Next Article in Journal
Identification and Structural Elucidation of Anti-Inflammatory Compounds from Chinese Olive (Canarium Album L.) Fruit Extracts
Next Article in Special Issue
In Vitro Studies of Fermented Korean Chung-Yang Hot Pepper Phenolics as Inhibitors of Key Enzymes Relevant to Hypertension and Diabetes
Previous Article in Journal
The Protective Effect of Brazilian Propolis against Glycation Stress in Mouse Skeletal Muscle
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Targeting Gut Microbiota for the Prevention and Management of Diabetes Mellitus by Dietary Natural Products

1
Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
2
Institute of Urban Agriculture, Chinese Academy of Agricultural Sciences, Chengdu 610213, China
3
Department of Food Science & Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
4
School of Food and Biological Engineering, Jiangsu University, Zhenjiang 212013, China
*
Authors to whom correspondence should be addressed.
Foods 2019, 8(10), 440; https://doi.org/10.3390/foods8100440
Submission received: 17 August 2019 / Revised: 20 September 2019 / Accepted: 23 September 2019 / Published: 25 September 2019
(This article belongs to the Special Issue The Benefits of Plant Extracts for Human Health)

Abstract

:
Diabetes mellitus is one of the biggest public health concerns worldwide, which includes type 1 diabetes mellitus, type 2 diabetes mellitus, gestational diabetes mellitus, and other rare forms of diabetes mellitus. Accumulating evidence has revealed that intestinal microbiota is closely associated with the initiation and progression of diabetes mellitus. In addition, various dietary natural products and their bioactive components have exhibited anti-diabetic activity by modulating intestinal microbiota. This review addresses the relationship between gut microbiota and diabetes mellitus, and discusses the effects of natural products on diabetes mellitus and its complications by modulating gut microbiota, with special attention paid to the mechanisms of action. It is hoped that this review paper can be helpful for better understanding of the relationships among natural products, gut microbiota, and diabetes mellitus.

1. Introduction

Over the past 20 years, the prevalence of diabetes mellitus (DM) and its complications has rapidly increased across the world, which poses a serious threat to global health [1,2,3]. The international diabetes federation (IDF) estimated that one in eleven adults aged 20 to 79 (415 million) have DM globally in 2015, and predicts that the number can rise to 642 million by 2040 [4]. DM is a complex metabolic disorder characterized with the functional impairment or a lack of insulin-producing β-cells, alone or in combination with insulin resistance [5]. DM has several types, including type 1 diabetes mellitus (T1DM), type 2 diabetes mellitus (T2DM), gestational diabetes mellitus (GDM), and other rare forms of diabetes mellitus [6,7]. T1DM is an autoimmune disease that is associated with the aberrant immune responses to specific β-cell autoantigens, which causes the reduction of insulin [8,9,10,11]. T2DM is due to insulin resistance in the target tissue and a relative lack of insulin secreted by islet β-cells, and is mainly affected by the combination of genetic susceptibility and lifestyle factors [12,13,14,15]. GDM is a kind of diabetes during pregnancy due to the increased severity of insulin resistance and an impairment of the compensatory increase in insulin secretion [16,17,18]. Other rare forms of diabetes mellitus, such as maturity-onset of diabetes of the young (MODY) resulted from mutations in a single gene. In addition, maternally inherited diabetes with deafness (MIDD) is caused by mitochondrial mutations, and rare forms resulted from insulin gene mutations [19,20]. In this review paper, only T2DM and T1DM will be discussed, because T2DM and T1DM are the main types of diabetes mellitus, accounting for 90%–95% and 5%–10%, respectively [4,21,22]. In addition, T2DM can induce various complications, which contributes to high morbidity and mortality, such as diabetic cardiovascular diseases, nephropathy, retinopathy, neuropathy, and erectile dysfunctions [23,24]. Furthermore, many studies have shown that some natural products and their bioactive components could prevent and manage diabetes mellitus and its complications by several possible mechanisms, such as enhancing insulin action, ameliorating insulin resistance, activating insulin signaling pathway, protecting islet β-cells, scavenging free radicals, decreasing inflammation, and modulating gut microbiota [25,26,27,28,29]. In recent years, the role of gut microbiota in the prevention and management of diabetes mellitus and its complications has been a hot topic in research [14,30,31,32]. Special attention will be paid to the role of gut microbiota in this paper.
Gut microbiota has been recently reported to be involved in the pathogenesis of several diseases, such as obesity [33,34,35,36,37], liver diseases [38,39,40,41], cardiovascular diseases [42,43,44], cancer [45,46], and DM [47,48,49,50]. The alteration of intestinal microbiota and the production of metabolites have been demonstrated to play a critical role in the initiation and development of DM [51,52,53,54]. Some bacteria like Saccharomyces boulardii Biocodex, Bifidobacterium animalis subsp. lactis 420, Lactobacillus casei CCFM419, and Clostridium butyricum CGMCC0313.1 have been shown to ameliorate DM [55,56,57,58,59]. However, it was reported that some bacteria, like Bacteroides and Candida albicans, promoted the incidence and progression of T2DM in animal studies [60,61]. On the other hand, natural products have attracted wide attention in recent years due to their diverse bioactivities, such as cardiovascular protective, hepatoprotective, anti-cancer, anti-obesity, and anti-diabetic effects [25,36,62,63,64,65,66,67,68,69,70,71]. Furthermore, natural products are also important factors in regulating gut microbiota [72,73,74]. A variety of dietary natural products and their bioactive components have shown anti-diabetic effects by regulating gut microbiota composition and abundance, the change of gut permeability, the production of short-chain fatty acids (SCFAs), the decrease of lipopolysaccharides (LPS), and the inhibition of inflammation [58,75,76,77,78,79,80]. In addition, natural products can be effective adjuvant agents in the therapy of T2DM with lower cost and less side effects [81,82]. In this review paper, we first address the relationship between gut microbiota and T1DM as well as T2DM based on animal and epidemiological studies, and then discuss the effects of natural products on T2DM and its complications via modulation of gut microbiota, based on the literature from animal and clinical studies in the last five years (2015–2019).

2. The Association Between Gut Microbiota and DM Based on Animal and Epidemiological Studies

Many animal studies have revealed that the gut microbiota plays an important role in the incidence and development of T1DM as well as T2DM [54,75,83,84]. In addition, increasing investigations have found that gut microbiota is closely associated with T1DM and T2DM patients [85,86,87], and they will be discussed in detail below.

2.1. The Association between Gut Microbiota and T1DM

The animal studies have shown that T1DM has been associated with the composition of the gut microbiota [88]. For example, the abundance of Proteobacteria showed a more marked change than that of the predominant phyla, such as Firmicutes and Bacteroidetes compared with the healthy control in the rat model of T1DM induced by streptozotocin [89]. In another study, the gut microbiota from non-obese diabetic (NOD) mice without myeloid differentiation primary response gene 88 (MyD88) was transferred to wild-type NOD mice, which reduced the intensity of insulitis and delayed the onset of autoimmune diabetes in recipients, by decreasing the amount of Lactobacillaceae and increasing the number of Lachnospiraceae and Clostridiaceae [59]. Moreover, the deficiency of TIR-domain-containing adapter-inducing interferon-β in NOD mice could prevent diabetes via the change in the gut microbiota, such as the proportion of Sutterella, Rikenella, and Turicibacter species [90]. In addition, the mouse β-defensin 14 (mBD14) in pancreatic endocrine cells in NOD mice could prevent autoimmune diabetes, while the microbiota dysbiosis could induce the deficiency of the pancreatic expression of mBD14, which results in the higher incidence of T1DM [91].
The relationship between gut microbiota and T1DM has been summarized in a review paper published in 2015 [54]. In addition, the epidemiological studies have reported that some intestinal microbiota like phylum Bacteroidetes, genus Clostridium, Bacteroides, and Veillonella as well as Candida albicans increased in T1DM patients, while some other gut microbiota like phylum Actinobacteria and Firmicutes, genus Lactobacillus, Bifidobacterium, and Faecalibacterium as well as the ratio of Firmicutes to Bacteroidetes decreased in T1DM patients [92,93,94,95,96].

2.2. The Association between Gut Microbiota and T2DM

The animal studies have found that there have been prominent differences in the composition of gut microbiota between T2DM animal models and control subjects. For example, an in vivo study found that the diabetic db/db mice had higher abundance of phyla Firmicutes, Proteobacteria, and Fibrobacteres [97]. Additionally, the streptozotocin-induced T2DM mice showed an increase in Brevibacterium, Corynebacterium, and Facklamia compared with wild type mice [98]. A decrease in the diversity of gut microbiota and a lack of butyrate-producing bacteria were observed in T2DM cats [99]. The level of serum fructosamine was inversely associated with the abundance of Prevotellaceae, and was positively associated with the abundance of Enterobacteriaceae. In addition, the transfer of Lachnospiraceae (strain AJ110941) from the feces of hyperglycemic obese mice to germ-free ob/ob mice contributed to the development of T2DM in ob/ob mice. The colonization of Lachnospiraceae could induce an increase in the level of fasting blood glucose (FBG) and a decrease in the level of plasma insulin as well as homeostasis model assessment-β (HOMA-β). The decrease in HOMA-β values indicated the dysfunction of pancreatic β-cells [100].
The epidemiological studies have also investigated the relationship between gut microbiota and T2DM patients [101,102,103,104]. For example, a case-control study has shown that the proportions of phylum Firmicutes and class Clostridia as well as the ratio of Firmicutes to Bacteroidetes decreased, whereas the abundance of class Betaproteobacteria increased in T2DM patients [101]. In addition, the concentration of Faecalibacterium prausnitzii and the abundance of genus Blautia was reduced in T2DM patients [102,103]. Moreover, a case-control study demonstrated that T2DM and obese patients had a lower quantity of Lactobacillus, especially its subgroups (L. acidophilus, L. plantarum, and L. reuteri) in comparison with controls [104].
Overall, several animal studies have revealed that the intestinal microbiota play an important role in T1DM and T2DM. In addition, many epidemiological investigations have demonstrated the closed association between gut microbiota and T2DM (Table 1). Furthermore, there are few prospective studies on the relationship between gut microbiota and DM, which should be further investigated in the future.

