Authors: Javeria Zaheer
Journal Name: Microbiology Archives, an International Journal
DOI: https://doi.org/10.51470/MA.2023.5.1.12
Keywords: Gut microbiota, Type 2 diabetes, Insulin resistance, Microbial dysbiosis, Probiotics, Prebiotics, Inflammation, Metabolic disorders, Microbial therapeutics,
Abstract
The gut microbiota has emerged as a central regulator of host metabolism, influencing numerous physiological processes that govern energy balance, immune function, and glucose homeostasis. A growing body of evidence links dysbiosis—disruptions in microbial composition and functionality—to the onset and progression of insulin resistance and type 2 diabetes (T2D). Changes in microbial diversity and the abundance of key bacterial taxa can influence the production of metabolites such as short-chain fatty acids (SCFAs), bile acids, and lipopolysaccharides (LPS), all of which play crucial roles in modulating inflammation, gut permeability, and insulin signaling. This review delves into the multifaceted relationship between gut microbiota and metabolic dysfunction, focusing on how microbial imbalances contribute to chronic low-grade inflammation, altered lipid metabolism, and impaired glucose uptake. Special emphasis is placed on the mechanisms involving SCFA production, dysregulated bile acid profiles, LPS-driven endotoxemia, and compromised intestinal barrier function. These disruptions collectively create an environment conducive to insulin resistance and β-cell dysfunction, the current and emerging microbiota-centered therapeutic strategies aimed at restoring gut eubiosis. Interventions such as probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT) are gaining traction for their potential to improve metabolic parameters and insulin sensitivity. By modulating the gut ecosystem, these therapies offer promising adjuncts to conventional diabetes management, the intricate gut–metabolism axis not only enhances our knowledge of T2D pathogenesis but also paves the way for innovative, microbiome-informed approaches to disease prevention and treatment.
1. Introduction
Type 2 diabetes mellitus (T2DM) is a complex, multifactorial metabolic disorder marked by chronic hyperglycemia due to a combination of insulin resistance and β-cell dysfunction. Globally, the prevalence of T2DM is rising at an alarming pace, with recent estimates by the World Health Organization (WHO) indicating that over 500 million people are currently affected [1-2]. This surge is primarily linked to lifestyle and environmental factors—such as poor dietary habits, reduced physical activity, and increasing rates of obesity—although genetic predisposition also plays a contributory role.
However, genetic factors alone cannot explain the rapid and widespread increase in T2DM incidence, prompting researchers to investigate alternative and complementary contributors to disease pathogenesis. Among these, the gut microbiota has emerged as a critical area of interest. The human gastrointestinal tract harbors trillions of microorganisms, including bacteria, archaea, fungi, viruses, and protozoa, forming a highly dynamic and metabolically active ecosystem [3-4]. Often referred to as a “hidden organ,” the gut microbiota is increasingly recognized for its integral role in host metabolic regulation and overall health.
This microbial community contributes to a wide range of essential physiological processes, such as the fermentation of non-digestible dietary fibers, production of bioactive metabolites like short-chain fatty acids (SCFAs), synthesis of vitamins, regulation of immune homeostasis, maintenance of gut epithelial integrity, and modulation of host metabolism via the gut-brain and gut-liver axes [5]. Recent advances in microbiome research have revealed that alterations in the composition and function of the gut microbiota—a condition known as dysbiosis—may significantly contribute to the pathogenesis of metabolic diseases, including T2DM. Dysbiosis is typically characterized by reduced microbial diversity, a decrease in health-promoting commensals, and a rise in potentially pathogenic microbes [6-7]. Notably, studies have identified distinct microbial signatures in individuals with T2DM, including a lower abundance of beneficial bacteria such as Akkermansia muciniphila and Faecalibacterium prausnitzii, along with an increased presence of genera like Ruminococcus and Clostridium, which have been linked to pro-inflammatory states and metabolic dysfunction.
Several mechanisms have been proposed to elucidate how the gut microbiota influences insulin sensitivity and glucose metabolism. A primary pathway involves the production of microbial-derived short-chain fatty acids (SCFAs)—notably acetate, propionate, and butyrate—which are generated through the fermentation of dietary fibers in the colon. These SCFAs play essential roles in maintaining intestinal barrier integrity, modulating immune responses, and regulating host energy metabolism via interaction with G-protein coupled receptors (GPCRs), such as GPR41 and GPR43. Among these, butyrate is particularly significant, serving as the main energy source for colonocytes and demonstrating strong anti-inflammatory and insulin-sensitizing properties [6].
