SciELO - Scientific Electronic Library Online

 
vol.39 número2Los posibles mecanismos de pardeamiento del tejido adiposo blanco: una diana novedosa para el tratamiento de la obesidadGuía Práctica ESPEN: nutrición clínica en las enfermedades del hígado índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Nutrición Hospitalaria

versión On-line ISSN 1699-5198versión impresa ISSN 0212-1611

Nutr. Hosp. vol.39 no.2 Madrid mar./abr. 2022  Epub 09-Mayo-2022

https://dx.doi.org/10.20960/nh.03866 

REVIEWS

Will intestinal flora therapy become a new target in type-2 diabetes mellitus? A review based on 13 clinical trials

¿Puede la terapia de flora intestinal convertirse en un nuevo objetivo para la diabetes mellitus de tipo 2? Revisión basada en 13 ensayos clínicos

Fujie Wang1  , Ting Zhao1  , Weiwei Wang1  , Qianqian Dai2  , Xianghua Ma1 

1Nutritional Department. The First Affiliated Hospital of Nanjing Medical University. Nanjing, People's Republic of China

2Nutritional Department. Xuzhou Cancer Hospital. Xuzhou, People's Republic of China

Abstract

Background:

diabetes mellitus (DM) is a chronic disease and its pathogenesis is still inconclusive. Current evidence suggests an association between intestinal flora and type-2 diabetes mellitus (T2DM). In this paper, we summarized the current research, determining whether intestinal flora may become a new method to treat T2DM, and providing a theoretical basis and literature references for the prevention of T2DM based on the regulation of intestinal flora.

Method:

we carried out a review based on 13 published clinical trials to determine the correlation between T2DM and intestinal flora, and between changes in clinical outcomes and in intestinal flora in the development of T2DM; to assess the pathological mechanisms; and to discuss the treatment of diabetes based on intestinal flora.

Results:

we found that intestinal flora is involved in the occurrence and development of T2DM. Several pathological mechanisms may be involved in the process, including improving the gut barrier, alleviating inflammation, increasing glucagon-like peptide (GLP) 1 and GLP 2, increasing the production of short-chain fatty acids (SCFAs), and so on. Several measures based on intestinal flora, including exercise, food, specific diets, drugs and probiotics, would be used to treat and even prevent T2DM.

Conclusions:

high-quality studies are required to better understand the clinical effects of intestinal flora in T2DM.

Keywords: Intestinal flora; T2DM; Therapy

Resumen

Antecedentes:

la diabetes mellitus (DM) es una enfermedad crónica cuya patogénesis no está clara. La evidencia actual sugiere una asociación entre la flora intestinal y la diabetes mellitus de tipo 2 (DMT2). Este artículo revisa la investigación actual para determinar si la flora intestinal puede ser un nuevo método de tratamiento de la diabetes mellitus de tipo 2 y proporciona la base teórica y las referencias de la literatura para la prevención de la diabetes mellitus de tipo 2 basada en la regulación de la flora intestinal.

Métodos:

se revisaron 13 ensayos clínicos publicados para determinar la correlación entre la DMT2 y la flora intestinal, los cambios en los resultados clínicos y los cambios en la flora intestinal durante el desarrollo de la DMT2; para resumir su mecanismo patogénico; y, desde el punto de vista de la flora intestinal, para explorar el tratamiento de la diabetes.

Resultados:

se encontró que la flora intestinal estaba involucrada en el desarrollo de la diabetes mellitus de tipo 2. Este proceso puede implicar una variedad de mecanismos patológicos, incluyendo la mejora de la barrera intestinal, la reducción de la inflamación, el aumento del péptido similar al glucagón (GLP) 1 y GLP 2, y el aumento del rendimiento de los ácidos grasos de cadena corta (SCFA). Algunas medidas basadas en la flora intestinal, como el ejercicio, los alimentos, las dietas especiales, los medicamentos y los probióticos, se utilizarán para tratar e incluso prevenir la DMT2.

Conclusión:

se necesitan estudios de alta calidad para comprender mejor los efectos clínicos de la flora intestinal en los pacientes con diabetes mellitus de tipo 2.

Palabras clave: Flora intestinal; Diabetes mellitus de tipo 2; Tratamiento

INTRODUCTION

Nowadays, diabetes mellitus has gradually become an increasingly striking social health problem around the world. In 2017, the eighth edition of the International Diabetes Federation (IDF) Diabetes Atlas showed that there were about 425 million diabetics worldwide and the number may increase to 700 million by 2045 (1). The incidence of diabetes mellitus (DM) in China is about 10 % (2), and the number has reached 114 million, amounting to 1/3 of the total of global diabetics. T2DM accounts for more than 90 % of total diabetes patients (3,4).

The intestinal flora is to date considered a complex organ composed of 500-1000 species and 1014 bacteria, which is more than 10 times the number of human cells, including bacteria, viruses, fungi, and protozoa, that are commensal with the human intestinal tract (5). Among these, bacteria represent the best studied group and will be the main focus of this review. Overall the predominant bacterial groups in the microbiome are Gram-positive Firmicutes and Gram-negative Bacteroidetes (6,7). Studies have shown that lots of chronic diseases may be related to intestinal microecological disorders (8), that intestinal flora is an important factor among environmental factors, and that its changes are related to a series of metabolic diseases such as obesity and DM (9,10).

At the same time, a lot of factors can contribute to changes in intestinal flora. Firstly, it is now understood that diet plays a significant role in shaping the microbiome, with experiments showing that dietary alterations can induce large, temporary microbial shifts within 24 h. The gut microbiota of patients with type-2 diabetes has been functionally characterized with diabetes-associated markers, showing enriched membrane transport of sugars and branched-chain amino acids, xenobiotic metabolism, and sulfate reduction along with decreased bacterial chemotaxis, butyrate synthesis, and metabolism of cofactors and vitamins (11). Revealed by metagenome analysis, gut microbiota composition transforms through early stages of human development, and is influenced by the diet (12). Secondly, regular exercise has an anti-inflammatory effect, which improves the immunological profile in type-2 diabetes mellitus (13). Thirdly, the intestinal flora changes with aging. Besides these, some other factors, such as drugs (16-17), lifestyle (30), etc., also play a role in the development of intestinal flora.

It is of huge importance to fully understand the changes of intestinal flora in T2DM, and the pathological mechanism of involvement of intestinal flora in the development of T2DM. Similarly, an in-depth study of risk factors associated with bacterial reduction in patients with T2DM is needed, including the impact of exercise, food, specific diets, drugs and probiotics. In this article, we carried out a review based on several published clinical trials to determine the correlation between T2DM and intestinal flora. We hope that this article may provide a theoretical basis and literature references for the regulation of intestinal flora in the treatment of T2DM and its complications.

MATERIALS AND METHODS

IDENTIFICATION OF ELIGIBLE STUDIES

One search strategy was run using "Intestinal flora" and "Type 2 diabetes mellitus" with no limitations. Besides, another search strategy was also run using the terms "Intestinal flora" and "Type 2 diabetes mellitus" limited to "humans" and "clinical trial". A broad search of the English-language literature for randomized controlled trials (RCTs) in patients with T2DM was performed by using Cochrane, Medline, PubMed, Central Register of Controlled Trials, Web of Science, and trial registry databases. All the relevant publications were reviewed, and duplications of articles from the two search strategies were avoided. The articles in reference lists were also hand-searched for potentially relevant publications. The search was conducted by two investigators. Any disagreements were resolved by consensus with involvement of the third author.

INCLUSION AND EXCLUSION CRITERIA

Human-associated studies, regardless of year of publication, would be included if they met the following criteria: 1) RCT; 2) patients had been diagnosed with T2DM; 3) were older than 18 years; 4) sufficient data were available of clinical outcomes. Studies would be excluded if they met the following criteria: animal experiment, review, mechanism research, case report, collection of papers, literature with incomplete data and/or duplicates, and no full manuscript availability. Participants would be excluded if they 1) had severe conditions, including digestive dysfunction, heart failure, renal failure, malignant tumors, severe cerebrovascular diseases, ketosis, hyperthyroidism, liver dysfunction, or severe gallbladder and pancreatic diseases; 2) were pregnant or lactating; 3) suffered from mental illness; 4) used anti-depressants, sedatives, neurological or psychiatric medications; 5) required insulin therapy or had arterial hypertension and dyslipidemia controlled by statins and either ace-inhibitors or angiotensin receptor blockers; 6) had diabetes-specific complications and ischemic heart disease; 7) lacked the ability to perform physical activities.

DATA EXTRACTION

Two investigators extracted the data independently and reached a consensus on all items. For each study, the following information was collected: first author, year of publication, sample size, mean age, gender, anthropometric measurements (weight, height, body mass index (BMI), waist circumference, and body composition by bioelectrical impedance analysis), dietary record, trial duration, and clinical outcomes. Clinical outcomes included: glycated hemoglobin (HbA1c), low-density lipoprotein-cholesterol (LDL), high-density lipoprotein-cholesterol (HDL), cholesterol (CHOL), triglycerides (TG), aspartate aminotransferase (AST) and alanine transaminase (ALT), free fatty acids, CRP, homeostasis model assessment of insulin resistance (HOMA-IR), biochemical analyses, fasting GLP-1 concentration, and gut microbiota. Data could be extracted separately as long as there was enough information available in the trials.

RESULTS

LITERATURE SEARCH

We used the terms "Intestinal flora" and "Type 2 diabetes me­llitus" with a broad search for randomized controlled trials (RCTs) in patients with T2DM. The results: Cochrane (700), Medline (733), PubMed (711), Central Register of Controlled Trials (688), Web of Science (699). In the end, a total of 744 articles (all published) were retrieved from the databases. A total of 360 articles about animal experiments, 262 articles with reviews, 9 articles on meta-analyses, 3 articles in letter format, 1 case report, 15 articles without full text, and 69 articles about non-randomized controlled trials were excluded, and 12 articles with incomplete data were also excluded. Thus, a total of 13 publications met the inclusion and exclusion criteria, and details from these trials were extracted separately. Figure 1 shows a flowchart of article selection and inclusion. Because of the heterogeneity found among patients and trial methods, and the large variety of outcome measurements used in these trials, the pooling of data for a meta-analysis was inappropriate. Results were, therefore, summarized qualitatively.

Figure 1.  Flow diagram illustrating the literature search and evaluation. 

STUDY CHARACTERISTICS

Details from 13 eligible published trials are discussed in table I. Table I summarizes the characteristics of the 13 trials. The number of participants in these trials ranged from 12 to 504, with median age ranging from 41 to 72 years. Trial duration ranged from 1 to 6 months, and intervention included exercise, food, specific diets, drugs, and probiotics. The intestinal flora of all patients in our study were classified in categories.

Table I.  Baseline characteristics of the references included 

NR: not reported; LCD: low carbohydrate diets; WTP: wholegrain, 80 traditional Chinese medicinal foods and prebiotics.

CLINICAL OUTCOMES

Effect of exercise on intestinal flora in diabetes

A total of 2 trials in table II demonstrated that the exercise group had shown significant improvements of lean mass from baseline amomg diabetes patients. Otherwise, fat mass and fasting glucose had decreased obviously. According to Evasio Pasini et al. (15), those who received exercise training had improved blood sugar, as well as functional and anthropometric variables. Moreover, chronic exercise reduced intestinal fungus overgrowth, leaky gut, and systemic inflammation. As well, in the research by Yan Liu et al. (22), the microbiome of responders exhibited an enhanced capacity for biosynthesis of short-chain fatty acids and catabolism of branched-chain amino acids.

Table II.  Raw outcomes for change of clinical indicators and intestinal flora through exercise by individual trials 

Data is expressed as mean ± SD.

Effect of food and specific diets on intestinal flora in diabetes

A total of 3 trials involved food and specific diets with effects measured on intestinal flora (Table III). They improved HbA1c (p < 0.01) and helped with weight loss. In the study of Mengxiao Ren et al. (14), A-LCD significantly increased the short-chain fatty acid (SCFAs)-producing bacteria Roseburia, Ruminococcus and Eubacterium. Liping Zhao et al. (19) showed that a select group of SCFA-producing strains was promoted by dietary fibers and that most other potential producers were either diminished or unchanged in patients with T2DM. However, in the trial by Yanislava Karusheva et al. (26), the oral glucose sensitivity index was 24 % (p < 0.01) and circulating growth factor 21 was 21 % higher (p < 0.05), whereas meal-derived insulin secretion was 28 % lower (p < 0.05). Then it could be seen that the effect of food and specific diets was obvious in improving glucose in diabetics, and may impact some pathological mechanisms.

Table III.  Raw outcomes for change of clinical indicators and intestinal flora through food and specific diets by individual trials 

Data is expressed as mean ± SD.

Effect of drugs on intestinal flora in diabetes

In the work by Hao Wu et al. (16), their findings provide support for the notion that an altered gut microbiota mediates some of metformin's antidiabetic effects. In the study of Xiaolin Tong et al. (17), they also found that both metformin and AMC significantly alleviated hyperglycemia and shifted gut microbiota structure in diabetic patients.

They significantly increased a co-abundant group represented by Blautiaspp, which significantly correlated with the improvements in glucose. Yifei Zhang et al. (20) measured alterations in gut microbiota using oral intake of probiotics or berberine (BBR), a bacteriostatic agent. All 3 trials demonstrated drugs had an impact on intestinal flora (Table IV). The three studies suggested that after the intervention with drugs, body weight loss and HOMA improve along with changes in intestinal flora.

Table IV.  Raw outcomes for change of clinical indicators and intestinal flora through drugs by individual trials 

Data is expressed as mean ± SD. BBR: berberine.

Effect of probiotics on intestinal flora in diabetes

The intestinal flora was significantly changed in diabetic patients after an intervention with Lactobacillus ADR-1 or ADR-3. According to Ming-Chia Hsieh et al. (18), the consumption of different strains of L. reuteri may influence changes in intestinal flora, which may lead to different outcomes after probiotic intake (Table V). In the study by Talia Palacios et al. (21), intentional manipulation of gastrointestinal microbial profiles may be useful for preventing and controlling type-2 diabetes mellitus and its associated metabolic complications. Somayyeh Firouzi et al. (24) proved that probiotics can also modestly improve HbA1c and fasting insulin in people with type-2 diabetes. Besides, Talia Palacios et al. (25) suggested that probiotics may act as an adjunctive to metformin by increasing the production of butyrate, which may consequently enhance glucose management.

Table V.  Raw outcomes for change of clinical indicators and intestinal flora through probiotics by individual trials 

Data is expressed as mean ± SD. Δ: after - before; NR: not reported

Effect of other methods on intestinal flora in diabetes

In the study by Yoriko Heianza et al. (23), they found the importance of changes in TMaO, choline and l-carnitine in improving insulin sensitivity during a weight-loss intervention for obese patients. Dietary fat intake may modify the associations of TMaO with insulin sensitivity and glucose metabolism.

SIDE EFFECTS

In these studies, more participants experienced gastrointestinal AEs in the treated groups, although the AEs did not affect the antidiabetic effect or gut microbiome features related to interventions in these studies. Again, this concern needs to be addressed in trials with longer intervention durations. Although the consumption of live probiotics products is generally considered safe for most human beings, some side effects have been reported under certain conditions. For example, live probiotics may become pathogenic when used in subjects with severe immune deficiency (27). In infants with short bowel or cardiac stenosis, bacteremia has been reported in some cases (28).

DISCUSSION

Research evidence from human intestinal microbial profiles demonstrates that each individual has a unique intestinal bacterial composition (in diversity and abundance). Modern pharmacological studies have shown that intestinal flora plays an important role in the occurrence and development of T2DM, ditto that for genetic, environmental, and dietary factors (29,30). However, the differences in intestinal flora may be due to different ethnicity, age, and gender. In addition, the mechanisms responsible for ecological imbalance, as well as the mechanistic link between altered gut flora and diabetes, is not yet clear.

EXERCISE AND INTESTINAL FLORA IN DIABETES

Exercise increased lean mass, reduced fat mass and inflammation with better glycemic control and physical performance. Some experimental research shows that physical activity may modify fecal short-chain fatty acids, which increases the presence of fecal butyrate and in turn butyrate-producer intestinal bacteria (31). Short-chain fatty acids activate muscular AMPK, an enzyme that regulates muscle metabolism of glucose and lipids. These metabolic effects may be important in diabetes and confirm cross-communication between microbiota, exercise, and global metabolism (32). And they suggest a link between microbiota, muscle, the brain, and human metabolism. The cure of intestinal microbiota with physical exercise and/or specific therapies could be an important step for tailored therapy allowing traditional therapy and global patient metabolism to function properly.

FOOD, SPECIFIC DIETS AND INTESTINAL FLORA IN DIABETES

An acute change in diet - for instance, one that is strictly animal-based or plant-based - alters microbial composition within just 24 h of initiation, with reversion to baseline within 48 h of diet discontinuation (33). Furthermore, the gut microbiome of animals fed a high-fat or high-sugar diet is more prone to circadian rhythm disruption (34). Studies also suggest that overwhelming systemic stress and inflammation - such as that induced via severe burn injury - can also produce characteristic acute changes in the gut microbiota within just one day of the sustained insult (35).

Several popular diets, including western, gluten-free, omnivore, vegetarian, vegan, and Mediterranean, have been studied for their ability to modulate the intestinal microbiota. In several studies, a western diet (high in animal protein and fat, low in fiber) led to a marked decrease in numbers of total bacteria and beneficial Bidobacterium and Eubacterium species (36-38).

Across the spectrum, the Mediterranean diet is highly regarded as a healthy balanced diet. It is distinguished by a beneficial fatty acid profile that is rich in both monounsaturated and polyunsaturated fatty acids, high levels of polyphenols and other antioxidants, high intake of fiber and other low glycemic carbohydrates, and relatively greater vegetable versus animal protein intake. Specifically, olive oil, assorted fruits, vegetables, cereals, legumes, and nuts, moderate consumption of fish, poultry, and red wine; and a lower intake of dairy products, red meat, processed meat and sweets characterize the traditional Mediterranean diet (39).

DRUGS AND INTESTINAL FLORA IN DIABETES

Wu et al. (16) showed that metformin interacts with different gut bacteria, possibly through the regulation of metal homeostasis. Furthermore, microbiota-based interventions may reduce gastrointestinal symptoms associated with metformin administration, with a consequent improvement in medication compliance (40).

Probiotics exhibit metabolic benefits by improving the gut barrier and alleviating inflammation (41), which are also key to the development of ageing-related diseases (42). Health-associated Bifidobacterium spp. have been shown to be depleted with ageing but enriched in extremely aged healthy subjects (43). It has been proposed that the intake of probiotics might improve the integrity of intestinal epithelium and diminish the toll-like receptor-4 pathways to reduce pro-inflammatory signaling and to enhance insulin sensitivity (44,45).

PROBIOTICS AND INTESTINAL FLORA IN DIABETES

Fermented foods containing lactic acid bacteria, such as cultured milk products and yogurt, represent a source of ingestible microorganisms that may beneficially regulate intestinal health (46). They are thought to accomplish this through their effects on the existing gut microbiome, in addition to a possible induction of anti-inflammatory cytokines such as IL-10 (47). Based on these properties, foods enriched for these modulatory microorganisms are referred to as probiotics. Several groups have reported increased total bacterial loads after regular consumption of fermented milk or yogurt (48-51). Notable increases in beneficial gut bifidobacteria and/or lactobacilli have also consistently been observed with several different types of probiotics (52).

Individuals with metabolic disorders such as obesity and diabetes have been shown to have an intestinal imbalance when compared to healthy individuals (53,54).

THE MECHANISMS OF PROBIOTICS IN DIABETES

Some possible mechanisms behind the effect of probiotics on glycemic control are presented. Researchers have hypothesized several mechanisms of action according to the previous literature. The effect of probiotics in improving glycemic control can be firstly explained through the action of primary bile acids on the farnesoid-X-receptor (FXR). Certain strains of Lactobacillus and Bifidobacterium are known to possess the bile salt hydrolase enzyme (BSH). This enzyme can directly increase the levels of primary bile acid, which in turn binds and activates the FXR, leading to increased storage of glucose, decreased production of glucose from non-glucose nutrients, increased synthesis of insulin and increased secretion of insulin (55,56). Secondly, probiotics are also known to increase glucagon-like peptide (GLP) 1 and GLP 2, which are able to decrease low-grade inflammation associated with diabetes, and to decrease insulin resistance, which in turn decreases ß-cell toxicity and improves glycemic control. Furthermore, GLP-1 and GLP-2 also decrease hunger and increase satiety, thus decreasing energy intake, which collectively improve glycemic control (57). Another possible mechanism is the increased production of short-chain fatty acids (SCFAs) by mainly Bifidobacterium in the colon via its action on insoluble dietary fibers. These SCFAs, especially butyrate, can decrease insulin resistance by promoting pancreatic ß-cell differentiation, proliferation and development; increase secretion of GLP-1, thus increasing secretion of insulin, and decrease the release of pro-inflammatory cytokines by adipose tissue (58,59).

In conclusion, notwithstanding some limitations, these findings may have important implications for managing T2DM in patients by treating the microecological imbalance. These studies will also provide empirical evidence to address currently unresolved issues with the efficacy and safety of probiotics. Future research should focus on identifying the role of and the complex interaction among probiotics and the proportion of the various phyla in the gut of individuals.

CONCLUSIONS

All in all, probiotics have emerged as a possible therapeutic option, and prospective clinical trials have shown promising results in T2D patients. But up to now, there is still a dearth of such studies, so there was a need for high-quality studies to better understand the clinical effects of the intestinal flora on the occurrence and development of diabetes. This issue remains unanswered at present, primarily because large, long-term prospective trials of probiotic therapy are absent. However, several problems such as the possible side effects of long-term use of probiotics remain to be solved. The effects of probiotics are also slow, and some patients and doctors may be afraid of delaying illness.

REFERENCES

1. Cho NH, Shaw JE, Karuranga S, Huang Y, Da Rocha Fernandes JD, Ohlrogge AW, et al. IDF Diabetes Atlas:Global estimates of diabetes prevalence for 2017 and projections for 2045. Diabetes Res Clin Pract 2018;138:271-81;DOI:10.1016/j.diabres.2018.02.023 [ Links ]

2. Wang L, Gao P, Zhang M, Huang Z, Zhang D, Deng Q, et al. Prevalence and ethnic pattern of diabetes and prediabetes in China in 2013. JAMA 2017;317(24):2515-23. DOI:10.1001/jama.2017.7596 [ Links ]

3. Holman N, Young B, Gadsby R. Current prevalence of Type 1 and Type 2 diabetes in adults and children in the UK. Diabet Med 2015;32(9):1119-20. DOI:10.1111/dme.12791 [ Links ]

4. Bruno G, Runzo C, Cavallo-Perin P, Merletti F, Rivetti M, Pinach S, et al. Incidence of type 1 and type 2 diabetes in adults aged 30-49 years:the population-based registry in the province of Turin, Italy. Diabetes Care 2005;28(11):2613-9. DOI:10.2337/diacare.28.11.2613 [ Links ]

5. Gill SR, Pop M, DeBoy RT, Eckburg PB, Turnbaugh PJ, Samuel BS, et al. Metagenomic Analysis of the human distal gut microbiome. Science (2006),312:1355-9. DOI:10.1126/science.1124234 [ Links ]

6. Flint HJ, Duncan SH, Scott KP, Louis P. Interactions and competition within the microbial community of the human colon:links between diet and health. Environ Microbiol 2007;9:1101-11. DOI:10.1111/j.1462-2920.2007.01281.x [ Links ]

7. Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, et al. Dominant and diet-responsive groups of bacteria within the human colonic microbiota. ISME J 2011;5:220-30. DOI:10.1038/ismej.2010.118 [ Links ]

8. Belkaid Y, Hand TW. Role of the microbiota in immunity and inflammation. Cell 2014;157:121-41. DOI:10.1016/j.cell.2014.03.011 [ Links ]

9. Noverr MC, Huffnagle GB. Does the microbiota regulate immune responses outside the gut? Trends Microbiol 2004;12:562-8. DOI:10.1016/j.tim.2004.10.008 [ Links ]

10. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505:559-63. DOI:10.1038/nature12820 [ Links ]

11. Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012;490:55-60. DOI:10.1038/nature11450 [ Links ]

12. Koenig JE, Spor A, Scalfone N, Fricker AD, Stombaugh J, Knight R, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci USA 2011;108(Suppl. 1):4578-85. DOI:10.1073/pnas.1000081107 [ Links ]

13. Balducci S, Zanuso S, Nicolucci A, Fernando F, Cavallo S, Cardelli P, et al. Anti-inflammatory effect of exercise training in subjects with type 2 diabetes and the metabolic syndrome is dependent on exercise modalities and independent of weight loss. Nutr Metab Cardiovasc Dis 2010;20:608-17. DOI:10.1016/j.numecd.2009.04.015 [ Links ]

14. Ren M, Zhang H, Qi J, Hu A, Jiang Q, Hou Y, et al. An Almond-Based Low Carbohydrate Diet Improves Depression and Glycometabolism in Patients with Type 2 Diabetes through Modulating Gut Microbiota and GLP-1:A Randomized Controlled Trial. Nutrients 2020;12(10):3036. DOI:10.3390/nu12103036 [ Links ]

15. Pasini E, Corsetti G, Assanelli D, Testa C, Romano C, Dioguardi FS, et al. Effects of chronic exercise on gut microbiota and intestinal barrier in human with type 2 diabetes. Minerva Med 2019;110(1):3-11. DOI:10.23736/S0026-4806.18.05589-1 [ Links ]

16. Wu H, Esteve E, Tremaroli V, Tanweer Khan M, Caesar R, Manner's-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 Med 2017;23(7):850-8. DOI:10.1038/nm.4345 [ Links ]

17. Tong X, Xu J, Lian F, Yu X, Zhao Y, Xu L, et al. Structural Alteration of Gut Microbiota during the Amelioration of Human Type 2 Diabetes with Hyperlipidemia by Metformin and a Traditional Chinese Herbal Formula:a Multicenter, Randomized, Open Label Clinical Trial. mBio 2018;9(3):e02392-17. DOI:10.1128/mBio.02392-17 [ Links ]

18. Hsieh MC, Tsai WH, Jheng YP, Su SL, Wang SY, Lin CC, et al. The beneficial effects of Lactobacillus reuteri ADR-1 or ADR-3 consumption on type 2 diabetes mellitus:a randomized, double-blinded, placebo-controlled trial. Sci Rep 2018;8(1):850-8. DOI:10.1038/s41598-018-35014-1 [ Links ]

19. 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 2018;359(6380):1151-6. DOI:10.1126/science.aao5774 [ Links ]

20. Zhang Y, Gu Y, Ren H, Wang S, Zhong H, Zhao X, et al. Gut microbiome-related effects of berberine and probiotics on type 2 diabetes (the PREMOTE study). Nat Commun 2020;11(1):5015. DOI:10.1038/s41467-020-18414-8 [ Links ]

21. Palacios T, Vitetta L, Coulson S, Madigan CD, Denyer GS, Caterson ID. The effect of a novel probiotic on metabolic biomarkers in adults with prediabetes and recently diagnosed type 2 diabetes mellitus:study protocol for a randomized controlled trial. Trials 2017;18(1):7. DOI:10.1186/s13063-016-1762-x [ Links ]

22. Liu Y, Wang Y, Ni Y, Cheung CKY, Lam KSL, Wang Y, et al. Gut Microbiome Fermentation Determines the Efficacy of Exercise for Diabetes Prevention. Cell Metab 2020;31(1):77-91.e5. DOI:10.1016/j.cmet.2019.11.001 [ Links ]

23. Heianza Y, Sun D, Li X, DiDonato JA, Bray GA, Sacks FM, et al. Gut microbiota metabolites, amino acid metabolites and improvements in insulin sensitivity and glucose metabolism:the POUNDS Lost trial. Gut 2019;68(2):263-70. DOI:10.1136/gutjnl-2018-316155 [ Links ]

24. Firouzi S, Majid HA, Ismail A, Kamaruddin NA, Barakatun-Nisak M-Y. Effect of multi-strain probiotics (multi-strain microbial cell preparation) on glycemic control and other diabetes-related outcomes in people with type 2 diabetes:a randomized controlled trial. Gut 2017;56(4):1535-50. DOI:10.1007/s00394-016-1199-8 [ Links ]

25. Palacios T, Vitetta L, Coulson S, Madigan CD, Lam YY, Manuel R, et al. Targeting the Intestinal Microbiota to Prevent Type 2 Diabetes and Enhance the Effect of Metformin on Glycaemia:A Randomised Controlled Pilot Study. Nutrients 2020;12(7):2041. DOI:10.3390/nu12072041 [ Links ]

26. Karusheva Y, Koessler T, Strassburger K, Markgraf D, Mastrototaro L, Jelenik T, et al. Short-term dietary reduction of branched-chain amino acids reduces meal-induced insulin secretion and modifies microbiome composition in type 2 diabetes:a randomized controlled crossover trial. Am J Clin 2019;110(5):1098-1107. DOI:10.1093/ajcn/nqz191 [ Links ]

27. Adams CA. The probiotic paradox:live and dead cells are biological response modifiers. Nutr Res Rev 2010;23(1):37-46. DOI:10.1017/S0954422410000090 [ Links ]

28. Didari T, Solki S, Mozaffari S, Nikfar S, Abdollahi M. A systematic review of the safety of probiotics. Expert Opin Drug Saf 2014;13(2):227-39. DOI:10.1517/14740338.2014.872627 [ Links ]

29. Song S, Lee J. Dietary patterns related to triglyceride and high-density lipoprotein cholesterol and the incidence of type 2 diabetes in Korean men and women, Nutrients 2019;11(1):8-21. DOI:10.3390/nu11010008 [ Links ]

30. Jeon J, Jang J, Park K. Effects of consuming calcium-rich foods on the incidence of type 2 diabetes mellitus. Nutrients 2019;11(1):31-41. DOI:10.3390/nu11010031 [ Links ]

31. Matsumoto M, Inoue R, Tsukahara T, Ushida K, Chiji H, Matsubara N, et al. Voluntary running exercise alters microbiota composition and increases n-butyrate concentration in the rat cecum. Biosci Biotechnol Biochem 2008;72:572-6. DOI:10.1271/bbb.70474 [ Links ]

32. Kasubuchi M, Hasegawa S, Hiramatsu T, Ichimura A, Kimura I. Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation. Nutrients 2015;7(4):2839-49. DOI:10.3390/nu7042839 [ Links ]

33. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014;505(7484):559-63. DOI:10.1038/nature12820 [ Links ]

34. Voigt RM, Forsyth CB, Green SJ, Mutlu E, Engen P, Vitaterna MH, et al. Circadian disorganization alters intestinal microbiota. PLoS ONE 2014;9(5):e97500. DOI:10.1371/journal.pone.0097500 [ Links ]

35. Earley ZM, Akhtar S, Green SJ, Naqib A, Khan O, Cannon AR, et al. Burn Injury alters the intestinal microbiome and increases gut permeability and bacterial translocation. PLoS ONE 2015;10(7):e0129996. DOI:10.1371/journal.pone.0129996 [ Links ]

36. Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, et al. Linking long-term dietary patterns with gut microbial enterotypes. Science 2011;334(6052):105-8. DOI:10.1126/science.1208344 [ Links ]

37. Reddy BS, Weisburger JH, Wynder EL. Effects of high risk and low risk diets for colon carcinogenesis on fecal microflora and steroids in man. J Nutr 1975;105(7):878-84. DOI:10.1093/jn/105.7.878 [ Links ]

38. Drasar BS, Crowther JS, Goddard P, Hawksworth G, Hill MJ, Peach S, et al. The relation between diet and the gut micro ora in man. Proc Nutr Soc 2007;32(2):49-52. DOI:10.1079/pns19730014 [ Links ]

39. Lopez-Legarrea P, Fuller NR, Zulet MA, Martinez JA, Caterson ID. The influence of Mediterranean, carbohydrate and high protein diets on gut microbiota composition in the treatment of obesity and associated inflammatory state. Asia Pac J Clin Nutr 2014;23(3):360-8. DOI:10.6133/apjcn.2014.23.3.16 [ Links ]

40. Greenway F, Wang S, Heiman M. A novel cobiotic containing a prebiotic and an antioxidant augments the glucose control and gastrointestinal tolerability of metformin:a case report. Benef Microbes 2014;5(1):29-32. DOI:10.3920/BM2012.0063 [ Links ]

41. Gomes AC, Bueno AA, de Souza RG, Mota JF. Gut microbiota, probiotics and diabetes. Nutr J 2014;13:60. DOI:10.1186/1475-2891-13-60 [ Links ]

42. Franceschi C, Garagnani P, Parini P, Giuliani C, Santoro A. Inflammaging:a new immune-metabolic viewpoint for age-related diseases. Nat Rev Endocrinol 2018;14(10):576-90. DOI:10.1038/s41574-018-0059-4 [ Links ]

43. Zmora N, Zilberman-Schapira G, Suez J, Mor U, Dori-Bachash M, Bashiardes S, et al. Personalized gut mucosal colonization resistance to empiric probiotics is associated with unique host and microbiome features. Cell 2018;174(6):1388-405.e21. DOI:10.1016/j.cell.2018.08.041 [ Links ]

44. Lee SK, Yang KM, Cheon JH, Kim TI, Kim WH. Anti-inflammatory mechanism of Lactobacillus rhamnosus GG in lipopolysaccharide-stimulated HT-29 cell. Korean J Gastroenterol 2012;60(2):86-93. DOI:10.4166/kjg.2012.60.2.86 [ Links ]

45. Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes:molecularmechanisms and probiotic treatment. EMBO Mol Med 2011;3(9):559-72. DOI:10.1002/emmm.201100159 [ Links ]

46. Shen J, Zuo ZX, Mao AP. Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn's disease, and pouchitis. Inflamm Bowel Dis 2014;20:21-35. DOI:10.1097/01.MIB.0000437495.30052.be [ Links ]

47. Foligne B, Parayre S, Cheddani R, Famelart MH, Madec MN, Ple C, et al. Immunomodulation properties of multi-species fermented milks. Food Microbiol 2016;53:60-9. DOI:10.1016/j.fm.2015.04.002 [ Links ]

48. Matsumoto K, Takada T, Shimizu K, Moriyama K, Kawakami K, Hirano K, et al. Effects of a probiotic fermented milk beverage containing Lactobacillus casei strain Shirota on defecation frequency, intestinal microbiota, and the intestinal environment of healthy individuals with soft stools. J Biosci Bioeng 2010;110:547-52. DOI:10.1016/j.jbiosc.2010.05.016 [ Links ]

49. He T, Priebe MG, Zhong Y, Huang C, Harmsen HJM, Raangs GC, et al. Effects of yogurt and bifidobacteria supplementation on the colonic microbiota in lactose-intolerant subjects. J Appl Microbiol 2008;104:595-604. DOI:10.1111/j.1365-2672.2007.03579.x [ Links ]

50. Zhong Y, Huang CY, He T, Harmsen HMJ. Effect of probiotics and yogurt on colonic microflora in subjects with lactose intolerance. Wei Sheng Yan Jiu 2006;35:587-91. [ Links ]

51. Goossens DAM, Jonkers DMAE, Russel MGVM, Stobberingh EE, Stockbrugger RW. The effect of a probiotic drink with Lactobacillus plantarum 299v on the bacterial composition in faeces and mucosal biopsies of rectum and ascending colon. Aliment Pharmacol Ther 2006;23:255-63. DOI:10.1111/j.1365-2036.2006.02749.x [ Links ]

52. Inoguchi S, Ohashi Y, Narai-Kanayama A, Aso K, Nakagaki T, Fujisawa T. Effects of non-fermented and fermented soybean milk intake on faecal microbiota and faecal metabolites in humans. Int J Food Sci Nutr 2012;63:402-10. DOI:10.3109/09637486.2011.630992 [ Links ]

53. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Microbial ecology:human gut microbes associated with obesity. Nature 2006;444:1022-3. DOI:10.1038/4441022a [ Links ]

54. Karlsson F, Tremaroli V, Nielsen J, Backhed F. Assessing the human gut microbiota in metabolic diseases. Diabetes 2013;62:3341-9. DOI:10.2337/db13-0844 [ Links ]

55. Renga B, Mencarelli A, Vavassori P, Brancaleone V, Fiorucci S. The bile acid sensor FXR regulates insulin transcription and secretion. Biochim Biophys Acta 2010;1802(3):363-72. DOI:10.1016/j.bbadis.2010.01.002 [ Links ]

56. Ding L, Yang L, Wang Z, Huang W. Bile acid nuclear receptor FXR and digestive system diseases. Acta Pharm Sin B 2015;5(2):135-44. DOI:10.1016/j.apsb.2015.01.004 [ Links ]

57. Shyangdan DS, Royle P, Clar C, Sharma P, Waugh N, Snaith A. Glucagon-like peptide analogues for type 2 diabetes mellitus. Cochrane Database Syst Rev 2011;2011(10):CD006423. DOI:10.1002/14651858.CD006423.pub2 [ Links ]

58. Tilg H, Moschen AR. Microbiota and diabetes:an evolving relationship. Gut 2014;63:1513-21. DOI:10.1136/gutjnl-2014-306928 [ Links ]

59. Puddu A, Sanguineti R, Montecucco F, Viviani GL. Evidence for the gut microbiota short-chain fatty acids as key pathophysiological molecules improving diabetes. Mediators Inflamm 2014;2014:162021. DOI:10.1155/2014/162021 [ Links ]

Received: September 06, 2021; Accepted: October 30, 2021

Correspondence: Xianghua Ma. Nutritional Department. The First Affiliated Hospital of Nanjing Medical University. 300 Guangzhou Road. 210029 Nanjing, People's Republic of China

Conflicts of interest:

the authors declare none.

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License