3. The Relationships among Natural Products, Gut Microbiota, and T2DM as well as T1DM Based on Animal Studies

Some natural products, such as vegetables, fruits, dietary fibers, and medicinal plants, have shown potential preventive effects against T2DM and T1DM as well as diabetic complications with mechanisms of action, at least partly, via the modulation of gut microbiota [80,105,106,107,108].

3.1. Diabetes Mellitus

The extracts of vegetables and their bioactive components have been demonstrated to alleviate T2DM by modulating the gut microbiota [75,79,80]. Capsaicin could improve the glucose homeostasis and insulin tolerance in obese diabetic ob/ob mice by increasing the production of SCFAs and modulating gut microbiota. It could increase the ratio of Firmicutes to Bacteroidetes and the quantity of Roseburia, and reduce the quantities of Bacteroides and Parabacteroides at the genus level, which could decrease the levels of pro-inflammatory cytokines, such as TNF-α and IL-6 [79]. Additionally, pumpkin polysaccharide alleviated the T2DM in mice fed with a high-fat diet (HFD). Its antidiabetic effects were also related to the increase in SCFAs production and selective enhancement of some bacteria, such as Bacteroidetes, Prevotella, and Deltaproteobacteria [75]. Moreover, the carrot juice fermented with Lactobacillus rhamnosus GG was demonstrated to ameliorate T2DM in rats via the increase in SCFAs in the cecum and the change in the composition and abundance of gut microbiota, such as Oscillibacter and Akkermansia [80]. In addition, the anti-diabetic activity of Momordica charantia (bitter melon) was enhanced by the fermentation of Lactobacillus, which could induce an increase in the abundance of Bacteroides caecigallinarwn, Bacteroides thetaiotaomicron, Prevotella loescheii, Prevotella oralis, and Prevotella melaninogenica as well as the level of SCFAs [109].
It has also been reported that the extracts of several fruits and their bioactive components could improve T2DM via gut microbiota [105,110,111]. The phlorizin in many fruits could competitively inhibit the sodium-glucose symporters and regulate the level of blood glucose by decreasing the level of serum LPS and insulin resistance, and increasing the level of SCFAs, especially butyric acid. It could also increase the quantity of Akkermansia muciniphila and Prevotella, which modulates the gut microbial community structure [110]. In addition, polysaccharides from the mulberry fruit have shown protective effects against T2DM via gut microbiota in db/db mice, and the treatment with polysaccharides could enrich the functional bacteria, such as Bacteroidales, Lactobacillus, Allobaculum, Bacteroides, and Akkermansia [105]. The extracts from cinnamon bark were reported to improve glucose tolerance and insulin resistance by reducing the abundance of genus Peptococcus, and the extracts from grape pomace could reduce Desulfovibrio and Lactococcus, and increase Allobaculum and Roseburia in diabetic mice [111].
The inulin was also found to alleviate different stages of T2DM in diabetic mice by modulating gut microbiota. It increased the relative abundance of Cyanobacteria and Bacteroides, and reduced the relative abundance of Deferribacteres and Tenericutes [112]. In another study, the oral administration with inulin-type fructan could reduce the FBG level, increase the glucagon-like peptide-1 (GLP-1) level, and alleviate glucose intolerance as well as blood lipid in T2DM rats induced by HFD and streptozotocin [108]. The treatment with inulin also increased the level of GLP-1 and improved the gut microbiota dysbiosis by enriching probiotic bacteria Lactobacillus and SCFAs-producing bacteria Lachnospiraceae, Phascolarctobacterium, and Bacteroides. Furthermore, the supplementation of the long-chain inulin-type fructan fibers from chicory root to female NOD mice could delay the progression of T1DM via the modulation of gut microbiota. This fiber could increase the ratio of Firmicutes to Bacteroidetes and the abundance of Ruminococcaceae and Lactobacilli, which might contribute to the increase in the expression of tight junction proteins occludin and claudin-2 and an antidiabetogenic effect [106].
Some traditional Chinese medicine have been found to be effective in preventing and treating T2DM. The administration of ethanol extract of Atractylodis macrocephalae rhizoma improved the glucose metabolism by regulating the gut microbiota in diabetic db/db mice, and it could increase the abundance of Bacteroides thetaiotaomicron and Methanobrevibacter smithii [113]. In addition, the extract of Alpinia oxyphylla Miq. had protective effects against diabetes in T2DM db/db mice. It decreased the blood glucose levels by modulating the intestinal microbiota composition, which increased the abundance of Akkermansia and decreased Helicobacter [114]. The water extract of Potentilla discolor Bunge could alleviate diabetes, which might be related to the change in the gut microbiota. The treatment reduced the relative abundance of Proteobacteria in T2DM mice induced by HFD and streptozotocin [77]. It was related to an increase in levels of fecal acetic acid, butyric acid, and their specific receptors including the expression of G-protein-coupled receptor 41 (GPR41) and G-protein-coupled receptor 43 (GPR43), which increased the insulin sensitivity and decreased the fat accumulation. Additionally, the expressions of toll-like receptor-4 (TLR4), MyD88, nuclear factor-kappa B (NF-κB), and gut mucosal tight junction proteins (caudin-3 and occludin) were decreased, which could reduce the gut permeability and inflammation. In addition, the oral administration of total saponins and polysaccharides in Polygonatum kingianum could prevent T2DM by regulating the gut microbiota [115]. The treatment improved micro-ecology in the gut by reducing the abundance of Bacteroidetes and Proteobacteria, and increasing Firmicutes. Furthermore, the baicalein showed anti-diabetic effects on diabetic rats, which reduced the levels of blood glucose, LPS, and insulin resistance. The treatment of baicalein could regulate the gut microbiota by increasing the relative abundance of Bacteroides and Bacteroidales S24-7 [78]. It could also increase the production of SCFAs as well as the thickness of the gut mucus layer.
Overall, many studies have focused on the effects of different natural products, such as vegetables, fruits, dietary fibers, and medicinal plants, on T2DM by regulating gut microbiota and improving microecology in the gut. On the other hand, there are few studies about the effect of natural products on T1DM by modulating gut microbiota. In the future, more studies are needed to investigate the effects of different natural products and bioactive components on T1DM via gut microbiota-related mechanisms.

3.2. Diabetic Complications

Long-term diabetes increases the likelihood of its complications like diabetic erectile dysfunctions, nephropathy, cardiovascular diseases, retinopathy, and neuropathy [116,117]. The gut microbiota has also been found to have a close relationship with the development of the diabetic complications. In the rat models with T2DM erectile dysfunction, there was a reduction in the relative abundance of beneficial bacteria like Allobaculum, Bifidobacterium, Eubacterium, and Anaerotruncus. In addition, the relative abundance of opportunistic pathogens increased, such as Enterococcus, Corynebacterium, Aerococcus, and Facklamia [118]. Some natural products have been reported to have protective effects on diabetic nephropathy or kidney injury in diabetic mice by modulating the gut microbiota balance [107,119]. For example, the polyphenolic extract of Dendrobium loddigesii could improve the symptoms of diabetes and complications in diabetic db/db mice. These effects were likely related to the improvement of the intestinal flora balance, with an increase in the relative abundance of Prevotella and Akkermansia, and the reduction in the relative abundance of S24-7/Rikenella/Escherichia coli [119]. Additionally, the treatment with the water-ethanolic extract of green macroalgae Enteromorpha prolifera reduced the inflammation in the liver and kidney by significantly increasing the abundance of Lachnospiraceae and Alisties as well as regulating the insulin signaling pathway in T2DM mice induced by HFD and high sucrose diet (HSD) and streptozotocin [107].
To sum up, several studies have demonstrated that the gut microbiota has a tight association with diabetic complications. The increase or decrease in different types of gut microbiota could exert different effects on the progression of diabetic complications. Moreover, several natural products have showed potential efficacy on the prevention and management against diabetic complications, and these effects were mainly associated with the modulation of gut microbiota composition and abundance, regulation of the insulin signaling pathway, and inhibition of inflammation.
Lastly, the relationship among gut microbiota, T2DM, and its complications is shown in Figure 1, and the association among natural products, gut microbiota, T2DM, and its complications is given in Table 2 and Figure 2.

