Next Article in Journal
Early Driving Pressure Is Associated with Major Adverse Kidney Events at 30 Days in ARDS Patients with SARS-CoV-2
Previous Article in Journal
Arthroscopic Flexor Hallux Brevis and Plantar Capsule Release (Cochrane Procedure) for Hallux Rigidus: Case Presentation with Long-Term Follow-Up
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Intestinal Microbiota Composition in Patients with Type 2 Diabetes and Effects of Oral Antidiabetics

by
Ahmet Toygar Kalkan
1,*,
Goknur Yorulmaz
1,
Aysen Akalin
1 and
Ener Cagri Dinleyici
2
1
Faculty of Medicine, Department of Endocrinology, Eskisehir Osmangazi University, Eskisehir 26040, Türkiye
2
Faculty of Medicine, Department of Pediatrics, Eskisehir Osmangazi University, Eskisehir 26040, Türkiye
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2025, 14(8), 2786; https://doi.org/10.3390/jcm14082786
Submission received: 12 March 2025 / Revised: 14 April 2025 / Accepted: 15 April 2025 / Published: 17 April 2025
(This article belongs to the Section Endocrinology & Metabolism)

Abstract

:
Introduction: The cause–effect relationships between microbiota composition changes and type 2 diabetes (T2D) are complex, likely involving two-way interactions, and require further elucidation. Few studies have examined the interactions of antidiabetic drugs with the gut microbiota. This study’s goal was to evaluate the gut microbiota of patients with type 2 diabetes at first diagnosis and again after 12 weeks of taking oral antidiabetic drugs. Methods: We performed a fecal microbiota analysis of adult patients who recently received a T2D diagnosis and healthy adults. We compared the microbiota compositions between the T2D patients and healthy controls; we also evaluated changes from baseline to 12 weeks of treatment in the total group receiving oral antidiabetics, as well as in the subgroups receiving metformin and linagliptin. Results: The alpha diversity and beta diversity indices were different at baseline between patients with type 2 diabetes and healthy controls. The LEfSe analysis showed that, at the genus level, the Lactobacillus, Rothia, Collinsella, and Eubacterium genera increased in relative abundance in the T2D group while, at the species level, the Rothia mucilaginosa, Collinsella aerofaciens, and Eubacterium bioforme strains were found to be dominant in the T2D group. Faecalibacterium at the genus level and Faecalibacterium prausnitzii at the strain level increased in relative abundance in the T2D group after 12 weeks. After 12 weeks of intervention, the alpha diversity indices were significantly lower in the T2D group compared to the control group. At the end of the 12th week, the Gemmiger and Collinsella genera were dominant in the T2D group, with Gemmiger formicilis and Collinsella aerofaciens being dominant at the species level; in the control group, Bacteroides and Alistipes were dominant at the genus level, and Prevotella stercorea and Alistipes finegoldii were dominant. There was no difference in the LEfSe analysis results between baseline and 12 weeks of linagliptin treatment. At the strain level, Gemmiger formicilis, Ruminococcus bromii, Rothia mucilaginosa, and Lactobacillus ruminis were predominant at the start of metformin treatment; however, after 12 weeks, Collinsella aerofaciens became predominant. Conclusions: We report that there is a substantial change in the composition of the gut microbiota in patients with T2D. Oral antidiabetic treatments, especially metformin, have some beneficial effects on microbiota composition. Few studies have explored the interaction of antidiabetic drugs with the gut microbiota; further research will elucidate the clinical impact of these changes in gut microbiota composition in diabetes.

1. Introduction

The human gut microbiota has recently emerged as having a significant influence on various diseases, particularly non-communicable diseases [1]. The gut microbiota has been widely associated with the onset of obesity and its related conditions, including type 2 diabetes (T2D) and insulin resistance [2]. Furthermore, data indicate that T2D can alter the composition of the gut microbiota. The causal links between dysbiosis and T2D are intricate and require further clarification. They likely form a bidirectional interaction [3,4,5]. A crucial aspect to consider when examining the involvement of the gut microbiota in T2D is the potential confounding influence of diabetes medications [6,7,8]. Antidiabetic drugs may influence the gut microbiota to enhance glucose homeostasis; hence, the gut microbiota could further impact the effectiveness of pharmacological treatment [3,9]. Recent studies suggest that the gut microbiota mediates the glucose-lowering effects of metformin, influencing the composition and functionality of the microbiome and augmenting the alterations associated with T2D [10,11,12]. Metformin enhances glucose homeostasis by modulating the gut microbiota, potentially exerting glucose-lowering effects through various mechanisms, including regulating intestinal glucose absorption, promoting the proliferation of short-chain fatty acid-producing bacteria that generate butyrate and propionate, stimulating the growth of mucin-degrading bacteria, augmenting the secretion of the gut hormone glucagone-like peptide-1 (GLP-1), regulating bile acid metabolism, and preserving the integrity of the intestinal barrier [3,10].
Besides metformin, various antidiabetic medications with distinct mechanisms of action are available. Limited research has investigated the relationships between these various antidiabetic medications and the gut microbiota [3,13,14]. With results comparable to those for metformin, existing research has suggested that the gut microbiota may serve as a potential modulator of these drugs. Dipeptidyl peptidase-4 (DPP-4) inhibitors, a new category of oral medications for T2D management, can successfully synergize with existing antidiabetic therapies to attain blood glucose targets [15]. There is limited information about the effects of DPP-4 inhibitors on microbiota composition [16,17].
In this study, we aimed to evaluate the intestinal microbiota composition at baseline and at week 12 in subjects diagnosed with T2D, receiving metformin or linagliptin, and to compare the groups within themselves and with healthy controls.

2. Materials and Methods

This was a case–control study involving patients with T2D, conducted from 2020 to 2021 at the Endocrinology and Metabolism Unit of Eskisehir Osmangazi University Faculty of Medicine in Türkiye. This research received financial support from the Eskişehir Osmangazi University Scientific Research Projects (TSA-2021-1587).

