Open access peer-reviewed chapter

Gut Microbiota Potential in Type 2 Diabetes

Written By

Shahzad Irfan, Humaira Muzaffar, Haseeb Anwar and Farhat Jabeen

Submitted: 02 May 2022 Reviewed: 31 May 2022 Published: 26 June 2022

DOI: 10.5772/intechopen.105616

From the Edited Volume

Effect of Microbiota on Health and Disease

Edited by Hoda El-Sayed

Chapter metrics overview

117 Chapter Downloads

View Full Metrics

Abstract

Appropriate metabolic regulation is vital for health. Multiple factors play important roles in maintaining the metabolic system in different physiological conditions. These factors range from intestinal metabolism of food and absorption of nutrients, pancreatic hormones and their interplay under feeding and fasting, hepatic regulation of macronutrient formation and metabolism storage of macronutrients in skeletal muscles. Intestinal metabolism of ingested food and subsequent nutrient absorption depends on the symbiotic microbial community residing in the gut. The specific ratio of different microbial phyla in the gut has proved to be extremely important for the beneficial role of the gut microbiome. The importance of gut microbiome in the regulation of metabolism has been highlighted with reports of the abnormal ratio of gut microbial community resulting in different metabolic disturbances ranging from obesity to the development of diabetes mellitus. The physiological impact of insulin on the metabolic regulation of macronutrients has recently been shown to be augmented by the secondary metabolites produced by anaerobic fermentation. The current chapter aims to highlight recent findings in the regulation of extraintestinal metabolism by gut microbiome with a specific emphasis on the physiology and pathophysiology of the pancreas in health and disease.

Keywords

  • Gut microbiota
  • diabetes mellitus
  • probiotics
  • pancreas

1. Introduction

Insulin is predominantly the most important endogenous protein responsible for the physiological regulation of metabolism [1]. Exogenous insulin is the only substantial treatment option for patients suffering from insulin deficiency since the initial discovery of insulin by Sir Frederick G Banting and its purification by James B. Collip in 1921 [2, 3]. The pancreatic gland is responsible for the regulated secretion of insulin to maintain glucose homeostasis under different physiological conditions [4, 5]. Islets of Langerhans present in the pancreas contain cells that secret specific hormones which help in maintaining glucose levels during feeding and fasting [5, 6, 7, 8, 9]. Islets of Langerhans are defined as closed areas containing multiple cell types with enormous vascular and nervous innervation [5]. Islets of Langerhans are designated as the endocrine portion of the pancreas. The exocrine part of the pancreas surrounds islets of Langerhans. Different cell types present in the islets secrete different types of hormones. Islets contain four different types of endocrine cells: alpha (α) cells (glucagon), beta (β) cells (insulin), delta (δ) cells (somatostatin) and PP cells (pancreatic polypeptide) [5]. Alpha cells are responsible for the secretion of glucagon hormone to enhance blood glucose levels under fasting conditions while β cells are responsible for insulin secretion which initiates postprandial glucose metabolism and thus controls the rising blood glucose levels [10, 11]. Apart from glucose metabolism, insulin is also involved in lipid and protein metabolism [12, 13, 14]. Blood glucose level acts as the main trigger for the release of insulin from β cells, a phenomenon known as glucose-stimulated insulin secretion (GSIS) [15, 16, 17]. Glucose at normal physiological levels not only induces insulin gene transcription by recruiting transcription factors (PDX-1, MafA and NeuroD) but also improves the insulin mRNA stability thus acting as a major physiologic regulator of insulin [18, 19, 20]. Glucose enters the β cells via glucose-specific channels present on the cell membrane commonly known as glucose transporters (GLUT) [4, 17]. Numerous types of glucose transporters are present in different tissue of the body [21]. But specifically, GLUT2 is most abundant and functional in the pancreas (β cells) and liver (hepatocytes) whereas GLUT4 is present on skeletal and cardiac muscles and adipocytes [21, 22].

Advertisement

2. Insulin and macronutrient metabolism

Insulin is the primary hormone responsible for initiating carbohydrate metabolism through phosphorylation of glucose and subsequent formation of glucose-6-phosphate inside the cells [23, 24]. Insulin activates the hexokinase enzymes in non-hepatic tissues and glucokinase (GCK) in β cells and hepatocytes to initiate glucose phosphorylation [25, 26, 27, 28]. The insulin hormone acts by binding to the cell surface insulin receptors which are vastly distributed in different tissues of the body [29]. The binding of insulin to its receptors activates adaptor proteins known as insulin receptor substrates (e.g. IRS1, IRS2) [30]. IRS protein converts the tyrosine phosphorylation signal into the lipid kinase by activating phosphoinositide2-kinase enzyme (PI3K). Activated PI3K further recruits ATP molecules which activates AKT (serine and threonine kinase) [31]. The Discovery of insulin’s primary role in activating AKT proved a landmark in explaining the conversion of tyrosine phosphorylation into serine/threonine phosphorylation signal. AKT activation also explains the insulin induced regulation of key steps in insulin signaling including (a) glucose uptake by glucose transporter (GLUT4), (b) glycogen synthesis by glycogen synthase kinase 3 (GSK3) inhibition, (c) synthesis of protein and fats via activation of the mechanistic target of rapamycin (mTOR), (d) gene expression regulation at the transcriptional levels by forkhead family box O (FOXO) transcription factor proteins. Insulin enhances GLUT4 activity in muscle and adipose tissue thus increasing the rate of glucose transport, glycolysis and subsequent glycogen synthesis in these tissues [32]. Insulin also prevents hepatic glucose synthesis by inhibiting hepatic glycogenolysis and gluconeogenesis [33, 34, 35].

Apart from glucose metabolism, insulin influences lipid and protein metabolism through multiple means. Insulin lowers the plasma fatty acid levels by decreasing adipocyte lipolysis and enhancing the hepatic formation of very low density lipoprotein (VLDL) [36, 37, 38, 39, 40, 41]. Insulin increases the protein synthesis in skeletal muscles and the liver by enhancing the amino acid transport inside the cells and reducing protein degradation and urea formation [13, 42, 43, 44, 45, 46, 47]. These metabolic effects of insulin on carbohydrates, lipids and proteins highlight the importance of insulin signaling in maintaining a nutritional consistency at the cellular level and ensuring a balanced physiological interplay between multiple tissues under diverse physiological conditions.

Advertisement

3. Glucose homeostasis: insulin-glucagon interplay

Alpha and β cells work together to maintain glucose homeostasis under feeding and fasting conditions through the periodic release of insulin and glucagon respectively [4, 25, 48]. Feeding results in increased plasma glucose levels. Rising plasma glucose levels demand immediate systemic activation of glucose metabolism by insulin. A delayed or deficient activation of glucose metabolism will result in abnormally high plasma and cellular glucose levels, a medical condition known as hyperglycemia. Glucose at higher-than-normal concentrations induces glucotoxic effects inside the cells. Rising postprandial glucose levels will trigger β cells to synthesize and secrete insulin. The postprandial rise in insulin levels activates glucokinase and hexokinase activity resulting in glucose phosphorylation in hepatocytes and muscle cells. Conversion of glucose into glucose-6-phosphate will result in the decline of plasma glucose levels over time. Physiologically because of GSIS the postprandial rise in the insulin secretion from β cells declines over time as the blood glucose level decline [15]. Thus, the rising plasma glucose levels provide positive feedback to enhance insulin secretion and the declining plasma glucose levels act as a negative feedback loop to lower insulin levels. Insulin negatively impacts glucagon secretion [49, 50, 51, 52]. Plasma glucose levels decline under fasting conditions. As glucose is the primary cellular source to generate ATP, a minimum threshold of plasma glucose levels must be maintained to avoid hypoglycemia.

Hypoglycemia is a serious medical condition characterized by very low plasma glucose levels. Fasting induced a decline in plasma glucose levels and subsequent diminished insulin levels initiate glucagon synthesis and secretion from alpha cells. To avoid hypoglycemia during fasting, glucagon enhances plasma glucose levels by activating hepatic gluconeogenesis/glucogenolysis thus forming glucose molecules from non-carbohydrate sources [10, 53, 54, 55, 56, 57]. Glucagon secretion from alpha cells and insulin secretion from β cells are also regulated by incretin hormones secreted from the intestines [58]. Incretin hormones are gut peptides secreted from the L and K cells of the small intestine and include glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1) [59, 60]. GIP and GLP-1 functional receptors are present on both alpha and β cells. In normal physiological conditions, the GIP induces glucagon secretion from alpha cells during fasting or hypoglycemic state whereas GLP-1 induces insulin secretion from β cells and inhibits glucagon secretion from alpha cells [59, 61].

Advertisement

4. Diabetes mellitus

Diabetes mellitus is primarily a metabolic dysfunction resulting in a significant reduction in the cellular ability to metabolize glucose because of either the lack of insulin or insulin inactivity (insulin resistance) [62, 63]. Diabetes mellitus is expected to affect 700 million worldwide by 2040 [64]. The compromised ability of the cells to metabolize glucose results in increased cellular and plasma levels of glucose, a condition known as hyperglycemia. Hyperglycemia induces tissue damage mainly through the increased influx of glucose through the polyol pathway and increased formation of advanced glycation end products (AGEs) and subsequent increased expression of AGE receptors and their ligand [65, 66]. Overproduction of reactive oxygen species (ROS) due to hyperglycemia through mitochondria acts as the main trigger for the activation of the polyol pathway, formation of AGEs and increase in AGE receptor expression [67].

