Open access peer-reviewed chapter

An Insight into the Changing Scenario of Gut Microbiome during Type 2 Diabetes

Written By

Alpana Mukhuty, Chandrani Fouzder, Snehasis Das and Dipanjan Chattopadhyay

Submitted: 12 June 2019 Reviewed: 28 November 2019 Published: 26 December 2019

DOI: 10.5772/intechopen.90697

From the Edited Volume

Parasitology and Microbiology Research

Edited by Gilberto Antonio Bastidas Pacheco and Asghar Ali Kamboh

Chapter metrics overview

932 Chapter Downloads

View Full Metrics

Abstract

The gut microbiome consists of bacteria, protozoans, viruses, and archaea collectively called as gut microbiota. Gut microbiome (GM) modulates a variety of physiological responses ranging from immune and inflammatory responses, neuronal signalling, gut barrier integrity and mobility, synthesis of vitamins, steroid hormones, neurotransmitters to metabolism of branched-chain aromatic amino acids, bile salts, and drugs. Type 2 diabetes mellitus (T2D) is a highly prevalent metabolic disorder that is featured by imbalance in blood glucose level, altered lipid profile, and their deleterious consequences. GM dysbiosis a major factor behind the incidence and progression of insulin resistance and is responsible for altering of intestinal barrier functions, host metabolic, and signaling pathways. The GM of type 2 diabetes (T2DM) patients is characterized by reduced levels of Firmicutes and Clostridia and an increased ratio of Bacteroidetes:Firmicutes. Endotoxemia stimulates a low-grade inflammatory response, which is known to trigger T2DM. Xenobiotics including dietary components, antibiotics, and nonsteroidal anti-inflammatory drugs strongly affect the gut microbial composition and can promote dysbiosis. However, the exact mechanisms behind the dynamics of gut microbes and their impact on host metabolism are yet to be deciphered. Interventions that can restore equilibrium in the GM have beneficial effects and can improve glycemic control.

Keywords

  • type 2 diabetes
  • inflammation
  • immune response
  • gut microbiome
  • xenobiotics

1. Introduction

Our quality of life and health status are modulated by our food habits and lifestyle. Hence several metabolic disorders and are the greatest global health issues are influenced by improper diet and lifestyle [1]. The other factors that are involved in the development of metabolic disorders and diseases are environmental factors, maternal health, and host genetic makeup. The resident microorganisms in our gastrointestinal tract are collectively collected as the gut microbiota (GM). GM consists of bacteria, fungi, Archaea, protozoa, and viruses. In case of mammals, GM comprises of four main phyla: Firmicutes (64%), Bacteroidetes (23%), Proteobacteria (8%), and Actinobacteria (3%). These phyla are important for the regulation of host metabolism and physiology [2]. The total number of both prokaryotic cells and host eukaryotic cells in the gut is approximately 100 trillion, which is three times that of the total number of human body cells [3]. Hence, our unique gut environment is considered as a functional and measurable organ [4]. However, the composition of GM varies along the gastrointestinal tract, and differs within and between individuals depending on the gestational age, mode of delivery, breastfeeding, antibiotic exposure, dietary lifestyle and nutritional status of the individual status of [5, 6]. The colonization of GM is limited in stomach and small intestine, but quite dense and diverse in the colon owing to the absence of digestive secretions, slow peristalsis, and rich nutrient supply [7]. This variety in composition of GM and its function is influenced by the consumption of improper diet, which in turn affects the health condition of the host. GM regulates the energy homeostasis, intestinal integrity and immunity against invading pathogens by participating in the digestive process and energy production, hampering pathogen colonization, and modulating the immune system; hence GM can modulate the overall health status of the host. Gut microbiome also influences an individual’s metabolic status such as calorie derived from indigestible dietary substances and storage of calories in adipose tissue, which regulates incidence of obesity in an individual. Studies from germ-free and wild type mice showed alteration in homeostasis in kidney, liver, and intestine in germ-free mice depicting the fact that GM influences whole body metabolism [8, 9, 10, 11, 12, 13]. GM also plays a vital role in vitamin production, energy harvest and storage, fermentation and absorption of undigested carbohydrates. The distribution of GM is determined by diet to a large extent as evident from individuals who follow a diet high in animal fat have dominance of Bacteroides in GM, whereas those who follow a carbohydrate-rich diet have a Prevotella dominant GM (Table 1) [14, 15, 16]. According to conventional theories the relationship between genetic and environmental factors such as high-calorie diet and lack of physical activities was considered as the major main contributor to obesity but recently GM has attracted much attention in relation to human health and disease. Recent scientific investigations have shown that GM can be considered as an important endogenous factor controlling obesity [17, 18].

Gut microbiota Facts and effects
Bifidobacteria Population reduces in high fat-fed mice gut increasing endotoxemia [14]
Bacteroidetes Population high in the gut of people consuming animal-based food rich diet [15]
Prevotella Population high in the gut of people consuming plant-based food rich diet [16]
Clostridium and Eubacterium Break down bile acid in the intestine to its secondary metabolites like deoxycholic acid and lithocholic acid. These metabolites bind to TGR5 receptor (G-protein-coupled receptor) present in the endocrine glands, adipocytes, muscles, immune organs, spinal cord and enteric nervous system, and stimulates the secretion of incretin hormone GLP-1 and insulin [19]
Lactobacillus reuteri GMNL-263 They are capable of reducing T2D markers like serum glucose, glycated hemoglobin and c-peptide in high-fructose-fed rats along with reduction in inflammatory cytokines IL-6 and TNF-α in adipose tissue and down-regulated forms of GLUT 4 and PPAR-γ [58]
Lactobacillus casei Shirota They can increase lipopolysaccharide-binding protein expression in plasma and diminishes endotoxemia [63]
Bifidobacterium animalis subsp. lactis They can restrict bacterial translocation in intestine alleviating bacteremia in early stages of T2D [64]
L. casei Zhang Oral administration can ameliorate impaired glucose tolerance in hyperinsulinemic rats induced by high-fructose [65]
Lactobacillus Oral administration is positively correlated with expression of CB2 receptor [76]
Clostridium Oral administration is negatively correlated with CB2 expression probiotics control GM through CB2 receptor expression [76]
Bifidobacterium infantis Impairs inflammation by altering the intestinal permeability [80, 81]
Bacteroidetes:Firmicutes ratio Low in GM of obese patients [112, 113]
Butyrate-producing bacteria (Roseburia species and Faecalibacterium prausnitzii) Low population in GM of T2DM patients [113]
Firmicutes (Gram-positive) and Bacteroidetes (Gram-negative) 90% of the bacterial species present in gut [15, 16]
Proteobacteria and particularly Escherichia coli High in T2D patients [113, 121]
Enterobacteriaceae Population elevated by T2D drugs [122]
Clostridium and Eubacterium Population lowered by T2D drugs [122]
Akkermansia sp.
Akkermansia muciniphila
Metformin increases the populations of Akkermansia sp. in high-fat diet-fed mice, hence improving glucose metabolism. Oral administration of Akkermansia muciniphila also improves metabolic dysfunctions like endotoxemia and adipose tissue inflammation [122]

Table 1.

