the role of gut microbiota in mediating obesity and ...obese animals, the proportions of the gut...

15
1548 Abstract. OBJECTIVE: This review inspects the relations between the microbiota and the in- testinal immune system in the advancement of metabolic illnesses, such as obesity and diabe- tes mellitus. The role of the microbiota in intes- tinal immune defense and the control of metab- olism are subject to examination. MATERIALS AND METHODS: In type 1 diabe- tes, the adhesion proteins prompt inside the in- testinal epithelium prompt a more significant im- mune response that may result in the destruc- tion of pancreatic β cells by CD8+ T-lympho- cytes, as well as increased articulation of inter- leukin-17, which is associated with autoimmuni- ty. Studies suggest that the beginning of meta- bolic ailments and certain co-morbidities can be viewed in light of the protection between the gut microbiota and the intestinal immune system. The gut microbiota is analyzed as a key regu- lator of metabolic ailments. Research demon- strates that obese patients with type 2 diabetes have a certain gut microbiota and that the micro- biota is translocated from the gut to the tissues in conjunction with the illness, which instigates inflammation. RESULTS: Research in animals and people suggests that a probiotic supplement may reg- ulate the gut microbiota, thereby improving the prognosis for diabetes. CONCLUSIONS: The mechanism underlying this phenomenon relates to a decrease in the inflammatory reaction and oxidative stress, as well as a decrease in leaky gut. Such reactions increase insulin sensitivity and reduce autoim- mune responses. Key Words: Diabetes mellitus, Microbiota, Obesity, Inflamma- tion, Oxidative stress. Introduction The intestinal tract is populated by massive amounts of microscopic organisms that are col- lectively known as the “gut microbiota”. The mi- crobiota is characterized as the intricate arrange- ment of intestinal microflora that live in harmony with the host, which has advantages for both parties. When the gut microbiota remains steady and separated from epithelial cells, it benefits the host, especially with regards to energy and immune development 1 . Estimates indicate that a single individual’s microbiota is comprised of more than 100 trillion microorganisms, which are partitioned into 500-1,000 species. This number of microorganisms is ten times higher than the to- tal number of cells that comprise the human body. The aggregate genome of the microbiota weighs 1.5 kg and has more than 3.3 million genes, which is 150 times the entire human genetic heritage. These genes are the basis for the survival of the different species, most of which live in the colon. Most of the microbiota is composed of mi- croscopic organisms, although it also includes viruses, fungi, and eukaryotic cells. Intesti- nal organisms are mostly anaerobic. More than 50 phyla are known, but two are predominant and cover 90% of the bacterial population in both humans and mice. The first are Firmicutes (gram-positive), which constitute 60-80% of the microbiota and include more than 200 genera (of which the most significant are Ruminococcus, Clostridium, and Lactobacillus ). The second are Bacteroidetes (gram-negative, including Bac- teroides, Prevotella, and Xylanibacter), which make up 20-30% of the microbiota, followed by Actinobacteria (gram-positive), which con- stitute around 10% of the microbiota (with a predominance of the genus Bifidobacterium). Proteobacteria, such as Escherichia and Entero- bacteriaceae, are less common. Researchers propose that the microbiota of most humans can be classified as one of three prevalent enterotypes, which are directed by European Review for Medical and Pharmacological Sciences 2020; 24: 1548-1562 D. PITOCCO 1,2 , M. DI LEO 1 , L. TARTAGLIONE 1 , F. DE LEVA 1 , C. PETRUZZIELLO 1 , A. SAVIANO 1 , A. PONTECORVI 1 , V. OJETTI 1,3 1 Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy 2 Department of Endocrinology, Università Cattolica del Sacro Cuore, Rome, Italy 3 Department of Emergency Medicine, Università Cattolica del Sacro Cuore, Rome, Italy Corresponding Author: Dario Pitocco, MD; e-mail: [email protected] The role of gut microbiota in mediating obesity and diabetes mellitus

Upload: others

Post on 27-Jan-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

  • 1548

    Abstract. – OBJECTIVE: This review inspects the relations between the microbiota and the in-testinal immune system in the advancement of metabolic illnesses, such as obesity and diabe-tes mellitus. The role of the microbiota in intes-tinal immune defense and the control of metab-olism are subject to examination.

    MATERIALS AND METHODS: In type 1 diabe-tes, the adhesion proteins prompt inside the in-testinal epithelium prompt a more significant im-mune response that may result in the destruc-tion of pancreatic β cells by CD8+ T-lympho-cytes, as well as increased articulation of inter-leukin-17, which is associated with autoimmuni-ty. Studies suggest that the beginning of meta-bolic ailments and certain co-morbidities can be viewed in light of the protection between the gut microbiota and the intestinal immune system. The gut microbiota is analyzed as a key regu-lator of metabolic ailments. Research demon-strates that obese patients with type 2 diabetes have a certain gut microbiota and that the micro-biota is translocated from the gut to the tissues in conjunction with the illness, which instigates inflammation.

    RESULTS: Research in animals and people suggests that a probiotic supplement may reg-ulate the gut microbiota, thereby improving the prognosis for diabetes.

    CONCLUSIONS: The mechanism underlying this phenomenon relates to a decrease in the inflammatory reaction and oxidative stress, as well as a decrease in leaky gut. Such reactions increase insulin sensitivity and reduce autoim-mune responses.

    Key Words:Diabetes mellitus, Microbiota, Obesity, Inflamma-

    tion, Oxidative stress.

    Introduction

    The intestinal tract is populated by massive amounts of microscopic organisms that are col-

    lectively known as the “gut microbiota”. The mi-crobiota is characterized as the intricate arrange-ment of intestinal microflora that live in harmony with the host, which has advantages for both parties. When the gut microbiota remains steady and separated from epithelial cells, it benefits the host, especially with regards to energy and immune development1. Estimates indicate that a single individual’s microbiota is comprised of more than 100 trillion microorganisms, which are partitioned into 500-1,000 species. This number of microorganisms is ten times higher than the to-tal number of cells that comprise the human body. The aggregate genome of the microbiota weighs 1.5 kg and has more than 3.3 million genes, which is 150 times the entire human genetic heritage. These genes are the basis for the survival of the different species, most of which live in the colon.

    Most of the microbiota is composed of mi-croscopic organisms, although it also includes viruses, fungi, and eukaryotic cells. Intesti-nal organisms are mostly anaerobic. More than 50 phyla are known, but two are predominant and cover 90% of the bacterial population in both humans and mice. The first are Firmicutes (gram-positive), which constitute 60-80% of the microbiota and include more than 200 genera (of which the most significant are Ruminococcus, Clostridium, and Lactobacillus). The second are Bacteroidetes (gram-negative, including Bac-teroides, Prevotella, and Xylanibacter), which make up 20-30% of the microbiota, followed by Actinobacteria (gram-positive), which con-stitute around 10% of the microbiota (with a predominance of the genus Bifidobacterium). Proteobacteria, such as Escherichia and Entero-bacteriaceae, are less common.

    Researchers propose that the microbiota of most humans can be classified as one of three prevalent enterotypes, which are directed by

    European Review for Medical and Pharmacological Sciences 2020; 24: 1548-1562

    D. PITOCCO1,2, M. DI LEO1, L. TARTAGLIONE1, F. DE LEVA1, C. PETRUZZIELLO1, A. SAVIANO1, A. PONTECORVI1, V. OJETTI1,3

    1Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy2Department of Endocrinology, Università Cattolica del Sacro Cuore, Rome, Italy3Department of Emergency Medicine, Università Cattolica del Sacro Cuore, Rome, Italy

    Corresponding Author: Dario Pitocco, MD; e-mail: [email protected]

    The role of gut microbiota in mediating obesity and diabetes mellitus

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1549

    three distinct genera – Bacteroides, Prevotella, and Ruminococcus. The enterotypes are inde-pendent from age, sexual orientation, ethnicity, and weight history (BMI)2. The microorganisms are closely interlinked with each other and the host3.

    In recent years, the prevalence of metabolic illnesses in developed nations has increased ex-ponentially. Certain simultaneous developments must be considered, including changes in the gut microbiota. One aim is to clarify the molec-ular source of metabolic infections, even though the huge contrasts and social differences among humans make this problematic. Over the past fifty years, advances in molecular science have enabled specialists to look at the hereditary qual-ities of metabolic ailments. The gut microbiota is related to an assortment of metabolic capacities, such as the maturation and ingestion of undigest-ed carbohydrates, the retention of electrolytes and minerals, the balance of gut motility, and the synthesis of certain micronutrients4. Significant attention has been paid to the role of lactic acid in producing bacteria that lower pH, produce anti-microbial substances that affect pathogens, stim-ulate the immune system, integrate vitamins, and have anti-allergy effects (e.g., Bifidobacterium and Lactobacillus). In the past ten years, studies have pointed to a relationship between gut micro-biota and the development of metabolic issues, especially diabetes mellitus (DM) and obesity. In addition, microbial adjustments in the human bowel have been presented as a potential reason for obesity5. Along with its metabolic capacities, the microbiota interfaces with the immune sys-tem, where it supports the development of resis-tant cells and ordinary microbiota. Studies6,7 also have suggested that certain parts of the intestinal microbiota play key roles in various ailments, such as food allergies, malignant growths, incen-diary intestinal ailments, cardiovascular diseases, and dyslipidaemia.

    Main TextThe growing interest in microbiota has led to

    an exponential increase in the number of publica-tions focused on this topic. Efforts are underway to examine the connection between the microbi-ota and the increase in such diseases as DM and obesity8. Some hypotheses suggest that changes in the intestinal microbiota lead to increased intestinal porousness and a mucosal immune response, thereby adding to the advancement of DM. The decrease in tight junction proteins

    prompts increased intestinal permeability, with microorganisms encouraging the translocation of bacterial lipopolysaccharide (LPS), possibly leading to metabolic endotoxemia and insulin obstruction9,10. In one of the main trials involving obese animals, the proportions of the gut phyla Bacteroidetes and Firmicutes were changed by reducing Bacteroidetes and increasing Firmic-utes11. In a recent study12, the microscopic organ-isms were evident in external areas of the bodily fluid layer in non-obese, non-diabetic subjects, while in obese people with DM, microorganisms could be found in the thick internal bodily fluid and in close proximity to the epithelium (Fig-ure 1). The overall advantages of these phyla are associated with an increased ability to draw energy from food and low-grade inflammation. An increase in Firmicutes and a decrease in Bacteroidetes has been seen in obese mice. This is associated with the presence of explicit genes that encode proteins that make polysaccharides indigestible, as well as the creation of mono-saccharides and short-chain fatty acids (SCFA) in the liver (Figure 2). This system may prompt increased supplement take-up and deposition by the gut microbiota with a comparable increase in metabolic disorders13.

    Both environmental and genetic compo-nents add to the pathogenesis of type 2 diabetes (T2D)14,15. T2D is the result of an increase in the generation of glucose in the liver and a decrease in the emission and activity of insulin. In T2D, a poor-quality part is communicated in tissues as-sociated with digestion control, such as the liver, adipose tissue, and muscle tissue16. This meta-bolic inflammation is characterized by moderate cytokine generation, including interleukin-(IL-) 6, IL-1, or tumor necrosis factor alpha (TNFα), which adversely influence cell insulin flags and prompt insulin resistance and DM17,18. Low-grade inflammation starts with increased weight. Some researches19,20 report that the gut microbiota is a significant independent factor in the improve-ment of DM. The human gut has trillions of microorganisms, including more than 1,000 mi-croscopic organisms in 1,000 animal groups21. In this paper, we present information on changes in the gut microbiota in both type 1 diabetes (T1D) and T2D. We also summarize the mechanisms that may link the gut microbiota to DM. Various investigations have found changes in the gut mi-crobiota in diabetic groups. However, there are no reliable findings concerning the species that are affected by these modifications in diabetic

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1550

    patients. In addition, detailed investigations into the mechanisms that connect the gut microbiota to DM are lacking.

    T1D is a proinflammatory issue that results in the pulverization of pancreatic islet beta cells and the loss of insulin generation22. Although the triggers of T1D have not yet been determined, studies suggest that, in addition to hereditary qualities, ecological factors and microbial con-taminations might be significant23. Recent inves-tigations propose that the intestinal microbiome plays a key role in the mechanism(s) behind this proinflammatory issue24.

    T2D is associated with intestinal mucosal harm9,25, which might have several sources. First, small intestinal mucosal wounds may be caused by gut microbial dysbiosis in diabetic patients.

    In metagenome-wide research into gut microbial substance, the proportions of some butyrate-pro-ducing microorganisms were found to be reduced in patients with T2D, while the proportions of opportunistic pathogens were increased26. In ad-dition, Sato et al27 found a higher rate of gut microbes in the circulatory system in diabetic patients, which led them to propose that micro-scopic organisms translocate from the digestive tract to the circulatory system. Second, one of the consequences of DM, microangiopathy, might account for inflammatory variations in the small intestine. Third, diabetic neuropathy could cause intestinal wounds. Diabetic patients have a high-er rate of abnormal intestinal motility, which is attributed to autonomic neuropathy28. Intestinal motility disorders are common after effects of gut

    Figure 1. Changes to the gut microbiota and intestinal barrier function in DM. Bacterial encroachment – a shrinking of the “bug-free” zone adjacent to the colonic epithelium – is a feature of human metabolic syndrome. Encroachment is associated with inflammatory bowel disease and with obesity in patients with diabetes. Under normal physiological conditions in the lean state, the gut microbiota is highly diversified. The intestinal barrier prevents pathogenic bacteria from penetrating the gut through the production of mucin. Intestinal epithelial cells produce tolerogenic responses to microbe-specific molecules of commensal bacteria bound to pattern-recognition receptors by secreting anti-inflammatory mediators. In obesity and/or diabetes mellitus, the gut flora is diversified and creates an imbalance in the ratio of bacterial species. It reduces the production of mucin and other anti-microbial factors, which allows for easier penetration of bacteria beyond the gut barrier. Pro-inflammatory cytokines produced by immune cells in response to the inflammatory milieu impair the epithelial barrier. A weakened gut barrier allows leakage of bacterial products, such as bacterial lipopolysaccharide, across the barrier and into the systemic circulation. High levels of bacterial lipopolysaccharide and bacterial products cause endotoxemia and systemic inflammation, which worsen metabolic disease.

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1551

    microbial dysbiosis and bacterial translocation, which may lead to mucosal damage29. Fourth, the capacity of intestinal stem cells may change profoundly in DM patients. Recent researches30 suggest that the proportions of insulin-like devel-opment factor 1 (IGF-1) and insulin-like develop-ment factor-binding protein (IGFBP-3) change in DM patients.

    The Role of the Microbiota in DiabetesThe gut microbiota is part of a dynamic eco-

    system, and it is modified at the phylum and class levels by a variety of natural and host factors that affect the gut and other organs. Consequently, the gut microbiota is related to several human diseases, including obesity and DM31,32. Even though an understanding of the connection be-tween the gut microbiota and T2D is essential, there are still several gaps in our knowledge that should be addressed. For instance, the entire

    microscopic bacteria population of the gut has not been characterized by subjects’ age, sex, or medical treatment in order to limit sources of variation. The resistance of muscle and fat tissue to insulin activities evident in T2D is chiefly the consequence of a mix of hereditary factors, body composition, nutritional variables, and environ-mental elements. Insulin-receptor, glucose-trans-porter, and post-receptor changes are apparent in T2D. Moreover, peripheral tissues known to in-terminable compensatory hyperinsulinemia rise as resistant to insulin33.

    The intestinal microbiota plays a key role in metabolic diseases. In obese and diabetic sub-jects, researchers have found changes in the pro-portions of Firmicutes, Bacteroidetes, and Pro-teobacteria8. Pyrosequencing procedures have made significant advances in our understanding of the human microbiota. Diabetic patients have lower counts of Bifidobacterium and Faecali-bacterium prausnitzii, which are Gram-positive34 with anti-inflammatory properties35. In contrast to non-diabetic subjects, diabetic patients have lower levels of butyrate-creating bacteria, such as Roseburia intestinalis and F. prausnitzii, and elevated amounts of Lactobacillus gasseri (L. gasseri), Streptococcus mutans (S. mutans), and some strains of Clostridium (Table I). They also have higher levels of Proteobacteria, and ex-panded articulation of the microbiota qualities associated with oxidative pressure and inflam-mation36,37. In studies that compare metabolical-ly healthy individuals to insulin-resistant, obese individuals, noteworthy changes are found in the latter38.

    A few studies have attempted to characterize the impact of the microbiota on insulin resistance. One study demonstrated that the reduced number

    Figure 2. The action of gut microbiota is needed to digest some polysaccharides. The gut microbiota converts polysaccharides into monosaccharides and short-chain fatty acids (SCFA). These SCFAs are able to bind and activate two G-protein-coupled receptors (GPR41 and GPR43) of the gut epithelial cells. The activation of these receptors induces peptide YY. Starch digestion is an example of this process: H2 is produced and its increase inhibits starch digestion, at which point other bacterial groups work to transform the H2 into methane.

    Table I. Dominant bacteria in human microbiota.

    Firmicutes (60-80%)RuminiococcusClostridiumLactobacillusBacteroidetes (20-30%)BacteroidesPrevotellaXylanibacterActinobacteria (< 10%)BifidobacteriumProteobacteria (< 1%)EscherichiaEnterobacteriaceae

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1552

    of Bifidobacterium and Lactobacillus and the diminished ratio of Firmicutes to Bacteroidetes were fundamentally associated with higher plas-ma glucose levels, while the higher quantities of Clostridium were associated with a higher HbA1c in T1D. These results39,40 contradict the findings for T2D patients (Table II). These outcomes sug-gest some level of gut microbial dysbiosis in DM, even though there are differences in the affect-ed species. Other distinctions may be identified in conjunction with certain topographical areas, ages, or sexual orientations, as well as different food habits.

    The gut microbiota hydrolyzes and ferments the dietary polysaccharides to create monosac-charides and short-chain unsaturated fats (SC-FAs) that the host can utilize for energy. SC-FAs, such as acetate, propionate, and butyrate, generally supply 5% to 10% of human energy resources41. The signaling activities of SCFAs are affected by endogenous ligands of the G protein-coupled receptors 41 (GPR41) and 43 (GPR43), which are mainly represented in adipo-cytes and recognized as receptors of unsaturated fats42,43. DM, especially T2D, is related to a group of important lipid metabolism abnormalities44. At the point when contrasted and GF mice, the degree of triglycerides in mice was higher in the adipose tissues and the liver, while it was lower in serum. This suggests that the gut microbiota plays a role in lipid metabolism45. The mechanism through which unsaturated fats are discharged from triglyceride-rich lipoproteins to the mus-cle and heart is affected by the fasting-induced adipose factor46. The gut microbiota also builds an amalgamation of hepatic triglycerides and increases the production of hepatic triglycerides. The low-grade, chronic systemic inflammation

    contributes to the establishment of insulin resis-tance, DM, and obesity29,30. The increased circu-lating concentration of plasma lipopolysaccharide (LPS), which is described as metabolic endotox-emia, is a trigger in the safeguarding of a low-tone continuous inflammatory state in the host in light of high-fat weight-control diet plans9,47.

    Increase in EndotoxemiaT2D corresponds to a pro-inflammatory state

    with increased production of cytokines, such as IL-6, IL-1, or tumor necrosis factor, which nega-tively affects insulin’s interaction with its recep-tors and adds to insulin resistance and DM. In-creased weight appears to be one variable produc-ing this low-grade inflammation. Experiments on animals have demonstrated that adjustments in the microbiota may change the grade of adipose tissue inflammation.

    Increased flowing lipopolysaccharide (LPS) levels have been found in subjects with more prominent fat intake48,49. Lipopolysaccharide binding protein (LBP) is an acute-phase protein created in the liver that flows in the blood. LBP starts the recognition of LPS and strengthens the host’s immune response to LPS50, which focuses on the proportion of powerful lipopolysaccha-rides and could be a dependable biomarker that connects the lipopolysaccharide load to the sub-sequent immune response51. Circulating LBP is expanded in glucose-intolerant men52, and it is an inflammatory marker associated with obesi-ty-related insulin resistance53. Sun et al54 found that an increase in circulating LBP is related to obesity, the Mets, and T2D. LPS are consumed by enterocytes and conveyed in plasma, where they are bound to chylomicrons55. In mice, achieving a balance in the intestinal microbiota using pre-

    Table II. Bacterial species related to occurrence of insulin resistance and type 2 diabetes mellitus (T2DM).

    Increased in T2DM Decreasedin T2DM

    Bacterial phylum Firmicutes X Bacteroidetes X

    Bacterial species Roseburia X Eubacterium halii X Faecalibacterium prauznitzii X Lactobacillus gasseri X Streptococcus mutans X Escherichia coli X X

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1553

    biotics acts positively on the intestinal barrier, as it lowers the high-fat-diet-initiated LPS endo-toxemia, as well as systemic and liver inflamma-tion33,56. LPS plays a causative role, as the addi-tion of LPS to regular mice feed leads to insulin resistance in the liver, glucose tolerance, and an increase in adipose tissue. LPS ties to the CD14/TLR4 receptor in macrophages and prompts an increase in the creation of pro-inflammatory mol-ecules. When LPS infusions were controlled in mice with hereditary nonappearance of the CD14/TLR4 receptor, the mice did not exhibit these metabolic qualities, and they did not develop T2D or obesity. This demonstrates the key role of the CD14/TLR4 receptor for LPS. Likewise, the knockout CD14/TLR4 mice were much more sensitive to insulin than wild-type controls57. This suggests a change in the extent of gram-negative bacteria in the bowel in certain circumstances. On the other hand, there might be a distinction in intestinal permeability, with the goal that LPS increments in serum, which is legitimately iden-tified with the level of insulin resistance. A con-nection between LPS and T2D advancement been exhibited in some clinical trials48,57-60. In accor-dance with metabolic endotoxemia, live bacteria are translocated from the intestinal wall into the blood as T2D advances61,62. The multiplication of bacteria in the intestinal mucosa does not require an increase in glucose in the short term, but it may be related to the chronic, low-grade inflam-mation that leads to the insulin obstruction that characterizes T2D12.

    Changes in Incretin Emission Identified with Insulin Resistance and Beta Cell Work

    Research has demonstrated that increases in Bifidobacterium spp. decrease inflammation in obese mice by increasing the generation of gluca-gon-like peptide-1 (GLP-1). This diminishes in-testinal permeability. There is also evidence that the increase in Bifidobacterium spp. prompted by some prebiotics is connected to higher emissions of GLP-1 and peptide YY by the bowel. These two molecules have beneficial impacts, as they decrease insulin resistance and enhance beta cell function63.

    Variations in Gut MicrobiotaThe value of the microbiota and the ability

    to adjust it for medical purposes are no longer discussed. Results can be accomplished using prebiotics, probiotics, and antibiotics. The dietary

    routine changes the synthesis of the microbiota and the metagenome’s expression independently from the host genome64,65. Studies focused on the Mediterranean diet have provided significant data on the effects of diet on the gut microbiota of obese subjects. These effects help counteract the progression of T2D66. When obese subjects consume a low-calorie diet that is low in fat and sugars, they experience an increase in Bacte-roidetes and a decrease in Firmicutes67,68. The use of probiotics and prebiotics appears to alter gut microbiota and improve starch digestion69. Bacteria-fermenting carbohydrates, such as Bi-fidobacterium and Lactobacillus, have been giv-en as a key component of prebiotic treatments in populations of various ages67. When mice on a high-fat regimen were fed prebiotics con-taining oligofructose, their Bifidobacteria levels were reestablished, their endotoxemia declined, and their glucose tolerance improved70. Other research indicates that red wine consumption might balance the development of intestinal flora in humans by increasing the quantities of En-terococcus, Prevotella, Bacteroides, Bifidobac-terium, Bacteroides uniformis, Eggerthella lenta, Blautia coccoides, and Eubacterium rectangle, while diminishing LPS. This suggests that red wine polyphenols may have prebiotic benefits71,72. Another line of mediation is the utilization of pro-biotics containing live bacterial strains, including Bifidobacterium and Lactobacillus species, as dietary enhancements. These probiotics can pos-itively affect intestinal flora. In mice, probiotics containing certain strains of Lactobacillus have been found to have an antidiabetic impact with a concomitant decrease in endotoxemia56,73,74. Treatment of hereditarily obese mice fed a fat-rich regimen with anti-microbials (ampicillin in addition to neomycin) led to changes in their mi-crobiota and reductions in their insulin resistance and weight. In addition, animals given anti-toxin treatments exhibited a significant reduction in the degree of inflammation, oxidative pressure, and macrophage penetration in adipose tissue75,76. Another study77 demonstrated an improvement in starch digestion following the destruction of Helicobacter pylori.

    Gut microbiota transplantation was first uti-lized in patients who developed pseudomembra-nous colitis after Clostridium difficile contamina-tions that were treated with anti-infection agents. In these patients, fecal material transplanted from healthy donors seemed to reestablish microbial equalization in the gut by replacing the more

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1554

    pathogenic intestinal bacterial strains with other, more useful strains78. This treatment has been used in connection with other gastrointestinal illnesses, which has opened up for new uses in such ailments as obesity, DM, and cardiovascu-lar disease79,80. Some researches concentrate on males with insulin resistance and metabolic dis-orders who receive an autologous fecal microbio-ta transplant or an allogeneic transplant from thin donors80. Subjects who received transplants from thin donors experienced a significant improve-ment in peripheral sensitivity to insulin. They also experienced enhanced microbial diversity in the bowel and an increase in butyrate-producing microorganisms. Vrieze et al81 found changes in gut microbiota with significant convergences of Lactobacillus gasseri and Streptococcus mutans (both living in the proximal bowel), as well as Escherichia coli, which could help predict the probability of insulin-resistance advancement in postmenopausal women. The outcomes of these small-scale studies have not been reproduced in other groups, but they have motivated the devel-opment of better systems for recognizing the gut microbiota and its potential properties.

    Recent studies suggest the presence of syner-gies between the intestinal microbiota and the immune system in DM. The intestinal mucosa is a significant site for pathogen aggression. When uninjured, it acts as the primary line of defense against antigens. The intestinal wall is comprised of a layer of bodily mucus, IgA-secreting cells, antimicrobial peptides, and a complex epithelial barrier system shaped by the grip and tight in-tersections82. The immune response is regulated by the gut microbiota, and intestinal bacteria have been shown to affect the pathogenesis of T1D83. T1D is a notable immune-system illness that is characterized by lymphocytic infiltration or inflammation in pancreatic islets, which is typically identified with the invasion of innate immune cells. Increased intestinal permeability may increase the assimilation of antigens, which can damage pancreatic β cells84. Individuals sus-ceptible to T1D and other autoimmune system diseases exhibit intestinal hindrances85, which prompt an increase in antigens. The cytokines created by innate immune cells in the gut could disable β cells by advancing β cell apoptosis and islet-explicit T cell infiltration. This suggests a connection between intestinal microbiota and the immune system in T1D86,87.

    After treatment with a mix of antibiotics (met-ronidazole, neomycin, and polymyxin), nonobese

    diabetic (NOD) pregnant mice show significant variations in the peripheral composition of the T cell compartment, alterations in their offspring, and a higher recurrence of groups of separation (CD3+CD8+ T cells) in the mesenteric lymph nodes. Lymphocytic infiltration into the pancre-atic islets is also somewhat lower in the descen-dants of NOD mice and there are changes in the groups of gut microbiota88. Other findings indi-cate that treatment with vancomycin shields NOD mice from T1D in the early postnatal period and builds the group of separation (CD 4+ T cells). The proportions of Gram-positive and Gram-neg-ative microorganisms decline with the exception of Akkermansia muciniphila89. Akkermansia mu-ciniphila is abundant in the gut microbiota of healthy people. It can act locally by fortifying the gut obstruction (traded off in non-obese dia-betic mice), thereby enhancing the generation of bodily fluid and thickening the bodily fluid layer. This could maintain a strategic distance from its infiltration by inflammatory symbionts, which could lead to the observed decrease in plasma en-dotoxaemia90. The increased population of Akker-mansia muciniphila initiates Foxp3 regulatory T cells in instinctive adipose tissues and essentially improves glucose tolerance. These outcomes in-dicate that balancing the immune system through the use of Akkermansia muciniphila may be a potential treatment for DM91.

    T2D has traditionally been seen as a metabolic illness. However, Depommier et al92 suggest a relationship between the immune system and the pathogenesis of T2D. The obesity-related chronic inflammation and β cell stress caused by glu-cose toxicity and lipotoxicity could lead to both intrinsic and versatile resistance in T2D. The local or generalized immune system responses activate TLRs and nucleotide-restricting oligom-erization area (NOD) receptors, and enhance the creation of cytokines, such as IL-1β, which crush β cells92-94.

    An in-depth understanding of the effect of the gut microbiota and the invulnerable framework on DM allows for further investigation. The nat-ural invulnerable framework could be actuated by TLR, which initiates dendritic cell develop-ment and inflammatory cytokine discharge and supports the activation of T cells95. TLR5, a part of the innate immune system that is commu-nicated in the intestinal mucosa, is significant in the improvement of metabolic syndromes96. TLR5-deficient mice display hyperphagia and develop metabolic diseases, including hyperlipid-

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1555

    emia, hypertension, insulin resistance, and obe-sity. After transplantation of the gut microbiota from TLR5-deficient mice to their wide-type GF partners, an increase in proinflammatory cytokines and aspects of metabolic illnesses, in-cluding insulin resistance and obesity, have been found in the recipients97. The increased inflam-matory mediators in DM produce oxidative and endoplasmic reticulum stress in pancreatic islet β cells, which determine insulin sensitivity and glucose homeostasis98. This demonstrates that the cooperation between the immune system and the intestinal microbiota helps to improve metabolic disorders. However, the underlying mechanisms remain poorly understood and reliable outcomes have not been described. For instance, two recent studies99,100 found no association among insuffi-ciency of TLR5 in mice, changes in the gut mi-crobiota, and metabolic disorders, such as obesity and intestinal inflammation.

    Novel Strategies Focusing on the Role of Gut Microbiota in Diabetes

    Experimental and clinical investigations have demonstrated that focusing on the gut microbiota may be a powerful method for anticipating and overseeing DM101,102. The gut microbiota is mostly affected by diet. Prebiotics are food components that offer a health advantage by modifying the gut microbiota. They include inulin, fructo-oli-gosaccharides, galacto-oligosaccharides, and lac-tulose103. The restorative impacts of prebiotics on metabolic infections were affirmed in a prelimi-nary clinical trial. Six obese volunteers with T2D and hypertension were fed a strict vegetarian diet for one month. Their metabolic parameters, including body weight, triglyceride levels, and HbA1c, fundamentally improved, as did their fasting glucose and postprandial glucose. The vegetarian diet prompted compositional changes in the gut microbiota, including a diminished ra-tio of Firmicutes to Bacteroidetes and an increase in Bacteroides fragilis and Clostridium, which reduced intestinal irritation and SCFA levels104. Another area of therapeutic interest is probiotics – live microorganisms ingested as either food or supplements. Probiotics are a class of live microorganisms that, when ingested, may offer health advantages105, including immune-system stimulation, lower blood cholesterol, protection against respiratory and intestinal illnesses, and a decrease in fiery reactions. This is because pro-biotics have the ability to discharge antimicrobial substances, contend with different pathogens, and

    fortify the intestinal barrier106. Bifidobacteria and Lactobacilli are the most widely utilized strains in supplements107. Yadav et al108 shows that the in-gestion of 300 grams of a probiotic yogurt (Lac-tobacillus acidophilus La5 and Bifidobacterium lactis Bb12) per day for about a month and a half decreases blood 11 glucose and HbA1c levels in T2D patients. An antidiabetic impact of probiot-ics was also found in mice fed with a supplement of Lactobacillus acidophilus and Lactobacillus cases108. Qin et al26 demonstrate that subjects with T2D experience moderate intestinal dysbiosis and an increase in the quantity of opportunistic gut pathogens (more than an adjustment in a par-ticular microbial animal variety) that are direct-ly connected with the pathophysiology of T2D. Moreover, they found a reduction in butyrate-cre-ating microbes. An analysis of genetic, bacteri-al capacities suggests an increase in capacities identified with the reaction to intestinal oxidative stress, which are associated with an inclination toward diabetic complications109. Moreover, an ongoing report indicates that metformin, which is widely used in the treatment of T2D, prompts an increase in Escherichia and Bifidobacterium and a decrease in Intestinibacter110.

    Fecal transplants have been subjected to ex-panding consideration as a therapeutic proce-dure111. Vrieze et al112 considered the impacts of moving the intestinal microbiota from thin sub-jects to male recipients with metabolic syndrome. A month and a half after the introduction of the gut microbiota, the insulin affectability of the recipients and their degrees of butyrate-creating intestinal microbiota had both increased funda-mentally111. The outcomes of the exchange of gut microbiota for DM in mice have also been exam-ined. After the exchange of microorganisms from MyD88-inadequate NOD mice, insulitis, and the beginning of DM improved. In addition, follow-ing oral transfers of fecal bacteria for more than three weeks, the gut microbiota was modified in terms of an increase in Lachnospiraceae and Clostridiaceae and a decrease in Lactobacillace-ae112. These outcomes demonstrate that the oral organization of gut microbiota may be a conceiv-able method for improving insulin affectability in DM. In any case, the safety of fecal microbiota transplantation should be explored, as negative impacts have also been found. For instance, two patients passed on in an examination, including 18 patients who experienced bacterial transplan-tations to treat intermittent Clostridium difficile colitis113. Clinical studies and basic discourse

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1556

    on fecal microbiota transplantation should help decide whether this method is useful for diabetic patients in general. In addition, more attention should be paid to the wellbeing, safety, and com-position of donor microbiota.

    Aas et al114 found that the intestinal microbi-ome in obese patients could not effectively utilize tryptophan (TRP), a significant amino acid con-

    stituent of human proteins and the reason for the generation of cell transmitters. In obese patients, TRP was utilized to deliver kynurenine instead of being utilized to create indoles (anti-inflammato-ry compounds), which had negative consequenc-es for glucose tolerance. Another situation for the connections among intestinal microbiome, in-flammatory, and cardiometabolic hazard has been

    Figure 3. Schematic reproduction of probiotic actions in Type 1 and Type 2 diabetes. Probiotic utilization increases the number of bifidobacteria and expanded expression of adhesion proteins reduces intestinal permeability, thereby damaging the activation of TLR4 by LPS. Consequently, NFkB activation pathways are impeded. The induction of TH17 cells is also repressed, which avoids pancreatic infiltration of CD8+ T cells.

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1557

    speculated. Interleukin-22 and the estimation of kynurenine/tryptophan are two new elements that could immediately be brought into clinical practice as a therapeutic agent (IL-22) and as a biomarker of intestinal dysbiosis (KYN/TRP) in the testing of metabolic syndrome.

    Conclusions

    The establishment of a link between gut micro-biota and metabolic ailments has recently moti-vated exponential advances in research. Multiple factors lead to changes in gut microbiota and affect the relationship between gut microbiota and T2D. Molecular mechanisms, including the anti-diabetic impacts of probiotics, have not been fully investigated, but they might include a de-crease in oxidative stress, immunomodulation, attenuation of inflammation, and modification of the intestinal microbiota (Figure 3)115. In the future, larger human investigations that consider the majority of the conceivable confounding vari-ables (e.g., age, gender, ethnicity, diet, and genetic factors) should enable us to effectively utilize the gut microbiota’s composition and develop novel indicative or therapeutic strategies for the treat-ment of metabolic diseases.

    Conflict of InterestThe Authors declare that they have no conflict of interests.

    References

    1) Hooper LV, Macpherson aJ. Immune adaptations that maintain homeostasis with the intestinal mi-crobiota. Nature Rev Immunol 2010; 10: 159-169.

    2) aruMugaM M, raes J, peLLetier e, Le pasLier D, YaMa-Da t, MenDe Dr, FernanDes gr, tap J, BruLs t, Bat-to JM, BertaLan M, BorrueL n, caseLLas F, FernanDez L, gautier L, hansen t, hattori M, haYashi t, KLeer-eBezeM M, KuroKawa K, LecLerc M, LeVenez F, Man-ichanh c, nieLsen hB, nieLsen t, pons n, pouLain J, Qin J, sicheritz-ponten t, tiMs s, torrents D, ugar-te e, zoetenDaL eg, wang J, guarner F, peDersen o, De Vos wM, BrunaK s, Doré J; Metahit consortiuM, antoLín M, artiguenaVe F, BLottiere hM, aLMeiDa M, Brechot c, cara c, cherVaux c, cuLtrone a, DeLo-rMe c, Denariaz g, DerVYn r, Foerstner Ku, Friss c, Van De guchte M, gueDon e, haiMet F, huBer w, Van hYLcKaMa-VLieg J, JaMet a, Juste c, Kaci g, KnoL J, LaKhDari o, LaYec s, Le roux K, Maguin e, Mérieux a, MeLo MinarDi r, M’rini c, MuLLer J, oozeer r, parKh-iLL J, renauLt p, rescigno M, sanchez n, sunagawa s,

    torreJon a, turner K, VanDeMeuLeBroucK g, VareLa e, winograDsKY Y, zeLLer g, weissenBach J, ehrLich sD, BorK p. Enterotypes of the human gut microbiome. Nature 2011; 473: 174-180.

    3) roDriguez-VaLera F, Martin-cuaDraDo aB, roDri-guez-Brito B, pasić L, thingstaD tF, rohwer F. Ex-plaining microbial population genomics though phage predation. Nature Rev Microbiol 2009; 7: 828-836.

    4) giLL r, pop M, DeBoY rt, ecKBurg pB, turnBaugh pJ, saMueL Bs. Metagenomic analysis of the human distal gut microbiome. Science 2006; 312: 1355-1359.

    5) LeY re, BacKheD F, turnBaugh p, Lozupone ca, Knight rD, gorDon Ji. Obesity alters gut micro-bial ecology. Proc Natl Acad Sci USA 2005; 102: 11070-11075.

    6) hoLMes e, Li JV, athanasiou t, ashraFian h, nich-oLson JK. Understanding the role of gut micro-biome-host metabolic signal disruption in health and disease. Trends Microbiol 2011; 19: 349-359.

    7) Larsen n, Vogensen FK, Berg Fw, nieLsen Ds, an-Dreasen as, peDersen BK, aL-souD wa, sørensen sJ, hansen Lh, JaKoBsen M. Gut microbiota in human adults with type 2 diabetes differs from non-dia-betic adults. PLoS One 2010; 5: e9085.

    8) LeY re, turnBaugh pJ, KLein s, gorDon Ji. Microbi-al ecology: human gut microbes associated with obesity. Nature 2006; 444, 1022-1023.

    9) cani pD, BiBiLoni r, KnauF c, waget a, neYrincK aM, DeLzenne nM, BurceLin r. Changes in gut microbiota control metabolic endotoxemia-in-duced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008; 57: 1470-1481.

    10) seconDuLFo M, LaFusco D, carratu r, DeMagistris L, sapone a, generoso M, MezzogioMo a, sasso Fc, cartenì M, De rosa r, prisco F, esposito V. Ultra-structural mucosal alterations and increased in-testinal permeability in non-celiac, type I diabetic patients. Dig Liver Dis 2004; 36: 35-45.

    11) LeY re. Obesity and the human microbiome. Curr Opin Gastroent 2010; 26: 5-11.

    12) chassaing B, raJa sM, Lewis JD, sriniVasan s, gewirtz at. Colonic microbiota encroachment correlates with dysglycemia in humans. Cell Mol Gastroen-terol Hepatol 2017; 4: 205-221.

    13) ereJuwa oo, suLaiMan sa, wahaB Msw. Modulation of gut microbiota in the management of metabolic disorders: the prospects and challenges. Int J Mol Sci 2014; 15: 4158-4188.

    14) rhoDes cJ. Type 2 diabetes-a matter of beta-cell life and death? Science 2005; 307: 380-384.

    15) hoFe cr, Feng L, zephYr D, stroMBerg aJ, hennig B, gaetKe LM. Fruit and vegetable intake, as re-flected by serum carotenoid concentrations, pre-dicts reduced probability of polychlorinated bi-phenyl-associated risk for type 2 diabetes: Na-tional Health and Nutrition Examination Survey 2003-2004. Nutr Res 2014; 34: 285-293.

    16) picKup Jc, crooK Ma. Is type II diabetes mellitus a

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1558

    disease of the innate immune system? Diabetolo-gia 1998; 41: 1241-1248.

    17) hotaMisLigiL gs. Inflammation and metabolic disor-ders. Nature 2006; 444: 860-867.

    18) shoeLson s, Lee J, goLDFine a. Inflammation and in-sulin resistance. J Clin Invest 2006; 116, 1793-1801.

    19) BurceLin r, serino M, chaBo c, BLasco-BaQue V, Am-ar J. Gut microbiota and diabetes: from patho-genesis to therapeutic perspective. Acta Diabetol 2011; 48, 257-273.

    20) nichoLson JK, hoLMes e, Kinross J, BurceLin r, giB-son g, Jia w, pettersson s. Host-gut microbiota metabolic interactions. Science 2012; 336: 1262-1267.

    21) neish as. Microbes in gastrointestinal health and disease. Gastroenterology 2009; 136: 65-80.

    22) roDriguez-VaLera F, Martin-cuaDraDo aB, roDri-guez-Brito B, pasi´c L, thingstaD tF, rohwer F, Mi-ra a. Explaining microbial population genomics though phage predation. Nature Rev Microbiol 2009; 7: 828-836.

    23) giLL sr, pop M, DeBoY rt, ecKBurg, turnBaugh pJ, saMueL Bs, gorDon Ji, reLMan Da, Fraser-Liggett cM, neLson Ke. Metagenomic analysis of the hu-man distal gut microbiome. Science 2006; 312: 1355-1359.

    24) cani pD, J. aMar J, M.a. igLesias Ma, poggi M, KnauF c, BasteLica D, neYrincK aM, FaVa F, tuohY KM, chaBo c, waget a, DeLMée e, cousin B, suLpice t, chaMontin B, Ferrières J, tanti JF, giBson gr, casteiLLa L, DeLzenne nM, aLessi Mc, BurceLin r. Metabolic endotoxemia initiates obesity and insu-lin resistance. Diabetes 2007; 56: 1761-1772.

    25) LeY re, BacKheD F, turnBaugh p, Lozupone ca, Knight rD, gorDon Ji. Obesity alters gut micro-bial ecology. Proc Natl Acad Sci USA 2005; 102: 11070-11075.

    26) Qin J, Li Y, cai z, Li s, zhu, zhang JF, Liang s, zhang w, guan Y, shen D, peng Y, zhang D, Jie z, wu w, Qin Y, xue w, Li J, han L, Lu D, wu p, Dai Y, sun x, Li z, tang a, zhong s, Li x, chen w, xu r, wang M, Feng Q, gong M, Yu J, zhang Y, zhang M, hansen t, sanchez g, raes J, FaLonY g, oKuDa s, aLMeiDa M, LechateLier e, renauLt p, pons n, Batto JM, zhang z, chen h, Yang r, zheng w, Li s, Yang h, wang J, eh-rLich sD, nieLsen r, peDersen o, Kristiansen K, wang J. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012; 490: 55-60.

    27) sato J, Kanazawa a, iKeDa F, Yoshihara Y, goto h, aBe h, KoMiYa K, Kawaguchi M, shiMizu t, ogihara t, taMura Y, saKurai Y, YaMaMoto r, Mita t, FuJitani Y, FuKuDa h, noMoto K, taKahashi t, asahara t, hi-rose t, nagata s, YaMashiro Y, wataDa h. Gut dys-biosis and detection of “live gut bacteria” in blood of Japanese patients with type 2 diabetes. Diabe-tes Care 2014; 37: 2343-2350.

    28) weLLen Ke, hotaMisLigiL gs. Inflammation, stress, and diabetes. J Clin Invest 2005; 115: 1111-1119.

    29) Ding s, chi MM, scuLL Bp, rigBY r, schwerBrocK nM,

    Magness s, JoBin c, LunD pK. High-fat diet: bacte-ria interactions promote intestinal inflammation which precedes and correlates with obesity and insulin resistance in mouse. PLoS One 2010; v5: e12191.

    30) zhong hJ, Yuan Y, xie wr, chen Mh, he xx. Type 2 diabetes mellitus is associated with more seri-ous small intestinal mucosal injuries. PLoS One 2016; 11: e0162354.

    31) Qin J, Li r, raes J, aruMugaM M, BurgDorF Ks, Man-ichanh c, nieLsen t, pons n, LeVenez F, YaMaDa t, MenDe Dr, Li J, xu J, Li s, Li D, cao J, wang B, Liang h, zheng h, xie Y, tap J, Lepage p, BertaLan M, Bat-to JM, hansen t, Le pasLier D, LinneBerg a, nieLsen hB, peLLetier e, renauLt p, sicheritz-ponten t, turn-er K, zhu h, Yu c, Li s, Jian M, zhou Y, Li Y, zhang x, Li s, Qin n, Yang h, wang J, BrunaK s, Doré J, guarner F, Kristiansen K, peDersen o, parKhiLL J, weis-senBach J; Metahit consortiuM, BorK p, ehrLich sD, wang J. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010; 464: 59-65.

    32) huMan MicroBioMe proJect c. Structure, function and diversity of the healthy human microbiome. Nature 2012; 486: 207-214.

    33) aBDuL-ghani Ma, DeFronzo ra. Pathogenesis of insulin resistance in skeletal muscle. J Biomedic Biotechnol 2010, 476279: 2010.

    34) Furet Jp, Kong Lc, tap J, poitou c, BasDeVant a, BouiLLot JL, Mariat D, corthier g, Doré J, henegar c, rizKaLLa s, cLéMent K. Differential adaptation of human gut microbiota to bariatric surgery-in-duced weight loss: links with metabolic and low-grade inflammation markers. Diabetes 2010; 59: 3049-3057.

    35) o’MahonY D, MurphY s, BoiLeau t, parK J, o’Brien F, groeger D, Konieczna p, ziegLer M, scuLLY p, shanahan F, KieLY B, o’MahonY L. Bifidobacteri-um animalis AHC7 protects against pathogen-in-duced NF-kappaB activation in vivo. BMC Immu-nol 2010; 11: 63.

    36) paLau-roDriguez M, tuLipan s, Queipo-ortuño Mi, urpi-sarDa M, tinahones FJ, anDres-LacueVa c. Me-tabolomic insight into the intricate gut microbial host interaction in the development of obesity and type 2 diabetes. Front Microbiol 2015; 6: 1151.

    37) tiLg h, Moschen ar. Microbiota and diabetes: an evolving relationship. Gut 2014; 63: 1513-1521.

    38) serino M, FernánDez-reaL JM, garcía-Fuentes e, Que-ipo-ortuño Mi, Moreno-naVarrete JM, sánchez a. The gut microbiota profile is associated with insu-lin action in humans. Acta Diabetol 2013; 50: 753-761.

    39) KarLsson Fh, treMaroLi V, nooKaew i, BergströM g, Behre cJ, FagerBerg B, nieLsen J, BäcKheD F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013; 498: 99-103.

    40) Murri M, LeiVa i, goMez-zuMaQuero JM, tinahones FJ, carDona F, soriguer F, Queipo-ortuño Mi. Gut

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1559

    microbiota in children with type 1 diabetes differs from that in healthy children: a case-control study. BMC Medicine 2013; 11: 46-50.

    41) McneiL ni. The contribution of the large intestine to energy supplies in man. Am J Clin Nutr 1984; 39: 338-342.

    42) Dentin r, J.p. pegorier, F. BenhaMeD, F. FouFeLLe, p. Ferré, V. FauVeau, Magnuson Ma, girarD J, postic c. Hepatic glucokinase is required for the syner-gistic action of ChREBP and SREBP-1c on glyco-lytic and lipogenic gene expression. J Biol Chem 2004; 279: 20314-20326.

    43) Brown aJ, goLDsworthY sM, Barnes aa, eiLert MM, tcheang L, DanieLs D, Muir ai, wiggLesworth MJ, Kinghorn i, Fraser nJ, piKe nB, struM Jc, stepLews-Ki KM, MurDocK pr, hoLDer Jc, MarshaLL Fh, sze-Keres pg, wiLson s, ignar DM, FoorD sM, wise a, DoweLL sJ. The Orphan G protein-coupled recep-tors GPR41 and GPR43 are activated by propio-nate and other short chain carboxylic acids. J Bi-ol Chem 2003; 278: 11312-11319.

    44) Krauss rM. Lipids and lipoproteins in patients with type 2 diabetes. Diabetes Care 2004; 27: 1496-1504.

    45) VeLagapuDi Vr, hezaVeh r, reigstaD cs, gopaLacha-rYuLu p, YetuKuri L, isLaM s, FeLin J, perKins r, Borén J, oresic M, BäcKheD F. The gut microbiota modu-lates host energy and lipid metabolism in mice. J Lip Res 2010; 51: 1101-1112.

    46) BäcKheD F, Manchester JK, seMenKoVich cF, gorDon Ji. Mechanisms underlying the resistance to di-et-induced obesity in germ-free mice. P Natl Acad Sci USA 2007; 104: 979-984.

    47) cherVonsKY a. Innate receptors and microbes in induction of autoimmunity. Curr Opin Immunol 2009; 21: 641-647.

    48) creeLY sJ, Mcternan pg, KusMinsKi cM, Fisher FM, Da siLVa nF, KhanoLKar M. Lipopolysaccharide ac-tivates an innate immune system response in hu-man adipose tissue in obesity and type 2 diabe-tes. Am J Phys-End Met 2007; 292: E740-E747.

    49) erriDge c, attina t, spicKett cM, weBB DJ. A high-fat meal induces low-grade endotoxemia: evidence of a novel mechanism of postprandial inflamma-tion. Am J Clin Nutr 2007; 86: 1286-1292.

    50) schuMann rr, Leong sr, FLaggs gw, graY pw, wright sD, Mathison Jc, toBias ps, uLeVitch rJ. Structure and function of lipopolysaccharide binding protein. Science 1990; 249: 1429-1433.

    51) Lepper p, schuMann c, triantaFiLou K, rasche F, schus-ter t, FranK h, schneiDer e, triantaFiLou M, Von eY-natten M. Association of lipopolysaccharide-bind-ing protein and coronary artery disease in men. J Am Coll Cardiol 2007; 50: 25-31.

    52) guBern c, Lopez-BerMeJo a, Biarnes J, VenDreLL J, ri-cart w, FernanDez-reaL JM. Natural antibiotics and insulin sensitivity: the role of bactericidal/perme-ability-increasing protein. Diabetes 2006; 55: 216-224.

    53) Moreno-naVarrete JM, ortega F, serino M, Luche e, waget a, parDo g, saLVaDor J, ricart w, FrühBecK g,

    BurceLin r, FernánDez-reaL JM. Circulating lipopoly-saccharide-binding protein (LBP) as a marker of obesity-related insulin resistance. J Obes 2012; 36: 1442-1449.

    54) sun L, z. Yu, x. Ye, s. zou, h. Li, D. Yu, wu h, chen Y, Dore J, cLéMent K, hu FB, Lin x. A mark-er of endotoxemia is associated with obesity and related metabolic disorders in apparently healthy Chinese. Diabetes Care 2010: 33: 1925-1932.

    55) cani pD, DeLzenne nM. The role of the gut microbi-ota in energy metabolism and metabolic disease. Curr Pharm Des 2009; 15, 1546-1558.

    56) cani pD, neYrincK aM, FaVa F, KnauF c, BurceLin rg, tuohY KM, giBson gr, DeLzenne nM. Selec-tive increases of bifidobacteria in gut micro-flora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007; 50: 2374-2383.

    57) pussinen pJ, haVuLinn as, Lehto M, sunDVaLL J, sa-LoMMa V. Endotoxemia is associated with an in-creased risk of incident diabetes. Diabetes Care 2011; 34: 392-397.

    58) Dixon an, VaLsaMaKis g, haniF Mw, FieLD a, Boutsi-aDis a, harte a, Mcternan pg, Barnett ah, KuMar s. Effect of the orlistat on serum endotoxin lipopoly-saccharide and adipocytokines in South Asian individuals with impaired glucose tolerance. Int J Clin Pract 2008; 62, 1124-1129.

    59) aL-attas ao, aL-Daghri nM, aL-ruBeaan K, Da siL-Va nF, saBico sL, KuMar s, Mcternan pg, harte aL. Changes in endotoxin levels in T2DM subjects on anti-diabetic therapies. Cardiovasc Diabetol 2009; 8: 20.

    60) ghaniM h, aBuaYsheh s, sia cL, KorzeniewsKi K, chauDhuri a, FernanDez-reaL JM, DanDona p. In-crease in plasma endotoxin concentrations and the expression of Toll-like receptors and suppres-sor of cytokine signaling-3 in mononuclear cells after a high-fat, high-carbohydrate meal: implica-tions for insulin resistance. Diabetes Care 2009; 32: 2281-2287.

    61) aMar J, chaBo c, waget a, KLopp p, Vachoux c, Ber-MúDez-huMarán Lg, sMirnoVa n, Bergé M, suLpice t, Lahtinen s, ouwehanD a, LangeLLa p, rautonen n, sansonetti pJ, BurceLin r. Intestinal mucosal ad-herence and translocation of commensal bacte-ria at the early onset of type 2 diabetes: molec-ular mechanisms and probiotic treatment. EMBO Mol Med 2011; 3: 559-572.

    62) aMar J, serino M, Lange c, chaBo c, iacoVoni J, MonDot s, Lepage p, KLopp c, Mariette J, Bouchez o, perez L, courtneY M, Marre M, KLopp p, Lantieri o, Doré J, charLes M, BaLKau B, BurceLin r; D.e.s.i.r. stuDY group. Involvement of tissue bacteria in the onset of diabetes in humans: evidence for a con-cept. Diabetologia 2011; 54: 3055-3061.

    63) VeLasQuez-ManoFF M. Gut microbiome: the peace-keepers. Nature 2015; 518: S3-S11.

    64) FaLK pg, hooper LV, MiDtVeDt t, gorDon Ji. Creat-ing and maintaining the gastrointestinal ecosys-

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1560

    tem: what we know and need to know from gno-tobiology. Microbiol Mol Biol Rev 1998; 62, 1157-1170.

    65) parashar a, uDaYaBanu M. Gut microbiota regulates key modulators of social behavior. Eur Neuropsy-chopharmacol 2016; 26: 78-91.

    66) haro c, Montes-Borrego M, rangeL-zúniga oa, aL-caLá-Díaz JF, góMez-DeLgaDo F, pérez-Martínez p, DeLgaDo-Lista J, Quintana-naVarro gM, tinahones FJ, LanDa BB, López-MiranDa J, caMargo a, pérez-JiMénez F. Two healthy diets modulate gut microbi-al community improving insulin sensitivity in hu-man obese population. J Clin Endocrinol Metab 2016; 101: 233-242.

    67) wu gD, chen J, hoFFMann c, Bittinger K, chen YY, KeiLBaugh sa, Bewtra M, Knights D, waLters wa, Knight r, sinha r, giLroY e, gupta K, BaLDassano r, nesseL L, Li h, BushMan FD, Lewis JD. Linking long-term dietary patterns with gut microbial entero-types. Science 2011; 334: 105-108.

    68) cotiLLarD a, KenneDY sp, Kong Lc, priFti e, pons n, Le chateLier e, aLMeiDa M, QuinQuis B, LeVenez F, gaLLeron n, gougis s, rizKaLLa s, Batto JM, renauLt p; anr MicrooBes consortiuM, Doré J, zucKer JD, cLéMent K, ehrLich sD. Dietary intervention impact on gut microbial gene richness. Nature 2013; 500: 585-588.

    69) rastaLL ra, giBson gr, giLL hs, guarner F, KLaenhaMMer tr, pot B, reiD g, rowLanD ir, sanD-ers Me. Modulation of the microbial ecology of the human colon by probiotics, prebiotics and sym-biotics to enhance human health: an overview of enabling science and potential applications. FEMS Microbiol Ecol 2005; 52: 145-152.

    70) MeYer D, stasse-woLthuis M. The bifidogenic effect of inulin and oligofructose and its consequenc-es for gut health. Eur J Clin Nutr 2009; 63: 1277-1289.

    71) Queipo-ortuño Mi, Boto-orDóñez M, Murri M, go-Mez-zuMaQuero JM, cLeMente-postigo M, estruch r, carDona Diaz F, anDrés-LacueVa c, tinahones FJ. In-fluence of red wine polyphenols and ethanol on the gut microbiota ecology and biochemical bio-markers. Am J Clin Nutr 2012; 95: 1323-1334.

    72) cLeMente-postigo M, Queipo-ortuño Mi, Boto-or-Doñez M, coin-araguez L, roca-roDriguez MM, DeL-gaDo-Lista J, carDona F, anDres-LacueVa c, tina-hones FJ. Effect of acute and chronic red wine consumption on lipopolysaccharide concentra-tions. Am J Clin Nutr 2013; 10, 1-9.

    73) YaDaV h, Jain s, sinha pr. Antidiabetic effect of pro-biotic dahi containing Lactobacillus acidophilus and Lactobacillus casei in high fructose fed rats. Nutrition 2007; 23: 62-68.

    74) Boto-orDóñez M, urpi-sarDa M, Queipo-ortuño Mi, tuLipani s, tinahones FJ, anDres-LacueVa c. High levels of bifidobacteria are associated with in-creased levels of anthocyanin microbial metabo-lites: a randomized clinical trial. Food Funct 2014; 5: 1932-1938.

    75) hernánDez e, BargieLa r, Diez Ms, FrieDrichs a, pérez-coBas ae, gosaLBes MJ, Knecht h, Martínez-

    Martínez M, seiFert J, Von Bergen M, artacho a, ruiz a, caMpoY c, Latorre a, ott sJ, MoYa a, suárez a, Martins Dos santos Va, Ferrer M. Functional con-sequences of microbial shifts in the human gas-trointestinal tract linked to antibiotic treatment and obesity. Gut Microbes 2013; 4: 306-315.

    76) MeMBrez M, BLancher F, JaQuet M, BiBiLoni r, cani pD, BurceLin rg, corthesY i, Macé K, chou cJ. Gut microbiota modulation with norfloxacin and ampi-cillin enhances glucose tolerance in mice. FASEB J 2008; 22: 2416-2426.

    77) roca-roDríguez MM, coín-aragüez L, corneJo-pare-Ja i, aLcaiDe J, cLu-FernánDez c, Muñoz-garach a, Durán-Martín e, Mora-naVas L, góMez-pérez aM, MoLina-Vega M, Díaz-perDigones c, Mancha-DoBLas i, tinahones FJ. Carbohydrate metabolism im-provement after Helicobacter pylori eradication. Diab Metab 2016; 42, 130-134.

    78) Van nooD e, Vrieze a, nieuwDorp M, Fuentes s, zoe-tenDaL eg, De Vos wM, Visser ce, KuiJper eJ, BarteLs-Man JF, tiJssen Jg, speeLMan p, DiJKgraaF Mg, KeLLer JJ. Duodenal infusion of donor feces for recurrent Clos-tridium difficile. N Engl J Med 2013; 368: 407-415.

    79) sMits Lp, Bouter Kec, De Vos wM, BoroDY tJ, nieu-wDorp M. Therapeutic potential of fecal microbi-ota transplantation. Gastroenterology 2013; 145: 946-953.

    80) Kootte rs, Vrieze a, hoLLeMan F, DaLLinga-thie gM, zoetenDaL eg, De Vos wM, groen aK, hoeKstra JB, stroes es, nieuwDorp M. The therapeutic potential of manipulating gut microbiotain obesity and type 2 diabetes mellitus. Diabetes Obes Metab 2012; 14: 112-120.

    81) Vrieze a, Van nooD e, hoLLeMan F, saLoJärVi J, Kootte rs, BarteLsMan JF, DaLLinga-thie gM, acKer-Mans Mt, serLie MJ, oozeer r, Derrien M, Druesne a, Van hYLcKaMa VLieg Je, BLoKs Vw, groen aK, heiL-ig hg, zoetenDaL eg, stroes es, De Vos wM, hoeK-stra JB, nieuwDorp M. Transfer of intestinal micro-biota from lean donors increases insulin sensitivi-ty in individuals with metabolic syndrome. Gastro-enterology 2012; 143: 913-916.

    82) ohLanD cL, Macnaughton wK. Probiotic bacte-ria and intestinal epithelial barrier function. Am J Physiol Gastrointest Liver Physiol 2010; 298: G807-G819.

    83) atKinson Ma, cherVonsKY a. Does the gut microbio-ta have a role in type 1 diabetes? Early evidence from humans and animal models of the disease. Diabetologia 2012; 55: 2868-2877.

    84) VehiK K, DaBeLea D. The changing epidemiology of type 1 diabetes: why is it going through the roof? Diabetes Metab Res Rev 2011; 27: 3-13.

    85) neu J, reVerte cM, MacKeY aD, LiBoni K, tuhaceK-te-nace LM, hatch M, Li n, caiceDo ra, schatz Da, at-Kinson M. Changes in intestinal morphology and permeability in the biobreeding rat before the on-set of type 1 diabetes. J Ped Gastroenterol Nutr 2005; 40: 589-595.

    86) szaBLewsKi L. Role of immune system in type 1 dia-betes mellitus pathogenesis. Int Immunopharma-col 2014; 22: 182-191.

  • The role of gut microbiota in mediating obesity and diabetes mellitus

    1561

    87) VaaraLa o. Is the origin of type 1 diabetes in the gut? Immunol Cell Biol 2012; 90: 271-276.

    88) torMo-BaDia n, håKansson å, VasuDeVan K, MoLin g, ahrné s, ciLio cM. Antibiotic treatment of preg-nant non-obese diabetic (NOD) mice leads to al-tered gut microbiota and intestinal immunological changes in the offspring. Scand J Immunol 2014; 80: 250-260.

    89) hansen ch, KrYch L, nieLsen Ds, Vogensen FK, han-sen Lh, sørensen sJ, BuscharD K, hansen aK. Ear-ly life treatment with vancomycin propagates Ak-kermansia muciniphila and reduces diabetes inci-dence in the NOD mouse. Diabetologia 2012; 55: 285-2294.

    90) hänninen a, toiVone r, pöYsti s, BeLzer c, pLoVier h, ouwerKerK Jp, eMani r, cani pD, De Vos wM. Ak-kermansia muciniphila induces gut microbiota re-modelling and controls islet autoimmunity in NOD mice. Gut 2018; 67: 1445-1453.

    91) Mithieux g. Does Akkermansia muciniphila play a role in type 1 diabetes? Gut 2018; 67: 1373-1374.

    92) DepoMMier c; eVerarD a; Druart c; pLoVier h; Van huL M; Vieira-siLVa s; FaLonY g; raes J; Maiter D; DeLzenne nM; De BarsY M; LouMaYe a; herMans Mp; thissen Jp; De Vos wM; cani pD. Supplemen-tation with Akkermansia muciniphila in over-weight and obese human volunteers: a proof-of-concept exploratory study. Nat Med 2019; 25: 1096-1103.

    93) Matzinger p. The danger model: a renewed sense of self. Science 2002; 296: 301-305.

    94) Donath MY, shoeLson se. Type 2 diabetes as an inflammatory disease. Nat Rev Immunol 2011; 11: 98-107.

    95) MeDzhitoV r. Origin and physiological roles of in-flammation. Nature 2011; 454: 428-435.

    96) Letran se, Lee sJ, atiF sM, ueMatsu s, aKira s, Mc-sorLeY sJ. TLR5 functions as an endocytic recep-tor to enhance flagellinspecific adaptive immuni-ty. Eur J Immunol 2011; 41: 29-38.

    97) ViJaY-KuMar M, aitKen JD, carVaLho Fa, cuLLenDer tc, Mwangi s, sriniVasan s, sitaraMan sV, Knight r, LeY re, gewirtz at. Metabolic syndrome and al-tered gut microbiota in mice lacking Toll-like re-ceptor 5. Science 2010; 328: 228-231.

    98) hasnain sz, Borg DJ, harcourt Be, tong h, sheng Yh, ng cp, Das i, wang r, chen ac, LouDoVaris t, KaY tw, thoMas he, whiteheaD Jp, ForBes JM, prins JB, McgucKin Ma. Glycemic control in diabetes is restored by therapeutic manipulation of cytokines that regulate beta cell stress. Nat Med 2014; 20: 1417-1426.

    99) chassaing B, LeY re, gewirtz at. Intestinal epithe-lial cell toll-like receptor 5 regulates the intesti-nal microbiota to prevent low-grade inflammation and metabolic syndrome in mice. Gastroenterol-ogy 2014; 147: 1363-1377.

    100) uBeDa c, LipuMa L, goBourne a, ViaLe a, Leiner i, eQuinDa M, Khanin r, paMer eg. Familial trans-mission rather than defective innate immuni-

    ty shapes the distinct intestinal microbiota of TLR deficient mice. J Exp Med 2012; 209: 1445-1456.

    101) stenMan LK, waget a, garret c, KLopp p, BurceLin r, Lahtinen s. Potential probiotic Bifidobacteri-um animalis ssp. lactis 420 prevents weight gain and glucose intolerance in diet-induced obese mice. Benef Microbes 2014; 5: 437-445.

    102) ogawa a, KaDooKa Y, Kato K, shirouchi B, sato M. Lactobacillus gasseri SBT2055 reduces post-prandial and fasting serum nonesterified fatty ac-id levels in Japanese hypertriacylglycerolemic subjects. Lipids Health Dis 2014; 13: 36-44.

    103) pineiro M, asp ng, reiD g, MacFarLane s, MoreL-Li L, Brunser o, tuohY K. FAO Technical Meet-ing on Prebiotics. J Clin Gastroenterol 2008; 42: S156-S159.

    104) KiM Ms, hwang ss, parK eJ, Bae Jw. Strict vege-tarian diet improves the risk factors associated with metabolic diseases by modulating gut mi-crobiota and reducing intestinal inflammation. Environ Microbiol Rep 2013, 5: 765-775.

    105) who-worLD heaLth organization: Evaluation of Health and Nutritional Properties of Probiotics in Food Including Powder Milk with Live Lactic Ac-id Bacteria, Joint WHO/FAO Expert Consultation on Geneva. Cordoba, Argentina: WHO; 2001.

    106) giLL h, prasaD J. Probiotics, immunomodulation, and health benefits. Adv Exp Med Biol 2008; 606: 423-454.

    107) prasaD J, giLL hs, sMart JB, gopaL pK. Selection and characterization of Lactobacillus and Bifido-bacterium strains for use as probiotics. Int Dairy J 1998; 8: 993-1002.

    108) YaDaV h, Jain s, sinha pr. Antidiabetic effect of probiotic dahi containing Lactobacillus acidoph-ilus and Lactobacillus casei in high fructose fed rats. Nutrition 2007; 23: 62-68.

    109) eJtaheD hs, MohtaDi-nia J, hoMaYouni-raD a, ni-aFar M, asghari-JaFaraBaDi M, MoFiD V. Probiotic yogurt improves antioxidant status in type 2 dia-betic patients. Nutrition 2012; 28, 539-543.

    110) KashYap p, Farrugia g. Oxidative stress: key play-er in gastrointestinal complications of diabetes. Neurogastroenterol Motil 2011; 23: 111-114.

    111) wu h, esteVe e, treMaroLi V, Khan Mt, caesar r, Mannerås-hoLM L, ståhLMan M, oLsson LM, serino M, pLanas-FèLix M, xiFra g, MercaDer JM, torrents D, BurceLin r, ricart w, perKins r, FernànDez-reaL JM, BäcKheD F. Metformin alters the gut microbi-ome of individuals with treatment-naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med 2017; 23: 850-858.

    112) Vrieze a, Van nooD e, hoLLeMan F, saLoJärVi J, Kootte rs, BarteLsMan JF, DaLLinga-thie gM, acKerMans Mt, serLie MJ, oozeer r, Derrien M, Druesne a, Van hYLcKaMa VLieg Je, BLoKs Vw, groen aK, heiLig hg, zoetenDaL eg, stroes es, De Vos wM, hoeKstra JB, nieuwDorp M. Transfer of intestinal microbiota from lean donors increas-es insulin sensitivity in individuals with meta-

  • D. Pitocco, M. Di Leo, L. Tartaglione, F. De Leva, C. Petruzziello, A. Saviano, A. Pontecorvi, V. Ojetti

    1562

    bolic syndrome. Gastroenterology 2012; 143: 913-916.

    113) peng J, narasiMhan s, Marchesi Jr, Benson a, wong Fs, wen L. Long term effect of gut micro-biota transfer on diabetes development. J Auto-immun 2014; 53: 85-94.

    114) aas J, gessert ce, BaKKen Js. Recurrent Clostridium difficile colitis: case series involving 18 patients treated with donor stool administered via a naso-gastric tube. Clin Infect Dis 2003; 36: 580-585.

    115) Laurans L, VentecLeF n, haDDaD Y, chaJaDine M, aLzaiD F, MetghaLchi s, soVran B, Denis rgp, Dai-rou J, carDeLLini M, Moreno-naVarrete JM, strauB M, Jegou s, McQuittY c, VieL t, esposito B, taVi-tian B, caLLeBert J, LuQuet sh, FeDerici M, Fernan-Dez-reaL JM, BurceLin r, LaunaY JM, teDgui a, MaL-Lat z, soKoL h, taLeB s. Genetic deficiency of in-doleamine 2,3-dioxygenase promotes gut mi-crobiota mediated metabolic health. Nat Med 2018; 24: 1113-1120.