Next Article in Journal
The Use of Mefenoxam to Treat Cutaneous and Gastrointestinal Pythiosis in Dogs: A Retrospective Study
Next Article in Special Issue
Enriched Aptamer Libraries in Fluorescence-Based Assays for Rikenella microfusus-Specific Gut Microbiome Analyses
Previous Article in Journal
Deciphering the Association between Campylobacter Colonization and Microbiota Composition in the Intestine of Commercial Broilers
Previous Article in Special Issue
Gut Bacterial Communities in HIV-Infected Individuals with Metabolic Syndrome: Effects of the Therapy with Integrase Strand Transfer Inhibitor-Based and Protease Inhibitor-Based Regimens
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiota and Critical Metabolites: Potential Target in Preventing Gestational Diabetes Mellitus?

1
Institute of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
2
Department of Integrated Traditional Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
*
Author to whom correspondence should be addressed.
Submission received: 22 May 2023 / Revised: 24 June 2023 / Accepted: 28 June 2023 / Published: 30 June 2023
(This article belongs to the Special Issue The Role of the Gut Microbiota in Human Health and Disease 2.0)

Abstract

:
Gestational diabetes mellitus (GDM) is an intractable issue that negatively impacts the quality of pregnancy. The incidence of GDM is on the rise, becoming a major health burden for both mothers and children. However, the specific etiology and pathophysiology of GDM remain unknown. Recently, the importance of gut microbiota and related metabolic molecules has gained prominence. Studies have indicated that women with GDM have significantly distinct gut microbiota and gut metabolites than healthy pregnant women. Given that the metabolic pathways of gut flora and related metabolites have a substantial impact on inflammation, insulin signaling, glucose, and lipid metabolism, and so on, gut microbiota or its metabolites, such as short-chain fatty acids, may play a significant role in both pathogenesis and progression of GDM. Whereas the role of intestinal flora during pregnancy is still in its infancy, this review aims to summarize the effects and mechanisms of gut microbiota and related metabolic molecules involved in GDM, thus providing potential intervention targets.

1. Introduction

Gestational diabetes mellitus (GDM) is the most common metabolic complication of pregnancy, affecting up to 25.5% of pregnant women [1,2]. It is defined as glucose intolerance provoking a transient state of hyperglycemia early or first detected during pregnancy [3]. GDM greatly increases the odds of polyhydramnios, pre-eclampsia, macrosomia, shoulder dystocia, cesarean section, and neonatal death [1,4,5,6,7]. Even with a successful birth, children may have a higher risk of developing obesity, diabetes, cardiometabolic disorders, and other metabolic disorders in the future [1,3]. Evidence suggests that women diagnosed with GDM in their first pregnancy have a 34.5% increased risk of developing GDM in subsequent pregnancies when compared to healthy women [8], and their prevalence of T2DM diagnosed within 5 years is approximately 20–50% [9,10,11]. Therefore, GDM has become a major health burden for both mothers and children.
Until now, the specific etiology and pathophysiology of GDM are still unknown. Profound hormonal, metabolic, and immunological changes that occur during pregnancy may be important contributors to GDM [7]. The first trimester is dominated by anabolism, storing adequate energy for fetal development. At this time, maternal insulin secretion elevates, glucose uptake by adipose tissue rises, and pregnant women begin to gain weight. As the pregnancy progresses, levels of placental and metabolic hormones, as well as proinflammatory cytokines, tend to increase, reducing maternal insulin sensitivity in the second half of pregnancy. Maternal insulin insensitivity stimulates gluconeogenesis and lipolysis during the third trimester, which is dominated by catabolism, resulting in elevated levels of maternal plasma glucose and free fatty acids (FFA) that are transported across the placenta and provide adequate energy for fetal development. However, hyperglycemia occurs during this process in susceptible pregnant women who are unable to compensate for insulin resistance (IR) and insufficient insulin secretion, thus resulting in GDM [3,4,7,12].
The 16S sequencing method, metabolomics, and next-generation sequencing (NGS) platforms all assist us in better comprehending the relationship between gut microecology and diseases [7]. The same holds true for GDM. Gut microecology means the great diversity of microorganisms and their surrounding environment. Gut microbiota is the most important component of gut microecology, although intestinal mucosa has a significant impact on gut microecology [13]. Through digestion, vitamin synthesis, metabolism, and systemic immunomodulation, the gut microbiota, also known as the “second genome,” play a crucial role in maintaining host physiology and homeostasis [7,14,15,16]. The gut metabolites, such as bile acids (BAs), amino acids, and short-chain fatty acids (SCFAs), are key mediators in host-microbiota cross-talk, modulating the integrity of the gut barrier, host physiology, and host pathological changes [16]. Studies have indicated that women with GDM have significantly distinct gut microbiota and gut metabolites than healthy pregnant women [9,17,18]. Moreover, some changes in maternal gut flora in GDM patients may be transmitted vertically to their children [19]. Given that gut flora and related metabolites substantially impact inflammation, insulin signaling, and glucose and lipid metabolism [15,16,20], GDM might be connected to disruptions in the gut microbiota and related metabolites, especially SCFAs. The purpose of this review is to summarize the impacts and mechanisms of gut microbiota and related metabolites in GDM, as well as to suggest prospective intervention targets, thereby expanding the chances for GDM prevention.

2. Gut Microbiota Changes during Normal Pregnancy

Firmicutes and Bacteroidetes are the most abundant forms of bacteria in gut flora, followed by Actinomycetes and Proteobacteria [21]. To prepare for the newborn, profound hormonal, immunological, and metabolic modifications occur throughout pregnancy [22,23], affecting the immunological state and energy storage of the intestinal mucosa [24,25], thus altering the composition of the gut microbiome and their metabolites [25,26,27].
Most studies agreed that the abundance of gut bacteria associated with energy storage promotion and inflammatory states might increase during pregnancy. Butyrate-producing bacteria such as Faecalibacterium and Eubacterium were identified to be over-represented in the first trimester [24]. At the same time, bacteria related to energy storage promotion, such as Akkermansia, Bifidobacterium, and Firmicutes, were hypothesized to be increased in the third trimester, and proinflammatory bacteria Proteobacteria and Actinobacteria were also augmented [25,28,29]. Santacruz et al. concurred that the abundance of gut bacteria associated with inflammatory states was increased in pregnancy [30]. Moreover, bacterial richness (α-diversity) was eventually diminished toward the third trimester [7,25]. Fecal samples from the first and third trimesters were pooled from five healthy-weight pregnant women, which were subsequently transplanted into the germ-free wild-type Swiss-Webster female mice. Shotgun metagenomics analysis revealed that the fraction of bacteria involved with evoking inflammatory states and energy storage was elevated during pregnancy. However, Digiulio et al. concluded minimal remodeling of gut microbiome associated with pregnancy [31] and intestinal flora in the first trimester was equivalent to that of non-pregnant women [30,32].