2.1. Inclusion and Exclusion Criteria

This study was planned to include adult patients older than 18 years with T2D and HbA1c levels of ≥6.5 and ≤10.0%, who had not received any glucose-lowering drug in the previous 12 weeks, who were followed up in the Endocrinology Department. We followed up with the patients with a single antidiabetic drug for a period of three months. Children, adolescents, and those with a body mass index (BMI) of >40, previous use of antibiotics or probiotics within the last 8 weeks, inflammatory or functional bowel disease, a history of gastrointestinal cancer or surgery (including bariatric surgery), acute or severe gastrointestinal symptoms requiring medical treatment, or a previous history of acute coronary syndrome or stroke were excluded from this study. The control group comprised persons, matched in age and gender to the research group, who were healthy, had not used antibiotics or probiotics in the preceding eight weeks and met the inclusion criteria. The serum HbA1c levels in the control group were within the normal limit.
We noted the patients’ age, gender, and clinical characteristics after obtaining their consent. The study team organized their treatment based on their disease status; eligible patients received metformin or linagliptin. The same study team in the Division of Endocrinology closely monitored the patients and performed clinical evaluations at the end of the 12th week. The study team asked patients to contact them if they needed any medication, especially antibiotics, during the 12-week period. Patients also received a diet and exercise counseling plan. The metformin dose was 2000 mg/day for 12 weeks of monotherapy, and the linagliptin dose was 5 mg once daily for 12 weeks of monotherapy.

2.2. Endpoints of This Study

The primary endpoint of this study was to evaluate the change in microbiota composition in T2D patients from baseline to the end of the 12th week and to compare this change with that in healthy controls. The secondary endpoint involved comparing the microbiota compositions of patients with those of healthy controls both at baseline and at the end of the 12th week. Additionally, it involved comparing the changes in intestinal microbiota composition after 12 weeks of metformin treatment or 12 weeks of linagliptin treatment to those observed for the healthy controls.

2.3. Fecal Samples and Microbiota Analysis

At least 5 mL of fresh stool sample was collected on Day 0 of this study and again 12 weeks later in a 15 mL Falcon tube. It was quickly frozen at −80 °C and kept upright until DNA extraction. At the end of the extraction process, an average of 50–60 ng of genomic DNA was obtained, diluted with 200 µL of CDT (elution buffer). We performed bacterial 16S ribosomal RNA (rRNA) gene target sequencing from the genetic materials obtained in our study. We amplified the genomic DNA using 16S V3-V4 314F-860R primer sets and prepared the libraries using the Nextera XT DNA library preparation kit and indexes (Illumina, San Diego, CA, USA). We cleaned the Amplicon libraries using AMPure XP from Beckman Coulter, carefully selecting large fragments. After preparing the library, we ran the sequencing on a NovoSeq 6000 (Illumina).

2.4. Bioinformatic and Statistical Analysis

We transferred paired-end Illumina reads (2 × 250) to Qiime2 media [18]. All the samples had an array depth exceeding 100X, and we did not exclude any sample from this study. Amplicon sequences with a low Phred score (<Q30) and chimeric amplicon sequences were identified and removed from the system via the Qiime2 Dada2 program (q2-dada2). We mapped the Amplicon Sequence Variants (ASVs) generated by Dada2 to the GreenGenes (/greengenes.lbl.gov (accessed on 8 February 2022)) database [19,20]. The R 4.1 environment created the Phyloseq object from the qiime2 structure files [21,22]. We interpreted the alpha diversity assessment, which evaluates the diversity of taxonomic units of interest in a sample, using three different indices: Chao1, Shannon, and Simpson. We calculated the p-values between groups using the Kruskal–Wallis test. We used a beta diversity analysis to assess taxonomic differences between individuals based on Jaccard, Bray–Curtis, weighted UniFrac, and unweighted UniFrac calculations. We determined specific differences between groups using a differential abundance analysis and the Deseq2 R package [23]. A Linear Discriminant Analysis Effect Size (LEFSe) analysis was used to find statistically significant (LDA threshold value > 2, p < 0.05) taxonomies among the groups [24]. We used the JASP package program to statistically analyze the patient and control groups’ demographic and laboratory characteristics. Spearman correlation tests were performed for the correlation between serum HbA1c levels and alpha diversity indices. p < 0.05 was considered to indicate statistical significance.

3. Results

In this case–control study, 44 T2D patients who met the inclusion criteria were included. Twelve patients were excluded because of the combined treatment requirement. Regarding the oral antidiabetic agent, the study population was subdivided into groups receiving linagliptin (n = 16) or metformin (n = 16). Some patients were excluded from the full analysis set due to protocol violations (not completing 12 weeks of treatment: metformin, n = 5; linagliptin, n = 3). After 12 weeks of follow-up, 24 T2D patients (14 men, 10 women, mean age 54.5 ± 8.7 years, 42–72 years) were considered eligible for the post-treatment evaluation. The control group consisted of 10 age- and gender-matched healthy adults (5 men, 5 women; mean age 50.9 ± 5.0 years, 45–61 years). The demographic and clinical characteristics were similar between the metformin and linagliptin groups and the healthy controls at baseline.
At baseline, the mean HbA1c levels were 8.21 ± 1.71, min–max 6.7–9.9, in the patient group. At 12 weeks of intervention, the mean HbA1c levels were significantly lower (6.81 ± 0.78, min–max 5.7–8.5) in the T2D group compared to the baseline group (p < 0.001). The 24 T2D patients included in this study were then evaluated separately with regard to the treatment received. Among them, 13 patients (7 men, 6 women; mean age 57.3 ± 10.1 years, 42–72 years) were treated with linagliptin, and 11 patients (7 men, 4 women; mean age 51.0 ± 4.7 years, 45–61 years) were treated with metformin as monotherapy oral antidiabetics. At baseline, the mean HbA1c level was 7.53 ± 1.29 (min–max 5.9–9.8) in the linagliptin group and 8.59 ± 1.06 (min–max 6.2–10) in the metformin group. At 12 weeks of intervention, the mean HbA1c level was 6.95 ± 0.94 (min–max 5.7–8.5) in the linagliptin group and 6.64 ± 0.52 (min–max 5.85–7.6) in the metformin group. The serum HbA1c levels were significantly lower at 12 weeks of intervention compared with the baseline levels for both the linagliptin and metformin groups (p < 0.05 and p < 0.01, respectively).