Facts and effects of various types of bacteria present in GM

Advertisement

2. Host-gut microbiota metabolite interaction

Several reports have shown that the metabolites derived by GM from fermentation of food play a key role in maintenance of the host metabolism. Clostridium and Eubacterium from our GM break down bile acid in the intestine to its secondary metabolites like deoxycholic acid and lithocholic acid. These metabolites bind to Takeda G protein coupled receptor-5 TGR5 receptor (G-protein-coupled bile receptor) present in the endocrine glands, adipocytes, muscles, immune organs, spinal cord and enteric nervous system, and stimulates the secretion of incretin hormone GLP-1 and insulin. Hence these metabolites in turn promote energy expenditure (Table 1) [19]. Long chain fatty acids, for example linoleic acid produced by the GM regulates our lipid profile finally resulting in obesity [20]. Short chain fatty acids (SCFs) another secondary metabolite of gut microbial fermentation is formed by the digestion of indigestible polysaccharides and oligosaccharides that are neither digested nor absorbed in the proximal jejunum [21]. SCFs mainly acetate and propionate contributed by Bacteroidetes and butyrate produced by Firmicutes balance the host metabolism by influencing energy homeostasis, lipid accumulation and appetite [22]. SCF produced in the gastrointestinal tract are also known to control the pH of the lumen by increasing the absorption of nutrients. SCFs also act as a source of nutrition for GM due to high carbon content [23]. Butyrate is the main source of energy for colonocytes. It aids in the proliferation, maturation, maintenance of colonocytes and also protects the colon by enhancing mucin expression and immune response [24]. Acetate and propionate can cross the liver epithelium, and propionate gets metabolized in the liver, whereas acetate stays in the peripheral circulation [25]. SCF also regulates epithelial barrier integrity by maintaining the tight junction proteins like claudin-1, occludin, and Zonula Occludens-1. Suppression of these proteins leads to invasion of bacteria and lipopolysaccharides (LPS) stimulating an inflammatory response [26]. Hence SCF acts as energy source and also regulates host biological responses including inflammation, oxidative stress, and immune response toward Crohn’s disease, ulcerative colitis, and colorectal cancer [27, 28]. Host metabolism is activated by SCFs by direct stimulation of G-coupled receptors like free fatty acid receptors 2 and 3 (FFAR2/GPR41 and FFAR3/GPR41) occurring mainly in the gut epithelial cells. They also activate host metabolism by inhibiting nuclear class I histone deacetylases (HDACs) present in the epithelial cells [27]. FFAR2 acts as the receptor for acetate and FFAR3 is the receptor for butyrate and propionate. Activation of these receptors regulates the level of satiety hormones like ghrelin (orexigenic peptide), glucagon like peptide-1 (GLP-1), and peptide YY (PYY) (anorexigenic peptide) [29]. Ghrelin secretion occurs pre-meal, while GLP-1 and PYY are secreted post-meal, which in turn stimulates insulin production in the pancreatic 𝛽 cells. GLP-1 and PYY also reduce food intake, normalizes weight loss and maintain the balance of energy intake. Increase in the production of SCFs enhances the secretion of PYY and GLP-1 but decreases secretion of ghrelin, which ultimately leads to increased satiety and reduction in food intake [30]. The other factors inducing reduced appetite is mediated by butyrate and propionate by (i) enhanced expression of leptin in adipocytes, direct regulation of body weight and energy homeostasis by decreased food intake and upregulated energy expenditure [31], (ii) promoting gluconeogenesis in the intestinal cells [32] and (iii) inhibition of histone acetyltransferase and deacetylases which exhibit anti-inflammatory responses, epigenetic modification necessary for proliferation and differentiation of immune cells, activated AMP-activated protein kinase (AMPK) pathway synchronised adiponectin secretion, induction of mitochondrial biogenesis and fatty acid oxidation [33]. In healthy subjects SCF regulates integrity of gut, secretion of hormones, and immune responses, while in metabolically unhealthy subjects SCF implements protection from diabetes, ulcerative colitis, colorectal cancer, and neurodegenerative disorders [24, 34].

2.1 Gut microbiota composition

Recent studies targeting metagenomics have disclosed that approximately 90% of the bacterial species in the GM of adult humans are Bacteroidetes (Gram-negative) and Firmicutes (Gram-positive) [35, 36]. A healthy person fosters 500–1000 bacterial species at a single time and almost 1012–1014 colony-forming units (CFU) with a total mass weight of about 1–2 kg in the total gut [37] with 109–1012 CFU/ml in the colon, 101–103 CFU/ml in jejunum and 104–108 CFU/ml in the ileum [38]. Transfer of microbiota from mother to embryo takes place in utero or during birth and attains strength by the 2 years. Composition of GM is shaped by host genetics, environmental factors and early exposure to microbes during birth. The other factors that regulate formation of a stale GM are exposure to vaginal microbiome during normal delivery, skin microbiota during cesarean sections, breast-feeding and antibiotics in neonatal or early childhood.

2.2 Role of gut microbiota in carbohydrate metabolism

Normal diet of a healthy human contains a considerable percentage of carbohydrates comprising of monosaccharides, disaccharides and complex polysaccharides. The difference lies in the absorption of the sugars, for example common sugars like cane sugar and fruit sugars are readily absorbed in the intestine, disaccharides like maltose, lactose and sucrose and complex polysaccharides like pectin, starch and hemicellulose are broken down into monosaccharides in the ileum with the help of bacterial enzymes like glycosidases before being absorbed [39]. After food intake consisting of carbohydrate-rich diet, glucose levels in the blood rise, and later are strongly regulated and kept at a homeostatic level by the help of two hormones, insulin and glucagon. Carbohydrate digestion and absorption occurs in the upper digestive tract via glucose transporters called GLUTs (glucose transporters) located on the epithelial cells [40]. GLUT proteins uptake glucose into the pancreatic β-cells. Metabolization of glucose stimulates insulin secretion due to increased ATP/ADP ratio, membrane depolarization and closure of potassium channels, resulting in calcium dependant exocytosis of insulin [41].

The role of gut environment and gut associated lymphoid tissue plays a pivotal role in T2D [42]. T2D is a chronic metabolic disorder characterized by fasting serum hyperglycemia, non-responsiveness of insulin and insulin insufficiency [43]. Insulin resistance or non-responsiveness occurs in the liver and skeletal muscle cells when they undergo failure to sense insulin. Other factors in T2D are non-responsiveness or deficiency of incretins, amplified lipid catabolism, increased glucagon levels in circulation and increased salt and water renal retention [43, 44]. High-fat-diet-fed germ-free mice, wild type mice and standard diet fed mice exhibit different metabolic and immunological characters depending on diet and GM [45, 46]. Also mice belonging to same genotype and diet exhibit different metabolism of glucose depending on their GM [47].

2.3 Role of gut microbiota and its association with diet

In the earlier sections it has been discusses that our GM plays a key role in digestion and absorption of food. Increased population of Bacteroidetes lead to increase in energy production. The population of Bifidobacteria reduce in high fat-fed mice gut increasing endotoxemia. Prebiotic supplementation can restore Bifidobacteria levels in the mouse gut [48, 49]. Bacteroidetes are more widespread in the gut of people consuming animal-based food rich diet. Prevotella is prevalent in people consuming plant-based food rich diet. In case of people consuming plant-based foods, the GM produce more SCFAs and increased synthesis of amylase, glutamate and riboflavin [50, 51]. On the contrary, people consuming animal-based foods have GM modified for increased catabolic processes as for example degradation of glycans and amino acids [52]. SCFAs like butyrate, propionate and acetate along with some gases like hydrogen are produced by the breakdown of these polysaccharides, are further used in colonic fermentation and yield energy [53]. Butyrates can decrease calorie intake of an individual by inducing satiety via production of GLP-1 and gastric inhibitory peptide-1 [54]. Butyrates are also involved in maintenance of gut integrity by supplying energy for regulating the survival and proliferation of enterocytes.

Low-grade inflammation is a key pathophysiological factor behind the progression of type 2 diabetes (T2D), and incidence of hyperglycemia and insulin resistance [55]. Progression of T2D occurs along with reduced GM diversity and increased gut inflammation. Gut inflammation includes innate immune responses via toll-like receptors, (TLRs) secretion of proinflammatory cytokines and increased endotoxemia. Also during high-fat diet induced obesity, intestinal Gram-negative bacteria translocates in the circulatory system, adipose tissue and cause endotoxemia [56].

2.4 Role of probiotics upon gut microbiota

Probiotics enhance production of interleukin-10 (IL-10) an important regulatory and anti-inflammatory cytokine in diabetic mice. Increased IL-10 down-regulates proinflammatory cytokines like interferon-γ (IFN-γ) and interleukin-2 (IL-2)/interleukin 1-β (IL-1β) preventing inflammation and incidence of diabetes [56, 57]. Lactobacillus reuteri GMNL-263 reduces T2D markers like serum glucose, glycated hemoglobin and c-peptide in high-fructose-fed rats along with reduction in inflammatory cytokines interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) in adipose tissue and down-regulated forms of GLUT 4 and peroxisome proliferator activated receptor-γ (PPAR-γ) (Figure 1 and Table 1) [58]. Methodical consumption of probiotic yoghurt reduces inflammatory markers such as high-sensitivity C-reactive protein levels in pregnant women and T2D [59, 60]. Probiotic strains decrease oxidative stress in pancreatic tissue, reducing inflammation and apoptosis of pancreatic cells [61]. Probiotic strains also lessen LDL cholesterol and total cholesterol in serum by regulating lipid metabolism, reducing the risk of T2D [62]. Consumption of Lactobacillus casei Shirota increase lipopolysaccharide-binding protein expression in plasma and diminishing endotoxemia (Table 1) [63]. Bifidobacterium animalis sub sp. lactis can restrict bacterial translocation in intestine alleviating bacteremia in early stages of T2D (Table 1) [64]. Oral administration of L. casei can also ameliorate impaired glucose tolerance in hyperinsulinemic rats induced by high-fructose (Table 1) [65].

Figure 1.

Influence of gut microbiota in various physiological responses [80, 81, 83, 84, 110, 111].

2.5 Role of gut microbiota in maintaining intestinal integrity and metabolic conditions

Increased gut permeability provides the relation between high-fat diet and LPS by causing LPS entry into circulation via the portal system in T2D patients [66]. Animal model studies have provided evidence between increased intestinal permeability and progression of obesity and insulin resistance [67, 68]. Consumption of prebiotics increase gut microbiota, rectify intestinal permeability, diminish inflammation, alleviate endotoxemia and ameliorate glucose tolerance [68]. High-fat diet induce decrease in tight junction proteins regulating epithelial integrity of gut lining and gut permeability such as zonula occluden-1 (ZO-1) and occludin. Dietary fatty acids activate toll-like receptor 2 (TLR-2) and toll like receptor 4 (TLR-4) signaling pathways. TLR-4 leads to LPS translocation into intestinal capillaries and induces insulin resistance in mice [69, 70, 71]. Altered gut permeability and plasma LPS levels are related with distribution of ZO-1 and occluding and endocannabinoid (eCB) system. Gut microbes selectively modify expression of the cannabinoid receptor 1 (CB1) in colon also affecting zona occluding ZO-1 and occludin [72]. Administration of probiotics changes the gut microbiota resulting in reduced gut permeability in obese mice. Antibiotic exposure induces metabolic endotoxemia in mice fed with high-fat diet, along with increased gut permeability, secretion of proinflammatory cytokines, and incidence of diabetes and obesity (Figure 1). Modulation of the eCB system is connected with inflammation and diabetes [72, 73]. Moderation of GM controls eCB expression in gut, thereby regulating gut permeability and plasma LPS levels through the CB1 receptor [72]. Changes in the gut microbiota due to prebiotic feeding reduce gut permeability in obese mice. Modulation of gut permeability occurs through the distribution of tight junction proteins through eCB systems [55]. Activation of cannabinoid CB2 receptor and blocking of CB1 receptor improves glucose tolerance [74, 75]. Lactobacillus administration is positively correlated with expression of CB2 receptor, and Clostridium spp. is negatively correlated with CB2 expression (Table 1) [76]. Also probiotics control GM through CB2 receptor expression [77].

Advertisement

3. Gut microbiota and obesity mediated type 2 diabetes

GM has a close association with host obesity, since the increase in total body fat in wild type mice is high when compared to germ free mice consuming more food. Transplanting of cecum-derived microbiota induced an increase in body fat mass and insulin resistance, adipocyte hypertrophy, and increased level of circulating leptin and glucose [78]. Germ free mice when fed with a diet rich in fat and sugar content showed lean phenotype however wild type mice who were fed with the high sugar and high fat diet turned obese. Also the germ free mice showed enhanced insulin sensitivity, leading to improved glucose tolerance and altered cholesterol metabolism diminishing cholesterol storage and increasing cholesterol excretion via fecal route. GM alters intestinal permeability, causes endotoxemia, enhances calorie provision, stimulates endocannabinoid system (eCB), regulates lipid metabolism by increasing activity of lipoprotein lipase and lipogenesis resulting in host obesity. Lipopolysaccharides (LPS), present in the cell membrane of Gram-negative bacteria, stimulate low-grade inflammation and incidence of insulin resistance (IR). LPS reaches the circulation from gut by diffusion either by enhanced intestinal permeability or absorption after association with chylomicron [79]. LPS acts as a ligand for toll-like receptors TLR-4 occurring in immune cells, liver and adipose tissue. LPS activated TLR-4 prompts conformational changes recruiting adapter molecules like myeloid differentiation primary factor MyD88 protein, IL-1 receptor associated kinase IRAK, TNF receptor associated factor TRAF6, and NF-κB inducing kinase NIK, phosphorylating and degrading inhibitor of nuclear factor kappa B kinase IKKB, inhibitor of nuclear factor kappa light chain enhancer of activated B cells NF-κB. Activated NF-κB translocates to the nucleus triggering expression of inflammatory proteins and various pathways like janus kinase JNK, p38 microtubule associated protein kinase MAPK, and extracelluar signal regulated kinase ERK finally resulting in insulin resistance (Figure 1). Colonization of Bifidobacterium infantis can impair inflammation by altering the intestinal permeability. Excess of lipid in diet enhances exposure to free fatty acids and their derivatives, facilitates endotoxin absorption and increases plasma LPS level termed as “metabolic endotoxemia” (Table 1) [80, 81]. Interaction between endogenous lipid and cannabinoid receptor (CB1 and CB2) stimulates adenylate cyclase and MAPK, ERK, and NF-κB pathways, triggering inflammation, insulin resistance and obesity [82]. On the whole GM stimulates the eCB system, enhances intestinal permeability, triggering LPS entry into circulatory system resulting in endotoxemia. Rise in LPS, modulates the integrity of the tight junctions of the intestinal membrane increasing LPS in circulation. Therefore, GM is a complex system having both advantageous and dangerous microbes, and understanding the GM and host integration system provides a generalized idea about the function of each unit of the GM-host system [83, 84].

3.1 Gut microbiota and carbohydrate metabolism during type 2 diabetes

Bile acids affect glucose homeostasis via activation of nuclear farnesoid X receptor (FXR) and the membrane-bound G protein coupled receptor, TGR5. These receptors are expressed in liver, ileum and pancreas [85]. Some bile acids act as agonists for FXR, and others are FXR antagonists [86, 87, 88]. Known FXR agonists are CDCA, lithocholic acid, deoxycholic acid, and cholic acid [89]. The antidiabetic effects exhibited by vertical sleeve gastrectomy, bariatric surgery, occurs through FXR [90]. Also, intestinal FXR agonist treatment can improve insulin sensitivity [91]. In the ileum, activation of FXR leads to the production of fibroblast growth factor-19, a hormone that affects glucose tolerance through mechanisms that are largely independent of insulin [92, 93]. Activation of TGR5 produces glucagon-like peptide-1 (GLP-1) from ileum improves both energy and glucose homeostasis [94]. Activation of FXR in pancreas regulates insulin transport and secretion [95], and protects the islets from lipotoxicity [96]. FXR activation in liver improves insulin sensitivity in T2D patients [97]. The GM can modulate the amount and type of secondary bile acids produced via FXR and TGR5 signaling. GM enzymes such as bile salt hydrolase for deconjugation, 7-alpha dehydroxylase for dihydroxylation and 7α-hydroxysteroid dehydrogenase for epimerization of bile acids are reduced in T2D patients compared to healthy controls [98]. Bile acid concentrations in the circulation show a diurnal pattern since they increase after food intake [99].

3.2 Gut microbiota and lipid metabolism during type 2 diabetes

Our body metabolism, inflammatory processes and innate immune system are regulated by dietary lipids [100]. The dietary lipids can also act as (proinflammatory) ligands which can bind to nuclear receptors [101]. The nuclear receptors are peroxisome-proliferator-activated receptors (PPAR) and liver X receptors (LXR) which regulate metabolic and inflammatory pathways. Hence the dietary lipids can improve insulin action and down-regulate secretion of pro-inflammatory cytokines [102, 103]. Lipids can also activate G-protein coupled receptors (Gpcr) such as Gpr43 when activated by dietary-metabolite acetate lipolysis in adipocytes is decreased leading to reduced plasma-free fatty acids. Gpr43 can be considered as a potential target for regulation of lipid metabolism [104]. Inflammation and lipid accumulation are characteristic features of atherosclerosis [105]. Recent evidences provide sufficient link between atherosclerosis and GM variety [106]. Short-term antibiotic administration can alter the composition of GM which can convert dietary choline and l-carnitine to trimethylamine (TMA). TMA is later oxidized into TMAO by the action of hepatic Flavin monooxygenases [107]. Dietary choline is highly available in foods rich in lipid phosphatidylcholine, lecithin, such as in eggs, red meat, milk, poultry, liver, and fish [108]. Bile acids are key modulators of lipid and cholesterol metabolism, and they facilitate intestinal absorption and transport of nutrients, vitamins, and lipids. Production of bile occurs in the liver and 95% of bile acids are reabsorbed in the ileum. Later the bile acids are re-absorbed in liver, entering the enterohepatic circulation. GM converts primary bile salts to secondary bile salts by bile acid de-hydroxylation [109]. Bile acids can also result in the release of GLP-1 from enteroendocrine L cells via activation of Takeda G protein coupled receptor-5 (TGR5) (Figure 1). This phenomenon affects insulin secretion sensitivity [110]. Bile acids have another receptor called farnesoid X receptor (FXR) present in liver, intestine, and pancreatic beta cells [111]. Hence, bile acids improve our metabolism in the long term after bariatric surgery by enhancing intestinal hormone secretion.

3.3 Gut microbiota composition during type 2 diabetes

The GM of T2D patients exhibit low population of Firmicutes and Clostridia and high ratio of Bacteroidetes:Firmicutes (Table 1) [112, 113]. However, the GM of T2DM and obese patients are not always identical because the GM of obese patients show decreased Bacteroidetes:Firmicutes ratio [113, 114, 115, 118]. GM of T2DM patients also show low population of butyrate-producing bacteria. Short-chain fatty acids (SCFAs) like butyrate, acetate, and propionate are fermented from dietary fiber in large intestine by GM. SCFAs regulate energy metabolism, immune responses and tumorigenesis in gut. Butyrate is the energy source for colonic epithelial cells. Butyrate perpetuates intestinal integrity and thereby avert translocation of Gram-negative intestinal bacteria across the lumen of the gut. This phenomenon ultimately leads to endotoxemia triggering a low-grade inflammation during T2D [15, 113, 115].

The major risk factors behind T2D are genetic predisposition, less physical activity, fetal programming, obesity and altered GM [114, 116]. Total weight of GM in the distal gut is about 1.5 kg and it is considered as a microbial organ. The GM consists of embers from Bacteria, Archaea, Eukarya and viruses, but a large part of the population includes anaerobic bacteria. 90% of the bacterial species present in gut are grouped into the two bacterial phyla Firmicutes (Gram-positive) and Bacteroidetes (Gram-negative) (Table 1) [15, 16]. An average adult fosters a minimum of 160 bacterial species and a set of genes in the GM is obligatory for proper functioning of the GM [15]. The GM gives protection from disease causing pathogens and facilitates the immune system. GM also help in production of vitamin K and many B-vitamins like folate, vitamin B12. Metagenomic studies about sequencing of T2D patients exhibit dysbiotic GM and less butyrate-producing bacteria (Roseburia species and Faecalibacterium prausnitzii). Metabolic disorders like obesity and impaired glucose metabolism are related with an altered ratio of Firmicutes and Bacteroidetes [118, 119, 120]. Populations of Proteobacteria and particularly Escherichia coli are also high in T2D patients (Table 1) [113, 121]. Gram-negative bacteria contribute to inflammatory lipopolysaccharides (LPS) stimulating pro-inflammation, during T2D and obesity. Oral administration of metformin, a widely used drug for T2D elevates populations of Enterobacteriaceae and lowers populations of Clostridium and Eubacterium. Metformin also increases the populations of Akkermansia sp. in high-fat diet-fed mice, hence improving glucose metabolism [122]. Oral administration of Akkermansia muciniphila also improves metabolic dysfunctions like endotoxemia and adipose tissue inflammation (Table 1) [122, 123]. Hence metformin can be used as a potent drug in improvising the GM content in T2D patients, managing glucose tolerance and inflammation.

Advertisement

4. Modulation of gut microbiota to cure type 2 diabetes

4.1 Antibiotics

Antibiotics have become very popular for elimination of pathogenic bacteria. However, antibiotics are also harmful to the local population of beneficial GM. Hence excess use of antibiotics must be prevented for healthy maintenance of GM. Bacterio-therapeutic use of antibiotics in farm animals has increased increase growth and food production, but has taken a toll of their metabolic pathways [115]. Excess of usage of antibiotics in early infancy show chronic effects on GM diversity, overweight in infants, obesity in adults. For example, excess of bacterio-therapy with vancomycin has increased the incidence of obesity in adults. Even, short-term treatment with vancomycin impeded peripheral insulin sensitivity and other related metabolic syndromes affecting GM (Table 2) [115]. Hence, even short-term treatment with oral antibiotics harness intense and chronic damage to GM diversity and function.

Types of cure Effects
Antibiotics
  • Affect GM diversity

  • Overweight in infant

  • Obesity in adult

  • Vancomycin impede insulin sensitivity [115]

Prebiotics and probiotics
  • Compete with pathogenic bacteria

  • Intensify intestinal barrier by secreting some antimicrobial substances

  • Enhances immune system [15, 16, 115]

Dietary modulation
  • Increase GM ecosystem diversity

  • Enhances SCFA

  • Reduces fasting and postprandial glucose, A1C, serum cholesterol, insulin resistance, BMI, waist and hip circumferences [124]

Metformin
  • Increases levels of butyrate-producing bacteria

  • Decreases levels of Lactobacillus [125]

Fecal microbiota transplant
  • Allogenic infusion from lean donors lead to significant rise in GM diversity, enhanced levels of butyrate producing bacteria and improved insulin sensitivity [114, 115]

Bariatric surgery
  • Proteobacteria rises and Firmicutes and Bacteroides lowers

  • BMI reduces by 15–32%

  • C-reactive protein decreases

  • T2DM is attenuated [112, 115]

Table 2.

Types of treatments for T2D involving modulation of GM and their effects.

4.2 Prebiotics and probiotics

Recently prebiotics and probiotics have gained a lot of popularity among individuals as a healthy substitute for antibiotics. Prebiotics are actually indigestible carbohydrates that improve the growth and function of colonic bacteria boosting host health. Prebiotics include oligosaccharides which cannot be digested in the upper GI tract. These oligosaccharides are fermented, producing SCFAs in the colon and result in stimulation of growth of colonic. Prebiotics can be obtained from a large number of dietary elements like barley, garlic, asparagus, wheat bran and onions and both prebiotics and probiotics can be obtained from pickled and fermented foods like sauerkraut, kimchi, miso, yogurt [15, 16]. Probiotics obtained from food and supplements contain some very popular strains like bifidobacteria and lactobacilli. These bacteria alter the composition and function of GM as well as host system activity. The prebiotics and probiotics compete with pathogenic bacteria, intensify the intestinal barrier by secreting some antimicrobial substances and enhances the immune system (Table 2) [15, 16, 115].

4.3 Dietary modulation

Changes in diet plan can modulate activity of GM and host metabolism. A fat and carbohydrate restricted diet increased the ratio of Bacteroidetes to Firmicutes in obese patients with T2D [118]. Also calorie deficient diet plans or diet plans rich in high-fiber macrobiotics like complex carbohydrates, legumes, fermented products, sea salt, and green tea and free of animal protein fat, and added sugar improved dysbiosis, increased GM ecosystem diversity, and enhanced SCFA producers in T2D patients. Macrobiotic diet can more efficiently reduce fasting and postprandial glucose, A1C, serum cholesterol, insulin resistance, BMI, waist and hip circumferences than the control diet. Also macrobiotic diet could effectively reduce pro-inflammatory bacterial strains (Table 2) [124].

4.4 Metformin

Metformin, already a well-established drug for T2D, has recently been known to have bacterio-therapeutic effects on microbial composition and production of SCFA. Several recent reports have shown that metformin affects GM of T2D patients like increasing the levels of butyrate-producing bacteria. Metformin can also decrease the levels of Lactobacillus which remains high in T2D patients (Table 2) [125].

4.5 Fecal microbiota transplant

Fecal microbiota transplant, or stool transplant also called bacteriotherapy, which is the process of replacing fecal bacteria from a healthy individual into a host individual has been quite effective in restoring GM composition. Fecal microbiota transplant is used in treating recurrent Clostridium difficile colitis recharging useful bacteria in the GI tract along with usage of antibiotics. Autologous infusion is reinfusion of one’s collected feces and allogenic infusion is infusion with feces from a donor. Insulin resistant adults when autologously transplanted did not alter the GM composition but when transplanted with allogenic infusion from lean donors exhibited significant rise in GM diversity, enhanced levels of butyrate producing bacteria and improved sensitivity to insulin (Table 2) [114, 115].

4.6 Bariatric surgery

Bariatric surgery, or Roux-en-Y gastric bypass (RYGBP), is removal of a portion of stomach and re-routing the small intestine to a small stomach pouch. It is performed on people as an efficient tool to treat obesity. After bariatric surgery huge changes occur in the GM, Proteobacteria rises and Firmicutes and Bacteroides lowers, BMI reduces by 15–32%, C-reactive protein decreases and T2DM is attenuated. However, increase in some bacteria are highly significant than the normal levels in lean controls, which means these alterations are linked with GM modification, and not body weight (Table 2) [112, 117, 118].

Advertisement

5. Conclusion

The GM makes one of the largest organs in human body and remains the reason behind various metabolic disorders such as obesity, atherosclerosis, type 2 diabetes and so on. The alterations in GM is very susceptible to changes in our diet and environment which makes them vulnerable and ultimately ends in the incidence of diseases. Reversal of the GM alterations can restore the normal physiological functions and health. Hence further investigation is required in order to get a detailed scenario of the composition of various GM and their detailed function. Scrutiny of the composition of the GM and the change in their population in various metabolic disorders can create new avenues in finding out the treatment for those diseases. Deeper insights in the composition and function of GM can also provide more ideas for development of various techniques and drugs for the enhancement of the GM for better physiological responses and treatment of diseases.

Advertisement

Acknowledgments

AM is thankful to the Science & Engineering Research Board (SERB), Department of Science & Technology, Govt. of India, for her JRF fellowship (Grant No. ECR/2017/001028). DC thankful DBT for JRF. SD thanks UGC, New Delhi for SRF. The authors are thankful to Dr. Rakesh Kundu for technical assistance and constant encouragement.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Notes/thanks/other declarations

The authors thank to the Head of the Department of Zoology, for providing the assistance in their research work.

References

  1. 1. Robson AA. Preventing diet induced disease: Bioavailable nutrient-rich, low-energy-dense diets. Nutrition and Health. 2009;20(2):135-166
  2. 2. Qin J et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature. 2010;464:59-65
  3. 3. Bianconi E et al. An estimation of the number of cells in the human body. Annals of Human Biology. 2013;40:463-471
  4. 4. Fukuda S, Ohno H. Gut microbiome and metabolic diseases. Seminars in Immunopathology. 2014;36:103-114
  5. 5. Collado MC et al. Factors influencing gastrointestinal tract and microbiota immune interaction in preterm infants. Pediatric Research. 2015;77(6):726-731
  6. 6. Xu J et al. Evolution of symbiotic bacteria in the distal human intestine. PLoS Biology. 2007;5:1574-1586
  7. 7. Sekirov I et al. Gut microbiota in health and disease. Physiological Reviews. 2010;90(3):859-904
  8. 8. Zhao L, Shen J. Whole-body systems approaches for gut microbiota-targeted, preventive healthcare. Journal of Biotechnology. 2010;149(3):183-190
  9. 9. Claus SP et al. Systemic multicompartmental effects of the gut microbiome on mouse metabolic phenotypes. Molecular Systems Biology. 2008;4:219
  10. 10. Swann JR et al. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(1):4523-4530
  11. 11. Bäckhed F et al. The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences of the United States of America. 2004;101(44):15718-15723
  12. 12. Bäckhed F et al. Host-bacterial mutualism in the human intestine. Science. 2005;307(5717):1915-1920
  13. 13. Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535(7610):56-64
  14. 14. Clemente JC et al. The impact of the gut microbiota on human health: An integrative view. Cell. 2012;148(6):1258-1270
  15. 15. Barengolts E. Gut microbiota, prebiotics, probiotics and synbiotics in management of obesity and prediabetes: Review of randomized controlled trials. Endodontics: Principles and Practice. 2016;22(10):1224-1234
  16. 16. Fujimura KE et al. Role of the gut microbiota in defining human health. Expert Review of Anti Infective Therapy. 2010;8(4):435-454
  17. 17. Murphy EF et al. Composition and energy harvesting capacity of the gut microbiota: Relationship to diet, obesity and time in mouse models. Gut. 2010;59(12):1635-1642
  18. 18. Lopez-Legarrea P et al. The influence of Mediterranean, carbohydrate and high protein diets on gut microbiota composition in the treatment of obesity and associated inflammatory state. Asia Pacific Journal of Clinical Nutrition. 2014;23(3):360-368
  19. 19. Kishino S et al. Polyunsaturated fatty acid saturation by gut lactic acid bacteria affecting host lipid composition. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(44):17808-17813
  20. 20. Miyamoto J et al. A gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. E. Journal of Biological Chemistry. 2015;290(5):2902-2918
  21. 21. Blachier F et al. Effects of amino acid-derived luminal metabolites on the colonic epithelium and physiopathological consequences. Amino Acids. 2007;33(4):547-562
  22. 22. Postler TS, Ghosh S. Understanding the holobiont: How microbial metabolites affect human health and shape the immune system. Cell Metabolism. 2017;26(1):110-130
  23. 23. Macfarlane GT, Macfarlane S. Bacteria, colonic fermentation, and gastrointestinal health. Journal of AOAC International. 2012;95(1):50-60
  24. 24. van der Beek CM et al. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutrition Reviews. 2017;75(4):286-305
  25. 25. Koh A et al. From dietary fiber to host physiology: Short-chain fatty acids as key bacterialmetabolites. Cell. 2016;165(6):1332-1345
  26. 26. Wang HB et al. Butyrate enhances intestinal epithelial barrier function via upregulation of tight junction protein claudin-1 transcription. Digestive Diseases and Sciences. 2012;57(12):3126-3135
  27. 27. Tan J et al. The role of short-chain fatty acids in health and disease. Advances in Immunology. 2014;121:91-119
  28. 28. Huang W et al. Short-chain fatty acids inhibit oxidative stress and inflammation in mesangial cells induced by high glucose and lipopolysaccharide. Experimental and Clinical Endocrinology & Diabetes. 2017;125(02):98-105
  29. 29. Tolhurst G et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-proteincoupled receptor FFAR2. Diabetes. 2012;61(2):364-371
  30. 30. Li X et al. Gut microbial metabolite short-chain fatty acids and obesity. Bioscience of Microbiota, Food and Health. 2017;36(4):135-140
  31. 31. Chambers ES et al. Effects of targeted delivery of propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut. 2015;64(11):1744-1754
  32. 32. de Vadder F et al. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014;156(1-2):84-96
  33. 33. Jian H et al. Butyrate alleviates high fat diet-induced obesity through activation of adiponectinmediated pathway and stimulation of mitochondrial function in the skeletal muscle of mice. Oncotarget. 2016;7(35):56071-56082
  34. 34. Bolognini D et al. The pharmacology and function of receptors for short-chain fatty acids. Molecular Pharmacology. 2016;89(3):388-398
  35. 35. Human Microbiome Project C. A framework for human microbiome research. Nature. 2012;486:215-221
  36. 36. Human Microbiome Project C. Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207-214
  37. 37. Xu J et al. Does canine inflammatory bowel disease influence gut microbial profile and host metabolism? BMC Veterinary Research. 2016;12:114
  38. 38. Blaut M, Clavel T. Metabolic diversity of the intestinal microbiota: Implications for health and disease. Journal of Nutrition. 2007;137:751S-755S
  39. 39. Flint HJ et al. Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 2012;3:289-306
  40. 40. Arumugam M et al. Enterotypes of the human gut microbiome. Nature. 2011;473:174-180
  41. 41. Hehemann JH et al. Transfer of carbohydrate-active enzymes from marine bacteria to Japanese gut microbiota. Nature. 2010;464:908-912
  42. 42. Mason KL et al. Overview of gut immunology. Advances in Experimental Medicine and Biology. 2008;635:1-1
  43. 43. Fonseca VA. Defining and characterizing the progression of type 2 diabetes. Diabetes Care. 2009;32(2):S151-S156
  44. 44. Musso G et al. Interactions between gut microbiota and host metabolism predisposing to obesity and diabetes. Annual Review of Medicine. 2011;62:361-380
  45. 45. Wen L, Duffy A. Factors influencing the gut microbiota, inflammation, and type 2 diabetes. Journal of Nutrition. 2017;147:1468S-1475S
  46. 46. Caesar R et al. Gut derived lipopolysaccharide augments adipose macrophage accumulation but is not essential for impaired glucose or insulin tolerance in mice. Gut. 2012;61:1701-1707
  47. 47. Backhed F et al. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proceedings of the National Academy of Sciences of the United States of America. 2007;104:979-984
  48. 48. Woting A et al. Alleviation of high fat diet-induced obesity by oligofructose in gnotobiotic mice is independent of presence of Bifidobacterium longum. Molecular Food and Nutrition Research. 2015;59:2267-2278
  49. 49. Cani PD et al. 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
  50. 50. De Filippo C et al. Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proceedings of the National Academy of Sciences of the United States of America. 2010;107:14691-14696
  51. 51. Fan W et al. Impact of diet in shaping gut microbiota revealed by a comparative study in infants during the six months of life. Journal of Microbiology and Biotechnology. 2014;24:133-143
  52. 52. Yatsunenko T et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222-227
  53. 53. Nicholson JK et al. Host-gut microbiota metabolic interactions. Science. 2012;336:1262-1267
  54. 54. den Besten G et al. The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism. The Journal of Lipid Research. 2013;54:2325-2340
  55. 55. Cani PD et al. Involvement of gut microbiota in the development of low-grade inflammation and type 2 diabetes associated with obesity. Gut Microbes. 2012;3:279-288
  56. 56. Sanz Y et al. Understanding the role of gut microbes and probiotics in obesity: How far are we? Pharmacological Research. 2013;69:144-155
  57. 57. Cano PG et al. Bifidobacterium CECT 7765 improves metabolic and immunological alterations associated with obesity in high-fat diet-fed mice. Obesity (Silver Spring). 2013;21:2310-2321
  58. 58. Hsieh FC et al. Oral administration of Lactobacillus reuteri GMNL-263 improves insulin resistance and ameliorates hepatic steatosis in high fructose-fed rats. Nutrition and Metabolsim (London). 2013;10:35
  59. 59. Asemi Z et al. Effects of daily consumption of probiotic yoghurt on inflammatory factors in pregnant women: A randomized controlled trial. Pakistan Journal of Biological Sciences. 2011;14:476-482
  60. 60. Asemi Z et al. Effect of daily consumption of probiotic yoghurt on insulin resistance in pregnant women: A randomized controlled trial. European Journal of Clinical Nutrition. 2013;67:71-74
  61. 61. Ejtahed HS et al. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition. 2012;28:539-543
  62. 62. Ooi LG, Liong MT. Cholesterol-lowering effects of probiotics and prebiotics: A review of in vivo and in vitro findings. International Journal of Molecular Sciences. 2010;11:2499-2522
  63. 63. Naito E et al. Beneficial effect of oral administration of Lactobacillus casei strain Shirota on insulin resistance in diet-induced obesity mice. Journal of Applied Microbiology. 2011;110:650-657
  64. 64. Amar J et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: Molecular mechanisms and probiotic treatment. EMBO Molecular Medicine. 2011;3:559-572
  65. 65. Zhang Y et al. Probiotic Lactobacillus casei Zhang ameliorates high-fructose-induced impaired glucose tolerance in hyperinsulinemia rats. European Journal of Nutrition. 2014;53:221-232
  66. 66. Horton F et al. Increased intestinal permeability to oral chromium (51 Cr)-EDTA in human type 2 diabetes. Diabetic Medicine. 2014;31:559-563
  67. 67. Ding S, Lund PK. Role of intestinal inflammation as an early event in obesity and insulin resistance. Current Opinion in Clinical Nutrition and Metabolic Care. 2011;14:328-333
  68. 68. Xiao S et al. A gut microbiota-targeted dietary intervention for amelioration of chronic inflammation underlying metabolic syndrome. FEMS Microbiology Ecology. 2014;87:357-367
  69. 69. Zhang X et al. Modulation of gut microbiota by berberine and metformin during the treatment of high-fat diet-induced obesity in rats. Scientific Reports. 2015;5:14405
  70. 70. Saberi M et al. Hematopoietic cell-specific deletion of toll-like receptor 4 ameliorates hepatic and adipose tissue insulin resistance in high-fat-fed mice. Cell Metabolsim. 2009;10:419-429
  71. 71. Poggi M et al. C3H/HeJ mice carrying a toll-like receptor 4 mutation are protected against the development of insulin resistance in white adipose tissue in response to a high-fat diet. Diabetologia. 2007;50:1267-1276
  72. 72. Muccioli GG et al. The endocannabinoid system links gut microbiota to adipogenesis. Molecular Systems Biology. 2010;6:392
  73. 73. Scherer T, Buettner C. The dysregulation of the endocannabinoid system in diabesity—A tricky problem. Journal of Molecular Medicine (Berl). 2009;87:663-668
  74. 74. Cani PD et al. Glucose metabolism: Focus on gut microbiota, the endocannabinoid system and beyond. Diabetes & Metabolism. 2014;40:246-257
  75. 75. Bermudez-Silva FJ et al. Role of cannabinoid CB2 receptors in glucose homeostasis in rats. European Journal of Pharmacology. 2007;565:207-211
  76. 76. Aguilera M et al. Stress and antibiotics alter luminal and wall adhered microbiota and enhance the local expression of visceral sensory-related systems in mice. Neurogastroenterology and Motility. 2013;25:e515-e529
  77. 77. Rousseaux C et al. Lactobacillus acidophilus modulates intestinal pain and induces opioid and cannabinoid receptors. Nature Medicine. 2007;13:35-37
  78. 78. Wostmann BS et al. Dietary intake, energy metabolism, and excretory losses of adult male germfree Wistar rats. Laboratory Animals. 1983;33(1):46-50
  79. 79. Manco M et al. Gut microbiota, lipopolysaccharides, and innate immunity in the pathogenesis of obesity and cardiovascular risk. Endocrine Reviews. 2010;31(6):817-844
  80. 80. Caesar R et al. Crosstalk between gut microbiota and dietary lipids aggravates WAT inflammation through TLR signalling. Cell Metabolism. 2015;22(4):658-668
  81. 81. Moreira APB, Alfenas RCG. The influence of endotoxemia on the molecular mechanisms of insulin resistance. Nutricion Hospitalaria. 2012;27:382-390
  82. 82. Muccioli GG et al. The endocannabinoid system links gut microbiota to adipogenesis. Molecular Systems Biology. 2010;6:392
  83. 83. Nicholson JK, Lindon JC. Systems biology: Metabonomics. Nature. 2008;455(7216):1054-1056
  84. 84. Knight R et al. The microbiome and human biology. Annual Review of Genomics and Human Genetics. 2017;18:65-86
  85. 85. Kuipers F, Bloks VW, Groen AK. Beyond intestinal soap–bile acids in metabolic control. Nature Reviews Endocrinology. 2014;10:488-498
  86. 86. Haeusler RA et al. Human insulin resistance is associated with increased plasma levels of 12alpha-hydroxylated bile acids. Diabetes. 2013;62:4184-4191
  87. 87. Wewalka M et al. Fasting serum taurine-conjugated bile acids are elevated in type 2 diabetes and do not change with intensification of insulin. Journal of Clinical Endocrinology and Metabolism. 2014;99:1442-1451
  88. 88. Sayin SI et al. Gut microbiota regulates bile acid metabolism by reducing the levels of taurobeta-muricholic acid, a naturally occurring FXR antagonist. Cell Metabolism. 2013;17:225-235
  89. 89. Ridlon JM et al. Bile acids and the gut microbiome. Current Opinion in Gastroenterology. 2014;30:332-338
  90. 90. Ryan KK et al. FXR is a molecular target for the effects of vertical sleeve gastrectomy. Nature. 2014;509:183-188
  91. 91. Fang S et al. Intestinal FXR agonism promotes adipose tissue browning and reduces obesity and insulin resistance. Nature Medicine. 2015;21:159-165
  92. 92. Schaap FG. Role of fibroblast growth factor 19 in the control of glucose homeostasis. Current Opinion in Clinical in Nutrition and Metabolic Care. 2012;15:386-391
  93. 93. Morton GJ et al. FGF19 action in the brain induces insulin-independent glucose lowering. Journal of Clinical Investigation. 2013;123:4799-4808
  94. 94. Sandoval DA, D’Alessio DA. Physiology of proglucagon peptides: Role of glucagon and GLP-1 in health and disease. Physiological Reviews. 2015;95:513-548
  95. 95. Renga B et al. The bile acid sensor FXR regulates insulin transcription and secretion. Biochimica et Biophysica Acta. 1802;2010:363-372
  96. 96. Popescu IR et al. The nuclear receptor FXR is expressed in pancreatic beta-cells and protects human islets from lipotoxicity. FEBS Letters. 2010;584:2845-2851
  97. 97. Mudaliar S et al. Efficacy and safety of the farnesoid X receptor agonist obeticholic acid in patients with type 2 diabetes and nonalcoholic fatty liver disease. Gastroenterology. 2013;145:574-582
  98. 98. Labbe A et al. Bacterial bile metabolising gene abundance in Crohn’s, ulcerative colitis and type 2 diabetes metagenomes. PLoS One. 2014;9:e115175
  99. 99. Steiner C et al. Bile acid metabolites in serum: Intraindividual variation and associations with coronary heart disease, metabolic syndrome and diabetes mellitus. PloS One. 2011;6:e25006
  100. 100. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-867
  101. 101. Chawla A et al. Nuclear receptors and lipid physiology: Opening the X-files. Science. 2001;294(5548):1866-1870
  102. 102. Glass CK, Ogawa S. Combinatorial roles of nuclear receptors in inflammation and immunity. Nature Reviews Immunology. 2006;6(1):44-55
  103. 103. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. Journal of Clinical Investigation. 2005;115(5):1111-1119
  104. 104. Ge H et al. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology. 2008;149(9):4519-4526
  105. 105. Semenkovich CF. Insulin resistance and atherosclerosis. Journal of Clinical Investigation. 2006;116(7):1813-1822
  106. 106. Shapiro H et al. Personalized microbiome-based approaches to metabolic syndrome management and prevention. Journal of Diabetes. 2017;9(3):226-236
  107. 107. Koeth RA et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nature Medicine. 2013;19(5):576-585
  108. 108. Wang Z et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature. 2011;472(7341):57-63
  109. 109. Chiang JY. Bile acids: Regulation of synthesis. Journal of Lipid Research. 2009;50(10):1955-1966
  110. 110. Thomas C et al. TGR5-mediated bile acid sensing controls glucose homeostasis. Cell Metabolism. 2009;10(3):167-177
  111. 111. Düfer M et al. Bile acids acutely stimulate insulin secretion of mouse b-cells via Farnesoid X receptor activation and KATP channel inhibition. Diabetes. 2012;61:1479-1489
  112. 112. Graessler J et al. Metagenomic sequencing of the human gut microbiome before and after bariatric surgery in obese patients with type 2 diabetes: Correlation with inflammatory and metabolic parameters. The Pharmacogenomics Journal. 2013;13(6):514-522
  113. 113. Larsen N et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5(2):e9085
  114. 114. Grarup N et al. Genetic susceptibility to type 2 diabetes and obesity: From genome-wide association studies to rare variants and beyond. Diabetologia. 2014;57:1528-1541
  115. 115. Hartsra AV et al. Insights into the role of the microbiome in obesity and type 2 diabetes. Diabetes Care. 2015;38(1):159-165
  116. 116. Gomes AC et al. The human gut microbiota: Metabolism and perspective in obesity. Gut Microbes. 2019;9:308-325
  117. 117. Aron-Wisnewsky et al. Major microbiota dysbiosis in severe obesity: Fate after bariatric surgery. Gut. 2019;68:70-82
  118. 118. Ley RE et al. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444:1022-1023
  119. 119. Heianza YD et al. Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism: The POUNDS lost trial. Gut. 2019;68:263-270
  120. 120. Turnbaugh PJ et al. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature. 2006;444:1027-1031
  121. 121. Qin J et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55-60
  122. 122. Shin NR et al. An increase in the Akkermansia spp. population induced by metformin treatment improves glucose homeostasis in diet-induced obese mice. Gut. 2014;63:727-735
  123. 123. Everard A et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proceedings of the National Academy of Sciences of the United States of America. 2013;110:9066-9071
  124. 124. Candela M et al. Modulation of gut microbiota dysbioses in type 2 diabetic patients by macrobiotic Ma-Pi 2 diet. British Journal of Nutrition. 2016;116(1):80-93
  125. 125. Forslund K et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature. 2015;528(7581):262-266

Written By

Alpana Mukhuty, Chandrani Fouzder, Snehasis Das and Dipanjan Chattopadhyay

Submitted: 12 June 2019 Reviewed: 28 November 2019 Published: 26 December 2019