Diabetes mellitus has been categorized in two primary forms: Type 1 Diabetes Mellitus (T1DM) and Type 2 Diabetes Mellitus (T2DM). T1DM has been characterized by a mutation in the insulin gene or immune cell-mediated destruction of β cell resulting in either the synthesis of abnormal insulin protein that fails to activate insulin receptors or a complete lack of endogenous insulin secretion [63]. T1DM patients are usually diagnosed early in their life. The only possible medical treatment referred to these patients is the multiple daily doses of synthetic insulin. T2DM on the other hand is much more complicated and requires a thorough diagnostic approach [62, 68, 69, 70]. T2DM is considered one of the most common metabolic disorders globally. Major risk factors for T2DM include a sedentary lifestyle, lack of exercise, excessive use of a high-carb and high-fat diet, overweight and obesity [71]. Poor lifestyle and dietary habits have been attributed to the global incidence of type 2 diabetes in the last 2 decades. Obesity, visceral fat deposition and increased body mass index (BMI) play a central role in the pathophysiology of type 2 diabetic patients [62]. Quality, quantity and type of food have been debated to be the primary cause of this global incident. A healthy diet with the appropriate amount of nutrients and fiber and a certain level of physical activity has been advised globally to counter the incidence of T2DM in young adults.

The development of T2DM is mainly caused by the significant decline in insulin secretion from β cells or the inability of insulin-responsive tissues (muscles, fat and liver) to respond to insulin, mainly because of defective insulin signaling resulting in hyperinsulinemia and subsequent insulin resistance [72, 73, 74, 75]. Failure of the insulin hormone to activate insulin receptors at the cellular level has been attributed to be the major cause of hyperinsulinemia and insulin resistance [76, 77]. Insulin binding to insulin receptors at the plasma membrane activates a signaling cascade that initiates glucose metabolism inside the cells. Insulin-bound insulin receptors or activated insulin receptors go through internalization at the plasma membrane, a phenomenon known as insulin receptor endocytosis [1, 78]. Following the activation, the endocytosis of the insulin receptor is the primary physiological mechanism through which the duration and intensity of insulin signaling are controlled. Hyperinsulinemia accelerates insulin receptor endocytosis and affects the presence of adequate functional insulin receptors at the plasma membrane resulting in insulin resistance [79]. Apart from accelerated insulin receptor endocytosis, insulin-stimulated insulin receptor kinase activity is also decreased in diabetic patients [80]. Compromised insulin signaling fails to activate glucose metabolic enzymes like glucokinase and hexokinase resulting in hyperglycemia. High plasma glucose levels initiate glucose-stimulated insulin secretory (GSIS) response from β cells resulting in the rise of plasma insulin levels. The rising insulin levels should be normalized over time because of the renal insulin clearance mechanism. But compromised renal insulin clearance rate in diabetic subjects results in abnormally high plasma levels of insulin (hyperinsulinemia) [81, 82].

Hyperinsulinemia and hyperglycemia in theory cannot trigger alpha cells to secrete glucagon. But it has been observed that T2DM patients with insulin resistance, hyperinsulinemia and hyperglycemia also have abnormally high plasma levels of glucagon [83]. Hinting toward the disturbance in the alpha and β cell interplay through the inability of the insulin to block glucagon gene transcription [84]. T2DM is also characterized by a decrease in GLP-1 secretion from L cells of the small intestine [85, 86]. Indicating a pathophysiological role of the gut in the development and progression of type 2 diabetes [87, 88]. GLP-1 receptor agonists which induce an increase in insulin secretion from β cells and inhibit glucagon secretion are the major treatment option for T2DM patients to combat hyperglycemic conditions [89, 90, 91].

Advertisement

5. Gut microbiota profile in Type 2 diabetes mellitus

The gut microbiome was first defined scientifically in 2001 as “an ecological community of commensal, symbiotic and pathogenic microorganisms that collectively share our body space” [92]. Approximately 100 trillion microbes are found in the human gastrointestinal tract (GIT) and strongly influence the health status of individuals either directly or indirectly [93, 94, 95, 96, 97]. The primary reason for the pathophysiological effect of the gut microbiome on human physiology has been attributed to the disruption of the stable communities of gut microbes through medication, diet and lifestyle. A normal, healthy gut microbiome profile is termed eubiosis and abnormal gut microbiome composition is called dysbiosis [98, 99, 100, 101, 102, 103, 104, 105, 106]. Eubiosis typically refers to an ideal bacterial population comprising 95% of Bacteroidetes and 5% Firmicutes producing abundant microbial metabolites like short-chain fatty acids (SCFAs), branched-chain amino acids (BCAAs) and impacting lipid metabolism. SCFAs like butyrate, acetate and propionate are produced by the anaerobic fermentation of non-digestible carbohydrates (dietary fiber) and promote gut integrity and protect gut epithelial lining by forming tight junctions and preventing gut permeability [107]. These microbial secondary metabolites act as central components in microbe to host signaling pathways activation. Much of the specific microbiota involved in the production of these important secondary metabolites are reduced in T2DM patients.

Substantial data from human studies support the possibility that dysbiosis triggers obesity, inflammation, insulin resistance and T2DM [108, 109, 110, 111]. Association of dysbiosis is also attributed to the pathogenesis of intestinal tissue. Intestinal disorders attributed to dysbiosis include inflammatory bowel disease, irritable bowel syndrome (IBD) and coeliac disease [102, 111, 112]. Whereas metabolic syndrome, obesity, and cardiovascular complications are attributed as extra-intestinal effects of dysbiosis. Dysbiosis has also been attributed not only to the initiation of the T2DM in humans (a condition known as prediabetes) but also during the progression and subsequent secondary complications of T2DM with several lines of evidence suggesting that manipulation of the gut microbiome helps to minimize or alleviate the T2DM conditions [98, 113, 114, 115, 116, 117, 118, 119, 120, 121].

The role of gut microbiota in health and disease and specifically the pathogenesis of T2DM has been experimentally investigated mainly by using rodent models as a limited amount of experimental data can be generated through human studies. Keeping in mind that the rodents and human physiology are not exactly similar and certain physiological differences exist. The non-human primates seem to be a much more appropriate animal model to study different aspects of primate physiology including the gut microbiome and its interaction with metabolic dysregulation [122, 123, 124, 125, 126, 127]. Nonetheless, the current understanding of the role of the gut microbiome in the context of metabolic syndrome or pathogenesis of diabetes mellitus has primarily originated from the data on rodent and human studies [94, 97, 116, 128, 129, 130, 131, 132, 133]. Interestingly efforts have been made in the past to characterize the gut microbiome in normal and diabetic individuals as well as some therapeutic approaches have been adopted [95, 98, 113, 116, 117, 120, 121, 134].

The attempts to characterize the normal human gut microbiome revealed four primary phyla which are responsible for the physiological role of gut in metabolic modulation [128, 132, 133, 135, 136, 137, 138, 139, 140, 141]. These four specific phyla/families of microbes present in the gut include Bacteroidetes (Bacteroidota), Firmicutes (Bacillota), Proteobacteria (Pseudomonadota) and actinobacteria (Actinomycetota) [95, 142]. The specific proportion for each of these phyla in normal physiological and homeostatic conditions indicates that the largest group of microbes is the Firmicutes which make up to 64% of the total gut microbiota. Followed by the Bacteroidetes, which make up the second-largest group, contributing up to 23% of the total gut microbiota. Proteobacteria and actinobacteria contribute the rest with 8% and 3% respectively. These specific percentage contributions of each phylum are extremely important physiologically. Increased prevalence of pro-inflammatory conditions such as obesity, T2DM, arthritis and even cancer have been attributed to the disruption of these specific percentage contributions of each phylum [132, 143]. Human and animal data have highlighted the unique compositional changes in the microbiota profiles at the phylum level in T2DM conditions [113, 128]. T2DM patients exhibit increased membrane transport of sugars, BCAA transportation, methane metabolism and sulfate reduction [128]. These patients also have reduced butyrate biosynthesis and cofactors/vitamins metabolism.

Although a certain level of discrepancy does exist in terms of phyla composition data between different T2DM patients which has been attributed to the specific geographical location, culture-specific diet and medication use [144]. Numerous independent research groups have reported widely contrasting microbiota findings in the context of phyla composition in T2DM patients [113, 114, 117, 119, 128, 145146]. It seems highly unlikely that a single microbe species can play a significant or dominant role in determining the risk of T2DM. The conflicting data from several independent groups also have some interesting similarities. Specifically, it was a common observation among T2DM patients that butyrate-producing microbes were particularly depleted [117, 128]. As human microbiome is comprised mainly of Bacteroidetes and Firmicutes with a specific ratio (B/F > 1) and obesity has been shown to impact this ratio and result in the increased prevalence of Firmicutes to that of Bacteroidetes [109, 147, 148, 149]. Implicating that a disrupted B/F ratio can contribute to obesity in humans. Similarly increased concentration of Bacteroidetes and Proteobacteria with a significant decline in Firmicutes has been reported in T2DM patients [113] T2DM also demonstrates an increase in pathogenic microbial species like Clostridium symbiosum, Clostridium ramosum, and Escherichia coli resulting in systemic inflammation [119, 128].