4. The Relationships among Natural Products, Gut Microbiota, and T2DM Based on Clinical Studies

Several clinical trials have evaluated the protective effects of natural products on T2DM via gut microbiota [120,121]. A double-blind, randomized, controlled clinical trial (RCT) involving 60 patients with T2DM found that the supplementation of 10 g/day inulin powder promoted the gut health by increasing the proportion of Akkermansia muciniphila [120]. Moreover, an RCT study found that the fiber-rich macrobiotic Ma-Pi 2 diet could regulate gut microbiome dysbiosis in obese T2DM patients by increasing the gut microbiota ecosystem diversity, recovering the SCFAs-producing bacteria community, such as Roseburia, Lachnospira, Faecalibacterium, Bacteroides, and Akkermansia, and inhibiting the increase of the pro-inflammatory group, such as Collinsella and Streptococcus [122]. Furthermore, an RCT study including 100 patients with T2DM reported that a Chinese herbal formula could ameliorate T2DM and improved the glucose and lipid homeostasis by increasing the abundance of Faecalibacterium spp. [116]. Another RCT study observed that the treatment with Gegen Qinlian decoction, which is a type of Chinese herbal formula, at the moderate and high doses, significantly reduced the mean alterations in adjusted FBG and glycated hemoglobin A1c (HbA1c) levels by enriching the beneficial bacteria, such as Faecalibacterium prausnitzii [121].
To sum up, several clinical trials demonstrate that natural products can ameliorate T2DM by regulating gut microbiota (Table 3). Some natural products, such as inulin and medicine plants, are able to enrich certain beneficial bacteria in T2DM patients, such as Akkermansia muciniphila, Faecalibacterium, and Lachnospira.

5. Conclusions

In conclusion, the relationship among diabetes mellitus (including T1DM and T2DM), gut microbiota, and natural products has been summarized and discussed. Animal and epidemiological studies found significant differences in intestinal microbiota composition and abundance between diabetic patients and healthy controls. The abundance of Bifidobacterium and Lactobacillus as well as the ratio of Firmicutes to Bacteroidetes decreased, while the abundance of Bacteroidetes and Proteobacteria increased in T1DM and T2DM patients. However, the consequences were not always consistent due to other factors like types of diets and progression of diseases. In addition, animal studies have demonstrated that gut microbiota is one of the most important factors for diabetes initiation and development. Several natural products possessing prebiotic effects like fruits, vegetables, and medicinal plants, have been found to ameliorate T2DM by modulating gut microbiota composition and abundance, reducing the gut permeability, increasing the production of SCFAs, decreasing the level of LPS, and inhibiting the inflammation. Moreover, clinical trials have further confirmed that several natural products are effective in preventing and treating T2DM, with regulation of gut microbiota as one of the potential mechanisms. In the future, it is necessary to explore the effects of more natural products and their bioactive components on DM via gut microbiota-related mechanisms. In addition, more well-designed clinical trials on natural products and their various bioactive compounds should be carried out to verify their effects on T2DM and its complications by regulating gut microbiota. Furthermore, functional foods based on natural products can be researched and developed by targeting gut microbiota for the prevention and management of T2DM. On the other hand, present studies mainly focus on modulating the action of natural products and their bioactive components on gut microbiota for preventing and managing T2DM. However, seldom studies have been carried out about the metabolism of natural products and their bioactive components by gut microbiota, even though the metabolites could directly play a role in the prevention and management of DM or, in return, could modulate gut microbiota. Thus, metabolisms of natural products and their bioactive components by gut microbiota should be widely studied in the future for the prevention and management of DM.

Author Contributions

Conceptualization, B.-Y.L., R.-Y.G., and H.-B.L. Writing—original draft preparation, B.-Y.L., X.-Y.X., J.-M.M., A.S., and Q.-Q.M. Writing—review and editing, R.-Y.G., Q.-C.S., and H.-B.L. Supervision, R.-Y.G. and H.-B.L. Funding acquisition, R.-Y.G. and H.-B.L.

Funding

The Shanghai Basic and Key Program (No. 18JC1410800), Technology Innovation Program of Chinese Academy of Agricultural Sciences (ASTIP), the Shanghai Pujiang Talent Plan (No. 18PJ1404600), the Agri-X Interdisciplinary Fund of Shanghai Jiao Tong University (No. Agri-X2017004), and the Key Project of Guangdong Provincial Science and Technology Program (No. 2014B020205002) supported this study.

Acknowledgments

Thanks Shi-Yu Cao for her assistance.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Zimmet, P.; Alberti, K.G.; Magliano, D.J.; Bennett, P.H. Diabetes mellitus statistics on prevalence and mortality: Facts and fallacies. Nat. Rev. Endocrinol. 2016, 12, 616–622. [Google Scholar] [CrossRef] [PubMed]
  2. Chen, Y.; Wang, T.; Liu, X.; Shankar, R.R. Prevalence of type 1 and type 2 diabetes among US pediatric population in the marketscan multi-state database, 2002 to 2016. Pediatr. Diabetes 2019, 20, 523–529. [Google Scholar] [CrossRef] [PubMed]
  3. Rosenbauer, J.; Icks, A.; Giani, G. Incidence and prevalence of childhood type 1 diabetes mellitus in germany--model-based national estimates. J. Pediatr. Endocrinol. Metab. 2002, 15, 1497–1504. [Google Scholar] [CrossRef]
  4. Zheng, Y.; Ley, S.H.; Hu, F.B. Global aetiology and epidemiology of type 2 diabetes mellitus and its complications. Nat. Rev. Endocrinol. 2018, 14, 88–98. [Google Scholar] [CrossRef] [PubMed]
  5. King, G.L. The role of inflammatory cytokines in diabetes and its complications. J. Periodontol. 2008, 79, 1527–1534. [Google Scholar] [CrossRef] [PubMed]
  6. Peltier, A.; Goutman, S.A.; Callaghan, B.C. Painful diabetic neuropathy. BMJ 2014, 348, g1799. [Google Scholar] [CrossRef] [PubMed]
  7. Wang, P.; Fiaschi-Taesch, N.M.; Vasavada, R.C.; Scott, D.K.; Garcia-Ocana, A.; Stewart, A.F. Diabetes mellitus--advances and challenges in human β-cell proliferation. Nat. Rev. Endocrinol. 2015, 11, 201–212. [Google Scholar] [CrossRef] [PubMed]
  8. Kleinberger, J.W.; Maloney, K.A.; Pollin, T.I. The genetic architecture of diabetes in pregnancy: Implications for clinical practice. Am. J. Perinatol. 2016, 33, 1319–1326. [Google Scholar] [CrossRef] [PubMed]
  9. Vana, D.R.; Adapa, D.; Prasad, V.S.S.; Choudhury, A. Diabetes mellitus types: Key genetic determinants and risk assessment. Genet. Mol. Res. 2019, 18. [Google Scholar] [CrossRef]
  10. Hober, D.; Sauter, P. Pathogenesis of type 1 diabetes mellitus: Interplay between enterovirus and host. Nat. Rev. Endocrinol. 2010, 6, 279–289. [Google Scholar] [CrossRef]
  11. Roep, B.O.; Tree, T.I. Immune modulation in humans: Implications for type 1 diabetes mellitus. Nat. Rev. Endocrinol. 2014, 10, 229–242. [Google Scholar] [CrossRef] [PubMed]
  12. Van Belle, T.L.; Coppieters, K.T.; von Herrath, M.G. Type 1 diabetes: Etiology, immunology, and therapeutic strategies. Physiol. Rev. 2011, 91, 79–118. [Google Scholar] [CrossRef] [PubMed]
  13. DiMeglio, L.A.; Evans-Molina, C.; Oram, R.A. Type 1 diabetes. Lancet 2018, 391, 2449–2462. [Google Scholar] [CrossRef]
  14. Canfora, E.E.; Meex, R.C.R.; Venema, K.; Blaak, E.E. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat. Rev. Endocrinol. 2019, 15, 261–273. [Google Scholar] [CrossRef] [PubMed]
  15. Gross, B.; Pawlak, M.; Lefebvre, P.; Staels, B. PPARs in obesity-induced T2DM, dyslipidaemia and NAFLD. Nat. Rev. Endocrinol. 2017, 13, 36–49. [Google Scholar] [CrossRef] [PubMed]
  16. Kootte, R.S.; Vrieze, A.; Holleman, F.; Dallinga-Thie, G.M.; Zoetendal, E.G.; de Vos, W.M.; Groen, A.K.; Hoekstra, J.B.L.; Stroes, E.S.; Nieuwdorp, M. The therapeutic potential of manipulating gut microbiota in obesity and type 2 diabetes mellitus. Diabetes Obes. Metab. 2012, 14, 112–120. [Google Scholar] [CrossRef]
  17. Mao, Q.Q.; Xu, X.Y.; Cao, S.Y.; Gan, R.Y.; Corke, H.; Beta, T.; Li, H.B. Bioactive compounds and bioactivities of ginger (Zingiber officinale roscoe). Foods 2019, 8, 185. [Google Scholar] [CrossRef]
  18. Buchanan, T.A.; Xiang, A.H.; Page, K.A. Gestational diabetes mellitus: Risks and management during and after pregnancy. Nat. Rev. Endocrinol. 2012, 8, 639–649. [Google Scholar] [CrossRef]
  19. Coustan, D.R. Gestational diabetes mellitus. Clin. Chem. 2013, 59, 1310–1321. [Google Scholar] [CrossRef]
  20. Johns, E.C.; Denison, F.C.; Norman, J.E.; Reynolds, R.M. Gestational diabetes mellitus: Mechanisms, treatment, and complications. Trends Endocrinol. Metab. 2018, 29, 743–754. [Google Scholar] [CrossRef]
  21. Chakraborty, C.; Bandyopadhyay, S.; Doss, C.G.; Agoramoorthy, G. Exploring the genomic roadmap and molecular phylogenetics associated with mody cascades using computational biology. Cell Biochem. Biophys. 2015, 71, 1491–1502. [Google Scholar] [CrossRef] [PubMed]
  22. Malecki, M.T. Genetics of type 2 diabetes mellitus. Diabetes Res. Clin. Pract. 2005, 68 (Suppl. 1), S10–S21. [Google Scholar] [CrossRef]
  23. American Diabetes, A. Diagnosis and classification of diabetes mellitus. Diabetes Care 2010, 33 (Suppl. 1), S62–S69. [Google Scholar] [CrossRef]
  24. Tauschmann, M.; Hovorka, R. Technology in the management of type 1 diabetes mellitus - current status and future prospects. Nat. Rev. Endocrinol. 2018, 14, 464–475. [Google Scholar] [CrossRef] [PubMed]
  25. Meng, J.M.; Cao, S.Y.; Wei, X.L.; Gan, R.Y.; Wang, Y.F.; Cai, S.X.; Xu, X.Y.; Zhang, P.Z.; Li, H.B. Effects and mechanisms of tea for the prevention and management of diabetes mellitus and diabetic complications: An updated review. Antioxidants 2019, 8, 170. [Google Scholar] [CrossRef] [PubMed]
  26. Xu, L.; Li, Y.; Dai, Y.; Peng, J. Natural products for the treatment of type 2 diabetes mellitus: Pharmacology and mechanisms. Pharmacol. Res. 2018, 130, 451–465. [Google Scholar] [CrossRef]
  27. Prabhakar, P.K.; Doble, M. Mechanism of action of natural products used in the treatment of diabetes mellitus. Chin. J. Integr. Med. 2011, 17, 563–574. [Google Scholar] [CrossRef]
  28. Patel, S.S.; Udayabanu, M. Effect of natural products on diabetes associated neurological disorders. Rev. Neurosci. 2017, 28, 271–293. [Google Scholar] [CrossRef]
  29. Dragan, S.; Andrica, F.; Serban, M.C.; Timar, R. Polyphenols-rich natural products for treatment of diabetes. Curr. Med. Chem. 2015, 22, 14–22. [Google Scholar] [CrossRef]
  30. He, C.X.; Shan, Y.J.; Song, W. Targeting gut microbiota as a possible therapy for diabetes. Nutr. Res. 2015, 35, 361–367. [Google Scholar] [CrossRef]
  31. Hu, Y.J.; Wong, F.S.; Wen, L. Antibiotics, gut microbiota, environment in early life and type 1 diabetes. Pharmacol. Res. 2017, 119, 219–226. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Hu, Y.J.; Peng, J.; Li, F.Y.; Wong, F.S.; Wen, L. Evaluation of different mucosal microbiota leads to gut microbiota-based prediction of type 1 diabetes in NOD mice. Sci. Rep. 2018, 8, 15451. [Google Scholar] [CrossRef] [PubMed]
  33. Seganfredo, F.B.; Blume, C.A.; Moehlecke, M.; Giongo, A.; Casagrande, D.S.; Spolidoro, J.V.N.; Padoin, A.V.; Schaan, B.D.; Mottin, C.C. Weight-loss interventions and gut microbiota changes in overweight and obese patients: A systematic review. Obes. Rev. 2017, 18, 832–851. [Google Scholar] [CrossRef] [PubMed]
  34. Bouter, K.E.; van Raalte, D.H.; Groen, A.K.; Nieuwdorp, M. Role of the gut microbiome in the pathogenesis of obesity and obesity-related metabolic dysfunction. Gastroenterology 2017, 152, 1671–1678. [Google Scholar] [CrossRef] [PubMed]
  35. Sun, L.; Ma, L.; Ma, Y.; Zhang, F.; Zhao, C.; Nie, Y. Insights into the role of gut microbiota in obesity: Pathogenesis, mechanisms, and therapeutic perspectives. Protein Cell 2018, 9, 397–403. [Google Scholar] [CrossRef] [PubMed]
  36. Cao, S.Y.; Zhao, C.N.; Yu Xu, X.Y.; Tang, G.Y.; Corke, H.; Gan, R.Y.; Li, H.B. Dietary plants, gut microbiota and obesity: Effects and mechanisms. Trends Food Sci. Technol. 2019, 92, 194–204. [Google Scholar] [CrossRef]
  37. Zhang, Y.J.; Li, S.; Gan, R.Y.; Zhou, T.; Xu, D.P.; Li, H.B. Impacts of gut bacteria on human health and diseases. Int. J. Mol. Sci. 2015, 16, 7493–7519. [Google Scholar] [CrossRef] [PubMed]
  38. Meng, X.; Li, S.; Li, Y.; Gan, R.Y.; Li, H.B. Gut microbiota’s relationship with liver disease and role in hepatoprotection by dietary natural products and probiotics. Nutrients 2018, 10, 1457. [Google Scholar] [CrossRef]
  39. Hrncir, T.; Hrncirova, L.; Kverka, M.; Tlaskalova-Hogenova, H. The role of gut microbiota in intestinal and liver diseases. Lab. Anim. 2019, 53, 271–280. [Google Scholar] [CrossRef]
  40. Petrov, P.D.; Garcia-Mediavilla, M.V.; Guzman, C.; Porras, D.; Nistal, E.; Martinez-Florez, S.; Castell, J.V.; Gonzalez-Gallego, J.; Sanchez-Campos, S.; Jover, R. A network involving gut microbiota, circulating bile acids, and hepatic metabolism genes that protects against non-alcoholic fatty liver disease. Mol. Nutr. Food Res. 2019, 1900487. [Google Scholar] [CrossRef]
  41. Ezzaidi, N.; Zhang, X.; Coker, O.O.; Yu, J. New insights and therapeutic implication of gut microbiota in non-alcoholic fatty liver disease and its associated liver cancer. Cancer Lett. 2019, 459, 186–191. [Google Scholar] [CrossRef] [PubMed]
  42. Li, J.; Zhao, F.; Wang, Y.; Chen, J.; Tao, J.; Tian, G.; Wu, S.; Liu, W.; Cui, Q.; Geng, B.; et al. Gut microbiota dysbiosis contributes to the development of hypertension. Microbiome 2017, 5, 14. [Google Scholar] [CrossRef] [PubMed]
  43. Tang, W.H.; Kitai, T.; Hazen, S.L. Gut microbiota in cardiovascular health and disease. Circ. Res. 2017, 120, 1183–1196. [Google Scholar] [CrossRef] [PubMed]
  44. Zuo, K.; Li, J.; Li, K.; Hu, C.; Gao, Y.; Chen, M.; Hu, R.; Liu, Y.; Chi, H.; Wang, H.; et al. Disordered gut microbiota and alterations in metabolic patterns are associated with atrial fibrillation. GigaScience 2019, 8, giz058. [Google Scholar] [CrossRef] [PubMed]
  45. Tao, J.; Li, Y.; Li, S.; Li, H.-B. Plant foods for the prevention and management of colon cancer. J. Funct. Food. 2018, 42, 95–110. [Google Scholar] [CrossRef]
  46. Xu, X.Y.; Zhao, C.N.; Cao, S.Y.; Tang, G.Y.; Gan, R.Y.; Li, H.B. Effects and mechanisms of tea for the prevention and management of cancers: An updated review. Crit. Rev. Food. Sci. Nutr. 2019, 1–13. [Google Scholar] [CrossRef]
  47. Li, X.; Watanabe, K.; Kimura, I. Gut microbiota dysbiosis drives and implies novel therapeutic strategies for diabetes mellitus and related metabolic diseases. Front. Immunol. 2017, 8, 1882. [Google Scholar] [CrossRef] [PubMed]
  48. Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia muciniphila protects against atherosclerosis by preventing metabolic endotoxemia-induced inflammation in Apoe(−/−) mice. Circulation 2016, 133, 2434–2446. [Google Scholar] [CrossRef] [PubMed]
  49. Goldacre, R.R. Associations between birthweight, gestational age at birth and subsequent type 1 diabetes in children under 12: A retrospective cohort study in England, 1998–2012. Diabetologia 2018, 61, 616–625. [Google Scholar] [CrossRef]
  50. Hanninen, A.; Toivonen, R.; Poysti, S.; Belzer, C.; Plovier, H.; Ouwerkerk, J.P.; Emani, R.; Cani, P.D.; De Vos, W.M. Akkermansia muciniphila induces gut microbiota remodelling and controls islet autoimmunity in NOD mice. Gut 2018, 67, 1445–1453. [Google Scholar] [CrossRef]
  51. Moossavi, S.; Bishehsari, F. Microbes: Possible link between modern lifestyle transition and the rise of metabolic syndrome. Obes. Rev. 2019, 20, 407–419. [Google Scholar] [CrossRef] [PubMed]
  52. Cao, Y.; Yao, G.W.; Sheng, Y.Y.; Yang, L.; Wang, Z.X.; Yang, Z.; Zhuang, P.W.; Zhang, Y.J. Jinqi jiangtang tablet regulates gut microbiota and improve insulin sensitivity in type 2 diabetes mice. J. Diabetes Res. 2019, 2019, 1872134. [Google Scholar] [CrossRef] [PubMed]
  53. Han, H.; Li, Y.Y.; Fang, J.; Liu, G.; Yin, J.; Li, T.J.; Yin, Y.L. Gut microbiota and type 1 diabetes. Int. J. Mol. Sci. 2018, 19, 995. [Google Scholar] [CrossRef] [PubMed]
  54. Gulden, E.; Wong, F.S.; Wen, L. The gut microbiota and type 1 diabetes. Clin. Immunol. 2015, 159, 143–153. [Google Scholar] [CrossRef] [PubMed]
  55. Wang, G.; Li, X.F.; Zhao, J.X.; Zhang, H.; Chen, W. Lactobacillus casei CCFM419 attenuates type 2 diabetes via a gut microbiota dependent mechanism. Food Funct. 2017, 8, 3155–3164. [Google Scholar] [CrossRef] [PubMed]
  56. Everard, A.; Matamoros, S.; Geurts, L.; Delzenne, N.M.; Cani, P.D. Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice. Mbio 2014, 5, e01011-14. [Google Scholar] [CrossRef] [PubMed]
  57. Jia, L.L.; Li, D.Y.; Feng, N.H.; Shamoon, M.; Sun, Z.H.; Ding, L.; Zhang, H.; Chen, W.; Sun, J.; Chen, Y.Q. Anti-diabetic effects of Clostridium butyricum CGMCC0313.1 through promoting the growth of gut butyrate-producing bacteria in type 2 diabetic mice. Sci. Rep. 2017, 7, 7046. [Google Scholar] [CrossRef] [PubMed]
  58. Amar, J.; Chabo, C.; Waget, A.; Klopp, P.; Vachoux, C.; Bermudez-Humaran, L.G.; Smirnova, N.; Berge, M.; Sulpice, T.; Lahtinen, S.; et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: Molecular mechanisms and probiotic treatment. EMBO Mol. Med. 2011, 3, 559–572. [Google Scholar] [CrossRef]
  59. Peng, J.; Narasimhan, S.; Marchesi, J.R.; Benson, A.; Wong, F.S.; Wen, L. Long term effect of gut microbiota transfer on diabetes development. J. Autoimmun. 2014, 53, 85–94. [Google Scholar] [CrossRef] [Green Version]
  60. Plotkin, B.J.; Paulson, D.; Chelich, A.; Jurak, D.; Cole, J.; Kasimos, J.; Burdick, J.R.; Casteel, N. Immune responsiveness in a rat model for type ii diabetes (Zucker rat, fa/fa): Susceptibility to candida albicans infection and leucocyte function. J. Med. Microbiol. 1996, 44, 277–283. [Google Scholar] [CrossRef]
  61. Raffel, L.; Pitsakis, P.; Levison, S.P.; Levison, M.E. Experimental candida albicans, staphylococcus aureus, and streptococcus faecalis pyelonephritis in diabetic rats. Infect. Immun. 1981, 34, 773–779. [Google Scholar] [PubMed]
  62. Tang, G.Y.; Meng, X.; Li, Y.; Zhao, C.N.; Liu, Q.; Li, H.B. Effects of vegetables on cardiovascular diseases and related mechanisms. Nutrients 2017, 9, 857. [Google Scholar] [CrossRef]
  63. Zhao, C.N.; Meng, X.; Li, Y.; Li, S.; Liu, Q.; Tang, G.Y.; Li, H.B. Fruits for prevention and treatment of cardiovascular diseases. Nutrients 2017, 9, 598. [Google Scholar] [CrossRef] [PubMed]
  64. Meng, X.; Li, Y.; Li, S.; Gan, R.-Y.; Li, H.-B. Natural products for prevention and treatment of chemical-induced liver injuries. Compr. Rev. Food. Sci. Food Saf. 2018, 17, 472–495. [Google Scholar] [CrossRef]
  65. Shang, A.; Cao, S.Y.; Xu, X.Y.; Gan, R.Y.; Tang, G.Y.; Corke, H.; Mavumengwana, V.; Li, H.B. Bioactive compounds and biological functions of garlic (Allium sativum L.). Foods 2019, 8, 246. [Google Scholar] [CrossRef] [PubMed]
  66. Cao, S.Y.; Li, Y.; Meng, X.; Zhao, C.N.; Li, S.; Gan, R.Y.; Li, H.B. Dietary natural products and lung cancer: Effects and mechanisms of action. J. Funct. Food. 2019, 52, 316–331. [Google Scholar] [CrossRef]
  67. Liu, Q.; Meng, X.; Li, Y.; Zhao, C.N.; Tang, G.Y.; Li, S.; Gan, R.Y.; Li, H.-B. Natural products for the prevention and management of Helicobacter pylori infection. Compr. Rev. Food. Sci. Food Saf. 2018, 17, 937–952. [Google Scholar] [CrossRef]
  68. Li, Y.; Li, S.; Meng, X.; Gan, R.Y.; Zhang, J.J.; Li, H.B. Dietary natural products for prevention and treatment of breast cancer. Nutrients 2017, 9, 728. [Google Scholar] [CrossRef]
  69. Zhou, Y.; Li, Y.; Zhou, T.; Zheng, J.; Li, S.; Li, H.B. Dietary natural products for prevention and treatment of liver cancer. Nutrients 2016, 8, 156. [Google Scholar] [CrossRef]
  70. Gan, R.Y.; Li, H.B.; Sui, Z.Q.; Corke, H. Absorption, metabolism, anti-cancer effect and molecular targets of epigallocatechin gallate (EGCG): An updated review. Crit. Rev. Food. Sci. Nutr. 2018, 58, 924–941. [Google Scholar] [CrossRef]
  71. Xu, X.Y.; Meng, X.; Li, S.; Gan, R.Y.; Li, Y.; Li, H.B. Bioactivity, health benefits, and related molecular mechanisms of curcumin: Current progress, challenges, and perspectives. Nutrients 2018, 10, 1553. [Google Scholar] [CrossRef] [PubMed]
  72. Arora, T.; Backhed, F. The gut microbiota and metabolic disease: Current understanding and future perspectives. J. Intern. Med. 2016, 280, 339–349. [Google Scholar] [CrossRef] [PubMed]
  73. Munch, N.S.; Fang, H.Y.; Ingermann, J.; Maurer, H.C.; Anand, A.; Kellner, V.; Sahm, V.; Wiethaler, M.; Baumeister, T.; Wein, F.; et al. High-fat diet accelerates carcinogenesis in a mouse model of barrett’s esophagus via interleukin 8 and alterations to the gut microbiome. Gastroenterology 2019, 157, 492–495. [Google Scholar] [CrossRef] [PubMed]
  74. Tadokoro, Y.; Hoshii, T.; Yamazaki, S.; Eto, K.; Ema, H.; Kobayashi, M.; Ueno, M.; Ohta, K.; Arai, Y.; Hara, E.; et al. Spred1 safeguards hematopoietic homeostasis against diet-induced systemic stress. Cell stem cell 2018, 22, 713–718. [Google Scholar] [CrossRef] [PubMed]
  75. Liu, G.M.; Liang, L.; Yu, G.Y.; Li, Q.H. Pumpkin polysaccharide modifies the gut microbiota during alleviation of type 2 diabetes in rats. Int. J. Biol. Macromol. 2018, 115, 711–717. [Google Scholar] [CrossRef] [PubMed]
  76. Han, J.L.; Lin, H.L.; Huang, W.P. Modulating gut microbiota as an anti-diabetic mechanism of berberine. Med. Sci. Monit. 2011, 17, RA164–RA167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Han, L.H.; Li, T.G.; Du, M.; Chang, R.; Zhan, B.Y.; Mao, X.Y. Beneficial effects of Potentilla discolor Bunge water extract on inflammatory cytokines release and gut microbiota in high-fat diet and streptozotocin-induced type 2 diabetic mice. Nutrients 2019, 11, 670. [Google Scholar] [CrossRef]