The Important mechanism centers on the gut microbiota’s regulation of systemic inflammation. Dysbiosis can compromise gut barrier function, resulting in increased intestinal permeability—commonly termed a “leaky gut.” This condition allows for the translocation of bacterial components, particularly lipopolysaccharides (LPS), into the systemic circulation. Elevated LPS levels induce chronic low-grade inflammation, known as metabolic endotoxemia, which disrupts insulin receptor signaling in peripheral tissues such as adipose tissue, liver, and skeletal muscle, thereby promoting insulin resistance, bile acid metabolism—closely regulated by gut microbes—has emerged as a vital link between the gut microbiota and metabolic homeostasis [7]. Through microbial biotransformation, primary bile acids are converted into secondary bile acids, which can activate nuclear receptors like the farnesoid X receptor (FXR) and the G-protein-coupled bile acid receptor (TGR5). These receptors play pivotal roles in the regulation of glucose and lipid metabolism. Disruption of this bile acid-microbiota axis has been associated with impaired glucose tolerance and the development of insulin resistance.
Diet is one of the most influential modulators of gut microbial composition and function. Western-style diets—characterized by high intake of fats and refined sugars and low consumption of dietary fiber—have been shown to rapidly disrupt microbial profiles, leading to a reduction in beneficial SCFA-producing bacteria. In contrast, dietary interventions that increase fiber intake and reduce saturated fat consumption have demonstrated positive effects on gut microbiota diversity and, consequently, metabolic health outcomes [8].
Understanding the gut microbiota’s role in the pathogenesis of type 2 diabetes mellitus (T2DM) opens new avenues for therapeutic intervention. Microbiota-targeted strategies—including probiotics, prebiotics, synbiotics, postbiotics, and fecal microbiota transplantation (FMT)—are under active investigation for their potential to restore microbial balance, enhance insulin sensitivity, and mitigate systemic inflammation. Although still in the early stages of clinical translation, these microbial-based therapies show promise as adjunctive or alternative approaches to conventional anti-diabetic treatments.
This review explores the intricate relationship between gut microbiota and T2DM, emphasizing the mechanisms through which microbial dysbiosis contributes to insulin resistance. Furthermore, we evaluate both current and emerging microbiome-modulating strategies as potential interventions for the prevention and management of T2DM [9]. A deeper understanding of the gut microbiome’s role may ultimately revolutionize the treatment of metabolic disorders, enabling the development of more personalized and effective therapeutic strategies.
Fig 1. This figure illustrates the intricate relationship between gut microbiota and insulin resistance in the development of Type 2 Diabetes Mellitus (T2DM). In the first panel, a healthy gut microbiota composition is shown, characterized by microbial diversity that supports glucose metabolism, enhances gut barrier function, and maintains anti-inflammatory responses. The second panel depicts gut dysbiosis commonly observed in T2DM patients, marked by a reduction in beneficial microbes and an increase in pathogenic bacteria, leading to increased intestinal permeability and systemic exposure to endotoxins such as lipopolysaccharides (LPS). The third panel highlights how these endotoxins contribute to systemic inflammation and impair insulin signaling pathways, thereby promoting insulin resistance. The final panel presents potential therapeutic strategies, including the use of probiotics, prebiotics, dietary modifications, and fecal microbiota transplantation, which aim to restore microbial balance, reduce inflammation, and improve insulin sensitivity in individuals with T2DM.
The human gut microbiota constitutes a densely populated and metabolically active microbial ecosystem that plays an indispensable role in regulating host metabolic functions. One of its most critical contributions is the fermentation of non-digestible carbohydrates—such as dietary fibers and resistant starches—into short-chain fatty acids (SCFAs), primarily acetate, propionate, and butyrate [10]. These SCFAs not only serve as major energy sources for colonocytes but also function as key signaling molecules that influence energy homeostasis, glucose metabolism, and immune responses [11].