Insulin resistance has also been attributed to disrupted Bacteroidetes and Firmicutes (B/F) ratio. An altered B/F ratio impacts intestinal permeability and lipopolysaccharide (LPS) from proteobacteria are translocated from inside the gut. LPS translocation activates immune response through interleukin-1 (IL-1), tumor necrosis factor (TNF), Jun N-terminal kinases (JNK) and IkB kinase (IKK). LPS-induced activation of JNK and IKK results in the phosphorylation of insulin receptor substrate (IRS) which fails to activate downstream effector molecules like PI3K and AKT thus rendering the insulin signaling cascade ineffective [150, 151]. IKK also activates the nuclear translocation of nuclear factor kappa B (NF-kB). NF-kB, a transcription factor, induces the expression of several genes involved in inflammatory and apoptotic responses [152, 153, 154, 155]. The inflammatory state also called metabolic endotoxemia is accompanied by insulin resistance and obesity.

Advertisement

6. Regulation of glucose homeostasis by Gut microbiota

Gut microbiota has been shown to impact the pancreas directly. Gut microbiota has been proposed to modulate glucose homeostasis through multiple mechanisms [115, 116, 118, 156, 157]. Experimental data support four specific mechanisms through which the gut microbiome influences glucose homeostasis; (1) the β cell modulating effects of metabolites that are formed due to gut anaerobic microbial fermentation [157, 158, 159], (2) induction of cytokine activity in the islets of Langerhans via inflammatory cascades [160, 161, 162, 163, 164], (3) direct islets signaling affecting insulin and glucagon secretion through incretins modulation [87, 165], (4) alteration in the gut permeability, thus permitting the influx of toxins through intestinal mucosal barrier [166]. Mechanisms 1 and 3 are mainly considered for increased T2DM susceptibility and whereas mechanisms 2 and 4 are particularly implicated in the development of T1DM in early life. As T1DM is characterized by a significant reduction in the number of functional β cells. Cytokine and toxin-induced β cell apoptosis or dedifferentiation are considered major risk factors for T1DM.

Abnormal gut microbiome composition alters the intestinal barrier which favors absorption and increased circulating levels of LPS and BCAA. LPS induces low-grade inflammation and insulin resistance while BCAA is associated with an increased risk of T2DM development. An altered intestinal barrier also reduces the absorption of beneficial SCFAs and secondary bile acids. Metabolically SCFAs are mainly produced as an energy source for the gut epithelium. Butyrate is used by colonic epithelial cells for energy, acetate is used as a fatty acid precursor like cholesterol and propionate is a precursor for the process of hepatic gluconeogenesis [167, 168, 169]. Animal data have shown the beneficial impact of acetate supplementation on insulin resistance and glucose tolerance in animals fed with a high-fat diet [170]. Acetate at high intravenous (i.v) dose has also been reported to acutely enhance circulating levels of GLP-1 in humans [171]. Butyrate supplementation has been reported to enhance insulin sensitivity in mice fed with a high-fat diet while obesity and insulin resistance fail to develop over the course of 16 weeks [165].

Functional modulation of β cells through secondary metabolites is highly important in maintaining homeostatic glucose levels. SCFA has been highlighted as an important signaling molecule as the recent findings of the presence of functional SCFA cell surface receptors on different tissues including gut and peripheral tissues [172, 173, 174, 175]. Gut microbial modulation of the host’s metabolism modulated by SCFA production has been demonstrated by the activation of G-protein coupled cell surface receptors (GPCRs) also known as free fatty acid receptors (FFAR). FFAR includes different GPCRs which bind fatty acids of different chain lengths. GPR40 (FFA1), GPR84, and GPR 120 (FFA4) bind with the medium and long-chain fatty acids. Whereas GPR43 (FFA20), GPR41 (FFA3), and GPR109 bind with SCFAs. Propionate and acetate are found to be the most potent agonists of FFA3/GPR41 while butyrate selectively binds with FFA2. FFARs have been shown to be present in different peripheral tissues including the gut, liver, and pancreas. SCFAs like butyrate and propionate along with secondary bile acids indirectly modulate β cells. SCFAs enhance insulin secretion by activating GLP-1 secretion from the intestines [158, 176]. Butyrate and propionate bind and activate G-protein coupled receptors (GPR43, GPR119) present on the enteroendocrine L cells and stimulate the release of GLP-1 in humans [177, 178]. Propionate also has been reported to influence β cell activity directly in humans. Propionate inhibits inflammatory cytokine-induced β cell apoptosis in human islets and enhances GSIS response from β cells independent of increased GLP-1 levels [179]. FFARs have been expressed by β cells and reported to modulate β cell activity in terms of GSIS [174, 180]. Apart from β cell modulation in terms of GSIS response, an interesting observation was made in these studies that high-fat diet-induced insulin resistance in a mouse model has shown to influence FFA2 receptor expression in β cells. Apart from these above-mentioned in vivo studies a recently published in vitro data further extends the notion that acetate, propionate, and butyrate separately enhance insulin secretion along with an increase in the expression levels of insulin genes from rat islets during long-term incubation [181]. Interestingly the authors noted that long-term incubation with butyrate induced a significant downregulation of β cell-specific key transcriptional factors and functional genes involved in the maturity-onset diabetes of the young (MODY). Another interesting finding which was made in this recent study was the significant suppression of the β cell identity genes like GLUT2, GCK, Pdx1, MafA, Nkx-6.1, and NeuroD1 after the long-term incubation with butyrate. The global suppression of the β cell identity gene was surprisingly independent of the deacetylase activity of butyrate indicating a non-DNA acetylation mechanism involved. A significant decrease in the gene expression pattern of GLUT2 and GCK in rat islets after the long-term incubation with butyrate indicates that glucose or GSIS was not involved in the increased mRNA levels of INS1 and secretion of insulin protein. Instead the basal levels of intracellular calcium ions [Ca2+]i was much higher in butyrate-treated islets as compared to the control. The combined effect of acetate, propionate, and butyrate on isolated rat or mouse islets in short- and long-term incubations needs to be examined in future studies. Along with in vivo approaches to fully characterize the impact of microbial metabolites on glucose homeostasis in different physiological conditions.

Advertisement

7. Prebiotics, probiotics and diet

Prebiotics are food ingredients that are non-digestible but fermentable oligosaccharides. The primary role of prebiotics in food is to stimulate the fermenting activity of gut microbes and eventually trigger the growth of beneficial gut microbes [182, 183, 184, 185]. Probiotics on the other hand are special foods that contain a certain amount of alive non-pathogenic bacteria which help to improve gut health and confer eubiosis [186, 187]. Bifidobacteria, lactobacilli streptococci and E. coli are the main bacterial strains that constitute most of the available probiotics. Prebiotics and probiotics supplementation has been shown to reduce inflammation and obesity in T2D patients [188, 189, 190, 191, 192]. The beneficial effect of prebiotics and probiotics on gut health, in general, is well accepted. However, the expected benefits of pre and probiotics in dysbiotic T2D patients is limited. Advanced stage or elderly diabetic patients fails to respond to pre and probiotic supplementation as compared to young and early-stage T2D patients [190, 193, 194]. An appropriate ratio of different gut microbes is extremely important for proper metabolic physiology. As dysbiosis has been attributed as an important permissive or causative factor in developing T2D. Extensive use of high-fat diets, diets which are also called western diets or fast food, have the ability to modulate gut microbiota. Specifically, the downregulation of beneficial Bifidobacteria (Actinobacteria), which helps to break down food, and nutrient absorption and helps to alleviate constipation and diarrhea by fighting off pathogenic microbes. As Actinobacteria only makes up to 3% of the total gut microbiota, the high fat diet-induced decrease in Bifidobacteria causes an acute pathological impact on metabolism and gut health. A high-fat diet also induces an unwanted increase in the proteobacteria, which usually accounts for a maximum 8%. An increase in the LPS containing proteobacteria causes inflammation and obesity, a condition known as endotoxemia which is accompanied by insulin resistance. Oligofructose-containing prebiotics has been shown to lower LPS containing Proteobacteria by enhancing Bifidobacterial thus modulating endotoxemia and via GLP-1 dependent pathway improves glucose tolerance [195]. Prebiotics, probiotics and fecal microbial transplantation (FMTs) are the main treatment options to enhance gut microbiota. The limited success of these treatment options to restore and maintain the eubiosis over time and the unique microbiology of the gut microbiome has called for a better understanding of gut microbial response and adaptation to different diets and lifestyles. An elegant recent report documenting the in vivo bacterial gene expression profile in the gut in different groups of mice indicates that bacterial gene expression is hugely impacted by the type of food present in the gut [196]. Lifestyles, cultures and specific diets have been shown to modulate the gut microbiome in healthy non-diabetic subjects (Figure 1). These are lifestyle/diet-induced effects that eventually cause metabolic disorders and obesity. Apart from these detrimental outcomes of certain lifestyles, medications especially antibiotics can severely damage the overall population and the specific ratio of the gut microbiome.

Figure 1.

Impact of the use of probiotics and prebiotics on the gut microbiome in terms of its functionality and improving the glycemic control through manipulation of multiple factors like improved incretin secretions, increase in the production of SCFAs, improved bile acid metabolism and the decrease in the LPs induced low grade inflammatory response.

