Home Impact of gut microbiota and associated mechanisms on postprandial glucose levels in patients with diabetes
Article Open Access

Impact of gut microbiota and associated mechanisms on postprandial glucose levels in patients with diabetes

  • Xinyuan Feng , Mingqun Deng , Lina Zhang and Qi Pan EMAIL logo
Published/Copyright: December 20, 2023

Abstract

Diabetes and its complications are serious medical and global burdens, often manifesting as postprandial hyperglycemia. In recent years, considerable research attention has focused on relationships between the gut microbiota and circulating postprandial glucose (PPG). Different population studies have suggested that PPG is closely related to the gut microbiota which may impact PPG via short-chain fatty acids (SCFAs), bile acids (BAs) and trimethylamine N-oxide (TMAO). Studies now show that gut microbiota models can predict PPG, with individualized nutrition intervention strategies used to regulate gut microbiota and improve glucose metabolism to facilitate the precision treatment of diabetes. However, few studies have been conducted in patients with diabetes. Therefore, little is known about the relationships between the gut microbiota and PPG in this cohort. Thus, more research is required to identify key gut microbiota and associated metabolites and pathways impacting PPG to provide potential therapeutic targets for PPG.

Introduction

Diabetes is one of the most common chronic non-communicable diseases which threatens global human health.[1, 2] The International Diabetes Federation (10th Diabetes Atlas, 2021) reports that global disease prevalence, between the ages of 20 to 79 years, is estimated to be 10.5% (536.6 million adults),[3] with disease-related health expenditure is estimated at 966 billion dollars and is expected to reach 10.54 trillion dollars by 2045.[4] Diabetic nephropathy, diabetic retinopathy, cardiovascular events and other diabetes-related complications seriously affect the quality of life of patients, increasing the hospitalization rate and mortality of patients.[5, 6, 7, 8, 9, 10, 11] In China, approximately 33% of diabetic outpatients achieve blood glucose targets.[12] Blood glucose fluctuation, especially postprandial hyperglycemia, is closely related to diabetes complications.[13] Glycemic control was assessed using the hemoglobin A1c (HbA1c) level.[14] The system evaluation showed that a decrease in postprandial glucose (PPG) accounted for nearly twice as much as fasting plasma glucose (FPG) for the decreases in HbA1c.[15] So PPG had a better correlation with HbA1c than FPG. The American Diabetes Association Guidelines 2021 suggests that PPG monitoring should be conducted in patients with diabetes who failed to reach satisfactory HbA1c levels but obtained target fasting blood glucose, and PPG levels should be maintained below 10.0 mmol/L to reduce HbA1c.[16] Gastric emptying, intestinal proinsulin system, and preprandial blood glucose levels affect postprandial blood glucose levels. In recent years, more studies have reported that the gut microbiota may exert important actions on blood glucose levels, in addition to the effects of islet functions, diets, exercise, and other factors.[17,18] Studies investigating relationships between diabetes and gut microbiota have successfully shown that gut microbiota composition and abundance are related to fasting blood glucose or HbA1c levels, but few studies have focused on relationships between PPG and the gut microbiota.[19, 20, 21] In this review, we address this issue and examine associated underlying mechanisms.

Gut microbiota overview

As the second gene bank of human beings,[22] the number of gut microbiota is 10 times the total number of human cells, and the number of genes carried by them is more than 100 times their own. At the same time, as the microbial organ of the host, it is closely related to various metabolic pathways.[23,24] Gut microbiota is established during infancy and develops to maturity across the first 2 years of life. Their stability decreases in one’s old age under the combined influence of genetic and environmental factors. [25] Gut microbiota can be classified by phylum, class, order, family, genus, and species, and mainly includes Firmicutes (Lactobacillus, Clostridium, Ruminococcus, etc.), Bacteroidetes (Bacteroides, Prevotella, etc.), Proteobacteria (Escherichia coli, etc.), and Actinobacteria (Bifidobacteria, etc.). Firmicutes contain the greatest number of genera, comprising more than 200 genera.[26] Regarded as a unique host microbial organ, intestinal microorganisms are closely associated with different metabolic pathways. Gut microbiota breaks down undigested plant polysaccharides, proteins or amino acids by encoding active enzymes of carbohydrates and protein,[27] and those resultant metabolites could exert a biological activity and affect the metabolism of body.[28]

PPG and its clinical significance

PPG is one of the main reasons for increased HbA1c levels.[29, 30] High PPG is closely related to the occurrence and development of chronic diabetic complications.[31, 32, 33] Hyperglycemia and its effect after acute myocardial infarction on cardiovascular outcomes in patients with type 2 diabetes mellitus (HEART2D) study compared the effects of different blood glucose control strategies on cardiovascular endpoints in 1115 type 2 diabetes (T2D) patients after acute myocardial infarction. The study showed that in patients older than 65.7 years, there is no significant differences in baseline characteristics including HbA1c, diabetic therapies, prior cardiovascular disease history, or other clinically relevant measures between different study arms.[34] The PPG control group recorded a significantly less time to the first cardiovascular event, and a significantly lower proportion of patients experienced a first cardiovascular event when compared with the fasting blood glucose control group (n = 56 [29.6%] vs. n = 85 [40.5%]; hazard ratio = 0.69 [95% confidence interval (CI): 0.49 to 0.96]; P = 0.029). Controlling PPG is important for promoting HbA1c levels and preventing microvascular and macrovascular diseases and heart events in diabetes.[35] After eating the same foods, PPG in an Asian population was higher when compared with Caucasians, and was putatively related to chewing habits, basic oral hygiene, high amylase activity,[36] oral physiological, anatomical parameters and other factors in these populations.[37] For these populations, PPG appeared to contribute to HbA1c. Therefore, more attention must be paid to PPG in this group.

Relationships between gut microbiota and PPG

In vitro research progress on gut microbiota and glucose lipid metabolism

The molecular mechanisms by which gut microbiota affects host metabolic balance mainly include the following two ways: (1) the role of gut microbiota itself; (2) the effects mediated by metabolites of gut microbiota.[38,39] Gut microbiota can have a direct impact on the host by disrupting the integrity of the intestinal mucosal barrier, allowing molecules such as lipopolysaccharides (LPS) to enter the host cycle. LPS of Akkermansia muciniphila can specifically activate the expression of Toll-like receptors 2 (TLR2).[40] A His-tagged Amuc_1100 produced in E. coli (hereafter called Amuc_1100*) could similarly signal TLR2-expressing cells to A. muciniphila. Plover et al. [41]’s research has shown that Amuc_ 1100 * improves the metabolic syndrome in obese and diabetic mice through TLR2 signaling. Compared with normal-fed mice, untreated high-fat fed mice exhibited lower phosphorylation of the protein kinase B (PKB/Akt) pathway, while mice fed with Amuc_1100 * treatment offset this impact and improved insulin sensitivity. In addition, gut microbiota can also have indirect effects through its metabolites, such as the production of short-chain fatty acids (SCFAs), bile acids (BAs), trimethylamine N-oxide (TMAO), etc., which affect the metabolism (Table 1).