3.1. Comparison of Microbiota Compositions of T2D Patients and Healthy Controls Before Treatment (Baseline)

In our study, the observed OTUs, Chao1 index, Simpson index, and Shannon index were used to evaluate the alpha diversity. At baseline, the median value of the Chao1 index was lower (p < 0.05), and the Shannon index was lower without statistical significance in the T2D group than in the control group. The median number of observed OTUs in the T2D group was lower than that in the control group (p < 0.05). The Bray–Curtis, Jaccard, weighted UniFrac, and unweighted UniFrac principal coordinate analysis (PCoA) results for T2D patients were different from those for the control group (p < 0.05). There was no correlation between the HbA1c levels and alpha diversity index parameters (p > 0.05).
At the phylum level, Firmicutes 61.4%, Bacteroidetes 27.4%, Proteobacteria 5.9%, Actinobacteria 2.7%, and Verrucomicrobia 1.4% were found in the T2D group, while Firmicutes 73.6%, Bacteroidetes 17.4%, Proteobacteria 4.0%, and Actinobacteria 3.4% were found in the healthy adult group. While the Firmicutes phylum was predominant in healthy adults (p < 0.001), Bacteroidetes (p < 0.001) and Tenericutes (p < 0.001) were higher in relative abundance in the T2D group compared to healthy adults. At the genus level, Faecalibacterium 18.2%, Prevotella 11.4%, Dialister 11.2%, Bacteroides 10.2%, Oscillospira 7.5%, Ruminococcus 5%, and Gemmiger 5% were dominant in the T2D group, while Faecalibacterium 22.1%, Bacteroides 16.5%, Prevotella 14.2%, Dialister 8.3%, Oscillospira 6.4%, Ruminococcus 5.1%, Succinovibrio 4.1%, Gemmiger 2.9%, Alistipes 2.4%, and Lachnospira 2.0% were dominant in healthy controls (Figure 1). In the T2D group compared to healthy adults, Ruminococcus (p < 0.0001), Blautia (p < 0.001), Bacteroides (p < 0.001), and Bifidobacterium (p < 0.05) were predominant at the genus level, and at the strain level, Ruminococcus bromii (p < 0.0001), Prevotella copri (p < 0.0001), Collinsella aerofaciens (p < 0.0001), Veillonella dispar (p < 0.0001), Eubacterium bioforme (p < 0.0001), Dorea formigenerans (p < 0.001), Lactobacillus ruminis (p < 0.001), Rothia mucilaginosa (p < 0.0001), and Gemmiger focilis (p < 0.0001) were higher in relative abundance. In the healthy control group, Faecalibacterium at the genus level (p < 0.05) and Ruminococcus callidus, Bacteroides uniformis, and Faecalibacterium prausnitzii at the strain level (p < 0.05) were statistically significantly higher in relative abundance than they were in the T2D group (Figure 1). The LEfSe analysis showed that, in the T2D group, at the genus level, Lactobacillus, Rothia, Collinsella, and Eubacterium increased in relative abundance, and at the species level, the Rothia mucilaginosa, Collinsella aerofaciens, and Eubacterium bioforme strains were found to be dominant.

3.2. Comparison of Microbial Compositions in Patients with T2D at Admission and 12 Weeks Later

In the T2D group, no statistically significant differences were found for the alpha and beta diversity indices at baseline and at the end of the 12th week (p > 0.05 for all). While Faecalibacterium at the genus level and Faecalibacterium prausnitzii at the strain level increased in relative abundance in the T2D group after 12 weeks of follow-up, the Succinovibrio genus was more dominant at the beginning of this study compared to the end of the 12th week. The LEfSe analysis results at baseline and at the end of the 12th week in T2D patients are shown in Figure 2.

3.3. Comparison of Microbial Compositions of T2D Patients and Healthy Controls at 12-Week Follow-Up

The median values of the Chao1 index, Shannon index, and observed number of OTUs were significantly lower in the T2D group compared to the control group at the beginning of this study and at the end of the 12th week (p < 0.01 for all three indices). There were no significant differences in the beta diversity index results (p > 0.05).
The LEfSe analysis results for the T2D cases and healthy controls after 12 weeks of follow-up are shown in Figure 2. At the end of the 12th week, in the T2D group, the Gemmiger and Collinsella genera were dominant, and Gemmiger formicilis and Collinsella aerofaciens were dominant at the species level; in the control group, Bacteroides and Alistipes were dominant at the genus level, and Prevotella stercorea and Alistipes finegoldii were the dominant species.