3. Gut Microbiota and Related Metabolic Changes during GDM

Pregnant women with GDM are characterized by higher IR and glucose intolerance during pregnancy as compared to healthy pregnant women [33]. Previous studies have shown that the gut microbiome can regulate metabolic homeostasis, affecting the composition of the gut microbiome [9,34]. Therefore, it is plausible to believe that the changed composition of intestinal flora and related metabolites in GDM patients may be the underlying mechanism of GDM.
Emerging studies have concentrated on alterations of intestinal flora and related metabolites during GDM. Most studies have revealed that women with GDM had significant differences in beta diversity (the overall structure of the microbiota) and lower alpha diversity (the number of species present in the given community) [17,18,35,36,37,38,39]. However, no difference in beta diversity and alpha diversity or an increase in alpha diversity across the pregnant trimesters was also reported [40,41,42,43].
Changes appear in early pregnancy. A case-control study with 75 overweight participants that thoroughly removed any confounding factors discovered that the relative abundance of the Ruminococcaceae family was significantly augmented during early pregnancy in women who later progressed to GDM compared to those without developing GDM [44]. Greater Ruminococcaceae family abundance was related to impaired glucose homeostasis, probably via boosting inflammatory state and consequent impairment of insulin metabolism [44,45]. It remains to be known whether the same changes would occur in pregnant women of normal weight who have GDM. Ma et al. carried out a case-control study and revealed that Eisenbergiella, Tyzzerella 4, and Lachnospiraceae NK4A136 were enriched in the GDM group compared to controls who were matched with age, gestational age, and sample collection date [46]. They also discovered that Eisenbergiella and Tyzzerella 4 were positively correlated with fasting blood glucose levels [46]. Functional annotation modules also showed that sphingolipid metabolism, starch, and sucrose metabolism were boosted, while lysine biosynthesis and nitrogen metabolism were decreased [46]. In a nested case-control study, it was demonstrated that Actinomyces, Adlercreutzia, Bifidobacterium, Coriobacteriaceae, Lachnospiraceae spp., and Rothia were considerably reduced, whereas Enterobacteriaceae, Ruminococcaceae spp., and Veillonellaceae were augmented in GDM women. Furthermore, Hu et al. affirmed the predictive power of microbial alterations; they demonstrated that the abundance of Staphylococcus relative to Clostridium, Coriobacteriaceae, and Roseburia was positively correlated with fasting blood glucose and postprandial glucose level [47].
Gut microbiota dysbiosis in women with GDM also occurs during the second trimester. Pathobionts, such as P. distasonis, Klebsiella variicola, and Catenibacterium mitsuokai, which were positively associated with maternal glucose levels, were revealed to be elevated during the second trimester in women with GDM compared to women without GDM. In comparison, the levels of beneficial butyrate-producing bacteria, including Alistipess spp., Bifidobacterium spp., Eubacterium spp., and Methanobrevibacter smithii, were lowered [17]. Moreover, functional analysis figured out that gut microbiota dysbiosis during the second trimester may be correlated with the upregulation of membrane transport, energy metabolism, lipopolysaccharide, and phosphotransferase activation, as well as the suppression of amino acid metabolic pathways [17]. Chen et al. confirmed that beneficial acetate-producing, lactate-producing, and butyrate-producing bacteria, including Bifidobacterium spp., Eubacterium spp., were depleted while Corynebacterium spp., Lactobacillus spp., and Blautia hydrogenotrophica were enriched in GDM patients [35]. Ye and Zhang reported a rise in the abundance of Blautia and Eubacterium hallii group, with a reduction in the richness of Faecalibacterium in GDM patients who failed to control glycemic [48]. It was affirmed that Gemmiger, Oscillospira, Enterococcaceae unassigned genera of Clostridiales, Ruminococcaceae, seven genera within the phylum Firmicutes, and two within the phylum Actinobacteria were depleted, while four genera within phylum Bacteroidetes were raised [36]. Simultaneously, the genera Bacteroides, Dialister, and Campylobacter, and an unassigned genus of Enterococcaceae appeared to be taxonomic biomarkers of GDM, whereas the genera Gemmiger and Bifidobacterium, as well as unassigned genera of Clostridiales and Ruminococcaceae, appeared to be markers of normal pregnancy [36]. Wei et al. carried out a cross-sectional study and discovered no difference in α-diversity, but they stated that Ruminococcus bromii, Clostridium colinum, and Streptococcus infantis were significantly associated with the GDM samples. Moreover, S. infantis were positively associated with blood glucose levels after adjusting for body mass index (BMI) [49]. A cross-sectional study in which BMI was matched in separate groups revealed no difference in β-diversity in women with GDM. However, the higher abundance of Bacteroides caccae, Bacteroides massiliensis, and Bacteroides thetaiotaomicron, with a lower abundance of Bacteroides vulgatus, Eubacterium eligens, Lactobacillus rogosae, and Prevotella copri were detected in women with GDM [42]. According to Zhang et al., women who later progressed to GDM showed decreased α-diversity and abundance of genera belonging to Ruminococcaceae, Coriobacteriales, and Lachnospiraceae [39]. Liang et al. explored that Bacteroides and Lachnoclostridium were more abundant, but Ruminococcaceae UCG-002, Ruminococcaceae UCG-005, Clostridium sensu stricto 1, and Streptococcus were reduced in the GDM group. Further, Paraprevotella, Roseburia, Faecalibacterium, and Ruminococcaceae_UCG-002 were significantly negatively correlated with glucose. Ruminococcaceae_UCG-002 was significantly negatively correlated with hemoglobin A1c. Bacteroides were significantly positively correlated with glucose, while Sutterella, Oscillibacter, and Bifidobacterium were significantly positively correlated with glucagon-like peptide-1 [50].
During the third trimester, the Akkermansia genus was enhanced in women with GDM [51], while the abundance of Faecalibacterium was found to decrease substantially [19]. According to Crusell et al., Actinobacteria at the phylum level and Collinsella, Desulfovibrio, and Rothia at the genus level were more abundant in GDM women [18]. After adjustment for BMI, OTUs allocated to Akkermansia were associated with decreased insulin sensitivity, while Christensenella OTUs were associated with greater fasting plasma glucose concentration in GDM women [18]. Crusell et al. also discovered that the aberrant composition of the gut microbiome could be observed till 8 months after birth [18]. According to Cortez et al., Ruminococcus and Prevotella were increased, while Bacteroides, Eubacterium, and Firmicutes were reduced in women with GDM during the third trimester [37,52]. In a cross-sectional study, increased Gammaproteobacteria and Haemophilus abundances were found in women with GDM compared to weight-matched controls [38]. However, Li and colleagues declared the greater α-diversity in women with GDM. They also affirmed greater BMI, a larger proportion of Firmicutes, and a lower proportion of Bacteroides than healthy controls [43]. Dynamic changes in intestinal flora from the first trimester to the second trimester were observed. It was also discovered that the GDM group had greater pregestational BMI, a consistent decrease in Coprococcus and Streptococcus levels, and suppressed interbacterial interactions, which may potentially serve as early biomarkers for GDM [53] (Table 1).
In a nutshell, despite many studies focusing on gut microbiota in GDM, the findings are inconclusive. Disparities in these findings can be attributed to sample size, geographical location, study design, and participant selection criteria (including population BMI, food habits, gestational age, and sequencing platform) [7]. Moreover, studies that dynamically investigate the changes through multi-point fecal sampling during different trimesters are rare. Still, it can be concluded that gut microbiota dysbiosis in GDM can be characterized by changes in α-diversity and β-diversity, an increase in gram-negative bacteria and part of gram-positive bacteria, alterations in SCFA-producing bacteria (including Akkermansia, Bifidobacterium bifidum, Coprococcus, Lactobacillus casei, Roseburia, and Ruminococcus), and a decrease in bacteria with probiotics properties [17,18,19,35,36,44,47,50,54].

4. GDM–Gut Interaction

4.1. Effect of Metabolic Overload and Chronic Inflammation on Gut Microecology

Currently, research into the underlying mechanisms involving GDM and gut microbiota dysbiosis is at the preliminary stage. Many studies have reported that changes in gut microecology were associated with decreased insulin sensitivity, higher plasma glucose concentration, and greater BMI in GDM women. These changes appear to begin in early pregnancy and last for several months after delivery, negatively affecting intestinal function.
Studies also indicated that pregnant women with GDM tend to have a significantly higher pre-pregnancy BMI than women without GDM [37,43]. Overweight and obesity before pregnancy are associated with a greater risk of developing GDM [55]. The main reason may be related to gut dysbiosis, which plays a role in the pathogenesis and progression of GDM [56]. Most studies revealed that in genetical obese ob/ob mice, diet-induced obese (DIO) mice, and obese people, the proportion of Akkermancia muciniphila, butyrate-producing bacteria, including Roseburia and Faecalibacterium prauznitzii, were reduced [57,58,59]. At the same time, the proportion of Bacteroidetes and Firmicutes was aberrant; though some studies agreed on a significant increase in Firmicutes, others reported a significant increase in Bacteroidetes and a decrease in Firmicutes [60,61,62,63,64,65,66]. Early white adipose tissue dilatation and chronic obesity can both activate the inflammatory programs, permanently skewing the immune system toward proinflammatory phenotypes [67].
GDM is usually accompanied by hyperglycemia and IR. Human metagenome-wide association studies in patients with type 2 diabetes demonstrated highly significant correlations between hyperglycemia and IR and specific intestinal bacteria, bacterial genes, and associated metabolic pathways [68]. Hyperglycemia affects goblet cell proliferation, maturation, and mucus biosynthesis, which will disrupt the intestinal barrier and intestinal mucus, allowing pathogenic bacteria and their elements, such as LPS, to pass through the epithelial barrier [69,70]. Subsequently, LPS binds to toll-like receptors on the surface of intestinal epithelial cells to recruit immune cells in the intestine [67,71]. IFN-γ, IL-1, and other proinflammatory cytokines are released by recruited immune cells, disrupting the enteral environment and increasing intestinal permeability, further eliciting an immune response and triggering chronic inflammation [70,72,73]. Hyperglycemia can also drive immune cell dysfunction via mitochondrial dysfunction [74,75]. Damage to the enteral environment and immune response will affect the value and growth of intestinal flora, leading to gut microbiota dysbiosis [67,71,76,77,78].

4.2. Roles of Gut Microecology in GDM

Hypotheses have been put forward that changes in microecology may be related to GDM by affecting intestinal barrier function, glucose metabolism disorder, energy accumulation, and other ways.

4.2.1. Gut Microbiota Dysbiosis

Zonulin, which is released by the liver and gut epithelial cells, is a physiological tight junction modulator and a potential predictor of gastrointestinal permeability [7,79]. Recent research has found that the zonulin level in plasma is significantly elevated in GDM women, indicating their greater gastrointestinal permeability and dysfunction of the gut barrier [44,80]. The gut barrier consists of mucus, intercellular tight junctions, immunoglobulins, antimicrobial peptides secreted by Paneth cells, microbes, and other components [79]. The mucus barrier is one of the first lines of the gut barrier. Gut microbiota dysbiosis may result in the thinning of the mucosal layer in GDM patients [81]. For example, Prevotella and Akkermansia levels are reported to be elevated in women with GDM. They are mucin-degrading pathobionts, which can increase mucin oligosaccharide degradation beyond the normal limit, leading to gut barrier dysfunction and leaky gut [57,82]. Additionally, several studies have proved a decrease in SCFA-producing bacteria in women with GDM during the second and third trimesters [24,35]. Elhaseen et al. discovered that SCFAs could significantly upregulate the tight junction protein genes (ZO-1 and occludin) of Caco-2 cells, implying that SCFAs have the potential to strengthen the gut barrier [83]. Therefore, a decrease in SCFAs may be associated with increased permeability of the gut epithelium.
The other feature of gut microbiota dysbiosis in women with GDM is the elevated level of gram-negative bacteria, including Prevotella, Haemophilus, and Desulfovibrio in both mid and late pregnancy [18,37,38,42,52]. Rises in Gram-negative pathobionts are linked to LPS biosynthesis. Accordingly, functional analysis revealed that pathways relating to LPS biosynthesis and transport system were increased in women with GDM [17,52]. Elevated levels of Gram-negative bacteria and LPS in women with GDM may weaken the intestinal epithelial barrier [7]. Elevated LPS and pathobionts, on the other hand, can adhere to the mucosal layer, cross the epithelial layer of the gut via toll-like receptor 2/4 (TLR2/4) activation, translocate via phagocytosis and dendritic cell (DC) co-localization, ultimately entering systemic circulation and causing metabolic endotoxemia [7,79]. Furthermore, the compromised intestinal barrier can facilitate the migration of LPS and pathobionts, which enter the systemic circulation and peripheral tissues, binding to and activating TLR [70,72,73]. TLR activation triggers macrophage infiltration and inflammation pathways, such as C-Jun N-terminal kinase (JNK), inhibitory B kinase (IKK), and nuclear factor kappa-B (NF-κB), which might evoke serine phosphorylation of the insulin receptor substrate-1+Ser307 (IRS-1+Ser307), resulting in suppression of phosphatidylinositol 3-kinase (PI3-K) and protein kinase BSer473 (AktSer473). This procedure will impair insulin signaling and reduce glucose uptake in peripheral tissues, resulting in hyperglycemia in GDM women [7,70,72,73] (Figure 1).