Table 1

The mechanism of the effect of gut microbiota metabolites on postprandial blood glucose

Gut microbiota metabolites Related signaling pathways/signaling molecules/key enzymes Microbiota Mechanism
SCFA (butyrate, formic, acetic, and propionic acids) FFAR2 FFAR3 Bacteroidetes Firmicutes Lachnospiraceae Ruminococcus SCFA binds to specific transmembrane receptors FFAR2 and FFAR3 stimulates GLP-1 and PYY secretion, inhibits appetite, and reduces PPG.
BA FXR TGR5 Clostridium Bacteroides Lactobacillus Bifidobacterium BA regulates PPG through signals from multiple parts of the body. Liver BA-FXR promotes glycogen synthesis; Intestinal BA-TGR5 promotes GLP-1 expression and secretion, while BA-FXR inhibits GLP-1 production; BA-TGR5 mediates satiety in the brain and increases energy consumption in skeletal muscle and brown adipose tissue; Pancreatic islet β BA-TGR5 and BA-FXR in cells induce insulin production.
TMAO, TMA FMO3 PKA IGF-2 PI3K/Akt Bacteroidetes, Firmicutes The levels of TMAO and TMA increase after meals, which affect the phosphorylation process of PKA and IGF-2, and block the PI3K/Akt insulin signaling pathway to increase PPG.
  1. SCFA: short-chain fatty acid, FFAR2: free fatty acid receptor 2, FFAR3: free fatty acid receptor 3, BA: bile acid, FXR: farnesoid X receptor, TGR5: Takeda G protein receptor 5, TMAO: trimethylamine oxide, TMA: trimethylamine, FMO3: Flavin containing monooxygenase 3, PI3K/Akt: phosphatidylinositol 3-kinase/protein kinase B, IGF-2: insulin-like growth factor 2, PKA: protein kinase A, GLP-1: glucagon-like peptide 1, PPG: postprandial glucose.

PPG in different populations

Postprandial glucose responses (PPGR) reflect increased areas under blood glucose response curves within 2 hours after eating.[42] Despite eating the same food, the PPGR of different individuals was significantly different, and the level of PPG was also different. In addition to food characteristics (e.g. carbohydrate content) and genetic factors, PPGR may be affected by intestinal microbiome differences in different individuals.[43,44]

PPG in non-diabetic individuals

In normal populations, PPG levels may be somewhat predicted by combined information such as diet, body composition, and gut microbiota. Zeevi et al.[45] integrated data from 800 non-diabetic subjects and generated a personalized PPGR prediction model, and showed that Proteobacteria and Enterobacter were positively associated with PPGR in standardized diets. Using a standardized PPGR prediction model in healthy Danish adults, it was suggested that intestinal metagenomic species abundance, specifically, Clostridia MGS.hg0341 and Bifidobacteria were negatively associated with PPG.[46] Nolte et al. reported similar findings that Faecalibacterium prausnitzii was negatively associated with PPG.[47] In the elderly, certain correlations exist between intestinal bacteria and PPG; research in elderly healthy individuals (> 65 years old) reported significant correlations between gut bacteria and peak glucose levels after dinner and the 4-hour area under the curve (AUC) period after dinner.[48] Bacteroidetes, Blautia, and Bilophila are positive, while Ruminococcus and Holdmannia are negatively associated with PPG.[49]

Relationships between gut microbiota and diabetic PPG

Intestinal microbial composition in patients with diabetes is distinct when compared with healthy individuals. A systematic review of 25 studies comprising 2209 type 1 diabetes (T1D) and T2D patients reported no significant changes in the number of gut microbiota in this population, but Bacteroides, Bifidobacterium, and Clostridium abundance had decreased and were negatively associated with blood sugar levels.[50]

For patients with T1D, decreased Firmicutes/Bacteroides ratios may be related to T1D incidences and increased HbA1c levels.[51] Gut microbiota diversity in T1D patients is associated with HbA1c levels.[52] Research has suggested that gut microbiota diversity in adult T1D patients who are not newly diagnosed and whose mean/median HbA1c levels are less than 8% are similar to those of normal individuals, but when HbA1c levels exceed 8%, gut microbiota diversity is distinct from healthy individuals. [53] A previous T1D study reported that gut microbiota affected PPG; a prediction model incorporated gut microbiota characteristics can predict PPGR in T1D patients, except for the effects of carbohydrate content, and the proportion of carbohydrate to fat on PPG.[54,55] Shilo et al. enrolled 121 T1D patients, measured 6377 PPG data points, and designed a prediction model, which integrated blood glucose levels, insulin doses, dietary habits, and gut microbiota to accurately predict PPGR and provide T1D patients with optimal meal insulin doses.[56]

For T2D patients, the study showed that Roseburia and Faecalibacterium abundance decreased, while Lactobacillus gasseri, Streptococcus mutans, and some Clostridium spp. abundance increased when compared with a healthy population.[57] Blood glucose levels in T2D patients are associated with gut microbiota abundance.[58] Enterobacteria and Enterococci abundance is less in patients with good blood glucose control (HbA1c < 6.5%) when compared with patients with poor blood glucose control (HbA1c ≥ 6.5%), while Bifidobacteria and Bacteroidetes abundance is higher when compared with patients with poor blood glucose control.[59] Studies have shown that intestinal microbial interventions can affect PPG. In one study, 102 T2D patients were randomly divided into two groups; the control group was given basic hypoglycemic drugs and the intervention group was given hypoglycemic drugs plus triple viable Bifidobacterium capsules. After 8 weeks, Bifidobacteria and Lactobacilli levels increased in the intervention group when compared with the control group, and Enterococcus and coccobacillus abundance decreased. Also, mean 2-hour PPG levels in the intervention group were 1.54 mmo/L lower when compared with the control group (P = 0.026).[60]

How the gut microbiota affects PPG

In addition to directly acting through its own LPS, gut microbiota can generate various bioactive metabolites such as short-chain fatty acids, bile acids, and trimethylamine oxide through the liver or intestines. These metabolites can serve as indirect regulatory factors, regulating host glucose metabolism and insulin signaling pathways, and affecting postprandial glucose through related metabolic pathways in different tissues and organs (Figure 1).

Figure 1 Mechanisms showing how the microbiota potentially reduce postprandial blood glucose. A. Gut microbiota mainly includes Firmicutes, Bacteroidetes, Proteobateria and Actinobacteria. B. Undigested dietary fiber generates SCFAs via microbiota fermentation in the intestine (blue arrow); BA is secreted into bile by liver cells, enters the intestine and participates in the hepatointestinal circulation (green arrow); After high-fat food is eaten, TMA is generated through gut microbiota enzymes, and TMA generates TMAO under the action of liver FMO3 (red arrow). C. Gut microbiota generates many signaling metabolites, such as SCFAs, BAs and TMAO, which participate in different metabolic pathways to ultimately affect PPG. SCFAs: short-chain fatty acids, BA: bile acid, TMA: trimethylamine, FMO3: Flavin containing monooxygenase 3, TMAO: trimethylamine oxide, PI3K/Akt: phosphatidylinositol 3-kinase/protein kinase B, FFAR2: free fatty acid receptor 2, FFAR3: free fatty acid receptor 3, TGR5: Takeda G protein receptor 5, FXR: farnesoid X receptor, GLP-1: glucagon-like peptide 1, PPG: postprandial glucose
Figure 1

Mechanisms showing how the microbiota potentially reduce postprandial blood glucose. A. Gut microbiota mainly includes Firmicutes, Bacteroidetes, Proteobateria and Actinobacteria. B. Undigested dietary fiber generates SCFAs via microbiota fermentation in the intestine (blue arrow); BA is secreted into bile by liver cells, enters the intestine and participates in the hepatointestinal circulation (green arrow); After high-fat food is eaten, TMA is generated through gut microbiota enzymes, and TMA generates TMAO under the action of liver FMO3 (red arrow). C. Gut microbiota generates many signaling metabolites, such as SCFAs, BAs and TMAO, which participate in different metabolic pathways to ultimately affect PPG. SCFAs: short-chain fatty acids, BA: bile acid, TMA: trimethylamine, FMO3: Flavin containing monooxygenase 3, TMAO: trimethylamine oxide, PI3K/Akt: phosphatidylinositol 3-kinase/protein kinase B, FFAR2: free fatty acid receptor 2, FFAR3: free fatty acid receptor 3, TGR5: Takeda G protein receptor 5, FXR: farnesoid X receptor, GLP-1: glucagon-like peptide 1, PPG: postprandial glucose

Role of short-chain fatty acids: Undigested dietary fiber generates SCFAs via bacterial fermentation in the distal ileum and colon.[61] SCFAs are organic fatty acids that contain 1–6 carbon atoms.[62] Butyrate is the main energy source for gut epithelial cells,[63] and is closely associated with metabolism. SCFAs also include formic, acetic, and propionic acids.[64] Many bacteria produce acetic acid,[65] while Bacteroidetes is the dominant propionic acid producer,[66] and Firmicutes is the dominant butyrate producer.[67] SCFAs are implicated in carbohydrate metabolism via different metabolic pathways to reduce PPG levels.[68] SCFAs and their specific transmembrane receptors, including the free fatty acid receptor 2 (FFAR2) and the free fatty acid receptor 3 (FFAR3), are involved in glucose and lipid metabolism.[69] It was reported that acetic acid selectively mediated Gq/11 or Gi/o pathways via FFAR2 and FFAR3 to increase or decrease glucose-induced insulin secretion.[70] FFAR3 has Gi/o coupling, and FFAR2 is doubly coupled through the Gi/o and Gq families.[71] Additionally, butyrate is mainly produced by Lachnospiraceae and Ruminococcus,[72] which reduces PPG and improves insulin sensitivity via epigenetic regulation, mitochondrial β-oxidation, and β-cell proliferation.[73]

The gut microbiota impacts PPG by producing SCFAs which stimulate glucagon-like peptide 1 (GLP-1) secretion via FFAR2 and FFAR3, induce glucose-dependent insulin secretion and inhibit glucagon secretion.[74] SCFAs also stimulate peptide YY secretion to inhibit the appetite and decrease PPG levels.[75] Vitale et al. reported that when compared with control meals, a Mediterranean diet group had significantly lower PPG and insulin responses, and that blood glucose and insulin sensitivity levels were improved after an 8-week dietary intervention.[53] Butyric acid levels in the Mediterranean diet group also increased significantly after meals (P = 0.019) and were directly related to insulin sensitivity (r = 0.397, P = 0.050). These metabolic changes were accompanied by significant changes in intestinal microbiota; when compared with the control group, Intestinimonas butyriciproducens and Akkermansia muciniphila abundance in the Mediterranean diet group increased.

Role of bile acids: BAs are diversified amphipathic steroid molecules which promote intestinal absorption and dietary lipid transportation,[76] with concentrations dependent on biosynthesis, enterohepatic circulation, and intestinal microbiota levels.[77] Clostridium, Bacteroides, Lactobacillus, Bifidobacterium and Enterococcus have been proven to be involved in the production of bile acid.[78] Recent research reported that BAs are key signal molecules in glucose, lipid, and energy metabolism as they combine with the farnesoid X receptor (FXR) and the Takeda G-protein-coupled receptor 5 (TGR5) in multiple tissues and organs to regulate GLP-1 secretion, gluconeogenesis, glycogen synthesis, inflammatory responses, and gut microbiome structures.[79, 80, 81, 82] FXR is widely expressed in various tissues and organs such as the intestine, liver, and white adipose tissue, which can form heterodimers to inhibit the expression of the rate limiting enzyme cholesterol 7α-hydroxylase (CYP7A1) in BA biosynthesis, weakening cholesterol liver conversion; The expression of CYP7A1 alleviated metabolic disorders associated with obesity, including glucose intolerance, insulin resistance, and dyslipidemia.[83,84] TGR5 is a G protein-coupled receptor, which can produce cAMP through BAs, and then activate the protein kinase A (PKA) pathway.[85] In the liver, BA-FXR signal transduction inhibits gluconeogenesis and promotes glycogen synthesis by negative modulation. In intestinal cells, BA-TGR5 signaling promotes GLP-1 expression and secretion, while BA-FXR signaling inhibits GLP-1 production. In addition, BA-TGR5 signal transduction can mediate satiety in the brain, and increase energy consumption in skeletal muscle and brown adipose tissue. In the pancreas, β BA-TGR5 and BA-FXR signaling in cells induce insulin production.[86, 87, 88] BA sequestrants (BAS) or bariatric surgery can significantly eliminate blood glucose abnormalities. A meta-analysis of 2950 T2D patients from 17 studies reported that HbA1c levels in a BAS group decreased when compared with a control group (mean difference -0.55%; 95% CI: -0.64 to -0.46).[89] Bariatric surgery alters the enterohepatic BA circulation, resulting in increased plasma bile levels as well as altered BA composition.[90] Weight loss surgery, especially Roux-en-Y gastric bypass surgery, can increase circulating BA concentrations.[91] Postoperative BA concentrations are positively correlated with serum GLP-1 concentrations but negatively correlated with PPG.[92] In addition to SCFAs and BAs actions, amino acids and their metabolites, especially tryptophan and associated derivatives, can affect glucose metabolism, but research suggests that amino acid metabolic pathways may affect diabetes and fasting blood glucose levels.[93, 94, 95] However, few studies have investigated the effects of gut microbiota on PPG via amino acid pathways.[96]

Effect of trimethylamine oxide: Ingestion of high-fat foods can generate the primary intestinal metabolite trimethylamine (TMA) through gut microbiota enzymes such as CutC/D,[97,98] CntA/B,[99] and YeaW/X.[100] TMA is mostly produced by Bacteroidetes or Firmicutes bacteria.[101] Flavin containing monooxygenase 3 (FMO3) in the liver can promote TMA to produce TMAO.[102,103] Previous studies have found that the TMAO pathway is associated with the occurrence and development of diseases such as heart failure, chronic kidney disease, and obesity.[104,105] Research has shown that TMA and TMAO peak levels occur approximately 4 hours after a single feeding of a high-fat diet in fasted mice, indicating that TMA and TMAO are produced after meals and exhibit hormonal oscillations related to TMA source nutrient intake. Some TMA and TMAO are involved in phosphorylation processes such as PKA and insulin like growth factor 2 (IGF-2), and regulate the cascade reaction of insulin signaling.[106] Animal experiments have shown that mice fed a high-fat diet have an increase in TMAO, exacerbating impaired glucose tolerance and insulin resistance, and leading to inflammation of adipose tissue in mice fed a high-fat diet. This process may be related to block the insulin signaling pathway through the phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway.[107] An 8-week dietary intervention was conducted on patients with abnormal blood glucose levels, and the results showed a decrease in TMAO levels after intervention with a purely vegetarian diet, accompanied by a decrease in postprandial blood glucose levels.[108] In summary, the gut microbiota can affect PPG through the metabolite TMAO of a high-fat diet.

Gut microbiota in PPG and its role in precision medicine

Under the combined effects of the intestinal flora and other factors, when the same foods are eaten, blood glucose levels are differentially affected in individuals.[109] The development of individualized hypoglycemic intervention strategies for different blood glucose responses may facilitate more stable blood glucose control strategies. T1D depends on insulin treatment, and appropriate insulin doses are important to control blood glucose levels in patients. The traditional method of calculating good glycemic indices to guide insulin doses is not enough to control T1D blood glucose levels.[110] Based on the tenet that the gut microbiota impacts PPG, prediction models incorporating gut microbiota are important methods for predicting PPGR and may provide personalized treatments for T1D in the future.[111] Individualized nutrition interventions can also affect the gut microbiota and improve PPG. Studies have suggested that environmental factors may have greater roles than genetics in shaping human gut microbiota composition.[112] Personalized nutrition regimens, based on the microbiota, have been used to predict and guide blood glucose levels to generate individualized diabetes prevention and treatment strategies.[113] The cohort study of Zeevi et al.[45] monitored the weekly blood glucose level of 800 people and measured the blood glucose response to 46898 meals. The prediction algorithm based on the above data integrates the blood parameters, eating habits, anthropometry, physical activity and intestinal microbiota measured in the queue and can accurately predict the PPGR to the real diet. In addition, they validated these predictions in an independent 100-participant cohort, the authors showed that PPG levels in the bad diet group were significantly higher when compared with the good diet group, and the bad diet group had greater glucose fluctuations evaluated by the continuous blood glucose monitoring system after 1 week. Gut microbiota analyses indicated that Bifidobacteria and Bacteroidetes abundance were higher in the healthy diet group.

Conclusion

While numerous studies have shown that gut microbiota is related to PPG and can predict PPGR in non-diabetic populations, limited research focuses on how it predicts and affects PPGR in diabetic patients. More research is required in this area to identify precise interventions and reduce complication risks in diabetic patients. Gut microbiota generates many signaling metabolites, such as SCFAs, BAs, and TMAO, which participate in different metabolic pathways to ultimately affect PPG. However, the precise mechanisms underpinning their impact on PPG remain unclear. Future research must identify key gut microbiota and associated metabolites and pathways impacting PPG and provide potential therapeutic targets for improving PPG outcomes.


Chinese Academy of Medical Sciences, Beijing 100050, China.

Funding statement: This project was supported by the National High Level Hospital Clinical Research Funding (BJ-2022-145) and CAMS Innovation Fund for Medical Sciences (2021-I2M-1-050).

  1. Author Contributions

    Pan Q made substantial contributions to the study concept and design and critically revised the manuscript for important intellectual content. Feng X drafted the manuscript. Deng M and Zhang L helped to improve the English of the paper. All authors have read and approved the final version to be published.

  2. Conflicts of Interest

    The authors declare no competing interest.

  3. Data Availability Statement

    No additional data is available.

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;138:271–281.10.1016/j.diabres.2018.02.023Search in Google Scholar PubMed

2 Chinese Elderly Type 2 Diabetes Prevention and Treatment of Clinical Guidelines Writing Group Geriatric Geriatric Endocrinology and Metabolism Branch of Chinese Geriatric Society Geriatric Geriatric Endocrinology and Metabolism Branch of Chinese Geriatric Health Care Society Geriatric Geriatric Professional Committee of Beijing Medical Award Foundation National Clinical Medical Research Center for Geriatric Diseases (PLA General Hospital). Clinical guidelines for prevention and treatment of type 2 diabetes mellitus in the elderly in China (2022 edition). Zhong hua Nei Ke Za Zhi. 2022;61:12–50.Search in Google Scholar

3 Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, et al. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: Results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract 2019;157:107843.10.1016/j.diabres.2019.107843Search in Google Scholar PubMed

4 Sun H, Saeedi P, Karuranga S, Pinkepank M, Ogurtsova K, Duncan BB, et al. IDF Diabetes Atlas: Global, regional and country-level diabetes prevalence estimates for 2021 and projections for 2045. Diabetes Res Clin Pract 2022;183:109119.10.1016/j.diabres.2021.109119Search in Google Scholar PubMed

5 Cagnin S, Brugnaro M, Millino C, Pacchioni B, Troiano C, Di Sante M, et al. Monoamine Oxidase-Dependent Pro-Survival Signaling in Diabetic Hearts Is Mediated by miRNAs. Cells 2022;11:2697.10.3390/cells11172697Search in Google Scholar PubMed PubMed Central

6 Ali MK, Pearson-Stuttard J, Selvin E, Gregg EW. Interpreting global trends in type 2 diabetes complications and mortality. Diabetologia 2022;65:3–13.10.1007/s00125-021-05585-2Search in Google Scholar PubMed PubMed Central

7 Zhang M, Wang X, Liu M, Liu D, Pan J, Tian J, et al. Inhibition of PHLPP1 ameliorates cardiac dysfunction via activation of the PI3K/Akt/mTOR signalling pathway in diabetic cardiomyopathy. J Cell Mol Med 2020;24:4612–4623.10.1111/jcmm.15123Search in Google Scholar PubMed PubMed Central

8 Rohm TV, Meier DT, Olefsky JM, Donath MY. Inflammation in obesity, diabetes, and related disorders. Immunity. 2022;55:31-55.10.1016/j.immuni.2021.12.013Search in Google Scholar PubMed PubMed Central

9 Magee C, Grieve DJ, Watson CJ, Brazil DP. Diabetic Nephropathy: a Tangled Web to Unweave. Cardiovasc Drugs Ther 2017;31:579-592.10.1007/s10557-017-6755-9Search in Google Scholar PubMed PubMed Central

10 Vujosevic S, Aldington SJ, Silva P, Hernández C, Scanlon P, Peto T, et al. Screening for diabetic retinopathy: new perspectives and challenges. Lancet Diabetes Endocrinol 2020;8:337–347.10.1016/S2213-8587(19)30411-5Search in Google Scholar PubMed

11 Li J, Chandra A, Liu L, Zhang L, Xu J, Zhao M. Ocular findings, surgery details and outcomes in proliferative diabetic retinopathy patients with chronic kidney disease. PLoS One 2022;17:e0273133.10.1371/journal.pone.0273133Search in Google Scholar PubMed PubMed Central

12 Chen R, Ji L, Chen L, Chen L, Cai D, Feng B, et al. Glycemic control rate of T2DM outpatients in China: a multi-center survey. Med Sci Monit 2015;21:1440–1446.10.12659/MSM.892246Search in Google Scholar PubMed PubMed Central

13 Tily H, Patridge E, Cai Y, Gopu V, Gline S, Genkin M, et al. Gut Microbiome Activity Contributes to Prediction of Individual Variation in Glycemic Response in Adults. Diabetes Ther 2022;13:89–111.10.1007/s13300-021-01174-zSearch in Google Scholar PubMed PubMed Central

14 AlHaidar AM, AlShehri NA, AlHussaini MA. Family Support and Its Association with Glycemic Control in Adolescents with Type 1 Diabetes Mellitus in Riyadh, Saudi Arabia. J Diabetes Res 2020;2020:5151604.10.1155/2020/5151604Search in Google Scholar PubMed PubMed Central

15 Ketema EB, Kibret KT. Correlation of fasting and postprandial plasma glucose with HbA1c in assessing glycemic control; systematic review and meta-analysis. Arch Public Health 2015;73:43.10.1186/s13690-015-0088-6Search in Google Scholar PubMed PubMed Central

16 American Diabetes Association. 6. Glycemic Targets: Standards of Medical Care in Diabetes-2021. Diabetes Care 2021;44:S73-S84.10.2337/dc21-S006Search in Google Scholar PubMed

17 Muller PA, Matheis F, Schneeberger M, Kerner Z, Jové V, Mucida D. Microbiota-modulated CART+ enteric neurons autonomously regulate blood glucose. Science 2020;370:314–321.10.1126/science.abd6176Search in Google Scholar PubMed PubMed Central

18 Martín-Peláez S, Fito M, Castaner O. Mediterranean Diet Effects on Type 2 Diabetes Prevention, Disease Progression, and Related Mechanisms. A Review. Nutrients 2020;12:2236.10.3390/nu12082236Search in Google Scholar PubMed PubMed Central

19 Benítez-Páez A, Gómez Del Pugar EM, López-Almela I, Moya-Pérez Á, Codoñer-Franch P, Sanz Y. Depletion of Blautia Species in the Microbiota of Obese Children Relates to Intestinal Inflammation and Metabolic Phenotype Worsening. mSystems 2020;5:e00857–00819.10.1128/mSystems.00857-19Search in Google Scholar PubMed PubMed Central