3.4. Metformin and Linagliptin Groups

At the beginning of treatment, there was no difference between the linagliptin group and the metformin group in terms of their alpha and beta diversity parameters (p > 0.05). In the linagliptin group, there was a decrease in the median values of the Shannon and Chao-1 indices between the baseline and the 12th week (p < 0.05 and p < 0.01). In the metformin group, there was no difference in the median values of the Shannon and Chao-1 indices between the baseline and the 12th week (p > 0.05).
The genus-level microbiota compositions of the linagliptin and metformin subgroups of T2D patients at the beginning of treatment and at the end of 12 weeks, in comparison with that of healthy controls, are shown in Figure 3.
There was no difference in the LEfSe analysis between baseline and 12 weeks of linagliptin treatment. When the microbiota composition was compared with that of healthy controls at the 12th week of linagliptin treatment, Bacteroides, Alistipes, and Parabacteroides were dominant at the genus level in the linagliptin group, while Alistipes finegoldii, Prevotella stercorea, and Parabacteroides distasonis were dominant at the strain level; in healthy adults, Collinsella and Eubacterium were dominant at the genus level, while Bifidobacterium adolescentis, Collinsella aerofaciens, and Eubacterium bioforme were the dominant strains (Figure 4).
When the microbiota at baseline and 12 weeks of metformin treatment and from healthy controls were evaluated by LEfSe analysis, Firmicutes and Actinobacteria at the phylum level and Gemmiger and Rothia at the genus level were dominant at the beginning of metformin treatment, while Coriobactericeae at the phylum level and Collinsella at the genus level were dominant at the end of the 12th week; Bacteroides at the phylum level and Alistipes at the genus level were dominant in the healthy control group. At the strain level, Gemmiger formicilis, Ruminococcus bromii, Rothia mucilaginosa, and Lactobacillus ruminis were dominant at the beginning of metformin treatment, while Collinsella aerofaciens was dominant after the 12th week of metformin treatment (Figure 5).

4. Discussion

In our study, we found that adult patients with T2D had a different intestinal microbiota composition at the time of diagnosis compared to healthy adults: their bacterial diversity and richness were decreased. After 12 weeks of diet and oral antidiabetic monotherapy (metformin or linagliptin), the patients’ HbA1c levels decreased, and there were some alterations or improvements in the intestinal microbiota composition. However, the microbiota composition remained different in T2D cases compared to healthy individuals. Many studies have shown the role of the microbiota in chronic metabolic diseases such as diabetes and obesity. Although data on whether the microbiota is the cause or effect of these diseases are still being evaluated, microbiota composition has been shown to play an important role in the development of T2D and its prognosis [25].
Research on T2D has demonstrated a disturbance in the composition of the intestinal microbiota. Our study revealed that Firmicutes was the most prevalent phylum in both the patient and control groups. However, Firmicutes was more common in healthy adults, while Bacteroidetes was more common in people with T2D than in healthy adults. In our study, at the genus and species levels, there were significant differences between patients with T2D and healthy controls. Ruminococcus, Blautia, Bacteroides, and Bifidobacterium at the genus level and Ruminococcus bromii, Prevotella copri, Collinsella aerofaciens, Veillonella dispar, Eubacterium bioforme, Dorea formigenerans, Lactobacillus ruminis, Rothia mucilaginosa, and Gemmiger focilis at the species level were common in patients with T2D, while at the strain level, Faecalibacterium prausnitzii, Ruminococcus callidus, and Bacteroides uniformis were common in healthy adults. Gurung et al. [26] conducted a systematic review and found that the gut microbiome of T2D subjects consistently showed a decrease in relative abundance of the genera Bifidobacterium, Bacteroides, and Akkermansia, along with a consistent increase in relative abundance of the genera Ruminococcus, Fusobacterium, and Blautia. Larssen et al. [27] found a connection between serum glucose levels and the Bacteroidetes-to-Firmicutes ratio, as well as the BacteroidesPrevotella to Clostridium coccoidesEubacterium rectale ratio, in patients with T2D. Previous studies have shown that butyrate-producing microorganisms, like Eubacterium rectale, Faecalibacterium prausnitzii, Roseburia intestinalis, and R. inulinivorans, are less common in people with T2D [28]. In Korea, a study on identical twins reported the potential use of Akkermansia muciniphila as a parameter for early diagnosis of T2D [29]. Reports also suggest that diabetes may be associated with the Acidaminococcus, Aggregatibacter, Anaerostipes, Blautia, Desulfovibrio, Dorea, and Faecalibacterium genera [30]. The mild decrease in alpha diversity observed in our study, both at the time of diagnosis and 12 weeks later, suggests that diabetes is associated with a moderate level of inflammation. In our study, after 12 weeks of medical treatment and diet, we saw a positive change in the microbiota composition. There was an increase in Faecalibacterium at the genus level and Faecalibacterium prausnitzii at the strain level. The blood sugar levels also stayed the same. The fact that alpha diversity indicators remained low at the end of the 12th week in T2D cases, the prevalence of the Gemmiger and Collinsella genera in the T2D group, and the dominance of Gemmiger formicilis and Collinsella aerofaciens at the species level demonstrated that the microbiota composition was still different to that of healthy controls. Previous studies indicated C. aerofaciens as a pathobiont that is also overabundant in metabolic syndrome, T2D, autoimmune polyendocrine syndrome type 1, and psoriatic arthritis. Our results highlighted that some predominant bacteria remained predominant at the 12th week of intervention, despite some improvement in HbA1c levels [31]. Hur and colleagues [32] showed that Gemmiger formicilis and Collinsella aerofaciens were higher in relative abundance in adults receiving a Westernized diet. Collinsella genus, including C. aerofaciens, a trimethylamine (TMA)-producing bacteria with atherogenic effects, was associated with cardiovascular risk in T2D patients [33]. In an experimental model, C. aerofaciens is also linked with increased ethanol production and liver inflammation contributes to the pathophysiology of non-alcoholic fatty liver disease [34]. Considering the effects of many factors on diabetes and its course, it seems unlikely that 3 months of diabetes and drug treatment will provide a complete improvement in the microbiota composition, as expected. Improvements in the microbiota composition over time with sustainable lifestyle changes and treatment compliance may be among the long-term treatment goals in these patients.
Metformin is the most common and first recommended oral antidiabetic drug for the treatment of T2D. Experimental and clinical studies have shown that the benefits of metformin are, at least partially, attributable to its impact on gut microbiota [35]. Metformin specifically enhances the prevalence of mucin-degrading Akkermansia muciniphila and short-chain fatty acid (SCFA)-producing genera, including Megasphaera and Blautia, thereby augmenting the advantageous effects of mucin and SCFAs on intestinal barrier integrity, bile acid metabolism, and glucose homeostasis. Metformin further enhances the prevalence of Bifidobacterium bifidum. Researchers have proposed that the confounding effects of metformin may account for the heightened prevalence of Lactobacillus species. Previous studies suggested that these bacteria could enhance the effects of metformin by producing more butyrate and propionate, and they hypothesized that an excess of Escherichia coli species in the gut could contribute to an inability to metabolize metformin [5,10,36]. Studies suggest that metformin benefits microbiota both directly and by regulating blood glucose [10,37,38,39]. Researchers suggest that the positive impact of metformin on the microbiota in T2D patients may stem from its beneficial effects on short-chain fatty acids like butyrate and propionate, which stimulate intestinal gluconeogenesis [10,12]. Researchers also suggest that stimulating gluconeogenesis in the intestines reduces appetite by influencing glucose production in the liver, potentially leading to positive effects on weight loss and metabolic control [40,41]. Studies have also shown that T2D patients who are not taking metformin have fewer microbial genes associated with glycine and tryptophan breakdown than patients who are taking metformin [42]. In our study, 12 weeks of metformin treatment had some beneficial effects on microbiota composition.
DPP-4 inhibitors are another group of oral antidiabetic drugs used in the treatment of T2D. Studies have shown changes in the gut microbiota with DPP-4 inhibitors. Unlike other DPP-4 inhibitors, linagliptin undergoes 90% elimination in feces following oral administration, rather than in the kidneys. The enterohepatic system converts a small portion of absorbed linagliptin to its pharmacologically inactive metabolite. Linagliptin binds 75–99% to plasma proteins and has a half-life of 12 h [43,44]. Researchers found that the DPP-4 inhibitors sitagliptin and vildagliptin alter the gut microbial composition in diabetic rats [45,46,47]. A study using vildagliptin, a DPP-4 inhibitor, revealed a decrease in Oscillibacter and an increase in Lactobacilli. Olivares et al. [46] suggested that inhibition of DPP-4 may have a direct effect on the gastrointestinal tract. Smith et al. [16] evaluated the effect of sitagliptin on intestinal microbiota composition. They added sitagliptin (100 mg) to metformin and/or sulfonylureas treatment in 19 adult patients with T2D and showed that sitagliptin had no effect on the alpha or beta diversity of the intestinal microbiota, nor were there any changes in microbial composition related to clinical parameters. Our study is a pivotal study showing the effect of linagliptin on microbiota composition. We found no changes in microbiota composition (with LEfSe analysis) between the baseline and 12th week of intervention in the linagliptin group. Regarding the abundance in F. prausnitzii, there is an increase (from 32% to 48%) at 12 weeks of linagliptin use, there are no statistical differences, and the low sample size might affect these results. When the microbiota composition was compared with that of healthy controls at the 12th week of linagliptin treatment, Bacteroides, Alistipes, and Parabacteroides were dominant at the genus level, while Alistipes finegoldii, Prevotella stercorea, and Parabacteroides distasonis were dominant at the strain level in the linagliptin group.
This study had some limitations. It is difficult to control each parameter related to microbiota composition in patients with T2D. Our enrollment number was considerably low; however, we enrolled patients receiving only monotherapy and excluded those with different clinical conditions related to T2D. Due to the study design and the nature of the disease, this study was not a placebo-controlled trial. While the patients received dietary and exercise advice, this study lacked any control or documentation of dietary intake, which can strongly influence gut microbiota composition. We did not show any correlation between HbA1c levels and alpha diversity indices; this might be related to the short course of therapy and needs to be evaluated in further studies.