4.2.2. Roles of Critical Metabolites

Gut-related metabolites include SCFAs, bile acids (BAs), amino acids, and so on. Metabolomics analysis has revealed that these metabolites were significantly altered in women with GDM, especially SCFAs.
SCFAs are derived from microbiota-accessible carbohydrates (MACs) fermented in the colon from dietary fibers and resistant starch fermentation [84,85]. They are extensively involved in processing undigested diets for additional energy, lipid metabolism, glucose metabolism, and inflammation, in addition to their potential role in gut barrier maintenance [52,83,86]. SCFAs promote adipogenesis in adipose tissue by increasing the expression of the peroxisome proliferator-activated receptor (PPAR) while decreasing serum FFA levels by suppressing lipolysis through activating GPR43 [87,88]. Propionate is an essential substrate for gluconeogenesis in the liver and skeletal muscle, which can also inhibit lipogenesis by suppressing fatty acid synthase [88]. Butyrate and acetate have been linked to lipogenesis in the liver [89]. SCFAs also endorse glycogen storage while inhibiting glycolysis in the liver and skeletal muscles [90]. In the gut, SCFAs can stimulate enteroendocrine L cells to release glucagon-like peptide-1 (GLP-1) and intestinal hormone peptide YY (PYY) [91,92,93]. They both are essential brain-gut peptides that help to control appetite, promote insulin secretion, and suppress the release of pancreatic glucagon [93,94]. On the other hand, butyrate constitutes a major energy source for intestinal epithelial cells and metabolic responses, while acetate regulates intestinal PH value and intestinal flora [54,95]. They are essential for enhancing epithelial barrier function and intestinal microecology. Moreover, SCFAs influence hematopoietic progenitors in the murine bone marrow and maintain the balance between anti-inflammatory and proinflammatory cells in mice by affecting peripheral DC cells and T cells, implying that they are important for the development of the innate and adaptive immune system [96,97].
As mentioned above, the amount and types of SCFAs are largely determined by the number of SCFA-producing bacteria. The Firmicutes phylum, particularly Faecalibacterium, Roseburia, and Bifidobacterium, produce butyrate, whereas Bacteroidetes produce acetate and propionate [98]. The majority of butyrate will be absorbed as an energy source by colonocytes (7), whereas the majority of acetate will be metabolized by the liver, adipose, muscle, heart, and kidney tissues [7,99].
Alterations in the abundance of SCFA-producing bacteria have been reported in women with GDM. The compositions of SCFA-producing genus Faecalibacterium, Bifidobacterium, Ruminococcus, Roseburia, Coprococcus, Akkermansia, Phascolarctobacterium, and Eubacterium were deficient in women with GDM and were inversely correlated with glucose tolerance [17,18,19,37,48,51,53]. As a result, acetate, butanoate, and propanoate production were revealed to be reduced during the second and third trimesters [95,100]. Inadequate SCFAs reduce adipose tissue lipid storage capacity, inhibit fatty acid oxidation, and increase lipolysis, which then raises serum FFA levels and increases lipid storage in the liver and muscle [7,48]. Simultaneously, insufficient SCFAs may be unable to maintain the balance of anti-inflammatory and proinflammatory cells, leading to low-grade inflammation. Elevated serum FFA levels, increased lipid storage in the liver and muscle, and low-grade inflammation may all contribute to insulin resistance and hyperglycemia in women with GDM [7,48]. At the same time, insufficient SCFAs may be unable to maintain the gut barrier and intestinal PH value, evoking gut leakage and gut dysbiosis and forming a vicious cycle [54]. On the other hand, elevations of SCFAs-producing bacteria such as Firmicutes, Phascolarctobacterium, Faecalibacterium, and Bacteroidetes also occur in women with GDM, which lead to excessive butyrate, isobutyrate, isovalerate, acetic and propionic production [52,101]. Excessive SCFAs production may lead to extra energy harvesting capacity and FFA overflow, increasing lipid storage in the liver and skeletal muscle and leading to obesity [102]. Additionally, it may upregulate gluconeogenesis pathways and suppress glycolysis pathways and insulin signaling in peripheral tissues, resulting in hyperglycemia and IR [7,102]. Both isobutyric and isovaleric acids are branched-chain SCFAs, which may be involved in the release of proinflammatory cytokines [101] (Figure 2).
Apart from the effect of SCFA on energy storage, changes in the gut microbiota of women with GDM may also have a direct impact on energy storage. It was reported that bacteria associated with energy storage promotion, such as Akkermansia, Bifidobacterium, and Firmicutes, were augmented during the third trimester in healthy pregnant women. However, bacterium levels appeared to be lower in the second trimester [35,36,47], whereas Akkermansia levels were higher in the third trimester in women with GDM [18,51]. Hence, women with GDM may experience energy storage disorders, eliciting energy metabolism disturbance, which may be associated with glucose and lipid metabolism disorders [58,62,86]. On the other hand, Bacteroidetes was reported to be elevated in GDM women and was positively correlated with blood glucose. Bacteroidetes is considered a gram-negative bacterium that can produce a proinflammatory marker—LPS [75]. It may be implicated in the pathogenesis and progression of metabolic disturbance via inflammation [17,38,103].
Moreover, aromatic amino acids (AAA)-degrading bacteria such as Clostridium, Fusobacterium, and Eubacterium were decreased in GDM women compared to healthy pregnant women [17,37]. That might be the reason for the notably delayed and blunted decrease in AAA in GDM women [104]. Since AAA catabolism by the gut microbiome yields numerous bioactive molecules that regulate murine immune, metabolic, and neuronal responses in the gut and distant organs, decreased AAA catabolism may disturb host–microbe metabolic axes [105]. This needs to be further clarified in human trials. Metabolomics analysis of meconium and serum showed that metabolic pathways, including taurine and hypotaurine metabolism, pyrimidine metabolism, beta-alanine metabolism, and BAs biosynthesis, were altered in GDM subjects [106]. However, the correlation between these changes in metabolites and intestinal flora and GDM still remains unclear.

5. Prospect and Implication

As GDM has become a major health burden for both mothers and children, it is critical to propose effective prevention and treatment strategies. Nonetheless, pregnancy is a unique time, and treatment must take the safety of the fetus into account. In the United States, insulin is currently the first-line agent recommended for the treatment of GDM, whereas metformin and glyburide are not because they can cross the placenta to the fetus and cause neonatal hypoglycemia [107]. Hence, the current options for pharmacologic therapy are extremely limited. Given the importance of gut microecology in GDM, regulating intestinal flora may be a potential strategy for GDM (Figure 3).
Lifestyle behavior change, including dietary restriction and exercise, is the first line of GDM management [107], which are well-established to have the greatest impact on gut microbiota [108]. Dietary carbohydrates (CHO) are an important source of energy for both the mother and the fetus, which can impact gut microbiota and blood glucose levels. Nondigestible CHO components (fiber) can serve as an energy source for colonic microbiota, modulating gut microbiota composition [109]. Lower fiber intake has been linked to decreased gut microbiota diversity and richness [110], increased abundance of Collinsella (a genus closely linked to higher blood glucose) and Sutterella (a Proteobacteria linked to inflammatory status), and higher serum zonulin levels [111,112,113,114]. Furthermore, monounsaturated fatty acids (MUFAs) have been associated with an increase in the abundance of Firmicutes, Proteobacteria, and Bacteroidetes [113]. In pregnant women, fat-soluble vitamins also appear to modulate gut microbiota. Mandal et al. conducted the cohort study and discovered that higher vitamin D intake was associated with decreased microbial α-diversity and a relative decrease in the abundance of proinflammatory Proteobacteria phylum [113]. Higher vitamin E intake was also coupled with a decrease in the abundance of Proteobacteria [113]. To control maternal fasting and postprandial glucose during GDM, current dietary guidelines recommend limiting CHO intake or replacing high glycemic/rapidly digesting CHO with those that are more slowly digesting, as well as taking enough vitamins [107].
Exercise, on the other hand, has been shown to strengthen gut health by increasing microbiome diversity and balancing beneficial and pathogenic bacterial communities [115,116,117]. Specifically, exercise increases butyrate-producing bacteria such as Roseburia hominis, which raises butyrate concentrations in both humans and mice [116,118]. It can also boost key antioxidant enzymes and anti-inflammatory cytokines in intestinal lymphocytes, assisting in the reduction of intestinal inflammation [116,118].
Probiotics and prebiotics are live microorganisms that benefit the host when administered in adequate amounts [119]. As previously stated, women with GDM had a decrease in bacteria with probiotic and prebiotic properties. Therefore, probiotic and prebiotic supplementation may be advantageous. Parallel, double-blind, randomized control trials (RCTs) were published by Dolatkhah et al. They enrolled women with GDM at 24 to 30 weeks of gestation and randomly assigned them to receive probiotics or placebo in capsule form for 6 to 8 weeks. Participants who received probiotics experienced a decrease in fasting blood sugar (FBS) and a significant decrease in the homeostatic model assessment of IR (HOMA-IR) after treatment, implying the potential roles of probiotics on glucose regulation [120,121]. In another RCT, women with GDM were randomly assigned to receive either Probiotic Mixture (VSL#3 probiotics) in a capsule or a placebo for 8 weeks. Hs-CRP, TNF-α, and IL-6 levels were significantly lower in participants who received probiotics, disclosing the potential effect of probiotics on the immune response [122]. However, no effect of probiotics on glycemic control was found in women with GDM receiving a single-strain probiotic capsule delivering 1 billion CFU of Lactobacillus salivarius UCC118 [123]. Differences between studies could be attributed to differences in sample size, probiotic dosage, and duration of probiotic/fermented food/antibiotic consumption prior to intervention [123,124]. Therefore, more RCTs are needed to determine which probiotic or prebiotic strains are optimal and how to utilize them for treating GDM. On the other hand, consuming probiotics and prebiotics requires constant monitoring to avoid adverse effects such as systemic infections and mild gastrointestinal upset [125].
The scope of this review is limited because research into the gut and GDM is still in its early stages. Moreover, the results may be affected by the different study designs and participant selection criteria (diet type, geographical environment, lifestyle, gestational age, and drug factors). We believe it is necessary to further investigate the changes in intestinal flora before the occurrence of hyperglycemia in GDM to provide new approaches for GDM prevention.

6. Conclusions

Although the abnormalities of the intestinal tract may not be the initial factor of GDM, it has been considered to participate in the pathological process of GDM greatly. Women with GDM have significantly distinct gut microbiota and gut metabolites than healthy pregnant women. In turn, the alerted gut microecology and related metabolites substantially impact intestinal barrier function, inflammation, insulin signaling, glucose metabolism, lipid metabolism, and energy accumulation, playing a significant role in both the pathogenesis and progression of GDM. Dietary restrictions, exercise, and probiotics (prebiotics), which can reverse the altered gut microbiota and metabolites, may help alleviate symptoms of GDM.

Author Contributions

M.Z. and Y.S. contributed to the conception of this review. R.H. wrote the manuscript. R.H., M.Z. and Y.S. revised the manuscript. R.H. and Z.L. designed and illustrated the figures. Y.G., Y.H., F.L., H.D. and W.M. performed the literature search and interpretation. R.H., Z.L., Y.G., Y.H., H.D., W.M., K.S., M.Z. and Y.S. reviewed and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

Thanks to BioRender, the figure is created with BioRender.com. URL: https://biorender.com (accessed on 24 February 2023).