Advertisement

8. Conclusion

Diabetes mellitus has emerged as the major metabolic disorder in the last two decades. A sedentary urban lifestyle, increased consumption of processed and fried foods and diets high in fat and protein have been indicated as the main reason for unhealthy weight gain causing obesity and disrupting the normal physiological pathways responsible for metabolic homeostasis. The role of the gut microbiome in ensuring a healthy metabolic and immune system is paramount. The remarkable research efforts made in the last two decades highlight gut microbial imbalance or dysbiosis as a common finding in diabetic patients. The direct and indirect regulatory influence of the gut microbial activity on the islet’s functionality has been experimentally characterized in rodent models. The experimental findings highlight the importance of a healthy gut microbial community and the use of the appropriate amount of dietary fiber to support fermentation and production of beneficial SCFAs which not only impact the intestinal permeability but also influence β cell activity directly as well as indirectly. The use of pre or probiotics along with a healthy diet comprising enough dietary fiber is a prerequisite for communities and individuals suffering from obesity and diabetes.

References

  1. 1. Petersen MC, Shulman GI. Mechanisms of insulin action and insulin resistance. Physiological Reviews. 2018;98(4):2133-2223
  2. 2. Hegele RA, Maltman GM. Insulin’s centenary: The birth of an idea. The Lancet Diabetes and Endocrinology. 2020;8(12):971-977
  3. 3. Gerstein HC, Rutty CJ. Insulin therapy: The discovery that shaped a century. Canadian Journal of Diabetes. 2021;45(8):798-803. Available from: https://pubmed.ncbi.nlm.nih.gov/34045148/
  4. 4. Thorens B. GLUT2, glucose sensing and glucose homeostasis. Diabetologia. 2015;58:221-232
  5. 5. Atkinson MA, Campbell-Thompson M, Kusmartseva I, Kaestner KH. Organisation of the human pancreas in health and in diabetes. Diabetologia. 2020;63:1966-1973
  6. 6. Tengholm A, Gylfe E. cAMP signalling in insulin and glucagon secretion. Diabetes, Obesity and Metabolism. 2017;19:42-53
  7. 7. Rutter GA, Hodson DJ. Minireview: Intraislet regulation of insulin secretion in humans. Molecular Endocrinology. 2013;27:1984-1995
  8. 8. Adeva-Andany MM, Funcasta-Calderón R, Fernández-Fernández C, Castro-Quintela E, Carneiro-Freire N. Metabolic effects of glucagon in humans. Journal of Clinical & Translational Endocrinology. 2019;15:45-53
  9. 9. Hughes JW, Ustione A, Lavagnino Z, Piston DW. Regulation of islet glucagon secretion: Beyond calcium. Diabetes, Obesity and Metabolism. 2018;20:127-136
  10. 10. Yu Q , Shuai H, Ahooghalandari P, Gylfe E, Tengholm A. Glucose controls glucagon secretion by directly modulating cAMP in alpha cells. Diabetologia. 2019;62(7):1212-1224
  11. 11. Kyung KM, Shin HM, Jung HS, Lee EJ, Kim TK, Kim TN, et al. Comparison of pancreatic beta cells and alpha cells under hyperglycemia: Inverse coupling in pAkt-FoxO1. Diabetes Research and Clinical Practice. 2017;131:1-11
  12. 12. Lee S, Dong HH. FoxO integration of insulin signaling with glucose and lipid metabolism. Journal of Endocrinology. 2017;233:R67-R79
  13. 13. Tessari P. Changes in protein, carbohydrate, and fat metabolism with aging: Possible role of insulin. Nutrition Reviews. 2000;58(1):11-19
  14. 14. Tessari P. Role of insulin in age-related changes in macronutrient metabolism. European Journal of Clinical Nutrition. 2000;54(Suppl 3):S126-S130. Available from: https://pubmed.ncbi.nlm.nih.gov/11041084/
  15. 15. Komatsu M, Takei M, Ishii H, Sato Y. Glucose-stimulated insulin secretion: A newer perspective. Journal of Diabetes Investigation. 2013;4(6):511-516. Available from: https://pubmed.ncbi.nlm.nih.gov/24843702/
  16. 16. Straub SG, Sharp GWG. Glucose-stimulated signaling pathways in biphasic insulin secretion. Diabetes/Metabolism Research and Reviews. 2002;18(6):451-463
  17. 17. Wang J, Gu W, Chen C. Knocking down insulin receptor in pancreatic beta cell lines with lentiviral-small hairpin RNA reduces glucose-stimulated insulin secretion via decreasing the gene expression of insulin, GLUT2 and Pdx1. International Journal of Molecular Sciences. 2018;19(4):985
  18. 18. Poitout V, Hagman D, Stein R, Artner I, Robertson RP, Harmon JS. Regulation of the insulin gene by glucose and fatty acids. Journal of Nutrition. 2006;136(4):873-876 Available from: https://pubmed.ncbi.nlm.nih.gov/16549443/
  19. 19. Tillmar L, Carlsson C, Welsh N. Control of insulin mRNA stability in rat pancreatic islets. Regulatory role of a 3′-untranslated region pyrimidine-rich sequence. The Journal of Biological Chemistry. 2002;277(2):1099-1106. Available from: https://pubmed.ncbi.nlm.nih.gov/11696543/
  20. 20. Tillmar L, Welsh N. Glucose-induced binding of the polypyrimidine tract-binding protein (PTB) to the 3′-untranslated region of the insulin mRNA (ins-PRS) is inhibited by rapamycin. Molecular and Cellular Biochemistry. 2004;260(1):85-90
  21. 21. Thorens B, Mueckler M. Glucose transporters in the 21st century. American Journal of Physiology. Endocrinology and Metabolism. 2010;298:141-145. Available from: http://www.ajpendo.org
  22. 22. Mueckler M, Thorens B. The SLC2 (GLUT) family of membrane transporters. Molecular Aspects of Medicine. 2013;34(2-3):121-138
  23. 23. Posner BI. Insulin signalling: The inside story. Can. Journal of Diabetes. 2017;41(1):108. Available from: /pmc/articles/PMC5272803/
  24. 24. Newsholme EA, Dimitriadis G. Integration of biochemical and physiologic effects of insulin on glucose metabolism. Experimental and Clinical Endocrinology and Diabetes. 2001;109(SUPPL. 2):S122-S134. Available from: http://www.thieme-connect.de/DOI/DOI?10.1055/s-2001-18575
  25. 25. Matschinsky FM, Wilson DF. The central role of glucokinase in glucose homeostasis: A perspective 50 years after demonstrating the presence of the enzyme in islets of Langerhans. Frontiers in Physiology. 2019;10:148
  26. 26. Massa ML, Gagliardino JJ, Francini F. Liver glucokinase: An overview on the regulatorymechanisms of its activity. IUBMB Life. 2011;63:1-6
  27. 27. Iynedjian PB. Molecular physiology of mammalian glucokinase. Cellular and Molecular Life Sciences. 2009;66:27-42
  28. 28. Matschinsky FM. Regulation of pancreatic-cell Glucokinase from basics to therapeutics. Diabetes. 2002;51(Suppl 3):S394-S404
  29. 29. Haeusler RA, McGraw TE, Accili D. Metabolic signalling: Biochemical and cellular properties of insulin receptor signalling. Nature Reviews Molecular Cell Biology. 2018;19:31-44
  30. 30. White MF. IRS proteins and the common path to diabetes. American Journal of Physiology—Endocrinology and Metabolism. 2002;283(3):46-43
  31. 31. Kohn AD, Kovacina KS, Roth RA. Insulin stimulates the kinase activity of RAC-PK, a pleckstrin homology domain containing ser/thr kinase. The EMBO Journal. 1995;14(17):4288-4295. Available from: https://onlinelibrary.wiley.com/doi/full/10.1002/j.1460-2075.1995.tb00103.x
  32. 32. Zorzano A, Palacín M, Gumà A. Mechanisms regulating GLUT4 glucose transporter expression and glucose transport in skeletal muscle. Acta Physiologica Scandinavica. 2005;183(1):43-58
  33. 33. Wang L, Liu Q , Kitamoto T, Hou J, Qin J, Accili D. Identification of insulin-responsive transcription factors that regulate glucose production by hepatocytes. Diabetes. 2019;68(6):1156-1167
  34. 34. den Boer MAM, Voshol PJ, Kuipers F, Romijn JA, Havekes LM. Hepatic glucose production is more sensitive to insulin-mediated inhibition than hepatic VLDL-triglyceride production. American Journal of Physiology. Endocrinology and Metabolism. 2006;291:1360-1364. Available from: http://www.ajpendo.org
  35. 35. Hatting M, Tavares CDJ, Sharabi K, Rines AK, Puigserver P. Insulin regulation of gluconeogenesis. Annals of the New York Academy of Sciences. 2018;1411:21-35
  36. 36. Kamagate A, Dong HH. FoxO1 integrates insulin signaling to VLDL production NIH public access. Cell Cycle. 2008;7(20):3162-3170. Available from: http://www.landesbioscience.com/journals/cc/article/6882