5. Conclusions

In conclusion, we observed a substantial change in the composition of the gut microbiota in patients with type 2 diabetes. Twelve weeks of an oral antidiabetic treatment, especially metformin, had some beneficial effects on microbiota composition. Limited research has investigated the relationship between antidiabetic medications and the gut microbiota. More research needs to be conducted to fully understand how these drugs change the microbiota in the gut and how such changes help to improve glucose homeostasis.

Author Contributions

G.Y., A.A. and E.C.D.: developed the concept and wrote this manuscript; G.Y., A.T.K. and A.A.: enrolled the patients; E.C.D.: analyzed the results. All authors have read and agreed to the published version of the manuscript.

Funding

This study has been supported by the Eskisehir Osmangazi University Research Grant (TSA-2021-1587).

Institutional Review Board Statement

Ethics approval was obtained from the Eskisehir Osmangazi University Local Ethical Committee (26 January 2021, numbered E-25403353-050.99-146275). This study is a prospective study; we obtained written informed consent from all participants, and none of the personally identifiable information has been shown in this study. All procedures performed in this trial were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki Declaration and its later amendments or comparable ethical standards.

Informed Consent Statement

Written informed consent was obtained from all subjects involved in this study.

Data Availability Statement

Data available on request from the authors.

Acknowledgments

The authors would like to thank all participants. We also thanks to Ahmet Varis and Mucahit Kaya for analysis.

Conflicts of Interest

The authors declare no potential conflicts of interest.