20 Li X, Zhang ZH, Zabed HM, Yun J, Zhang G, Qi X. An Insight into the Roles of Dietary Tryptophan and Its Metabolites in Intestinal Inflammation and Inflammatory Bowel Disease. Mol Nutr Food Res 2021;65:e2000461.10.1002/mnfr.202000461Search in Google Scholar PubMed

21 Hyland NP, Cavanaugh CR, Hornby PJ. Emerging effects of tryptophan pathway metabolites and intestinal microbiota on metabolism and intestinal function. Amino Acids 2022;54:57–70.10.1007/s00726-022-03123-xSearch in Google Scholar PubMed

22 Yang X, Xie L, Li Y, Wei C. More than 9,000,000 unique genes in human gut bacterial community: estimating gene numbers inside a human body. PLoS One 2009;4:e6074.10.1371/journal.pone.0006074Search in Google Scholar PubMed PubMed Central

23 Martin AM, Sun EW, Rogers GB, Keating DJ. The Influence of the Gut Microbiome on Host Metabolism Through the Regulation of Gut Hormone Release. Front Physiol 2019;10:428.10.3389/fphys.2019.00428Search in Google Scholar PubMed PubMed Central

24 Stott KJ, Phillips B, Parry L, May S. Recent advancements in the exploitation of the gut microbiome in the diagnosis and treatment of colorectal cancer. Biosci Rep 2021;41:BSR20204113.10.1042/BSR20204113Search in Google Scholar PubMed PubMed Central

25 Dominguez-Bello MG, Godoy-Vitorino F, Knight R, Blaser MJ. Role of the microbiome in human development. Gut 2019;68:1108–1114.10.1136/gutjnl-2018-317503Search in Google Scholar PubMed PubMed Central

26 Sikalidis AK, Maykish A. The Gut Microbiome and Type 2 Diabetes Mellitus: Discussing a Complex Relationship. Biomedicines 2020;8:8.10.3390/biomedicines8010008Search in Google Scholar PubMed PubMed Central

27 Zhao J, Zhang X, Liu H, Brown MA, Qiao S. Dietary Protein and Gut Microbiota Composition and Function. Curr Protein Pept Sci 2019;20:145–154.10.2174/1389203719666180514145437Search in Google Scholar PubMed

28 Cani PD. Microbiota and metabolites in metabolic diseases. Nat Rev Endocrinol 2019;15:69–70.10.1038/s41574-018-0143-9Search in Google Scholar PubMed

29 Woerle HJ, Neumann C, Zschau S, Tenner S, Irsigler A, Schirra J, et al. Impact of fasting and postprandial glycemia on overall glycemic control in type 2 diabetes. Importance of postprandial glycemia to achieve target HbA1C levels. Diabetes Res Clin Pract 2007;77:280-285.10.1016/j.diabres.2006.11.011Search in Google Scholar PubMed

30 Monnier L, Colette C. Contributions of fasting and postprandial glucose to hemoglobin A1c. Endocr Pract 2006;12 Suppl 1:42-6.10.4158/EP.12.S1.42Search in Google Scholar PubMed

31 Deguchi S, Ogata F, Isaka T, Otake H, Nakazawa Y, Kawasaki N, et al. Prevention of Postprandial Hyperglycemia by Ophthalmic Nanoparticles Based on Protamine Zinc Insulin in the Rabbit. Pharmaceutics 2021;13:375.10.3390/pharmaceutics13030375Search in Google Scholar PubMed PubMed Central

32 Takao T, Takahashi K, Suka M, Suzuki N, Yanagisawa H. Association between postprandial hyperglycemia at clinic visits and all-cause and cancer mortality in patients with type 2 diabetes: A long-term historical cohort study in Japan. Diabetes Res Clin Pract 2019;148:152–159.10.1016/j.diabres.2019.01.006Search in Google Scholar PubMed

33 Aryangat AV, Gerich JE. Type 2 diabetes: postprandial hyperglycemia and increased cardiovascular risk. Vasc Health Risk Manag 2010;6:145–155.10.2147/VHRM.S8216Search in Google Scholar PubMed PubMed Central

34 Raz I, Ceriello A, Wilson PW, Battioui C, Su EW, Kerr L, et al. Post hoc subgroup analysis of the HEART2D trial demonstrates lower cardiovascular risk in older patients targeting postprandial versus fasting/premeal glycemia. Diabetes Care 2011;34:1511–1513.10.2337/dc10-2375Search in Google Scholar PubMed PubMed Central

35 Hassanein M, Akbar MAJ, Al-Shamiri M, Amir A, Amod A, Chudleigh R, et al. Management of Diabetes and Hypertension within the Gulf Region: Updates on Treatment Practices and Therapies. Diabetes Ther 2022;13:1253–1280.10.1007/s13300-022-01282-4Search in Google Scholar PubMed PubMed Central

36 Jin X, Lin S, Gao J, Kim EH, Morgenstern MP, Wilson AJ, et al. Ethnicity impact on oral processing behaviour and glycemic response to noodles: Chinese (Asian) vs. New Zealander (Caucasian). Food Funct 2022;13:3840–3852.10.1039/D1FO04078BSearch in Google Scholar PubMed

37 Ketel EC, de Wijk RA, de Graaf C, Stieger M. Relating oral physiology and anatomy of consumers varying in age, gender and ethnicity to food oral processing behavior. Physiol Behav 2020;215:112766.10.1016/j.physbeh.2019.112766Search in Google Scholar PubMed

38 Gribble FM, Reimann F. Function and mechanisms of enteroendocrine cells and gut hormones in metabolism. Nat Rev Endocrinol 2019;15:226–237.10.1038/s41574-019-0168-8Search in Google Scholar PubMed

39 Brønden A, Knop FK. Gluco-Metabolic Effects of Pharmacotherapy-Induced Modulation of Bile Acid Physiology. J Clin Endocrinol Metab 2020;105:dgz025.10.1210/clinem/dgz025Search in Google Scholar PubMed

40 Reunanen J, Kainulainen V, Huuskonen L, Ottman N, Belzer C, Huhtinen H, et al. Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer. Appl Environ Microbiol 2015;81:3655–3662.10.1128/AEM.04050-14Search in Google Scholar PubMed PubMed Central

41 Plovier H, Everard A, Druart C, Depommier C, Van Hul M, Geurts L, et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat Med 2017;23:107–113.10.1038/nm.4236Search in Google Scholar PubMed

42 Pustozerov E, Tkachuk A, Vasukova E, Dronova A, Shilova E, Anopova A, et al. The Role of Glycemic Index and Glycemic Load in the Development of Real-Time Postprandial Glycemic Response Prediction Models for Patients With Gestational Diabetes. Nutrients 2020;12:302.10.3390/nu12020302Search in Google Scholar PubMed PubMed Central

43 Rein M, Ben-Yacov O, Godneva A, Shilo S, Zmora N, Kolobkov D, et al. Effects of personalized diets by prediction of glycemic responses on glycemic control and metabolic health in newly diagnosed T2DM: a randomized dietary intervention pilot trial. BMC Med 2022;20:56.10.1186/s12916-022-02254-ySearch in Google Scholar PubMed PubMed Central

44 Attaye I, Pinto-Sietsma SJ, Herrema H, Nieuwdorp M. A Crucial Role for Diet in the Relationship Between Gut Microbiota and Cardiometabolic Disease. Annu Rev Med 2020;71:149–161.10.1146/annurev-med-062218-023720Search in Google Scholar PubMed

45 Zeevi D, Korem T, Zmora N, Israeli D, Rothschild D, Weinberger A, et al. Personalized Nutrition by Prediction of Glycemic Responses. Cell 2015;163:1079–1094.10.1016/j.cell.2015.11.001Search in Google Scholar PubMed

46 Søndertoft NB, Vogt JK, Arumugam M, Kristensen M, Gøbel RJ, Fan Y, et al. The intestinal microbiome is a co-determinant of the postprandial plasma glucose response. PLoS One 2020;15:e0238648.10.1371/journal.pone.0238648Search in Google Scholar PubMed PubMed Central

47 Nolte Fong JV, Miketinas D, Moore LW, Nguyen DT, Graviss EA, Ajami N, et al. Precision Nutrition Model Predicts Glucose Control of Overweight Females Following the Consumption of Potatoes High in Resistant Starch. Nutrients 2022;14:268.10.3390/nu14020268Search in Google Scholar PubMed PubMed Central

48 Jiang Z, Sun TY, He Y, Gou W, Zuo LS, Fu Y, et al. Dietary fruit and vegetable intake, gut microbiota, and type 2 diabetes: results from two large human cohort studies. BMC Med 2020;18:371.10.1186/s12916-020-01842-0Search in Google Scholar PubMed PubMed Central

49 Mineshita Y, Sasaki H, Kim HK, Shibata S. Relationship between Fasting and Postprandial Glucose Levels and the Gut Microbiota. Metabolites 2022;12:669.10.3390/metabo12070669Search in Google Scholar PubMed PubMed Central

50 He Xue D, Wang F, Wang Y, Xiao Y, Lv XG, Yi J. Systematic evaluation of intestinal flora characteristics and their correlation in patients with diabetes. Zhongguo Weishengtaixue Zazhi 2020;32:397-403.Search in Google Scholar

51 Sofi MH, Johnson BM, Gudi RR, Jolly A, Gaudreau MC, Vasu C. Polysaccharide A-Dependent Opposing Effects of Mucosal and Systemic Exposures to Human Gut Commensal Bacteroides fragilis in Type 1 Diabetes. Diabetes 2019;68:1975–1989.10.2337/db19-0211Search in Google Scholar PubMed PubMed Central

52 Leiva-Gea I, Sánchez-Alcoholado L, Martín-Tejedor B, Castellano-Castillo D, Moreno-Indias I, Urda-Cardona A, et al. Gut Microbiota Differs in Composition and Functionality Between Children With Type 1 Diabetes and MODY2 and Healthy Control Subjects: A Case-Control Study. Diabetes Care 2018;41:2385–2395.10.2337/dc18-0253Search in Google Scholar PubMed

53 Vitale M, Giacco R, Laiola M, Della Pepa G, Luongo D, Mangione A, et al. Acute and chronic improvement in postprandial glucose metabolism by a diet resembling the traditional Mediterranean dietary pattern: Can SCFAs play a role?Clin Nutr 2021;40:428–437.10.1016/j.clnu.2020.05.025Search in Google Scholar PubMed

54 Kim JS, Nam K, Chung SJ. Effect of nutrient composition in a mixed meal on the postprandial glycemic response in healthy people: a preliminary study. Nutr Res Pract 2019;13:126–133.10.4162/nrp.2019.13.2.126Search in Google Scholar PubMed PubMed Central

55 Meng H, Matthan NR, Ausman LM, Lichtenstein AH. Effect of macronutrients and fiber on postprandial glycemic responses and meal glycemic index and glycemic load value determinations. Am J Clin Nutr 2017;105:842–853.10.3945/ajcn.116.144162Search in Google Scholar PubMed PubMed Central

56 Shilo S, Godneva A, Rachmiel M, Korem T, Kolobkov D, Karady T, et al. Prediction of Personal Glycemic Responses to Food for Individuals With Type 1 Diabetes Through Integration of Clinical and Microbial Data. Diabetes Care 2022;45:502–511.10.2337/dc21-1048Search in Google Scholar PubMed

57 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;490:55–60.10.1038/nature11450Search in Google Scholar PubMed

58 Canfora EE, Meex RCR, Venema K, Blaak EE. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat Rev Endocrinol 2019;15:261–273.10.1038/s41574-019-0156-zSearch in Google Scholar PubMed

59 Xu HB. Analysis of intestinal flora characteristics and blood glucose control level in 315 elderly patients with type 2 diabetes. Chin J Physician 2017;19:130-132.Search in Google Scholar

60 Feng G, Zhong WZ, Zhao J, Wang XL. Effects of probiotics on constant natural killer T cells, blood glucose indicators, inflammatory factors and intestinal flora in patients with type 2 diabetes. Zhongguo Yaoye 2022;31: 50-54.Search in Google Scholar

61 Meyer D. Health benefits of prebiotic fibers. Adv Food Nutr Res 2015;74:47–91.10.1016/bs.afnr.2014.11.002Search in Google Scholar PubMed

62 Belén Sanz-Martos A, Fernández-Felipe J, Merino B, Cano V, Ruiz-Gayo M, Del Olmo N. Butyric Acid Precursor Tributyrin Modulates Hippocampal Synaptic Plasticity and Prevents Spatial Memory Deficits: Role of PPARγ and AMPK. Int J Neuropsychopharmacol 2022;25:498–511.10.1093/ijnp/pyac015Search in Google Scholar PubMed PubMed Central

63 Roh TT, Chen Y, Rudolph S, Gee M, Kaplan DL. InVitro Models of Intestine Innate Immunity. Trends Biotechnol 2021;39:274–285.10.1016/j.tibtech.2020.07.009Search in Google Scholar PubMed PubMed Central

64 Kwek E, Yan C, Ding H, Hao W, He Z, Ma KY, et al. Effects of Thermally-Oxidized Frying Oils (Corn Oil and Lard) on Gut Microbiota in Hamsters. Antioxidants (Basel) 2022;11:1732.10.3390/antiox11091732Search in Google Scholar PubMed PubMed Central

65 Hernández-Maldonado LM, Blancas-Benítez FJ, Zamora-Gasga VM, Cárdenas-Castro AP, Tovar J, Sáyago-Ayerdi SG. In Vitro Gastrointestinal Digestion and Colonic Fermentation of High Dietary Fiber and Antioxidant-Rich Mango (Mangifera indica L.) "Ataulfo"-Based Fruit Bars. Nutrients 2019;11:1564.10.3390/nu11071564Search in Google Scholar PubMed PubMed Central

66 Shang H, Zhao J, Dong X, Guo Y, Zhang H, Cheng J, et al. Inulin improves the egg production performance and affects the cecum microbiota of laying hens. Int J Biol Macromol 2020;155:1599–1609.10.1016/j.ijbiomac.2019.11.137Search in Google Scholar PubMed

67 Ting CH, Pan CY, Chen YC, Lin YC, Chen TY, Rajanbabu V, et al. Impact of Tilapia hepcidin 2-3 dietary supplementation on the gut microbiota profile and immunomodulation in the grouper (Epinephelus lanceolatus). Sci Rep 2019;9:19047.10.1038/s41598-019-55509-9Search in Google Scholar PubMed PubMed Central

68 Szczuko M, Kikut J, Maciejewska D, Kulpa D, Celewicz Z, Ziętek M. The Associations of SCFA with Anthropometric Parameters and Carbohydrate Metabolism in Pregnant Women. Int J Mol Sci 2020;21:9212.10.3390/ijms21239212Search in Google Scholar PubMed PubMed Central

69 Tolhurst G, Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, et al. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012;61:364–371.10.2337/db11-1019Search in Google Scholar PubMed PubMed Central

70 Tang C, Ahmed K, Gille A, Lu S, Gröne HJ, Tunaru S, et al. Loss of FFA2 and FFA3 increases insulin secretion and improves glucose tolerance in type 2 diabetes. Nat Med 2015;21:173–177.10.1038/nm.3779Search in Google Scholar PubMed

71 Shimizu H, Masujima Y, Ushiroda C, Mizushima R, Taira S, Ohue-Kitano R, et al. Dietary short-chain fatty acid intake improves the hepatic metabolic condition via FFAR3. Sci Rep 2019;9:16574.10.1038/s41598-019-53242-xSearch in Google Scholar PubMed PubMed Central

72 Louis P, Young P, Holtrop G, Flint HJ. Diversity of human colonic butyrate-producing bacteria revealed by analysis of the butyryl- CoA:acetate CoA-transferase gene. Environ Microbiol 2010;12:304–314.10.1111/j.1462-2920.2009.02066.xSearch in Google Scholar PubMed

73 Lu Y, Fan C, Liang A, Fan X, Wang R, Li P, et al. Effects of SCFA on the DNA methylation pattern of adiponectin and resistin in high-fat-diet-induced obese male mice. Br J Nutr 2018;120:385–392.10.1017/S0007114518001526Search in Google Scholar PubMed

74 Nishitsuji K, Xiao J, Nagatomo R, Umemoto H, Morimoto Y, Akatsu H, et al. Analysis of the gut microbiome and plasma short-chain fatty acid profiles in a spontaneous mouse model of metabolic syndrome. Sci Rep 2017;7:15876.10.1038/s41598-017-16189-5Search in Google Scholar PubMed PubMed Central

75 Larraufie P, Martin-Gallausiaux C, Lapaque N, Dore J, Gribble FM, Reimann F, et al. SCFAs strongly stimulate PYY production in human enteroendocrine cells. Sci Rep 2018;8:74.10.1038/s41598-017-18259-0Search in Google Scholar PubMed PubMed Central

76 Ye P, Xi Y, Huang Z, Xu P. Linking Obesity with Colorectal Cancer: Epidemiology and Mechanistic Insights. Cancers (Basel) 2020;12:1408.10.3390/cancers12061408Search in Google Scholar PubMed PubMed Central

77 Proungvitaya S, Sombattheera S, Boonsiri P, Limpaiboon T, Wongkham S, Wongkham C, et al. Diagnostic value of serum bile acid composition patterns and serum glycocholic acid levels in cholangiocarcinoma. Oncol Lett 2017;14:4943–4948.10.3892/ol.2017.6763Search in Google Scholar PubMed PubMed Central

78 Lin H, An Y, Hao F, Wang Y, Tang H. Correlations of Fecal Metabonomic and Microbiomic Changes Induced by High-fat Diet in the Pre-Obesity State. Sci Rep 2016;6:21618.Search in Google Scholar

79 Li M, Hu X, Xu Y, Hu X, Zhang C, Pang S. A Possible Mechanism of Metformin in Improving Insulin Resistance in Diabetic Rat Models. Int J Endocrinol 2019;2019:3248527.10.1155/2019/3248527Search in Google Scholar PubMed PubMed Central

80 Thomas C, Pellicciari R, Pruzanski M, Auwerx J, Schoonjans K. Targeting bile-acid signalling for metabolic diseases. Nat Rev Drug Discov 2008;7:678–693.10.1038/nrd2619Search in Google Scholar PubMed

81 Kiriyama Y, Nochi H. The Biosynthesis, Signaling, and Neurological Functions of Bile Acids. Biomolecules 2019;9:232.10.3390/biom9060232Search in Google Scholar PubMed PubMed Central

82 Kim KH, Choi S, Zhou Y, Kim EY, Lee JM, Saha PK, et al. Hepatic FXR/SHP axis modulates systemic glucose and fatty acid homeostasis in aged mice. Hepatology 2017;66:498–509.10.1002/hep.29199Search in Google Scholar PubMed PubMed Central

83 Liu H, Pathak P, Boehme S, Chiang JL. Cholesterol 7α-hydroxylase protects the liver from inflammation and fibrosis by maintaining cholesterol homeostasis. J Lipid Res 2016;57:1831–1844.10.1194/jlr.M069807Search in Google Scholar PubMed PubMed Central

84 Li T, Owsley E, Matozel M, Hsu P, Novak CM, Chiang JY. Transgenic expression of cholesterol 7alpha-hydroxylase in the liver prevents high-fat diet-induced obesity and insulin resistance in mice. Hepatology 2010;52:678–690.10.1002/hep.23721Search in Google Scholar PubMed PubMed Central

85 Katsuma S, Hirasawa A, Tsujimoto G. Bile acids promote glucagon-like peptide-1 secretion through TGR5 in a murine enteroendocrine cell line STC-1. Biochem Biophys Res Commun 2005;329:386–390.10.1016/j.bbrc.2005.01.139Search in Google Scholar PubMed

86 Shapiro H, Kolodziejczyk AA, Halstuch D, Elinav E. Bile acids in glucose metabolism in health and disease. J Exp Med 2018;215:383–396.10.1084/jem.20171965Search in Google Scholar PubMed PubMed Central

87 Schittenhelm B, Wagner R, Kähny V, Peter A, Krippeit-Drews P, Düfer M, et al. Role of FXR in β-cells of lean and obese mice. Endocrinology 2015;156:1263–1271.10.1210/en.2014-1751Search in Google Scholar PubMed

88 Kumar DP, Asgharpour A, Mirshahi F, Park SH, Liu S, Imai Y, et al. Activation of Transmembrane Bile Acid Receptor TGR5 Modulates Pancreatic Islet α Cells to Promote Glucose Homeostasis. J Biol Chem 2016;291:6626–6640.10.1074/jbc.M115.699504Search in Google Scholar PubMed PubMed Central

89 Hansen M, Sonne DP, Mikkelsen KH, Gluud LL, Vilsbøll T, Knop FK. Bile acid sequestrants for glycemic control in patients with type 2 diabetes: A systematic review with meta-analysis of randomized controlled trials. J Diabetes Complications 2017;31:918–927.10.1016/j.jdiacomp.2017.01.011Search in Google Scholar PubMed

90 Bozadjieva N, Heppner KM, Seeley RJ. Targeting FXR and FGF 19 to Treat Metabolic Diseases-Lessons Learned From Bariatric Surgery. Diabetes 2018;67:1720–1728.10.2337/dbi17-0007Search in Google Scholar PubMed PubMed Central

91 So SSY, Yeung CHC, Schooling CM, El-Nezami H. Targeting bile acid metabolism in obesity reduction: A systematic review and meta-analysis. Obes Rev 2020;21:e13017.10.1111/obr.13017Search in Google Scholar PubMed

92 Kaska L, Sledzinski T, Chomiczewska A, Dettlaff-Pokora A, Swierczynski J. Improved glucose metabolism following bariatric surgery is associated with increased circulating bile acid concentrations and remodeling of the gut microbiome. World J Gastroenterol 2016;22:8698–8719.10.3748/wjg.v22.i39.8698Search in Google Scholar PubMed PubMed Central

93 Lee JH, Lee J. Indole as an intercellular signal in microbial communities. FEMS Microbiol Rev 2010;34:426–444.10.1111/j.1574-6976.2009.00204.xSearch in Google Scholar PubMed

94 Wyatt M, Greathouse KL. Targeting Dietary and Microbial Tryptophan-Indole Metabolism as Therapeutic Approaches to Colon Cancer. Nutrients 2021;13:1189.10.3390/nu13041189Search in Google Scholar PubMed PubMed Central

95 Abildgaard A, Elfving B, Hokland M, Wegener G, Lund S. The microbial metabolite indole-3-propionic acid improves glucose metabolism in rats, but does not affect behaviour. Arch Physiol Biochem 2018;124:306–312.10.1080/13813455.2017.1398262Search in Google Scholar PubMed

96 Qi Q, Li J, Yu B, Moon JY, Chai JC, Merino J, et al. Host and gut microbial tryptophan metabolism and type 2 diabetes: an integrative analysis of host genetics, diet, gut microbiome and circulating metabolites in cohort studies. Gut 2022;71:1095–1105.10.1136/gutjnl-2021-324053Search in Google Scholar PubMed PubMed Central

97 Craciun S, Balskus EP. Microbial conversion of choline to trimethylamine requires a glycyl radical enzyme. Proc Natl Acad Sci U S A 2012;109:21307–21312.10.1073/pnas.1215689109Search in Google Scholar PubMed PubMed Central

98 Kalnins G, Kuka J, Grinberga S, Makrecka-Kuka M, Liepinsh E, Dambrova M, et al. Structure and Function of CutC Choline Lyase from Human Microbiota Bacterium Klebsiella pneumoniae. J Biol Chem 2015;290:21732–21740.10.1074/jbc.M115.670471Search in Google Scholar PubMed PubMed Central

99 Zhu Y, Jameson E, Crosatti M, Schäfer H, Rajakumar K, Bugg TD, et al. Carnitine metabolism to trimethylamine by an unusual Riesketype oxygenase from human microbiota. Proc Natl Acad Sci U S A 2014;111:4268–4273.10.1073/pnas.1316569111Search in Google Scholar PubMed PubMed Central

100 Koeth RA, Levison BS, Culley MK, Buffa JA, Wang Z, Gregory JC, et al. γ-Butyrobetaine is a proatherogenic intermediate in gut microbial metabolism of L-carnitine to TMAO. Cell Metab 2014;20:799–812.10.1016/j.cmet.2014.10.006Search in Google Scholar PubMed PubMed Central

101 Lin H, An Y, Hao F, Wang Y, Tang H. Correlations of Fecal Metabonomic and Microbiomic Changes Induced by High-fat Diet in the Pre-Obesity State. Sci Rep 2016;6:21618.10.1038/srep21618Search in Google Scholar PubMed PubMed Central

102 Tang WH, Hazen SL. The contributory role of gut microbiota in cardiovascular disease. J Clin Invest 2014;124:4204–4211.10.1172/JCI72331Search in Google Scholar PubMed PubMed Central

103 Aron-Wisnewsky J, Clément K. The gut microbiome, diet, and links to cardiometabolic and chronic disorders. Nat Rev Nephrol 2016;12:169–181.10.1038/nrneph.2015.191Search in Google Scholar PubMed

104 Warrier M, Shih DM, Burrows AC, Ferguson D, Gromovsky AD, Brown AL, et al. The TMAO-Generating Enzyme Flavin Monooxygenase 3 Is a Central Regulator of Cholesterol Balance. Cell Rep 2015;10:326–338.10.1016/j.celrep.2014.12.036Search in Google Scholar PubMed PubMed Central

105 Miao J, Ling AV, Manthena PV, Gearing ME, Graham MJ, Crooke RM, et al. Flavin-containing monooxygenase 3 as a potential player in diabetes-associated atherosclerosis. Nat Commun 2015;6:6498.10.1038/ncomms7498Search in Google Scholar PubMed PubMed Central

106 Schugar RC, Willard B, Wang Z, Brown JM. Postprandial gut microbiotadriven choline metabolism links dietary cues to adipose tissue dysfunction. Adipocyte 2018;7:49–56.10.1080/21623945.2017.1398295Search in Google Scholar PubMed PubMed Central

107 Gao X, Liu X, Xu J, Xue C, Xue Y, Wang Y. Dietary trimethylamine N-oxide exacerbates impaired glucose tolerance in mice fed a high fat diet. J Biosci Bioeng 2014;118:476–481.10.1016/j.jbiosc.2014.03.001Search in Google Scholar PubMed

108 Argyridou S, Davies MJ, Biddle GJH, Bernieh D, Suzuki T, Dawkins NP, et al. Evaluation of an 8-Week Vegan Diet on Plasma Trimethylamine-N-Oxide and Postchallenge Glucose in Adults with Dysglycemia or Obesity. J Nutr 2021;151:1844–1853.10.1093/jn/nxab046Search in Google Scholar PubMed PubMed Central

109 Huda MN, Salvador AC, Barrington WT, Gacasan CA, D'Souza EM, Deus Ramirez L, et al. Gut microbiota and host genetics modulate the effect of diverse diet patterns on metabolic health. Front Nutr 2022;9:896348.10.3389/fnut.2022.896348Search in Google Scholar PubMed PubMed Central

110 Krzymien J, Ladyzynski P. Insulin in Type 1 and Type 2 Diabetes-Should the Dose of Insulin Before a Meal be Based on Glycemia or Meal Content?Nutrients 2019;11:607.10.3390/nu11030607Search in Google Scholar PubMed PubMed Central

111 Dedrick S, Sundaresh B, Huang Q, Brady C, Yoo T, Cronin C, et al. The Role of Gut Microbiota and Environmental Factors in Type 1 Diabetes Pathogenesis. Front Endocrinol (Lausanne) 2020;11:78.10.3389/fendo.2020.00078Search in Google Scholar PubMed PubMed Central

112 Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature 2018;555:210–215.10.1038/nature25973Search in Google Scholar PubMed

113 Kolodziejczyk AA, Zheng D, Elinav E. Diet-microbiota interactions and personalized nutrition. Nat Rev Microbiol 2019;17:742–753.10.1038/s41579-019-0256-8Search in Google Scholar PubMed

Published Online: 2023-12-20

© 2023 Xinyuan Feng, Mingqun Deng, Lina Zhang, Qi Pan, published by De Gruyter on behalf of Scholar Media Publishing

This work is licensed under the Creative Commons Attribution 4.0 International License.

Articles in the same Issue

  1. Perspective
  2. Mucus hypersecretion in chronic obstructive pulmonary disease: From molecular mechanisms to treatment
  3. Timing of TIPS for the management of portal vein thrombosis in liver cirrhosis
  4. Commentary
  5. Could unpublishing negative results be harmful to the general public?
  6. Review Article
  7. Oncogenic KRAS triggers metabolic reprogramming in pancreatic ductal adenocarcinoma
  8. Mitochondrial damage-associated molecular patterns in chronic obstructive pulmonary disease: Pathogenetic mechanism and therapeutic target
  9. Exosomes and their derivatives as biomarkers and therapeutic delivery agents for cardiovascular diseases: Situations and challenges
  10. Autophagy-dependent ferroptosis in infectious disease
  11. Impact of gut microbiota and associated mechanisms on postprandial glucose levels in patients with diabetes
  12. Circular RNA: A promising new star of vaccine
  13. Crosstalk between gut microbiota and gut resident macrophages in inflammatory bowel disease
  14. Original Article
  15. Hypervolemia suppresses dilutional anaemic injury in a rat model of haemodilution
  16. Prognostic value of serum ammonia in critical patients with non-hepatic disease: A prospective, observational, multicenter study
  17. Global lineage evolution pattern of sars-cov-2 in Africa, America, Europe, and Asia: A comparative analysis of variant clusters and their relevance across continents
  18. Efficacy and safety of QL0911 in adult patients with chronic primary immune thrombocytopenia: A multicenter, randomized, double-blind, placebo-controlled, phase III trial
  19. A pan-cancer analysis of the oncogenic role of Golgi transport 1B in human tumors
  20. Short-term duration of diabetic retinopathy as a predictor for development of diabetic kidney disease
  21. Cardiac magnetic resonance imaging-derived septum swing index detects pulmonary hypertension: A diagnostic study
  22. Letter to Editor
  23. Temporal trend of acute myocardial infarction-related mortality and associated racial/ethnic disparities during the omicron outbreak
Downloaded on 11.9.2025 from https://www.degruyterbrill.com/document/doi/10.2478/jtim-2023-0116/html
Scroll to top button