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

  1. Choudhury, A.A.; Devi Rajeswari, V. Gestational diabetes mellitus—A metabolic and reproductive disorder. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 143, 112183. [Google Scholar] [CrossRef] [PubMed]
  2. Sacks, D.A.; Hadden, D.R.; Maresh, M.; Deerochanawong, C.; Dyer, A.R.; Metzger, B.E.; Lowe, L.P.; Coustan, D.R.; Hod, M.; Oats, J.J.; et al. Frequency of gestational diabetes mellitus at collaborating centers based on IADPSG consensus panel-recommended criteria: The Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study. Diabetes Care 2012, 35, 526–528. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Saravanan, P. Gestational diabetes: Opportunities for improving maternal and child health. Lancet Diabetes Endocrinol. 2020, 8, 793–800. [Google Scholar] [CrossRef] [PubMed]
  4. Kc, K.; Shakya, S.; Zhang, H. Gestational diabetes mellitus and macrosomia: A literature review. Ann. Nutr. Metab. 2015, 66 (Suppl. S2), 14–20. [Google Scholar] [CrossRef]
  5. Catalano, P.M.; McIntyre, H.D.; Cruickshank, J.K.; McCance, D.R.; Dyer, A.R.; Metzger, B.E.; Lowe, L.P.; Trimble, E.R.; Coustan, D.R.; Hadden, D.R.; et al. The hyperglycemia and adverse pregnancy outcome study: Associations of GDM and obesity with pregnancy outcomes. Diabetes Care 2012, 35, 780–786. [Google Scholar] [CrossRef] [Green Version]
  6. Kim, C. Maternal outcomes and follow-up after gestational diabetes mellitus. Diabet. Med. J. Br. Diabet. Assoc. 2014, 31, 292–301. [Google Scholar] [CrossRef] [Green Version]
  7. Hasain, Z.; Mokhtar, N.M.; Kamaruddin, N.A.; Mohamed Ismail, N.A.; Razalli, N.H.; Gnanou, J.V.; Raja Ali, R.A. Gut Microbiota and Gestational Diabetes Mellitus: A Review of Host-Gut Microbiota Interactions and Their Therapeutic Potential. Front. Cell. Infect. Microbiol. 2020, 10, 188. [Google Scholar] [CrossRef]
  8. Ehrlich, S.F.; Hedderson, M.M.; Feng, J.; Davenport, E.R.; Gunderson, E.P.; Ferrara, A. Change in body mass index between pregnancies and the risk of gestational diabetes in a second pregnancy. Obstet. Gynecol. 2011, 117, 1323–1330. [Google Scholar] [CrossRef] [Green Version]
  9. Wang, X.; Liu, H.; Li, Y.; Huang, S.; Zhang, L.; Cao, C.; Baker, P.N.; Tong, C.; Zheng, P.; Qi, H. Altered gut bacterial and metabolic signatures and their interaction in gestational diabetes mellitus. Gut Microbes 2020, 12, 1840765. [Google Scholar] [CrossRef]
  10. Eades, C.E.; Styles, M.; Leese, G.P.; Cheyne, H.; Evans, J.M. Progression from gestational diabetes to type 2 diabetes in one region of Scotland: An observational follow-up study. BMC Pregnancy Childbirth 2015, 15, 11. [Google Scholar] [CrossRef] [Green Version]
  11. Allalou, A.; Nalla, A.; Prentice, K.J.; Liu, Y.; Zhang, M.; Dai, F.F.; Ning, X.; Osborne, L.R.; Cox, B.J.; Gunderson, E.P.; et al. A Predictive Metabolic Signature for the Transition from Gestational Diabetes Mellitus to Type 2 Diabetes. Diabetes 2016, 65, 2529–2539. [Google Scholar] [CrossRef] [Green Version]
  12. Kampmann, U.; Madsen, L.R.; Skajaa, G.O.; Iversen, D.S.; Moeller, N.; Ovesen, P. Gestational diabetes: A clinical update. World J. Diabetes 2015, 6, 1065–1072. [Google Scholar] [CrossRef]
  13. Lu, D.; Huang, Y.; Kong, Y.; Tao, T.; Zhu, X. Gut microecology: Why our microbes could be key to our health. Biomed. Pharmacother. Biomed. Pharmacother. 2020, 131, 110784. [Google Scholar] [CrossRef]
  14. Flint, H.J.; Scott, K.P.; Duncan, S.H.; Louis, P.; Forano, E. Microbial degradation of complex carbohydrates in the gut. Gut Microbes 2012, 3, 289–306. [Google Scholar] [CrossRef] [Green Version]
  15. The Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [Green Version]
  16. Agus, A.; Clément, K.; Sokol, H. Gut microbiota-derived metabolites as central regulators in metabolic disorders. Gut 2021, 70, 1174–1182. [Google Scholar] [CrossRef]
  17. Kuang, Y.S.; Lu, J.H.; Li, S.H.; Li, J.H.; Yuan, M.Y.; He, J.R.; Chen, N.N.; Xiao, W.Q.; Shen, S.Y.; Qiu, L.; et al. Connections between the human gut microbiome and gestational diabetes mellitus. GigaScience 2017, 6, gix058. [Google Scholar] [CrossRef] [Green Version]
  18. Crusell, M.K.W.; Hansen, T.H.; Nielsen, T.; Allin, K.H.; Rühlemann, M.C.; Damm, P.; Vestergaard, H.; Rørbye, C.; Jørgensen, N.R.; Christiansen, O.B.; et al. Gestational diabetes is associated with change in the gut microbiota composition in third trimester of pregnancy and postpartum. Microbiome 2018, 6, 89. [Google Scholar] [CrossRef]
  19. Wang, J.; Zheng, J.; Shi, W.; Du, N.; Xu, X.; Zhang, Y.; Ji, P.; Zhang, F.; Jia, Z.; Wang, Y.; et al. Dysbiosis of maternal and neonatal microbiota associated with gestational diabetes mellitus. Gut 2018, 67, 1614–1625. [Google Scholar] [CrossRef]
  20. Li, R.; Andreu-Sánchez, S.; Kuipers, F.; Fu, J. Gut microbiome and bile acids in obesity-related diseases. Best Pract. Res. Clin. Endocrinol. Metab. 2021, 35, 101493. [Google Scholar] [CrossRef]
  21. Ley, R.E.; Peterson, D.A.; Gordon, J.I. Ecological and evolutionary forces shaping microbial diversity in the human intestine. Cell 2006, 124, 837–848. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Gohir, W.; Whelan, F.J.; Surette, M.G.; Moore, C.; Schertzer, J.D.; Sloboda, D.M. Pregnancy-related changes in the maternal gut microbiota are dependent upon the mother’s periconceptional diet. Gut Microbes 2015, 6, 310–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Edwards, S.M.; Cunningham, S.A.; Dunlop, A.L.; Corwin, E.J. The Maternal Gut Microbiome During Pregnancy. MCN Am. J. Matern. Child Nurs. 2017, 42, 310–317. [Google Scholar] [CrossRef] [PubMed]
  24. Koren, O.; Goodrich, J.K.; Cullender, T.C.; Spor, A.; Laitinen, K.; Bäckhed, H.K.; Gonzalez, A.; Werner, J.J.; Angenent, L.T.; Knight, R.; et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell 2012, 150, 470–480. [Google Scholar] [CrossRef] [Green Version]
  25. Gorczyca, K.; Obuchowska, A.; Kimber-Trojnar, Ż.; Wierzchowska-Opoka, M.; Leszczyńska-Gorzelak, B. Changes in the Gut Microbiome and Pathologies in Pregnancy. Int. J. Environ. Res. Public Health 2022, 19, 9961. [Google Scholar] [CrossRef]
  26. Stanislawski, M.A.; Dabelea, D.; Wagner, B.D.; Sontag, M.K.; Lozupone, C.A.; Eggesbø, M. Pre-pregnancy weight, gestational weight gain, and the gut microbiota of mothers and their infants. Microbiome 2017, 5, 113. [Google Scholar] [CrossRef]
  27. Rizzetto, L.; Fava, F.; Tuohy, K.M.; Selmi, C. Connecting the immune system, systemic chronic inflammation and the gut microbiome: The role of sex. J. Autoimmun. 2018, 92, 12–34. [Google Scholar] [CrossRef]
  28. Rodríguez, J.M.; Murphy, K.; Stanton, C.; Ross, R.P.; Kober, O.I.; Juge, N.; Avershina, E.; Rudi, K.; Narbad, A.; Jenmalm, M.C.; et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb. Ecol. Health Dis. 2015, 26, 26050. [Google Scholar] [CrossRef]
  29. Donaldson, G.P.; Lee, S.M.; Mazmanian, S.K. Gut biogeography of the bacterial microbiota. Nat. Rev. Microbiol. 2016, 14, 20–32. [Google Scholar] [CrossRef] [Green Version]
  30. Santacruz, A.; Collado, M.C.; García-Valdés, L.; Segura, M.T.; Martín-Lagos, J.A.; Anjos, T.; Martí-Romero, M.; Lopez, R.M.; Florido, J.; Campoy, C.; et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br. J. Nutr. 2010, 104, 83–92. [Google Scholar] [CrossRef] [Green Version]
  31. DiGiulio, D.B.; Callahan, B.J.; McMurdie, P.J.; Costello, E.K.; Lyell, D.J.; Robaczewska, A.; Sun, C.L.; Goltsman, D.S.; Wong, R.J.; Shaw, G.; et al. Temporal and spatial variation of the human microbiota during pregnancy. Proc. Natl. Acad. Sci. USA 2015, 112, 11060–11065. [Google Scholar] [CrossRef] [Green Version]
  32. Chassaing, B.; Gewirtz, A.T. Gut microbiota, low-grade inflammation, and metabolic syndrome. Toxicol. Pathol. 2014, 42, 49–53. [Google Scholar] [CrossRef]
  33. 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]
  34. Wang, L.; Zhou, J.; Gober, H.J.; Leung, W.T.; Huang, Z.; Pan, X.; Li, C.; Zhang, N.; Wang, L. Alterations in the intestinal microbiome associated with PCOS affect the clinical phenotype. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 133, 110958. [Google Scholar] [CrossRef]
  35. Chen, F.; Gan, Y.; Li, Y.; He, W.; Wu, W.; Wang, K.; Li, Q. Association of gestational diabetes mellitus with changes in gut microbiota composition at the species level. BMC Microbiol. 2021, 21, 147. [Google Scholar] [CrossRef]
  36. Chen, T.; Zhang, Y.; Zhang, Y.; Shan, C.; Zhang, Y.; Fang, K.; Xia, Y.; Shi, Z. Relationships between gut microbiota, plasma glucose and gestational diabetes mellitus. J. Diabetes Investig. 2021, 12, 641–650. [Google Scholar] [CrossRef]
  37. Cortez, R.V.; Taddei, C.R.; Sparvoli, L.G.; Ângelo, A.G.S.; Padilha, M.; Mattar, R.; Daher, S. Microbiome and its relation to gestational diabetes. Endocrine 2019, 64, 254–264. [Google Scholar] [CrossRef]