References

  1. Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef] [PubMed]
  2. Sasidharan Pillai, S.; Gagnon, C.A.; Foster, C.; Ashraf, A.P. Exploring the Gut Microbiota: Key Insights Into Its Role in Obesity, Metabolic Syndrome, and Type 2 Diabetes. J. Clin. Endocrinol. Metab. 2024, 109, 2709–2719. [Google Scholar] [CrossRef] [PubMed]
  3. Chen, R.; Pan, S. Interaction of anti-diabetic medications and gut microbiota. Front. Biosci. 2021, 26, 673–674. [Google Scholar] [CrossRef] [PubMed]
  4. Huda, M.N.; Kim, M.; Bennett, B.J. Modulating the Microbiota as a Therapeutic Intervention for Type 2 Diabetes. Front. Endocrinol. 2021, 12, 632335. [Google Scholar] [CrossRef]
  5. Crudele, L.; Gadaleta, R.M.; Cariello, M.; Moschetta, A. Gut microbiota in the pathogenesis and therapeutic approaches of diabetes. eBioMedicine 2023, 97, 104821. [Google Scholar] [CrossRef]
  6. Barlow, G.M.; Mathur, R. Type 2 Diabetes and the Microbiome. J. Endocr. Soc. 2022, 7, bvac184. [Google Scholar] [CrossRef]
  7. Whang, A.; Nagpal, R.; Yadav, H. Bi-directional drug-microbiome interactions of anti-diabetics. eBioMedicine 2019, 39, 591–602. [Google Scholar] [CrossRef]
  8. Adeshirlarijaney, A.; Gewirtz, A.T. Considering gut microbiota in treatment of type 2 diabetes mellitus. Gut Microbes 2020, 11, 253–264. [Google Scholar] [CrossRef]
  9. Liu, W.; Luo, Z.; Zhou, J.; Sun, B. Gut Microbiota and Antidiabetic Drugs: Perspectives of Personalized Treatment in Type 2 Diabetes Mellitus. Front. Cell. Infect. Microbiol. 2022, 12, 853771. [Google Scholar] [CrossRef]
  10. Szymczak-Pajor, I.; Drzewoski, J.; Kozłowska, M.; Krekora, J.; Śliwińska, A. The Gut Microbiota-Related Antihyperglycemic Effect of Metformin. Pharmaceuticals 2025, 18, 55. [Google Scholar] [CrossRef]
  11. Lee, C.B.; Chae, S.U.; Jo, S.J.; Jerng, U.M.; Bae, S.K. The Relationship between the Gut Microbiome and Metformin as a Key for Treating Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2021, 22, 3566. [Google Scholar] [CrossRef] [PubMed]
  12. Rosell-Díaz, M.; Fernández-Real, J.M. Metformin, Cognitive Function, and Changes in the Gut Microbiome. Endocr. Rev. 2024, 45, 210–226. [Google Scholar] [CrossRef] [PubMed]
  13. Wang, D.; Liu, J.; Zhou, L.; Zhang, Q.; Li, M.; Xiao, X. Effects of Oral Glucose-Lowering Agents on Gut Microbiota and Microbial Metabolites. Front. Endocrinol. 2022, 13, 905171. [Google Scholar] [CrossRef]
  14. Mindrescu, N.M.; Guja, C.; Jinga, V.; Ispas, S.; Curici, A.; Nelson Twakor, A.; Pantea Stoian, A.M. Interactions Between Gut Microbiota and Oral Antihyperglycemic Drugs: A Systematic Review. Int. J. Mol. Sci. 2024, 25, 3540. [Google Scholar] [CrossRef]
  15. Gallwitz, B. Clinical Use of DPP-4 Inhibitors. Front. Endocrinol. 2019, 10, 389. [Google Scholar] [CrossRef]
  16. Smits, M.M.; Fluitman, K.S.; Herrema, H.; Davids, M.; Kramer, M.H.H.; Groen, A.K.; Belzer, C.; de Vos, W.M.; Cahen, D.L.; Nieuwdorp, M.; et al. Liraglutide and sitagliptin have no effect on intestinal microbiota composition: A 12-week randomized placebo-controlled trial in adults with type 2 diabetes. Diabetes Metab. 2021, 47, 101223. [Google Scholar] [CrossRef]
  17. Martínez-López, Y.E.; Neri-Rosario, D.; Esquivel-Hernández, D.A.; Padron-Manrique, C.; Vázquez-Jiménez, A.; Sánchez-Castañeda, J.P.; Girón-Villalobos, D.; Mendoza-Ortíz, C.; Reyes-Escogido, M.L.; Evia-Viscarra, M.L.; et al. Effect of metformin and metformin/linagliptin on gut microbiota in patients with prediabetes. Sci. Rep. 2024, 14, 9678. [Google Scholar] [CrossRef]
  18. Bolyen, E.; Rideout, J.R.; Dillon, M.R.; Bokulich, N.A.; Abnet, C.C.; Al-Ghalith, G.A.; Alexander, H.; Alm, E.J.; Arumugam, M.; Asnicar, F.; et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 2019, 37, 852–857. [Google Scholar] [CrossRef]
  19. Schloss, P.D. Amplicon sequence variants artificially split bacterial genomes into separate clusters. mSphere 2021, 6, e0019121. [Google Scholar] [CrossRef]
  20. Werner, J.J.; Koren, O.; Hugenholtz, P.; DeSantis, T.Z.; Walters, W.A.; Caporaso, J.G.; Angenent, L.T.; Knight, R.; Ley, R.E. Impact of training sets on classification of high-throughput bacterial 16s rRNA gene surveys. ISME J. 2012, 6, 94–103. [Google Scholar] [CrossRef]
  21. McMurdie, P.J.; Holmes, S. Phyloseq: An R package for reproducible interactive analysis and graphics of microbiome census data. PLoS ONE 2013, 8, e61217. [Google Scholar] [CrossRef] [PubMed]
  22. R Core Team. A Language and Environment for Statistical Computing; R Foundation for Statistical Computing: Vienna, Austria, 2017. [Google Scholar]
  23. Love, M.I.; Huber, W.; Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 2014, 15, 550. [Google Scholar] [CrossRef] [PubMed]
  24. Segata, N.; Izard, J.; Waldron, L.; Gevers, D.; Miropolsky, L.; Garrett, W.S.; Huttenhower, C. Metagenomic biomarker discovery and explanation. Genome Biol. 2011, 12, R60. [Google Scholar] [CrossRef] [PubMed]
  25. Razavi, S.; Amirmozafari, N.; Zahedi Bialvaei, A.; Navab-Moghadam, F.; Khamseh, M.E.; Alaei-Shahmiri, F.; Sedighi, M. Gut microbiota composition and type 2 diabetes: Are these subjects linked together? Heliyon 2024, 10, e39464. [Google Scholar] [CrossRef]
  26. Gurung, M.; Li, Z.; You, H.; Rodrigues, R.; Jump, D.B.; Morgun, A.; Shulzhenko, N. Role of gut microbiota in type 2 diabetes pathophysiology. eBioMedicine 2020, 51, 102590. [Google Scholar] [CrossRef]
  27. Larsen, N.; Vogensen, F.K.; van den Berg, F.W.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; Al-Soud, W.A.; Sørensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010, 5, e9085. [Google Scholar] [CrossRef]
  28. Fliegerová, K.O.; Mahayri, T.M.; Sechovcová, H.; Mekadim, C.; Mrázek, J.; Jarošíková, R.; Dubský, M.; Fejfarová, V. Diabetes and gut microbiome. Front. Microbiol. 2025, 15, 1451054. [Google Scholar] [CrossRef]
  29. Yassour, M.; Lim, M.Y.; Yun, H.S.; Tickle, T.L.; Sung, J.; Song, Y.M.; Lee, K.; Franzosa, E.A.; Morgan, X.C.; Gevers, D.; et al. Sub-clinical detection of gut microbial biomarkers of obesity and type 2 diabetes. Genome Med. 2016, 8, 17. [Google Scholar] [CrossRef]
  30. Yang, Q.; Lin, S.L.; Kwok, M.K.; Leung, G.M.; Schooling, C.M. The Roles of 27 Genera of Human Gut Microbiota in Ischemic Heart Disease, Type 2 Diabetes Mellitus, and Their Risk Factors: A Mendelian Randomization Study. Am. J. Epidemiol. 2018, 187, 1916–1922. [Google Scholar] [CrossRef]
  31. Gargari, G.; Mantegazza, G.; Cremon, C.; Taverniti, V.; Valenza, A.; Barbaro, M.R.; Marasco, G.; Duncan, R.; Fiore, W.; Ferrari, R.; et al. Collinsella aerofaciens as a predictive marker of response to probiotic treatment in non-constipated irritable bowel syndrome. Gut Microbes. 2024, 16, 2298246. [Google Scholar] [CrossRef]
  32. Hur, H.J.; Wu, X.; Yang, H.J.; Kim, M.J.; Lee, K.H.; Hong, M.; Park, S.; Kim, M.S. Beneficial Effects of a Low-Glycemic Diet on Serum Metabolites and Gut Microbiota in Obese Women With Prevotella and Bacteriodes Enterotypes: A Randomized Clinical Trial. Front. Nutr. 2022, 9, 861880. [Google Scholar] [CrossRef] [PubMed]
  33. Siegel, E.G.; Bermejo, J.L.; Flade, I.; Hasslacher, C. Cardiovascular Complications and Composition of the Intestinal Microbiome in Patients with Type 2 Diabetes. Int. J. Diabetes Clin. Res. 2018, 5, 086. [Google Scholar] [CrossRef]
  34. Purohit, A.; Kandiyal, B.; Kumar, S.; Pragasam, A.K.; Kamboj, P.; Talukdar, D.; Verma, J.; Sharma, V.; Sarkar, S.; Mahajan, D.; et al. Collinsella aerofaciens linked with increased ethanol production and liver inflammation contribute to the pathophysiology of NAFLD. iScience 2023, 27, 108764. [Google Scholar] [CrossRef] [PubMed]
  35. Pavlo Petakh Kamyshna, I.; Kamyshnyi, A. Effects of metformin on the gut microbiota: A systematic review. Mol. Metab. 2023, 77, 101805. [Google Scholar] [CrossRef]
  36. Facchin, S.; Bertin, L.; Bonazzi, E.; Lorenzon, G.; De Barba, C.; Barberio, B.; Zingone, F.; Maniero, D.; Scarpa, M.; Ruffolo, C.; et al. Short-Chain Fatty Acids and Human Health: From Metabolic Pathways to Current Therapeutic Implications. Life 2024, 14, 559. [Google Scholar] [CrossRef]
  37. Zhang, Q.; Hu, N. Effects of Metformin on the Gut Microbiota in Obesity and Type 2 Diabetes Mellitus. Diabetes Metab. Syndr. Obes. 2020, 13, 5003–5014. [Google Scholar] [CrossRef]
  38. Li, L.; Chen, Y.; Tang, Z.; You, Y.; Guo, Y.; Liao, Y. Effect of metformin on gut microbiota imbalance in patients with T2DM, and the value of probiotic supplementation. Allergol. Immunopathol. 2024, 52, 84–90. [Google Scholar] [CrossRef]
  39. Cheng, M.; Ren, L.; Jia, X.; Wang, J.; Cong, B. Understanding the action mechanisms of metformin in the gastrointestinal tract. Front. Pharmacol. 2024, 15, 1347047. [Google Scholar] [CrossRef]
  40. Soty, M.; Gautier-Stein, A.; Rajas, F.; Mithieux, G. Gut-Brain Glucose Signaling in Energy Homeostasis. Cell Metab. 2017, 25, 1231–1242. [Google Scholar] [CrossRef]
  41. LaMoia, T.E.; Shulman, G.I. Cellular and Molecular Mechanisms of Metformin Action. Endocr. Rev. 2021, 42, 77–96. [Google Scholar] [CrossRef]
  42. Mardinoglu, A.; Boren, J.; Smith, U. Confounding Effects of Metformin on the Human Gut Microbiome in Type 2 Diabetes. Cell Metab. 2016, 3, 10–12. [Google Scholar] [CrossRef] [PubMed]
  43. Graefe-Mody, U.; Retlich, S.; Friedrich, C. Clinical pharmacokinetics and pharmacodynamics of linagliptin. Clin. Pharmacokinet. 2012, 51, 411–427. [Google Scholar] [CrossRef]
  44. Guedes, E.P.; Hohl, A.; de Melo, T.G.; Lauand, F. Linagliptin: Farmacology, efficacy and safety in type 2 diabetes treatment. Diabetol. Metab. Syndr. 2013, 5, 25. [Google Scholar] [CrossRef] [PubMed]
  45. Yan, X.; Feng, B.; Li, P.; Tang, Z.; Wang, L. Microflora Disturbance during Progression of Glucose Intolerance and Effect of Sitagliptin: An Animal Study. J. Diabetes Res. 2016, 2016, 2093171. [Google Scholar] [CrossRef] [PubMed]
  46. Olivares, M.; Neyrinck, A.M.; Pötgens, S.A.; Beaumont, M.; Salazar, N.; Cani, P.D.; Bindels, L.B.; Delzenne, N.M. The DPP-4 inhibitor vildagliptin impacts the gut microbiota and prevents disruption of intestinal homeostasis induced by a Western diet in mice. Diabetologia 2018, 61, 1838–1848. [Google Scholar] [CrossRef]
  47. Liu, B.; Zhang, L.; Yang, H.; Zheng, H.; Liao, X. Microbiota: A potential orchestrator of antidiabetic therapy. Front. Endocrinol. 2023, 14, 973624. [Google Scholar] [CrossRef]
Figure 1. Comparison of fecal microbiota compositions of healthy controls and patients with type 2 diabetes at the genus and species levels at admission. Maroon: healthy controls. Blue: T2D.
Figure 1. Comparison of fecal microbiota compositions of healthy controls and patients with type 2 diabetes at the genus and species levels at admission. Maroon: healthy controls. Blue: T2D.
Jcm 14 02786 g001aJcm 14 02786 g001b
Figure 2. LEfSe analysis results: A cladogram showing that each link of the radial phylogenetic tree is a different taxonomic level in stool samples for the T2D group and healthy controls at 12 weeks of intervention. Horizontal bars represent log10-transformed LDA scores indicated by vertical dotted lines. Maroon dots indicate T2D cases, and no imprinting microorganisms were detected in healthy adults in the LEfSe analysis. Maroon: healthy controls. Blue: T2D after 12 weeks of treatment.
Figure 2. LEfSe analysis results: A cladogram showing that each link of the radial phylogenetic tree is a different taxonomic level in stool samples for the T2D group and healthy controls at 12 weeks of intervention. Horizontal bars represent log10-transformed LDA scores indicated by vertical dotted lines. Maroon dots indicate T2D cases, and no imprinting microorganisms were detected in healthy adults in the LEfSe analysis. Maroon: healthy controls. Blue: T2D after 12 weeks of treatment.
Jcm 14 02786 g002
Figure 3. Comparison of the genus-level microbiota compositions of the stool of T2D patients receiving linagliptin (blue) or metformin (green), at the beginning of treatment and after 12 weeks of treatment, with that of healthy controls (maroon).
Figure 3. Comparison of the genus-level microbiota compositions of the stool of T2D patients receiving linagliptin (blue) or metformin (green), at the beginning of treatment and after 12 weeks of treatment, with that of healthy controls (maroon).
Jcm 14 02786 g003
Figure 4. LEfSe analysis of fecal samples from the linagliptin group at Week 12 (green) and healthy controls (red). Horizontal bars represent log10-transformed LDA scores indicated by vertical dotted lines. p, family; c, class; o, team; f, family; g, genus; and s, species. (LDA threshold value > 2, p < 0.05.).
Figure 4. LEfSe analysis of fecal samples from the linagliptin group at Week 12 (green) and healthy controls (red). Horizontal bars represent log10-transformed LDA scores indicated by vertical dotted lines. p, family; c, class; o, team; f, family; g, genus; and s, species. (LDA threshold value > 2, p < 0.05.).
Jcm 14 02786 g004
Figure 5. LEfSe analysis of fecal samples from the metformin group at baseline (red) and Week 12 (blue) and from healthy controls (Group 3, green). Horizontal bars represent log10-transformed LDA scores indicated by vertical dotted lines. p, family; c, class; o, team; f, family; g, genus; and s, species. (LDA threshold value > 2, p < 0.05).
Figure 5. LEfSe analysis of fecal samples from the metformin group at baseline (red) and Week 12 (blue) and from healthy controls (Group 3, green). Horizontal bars represent log10-transformed LDA scores indicated by vertical dotted lines. p, family; c, class; o, team; f, family; g, genus; and s, species. (LDA threshold value > 2, p < 0.05).
Jcm 14 02786 g005
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

Kalkan, A.T.; Yorulmaz, G.; Akalin, A.; Dinleyici, E.C. Intestinal Microbiota Composition in Patients with Type 2 Diabetes and Effects of Oral Antidiabetics. J. Clin. Med. 2025, 14, 2786. https://doi.org/10.3390/jcm14082786

AMA Style

Kalkan AT, Yorulmaz G, Akalin A, Dinleyici EC. Intestinal Microbiota Composition in Patients with Type 2 Diabetes and Effects of Oral Antidiabetics. Journal of Clinical Medicine. 2025; 14(8):2786. https://doi.org/10.3390/jcm14082786

Chicago/Turabian Style

Kalkan, Ahmet Toygar, Goknur Yorulmaz, Aysen Akalin, and Ener Cagri Dinleyici. 2025. "Intestinal Microbiota Composition in Patients with Type 2 Diabetes and Effects of Oral Antidiabetics" Journal of Clinical Medicine 14, no. 8: 2786. https://doi.org/10.3390/jcm14082786

APA Style

Kalkan, A. T., Yorulmaz, G., Akalin, A., & Dinleyici, E. C. (2025). Intestinal Microbiota Composition in Patients with Type 2 Diabetes and Effects of Oral Antidiabetics. Journal of Clinical Medicine, 14(8), 2786. https://doi.org/10.3390/jcm14082786

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