  37. 37. Kamagate A, Qu S, Perdomo G, Su D, Dae HK, Slusher S, et al. FoxO1 mediates insulin-dependent regulation of hepatic VLDL production in mice. Journal of Clinical Investigation. 2008;118(6):2347-2364
  38. 38. Kim DH, Zhang T, Lee S, Calabuig-Navarro V, Yamauchi J, Piccirillo A, et al. FoxO6 integrates insulin signaling with MTP for regulating VLDL production in the liver. Endocrinology. 2014;155(4):1255-1267
  39. 39. Scherer T, Ohare J, Diggs-Andrews K, Schweiger M, Cheng B, Lindtner C, et al. Brain insulin controls adipose tissue lipolysis and lipogenesis. Cell Metabolism. 2011;13(2):183-194
  40. 40. Iwen KA, Scherer T, Heni M, Sayk F, Wellnitz T, Machleidt F, et al. Intranasal insulin suppresses systemic but not subcutaneous lipolysis in healthy humans. Journal of Clinical Endocrinology and Metabolism. 2014;99(2):E246-E251
  41. 41. Saltiel AR. Insulin signaling in the control of glucose and lipid homeostasis. Handbook of Experimental Pharmacology. 2016;233:51-71. Available from: https://pubmed.ncbi.nlm.nih.gov/26721672/
  42. 42. Charlton M, Nair KS. Protein metabolism in insulin-dependent diabetes mellitus. The Journal of Nutrition. 1998;128(2):323S-327S. Available from: https://academic.oup.com/jn/article/128/2/323S/4723974
  43. 43. Fukagawa NK, Minaker KL, Rowe JW, Goodman MN, Matthews DE, Bier DM, et al. Insulin-mediated reduction of whole body protein breakdown. Dose-response effects on leucine metabolism in postabsorptive men. Journal of Clinical Investigation. 1985;76(6):2306-2311
  44. 44. Newgard CB, An J, Bain JR, Muehlbauer MJ, Stevens RD, Lien LF, et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metabolism. 2009;9(4):311-326
  45. 45. Tessari P, Nosadini R, Trevisan R, de Kreutzenberg SV, Inchiostro S, Duner E, et al. Defective suppression by insulin of leucine-carbon appearance and oxidation in type 1, insulin-dependent diabetes mellitus. Evidence for insulin resistance involving glucose and amino acid metabolism. Journal of Clinical Investigation. 1986;77(6):1797-1804
  46. 46. Yoon MS. The emerging role of branched-chain amino acids in insulin resistance and metabolism. Nutrients. 2016;8(7):405
  47. 47. Asghari G, Farhadnejad H, Teymoori F, Mirmiran P, Tohidi M, Azizi F. High dietary intake of branched-chain amino acids is associated with an increased risk of insulin resistance in adults. Journal of Diabetes. 2018;10(5):357-364
  48. 48. Holst JJ, Holland W, Gromada J, Lee Y, Unger RH, Yan H, et al. Insulin and glucagon: Partners for life. Endocrinology. 2017;158(4):696-701
  49. 49. Vergari E, Knudsen JG, Ramracheya R, Salehi A, Zhang Q , Adam J, et al. Insulin inhibits glucagon release by SGLT2-induced stimulation of somatostatin secretion. Nature Communications. 2019;10(1):139
  50. 50. Wan CKN, Giacca A, Matsuhisa M, El-Bahrani B, Lam L, Rodgers C, et al. Increased responses of glucagon and glucose production to hypoglycemia with intraperitoneal versus subcutaneous insulin treatment. Metabolism, Clinical and Experimental. 2000;49(8):984-989
  51. 51. Ito K, Maruyama H, Hirose H, Kido K, Koyama K, Kataoka K, et al. Exogenous insulin dose-dependently suppresses glucopenia-induced glucagon secretion from perfused rat pancreas. Metabolism. 1995;44(3):358-362
  52. 52. Göbl C, Morettini M, Salvatori B, Alsalim W, Kahleova H, Ahrén B, et al. Temporal patterns of glucagon and its relationships with glucose and insulin following ingestion of different classes of macronutrients. Nutrients. 2022;14(2):376. Available from: http://www.ncbi.nlm.nih.gov/pubmed/35057557
  53. 53. Walker JN, Ramracheya R, Zhang Q , Johnson PRV, Braun M, Rorsman P. Regulation of glucagon secretion by glucose: Paracrine, intrinsic or both? Diabetes, Obesity and Metabolism. 2011;13(SUPPL. 1):95-105. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/j.1463-1326.2011.01450.x
  54. 54. Onyango AN. Mechanisms of the regulation and dysregulation of glucagon secretion. Oxidative Medicine and Cellular Longevity. 2020;2020:3089139
  55. 55. Barthel A, Schmoll D. Novel concepts in insulin regulation of hepatic gluconeogenesis. American Journal of Physiology-Endocrinology and Metabolism. 2003;285(4):E685-E692. Available from: http://www.ajpendo.org
  56. 56. Cooperberg BA, Cryer PE. Insulin reciprocally regulates glucagon secretion in humans. Diabetes. 2010;59(11):2936-2940
  57. 57. Bansal P, Wang Q. Insulin as a physiological modulator of glucagon secretion. American Journal of Physiology. Endocrinology and Metabolism. 2008;295(4):E751-E761
  58. 58. Holst J, Christensen M, Lund A, de Heer J, Svendsen B, Kielgast U, et al. Regulation of glucagon secretion by incretins. Diabetes, Obesity and Metabolism. 2011;13(SUPPL. 1):89-94. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/j.1463-1326.2011.01452.x
  59. 59. El K, Campbell JE. The role of GIP in α-cells and glucagon secretion. Peptides (NY). 2020;125:170213
  60. 60. Seino Y, Fukushima M, Yabe D. GIP and GLP-1, the two incretin hormones: Similarities and differences. Journal of Diabetes Investigation. 2010;1:8-23
  61. 61. Drucker DJ. Mechanisms of action and therapeutic application of glucagon-like peptide-1. Cell Metabolism. 2018;27(4):740-756
  62. 62. Galicia-Garcia U, Benito-Vicente A, Jebari S, Larrea-Sebal A, Siddiqi H, Uribe KB, et al. Pathophysiology of type 2 diabetes mellitus. International Journal of Molecular Sciences. 2020;21:1-34
  63. 63. Harrison LC. The dark side of insulin: A primary autoantigen and instrument of self-destruction in type 1 diabetes. Molecular Metabolism. 2021;52:101288
  64. 64. Maiese K. New insights for oxidative stress and diabetes mellitus. Oxidative Medicine and Cellular Longevity. 2015;2015:875961
  65. 65. Brownlee M. The pathobiology of diabetic complications a unifying mechanism. Banting Lecture. Diabetes. 2005;54(6):1615-1625
  66. 66. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circulation Research. 2010;107:1058-1070
  67. 67. Pi J, Zhang Q , Fu J, Woods CG, Hou Y, Corkey BE, et al. ROS signaling, oxidative stress and Nrf2 in pancreatic beta-cell function. Toxicology and Applied Pharmacology. 2010;244:77-83
  68. 68. Hanley SC, Austin E, Assouline-Thomas B, Kapeluto J, Blaichman J, Moosavi M, et al. β-Cell mass dynamics and islet cell plasticity in human type 2 diabetes. Endocrinology. 2010;151(4):1462-1472
  69. 69. Satin LS, Butler PC, Ha J, Sherman AS. Pulsatile insulin secretion, impaired glucose tolerance and type 2 diabetes. Molecular Aspects of Medicine. 2015;42:61-77
  70. 70. Moin ASM, Dhawan S, Cory M, Butler PC, Rizza RA, Butler AE. Increased frequency of hormone negative and polyhormonal endocrine cells in lean individuals with type 2 diabetes. Journal of Clinical Endocrinology and Metabolism. 2016;101(10):3628-3636
  71. 71. Roden M, Shulman GI. The integrative biology of type 2 diabetes. Nature. 2019;576(7785):51-60
  72. 72. Zick Y. Insulin resistance: A phosphorylation-based uncoupling of insulin signaling. Trends in Cell Biology. 2001;11(11):437-441
  73. 73. Chatterjee S, Khunti K, Davies MJ. Type 2 diabetes. The Lancet. 2017;389(10085):2239-2251
  74. 74. Lewis GF, Carpentier A, Vranic M, Giacca A. Resistance to insulin’s acute direct hepatic effect in suppressing steady-state glucose production in individuals with type 2 diabetes. Diabetes. 1999;48(3):570-576
  75. 75. Weyer C, Bogardus C, Mott DM, Pratley RE. Introduction the natural history of insulin secretory dysfunction and insulin resistance in the pathogenesis of type 2 diabetes mellitus. Journal of Clinical Investigation. 1999;104(6):787-794
  76. 76. Haeusler RA, McGraw TE, Accili D. Biochemical and cellular properties of insulin receptor signalling. Nature Reviews. Molecular Cell Biology. 2018;19:31-44
  77. 77. Boucher J, Kleinridders A, Ronald KC. Insulin receptor signaling in normal and insulin-resistant states. Cold Spring Harbor Perspectives in Biology. 2014;6(1):a009191
  78. 78. Kaksonen M, Roux A. Mechanisms of clathrin-mediated endocytosis. Nature Reviews. Molecular Cell Biology. 2018;19(5):313-326. Available from: https://pubmed.ncbi.nlm.nih.gov/29410531/