SCFAs exert their metabolic effects through multiple pathways. A primary mechanism involves the activation of G-protein-coupled receptors (GPCRs), including GPR41 (also known as FFAR3) and GPR43 (FFAR2), which are expressed on intestinal epithelial cells, adipocytes, and various immune cells. The activation of these receptors stimulates the secretion of anorexigenic gut hormones such as glucagon-like peptide-1 (GLP-1) and peptide YY (PYY) [12]. These hormones play a pivotal role in metabolic regulation by enhancing insulin secretion, delaying gastric emptying, and suppressing appetite—ultimately contributing to improved glucose tolerance and energy balance.
butyrate, among the major SCFAs, plays a crucial role in maintaining intestinal barrier integrity by upregulating the expression of tight junction proteins. A well-functioning gut barrier prevents the translocation of microbial products such as lipopolysaccharide (LPS)—a pro-inflammatory endotoxin derived from Gram-negative bacteria—into systemic circulation. However, under conditions of dysbiosis, often triggered by high-fat and low-fiber diets, the abundance of SCFA-producing bacteria declines, compromising gut barrier function [13]. This condition, commonly referred to as “leaky gut,” facilitates the entry of LPS into the bloodstream, leading to metabolic endotoxemia. The resulting chronic low-grade inflammation is a well-recognized contributor to insulin resistance, particularly through the activation of Toll-like receptor 4 (TLR4) signaling pathways in adipose and hepatic tissues.
In addition to SCFAs, the gut microbiota plays a pivotal role in bile acid metabolism. Primary bile acids synthesized from cholesterol in the liver are released into the intestine for lipid digestion. Gut bacteria then deconjugate and convert these into secondary bile acids, which act as signaling molecules through nuclear receptors such as farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5). Activation of FXR and TGR5 regulates key metabolic processes, including hepatic gluconeogenesis, lipogenesis, and insulin sensitivity. Dysbiosis-induced alterations in the bile acid pool and signaling pathways can disrupt these metabolic processes, contributing to glucose intolerance.
Furthermore, gut microbes influence lipid metabolism by modulating host enzymes involved in fatty acid oxidation and lipogenesis. Certain microbial populations have been associated with increased hepatic triglyceride synthesis, promoting hepatic steatosis and features of the metabolic syndrome [14]. Additionally, microbial-derived metabolites such as trimethylamine-N-oxide (TMAO)—produced from dietary choline and carnitine—have been linked to cardiovascular disease risk and insulin resistance.
Altogether, the gut microbiota serves as a critical intermediary between diet and host metabolism. The balance of microbial species and their metabolic outputs has profound implications for energy homeostasis, inflammatory tone, and insulin sensitivity [15]. Disruptions in microbial composition and function, frequently observed in individuals with type 2 diabetes, impair these regulatory pathways, emphasizing the central role of the microbiome in metabolic disease progression. Understanding these complex host–microbe interactions provides valuable opportunities for the development of microbiota-targeted therapies aimed at restoring eubiosis and improving metabolic outcomes.
3. Dysbiosis and Insulin Resistance
The gut microbiota plays a pivotal role in regulating metabolic homeostasis. However, in metabolic disorders such as type 2 diabetes mellitus (T2DM), notable alterations in the composition and functional capacity of the gut microbial community—collectively termed dysbiosis—have been widely documented. This microbial imbalance has emerged as a key contributor to the development of insulin resistance, a central hallmark of T2DM. Multiple clinical and preclinical studies have shown that individuals with T2DM often exhibit reduced microbial diversity, a condition associated with decreased ecological stability and resilience of the gut ecosystem. Specifically, there is a marked decline in beneficial taxa such as Akkermansia muciniphila, Faecalibacterium prausnitzii, and Bifidobacterium spp., alongside an increased abundance of potentially pathogenic bacteria, including species from the Ruminococcus and Clostridium genera [16]. These compositional shifts compromise critical microbial functions that support host metabolism, thereby contributing to metabolic dysregulation.
A major downstream effect of dysbiosis is the disruption of intestinal barrier integrity. Under healthy conditions, the gut epithelium forms a selective barrier that permits nutrient absorption while preventing the translocation of microbial products. Beneficial bacteria such as A. muciniphila support this barrier by stimulating mucin production and upregulating tight junction protein expression. In the context of dysbiosis, the diminished presence of such microbes leads to increased intestinal permeability, often referred to as “leaky gut.” This compromised barrier permits the translocation of microbial components, notably lipopolysaccharides (LPS)—endotoxins from Gram-negative bacteria—into the systemic circulation.
Circulating LPS interacts with Toll-like receptor 4 (TLR4) on immune cells and metabolic tissues such as the adipose tissue and liver, initiating an inflammatory signaling cascade. This includes activation of the nuclear factor kappa B (NF-κB) pathway and subsequent release of pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6) [17]. This chronic, low-grade inflammation, also referred to as metabolic endotoxemia, plays a critical role in impairing insulin receptor signaling, thereby exacerbating insulin resistance and promoting the progression of T2DM.
Chronic inflammation exerts a detrimental impact on insulin receptor signaling, representing a key mechanistic link between gut dysbiosis and insulin resistance. Pro-inflammatory cytokines, such as TNF-α and IL-6, interfere with the insulin signaling cascade by promoting serine phosphorylation of insulin receptor substrate (IRS) proteins. This aberrant phosphorylation impairs downstream signaling through the phosphoinositide 3-kinase (PI3K)-Akt pathway, ultimately reducing glucose uptake by peripheral tissues and contributing to systemic insulin resistance [18]. In parallel, dysbiosis significantly influences lipid metabolism, promoting hepatic lipogenesis and fat accumulation, which are closely associated with insulin resistance and non-alcoholic fatty liver disease (NAFLD) or hepatic steatosis. Another critical consequence of dysbiosis is the reduced production of beneficial microbial metabolites, particularly butyrate, a key short-chain fatty acid (SCFA) with potent anti-inflammatory and gut-protective properties. Butyrate supports tight junction integrity, promotes mucosal healing, and regulates immune responses. A deficiency in butyrate-producing bacteria exacerbates intestinal permeability, fuels inflammation, and perpetuates the metabolic dysfunction seen in T2DM. Moreover, bile acid metabolism, which is intricately regulated by the gut microbiota, is significantly altered in dysbiosis. Microbial transformations of primary bile acids into secondary bile acids influence host metabolic regulation via nuclear receptors such as the farnesoid X receptor (FXR) and G protein-coupled bile acid receptor 1 (TGR5). These receptors play central roles in glucose homeostasis, lipid metabolism, and insulin sensitivity. Dysregulation of bile acid profiles and signaling pathways due to microbial imbalance further exacerbates metabolic disturbances and contributes to the development of insulin resistance. Taken together, the evidence strongly indicates that gut microbial dysbiosis contributes to insulin resistance through multiple interrelated mechanisms, including:
- Compromised intestinal barrier function
- Metabolic endotoxemia
- Chronic systemic inflammation
- Impaired insulin receptor signaling
- Altered production of microbial metabolites such as SCFAs and bile acids [19]
These insights underscore the potential of microbiota-targeted interventions—such as dietary modulation, probiotics, prebiotics, synbiotics, and fecal microbiota transplantation (FMT)—as novel therapeutic strategies for restoring microbial balance and mitigating metabolic dysfunction in type 2 diabetes mellitus.
4. Mechanisms Linking Gut Microbiota to Type 2 Diabetes Mellitus
A growing body of evidence supports the pivotal role of the gut microbiota in the development and progression of Type 2 Diabetes Mellitus (T2DM). Alterations in microbial composition and function—collectively referred to as dysbiosis—are associated with several pathophysiological mechanisms that contribute to insulin resistance and metabolic dysfunction. These mechanisms can be broadly categorized into four interrelated domains: inflammation and immune modulation, microbial metabolite alterations, gut barrier dysfunction, and bile acid signaling disruptions.
4.1 Inflammation and Immune Modulation
One of the central mechanisms linking gut microbiota dysbiosis to insulin resistance in T2DM is chronic low-grade inflammation, driven by innate immune activation. A key contributor to this inflammatory state is lipopolysaccharide (LPS), an endotoxin derived from the outer membrane of Gram-negative bacteria. Under healthy conditions, the intestinal epithelium acts as a selective barrier, preventing LPS from entering the systemic circulation. However, dysbiosis and increased intestinal permeability—often induced by high-fat, low-fiber diets—facilitate the translocation of LPS into the bloodstream, a condition referred to as metabolic endotoxemia.
Circulating LPS activates Toll-like receptor 4 (TLR4), which is expressed on immune cells and insulin-sensitive tissues such as adipose tissue, liver, and skeletal muscle. This activation triggers the NF-κB signaling cascade, resulting in the production and release of pro-inflammatory cytokines including tumor necrosis factor-alpha (TNF-α), interleukin-1β (IL-1β), and interleukin-6 (IL-6) [20]. These cytokines interfere with insulin signaling pathways by promoting serine phosphorylation of insulin receptor substrate (IRS) proteins, thereby inhibiting downstream activation of the PI3K-Akt pathway, a crucial mediator of glucose uptake and insulin sensitivity.
