Hostname: page-component-76fb5796d-25wd4 Total loading time: 0 Render date: 2024-04-27T08:28:20.328Z Has data issue: false hasContentIssue false

Probiotics, prebiotics, synbiotics and insulin sensitivity

Published online by Cambridge University Press:  17 October 2017

Y. A. Kim
Affiliation:
School of Pharmacy and Medical Science and Sansom Institute for Health Research, University of South Australia, GPO Box 2471, Adelaide SA 5000, Australia
J. B. Keogh
Affiliation:
School of Pharmacy and Medical Science and Sansom Institute for Health Research, University of South Australia, GPO Box 2471, Adelaide SA 5000, Australia
P. M. Clifton*
Affiliation:
School of Pharmacy and Medical Science and Sansom Institute for Health Research, University of South Australia, GPO Box 2471, Adelaide SA 5000, Australia
*
*Corresponding author: Professor Peter Clifton, fax +61 8 8302 2389, email Peter.Clifton@unisa.edu.au
Rights & Permissions [Opens in a new window]

Abstract

Animal studies indicate that the composition of gut microbiota may be involved in the progression of insulin resistance to type 2 diabetes. Probiotics and/or prebiotics could be a promising approach to improve insulin sensitivity by favourably modifying the composition of the gut microbial community, reducing intestinal endotoxin concentrations and decreasing energy harvest. The aim of the present review was to investigate the effects of probiotics, prebiotics and synbiotics (a combination of probiotics and prebiotics) on insulin resistance in human clinical trials and to discuss the potential mechanisms whereby probiotics and prebiotics improve glucose metabolism. The anti-diabetic effects of probiotics include reducing pro-inflammatory cytokines via a NF-κB pathway, reduced intestinal permeability, and lowered oxidative stress. SCFA play a key role in glucose homeostasis through multiple potential mechanisms of action. Activation of G-protein-coupled receptors on L-cells by SCFA promotes the release of glucagon-like peptide-1 and peptide YY resulting in increased insulin and decreased glucagon secretion, and suppressed appetite. SCFA can decrease intestinal permeability and decrease circulating endotoxins, lowering inflammation and oxidative stress. SCFA may also have anti-lipolytic activities in adipocytes and improve insulin sensitivity via GLUT4 through the up-regulation of 5'-AMP-activated protein kinase signalling in muscle and liver tissues. Resistant starch and synbiotics appear to have favourable anti-diabetic effects. However, there are few human interventions. Further well-designed human clinical studies are required to develop recommendations for the prevention of type 2 diabetes with pro- and prebiotics.

Type
Review Article
Copyright
© The Authors 2017 

Introduction

The global diabetic population is rapidly growing from 382 million in 2013 to an estimated 592 million by 2035( Reference Guariguata, Whiting and Hambleton 1 ). This situation imposes a great socio-economic burden on public health( Reference Sherwin, Anderson and Buse 2 ). Type 2 diabetes mellitus (T2DM) is a chronic metabolic disorder of abnormal glucose and lipid metabolism, resulting in CVD, retinopathy, nephropathy, neuropathy, leg ulcers and gangrene( Reference Nathan 3 ). The risk factors for T2DM include obesity, age, genetics, smoking, sedentary lifestyle and hypertension( Reference Sherwin, Anderson and Buse 2 ). Recently, it has been proposed that changes in gut microbiota composition resulting from obesity could contribute to the pathogenesis of T2DM( Reference Backhed, Ding and Wang 4 Reference Ley, Turnbaugh and Klein 8 ).

Probiotics and prebiotics may exert anti-diabetic effects through changes in microbiota( Reference Yadav, Lee and Lloyd 9 Reference Tajabadi-Ebrahimi, Sharifi and Farrokhian 13 ). Beneficial modification of the gut flora by probiotic and/or prebiotic treatment could be one dietary therapy for the prevention and treatment of T2DM.

PubMed, EMBASE, Cochrane and Scopus online database were searched for human intervention studies using the following terms: probiotic OR fermented OR yogurt OR cheese OR prebiotic OR inulin OR fructo-oligosaccharide OR synbiotic OR resistant starch OR gut microbiota, PLUS glucose OR glycemic OR hyperglycemia OR insulin OR insulin sensitivity OR type 2 diabetes Plus Human trial. Reviews were also utilised to clarify the potential mechanisms by which probiotics, prebiotics and synbiotics may alter insulin sensitivity.

Gut microbiota in individuals with type 2 diabetes mellitus and obesity

Most gut micro-organisms inhabit the large intestine that contains an estimated 1011−12 bacteria per g( Reference Moreno-Indias, Cardona and Tinahones 14 ). Gut microbiota can influence host adiposity and regulate fat storage( Reference Backhed, Ding and Wang 4 , Reference Ley, Backhed and Turnbaugh 7 , Reference Jumpertz, Le and Turnbaugh 15 Reference Shavakhi, Minakari and Firouzian 17 ).

The Bacteroidetes and the Firmicutes are groups of bacteria dominant in the human gut( Reference Ley, Turnbaugh and Klein 8 ). A correlation between changes in gut microbiota composition and obesity was reported in obese human subjects( Reference Ley, Turnbaugh and Klein 8 , Reference Turnbaugh, Hamady and Yatsunenko 18 ) and ob/ob mice( Reference Turnbaugh, Ley and Mahowald 19 ), with lower microbial diversity, increased Firmicutes and decreased Bacteroidetes, and this obesity-associated gut microbiota had an increased capacity for energy harvest from the diet( Reference Turnbaugh, Ley and Mahowald 19 ). Germ-free wild-type C57BL/6J mice colonised with caecal microbiota from obese donors showed a significant increase (47 (sd 8·3) %) in body fat, compared with 27 (sd 3·6) % in mice colonised with a microbiota from lean donors over 2 weeks. During the 2 weeks, obese microbiota recipients consumed 55·4 (sd 2·5) g chow and gained 1·3 (sd 0·2) g fat, while the lean microbiota recipients consumed 54·0 (sd 1·2) g chow and gained 0·86 (sd 0·1) g fat, a difference of 2 % of total energy consumed( Reference Turnbaugh, Ley and Mahowald 19 ).

Furthermore, composition of the intestinal microbiota in adults with T2DM was different from that in non-diabetic adults. The proportion of Firmicutes, Clostridia and bifidobacteria was significantly lower in diabetic adults than in non-diabetic adults( Reference Larsen, Vogensen and van den Berg 20 , Reference Wu, Ma and Han 21 ).

Mucin-degrading bacteria such as Akkermansia muciniphila and Desulfovibrio were enriched in samples derived from T2DM patients( Reference Qin, Li and Cai 22 ). In contrast, several recent studies showed that in mice, direct administration of Akkermansia muciniphila ( Reference Everard, Belzer and Geurts 23 Reference Schneeberger, Everard and Gomez-Valades 27 ) or specific proteins isolated from the outer of membrane of Akkermansia muciniphila could prevent obesity, insulin resistance as well as atherosclerosis and a human study also showed that the abundance of Akkermansia muciniphila was associated with glucose homeostasis and body fat composition( Reference Dao, Everard and Aron-Wisnewsky 28 ).

Vrieze et al. ( Reference Vrieze, Van Nood and Holleman 29 ) investigated the effect of altering the gut microbiota on insulin sensitivity in subjects with the metabolic syndrome. Obese subjects given a small-intestinal infusion of faeces from lean donors (n 9) have shown improved peripheral insulin sensitivity after 6 weeks (median rate of glucose disappearance changed from 26·2 to 45·3 µmol/kgpermin; P<0·05), as assessed by the two-step hyperinsulinaemic–euglycaemic clamp method. These subjects also have shown a 2·5-fold increase in butyrate-producing gut microbiota, Roseburia intestinalis, compared with obese subjects reinfused with their own faeces (n 9). The faecal microbiota of obese subjects had lower microbial diversity, and contained higher amounts of Bacteroidetes and lower amounts of Clostridium cluster XIVa compared with faecal microbiota after lean donor infusion at 6 weeks( Reference Vrieze, Van Nood and Holleman 29 ).

Germ-free mice which are protected from diet-induced obesity had increased phosphorylated 5'-AMP-activated protein kinase (AMPK) in skeletal muscle and liver and increased fatty acid oxidation enzymes (acetyl CoA carboxylase; carnitine palmitoyltransferase), while germ-free knockout mice deficient in fasting-induced adipose factor (FIAF), a circulating lipoprotein lipase inhibitor, were not resistant to diet-induced obesity. Germ-free FIAF-deficient animals fed a Western diet showed decreased expression of the peroxisomal proliferator activated receptor-γ coactivator 1α (PGC1α) which is known to increase genes encoding regulators of mitochondrial fatty acid oxidation in the gastrocnemius muscle compared with germ-free FIAF+/+ littermates, while there were no differences in phosphorylated AMPK levels between both groups. Consequently, germ-free mice were protected from diet-induced obesity through increased FIAF by inducing PGClα and also through elevated AMPK activity, implicating that obese gut microbiota can be responsible for decreased fatty acid oxidation and decreased FIAF/AMPK within the adipose tissue and liver( Reference Bäckhed, Manchester and Semenkovich 30 ).

The lipopolysaccharide (LPS) endotoxins, found in the outer membrane of some species of Gram-negative bacteria (for example, Neisseria spp. and Haemophilus spp.), induce signalling by binding to Toll-like receptor-4 present on endothelial cells, macrophages and monocytes. This promotes pro-inflammatory cytokines, chemokines, adhesion molecules and reactive oxygen species. An increase in LPS has been directly associated with insulin resistance( Reference Cani, Amar and Iglesias 31 ). Cani et al. ( Reference Cani, Bibiloni and Knauf 32 ) found in a mouse model that high-fat feeding changes gut microbiota with a marked reduction in some bacteria (Lactobacillus spp. and Bacteroides–Prevotella spp.), leading to an increased intestinal permeability, and LPS absorption. This increased metabolic endotoxaemia initiates adipose tissue inflammation (plasminogen activator inhibitor-1 and IL-1 mRNA), macrophage infiltration markers (macrophage chemoattractant protein-1 (MCP-1) mRNA, F4/80 mRNA) and oxidative stress (NADPHox mRNA, visceral adipose tissue and six transmembrane protein of prostate 2 (STAMP2, known to regulate nutrient-derived and inflammatory signals coordinately for metabolic homeostasis( Reference Wellen, Fucho and Gregor 33 , Reference Waki and Tontonoz 34 ))).

Even though intestinal microbiota may play a role in the aetiology of obesity and insulin resistance, the relationship between the bacteria and these metabolic disorders remains a matter of debate and most publications merely report associations between intestinal microbial composition and metabolic disorders such as obesity and T2DM( Reference Bouter, van Raalte and Groen 35 ).

Probiotics and effects of probiotics on glucose metabolism in human interventions

According to the FAO/WHO, probiotics are ‘live microorganisms which, when administered in adequate amounts, confer a health benefit on the host’( Reference Araya, Morelli and Reid 36 ). The requirement of adequate amounts differs between countries: products must contain at least 107 colony-forming units (CFU)/g of probiotic bacteria in Japan, at least 108 CFU/g probiotic bacteria in USA and 109 CFU/g probiotic bacteria in Canada. In general, >106–108 CFU/g, or >108–1010 CFU/d of viable cells are regarded efficacious( Reference Homayoni Rad, Mehrabany and Alipoor 37 , Reference Champagne, Ross and Saarela 38 ), but the cell count levels recognised do not guarantee a health effect( Reference Champagne, Ross and Saarela 38 , Reference Reid 39 ). Moreover, it is suggested that the recommendation for CFU determination should be established using accurate and frequent assessments because the number of viable cells is reduced during production, processing and formulation( Reference Champagne, Ross and Saarela 38 ).