  38. Xu, Y.; Zhang, M.; Zhang, J.; Sun, Z.; Ran, L.; Ban, Y.; Wang, B.; Hou, X.; Zhai, S.; Ren, L.; et al. Differential intestinal and oral microbiota features associated with gestational diabetes and maternal inflammation. Am. J. Physiol. Endocrinol. Metab. 2020, 319, E247–E253. [Google Scholar] [CrossRef]
  39. Zhang, Y.; Chen, T.; Zhang, Y.; Hu, Q.; Wang, X.; Chang, H.; Mao, J.H.; Snijders, A.M.; Xia, Y. Contribution of trace element exposure to gestational diabetes mellitus through disturbing the gut microbiome. Environ. Int. 2021, 153, 106520. [Google Scholar] [CrossRef]
  40. Dualib, P.M.; Taddei, C.R.; Fernandes, G.; Carvalho, C.R.S.; Sparvoli, L.G.; Silva, I.T.; Mattar, R.; Ferreira, S.R.G.; Dib, S.A.; Almeida-Pititto, B. Gut Microbiota across Normal Gestation and Gestational Diabetes Mellitus: A Cohort Analysis. Metabolites 2022, 12, 796. [Google Scholar] [CrossRef]
  41. Qin, S.; Liu, Y.; Wang, S.; Ma, J.; Yang, H. Distribution characteristics of intestinal microbiota during pregnancy and postpartum in healthy women. J. Matern.-Fetal Neonatal Med. Off. J. Eur. Assoc. Perinat. Med. Fed. Asia Ocean. Perinat. Soc. Int. Soc. Perinat. Obs. 2022, 35, 2915–2922. [Google Scholar] [CrossRef] [PubMed]
  42. Festa, C.; Drago, L.; Martorelli, M.; Di Marino, V.P.; Bitterman, O.; Corleto, C.C.; Corleto, V.D.; Napoli, A. Flash on gut microbiome in gestational diabetes: A pilot study. New Microbiol. 2020, 43, 195–197. [Google Scholar] [PubMed]
  43. Li, G.; Yin, P.; Chu, S.; Gao, W.; Cui, S.; Guo, S.; Xu, Y.; Yuan, E.; Zhu, T.; You, J.; et al. Correlation Analysis between GDM and Gut Microbial Composition in Late Pregnancy. J. Diabetes Res. 2021, 2021, 8892849. [Google Scholar] [CrossRef] [PubMed]
  44. Mokkala, K.; Houttu, N.; Vahlberg, T.; Munukka, E.; Rönnemaa, T.; Laitinen, K. Gut microbiota aberrations precede diagnosis of gestational diabetes mellitus. Acta Diabetol. 2017, 54, 1147–1149. [Google Scholar] [CrossRef] [PubMed]
  45. Gomez-Arango, L.F.; Barrett, H.L.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Dekker Nitert, M. Connections Between the Gut Microbiome and Metabolic Hormones in Early Pregnancy in Overweight and Obese Women. Diabetes 2016, 65, 2214–2223. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Ma, S.; You, Y.; Huang, L.; Long, S.; Zhang, J.; Guo, C.; Zhang, N.; Wu, X.; Xiao, Y.; Tan, H. Alterations in Gut Microbiota of Gestational Diabetes Patients During the First Trimester of Pregnancy. Front. Cell. Infect. Microbiol. 2020, 10, 58. [Google Scholar] [CrossRef] [Green Version]
  47. Hu, P.; Chen, X.; Chu, X.; Fan, M.; Ye, Y.; Wang, Y.; Han, M.; Yang, X.; Yuan, J.; Zha, L.; et al. Association of Gut Microbiota during Early Pregnancy with Risk of Incident Gestational Diabetes Mellitus. J. Clin. Endocrinol. Metab. 2021, 106, e4128–e4141. [Google Scholar] [CrossRef]
  48. Ye, G.; Zhang, L.; Wang, M.; Chen, Y.; Gu, S.; Wang, K.; Leng, J.; Gu, Y.; Xie, X. The Gut Microbiota in Women Suffering from Gestational Diabetes Mellitus with the Failure of Glycemic Control by Lifestyle Modification. J. Diabetes Res. 2019, 2019, 6081248. [Google Scholar] [CrossRef] [Green Version]
  49. Wei, J.; Qing, Y.; Zhou, H.; Liu, J.; Qi, C.; Gao, J. 16S rRNA gene amplicon sequencing of gut microbiota in gestational diabetes mellitus and their correlation with disease risk factors. J. Endocrinol. Investig. 2022, 45, 279–289. [Google Scholar] [CrossRef]
  50. 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]
  51. Liu, H.; Pan, L.L.; Lv, S.; Yang, Q.; Zhang, H.; Chen, W.; Lv, Z.; Sun, J. Alterations of Gut Microbiota and Blood Lipidome in Gestational Diabetes Mellitus with Hyperlipidemia. Front. Physiol. 2019, 10, 1015. [Google Scholar] [CrossRef] [Green Version]
  52. Ferrocino, I.; Ponzo, V.; Gambino, R.; Zarovska, A.; Leone, F.; Monzeglio, C.; Goitre, I.; Rosato, R.; Romano, A.; Grassi, G.; et al. Changes in the gut microbiota composition during pregnancy in patients with gestational diabetes mellitus (GDM). Sci. Rep. 2018, 8, 12216. [Google Scholar] [CrossRef]
  53. Zheng, W.; Xu, Q.; Huang, W.; Yan, Q.; Chen, Y.; Zhang, L.; Tian, Z.; Liu, T.; Yuan, X.; Liu, C.; et al. Gestational Diabetes Mellitus Is Associated with Reduced Dynamics of Gut Microbiota during the First Half of Pregnancy. mSystems 2020, 5, e00109–e00120. [Google Scholar] [CrossRef] [Green Version]
  54. Huang, L.; Thonusin, C.; Chattipakorn, N.; Chattipakorn, S.C. Impacts of gut microbiota on gestational diabetes mellitus: A comprehensive review. Eur. J. Nutr. 2021, 60, 2343–2360. [Google Scholar] [CrossRef]
  55. Sun, Y.; Shen, Z.; Zhan, Y.; Wang, Y.; Ma, S.; Zhang, S.; Liu, J.; Wu, S.; Feng, Y.; Chen, Y.; et al. Effects of pre-pregnancy body mass index and gestational weight gain on maternal and infant complications. BMC Pregnancy Childbirth 2020, 20, 390. [Google Scholar] [CrossRef]
  56. Saad, M.J.; Santos, A.; Prada, P.O. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology 2016, 31, 283–293. [Google Scholar] [CrossRef] [Green Version]
  57. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
  58. Jumpertz, R.; Le, D.S.; Turnbaugh, P.J.; Trinidad, C.; Bogardus, C.; Gordon, J.I.; Krakoff, J. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am. J. Clin. Nutr. 2011, 94, 58–65. [Google Scholar] [CrossRef] [Green Version]
  59. Pauli, J.R.; Ropelle, E.R.; Cintra, D.E.; Carvalho-Filho, M.A.; Moraes, J.C.; De Souza, C.T.; Velloso, L.A.; Carvalheira, J.B.; Saad, M.J. Acute physical exercise reverses S-nitrosation of the insulin receptor, insulin receptor substrate 1 and protein kinase B/Akt in diet-induced obese Wistar rats. J. Physiol. 2008, 586, 659–671. [Google Scholar] [CrossRef]
  60. Caricilli, A.M.; Picardi, P.K.; de Abreu, L.L.; Ueno, M.; Prada, P.O.; Ropelle, E.R.; Hirabara, S.M.; Castoldi, Â.; Vieira, P.; Camara, N.O.; et al. Retraction: Gut Microbiota Is a Key Modulator of Insulin Resistance in TLR 2 Knockout Mice. PLoS Biol. 2016, 14, e1002479. [Google Scholar] [CrossRef] [Green Version]
  61. Mujico, J.R.; Baccan, G.C.; Gheorghe, A.; Díaz, L.E.; Marcos, A. Changes in gut microbiota due to supplemented fatty acids in diet-induced obese mice. Br. J. Nutr. 2013, 110, 711–720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef] [PubMed]
  63. Arthur, J.C.; Perez-Chanona, E.; Mühlbauer, M.; Tomkovich, S.; Uronis, J.M.; Fan, T.J.; Campbell, B.J.; Abujamel, T.; Dogan, B.; Rogers, A.B.; et al. Intestinal inflammation targets cancer-inducing activity of the microbiota. Science 2012, 338, 120–123. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Collado, M.C.; Isolauri, E.; Laitinen, K.; Salminen, S. Distinct composition of gut microbiota during pregnancy in overweight and normal-weight women. Am. J. Clin. Nutr. 2008, 88, 894–899. [Google Scholar] [CrossRef] [Green Version]
  65. Henao-Mejia, J.; Elinav, E.; Jin, C.; Hao, L.; Mehal, W.Z.; Strowig, T.; Thaiss, C.A.; Kau, A.L.; Eisenbarth, S.C.; Jurczak, M.J.; et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 2012, 482, 179–185. [Google Scholar] [CrossRef] [Green Version]
  66. Schwiertz, A.; Taras, D.; Schäfer, K.; Beijer, S.; Bos, N.A.; Donus, C.; Hardt, P.D. Microbiota and SCFA in lean and overweight healthy subjects. Obesity 2010, 18, 190–195. [Google Scholar] [CrossRef]
  67. Saltiel, A.R.; Olefsky, J.M. Inflammatory mechanisms linking obesity and metabolic disease. J. Clin. Investig. 2017, 127, 1–4. [Google Scholar] [CrossRef] [Green Version]
  68. Velmurugan, G.; Ramprasath, T.; Gilles, M.; Swaminathan, K.; Ramasamy, S. Gut Microbiota, Endocrine-Disrupting Chemicals, and the Diabetes Epidemic. Trends Endocrinol. Metab. TEM 2017, 28, 612–625. [Google Scholar] [CrossRef]
  69. Watanabe, R.; Ashida, H.; Kobayashi-Miura, M.; Yokota, A.; Yodoi, J. Effect of chronic administration with human thioredoxin-1 transplastomic lettuce on diabetic mice. Food Sci. Nutr. 2021, 9, 4232–4242. [Google Scholar] [CrossRef]
  70. Zhang, X.; Monnoye, M.; Mariadassou, M.; Beguet-Crespel, F.; Lapaque, N.; Heberden, C.; Douard, V. Glucose but Not Fructose Alters the Intestinal Paracellular Permeability in Association with Gut Inflammation and Dysbiosis in Mice. Front. Immunol. 2021, 12, 742584. [Google Scholar] [CrossRef]
  71. Cox, A.J.; West, N.P.; Cripps, A.W. Obesity, inflammation, and the gut microbiota. Lancet Diabetes Endocrinol. 2015, 3, 207–215. [Google Scholar] [CrossRef]
  72. Arnone, D.; Chabot, C.; Heba, A.C.; Kökten, T.; Caron, B.; Hansmannel, F.; Dreumont, N.; Ananthakrishnan, A.N.; Quilliot, D.; Peyrin-Biroulet, L. Sugars and Gastrointestinal Health. Clin. Gastroenterol. Hepatol. Off. Clin. Pract. J. Am. Gastroenterol. Assoc. 2022, 20, 1912–1924.e7. [Google Scholar] [CrossRef]
  73. Lv, W.; Graves, D.T.; He, L.; Shi, Y.; Deng, X.; Zhao, Y.; Dong, X.; Ren, Y.; Liu, X.; Xiao, E.; et al. Depletion of the diabetic gut microbiota resistance enhances stem cells therapy in type 1 diabetes mellitus. Theranostics 2020, 10, 6500–6516. [Google Scholar] [CrossRef]
  74. Van Niekerk, G.; Davis, T.; Patterton, H.G.; Engelbrecht, A.M. How Does Inflammation-Induced Hyperglycemia Cause Mitochondrial Dysfunction in Immune Cells? BioEssays News Rev. Mol. Cell. Dev. Biol. 2019, 41, e1800260. [Google Scholar] [CrossRef]
  75. Monteiro, R.; Azevedo, I. Chronic inflammation in obesity and the metabolic syndrome. Mediat. Inflamm. 2010, 2010, 289645. [Google Scholar] [CrossRef]
  76. Kayama, H.; Okumura, R.; Takeda, K. Interaction Between the Microbiota, Epithelia, and Immune Cells in the Intestine. Annu. Rev. Immunol. 2020, 38, 23–48. [Google Scholar] [CrossRef]
  77. Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
  78. Buford, T.W. (Dis)Trust your gut: The gut microbiome in age-related inflammation, health, and disease. Microbiome 2017, 5, 80. [Google Scholar] [CrossRef] [Green Version]
  79. Stephens, R.W.; Arhire, L.; Covasa, M. Gut Microbiota: From Microorganisms to Metabolic Organ Influencing Obesity. Obesity (Silver Spring Md.) 2018, 26, 801–809. [Google Scholar] [CrossRef] [Green Version]
  80. Demir, E.; Ozkan, H.; Seckin, K.D.; Sahtiyancı, B.; Demir, B.; Tabak, O.; Kumbasar, A.; Uzun, H. Plasma Zonulin Levels as a Non-Invasive Biomarker of Intestinal Permeability in Women with Gestational Diabetes Mellitus. Biomolecules 2019, 9, 24. [Google Scholar] [CrossRef] [Green Version]
  81. Paone, P.; Cani, P.D. Mucus barrier, mucins and gut microbiota: The expected slimy partners? Gut 2020, 69, 2232–2243. [Google Scholar] [CrossRef] [PubMed]
  82. Wright, D.P.; Rosendale, D.I.; Robertson, A.M. Prevotella enzymes involved in mucin oligosaccharide degradation and evidence for a small operon of genes expressed during growth on mucin. FEMS Microbiol. Lett. 2000, 190, 73–79. [Google Scholar] [CrossRef] [PubMed]
  83. Elamin, E.E.; Masclee, A.A.; Dekker, J.; Pieters, H.J.; Jonkers, D.M. Short-chain fatty acids activate AMP-activated protein kinase and ameliorate ethanol-induced intestinal barrier dysfunction in Caco-2 cell monolayers. J. Nutr. 2013, 143, 1872–1881. [Google Scholar] [PubMed] [Green Version]
  84. Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef] [Green Version]
  85. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Bäckhed, F. From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites. Cell 2016, 165, 1332–1345. [Google Scholar] [CrossRef] [Green Version]
  86. Blaut, M. Gut microbiota and energy balance: Role in obesity. Proc. Nutr. Soc. 2015, 74, 227–234. [Google Scholar] [CrossRef] [Green Version]
  87. Hong, Y.H.; Nishimura, Y.; Hishikawa, D.; Tsuzuki, H.; Miyahara, H.; Gotoh, C.; Choi, K.C.; Feng, D.D.; Chen, C.; Lee, H.G.; et al. Acetate and propionate short chain fatty acids stimulate adipogenesis via GPCR43. Endocrinology 2005, 146, 5092–5099. [Google Scholar] [CrossRef] [Green Version]
  88. Ge, H.; Li, X.; Weiszmann, J.; Wang, P.; Baribault, H.; Chen, J.L.; Tian, H.; Li, Y. Activation of G protein-coupled receptor 43 in adipocytes leads to inhibition of lipolysis and suppression of plasma free fatty acids. Endocrinology 2008, 149, 4519–4526. [Google Scholar] [CrossRef] [Green Version]
  89. Den Besten, G.; Bleeker, A.; Gerding, A.; van Eunen, K.; Havinga, R.; van Dijk, T.H.; Oosterveer, M.H.; Jonker, J.W.; Groen, A.K.; Reijngoud, D.J.; et al. Short-Chain Fatty Acids Protect Against High-Fat Diet-Induced Obesity via a PPARγ-Dependent Switch From Lipogenesis to Fat Oxidation. Diabetes 2015, 64, 2398–2408. [Google Scholar] [CrossRef] [Green Version]
  90. Fushimi, T.; Tayama, K.; Fukaya, M.; Kitakoshi, K.; Nakai, N.; Tsukamoto, Y.; Sato, Y. Acetic acid feeding enhances glycogen repletion in liver and skeletal muscle of rats. J. Nutr. 2001, 131, 1973–1977. [Google Scholar] [CrossRef] [Green Version]
  91. Bolognini, D.; Tobin, A.B.; Milligan, G.; Moss, C.E. The Pharmacology and Function of Receptors for Short-Chain Fatty Acids. Mol. Pharmacol. 2016, 89, 388–398. [Google Scholar] [CrossRef] [Green Version]
  92. Kim, Y.A.; Keogh, J.B.; Clifton, P.M. Probiotics, prebiotics, synbiotics and insulin sensitivity. Nutr. Res. Rev. 2018, 31, 35–51. [Google Scholar] [CrossRef]
  93. De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Bäckhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [Green Version]
  94. Zhou, J.; Martin, R.J.; Raggio, A.M.; Shen, L.; McCutcheon, K.; Keenan, M.J. The importance of GLP-1 and PYY in resistant starch’s effect on body fat in mice. Mol. Nutr. Food Res. 2015, 59, 1000–1003. [Google Scholar] [CrossRef] [Green Version]
  95. Wang, S.; Liu, Y.; Qin, S.; Yang, H. Composition of Maternal Circulating Short-Chain Fatty Acids in Gestational Diabetes Mellitus and Their Associations with Placental Metabolism. Nutrients 2022, 14, 3727. [Google Scholar] [CrossRef]
  96. Dang, A.T.; Marsland, B.J. Microbes, metabolites, and the gut-lung axis. Mucosal Immunol. 2019, 12, 843–850. [Google Scholar] [CrossRef] [Green Version]
  97. Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef]
  98. Macfarlane, S.; Macfarlane, G.T. Regulation of short-chain fatty acid production. Proc. Nutr. Soc. 2003, 62, 67–72. [Google Scholar] [CrossRef]
  99. Bloemen, J.G.; Venema, K.; van de Poll, M.C.; Damink, S.W.O.; Buurman, W.A.; Dejong, C.H. Short chain fatty acids exchange across the gut and liver in humans measured at surgery. Clin. Nutr. 2009, 28, 657–661. [Google Scholar] [CrossRef]
  100. Wu, Y.; Bible, P.W.; Long, S.; Ming, W.K.; Ding, W.; Long, Y.; Wen, X.; Li, X.; Deng, X.; Deng, Y.; et al. Metagenomic analysis reveals gestational diabetes mellitus-related microbial regulators of glucose tolerance. Acta Diabetol. 2020, 57, 569–581. [Google Scholar] [CrossRef]
  101. Chen, S.; Li, J.; Ren, S.; Gao, Y.; Zhou, Y.; Xuan, R. Expression and clinical significance of short-chain fatty acids in pregnancy complications. Front. Cell. Infect. Microbiol. 2022, 12, 1071029. [Google Scholar] [CrossRef] [PubMed]
  102. Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef] [PubMed]
  103. Rajilić-Stojanović, M.; de Vos, W.M. The first 1000 cultured species of the human gastrointestinal microbiota. FEMS Microbiol. Rev. 2014, 38, 996–1047. [Google Scholar] [CrossRef]
  104. Gao, B.; Shen, Q.; Wu, Y.; Cao, M.; Zhang, Q.; Chen, L. Branched-chain and aromatic amino acid levels response to an oral glucose load associated with gestational diabetes mellitus. Sci. Rep. 2022, 12, 12257. [Google Scholar] [CrossRef] [PubMed]
  105. Liu, Y.; Hou, Y.; Wang, G.; Zheng, X.; Hao, H. Gut Microbial Metabolites of Aromatic Amino Acids as Signals in Host-Microbe Interplay. Trends Endocrinol. Metab. TEM 2020, 31, 818–834. [Google Scholar] [CrossRef]
  106. Chen, T.; Qin, Y.; Chen, M.; Zhang, Y.; Wang, X.; Dong, T.; Chen, G.; Sun, X.; Lu, T.; White, R.A.; et al. Gestational diabetes mellitus is associated with the neonatal gut microbiota and metabolome. BMC Med. 2021, 19, 120. [Google Scholar] [CrossRef]
  107. American Diabetes Association. 14. Management of Diabetes in Pregnancy: Standards of Medical Care in Diabetes-2020. Diabetes Care 2020, 43, S183–S192. [Google Scholar] [CrossRef] [Green Version]
  108. Nie, Q.; Chen, H.; Hu, J.; Fan, S.; Nie, S. Dietary compounds and traditional Chinese medicine ameliorate type 2 diabetes by modulating gut microbiota. Crit. Rev. Food Sci. Nutr. 2019, 59, 848–863. [Google Scholar] [CrossRef]
  109. Mustad, V.A.; Huynh, D.T.T.; López-Pedrosa, J.M.; Campoy, C.; Rueda, R. The Role of Dietary Carbohydrates in Gestational Diabetes. Nutrients 2020, 12, 385. [Google Scholar] [CrossRef] [Green Version]
  110. Röytiö, H.; Mokkala, K.; Vahlberg, T.; Laitinen, K. Dietary intake of fat and fibre according to reference values relates to higher gut microbiota richness in overweight pregnant women. Br. J. Nutr. 2017, 118, 343–352. [Google Scholar] [CrossRef]
  111. Gomez-Arango, L.F.; Barrett, H.L.; Wilkinson, S.A.; Callaway, L.K.; McIntyre, H.D.; Morrison, M.; Dekker Nitert, M. Low dietary fiber intake increases Collinsella abundance in the gut microbiota of overweight and obese pregnant women. Gut Microbes 2018, 9, 189–201. [Google Scholar] [CrossRef] [Green Version]
  112. Barrett, H.L.; Gomez-Arango, L.F.; Wilkinson, S.A.; McIntyre, H.D.; Callaway, L.K.; Morrison, M.; Dekker Nitert, M. A Vegetarian Diet Is a Major Determinant of Gut Microbiota Composition in Early Pregnancy. Nutrients 2018, 10, 890. [Google Scholar] [CrossRef] [Green Version]
  113. Mandal, S.; Godfrey, K.M.; McDonald, D.; Treuren, W.V.; Bjørnholt, J.V.; Midtvedt, T.; Moen, B.; Rudi, K.; Knight, R.; Brantsæter, A.L.; et al. Fat and vitamin intakes during pregnancy have stronger relations with a pro-inflammatory maternal microbiota than does carbohydrate intake. Microbiome 2016, 4, 55. [Google Scholar] [CrossRef] [Green Version]