  78. Zhang, B.W.; Sun, W.L.; Yu, N.; Sun, J.; Yu, X.X.; Li, X.; Xing, Y.; Yan, D.; Ding, Q.Z.; Xiu, Z.L.; et al. Anti-diabetic effect of baicalein is associated with the modulation of gut microbiota in streptozotocin and high-fat-diet induced diabetic rats. J. Funct. Food. 2018, 46, 256–267. [Google Scholar] [CrossRef]
  79. Song, J.X.; Ren, H.; Gao, Y.F.; Lee, C.Y.; Li, S.F.; Zhang, F.; Li, L.; Chen, H. Dietary capsaicin improves glucose homeostasis and alters the gut microbiota in obese diabetic ob/ob mice. Front. Physiol. 2017, 8, 602. [Google Scholar] [CrossRef]
  80. Hu, R.K.; Zeng, F.; Wu, L.X.; Wan, X.Z.; Chen, Y.F.; Zhang, J.C.; Liu, B. Fermented carrot juice attenuates type 2 diabetes by mediating gut microbiota in rats. Food Funct. 2019, 10, 2935–2946. [Google Scholar] [CrossRef]
  81. Alam, F.; Islam, M.A.; Kamal, M.A.; Gan, S.H. Updates on managing type 2 diabetes mellitus with natural products: Towards antidiabetic drug development. Curr. Med. Chem. 2018, 25, 5395–5431. [Google Scholar] [CrossRef] [PubMed]
  82. Buchholz, T.; Melzig, M.F. Polyphenolic compounds as pancreatic lipase inhibitors. Planta Med. 2015, 81, 771–783. [Google Scholar] [CrossRef]
  83. Bibbo, S.; Dore, M.P.; Pes, G.M.; Delitala, G.; Delitala, A.P. Is there a role for gut microbiota in type 1 diabetes pathogenesis? Ann. Med. 2017, 49, 11–22. [Google Scholar] [CrossRef] [PubMed]
  84. Sircana, A.; Framarin, L.; Leone, N.; Berrutti, M.; Castellino, F.; Parente, R.; De Michieli, F.; Paschetta, E.; Musso, G. Altered gut microbiota in type 2 diabetes: Just a coincidence? Curr. Diabetes Rep. 2018, 18, 98. [Google Scholar] [CrossRef] [PubMed]
  85. Vatanen, T.; Franzosa, E.A.; Schwager, R.; Tripathi, S.; Arthur, T.D.; Vehik, K.; Lernmark, A.; Hagopian, W.A.; Rewers, M.J.; She, J.X.; et al. The human gut microbiome in early-onset type 1 diabetes from the teddy study. Nature 2018, 562, 589–594. [Google Scholar] [CrossRef] [PubMed]
  86. Gulden, E.; Palm, N.; Herold, K.C. Mait cells: A link between gut integrity and type 1 diabetes. Cell. Metab. 2017, 26, 813–815. [Google Scholar] [CrossRef] [PubMed]
  87. Sanna, S.; van Zuydam, N.R.; Mahajan, A.; Kurilshikov, A.; Vich Vila, A.; Vosa, U.; Mujagic, Z.; Masclee, A.A.M.; Jonkers, D.; Oosting, M.; et al. Causal relationships among the gut microbiome, short-chain fatty acids and metabolic diseases. Nat. Genet. 2019, 51, 600–605. [Google Scholar] [CrossRef] [PubMed]
  88. Wen, L.; Ley, R.E.; Volchkov, P.Y.; Stranges, P.B.; Avanesyan, L.; Stonebraker, A.C.; Hu, C.; Wong, F.S.; Szot, G.L.; Bluestone, J.A.; et al. Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature 2008, 455, 1109–1113. [Google Scholar] [CrossRef] [PubMed]
  89. Wirth, R.; Bodi, N.; Maroti, G.; Bagyanszki, M.; Talapka, P.; Fekete, E.; Bagi, Z.; Kovacs, K.L. Regionally distinct alterations in the composition of the gut microbiota in rats with streptozotocin-induced diabetes. PLoS ONE 2014, 9, e110440. [Google Scholar] [CrossRef]
  90. Gulden, E.; Chao, C.; Tai, N.W.; Pearson, J.A.; Peng, J.; Majewska-Szczepanik, M.; Zhou, Z.G.; Wong, F.S.; Wen, L. TRIF deficiency protects non-obese diabetic mice from type 1 diabetes by modulating the gut microbiota and dendritic cells. J. Autoimmun. 2018, 93, 57–65. [Google Scholar] [CrossRef] [PubMed]
  91. Miani, M.; Le Naour, J.; Waeckel-Enee, E.; Verma, S.C.; Straube, M.; Emond, P.; Ryffel, B.; Van Endert, P.; Sokol, H.; Diana, J. Gut microbiota-stimulated innate lymphoid cells support beta-defensin 14 expression in pancreatic endocrine cells, preventing autoimmune diabetes. Cell Metab. 2018, 28, 557–562. [Google Scholar] [CrossRef] [PubMed]
  92. De Goffau, M.C.; Fuentes, S.; van den Bogert, B.; Honkanen, H.; de Vos, W.M.; Welling, G.W.; Hyoty, H.; Harmsen, H.J.M. Aberrant gut microbiota composition at the onset of type 1 diabetes in young children. Diabetologia 2014, 57, 1569–1577. [Google Scholar] [CrossRef] [PubMed]
  93. Salamon, D.; Sroka-Oleksiak, A.; Kapusta, P.; Szopa, M.; Mrozinska, S.; Ludwig-Slomczynska, A.H.; Wolkow, P.P.; Bulanda, M.; Klupa, T.; Malecki, M.T.; et al. Characteristics of gut microbiota in adult patients with type 1 and type 2 diabetes based on next-generation sequencing of the 16s rRNA gene fragment. Pol. Arch. Intern. Med. 2018, 128, 336–343. [Google Scholar] [CrossRef] [PubMed]
  94. Huang, Y.; Li, S.C.; Hu, J.; Ruan, H.B.; Guo, H.M.; Zhang, H.H.; Wang, X.; Pei, Y.F.; Pan, Y.; Fang, C. Gut microbiota profiling in Han Chinese with type 1 diabetes. Diabetes Res. Clin. Pract. 2018, 141, 256–263. [Google Scholar] [CrossRef] [PubMed]
  95. Murri, M.; Leiva, I.; Gomez-Zumaquero, J.M.; Tinahones, F.J.; Cardona, F.; Soriguer, F.; Queipo-Ortuno, M.I. Gut microbiota in children with type 1 diabetes differs from that in healthy children: A case-control study. BMC Med. 2013, 11, 46. [Google Scholar] [CrossRef] [PubMed]
  96. Soyucen, E.; Gulcan, A.; Aktuglu-Zeybek, A.C.; Onal, H.; Kiykim, E.; Aydin, A. Differences in the gut microbiota of healthy children and those with type 1 diabetes. Pediatr. Int. 2014, 56, 336–343. [Google Scholar] [CrossRef] [PubMed]
  97. Geurts, L.; Lazarevic, V.; Derrien, M.; Everard, A.; Van Roye, M.; Knauf, C.; Valet, P.; Girard, M.; Muccioli, G.G.; Francois, P.; et al. Altered gut microbiota and endocannabinoid system tone in obese and diabetic leptin-resistant mice: Impact on apelin regulation in adipose tissue. Front. Microbiol. 2011, 2, 149. [Google Scholar] [CrossRef]
  98. Ohtsu, A.; Takeuchi, Y.; Katagiri, S.; Suda, W.; Maekawa, S.; Shiba, T.; Komazaki, R.; Udagawa, S.; Sasaki, N.; Hattori, M.; et al. Influence of Porphyromonas gingivalis in gut microbiota of streptozotocin-induced diabetic mice. Oral Dis. 2019, 25, 868–880. [Google Scholar] [CrossRef]
  99. Kieler, I.N.; Osto, M.; Hugentobler, L.; Puetz, L.; Gilbert, M.T.P.; Hansen, T.; Pedersen, O.; Reusch, C.E.; Zini, E.; Lutz, T.A.; et al. Diabetic cats have decreased gut microbial diversity and a lack of butyrate producing bacteria. Sci. Rep. 2019, 9, 4822. [Google Scholar] [CrossRef]
  100. Kameyama, K.; Itoii, K. Intestinal colonization by a Lachnospiraceae bacterium contributes to the development of diabetes in obese mice. Microbes Environ. 2014, 29, 427–430. [Google Scholar] [CrossRef]
  101. Larsen, N.; Vogensen, F.K.; van den Berg, F.W.J.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Abu Al-Soud, W.; Sorensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from NOD-diabetic adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef] [PubMed]
  102. Navab-Moghadam, F.; Sedighi, M.; Khamseh, M.E.; Alaei-Shahmiri, F.; Talebi, M.; Razavi, S.; Amirmozafari, N. The association of type II diabetes with gut microbiota composition. Microb. Pathog. 2017, 110, 630–636. [Google Scholar] [CrossRef] [PubMed]
  103. Inoue, R.; Ohue-Kitano, R.; Tsukahara, T.; Tanaka, M.; Masuda, S.; Inoue, T.; Yamakage, H.; Kusakabe, T.; Hasegawa, K.; Shimatsu, A.; et al. Prediction of functional profiles of gut microbiota from 165 rRNA metagenomic data provides a more robust evaluation of gut dysbiosis occurring in Japanese type 2 diabetic patients. J. Clin. Biochem. Nutr. 2017, 61, 217–221. [Google Scholar] [CrossRef] [PubMed]
  104. Suceveanu, A.I.; Stoian, A.P.; Parepa, I.; Voinea, C.; Hainarosie, R.; Manuc, D.; Nitipir, C.; Mazilu, L.; Suceveanu, A.P. Gut microbiota patterns in obese and type 2 diabetes (T2D) patients from Romanian Black Sea Coast Region. Rev. Chim. 2018, 69, 2260–2267. [Google Scholar]
  105. Chen, C.; You, L.J.; Huang, Q.; Fu, X.; Zhang, B.; Liu, R.H.; Li, C. Modulation of gut microbiota by mulberry fruit polysaccharide treatment of obese diabetic db/db mice. Food Funct. 2018, 9, 3732–3742. [Google Scholar] [CrossRef] [PubMed]
  106. Chen, K.; Chen, H.; Faas, M.M.; de Haan, B.J.; Li, J.H.; Xiao, P.; Zhang, H.; Diana, J.; de Vos, P.; Sun, J. Specific inulin-type fructan fibers protect against autoimmune diabetes by modulating gut immunity, barrier function, and microbiota homeostasis. Mol. Nutr. Food Res. 2017, 61, 1601006. [Google Scholar] [CrossRef] [PubMed]
  107. Yan, X.; Yang, C.F.; Lin, G.P.; Chen, Y.Q.; Miao, S.; Liu, B.; Zhao, C. Antidiabetic potential of green seaweed Enteromorpha prolifera flavonoids regulating insulin signaling pathway and gut microbiota in type 2 diabetic mice. J. Food Sci. 2019, 84, 165–173. [Google Scholar] [CrossRef] [PubMed]
