Skip Navigation
Skip to contents

Diabetes Metab J : Diabetes & Metabolism Journal

Search
OPEN ACCESS

Articles

Page Path
HOME > Diabetes Metab J > Volume 39(4); 2015 > Article
Review
Obesity and Metabolic Syndrome Probiotics as Complementary Treatment for Metabolic Disorders
Mélanie Le Barz1,2,3, Fernando F. Anhê1,2, Thibaut V. Varin2, Yves Desjardins2, Emile Levy2,4,5, Denis Roy2, Maria C. Urdaci3, André Marette1,2
Diabetes & Metabolism Journal 2015;39(4):291-303.
DOI: https://doi.org/10.4093/dmj.2015.39.4.291
Published online: August 17, 2015
  • 7,570 Views
  • 134 Download
  • 80 Web of Science
  • 78 Crossref
  • 97 Scopus

1Department of Medicine, Faculty of Medicine, Cardiology Axis of the Québec Heart and Lung Institute, Québec, QC, Canada.

2Institute of Nutrition and Functional Foods, Laval University, Québec, QC, Canada.

3University of Bordeaux, UMR 5248, CBMN, Bordeaux, France.

4Research Centre, Sainte-Justine Hospital, Montreal, QC, Canada.

5Department of Nutrition, University of Montreal Faculty of Medicine, Montreal, QC, Canada.

Corresponding author: André Marette. Cardiology Axis of the Quebec Heart and Lung Institute, Laval University, Hôpital Laval, Pavillon Marguerite d'Youville, Bureau Y4340, Québec City, QC G1V 4G5, Canada. andre.marette@criucpq.ulaval.ca