  114. Mokkala, K.; Röytiö, H.; Munukka, E.; Pietilä, S.; Ekblad, U.; Rönnemaa, T.; Eerola, E.; Laiho, A.; Laitinen, K. Gut Microbiota Richness and Composition and Dietary Intake of Overweight Pregnant Women Are Related to Serum Zonulin Concentration, a Marker for Intestinal Permeability. J. Nutr. 2016, 146, 1694–1700. [Google Scholar] [CrossRef] [Green Version]
  115. Portincasa, P.; Bonfrate, L.; Vacca, M.; De Angelis, M.; Farella, I.; Lanza, E.; Khalil, M.; Wang, D.Q.; Sperandio, M.; Di Ciaula, A. Gut Microbiota and Short Chain Fatty Acids: Implications in Glucose Homeostasis. Int. J. Mol. Sci. 2022, 23, 1105. [Google Scholar] [CrossRef]
  116. Allen, J.M.; Mailing, L.J.; Cohrs, J.; Salmonson, C.; Fryer, J.D.; Nehra, V.; Hale, V.L.; Kashyap, P.; White, B.A.; Woods, J.A. Exercise training-induced modification of the gut microbiota persists after microbiota colonization and attenuates the response to chemically-induced colitis in gnotobiotic mice. Gut Microbes 2018, 9, 115–130. [Google Scholar] [CrossRef] [Green Version]
  117. Gubert, C.; Kong, G.; Renoir, T.; Hannan, A.J. Exercise, diet and stress as modulators of gut microbiota: Implications for neurodegenerative diseases. Neurobiol. Dis. 2020, 134, 104621. [Google Scholar] [CrossRef]
  118. Allen, J.M.; Mailing, L.J.; Niemiro, G.M.; Moore, R.; Cook, M.D.; White, B.A.; Holscher, H.D.; Woods, J.A. Exercise Alters Gut Microbiota Composition and Function in Lean and Obese Humans. Med. Sci. Sport. Exerc. 2018, 50, 747–757. [Google Scholar] [CrossRef]
  119. Salminen, S.; Collado, M.C.; Endo, A.; Hill, C.; Lebeer, S.; Quigley, E.M.M.; Sanders, M.E.; Shamir, R.; Swann, J.R.; Szajewska, H.; et al. The International Scientific Association of Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of postbiotics. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 649–667. [Google Scholar] [CrossRef]
  120. Dolatkhah, N.; Hajifaraji, M.; Abbasalizadeh, F.; Aghamohammadzadeh, N.; Mehrabi, Y.; Abbasi, M.M. Is there a value for probiotic supplements in gestational diabetes mellitus? A randomized clinical trial. J. Health Popul. Nutr. 2015, 33, 25. [Google Scholar] [CrossRef] [Green Version]
  121. Karamali, M.; Dadkhah, F.; Sadrkhanlou, M.; Jamilian, M.; Ahmadi, S.; Tajabadi-Ebrahimi, M.; Jafari, P.; Asemi, Z. Effects of probiotic supplementation on glycaemic control and lipid profiles in gestational diabetes: A randomized, double-blind, placebo-controlled trial. Diabetes Metab. 2016, 42, 234–241. [Google Scholar] [CrossRef] [PubMed]
  122. Jafarnejad, S.; Saremi, S.; Jafarnejad, F.; Arab, A. Effects of a Multispecies Probiotic Mixture on Glycemic Control and Inflammatory Status in Women with Gestational Diabetes: A Randomized Controlled Clinical Trial. J. Nutr. Metab. 2016, 2016, 5190846. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Lindsay, K.L.; Brennan, L.; Kennelly, M.A.; Maguire, O.C.; Smith, T.; Curran, S.; Coffey, M.; Foley, M.E.; Hatunic, M.; Shanahan, F.; et al. Impact of probiotics in women with gestational diabetes mellitus on metabolic health: A randomized controlled trial. Am. J. Obstet. Gynecol. 2015, 212, 496.e1–496.e11. [Google Scholar]
  124. Taylor, B.L.; Woodfall, G.E.; Sheedy, K.E.; O’Riley, M.L.; Rainbow, K.A.; Bramwell, E.L.; Kellow, N.J. Effect of Probiotics on Metabolic Outcomes in Pregnant Women with Gestational Diabetes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2017, 9, 461. [Google Scholar] [CrossRef] [Green Version]
  125. Sotoudegan, F.; Daniali, M.; Hassani, S.; Nikfar, S.; Abdollahi, M. Reappraisal of probiotics’ safety in human. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2019, 129, 22–29. [Google Scholar] [CrossRef]
Figure 1. Roles of gut microbiota dysbiosis in GDM. Figure legend: AktSer473: Protein kinase BSerine473; IKK: Inhibitory B kinase; IRS1: Insulin receptor substrate-1; JNK: C-Jun N-terminal kinase; LPS: Lipopolysaccharide; NF-κB: Nuclear factor kappa-B; PI3-K: Phosphatidylinositol 3-kinase; TLR: Toll-like receptor. The gut barrier consists of mucus, intercellular tight junctions, immunoglobulins, antimicrobial peptides secreted by Paneth cells, microbes, and other components. Raises in Gram-negative pathobionts are linked to LPS biosynthesis. Elevated LPS and pathobionts can adhere to the mucosal layer, cross the epithelial layer of the gut via TLR2/4 activation, and translocate via phagocytosis and DC co-localization, ultimately entering systemic circulation and causing metabolic endotoxemia. Further, gut microbiota dysbiosis may result in thinning of the mucosal layer and disruption of the intercellular tight junctions in GDM patients. The compromised intestinal barrier can facilitate the migration of LPS and pathobionts. LPS and pathobionts bind to and activate TLR. TLR activation triggers macrophage infiltration and inflammation pathways, such as JNK, IKK, and NF-κB, which might evoke serine phosphorylation of IRS-1+Ser307, resulting in suppression of PI3-K and AktSer473. This procedure will impair insulin signaling and reduce glucose uptake in peripheral tissues, resulting in hyperglycemia in GDM women. The figure was created with BioRender.com. URL: https://biorender.com (accessed on 24 June 2023).
Figure 1. Roles of gut microbiota dysbiosis in GDM. Figure legend: AktSer473: Protein kinase BSerine473; IKK: Inhibitory B kinase; IRS1: Insulin receptor substrate-1; JNK: C-Jun N-terminal kinase; LPS: Lipopolysaccharide; NF-κB: Nuclear factor kappa-B; PI3-K: Phosphatidylinositol 3-kinase; TLR: Toll-like receptor. The gut barrier consists of mucus, intercellular tight junctions, immunoglobulins, antimicrobial peptides secreted by Paneth cells, microbes, and other components. Raises in Gram-negative pathobionts are linked to LPS biosynthesis. Elevated LPS and pathobionts can adhere to the mucosal layer, cross the epithelial layer of the gut via TLR2/4 activation, and translocate via phagocytosis and DC co-localization, ultimately entering systemic circulation and causing metabolic endotoxemia. Further, gut microbiota dysbiosis may result in thinning of the mucosal layer and disruption of the intercellular tight junctions in GDM patients. The compromised intestinal barrier can facilitate the migration of LPS and pathobionts. LPS and pathobionts bind to and activate TLR. TLR activation triggers macrophage infiltration and inflammation pathways, such as JNK, IKK, and NF-κB, which might evoke serine phosphorylation of IRS-1+Ser307, resulting in suppression of PI3-K and AktSer473. This procedure will impair insulin signaling and reduce glucose uptake in peripheral tissues, resulting in hyperglycemia in GDM women. The figure was created with BioRender.com. URL: https://biorender.com (accessed on 24 June 2023).
Microorganisms 11 01725 g001
Figure 2. Roles of SCFAs in GDM. Figure legend: Inadequate SCFAs may be associated with increased permeability of gut epithelium. Inadequate SCFAs also reduce adipose tissue lipid storage capacity, inhibit fatty acid oxidation, and increase lipolysis, which then raises serum FFA levels and increases lipid storage in the liver and muscle. Simultaneously, insufficient SCFAs may be unable to maintain the balance of anti-inflammatory and proinflammatory cells, leading to low-grade inflammation. Elevated serum FFA levels, increased lipid storage in the liver and muscle, and low-grade inflammation may all contribute to insulin resistance and hyperglycemia in women with GDM. On the other hand, excessive SCFA production may lead to extra energy harvesting capacity and FFA overflow, increasing lipid storage in the liver and skeletal muscle and leading to obesity. Excessive SCFAs may upregulate gluconeogenesis pathways and suppress glycolysis pathways and insulin signaling in peripheral tissues, resulting in hyperglycemia and insulin resistance. The figure was created with BioRender.com. URL: https://biorender.com (accessed on 24 June 2023).
Figure 2. Roles of SCFAs in GDM. Figure legend: Inadequate SCFAs may be associated with increased permeability of gut epithelium. Inadequate SCFAs also reduce adipose tissue lipid storage capacity, inhibit fatty acid oxidation, and increase lipolysis, which then raises serum FFA levels and increases lipid storage in the liver and muscle. Simultaneously, insufficient SCFAs may be unable to maintain the balance of anti-inflammatory and proinflammatory cells, leading to low-grade inflammation. Elevated serum FFA levels, increased lipid storage in the liver and muscle, and low-grade inflammation may all contribute to insulin resistance and hyperglycemia in women with GDM. On the other hand, excessive SCFA production may lead to extra energy harvesting capacity and FFA overflow, increasing lipid storage in the liver and skeletal muscle and leading to obesity. Excessive SCFAs may upregulate gluconeogenesis pathways and suppress glycolysis pathways and insulin signaling in peripheral tissues, resulting in hyperglycemia and insulin resistance. The figure was created with BioRender.com. URL: https://biorender.com (accessed on 24 June 2023).
Microorganisms 11 01725 g002
Figure 3. Treatment for GDM. Figure legend: CHO: Carbohydrates. Dietary restrictions, exercise, probiotics (prebiotics), and insulin may help alleviate symptoms of GDM. The figure was created with BioRender.com. URL: https://biorender.com (accessed on 24 June 2023).
Figure 3. Treatment for GDM. Figure legend: CHO: Carbohydrates. Dietary restrictions, exercise, probiotics (prebiotics), and insulin may help alleviate symptoms of GDM. The figure was created with BioRender.com. URL: https://biorender.com (accessed on 24 June 2023).
Microorganisms 11 01725 g003
Table 1. Characteristics of included studies of gut microbiota and related metabolic changes during GDM.
Table 1. Characteristics of included studies of gut microbiota and related metabolic changes during GDM.
Author, YearStudy DesignGroup SettingTimeMeasurementsConclusions
During the first trimester
Mokkala, 2017 [44]Case-control study15 Women who developed GDM VS. 60 healthy pregnant women10.4–15.4 weeks of gestation16S RNA gene sequencing, QIIME pipeline1. Ruminococcaceae family were increased in GDM women;
2. Ruminococcaceae family ≥26.8% predicted positive GDM diagnosis (sensitivity: 60%, specificity: 88%).
Ma, 2020 [46]Case-control study98 Women who developed GDM VS. 98 healthy pregnant women10–15 weeks of gestation16S rRNA microarray, Shotgun metagenomics sequencing1. Eisenbergiella, Tyzzerella 4, and Lachnospiraceae NK4A136 were enriched in the GDM group;
2. Eisenbergiella and Tyzzerella 4 were positively correlated with FBS.
Hu, 2021 [47]Case-control study201 Women who developed GDM VS. 201 matched controls6–15 weeks of gestation16S rRNA microarray1. Actinomyces, Adlercreutzia, Bifidobacterium, Coriobacteriaceae, Lachnospiraceae spp., and Rothia were reduced in GDM women;
2. Enterobacteriaceae, Ruminococcaceae spp., and Veillonellaceae were augmented in GDM women
3. Abundances of Staphylococcus relative to Clostridium, Coriobacteriaceae, and Roseburia were positively correlated with FBS and postprandial glucose levels.
During the second trimester
Kuang, 2017 [17]Case-control study43 GDM patients VS. 81 healthy pregnant women21–29 weeks of gestationWhole-metagenome shotgun sequencing1. P. distasonis, Klebsiella variicola, and Catenibacterium mitsuokai were elevated in women with GDM, which were positively associated with maternal glucose levels;
2. Alistipess spp., Bifidobacterium spp., Eubacterium spp., and Methanobrevibacter smithii were lowered.
Chen, 2021 [35]Case-control study30 GDM patients VS. 28 healthy pregnant women24–28 weeks of gestation16S rRNA microarray1. Bifidobacterium spp., and Eubacterium spp., were depleted in GDM patients;
2. Corynebacterium spp., Lactobacillus spp., and Blautia hydrogenotrophica were enriched in GDM patients.
Ye, 2019 [48]Case-control study24 GDM patients with successful glycemic control vs. 12 failure of glycemic control24–28 weeks of gestation16S rRNA sequencing1. The abundance of Blautia and Eubacterium hallii group was augmented in GDM patients who failed to control glycemic;
2. The richness of Faecalibacterium was reduced in GDM patients who failed to control glycemic.
Chen, 2021
[36]
Case-control study110 GDM patients VS. 220 healthy pregnant women22–24 weeks of gestation16S rRNA sequencing1. GDM patients had lower α-diversity that was significantly associated with glycemic;
2. Seven genera within the phylum Firmicutes and two within the phylum Actinobacteria were decreased in GDM patients;
3. Four genera within phylum Bacteroidetes were elevated in GDM patients.
Wei, 2022 [49]Cross-sectional study15 GDM patients VS. 18 healthy pregnant women24–28 weeks of gestation16S rRNA gene amplicon sequencing1. No difference in α-diversity;
2. Ruminococcus bromii, Clostridium colinum, and Streptococcus infantis were increased in GDM patients;
3. S. infantis were positively associated with blood glucose levels after adjusting for BMI.
Festa, 2020 [42]Cross-sectional study14 GDM patients VS. 15 healthy pregnant women matched on BMI24–28 weeks of gestation16S rRNA microarray1. No difference in β-diversity in women with GDM;
2. Abundances of Bacteroides caccae, Bacteroides massiliensis, and Bacteroides thetaiotaomicron were elevated in GDM patients;
3. Abundance of Bacteroides vulgatus, Eubacterium eligens, Lactobacillus rogosae, and Prevotella copri were suppressed in women with GDM.
Zhang, 2021 [39]Prospective cohort study128 GDM patients VS. 709 healthy pregnant women22–24 weeks of gestation16S rRNA gene sequencing1. Women who later progressed to GDM showed decreased α-diversity;
2. Genera belonging to Ruminococcaceae, Coriobacteriales, and Lachnospiraceae were decreased in GDM patients
Liang, 2022 [50]Case-control study35 GDM patients VS. 25 healthy pregnant women24–28 weeks of gestation16S rRNA gene sequencing1. Abundances of Bacteroides and Lachnoclostridium were elevated in GDM patients;
2. Ruminococcaceae UCG-002, Ruminococcaceae UCG-005, Clostridium sensu stricto 1, and Streptococcus were reduced in GDM patients;
3. Paraprevotella, Roseburia, Faecalibacterium, and Ruminococcaceae_UCG-002 were negatively correlated with glucose;
4. Ruminococcaceae_UCG-002 was negatively correlated with hemoglobin A1c;
5. Bacteroides was positively correlated with glucose;
6. Sutterella, Oscillibacter, and Bifidobacterium were positively correlated with GLP-1.
During the third trimester
Wang, 2018 [19]Cross-sectional study581 maternal and 248 neonatal samplesDuring the third trimester16S rRNA gene sequencingThe abundance of Faecalibacterium was reduced in GDM patients.
Crusel, 2018 [18]Case-control study50 GDM patients VS. 157 healthy pregnant womenDuring the third trimester and 8 months postpartum16S rRNA gene amplicon sequencing1. Actinobacteria at the phylum level and Collinsella, Desulfovibrio, and Rothia at the genus level were enriched in GDM women;
2. After adjustment for BMI, OTUs allocated to Akkermansia were associated with decreased insulin sensitivity, while Christensenella OTUs were associated with greater fasting plasma glucose concentration in GDM women;
3. Aberrant composition of the gut microbiome can be observed till 8 months after birth.
Cortez, 2019 [37]Cross-sectional study26 GDM patients VS. 42 healthy pregnant women28–36 weeks of gestationNext-generation sequencing1. Abundances of Firmicutes phylum were increased in the GDM group, while that of the Bacteroidetes phylum was increased in the control group;
2. GDM patients have a higher ratio of Firmicutes/Bacteroidetes.
Ferrocino, 2018 [52]Cohort study41 GDM patients24–28 weeks and at 38 weeks of gestation16S amplicon-based sequencingFirmicutes were enriched, and Bacteroidetes and Actinobacteria were decreased in GDM patients.
Xu, 2020 [38]Cross-sectional study30 GDM patients VS. 31 healthy pregnant womenDuring the third trimester16S rRNA sequencing1. GDM cases showed lower α-diversity;
2. Abundances of Gammaproteobacteria and Haemophilus were augmented in women with GDM
Li, 2021 [43]Cross-sectional study23 GDM patients VS. 29 healthy pregnant women>28 weeks of gestation16S rRNA sequencing1. Greater α-diversity in women with GDM;
2. A larger proportion of Firmicutes and a lower proportion of Bacteroides in women with GDM.
From the first trimester to the second trimester
Zheng, 2020 [53]Cross-sectional study31 GDM patients VS. 103 healthy pregnant womenDuring the first half of pregnancy16S rRNA gene amplicon sequencingCoprococcus and Streptococcus levels were consistently reduced in women with GDM.
BMI: body mass index, FBS: fasting blood glucose, rRNA: ribosomal ribonucleic 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

Hu, R.; Liu, Z.; Geng, Y.; Huang, Y.; Li, F.; Dong, H.; Ma, W.; Song, K.; Zhang, M.; Song, Y. Gut Microbiota and Critical Metabolites: Potential Target in Preventing Gestational Diabetes Mellitus? Microorganisms 2023, 11, 1725. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms11071725

AMA Style

Hu R, Liu Z, Geng Y, Huang Y, Li F, Dong H, Ma W, Song K, Zhang M, Song Y. Gut Microbiota and Critical Metabolites: Potential Target in Preventing Gestational Diabetes Mellitus? Microorganisms. 2023; 11(7):1725. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms11071725

Chicago/Turabian Style

Hu, Runan, Zhuo Liu, Yuli Geng, Yanjing Huang, Fan Li, Haoxu Dong, Wenwen Ma, Kunkun Song, Mingmin Zhang, and Yufan Song. 2023. "Gut Microbiota and Critical Metabolites: Potential Target in Preventing Gestational Diabetes Mellitus?" Microorganisms 11, no. 7: 1725. https://0-doi-org.brum.beds.ac.uk/10.3390/microorganisms11071725

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

Article Metrics

Back to TopTop