  108. Zhang, Q.; Yu, H.Y.; Xiao, X.H.; Hu, L.; Xin, F.J.; Yu, X.B. Inulin-type fructan improves diabetic phenotype and gut microbiota profiles in rats. PeerJ 2018, 6, e4446. [Google Scholar] [CrossRef]
  109. Gao, H.; Wen, J.J.; Hu, J.L.; Nie, Q.X.; Chen, H.H.; Xiong, T.; Nie, S.P.; Xie, M.Y. Fermented Momordica charantia L. Juice modulates hyperglycemia, lipid profile, and gut microbiota in type 2 diabetic rats. Food Res. Int. 2019, 121, 367–378. [Google Scholar] [CrossRef]
  110. Mei, X.R.; Zhang, X.Y.; Wang, Z.G.; Gao, Z.Y.; Liu, G.; Hu, H.L.; Zou, L.; Li, X.L. Insulin sensitivity-enhancing activity of phlorizin is associated with lipopolysaccharide decrease and gut microbiota changes in obese and type 2 diabetes (db/db) mice. J. Agric. Food Chem. 2016, 64, 7502–7511. [Google Scholar] [CrossRef]
  111. Van Hul, M.; Geurts, L.; Plovier, H.; Druart, C.; Everard, A.; Stahlman, M.; Rhimi, M.; Chira, K.; Teissedre, P.L.; Delzenne, N.M.; et al. Reduced obesity, diabetes, and steatosis upon cinnamon and grape pomace are associated with changes in gut microbiota and markers of gut barrier. Am. J. Physiol. Endocrinol. Metab. 2018, 314, E334–E352. [Google Scholar] [CrossRef] [PubMed]
  112. Li, K.; Zhang, L.; Xue, J.; Yang, X.L.; Dong, X.Y.; Sha, L.P.; Lei, H.; Zhang, X.X.; Zhu, L.L.; Wang, Z.; et al. Dietary inulin alleviates diverse stages of type 2 diabetes mellitus via anti-inflammation and modulating gut microbiota in db/db mice. Food Funct. 2019, 10, 1915–1927. [Google Scholar] [CrossRef] [PubMed]
  113. Zhang, W.Y.; Zhang, H.H.; Yu, C.H.; Fang, J.; Ying, H.Z. Ethanol extract of Atractylodis macrocephalae rhizoma ameliorates insulin resistance and gut microbiota in type 2 diabetic db/db mice. J. Funct. Food. 2017, 39, 139–151. [Google Scholar] [CrossRef]
  114. Xie, Y.Q.; Xiao, M.; Ni, Y.L.; Jiang, S.F.; Feng, G.Z.; Sang, S.G.; Du, G.K. Alpinia oxyphylla Miq. extract prevents diabetes in mice by modulating gut microbiota. J. Diabetes Res. 2018, 2018, 4230590. [Google Scholar] [CrossRef] [PubMed]
  115. Yan, H.L.; Lu, J.M.; Wang, Y.F.; Gu, W.; Yang, X.X.; Yu, J. Intake of total saponins and polysaccharides from Polygonatum kingianum affects the gut microbiota in diabetic rats. Phytomedicine 2017, 26, 45–54. [Google Scholar] [CrossRef] [PubMed]
  116. Singh, D.K.; Winocour, P.; Farrington, K. Oxidative stress in early diabetic nephropathy: Fueling the fire. Nat. Rev. Endocrinol. 2011, 7, 176–184. [Google Scholar] [CrossRef] [PubMed]
  117. Thorve, V.S.; Kshirsagar, A.D.; Vyawahare, N.S.; Joshi, V.S.; Ingale, K.G.; Mohite, R.J. Diabetes-induced erectile dysfunction: Epidemiology, pathophysiology and management. J. Diabetes Complicat. 2011, 25, 129–136. [Google Scholar] [CrossRef]
  118. Li, H.; Qi, T.; Huang, Z.S.; Ying, Y.; Zhang, Y.; Wang, B.; Ye, L.; Zhang, B.; Chen, D.L.; Chen, J. Relationship between gut microbiota and type 2 diabetic erectile dysfunction in Sprague-Dawley rats. J. Huazhong Univ. Sci. Tech. Med. Sci. 2017, 37, 523–530. [Google Scholar] [CrossRef]
  119. Li, X.W.; Chen, H.P.; He, Y.Y.; Chen, W.L.; Chen, J.W.; Gao, L.; Hu, H.Y.; Wang, J. Effects of rich-polyphenols extract of Dendrobium loddigesii on anti-diabetic, anti-inflammatory, anti-oxidant, and gut microbiota modulation in db/db mice. Molecules 2018, 23, 3245. [Google Scholar] [CrossRef]
  120. Roshanravan, N.; Mahdavi, R.; Jafarabadi, M.A.; Alizadeh, E.; Ghavami, A.; Saadat, Y.R.; Alamdari, N.M.; Dastouri, M.R.; Alipour, S.; Ostadrahimi, A. The effects of sodium butyrate and high-performance inulin supplementation on the promotion of gut bacterium Akkermansia muciniphila growth and alterations in miR-375 and KLF5 expression in type 2 diabetic patients: A randomized, double-blind, placebo-controlled trial. Eur. J. Integr. Med. 2018, 18, 1–7. [Google Scholar]
  121. Xu, J.; Lian, F.M.; Zhao, L.H.; Zhao, Y.F.; Chen, X.Y.; Zhang, X.; Guo, Y.; Zhang, C.H.; Zhou, Q.; Xue, Z.S.; et al. Structural modulation of gut microbiota during alleviation of type 2 diabetes with a chinese herbal formula. ISME J. 2015, 9, 552–562. [Google Scholar] [CrossRef] [PubMed]
  122. Candela, M.; Biagi, E.; Soverini, M.; Consolandi, C.; Quercia, S.; Severgnini, M.; Peano, C.; Turroni, S.; Rampelli, S.; Pozzilli, P.; et al. Modulation of gut microbiota dysbioses in type 2 diabetic patients by macrobiotic Ma-Pi 2 diet. Br. J. Nutr. 2016, 116, 80–93. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Tong, X.L.; Xu, J.; Lian, F.M.; Yu, X.T.; Zhao, Y.F.; Xu, L.P.; Zhang, M.H.; Zhao, X.Y.; Shen, J.; Wu, S.P.; 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, e02392-17. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The association among gut microbiota, T2DM, and its complications. The changes of gut microbiota caused the increase in LPS, which could cause inflammation and insulin resistance. It indicates that gut microbiota would play an important role in the initiation and development of T2DM and its complications. Abbreviations: LPS, lipopolysaccharides; T2DM, type 2 diabetes mellitus.
Figure 1. The association among gut microbiota, T2DM, and its complications. The changes of gut microbiota caused the increase in LPS, which could cause inflammation and insulin resistance. It indicates that gut microbiota would play an important role in the initiation and development of T2DM and its complications. Abbreviations: LPS, lipopolysaccharides; T2DM, type 2 diabetes mellitus.
Foods 08 00440 g001
Figure 2. The relationship among natural products, gut microbiota, T2DM, and its complications. Some natural products and their bioactive components alleviate T2DM by changing the composition and abundance of gut microbiota. It decreases the permeability of gut and the level of LPS, increases the production of SCFAs, and inhibits the inflammation. The arrows out of the box indicates the direction of action, and the up arrows in the box mean the upregulation or activation, and the down arrows in the box mean the downregulation or inhibition. Abbreviations: LPS, lipopolysaccharides. SCFAs, short-chain fatty acids. T2DM, type 2 diabetes mellitus. GPR41/43, G-protein-coupled receptor41/43. TLR4, toll-like receptor-4. GLP-1, glucagon-like peptide-1. PYY, peptide YY. MyD88, myeloid differentiation primary response gene 88. PI3K, phosphatidylinositol-3-kinase. NF-κB, nuclear factor-kappa B. IL-6, interleukin 6. IL-8, interleukin 8. TNF-α, tumor necrosis factor-α.
Figure 2. The relationship among natural products, gut microbiota, T2DM, and its complications. Some natural products and their bioactive components alleviate T2DM by changing the composition and abundance of gut microbiota. It decreases the permeability of gut and the level of LPS, increases the production of SCFAs, and inhibits the inflammation. The arrows out of the box indicates the direction of action, and the up arrows in the box mean the upregulation or activation, and the down arrows in the box mean the downregulation or inhibition. Abbreviations: LPS, lipopolysaccharides. SCFAs, short-chain fatty acids. T2DM, type 2 diabetes mellitus. GPR41/43, G-protein-coupled receptor41/43. TLR4, toll-like receptor-4. GLP-1, glucagon-like peptide-1. PYY, peptide YY. MyD88, myeloid differentiation primary response gene 88. PI3K, phosphatidylinositol-3-kinase. NF-κB, nuclear factor-kappa B. IL-6, interleukin 6. IL-8, interleukin 8. TNF-α, tumor necrosis factor-α.
Foods 08 00440 g002
Table 1. The epidemiological studies on the association between gut microbiota and T2DM.
Table 1. The epidemiological studies on the association between gut microbiota and T2DM.
Study TypesParticipantsAlterations in Gut Microbiota CompositionReference
Case-control studyAdults with T2DM (n = 18)
Healthy male adults (n = 18)
↓ The proportions of phylum Firmicutes and class Clostridia, and the ratios of Firmicutes to Bacteroidetes
↑ The proportion of betaproteobacteria
[101]
Case-control studyPatients with T2DM (n = 18)
Healthy individuals (n = 18)
↓ The concentration of Faecalibacterium prausnitzii[102]
Case-control studyPatients with T2DM (n = 10)
Healthy individuals (n = 12)
↓ The abundance of genus Blautia[103]
Case-control studyPatients with T2DM (n = 100)
Healthy individuals (n = 100)
↓ The counts of Lactobacillus sp. (L. acidophilus, L. plantarum, and L. reuteri) and Bifidobacterium[104]