Copyright © 2015 Korean Diabetes Association

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

  • Over the past decade, growing evidence has established the gut microbiota as one of the most important determinants of metabolic disorders such as obesity and type 2 diabetes. Indeed, obesogenic diet can drastically alter bacterial populations (i.e., dysbiosis) leading to activation of pro-inflammatory mechanisms and metabolic endotoxemia, therefore promoting insulin resistance and cardiometabolic disorders. To counteract these deleterious effects, probiotic strains have been developed with the aim of reshaping the microbiome to improve gut health. In this review, we focus on benefits of widely used probiotics describing their potential mechanisms of action, especially their ability to decrease metabolic endotoxemia by restoring the disrupted intestinal mucosal barrier. We also discuss the perspective of using new bacterial strains such as butyrate-producing bacteria and the mucolytic Akkermansia muciniphila, as well as the use of prebiotics to enhance the functionality of probiotics. Finally, this review introduces the notion of genetically engineered bacterial strains specifically developed to deliver anti-inflammatory molecules to the gut.
Trillions of microorganisms among bacteria, archaea, viruses, and fungi inhabit our gastrointestinal tract (GIT). Bacterial cells largely outnumber our own cells, and the gut microbiota is a prolific example of a symbiotic relationship as it plays a crucial role in host's physiology and health. Studies based on gnotobiotic models and fecal microbial transplants (FMT) have provided unequivocal evidence that perturbations in bacterial communities play a key role in the pathophysiology of obesity and insulin resistance [12]. The gut microbiota is the product of a complex interaction between host's genetics and environment, and diet is one of the main driving forces shaping intestinal bacterial communities [3]. The so-called western obesogenic diet (i.e., rich in saturated/trans fat and simple sugars and poor in fibers) is associated with specific modulation of taxonomic profiles that are functionally linked with a more proinflammatory milieu and disrupted intestinal barrier. Disturbance of intestinal homeostasis then leads to excessive bacterial fragments/products internal diffusion, which promotes inflammation in key insulin-responsive tissues, resulting in insulin resistance [4].
The current knowledge suggests that gut bacterial profiles may represent new disease predictors and that manipulation of the gut microbiota could be a promising approach for the prevention and management of metabolic diseases [5]. Indeed, Cani et al. [6] were the first group to demonstrate a positive correlation between alteration of gut microbiota population, the increase of intestinal permeability and the development of metabolic endotoxemia that is characterized by the translocation of bacterial lipopolysaccharides (LPSs) into the systemic circulation and induction of inflammatory pathways in mice fed obesogenic diet.
Since Metchnikoff's era, the field of probiotics-live microorganisms that, when administered in adequate amounts, confers a health benefit on the host (Food and Agriculture Organization of the United Nations, 2002; updated by Hill et al. [7])-continues to grow thanks to the recent access to investigate the role of an increasing number of potential probiotic strains in host's physiology. According to this definition, the safety and efficacy of a given strain must be scientifically demonstrated in order to be considered as a probiotic. Here, we propose a critical review of the most recent studies concerning the effects of probiotic bacterial strains in the prevention or treatment of metabolic disorders such as obesity, insulin resistance, diabetes mellitus and its comorbidities.
Most currently used probiotics belong to bifidobacteria, lactic acid bacteria (LAB), dairy propionibacteria, yeasts (Saccharomyces boulardii), Bacillus, and the gram-negative Escherichia coli strain Nissle 1917 [8]. LAB represent a heterogeneous group of microorganisms broadly present in the diet, particularly by the use of non-human strains in the fermentation of dairy products being also normal inhabitants of the gastrointestinal and urogenital tract [9]. Most of them are members of the phylum Firmicutes, while Bifidobacterium, also considered as lactic-producing bacteria, belong to Actinobacteria phylum.
Probiotic administration has been shown to stimulate the immune response, improve lactose tolerance, help prevent diarrhea, have an anti-inflammatory effect and even restore obesity-linked gut dysbiosis [10]. Given the relationship between obesity-related disorders and gut homeostasis, probiotics may be of interest to supplement the limited arsenal of therapies against the metabolic syndrome. The diversity of reported studies in Tables 1 and 2, shows that the positive effects of probiotics are strain-specific and the idea of a "universal strain," that would provide at once all the benefits associated with probiotics, is unrealistic, even for strains of the same species [7]. In the context of obesity and metabolic disorders, probiotic supplementation may help to reduce hyperphagia [11], improving control of weight gain, fat mass loss and glucose tolerance. On the contrary, such positive effects could also be obtained without modulation of caloric intake, as demonstrated by most of the reported studies [12131415161718]. To demonstrate the beneficial effect of probiotics in improvement of metabolic disorders, researchers have access to a variety of assays such as plasma and liver cholesterol, free-fatty acids, alanine and aspartate transaminases (hepatotoxicity markers), gene and protein expressions (involved in inflammatory and metabolic pathways), etc. (Tables 1 and 2 for details).
For example, dairy products supplemented with Propionibacterium, a well-known promising non-LAB genus, may exert a probiotic effect in the colon by producing metabolites such as short-chain fatty acids (SCFA), vitamins (B8, B9, and B12), and 1,4-dihydroxy-2-naphtoic acid, bifidogenic and anti-inflammatory product (DHNA) [19]. Oksaharju et al. [20] demonstrated that Propionibacterium freudenreichii ssp. shermani JS has anti-inflammatory effects on high fat diet-induced inflammation in ApoE*3Leiden mice, with a decrease of intestinal mast cell numbers and a demonstrated intestinal but also systemic anti-inflammatory potential.
Finally, although it needs further investigation, multiple strain probiotics could confer a more effective strategy than single-strain probiotics against diet-induced obesity (DIO) [12]. Interestingly, VSL#3, a mixture of eight different strains of bacteria, has shown efficacy in prevention but also in the treatment of obesity and type 2 diabetes [11].
Probiotic administration is frequently associated with important shifts in gut bacterial composition, along with beneficial effects on metabolism and inflammatory tone [1617182122]. Indeed, within the gut, probiotic are in competition for nutrients, metabolites and also for antimicrobial proteins, altering gut microbiota population diversity in several ways [12]. However, it remains unclear if the modulation of gut microbiota is the cause or the consequence of probiotic treatment, or whether the mechanisms are partially or totally interdependent. The probiotic components associated with positive effects are a variety of cell constituents as polysaccharides, peptidoglycan, DNA, teichoic acids and certain cell-surface bound and secreted proteins as well as organic acids, bacteriocins, polyphosphate, and fatty acids (FA), which can modulate host responses, inhibit pathogens or interact with the intestinal microbiota [23]. Furthermore, whereas a disrupted intestinal barrier contributes to the pathogenesis of metabolic diseases, the underlying causes remain unclear. Indeed, it may include changes in nutritional factors, infections (i.e., Helicobacter pylori infection leading to an increased rate of incident diabetes), exposure to toxins, lack of exposure to microbes in early childhood, as well as impaired function and diversity of the gut microbiota [24]. Moreover, probiotic strains can not only affect the intestinal microbiota directly but also affect other organs by modulating intestinal inflammation and permeability [25]. Several potential mechanisms underlying the beneficial effects of probiotics are illustrated in Figs 1 and 2.
The "obese microbiome" is thought to display an increased capacity to harvest energy from the diet along with a decreased ability to stimulate the production of gut factors that inhibit fat deposition [1]. Furthermore, the beneficial effect of probiotics to decrease DIO is both highly strain- and model-specific [1318262728]. For example, the gut microbiota could promote storage of triglyceride in adipocytes through suppression of intestinal expression of a circulating lipoprotein lipase (LPL) inhibitor, the angiopoietin-like 4 [2]. Nevertheless, storage of excess FA is the result of unbalanced lipid absorption involving LPL and lipid catabolism [29]. In fact, Yoo et al. [12], treated DIO-mice with a combination of probiotics that resulted in a decreased expression of genes involved in FA transport and β-oxidation (Table 1). Another potential mechanism by which probiotics can counteract the negative effect of obesogenic diet is by interaction with commensal bacteria and altering expressions of microbial enzymes, especially those involved in carbohydrate metabolism or butyrate synthesis pathways [3031]. Butyrate, with acetate and propionate, are the most abundant SCFA produced by some colonic bacteria as end-products from the breakdown of non-digestible carbohydrates that pass unaffected through the small intestine [5]. Among major bacterial phyla, Bacteroidetes are recognized as acetate and propionate producers, whereas Firmicutes are more butyrate-producing bacteria. The butyrate-producer strain miyairi 588 has shown promising effects on liver homeostasis and insulin resistance in a rat model of choline-deficient diet-induced non-alcoholic fatty liver disease (NAFLD) [32]. As reported in Table 1, Ritze et al. [21] have also shown that Lactobacillus rhamnosus GG protects against NAFLD through specifically reducing liver fat mass loss in association with modulation of the carbohydrate-responsive element-binding protein pathway.
Moreover, in many studies, the beneficial effects allocated to probiotics on body fat mass, could be explained by complex and still unclear mechanisms that may or may not involve change in caloric intake (Tables 1 and 2). Yadav et al. [11] have demonstrated that the VSL#3 probiotic promoted the release of the hormone glucagon-like protein-1 (GLP-1), resulting in reduced food intake and improved glucose tolerance, which was correlated with SCFA production leading to L-cell stimulation and GLP-1 production and the modulation of several genes involved in food intake regulation.
The modulation of the intestinal immune system is also thought to ameliorate insulin sensitivity even without decreased fat mass accumulation [3334]. The intestinal barrier is a functional entity separating the gut lumen from the inner host. It comprises elements that are mechanical (mucus, epithelial layer), humoral (defensins, immunoglobulin A), cellular or cell-mediated (lymphocytes, innate immune cells), muscular and neurological [24]. This barrier is maintained by the expression of adherens junctions and tight junctions (TJ) molecules, including cadherins, claudins, occludin, and junctional adhesion proteins, which seal adjacent cells together [35]. Moreover, the intestinal mucosa is the primary site where the mucosa-associated lymphoid tissue is exposed to and interacts with the external environment. Gut barrier integrity is influenced by both exogenous (i.e., toxins, stress, diet, vitamins, pro- and prebiotics, antibiotics, exercise [24]) and endogenous factors (i.e., inflammatory mediators, defensins, serotonin, proteases, mucus quality, and the endocannabinoid system) [36]. In obese individuals, decrease in TJ protein abundance, myosin light chain kinase activation and cytoskeletal modulation (ZO1 interacts directly with actin, occludin, claudins, or other proteins) have all been proposed to mediate cytokine-induced loss of TJ barrier function [37].
It is well documented that LAB are able to sense the environment, to produce bacteriocins which can directly modulate gut microbiota populations (Figs 1 and 2), but also organic acids (i.e., lactic and acetic acids) that indirectly inhibit pathogen colonization by decreasing intestinal pH or increasing peristalsis [38]. By preventing the invasion of undesirable microorganisms, beneficial probiotic effects can also reinforce intestinal barrier integrity. Indeed, mucosal permeability is adaptable and may be directly regulated in response to extracellular stimuli, such as nutrients and bacteria. These interactions can result in the variation of gene expression of receptors involved in numerous and diverse pathways leading to the production of cytokines and other active molecules, secreted from epithelial cells into the lumen inducing gut microbiota modulations. It was recently demonstrated that DIO-mice display low-grade systemic inflammation and metabolic perturbations, in association with reduced intestinal bifidobacteria and increased plasma levels of endotoxin (LPS), a trait strongly correlated with disrupted intestinal barrier integrity [6]. Moreover, plasma citrulline and intestinal FA-binding protein levels (markers of gut barrier integrity) are significantly elevated in severely obese diabetic individuals, which was associated with increased small-intestinal enterocyte mass and increased enterocyte turnover [39]. Furthermore, there are several families of innate receptors which are involved in the recognition of microbe-associated molecular patterns (including Toll-like receptors, NOD-like receptors, or inflammasomes) [40]. Moreover, changes in gut microbiota modulate endotoxemia by a mechanism that affects gut barrier function and increases intestinal permeability, which may involve the disruption of TJ [641]. Cani's group recently focused on the probiotic effect of Akkermansia muciniphila, an interesting mucin-degrading member of the Verrucomicrobia phylum, and found that its administration to DIO mice decreases metabolic endotoxemia and adipose tissue inflammation by improving intestinal mucosal barrier function, a trait linked to an increased mucus layer thickness [15].
Among other potential mechanisms involved in the maintenance of intestinal homeostasis, butyrate production has also been suggested to alleviate intestinal bowel diseases (IBDs) through its ability to inhibit histone deacetylases [42] and to activate G-coupled protein receptors [43], leading to enhanced protective immunity and improved gut barrier. Inoculation of mice with the butyrate-producers Clostridium cluster IV and XIVa or butyrate administration per se were both capable of expanding the colonic population of regulatory T cells (Treg), which increases the production of the anti-inflammatory cytokine interleukin 10 and reduces the colonic population of the pro-inflammatory CD4+ T cells [44]. Similarly, oral administration of Butyricicoccus pullicaecorum, whose presence was found to be lower in IBD patients compared with healthy subjects, attenuated intestinal inflammation in a rat model of colitis [45]. While the resolution of obesity-induced intestinal inflammation is a valuable strategy to improve whole-body metabolism [33], butyrate can also act at the systemic level to exert anti-obesity and anti-inflammatory effects [46]. Interestingly, given the large body of literature supporting the beneficial effects of butyrate, the administration of butyrate producer strains such as Faecalibacterium prausnitzii, Roseburia intestinalis, or Anaerostipes caccae may confer predictability and safeness to potential probiotic-based treatments of several pro-inflammatory disorders [4748].
The concomitant use of probiotics with specific prebiotics, known as sources of "non-digestible compound that, through its metabolization by microorganisms in the gut, modulates composition and/or activity of the gut microbiota, thus conferring a beneficial physiological effect on the host" [49] should also be considered as mean of improving health status. Prebiotics can improve probiotic effects on body weight loss and maintenance, when they are co-administrated to the host organism [50]. Moreover, inulin-type fructans (ITFs) have been shown to affect gut ecology and stimulate immune cell activity, as well as decreasing body weight gain and fat mass in obese individuals [51]. It also appears that polyphenols can, in conjunction with a probiotic strain of Bacillus, stimulate the growth of anti-inflammatory bacterial species belonging to the genus Barnesiella and improve the bioavailability of certain health beneficial polyphenols [52]. In the context of obesity, the use of relatively new prebiotics such as arabinoxylan (AX) and arabinoxylan oligo-saccharides (AXOS) may be promising candidates to counteract related metabolic disorders, since AX and AXOS have been linked to adiposity reduction [53] and lower metabolic endotoxemia [54] in obese mice, respectively. Furthermore, there is growing evidence that the bifidogenic and butyrogenic effects of AX and AXOS are reflected in potential cross-feeding mechanisms, such as for ITF, in which primary degraders such as Bifidobacterium selectively and competitively degrade these fructose polymers to produce acetate and lactate that are consumed by secondary degraders such as Roseburia to produce butyrate [55]. Interestingly, AX administration in rodents has also been involved in gut microbiota modulation; firstly by increasing Bifidobacterium and Roseburia in DIO mice [56], and secondly by shifting mucin degradation from the caecum to the colon where a higher abundance of mucolytic A. muciniphila may locally produce beneficial metabolites such as propionate [57].
Another growing concept is to genetically engineer bacterial strains in order to reinforce a pre-existing probiotic capacity or to increase their effectiveness. In fact, LAB have been genetically manipulated in order to target the delivery of antioxidant and anti-inflammatory molecules produced by probiotics (i.e., enzymes, cytokines) for the treatment of IBD [58]. Since the use of gut anti-inflammatory agents is promising against the metabolic syndrome [33], it would be interesting to test whether these engineered LAB originally conceived to counter IBD could also exert positive effects on obesity and associated metabolic disorders. Indeed, Duan et al. [59] recently reported the successful application of an engineered probiotic that secretes the inactive full-length form of GLP-1 to reprogram intestinal cells into glucose-responsive insulin-secreting cells for the treatment of type 1 diabetes. Another interesting potential strategy is the genetic modification of the probiotic E. coli Nissle 1917 to produce N-acylphosphatidylethanolamines, which is converted quickly after meals into potent appetite-suppressing lipids, know as N-acylethanolamines [60]. The aforementioned examples show the great potential of engineered strains as a strategy to treat obesity and its metabolic consequences.
Altogether, various studies (Tables 1 and 2) demonstrate that probiotic administration may confer beneficial effects in the prevention and treatment of obesity, inflammation and other associated metabolic disorders through various mechanisms including direct effects on mucosal barrier and surrounding cells in particular, that can impede on chronic inflammation (Figs. 1 and 2). Currently, researchers are on the path to uncover beneficial and detrimental gut microbiota phylotypes that could lead to the use of living probiotics in order to reshape gut bacterial communities in beneficial ways to the host. The major issue that hampers a meta-analysis comparison of all the potential probiotic strains is the considerable heterogeneity between protocols used in many studies (model, dose, treatment, and times). For the same reason, research on probiotics are still confronted with an apparent lack of conclusive results, further limited by the small number of trials where the application of probiotics was evaluated in double-blinded large-scale cohorts studies, particularly in the context of obesity prevention. Indeed, even if FMT showed very good results in recent human trials, the fact that potential adverse effects have also been reported, calls for caution because probiotics are already used for obesity management. This is particularly true for specific groups (i.e., neonates infants or individuals with immune deficiency) that may be a greater risk for adverse effects of probiotics. Moreover, a better understanding of how environmental factors (i.e., culture conditions, product formulations, storage time, host metagenome and genotype and variability of consumer-associated factors) influence probiotic function would ultimately be useful for unraveling the significant inter-individual variation in response to probiotic bacteria among human subjects and for comparing outcomes of different clinical studies. Despite the methodological and regulatory issues raised above, the field of probiotics is evolving based on a growing body of research, which is paving the way for a successful strategy against obesity and its related comorbidities, using strains capable of producing well characterized molecules, or using engineered bacteria that ensures safety of use. Moreover, the increased interest in the role of the gut microbiota in host's physiology is revealing novel potential probiotic strains while triggering a regain of interest in probiotics as a tool to manipulate intestinal bacterial communities and therefore treat/prevent intestinal and systemic diseases.