  79. 79. Hall C, Yu H, Choi E, Insulin receptor endocytosis in the pathophysiology of insulin resistance. Experimental & Molecular Medicine. 2020;52(6):911-920. Available from: https://www.nature.com/articles/s12276-020-0456-3
  80. 80. Caro JF, Ittoop O, Pories WJ, Meelheim D, Flickinger EG, Thomas F, et al. Studies on the mechanism of insulin resistance in the liver from humans with noninsulin-dependent diabetes. Insulin action and binding in isolated hepatocytes, insulin receptor structure, and kinase activity. The Journal of Clinical Investigation. 1986;78(1):249-258. Available from: https://pubmed.ncbi.nlm.nih.gov/3522628/
  81. 81. Thomas DD, Corkey BE, Istfan NW, Apovian CM. Hyperinsulinemia: An early indicator of metabolic dysfunction. Journal of the Endocrine Society. 2019;3(9):1727. Available from: /pmc/articles/PMC6735759/
  82. 82. Kurauti MA, Ferreira SM, Soares GM, Vettorazzi JF, Carneiro EM, Boschero AC, et al. Hyperinsulinemia is associated with increasing insulin secretion but not with decreasing insulin clearance in an age-related metabolic dysfunction mice model. Journal of Cellular Physiology. 2019;234(6):9802-9809. Available from: https://pubmed.ncbi.nlm.nih.gov/30370604/
  83. 83. D’alessio D. The role of dysregulated glucagon secretion in type 2 diabetes. Diabetes, Obesity and Metabolism. 2011;13(SUPPL. 1):126-132
  84. 84. Philippe J, Knepel W, Waeber G. Insulin regulation of the glucagon gene is mediated by an insulin-responsive DNA element. Proceedings of the National Academy of Sciences of the United States of America. 1991;88(16):7224. Available from: /pmc/articles/PMC52266/?report=abstract
  85. 85. Eissele R, Göke R, Willemer S. Glucagon-like peptide-1 cells in the gastrointestinal tract and pancreas of rat, pig and man. European Journal of Clinical Investigation. 1992;22:283-291
  86. 86. Holst JJ. The physiology of glucagon-like peptide 1. Physiological Reviews. 2007;87(4):1409-1439
  87. 87. Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes. 2012;61(2):364-371
  88. 88. Lund A. On the role of the gut in diabetic hyperglucagonaemia. Danish Medical Journal. 2017;64(4):B5340
  89. 89. Drucker DJ. Glucagon-like peptide-1 and the islet β-cell: Augmentation of cell proliferation and inhibition of apoptosis. Endocrinology. 2003;144:5145-5148
  90. 90. Drucker DJ, Nauck MA. The incretin system: Glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet. 2006;368(9548):1696-1705
  91. 91. Gloyn AL, Drucker DJ. Precision medicine in the management of type 2 diabetes. The Lancet Diabetes and Endocrinology. 2018;6(11):891-900
  92. 92. Lederberg J, McCray AT. Ome SweetOmics—A genealogical treasury of words. The Scientist. 2001;15(7):8. Available from: https://go.gale.com/ps/i.do?id=GALE%7CA73535513&sid=googleScholar&v=2.1&it=r&linkaccess=abs&issn=08903670&p=AONE&sw=w
  93. 93. Relman DA, Falkow S. The meaning and impact of the human genome sequence for microbiology. Trends in Microbiology. 2001;9(5):206-208
  94. 94. Clavel T, Desmarchelier C, Haller D, Gérard P, Rohn S, Lepage P, et al. Intestinal microbiota in metabolic diseases: From bacterial community structure and functions to species of pathophysiological relevance. Gut Microbes. 2014;5(4):544-551
  95. 95. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The human microbiome project. Nature. 2007;449(7164):804-810
  96. 96. Clemente JC, Ursell LK, Parfrey LW, Knight R. The impact of the gut microbiota on human health: An integrative view. Cell. 2012;148(6):1258-1270
  97. 97. Kau AL, Ahern PP, Griffin NW, Goodman AL, Gordon JI. Human nutrition, the gut microbiome and the immune system. Nature. 2011;474(7351):327-336
  98. 98. Sato J, Kanazawa A, Ikeda F, Yoshihara T, Goto H, Abe H, et al. Gut dysbiosis and detection of “live gut bacteria” in blood of Japanese patients with type 2 diabetes. Diabetes Care. 2014;37(8):2343-2350
  99. 99. Candela M, Biagi E, Soverini M, Consolandi C, Quercia S, Severgnini 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
  100. 100. Hooks KB, O’Malley MA. Dysbiosis and its discontents. mBio. 2017;8:5
  101. 101. Carding S, Verbeke K, Vipond DT, Corfe BM, Owen LJ. Dysbiosis of the gut microbiota in disease. Microbial Ecology in Health and Disease. 2015;26. DOI: 10.3402/mehd.v26.26191
  102. 102. Toor D, Wasson MK, Kumar P, Karthikeyan G, Kaushik NK, Goel C, et al. Dysbiosis disrupts gut immune homeostasis and promotes gastric diseases. International Journal of Molecular Sciences. 2019;20(10):2432
  103. 103. Martinez JE, Kahana DD, Ghuman S, Wilson HP, Wilson J, Kim SCJ, et al. Unhealthy lifestyle and gut dysbiosis: A better understanding of the effects of poor diet and nicotine on the intestinal microbiome. Frontiers in Endocrinology. 2021;12:649
  104. 104. Magne F, Gotteland M, Gauthier L, Zazueta A, Pesoa S, Navarrete P, et al. The firmicutes/bacteroidetes ratio: A relevant marker of gut dysbiosis in obese patients? Nutrients. 2020;12(5):1474
  105. 105. Rogers GB, Keating DJ, Young RL, Wong ML, Licinio J, Wesselingh S. From gut dysbiosis to altered brain function and mental illness: Mechanisms and pathways. Molecular Psychiatry. 2016;21(6):738-748
  106. 106. Bistoletti M, Caputi V, Baranzini N, Marchesi N, Filpa V, Marsilio I, et al. Antibiotic treatment-induced dysbiosis differently affects BDNF and TrkB expression in the brain and in the gut of juvenile mice. PLoS One. 2019;14(2):e0212856
  107. 107. Macfarlane GT, Macfarlane S. Bacteria, colonic fermentation, and gastrointestinal health. Journal of AOAC International. 2012;95(1):50-60
  108. 108. Turnbaugh PJ, Gordon JI. The core gut microbiome, energy balance and obesity. Journal of Physiology. 2009;587(17):4153-4158
  109. 109. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology: Human gut microbes associated with obesity. Nature. 2006;444(7122):1022-1023
  110. 110. Chakraborti CK. New-found link between microbiota and obesity. World Journal of Gastrointestinal Pathophysiology. 2015;6(4):110
  111. 111. Petersen C, Round JL. Defining dysbiosis and its influence on host immunity and disease. Cellular Microbiology. 2014;16(7):1024-1033
  112. 112. Wei L, Singh R, Ro S, Ghoshal UC. Gut microbiota dysbiosis in functional gastrointestinal disorders: Underpinning the symptoms and pathophysiology. JGH Open. 2021;5(9):976-987. Available from: http://www.ncbi.nlm.nih.gov/pubmed/34584964
  113. 113. Larsen N, Vogensen FK, van den Berg FWJ, Nielsen DS, Andreasen AS, Pedersen BK, et al. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS One. 2010;5(2):e9085
  114. 114. Zhao L, Lou H, Peng Y, Chen S, Zhang Y, Li X. Comprehensive relationships between gut microbiome and faecal metabolome in individuals with type 2 diabetes and its complications. Endocrine. 2019;66(3):526-537
  115. 115. Harsch I, Konturek P. The role of Gut microbiota in obesity and type 2 and type 1 diabetes mellitus: New insights into “old” diseases. Medical Science. 2018;6(2):32
  116. 116. Kootte RS, Vrieze A, Holleman F, Dallinga-Thie GM, Zoetendal EG, de Vos WM, et al. The therapeutic potential of manipulating gut microbiota in obesity and type 2 diabetes mellitus. Diabetes, Obesity and Metabolism. 2012;14(2):112-120
  117. 117. Sedighi M, Razavi S, Navab-Moghadam F, Khamseh ME, Alaei-Shahmiri F, Mehrtash A, et al. Comparison of gut microbiota in adult patients with type 2 diabetes and healthy individuals. Microbial Pathogenesis. 2017;111:362-369
  118. 118. Aydin Ö, Nieuwdorp M, Gerdes V. The Gut microbiome as a target for the treatment of type 2 diabetes. Current Diabetes Reports. 2018;18(8):55
  119. 119. Lambeth SM, Carson T, Lowe J, Ramaraj T, Leff JW, Luo L, et al. Composition, diversity and abundance of gut microbiome in prediabetes and type 2 diabetes. Journal of Diabetes and Obesity. 2015;2(3):1-7
  120. 120. Zhao L, Zhang F, Ding X, Wu G, Lam YY, Wang X, et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science (1979). 2018;359(6380):1151-1156
  121. 121. Zhang F, Wang M, Yang J, Xu Q , Liang C, Chen B, et al. Response of gut microbiota in type 2 diabetes to hypoglycemic agents. Endocrine. 2019;66(3):485-493
  122. 122. Mallott EK, Amato KR. Butyrate production pathway abundances are similar in human and nonhuman primate gut microbiomes. Molecular Biology and Evolution. 2022;39(1):msab279. Available from: https://academic.oup.com/mbe/article/39/1/msab279/6372698
  123. 123. Nagpal R, Shively CA, Appt SA, Register TC, Michalson KT, Vitolins MZ, et al. Gut microbiome composition in non-human primates consuming a Western or Mediterranean diet. Frontiers in Nutrition. 2018;5:28
  124. 124. Clayton JB, Gomez A, Amato K, Knights D, Travis DA, Blekhman R, et al. The gut microbiome of nonhuman primates: Lessons in ecology and evolution. American Journal of Primatology. 2018;80(6):e22867. Available from: https://pubmed.ncbi.nlm.nih.gov/29862519/
  125. 125. Amato KR. Incorporating the gut microbiota into models of human and non-human primate ecology and evolution. American Journal of Physical Anthropology. 2016;159:S196-S215
  126. 126. Amato KR, Sanders JG, Song SJ, Nute M, Metcalf JL, Thompson LR, et al. Evolutionary trends in host physiology outweigh dietary niche in structuring primate gut microbiomes. ISME Journal. 2019;13(3):576-587
  127. 127. Manara S, Asnicar F, Beghini F, Bazzani D, Cumbo F, Zolfo M, et al. Microbial genomes from non-human primate gut metagenomes expand the primate-associated bacterial tree of life with over 1000 novel species. Genome Biology. 2019;20(1)
  128. 128. Wang J, Qin J, Li Y, Cai Z, Li S, Zhu J, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490(7418):55-60
  129. 129. Lazar V, Ditu LM, Pircalabioru GG, Picu A, Petcu L, Cucu N, et al. Gut microbiota, host organism, and diet trialogue in diabetes and obesity. Frontiers in Nutrition. 2019;6:21
  130. 130. Meijnikman AS, Gerdes VE, Nieuwdorp M, Herrema H. Evaluating causality of gut microbiota in obesity and diabetes in humans. Endocrine Reviews. 2018;39(2):133-153
  131. 131. Smits SA, Marcobal A, Higginbottom S, Sonnenburg JL, Kashyap PC. Individualized responses of gut microbiota to dietary intervention modeled in humanized mice. mSystems. 2016;1(5):e00098-e00116
  132. 132. le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500(7464):541-546
  133. 133. Gomez A, Sharma AK, Mallott EK, Petrzelkova KJ, Jost Robinson CA, Yeoman CJ, et al. Plasticity in the human gut microbiome defies evolutionary constraints. mSphere. 2019;4(4):e00271-e00319
  134. 134. Wu H, Esteve E, Tremaroli V, Khan MT, Caesar R, Mannerås-Holm L, et al. Metformin alters the gut microbiome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nature Medicine. 2017;23(7):850-858
  135. 135. Tito RY, Knights D, Metcalf J, Obregon-Tito AJ, Cleeland L, Najar F, et al. Insights from characterizing extinct human Gut microbiomes. PLoS One. 2012;7(12):e51146
  136. 136. Koo H, Hakim JA, Crossman DK, Kumar R, Lefkowitz EJ, Morrow CD. Individualized recovery of gut microbial strains post antibiotics. npj Biofilms and Microbiomes. 2019;5(1):30
  137. 137. Murphy EF, Cotter PD, Healy S, Marques TM, O’Sullivan O, Fouhy F, 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
  138. 138. Ochman H, Worobey M, Kuo CH, Ndjango JBN, Peeters M, Hahn BH, et al. Evolutionary relationships of wild hominids recapitulated by gut microbial communities. PLos Biol. 2010;8(11):e1000546
  139. 139. Kushugulova A, Forslund SK, Costea PI, Kozhakhmetov S, Khassenbekova Z, Urazova M, et al. Metagenomic analysis of gut microbial communities from a central Asian population. BMJ Open. 2018;8(7):e021682
  140. 140. Falony G, Joossens M, Vieira-Silva S, Wang J, Darzi Y, Faust K, et al. Population-level analysis of gut microbiome variation. Science (1979). 2016;352(6285):560-564
  141. 141. Karlsson FH, Tremaroli V, Nookaew I, Bergström G, Behre CJ, Fagerberg B, et al. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature. 2013;498(7452):99-103
  142. 142. Mahowald MA, Rey FE, Seedorf H, Turnbaugh PJ, Fulton RS, Wollam A, et al. Characterizing a model human gut microbiota composed of members of its two dominant bacterial phyla. Proceedings of the National academy of Sciences of the United States of America. 2009;106(14):5859-5864
  143. 143. Guarner F, Malagelada JR. Gut flora in health and disease. Lancet. 2003;361(9356):512-519
  144. 144. He Y, Wu W, Zheng HM, Li P, McDonald D, Sheng HF, et al. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nature Medicine. 2018;24(10):1532-1535
  145. 145. Furet JP, Kong LC, Tap J, Poitou C, Basdevant A, Bouillot JL, et al. Differential adaptation of human gut microbiota to bariatric surgery-induced weight loss: Links with metabolic and low-grade inflammation markers. Diabetes. 2010;59(12):3049-3057
  146. 146. Zhang X, Shen D, Fang Z, Jie Z, Qiu X, Zhang C, et al. Human gut microbiota changes reveal the progression of glucose intolerance. PLoS One. 2013;8(8):e71108
  147. 147. Koliada A, Syzenko G, Moseiko V, Budovska L, Puchkov K, Perederiy V, et al. Association between body mass index and Firmicutes/Bacteroidetes ratio in an adult Ukrainian population. BMC Microbiology. 2017;17(1):120
  148. 148. Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD, Gordon JI. Obesity alters gut microbial ecology. Proceedings of the National academy of Sciences of the United States of America. 2005;102(31):11070-11075
  149. 149. Turnbaugh PJ, Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, et al. A core gut microbiome in obese and lean twins. Nature. 2008;457(7228):480-484. Available from: https://www.nature.com/articles/nature07540
  150. 150. Velloso LA, Folli F, Sun XJ, White MF, Saad MJA, Kahn CR. Cross-talk between the insulin and angiotensin signaling systems. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(22):12490-12495. Available from: https://pubmed.ncbi.nlm.nih.gov/8901609/
  151. 151. Folli F, Kahn CR, Hansen H, Bouchie JL, Feener EP. Angiotensin II inhibits insulin signaling in aortic smooth muscle cells at multiple levels. A potential role for serine phosphorylation in insulin/angiotensin II crosstalk. The Journal of Clinical Investigation. 1997;100(9):2158-2169. Available from: https://pubmed.ncbi.nlm.nih.gov/9410892/
  152. 152. Li X, Wu Y, Song Y, Ding N, Lu M, Jia L, et al. Activation of NF-κB-inducing kinase in islet β cells causes β cell failure and diabetes. Molecular Therapy. 2020;28(11):2430-2441
  153. 153. Malle EK, Zammit NW, Walters SN, Koay YC, Wu J, Tan BM, et al. Nuclear factor κB-inducing kinase activation as a mechanism of pancreatic β cell failure in obesity. Journal of Experimental Medicine. 2015;212(8):1239-1254
  154. 154. Karin M, Delhase M. The IκB kinase (IKK) and NF-κB: Key elements of proinflammatory signalling. Seminars in Immunology. 2000;12(1):85-98
  155. 155. Meyerovich K, Fukaya M, Terra LF, Ortis F, Eizirik DL, Cardozo AK. The non-canonical NF-κB pathway is induced by cytokines in pancreatic beta cells and contributes to cell death and proinflammatory responses in vitro. Diabetologia. 2016;59(3):512-521
  156. 156. Aw W, Fukuda S. Understanding the role of the gut ecosystem in diabetes mellitus. Journal of Diabetes Investigation. 2018;9(1):5-12. Available from: https://onlinelibrary.wiley.com/doi/full/10.1111/jdi.12673
  157. 157. Gérard C, Vidal H. Impact of gut microbiota on host glycemic control. Frontiers in Endocrinology. 2019;10(JAN):29
  158. 158. Priyadarshini M, Navarro G, Layden BT. Gut microbiota: FFAR reaching effects on islets. Endocrinology. 2018;159(6):2495-2505. Available from: https://pubmed.ncbi.nlm.nih.gov/29846565/
  159. 159. Brubaker PL. Linking the gut microbiome to metabolism through endocrine hormones. Endocrinology. 2018;159(8):2978-2979
  160. 160. Kamada N, Seo SU, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nature Reviews. Immunology. 2013;13(5):321-335
  161. 161. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461(7268):1282-1286
  162. 162. Åkerfeldt MC, Howes J, Chan JY, Stevens VA, Boubenna N, McGuire HM, et al. Cytokine-induced β-cell death is independent of endoplasmic reticulum stress signaling. Diabetes. 2008;57(11):3034-3044. Available from: https://diabetesjournals.org/diabetes/article/57/11/3034/13408/Cytokine-Induced-Cell-Death-Is-Independent-of
  163. 163. Ehses JA, Böni-Schnetzler M, Faulenbach M, Donath MY. Macrophages, cytokines and β-cell death in type 2 diabetes. Biochemical Society Transactions. 2008;36(3):340-342
  164. 164. Tesi M, Bugliani M, Ferri G, Suleiman M, de Luca C, Bosi E, et al. Pro-inflammatory cytokines induce insulin and glucagon double positive human islet cells that are resistant to apoptosis. Biomolecules. 2021;11(2):1-11
  165. 165. Gao Z, Yin J, Zhang J, Ward RE, Martin RJ, Lefevre M, et al. Butyrate improves insulin sensitivity and increases energy expenditure in mice. Diabetes. 2009;58(7):1509-1517
  166. 166. Myers MA, Hettiarachchi KD, Ludeman JP, Wilson AJ, Wilson CR, Zimmet PZ. Dietary microbial toxins and type 1 diabetes. Annals of the New York Academy of Sciences. 2003;1005(1):418-422. Available from: https://onlinelibrary.wiley.com/doi/full/10.1196/annals.1288.071
  167. 167. Cummings JH, Pomare EW, Branch HWJ, Naylor CPE, MacFarlane GT. Short chain fatty acids in human large intestine, portal, hepatic and venous blood. Gut. 1987;28(10):1221-1227
  168. 168. Lin H, Frassetto A, Kowalik EJ, Nawrocki AR, Lu MM, Kosinski JR, et al. Butyrate and propionate protect against diet-induced obesity and regulate gut hormones via free fatty acid receptor 3-independent mechanisms. PLoS One. 2012;7(4):e35240
  169. 169. Al-Lahham SH, Peppelenbosch MP, Roelofsen H, Vonk RJ, Venema K. Biological effects of propionic acid in humans; Metabolism, potential applications and underlying mechanisms. Biochimica et Biophysica Acta. 2010;1801(11):1175-1183. Available from: https://pubmed.ncbi.nlm.nih.gov/20691280/
  170. 170. Sakakibara S, Yamauchi T, Oshima Y, Tsukamoto Y, Kadowaki T. Acetic acid activates hepatic AMPK and reduces hyperglycemia in diabetic KK-A(y) mice. Biochemical and Biophysical Research Communications. 2006;344(2):597-604. Available from: https://pubmed.ncbi.nlm.nih.gov/16630552/
  171. 171. Freeland KR, Wolever TMS. Acute effects of intravenous and rectal acetate on glucagon-like peptide-1, peptide YY, ghrelin, adiponectin and tumour necrosis factor-α. British Journal of Nutrition. 2010;103(3):460-466. Available from: https://www.cambridge.org/core/journals/british-journal-of-nutrition/article/acute-effects-of-intravenous-and-rectal-acetate-on-glucagonlike-peptide1-peptide-yy-ghrelin-adiponectin-and-tumour-necrosis-factor/69D7F8BC56D56DB34521824AE54923B0
  172. 172. Tang C, Offermanns S. FFA2 and FFA3 in metabolic regulation. Handbook of Experimental Pharmacology. 2017;236:205-220
  173. 173. Ulven T. Short-chain free fatty acid receptors FFA2/GPR43 and FFA3/GPR41 as new potential therapeutic targets. Frontiers in Endocrinology. 2012;3:111
  174. 174. Priyadarshini M, Villa SR, Fuller M, Wicksteed B, Mackay CR, Alquier T, et al. An acetate-specific GPCR, FFAR2, regulates insulin secretion. Molecular Endocrinology. 2015;29(7):1055-1066
  175. 175. Remely M, Aumueller E, Merold C, Dworzak S, Hippe B, Zanner J, et al. Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity. Gene. 2014;537(1):85-92
  176. 176. Priyadarshini M, Wicksteed B, Schiltz GE, Gilchrist A, Layden BT. SCFA receptors in pancreatic β cells: Novel diabetes targets? Trends in Endocrinology and Metabolism. 2016;27(9):653-664
  177. 177. Li NX, Brown S, Kowalski T, Wu M, Yang L, Dai G, et al. GPR119 agonism increases glucagon secretion during insulin-induced hypoglycemia. Diabetes. 2018;67(7):1401-1413
  178. 178. Mandøe MJ, Hansen KB, Hartmann B, Rehfeld JF, Holst JJ, Hansen HS. The 2-monoacylglycerol moiety of dietary fat appears to be responsible for the fat-induced release of GLP-1 in humans. The American Journal of Clinical Nutrition. 2015;102(3):548-555. Available from: https://pubmed.ncbi.nlm.nih.gov/26178726/
  179. 179. Pingitore A, Chambers ES, Hill T, Maldonado IR, Liu B, Bewick G, et al. The diet-derived short chain fatty acid propionate improves beta-cell function in humans and stimulates insulin secretion from human islets in vitro. Diabetes, Obesity & Metabolism. 2017;19(2):257-265
  180. 180. McNelis JC, Lee YS, Mayoral R, van der Kant R, Johnson AMF, Wollam J, et al. GPR43 potentiates β-cell function in obesity. Diabetes. 2015;64(9):3203-3217
  181. 181. Wang S, Yuan M, Zhang L, Zhu K, Sheng C, Zhou F, et al. Sodium butyrate potentiates insulin secretion from rat islets at the expense of compromised expression of β cell identity genes. Cell Death and Disease. 2011;13(1):67. Available from: /pmc/articles/PMC8770496/
  182. 182. Mitchell CM, Davy BM, Ponder MA, McMillan RP, Hughes MD, Hulver MW, et al. Prebiotic inulin supplementation and peripheral insulin sensitivity in adults at elevated risk for type 2 diabetes: A pilot randomized controlled trial. Nutrients. 2021;13(9):3235. Available from: https://pubmed.ncbi.nlm.nih.gov/34579112/
  183. 183. Ahluwalia B, Magnusson MK, Böhn L, Störsrud S, Larsson F, Öhman L, et al. Aloe barbadensis mill. Extract improves symptoms in IBS patients with diarrhoea: Post hoc analysis of two randomized double-blind controlled studies. Therapeutic Advances in Gastroenterology. 2021;14:17562848211048133. Available from: https://pubmed.ncbi.nlm.nih.gov/34646359/
  184. 184. Meyer D, Stasse-Wolthuis M. The bifidogenic effect of inulin and oligofructose and its consequences for gut health. European Journal of Clinical Nutrition. 2009;63(11):1277-1289. Available from: https://pubmed.ncbi.nlm.nih.gov/19690573/
  185. 185. Kimura I, Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, et al. The gut microbiota suppresses insulin-mediated fat accumulation via the short-chain fatty acid receptor GPR43. Nature Communications. 2013;4:1829
  186. 186. George Kerry R, Patra JK, Gouda S, Park Y, Shin HS, Das G. Benefaction of probiotics for human health: A review. Journal of Food and Drug Analysis. 2018;26(3):927-939
  187. 187. Abenavoli L, Scarpellini E, Colica C, Boccuto L, Salehi B, Sharifi-Rad J, et al. Gut microbiota and obesity: A role for probiotics. Nutrients. 2019;11(11):2690
  188. 188. Rastall RA, Gibson GR, Gill HS, Guarner F, Klaenhammer TR, Pot B, et al. Modulation of the microbial ecology of the human colon by probiotics, prebiotics and synbiotics to enhance human health: An overview of enabling science and potential applications. FEMS Microbiology Ecology. 2005;52(2):145-152
  189. 189. Tao YW, Gu YL, Mao XQ , Zhang L, Pei YF. Effects of probiotics on type II diabetes mellitus: A meta-analysis. Journal of Translational Medicine. 2020;18(1):30
  190. 190. Uusitalo U, Liu X, Yang J, Aronsson CA, Hummel S, Butterworth M, et al. Association of early exposure of probiotics and islet autoimmunity in the TEDDY study. JAMA Pediatrics. 2016;170(1):20-28
  191. 191. Ardeshirlarijani E, Tabatabaei-Malazy O, Mohseni S, Qorbani M, Larijani B, Baradar JR. Effect of probiotics supplementation on glucose and oxidative stress in type 2 diabetes mellitus: A meta-analysis of randomized trials. DARU, Journal of Pharmaceutical Sciences. 2019;27(2):827-837
  192. 192. Everard A, Lazarevic V, Derrien M, Girard M, Muccioli GM, Neyrinck AM, et al. Responses of gut microbiota and glucose and lipid metabolism to prebiotics in genetic obese and diet-induced leptin-resistant mice. Diabetes. 2011;60(11):2775-2786
  193. 193. Sun Z, Sun X, Li J, Li Z, Hu Q , Li L, et al. Using probiotics for type 2 diabetes mellitus intervention: Advances, questions, and potential. Critical Reviews in Food Science and Nutrition. 2020;60(4):670-683
  194. 194. Sun J, Buys NJ. Glucose- and glycaemic factor-lowering effects of probiotics on diabetes: A meta-analysis of randomised placebo-controlled trials. British Journal of Nutrition. 2016;115(7):1167-1177
  195. 195. Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM, Burcelin R. Improvement of glucose tolerance and hepatic insulin sensitivity by Oligofructose requires a functional glucagon-like peptide 1 receptor. Diabetes. 2006;55(5):1484-1490
  196. 196. Schmidt F, Zimmermann J, Tanna T, Farouni R, Conway T, Macpherson AJ. Noninvasive assessment of gut function using transcriptional recording sentinel cells. Science. 2022;376(6594):eabm6038

Written By

Shahzad Irfan, Humaira Muzaffar, Haseeb Anwar and Farhat Jabeen

Submitted: 02 May 2022 Reviewed: 31 May 2022 Published: 26 June 2022