4.2. Short-Chain Fatty Acid (SCFA) Production
Short-chain fatty acids (SCFAs)—primarily butyrate, propionate, and acetate—are essential microbial metabolites produced through the fermentation of dietary fibers by specific gut bacteria, including Faecalibacterium prausnitzii, Roseburia, and Bifidobacterium species. Among these, butyrate stands out for its potent anti-inflammatory and insulin-sensitizing effects. It serves as the main energy source for colonocytes and plays a crucial role in maintaining gut barrier integrity by strengthening tight junctions. Butyrate also exerts immunomodulatory effects by promoting the differentiation of regulatory T cells (Tregs) and inhibiting the production of pro-inflammatory cytokines [21]. In addition to its local effects, SCFAs influence host metabolism systemically. They activate G-protein-coupled receptors (GPR41 and GPR43) on enteroendocrine and immune cells, stimulating the secretion of glucagon-like peptide-1 (GLP-1) and peptide YY (PYY)—hormones that enhance insulin secretion, improve glucose tolerance, and regulate appetite. In individuals with T2DM, reduced populations of SCFA-producing bacteria are commonly observed, resulting in decreased SCFA levels. This deficiency contributes to gut barrier dysfunction, elevated systemic inflammation, and impaired glucose metabolism, thereby exacerbating insulin resistance and disease progression.
4.3. Gut Barrier Integrity
The intestinal epithelial barrier serves as a critical line of defense, regulating the selective passage of nutrients while preventing the translocation of harmful microbial products into systemic circulation. This barrier is maintained by intercellular tight junction proteins—such as occludin and claudins—and is strongly supported by butyrate-producing bacteria. In individuals with type 2 diabetes mellitus (T2DM), microbial dysbiosis leads to a reduction in these beneficial microbial populations, thereby compromising the integrity of the epithelial barrier and increasing intestinal permeability, a condition commonly referred to as “leaky gut.”
As a result, lipopolysaccharide (LPS) and other microbial metabolites can translocate into the bloodstream, where they activate immune responses and promote chronic, low-grade systemic inflammation [22]. This inflammatory cascade further impairs insulin receptor signaling and exacerbates insulin resistance. Moreover, the loss of barrier function allows for increased antigen trafficking, which can trigger both innate and adaptive immune responses, perpetuating metabolic inflammation and contributing to the progression of T2DM.
4.4. Bile Acid Metabolism
The gut microbiota plays a pivotal role in shaping bile acid composition and modulating bile acid-mediated signaling pathways. Primary bile acids, synthesized from cholesterol in the liver, are transformed into secondary bile acids by specific microbial enzymes in the colon. These secondary bile acids function as signaling molecules by activating nuclear and membrane-bound receptors, including the farnesoid X receptor (FXR) and the G-protein-coupled bile acid receptor 1 (TGR5). Activation of FXR influences key metabolic processes such as hepatic gluconeogenesis, lipogenesis, and insulin sensitivity, while TGR5 signaling enhances glucagon-like peptide-1 (GLP-1) secretion, promotes energy expenditure, and attenuates inflammatory cytokine production.
In a dysbiotic state, the microbial conversion of bile acids becomes altered, resulting in an imbalanced bile acid pool and impaired receptor signaling. This disruption contributes to metabolic dysfunctions including glucose intolerance, hepatic steatosis, and systemic insulin resistance. These mechanisms highlight the complex and multifactorial role of the gut microbiota in regulating host metabolic pathways central to the pathogenesis of type 2 diabetes mellitus (T2DM) [9–12]. A deeper understanding of these interactions offers promising avenues for microbiota-targeted therapies—such as prebiotics, probiotics, and fecal microbiota transplantation (FMT)—in the prevention and management of T2DM.
5.1. Probiotics
Probiotics are defined as live microorganisms that, when administered in adequate amounts, confer health benefits to the host by positively influencing gut microbial composition and function. In the context of type 2 diabetes mellitus (T2DM), accumulating evidence from both clinical and preclinical studies supports the role of probiotic supplementation in improving metabolic outcomes, particularly through modulation of inflammation, gut barrier integrity, and glucose metabolism. Specific probiotic strains, especially those belonging to the genera Lactobacillus and Bifidobacterium, have demonstrated efficacy in enhancing gut epithelial barrier function by upregulating tight junction proteins and reducing systemic endotoxemia. For example, Lactobacillus casei and Bifidobacterium lactis have been shown to fortify intestinal barrier integrity and decrease circulating levels of lipopolysaccharides (LPS), thereby mitigating metabolic endotoxemia. Probiotics also influence the production of beneficial microbial metabolites, notably short-chain fatty acids (SCFAs) such as butyrate. Butyrate exerts potent anti-inflammatory and insulin-sensitizing effects by maintaining epithelial health, promoting regulatory T-cell differentiation, and modulating host energy metabolism. In addition, some probiotic strains enhance the secretion of incretin hormones, particularly glucagon-like peptide-1 (GLP-1), which stimulates insulin release, suppresses glucagon secretion, and improves postprandial glucose control, these mechanisms underscore the therapeutic potential of probiotics in modulating gut microbiota and alleviating insulin resistance in individuals with T2DM. However, strain-specific effects and interindividual variability necessitate further well-controlled clinical trials to identify optimal formulations and dosing regimens.
5.2. Prebiotics
Prebiotics are defined as non-digestible food components that selectively promote the growth and activity of beneficial gut microorganisms, particularly short-chain fatty acid (SCFA)-producing bacteria such as Bifidobacterium and Faecalibacterium. Commonly studied prebiotics include inulin, fructooligosaccharides (FOS), and galactooligosaccharides (GOS), which serve as fermentable substrates in the colon.
Prebiotic supplementation has been shown to enhance the production of SCFAs—especially butyrate and propionate—leading to improved intestinal epithelial integrity and reduced translocation of microbial endotoxins. These metabolites also exert anti-inflammatory effects by modulating immune cell function and downregulating pro-inflammatory cytokine production. Clinical trials have reported that prebiotics can improve glycemic parameters, such as fasting plasma glucose and insulin sensitivity (as measured by HOMA-IR), while simultaneously reducing circulating levels of tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6). Additionally, prebiotics may influence satiety and energy intake by modulating gut-brain axis signaling through incretin hormones and other neuroactive compounds.
5.3. Synbiotics
Synbiotics combine probiotics and prebiotics in a synergistic formulation aimed at enhancing the survival, colonization, and functional activity of beneficial microbes in the gastrointestinal tract. This dual approach provides both exogenous microbial strains and their preferred fermentable substrates, creating a favorable environment for microbial persistence and metabolic activity.
Studies have demonstrated that synbiotic supplementation—such as Lactobacillus acidophilus combined with inulin—can lead to significant improvements in metabolic markers among individuals with T2DM. These include better glycemic control, improved lipid profiles, enhanced antioxidant capacity, and reductions in markers of systemic inflammation. The synergistic effect of synbiotics also enhances SCFA production and gut barrier function, promoting a more robust and balanced microbial ecosystem. Compared to probiotics or prebiotics alone, synbiotics may offer greater therapeutic efficacy due to their dual mechanism of action.
5.4. Dietary Interventions
Dietary patterns play a foundational role in shaping gut microbiota composition and metabolic health. Diets rich in fermentable fibers, complex carbohydrates, and polyphenols—such as the Mediterranean and DASH (Dietary Approaches to Stop Hypertension) diets—have been consistently associated with increased microbial diversity and abundance of beneficial taxa, including Akkermansia muciniphila, Roseburia, and Faecalibacterium prausnitzii.
These plant-based dietary patterns promote SCFA production, support epithelial barrier function, and exert anti-inflammatory effects, all of which contribute to improved insulin sensitivity and glycemic control. Evidence from cohort studies and randomized controlled trials indicates that adherence to high-fiber diets leads to reduced HbA1c levels, improved postprandial glucose responses, and lower systemic inflammation in T2DM populations. Conversely, diets high in saturated fats, refined sugars, artificial sweeteners, and ultra-processed foods are associated with dysbiosis, impaired microbial resilience, and increased risk of metabolic dysfunction. Thus, long-term dietary modulation remains a cornerstone strategy for microbiota-directed management of type 2 diabetes.
5.5. Fecal Microbiota Transplantation (FMT)
Fecal microbiota transplantation (FMT) entails the transfer of processed fecal material from a healthy donor into the gastrointestinal tract of a recipient to reestablish a balanced gut microbial community. Initially established as an effective treatment for recurrent Clostridioides difficile infection, FMT has garnered attention as a potential therapeutic strategy for metabolic disorders, including type 2 diabetes mellitus (T2DM). Emerging clinical studies suggest that FMT from lean, metabolically healthy donors to individuals with metabolic syndrome or T2DM may transiently improve insulin sensitivity, increase gut microbial diversity, and enhance the production of beneficial metabolites such as short-chain fatty acids (SCFAs). Notably, post-FMT elevations in taxa such as Akkermansia muciniphila and Bacteroides spp. have been positively associated with improved glucose homeostasis and reduced systemic inflammation. However, the long-term efficacy, safety, and reproducibility of FMT remain under investigation. Critical challenges include variability in donor microbiota composition, lack of standardized transplantation protocols, and the potential for adverse events. Moreover, the durability of beneficial microbial colonization post-FMT is uncertain, and results across studies are inconsistent.While FMT represents a promising avenue for microbiota-based intervention in T2DM, it remains an experimental approach requiring rigorous clinical validation. Future advancements may involve the development of defined microbial consortia or next-generation probiotics that offer the benefits of FMT with greater safety and standardization.
6. Future Directions and Challenges
The growing promise of gut microbiota-targeted strategies in the management of T2DM, several critical challenges must be addressed to facilitate their translation into routine clinical practice. One major obstacle is the pronounced inter-individual variability in microbiota composition, influenced by genetics, diet, environment, and medication use. This heterogeneity results in differential responses to interventions such as probiotics, prebiotics, dietary changes, and FMT. Another challenge lies in the limited colonization and persistence of administered microbes, which may reduce the long-term efficacy of microbial therapies. Additionally, the lack of large-scale, randomized, and long-duration clinical trials hinders the establishment of definitive evidence for the safety, consistency, and sustainability of these approaches.
Future research should prioritize the integration of multi-omics technologies—such as metagenomics, metabolomics, proteomics, and transcriptomics—combined with advanced bioinformatics and machine learning. These platforms can facilitate the development of personalized therapeutic regimens by identifying individual microbial signatures and predicting treatment responses. Furthermore, regulatory guidelines and quality control measures are needed to ensure the safety and standardization of microbiome-based products and protocols. Precision microbiome modulation, tailored to each patient’s unique gut ecosystem and metabolic profile, holds transformative potential for T2DM prevention and treatment. However, realizing this potential will require a multidisciplinary effort involving microbiology, clinical medicine, systems biology, and computational modeling.
Conclusion
The gut microbiota plays a critical role in the pathogenesis of insulin resistance and type 2 diabetes mellitus (T2DM), primarily by modulating metabolic, inflammatory, and hormonal pathways. Growing evidence supports that dysbiosis—an imbalance in microbial composition—contributes to chronic systemic inflammation, impaired glucose metabolism, and diminished insulin sensitivity. Conversely, restoring a healthy gut microbial ecosystem offers significant therapeutic potential.
Microbiota-targeted interventions—including probiotics, prebiotics, synbiotics, dietary strategies, and fecal microbiota transplantation (FMT)—have shown varying degrees of success in enhancing glycemic control, improving insulin sensitivity, and attenuating low-grade inflammation. These approaches represent promising, non-pharmacological adjuncts to conventional diabetes management, with the potential to address disease mechanisms at their origin, the clinical translation of these strategies remains in its infancy. Challenges such as inter-individual variability in microbiome composition, inconsistent therapeutic outcomes, and the lack of long-term safety and efficacy data must be addressed. To optimize outcomes, future research should focus on personalized approaches supported by multi-omics integration—metagenomics, metabolomics, transcriptomics—and large-scale, longitudinal clinical trials. Modulation of the gut microbiota stands at the frontier of innovative therapeutic strategies for T2DM. Realizing its full potential will require interdisciplinary collaboration across microbiology, nutrition science, endocrinology, and systems biology, paving the way for precision medicine in metabolic disease prevention and treatment.
- Han, J. L., & Lin, H. L. (2014). Intestinal microbiota and type 2 diabetes: from mechanism insights to therapeutic perspective. World journal of gastroenterology: WJG, 20(47), 17737.
- Diamant, M., Blaak, E. E., & De Vos, W. M. (2011). Do nutrient–gut–microbiota interactions play a role in human obesity, insulin resistance and type 2 diabetes?. Obesity reviews, 12(4), 272-281.
- Burcelin, R., Serino, M., Chabo, C., Blasco-Baque, V., & Amar, J. (2011). Gut microbiota and diabetes: from pathogenesis to therapeutic perspective. Acta diabetologica, 48, 257-273.
- Zhang, L., Chu, J., Hao, W., Zhang, J., Li, H., Yang, C., … & Wang, H. (2021). Gut microbiota and type 2 diabetes mellitus: Association, mechanism, and translational applications. Mediators of inflammation, 2021(1), 5110276.
- Adeshirlarijaney, A., & Gewirtz, A. T. (2020). Considering gut microbiota in treatment of type 2 diabetes mellitus. Gut microbes, 11(3), 253-264.
- Shen, J., Obin, M. S., & Zhao, L. (2013). The gut microbiota, obesity and insulin resistance. Molecular aspects of medicine, 34(1), 39-58.
- Liu, L., Zhang, J., Cheng, Y., Zhu, M., Xiao, Z., Ruan, G., & Wei, Y. (2022). Gut microbiota: A new target for T2DM prevention and treatment. Frontiers in endocrinology, 13, 958218.
- Jiang, H., Cai, M., Shen, B., Wang, Q., Zhang, T., & Zhou, X. (2022). Synbiotics and gut microbiota: new perspectives in the treatment of type 2 diabetes mellitus. Foods, 11(16), 2438.
- Delzenne, N. M., Cani, P. D., Everard, A., Neyrinck, A. M., & Bindels, L. B. (2015). Gut microorganisms as promising targets for the management of type 2 diabetes. Diabetologia, 58, 2206-2217.
- Zhou, Z., Sun, B., Yu, D., & Zhu, C. (2022). Gut microbiota: an important player in type 2 diabetes mellitus. Frontiers in cellular and infection microbiology, 12, 834485.
- Harsch, I. A., & Konturek, P. C. (2018). The role of gut microbiota in obesity and type 2 and type 1 diabetes mellitus: new insights into “old” diseases. Medical sciences, 6(2), 32.
- Wang, H., Lu, Y., Yan, Y., Tian, S., Zheng, D., Leng, D., … & Bai, Y. (2020). Promising treatment for type 2 diabetes: fecal microbiota transplantation reverses insulin resistance and impaired islets. Frontiers in cellular and infection microbiology, 9, 455.
- Brunkwall, L., & Orho-Melander, M. (2017). The gut microbiome as a target for prevention and treatment of hyperglycaemia in type 2 diabetes: from current human evidence to future possibilities. Diabetologia, 60(6), 943-951.
- Ortega, M. A., Fraile-Martínez, O., Naya, I., García-Honduvilla, N., Álvarez-Mon, M., Buján, J., & de la Torre, B. (2020). Type 2 diabetes mellitus associated with obesity (diabesity). The central role of gut microbiota and its translational applications. Nutrients, 12(9), 2749.
- Huda, M. N., Kim, M., & Bennett, B. J. (2021). Modulating the microbiota as a therapeutic intervention for type 2 diabetes. Frontiers in endocrinology, 12, 632335.
- Bielka, W., Przezak, A., & Pawlik, A. (2022). The role of the gut microbiota in the pathogenesis of diabetes. International journal of molecular sciences, 23(1), 480.
- Zhai, L., Wu, J., Lam, Y. Y., Kwan, H. Y., Bian, Z. X., & Wong, H. L. X. (2021). Gut-microbial metabolites, probiotics and their roles in type 2 diabetes. International Journal of Molecular Sciences, 22(23), 12846.
- Tanase, D. M., Gosav, E. M., Neculae, E., Costea, C. F., Ciocoiu, M., Hurjui, L. L., & Serban, I. L. (2020). Role of gut microbiota on onset and progression of microvascular complications of type 2 diabetes (T2DM). Nutrients, 12(12), 3719.
- Chen, Z., Radjabzadeh, D., Chen, L., Kurilshikov, A., Kavousi, M., Ahmadizar, F., & Voortman, T. (2021). Association of insulin resistance and type 2 diabetes with gut microbial diversity: a microbiome-wide analysis from population studies. JAMA network Open, 4(7), e2118811-e2118811.
- Pascale, A., Marchesi, N., Govoni, S., Coppola, A., & Gazzaruso, C. (2019). The role of gut microbiota in obesity, diabetes mellitus, and effect of metformin: new insights into old diseases. Current opinion in pharmacology, 49, 1-5.
- Ejtahed, H. S., Soroush, A. R., Angoorani, P., Larijani, B., & Hasani-Ranjbar, S. (2016). Gut microbiota as a target in the pathogenesis of metabolic disorders: a new approach to novel therapeutic agents. Hormone and Metabolic Research, 48(06), 349-358.
- Zheng, Y., Gou, X., Zhang, L., Gao, H., Wei, Y., Yu, X., & Li, M. (2020). Interactions between gut microbiota, host, and herbal medicines: a review of new insights into the pathogenesis and treatment of type 2 diabetes. Frontiers in Cellular and Infection Microbiology, 10, 360.
- Delzenne, N. M., & Cani, P. D. (2011). Gut microbiota and the pathogenesis of insulin resistance. Current diabetes reports, 11, 154-159.
- Panwar, H., Rashmi, H. M., Batish, V. K., & Grover, S. (2013). Probiotics as potential biotherapeutics in the management of type 2 diabetes–prospects and perspectives. Diabetes/metabolism research and reviews, 29(2), 103-112.