CONFLICTS OF INTEREST: No potential conflict of interest relevant to this article was reported.

  • 1. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006;444:1027-1031. ArticlePubMedPDF
  • 2. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, Semenkovich CF, Gordon JI. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 2004;101:15718-15723. ArticlePubMedPMC
  • 3. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Turnbaugh PJ. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505:559-563. ArticlePubMedPDF
  • 4. Khan MT, Nieuwdorp M, Backhed F. Microbial modulation of insulin sensitivity. Cell Metab 2014;20:753-760. ArticlePubMed
  • 5. Hur KY, Lee MS. Gut microbiota and metabolic disorders. Diabetes Metab J 2015;39:198-203. ArticlePubMedPMC
  • 6. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, 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. ArticlePubMedPDF
  • 7. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB, Flint HJ, Salminen S, Calder PC, Sanders ME. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol 2014;11:506-514. ArticlePubMedPDF
  • 8. Gareau MG, Sherman PM, Walker WA. Probiotics and the gut microbiota in intestinal health and disease. Nat Rev Gastroenterol Hepatol 2010;7:503-514. ArticlePubMedPMCPDF
  • 9. de Moreno de LeBlanc A, LeBlanc JG. Effect of probiotic administration on the intestinal microbiota, current knowledge and potential applications. World J Gastroenterol 2014;20:16518-16528. ArticlePubMedPMC
  • 10. Champagne CP, Gardner NJ, Roy D. Challenges in the addition of probiotic cultures to foods. Crit Rev Food Sci Nutr 2005;45:61-84. ArticlePubMed
  • 11. Yadav H, Lee JH, Lloyd J, Walter P, Rane SG. Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion. J Biol Chem 2013;288:25088-25097. ArticlePubMedPMC
  • 12. Yoo SR, Kim YJ, Park DY, Jung UJ, Jeon SM, Ahn YT, Huh CS, McGregor R, Choi MS. Probiotics L. plantarum and L. curvatus in combination alter hepatic lipid metabolism and suppress diet-induced obesity. Obesity (Silver Spring) 2013;21:2571-2578. ArticlePubMedPDF
  • 13. Stenman LK, Waget A, Garret C, Klopp P, Burcelin R, Lahtinen S. Potential probiotic Bifidobacterium animalis ssp. lactis 420 prevents weight gain and glucose intolerance in diet-induced obese mice. Benef Microbes 2014;5:437-445. ArticlePubMed
  • 14. Kang JH, Yun SI, Park MH, Park JH, Jeong SY, Park HO. Anti-obesity effect of Lactobacillus gasseri BNR17 in high-sucrose diet-induced obese mice. PLoS One 2013;8:e54617ArticlePubMedPMC
  • 15. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, de Vos WM, Cani PD. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 2013;110:9066-9071. ArticlePubMedPMC
  • 16. Everard A, Matamoros S, Geurts L, Delzenne NM, Cani PD. 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-e01014. PubMedPMC
  • 17. Wang J, Tang H, Zhang C, Zhao Y, Derrien M, Rocher E, van-Hylckama Vlieg JE, Strissel K, Zhao L, Obin M, Shen J. Modulation of gut microbiota during probiotic-mediated attenuation of metabolic syndrome in high fat diet-fed mice. ISME J 2015;9:1-15. ArticlePubMedPDF
  • 18. Wu CC, Weng WL, Lai WL, Tsai HP, Liu WH, Lee MH, Tsai YC. Effect of Lactobacillus plantarum strain K21 on high-fat diet-fed obese mice. Evid Based Complement Alternat Med 2015;2015:391767ArticlePubMedPMCPDF
  • 19. Cousin FJ, Deutsch SM, Perez Chaia A, Foligne B, Jan G. Interactions between probiotic dairy propionibacteria and the intestinal epithelium. Curr Immunol Rev 2012;8:216-226.Article
  • 20. Oksaharju A, Kooistra T, Kleemann R, van Duyvenvoorde W, Miettinen M, Lappalainen J, Lindstedt KA, Kovanen PT, Korpela R, Kekkonen RA. Effects of probiotic Lactobacillus rhamnosus GG and Propionibacterium freudenreichii ssp. shermanii JS supplementation on intestinal and systemic markers of inflammation in ApoE*3Leiden mice consuming a high-fat diet. Br J Nutr 2013;110:77-85. ArticlePubMed
  • 21. Ritze Y, Bardos G, Claus A, Ehrmann V, Bergheim I, Schwiertz A, Bischoff SC. Lactobacillus rhamnosus GG protects against non-alcoholic fatty liver disease in mice. PLoS One 2014;9:e80169ArticlePubMedPMC
  • 22. Park DY, Ahn YT, Park SH, Huh CS, Yoo SR, Yu R, Sung MK, McGregor RA, Choi MS. Supplementation of Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032 in diet-induced obese mice is associated with gut microbial changes and reduction in obesity. PLoS One 2013;8:e59470ArticlePubMedPMC
  • 23. O'Shea EF, Cotter PD, Stanton C, Ross RP, Hill C. Production of bioactive substances by intestinal bacteria as a basis for explaining probiotic mechanisms: bacteriocins and conjugated linoleic acid. Int J Food Microbiol 2012;152:189-205. ArticlePubMed
  • 24. Thomas LV, Ockhuizen T, Suzuki K. Exploring the influence of the gut microbiota and probiotics on health: a symposium report. Br J Nutr 2014;112(Suppl 1):S1-S18. ArticlePMC
  • 25. Moya-Perez A, Romo-Vaquero M, Tomas-Barberan F, Sanz Y, Garcia-Conesa MT. Hepatic molecular responses to Bifidobacterium pseudocatenulatum CECT 7765 in a mouse model of diet-induced obesity. Nutr Metab Cardiovasc Dis 2014;24:57-64. ArticlePubMed
  • 26. Hariri M, Salehi R, Feizi A, Mirlohi M, Kamali S, Ghiasvand R. The effect of probiotic soy milk and soy milk on anthropometric measures and blood pressure in patients with type II diabetes mellitus: a randomized double-blind clinical trial. ARYA Atheroscler 2015;11(1 Suppl):74-80. PubMedPMC
  • 27. Minami J, Kondo S, Yanagisawa N, Odamaki T, Xiao JZ, Abe F, Nakajima S, Hamamoto Y, Saitoh S, Shimoda T. Oral administration of Bifidobacterium breve B-3 modifies metabolic functions in adults with obese tendencies in a randomised controlled trial. J Nutr Sci 2015;4:e17ArticlePubMedPMC
  • 28. Nabavi S, Rafraf M, Somi MH, Homayouni-Rad A, Asghari-Jafarabadi M. Effects of probiotic yogurt consumption on metabolic factors in individuals with nonalcoholic fatty liver disease. J Dairy Sci 2014;97:7386-7393. ArticlePubMed
  • 29. Ahn HY, Kim M, Ahn YT, Sim JH, Choi ID, Lee SH, Lee JH. The triglyceride-lowering effect of supplementation with dual probiotic strains, Lactobacillus curvatus HY7601 and Lactobacillus plantarum KY1032: Reduction of fasting plasma lysophosphatidylcholines in nondiabetic and hypertriglyceridemic subjects. Nutr Metab Cardiovasc Dis 2015;25:724-733. ArticlePubMed
  • 30. McNulty NP, Yatsunenko T, Hsiao A, Faith JJ, Muegge BD, Goodman AL, Henrissat B, Oozeer R, Cools-Portier S, Gobert G, Chervaux C, Knights D, Lozupone CA, Knight R, Duncan AE, Bain JR, Muehlbauer MJ, Newgard CB, Heath AC, Gordon JI. The impact of a consortium of fermented milk strains on the gut microbiome of gnotobiotic mice and monozygotic twins. Sci Transl Med 2011;3:106ra106.ArticlePubMedPMC
  • 31. Veiga P, Pons N, Agrawal A, Oozeer R, Guyonnet D, Brazeilles R, Faurie JM, van Hylckama, Houghton LA, Whorwell PJ, Ehrlich SD, Kennedy SP. Changes of the human gut microbiome induced by a fermented milk product. Sci Rep 2014;4:6328ArticlePubMedPMCPDF
  • 32. Endo H, Niioka M, Kobayashi N, Tanaka M, Watanabe T. Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progression in rats: new insight into the probiotics for the gut-liver axis. PLoS One 2013;8:e63388ArticlePubMedPMC
  • 33. Luck H, Tsai S, Chung J, Clemente-Casares X, Ghazarian M, Revelo XS, Lei H, Luk CT, Shi SY, Surendra A, Copeland JK, Ahn J, Prescott D, Rasmussen BA, Chng MH, Engleman EG, Girardin SE, Lam TK, Croitoru K, Dunn S, Philpott DJ, Guttman DS, Woo M, Winer S, Winer DA. Regulation of obesity-related insulin resistance with gut anti-inflammatory agents. Cell Metab 2015;21:527-542. ArticlePubMed
  • 34. Hulston CJ, Churnside AA, Venables MC. Probiotic supplementation prevents high-fat, overfeeding-induced insulin resistance in human subjects. Br J Nutr 2015;113:596-602. ArticlePubMedPMC
  • 35. Laukoetter MG, Bruewer M, Nusrat A. Regulation of the intestinal epithelial barrier by the apical junctional complex. Curr Opin Gastroenterol 2006;22:85-89. ArticlePubMed
  • 36. Rousseaux C, Thuru X, Gelot A, Barnich N, Neut C, Dubuquoy L, Dubuquoy C, Merour E, Geboes K, Chamaillard M, Ouwehand A, Leyer G, Carcano D, Colombel JF, Ardid D, Desreumaux P. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nat Med 2007;13:35-37. ArticlePubMedPDF
  • 37. Turner JR. Intestinal mucosal barrier function in health and disease. Nat Rev Immunol 2009;9:799-809. ArticlePubMedPDF
  • 38. Kailasapathy K, Chin J. Survival and therapeutic potential of probiotic organisms with reference to Lactobacillus acidophilus and Bifidobacterium spp. Immunol Cell Biol 2000;78:80-88. ArticlePubMedPDF
  • 39. Verdam FJ, Greve JW, Roosta S, van Eijk H, Bouvy N, Buurman WA, Rensen SS. Small intestinal alterations in severely obese hyperglycemic subjects. J Clin Endocrinol Metab 2011;96:E379-E383. ArticlePubMedPDF
  • 40. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature 2011;474:327-336. ArticlePubMedPMCPDF
  • 41. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, Gibson GR, Delzenne NM. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007;50:2374-2383. ArticlePubMedPDF
  • 42. Felice C, Lewis A, Armuzzi A, Lindsay JO, Silver A. Review article: selective histone deacetylase isoforms as potential therapeutic targets in inflammatory bowel diseases. Aliment Pharmacol Ther 2015;41:26-38. ArticlePubMed
  • 43. Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 2013;145:396-406. 406.e1-406.e10. ArticlePubMed
  • 44. Smith PM, Howitt MR, Panikov N, Michaud M, Gallini CA, Bohlooly YM, Glickman JN, Garrett WS. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013;341:569-573. ArticlePubMed
  • 45. Eeckhaut V, Machiels K, Perrier C, Romero C, Maes S, Flahou B, Steppe M, Haesebrouck F, Sas B, Ducatelle R, Vermeire S, Van Immerseel F. Butyricicoccus pullicaecorum in inflammatory bowel disease. Gut 2013;62:1745-1752. ArticlePubMed
  • 46. den Besten G, Bleeker A, Gerding A, van Eunen K, Havinga R, van Dijk TH, Oosterveer MH, Jonker JW, Groen AK, Reijngoud DJ, Bakker BM. Short-chain fatty acids protect against high-fat diet-induced obesity via a PPARgamma-dependent switch from lipogenesis to fat oxidation. Diabetes 2015;64:2398-2408. PubMed
  • 47. Quevrain E, Maubert MA, Michon C, Chain F, Marquant R, Tailhades J, Miquel S, Carlier L, Bermudez-Humaran LG, Pigneur B, Lequin O, Kharrat P, Thomas G, Rainteau D, Aubry C, Breyner N, Afonso C, Lavielle S, Grill JP, Chassaing G, Chatel JM, Trugnan G, Xavier R, Langella P, Sokol H, Seksik P. Identification of an anti-inflammatory protein from Faecalibacterium prausnitzii, a commensal bacterium deficient in Crohn's disease. Gut 2015 6 04 [Epub].
  • 48. Falony G, Vlachou A, Verbrugghe K, De Vuyst L. Cross-feeding between Bifidobacterium longum BB536 and acetate-converting, butyrate-producing colon bacteria during growth on oligofructose. Appl Environ Microbiol 2006;72:7835-7841. ArticlePubMedPMCPDF
  • 49. Bindels LB, Neyrinck AM, Salazar N, Taminiau B, Druart C, Muccioli GG, Francois E, Blecker C, Richel A, Daube G, Mahillon J, de Los Reyes-Gavilan CG, Cani PD, Delzenne NM. Non digestible oligosaccharides modulate the gut microbiota to control the development of leukemia and associated cachexia in mice. PLoS One 2015;10:e0131009ArticlePubMedPMC
  • 50. Sanchez M, Darimont C, Drapeau V, Emady-Azar S, Lepage M, Rezzonico E, Ngom-Bru C, Berger B, Philippe L, Ammon-Zuffrey C, Leone P, Chevrier G, St-Amand E, Marette A, Dore J, Tremblay A. Effect of Lactobacillus rhamnosus CGMCC1.3724 supplementation on weight loss and maintenance in obese men and women. Br J Nutr 2014;111:1507-1519. ArticlePubMed
  • 51. Parnell JA, Reimer RA. Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am J Clin Nutr 2009;89:1751-1759. ArticlePubMedPMC
  • 52. Dudonne S, Varin TV, Forato Anhe F, Dube P, Roy D, Pilon G, Marette A, Levy E, Jacquot C, Urdaci M, Desjardins Y. Modulatory effects of a cranberry extract co-supplementation with Bacillus subtilis CU1 probiotic on phenolic compounds bioavailability and gut microbiota composition in high-fat diet-fed mice. PharmaNutrition 2015 5 01 [Epub].
  • 53. Delzenne NM, Neyrinck AM, Cani PD. Gut microbiota and metabolic disorders: how prebiotic can work? Br J Nutr 2013;109(Suppl 2):S81-S85. ArticlePubMed
  • 54. Neyrinck AM, Van Hee VF, Piront N, De Backer F, Toussaint O, Cani PD, Delzenne NM. Wheat-derived arabinoxylan oligosaccharides with prebiotic effect increase satietogenic gut peptides and reduce metabolic endotoxemia in diet-induced obese mice. Nutr Diabetes 2012;2:e28ArticlePubMedPMCPDF
  • 55. De Vuyst L, Leroy F. Cross-feeding between bifidobacteria and butyrate-producing colon bacteria explains bifdobacterial competitiveness, butyrate production, and gas production. Int J Food Microbiol 2011;149:73-80. ArticlePubMed
  • 56. Neyrinck AM, Possemiers S, Druart C, Van de Wiele T, De Backer F, Cani PD, Larondelle Y, Delzenne NM. Prebiotic effects of wheat arabinoxylan related to the increase in bifidobacteria, Roseburia and Bacteroides/Prevotella in diet-induced obese mice. PLoS One 2011;6:e20944ArticlePubMedPMC
  • 57. Van den Abbeele P, Gerard P, Rabot S, Bruneau A, El Aidy S, Derrien M, Kleerebezem M, Zoetendal EG, Smidt H, Verstraete W, Van de Wiele T, Possemiers S. Arabinoxylans and inulin differentially modulate the mucosal and luminal gut microbiota and mucin-degradation in humanized rats. Environ Microbiol 2011;13:2667-2680. ArticlePubMed
  • 58. de Moreno de LeBlanc A, Del Carmen S, Chatel JM, Miyoshi A, Azevedo V, Langella P, Bermudez-Humaran LG, LeBlanc JG. Current review of genetically modified lactic acid bacteria for the prevention and treatment of colitis using murine models. Gastroenterol Res Pract 2015;2015:146972PubMedPMC
  • 59. Duan FF, Liu JH, March JC. Engineered commensal bacteria reprogram intestinal cells into glucose-responsive insulin-secreting cells for the treatment of diabetes. Diabetes 2015;64:1794-1803. ArticlePubMedPMCPDF
  • 60. Chen Z, Guo L, Zhang Y, Walzem RL, Pendergast JS, Printz RL, Morris LC, Matafonova E, Stien X, Kang L, Coulon D, McGuinness OP, Niswender KD, Davies SS. Incorporation of therapeutically modified bacteria into gut microbiota inhibits obesity. J Clin Invest 2014;124:3391-3406. ArticlePubMedPMC
Fig. 1

Potential beneficial effects of probiotic supplementation against metabolic disorders. GPR, G protein-coupled receptor; SCFA, short-chain fatty acid; ChREBP, carbohydrate-responsive element-binding protein; SREBP, sterol regulatory element-binding protein; AMPK, AMP-activated protein kinase; ANGPTL4, angiopoietin-like protein 4; TJ, tight junction; AJ, adherens junction.

dmj-39-291-g001.jpg
Fig. 2

Potential direct effects of probiotics to protect gut microbiota and intestinal barrier integrity. Obesogenic diet or "Western diet" alter gut microbiota population diversity and intestinal barrier integrity. Cross-talk between ingested probiotics, gut microbiota (commensal bacteria) and epithelial cells (1). Probiotics produce metabolites that could serve to increase both the diversity of commensal bacteria and the availability of nutrients used by intestinal epithelial cells (IEC). Commensal bacteria multiply and in turn, also produce metabolites that could be used by surrounding cells. In patients suffering from metabolic disorders, intestinal permeability is altered leading to an increase of low-grade inflammation and metabolic endotoxemia (2). Probiotics can increase production of tight- and adherens junction (TJ and AJ) proteins (3), improving gut permeability and inhibiting the passage of lipopolysaccharides (LPS) into systemic circulation that decreases metabolic endotoxemia. Moreover, probiotics express microorganism-associated molecular patterns (MAMPs) that could bind to host pattern recognition receptors (PRRs) located at cell surface of IEC (1) and dendritic cells (4), and induce the activation/inhibition of signaling pathways. For example (5), probiotics can stimulate dendritic cells leading to inhibition of pro-inflammatory CD4+ cell proliferation and activation of anti-inflammatory pathways (Treg and plasma cell proliferation, resulting in production of anti-inflammatory cytokines and IgA immunoglobulins (6), respectively). Mainly present in the mucus layer, IgA reinforce the protective role of mucosal barrier. Mucus production can also be increased by probiotics that stimulate goblet cells leading to activation of mucin gene expression and therefore production of mucin glycoproteins (7). Once assembled, these proteins are excreted and form the mucus layer, which acts as barrier against pathogen colonization. However, probiotics can also induce physical barrier against pathogens or produce bacteriocins that inhibits undesirable microorganism invasion (8). TLR, Toll-like receptor; NLR, NOD-like receptor; TNF-α, tumor necrosis factor α; IFN-γ, interferon γ; TGF-β, transforming growth factor β; IL-10, interleukin 10.

dmj-39-291-g002.jpg
Table 1

Recent studies about beneficial probiotic effects on metabolic disorders in mice

dmj-39-291-i001.jpg
Probiotic strains Dose Host organism Diet Treatment Principal findings Reference
VSL#3 (L. acidophilus MB 443, L. delbrueckii subsp. bulgaricus MB 453, L. casei MB 451, L. plantarum MB 452, B. longum Y10, B. infantis Y1, B. breve Y8, and S. salivarius subsp. thermophilus MB 455) 5 mg/kg of body weight Mice (C57BL6) HFD Preventive: 8 wk ↓Body weight gain; ↓fat mass accumulation in WAT and liver, and adipocyte size; ↑insulin and glucose tolerance; ↓plasma pro-inf. cytokines, TG and FFA; ↓resistin; ↑adiponectin; ↑Stat3 phosphorylation in the hypothalamus; expression levels of food intake regulatory genes : ↓AgRP, ↓NpY, and ↑POMC; ↓Firmicutes; ↑Bacteroidetes; ↑Bifidobactericeae; ↑butyrate production; ↑GLP-1 secretion Yadav et al. (2013) [11]
Therapeutic: 13 wk on HFD+8 wk of treatment
L. plantarum KY1032; L. curvatus HY7601 1010 cfu/day for each strain Mice (C57BL6) HFHC 9 wk Probiotic strains combinaison ↓plasma and liver chol; ↓fat mass accumulation in WAT and liver; ↓transcriptional regulators of lipid metabolism genes; ↓FA synthesis-related genes and FA β-oxidation genes; ↓LPL in WAT; multi-strain probiotics may prove more beneficial than single-strain probiotics Yoo et al. (2013) [12]
L. plantarum KY1032; L. curvatus HY7601 5.109 cfu/day for each strain Mice (C57BL6) HFD 10 wk of treatment after DIO in 8 wk ↓Body weight gain; ↓fat mass accumulation; ↓ALT; ↓plasma chol, leptin, and insulin; ↓eWAT pro-inf. gene expression; ↓gut microbiota diversity; ↓A. muciniphila; ↑Ruminococcaceae; ↑Lachnospiraceae Park et al. (2013) [22]
Propionibacterium freudenreichii spp. shermanii JS (PJS) or L. rhamnosus GG (GG) 109 cfu/day for each strain Heterozygous ApoE*3Leiden C57B1/6 mice HFD 4 wk PJS ↓body weight gain and gut TNF-α intensity; PJS and GG ↓intestinal mast cell numbers and ↓plasma ALT; GG ↑gut IL-10 intensity Oksaharju et al. (2013) [20]
Bifidobacterium pseudocatenulatum CECT 7765 109 cfu/day Mice (C57BL6) HFD 7 wk ↓Serum chol, TG, glucose, insulin, and leptin levels; modulation of liver expression of key proteins involved in the energy metabolism and lipid transports Moya-Perez et al. (2014) [25]
B. animalis subsp. lactis 420 109 cfu/day Mice (C57BL6) HFD 12 wk ↓Fat mass accumulation in obese and diabetic mice; ↓body weight gain; ↓metabolic endotoxemia Stenman et al. (2014) [13]
Lactobacillus gasseri BNR17 109-1010 cfu/day Mice (C57BL6) HFD 10 wk ↓Body weight gain; ↓fat mass accumulation; ↑GLUT4 mRNA expression in WAT; ↑mRNA expressions of ACO, CPT1, PPARα, PPARδ, and ANGPTL4 in the liver; ↓serum insulin and leptin Kang et al. (2013) [14]
A. muciniphila 2.108 cfu/day Mice (C57BL6) HFD 4 wk ↓s.c., mesenteric, epididymal fat mass, body weight gain, insulin resistance, endotoxemia; ↑mRNA expression of markers of adipocyte differentiation and lipid oxydation; A. muciniphila treatment restored mucus layer thickness Everard et al. (2013) [15]
S. boulardii Biocodex (yeast) 120 mg/day Mice (db/db) Chow diet 4 wk ↓Body weight gain; ↓fat mass accumulation; ↓hepatic steatosis (lipid content, liver weight); ↓hepatic and systemic inflammation (MCP1, IL-1β, IL-4, IL-6, TNF-α); ↑caecum weight; ↑Bacteroidetes; ↓Firmicutes; ↓Proteobacteria; ↓Tenericutes; S. boulardii treatment ↓genera associated with diabetes and inflammation in db/db mice (i.e., Odoribacter, Ruminococcus, Prevotella) Everard et al. (2014) [16]
L. rhamnosus GG 5.107 cfu/g of body weight Mice (C57BL6, female) HFrD 8 wk ↓Development of high-fructose induced NAFLD; ↓plasma ALT; ↓liver fat accumulation; ↓liver expression of ChREBP, ACC1 and FAS; ↓liver inflammation (TNF-α, IL-1β); ↑gut expression of occludin and claudin-1; LGG almost normalized the elevated portal LPS levels in HFrD fed mice; ↑Bacteroidetes; ↑Firmicutes Ritze et al. (2014) [21]
Lactobacillus paracasei CNCM I-4270 (LC), L. rhamnosus I-3690 (LR) or B. animalis subsp. lactis I-2494 (BA) 108 cells/day (individually, not in combination) Mice (C57BL6) HFD 12 wk Each strain attenuated weight gain, macrophage infiltration into eWAT, markedly ↑glucose-insulin homeostasis and hepatic steatosis; BA more robustly attenuated inflammatory effect of HFD (↓TNF-α expression in eWAT and liver, ↓LBP (a marker of endotoxin load), ↑anti-inf. adiponectin levels); Lactobacillus and Bifidobacterium strains differentially affected host inflammation, gut microbial fermentation and gut microbiota composition Wang et al. (2015) [17]
L. plantarum K21 109 cfu/day Mice (C57Bl/6J) HFD 8 wk ↓Body weight gain; ↓visceral fat mass accumulation; ↓hepatic lipid, chol content; ↓plasma leptin, ALT, AST, TG, and chol; regulation of hepatic PPAR-γ expression; ↓gut permeability; ↓Clostridium perfringens; ↑Bifidobacterium; ↑Lactobacillus Wu et al. (2015) [18]

L. acidophilus, Lactobacillus acidophilus; L. delbrueckii, Lactobacillus delbrueckii; L. casei, Lactobacillus casei; L. plantarum, Lactobacillus plantarum; B. longum, Bifidobacterium longum; B. infantis, Bifidobacterium infantis; B. breve, Bifidobacterium breve; S. Salivarius, Streptococcus salivarius; HFD, high fat diet; WAT, white adipose tissue; pro-inf., pro-inflammatory; TG, triglyceride; FFA, free fatty acid; Stat 3, signal transducer and activator of transcription 3; AgRP, agouti-related peptide; NpY, neuropeptide Y; POMC, pro-opiomelanocortin; GLP-1, glucagon-like protein-1; L. curvatus, Lactobacillus curvatus; cfu, colony forming unit; HFHC, high fat/high cholesterol diet; chol, cholesterol; FA, fatty acid; LPL, lipoprotein lipase; DIO, diet-induced obesity; ALT, alanine transaminase; eWAT, epididymal white adipose tissue; A. muciniphila, Akkermansia muciniphila; L. rhamnosus, Lactobacillus rhamnosus; TNF-α, tumor necrosis factor α; IL-10, interleukin 10; B. animalis, Bifidobacterium animalis; HSD, high-sucrose diet; Glut4, glucose transporter 4; ACO, acetyl-Coa oxidase; CPT, carnitine palmytoiltransferase; PPAR, peroxisome proliferator-activated receptor; ANGPTL4, angiopoietin-like 4; s.c., subcutaneous; S. boulardii, Saccharomyces boulardii; MCP1, monocyte chemoattractant protein 1; HFrD, high-fructose diet; NAFLD, non-alcoholic fatty liver disease; ChREBP, carbohydrate-responsive element-binding protein; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; LGG, Lactobacillus rhamnosus GG; LPS, lipopolysaccharide; LBP, lipopolysaccharide binding protein; AST, aspartate transaminase.

Table 2

Most recent double-blinded studies of probiotic effects on metabolic disorders in humans

dmj-39-291-i002.jpg
Probiotic strains Dose Host organism Diet Treatment Principal findings Reference
Lactobacillus acidophilus La5; Bifidobacterium lactis Bb12 300 g of low fat (2.5% fat) yogurt/day corresponding to more than 5.108 cfu/dose of each strain NAFLD patients Own regular lifestyles (without other yogurt) 8 wk ↓Body weight and BMI; ↓serum ALT and AST, total chol and LDL-C Nabavi et al. (2014) [28]
Lactobacillus rhamnosus CGMCC1.3724+ prebiotics 1, 6.108 cfu/capsule with oligofructose and inulin (2 capsules/day) Healthy overweight men and women Energy restriction Phase 1: 12 wk of dietary restriction +/- probiotic Prebiotics improve probiotic survival; ↓of body weight gain and body fat mass in women; ↓Lachnospiraceae family in women but not in men; ↓leptin concentration in plasma Sanchez et al. (2014) [50]
Phase 2: 12 wk of weight maintenance +/probiotic
Lactobacillus curvatus HY7601; L. plantarum KY1032 0, 5.1010 cfu/day of each strain in 2 g of powder Non-diabetic and hyper-triglyceridemic subjects Own regular lifestyles 12 wk ↓Serum TG; ↑plasma apolipoprotein A-V and LDL particule size Ahn et al. (2015) [29]
Lactobacillus casei Shirota 65 mL of Yakult Light twice each day Healthy human subjects HFD (only 7 day) 4 wk (normal diet during 3 wk followed by a high-fat high-energy diet during 7 day) Trend to reduce body weight gain; prevention of ↓insulin sensitivity induced by HFD; preservation of glycaemia and insulin action Hulston et al. (2015) [34]
L. plantarum A7 200 mL soy milk/day Type 2 diabetes patients Own regular lifestyles (without consumption of other dairy products) 8 wk ↓Systolic and diastolic blood pressure Hariri et al. (2015) [26]
Bifidobacterium breve B-3 5.1010 cfu/day Overweight human subjects Own regular lifestyles 12 wk ↓Fat mass accumulation; ↓plasma HbA1c; ↑γ-GTP; ↓hCRP levels Minami et al. (2015) [27]

cfu, colony forming unit; NAFLD, non-alcoholic fatty liver disease; BMI, body mass index; ALT, alanine transaminase; AST, aspartate transaminase; chol, cholesterol; LDL-C, low density lipoprotein cholesterol; L. plantarum, Lactobacillus plantarum; TG, triglyceride; HFD, high fat diet; HbA1c, glycosylated hemoglobin; γ-GTP, γ-glutamin transpeptidase; hCRP, human C-reactive protein.

Figure & Data

References

    Citations

    Citations to this article as recorded by  
    • Akkermansia muciniphila and Alcohol‐Related Liver Diseases. A Systematic Review
      Lydie Sparfel, Sandy Ratodiarivony, Elisa Boutet‐Robinet, Sandrine Ellero‐Simatos, Anne Jolivet‐Gougeon
      Molecular Nutrition & Food Research.2024;[Epub]     CrossRef
    • Next generation probiotics: Engineering live biotherapeutics
      Sanjeeva Kumar Murali, Thomas J. Mansell
      Biotechnology Advances.2024; 72: 108336.     CrossRef
    • metaProbiotics: a tool for mining probiotic from metagenomic binning data based on a language model
      Shufang Wu, Tao Feng, Waijiao Tang, Cancan Qi, Jie Gao, Xiaolong He, Jiaxuan Wang, Hongwei Zhou, Zhencheng Fang
      Briefings in Bioinformatics.2024;[Epub]     CrossRef
    • Dietary supplementation of Lactobacillus brevis SAD ameliorates high-fat diet-induced hyperglycemia and associated metabolic issues in Swiss albino mice
      Amlan Jyoti Ghosh, Supriyo Ghosh, Rejuan Islam, Sagar Sarkar, Tilak Saha
      Egyptian Journal of Basic and Applied Sciences.2024; 11(1): 148.     CrossRef
    • Systematic review of probiotics as an adjuvant treatment for psychiatric disorders
      Evan Forth, Benjamin Buehner, Ana Storer, Cassandra Sgarbossa, Roumen Milev, Arthi Chinna Meyyappan
      Frontiers in Behavioral Neuroscience.2023;[Epub]     CrossRef
    • Antimicrobial-prebiotic: Novel dual approach of pomegranate peel extract in vitro and in food system
      Marwa Al-Moghazy, Doha H. Abou baker, Hoda S. El-Sayed
      Biocatalysis and Agricultural Biotechnology.2023; 49: 102664.     CrossRef
    • Pediococcus acidilactici pA1c® Improves the Beneficial Effects of Metformin Treatment in Type 2 Diabetes by Controlling Glycaemia and Modulating Intestinal Microbiota
      Miriam Cabello-Olmo, María Oneca, Raquel Urtasun, María J. Pajares, Saioa Goñi, José I. Riezu-Boj, Fermín I. Milagro, Josune Ayo, Ignacio J. Encio, Miguel Barajas, Miriam Araña
      Pharmaceutics.2023; 15(4): 1203.     CrossRef
    • Anti-Inflammatory, Antioxidant, Metabolic and Gut Microbiota Modulation Activities of Probiotic in Cardiac Remodeling Condition: Evidence from Systematic Study and Meta-Analysis of Randomized Controlled Trials
      Nurpudji Astuti Taslim, Muhammad Yusuf, Ade Meidian Ambari, Imke Maria Del Rosario Puling, Filzatuz Zahro Ibrahim, Hardinsyah Hardinsyah, Rudy Kurniawan, William Ben Gunawan, Nelly Mayulu, Victor F. F. Joseph, Nindy Sabrina, Mochammad Rizal, Trina Ekawati
      Probiotics and Antimicrobial Proteins.2023; 15(4): 1049.     CrossRef
    • Probiotics and Health: A Review
      Priti Tomar, Shikha Sharma, Neha Dangi
      Current Probiotics.2023;[Epub]     CrossRef
    • SCD Probiotics mitigate cafeteria diet‐induced liver damage in Wistar rats during development
      Taha Ceylani, Harun Önlü, Seda Keskin, Hüseyin Allahverdi, Hikmet Taner Teker
      Journal of Gastroenterology and Hepatology.2023; 38(12): 2142.     CrossRef
    • Lactobacillus acidophilus ameliorates obesity in mice through modulation of gut microbiota dysbiosis and intestinal permeability
      Yongbo Kang, Xing Kang, Hao Yang, Haixia Liu, Xiaodan Yang, Qingqing Liu, Haixia Tian, Yang Xue, Peng Ren, Xiaoyu Kuang, Yue Cai, Mingwei Tong, Lin Li, Weiping Fan
      Pharmacological Research.2022; 175: 106020.     CrossRef
    • Role of probiotics and prebiotics in mitigation of different diseases
      Sabeena Manzoor, Shoib Mohmad Wani, Sajad Ahmad Mir, Danish Rizwan
      Nutrition.2022; 96: 111602.     CrossRef
    • Balancing reactive oxygen species generation by rebooting gut microbiota
      Vandna Singh, Shruti Ahlawat, Hari Mohan, Sarvajeet Singh Gill, Krishna Kant Sharma
      Journal of Applied Microbiology.2022; 132(6): 4112.     CrossRef
    • Grape seed and skin extract, a potential prebiotic with anti-obesity effect through gut microbiota modulation
      Mohamed Mokrani, Kamel Charradi, Ferid Limam, Ezzedine Aouani, Maria C. Urdaci
      Gut Pathogens.2022;[Epub]     CrossRef
    • Anticandidal and Antibiofilm Effect of Synbiotics including Probiotics and Inulin-Type Fructans
      Ricardo García-Gamboa, Miguel Domínguez-Simi, Misael S. Gradilla-Hernández, Jorge Bravo, Andrés Moya, Blanca Ruiz-Álvarez, Marisela González-Avila
      Antibiotics.2022; 11(8): 1135.     CrossRef
    • The Biotics Family: Current Knowledge and Future Perspectives in Metabolic Diseases
      Codrina-Madalina Palade, Georgiana-Anca Vulpoi, Radu-Alexandru Vulpoi, Vasile Liviu Drug, Oana-Bogdana Barboi, Manuela Ciocoiu
      Life.2022; 12(8): 1263.     CrossRef
    • The role of probiotic supplementation on insulin resistance in obesity associated diabetes: A mini review
      Seeme Saha, S M Niazur Rahman, Nazmun Nahar Alam
      Biomedicine.2022; 42(4): 651.     CrossRef
    • In Vivo and In Silico Investigation of the Anti-Obesity Effects of Lactiplantibacillus plantarum Combined with Chia Seeds, Green Tea, and Chitosan in Alleviating Hyperlipidemia and Inflammation
      Dalia Elebeedy, Aml Ghanem, Asmaa Saleh, Mona H. Ibrahim, Omkulthom Al Kamaly, Mohammed A. S. Abourehab, Mohamed A. Ali, Ahmed I. Abd El Maksoud, Mahmoud A. El Hassab, Wagdy M. Eldehna
      International Journal of Molecular Sciences.2022; 23(20): 12200.     CrossRef
    • Machine learning for data integration in human gut microbiome
      Peishun Li, Hao Luo, Boyang Ji, Jens Nielsen
      Microbial Cell Factories.2022;[Epub]     CrossRef
    • Probiotics Supplementation on Cardiac Remodeling Following Myocardial Infarction: a Single-Center Double-Blind Clinical Study
      Jalal Moludi, Somaieh Saiedi, Behzad Ebrahimi, Mohammad Alizadeh, Yaser Khajebishak, Sevda Saleh Ghadimi
      Journal of Cardiovascular Translational Research.2021; 14(2): 299.     CrossRef
    • Draft genome sequence of Lactobacillus plantarum strain DMR17 isolated from homemade cow dahi of Sikkim Himalayan region: an evaluation of lactate fermentation and secondary metabolism
      Ranjan Kaushal Tirwa, Ishfaq Nabi Najar, Nagendra Thakur, Lalit Kumar Chaurasia, Buddhiman Tamang
      Archives of Microbiology.2021; 203(1): 305.     CrossRef
    • Role of Metabolic Endotoxemia in Systemic Inflammation and Potential Interventions
      Shireen Mohammad, Christoph Thiemermann
      Frontiers in Immunology.2021;[Epub]     CrossRef
    • Probiotics ameliorate chronic low-grade inflammation and fat accumulation with gut microbiota composition change in diet-induced obese mice models
      Hyunchae Joung, Jaeryang Chu, Byoung-Kook Kim, In-Suk Choi, Woosang Kim, Tae-Sik Park
      Applied Microbiology and Biotechnology.2021; 105(3): 1203.     CrossRef
    • Probiotics and Prebiotics Having Broad Spectrum Anticancer Therapeutic Potential: Recent Trends and Future Perspectives
      Varruchi Sharma, Nitin Sharma, Imran Sheikh, Vikas Kumar, Nirmala Sehrawat, Mukesh Yadav, Gobind Ram, Atul Sankhyan, Anil K. Sharma
      Current Pharmacology Reports.2021; 7(2): 67.     CrossRef
    • Metagenomic and metatranscriptomic profiling of Lactobacillus casei Zhang in the human gut
      Jicheng Wang, Jiachao Zhang, Wenjun Liu, Heping Zhang, Zhihong Sun
      npj Biofilms and Microbiomes.2021;[Epub]     CrossRef
    • A Brief Overview on Probiotics: The Health Friendly Microbes
      Sanjukta Mishra, Swastik Acharya
      Biomedical and Pharmacology Journal.2021; 14(4): 1869.     CrossRef
    • Administration of indigenous probiotics modulate high-fat diet-induced metabolic syndrome in Sprague Dawley rats
      Sakshi Khanna, Sanisha Walia, Kanthi Kiran Kondepudi, Geeta Shukla
      Antonie van Leeuwenhoek.2020; 113(9): 1345.     CrossRef
    • Probiotic-directed modulation of gut microbiota is basal microbiome dependent
      Qiangchuan Hou, Feiyan Zhao, Wenjun Liu, Ruirui Lv, Wei Wei Thwe Khine, Jia Han, Zhihong Sun, Yuan-Kun Lee, Heping Zhang
      Gut Microbes.2020; 12(1): 1736974.     CrossRef
    • Bifidobacterium pseudolongum reduces triglycerides by modulating gut microbiota in mice fed high-fat food
      Ting-bei Bo, Jing Wen, Yuan-chun Zhao, Shuang-jie Tian, Xue-ying Zhang, De-hua Wang
      The Journal of Steroid Biochemistry and Molecular Biology.2020; 198: 105602.     CrossRef
    • Light at night affects gut microbial community and negatively impacts host physiology in diurnal animals: Evidence from captive zebra finches
      Indu Malik, Twinkle Batra, Subhajit Das, Vinod Kumar
      Microbiological Research.2020; 241: 126597.     CrossRef
    • Lactobacillus rhamnosus2016SWU.05.0601 regulates immune balance in ovalbumin‐sensitized mice by modulating expression of the immune‐related transcription factors and gut microbiota
      Jiajia Song, Yang Li, Jian Li, Hongwei Wang, Yu Zhang, Huayi Suo
      Journal of the Science of Food and Agriculture.2020; 100(13): 4930.     CrossRef
    • Probiotic Cocktail Identified by Microbial Network Analysis Inhibits Growth, Virulence Gene Expression, and Host Cell Colonization of Vancomycin-Resistant Enterococci
      Wei-Sheng Sun, Yuarn-Jang Lee, Kun-Nan Tsai, Yu-Hsuan Ho, Shiuh-Bin Fang
      Microorganisms.2020; 8(6): 816.     CrossRef
    • Potential therapeutic applications of the gut microbiome in obesity: from brain function to body detoxification
      Béatrice S.-Y. Choi, Laurence Daoust, Geneviève Pilon, André Marette, Angelo Tremblay
      International Journal of Obesity.2020; 44(9): 1818.     CrossRef
    • The Multispecies Probiotic Effectively Reduces Homocysteine Concentration in Obese Women: A Randomized Double-Blind Placebo-Controlled Study
      Karolina Majewska, Matylda Kręgielska-Narożna, Hieronim Jakubowski, Monika Szulińska, Paweł Bogdański
      Journal of Clinical Medicine.2020; 9(4): 998.     CrossRef
    • Impact of probiotics and prebiotics targeting metabolic syndrome
      Douglas Xavier-Santos, Raquel Bedani, Egidio Dorea Lima, Susana Marta Isay Saad
      Journal of Functional Foods.2020; 64: 103666.     CrossRef
    • Composite probiotics alleviate type 2 diabetes by regulating intestinal microbiota and inducing GLP-1 secretion in db/db mice
      Yanming Wang, Dinareer Dilidaxi, Yuche Wu, Jialehasibieke Sailike, Xin Sun, Xin-hua Nabi
      Biomedicine & Pharmacotherapy.2020; 125: 109914.     CrossRef
    • Lactic Acid Bacteria: Food Safety and Human Health Applications
      Raphael D. Ayivi, Rabin Gyawali, Albert Krastanov, Sulaiman O. Aljaloud, Mulumebet Worku, Reza Tahergorabi, Roberta Claro da Silva, Salam A. Ibrahim
      Dairy.2020; 1(3): 202.     CrossRef
    • Probiotics supplementation and insulin resistance: a systematic review
      Bárbara Izabel Moraes Salles, Débora Cioffi, Sandra Roberta G. Ferreira
      Diabetology & Metabolic Syndrome.2020;[Epub]     CrossRef
    • Impact of Gut Microbiota Composition on Onset and Progression of Chronic Non-Communicable Diseases
      Annalisa Noce, Giulia Marrone, Francesca Di Daniele, Eleonora Ottaviani, Georgia Wilson Jones, Roberta Bernini, Annalisa Romani, Valentina Rovella
      Nutrients.2019; 11(5): 1073.     CrossRef
    • Anti-Obesity Effects of Lactobacillus fermentum CQPC05 Isolated from Sichuan Pickle in High-Fat Diet-Induced Obese Mice through PPAR-α Signaling Pathway
      Kai Zhu, Fang Tan, Jianfei Mu, Ruokun Yi, Xianrong Zhou, Xin Zhao
      Microorganisms.2019; 7(7): 194.     CrossRef
    • Gut Microbiota Alteration After Long-Term Consumption of Probiotics in the Elderly
      Renyuan Gao, Xiaohui Zhang, Linsheng Huang, Rongrong Shen, Huanlong Qin
      Probiotics and Antimicrobial Proteins.2019; 11(2): 655.     CrossRef
    • Effects of Regular Kefir Consumption on Gut Microbiota in Patients with Metabolic Syndrome: A Parallel-Group, Randomized, Controlled Study
      Ezgi BELLIKCI-KOYU, Banu Pınar SARER-YUREKLI, Yakut AKYON, Fadime AYDIN-KOSE, Cem KARAGOZLU, Ahmet Gokhan OZGEN, Annika BRINKMANN, Andreas NITSCHE, Koray ERGUNAY, Engin YILMAZ, Zehra BUYUKTUNCER
      Nutrients.2019; 11(9): 2089.     CrossRef
    • In vivo screening of multiple bacterial strains identifies Lactobacillus rhamnosus Lb102 and Bifidobacterium animalis ssp. lactis Bf141 as probiotics that imp
      Mélanie Le Barz, Noëmie Daniel, Thibault V. Varin, Sabrine Naimi, Véronique Demers‐Mathieu, Geneviève Pilon, Julie Audy, Émilie Laurin, Denis Roy, Maria C. Urdaci, Daniel St‐Gelais, Ismail Fliss, André Marette
      The FASEB Journal.2019; 33(4): 4921.     CrossRef
    • Dose-dependent and strain-dependent anti-obesity effects ofLactobacillus sakeiin a diet induced obese murine model
      Yosep Ji, Young Mee Chung, Soyoung Park, Dahye Jeong, Bongjoon Kim, Wilhelm Heinrich Holzapfel
      PeerJ.2019; 7: e6651.     CrossRef
    • Influence of a multistrain probiotic on body composition and mood in female occupational shift workers
      Abbie E. Smith-Ryan, Meredith G. Mock, Eric T. Trexler, Katie R. Hirsch, Malia N.M. Blue
      Applied Physiology, Nutrition, and Metabolism.2019; 44(7): 765.     CrossRef
    • Gut Microbiome Modulation Based on Probiotic Application for Anti-Obesity: A Review on Efficacy and Validation
      Kaliyan Barathikannan, Ramachandran Chelliah, Momna Rubab, Eric Banan-Mwine Daliri, Fazle Elahi, Dong-Hwan Kim, Paul Agastian, Seong-Yoon Oh, Deog Hwan Oh
      Microorganisms.2019; 7(10): 456.     CrossRef
    • Current Understanding of the Gut Microflora in Subjects with Nutrition-Associated Metabolic Disorder Such as Obesity and/or Diabetes: Is There Any Relevance with Oral Microflora?
      Hiromichi Yumoto, Takashi Uebanso, Takaaki Shimohata, Akira Takahashi
      Current Oral Health Reports.2019; 6(2): 100.     CrossRef
    • Probiotic Supplementation in a Clostridium difficile-Infected Gastrointestinal Model Is Associated with Restoring Metabolic Function of Microbiota
      Mohd Baasir Gaisawat, Chad W. MacPherson, Julien Tremblay, Amanda Piano, Michèle M. Iskandar, Thomas A. Tompkins, Stan Kubow
      Microorganisms.2019; 8(1): 60.     CrossRef
    • Benefaction of probiotics for human health: A review
      Rout George Kerry, Jayanta Kumar Patra, Sushanto Gouda, Yooheon Park, Han-Seung Shin, Gitishree Das
      Journal of Food and Drug Analysis.2018; 26(3): 927.     CrossRef
    • Regulation of immunometabolism in adipose tissue
      Manju Kumari, Joerg Heeren, Ludger Scheja
      Seminars in Immunopathology.2018; 40(2): 189.     CrossRef
    • Dose-Dependent Effects of Multispecies Probiotic Supplementation on the Lipopolysaccharide (LPS) Level and Cardiometabolic Profile in Obese Postmenopausal Women: A 12-Week Randomized Clinical Trial
      Monika Szulińska, Igor Łoniewski, Saskia van Hemert, Magdalena Sobieska, Paweł Bogdański
      Nutrients.2018; 10(6): 773.     CrossRef
    • Synthetic Biology Approaches to Engineer Probiotics and Members of the Human Microbiota for Biomedical Applications
      Josef R. Bober, Chase L. Beisel, Nikhil U. Nair
      Annual Review of Biomedical Engineering.2018; 20(1): 277.     CrossRef
    • The journey of gut microbiome – An introduction and its influence on metabolic disorders
      Ankita Chattopadhyay, S. Mythili
      Frontiers in Biology.2018; 13(5): 327.     CrossRef
    • Immunomodulatory Effects of Probiotics on Cytokine Profiles
      Md. Abul Kalam Azad, Manobendro Sarker, Dan Wan
      BioMed Research International.2018; 2018: 1.     CrossRef
    • Polysaccharide extracted from Enteromorpha ameliorates Cisplastin-induced small intestine injury in mice
      Xinxiu Ren, Lei Liu, Pingkun Liu, Yaser Gamallat, Yi Xin, Dong Shang
      Journal of Functional Foods.2018; 49: 154.     CrossRef
    • Implications of the Intestinal Microbiota in Diagnosing the Progression of Diabetes and the Presence of Cardiovascular Complications
      Alina Mihaela Leustean, Manuela Ciocoiu, Anca Sava, Claudia Florida Costea, Mariana Floria, Claudia Cristina Tarniceriu, Daniela Maria Tanase
      Journal of Diabetes Research.2018; 2018: 1.     CrossRef
    • Adaptation and Probiotic Potential of Lactobacilli, Isolated from the Oral Cavity and Intestines of Healthy People
      Yulia Chervinets, Vyacheslav Chervinets, Boris Shenderov, Ekaterina Belyaeva, Andrey Troshin, Sergey Lebedev, Valery Danilenko
      Probiotics and Antimicrobial Proteins.2018; 10(1): 22.     CrossRef
    • Microbiota intestinal, sobrepeso y obesidad
      Noé González-Gallegos, Yesica Sughey González-Torres, Luis Felipe Padilla-Durán
      RESPYN Revista Salud Pública y Nutrición.2017; 16(3): 23.     CrossRef
    • Effects of weight loss using supplementation with Lactobacillus strains on body fat and medium-chain acylcarnitines in overweight individuals
      Minkyung Kim, Minjoo Kim, Miso Kang, Hye Jin Yoo, Min Sun Kim, Young-Tae Ahn, Jae-Hun Sim, Sun Ha Jee, Jong Ho Lee
      Food & Function.2017; 8(1): 250.     CrossRef
    • Impact of supplementation with a food-derived microbial community on obesity-associated inflammation and gut microbiota composition
      Marianna Roselli, Chiara Devirgiliis, Paola Zinno, Barbara Guantario, Alberto Finamore, Rita Rami, Giuditta Perozzi
      Genes & Nutrition.2017;[Epub]     CrossRef
    • Heat-killed Bifidobacterium animalis subsp. Lactis CECT 8145 increases lean mass and ameliorates metabolic syndrome in cafeteria-fed obese rats
      Antoni Caimari, Josep Maria del Bas, Noemí Boqué, Anna Crescenti, Francesc Puiggròs, Empar Chenoll, Patricia Martorell, Daniel Ramón, Salvador Genovés, Lluís Arola
      Journal of Functional Foods.2017; 38: 251.     CrossRef
    • Significance of Microbiota in Obesity and Metabolic Diseases and the Modulatory Potential by Medicinal Plant and Food Ingredients
      Hoda M. Eid, Michelle L. Wright, N. V. Anil Kumar, Abdel Qawasmeh, Sherif T. S. Hassan, Andrei Mocan, Seyed M. Nabavi, Luca Rastrelli, Atanas G. Atanasov, Pierre S. Haddad
      Frontiers in Pharmacology.2017;[Epub]     CrossRef
    • Effects of a multi-strain probiotic supplement for 12 weeks in circulating endotoxin levels and cardiometabolic profiles of medication naïve T2DM patients: a randomized clinical trial
      Shaun Sabico, Ayah Al-Mashharawi, Nasser M. Al-Daghri, Sobhy Yakout, Abdullah M. Alnaami, Majed S. Alokail, Philip G. McTernan
      Journal of Translational Medicine.2017;[Epub]     CrossRef
    • Modulation of Tryptophan/Serotonin Pathway by Probiotic Supplementation in Human Immunodeficiency Virus–Positive Patients: Preliminary Results of a New Study Approach
      Giuseppe Corano Scheri, Saeid Najafi Fard, Ivan Schietroma, Andrea Mastrangelo, Claudia Pinacchio, Noemi Giustini, Sara Serafino, Gabriella De Girolamo, Eugenio Nelson Cavallari, Maura Statzu, Luca Laghi, Annamaria Vullo, Giancarlo Ceccarelli, Vincenzo Vu
      International Journal of Tryptophan Research.2017; 10: 117864691771066.     CrossRef
    • Dairy probiotics: Beyond the role of promoting gut and immune health
      Georgia Zoumpopoulou, Bruno Pot, Effie Tsakalidou, Konstantinos Papadimitriou
      International Dairy Journal.2017; 67: 46.     CrossRef
    • Microbiome and NAFLD: potential influence of aerobic fitness and lifestyle modification
      Matthew R. Panasevich, Willem T. Peppler, Daniel B. Oerther, David C. Wright, R. Scott Rector
      Physiological Genomics.2017; 49(8): 385.     CrossRef
    • Pterostilbene‐induced changes in gut microbiota composition in relation to obesity
      Usune Etxeberria, Elizabeth Hijona, Leixuri Aguirre, Fermin I. Milagro, Luis Bujanda, Agnes M. Rimando, José Alfredo Martínez, María P. Portillo
      Molecular Nutrition & Food Research.2017;[Epub]     CrossRef
    • Microbiota manipulation for weight change
      Tal Dror, Yaakov Dickstein, Grégory Dubourg, Mical Paul
      Microbial Pathogenesis.2017; 106: 146.     CrossRef
    • Targeting the gastrointestinal tract to treat type 2 diabetes
      Paige V Bauer, Frank A Duca
      Journal of Endocrinology.2016; 230(3): R95.     CrossRef
    • Novel perspectives on therapeutic modulation of the gut microbiota
      Justin L. McCarville, Alberto Caminero, Elena F. Verdu
      Therapeutic Advances in Gastroenterology.2016; 9(4): 580.     CrossRef
    • Alterations in fecal microbiota composition by probiotic supplementation in healthy adults: a systematic review of randomized controlled trials
      Nadja B. Kristensen, Thomas Bryrup, Kristine H. Allin, Trine Nielsen, Tue H. Hansen, Oluf Pedersen
      Genome Medicine.2016;[Epub]     CrossRef
    • Gut Microbiota and Obesity
      Nicole D. White
      American Journal of Lifestyle Medicine.2016; 10(2): 104.     CrossRef
    • The antioxidative effects of three lactobacilli on high-fat diet induced obese mice
      Wei Song, Chen Song, Yujuan Shan, Weihong Lu, Jiliang Zhang, Panpan Hu, Xianfan Wu, L. Li, Shouli Guo
      RSC Advances.2016; 6(70): 65808.     CrossRef
    • Intestinal removal of free fatty acids from hosts by Lactobacilli for the treatment of obesity
      Hea‐Jong Chung, Jae G. Yu, In‐Ah Lee, Ming‐Jie Liu, Yan‐Fei Shen, Satya P. Sharma, Mohammad A. H. M. Jamal, Jun‐Hyun Yoo, Hyeon‐Jin Kim, Seong‐Tshool Hong
      FEBS Open Bio.2016; 6(1): 64.     CrossRef
    • Gnotobiotic mouse model’s contribution to understanding host–pathogen interactions
      Klara Kubelkova, Milota Benuchova, Hana Kozakova, Marek Sinkora, Zuzana Krocova, Jaroslav Pejchal, Ales Macela
      Cellular and Molecular Life Sciences.2016; 73(20): 3961.     CrossRef
    • Gut Endotoxin Leading to a Decline IN Gonadal function (GELDING) - a novel theory for the development of late onset hypogonadism in obese men
      Kelton Tremellen
      Basic and Clinical Andrology.2016;[Epub]     CrossRef
    • A typical day working in a laboratory in 2050: are microbiologists becoming chemists and serene workers?
      Daniele Sohier, Armelle Riou, Florence Postollec
      Current Opinion in Food Science.2016; 10: 1.     CrossRef
    • Nutritional advice for community patients: insights from a panel discussion
      Linda V Thomas, Gill Jenkins, Julie Belton, Suzie Clements, Ciara Jacob, Naomi Johnson, Deirdre Joy, Jennifer Low, Eileen Munson, Jessica Sheppard
      British Journal of Community Nursing.2016; 21(3): 130.     CrossRef

    • PubReader PubReader
    • Cite
      CITE
      export Copy
      Close
      Download Citation
      Download a citation file in RIS format that can be imported by all major citation management software, including EndNote, ProCite, RefWorks, and Reference Manager.

      Format:
      • RIS — For EndNote, ProCite, RefWorks, and most other reference management software
      • BibTeX — For JabRef, BibDesk, and other BibTeX-specific software
      Include:
      • Citation for the content below
      Probiotics as Complementary Treatment for Metabolic Disorders
      Diabetes Metab J. 2015;39(4):291-303.   Published online August 17, 2015
      Close
    • XML DownloadXML Download
    Figure
    Related articles

    Diabetes Metab J : Diabetes & Metabolism Journal