Abbreviations: T2DM, Type 2 diabetes mellitus. ↓, Decrease. ↑, Increase.
Table 2. The animal studies of natural products on DM and its complications by modulating gut microbiota.
Table 2. The animal studies of natural products on DM and its complications by modulating gut microbiota.
Natural ProductsDiseaseStudy TypesModelsEffectsMechanismsReference
Long-chain inulin-type fructans fibers T1DMIn vivoNOD diabetic micePromoting modulatory T-cell responses.
Delaying the development of T1DM.
↑ The ratio of Firmicutes to Bacteroidetes
↑ The abundance of Ruminococcaceae and Lactobacilli
↑ The production of SCFAs
↑ The expression of tight junction proteins occludin and claudin-2
[106]
Capsaicin T2DMIn vivoObese T2DM ob/ob miceInhibiting the levels of FBG and insulin.
Improving the glucose homeostasis and insulin tolerance.
↑ The ratio of Firmicutes to Bacteroidetes
↑ The abundance of Roseburia
↑ The production of butyrate
↓ The quantities of Bacteroides and Parabacteroides
↓ The levels of TNF-α and IL-6
[79]
Pumpkin polysaccharideT2DMIn vivoT2DM ratsImproving insulin tolerance.
Decreasing the levels of glucose, TC, LDL-C.
Increasing the level of HDL-C.
↑ The quantities of some bacteria, such as Bacteroidetes, Prevotella, Deltaproteobacteria, Oscillospira, Veillonellaceae, Phascolarctobacterium, Sutterella, and Bilophila
↑ The production of SCFAs
[75]
Fermented carrot juiceT2DMIn vivoT2DM ratsRegulating the levels of blood glucose and insulin as well as the morphology of the pancreas and kidney.↑ The quantities of Christensenellaceae, Oscillibacter, Ruminococcaceae, Lachnospiraceae, and Akkermansia
↑ The level of SCFAs
[80]
Fermented Momordica charantia juiceT2DMIn vivoT2DM rats induced by HFD and STZRelieving the hyperglycemia, hyperinsulinemia, and hyperlipidemia.↑ The abundance of Bacteroides caecigallinarwn, Bacteroides thetaiotaomicron, Prevotella loescheii, Prevotella oralis, and Prevotella melaninogenica
↑ The concentrations of acetic acid, propionic acid, and butyric acid
[109]
Phlorizin T2DMIn vivoT2DM db/db miceReducing insulin resistance.
Regulating the level of blood glucose.
↑ The abundance of Akkermansia muciniphila and Prevotella
↑ The gut microbial diversity
↑ The production of butyric acid
↓ The level of LPS
[110]
Mulberry fruit polysaccharideT2DMIn vivoT2DM db/db miceImproving glucose tolerance. Increasing the level of HDL-C.
Decreasing the levels of TC TG, LDL-C, and FFA.
Inhibiting body weight gain.
↑ The abundance of Bacteroidales, Lactobacillus, Allobaculum, Bacteroides, and Akkermansia
↓ The gut microbial diversity
[105]
Cinnamon bark and grape pomace extractsT2DMIn vivoT2DM C57BL/6J mice induced by HFDImproving glucose tolerance and insulin resistance. Decreasing fat mass gain and adipose tissue inflammation.↑ The abundances of Allobaculum and Roseburia,
↑ The expression of tight junction proteins.
↓ The abundance of genus Peptococcus, Desulfovibrio, and Lactococcus
[111]
InulinT2DMIn vivoMiceDecreasing the levels of FBG and glycated hemoglobin, and body weight.↑ The relative abundance of Cyanobacteria and Bacteroides
↓ The relative abundance of Deferribacteres and Tenericutes
[112]
Inulin-type fructanT2DMIn vivoT2DM rats induced by HFD/STZReducing the levels of FBG, IL-6, and alleviated glucose intolerance.↑ The abundance of Lactobacillus and SCFAs-producing bacteria, such as Lachnospiraceae, Phascolarctobacterium, and Bacteroides
↑ The level of GLP-1
[108]
Atractylodis macrocephalae Rhizoma ethanol extractT2DMIn vivoT2DM db/db miceDecreasing the levels of blood glucose, TG, TC, endotoxin, and IL-10↑ The abundance of Bacteroides thetaiotaomicron and Methanobrevibacter smithii[113]
Alpinia oxyphylla Miq. extractT2DMIn vivoT2DM db/db miceImproving glycemic control and renal function↑ The ratio of Bacteroidetes to Firmicutes and ↑ The abundance of Akkermansia
↓ The abundance of Helicobacter
[114]
Potentilla discolor Bunge water extractT2DMIn vivoT2DM C57BL/6J mice induced by HFD and STZDecreasing the level of pro-inflammatory cytokines. Improving inflammation.↑ The concentrations of fecal acetic acid butyric acid and their specific receptors including the expression of GPR41 and GPR43
↓ The ratio of Firmicutes to Bacteroidetes and ↓ The abundance of Proteobacteria
↓ The expression of gut mucosal tight junction proteins (Claudin3, ZO-1, and Occludin), TLR4, MyD88, and NF-κB
[77]
Polygonatum kingianuT2DMIn vivoT2DM SD rats induced by HFD and STZIncreasing the level of fasting insulin.
Preventing the increase of FBG.
Improving the intestinal microecology.
↑ The abundance of Firmicutes
↓The abundances of Bacteroidetes and Proteobacteria
[115]
BaicaleinT2DMIn vivoDiabetic rats induced by HFD, HSD, and STZDecreasing the level of blood glucose.
Improved inflammation and lipid metabolism.
↑ The relative abundances of Bacteroides and Bacteroidales S24-7
↑ The production of SCFAs
↑ The thickness of the gut mucus layer
[78]
Dendrobium loddigesii polyphenols extractDiabetic nephropathyIn vivoDiabetic nephropathy db/db miceImproving diabetic nephropathy.
Decreasing blood glucose level.
Increasing the level of insulin.
↑ The relative abundance of Prevotella/Akkermansia
↓ The relative abundance of S24-7/Rikenella/Escherichia coli
[119]
Green seaweed Enteromorpha prolifera flavonoidsT2DMIn vivoT2DM mice induced by HFD and HSD and STZDecreasing the level of FBG. Improving glucose tolerance. Reducing inflammation. Preventing liver and kidney injury.↑ The abundances of Lachnospiraceae and Alisties
↑ The IRS1/PI3K/AKT pathway
↓ The JNK1/2 insulin pathway in liver
[107]
Abbreviations: DM, diabetes mellitus. T1DM, type 1 diabetes mellitus. T2DM, type 2 diabetes mellitus. NOD mice, non-obese diabetic mice. HFD, high-fat diet. HSD, high sucrose diet. STZ, streptozotocin. LPS, lipopolysaccharides. FBG, fasting blood glucose. GLP-1, glucagon-like peptide-1. TG, triglyceride. HOMA-IR, homeostasis model assessment of insulin resistance. TC, total cholesterol. FFA, free fatty acid. SCFAs, short-chain fatty acids. IL-6, interleukin 6.IL-10, interleukin 10. TLR4, toll-like receptor-4. TNF-α, tumor necrosis factor-α. MyD88, myeloid differentiation primary response gene 88. HDL-C, high-density lipoprotein cholesterol. LDL-C, low-density lipoprotein cholesterol. NF-κB, nuclear factor-kappa B. GPR41/43, G-protein-coupled receptor 41/43. IRS1, insulin receptor substrate 1. PI3K, phosphatidylinositol-3-kinase. AKT, protein kinase B. JNK, Jun N-terminal kinase.
Table 3. The clinical trials of natural products on T2DM by modulating gut microbiota.
Table 3. The clinical trials of natural products on T2DM by modulating gut microbiota.
Natural ProductsStudy TypesParticipantsDose DurationResultsReference
InulinRCTT2DM patients
(n = 60)
10 g/dayN/A↑ The proportion of Akkermansia muciniphila[120]
Macrobiotic Ma-Pi 2 diet enriched fiberRCTObese T2DM patients
(n = 56)
N/AN/A↑ Gut microbiota ecosystem diversity
↑ SCFA-producing bacteria, such as Faecalibacterium, Roseburia, Lachnospira, Bacteroides and Akkermansia
↓ Pro-inflammatory bacteria, such as Collinsella and Streptococcus
[122]
Chinese herbal formulaRCTT2DM patients
(n = 100)
N/A12 weeks↑ The abundance of Faecalibacterium spp
↓ Hyperglycemia and hyperlipidemia
[123]
Ge gen Qin lian decoctionRCTT2DM patients
(n = 187)
N/A12 weeks↑ The number of Faecalibacterium prausnitzii
↓ The mean changes of FBG and HbA1c levels
[121]
Abbreviations: N/A, not available. RCT, randomized control clinical trial. FBG, fasting blood glucose. T2DM, type 2 diabetes mellitus. HbA1c, hemoglobin A1c. SCFA, short-chain fatty acids.

Share and Cite

MDPI and ACS Style

Li, B.-Y.; Xu, X.-Y.; Gan, R.-Y.; Sun, Q.-C.; Meng, J.-M.; Shang, A.; Mao, Q.-Q.; Li, H.-B. Targeting Gut Microbiota for the Prevention and Management of Diabetes Mellitus by Dietary Natural Products. Foods 2019, 8, 440. https://doi.org/10.3390/foods8100440

AMA Style

Li B-Y, Xu X-Y, Gan R-Y, Sun Q-C, Meng J-M, Shang A, Mao Q-Q, Li H-B. Targeting Gut Microbiota for the Prevention and Management of Diabetes Mellitus by Dietary Natural Products. Foods. 2019; 8(10):440. https://doi.org/10.3390/foods8100440

Chicago/Turabian Style

Li, Bang-Yan, Xiao-Yu Xu, Ren-You Gan, Quan-Cai Sun, Jin-Ming Meng, Ao Shang, Qian-Qian Mao, and Hua-Bin Li. 2019. "Targeting Gut Microbiota for the Prevention and Management of Diabetes Mellitus by Dietary Natural Products" Foods 8, no. 10: 440. https://doi.org/10.3390/foods8100440

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop