Next Article in Journal
Dietary and Behavioral Interventions for Diabetes: Current Evidence, Challenges, and Opportunities
Previous Article in Journal
Sweetness Ratings of U.S. Infant Formulas
Previous Article in Special Issue
Microencapsulated Propionate and Butyrate Improved Energy Balance and Gut Microbiota Composition in Diet-Induced Obese Rats
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota and Metabolites: Biomarkers and Therapeutic Targets for Diabetes Mellitus and Its Complications

1
Special Key Laboratory of Ocular Diseases of Guizhou Province, Department of Immunology, Zunyi Medical University, Zunyi 563000, China
2
Special Key Laboratory of Gene Detection and Therapy of Guizhou Province, School of Basic Medical Sciences, Zunyi Medical University, Zunyi 563000, China
*
Author to whom correspondence should be addressed.
Nutrients 2025, 17(16), 2603; https://doi.org/10.3390/nu17162603
Submission received: 4 July 2025 / Revised: 7 August 2025 / Accepted: 7 August 2025 / Published: 11 August 2025

Abstract

Diabetes mellitus (DM) is a complex metabolic disease characterized by significantly elevated blood glucose levels as a result of dysfunctional or impaired pancreatic β-cells, leading to insulin deficiency. This condition can result in severe complications, including cardiovascular diseases, kidney failure, vision impairment, and nerve damage. Currently available anti-diabetic drugs do not fully prevent the progression of these complications. Moreover, they often have significant side effects. The gut microbiota plays a crucial role in influencing diet, energy metabolism, and blood glucose levels. Research shows a strong link between microbiota dysbiosis and DM, as well as the severity of its complications. Commensal bacteria can help manage blood glucose levels, reduce inflammation, regulate metabolism, and enhance the gut barrier. Conversely, opportunistic pathogens can worsen insulin resistance, promote metabolic disorders, disrupt gut integrity, and affect appetite and weight. This article describes the characteristics of gut microbiota in various types of DM and explores the role of the “gut microbiota–metabolite–signaling pathway” axis in DM and its complications. In addition, it highlights the therapeutic potential of traditional Chinese medicine and dietary interventions through modulation of the gut microbiota and metabolites. The aim is to provide comprehensive evidence supporting the integration of TCM dietary therapy, targeted dietary strategies, and specific probiotics as alternative and complementary therapies for DM and its complications.

1. Introduction

Studies show that between 1990 and 2019, the global incidence rate of diabetes mellitus (DM) was on the rise year by year [1]. The incidence of type 2 diabetes mellitus (T2DM) in adolescents nearly tripled compared to pre-pandemic levels during COVID-19 [2], and the adult T2DM incidence rate also shows the same trend [3]. This may be related to virus-induced pancreatic beta-cell damage through the ACE2 receptor [4]. DM mainly includes type 1 diabetes mellitus (T1DM), T2DM, and gestational diabetes mellitus (GDM). T1DM is primarily caused by the immune system mistakenly attacking pancreatic beta cells (β-cells), leading to insufficient insulin secretion. This process may be triggered by factors such as viral infections, dietary components, and chemical toxins. T2DM is characterized by insulin resistance (IR), and during pregnancy, hormones secreted by the placenta, such as placental lactogen, growth hormone, estrogen, and progesterone, increase IR. When the body’s insulin cannot lower blood glucose to the required level, it will lead to the occurrence of GDM [5]. Continuous elevation of blood glucose levels in the body leads to protein glycation and tissue damage, resulting in various complications. Diabetic retinopathy (DR) and diabetic kidney disease (DKD) are mainly caused by long-term hyperglycemia leading to microvascular damage. Diabetes neuropathy (DN) is a neuropathy caused by hyperglycemia, while diabetic cardiovascular disease (DCD) is the result of hyperglycemia, hypertension, and hyperlipidemia [6]. The management of DM primarily involves pharmacological treatment, lifestyle changes, and nutritional interventions [7]. Pharmacological treatment includes oral hypoglycemic agents and insulin injections, which help control blood glucose by reducing hepatic glucose production and increasing insulin sensitivity [8]. Lifestyle changes focus on maintaining a healthy weight and engaging in moderate-intensity physical activities such as walking, cycling, and swimming. Nutritional intervention emphasizes a balanced diet such as a low-sugar, low-fat, and high-fiber diet (HFD), as well as fat-soluble active components like prebiotics, probiotics, and coenzyme Q10, enabling patients to self-manage blood glucose levels [9].
Gut microbiota is closely related to DM; glycated hemoglobin (HbA1c) levels are correlated with Prevotella, Clostridia, and Ruminococcaceae, and show a positive correlation with Dorea, Bacteroidetes, Lactobacillus, and Bifidobacteria. Further research has shown that Rikenellaceae and Enterobacteriaceae are positively correlated with hyperglycemia development, while Synechococcus sp., Bifidobacterium adolescentis, and Chlorobium phaeovibrioides are negatively correlated with IR [10]. Commensal bacteria support gut metabolism, while an increase in pathogenic bacteria can lead to dysfunction of the gut barrier, thereby affecting digestion and absorption [11]. Clostridium increases intestinal permeability by disrupting tight junction proteins in the gut, allowing lipopolysaccharides (LPSs) to enter the bloodstream and exert pro-inflammatory effects under conditions of high intestinal permeability [12]. Escherichia coli, Klebsiella, and Desulfovibrio can promote the production of LPSs, attacking pancreatic beta cells [13]. Streptococcus releases peptidoglycan and lipoteichoic acid, activating the NLRP3 inflammasome and enhancing the inflammatory response [14]. Prevotella copri (P. copri) is enriched in pregnant women with GDM and can elevate branched-chain amino acid (BCAA) levels, increasing the risk of developing GDM [15]. Despite numerous studies indicating that the gut microbiota is involved in the occurrence and development of DM, the specific mechanisms by which the microbiota affects different types of DM and its complications remain unclear. Existing research has confirmed that the gut microbiota plays a dual role in the production and regulation of metabolites such as short-chain fatty acids (SCFAs), BCAAs, lipopolysaccharides (LPSs), and indolepropionic acid (IPA). Dysregulation of metabolite expression can further exacerbate metabolic disorders. For example, low levels of SCFAs and high levels of BCAAs and LPSs may impair insulin signaling, leading to β-cell dysfunction, reduced glucose responsiveness, and insufficient insulin secretion, thus resulting in hyperglycemia (Figure 1). However, current research mainly focuses on a single type of DM, lacking comprehensive cross-type analysis.
Therefore, this review aims to extract experimental data on differences in gut microbiota abundance from the published literature, and to elucidate the unique characteristics of the gut microbiota in patients with different types of DM and its complications through a narrative review, and it attempts to construct the interaction relationships among gut microbiota, metabolites, and disease. Further evaluation of the potential effects of dietary adjustments, traditional Chinese medicine (TCM) treatments, and probiotics is conducted to provide new insights and therapeutic strategies for DM and its complications.

2. Characteristics of Gut Microbiota in DM and Its Complications

2.1. Gut Microbiota in T1DM

Gut microbiota plays a regulatory role through different abundances, and there is a significant correlation between the abundance of different intestinal microbiota and the incidence rate of T1DM. The Veillonellaceae is associated with a decreased susceptibility to T1DM, while the Eubacterium coprostanoligenes group significantly impacts T1DM complications. The external validation phase confirms that Veillonellaceae can reduce susceptibility to T1DM [16]. Mendelian randomization (MR) studies using the inverse-variance weighted method found that the Saccharomyces and Bacteroides have a causal relationship with T1DM [17]. The composition and abundance of gut microbiota vary with age in T1DM patients, exhibiting different disease characteristics. In adults with T1DM, the gut microorganisms Blautia, Roseburia, and Faecalibacterium that produce SCFAs are significantly reduced [18], and there is lower abundance of Veillonella [19]. Conversely, there is an increase in Anaerobic Clostridia, Desulfovibrio, Ruminococcus, Bacteroides, and Lactobacillus johnsonii [20]. Among middle-aged and elderly individuals, the relative abundance of Bacteroidetes, Akkermansia, and Faecalibacterium remains stable [21,22]. Children with T1DM exhibit unique microbial profiles, with increased levels of Cyanobacteria, Fusarium, Bacteroides, and the Eubacterium hallii group in the gut [23]. Although omics sequencing and statistical analysis indicate a close relationship between the gut microbiota and T1DM, there are no reports on the implementation of microbiota transplantation or supplementation in clinical diabetic patients. Streptozotocin is the primary research method for constructing diabetic animal models [24]. In animal model research, the imbalance between Th17 and Treg cells in the pancreas decreases after supplementation with Akkermansia muciniphila (A. muciniphila), and the upregulated Treg cells exert anti-inflammatory effects, thereby reducing islet damage [25,26]. The gut microbiota, in addition to regulating intestinal barrier functions and inflammation, also participates in the development of diabetes and its complications by modulating the host’s metabolite expression levels. Stroke is a common complication in patients with T1DM. Faecalibacterium, Roseburia, and Blautia are the main contributing bacteria for butyrate. Administration of butyrate to T1DM mice can regulate the response and polarization of BV2 cells and alleviate neural damage after middle cerebral artery occlusion by downregulating MyD88 [27]. Therefore, the gut microbiota plays a dual role in T1DM through immune cross-activation, barrier disruption, and metabolic regulation.

2.2. Gut Microbiota in T2DM

In T2DM, chronic hyperglycemia exacerbates the body’s inflammatory response, damages organ function, and leads to serious complications [28]. Compared to healthy individuals, T2DM patients exhibit elevated levels of Clostridium, Sutterella, Dorea, Bacteroides, Eggerthella, Hungatella, Peptostreptococcus, Ruminococcus, Lactobacillus, and Parvimonas [29,30]. Conversely, bacteria such as Dehalobacterium, Anaeroplasma, Akkermansia, Roseburia, Adlercreutzia, and Oscillospira, along with Butyricicoccus, Lactobacillus, Bifidobacterium, Faecalibacterium, and Paraprevotella are found in reduced abundance [29,31,32]. Certain bacteria, including Roseburia, Bilophila, Oscillibacter, and Lactobacillus, may serve as markers for T2DM [33], while Flavonifractor and Haemophilus are protective, and Actinomyces and Candida are risk factors [34]. Prediabetic individuals exhibit significant changes in anaerobic bacteria, including Enterobacteriaceae, Prevotella 9, Blautia, Granulicatella, and Veillonella [35]. These changes in the gut microbiota are closely associated with impaired glucose tolerance in prediabetes; for example, FGF15/19 inhibits hepatic gluconeogenesis and promotes glycogen synthesis, while a high abundance of Bacteroides downregulates the expression of the intestinal FXR signaling pathway, thereby reducing FGF15/19 levels, which may lead to hepatic IR and impaired glucose tolerance [36]. Moreover, animal models of T2DM play an important role in the study of disease treatment; the diabetic group shows significant increases in opportunistic pathogens such as Eubacterium, Bilophila, and Mucispirillum. Meanwhile, commensal bacteria like Lactobacillus were markedly reduced [37]. In T2DM mice, dapagliflozin promotes the production of GLP-1 by modulating the gut microbiota and tryptophan metabolism, thereby facilitating beta-cell regeneration and slowing the progression of DM [38,39]. Therefore, the gut microbiota may improve T2DM by regulating metabolites, intestinal barrier function, and insulin sensitivity [40].

2.3. Gut Microbiota in GDM

GDM is a type of DM first recognized during pregnancy [41]. GDM poses several risks to mothers, including gestational hypertension, preeclampsia, and preterm delivery. It may also lead to complications such as difficult labor, postpartum hemorrhage, and postpartum T2DM [42,43]. In women with GDM, the increase in estrogen, progesterone, and human placental lactogen during pregnancy leads to IR and pancreatic β-cell dysfunction [44]. After the birth of the fetus, the decrease in these hormone levels may temporarily restore compensatory balance of blood glucose, but the damage to pancreatic β-cells is usually irreversible. Under the influence of chronic inflammation and oxidative stress, IR and β-cell function impairment may further worsen, eventually leading to the development of T2DM [45]. It potentially impacts both maternal and fetal health. Compared to healthy individuals without GDM, GDM patients showed a decrease in Lachnospiraceae, Blautia, Collinsella, Parabacteroides, Eubacterium hallii, Lactobacillus, Romboutsia, and Ruminococcus, while Bacteroides, Akkermensia, Acidaminococcus, and Lachnospiraceae NK4A136 group showed a significant increase [46,47,48,49]. MR analysis links Olsenella, Lachnoclostridium, Prevotella 9, Ruminococcus 2, and Oscillibacter with GDM [50]. Oral probiotics can reverse these changes by altering the biosynthesis and metabolism of L-asparagine and L-aspartate, thereby alleviating the symptoms of GDM [51]. In patients with GDM, the levels of 2-Hydroxybutyrate (2-HB) and L-α-aminobutyrate are increased, whereas methionine sulfoxide and allantoin are significantly decreased. 2-HB not only indicates the loss of pancreatic function but also signifies IR [52]. Ruminococcus, Eubacterium, Prevotella, and Parabacteroides increase the synthesis of 2-HB [53]. Overall, the gut microbiota regulates the metabolic imbalance associated with GDM through metabolite interactions. Targeting specific microbiota for modulation holds promise as a potential therapeutic approach to alleviate GDM and offers significant research potential [54].

2.4. Gut Microbiota in DR

In healthcare institutions in Spain, the incidence of DR among patients with T2DM is 6.99 per 1000 person-years [55]. In patients with DR, levels of Bacteroides, Roseburia, Lactobacillus, Ruminococcus, and Bifidobacterium are elevated, whereas Blautia, Faecalibacterium, Akkermansia, Clostridium, Romboutsia, and Escherichia-Shigella are depleted [56,57,58]. MR studies show that Peptococcaceae and Christensenellaceae act as protective factors against DR, while Ruminococcaceae, Adlercreutzia, and Eubacterium increase the risk of DR [59]. Gut microbiome sequencing studies using a streptozotocin-induced diabetic rat model reveal similar microbiota profiles between diabetic rats and those with DR [60]. At the genus level, Coriobacteriaceae, Veillonellaceae, Streptococcaceae, and Senegalimassilia are significantly reduced, while Burkholderiaceae, Fusobacterium, Pseudomonas, and Adlercreutzia are significantly enriched [61,62]. Vascular endothelial growth factor (VEGF) promotes angiogenesis, increases vascular permeability, and induces inflammation and oxidative stress, thereby exacerbating the inflammatory response and microvascular damage in DR [63]. In the T2DM mouse model with hindlimb ischemia, VEGF signaling molecules were positively correlated with Bifidobacterium, Clostridium sensu stricto 1, Lachnospiraceae NK4A136 group, and Coriobacteriaceae UCG 002. On the other hand, Lactococcus, Lachnoclostridium, Eubacterium brachy group, Kurthia, Weissella, Escherichia-Shigella, and Staphylococcus were negatively correlated with VEGF signaling molecules [64].

2.5. Gut Microbiota in DKD

DKD is a severe complication of DM that primarily affects the kidneys, leading to a progressive decline in their function. DKD patients have higher abundances of Blautia, Lactobacillus, Romboutsia, Turicibacter, Bacteroides, Akkermansia, Ruminococcus, Escherichia, Bilophila, and Bilophila [65,66]. Conversely, the abundances of Roseburia intestinalis, Lachnospiraceae, Faecalibacterium, and Prevotella are significantly reduced [66,67]. Certain bacteria, such as the Eubacterium nodatum group, Lactobacillus, and Faecalibaculum, show a strong correlation with metabolic disorders in DKD [68]. Rikenella and Bacilli help decrease urinary protein levels, while Lachnospiraceae increases serum creatinine and indoxyl sulfate levels [69]. In DKD animal models, Lycoperoside H inhibits the abundances of Turicibacter, Clostridium, and Bifidobacterium while upregulating Blautia, exerting therapeutic effects and reducing DKD symptoms [70]. Metabolomics studies have revealed that DKD patients exhibit significantly higher levels of D-mannose, galacturonic acid, and citric acid. In contrast, levels of 3-methylindole, 3-(2-hydroxyethyl)indole, and indole propionic acid decrease, along with reductions in selenium metabolism and BCAA synthesis pathways [71].

2.6. Gut Microbiota in DN and DCD

DN and diabetic peripheral neuropathy (DPN) are both primarily characterized by nerve damage [72], leading to sensory and motor dysfunction. Lachnospiraceae MK4B4 group, Parabacteroides, and Anaerotruncus were reduced in DN patients [73]. Conversely, there is a notable increase in the abundances of Enterococcus, Bifidobacterium, Allobaculum, Ruminococcaceae UCG 010, Anaeroplasma, Parasutterella, Dubosiella, Lactobacillus, and Turicibacter [73,74]. MR studies have indicated that Ruminococcaceae UCG 013 and Eggerthella are associated with a higher risk of developing DN [75]. In DM complicated by large vessel inflammatory lesions and hypertension, dietary phosphatidylcholine and L-carnitine are broken down by intestinal flora to produce trimethylamine N-oxide (TMAO) [76]. TMAO promotes vasoconstriction induced by angiotensin II, leading to hypertension [77]. Although oral antibiotics are a common treatment for inflammation, they reduce the beneficial effects of bacterial groups such as Bacteroidetes and Clostridium [78,79]. This reduction in gut diversity and alteration in microbial metabolic function can lead to decreased tryptophan levels and disrupted lipid metabolism, negatively impacting serum metabolism and worsening vascular sclerosis [80]. Bacteroides, Lactobacillaceae, and Desulfovibrionaceae are involved in lipid and glucose metabolism, and downregulating Bacteroides fragilis can slow atherosclerosis progression [81]. Twenty male patients with stable coronary artery disease consumed a daily beverage containing Lactobacillus reuteri 299v (Lp299v). The results indicated that Lp299v significantly reduced IL-8, IL-12, and leptin levels, and improved endothelial function [82].

2.7. The Gut Microbiome Is a Potential Target for DM Treatment

Expression levels of gut microbiota vary between different types of DM and its complications. For example, the abundance of Faecalibacterium is reduced in all DM and its complications (Table 1). Ruminococcus increases in conditions like T1DM, T2DM, DR, DKD, and DN. Regenerating islet-derived protein 3γ, a gut antimicrobial peptide with bactericidal activity, is effectively induced by Ruminococcus gnavus (R. gnavus), leading to rapid microbial death [83]. Implantation of a single strain of R. gnavus from lupus patients into C57BL/6 mice increased intestinal permeability [84]. Oral administration of R. gnavus in DN mice exacerbated renal lesions by upregulating IL-6, increasing levels of creatinine, urinary protein, and blood urea nitrogen levels. At the same time, it downregulated the tight junction proteins ZO-1, Occludin, and Claudin-1 [85]. In mice with gastric mucosal inflammation, R. gnavus led to plasma cell and lymphocyte infiltration into the gastric mucosa lamina propria, increasing the proportion of neutrophil infiltration [86]. Similarly, Bacteroides increases in both GDM, DR, DKD, and T2DM, while Lactobacillus rises in T1DM, T2DM, DR, DKD, and DPN, but decreases in GDM. Bifidobacteria decreases in T1DM, T2DM, and GDM but increases in DR and DN. Probiotic preparations containing Bifidobacterium and inulin (INU) have significantly and persistently reduced visceral fat area and total fat area in animals [87]. Lactobacillus exerts anti-inflammatory effects in the early stages of intestinal inflammation, protecting the intestinal barrier [88]. Clostridium decreases in both T1DM, GDM, and DR, and Blautia decreases in T1DM and DR but increases in GDM, DKD, and DN. Roseburia decreases in T2DM, GDM, and DKD, and Romboutsia decreases in T1DM, T2DM, GDM, DR, and DKD, while Akkermansia increases in DKD but decreases in T2DM, GDM, DR, and DN. We observed that the abundances of Akkermansia, Clostridium, and Lactobacillus at the genus level exhibited different trends. However, the microbial effects of Akkermansia at the species level were inconsistent [89], which might be due to the limitations of the current 16S rRNA sequencing technology. Moreover, factors such as genetics, diet, and chronic inflammatory stimuli may also contribute to these variations [90].
The gut microbiota plays a crucial role in the progression of DM and its complications, as well as in immune regulation [91]. In the T1DM model, following human amniotic mesenchymal stem cell treatment, the abundance of Alcaligenes, Bifidobacterium, and Prevotella increased, promoting the frequency of Th2 and Treg cells in the mesenteric lymph nodes, reducing the frequency of Th1 and Th17 cells, and significantly improving blood glucose and insulin secretion [92]. A study on human microbiota-associated mouse models found that transplanting fecal microbiota from T1DM patients and healthy individuals into germ-free mice resulted in phenotype reproducibility, providing feasibility and new research directions for microbiota transplantation [93]. A clinical controlled trial report shows that after receiving oral administration of A. muciniphila, T2DM patients experienced reduced weight and glycated HbA1c compared to the control group. Although the intergroup differences were not significant, this provides guidance for the clinical supplementation of gut microbiota [94]. As a new non-pharmacological treatment, the gut microbiota treats and improves DM and its complications by regulating gut permeability and insulin sensitivity, with significant potential clinical value [95]. By intervening in the abundance of these flora, the disease severity can be improved. These specific differences in gut microbiota are not only potential biomarkers for early detection and risk stratification but also provide promising therapeutic targets for modulating the gut microbiome to prevent or treat DM and its complications.
Table 1. Abundance and potential function of gut microbiota in DM and its complications.
Table 1. Abundance and potential function of gut microbiota in DM and its complications.
Gut MicrobiotaDM and Its ComplicationsFunctionReferences
T1DMT2DMGDMDRDKDDN/DPN
BacteroidesFacilitates carbohydrate breakdown, improving insulin sensitivity and blood glucose regulation; dual inflammatory control[29,47,56,65,96]
FaecalibacteriumButyrate production, enhances gut barrier integrity and insulin sensitivity[29,32,47,57,66,96]
LactobacillusProbiotics; maintaining intestinal homeostasis, regulating metabolism, and immunity; SCFA production[29,48,57,66,74]
RuminococcusGenerate acetic acid and butyric acid, dual insulin resistance, blood glucose and inflammation regulation[20,28,47,58,66,96]
ClostridiumButyrate production, improves insulin sensitivity, and lowers blood glucose; dual inflammatory control[18,29,47,57,67,74]
Blautia Generate SCFAs, reduce inflammation, and improve insulin sensitivity and glucose metabolism[18,46,56,67,96]
Roseburia Butyrate production, anti-inflammatory effects, and improves insulin sensitivity[20,28,47,58,66]
Romboutsia Modulating gut barrier, glycemia, and inflammation[29,49,56,67]
Akkermansia Enhances gut barrier integrity and insulin sensitivity[28,46,58,67]
Bifidobacterium Acetate and lactate production, improve glycemic control and insulin sensitivity[29,46,57,73]
Prevotella SCFA production, dual insulin sensitivity, and glycemic control[29,47,67]
T1DM, Type 1 diabetes mellitus. T2DM, Type 2 diabetes mellitus. GDM, Gestational diabetes mellitus. DR, Diabetic retinopathy. DKD, Diabetic kidney disease. DN, Diabetic neuropathy. DPN, Diabetic peripheral neuropathy. ↑, This abundance increases. ↓, This abundance decreased. SCFAs, Short-chain fatty acids.

3. Targeting the Gut Microbiota–Metabolite–Signaling Pathway Axis to Improve DM

3.1. Gut Microbiota Participates in DM by Regulating Metabolites

The gut microbiota not only plays a key role in digestion and nutrient absorption but also engages in complex interactions with the host’s metabolic pathways through its metabolites, thus affecting the expression levels of multiple systems within the body, including energy metabolism, fat metabolism, glucose metabolism, and immune response [97]. However, the specific mechanisms remain unclear. Studies have shown that certain bacteria, such as Lactobacillus and Prevotella UCG 001, are closely associated with sphingolipid metabolism as well as the biosynthesis of tryptophan and tyrosine [98]. Blautia and Clostridium are positively linked to glutamine levels, while showing negative correlations with aspartic acid, methionine, lysine, and 1-methyl-L-histidine [99]. On the other hand, Alloprevotella is inversely related to γ-aminobutyric acid and hexanoylglycine, whereas Prevotella is positively correlated with compounds like 5-hydroxyindoleacetic acid, O-acetyl-L-serine, glutamic acid, and aspartic acid [100]. Furthermore, the families Christensenellaceae R7 and Ruminococcaceae UCG 002 are strongly linked with higher expression of ornithine and L-arginine [101]. L-isoleucine and phenylalanine are positively correlated with Enterobacteriaceae, Clostridiaceae, and Romboutsia [102]. These complex interactions between microbiota metabolites underscore the critical role the gut microbiota plays in regulating metabolic processes. Specific microbiota may exert bidirectional effects in the pathogenesis of metabolic diseases by regulating fatty acid, amino acid, and lipid metabolism pathways (Figure 2).
Previous studies have confirmed that balancing SCFAs is beneficial for the human body. Oscillibacter, a producer of LPSs, along with Bacteroides and Odoribacter, which produce SCFAs, are less abundant in DKD patients [71]. Blautia coccoides (B. coccoides), Prevotella, along with Ruminococcaceae and Lachnospiraceae, are involved in producing SCFAs [103]. Clostridium sensu stricto, Lactobacillus, and Bacteroides primarily produce acetic acid [104]. Additionally, combined interventions of Veillonella and Lactobacillus increased the abundance of commensal bacteria such as Akkermansia and the total content of SCFAs, while reducing the abundance of opportunistic pathogens like Desulfovibrio and Escherichia-Shigella. These changes collectively exerting anti-inflammatory effects [105]. In mice with T2DM, anaerobic bacteria such as Bacteroides, Cyanobacteria, Colidextribacter, Lachnospiraceae NK4A136, Roseburia, Dubosiella, Oscillibacter, and Lachnoclostridium are involved in the biosynthesis of BCAAs [106]. In diet-induced obese mice, gavage administration of Bacteroides spp. improved imbalanced BCAA metabolism and reversed the increased body weight [107].
In an inflammatory bowel disease mouse model, Fournierella, Clostridium, and Peptostreptococcus promote the synthesis of IPA, while Lachnoclostridium, Erysipelatoclostridium, and Parabacteroides reduce its abundance by consuming IPA [108]. Clostridium species promote the synthesis of IPA, exerting inhibitory effects that significantly improve glucose metabolism and reduce body weight [109]. TMAO has regulatory effects on insulin sensitivity, inflammatory response, and lipid metabolism [110]. Bacteroides and Clostridium promote the production of trimethylamine N-oxide [111]. Quinic acid modulates the abundance of gut microbiota by downregulating Streptococcus danieliae and upregulating Ileibacterium valens and Lactobacillus intestinalis. It regulates TMA/TMAO-mediated hepatic lipid metabolism, improving atherosclerosis in Apoe−/− mice [112]. 16S sequencing results of the gut microbiota in patients with impaired glucose tolerance show a decrease in Faecalibacterium, which is capable of producing taurodeoxycholic acid and carnosine. This reduction weakens the inhibitory effect on insulin-like growth factor binding protein 3, exacerbating hyperglycemia and related vascular lesions [113]. Therefore, the gut microbiota demonstrates new research potential by reshaping SCFAs, BCAAs, and other regulatory metabolites to regulate metabolic diseases.

3.2. BCAAs, SCFAs, and IPA Regulate Signaling Pathways to Improve DM and Its Complications

The insulin signaling and AMPK pathways are primarily responsible for glucose uptake, glycogen storage, and endocrine metabolism [114]. Excessive activation of the mTOR pathway, however, can exacerbate IR [115]. The NF-κB pathway is closely related to the body’s inflammation levels. High expression of NF-κB leads to a high inflammatory response in DM, promoting the occurrence of complications [116]. Additionally, the binding of advanced glycation end-products (AGEs) to their receptor for advanced glycation end-products (RAGE) initiates a series of downstream signals that contribute to the development and progression of DM. Studies have confirmed that different expression levels of metabolites can regulate DM-related signaling pathways. BCAAs, significant components of AAs in the body, regulate the Akt2 signaling pathway, impacting lipid metabolism and glucose homeostasis [117] (Figure 3). Supplementing BCAAs may activate the INFGR1/JAK1/STAT1 pathway, promoting pro-inflammatory macrophages and IR [118]. BCAAs enhance the fatty acid oxidation function by modulating the GCN2/ATF6/PPAR-α signaling pathway [119]. The liver plays a crucial role in regulating diabetes homeostasis. In a cirrhotic rat model treated with BCAAs, BCAAs reduced the protein expression of lipopolysaccharide-binding protein, Toll-like receptor 4, and STAT3, thereby protecting liver function [120]. However, in T1D mice, the gut microbiota shows insufficient degradation of BCAAs. This activates the mTOR signaling pathway, leading to mitochondrial damage and cardiomyocyte apoptosis [121].
Research has found that Streptococcus, Prevotella, and Faecalibacterium, along with species like Bacteroides and Eubacterium, are significantly reduced [122,123], leading to decreased SCFA production in GDM patients. In diabetic neuropathy, SCFAs exert antioxidant protection through the G protein-coupled receptors (GPR)43 pathway (Figure 4); under oxidative stress induction, neuronal cell lines undergo dysfunction, whereas GPR43 exerts a protective effect by inhibiting H2O2-induced neuronal damage [124]. Additionally, the NF-kappaB/MAPKs signaling pathway further reduces inflammation in the body under the inhibition of GPR43 [125]. The NLRP3/Caspase-1 pathway regulates THP-1 cell metabolism inhibited by SCFAs to alleviate cellular inflammation [126]. The GLP-1/GLP-1R/cAMP/PKA/CREB/INS pathway enhances the management of T2DM under the influence of SCFAs [127]; SCFAs inhibit the HDAC3-H3K27ac-PPAR-γ axis, thereby reducing lipid storage in the body [128]. Additionally, SCFAs decrease NF-κB transcriptional activity and restore apolipoprotein A-I transcription levels in HepG2 cells [129]. Butyrate reduces apoptosis, thereby ameliorating DKD, primarily by inhibiting the NLRP3 inflammasome via the STING/NF-κB/p65 pathway [130]. Restoration of gut microbiota and immune regulation imbalance was observed in T1DM patients receiving oral SCFAs biotherapy. Transplanting the fecal microbiota from these patients into humanized GF mice could delay the progression of DM [131]. Therefore, restoring the balance of SCFA expression levels can further alleviate DM and its complications. T2DM is often accompanied by cognitive decline. Studies have shown that IPA can prevent neuronal death and restore mitochondrial function. Under intermittent fasting treatment, the levels of SCFAs and IPA are upregulated in db/db mice. However, antibiotic treatment exacerbates cognitive impairment by inhibiting gut microbiota and reducing IPA [132]. Supplementation with IPA restored colonic barrier function by upregulating IL-25 and alleviated obesity and metabolic disorders induced by a high-fat (HF) diet [133]. Furthermore, network pharmacology studies have revealed that IPA can modulate signaling pathways such as NF-κB, VEGF, and TNF. These pathways are closely associated with DM and its complications [134,135,136].

3.3. Other Metabolite Regulation Pathways Improve DM and Its Complications

The PI3K/AKT/mTOR, NF-κB, and MAPK signaling pathways, along with oxidative stress, play crucial roles in diabetes and its complications by regulating glucose metabolism and insulin sensitivity. In DM rats, TMAO accelerates wound healing by modulating the PI3K/AKT/mTOR pathway [137]. Increased expression of neutrophil extracellular traps (NETs) can inhibit the function and angiogenesis of HTR-8/Svneo cells in patients with GDM. However, feeding PAD4−/− mice with TMAO inhibits NET formation, thereby promoting the development of the placenta and fetus [138]. In other metabolites such as phytosphingosine, the highly inflammatory response induced by pathogenic cytokines is improved through the MAPK and NF-κB pathways [139]. High inflammatory response mediated by LPSs can be reversed by the inhibition of NF-κB and downstream pathways by madecassic acid [140]. On the other hand, lupeol alleviates oxidative stress-induced islet inflammation and apoptosis in streptozotocin-induced hyperglycemic mouse models [141]. Ginsenosides exhibit unique biological effects in DM metabolism, such as ginsenoside Rk3 improving metabolic disorders by upregulating the AMPK/Akt pathway [142]. Ginsenoside Rg3 exerts cardioprotective effects by activating the PPAR-γ signaling pathway [143]. Ginsenoside Rd alleviates hyperglycemia complications through the Akt pathway [144], and ginsenoside Rb1 mitigates diabetic atherosclerosis via the AMPK pathway [145]. IR in DM can be improved by ginsenoside Rg5 upregulating the Sirt1/PGC-1α pathway [146].
HepG2 cells have a regulatory effect on IR, and eicosapentaenoic acid (EPA) can reverse the imbalance of HepG2 cells through the ROS/JUN pathway [147] and alleviate inflammation and oxidative stress by miR-1a-3p/sFRP1/Wnt/PCP-JNK [148]. Furthermore, EPA activates the AMPK pathway to optimize glucose metabolism [149], and promotes the generation of docosahexaenoic acid, providing antioxidant protection in retinopathy [150]. Chenodeoxycholic acid has multiple biological effects in the human body by upregulating ROS/p38 MAPK/DGAT1 to catalyze lipid peroxidation in DM [151]. Chenodeoxycholic acid can also activate the FXR-MLCK pathway, further reducing LPS damage to the intestinal epithelial barrier [152]. In addition, the AMPK/NF-κB and TGFβ1-Nrf2 pathway can promote ferritin deposition and cell apoptosis, while Lupeol exerts anti-inflammatory effects by inducing oxidative stress and apoptosis through the AMPK/NF-κB pathway [153], further improving bile acid metabolism [154]. We believe that targeting the gut microbiota, metabolites, and disease-related signaling pathways within the “gut microbiota–metabolite–signaling pathway” network can restore metabolic balance in diabetic patients, thereby potentially improving DM management and reducing complications (Table 2).

4. TCM Therapeutic Strategies Targeting Gut Microbiota and Metabolites

4.1. TCM Treats DM Through Gut Microbiota and Metabolites

Gut microbiota and their metabolites have become new avenues for TCM treatment [155]. Polysaccharides and polyphenolic compounds in TCM regulate gut microbiota and their metabolites, reduce inflammatory responses, and improve gut health. Additionally, the bioactive components of TCM can be further activated by the gut microbiota, enhancing pharmacological activity. TCM regulates SCFA expression levels through gut microbiota, controlling hyperglycemia and reducing inflammation (Table 3). In T1DM, supplementation with extra virgin olive oil can slow gastric emptying, enhance the abundance of Lachnoclostridium and Ruminococcaceae UCG 005, and promote the production of SCFAs [156]. Additionally, prebiotic ham combined with high-amylose maize starch improves beta-cell function and blood glucose regulation through SCFA modulation [157]. Astragalus polysaccharide can increase the levels of propionic acid, acetate, and butyrate salts in T1D-diabetic mice, thus helping to restore the gut microbiota and suppress the inflammatory response [158]. INU and soluble fiber inulin and omega 3-PUFA promote the production of essential SCFAs by commensal bacteria, which helps regulate the immune system by recruiting regulatory T cells to the pancreas via CCL17, enhancing the immune response [159]. TCM has shown beneficial effects in the treatment of T2DM. Fuzhuan brick tea increases Ruminococcus, lactic acid bacteria, and Lachnospiraceae NK4A136 group, while reducing Prevotella and Faecalibacterium, thereby enhancing SCFA levels [160]. Phellinus linteus enhances the levels of Bacteroides, ParaBacteroides, and Alistipes, which are associated with SCFAs and bile acid metabolism [161]. Canagliflozin upregulates Muribaculaceae and Bacteroidaceae, promoting an increase in SCFA levels [162]. Carvacrol restored blood glucose and lipid disorders in T2DM rat models by increasing SCFA levels and upregulating GPR 43/41 expression [163]. In a GDM mouse model, high-fermentation dietary fiber like konjac reduces intestinal permeability and LPS by increasing abundances of Lachnospiraceae and butyrate [164].
The study of TCM extracts has a long history in China. Cinnamaldehyde is the main component of the cinnamon tree. In T1D mice, blood glucose levels are regulated by cinnamaldehyde through modulation of the gut microbiota and bile acids, leading to a reduction in blood glucose [165]. In T2DM mice, mulberry leaf water extract restores intestinal permeability and glucose and lipid metabolism, and this effect is achieved through the gut microbiota [166]. Jiang-Tang-San-Huang promotes the abundance of Bacteroides and Bifidobacteria, which helps to correct gut dysbiosis and regulate inflammatory pathways [167]. Polysaccharides from Dendrobium officinale boost the abundance of Eubacterium, Bifidobacterium, and lactic acid bacteria, inhibit Helicobacter pylori, and significantly enhance the intestinal barrier function [168]. Components of Tribulus terrestris help regulate imbalanced microbiota and increase the levels of commensal bacteria [169]. Ganzhou orange peel pectin, a plant extract, reduced opportunistic pathogens like Alistipes, Oscillibacter, and Helicobacter in diabetic mice, while commensal bacteria like Dubosiella, Akkermansiaceae, and Atopobiaceae increased [170].
Polysaccharides have become a prominent focus in TCM [171]. Sequencing of the gut microbiota in a T1D rat model revealed that the polysaccharides of D. huoshanense promote the abundance of Firmicutes and the expression levels of SCFAs [172]. Crude polysaccharides from D. divaricata regulate the levels of mucin-2 and tight junction proteins, restoring intestinal function, and enhance the regulatory role of insulin receptor substrate-1 on insulin [173]. Licorice-derived polysaccharides administered to mice increased levels of Romboutsia, Akkermansia, lactic acid bacteria, and other commensal bacteria, while inhibiting the pathogenic bacterium Bacteroides [174]. Superoxide dismutase and catalase, under the action of golden flowers containing polysaccharides, polyphenols, proteins, and amino acids, downregulate the expression of TNF-α, IL-4, and IL-6 to alleviate oxidative stress in the body [175]. Modern technology has also facilitated the discovery of traditional medicine. For example, low-methoxyl pectin opens up a realistic option for controlling gut microbiota and preventing T1DM [176]. Flavonoids from cactus upregulate the expression of SCFAs to exert resistance against DKD [177].
Table 3. Regulatory effects of TCM on gut microbiota and metabolites in different types of DM.
Table 3. Regulatory effects of TCM on gut microbiota and metabolites in different types of DM.
TypesDrugResearch SubjectGut MicrobiotaMetabolitesReferences
IncreaseDecreaseUpDown
T1DMExtra virgin olive oilNOD miceLachnoclostridium,
Ruminococcaceae UCG 005
Lachnospira,
Eubacterium
Madecassic acid, LupeolGinsenoside, Oleamide[156]
Astragalus polysaccharidesT1D miceMuribaculum, Lactobacillus,
Bacteroides
Corynebacterium,
Brevibacterium,
Brachybacterium
Acetic acid, PA,
BA, SCFAs
Isobutyric acid,
Isopentanoic acid, BCFAs
[158]
Soluble fiber inulin and omega 3-PUFANOD miceAkkermansiaBacteroides intestinalis, Streptococcus sp.Docosapentaenoic acid,
Docesahexaenoic acid,
Eicosapentaenoic acid
2-Hydroxybutyric acid[159]
CinnamaldehydeT1DM model miceParasutterella, Odoribacter, BurkholderialesDorea, MucispirillumMyristoleic acid,
3-hydroxybutyric acid
Hydrocinnamic acid, 2-phenylpropionate[165]
Polysaccharides of
D. huoshanense
T1D miceLactobacillus, MegasphaeraBacteroides,
Parabacteroides,
Dorea, Enterocloser
Acetic acid, PA, Butyrate/[172]
Crude polysaccharidesT1D miceLactobacillusRuminococcaceae, Lachnospiraceae, Rikenellaceae//[173]
“Golden-flower” Tibetan teaT1D miceLactobacillus,
Lachnospiraceae NK4A136 group
BacteroidesSCFAs, Superoxide dismutase, Catalase/[175]
Low-methoxyl
pectin
NOD miceFirmicutes, TM7, ProteobacteriaBacteroidetesCetate, Propionate, Butyrate, SCFAs/[176]
T2DMEthanol extract of propolisT2D miceParasutterella, Bifidobacterium,
Faecalibaculum, Dubosiella,
Lachnoclostridium
N-Acetyl-L-glutamic acid, D-(+)-Galactose, (R)-Lactate, L-(+) Lactic acidLactulose, L-Proline,
O-Acetyl-L-serine,
S-Adenosyl-L-homocysteine
[37]
Polysaccharides from Phellinus linteusT2D rat modelAlistipes, Prevotellaceae,
Bacteroides,
Parabacteroides
Faecalibaculum, LachnospiraceaeSCFAs,
Primary bile acids
Aspartate aminotransferase alanine aminotransferase,
Primary bile acids
[161]
Morus alba L. water extractsT2D miceDubosiellaAnaerovorax, Bilophila, Blautia, LachnoclostridiumBranched-chain ketoacid,
Dehydrogenase E1α
Amino acid[166]
Jiang-Tang-San-HuangT2D rat modelRomboutsia, Lactobacillus,
Bacteroides,
Bifidobacterium
EnterococcsPrimary bile acids, Chenodeoxycholic acidTaurocholic acid[167]
Navel orange peel pectindiabetic mouseDubosiella, Akkermansia,
Lachnospiraceae, Atopobiaceae
Muribaculaceae,
Lachnospiraceae NK4A136 group
Acetic acid, Total acid, BAPA[170]
Polysaccharide
extract
T2D miceAkkermansia, Lactobacillus, Alistipes,
Romboutsia,
Faecalibaculum
Bacteroides, Alloprevotella,
Escherichia-Shigella, Clostridium
Propionate, ButyrateTriglycerides, Total cholesterol [174]
GDMInulin-type fructansGDM miceAkkermansia, BifidobacteriumDubosiellaBA, Acetic acid/[122]
KonjacGDM miceDubosiella, MonoglobuBavteroides, Romboutsia,
Faecalibaculum
PhenylalanineDiamine oxidase, LPS, Valine, Leucine, Isoleucine[164]
T1DM, Type 1 diabetes mellitus. NOD mice, Non-obese diabetic mice. T2DM, Type 2 diabetes mellitus. GDM, Gestational diabetes mellitus. PA, Propionic acid. BA, Butyric acid. BCFAs, Branched-Chain Fatty Acids. SCFAs, Short-chain fatty acids.

4.2. TCM Treats DM Complications Through Gut Microbiota and Metabolites

DM complications seriously threaten human health. The Luo Tong Formula improves intestinal microecological balance in diabetic rats by reversing changes in gut microbiota, such as Enterobacteriaceae, Bacteroidetes, Prevotellaceae, Enterococcaceae, and Klebsiella, thereby mitigating the hyperglycemic and inflammatory state of DR [178]. Quercetin improves DR by balancing gut microbiota and intestinal permeability [179]. In the DKD mouse model, the Qing-Re-Xiao-Zheng formula inhibits the expression of NF-κB and Toll-like receptor 4 [180]. The polysaccharides in the Fufang-zhenzhu-tiaozhi formula and polysaccharides from Moutan Cortex increased the expression levels of short-chain fatty acids, thereby alleviating renal injury [181,182]. Fructooligosaccharides, a type of soluble dietary fiber, act as prebiotics that alleviate DKD by preventing LPSs from entering the circulatory system and reducing their expression levels within the body [183]. A. muciniphila and Lactobacillus murinus are closely related to glucose and lipid metabolism. Astragalus membranaceus and Salvia miltiorrhiza improve the DKD rat model by upregulating the abundance of A. muciniphila and Lactobacillus murinus [184]. INU-type fructans enhance the abundance of Akkermansia and Candidatus Saccharimonas, thereby alleviating mitochondrial dysfunction and downregulating toxic glucose metabolites, which further reduce glomerular injury and renal fibrosis [185]. These microbial communities significantly regulate metabolite levels in the body, thereby treating and alleviating DKD (Table 4).
In DN, corn silk polysaccharides improve DN by adjusting the abundance of Dubosiella, Bacteroidetes, and Firmicutes, enhancing the synthesis and metabolism of bile acids, FAs, tryptophan, and tyrosine [186]. San-Huang-Yi-Shen Capsules affect the abundance of Ruminococcaceae UCG 005, Lactobacillus, Anaerostipes, and Anaerococcus, and affect the expression levels of metabolites such as L-carnitine and threonine [187]. Clostridium, Bifidobacterium, and Fusobacterium inhibit p-cresol formation, while magnesium lithospermate B and its metabolite danshensu improve DN via gut microbiota [188]. In animal models, sanziguben polysaccharides reduce the abundance of Proteobacteria and Klebsiella, thereby modulating LPS levels in DN mice. TCM also has therapeutic effects on DPN. In the DPN rat model, Jinmaitong restores gut microbiota imbalance and upregulates serum NRG1 levels to protect against DPN [189]. Similarly, gingerol-enriched ginger and quercetin also have preventive and therapeutic effects in DPN [190,191]. Agarotetraose increased the abundance of Muribaculaceae, Lachnospiraceae, and Blautia, while decreasing the abundance of Faecalibaculum and Desulfovibrio. Further studies showed that the total bile acid content in feces is negatively correlated with atherosclerosis progression [192]. Huangqi Guizhi Wuwu Decoction has a significant therapeutic effect on diabetic peripheral neuropathy by regulating the abundances of Lactobacillus, Prevotella, Bacteroidetes, and Desulfovibrio. Correlation analysis has suggested that it may regulate the biosynthesis of unsaturated FAs and the metabolic functions of valine, leucine, and isoleucine [193]. Berberine is a prominent topic in pharmacological research [194]. Animal studies have found that berberine regulates the Lachnospiraceae NK4A136 group, Eubacterium, and Bacteroidales S24 7 group, inhibiting the production of trimethylamine and trimethylamine N-oxide, thereby alleviating choline-induced atherosclerosis.
Table 4. Regulatory effects of TCM on gut microbiota and metabolites in different types of DM complications.
Table 4. Regulatory effects of TCM on gut microbiota and metabolites in different types of DM complications.
TypesDrugResearch
Subject
Gut MicrobiotaMetabolitesReferences
IncreaseDecreaseUpDown
DRLuo Tong formulaDR rat modelCandidatus_Saccharimonas,
Romboutsia, Enterorhabdus
Prevotella//[178]
QuercetinSprague Dawley miceTuricibacter, Roseburia, BifidobacteriumStreptococcus, Veillonella, PrevotellaAcetic acid, PA, BA/[179]
DKDTangshen FormulaDKD miceBarnesiellaRomboutsia, Akkermansia, CollinsellaTryptophan, 5-hydroxyindoleacetate, Glutamic acid, AspartateIndole-3-acetic acid, Xanthurenic acid[100]
Qing-Re-Xiao-Zheng formulaDKD miceRikenellaceae, AkkermansiaDesulfovibrioSCFAsLPSs[180]
Fufang-zhenzhu-tiaozhi formulaDKD miceBacteroidota, Actinobacteriota, PseudonocardiaWeissella, Enterococcus,
Akkermansia
PA, Methylmalonic acid, Butanoic acid3-hydroxybutyrylcamitine, Gamma-muricholic acid[181]
PolysaccharidesDKD rat modelMollicutes, Bacteroidota,
Ruminococcaceae_UCG-014
LactobacillusAcetic acid, PA, BAIsovaleric acid, BCFAs[182]
Salvia miltiorrhizaDKD rat modelAkkermansia, Lactobacillus,
A. musciniphila
Prevotellaceae UCG 001Phytosphingosine, SphinganineIndolyl sulfate, P-cresolsulfate, Myo-inositol[184]
DNSan-Huang-Yi-Shen capsuleDN rat modelLactobacillus, Allobaculum,
Ruminococcaceae UCG 005,
Anaerovibrio, Bacteroides
Candidatus SaccharimonasAmino sugar,
Pyruvate metabolism,
Nucleotide sugar metabolism
TCA cycle,
Arachidonic acid,
Mannose metabolism
[187]
Magnesium lithospermate Bdiabetic mouseBifidobacterium, Lachnospiraceae,
Aerococcus, Bacteroidales
Alistipes,
Lachnospiraceae NK4A136 group
BA, Isobutyric acid, Pentanoic acid, Alanine, Threonine, Glycine, LysineTyrosine[188]
DPNJinmaitongDPN ratsHelicobacterae, Blautia, Escherichia-ShigellaClostridium, Oscillibacter//[189]
QuercetinDPN ratsPrevotella, Escherichia-Shigella, BifidobacteriumDesulfovibrio, Lactobacillus//[190]
Gingerol-enriched gingerDPN ratsLachnospiraceae //[191]
Huangqi Guizhi Wuwu Decoctiondb/db miceLactobacillus, Alloprevotella,
Bacteroides
Lachnoclostridium, Blautia,
Desulfovibrio Ruminococcus,
Akkermansia, Caproiciproducens
/Sphinganine,
Sphingosine 1-phosphate, Phytosphingosine
[193]
DR, Diabetic retinopathy. DKD, Diabetic kidney disease. DN, Diabetic neuropathy. DPN, Diabetic peripheral neuropathy. PA, Propionic acid. BA, Butyric acid. BCFAs, Branched-Chain Fatty Acids. SCFAs, Short-chain fatty acids. LPSs, Lipopolysaccharides.

4.3. Future Treatment Strategies of TCM in DM

TCM exerts unique therapeutic advantages in improving glucose and lipid metabolism and alleviating inflammation through multi-target modulation of gut microbiota and its metabolites [184]. Due to their genetic similarity to humans, rodent models, particularly mice [195], are widely used in preclinical studies of TCM for the treatment of DM. Researchers have developed various modeling strategies to induce acute or progressive β-cell destruction, hyperglycemia, and IR in these animals. For instance, Kachapati et al. [196] and Sharma et al. [197] found that mice exhibit many characteristics similar to humans regarding hyperglycemia and renal pathology. However, there are significant physiological and metabolic differences between rodents and humans. These differences include variations in lifespan, immune system responses, and metabolic rates, which may affect the translation of research findings to human clinical applications. Nevertheless, rodent models remain important tools for advancing our understanding and treatment of DM. Liu et al. [198] and Takaichi et al. [199] successfully transplanted human stem cells into DM models induced in C57BL/6J mice without encountering issues of immune rejection in transplantation therapy. Therefore, it is important to critically analyze TCM research findings based on animal models.
TCM mainly includes herbal medicine, acupuncture, dietary therapy, and physical exercise. Among these, Chinese herbal medicine is the most commonly used [200]. However, Chinese herbal medicine involves potential risks, such as the use of toxic substances, ingredients from endangered species, and poorly characterized bioactive compounds [201]. Therefore, the use of herbal medicine outside the framework of TCM diagnosis is not considered a standardized medical procedure. Although TCM has shown significant protective effects against diseases such as DM, there is a lack of critical analysis and standardized reporting to guide its use [202]. Therefore, it is essential to rigorously evaluate the role of TCM in nutritional intervention and precision medicine when exploring the effectiveness of TCM and its components in improving and treating DM and its complications [203]. Although the mechanisms of bioactive components such as polysaccharides remain unclear, we believe that future treatment strategies for DM and its complications should focus on regulating specific microbiota, enhancing intestinal barrier function [204], and reducing the entry of harmful bacteria and LPSs into the bloodstream, thus mitigating metabolic inflammation [205]. Furthermore, TCM can modulate metabolites such as SCFAs, BCAAs, bile acids, and TMAO, improving insulin sensitivity, microvascular complications, and glucose homeostasis. It also exerts anti-inflammatory effects by inhibiting the activity of inflammatory factors like TNF-α, IL-6, and signaling pathways such as NF-κB and AGEs/RAGE [206]. Targeting multiple points in the “gut microbiota–host metabolite” network to conduct clinical translational research on TCM derivatives (such as TCM dietary therapy, medicinal herbal beverages, and herbal medicine extracts) in the treatment of DM and its complications may become a promising new treatment approach.

5. Diet as Medicine: Treatment Strategies for DM and Its Complications

5.1. Adjust Dietary Structure to Improve DM and Its Complications

Eating habits exhibit regional, cultural, and ethnic characteristics; dietary intake is broken down and digested with the help of gut microbiota. The Mediterranean diet (MedDiet) is characterized by an increased intake of legumes, fruits, whole grains, and vegetables, and reduced intake of red meat and sweets [207]. In MedDiet interventions involving increased physical activity and reduced caloric intake (Table 5), the Eubacterium hallii group and Dorea decreased, while alpha diversity significantly increased [208]. The Japanese diet is similar to the MedDiet, with a higher intake of vegetables, fish, and soy products, and a lower intake of meat. This diet leads to an increase in Lachnospiracea incertae sedis, Gemmiger, and Faecalibacterium, while SCFA-producing bacteria significantly decline [209]. Time-restricted eating encourages regular eating patterns, whereas Ramadan fasting involves a reversal of typical eating times. Significant differences in the abundances of Roseburia, Akkermansia, Bacteroides, Prevotella 9, and Megamonas were observed between these two dietary patterns [210]. Floral tea is commonly part of daily life. Feeding mice an HF and high-sucrose (HS) diet along with alcohol increased opportunistic pathogens; however, supplementation with water extract of Chrysanthemum morifolium Ramat. reversed this effect, significantly increasing the abundances of Clostridium and Faecalibaculum [211]. Capsaicin is commonly present in Chinese cuisine. Studies have found that capsaicin can reduce the abundance of Streptococcus, Enterococcus, Barnesiella intestinihominis, and Eubacterium uniformis, which are closely related to the generation of SCFAs [212].
Cold drinks and HF diets are associated with increased systemic inflammatory responses and mucosal barrier dysfunction [213]. Eggs are nutrient-rich, and in an aging mouse model, the abundances of Blautia, Odoribacter, and Alistipes significantly increased in mice fed with eggs [214]. Phenylalanine and tryptophan from diet are metabolized by R. gnavus to produce phenylethylamine and indole, which activate enterochromaffin cells in the gut to synthesize serotonin [215]. Studies confirm that the diversity of the gut microbiota is modulated by diet (Figure 5). HF and HS diets are an important factor in the occurrence and development of DM and its complications. A diet rich in fat and sugar can aggravate DM, leading to a decrease in Turicibacter, Ileibacterium, and Bifidobacterium, while increasing the abundances of Romboutsia, Lactococcus, and Enterococcus [216]. An HFD and high-carbohydrate diet can increase the production of SCFAs by promoting the fermentation process of gut microbiota [217]. In HF diet-fed mice, resistant starch type 1 derived from potatoes increased the abundance of Akkermansia, while decreasing the abundance of Muribaculaceae [218]. Therefore, dietary interventions that regulate microbial balance are feasible and promising strategies for disease management and treatment [219,220,221,222].
Table 5. The impact of dietary types on gut microbiota and its potential functions.
Table 5. The impact of dietary types on gut microbiota and its potential functions.
Type of DietGut Microbiota AbundanceFunctionReferences
IncreaseDecrease
MedDietRoseburia, Bacteroides, Faecalibacterium, Akkermansia, Bifidobacterium, Lachnospiraceae_UCG.001Eubacterium hallii group and Dorea, Blautia, Romboutsia, Ruminococcus, Prevotella 9Promotes the growth of probiotics, lowers blood glucose, and has anti-inflammatory effects[208]
Japanese dietLachnospiracea, Gemmiger, FaecalibacteriumAlloprevotella, Bifidobacterium, Actinomyces, ParabacteroidesReduces the risk of diabetes, improves blood glucose control[209]
TREFaecalibacterium, DialisterAlloprevotella, PrevotellaEnhances insulin sensitivity, reduces body fat, and optimizes metabolism[210]
RFFaecalibacterium, Roseburia, Akkermansia, Bacteroides, Allobaculum, BlautiaPrevotella 9Improves insulin sensitivity, promotes weight loss, and reduces inflammation[210]
C. morifoliumAkkermansia, Bacteroidales, RikenellaceaeClostridium, FaecalibaculumReduces the risk of diabetes, improves blood glucose control[211]
CapsaicinAkkermansia, AnaerotruncusStreptococcus, Alistipes, Faecalibacterium, Barnesiella intestinihominisImproves glucose and lipid metabolism disorders[212]
CDHFD/Muribaculum, OdoribacterAggravates metabolic disorders associated with diabetes[213]
Eggs (selenium and/or zinc)BlautiaAlistipes, OdoribacterAntioxidant effects and enhances insulin sensitivity[214]
HF and HS dietRomboutsia, Lactococcus, and EnterococcusTuricibacter, Ileibacterium, BifidobacteriumPromotes insulin resistance, β-cell damage, and inflammation[216]
HFDRoseburia, Ruminococcus gnavusBacteroides, AlistipesPromotes the proliferation of beneficial bacteria, blood glucose regulation, and anti-inflammatory effects[217]
Vegetarian and vegan dietsFaecalibacterium prausnitziiBacteroides fragilisEffective weight management, reduction of diabetes and metabolic syndrome risk[220]
Ketogenic DietAkkermansia, Clostridia_UCG 014Bacteroides, Anaerostipes, RuminococcusEffective weight management and promotion of glucose metabolism[221]
RSBifidobacterium adolescentis, Bifidobacterium longum, Ruminococcus bromiiAlisipes putredinis, Bacteroides vulgatus, Odoribacter sp. lanchnicusEffective weight management and promotion of glucose metabolism[222]
MedDiet, Mediterranean diet. TRE, Time-restricted eating. RF, Ramadan fasting. C. morifolium, Chrysanthemum morifolium flower. CDHFD, Cold drink and high-fat diet. HF, High-fat. HS, High-sucrose. HFD, High-fiber diet. RS, resistant starch.

5.2. Supplement Probiotics to Improve DM and Its Complications

Probiotics can improve the balance of gut microbiota [223]. Bifidobacteria, a major component of breast milk, when supplemented in infants, results in abundant indole-3-lactic acid and indole lactic acid in the gut, both of which are linked to immune regulation [224]. Bifidobacterium also produces high concentrations of acetate in the gut lumen, inducing acidification [225]. Probiotic preparations containing Bifidobacterium and INU significantly upregulate the levels of 3-hydroxybutyric acid and glycolic acid [226]. Although clinical studies are limited to statistical analyses of the correlation between microbiota and metabolite levels, Bacteroides thetaiotaomicron significantly increased the proportion of polyunsaturated fatty acids in the liver of mice in a DN animal model [227]. In a mouse model of metabolic-associated fatty liver disease, A. muciniphila was found to regulate the metabolism of L-aspartic acid [228]. In addition, Probiotics have been demonstrated to lower fasting insulin, blood glucose, and homeostatic model assessment of IR levels in patients with GDM, while also enhancing the quantitative insulin sensitivity check index [229].
Probiotics and lactic acid bacteria improve glucose metabolism disorders by increasing the expression of glucose transport proteins and regulating blood glucose and insulin levels [230]. Lactobacillus paracasei IMC 502 alleviates T2DM by modulating SCFAs [231]. Additionally, GPR43/41 and glucagon enhance insulin secretion by activating GLP-1, a process that can be initiated by probiotics [232]. Increasing levels of Bifidobacterium, Lactobacillus, and Akkermansia can improve glucose homeostasis. In HF diet-induced obese mice, elevated fasting blood glucose levels were observed. However, fecal transplantation of P. copri successfully reversed the hyperglycemia [233]. In mice fed an HF and HS diet, the abundance of Bifidobacterium is reduced. Nevertheless, feeding these mice Bifidobacterium longum 070103 fermented dairy products can reverse this condition, leading to significant decrease in fasting blood glucose and leptin levels [234]. Lactobacillus plantarum strain OLL2712 induces IL-10 production by dendritic cells derived from mice. IL-10 strongly promotes the expression of anti-inflammatory macrophages. Additionally, oral administration of OLL2712 to type 2 diabetic KKAy mice inhibits serum pro-inflammatory cytokines [235]. Therefore, the implementation of probiotics-based interventions may regulate blood glucose through intestinal microbiota, metabolites (Figure 6), insulin sensitivity, and other channels to improve DM and its complications [236].

6. Conclusions

This review comprehensively synthesizes evidence on T1DM, T2DM, GDM, and their complications (DR, DKD, DN), including the relationship between the gut microbiota and/or gut-microbiota-related metabolites and IR among them. This review also summarizes how TCM, dietary strategies, and probiotics target host metabolites via the gut microbiota to treat and improve DM and its complications. Future research should focus on interdisciplinary collaboration among nutritionists, microbiologists, and clinicians to develop and implement intervention strategies providing novel approaches for precision nutrition therapies for DM and related metabolic disorders.

Author Contributions

K.Y.: Visualization, Writing—original draft, Writing—review and editing. X.S.: Writing—review and editing. X.W.: Writing—review and editing. J.Z.: Writing—review and editing. H.Y.: Funding acquisition, Writing—review and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Natural Science Foundation Project of China (grant numbers: 82160154, 81670844), the Hundred-level Innovative Talent Foundation of Guizhou Province (grant number: QKH-PTRC-GCC [2023]041), the Training Program Foundation for Young Talents of Zunyi Medical University (grant number: QKH-PTRC-2021-035), the Key Construction Discipline of Immunology and Pathogen Biology in Zhuhai Campus of Zunyi Medical University (grant number: ZHGF2024-1), the Program for Excellent Young Talents of Zunyi Medical University (grant number: 18-ZY-001), and the graduate research fund project of Guizhou Province (grant number: 2024YJSKYJJ338).

Data Availability Statement

No new data were created or analyzed in this study.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

DMDiabetes mellitus
T2DMType 2 diabetes mellitus
T1DMType 1 diabetes mellitus
GDMGestational diabetes mellitus
β-cellsBeta cells
IRInsulin resistance
DRDiabetic retinopathy
DKDDiabetic kidney disease
DNDiabetes neuropathy
DCDDiabetic cardiovascular disease
HFDHigh-fiber diet
HbA1cGlycated hemoglobin
LPSLipopolysaccharides
SCFAsShort-chain fatty acids
BCAAsBranched-chain amino acids
IPAIndolepropionic acid
TCMTraditional Chinese medicine
MRMendelian randomization
A. muciniphilaAkkermansia muciniphila
2-HB2-Hydroxybutyrate
VEGFVascular endothelial growth factor
DPNDiabetic peripheral neuropathy
Lp299vLactobacillus reuteri 299v
R. gnavusRuminococcus gnavus
TMAOTrimethylamine N-oxide
B. coccoidesBlautia coccoides
AGEsAdvanced glycation end-products
RAGEReceptor for advanced glycation end-products
GPRG protein-coupled receptors
HFHigh-fat
NETsNeutrophil extracellular traps
EPAEicosapentaenoic acid
NOD miceNon-obese diabetic mice
PAPropionic acid
BAButyric acid.
MedDietMediterranean diet
C. morifoliumChrysanthemum morifolium flower
HSHigh-sugar
CDHFDCold drink and high-fat diet
TRETime-restricted eating
RFRamadan fasting
FFAFree fatty acids
GABAGamma aminobutyric acid
GLP-1Glucagon-like peptide-1
GIPGlucose-dependent insulin-dependent polypeptide
n-3 PUFAOmega-3 polyunsaturated fatty acids

References

  1. Balooch Hasankhani, M.; Mirzaei, H.; Karamoozian, A. Global trend analysis of diabetes mellitus incidence, mortality, and mortality-to-incidence ratio from 1990 to 2019. Sci. Rep. 2023, 13, 21908. [Google Scholar] [CrossRef]
  2. Sasidharan Pillai, S.; Has, P.; Quintos, J.B.; Serrano Gonzalez, M.; Kasper, V.L.; Topor, L.S.; Fredette, M.E. Incidence, Severity, and Presentation of Type 2 Diabetes in Youth During the First and Second Year of the COVID-19 Pandemic. Diabetes Care 2023, 46, 953–958. [Google Scholar] [CrossRef]
  3. Rathmann, W.; Kuss, O.; Kostev, K. Incidence of newly diagnosed diabetes after COVID-19. Diabetologia 2022, 65, 949–954. [Google Scholar] [CrossRef]
  4. Yang, S.; Cao, J.; Wang, Y.; Chen, Q.; Li, F.; Gao, Y.; Li, R.; Yuan, L. Small Intestinal Endocrine Cell Derived Exosomal ACE2 Protects Islet β-Cell Function by Inhibiting the Activation of NLRP3 Inflammasome and Reducing β-Cell Pyroptosis. Int. J. Nanomed. 2024, 19, 4957–4976. [Google Scholar] [CrossRef]
  5. Kautzky-Willer, A.; Winhofer, Y.; Kiss, H.; Falcone, V.; Berger, A.; Lechleitner, M.; Weitgasser, R.; Harreiter, J. Gestational diabetes mellitus (Update 2023). Wien. Klin. Wochenschr. 2023, 135, 115–128. [Google Scholar] [CrossRef]
  6. Mora, T.; Roche, D.; Rodríguez-Sánchez, B. Predicting the onset of diabetes-related complications after a diabetes diagnosis with machine learning algorithms. Diabetes Res. Clin. Pr. 2023, 204, 110910. [Google Scholar] [CrossRef]
  7. American Diabetes Association Professional Practice Committee. Summary of Revisions: Standards of Care in Diabetes-2025. Diabetes Care 2025, 48, S6–S13. [Google Scholar] [CrossRef]
  8. Holt, R.I.G.; DeVries, J.H.; Hess-Fischl, A.; Hirsch, I.B.; Kirkman, M.S.; Klupa, T.; Ludwig, B.; Nørgaard, K.; Pettus, J.; Renard, E.; et al. The management of type 1 diabetes in adults. A consensus report by the American Diabetes Association (ADA) and the European Association for the Study of Diabetes (EASD). Diabetologia 2021, 64, 2609–2652. [Google Scholar] [CrossRef]
  9. Patiño-Cardona, S.; Garrido-Miguel, M.; Pascual-Morena, C.; Berlanga-Macías, C.; Lucerón-Lucas-Torres, M.; Alfaro-González, S.; Martínez-García, I. Effect of Coenzyme Q10 Supplementation on Lipid and Glycaemic Profiles: An Umbrella Review. J. Cardiovasc. Dev. Dis. 2024, 11, 377. [Google Scholar] [CrossRef]
  10. Wu, Z.; Zhang, B.; Chen, F.; Xia, R.; Zhu, D.; Chen, B.; Lin, A.; Zheng, C.; Hou, D.; Li, X.; et al. Fecal microbiota transplantation reverses insulin resistance in type 2 diabetes: A randomized, controlled, prospective study. Front. Cell. Infect. Microbiol. 2022, 12, 1089991. [Google Scholar] [CrossRef]
  11. Tan, H.; Shi, Y.; Yue, T.; Zheng, D.; Luo, S.; Weng, J.; Zheng, X. Machine learning approach reveals microbiome, metabolome, and lipidome profiles in type 1 diabetes. J. Adv. Res. 2024, 64, 213–221. [Google Scholar] [CrossRef]
  12. Du, E.; Wang, W.; Gan, L.; Li, Z.; Guo, S.; Guo, Y. Effects of thymol and carvacrol supplementation on intestinal integrity and immune responses of broiler chickens challenged with Clostridium perfringens. J. Anim. Sci. Biotechnol. 2016, 7, 19. [Google Scholar] [CrossRef]
  13. Zhao, Z.; Ning, J.; Bao, X.Q.; Shang, M.; Ma, J.; Li, G.; Zhang, D. Fecal microbiota transplantation protects rotenone-induced Parkinson’s disease mice via suppressing inflammation mediated by the lipopolysaccharide-TLR4 signaling pathway through the microbiota-gut-brain axis. Microbiome 2021, 9, 226. [Google Scholar] [CrossRef] [PubMed]
  14. Wang, X.; Zhao, Y.; Wang, D.; Liu, C.; Qi, Z.; Tang, H.; Liu, Y.; Zhang, S.; Cui, Y.; Li, Y.; et al. ALK-JNK signaling promotes NLRP3 inflammasome activation and pyroptosis via NEK7 during Streptococcus pneumoniae infection. Mol. Immunol. 2023, 157, 78–90. [Google Scholar] [CrossRef]
  15. Rinott, E.; Meir, A.Y.; Tsaban, G.; Zelicha, H.; Kaplan, A.; Knights, D.; Tuohy, K.; Scholz, M.U.; Koren, O.; Stampfer, M.J.; et al. The effects of the Green-Mediterranean diet on cardiometabolic health are linked to gut microbiome modifications: A randomized controlled trial. Genome Med. 2022, 14, 29. [Google Scholar] [CrossRef]
  16. Guo, K.; Ye, J.; Li, J.; Huang, J.; Zhou, Z. Effects of gut microbiome on type 1 diabetes susceptibility and complications: A large-scale bidirectional Mendelian randomization and external validation study. Diabetes Obes. Metab. 2024, 26, 3306–3317. [Google Scholar] [CrossRef]
  17. Luo, M.; Sun, M.; Wang, T.; Zhang, S.; Song, X.; Liu, X.; Wei, J.; Chen, Q.; Zhong, T.; Qin, J. Gut microbiota and type 1 diabetes: A two-sample bidirectional Mendelian randomization study. Front. Cell Infect. Microbiol. 2023, 13, 1163898. [Google Scholar] [CrossRef]
  18. Hu, J.; Ding, J.; Li, X.; Li, J.; Zheng, T.; Xie, L.; Li, C.; Tang, Y.; Guo, K.; Huang, J.; et al. Distinct signatures of gut microbiota and metabolites in different types of diabetes: A population-based cross-sectional study. eClinicalMedicine 2023, 62, 102132. [Google Scholar] [CrossRef] [PubMed]
  19. Allakany, A.I.; Elbanna, A.A.; Rohoma, K.H.; Ahmed, S.M.; Ibrahim, A.E.; Fawzy, M.A.; Header, D.A. Study of the gut microbiome in Egyptian patients with type 1 diabetes mellitus. Prz. Gastroenterol. 2023, 18, 190–197. [Google Scholar] [CrossRef] [PubMed]
  20. Chukhlovin, A.B.; Dudurich, V.V.; Kusakin, A.V.; Polev, D.E.; Ermachenko, E.D.; Aseev, M.V.; Zakharov, Y.A.; Eismont, Y.A.; Danilov, L.G.; Glotov, O.S. Evaluation of Gut Microbiota in Healthy Persons and Type 1 Diabetes Mellitus Patients in North-Western Russia. Microorganisms 2023, 11, 1813. [Google Scholar] [CrossRef]
  21. Moreira, L.A.A.; da Paz Lima, L.; de Oliveira Falcão, M.A.; Rosado, E.L. Profile of Gut Microbiota of Adults with Diabetes Mellitus Type 1: A Systematic Review. Curr. Diabetes Rev. 2023, 19, e280322202706. [Google Scholar] [CrossRef]
  22. Hoseini Tavassol, Z.; Ejtahed, H.S.; Atlasi, R.; Saghafian, F.; Khalagi, K.; Hasani-Ranjbar, S.; Siadat, S.D.; Nabipour, I.; Ostovar, A.; Larijani, B. Alteration in Gut Microbiota Composition of Older Adults Is Associated with Obesity and Its Indices: A Systematic Review. J. Nutr. Health Aging 2023, 27, 817–823. [Google Scholar] [CrossRef]
  23. Wang, L.; Gong, C.; Wang, R.; Wang, J.; Yang, Z.; Wang, X. A pilot study on the characterization and correlation of oropharyngeal and intestinal microbiota in children with type 1 diabetes mellitus. Front. Pediatr. 2024, 12, 1382466. [Google Scholar] [CrossRef]
  24. Zhu, B.T. Pathogenic Mechanism of Autoimmune Diabetes Mellitus in Humans: Potential Role of Streptozotocin-Induced Selective Autoimmunity against Human Islet β-Cells. Cells 2022, 11, 492. [Google Scholar] [CrossRef] [PubMed]
  25. Rodrigues, V.F.; Elias-Oliveira, J.; Pereira, Í.S.; Pereira, J.A.; Barbosa, S.C.; Machado, M.S.G.; Guimarães, J.B.; Pacheco, T.C.F.; Bortolucci, J.; Zaramela, L.S.; et al. Akkermansia muciniphila restrains type 1 diabetes onset by eliciting cDC2 and Treg cell differentiation in NOD and STZ-induced experimental models. Life Sci. 2025, 372, 123624. [Google Scholar] [CrossRef]
  26. Spanier, J.A.; Fung, V.; Wardell, C.M.; Alkhatib, M.H.; Chen, Y.; Swanson, L.A.; Dwyer, A.J.; Weno, M.E.; Silva, N.; Mitchell, J.S.; et al. Tregs with an MHC class II peptide-specific chimeric antigen receptor prevent autoimmune diabetes in mice. J. Clin. Investig. 2023, 133, e168601. [Google Scholar] [CrossRef] [PubMed]
  27. Zeng, X.; Ma, C.; Fu, W.; Xu, Y.; Wang, R.; Liu, D.; Zhang, L.; Hu, N.; Li, D.; Li, W. Changes in Type 1 Diabetes-Associated Gut Microbiota Aggravate Brain Ischemia Injury by Affecting Microglial Polarization Via the Butyrate-MyD88 Pathway in Mice. Mol. Neurobiol. 2025, 62, 3764–3780. [Google Scholar] [CrossRef]
  28. Fan, G.; Cao, F.; Kuang, T.; Yi, H.; Zhao, C.; Wang, L.; Peng, J.; Zhuang, Z.; Xu, T.; Luo, Y.; et al. Alterations in the gut virome are associated with type 2 diabetes and diabetic nephropathy. Gut Microbes 2023, 15, 2226925. [Google Scholar] [CrossRef]
  29. Yarmohammadi, H.; Soltanipur, M.; Rezaei, M.; Ejtahed, H.S.; Raei, M.; Razavi, A.; Mirhosseini, S.M.; Zangeneh, M.; Doroud, D.; Fateh, A.; et al. The Comparison of the Gut Microbiome Composition, Serum Inflammatory Markers and Faecal Short-Chain Fatty Acids Among Individuals With Type 1 and 2 Diabetes Mellitus With Healthy Controls: A Case-Control Study. Endocrinol. Diabetes Metab. 2025, 8, e70071. [Google Scholar] [CrossRef] [PubMed]
  30. Letchumanan, G.; Abdullah, N.; Marlini, M.; Baharom, N.; Lawley, B.; Omar, M.R.; Mohideen, F.B.S.; Addnan, F.H.; Nur Fariha, M.M.; Ismail, Z.; et al. Gut Microbiota Composition in Prediabetes and Newly Diagnosed Type 2 Diabetes: A Systematic Review of Observational Studies. Front. Cell Infect. Microbiol. 2022, 12, 943427. [Google Scholar] [CrossRef]
  31. Umirah, F.; Neoh, C.F.; Ramasamy, K.; Lim, S.M. Differential gut microbiota composition between type 2 diabetes mellitus patients and healthy controls: A systematic review. Diabetes Res. Clin. Pr. 2021, 173, 108689. [Google Scholar] [CrossRef]
  32. Alvarez-Silva, C.; Kashani, A.; Hansen, T.H.; Pinna, N.K.; Anjana, R.M.; Dutta, A.; Saxena, S.; Støy, J.; Kampmann, U.; Nielsen, T.; et al. Trans-ethnic gut microbiota signatures of type 2 diabetes in Denmark and India. Genome Med. 2021, 13, 37. [Google Scholar] [CrossRef]
  33. Li, H.; Li, C. Causal relationship between gut microbiota and type 2 diabetes: A two-sample Mendelian randomization study. Front. Microbiol. 2023, 14, 1184734. [Google Scholar] [CrossRef]
  34. Sun, K.; Gao, Y.; Wu, H.; Huang, X. The causal relationship between gut microbiota and type 2 diabetes: A two-sample Mendelian randomized study. Front. Public Health 2023, 11, 1255059. [Google Scholar] [CrossRef]
  35. Neri-Rosario, D.; Martínez-López, Y.E.; Esquivel-Hernández, D.A.; Sánchez-Castañeda, J.P.; Padron-Manrique, C.; Vázquez-Jiménez, A.; Giron-Villalobos, D.; Resendis-Antonio, O. Dysbiosis signatures of gut microbiota and the progression of type 2 diabetes: A machine learning approach in a Mexican cohort. Front. Endocrinol. 2023, 14, 1170459. [Google Scholar] [CrossRef]
  36. Wang, Y.; Xing, X.; Ma, Y.; Fan, Y.; Zhang, Y.; Nan, B.; Li, X.; Wang, Y.; Liu, J. Prevention of High-Fat-Diet-Induced Dyslipidemia by Lactobacillus plantarum LP104 through Mediating Bile Acid Enterohepatic Axis Circulation and Intestinal Flora. J. Agric. Food Chem. 2023, 71, 7334–7347. [Google Scholar] [CrossRef] [PubMed]
  37. Guan, R.; Ma, N.; Liu, G.; Wu, Q.; Su, S.; Wang, J.; Geng, Y. Ethanol extract of propolis regulates type 2 diabetes in mice via metabolism and gut microbiota. J. Ethnopharmacol. 2023, 310, 116385. [Google Scholar] [CrossRef] [PubMed]
  38. Jiang, Y.; Yang, J.; Xia, L.; Wei, T.; Cui, X.; Wang, D.; Jin, Z.; Lin, X.; Li, F.; Yang, K.; et al. Gut Microbiota-Tryptophan Metabolism-GLP-1 Axis Participates in β-Cell Regeneration Induced by Dapagliflozin. Diabetes 2024, 73, 926–940. [Google Scholar] [CrossRef] [PubMed]
  39. Gofron, K.K.; Wasilewski, A.; Małgorzewicz, S. Effects of GLP-1 Analogues and Agonists on the Gut Microbiota: A Systematic Review. Nutrients 2025, 17, 1303. [Google Scholar] [CrossRef]
  40. Sawicki, C.M.; Pacheco, L.S.; Rivas-Tumanyan, S.; Cao, Z.; Haslam, D.E.; Liang, L.; Tucker, K.L.; Joshipura, K.; Bhupathiraju, S.N. Association of Gut Microbiota-Related Metabolites and Type 2 Diabetes in Two Puerto Rican Cohorts. Nutrients 2024, 16, 959. [Google Scholar] [CrossRef]
  41. Hasain, Z.; Raja Ali, R.A.; Ahmad, H.F.; Abdul Rauf, U.F.; Oon, S.F.; Mokhtar, N.M. The Roles of Probiotics in the Gut Microbiota Composition and Metabolic Outcomes in Asymptomatic Post-Gestational Diabetes Women: A Randomized Controlled Trial. Nutrients 2022, 14, 3878. [Google Scholar] [CrossRef]
  42. Yang, J.; Wang, J.; Wu, W.; Su, C.; Wu, Y.; Li, Q. Xylooligosaccharides ameliorate insulin resistance by increasing Akkermansia muciniphila and improving intestinal barrier dysfunction in gestational diabetes mellitus mice. Food Funct. 2024, 15, 3122–3129. [Google Scholar] [CrossRef]
  43. Sweeting, A.; Wong, J.; Murphy, H.R.; Ross, G.P. A Clinical Update on Gestational Diabetes Mellitus. Endocr. Rev. 2022, 43, 763–793. [Google Scholar] [CrossRef]
  44. Juan, J.; Sun, Y.; Wei, Y.; Wang, S.; Song, G.; Yan, J.; Zhou, P.; Yang, H. Progression to type 2 diabetes mellitus after gestational diabetes mellitus diagnosed by IADPSG criteria: Systematic review and meta-analysis. Front. Endocrinol. 2022, 13, 1012244. [Google Scholar] [CrossRef] [PubMed]
  45. Van, J.A.D.; Luo, Y.; Danska, J.S.; Dai, F.; Alexeeff, S.E.; Gunderson, E.P.; Rost, H.; Wheeler, M.B. Postpartum defects in inflammatory response after gestational diabetes precede progression to type 2 diabetes: A nested case-control study within the SWIFT study. Metabolism 2023, 149, 155695. [Google Scholar] [CrossRef]
  46. Liu, N.; Sun, Y.; Wang, Y.; Ma, L.; Zhang, S.; Lin, H. Composition of the intestinal microbiota and its variations between the second and third trimesters in women with gestational diabetes mellitus and without gestational diabetes mellitus. Front. Endocrinol. 2023, 14, 1126572. [Google Scholar] [CrossRef]
  47. Liang, Y.Y.; Liu, L.Y.; Jia, Y.; Li, Y.; Cai, J.N.; Shu, Y.; Tan, J.Y.; Chen, P.Y.; Li, H.W.; Cai, H.H.; et al. Correlation between gut microbiota and glucagon-like peptide-1 in patients with gestational diabetes mellitus. World J. Diabetes 2022, 13, 861–876. [Google Scholar] [CrossRef] [PubMed]
  48. Wang, J.; Xie, Z.; Chen, P.; Wang, Y.; Li, B.; Dai, F. Effect of dietary pattern on pregnant women with gestational diabetes mellitus and its clinical significance. Open Life Sci. 2022, 17, 202–207. [Google Scholar] [CrossRef]
  49. Mei, S.; Chen, Y.; Long, Y.; Cen, X.; Zhao, X.; Zhang, X.; Ye, J.; Gao, X.; Zhu, C. Association of gut microbiota with overweight/obesity combined with gestational diabetes mellitus. J. Med. Microbiol. 2025, 74, 002010. [Google Scholar] [CrossRef] [PubMed]
  50. Wu, X.; Lin, D.; Li, Q.; Cai, J.; Huang, H.; Xiang, T.; Tan, H. Investigating causal associations among gut microbiota, gut microbiota-derived metabolites, and gestational diabetes mellitus: A bidirectional Mendelian randomization study. Aging 2023, 15, 8345–8366. [Google Scholar] [CrossRef]
  51. Liang, W.; Feng, Y.; Yang, D.; Qin, J.; Zhi, X.; Wu, W.; Jie, Q. Oral probiotics increased the proportion of Treg, Tfr, and Breg cells to inhibit the inflammatory response and impede gestational diabetes mellitus. Mol. Med. 2023, 29, 122. [Google Scholar] [CrossRef]
  52. Bae, G.; Berezhnoy, G.; Flores, A.; Cannet, C.; Schäfer, H.; Dahlke, M.H.; Michl, P.; Löffler, M.W.; Königsrainer, A.; Trautwein, C. Quantitative Metabolomics and Lipoprotein Analysis of PDAC Patients Suggests Serum Marker Categories for Pancreatic Function, Pancreatectomy, Cancer Metabolism, and Systemic Disturbances. J. Proteome Res. 2024, 23, 1249–1262. [Google Scholar] [CrossRef] [PubMed]
  53. Qin, F.; Li, J.; Mao, T.; Feng, S.; Li, J.; Lai, M. 2 Hydroxybutyric Acid-Producing Bacteria in Gut Microbiome and Fusobacterium nucleatum Regulates 2 Hydroxybutyric Acid Level In Vivo. Metabolites 2023, 13, 451. [Google Scholar] [CrossRef]
  54. Beldie, L.A.; Dica, C.C.; Moța, M.; Pirvu, B.F.; Burticală, M.A.; Mitrea, A.; Clenciu, D.; Efrem, I.C.; Vladu, B.E.; Timofticiuc, D.C.P.; et al. The Interactions Between Diet and Gut Microbiota in Preventing Gestational Diabetes Mellitus: A Narrative Review. Nutrients 2024, 16, 4131. [Google Scholar] [CrossRef] [PubMed]
  55. Hernández-Teixidó, C.; Barrot de la Puente, J.; Miravet Jiménez, S.; Fernández-Camins, B.; Mauricio, D.; Romero Aroca, P.; Vlacho, B.; Franch-Nadal, J. Incidence of Diabetic Retinopathy in Individuals with Type 2 Diabetes: A Study Using Real-World Data. J. Clin. Med. 2024, 13, 7083. [Google Scholar] [CrossRef] [PubMed]
  56. Bai, J.; Wan, Z.; Zhang, Y.; Wang, T.; Xue, Y.; Peng, Q. Composition and diversity of gut microbiota in diabetic retinopathy. Front. Microbiol. 2022, 13, 926926. [Google Scholar] [CrossRef]
  57. Huang, Y.; Wang, Z.; Ma, H.; Ji, S.; Chen, Z.; Cui, Z.; Chen, J.; Tang, S. Dysbiosis and Implication of the Gut Microbiota in Diabetic Retinopathy. Front. Cell Infect. Microbiol. 2021, 11, 646348. [Google Scholar] [CrossRef]
  58. Zhou, Z.; Zheng, Z.; Xiong, X.; Chen, X.; Peng, J.; Yao, H.; Pu, J.; Chen, Q.; Zheng, M. Gut Microbiota Composition and Fecal Metabolic Profiling in Patients With Diabetic Retinopathy. Front. Cell Dev. Biol. 2021, 9, 732204. [Google Scholar] [CrossRef]
  59. Liu, K.; Zou, J.; Fan, H.; Hu, H.; You, Z. Causal effects of gut microbiota on diabetic retinopathy: A Mendelian randomization study. Front. Immunol. 2022, 13, 930318. [Google Scholar] [CrossRef]
  60. Padakandla, S.R.; Das, T.; Sai Prashanthi, G.; Angadi, K.K.; Reddy, S.S.; Reddy, G.B.; Shivaji, S. Gut mycobiome dysbiosis in rats showing retinal changes indicative of diabetic retinopathy. PLoS ONE 2022, 17, e0267080. [Google Scholar] [CrossRef]
  61. Ye, P.; Zhang, X.; Xu, Y.; Xu, J.; Song, X.; Yao, K. Alterations of the Gut Microbiome and Metabolome in Patients With Proliferative Diabetic Retinopathy. Front. Microbiol. 2021, 12, 667632. [Google Scholar] [CrossRef]
  62. Gu, X.M.; Lu, C.Y.; Pan, J.; Ye, J.Z.; Zhu, Q.H. Alteration of intestinal microbiota is associated with diabetic retinopathy and its severity: Samples collected from southeast coast Chinese. World J. Diabetes 2023, 14, 862–882. [Google Scholar] [CrossRef]
  63. Ai, X.; Yu, P.; Luo, L.; Sun, J.; Tao, H.; Wang, X.; Meng, X. Berberis dictyophylla F. inhibits angiogenesis and apoptosis of diabetic retinopathy via suppressing HIF-1α/VEGF/DLL-4/Notch-1 pathway. J. Ethnopharmacol. 2022, 296, 115453. [Google Scholar] [CrossRef]
  64. Zheng, J.; Chen, M.; Ye, C.; Sun, X.; Jiang, N.; Zou, X.; Yang, H.; Liu, H. BuZangTongLuo decoction improved hindlimb ischemia by activating angiogenesis and regulating gut microbiota in diabetic mice. J. Ethnopharmacol. 2020, 248, 112330. [Google Scholar] [CrossRef]
  65. He, X.; Sun, J.; Liu, C.; Yu, X.; Li, H.; Zhang, W.; Li, Y.; Geng, Y.; Wang, Z. Compositional Alterations of Gut Microbiota in Patients with Diabetic Kidney Disease and Type 2 Diabetes Mellitus. Diabetes Metab. Syndr. Obes. 2022, 15, 755–765. [Google Scholar] [CrossRef]
  66. Han, S.; Chen, M.; Cheng, P.; Zhang, Z.; Lu, Y.; Xu, Y.; Wang, Y. A systematic review and meta-analysis of gut microbiota in diabetic kidney disease: Comparisons with diabetes mellitus, non-diabetic kidney disease, and healthy individuals. Front. Endocrinol. 2022, 13, 1018093. [Google Scholar] [CrossRef]
  67. Zhang, L.; Wang, Z.; Zhang, X.; Zhao, L.; Chu, J.; Li, H.; Sun, W.; Yang, C.; Wang, H.; Dai, W.; et al. Alterations of the Gut Microbiota in Patients with Diabetic Nephropathy. Microbiol. Spectr. 2022, 10, e0032422. [Google Scholar] [CrossRef]
  68. Zhang, B.; Wan, Y.; Zhou, X.; Zhang, H.; Zhao, H.; Ma, L.; Dong, X.; Yan, M.; Zhao, T.; Li, P. Characteristics of Serum Metabolites and Gut Microbiota in Diabetic Kidney Disease. Front. Pharmacol. 2022, 13, 872988. [Google Scholar] [CrossRef] [PubMed]
  69. Hong, J.; Fu, T.; Liu, W.; Du, Y.; Bu, J.; Wei, G.; Yu, M.; Lin, Y.; Min, C.; Lin, D. Jiangtang Decoction Ameliorates Diabetic Kidney Disease Through the Modulation of the Gut Microbiota. Diabetes Metab. Syndr. Obes. Targets Ther. 2023, 16, 3707–3725. [Google Scholar] [CrossRef] [PubMed]
  70. Zhang, Z.; Li, Q.; Liu, F.; Wang, D. Lycoperoside H protects against diabetic nephropathy via alteration of gut microbiota and inflammation. J. Biochem. Mol. Toxicol. 2022, 36, e23216. [Google Scholar] [CrossRef] [PubMed]
  71. Deng, L.; Yang, Y.; Xu, G. Empagliflozin ameliorates type 2 diabetes mellitus-related diabetic nephropathy via altering the gut microbiota. Biochim. Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2022, 1867, 159234. [Google Scholar] [CrossRef]
  72. Noureldein, M.H.; Rumora, A.E.; Teener, S.J.; Rigan, D.M.; Hayes, J.M.; Mendelson, F.E.; Carter, A.D.; Rubin, W.G.; Savelieff, M.G.; Feldman, E.L. Dietary Fatty Acid Composition Alters Gut Microbiome in Mice with Obesity-Induced Peripheral Neuropathy. Nutrients 2025, 17, 737. [Google Scholar] [CrossRef]
  73. Huang, W.; Lin, Z.; Sun, A.; Deng, J.; Manyande, A.; Xiang, H.; Zhao, G.F.; Hong, Q. The role of gut microbiota in diabetic peripheral neuropathy rats with cognitive dysfunction. Front. Microbiol. 2023, 14, 1156591. [Google Scholar] [CrossRef]
  74. Hong, J.; Fu, T.; Liu, W.; Du, Y.; Min, C.; Lin, D. Specific alterations of gut microbiota in diabetic microvascular complications: A systematic review and meta-analysis. Front. Endocrinol. 2022, 13, 1053900. [Google Scholar] [CrossRef]
  75. Tang, F.; Shen, L.; Gu, Z.; Zhang, L.; Fang, L.; Sun, H.; Ma, D.; Guo, Y.; Yang, Y.; Lu, B.; et al. Causal relationships between gut microbiota, gut metabolites, and diabetic neuropathy: A mendelian randomization study. Clin. Nutr. ESPEN 2024, 62, 128–136. [Google Scholar] [CrossRef]
  76. Wu, Y.; Bai, H.; Lu, Y.; Peng, R.; Qian, M.; Yang, X.; Cai, E.; Ruan, W.; Zhang, Q.; Zhang, J.; et al. Associations of Plasma Gut Microbiota-Derived TMAO and Precursors in Early Pregnancy with Gestational Diabetes Mellitus Risk: A Nested Case-Control Study. Nutrients 2025, 17, 810. [Google Scholar] [CrossRef]
  77. Jiang, S.; Shui, Y.; Cui, Y.; Tang, C.; Wang, X.; Qiu, X.; Hu, W.; Fei, L.; Li, Y.; Zhang, S.; et al. Gut microbiota dependent trimethylamine N-oxide aggravates angiotensin II-induced hypertension. Redox Biol. 2021, 46, 102115. [Google Scholar] [CrossRef]
  78. Yutani, M.; Matsumura, T.; Fujinaga, Y. Effects of antibiotics on the viability of and toxin production by Clostridium botulinum. Microbiol. Immunol. 2021, 65, 432–437. [Google Scholar] [CrossRef]
  79. Eck, A.; Rutten, N.; Singendonk, M.M.J.; Rijkers, G.T.; Savelkoul, P.H.M.; Meijssen, C.B.; Crijns, C.E.; Oudshoorn, J.H.; Budding, A.E.; Vlieger, A.M. Neonatal microbiota development and the effect of early life antibiotics are determined by two distinct settler types. PLoS ONE 2020, 15, e0228133. [Google Scholar] [CrossRef]
  80. Kappel, B.A.; De Angelis, L.; Heiser, M.; Ballanti, M.; Stoehr, R.; Goettsch, C.; Mavilio, M.; Artati, A.; Paoluzi, O.A.; Adamski, J.; et al. Cross-omics analysis revealed gut microbiome-related metabolic pathways underlying atherosclerosis development after antibiotics treatment. Mol. Metab. 2020, 36, 100976. [Google Scholar] [CrossRef]
  81. Shi, G.; Lin, Y.; Wu, Y.; Zhou, J.; Cao, L.; Chen, J.; Li, Y.; Tan, N.; Zhong, S. Bacteroides fragilis Supplementation Deteriorated Metabolic Dysfunction, Inflammation, and Aorta Atherosclerosis by Inducing Gut Microbiota Dysbiosis in Animal Model. Nutrients 2022, 14, 2199. [Google Scholar] [CrossRef]
  82. Malik, M.; Suboc, T.M.; Tyagi, S.; Salzman, N.; Wang, J.; Ying, R.; Tanner, M.J.; Kakarla, M.; Baker, J.E.; Widlansky, M.E. Lactobacillus plantarum 299v Supplementation Improves Vascular Endothelial Function and Reduces Inflammatory Biomarkers in Men With Stable Coronary Artery Disease. Circ. Res. 2018, 123, 1091–1102. [Google Scholar] [CrossRef] [PubMed]
  83. Ramirez, Z.E.; Surana, N.K. Ruminococcus gnavus and Limosilactobacillus reuteri Regulate Reg3γ Expression through Multiple Pathways. Immunohorizons 2023, 7, 228–234. [Google Scholar] [CrossRef]
  84. Silverman, G.J.; Deng, J.; Azzouz, D.F. Sex-dependent Lupus Blautia (Ruminococcus) gnavus strain induction of zonulin-mediated intestinal permeability and autoimmunity. Front. Immunol. 2022, 13, 897971. [Google Scholar] [CrossRef]
  85. Hong, J.; Fu, T.; Liu, W.; Du, Y.; Bu, J.; Wei, G.; Yu, M.; Lin, Y.; Min, C.; Lin, D. Specific Alternation of Gut Microbiota and the Role of Ruminococcus gnavus in the Development of Diabetic Nephropathy. J. Microbiol. Biotechnol. 2024, 34, 547–561. [Google Scholar] [CrossRef]
  86. Shen, S.; Ren, F.; Qin, H.; Bukhari, I.; Yang, J.; Gao, D.; Ouwehand, A.C.; Lehtinen, M.J.; Zheng, P.; Mi, Y. Lactobacillus acidophilus NCFM and Lactiplantibacillus plantarum Lp-115 inhibit Helicobacter pylori colonization and gastric inflammation in a murine model. Front. Cell Infect. Microbiol. 2023, 13, 1196084. [Google Scholar] [CrossRef] [PubMed]
  87. Baba, Y.; Saito, Y.; Kadowaki, M.; Azuma, N.; Tsuge, D. Effect of Continuous Ingestion of Bifidobacteria and Inulin on Reducing Body Fat: A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Comparison Study. Nutrients 2023, 15, 5025. [Google Scholar] [CrossRef]
  88. Magryś, A.; Pawlik, M. Postbiotic Fractions of Probiotics Lactobacillus plantarum 299v and Lactobacillus rhamnosus GG Show Immune-Modulating Effects. Cells 2023, 12, 2538. [Google Scholar] [CrossRef] [PubMed]
  89. Panzetta, M.E.; Valdivia, R.H. Akkermansia in the gastrointestinal tract as a modifier of human health. Gut Microbes 2024, 16, 2406379. [Google Scholar] [CrossRef]
  90. Parizadeh, M.; Arrieta, M.C. The global human gut microbiome: Genes, lifestyles, and diet. Trends Mol. Med. 2023, 29, 789–801. [Google Scholar] [CrossRef]
  91. Reis, F.; Ferreira, L.M.R.; Ortega, E.; Viana, S. Nutrition and Gut Microbiota-Immune System Interplay in Chronic Diseases. Nutrients 2025, 17, 1330. [Google Scholar] [CrossRef] [PubMed]
  92. Zheng, S.J.; Luo, Y.; Wang, J.B.; Chen, X.M.; Xu, Y.; Xiao, J.H. Regulated intestinal microbiota and gut immunity to ameliorate type 1 diabetes mellitus: A novel mechanism for stem cell-based therapy. Biomed. Pharmacother. 2024, 170, 116033. [Google Scholar] [CrossRef]
  93. Wang, Z.; Gong, M.; Fang, Y.; Yuan, H.; Zhang, C. Reconstruction characteristics of gut microbiota from patients with type 1 diabetes affect the phenotypic reproducibility of glucose metabolism in mice. Sci. China Life Sci. 2025, 68, 176–188. [Google Scholar] [CrossRef]
  94. Zhang, Y.; Liu, R.; Chen, Y.; Cao, Z.; Liu, C.; Bao, R.; Wang, Y.; Huang, S.; Pan, S.; Qin, L.; et al. Akkermansia muciniphila supplementation in patients with overweight/obese type 2 diabetes: Efficacy depends on its baseline levels in the gut. Cell Metab. 2025, 37, 592–605.e6. [Google Scholar] [CrossRef] [PubMed]
  95. Donati Zeppa, S.; Gervasi, M.; Bartolacci, A.; Ferrini, F.; Patti, A.; Sestili, P.; Stocchi, V.; Agostini, D. Targeting the Gut Microbiota for Prevention and Management of Type 2 Diabetes. Nutrients 2024, 16, 3951. [Google Scholar] [CrossRef]
  96. Wang, Y.; Ye, X.; Ding, D.; Lu, Y. Characteristics of the intestinal flora in patients with peripheral neuropathy associated with type 2 diabetes. J. Int. Med. Res. 2020, 48, 300060520936806. [Google Scholar] [CrossRef]
  97. González, A.; Fullaondo, A.; Odriozola, A. In Search of Healthy Ageing: A Microbiome-Based Precision Nutrition Approach for Type 2 Diabetes Prevention. Nutrients 2025, 17, 1877. [Google Scholar] [CrossRef]
  98. Han, C.; Shen, Z.; Cui, T.; Ai, S.S.; Gao, R.R.; Liu, Y.; Sui, G.Y.; Hu, H.Z.; Li, W. Yi-Shen-Hua-Shi granule ameliorates diabetic kidney disease by the “gut-kidney axis”. J. Ethnopharmacol. 2023, 307, 116257. [Google Scholar] [CrossRef] [PubMed]
  99. Guo, Q.; Gao, Z.; Zhao, L.; Wang, H.; Luo, Z.; Vandeputte, D.; He, L.; Li, M.; Di, S.; Liu, Y.; et al. Multiomics Analyses With Stool-Type Stratification in Patient Cohorts and Blautia Identification as a Potential Bacterial Modulator in Type 2 Diabetes Mellitus. Diabetes 2024, 73, 511–527. [Google Scholar] [CrossRef]
  100. Chen, D.Q.; Zhang, H.J.; Zhang, W.; Feng, K.; Liu, H.; Zhao, H.L.; Li, P. Tangshen Formula alleviates inflammatory injury against aged diabetic kidney disease through modulating gut microbiota composition and related amino acid metabolism. Exp. Gerontol. 2024, 188, 112393. [Google Scholar] [CrossRef] [PubMed]
  101. Jiang, S.Q.; Ye, S.N.; Huang, Y.H.; Ou, Y.W.; Chen, K.Y.; Chen, J.S.; Tang, S.B. Gut microbiota induced abnormal amino acids and their correlation with diabetic retinopathy. Int. J. Ophthalmol. 2024, 17, 883–895. [Google Scholar] [CrossRef]
  102. Lee, Y.; Kim, A.H.; Kim, E.; Lee, S.; Yu, K.S.; Jang, I.J.; Chung, J.Y.; Cho, J.Y. Changes in the gut microbiome influence the hypoglycemic effect of metformin through the altered metabolism of branched-chain and nonessential amino acids. Diabetes Res. Clin. Pract. 2021, 178, 108985. [Google Scholar] [CrossRef]
  103. Chen, Y.; Song, L.; Chen, M.; Huang, Y.; Wang, Z.; Ren, Z.; Xu, J. Pediococcus pentosaceus MIANGUAN2 Alleviates Influenza Virus Infection by Modulating Gut Microbiota and Enhancing Short-Chain Fatty Acid Production. Nutrients 2024, 16, 1923. [Google Scholar] [CrossRef] [PubMed]
  104. Zheng, X.; Xu, X.; Liu, M.; Yang, J.; Yuan, M.; Sun, C.; Zhou, Q.; Chen, J.; Liu, B. Bile acid and short chain fatty acid metabolism of gut microbiota mediate high-fat diet induced intestinal barrier damage in Macrobrachium rosenbergii. Fish. Shellfish. Immunol. 2024, 146, 109376. [Google Scholar] [CrossRef]
  105. Li, N.; Wang, H.; Zhao, H.; Wang, M.; Cai, J.; Hao, Y.; Yu, J.; Jiang, Y.; Lü, X.; Liu, B. Cooperative interactions between Veillonella ratti and Lactobacillus acidophilus ameliorate DSS-induced ulcerative colitis in mice. Food Funct. 2023, 14, 10475–10492. [Google Scholar] [CrossRef]
  106. Zheng, X.X.; Li, D.X.; Li, Y.T.; Chen, Y.L.; Zhao, Y.L.; Ji, S.; Guo, M.Z.; Du, Y.; Tang, D.Q. Mulberry leaf water extract alleviates type 2 diabetes in mice via modulating gut microbiota-host co-metabolism of branched-chain amino acid. Phytother. Res. PTR 2023, 37, 3195–3210. [Google Scholar] [CrossRef]
  107. Yoshida, N.; Yamashita, T.; Osone, T.; Hosooka, T.; Shinohara, M.; Kitahama, S.; Sasaki, K.; Sasaki, D.; Yoneshiro, T.; Suzuki, T.; et al. Bacteroides spp. promotes branched-chain amino acid catabolism in brown fat and inhibits obesity. iScience 2021, 24, 103342. [Google Scholar] [CrossRef] [PubMed]
  108. Gao, H.; Sun, M.; Li, A.; Gu, Q.; Kang, D.; Feng, Z.; Li, X.; Wang, X.; Chen, L.; Yang, H.; et al. Microbiota-derived IPA alleviates intestinal mucosal inflammation through upregulating Th1/Th17 cell apoptosis in inflammatory bowel disease. Gut Microbes 2025, 17, 2467235. [Google Scholar] [CrossRef]
  109. Wang, Z.; Yang, S.; Liu, L.; Mao, A.; Kan, H.; Yu, F.; Ma, X.; Feng, L.; Zhou, T. The gut microbiota-derived metabolite indole-3-propionic acid enhances leptin sensitivity by targeting STAT3 against diet-induced obesity. Clin. Transl. Med. 2024, 14, e70053. [Google Scholar] [CrossRef]
  110. Kong, L.; Zhao, Q.; Jiang, X.; Hu, J.; Jiang, Q.; Sheng, L.; Peng, X.; Wang, S.; Chen, Y.; Wan, Y.; et al. Trimethylamine N-oxide impairs β-cell function and glucose tolerance. Nat. Commun. 2024, 15, 2526. [Google Scholar] [CrossRef] [PubMed]
  111. Li, D.; Lu, Y.; Yuan, S.; Cai, X.; He, Y.; Chen, J.; Wu, Q.; He, D.; Fang, A.; Bo, Y.; et al. Gut microbiota-derived metabolite trimethylamine-N-oxide and multiple health outcomes: An umbrella review and updated meta-analysis. Am. J. Clin. Nutr. 2022, 116, 230–243. [Google Scholar] [CrossRef]
  112. Jin, Q.; Zhang, C.; Chen, R.; Jiang, L.; Li, H.; Wu, P.; Li, L. Quinic acid regulated TMA/TMAO-related lipid metabolism and vascular endothelial function through gut microbiota to inhibit atherosclerotic. J. Transl. Med. 2024, 22, 352. [Google Scholar] [CrossRef]
  113. Zhen, J.; Zhang, Y.; Li, Y.; Zhou, Y.; Cai, Y.; Huang, G.; Xu, A. The gut microbiota intervenes in glucose tolerance and inflammation by regulating the biosynthesis of taurodeoxycholic acid and carnosine. Front. Cell Infect. Microbiol. 2024, 14, 1423662. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, Y.; Li, L.; Chai, T.; Xu, H.; Du, H.Y.; Jiang, Y. Mulberry leaf multi-components exert hypoglycemic effects through regulation of the PI-3K/Akt insulin signaling pathway in type 2 diabetic rats. J. Ethnopharmacol. 2024, 319, 117307. [Google Scholar] [CrossRef] [PubMed]
  115. Bodur, C.; Kazyken, D.; Huang, K.; Tooley, A.S.; Cho, K.W.; Barnes, T.M.; Lumeng, C.N.; Myers, M.G.; Fingar, D.C. TBK1-mTOR Signaling Attenuates Obesity-Linked Hyperglycemia and Insulin Resistance. Diabetes 2022, 71, 2297–2312. [Google Scholar] [CrossRef]
  116. Wang, H.; Pan, F.; Liu, J.; Zhang, J.; Fuli, Z.; Wang, Y. Huayuwendan decoction ameliorates inflammation via IL-17/NF-κB signaling pathway in diabetic rats. J. Ethnopharmacol. 2024, 319, 117328. [Google Scholar] [CrossRef] [PubMed]
  117. Zhao, H.; Zhang, F.; Sun, D.; Wang, X.; Zhang, X.; Zhang, J.; Yan, F.; Huang, C.; Xie, H.; Lin, C.; et al. Branched-Chain Amino Acids Exacerbate Obesity-Related Hepatic Glucose and Lipid Metabolic Disorders via Attenuating Akt2 Signaling. Diabetes 2020, 69, 1164–1177. [Google Scholar] [CrossRef]
  118. Huang, H.; Chen, H.; Yao, Y.; Lou, X. Branched-chain amino acids supplementation induces insulin resistance and pro-inflammatory macrophage polarization via INFGR1/JAK1/STAT1 signal pathway. Mol. Med. 2024, 30, 149. [Google Scholar] [CrossRef]
  119. Li, Y.; Xiong, Z.; Yan, W.; Gao, E.; Cheng, H.; Wu, G.; Liu, Y.; Zhang, L.; Li, C.; Wang, S.; et al. Branched chain amino acids exacerbate myocardial ischemia/reperfusion vulnerability via enhancing GCN2/ATF6/PPAR-α pathway-dependent fatty acid oxidation. Theranostics 2020, 10, 5623–5640. [Google Scholar] [CrossRef]
  120. Eguchi, A.; Iwasa, M.; Tamai, Y.; Tempaku, M.; Takamatsu, S.; Miyoshi, E.; Hasegawa, H.; Kobayashi, Y.; Takei, Y. Branched-chain amino acids protect the liver from cirrhotic injury via suppression of activation of lipopolysaccharide-binding protein, toll-like receptor 4, and signal transducer and activator of transcription 3, as well as Enterococcus faecalis translocation. Nutrition 2021, 86, 111194. [Google Scholar] [CrossRef]
  121. Zheng, H.; Zhang, X.; Li, C.; Wang, D.; Shen, Y.; Lu, J.; Zhao, L.; Li, X.; Gao, H. BCAA mediated microbiota-liver-heart crosstalk regulates diabetic cardiomyopathy via FGF21. Microbiome 2024, 12, 157. [Google Scholar] [CrossRef]
  122. Miao, M.; Wang, Q.; Wang, X.; Fan, C.; Luan, T.; Yan, L.; Zhang, Y.; Zeng, X.; Dai, Y.; Li, P. The Protective Effects of Inulin-Type Fructans Against High-Fat/Sucrose Diet-Induced Gestational Diabetes Mice in Association With Gut Microbiota Regulation. Front. Microbiol. 2022, 13, 832151. [Google Scholar] [CrossRef]
  123. Giampieri, F.; Mazzoni, L.; Cianciosi, D.; Alvarez-Suarez, J.M.; Regolo, L.; Sánchez-González, C.; Capocasa, F.; Xiao, J.; Mezzetti, B.; Battino, M. Organic vs conventional plant-based foods: A review. Food Chem. 2022, 383, 132352. [Google Scholar] [CrossRef]
  124. Saikachain, N.; Sungkaworn, T.; Muanprasat, C.; Asavapanumas, N. Neuroprotective effect of short-chain fatty acids against oxidative stress-induced SH-SY5Y injury via GPR43-dependent pathway. J. Neurochem. 2023, 166, 201–214. [Google Scholar] [CrossRef]
  125. Zhao, Z.; Tong, Y.; Kang, Y.; Qiu, Z.; Li, Q.; Xu, C.; Wu, G.; Jia, W.; Wang, P. Sodium butyrate (SB) ameliorated inflammation of COPD induced by cigarette smoke through activating the GPR43 to inhibit NF-κB/MAPKs signaling pathways. Mol. Immunol. 2023, 163, 224–234. [Google Scholar] [CrossRef]
  126. Yi, C.; Sun, W.; Ding, L.; Yan, M.; Sun, C.; Qiu, C.; Wang, D.; Wu, L. Short-Chain Fatty Acids Weaken Ox-LDL-Induced Cell Inflammatory Injury by Inhibiting the NLRP3/Caspase-1 Pathway and Affecting Cellular Metabolism in THP-1 Cells. Molecules 2022, 27, 8801. [Google Scholar] [CrossRef]
  127. Xia, T.; He, W.; Luo, Z.; Wang, K.; Tan, X. Achyranthes bidentata polysaccharide ameliorates type 2 diabetes mellitus by gut microbiota-derived short-chain fatty acids-induced activation of the GLP-1/GLP-1R/cAMP/PKA/CREB/INS pathway. Int. J. Biol. Macromol. 2024, 270, 132256. [Google Scholar] [CrossRef] [PubMed]
  128. Chen, H.; Wang, S.H.; Li, H.L.; Zhou, X.B.; Zhou, L.W.; Chen, C.; Mansell, T.; Novakovic, B.; Saffery, R.; Baker, P.N.; et al. The attenuation of gut microbiota-derived short-chain fatty acids elevates lipid transportation through suppression of the intestinal HDAC3-H3K27ac-PPAR-γ axis in gestational diabetes mellitus. J. Nutr. Biochem. 2024, 133, 109708. [Google Scholar] [CrossRef] [PubMed]
  129. Tayyeb, J.Z.; Popeijus, H.E.; Mensink, R.P.; Konings, M.; Mokhtar, F.B.A.; Plat, J. Short-Chain Fatty Acids (Except Hexanoic Acid) Lower NF-kB Transactivation, Which Rescues Inflammation-Induced Decreased Apolipoprotein A-I Transcription in HepG2 Cells. Int. J. Mol. Sci. 2020, 21, 5088. [Google Scholar] [CrossRef] [PubMed]
  130. Tian, X.; Zeng, Y.; Tu, Q.; Jiao, Y.; Yao, S.; Chen, Y.; Sun, L.; Xia, Q.; Luo, Y.; Yuan, L.; et al. Butyrate alleviates renal fibrosis in CKD by regulating NLRP3-mediated pyroptosis via the STING/NF-κB/p65 pathway. Int. Immunopharmacol. 2023, 124, 111010. [Google Scholar] [CrossRef]
  131. Tillett, B.J.; Dwiyanto, J.; Secombe, K.R.; George, T.; Zhang, V.; Anderson, D.; Duggan, E.; Giri, R.; Loo, D.; Stoll, T.; et al. SCFA biotherapy delays diabetes in humanized gnotobiotic mice by remodeling mucosal homeostasis and metabolome. Nat. Commun. 2025, 16, 2893. [Google Scholar] [CrossRef]
  132. Liu, Z.; Dai, X.; Zhang, H.; Shi, R.; Hui, Y.; Jin, X.; Zhang, W.; Wang, L.; Wang, Q.; Wang, D.; et al. Gut microbiota mediates intermittent-fasting alleviation of diabetes-induced cognitive impairment. Nat. Commun. 2020, 11, 855. [Google Scholar] [CrossRef]
  133. Chen, L.; Yang, Y.; Sun, S.; Xie, Y.; Pan, C.; Li, M.; Li, C.; Liu, Y.; Xu, Z.; Liu, W.; et al. Indolepropionic acid reduces obesity-induced metabolic dysfunction through colonic barrier restoration mediated via tuft cell-derived IL-25. FEBS J. 2022, 289, 5985–6004. [Google Scholar] [CrossRef]
  134. Yao, W.; Huo, J.; Liu, K.; Tao, P. Exploring the beneficial effect of gut microbiota metabolites on diabetic nephropathy via network pharmacology study. Sci. Rep. 2025, 15, 11027. [Google Scholar] [CrossRef]
  135. Lee, H.; Park, S.; Ju, S.; Kim, S.; Yoo, J.W.; Yoon, I.S.; Min, D.S.; Jung, Y. Preparation and Evaluation of Colon-Targeted Prodrugs of the Microbial Metabolite 3-Indolepropionic Acid as an Anticolitic Agent. Mol. Pharm. 2021, 18, 1730–1741. [Google Scholar] [CrossRef]
  136. Zeng, Y.; Guo, M.; Wu, Q.; Tan, X.; Jiang, C.; Teng, F.; Chen, J.; Zhang, F.; Ma, X.; Li, X.; et al. Gut microbiota-derived indole-3-propionic acid alleviates diabetic kidney disease through its mitochondrial protective effect via reducing ubiquitination mediated-degradation of SIRT1. J. Adv. Res. 2025, 73, 607–630. [Google Scholar] [CrossRef]
  137. Rybka, M.; Mazurek, Ł.; Jurak, J.; Laskowska, A.; Zajdel, M.; Czuwara, J.; Sulejczak, D.; Szudzik, M.; Samborowska, E.; Schwartz, R.A.; et al. Keratin-TMAO dressing accelerates full-thickness skin wound healing in diabetic rats via M2-macrophage polarization and the activation of PI3K/AKT/mTOR signaling pathway. Int. J. Biol. Macromol. 2025, 310, 143313. [Google Scholar] [CrossRef]
  138. Lin, X.; Zhang, Y.; He, X.; Chen, Y.; Chen, N.; Liu, J.; Wang, M.; Li, Y.; Yang, H.; Fan, L.; et al. The Choline Metabolite TMAO Inhibits NETosis and Promotes Placental Development in GDM of Humans and Mice. Diabetes 2021, 70, 2250–2263. [Google Scholar] [CrossRef] [PubMed]
  139. Sung, M.; Lim, S.; Park, S.; Choi, Y.; Kim, S. Anti-inflammatory effects of phytosphingosine-regulated cytokines and NF-kB and MAPK mechanism. Cell. Mol. Biol. 2024, 70, 22–30. [Google Scholar] [CrossRef] [PubMed]
  140. Won, J.H.; Shin, J.S.; Park, H.J.; Jung, H.J.; Koh, D.J.; Jo, B.G.; Lee, J.Y.; Yun, K.; Lee, K.T. Anti-inflammatory effects of madecassic acid via the suppression of NF-kappaB pathway in LPS-induced RAW 264.7 macrophage cells. Planta Med. 2010, 76, 251–257. [Google Scholar] [CrossRef] [PubMed]
  141. Das, A.K.; Hossain, U.; Ghosh, S.; Biswas, S.; Mandal, M.; Mandal, B.; Brahmachari, G.; Bagchi, A.; Sil, P.C. Amelioration of oxidative stress mediated inflammation and apoptosis in pancreatic islets by Lupeol in STZ-induced hyperglycaemic mice. Life Sci. 2022, 305, 120769. [Google Scholar] [CrossRef]
  142. Liu, Y.; Deng, J.; Fan, D. Ginsenoside Rk3 ameliorates high-fat-diet/streptozocin induced type 2 diabetes mellitus in mice via the AMPK/Akt signaling pathway. Food Funct. 2019, 10, 2538–2551. [Google Scholar] [CrossRef] [PubMed]
  143. Zhang, C.; Yu, H.; Ye, J.; Tong, H.; Wang, M.; Sun, G. Ginsenoside Rg3 Protects against Diabetic Cardiomyopathy and Promotes Adiponectin Signaling via Activation of PPAR-γ. Int. J. Mol. Sci. 2023, 24, 16736. [Google Scholar] [CrossRef]
  144. Wang, W.; Guan, F.; Sagratini, G.; Yan, J.; Xie, J.; Jin, Z.; Liu, M.; Liu, H.; Liu, J. Ginsenoside Rd attenuated hyperglycemia via Akt pathway and modulated gut microbiota in streptozotocin-induced diabetic rats. Curr. Res. Food Sci. 2023, 6, 100491. [Google Scholar] [CrossRef]
  145. Zhang, X.; Wang, L.; Guo, R.; Xiao, J.; Liu, X.; Dong, M.; Luan, X.; Ji, X.; Lu, H. Ginsenoside Rb1 Ameliorates Diabetic Arterial Stiffening via AMPK Pathway. Front. Pharmacol. 2021, 12, 753881. [Google Scholar] [CrossRef]
  146. Zhu, Y.; Yang, H.; Deng, J.; Fan, D. Ginsenoside Rg5 Improves Insulin Resistance and Mitochondrial Biogenesis of Liver via Regulation of the Sirt1/PGC-1α Signaling Pathway in db/db Mice. J. Agric. Food Chem. 2021, 69, 8428–8439. [Google Scholar] [CrossRef]
  147. Sun, Y.; Wang, J.; Guo, X.; Zhu, N.; Niu, L.; Ding, X.; Xie, Z.; Chen, X.; Yang, F. Oleic Acid and Eicosapentaenoic Acid Reverse Palmitic Acid-induced Insulin Resistance in Human HepG2 Cells via the Reactive Oxygen Species/JUN Pathway. Genom. Proteom. Bioinform. 2021, 19, 754–771. [Google Scholar] [CrossRef]
  148. Zang, T.; Chen, H.; Shen, S.; Xu, F.; Wang, R.; Yin, J.; Chen, X.; Guan, M.; Shen, L.; Pan, H.; et al. Highly Purified Eicosapentaenoic Acid Alleviates the Inflammatory Response and Oxidative Stress in Macrophages during Atherosclerosis via the miR-1a-3p/sFRP1/Wnt/PCP-JNK Pathway. Oxidative Med. Cell. Longev. 2022, 2022, 9451058. [Google Scholar] [CrossRef] [PubMed]
  149. Kim, N.; Kang, M.S.; Nam, M.; Kim, S.A.; Hwang, G.S.; Kim, H.S. Eicosapentaenoic Acid (EPA) Modulates Glucose Metabolism by Targeting AMP-Activated Protein Kinase (AMPK) Pathway. Int. J. Mol. Sci. 2019, 20, 4751. [Google Scholar] [CrossRef]
  150. Simón, M.V.; Agnolazza, D.L.; German, O.L.; Garelli, A.; Politi, L.E.; Agbaga, M.P.; Anderson, R.E.; Rotstein, N.P. Synthesis of docosahexaenoic acid from eicosapentaenoic acid in retina neurons protects photoreceptors from oxidative stress. J. Neurochem. 2016, 136, 931–946. [Google Scholar] [CrossRef] [PubMed]
  151. Liu, J.; Wei, Y.; Jia, W.; Can, C.; Wang, R.; Yang, X.; Gu, C.; Liu, F.; Ji, C.; Ma, D. Chenodeoxycholic acid suppresses AML progression through promoting lipid peroxidation via ROS/p38 MAPK/DGAT1 pathway and inhibiting M2 macrophage polarization. Redox Biol. 2022, 56, 102452. [Google Scholar] [CrossRef]
  152. Song, M.; Ye, J.; Zhang, F.; Su, H.; Yang, X.; He, H.; Liu, F.; Zhu, X.; Wang, L.; Gao, P.; et al. Chenodeoxycholic Acid (CDCA) Protects against the Lipopolysaccharide-Induced Impairment of the Intestinal Epithelial Barrier Function via the FXR-MLCK Pathway. J. Agric. Food Chem. 2019, 67, 8868–8874. [Google Scholar] [CrossRef]
  153. Zhou, J.C.; Wu, B.; Zhang, J.J.; Zhang, W. Lupeol triggers oxidative stress, ferroptosis, apoptosis and restrains inflammation in nasopharyngeal carcinoma via AMPK/NF-κB pathway. Immunopharmacol. Immunotoxicol. 2022, 44, 621–631. [Google Scholar] [CrossRef]
  154. Qin, D.; Pan, P.; Lyu, B.; Chen, W.; Gao, Y. Lupeol improves bile acid metabolism and metabolic dysfunction-associated steatotic liver disease in mice via FXR signaling pathway and gut-liver axis. Biomed. Pharmacother. 2024, 177, 116942. [Google Scholar] [CrossRef] [PubMed]
  155. Tian, Y.; Jing, G.; Yin, R.; Ma, M.; Cao, W.; Zhang, M. Neuroprotective effects of traditional Chinese medicine Naofucong on diabetic cognitive impairment: Mechanisms involving insulin-degrading enzyme-mediated degradation of Amyloid-β and inhibition of ERK/JNK/p38 MAPK signaling pathway. Brain Res. 2025, 1849, 149365. [Google Scholar] [CrossRef]
  156. Wang, Y.; Shen, Y.; Lu, S.; Wu, J. EVOO supplement prevents type 1 diabetes by modulating gut microbiota and serum metabolites in NOD mice. Life Sci. 2023, 335, 122274. [Google Scholar] [CrossRef]
  157. Ismail, H.M.; Spall, M.; Evans-Molina, C.; DiMeglio, L.A. Evaluating the effect of prebiotics on the gut microbiome profile and β cell function in youth with newly diagnosed type 1 diabetes: Protocol of a pilot randomized controlled trial. Pilot. Feasibility Stud. 2023, 9, 150. [Google Scholar] [CrossRef]
  158. Yang, B.; Xiong, Z.; Lin, M.; Yang, Y.; Chen, Y.; Zeng, J.; Jia, X.; Feng, L. Astragalus polysaccharides alleviate type 1 diabetes via modulating gut microbiota in mice. Int. J. Biol. Macromol. 2023, 234, 123767. [Google Scholar] [CrossRef]
  159. Lo Conte, M.; Antonini Cencicchio, M.; Ulaszewska, M.; Nobili, A.; Cosorich, I.; Ferrarese, R.; Massimino, L.; Andolfo, A.; Ungaro, F.; Mancini, N.; et al. A diet enriched in omega-3 PUFA and inulin prevents type 1 diabetes by restoring gut barrier integrity and immune homeostasis in NOD mice. Front. Immunol. 2022, 13, 1089987. [Google Scholar] [CrossRef] [PubMed]
  160. Zhang, X.; Li, Q.; Han, N.; Song, C.; Lin, Y.; Zhang, L.; Ren, D.; Zhao, Y.; Yang, X.; Li, T. Effects of Fu brick tea polysaccharides on gut microbiota and fecal metabolites of HFD/STZ-induced type 2 diabetes rats. Food Funct. 2023, 14, 10910–10923. [Google Scholar] [CrossRef] [PubMed]
  161. Liu, T.; Zhao, M.; Zhang, Y.; Xu, R.; Fu, Z.; Jin, T.; Song, J.; Huang, Y.; Wang, M.; Zhao, C. Polysaccharides from Phellinus linteus attenuate type 2 diabetes mellitus in rats via modulation of gut microbiota and bile acid metabolism. Int. J. Biol. Macromol. 2024, 262, 130062. [Google Scholar] [CrossRef]
  162. Wang, L.; Liang, C.; Song, X.; Jia, X.; Wang, X.; Zhang, Y.; Xie, Q.; Zheng, N.; Yuan, H. Canagliflozin alters the gut, oral, and ocular surface microbiota of patients with type 2 diabetes mellitus. Front. Endocrinol. 2023, 14, 1256292. [Google Scholar] [CrossRef]
  163. Sun, Y.; Qu, H.; Niu, X.; Li, T.; Wang, L.; Peng, H. Carvacrol improves blood lipid and glucose in rats with type 2 diabetes mellitus by regulating short-chain fatty acids and the GPR41/43 pathway. Korean J. Physiol. Pharmacol. 2024, 28, 1–10. [Google Scholar] [CrossRef]
  164. Huang, S.; Chen, J.; Cui, Z.; Ma, K.; Wu, D.; Luo, J.; Li, F.; Xiong, W.; Rao, S.; Xiang, Q.; et al. Lachnospiraceae-derived butyrate mediates protection of high fermentable fiber against placental inflammation in gestational diabetes mellitus. Sci. Adv. 2023, 9, eadi7337. [Google Scholar] [CrossRef] [PubMed]
  165. Zhao, H.; Wu, H.; Duan, M.; Liu, R.; Zhu, Q.; Zhang, K.; Wang, L. Cinnamaldehyde Improves Metabolic Functions in Streptozotocin-Induced Diabetic Mice by Regulating Gut Microbiota. Drug Des. Dev. Ther. 2021, 15, 2339–2355. [Google Scholar] [CrossRef]
  166. Du, Y.; Zhang, R.; Zheng, X.X.; Zhao, Y.L.; Chen, Y.L.; Ji, S.; Guo, M.Z.; Tang, D.Q. Mulberry (Morus alba L.) leaf water extract attenuates type 2 diabetes mellitus by regulating gut microbiota dysbiosis, lipopolysaccharide elevation and endocannabinoid system disorder. J. Ethnopharmacol. 2024, 323, 117681. [Google Scholar] [CrossRef] [PubMed]
  167. Tawulie, D.; Jin, L.; Shang, X.; Li, Y.; Sun, L.; Xie, H.; Zhao, J.; Liao, J.; Zhu, Z.; Cui, H.; et al. Jiang-Tang-San-Huang pill alleviates type 2 diabetes mellitus through modulating the gut microbiota and bile acids metabolism. Phytomed. Int. J. Phytother. Phytopharm. 2023, 113, 154733. [Google Scholar] [CrossRef] [PubMed]
  168. Chen, X.; Chen, C.; Fu, X. Dendrobium officinale Polysaccharide Alleviates Type 2 Diabetes Mellitus by Restoring Gut Microbiota and Repairing Intestinal Barrier via the LPS/TLR4/TRIF/NF-kB Axis. J. Agric. Food Chem. 2023, 71, 11929–11940. [Google Scholar] [CrossRef]
  169. Meng, X.; Shi, M.; Guo, G.; Xing, J.; Liu, Z.; Song, F.; Liu, S. In-depth investigation of the therapeutic effect of Tribulus terrestris L. on type 2 diabetes based on intestinal microbiota and feces metabolomics. J. Ethnopharmacol. 2024, 325, 117815. [Google Scholar] [CrossRef]
  170. Du, C.; Zuo, F.; Cao, Y.; Zang, Y. Anti-diabetic effects of natural and modified ‘Ganzhou’ navel orange peel pectin on type 2 diabetic mice via gut microbiota. Food Funct. 2023, 14, 10977–10990. [Google Scholar] [CrossRef]
  171. Pi, Y.; Fang, M.; Li, Y.; Cai, L.; Han, R.; Sun, W.; Jiang, X.; Chen, L.; Du, J.; Zhu, Z.; et al. Interactions between Gut Microbiota and Natural Bioactive Polysaccharides in Metabolic Diseases: Review. Nutrients 2024, 16, 2838. [Google Scholar] [CrossRef]
  172. Xu, H.; Liu, Z.; Xu, W.; Zhang, Y. Beneficial In Vitro Effects of Polysaccharide and Non-Polysaccharide Components of Dendrobium huoshanense on Gut Microbiota of Rats with Type 1 Diabetes as Opposed to Metformin. Molecules 2024, 29, 2791. [Google Scholar] [CrossRef]
  173. Siddiqui, N.Z.; Rehman, A.U.; Yousuf, W.; Khan, A.I.; Farooqui, N.A.; Zang, S.; Xin, Y.; Wang, L. Effect of crude polysaccharide from seaweed, Dictyopteris divaricata (CDDP) on gut microbiota restoration and anti-diabetic activity in streptozotocin (STZ)-induced T1DM mice. Gut Pathog. 2022, 14, 39. [Google Scholar] [CrossRef]
  174. Ye, J.; Ma, J.; Rozi, P.; Kong, L.; Zhou, J.; Luo, Y.; Yang, H. The polysaccharides from seeds of Glycyrrhiza uralensis ameliorate metabolic disorders and restructure gut microbiota in type 2 diabetic mice. Int. J. Biol. Macromol. 2024, 264, 130622. [Google Scholar] [CrossRef]
  175. Deng, J.; Luo, K.; Xia, C.; Zhu, Y.; Xiang, Z.; Zhu, B.; Tang, X.; Zhang, T.; Shi, L.; Lyu, X.; et al. Phytochemical composition of Tibetan tea fermented by Eurotium cristatum and its effects on type 1 diabetes mice and gut microbiota. Heliyon 2024, 10, e27145. [Google Scholar] [CrossRef] [PubMed]
  176. Wu, C.; Pan, L.L.; Niu, W.; Fang, X.; Liang, W.; Li, J.; Li, H.; Pan, X.; Chen, W.; Zhang, H.; et al. Modulation of Gut Microbiota by Low Methoxyl Pectin Attenuates Type 1 Diabetes in Non-obese Diabetic Mice. Front. Immunol. 2019, 10, 1733. [Google Scholar] [CrossRef] [PubMed]
  177. Liao, H.; Zhao, Y.; Liang, Y.; Zou, K. Flavonoids Derived from Opuntia ficus-indica Fruit Alleviate Renal Injury in Diabetic Nephropathy Mice by Altering Gut Microbiota and Promoting the Production of SCFAs. Nutrients 2025, 17, 1800. [Google Scholar] [CrossRef] [PubMed]
  178. Di, S.; Yao, C.; Qiao, L.; Li, X.; Pang, B.; Lin, J.; Wang, J.; Li, M.; Tong, X. Exploration of the mechanisms underlying the beneficial effect of Luo Tong formula on retinal function in diabetic rats via the “gut microbiota-inflammation-retina” axis. Chin. Med. 2022, 17, 133. [Google Scholar] [CrossRef]
  179. Liu, Y.; Gong, Y.; Li, M.; Li, J. Quercetin protects against hyperglycemia-induced retinopathy in Sprague Dawley rats by regulating the gut-retina axis and nuclear factor erythroid-2-related factor 2 pathway. Nutr. Res. 2024, 122, 55–67. [Google Scholar] [CrossRef]
  180. Gao, Y.; Yang, R.; Guo, L.; Wang, Y.; Liu, W.J.; Ai, S.; Woon, T.H.; Wang, Z.; Zhai, Y.; Wang, Z.; et al. Qing-Re-Xiao-Zheng Formula Modulates Gut Microbiota and Inhibits Inflammation in Mice With Diabetic Kidney Disease. Front. Med. 2021, 8, 719950. [Google Scholar] [CrossRef]
  181. Lan, T.; Tang, T.; Li, Y.; Duan, Y.; Yuan, Q.; Liu, W.; Ren, Y.; Li, N.; Liu, X.; Zhang, Y.; et al. FTZ polysaccharides ameliorate kidney injury in diabetic mice by regulating gut-kidney axis. Phytomedicine 2023, 118, 154935. [Google Scholar] [CrossRef]
  182. Zhang, M.; Yang, L.; Zhu, M.; Yang, B.; Yang, Y.; Jia, X.; Feng, L. Moutan Cortex polysaccharide ameliorates diabetic kidney disease via modulating gut microbiota dynamically in rats. Int. J. Biol. Macromol. 2022, 206, 849–860. [Google Scholar] [CrossRef] [PubMed]
  183. Pengrattanachot, N.; Thongnak, L.; Promsan, S.; Phengpol, N.; Sutthasupha, P.; Tocharus, J.; Lungkaphin, A. Fructooligosaccharides Ameliorate Renal Injury and Dysfunction Through the Modulation of Gut Dysbiosis, Inhibition of Renal Inflammation, Oxidative Stress, Fibrosis, and Improve Organic Anion Transporter 3 Function in an Obese Rat Model. Mol. Nutr. Food Res. 2024, 68, e2400191. [Google Scholar] [CrossRef]
  184. Shen, Z.; Cui, T.; Liu, Y.; Wu, S.; Han, C.; Li, J. Astragalus membranaceus and Salvia miltiorrhiza ameliorate diabetic kidney disease via the “gut-kidney axis”. Phytomed. Int. J. Phytother. Phytopharm. 2023, 121, 155129. [Google Scholar] [CrossRef]
  185. Luo, L.; Luo, J.; Cai, Y.; Fu, M.; Li, W.; Shi, L.; Liu, J.; Dong, R.; Xu, X.; Tu, L.; et al. Inulin-type fructans change the gut microbiota and prevent the development of diabetic nephropathy. Pharmacol. Res. 2022, 183, 106367. [Google Scholar] [CrossRef]
  186. Dong, W.; Zhao, Y.; Li, X.; Huo, J.; Wang, W. Corn silk polysaccharides attenuate diabetic nephropathy through restoration of the gut microbial ecosystem and metabolic homeostasis. Front. Endocrinol. 2023, 14, 1232132. [Google Scholar] [CrossRef] [PubMed]
  187. Su, X.; Yu, W.; Liu, A.; Wang, C.; Li, X.; Gao, J.; Liu, X.; Jiang, W.; Yang, Y.; Lv, S. San-Huang-Yi-Shen Capsule Ameliorates Diabetic Nephropathy in Rats Through Modulating the Gut Microbiota and Overall Metabolism. Front. Pharmacol. 2021, 12, 808867. [Google Scholar] [CrossRef] [PubMed]
  188. Zhu, N.; Duan, H.; Feng, Y.; Xu, W.; Shen, J.; Wang, K.; Liu, J. Magnesium lithospermate B ameliorates diabetic nephropathy by suppressing the uremic toxin formation mediated by gut microbiota. Eur. J. Pharmacol. 2023, 953, 175812. [Google Scholar] [CrossRef]
  189. Xie, J.; Song, W.; Liang, X.; Zhang, Q.; Shi, Y.; Liu, W.; Shi, X. Jinmaitong ameliorates diabetic peripheral neuropathy in streptozotocin-induced diabetic rats by modulating gut microbiota and neuregulin 1. Aging 2020, 12, 17436–17458. [Google Scholar] [CrossRef]
  190. Xie, J.; Song, W.; Liang, X.; Zhang, Q.; Shi, Y.; Liu, W.; Shi, X. Protective effect of quercetin on streptozotocin-induced diabetic peripheral neuropathy rats through modulating gut microbiota and reactive oxygen species level. Biomed. Pharmacother. 2020, 127, 110147. [Google Scholar] [CrossRef]
  191. Shen, C.L.; Wang, R.; Santos, J.M.; Elmassry, M.M.; Stephens, E.; Kim, N.; Neugebauer, V. Ginger alleviates mechanical hypersensitivity and anxio-depressive behavior in rats with diabetic neuropathy through beneficial actions on gut microbiome composition, mitochondria, and neuroimmune cells of colon and spinal cord. Nutr. Res. 2024, 124, 73–84. [Google Scholar] [CrossRef]
  192. Li, J.; Yang, S.; Liu, D.; Yan, Q.; Guo, H.; Jiang, Z. Neoagarotetraose Alleviates Atherosclerosis via Modulating Cholesterol and Bile Acid Metabolism in ApoE−/− Mice. Nutrients 2024, 16, 1502. [Google Scholar] [CrossRef]
  193. Zhang, K.; Peng, P.; Huang, J.; Chen, M.; Liu, F.; Zhu, C.; Lu, Q.; Wang, M.; Lin, C. Integrating plasma metabolomics and gut microbiome to reveal the mechanisms of Huangqi Guizhi Wuwu Decoction intervene diabetic peripheral neuropathy. J. Ethnopharmacol. 2024, 319, 117301. [Google Scholar] [CrossRef]
  194. Pan, L.; Yu, H.; Fu, J.; Hu, J.; Xu, H.; Zhang, Z.; Bu, M.; Yang, X.; Zhang, H.; Lu, J.; et al. Berberine ameliorates chronic kidney disease through inhibiting the production of gut-derived uremic toxins in the gut microbiota. Acta Pharm. Sinica. B 2023, 13, 1537–1553. [Google Scholar] [CrossRef]
  195. Eppig, J.T.; Blake, J.A.; Bult, C.J.; Kadin, J.A.; Richardson, J.E. The Mouse Genome Database (MGD): Facilitating mouse as a model for human biology and disease. Nucleic Acids Res. 2015, 43, D726–D736. [Google Scholar] [CrossRef] [PubMed]
  196. Kachapati, K.; Adams, D.; Bednar, K.; Ridgway, W.M. The non-obese diabetic (NOD) mouse as a model of human type 1 diabetes. Methods Mol. Biol. 2012, 933, 3–16. [Google Scholar] [CrossRef]
  197. Sharma, K.; McCue, P.; Dunn, S.R. Diabetic kidney disease in the db/db mouse. Am. J. Physiol. Ren. Physiol. 2003, 284, F1138–F1144. [Google Scholar] [CrossRef] [PubMed]
  198. Liu, B.; Wei, Y.; He, J.; Feng, B.; Chen, Y.; Guo, R.; Griffin, M.D.; Hynes, S.O.; Shen, S.; Liu, Y.; et al. Human umbilical cord-derived mesenchymal stromal cells improve myocardial fibrosis and restore miRNA-133a expression in diabetic cardiomyopathy. Stem Cell Res. Ther. 2024, 15, 120. [Google Scholar] [CrossRef]
  199. Takaichi, S.; Tomimaru, Y.; Akagi, T.; Kobayashi, S.; Fukuda, Y.; Toya, K.; Asaoka, T.; Iwagami, Y.; Yamada, D.; Akita, H.; et al. Three-dimensional Vascularized β-cell Spheroid Tissue Derived From Human Induced Pluripotent Stem Cells for Subcutaneous Islet Transplantation in a Mouse Model of Type 1 Diabetes. Transplantation 2022, 106, 48–59. [Google Scholar] [CrossRef] [PubMed]
  200. Zhang, J.; Wu, Y.; Tian, Y.; Xu, H.; Lin, Z.X.; Xian, Y.F. Chinese herbal medicine for the treatment of intestinal cancer: Preclinical studies and potential clinical applications. Mol. Cancer 2024, 23, 217. [Google Scholar] [CrossRef]
  201. Dickman, K.G.; Chen, C.H.; Grollman, A.P.; Pu, Y.S. Aristolochic acid-containing Chinese herbal medicine and upper urinary tract urothelial carcinoma in Taiwan: A narrative review. World J. Urol. 2023, 41, 899–907. [Google Scholar] [CrossRef] [PubMed]
  202. Chen, Y.K.; Liu, T.T.; Teia, F.K.F.; Xie, M.Z. Exploring the underlying mechanisms of obesity and diabetes and the potential of Traditional Chinese Medicine: An overview of the literature. Front. Endocrinol. 2023, 14, 1218880. [Google Scholar] [CrossRef]
  203. Luo, W.; Zhou, J.; Yang, X.; Wu, R.; Liu, H.; Shao, H.; Huang, B.; Kang, X.; Yang, L.; Liu, D. A Chinese medical nutrition therapy diet accompanied by intermittent energy restriction alleviates type 2 diabetes by enhancing pancreatic islet function and regulating gut microbiota composition. Food Res. Int. 2022, 161, 111744. [Google Scholar] [CrossRef]
  204. Song, Q.; Wang, Y.; Huang, L.; Shen, M.; Yu, Y.; Yu, Q.; Chen, Y.; Xie, J. Review of the relationships among polysaccharides, gut microbiota, and human health. Food Res. Int. 2021, 140, 109858. [Google Scholar] [CrossRef] [PubMed]
