6], in obese individuals, high levels of this circulating metabolite are documented[5]. Furthermore, the imbalance of higher relative abundance of succinate-producing bacteria (Prevotellaceae and Veillonellaceae) and lower relative abundance of succinate-consuming bacteria Odoribacteraceae and Clostridaceae) may promote an increase in succinate levels and, ultimately, impaired glucose metabolism. These authors also pointed out succinate as having a potential role in metabolic-associated cardiovascular disorders and obesity. Additionally, succinate acts as an immunogenic molecule, identified as damage-associated molecular patterns. This molecule is recognized by immune cells and stabilizes hypoxia-inducible factor-1α through its G-protein coupled receptor (succinate receptor 1/SUCNR1 or GPR19), which leads to the proinflammatory differentiation of T lymphocytes, and production of cytokines through interaction with Toll-like receptor ligands in dendritic cells[7,8]. Collectively, these findings may promote an enhancement of insulin resistance and DM burden.

Furthermore, hydrogen sulfide (H2S) and the role of sulfur-reducing bacteria from the intestinal microbiota have gained insights into the physiological implications of host glycemic control[9]. Thus, H2S metabolite may protect against oxidative stress by restoring reduced glutathione (GSH) and scavenging of mitochondrial reactive oxygen species, inducing pro-survival/angiogenesis signaling pathway (STAT3, signal transducer and activator of transcription 3), and promoting immunomodulation (inhibition/activation of nuclear factor-κB) and vasodilation (activation of KATP ion channel)[10]. However, the balance between therapeutic and harmful effects of H2S should be considered when targeting that metabolite, as H2S either endogenous or exogenous, as well as that produced by the gut microbiota, promotes or inhibits a variety of characteristics in mucosal microbiota biofilms[11]. Depending on H2S concentration, in particular, when the gut microbiota produces an excessive amount, it may cause mucus disruption and inflammation in the colon and contribute to cancer. Conversely, low levels of H2S directly stabilize mucus layers, prevent fragmentation and adherence of the microbiota biofilm to the epithelium, inhibit the release of invasive opportunistic pathogens or pathobionts, and prevent inflammation and tissue injury[11]. Moreover, H2S overproduction is a causative factor in the pathogenesis of β-cell death in DM due to increased levels of reactive oxygen and nitrogen species, whereas its deficiency, as a result of increased H2S consumption by hyperglycemic cells, may lead to endothelial dysfunction, and kidney and heart diseases[12].

As we learn more about gut-microbiota-derived metabolites, we will better understand how to target these metabolites. Thus, acetate, which is involved in host energy, substrate metabolism, and appetite via secretion of the gut hormones [glucagon-like peptide (GLP) and peptide YY], may be increased by oral acetate administration (vinegar intake), colonic acetate infusions, acetogenic fibers and acetogenic probiotic administration[13]. These strategies may both decrease whole-body lipolysis and systemic proinflammatory cytokine levels, and increase energy expenditure, insulin sensitivity, and fat oxidation, which contributes to weight control and glucose homeostasis. Probiotics (live microorganisms) act as microbiome modulators and confer a health benefit, as demonstrated by the capacity of selected probiotic strains (lactobacilli and enterococci) to increase SCFA production; in particular, propionate and butyrate[14]. As reviewed elsewhere, probiotic administration (Bifidobacterium pseudocatenulatum, Lactobacillus plantarum, or the formula VSL#3) in preclinical models of obesity led to an increase in the intestinal barrier function, a reduction in the endotoxemia, acceleration in metabolism, and suppression of body weight gain and insulin resistance via modulation of the gut microbiota composition and SCFA production[15]. Probiotics may also ameliorate glucose homeostasis and lipid profile in diabetic mice[15].

From a clinical point of view, obese children treated with the probiotic Lactobacillus casei shirota for 6 mo presented with loss of weight, improved lipid metabolism, and an increase in the number of Bifidobacterium spp. and acetate concentration in the feces[16]. Likewise, patients with type 2 DM treated with probiotics containing Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for 6 wk had improved glucose and lipid profiles, which were associated with lower levels of systemic inflammation and increased concentration of acetate[17]. Additionally, modification of gut microbiota by dietary weight loss intervention decreased circulating succinate levels and improved the metabolic profile in a cohort of individuals with type 2 DM and obesity[6].

Pharmacological interventions or xenobiotics may also have effects on gut microbiota. Metformin is the most frequently administered medication to treat patients with insulin resistance and type 2 DM. This drug may alter the gut microbiota composition through an increase in the Bacteroidetes and Verrucomicrobia phyla and the mucin-degrading Akkermansia muciniphila, Bacteroides, and Escherichia genera, as well as in butyrate and propionate production, emphasizing maintenance of the integrity of the intestinal barrier, regulation of bile acid metabolism and improvement in glucose homeostasis[18,19]. Importantly, metformin may have these benefits in newly diagnosed DM[20].

Sodium-glucose cotransporter 2 inhibitors represent the most recently approved class of oral medications for the treatment of type 2 DM. Dapagliflozin decreased the Firmicutes-to-Bacteriodetes ratio in diabetic mice, which was correlated with improvement in vascular function[21]. In a rodent model of type 1 DM, inhibition of SGLT2 reduced the intermediate metabolite succinate and increased butyrate levels, as well as decreased norepinephrine content in the kidney[22]. Hence, the impact of SGLT2 inhibitors on the gut microbiota is an area of active research.

Likewise, GLP-1 agonists reduced the abundance of the species of the Firmicutes phylum (Lachnospiraceae and Clostridiales) and increased the abundance of the species representing the Proteobacteria (Burkholderiales bacterium YL45) and Verrucomicrobia (Akkermansia muciniphila), as well as Firmicutes (Clostridiales and Oscillospiraceae) phyla in obese mice[23]. In particular, body weight loss was associated with increased abundance of Akkermansia muciniphila, a mucin-degrading SCFA-producing species, whose abundance is decreased in obesity and has a negative correlation with markers of gut permeability and inflammation. Notably, the GLP-1 agonist liraglutide can prevent weight gain by modulating gut microbiota composition in both obese and diabetic obese animals[24].

In the cardiometabolic disease setting, lipid-lowering drugs, such as statins, may also play an important role in modulating gut microbiota. In vitro studies have documented increased levels of SCFA production, including propionate, butyrate and acetate[25]. These drugs may increase the abundance of the Bacteroides, Butyricimonas and Mucispirillum genera, which is associated with a decrease in the inflammatory response, including lower levels of interleukin (IL)-1β and IL-6, and higher levels of transforming growth factor β-1 in the ileum, and improved hyperglycemia[26]. In humans, obesity is associated with a microbiota signature based on the abundance of the Bacteroides genus profile, displaying the lowest abundances of Akkermansia and Faecalibacterium, as well as a decrease in the butyrate production potential[27]. Importantly, statin therapy resulted in a lower prevalence of a proinflammatory microbial community type in obese individuals.

In conclusion, the gut microbiota imbalances and maladaptive responses have been implicated in the pathology of insulin resistance, DM, and obesity[28]. Host-gut microbiota interaction is suggested to play a contributory role in the therapeutic effects of antidiabetics, statins, and weight-loss-promoting drugs. Therefore, additional studies combining untargeted metabolomics and proteomics are essential to identify further microbial metabolites or proteins and to determine how they interact with the host targets in improving host metabolism.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Endocrinology and metabolism

Country/Territory of origin: Brazil

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): 0

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Velikova TV, Yang M S-Editor: Gao CC L-Editor: Kerr C P-Editor: Gao CC

References
1.  Jang HR, Lee HY. Mechanisms linking gut microbial metabolites to insulin resistance. World J Diabetes. 2021;12:730-744.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 7]  [Cited by in F6Publishing: 13]  [Article Influence: 4.3]  [Reference Citation Analysis (2)]
2.  Wang H, Lu Y, Yan Y, Tian S, Zheng D, Leng D, Wang C, Jiao J, Wang Z, Bai Y. Promising Treatment for Type 2 Diabetes: Fecal Microbiota Transplantation Reverses Insulin Resistance and Impaired Islets. Front Cell Infect Microbiol. 2019;9:455.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 95]  [Article Influence: 23.8]  [Reference Citation Analysis (0)]
3.  Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W, Guan Y, Shen D, Peng Y, Zhang D, Jie Z, Wu W, Qin Y, Xue W, Li J, Han L, Lu D, Wu P, Dai Y, Sun X, Li Z, Tang A, Zhong S, Li X, Chen W, Xu R, Wang M, Feng Q, Gong M, Yu J, Zhang Y, Zhang M, Hansen T, Sanchez G, Raes J, Falony G, Okuda S, Almeida M, LeChatelier E, Renault P, Pons N, Batto JM, Zhang Z, Chen H, Yang R, Zheng W, Yang H, Wang J, Ehrlich SD, Nielsen R, Pedersen O, Kristiansen K. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55-60.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3971]  [Cited by in F6Publishing: 4291]  [Article Influence: 357.6]  [Reference Citation Analysis (0)]
4.  Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ, Fagerberg B, Nielsen J, Bäckhed F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498:99-103.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1766]  [Cited by in F6Publishing: 1823]  [Article Influence: 165.7]  [Reference Citation Analysis (0)]
5.  Serena C, Ceperuelo-Mallafré V, Keiran N, Queipo-Ortuño MI, Bernal R, Gomez-Huelgas R, Urpi-Sarda M, Sabater M, Pérez-Brocal V, Andrés-Lacueva C, Moya A, Tinahones FJ, Fernández-Real JM, Vendrell J, Fernández-Veledo S. Elevated circulating levels of succinate in human obesity are linked to specific gut microbiota. ISME J. 2018;12:1642-1657.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 207]  [Cited by in F6Publishing: 212]  [Article Influence: 35.3]  [Reference Citation Analysis (0)]
6.  De Vadder F, Kovatcheva-Datchary P, Zitoun C, Duchampt A, Bäckhed F, Mithieux G. Microbiota-Produced Succinate Improves Glucose Homeostasis via Intestinal Gluconeogenesis. Cell Metab. 2016;24:151-157.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 385]  [Cited by in F6Publishing: 408]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
7.  Garcia-Martinez I, Shaker ME, Mehal WZ. Therapeutic Opportunities in Damage-Associated Molecular Pattern-Driven Metabolic Diseases. Antioxid Redox Signal. 2015;23:1305-1315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
8.  Rodríguez-Nuevo A, Zorzano A. The sensing of mitochondrial DAMPs by non-immune cells. Cell Stress. 2019;3:195-207.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 62]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
9.  Pichette J, Fynn-Sackey N, Gagnon J. Hydrogen Sulfide and Sulfate Prebiotic Stimulates the Secretion of GLP-1 and Improves Glycemia in Male Mice. Endocrinology. 2017;158:3416-3425.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 60]  [Article Influence: 8.6]  [Reference Citation Analysis (0)]
10.  Pal VK, Bandyopadhyay P, Singh A. Hydrogen sulfide in physiology and pathogenesis of bacteria and viruses. IUBMB Life. 2018;70:393-410.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 55]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
11.  Buret AG, Allain T, Motta JP, Wallace JL. Effects of Hydrogen Sulfide on the Microbiome: From Toxicity to Therapy. Antioxid Redox Signal. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 44]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
12.  Szabo C. Roles of hydrogen sulfide in the pathogenesis of diabetes mellitus and its complications. Antioxid Redox Signal. 2012;17:68-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 152]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
13.  Hernández MAG, Canfora EE, Jocken JWE, Blaak EE. The Short-Chain Fatty Acid Acetate in Body Weight Control and Insulin Sensitivity. Nutrients. 2019;11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 162]  [Cited by in F6Publishing: 266]  [Article Influence: 53.2]  [Reference Citation Analysis (0)]
14.  Nagpal R, Wang S, Ahmadi S, Hayes J, Gagliano J, Subashchandrabose S, Kitzman DW, Becton T, Read R, Yadav H. Human-origin probiotic cocktail increases short-chain fatty acid production via modulation of mice and human gut microbiome. Sci Rep. 2018;8:12649.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 140]  [Cited by in F6Publishing: 166]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
15.  Markowiak-Kopeć P, Śliżewska K. The Effect of Probiotics on the Production of Short-Chain Fatty Acids by Human Intestinal Microbiome. Nutrients. 2020;12.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 194]  [Cited by in F6Publishing: 381]  [Article Influence: 95.3]  [Reference Citation Analysis (0)]
16.  Nagata S, Chiba Y, Wang C, Yamashiro Y. The effects of the Lactobacillus casei strain on obesity in children: a pilot study. Benef Microbes. 2017;8:535-543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 38]  [Article Influence: 5.4]  [Reference Citation Analysis (0)]
17.  Tonucci LB, Olbrich Dos Santos KM, Licursi de Oliveira L, Rocha Ribeiro SM, Duarte Martino HS. Clinical application of probiotics in type 2 diabetes mellitus: A randomized, double-blind, placebo-controlled study. Clin Nutr. 2017;36:85-92.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 174]  [Cited by in F6Publishing: 189]  [Article Influence: 21.0]  [Reference Citation Analysis (0)]
18.  Vallianou NG, Stratigou T, Tsagarakis S. Metformin and gut microbiota: their interactions and their impact on diabetes. Hormones (Athens). 2019;18:141-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 72]  [Article Influence: 14.4]  [Reference Citation Analysis (0)]
19.  Zhang Q, Hu N. Effects of Metformin on the Gut Microbiota in Obesity and Type 2 Diabetes Mellitus. Diabetes Metab Syndr Obes. 2020;13:5003-5014.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 88]  [Article Influence: 22.0]  [Reference Citation Analysis (0)]
20.  Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Mannerås-Holm L, Ståhlman M, Olsson LM, Serino M, Planas-Fèlix M, Xifra G, Mercader JM, Torrents D, Burcelin R, Ricart W, Perkins R, Fernàndez-Real JM, Bäckhed F. 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.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 824]  [Cited by in F6Publishing: 846]  [Article Influence: 120.9]  [Reference Citation Analysis (0)]
21.  Lee DM, Battson ML, Jarrell DK, Hou S, Ecton KE, Weir TL, Gentile CL. SGLT2 inhibition via dapagliflozin improves generalized vascular dysfunction and alters the gut microbiota in type 2 diabetic mice. Cardiovasc Diabetol. 2018;17:62.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 150]  [Article Influence: 25.0]  [Reference Citation Analysis (0)]
22.  Herat LY, Ward NC, Magno AL, Rakoczy EP, Kiuchi MG, Schlaich MP, Matthews VB. Sodium glucose co-transporter 2 inhibition reduces succinate levels in diabetic mice. World J Gastroenterol. 2020;26:3225-3235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
23.  Madsen MSA, Holm JB, Pallejà A, Wismann P, Fabricius K, Rigbolt K, Mikkelsen M, Sommer M, Jelsing J, Nielsen HB, Vrang N, Hansen HH. Metabolic and gut microbiome changes following GLP-1 or dual GLP-1/GLP-2 receptor agonist treatment in diet-induced obese mice. Sci Rep. 2019;9:15582.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 46]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
24.  Zhao 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 (Lausanne). 2018;9:233.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 70]  [Article Influence: 11.7]  [Reference Citation Analysis (0)]
25.  Zhao C, Hu Y, Chen H, Li B, Cao L, Xia J, Yin Y. An in vitro evaluation of the effects of different statins on the structure and function of human gut bacterial community. PLoS One. 2020;15:e0230200.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
26.  Kim J, Lee H, An J, Song Y, Lee CK, Kim K, Kong H. Alterations in Gut Microbiota by Statin Therapy and Possible Intermediate Effects on Hyperglycemia and Hyperlipidemia. Front Microbiol. 2019;10:1947.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 96]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
27.  Vieira-Silva S, Falony G, Belda E, Nielsen T, Aron-Wisnewsky J, Chakaroun R, Forslund SK, Assmann K, Valles-Colomer M, Nguyen TTD, Proost S, Prifti E, Tremaroli V, Pons N, Le Chatelier E, Andreelli F, Bastard JP, Coelho LP, Galleron N, Hansen TH, Hulot JS, Lewinter C, Pedersen HK, Quinquis B, Rouault C, Roume H, Salem JE, Søndertoft NB, Touch S; MetaCardis Consortium, Dumas ME, Ehrlich SD, Galan P, Gøtze JP, Hansen T, Holst JJ, Køber L, Letunic I, Nielsen J, Oppert JM, Stumvoll M, Vestergaard H, Zucker JD, Bork P, Pedersen O, Bäckhed F, Clément K, Raes J. Statin therapy is associated with lower prevalence of gut microbiota dysbiosis. Nature. 2020;581:310-315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 147]  [Cited by in F6Publishing: 229]  [Article Influence: 57.3]  [Reference Citation Analysis (0)]
28.  Fan Y, Pedersen O. Gut microbiota in human metabolic health and disease. Nat Rev Microbiol. 2021;19:55-71.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 729]  [Cited by in F6Publishing: 1602]  [Article Influence: 400.5]  [Reference Citation Analysis (0)]