Some of the species are: (1) lactic acid-producing bacteria (Lactobacillus, Bifidobacterium, Streptococcus); (2) non-lactic acid-producing bacterial species (Bacillus, Propionibacterium); (3) non-pathogenic yeasts (Saccharomyces; for example, Saccharomyces boulfecesardii, a non-colonising lactic acid-producing yeast); (4) non-spore-forming and non-flagellated rod or coccobacilli( Reference Saraf, Shashikanth and Priy 40 ). Over 100 Lactobacillus and over thirty Bifidobacterium species have been identified( Reference Saraf, Shashikanth and Priy 40 ). Lactic-producing Bifidobacterium and Lactobacillus are the predominant and subdominant probiotic groups( Reference Bermudez-Brito, Plaza-Diaz and Munoz-Quezada 41 ).

Human interventions with probiotics are shown in Table 1. Ten interventions( Reference Ejtahed, Mohtadi-Nia and Homayouni-Rad 10 , Reference Hove, Brons and Faerch 12 , Reference Laitinen, Poussa and Isolauri 42 Reference Barreto, Colado Simao and Morimoto 49 ) have shown positive effects of probiotics on glucose control. Ten interventions( Reference Lindsay, Kennelly and Culliton 6 , Reference Naruszewicz, Johansson and Zapolska-Downar 50 Reference Ogawa, Kadooka and Kato 58 ) have shown no effect and two interventions have shown negative effects( Reference Ivey, Hodgson and Kerr 59 ).

Table 1 Summary of probiotic human intervention studies

CFU, colony-forming unit; C, control; PC, placebo–control; P, parallel; RD, randomised; DB, double blind; SBP, systolic blood pressure; BP, blood pressure; SB, single blind; TC, total cholesterol; LDL-C, LDL-cholesterol; HDL-C, HDL-cholesterol; HC, hypercholesterolaemia; HOMA-IR, homeostasis model assessment of insulin resistance; QUICKI, quantitative insulin sensitivity check index; ATCC, American Type Culture Collection; NGT, normal glucose tolerance; IGT, impaired glucose tolerance; T2DM, type 2 diabetes mellitus; IL-Ira, IL-1 receptor antagonist; hs-CRP, high-sensitivity C-reactive protein; HbA1c, glycated Hb; FOS, fructo-oligosaccharide; MetS, metabolic syndrome; VLDL-C, VLDL-cholesterol.

Patients with T2DM supplemented with probiotic yogurt experienced attenuated fasting glucose and glycated Hb (HbA1c) concentrations and increased erythrocyte superoxide dismutase (SOD), glutathione peroxidase (GPx) activities and total antioxidants, compared with the control group( Reference Ejtahed, Mohtadi-Nia and Homayouni-Rad 10 ). Pregnant women given a probiotic supplement (Bifidobacterium lactis Bb12 and Lactobacillus rhamnosus GG) and dietary counselling together had improved glycaemic control during and after pregnancy compared with the control/placebo group( Reference Laitinen, Poussa and Isolauri 42 ).

In sixty women with gestational diabetes, the daily supplement of a probiotic capsule, containing three viable freeze-dried strains including Bifidobacterium bifidum (2×109 CFU/g), Lactobacillus acidophilus (2×109 CFU/g) and L. casei (2×109 CFU/g), for 6 weeks showed improved insulin sensitivity as assessed by the homoeostasis model assessment for insulin resistance (HOMA-IR) and quantitative insulin sensitivity check index (QUICKI), compared with a placebo capsule (cellulose). This probiotic supplementation also lowered TAG and VLDL levels, but this study did not measure HbA1c( Reference Karamali, Dadkhah and Sadrkhanlou 48 ). Moreover, oral supplementation of L. acidophilus NCFM (progenitor of the strain being used for complete chromosome sequencing in order to identify the relationship between genetics and probiotic functionality( Reference Sanders and Klaenhammer 60 )) for 4 weeks improved insulin sensitivity as assessed by hyperinsulinaemic–euglycaemic clamp, compared with a placebo group, without changes in inflammatory markers such as TNF-α, IL-6, IL-Ira and high-sensitivity C-reactive protein( Reference Andreasen, Larsen and Pedersen-Skovsgaard 43 ).

On the other hand, one study of overweight adults( Reference Ivey, Hodgson and Kerr 59 ) has shown conflicting results in that the intake of probiotic yogurt increased HOMA-IR (P=0·038) and probiotic capsules significantly increased fasting glucose (P=0·037) with no change in HOMA-IR( Reference Ivey, Hodgson and Kerr 59 ) (n 77 for the probiotic group and n 79 for the probiotic capsule; the specific study design is shown in Table 1). The probiotics used were Lactobacillus acidophilus La5 and Bifidobacterium animalis subsp. lactis Bb12 (dose of 3·0×109 CFU/d)( Reference Ivey, Hodgson and Kerr 59 ).

A single-blinded clinical trial of thirty-four subjects with T2DM showed no effects on glycaemic control, lipid profiles and inflammatory markers between a placebo group (n 18; 1000 mg magnesium stearate) and a treatment group (n 16) who received 1500 mg probiotic capsules containing Lactobacillus acidophilus, L. bulgaricus, L. bifidum and L. casei twice daily for 6 weeks( Reference Yousefinejad, Mazloom and Dabbaghmanesh 54 ). However, this study provided no counts of probiotics used.

In summary, given the mixed results from human interventions, it is still unclear if probiotics favourably influence glucose control. More human interventions are needed with more comprehensive and dynamic measures of insulin sensitivity, as most studies did not use these. It is also required to investigate the treatment effects of specific strain(s) at different dosages and durations on insulin resistance.

Other fermented food

Kimchi, made with napa cabbage and various ingredients (garlic, red pepper powder, onion, ginger, radish, fermented fish sauce and starch syrup), is a fermented traditional Korean food. Kimchi can give health benefits due to its high nutritional value and abundant bioactive compounds including dietary fibres, minerals, amino acids, vitamins, carotenoids, glucosinolates and polyphenols. Kimchi can be improved by additional ingredients and altered fermentation conditions( Reference Patra, Das and Paramithiotis 61 ). Fermented kimchi mostly contains lactic acid-producing bacteria including Lactobacillus plantarum, Lactobacillus brevis, Pediococcus cerevisiae, Streptococcus faecalis and Leuconostoc mesenteroides, which could exert a probiotic effect( Reference Patra, Das and Paramithiotis 61 ).

Studies have shown beneficial effects of fermented kimchi on glucose metabolism in obese( Reference Kim, An and Lee 62 ) and prediabetic individuals( Reference An, Lee and Jeon 63 ).

Fermented kimchi intake for 4 weeks decreased fasting glucose, fasting insulin, total cholesterol, MCP-1, leptin and the waist:hip ratio compared with fresh kimchi in a cross-over design of twenty-two overweight and obese patients. Fresh kimchi was defined as 1 d-old kimchi and fermented kimchi was defined as 10 d-old kimchi. The number of Lactobacilli in fermented kimchi was higher than in fresh kimchi (4·3×109 (sd 1·2×109)/ml v. 1·4×107 (sd 3×106)/ml)( Reference Kim, An and Lee 62 ). The consumption of fermented kimchi for 8 weeks decreased HbA1c, fasting insulin, HOMA and increased quantitative insulin sensitivity check index (QUICKI) and β-cell function compared with before fermented kimchi intake, in twenty-two adults with prediabetes( Reference An, Lee and Jeon 63 ).

A randomised controlled clinical trial in twenty-four obese women showed that fermented kimchi intake (180 g/d) for 8 weeks altered gut microbiota composition, with a decrease in genus Blautia and an increase in Prevotella and Bacteroides, compared with fresh kimchi intake and up-regulated expression of genes related to the metabolic syndrome such as acyl-CoA synthetase long-chain family member 1 (ACSL1; involved in enhancing fatty acid degradation) and aminopeptidase N (ANPEP; involved in the regulation of pain, angiogenesis, inflammation and apoptosis)( Reference Han, Bose and Wang 64 ).

Very recently, Shin et al. ( Reference Shin, Kang and Jang 65 ) demonstrated metabolic pathways of kimchi action based on in silico modelling of published data. A total of 4351 genes were associated with kimchi metabolites. Of these, 283 genes were associated with carbohydrate metabolism. In all, 309 genes were associated with lipid metabolism and twenty-seven genes (especially GNAS, CTNNB1, EDN1, RAC1 and adenyl cyclases (ADCY1, ADCY2, ADCY5) known to act as regulators of metabolic and cardiovascular function) are directly related with CVD. Twenty-three genes (especially PTPRC, LCK, JAK3, ZAP70 and VEGFA) were related to immune diseases and twenty-five genes were related to endocrine and metabolic diseases( Reference Shin, Kang and Jang 65 ). In summary, these inconsistent findings with probiotic interventions might result from heterogeneity in probiotic strains and populations. Intervention studies should be designed with a specific group and a specific strain( Reference Panwar, Rashmi and Batish 66 ).

Potential mechanisms of action of probiotics

One potential mechanism of anti-diabetic effects is that certain probiotics facilitate production of SCFA (acetate (C2), propionate (C3) and butyrate (C4)), leading to the secretion of incretin hormones which may influence glucose levels( Reference Yadav, Lee and Lloyd 9 , Reference Belenguer, Duncan and Calder 67 ). Yadav et al. ( Reference Yadav, Lee and Lloyd 9 ) have demonstrated a potential mechanism of probiotics through butyrate-induced secretion of glucagon-like peptide 1 (GLP-1) in mouse models. In this study( Reference Yadav, Lee and Lloyd 9 ), VSL#3 consisting of Lactobacillus casei, L. plantarum, L. acidophilus and L. delbrueckii subsp. bulgaricus, Bifidobacterium longum, B. breve and B. infantis and Streptococcus salivarius subsp. thermophilus was used. The levels of the SCFA butyrate in the mouse faecal samples significantly increased after VSL#3 (daily oral dose of 5 mg/kg body weight), as measured by liquid chromatography–electrospray ionisation–tandem MS. Significantly increased plasma butyrate levels were observed in VSL#3-treated mice compared with PBS-treated control mice. For the measurement of butyrate-producing bacteria, gene expression of butyrate kinase was assayed after 2 and 4 weeks of oral administration. Gene expression of butyrate kinase increased at 2 weeks in VSL#3-treated mice. An increase in GLP-1 was observed in the human intestinal L-cell line NCI-H716 treated with butyrate( Reference Yadav, Lee and Lloyd 9 ). GLP-1, an incretin hormone secreted by L-cells mainly in the ileum and large intestine, increases insulin secretion while glucagon is suppressed. GLP1 secretion results in delayed gastric emptying and reduced appetite, food intake and body-weight gain( Reference Drucker and Nauck 68 ). Lactic acid produced by lactic acid-producing bacteria can be converted to acetate or propionate by Clostridium propionicum, Propionibacterium ssp., Desulfovibrio ssp, Veillonella ssp. and Selenomonas ssp.( Reference Seeliger, Janssen and Schink 69 ) via methylmalonyl-CoA or acrylyl-CoA, and then to butyrate via acetyl-CoA by Eubacterium hallii (butyrate-producing species)( Reference Belenguer, Duncan and Calder 67 ).

Other potential mechanisms of anti-diabetic effects of probiotics could be associated with enhanced immunity and increased anti-inflammatory cytokine production, reduced intestinal permeability and reduced oxidative stress( Reference Ejtahed, Mohtadi-Nia and Homayouni-Rad 10 , Reference Yadav, Jain and Sinha 11 , Reference Ma, Forsythe and Bienenstock 70 , Reference Paszti-Gere, Szeker and Csibrik-Nemeth 71 ). In a randomised, double-blind, controlled intervention, patients with T2DM consumed 300 g/d of yogurt (L. acidophilus La5, B. lactis Bb12 with a dose of 3·98×109 CFU) for 6 weeks and experienced a reduction in fasting glucose and HbA1c and an increase in GPx and erythrocyte SOD activities and total antioxidant status, compared with the control (300 g/d of conventional yogurt). GPx and SOD are scavengers of reactive oxygen species( Reference Ejtahed, Mohtadi-Nia and Homayouni-Rad 10 ).

Pre-incubation of HeLa cells with live Lactobacillus reuteri cells for 1–2 h inhibited translocation of NF-κB to the nucleus, inhibited degradation of IKKB (inhibitor of NF-κB kinase subunit β) and prevented expression of pro-inflammatory cytokines under NF-κB regulation. Live L. reuteri up-regulated nerve growth factor and inhibited constitutive synthesis and secretion of IL-8 induced by TNF-α in T84 and HT29 cells (human colonic adenocarcinoma)( Reference Ma, Forsythe and Bienenstock 70 ). Nerve growth factor is known to play roles in the regulation of inflammation( Reference Lambiase, Bracci-Laudiero and Bonini 72 , Reference Ma, Wolvers and Stanisz 73 ) and proliferation of pancreatic β-cells( Reference Pierucci, Cicconi and Bonini 74 ). Metabolites of Lactobacillus plantarum 2142 down-regulated peroxide-induced elevation in proinflammatory cytokines IL-8 and TNF-α in the IPEC-J2 cell line (jejunal epithelia isolated from neonatal piglet)( Reference Paszti-Gere, Szeker and Csibrik-Nemeth 71 ). In streptozotocin-induced diabetic rats, probiotic dahi containing Lactobacillus casei and Lactobacillus acidophilus suppressed streptozotocin-induced oxidative stress in pancreatic tissues by preventing the depletion of glutathione, GPx and SOD, as well as by decreasing thiobarbituric acid-reactive substances and nitrite( Reference Yadav, Jain and Sinha 11 ). This finding implicates that probiotic dahi could delay streptozotocin-induced alteration in glucose homeostasis by exerting an antioxidant effect on β-cells( Reference Yadav, Jain and Sinha 11 ).

Prebiotics and effects of prebiotics on glucose metabolism in human interventions

Prebiotics are non-digestible food ingredients that are not metabolised or absorbed while passing through the upper gastrointestinal tract and are fermented by bacteria in the colon and selectively enhance the growth and/or activity of one or more potential beneficial bacteria (for example, Bifidobacterium and Lactobacillus) in the digestive system( Reference Yoo and Kim 75 Reference Flesch, Poziomyck and Damin 77 ).

Food sources of prebiotics are seeds, whole grains, legumes, chicory roots, Jerusalem artichokes, onions, garlic and some vegetables. Some prebiotics can be produced during the process of enzymic action or alcohol or cooking( Reference Flesch, Poziomyck and Damin 77 ).

Prebiotics include fructo-oligosaccharides (FOS), galacto-oligosaccharides, lactulose and large polysaccharides (inulin, resistant starches, cellulose, hemicellulose, pectin and gum)( Reference Yoo and Kim 75 ). Of these, researchers have given more attention to FOS( Reference Flesch, Poziomyck and Damin 77 ). Synthetic oligosaccharides such as galacto-oligosaccharies have shown better effects and fewer side effects than natural forms( Reference Flesch, Poziomyck and Damin 77 ). Oligofructose-enriched inulin can act across the whole colon. Oligofructose is a short-chain fructan (a polymer of fructose molecules) containing three to ten monosaccharides linked together. It is quickly fermented and completely metabolised in the ascending part of the colon, whereas inulin is a long-chain fructan containing nine to sixty-four monosaccharides linked together. It is fermented and metabolised in the descending colon( Reference Flesch, Poziomyck and Damin 77 , Reference Dehghan, Pourghassem Gargari and Asghari Jafar-abadi 78 ).

Inulin-type fructans of 10–20 g/d can normalise glucose tolerance or lipid profiles( Reference Sanz and Santacruz 79 Reference Letexier, Diraison and Beylot 84 ). FOS or inulin of 4 g/d is the minimal requirement for the enhancement of bifidobacteria growth but 14 g/d or more of inulin can cause intestinal discomfort( Reference Flesch, Poziomyck and Damin 77 ).

In various animal studies, prebiotics have shown improved glucose metabolism( Reference Kok, Morgan and Williams 85 , Reference Everard, Lazarevic and Derrien 86 ). However, a few human studies have demonstrated inconsistent findings. Human interventions of prebiotics are shown in Table 2.

Table 2 Summary of prebiotic human intervention studies

CO, cross-over; TC, total cholesterol; RD, randomised; DB, double blind; PC, placebo–control; P, parallel; HbA1c, glycated Hb; HOMA-IR, homeostasis model assessment of insulin resistance; PMR, partial meal replacement; C, control; L, longitudinal; HDL-C, HDL-cholesterol; HC, hypercholesterolaemia; GLP-1, glucagon-like peptide-1; PYY, peptide YY; FOS, fructo-oligosaccharide; LDL-C, LDL-cholesterol; OGTT, oral glucose tolerance test; LPS, lipopolysaccharide; hs-CRP, high-sensitivity C-reactive protein; T2DM, type 2 diabetes mellitus; SB, single blind; sc-FOS, short-chain fructo-oligosaccharide; AST, aspartate aminotransferase; GIP, glucose-dependent insulinotropic peptide; ALT, alanine aminotransferase; GGT, γ-glutamyltranspeptidase; RS, resistant starch; MTT, meal tolerance test; HAM-RS2, high-amylose maize type 2 resistant starch; MetS, metabolic syndrome.

Fructo-oligosaccharides

Seven studies have shown a favourable effect of FOS( Reference Yamashita, Kawai and Itakura 87 Reference Scheid, Genaro and Moreno 93 ) on glycaemic control, while three studies of FOS( Reference Alles, de Roos and Bakx 94 Reference Daud, Ismail and Thomas 96 ) have shown no effect.

Forty-eight overweight or obese adults (BMI>25 kg/m2) in a randomised, double-blind, placebo-controlled trial received 21 g oligofructose per d or a placebo (maltodextrin) for 12 weeks. FOS supplementation decreased ghrelin, glucose and insulin, and increased peptide YY (PYY) compared with a placebo( Reference Parnell and Reimer 90 ). Yamashita et al. ( Reference Yamashita, Kawai and Itakura 87 ) have also demonstrated a beneficial effect of supplementation of 8 g FOS per d for 14 d on glucose metabolism in individuals with T2DM. They found reductions in fasting glucose, total cholesterol and LDL-cholesterol. The intake of short-chain FOS of 10·6 g/d for 2 months reduced postprandial insulin response with no significant alteration in postprandial responses of glucose, NEFA and TAG in mild hypercholesterolaemic adults, compared with placebo( Reference Giacco, Clemente and Luongo 88 ). In a double-blind cross-over design, a daily consumption of 20 g FOS for 4 weeks decreased basal hepatic glucose production with no change in insulin-suppressed hepatic glucose production or insulin-stimulated glucose uptake using a hyperinsulinaemic clamp, compared with a daily consumption of 20 g sucrose in twelve healthy subjects( Reference Luo, Rizkalla and Alamowitch 89 ). However, in subjects with T2DM, supplementation of 20 g FOS had no effect on basal hepatic glucose production, fasting glucose and insulin concentrations( Reference Luo, Van Yperselle and Rizkalla 95 ), and on blood glucose and serum lipids( Reference Alles, de Roos and Bakx 94 ).

Inulin

Effects of inulin on glycaemic control have shown mixed results, with three interventions( Reference Jackson, Taylor and Clohessy 80 Reference Cani, Lecourt and Dewulf 82 ) showing a positive effect and four interventions( Reference Brighenti, Casiraghi and Canzi 83 , Reference Letexier, Diraison and Beylot 84 , Reference Tovar, Caamano Mdel and Garcia-Padilla 97 , Reference Causey, Feirtag and Gallaher 98 ) showing no effect.

In a parallel study of fifty-four subjects receiving 10 g inulin (n 27) or maltodextrin (n 27) daily for 8 weeks, insulin concentrations were lower at 4 weeks within the inulin group compared with baseline, but no differences were observed at weeks 4 and 8 in comparison with a placebo. No effect of inulin on fasting glucose concentrations was observed compared with a placebo( Reference Jackson, Taylor and Clohessy 80 ). However, in a cross-over study of twelve men with hypercholesterolaemia, a diet supplemented with 1 pint (0·4732 litres) of vanilla ice cream made with 20 g inulin for 3 weeks decreased total cholesterol and TAG but did not alter glucose and insulin, compared with the same diet supplemented with 1 pint of vanilla ice cream made with sucrose( Reference Causey, Feirtag and Gallaher 98 ).

Oligofructose-enriched inulin

Three interventions have shown favourable effects on glycaemic control when a combination of FOS and inulin was used( Reference Dehghan, Pourghassem Gargari and Asghari Jafar-abadi 78 , Reference Genta, Cabrera and Habib 92 , Reference Dewulf, Cani and Claus 99 ). In a randomised controlled study of fifty-two women with T2DM, 10 g FOS-enriched inulin per d (n 27) for 8 weeks lowered fasting glucose and glycosylated Hb and improved inflammatory markers (IL-6, TNF-α) and decreased LPS, compared with a placebo (maltodextrin; n 25)( Reference Dehghan, Pourghassem Gargari and Asghari Jafar-abadi 78 ). Further research is necessary to clarify the effects of oligofructose-enriched inulin on glucose metabolism.

Resistant starch

Consumption of resistant starch improved insulin sensitivity in healthy subjects( Reference Robertson, Bickerton and Dennis 100 Reference Robertson, Currie and Morgan 102 ) or in subjects with the metabolic syndrome( Reference Johnston, Thomas and Bell 103 , Reference Maki, Pelkman and Finocchiaro 104 ), and lowered postprandial glucose or insulin in individuals with T2DM( Reference Bodinham, Smith and Thomas 105 ) and women( Reference Behall, Scholfield and Hallfrisch 106 ). One study( Reference Nichenametla, Weidauer and Wey 107 ) showed no difference in glycaemic control. The 3 d intake of barley kernel-based bread rich in resistant starch and NSP increased fasting SCFA levels, gut hormones (fasting GLP-1, postprandial PYY and GLP-2) secretion and breath H2 excretion, and improved insulin sensitivity (the Matsuda index) after consuming a standardised breakfast, compared with white wheat bread( Reference Nilsson, Johansson-Boll and Bjorck 101 ).

In summary, the effects of prebiotics (inulin or FOS or oligofructose-enriched inulin administration) on glucose and lipid metabolism are not clear, but resistant starch appears to have a favourable effect on insulin sensitivity.

Other potential prebiotics

Costabile et al. ( Reference Costabile, Klinder and Fava 108 ) suggested that whole-grain wheat could exert a prebiotic effect on gut microbiota composition. This double-blind, randomised, cross-over trial comparing 100 % whole-grain breakfast cereal of 48 g/d with wheat bran breakfast cereal of 48 g/d for 3 weeks showed that whole grain significantly increased the number of faecal bifidobacteria and lactobacilli compared with the wheat bran cereal. However, there were no significant differences in faecal SCFA, fasting glucose, insulin, total cholesterol, TAG or HDL-cholesterol for whole grain intake compared with whole bran( Reference Costabile, Klinder and Fava 108 ).

Potential mechanisms of action of prebiotic-derived SCFA in insulin sensitivity

Microbial fermentation of prebiotics facilitates the production of SCFA (essential endproducts of carbohydrate metabolism) and enhances gut barrier function( Reference Cani, Possemiers and Van de Wiele 109 ). In a mouse model, a prebiotic treatment decreased intestinal permeability and increased GLP-2 secretion, and reducing the hepatic expression of inflammatory and oxidative stress markers and decreasing LPS during obesity and diabetes( Reference Cani, Possemiers and Van de Wiele 109 ).

SCFA and free fatty acid receptors

SCFA in the intestine activate G-protein-coupled receptors (GPR), such as GPR41 (namely, free fatty acid receptor 3; FFAR3) and GPR43 (namely, free fatty acid receptor 2; FFAR2). These receptors are present on ileal and colonic enteroendocrine L-cells, adipocytes and immune cells( Reference Woting and Blaut 110 ). Both GPR41 and GPR43 on intestinal epithelia L-cells trigger the secretion of gut hormones (GLP-1 and PYY). Leptin is also released from adipocytes when SCFA bind to GPR41. PYY, GLP1 and leptin can decrease appetite( Reference Dailey and Moran 111 Reference Meier and Gressner 114 ). GLP-1 increases insulin secretion from pancreatic β-cells and decreases glucagon secretion from the pancreatic islets, which leads to lower glucose output from the liver and enhanced peripheral uptake of glucose. GLP1 may suppress appetite and food intake via the autonomic nervous system or the brain( Reference Yoo and Kim 75 , Reference Diamant, Blaak and De Vos 115 , Reference Kaji, Karaki and Kuwahara 116 ). It is known that FFAR3 is activated by butyrate and propionate while FFAR2 is activated by acetate and propionate( Reference Layden, Angueira and Brodsky 117 ). FFAR3 knockout mice showed that butyrate and propionate inhibited food intake, reduced high-fat diet-induced weight gain and glucose intolerance and enhanced gut hormone release. FFAR3 was necessary for the maximal induction of GLP-1 by butyrate, whereas FFAR3 was unnecessary for the effects on body weight and glucose-dependent insulinotropic peptide secretion( Reference Lin, Frassetto and Kowalik 118 ). FFAR3 and FFAR2 can be expressed in several cells such as adipocytes, endocrine cells (for example, pancreatic islets) and immune cells( Reference Delzenne and Cani 119 ). FFAR2 is highly expressed in immune cells (neutrophils and monocytes) and haematopoietic tissues, compared with FFAR3( Reference Cox, Jackson and Stanton 120 ). SCFA can exert potent roles in inhibiting lipolysis and inflammation, and regulating energy metabolism( Reference Delzenne and Cani 119 , Reference Cox, Jackson and Stanton 120 Reference Zaibi, Stocker and O’Dowd 122 ). Ge et al. ( Reference Ge, Li and Weiszmann 121 ) demonstrated that when FFAR2 on adipocytes was activated by SCFA (acetate and propionate), adipocyte lipolysis and differentiation were inhibited, while in GPR43 knockout animals, this was not observed, suggesting that prebiotic fermentation could be detrimental with regard to obesity( Reference Ge, Li and Weiszmann 121 ). However, obese mice and human studies of prebiotics (especially, inulin-type fructans) have not shown this( Reference Dewulf, Cani and Neyrinck 123 , Reference Abrams, Griffin and Hawthorne 124 ). SCFA inhibited the production of MCP-1 and LPS-induced IL-10 in human monocytes, as well as LPS-induced TNF-α and interferon-γ in human peripheral blood mononuclear cells (PBMC: monocytes and lymphocytes (T-cells, B-cells and natural killer cells))( Reference Cox, Jackson and Stanton 120 ).

Anti-inflammatory effects

Elevated pro inflammatory makers such as high-sensitivity C-reactive protein, TNF-α and IL-6 are increased in T2DM( Reference Dehghan, Pourghassem Gargari and Asghari Jafar-abadi 78 ). SCFA can suppress these inflammatory mediators( Reference Cox, Jackson and Stanton 120 , Reference Vinolo, Rodrigues and Hatanaka 125 Reference Park, Lee and Lee 128 ). SCFA (acetate, propionate, and butyrate) decrease NO( Reference Vinolo, Rodrigues and Hatanaka 125 ). NO is produced by NO synthase which converts oxygen and arginine to citrulline and NO. NO acts as a vasodilator with beneficial effects on vascular health( Reference De Caterina, Libby and Peng 129 Reference Rudic, Shesely and Maeda 132 ) and has an anti-inflammatory effect under normal physiological conditions( Reference Sharma, Al-Omran and Parvathy 133 ). However, NO participates in immune responses by cytokine-activated macrophages which produce NO in high concentrations( Reference Sharma, Al-Omran and Parvathy 133 ).

SCFA suppressed LPS-stimulated TNF-α( Reference Tedelind, Westberg and Kjerrulf 126 ) from neutrophils and also suppressed TNF-α, IL-1β, IL-6 and NO in RAW 264·7 murine macrophage cells( Reference Liu, Li and Liu 127 ). Moreover, SCFA (0·2–20 mmol/l) lowered the LPS-induced production of TNF-α and interferon-γ in human PBMC in a dose-dependent manner( Reference Cox, Jackson and Stanton 120 ). Butyrate suppressed IL-6 and TNF-α in interferon-γ-stimulated RAW 264·7 murine macrophage cells( Reference Park, Lee and Lee 128 ). Studies( Reference Vinolo, Rodrigues and Hatanaka 125 , Reference Tedelind, Westberg and Kjerrulf 126 , Reference Park, Lee and Lee 128 ) showed that anti-inflammatory effects of SCFA could be mediated by inhibiting the activation of NF-κB (a transcriptional factor involved in the inflammatory response and cell proliferation and TNF-α production( Reference Usami, Kishimoto and Ohata 134 )). Butyrate is a histone deacetylase inhibitor( Reference Usami, Kishimoto and Ohata 134 , Reference Park, Joo and Pedchenko 135 ). SCFA (propionate and butyrate) suppressed the release of LPS-stimulated TNF-α and down-regulated NF-κB by facilitating PGE2 levels and cyclo-oxygenase-2 activities through inhibiting histone deacetylase in PBMC( Reference Usami, Kishimoto and Ohata 134 ) and murine macrophage cell line RAW 264·7 cells( Reference Park, Joo and Pedchenko 135 ). Therefore, SCFA, especially butyrate, exert an anti-inflammatory effect via two potential signalling pathways of NF-κB and histone deacetylase inhibition.

Butyrate also decreased the levels of MCP-1 in a dose-dependent manner with or without LPS in human PBMC( Reference Cox, Jackson and Stanton 120 ). SCFA inhibited the expression of vascular cell adhesion molecule-1 induced by TNF-α and IL-1β in human umbilical vein endothelial cells( Reference Zapolska-Downar, Siennicka and Kaczmarczyk 136 Reference Zapolska-Downar and Naruszewicz 138 ). Butyrate suppressed T-cell activation stimulated by antigen-presenting cells by down-regulating the expression of intracellular cell adhesion molecule-1 and lymphocyte function-associated antigen-3 in monocytes( Reference Bohmig, Krieger and Saemann 139 ).

SCFA and angiopoietin-like protein 4

Angiopoietin-like protein 4 (ANGPTL4) is a 50 kDa pro-hormone secreted from brown and white adipose tissues, liver, skeletal muscle, intestine and heart. Human ANGPTL4 is mainly expressed in the liver. It is known as fasting-induced adipose factor because ANGPTL4 is up-regulated in white adipose tissue and liver during fasting( Reference Woting and Blaut 110 , Reference Alex, Lichtenstein and Dijk 140 ), while human plasma ANGPTL4 concentrations are reduced after meal consumption( Reference Alex, Lichtenstein and Dijk 140 ). ANGPTL4 is a lipoprotein lipase inhibitor and thus causes decreased uptake of fatty acids into tissue( Reference Woting and Blaut 110 ). In mice, overexpression of ANGPTL4 decreased clearance of TAG-rich lipoproteins and increased circulating TAG levels( Reference Köster, Chao and Mosior 141 ). A very recent study showed that inhibition of or a lower level of ANGPTL4 is associated with lower risk of CVD in mice and non-human primate models( Reference Dewey, Gusarova and O’Dushlaine 142 ). The suppressed ANGPTL4 may result in increased lipoprotein lipase activity and lipolysis( Reference Yoo and Kim 75 , Reference Diamant, Blaak and De Vos 115 ).

On the other hand, lower serum ANGPTL4 levels are observed in subjects with T2DM compared with normal subjects. An inverse association between plasma glucose levels and HOMA-IR, and serum ANGPTL4 levels was found. These findings suggest that decreased ANGPTL4 could play a role in glucose tolerance( Reference Xu, Lam and Chan 143 ).

ANGPTL4 induced by fatty acids via PPAR in various tissues appears to reduce cellular lipid overload, oxidative stress and inflammation( Reference Lichtenstein, Mattijssen and de Wit 144 , Reference Georgiadi, Lichtenstein and Degenhardt 145 ). The SCFA, especially propionate, inhibited lipid synthesis in the presence of acetate as a source of acetyl-CoA in hepatocytes( Reference Demigné, Morand and Levrat 146 ). Propionate and/or butyrate, but not acetate, activated ANGPTL4 production in intestinal (Caco-2, HT-29 and HCT-116) and hepatic (HepG2) cancer cell lines( Reference Grootaert, Van de Wiele and Van Roosbroeck 147 ) and the entero-endocrine cell line HuTu-80 from the human small intestine( Reference Alex, Lichtenstein and Dijk 140 ).

ANGPTL4 is a downstream target gene of PPAR( Reference Yoo and Kim 75 ). PPAR, transcription factors with three isoforms (α, β and γ) are a superfamily of nuclear receptors( Reference Ferré 148 ). Fatty acids and lipid-derived substrates are their ligands. PPAR-γ agonists are used as T2DM treatment drugs. PPAR-α, present in liver, heart and skeletal muscle, promotes primarily hepatic fatty acid oxidation, ketone body synthesis and glucose sparing, while PPAR-γ, expressed in the lower intestine, adipose tissue and immunity cells, facilitates an increase in fatty acid storage in adipocytes( Reference Ferré 148 ).

Thiazolidinediones (TZD) are strong activators of PPAR-γ which improve insulin sensitivity and facilitate insulin-mediated suppression of gluconeogenesis in the liver and glucose uptake in the skeletal muscle( Reference Ferré 148 , Reference Bouskila, Pajvani and Scherer 149 ). However, PPAR-γ is expressed in adipose tissues but not in muscle, the main insulin-sensitive tissue( Reference Ferré 148 ). Activation of PPAR-γ causes release of adiponectin from mature adipocytes, which stimulates AMP involved in the up-regulation of glucose transporters (especially, GLUT4) in skeletal muscle, the stimulation of increased fatty acid oxidation in mitochondria( Reference Ferré 148 ), as well as the down-regulation of gluconeogenesis in the liver( Reference Rutter, Da Silva Xavier and Leclerc 150 ), consequently leading to improved insulin sensitivity in skeletal muscle( Reference Ferré 148 ) and in the liver( Reference Bouskila, Pajvani and Scherer 149 ). Metformin, a T2DM treatment medicine, is a stimulator of AMPK( Reference Ferré 148 ). It is suggested that the combined use of PPAR ligands (for example, TZD) and SCFA could minimise weight gain from TZD releasing ANGPTL4( Reference Korecka, De Wouters and Cultrone 151 ).

SCFA and intestinal gluconeogenesis

One potential mechanism for SCFA to prevent T2DM involves intestinal gluconeogenesis (IGN) which is mediated by signalling of the periportal nervous system( Reference De Vadder, Kovatcheva-Datchary and Goncalves 152 ). Hepatic gluconeogenesis and IGN play opposite roles in glucose homeostasis. IGN might be inversely associated with the risk of T2DM, as beneficial effects of IGN on a reduction in food intake, weight gain and hepatic glucose output, and on improvement in glycaemic control, have been shown( Reference Troy, Soty and Ribeiro 153 Reference Gautier-Stein, Zitoun and Lalli 155 ). In contrast, increased hepatic gluconeogenesis is related to the risk of T2DM( Reference DeFronzo 156 , Reference Magnusson, Rothman and Katz 157 ). The intestine produces approximately 20–25 % of total endogenous glucose in the fasted state( Reference Mithieux 158 ). Glucose produced by the intestine is sent to the portal vein. The periportal neural system in the portal vein walls detects glucose and sends a signal to the brain for the modulation of energy and glucose metabolism( Reference Mithieux 158 ). Interestingly, butyrate directly promotes IGN gene expression in enterocytes by increasing intracellular cyclic AMP levels in an FFAR2-independent manner( Reference De Vadder, Kovatcheva-Datchary and Goncalves 152 ). Propionate binding to FFAR3 present in the portal nerves increases IGN gene expression through a portal hypothalamic neural circuit( Reference De Vadder, Kovatcheva-Datchary and Goncalves 152 ). The benefit of this gut–brain neural circuits has been shown for portal glucose sensing initiated by a protein-enriched diet, resulting in decreased food intake( Reference Mithieux, Misery and Magnan 154 ).

IGN-deficient mice (with disruption of the glucose 6-phosphatase (G6Pase) catalytic subunit in the intestine) fed a SCFA- or FOS-rich diet showed no favourable effect on glucose and insulin with no change in body weight, compared with normal mice fed a SCFA- or FOS-rich diet( Reference De Vadder, Kovatcheva-Datchary and Goncalves 152 ). Moreover, normal mice fed a high-fat/high-sucrose diet supplemented with FOS showed improved glucose and insulin tolerance and decreased fat mass, whereas these metabolic benefits were absent in IGN-deficient mice fed the same diet with FOS( Reference De Vadder, Kovatcheva-Datchary and Goncalves 152 ). Therefore, IGN appears to be essential for the effect of SCFA on glucose homeostasis.

Synbiotics and effects of synbiotics on glucose metabolism in human interventions

A combination of probiotics and prebiotics is described as a synbiotic( Reference Flesch, Poziomyck and Damin 77 ). Lactobacillus acidophilus DSM20079 induced 14·5-fold more butyrate in the presence of inulin or pectin than in the presence of glucose( Reference Nazzaro, Fratianni and Nicolaus 159 ).

Human interventions of synbiotics are shown in Table 3. Eleven of twelve studies of synbiotics have shown favourable effects on glucose metabolism( Reference Tajabadi-Ebrahimi, Sharifi and Farrokhian 13 , Reference Shavakhi, Minakari and Firouzian 17 , Reference Moroti, Souza Magri and de Rezende Costa 160 Reference Mofidi, Poustchi and Yari 169 ). Three( Reference Shavakhi, Minakari and Firouzian 17 , Reference Eslamparast, Poustchi and Zamani 168 , Reference Mofidi, Poustchi and Yari 169 ) of four( Reference Shavakhi, Minakari and Firouzian 17 , Reference Wong, Won and Chim 167 Reference Mofidi, Poustchi and Yari 169 ) studies in subjects with non-alcoholic fatty liver disease have shown a positive effect on glucose control. The Asemi research group conducted a randomised, double-blind, placebo-controlled trial in subjects with T2DM; the consumption of synbiotic bread (containing the probiotic Lactobacillus sporogenes (1×108 CFU) and 0·07 g inulin per 1 g as prebiotic) for 8 weeks improved insulin metabolism, lipid profiles and plasma NO and malondialdehyde levels, compared with the probiotic alone (Lactobacillus sporogenes; 1×108 CFU) and a control bread( Reference Tajadadi-Ebrahimi, Bahmani and Shakeri 164 , Reference Bahmani, Tajadadi-Ebrahimi and Kolahdooz 170 , Reference Shakeri, Hadaegh and Abedi 171 ).

Table 3 Summary of synbiotic human intervention studies

CFU, colony-forming unit; FOS, fructo-oligosaccharide; T2DM, type 2 diabetes mellitus; RD, randomised; P, parallel; DB, double blind; PC, placebo–control; HDL-C, HDL-cholesterol; TC, total cholesterol; hs-CRP, high-sensitivity C-reactive protein; GSH, glutathione; NASH, non-alcoholic steatohepatitis; ALT, alanine aminotransferase; AST, aspartate aminotransferase; OL, open label; C, control; LDL-C, LDL-cholesterol; NAFLD, non-alcoholic fatty liver disease; HOMA-IR, homeostasis model assessment of insulin resistance; GGT, γ-glutamyltransferase; MetS, metabolic syndrome; QUICKI, quantitative insulin sensitivity check index; VLDL-C, VLDL-cholesterol; MDA, malondialdehyde; TAC, total antioxidant capacity; BP, blood pressure; CO, cross-over; SB, single blind.

In summary, a very limited number of interventions have shown beneficial effects on glucose metabolism. More pronounced effects of synbiotics on glycaemic control and inflammation have been observed than with the use of probiotics alone( Reference Jumpertz, Le and Turnbaugh 15 ).

Conclusion

Individuals with obesity or T2DM have been observed to have a different composition of gut microbiota. Altered gut microbiota may contribute to the development of T2DM. The composition of gut microbiota can be beneficially modified by probiotics and/or prebiotics to maintain glucose homeostasis. The potential mechanisms of action could involve insulinotropic and satiety effects mediated by gut hormones, GLP-1 and PYY, a β-cell-protective effect by reduced oxidative stress and lowered pro-inflammatory cytokines, anti-lipolytic activities and enhanced insulin sensitivity via GLUT4 through the up-regulation of AMPK signalling in tissues. An additional role of SCFA is in glycaemic control through IGN and mediated by the periportal nervous system. The antidiabetic effects of SCFA require further research. Use of resistant starch and synbiotics may become a diabetic nutritional strategy. Overall human interventions of probiotic and prebiotics showed mixed findings, so further work is required.

Acknowledgements

P. M. C. is supported by a National Health and Medical Research Council (NHMRC) Principal Research Fellowship, University of South Australia. Y. A. K. is supported by an Australian Postgraduate Award.

All authors conceived of the manuscript structure and contributed to the writing and editing.

The authors have no conflicts of interest related to this paper.

References

1. Guariguata, L, Whiting, DR, Hambleton, I, et al. (2014) Global estimates of diabetes prevalence for 2013 and projections for 2035. Diabetes Res Clin Pract 103, 137149.Google Scholar
2. Sherwin, RS, Anderson, RM, Buse, JB, et al. (2003) The prevention or delay of type 2 diabetes. Diabetes Care 26, S62S69.Google Scholar
3. Nathan, DM (1993) Long-term complications of diabetes mellitus. N Engl J Med 328, 16761685.CrossRefGoogle ScholarPubMed
4. Backhed, F, Ding, H, Wang, T, et al. (2004) The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A 101, 1571815723.CrossRefGoogle ScholarPubMed
5. Rabot, S, Membrez, M, Bruneau, A, et al. (2010) Germ-free C57BL/6J mice are resistant to high-fat-diet-induced insulin resistance and have altered cholesterol metabolism. FASEB J 24, 49484959.Google ScholarPubMed
6. Lindsay, KL, Kennelly, M, Culliton, M, et al. (2014) Probiotics in obese pregnancy do not reduce maternal fasting glucose: a double-blind, placebo-controlled, randomized trial (Probiotics in Pregnancy Study). Am J Clin Nutr 99, 14321439.CrossRefGoogle Scholar
7. Ley, RE, Backhed, F, Turnbaugh, P, et al. (2005) Obesity alters gut microbial ecology. Proc Natl Acad Sci U S A 102, 1107011075.CrossRefGoogle ScholarPubMed
8. Ley, RE, Turnbaugh, PJ, Klein, S, et al. (2006) Microbial ecology: human gut microbes associated with obesity. Nature 444, 10221023.CrossRefGoogle ScholarPubMed
9. Yadav, H, Lee, J-H, Lloyd, J, et al. (2013) Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion. J Biol Chem 288, 2508825097.Google Scholar
10. Ejtahed, HS, Mohtadi-Nia, J, Homayouni-Rad, A, et al. (2012) Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 28, 539543.Google Scholar
11. Yadav, H, Jain, S & Sinha, PR (2008) Oral administration of dahi containing probiotic Lactobacillus acidophilus and Lactobacillus casei delayed the progression of streptozotocin-induced diabetes in rats. J Dairy Res 75, 189195.Google Scholar
12. Hove, KD, Brons, C, Faerch, K, et al. (2015) Effects of 12 weeks of treatment with fermented milk on blood pressure, glucose metabolism and markers of cardiovascular risk in patients with type 2 diabetes: a randomised double-blind placebo-controlled study. Eur J Endocrinol 172, 1120.CrossRefGoogle ScholarPubMed
13. Tajabadi-Ebrahimi, M, Sharifi, N, Farrokhian, A, et al. (2017) A randomized controlled clinical trial investigating the effect of synbiotic administration on markers of insulin metabolism and lipid profiles in overweight type 2 diabetic patients with coronary heart disease. Exp Clin Endocrinol Diabetes 125, 2127.Google ScholarPubMed
14. Moreno-Indias, I, Cardona, F, Tinahones, FJ, et al. (2014) Impact of the gut microbiota on the development of obesity and type 2 diabetes mellitus. Front Microbiol 5, 190.Google Scholar
15. Jumpertz, R, Le, DS, Turnbaugh, PJ, et al. (2011) Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am J Clin Nutr 94, 5865.Google Scholar
16. Turnbaugh, PJ, Backhed, F, Fulton, L, et al. (2008) Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 3, 213223.Google Scholar
17. Shavakhi, A, Minakari, M, Firouzian, H, et al. (2013) Effect of a probiotic and metformin on liver aminotransferases in non-alcoholic steatohepatitis: a double blind randomized clinical trial. Int J Prev Med 4, 531537.Google ScholarPubMed
18. Turnbaugh, PJ, Hamady, M, Yatsunenko, T, et al. (2009) A core gut microbiome in obese and lean twins. Nature 457, 480484.CrossRefGoogle ScholarPubMed
19. Turnbaugh, PJ, Ley, RE, Mahowald, MA, et al. (2006) An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 444, 10271131.CrossRefGoogle ScholarPubMed
20. Larsen, N, Vogensen, FK, van den Berg, FW, et al. (2010) Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 5, e9085.Google Scholar
21. Wu, X, Ma, C, Han, L, et al. (2010) Molecular characterisation of the faecal microbiota in patients with type II diabetes. Curr Microbiol 61, 6978.Google Scholar
22. Qin, J, Li, Y, Cai, Z, et al. (2012) A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 490, 5560.Google Scholar
23. Everard, A, Belzer, C, Geurts, L, et al. (2013) Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc Natl Acad Sci U S A 110, 90669071.Google Scholar
24. Shin, NR, Lee, JC, Lee, HY, et al. (2014) An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut 63, 727735.Google Scholar
25. Org, E, Parks, BW, Joo, JW, et al. (2015) Genetic and environmental control of host–gut microbiota interactions. Genome Res 25, 15581569.CrossRefGoogle Scholar
26. Li, J, Lin, S, Vanhoutte, PM, et al. (2016) Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe -/- mice. Circulation 133, 24342446.CrossRefGoogle ScholarPubMed
27. Schneeberger, M, Everard, A, Gomez-Valades, AG, et al. (2015) Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci Rep 5, 16643.CrossRefGoogle Scholar
28. Dao, MC, Everard, A, Aron-Wisnewsky, J, et al. (2016) Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: relationship with gut microbiome richness and ecology. Gut 65, 426436.Google Scholar
29. Vrieze, A, Van Nood, E, Holleman, F, et al. (2012) Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 143, 913916.e7.Google Scholar
30. Bäckhed, F, Manchester, JK, Semenkovich, CF, et al. (2007) Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A 104, 979984.CrossRefGoogle ScholarPubMed
31. Cani, PD, Amar, J, Iglesias, MA, et al. (2007) Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 56, 17611772.Google Scholar
32. Cani, PD, Bibiloni, R, Knauf, C, et al. (2008) Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet–induced obesity and diabetes in mice. Diabetes 57, 14701481.Google Scholar
33. Wellen, KE, Fucho, R, Gregor, MF, et al. (2007) Coordinated regulation of nutrient and inflammatory responses by STAMP2 is essential for metabolic homeostasis. Cell 129, 537548.CrossRefGoogle ScholarPubMed
34. Waki, H & Tontonoz, P (2007) STAMPing out inflammation. Cell 129, 451452.CrossRefGoogle ScholarPubMed
35. Bouter, KE, van Raalte, DH, Groen, AK, et al. (2017) Role of the gut microbiome in the pathogenesis of obesity and obesity-related metabolic dysfunction. Gastroenterology 152, 16711678.Google Scholar
36. Araya, M, Morelli, L, Reid, G, et al. (2002) Guidelines for the evaluation of probiotics in food. Joint FAO/WHO Working Group Report on Drafting Guidelines for the Evaluation of Probiotics in Food, London, Ontario, Canada, April 30 and May 1, 2002. http://www.who.int/foodsafety/fs_management/en/probiotic_guidelines.pdf Google Scholar
37. Homayoni Rad, A, Mehrabany, EV, Alipoor, B, et al. (2012) Do probiotics act more efficiently in foods than in supplements? Nutrition 28, 733736.Google Scholar
38. Champagne, CP, Ross, RP, Saarela, M, et al. (2011) Recommendations for the viability assessment of probiotics as concentrated cultures and in food matrices. Int J Food Microbiol 149, 185193.Google Scholar
39. Reid, G (2008) Probiotics and prebiotics – progress and challenges. Int Dairy J 18, 969975.Google Scholar
40. Saraf, K, Shashikanth, M, Priy, T, et al. (2010) Probiotics – do they have a role in medicine and dentistry? JAPI 58, 488492.Google Scholar
41. Bermudez-Brito, M, Plaza-Diaz, J, Munoz-Quezada, S, et al. (2012) Probiotic mechanisms of action. Ann Nutr Metab 61, 160174.Google Scholar
42. Laitinen, K, Poussa, T & Isolauri, E (2009) Probiotics and dietary counselling contribute to glucose regulation during and after pregnancy: a randomised controlled trial. Br J Nutr 101, 16791687.Google Scholar
43. Andreasen, AS, Larsen, N, Pedersen-Skovsgaard, T, et al. (2010) Effects of Lactobacillus acidophilus NCFM on insulin sensitivity and the systemic inflammatory response in human subjects. Br J Nutr 104, 18311838.Google Scholar
44. Mohamadshahi, M, Veissi, M, Haidari, F, et al. (2014) Effects of probiotic yogurt consumption on inflammatory biomarkers in patients with type 2 diabetes. Bioimpacts 4, 8388.Google Scholar
45. Asemi, Z, Samimi, M, Tabassi, Z, et al. (2013) Effect of daily consumption of probiotic yoghurt on insulin resistance in pregnant women: a randomized controlled trial. Eur J Clin Nutr 67, 7174.Google ScholarPubMed
46. Rajkumar, H, Mahmood, N, Kumar, M, et al. (2014) Effect of probiotic (VSL#3) and omega-3 on lipid profile, insulin sensitivity, inflammatory markers, and gut colonization in overweight adults: a randomized, controlled trial. Mediators Inflamm 2014, 348959.Google Scholar
47. Ostadrahimi, A, Taghizadeh, A, Mobasseri, M, et al. (2015) Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: a randomized double-blind placebo-controlled clinical trial. Iran J Public Health 44, 228.Google ScholarPubMed
48. Karamali, M, Dadkhah, F, Sadrkhanlou, M, et al. (2016) Effects of probiotic supplementation on glycaemic control and lipid profiles in gestational diabetes: a randomized, double-blind, placebo-controlled trial. Diabetes Metab 42, 234241.Google Scholar
49. Barreto, FM, Colado Simao, AN, Morimoto, HK, et al. (2014) Beneficial effects of Lactobacillus plantarum on glycemia and homocysteine levels in postmenopausal women with metabolic syndrome. Nutrition 30, 939942.Google Scholar
50. Naruszewicz, M, Johansson, ML, Zapolska-Downar, D, et al. (2002) Effect of Lactobacillus plantarum 299v on cardiovascular disease risk factors in smokers. Am J Clin Nutr 76, 12491255.Google Scholar
51. Xiao, JZ, Kondo, S, Takahashi, N, et al. (2003) Effects of milk products fermented by Bifidobacterium longum on blood lipids in rats and healthy adult male volunteers. J Dairy Sci 86, 24522461.Google Scholar
52. Simons, LA, Amansec, SG & Conway, P (2006) Effect of Lactobacillus fermentum on serum lipids in subjects with elevated serum cholesterol. Nutr Metab Cardiovasc Dis 16, 531535.Google Scholar
53. Jones, ML, Martoni, CJ, Di Pietro, E, et al. (2012) Evaluation of clinical safety and tolerance of a Lactobacillus reuteri NCIMB 30242 supplement capsule: a randomized control trial. Regul Toxicol Pharmacol 63, 313320.Google Scholar
54. Yousefinejad, A, Mazloom, Z & Dabbaghmanesh, MH (2013) Effect of probiotics on lipid profile, glycemic control, insulin action, oxidative stress, and inflammatory markers in patients with type 2 diabetes: a clinical trial. Iran J Med Sci 38, 3843.Google Scholar
55. Jung, SP, Lee, KM, Kang, JH, et al. (2013) Effect of Lactobacillus gasseri BNR17 on overweight and obese adults: a randomized, double-blind clinical trial. Korean J Fam Med 34, 8089.CrossRefGoogle ScholarPubMed
56. Sharafedtinov, KK, Plotnikova, OA, Alexeeva, RI, et al. (2013) Hypocaloric diet supplemented with probiotic cheese improves body mass index and blood pressure indices of obese hypertensive patients – a randomized double-blind placebo-controlled pilot study. Nutr J 12, 138.CrossRefGoogle ScholarPubMed
57. Gøbel, RJ, Larsen, N, Jakobsen, M, et al. (2012) Probiotics to adolescents with obesity: effects on inflammation and metabolic syndrome. J Pediatr Gastroenterol Nutr 55, 673678.Google Scholar
58. Ogawa, A, Kadooka, Y, Kato, K, et al. (2014) Lactobacillus gasseri SBT2055 reduces postprandial and fasting serum non-esterified fatty acid levels in Japanese hypertriacylglycerolemic subjects. Lipids Health Dis 13, 36.Google Scholar
59. Ivey, KL, Hodgson, JM, Kerr, DA, et al. (2014) The effects of probiotic bacteria on glycaemic control in overweight men and women: a randomised controlled trial. Eur J Clin Nutr 68, 447452.Google Scholar
60. Sanders, ME & Klaenhammer, TR (2001) Invited review: the scientific basis of Lactobacillus acidophilus NCFM functionality as a probiotic. J Dairy Sci 84, 319331.Google Scholar
61. Patra, JK, Das, G, Paramithiotis, S, et al. (2016) Kimchi and other widely consumed traditional fermented foods of Korea: a review. Front Microbiol 7, 1493.CrossRefGoogle ScholarPubMed
62. Kim, EK, An, SY, Lee, MS, et al. (2011) Fermented kimchi reduces body weight and improves metabolic parameters in overweight and obese patients. Nutr Res 31, 436443.CrossRefGoogle ScholarPubMed
63. An, SY, Lee, MS, Jeon, JY, et al. (2013) Beneficial effects of fresh and fermented kimchi in prediabetic individuals. Ann Nutr Metab 63, 111119.Google Scholar
64. Han, K, Bose, S, Wang, JH, et al. (2015) Contrasting effects of fresh and fermented kimchi consumption on gut microbiota composition and gene expression related to metabolic syndrome in obese Korean women. Mol Nutr Food Res 59, 10041008.CrossRefGoogle ScholarPubMed
65. Shin, GH, Kang, BC & Jang, DJ (2016) Metabolic pathways associated with kimchi, a traditional Korean food, based on in silico modeling of published data. Genomics Inform 14, 222229.Google Scholar
66. Panwar, H, Rashmi, HM, Batish, VK, et al. (2013) Probiotics as potential biotherapeutics in the management of type 2 diabetes – prospects and perspectives. Diabetes Metab Res Rev 29, 103112.Google Scholar
67. Belenguer, A, Duncan, SH, Calder, AG, et al. (2006) Two routes of metabolic cross-feeding between Bifidobacterium adolescentis and butyrate-producing anaerobes from the human gut. Appl Environ Microbiol 72, 35933599.Google Scholar
68. Drucker, DJ & Nauck, MA (2006) The incretin system: glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 368, 16961705.CrossRefGoogle ScholarPubMed
69. Seeliger, S, Janssen, PH & Schink, B (2002) Energetics and kinetics of lactate fermentation to acetate and propionate via methylmalonyl-CoA or acrylyl-CoA. FEMS Microbiol Lett 211, 6570.CrossRefGoogle ScholarPubMed
70. Ma, D, Forsythe, P & Bienenstock, J (2004) Live Lactobacillus rhamnosus [corrected] is essential for the inhibitory effect on tumor necrosis factor α-induced interleukin-8 expression. Infect Immun 72, 53085314.CrossRefGoogle ScholarPubMed
71. Paszti-Gere, E, Szeker, K, Csibrik-Nemeth, E, et al. (2012) Metabolites of Lactobacillus plantarum 2142 prevent oxidative stress-induced overexpression of proinflammatory cytokines in IPEC-J2 cell line. Inflammation 35, 14871499.CrossRefGoogle ScholarPubMed
72. Lambiase, A, Bracci-Laudiero, L, Bonini, S, et al. (1997) Human CD4+ T cell clones produce and release nerve growth factor and express high-affinity nerve growth factor receptors. J Allergy Clin Immunol 100, 408414.Google Scholar
73. Ma, D, Wolvers, D, Stanisz, AM, et al. (2003) Interleukin-10 and nerve growth factor have reciprocal upregulatory effects on intestinal epithelial cells. Am J Physiol Regul Integr Comp Physiol 284, R1323R1329.Google Scholar
74. Pierucci, D, Cicconi, S, Bonini, P, et al. (2001) NGF-withdrawal induces apoptosis in pancreatic β cells in vitro . Diabetologia 44, 12811295.Google Scholar
75. Yoo, JY & Kim, SS (2016) Probiotics and prebiotics: present status and future perspectives on metabolic disorders. Nutrients 8, 173.Google Scholar
76. Gibson, GR, Probert, HM, Loo, JV, et al. (2004) Dietary modulation of the human colonic microbiota: updating the concept of prebiotics. Nutr Res Rev 17, 259275.Google Scholar
77. Flesch, AG, Poziomyck, AK & Damin, DC (2014) The therapeutic use of symbiotics. Arq Bras Cir Dig 27, 206209.Google Scholar
78. Dehghan, P, Pourghassem Gargari, B & Asghari Jafar-abadi, M (2014) Oligofructose-enriched inulin improves some inflammatory markers and metabolic endotoxemia in women with type 2 diabetes mellitus: a randomized controlled clinical trial. Nutrition 30, 418423.Google Scholar
79. Sanz, Y & Santacruz, A (2010) Probiotics and prebiotics in metabolic disorders and obesity. In Bioactive Foods in Promoting Health: Probiotics and Prebiotics, , pp. 237258 [RR Watson and VR Preedy, editors]. Boston, MA: Academic Press.Google Scholar
80. Jackson, KG, Taylor, GR, Clohessy, AM, et al. (1999) The effect of the daily intake of inulin on fasting lipid, insulin and glucose concentrations in middle-aged men and women. Br J Nutr 82, 2330.Google Scholar
81. Russo, F, Riezzo, G, Chiloiro, M, et al. (2010) Metabolic effects of a diet with inulin-enriched pasta in healthy young volunteers. Curr Pharm Des 16, 825831.Google Scholar
82. Cani, PD, Lecourt, E, Dewulf, EM, et al. (2009) Gut microbiota fermentation of prebiotics increases satietogenic and incretin gut peptide production with consequences for appetite sensation and glucose response after a meal. Am J Clin Nutr 90, 12361243.Google Scholar
83. Brighenti, F, Casiraghi, M, Canzi, E, et al. (1999) Effect of consumption of a ready-to-eat breakfast cereal containing inulin on the intestinal milieu and blood lipids in healthy male volunteers. Eur J Clin Nutr 53, 726733.Google Scholar
84. Letexier, D, Diraison, F & Beylot, M (2003) Addition of inulin to a moderately high-carbohydrate diet reduces hepatic lipogenesis and plasma triacylglycerol concentrations in humans. Am J Clin Nutr 77, 559564.CrossRefGoogle ScholarPubMed
85. Kok, NN, Morgan, LM, Williams, CM, et al. (1998) Insulin, glucagon-like peptide 1, glucose-dependent insulinotropic polypeptide and insulin-like growth factor I as putative mediators of the hypolipidemic effect of oligofructose in rats. J Nutr 128, 10991103.Google Scholar
86. Everard, A, Lazarevic, V, Derrien, M, et al. (2011) Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes 60, 27752786.CrossRefGoogle ScholarPubMed
87. Yamashita, K, Kawai, K & Itakura, M (1984) Effects of fructo-oligosaccharides on blood glucose and serum lipids in diabetic subjects. Nutr Res 4, 961966.Google Scholar
88. Giacco, R, Clemente, G, Luongo, D, et al. (2004) Effects of short-chain fructo-oligosaccharides on glucose and lipid metabolism in mild hypercholesterolaemic individuals. Clin Nutr 23, 331340.Google Scholar
89. Luo, J, Rizkalla, SW, Alamowitch, C, et al. (1996) Chronic consumption of short-chain fructooligosaccharides by healthy subjects decreased basal hepatic glucose production but had no effect on insulin-stimulated glucose metabolism. Am J Clin Nutr 63, 939945.Google Scholar
90. Parnell, JA & Reimer, RA (2009) Weight loss during oligofructose supplementation is associated with decreased ghrelin and increased peptide YY in overweight and obese adults. Am J Clin Nutr 89, 17511759.Google Scholar
91. Daubioul, CA, Horsmans, Y, Lambert, P, et al. (2005) Effects of oligofructose on glucose and lipid metabolism in patients with nonalcoholic steatohepatitis: results of a pilot study. Eur J Clin Nutr 59, 723726.Google Scholar
92. Genta, S, Cabrera, W, Habib, N, et al. (2009) Yacon syrup: beneficial effects on obesity and insulin resistance in humans. Clin Nutr 28, 182187.Google Scholar
93. Scheid, MM, Genaro, PS, Moreno, YM, et al. (2014) Freeze-dried powdered yacon: effects of FOS on serum glucose, lipids and intestinal transit in the elderly. Eur J Nutr 53, 14571464.Google Scholar
94. Alles, MS, de Roos, NM, Bakx, JC, et al. (1999) Consumption of fructooligosaccharides does not favorably affect blood glucose and serum lipid concentrations in patients with type 2 diabetes. Am J Clin Nutr 69, 6469.CrossRefGoogle Scholar
95. Luo, J, Van Yperselle, M, Rizkalla, SW, et al. (2000) Chronic consumption of short-chain fructooligosaccharides does not affect basal hepatic glucose production or insulin resistance in type 2 diabetics. J Nutr 130, 15721577.CrossRefGoogle ScholarPubMed
96. Daud, NM, Ismail, NA, Thomas, EL, et al. (2014) The impact of oligofructose on stimulation of gut hormones, appetite regulation and adiposity. Obesity 22, 14301438.Google Scholar
97. Tovar, AR, Caamano Mdel, C, Garcia-Padilla, S, et al. (2012) The inclusion of a partial meal replacement with or without inulin to a calorie restricted diet contributes to reach recommended intakes of micronutrients and decrease plasma triglycerides: a randomized clinical trial in obese Mexican women. Nutr J 11, 44.CrossRefGoogle ScholarPubMed
98. Causey, JL, Feirtag, JM, Gallaher, DD, et al. (2000) Effects of dietary inulin on serum lipids, blood glucose and the gastrointestinal environment in hypercholesterolemic men. Nutr Res 20, 191201.Google Scholar
99. Dewulf, EM, Cani, PD, Claus, SP, et al. (2013) Insight into the prebiotic concept: lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 62, 11121121.Google Scholar
100. Robertson, MD, Bickerton, AS, Dennis, AL, et al. (2005) Insulin-sensitizing effects of dietary resistant starch and effects on skeletal muscle and adipose tissue metabolism. Am J Clin Nutr 82, 559567.Google Scholar
101. Nilsson, AC, Johansson-Boll, EV & Bjorck, IM (2015) Increased gut hormones and insulin sensitivity index following a 3-d intervention with a barley kernel-based product: a randomised cross-over study in healthy middle-aged subjects. Br J Nutr 114, 899907.Google Scholar
102. Robertson, MD, Currie, JM, Morgan, LM, et al. (2003) Prior short-term consumption of resistant starch enhances postprandial insulin sensitivity in healthy subjects. Diabetologia 46, 659665.Google Scholar
103. Johnston, KL, Thomas, EL, Bell, JD, et al. (2010) Resistant starch improves insulin sensitivity in metabolic syndrome. Diabet Med 27, 391397.Google Scholar
104. Maki, KC, Pelkman, CL, Finocchiaro, ET, et al. (2012) Resistant starch from high-amylose maize increases insulin sensitivity in overweight and obese men. J Nutr 142, 717723.CrossRefGoogle ScholarPubMed
105. Bodinham, CL, Smith, L, Thomas, EL, et al. (2014) Efficacy of increased resistant starch consumption in human type 2 diabetes. Endocr Connect 3, 7584.Google Scholar
106. Behall, KM, Scholfield, DJ, Hallfrisch, JG, et al. (2006) Consumption of both resistant starch and β-glucan improves postprandial plasma glucose and insulin in women. Diabetes Care 29, 976981.Google Scholar
107. Nichenametla, SN, Weidauer, LA, Wey, HE, et al. (2014) Resistant starch type 4-enriched diet lowered blood cholesterols and improved body composition in a double blind controlled cross-over intervention. Mol Nutr Food Res 58, 13651369.Google Scholar
108. Costabile, A, Klinder, A, Fava, F, et al. (2008) Whole-grain wheat breakfast cereal has a prebiotic effect on the human gut microbiota: a double-blind, placebo-controlled, crossover study. Br J Nutr 99, 110120.CrossRefGoogle Scholar
109. Cani, PD, Possemiers, S, Van de Wiele, T, et al. (2009) Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 58, 10911103.CrossRefGoogle ScholarPubMed
110. Woting, A & Blaut, M (2016) The intestinal microbiota in metabolic disease. Nutrients 8, 202.Google Scholar
111. Dailey, MJ & Moran, TH (2013) Glucagon-like peptide 1 and appetite. Trends Endocrinol Metab 24, 8591.CrossRefGoogle ScholarPubMed
112. Verdich, C, Flint, A, Gutzwiller, JP, et al. (2001) A meta-analysis of the effect of glucagon-like peptide-1 (7–36) amide on ad libitum energy intake in humans. J Clin Endocrinol Metab 86, 43824389.Google ScholarPubMed
113. De Silva, A & Bloom, SR (2012) Gut hormones and appetite control: a focus on PYY and GLP-1 as therapeutic targets in obesity. Gut Liver 6, 1020.Google Scholar
114. Meier, U & Gressner, AM (2004) Endocrine regulation of energy metabolism: review of pathobiochemical and clinical chemical aspects of leptin, ghrelin, adiponectin, and resistin. Clin Chem 50, 15111525.Google Scholar
115. Diamant, M, Blaak, E & De Vos, W (2011) Do nutrient–gut–microbiota interactions play a role in human obesity, insulin resistance and type 2 diabetes? Obes Rev 12, 272281.CrossRefGoogle ScholarPubMed
116. Kaji, I, Karaki, S & Kuwahara, A (2014) Short-chain fatty acid receptor and its contribution to glucagon-like peptide-1 release. Digestion 89, 3136.Google Scholar
117. Layden, BT, Angueira, AR, Brodsky, M, et al. (2013) Short chain fatty acids and their receptors: new metabolic targets. Transl Res 161, 131140.Google Scholar
118. Lin, HV, Frassetto, A, Kowalik, EJ Jr, et al. (2012) Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLOS ONE 7, e35240.Google Scholar
119. Delzenne, NM & Cani, PD (2011) Interaction between obesity and the gut microbiota: relevance in nutrition. Annu Rev Nutr 31, 1531.Google Scholar
120. Cox, MA, Jackson, J, Stanton, M, et al. (2009) Short-chain fatty acids act as antiinflammatory mediators by regulating prostaglandin E2 and cytokines. World J Gastroenterol 15, 55495557.Google Scholar
121. Ge, H, Li, X, Weiszmann, J, et al. (2008) Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology 149, 45194526.Google Scholar
122. Zaibi, MS, Stocker, CJ, O’Dowd, J, et al. (2010) Roles of GPR41 and GPR43 in leptin secretory responses of murine adipocytes to short chain fatty acids. FEBS Lett 584, 23812386.Google Scholar
123. Dewulf, EM, Cani, PD, Neyrinck, AM, et al. (2011) Inulin-type fructans with prebiotic properties counteract GPR43 overexpression and PPARγ-related adipogenesis in the white adipose tissue of high-fat diet-fed mice. J Nutr Biochem 22, 712722.Google Scholar
124. Abrams, SA, Griffin, IJ, Hawthorne, KM, et al. (2007) Effect of prebiotic supplementation and calcium intake on body mass index. J Pediatr 151, 293298.Google Scholar
125. Vinolo, MA, Rodrigues, HG, Hatanaka, E, et al. (2011) Suppressive effect of short-chain fatty acids on production of proinflammatory mediators by neutrophils. J Nutr Biochem 22, 849855.CrossRefGoogle ScholarPubMed
126. Tedelind, S, Westberg, F, Kjerrulf, M, et al. (2007) Anti-inflammatory properties of the short-chain fatty acids acetate and propionate: a study with relevance to inflammatory bowel disease. World J Gastroenterol 13, 28262832.Google Scholar
127. Liu, T, Li, J, Liu, Y, et al. (2012) Short-chain fatty acids suppress lipopolysaccharide-induced production of nitric oxide and proinflammatory cytokines through inhibition of NF-κB pathway in RAW264.7 cells. Inflammation 35, 16761684.Google Scholar
128. Park, JS, Lee, EJ, Lee, JC, et al. (2007) Anti-inflammatory effects of short chain fatty acids in IFN-γ-stimulated RAW 264.7 murine macrophage cells: involvement of NF-κB and ERK signaling pathways. Int Immunopharmacol 7, 7077.Google Scholar
129. De Caterina, R, Libby, P, Peng, HB, et al. (1995) Nitric oxide decreases cytokine-induced endothelial activation. Nitric oxide selectively reduces endothelial expression of adhesion molecules and proinflammatory cytokines. J Clin Invest 96, 6068.Google Scholar
130. Moncada, S & Higgs, A (1993) The l-arginine–nitric oxide pathway. N Engl J Med 329, 20022012.Google Scholar
131. Murohara, T, Asahara, T, Silver, M, et al. (1998) Nitric oxide synthase modulates angiogenesis in response to tissue ischemia. J Clin Invest 101, 25672578.Google Scholar
132. Rudic, RD, Shesely, EG, Maeda, N, et al. (1998) Direct evidence for the importance of endothelium-derived nitric oxide in vascular remodeling. J Clin Invest 101, 731736.Google Scholar
133. Sharma, JN, Al-Omran, A & Parvathy, SS (2007) Role of nitric oxide in inflammatory diseases. Inflammopharmacology 15, 252259.Google Scholar
134. Usami, M, Kishimoto, K, Ohata, A, et al. (2008) Butyrate and trichostatin A attenuate nuclear factor κB activation and tumor necrosis factor α secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr Res 28, 321328.Google Scholar
135. Park, GY, Joo, M, Pedchenko, T, et al. (2004) Regulation of macrophage cyclooxygenase-2 gene expression by modifications of histone H3. Am J Physiol Lung Cell Mol Physiol 286, L956L962.Google Scholar
136. Zapolska-Downar, D, Siennicka, A, Kaczmarczyk, M, et al. (2004) Butyrate inhibits cytokine-induced VCAM-1 and ICAM-1 expression in cultured endothelial cells: the role of NF-κB and PPARα. J Nutr Biochem 15, 220228.Google Scholar
137. Menzel, T, Luhrs, H, Zirlik, S, et al. (2004) Butyrate inhibits leukocyte adhesion to endothelial cells via modulation of VCAM-1. Inflamm Bowel Dis 10, 122128.Google Scholar
138. Zapolska-Downar, D & Naruszewicz, M (2009) Propionate reduces the cytokine-induced VCAM-1 and ICAM-1 expression by inhibiting nuclear factor-κB (NF-κB) activation. J Physiol Pharmacol 60, 123131.Google Scholar
139. Bohmig, GA, Krieger, PM, Saemann, MD, et al. (1997) n-Butyrate downregulates the stimulatory function of peripheral blood-derived antigen-presenting cells: a potential mechanism for modulating T-cell responses by short-chain fatty acids. Immunology 92, 234243.Google Scholar
140. Alex, S, Lichtenstein, L, Dijk, W, et al. (2014) ANGPTL4 is produced by entero-endocrine cells in the human intestinal tract. Histochem Cell Biol 141, 383391.Google Scholar
141. Köster, A, Chao, YB, Mosior, M, et al. (2005) Transgenic angiopoietin-like (Angptl)4 overexpression and targeted disruption of Angptl4 and Angptl3: regulation of triglyceride metabolism. Endocrinology 146, 49434950.CrossRefGoogle ScholarPubMed
142. Dewey, FE, Gusarova, V, O’Dushlaine, C, et al. (2016) Inactivating variants in ANGPTL4 and risk of coronary artery disease. N Engl J Med 374, 11231133.Google Scholar
143. Xu, A, Lam, MC, Chan, KW, et al. (2005) Angiopoietin-like protein 4 decreases blood glucose and improves glucose tolerance but induces hyperlipidemia and hepatic steatosis in mice. Proc Natl Acad Sci U S A 102, 60866091.Google Scholar
144. Lichtenstein, L, Mattijssen, F, de Wit, NJ, et al. (2010) Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages. Cell Metab 12, 580592.Google Scholar
145. Georgiadi, A, Lichtenstein, L, Degenhardt, T, et al. (2010) Induction of cardiac Angptl4 by dietary fatty acids is mediated by peroxisome proliferator-activated receptor β/δ and protects against fatty acid-induced oxidative stress. Circ Res 106, 17121721.Google Scholar
146. Demigné, C, Morand, C, Levrat, M-A, et al. (1995) Effect of propionate on fatty acid and cholesterol synthesis and on acetate metabolism in isolated rat hepatocytes. Br J Nutr 74, 209219.Google Scholar
147. Grootaert, C, Van de Wiele, T, Van Roosbroeck, I, et al. (2011) Bacterial monocultures, propionate, butyrate and H2O2 modulate the expression, secretion and structure of the fasting-induced adipose factor in gut epithelial cell lines. Environ Microbiol 13, 17781789.Google Scholar
148. Ferré, P (2004) The biology of peroxisome proliferator-activated receptors relationship with lipid metabolism and insulin sensitivity. Diabetes 53, S43S50.Google Scholar
149. Bouskila, M, Pajvani, U & Scherer, P (2005) Adiponectin: a relevant player in PPARγ-agonist-mediated improvements in hepatic insulin sensitivity? Int J Obes 29, S17S23.Google Scholar
150. Rutter, GA, Da Silva Xavier, G & Leclerc, I (2003) Roles of 5'-AMP-activated protein kinase (AMPK) in mammalian glucose homoeostasis. Biochem J 375, 116.Google Scholar
151. Korecka, A, De Wouters, T, Cultrone, A, et al. (2013) ANGPTL4 expression induced by butyrate and rosiglitazone in human intestinal epithelial cells utilizes independent pathways. Am J Physiol Gastrointest Liver Physiol 304, G1025G1037.Google Scholar
152. De Vadder, F, Kovatcheva-Datchary, P, Goncalves, D, et al. (2014) Microbiota-generated metabolites promote metabolic benefits via gut–brain neural circuits. Cell 156, 8496.Google Scholar
153. Troy, S, Soty, M, Ribeiro, L, et al. (2008) Intestinal gluconeogenesis is a key factor for early metabolic changes after gastric bypass but not after gastric lap-band in mice. Cell Metab 8, 201211.CrossRefGoogle Scholar
154. Mithieux, G, Misery, P, Magnan, C, et al. (2005) Portal sensing of intestinal gluconeogenesis is a mechanistic link in the diminution of food intake induced by diet protein. Cell Metab 2, 321329.Google Scholar
155. Gautier-Stein, A, Zitoun, C, Lalli, E, et al. (2006) Transcriptional regulation of the glucose-6-phosphatase gene by cAMP/vasoactive intestinal peptide in the intestine. Role of HNF4α, CREM, HNF1α, and C/EBPα. J Biol Chem 281, 3126831278.Google Scholar
156. DeFronzo, R (1992) Pathogenesis of type 2 (non-insulin dependent) diabetes mellitus: a balanced overview. Diabetologia 35, 389397.CrossRefGoogle ScholarPubMed
157. Magnusson, I, Rothman, D, Katz, L, et al. (1992) Increased rate of gluconeogenesis in type II diabetes mellitus. A 13C nuclear magnetic resonance study. J Clin Invest 90, 1323.Google Scholar
158. Mithieux, G (2014) Nutrient control of energy homeostasis via gut–brain neural circuits. Neuroendocrinology 100, 8994.Google Scholar
159. Nazzaro, F, Fratianni, F, Nicolaus, B, et al. (2012) The prebiotic source influences the growth, biochemical features and survival under simulated gastrointestinal conditions of the probiotic Lactobacillus acidophilus . Anaerobe 18, 280285.Google Scholar
160. Moroti, C, Souza Magri, LF, de Rezende Costa, M, et al. (2012) Effect of the consumption of a new symbiotic shake on glycemia and cholesterol levels in elderly people with type 2 diabetes mellitus. Lipids Health Dis 11, 29.Google Scholar
161. Asemi, Z, Zare, Z, Shakeri, H, et al. (2013) Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP, and oxidative stress in patients with type 2 diabetes. Ann Nutr Metab 63, 19.Google Scholar
162. Rajkumar, H, Kumar, M, Das, N, et al. (2015) Effect of probiotic Lactobacillus salivarius UBL S22 and prebiotic fructo-oligosaccharide on serum lipids, inflammatory markers, insulin sensitivity, and gut bacteria in healthy young volunteers: a randomized controlled single-blind pilot study. J Cardiovasc Pharmacol Ther 20, 289298.CrossRefGoogle ScholarPubMed
163. Eslamparast, T, Zamani, F, Hekmatdoost, A, et al. (2014) Effects of synbiotic supplementation on insulin resistance in subjects with the metabolic syndrome: a randomised, double-blind, placebo-controlled pilot study. Br J Nutr 112, 438445.Google Scholar
164. Tajadadi-Ebrahimi, M, Bahmani, F, Shakeri, H, et al. (2014) Effects of daily consumption of synbiotic bread on insulin metabolism and serum high-sensitivity C-reactive protein among diabetic patients: a double-blind, randomized, controlled clinical trial. Ann Nutr Metab 65, 3441.Google Scholar
165. Asemi, Z, Khorrami-Rad, A, Alizadeh, SA, et al. (2014) Effects of synbiotic food consumption on metabolic status of diabetic patients: a double-blind randomized cross-over controlled clinical trial. Clin Nutr 33, 198203.Google Scholar
166. Taghizadeh, M & Asemi, Z (2014) Effects of synbiotic food consumption on glycemic status and serum hs-CRP in pregnant women: a randomized controlled clinical trial. Hormones (Athens) 13, 398406.Google Scholar
167. Wong, VW, Won, GL, Chim, AM, et al. (2013) Treatment of nonalcoholic steatohepatitis with probiotics. A proof-of-concept study. Ann Hepatol 12, 256262.Google Scholar
168. Eslamparast, T, Poustchi, H, Zamani, F, et al. (2014) Synbiotic supplementation in nonalcoholic fatty liver disease: a randomized, double-blind, placebo-controlled pilot study. Am J Clin Nutr 99, 535542.Google Scholar
169. Mofidi, F, Poustchi, H, Yari, Z, et al. (2017) Synbiotic supplementation in lean patients with non-alcoholic fatty liver disease: a pilot, randomised, double-blind, placebo-controlled, clinical trial. Br J Nutr 117, 662668.Google Scholar
170. Bahmani, F, Tajadadi-Ebrahimi, M, Kolahdooz, F, et al. (2016) The consumption of synbiotic bread containing Lactobacillus sporogenes and inulin affects nitric oxide and malondialdehyde in patients with type 2 diabetes mellitus: randomized, double-blind, placebo-controlled trial. J Am Coll Nutr 35, 506513.Google Scholar
171. Shakeri, H, Hadaegh, H, Abedi, F, et al. (2014) Consumption of synbiotic bread decreases triacylglycerol and VLDL levels while increasing HDL levels in serum from patients with type-2 diabetes. Lipids 49, 695701.Google Scholar
Figure 0

Table 1 Summary of probiotic human intervention studies

Figure 1

Table 2 Summary of prebiotic human intervention studies

Figure 2

Table 3 Summary of synbiotic human intervention studies