  205. Coutiño-Hernández, D.; Sánchez-Tapia, M.; Leal-Vega, F.; Bobadilla Del Valle, M.; Ledezma, H.; Cervantes, R.; Pedraza-Chaverri, J.; Granados-Portillo, O.; Díaz, D.; Antunes-Ricardo, M.; et al. Modulation of gut microbiota by Mantequilla and Melipona honeys decrease low-grade inflammation caused by high fructose corn syrup or sucrose in rats. Food Res. Int. 2022, 151, 110856. [Google Scholar] [CrossRef] [PubMed]
  206. González, I.; Morales, M.A.; Rojas, A. Polyphenols and AGEs/RAGE axis. Trends and challenges. Food Res. Int. 2020, 129, 108843. [Google Scholar] [CrossRef]
  207. Barber, T.M.; Kabisch, S.; Pfeiffer, A.F.H.; Weickert, M.O. The Effects of the Mediterranean Diet on Health and Gut Microbiota. Nutrients 2023, 15, 2150. [Google Scholar] [CrossRef]
  208. García-Gavilán, J.F.; Atzeni, A.; Babio, N.; Liang, L.; Belzer, C.; Vioque, J.; Corella, D.; Fitó, M.; Vidal, J.; Moreno-Indias, I.; et al. Effect of 1-year lifestyle intervention with energy-reduced Mediterranean diet and physical activity promotion on the gut metabolome and microbiota: A randomized clinical trial. Am. J. Clin. Nutr. 2024, 119, 1143–1154. [Google Scholar] [CrossRef]
  209. Sato, S.; Chinda, D.; Iino, C.; Sawada, K.; Mikami, T.; Nakaji, S.; Sakuraba, H.; Fukuda, S. A Cohort Study of the Influence of the 12-Component Modified Japanese Diet Index on Oral and Gut Microbiota in the Japanese General Population. Nutrients 2024, 16, 524. [Google Scholar] [CrossRef]
  210. Pieczyńska-Zając, J.M.; Malinowska, A.; Łagowska, K.; Leciejewska, N.; Bajerska, J. The effects of time-restricted eating and Ramadan fasting on gut microbiota composition: A systematic review of human and animal studies. Nutr. Rev. 2024, 82, 777–793. [Google Scholar] [CrossRef]
  211. Liu, D.; Zhan, J.; Wang, S.; Chen, L.; Zhu, Q.; Nie, R.; Zhou, X.; Zheng, W.; Luo, X.; Wang, B.; et al. Chrysanthemum morifolium attenuates metabolic and alcohol-associated liver disease via gut microbiota and PPARα/γ activation. Phytomed. Int. J. Phytother. Phytopharm. 2024, 130, 155774. [Google Scholar] [CrossRef] [PubMed]
  212. Zhang, X.; Hu, H.; Zhang, Y.; Hu, S.; Lu, J.; Peng, W.; Luo, D. Dietary Capsaicin Exacerbates Gut Microbiota Dysbiosis and Mental Disorders in Type 1 Diabetes Mice. Nutrients 2025, 17, 593. [Google Scholar] [CrossRef] [PubMed]
  213. Tian, Y.; Fu, M.; Su, J.; Yan, M.; Yu, J.; Wang, C.; Niu, Z.; Du, Y.; Hu, X.; Zheng, J.; et al. Gut microbiota dysbiosis and intestinal barrier impairment in diarrhea caused by cold drink and high-fat diet. Toxicology 2024, 502, 153728. [Google Scholar] [CrossRef] [PubMed]
  214. Liu, Q.; Wang, Y.; Wan, Y.; Liang, Y.; Tan, Y.; Wei, M.; Hou, T. Selenium- and/or Zinc-Enriched Egg Diet Improves Oxidative Damage and Regulates Gut Microbiota in D-Gal-Induced Aging Mice. Nutrients 2024, 16, 512. [Google Scholar] [CrossRef]
  215. Zhai, L.; Huang, C.; Ning, Z.; Zhang, Y.; Zhuang, M.; Yang, W.; Wang, X.; Wang, J.; Zhang, L.; Xiao, H.; et al. Ruminococcus gnavus plays a pathogenic role in diarrhea-predominant irritable bowel syndrome by increasing serotonin biosynthesis. Cell Host Microbe 2023, 31, 33–44.e5. [Google Scholar] [CrossRef]
  216. Aggarwal, H.; Gautam, J.; Kumari, D.; Gupta, S.K.; Bajpai, S.; Chaturvedi, K.; Kumar, Y.; Dikshit, M. Comparative profiling of gut microbiota and metabolome in diet-induced obese and insulin-resistant C57BL/6J mice. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2024, 1871, 119643. [Google Scholar] [CrossRef]
  217. Igudesman, D.; Crandell, J.L.; Corbin, K.D.; Hooper, J.; Thomas, J.M.; Bulik, C.M.; Pence, B.W.; Pratley, R.E.; Kosorok, M.R.; Maahs, D.M.; et al. Associations of Dietary Intake with the Intestinal Microbiota and Short-Chain Fatty Acids Among Young Adults with Type 1 Diabetes and Overweight or Obesity. J. Nutr. 2023, 153, 1178–1188. [Google Scholar] [CrossRef]
  218. Zhang, W.; Zhang, N.; Guo, X.; Fan, B.; Cheng, S.; Wang, F. Potato Resistant Starch Type 1 Promotes Obesity Linked with Modified Gut Microbiota in High-Fat Diet-Fed Mice. Molecules 2024, 29, 370. [Google Scholar] [CrossRef]
  219. Schwartz, L.T.; Ladouceur, J.G.; Russell, M.M.; Xie, S.Y.L.; Bu, S.; Kerver, J.M.; Comstock, S.S. The Relationship Between Fiber Intake and Gut Bacterial Diversity and Composition During the Third Trimester of Pregnancy. Nutrients 2025, 17, 773. [Google Scholar] [CrossRef]
  220. Kahleova, H.; Rembert, E.; Alwarith, J.; Yonas, W.N.; Tura, A.; Holubkov, R.; Agnello, M.; Chutkan, R.; Barnard, N.D. Effects of a Low-Fat Vegan Diet on Gut Microbiota in Overweight Individuals and Relationships with Body Weight, Body Composition, and Insulin Sensitivity. A Randomized Clinical Trial. Nutrients 2020, 12, 2917. [Google Scholar] [CrossRef]
  221. Deledda, A.; Palmas, V.; Heidrich, V.; Fosci, M.; Lombardo, M.; Cambarau, G.; Lai, A.; Melis, M.; Loi, E.; Loviselli, A.; et al. Dynamics of Gut Microbiota and Clinical Variables after Ketogenic and Mediterranean Diets in Drug-Naïve Patients with Type 2 Diabetes Mellitus and Obesity. Metabolites 2022, 12, 1092. [Google Scholar] [CrossRef]
  222. Li, H.; Zhang, L.; Li, J.; Wu, Q.; Qian, L.; He, J.; Ni, Y.; Kovatcheva-Datchary, P.; Yuan, R.; Liu, S.; et al. Resistant starch intake facilitates weight loss in humans by reshaping the gut microbiota. Nat. Metab. 2024, 6, 578–597. [Google Scholar] [CrossRef] [PubMed]
  223. Zikou, E.; Dovrolis, N.; Dimosthenopoulos, C.; Gazouli, M.; Makrilakis, K. The Effect of Probiotic Supplements on Metabolic Parameters of People with Type 2 Diabetes in Greece-A Randomized, Double-Blind, Placebo-Controlled Study. Nutrients 2023, 15, 4663. [Google Scholar] [CrossRef] [PubMed]
  224. Henrick, B.M.; Rodriguez, L.; Lakshmikanth, T.; Pou, C.; Henckel, E.; Arzoomand, A.; Olin, A.; Wang, J.; Mikes, J.; Tan, Z.; et al. Bifidobacteria-mediated immune system imprinting early in life. Cell 2021, 184, 3884–3898.e11. [Google Scholar] [CrossRef]
  225. Odenwald, M.A.; Lin, H.; Lehmann, C.; Dylla, N.P.; Cole, C.G.; Mostad, J.D.; Pappas, T.E.; Ramaswamy, R.; Moran, A.; Hutchison, A.L.; et al. Bifidobacteria metabolize lactulose to optimize gut metabolites and prevent systemic infection in patients with liver disease. Nat. Microbiol. 2023, 8, 2033–2049. [Google Scholar] [CrossRef]
  226. Biggio, F.; Fattuoni, C.; Mostallino, M.C.; Follesa, P. Effects of Chronic Bifidobacteria Administration in Adult Male Rats on Plasma Metabolites: A Preliminary Metabolomic Study. Metabolites 2022, 12, 762. [Google Scholar] [CrossRef]
  227. Li, H.; Wang, X.K.; Tang, M.; Lei, L.; Li, J.R.; Sun, H.; Jiang, J.; Dong, B.; Li, H.Y.; Jiang, J.D.; et al. Bacteroides thetaiotaomicron ameliorates mouse hepatic steatosis through regulating gut microbial composition, gut-liver folate and unsaturated fatty acids metabolism. Gut Microbes 2024, 16, 2304159. [Google Scholar] [CrossRef] [PubMed]
  228. Rao, Y.; Kuang, Z.; Li, C.; Guo, S.; Xu, Y.; Zhao, D.; Hu, Y.; Song, B.; Jiang, Z.; Ge, Z.; et al. Gut Akkermansia muciniphila ameliorates metabolic dysfunction-associated fatty liver disease by regulating the metabolism of L-aspartate via gut-liver axis. Gut Microbes 2021, 13, 1927633. [Google Scholar] [CrossRef]
  229. Lan, X.; Li, B.; Zhao, J.; Stanton, C.; Ross, R.P.; Chen, W.; Yang, B. Probiotic intervention improves metabolic outcomes in gestational diabetes mellitus: A meta-analysis of randomized controlled trials. Clin. Nutr. 2024, 43, 1683–1695. [Google Scholar] [CrossRef]
  230. Song, H.; Xue, H.; Zhang, Z.; Wang, J.; Li, A.; Zhang, J.; Luo, P.; Zhan, M.; Zhou, X.; Chen, L.; et al. Amelioration of Type 2 Diabetes Using Four Strains of Lactobacillus Probiotics: Effects on Gut Microbiota Reconstitution-Mediated Regulation of Glucose Homeostasis, Inflammation, and Oxidative Stress in Mice. J. Agric. Food Chem. 2023, 71, 20801–20814. [Google Scholar] [CrossRef]
  231. Gu, Y.; Chen, H.; Li, X.; Li, D.; Sun, Y.; Yang, L.; Ma, Y.; Chan, E.C.Y. Lactobacillus paracasei IMC 502 ameliorates type 2 diabetes by mediating gut microbiota-SCFA-hormone/inflammation pathway in mice. J. Sci. Food Agric. 2023, 103, 2949–2959. [Google Scholar] [CrossRef] [PubMed]
  232. Wang, Y.; Dilidaxi, D.; Wu, Y.; Sailike, J.; Sun, X.; Nabi, X.H. Composite probiotics alleviate type 2 diabetes by regulating intestinal microbiota and inducing GLP-1 secretion in db/db mice. Biomed. Pharmacother. 2020, 125, 109914. [Google Scholar] [CrossRef] [PubMed]
  233. Gong, J.; Zhang, Q.; Hu, R.; Yang, X.; Fang, C.; Yao, L.; Lv, J.; Wang, L.; Shi, M.; Zhang, W.; et al. Effects of Prevotella copri on insulin, gut microbiota and bile acids. Gut Microbes 2024, 16, 2340487. [Google Scholar] [CrossRef] [PubMed]
  234. Jiang, T.; Li, Y.; Li, L.; Liang, T.; Du, M.; Yang, L.; Yang, J.; Yang, R.; Zhao, H.; Chen, M.; et al. Bifidobacterium longum 070103 Fermented Milk Improve Glucose and Lipid Metabolism Disorders by Regulating Gut Microbiota in Mice. Nutrients 2022, 14, 4050. [Google Scholar] [CrossRef]
  235. Toshimitsu, T.; Mochizuki, J.; Ikegami, S.; Itou, H. Identification of a Lactobacillus plantarum strain that ameliorates chronic inflammation and metabolic disorders in obese and type 2 diabetic mice. J. Dairy Sci. 2016, 99, 933–946. [Google Scholar] [CrossRef]
  236. Dimba, N.R.; Mzimela, N.; Sosibo, A.M.; Khathi, A. Effectiveness of Prebiotics and Mediterranean and Plant-Based Diet on Gut Microbiota and Glycemic Control in Patients with Prediabetes or Type 2 Diabetes: A Systematic Review and Meta-Analysis. Nutrients 2024, 16, 3272. [Google Scholar] [CrossRef]
Figure 1. The gut microbiota in different types of DM regulates blood glucose levels by modulating the expression of metabolites. The differences in microbial community abundance, as shown in the figure, may be key factors for distinguishing between types of DM. These microbial communities influence the host’s metabolic profile, affecting glucose and lipid metabolism, and contributing to the onset and progression of the disease. T1DM, Type 1 diabetes mellitus. T2DM, Type 2 diabetes mellitus. GDM, Gestational diabetes mellitus. SCFAs, Short-chain fatty acids. BCAAs, Branched-chain amino acids. FFA, Free fatty acid. 2-HB, 2-Hydroxybutyrate. DAG, Diacylglycerol. AGEs, Glycation end-products. GABA, Gamma aminobutyric acid. GLP-1, Glucagon-like peptide-1. GIP, Glucose-dependent insulin-dependent polypeptide.
Figure 1. The gut microbiota in different types of DM regulates blood glucose levels by modulating the expression of metabolites. The differences in microbial community abundance, as shown in the figure, may be key factors for distinguishing between types of DM. These microbial communities influence the host’s metabolic profile, affecting glucose and lipid metabolism, and contributing to the onset and progression of the disease. T1DM, Type 1 diabetes mellitus. T2DM, Type 2 diabetes mellitus. GDM, Gestational diabetes mellitus. SCFAs, Short-chain fatty acids. BCAAs, Branched-chain amino acids. FFA, Free fatty acid. 2-HB, 2-Hydroxybutyrate. DAG, Diacylglycerol. AGEs, Glycation end-products. GABA, Gamma aminobutyric acid. GLP-1, Glucagon-like peptide-1. GIP, Glucose-dependent insulin-dependent polypeptide.
Nutrients 17 02603 g001
Figure 2. The gut microbiota plays a key role in the synthesis and metabolism of metabolites. As shown in the figure, the microbial community regulates the levels of specific metabolites, targeting one or more metabolites to influence the host’s metabolic balance. SCFAs, Short-chain fatty acids. BCAAs, Branched-chain amino acids.
Figure 2. The gut microbiota plays a key role in the synthesis and metabolism of metabolites. As shown in the figure, the microbial community regulates the levels of specific metabolites, targeting one or more metabolites to influence the host’s metabolic balance. SCFAs, Short-chain fatty acids. BCAAs, Branched-chain amino acids.
Nutrients 17 02603 g002
Figure 3. Metabolites’ involvement in DM and its complications through the regulation of signaling pathways. Metabolites such as BCAAs, ginsenosides, and IPA modulate signaling pathways including AMPK, Akt, and ROS, thereby influencing the onset and progression of various diseases. BCAAs, Branched-chain amino acids. G-Rk3, Ginsenoside Rk3. EPA, Eicosapentaenoic Acid. IPA, Indolepropionic acid. G-Rg5, Ginsenoside Rg5. G-RD, Ginsenoside Rd. G-Rg3, Ginsenoside Rg3. G-Rb1, Ginsenoside Rb1.
Figure 3. Metabolites’ involvement in DM and its complications through the regulation of signaling pathways. Metabolites such as BCAAs, ginsenosides, and IPA modulate signaling pathways including AMPK, Akt, and ROS, thereby influencing the onset and progression of various diseases. BCAAs, Branched-chain amino acids. G-Rk3, Ginsenoside Rk3. EPA, Eicosapentaenoic Acid. IPA, Indolepropionic acid. G-Rg5, Ginsenoside Rg5. G-RD, Ginsenoside Rd. G-Rg3, Ginsenoside Rg3. G-Rb1, Ginsenoside Rb1.
Nutrients 17 02603 g003
Figure 4. Metabolites like SCFAs, EPA, leucine, and lupeol play significant roles in the development of DM and its complications by regulating key signaling pathways, including NF-κB, AMPK, and mTOR. These pathways are essential for maintaining metabolic homeostasis. EPA, Eicosapentaenoic Acid. SCFAs, short-chain fatty acids.
Figure 4. Metabolites like SCFAs, EPA, leucine, and lupeol play significant roles in the development of DM and its complications by regulating key signaling pathways, including NF-κB, AMPK, and mTOR. These pathways are essential for maintaining metabolic homeostasis. EPA, Eicosapentaenoic Acid. SCFAs, short-chain fatty acids.
Nutrients 17 02603 g004
Figure 5. Different dietary structures and eating habits have a regulatory effect on the gut microbiota. A diet rich in vegetables, high in fiber, and low in sugar can promote the abundance of beneficial bacteria, while an HF diet can increase the abundance of pathogenic bacteria. MedDiet, Mediterranean diet. TRE, Time-restricted eating. RF, Ramadan fasting. CDHFD, Cold drink and high-fat diet. HF, High-fat. HS, High-sucrose.
Figure 5. Different dietary structures and eating habits have a regulatory effect on the gut microbiota. A diet rich in vegetables, high in fiber, and low in sugar can promote the abundance of beneficial bacteria, while an HF diet can increase the abundance of pathogenic bacteria. MedDiet, Mediterranean diet. TRE, Time-restricted eating. RF, Ramadan fasting. CDHFD, Cold drink and high-fat diet. HF, High-fat. HS, High-sucrose.
Nutrients 17 02603 g005
Figure 6. Different dietary structures affect the gut microbiota and metabolism and regulate blood glucose. In the diagram, arrows represent regulatory effects, and different background colors represent the direct impact of dietary structure on the gut microbiota. Reasonable diet on the left side and balanced gut, blood glucose homeostasis. In contrast, the right side indicates that poor dietary habits lead to gut homeostasis imbalance and opportunistic pathogens. Subsequently, under the influence of the microbiota, the body’s metabolism changes its function, thereby participating in blood glucose regulation. LPSs, Lipopolysaccharides. SCFAs, Short-chain fatty acids. BCAAs, Branched-chain amino acids. FFAs, Free fatty acids. GABA, Gamma aminobutyric acid. IPA, Indolepropionic acid. TMAO, Trimethylamine N-oxide. MedDiet, Mediterranean diet. TRE, Time-restricted eating. CDHFD, Cold drink and high-fat diet. HF, High-fat. HS, High-sucrose. HFD, High-fiber diet. RS, resistant starch. n-3 PUFA, Omega-3 polyunsaturated fatty acids.
Figure 6. Different dietary structures affect the gut microbiota and metabolism and regulate blood glucose. In the diagram, arrows represent regulatory effects, and different background colors represent the direct impact of dietary structure on the gut microbiota. Reasonable diet on the left side and balanced gut, blood glucose homeostasis. In contrast, the right side indicates that poor dietary habits lead to gut homeostasis imbalance and opportunistic pathogens. Subsequently, under the influence of the microbiota, the body’s metabolism changes its function, thereby participating in blood glucose regulation. LPSs, Lipopolysaccharides. SCFAs, Short-chain fatty acids. BCAAs, Branched-chain amino acids. FFAs, Free fatty acids. GABA, Gamma aminobutyric acid. IPA, Indolepropionic acid. TMAO, Trimethylamine N-oxide. MedDiet, Mediterranean diet. TRE, Time-restricted eating. CDHFD, Cold drink and high-fat diet. HF, High-fat. HS, High-sucrose. HFD, High-fiber diet. RS, resistant starch. n-3 PUFA, Omega-3 polyunsaturated fatty acids.
Nutrients 17 02603 g006
Table 2. The potential roles and regulatory signaling pathways of Key metabolites in DM and its complications.
Table 2. The potential roles and regulatory signaling pathways of Key metabolites in DM and its complications.
MetaboliteRegulating Signal PathwaysFunctionReferences
BCAAsAkt2Promoting insulin secretion and β-cell dysfunction, dual inflammatory regulation[117,118,119,121]
INFGR1/JAK1/STAT1
GCN2/ATF6/PPAR-α
mTOR
SCFAsGPR 43/NF-kappaB/MAPKEnhancing insulin sensitivity, β-cell function, lowering blood glucose, and anti-inflammatory effects[124,125,126,127,128,130]
NLRP3/Caspase-1
STING/NF-κB/p65
GLP-1/GLP-1R/cAMP/PKA/CREB/INS
HDAC3-H3K27ac-PPAR-γ
IPAGPR109AMaintain intestinal homeostasis, enhance insulin secretion, and reduce insulin resistance[132,135,136]
AHR/NF-κB
SIRT1/PGC-1α
PXR
TMAOPI3K/Akt/mTORPromoting inflammation, inhibiting insulin signaling suppression, and damaging β-cells[137,138]
MAPK/NF-κB
PERK-FoxO1
PhytosphingosineMAPKImproving metabolic disorders in diabetes and anti-inflammatory effects[139]
NF-κB
Madecassic acidNF-κBAnti-inflammatory effects and regulating lipid metabolism[140]
GinsenosidesAMPK/AktRegulating hepatic glucose metabolism, alleviating inflammation and oxidative stress[142,143,146]
PPAR-γ
Sirt1/PGC-1α
EPAROS/JUNIncreasing insulin sensitivity, promoting pancreatic β-cell function, and anti-inflammatory effects[147,148,149]
miR-1a-3p/sFRP1/Wnt/PCP-JNK
AMPK
Chenodeoxycholic acidROS/p38 MAPK/DGAT1Improving glucose and lipid metabolism, protecting pancreatic β-cell function[151,152]
FXR-MLCK
LupeolAMPK/NF-κBAntioxidant, anti-inflammatory, and protective effects on pancreatic β-cells[153,154]
FXR
BCAAs, Branched-chain amino acids. SCFAs, Short-chain fatty acids. IPA, Indolepropionic acid. TMAO, Trimethylamine N-oxide. EPA, Eicosapentaenoic acid.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yan, K.; Sun, X.; Wang, X.; Zheng, J.; Yu, H. Gut Microbiota and Metabolites: Biomarkers and Therapeutic Targets for Diabetes Mellitus and Its Complications. Nutrients 2025, 17, 2603. https://doi.org/10.3390/nu17162603

AMA Style

Yan K, Sun X, Wang X, Zheng J, Yu H. Gut Microbiota and Metabolites: Biomarkers and Therapeutic Targets for Diabetes Mellitus and Its Complications. Nutrients. 2025; 17(16):2603. https://doi.org/10.3390/nu17162603

Chicago/Turabian Style

Yan, Kai, Xin Sun, Xin Wang, Jing Zheng, and Hongsong Yu. 2025. "Gut Microbiota and Metabolites: Biomarkers and Therapeutic Targets for Diabetes Mellitus and Its Complications" Nutrients 17, no. 16: 2603. https://doi.org/10.3390/nu17162603

APA Style

Yan, K., Sun, X., Wang, X., Zheng, J., & Yu, H. (2025). Gut Microbiota and Metabolites: Biomarkers and Therapeutic Targets for Diabetes Mellitus and Its Complications. Nutrients, 17(16), 2603. https://doi.org/10.3390/nu17162603

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

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop