Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Oncology Letters
      • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Biomedical Reports
      • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • Information for Authors
    • Information for Reviewers
    • Information for Librarians
    • Information for Advertisers
    • Conferences
  • Language Editing
Spandidos Publications Logo
  • About
    • About Spandidos
    • Aims and Scopes
    • Abstracting and Indexing
    • Editorial Policies
    • Reprints and Permissions
    • Job Opportunities
    • Terms and Conditions
    • Contact
  • Journals
    • All Journals
    • Biomedical Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Experimental and Therapeutic Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Epigenetics
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Functional Nutrition
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Molecular Medicine
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • International Journal of Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Medicine International
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular and Clinical Oncology
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Molecular Medicine Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Letters
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • Oncology Reports
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
    • World Academy of Sciences Journal
      • Information for Authors
      • Editorial Policies
      • Editorial Board
      • Aims and Scope
      • Abstracting and Indexing
      • Bibliographic Information
      • Archive
  • Articles
  • Information
    • For Authors
    • For Reviewers
    • For Librarians
    • For Advertisers
    • Conferences
  • Language Editing
Login Register Submit
  • This site uses cookies
  • You can change your cookie settings at any time by following the instructions in our Cookie Policy. To find out more, you may read our Privacy Policy.

    I agree
Search articles by DOI, keyword, author or affiliation
Search
Advanced Search
presentation
Biomedical Reports
Join Editorial Board Propose a Special Issue
Print ISSN: 2049-9434 Online ISSN: 2049-9442
Journal Cover
January-2026 Volume 24 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

Journals

International Journal of Molecular Medicine

International Journal of Molecular Medicine

International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.

International Journal of Oncology

International Journal of Oncology

International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.

Molecular Medicine Reports

Molecular Medicine Reports

Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.

Oncology Reports

Oncology Reports

Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine

Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.

Oncology Letters

Oncology Letters

Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.

Biomedical Reports

Biomedical Reports

Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.

Molecular and Clinical Oncology

Molecular and Clinical Oncology

International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.

World Academy of Sciences Journal

World Academy of Sciences Journal

Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.

International Journal of Functional Nutrition

International Journal of Functional Nutrition

Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.

International Journal of Epigenetics

International Journal of Epigenetics

Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.

Medicine International

Medicine International

An International Open Access Journal Devoted to General Medicine.

Journal Cover
January-2026 Volume 24 Issue 1

Full Size Image

Sign up for eToc alerts
Recommend to Library

  • Article
  • Citations
    • Cite This Article
    • Download Citation
    • Create Citation Alert
    • Remove Citation Alert
    • Cited By
  • Similar Articles
    • Related Articles (in Spandidos Publications)
    • Similar Articles (Google Scholar)
    • Similar Articles (PubMed)
  • Download PDF
  • Download XML
  • View XML
Article Open Access

Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients

  • Authors:
    • Amalia Ioana Arhire
    • Teodora Papuc
    • Sorin Ioacără
    • Gratiela Gradisteanu Pircalabioru
    • Carmen Gabriela Barbu
  • View Affiliations / Copyright

    Affiliations: Department of General Medicine, The University of Medicine and Pharmacy ‘Carol Davila’, 020021 Bucharest, Romania, Department of Botany and Microbiology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania
    Copyright: © Arhire et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 1
    |
    Published online on: October 27, 2025
       https://doi.org/10.3892/br.2025.2074
  • Expand metrics +
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Metrics: Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )
Cited By (CrossRef): 0 citations Loading Articles...

This article is mentioned in:



Abstract

With the increasing occurrence of type 1 diabetes mellitus (T1DM) in younger individuals, it is important to identifying contributing factors. A retrospective case control pilot study involving 31 children within the first 6 months after diagnosis, was conducted in the Elias Hospital in Bucharest, Romania) between January 2019‑ December 2021. Spearman correlation analysis was performed, evaluating the association between microbiota and early onset T1DM, high‑level onset glycaemia, ketoacidosis, thyroid autoimmunity and clinical parameters involving growth. Gut dysbiosis with an overabundance of bacteria and fungi was observed compared with the healthy control group. Butyricicoccus was positively associated with younger age at onset (r=0.6276, P=0.0018) but negatively correlated with markers of disease progression, such as T1DM recent diagnosis (r=‑0.517, P=0.0164), homeostasis model assessment of β‑cell function) (r=‑0.5962, P=0.0034) and C‑peptide values (r=‑0.5005, P=0.0344). Bacteroides showed a negative association with early‑stage diabetes (r=‑0.4431, P=0.0442) and Clostridium leptum was negatively associated with recent diagnosis (r=‑0.6278, P=0.0023). Antibiotic use prior to disease diagnosis was positively correlated with Candida albicans (r=0.4431, P=0.0442). Inflammation, determined by the systemic inflammatory index, was positively correlated with Enterobacteriaceae (r=0.4331, P=0.0441) and negatively correlated with Clostridium coccoides (r=‑0.4241, P=0.0492). C‑reactive protein levels were negatively associated with Bacteroides (r=‑0.4331, P=0.0498). These findings highlighted the role of gut microbiota in pancreatic inflammation and T1DM progression. In conclusion, dysbiosis of children with T1DM was correlated with younger age and a more severe onset. Microbial metabolites levels differed in these patients compared with the healthy control group. To the best of our knowledge, the present study is the first to assess gastrointestinal dysbiosis in T1DM.

Introduction

The total number of patients with type 1 diabetes mellitus (T1DM) is steadily rising. In 2025, an estimated 9.5 million people worldwide are living with T1DM, representing a 13% increase compared with the 8.4 million reported in 2021. Since the genetic transmission of this disease has remained relatively constant, this growing prevalence suggests the involvement of alternative risk factors, including environmental triggers, lifestyle influences, and changes in early-life exposures. These include pollutants, endocrine disruptors, vitamin D deficiency and pathological colonization of the gastrointestinal tract (1,2).

The gastrointestinal microbiota (the colonization of the gastrointestinal tract) comprises all bacteria, viruses and fungi that inhabit this environment. An imbalance of this microbiota (dysbiosis) with more proinflammatory bacteria that affect the immune system is hypothesized to explain the global rise in T1DM, especially the early onset of this disease (1-3).

The balance of the gut microbiota is disrupted by various factors, leading to dysbiosis, which serves a vital role in the development and progression of numerous types of disease, including type 2 diabetes mellitus, autoimmune diseases such as Hashimoto's thyroiditis and Chon's disease (4). The onset of T1DM, especially with onset at younger ages, represents a global health challenge, as it does not affect all individuals with genetic susceptibility (5). The alterations in microbiota and chronic inflammation, due to environmental influences, are hypothesized as explanations for the shifts in the prevalence of this disease (5,6).

Several mouse studies have provided evidence of a potential link between microbiota and T1DM (7-9). For example, exposure to antibiotics in the prenatal period or in the first weeks after birth influences the gut microbiota and the T1DM incidence by reducing the pancreatic inflammation and levels of pro-inflammatory cytokines (10-14).

In the context of T1DM, changes in the gut microbiota have been observed prior to the emergence of systemic signs of islet autoimmunity (15,16). This shift in microbiota could be attributed to the fact that previous studies primarily identified these modifications through gene analysis of 16S rRNA, which may not capture specific structural and functional characteristics potentially involved in disease progression (17,18). Later studies used specialized designs to control all known factors influencing T1DM susceptibility and analyzed microbiome characteristics using longitudinal metagenomic sequencing of stool samples (19,20). Patients with T1DM harbor a proinflammatory environment, irrespective of geographical location, with a higher abundance of Bacteroidetes and a lower abundance of Firmicutes (21,22).

Decreased levels of Firmicutes (comprising strains of Clostridium and Eubacterium) may pose a risk to the host as this phylum encompasses numerous producers of the short-chain fatty acid (SCFA) butyrate (23). Butyrate is key for intestinal homeostasis, serving as an energy source for colonocytes and promoting the secretion of mucins. This aids in reducing gut permeability by facilitating the formation of tight junctions (13,17,18,22,23) and protecting against pathogens and harmful substances (21,24,25). The Bacteroidetes phylum contains strains of Bacteroides and Prevotella. Studies have shown that T1DM is characterized by a dominance of Bacteroides, taxa associated with intestinal inflammation, while levels of protective Prevotella are decreased (26-28).

Species within the Bacteroides genus ferment glucose and lactate to produce propionate, acetate and succinate, but they lack the ability to generate butyrate (29). Additionally, lactate-producing bacteria, including certain probiotic strains such as Lactobacillus rhamnosus, L. reuteri, L. johnsonii N6.2, L. plantarum and Bifidobacterium lactis, synthesize butyrate, thereby reinforcing the intestinal barrier function (26). These findings underscore the role of the gut microbiota in T1DM and suggest that targeting specific microbial components may have potential for interventions aimed at supporting intestinal health in individuals with T1DM (1,21,27-30).

Similar to autoimmune conditions, T1DM prevalence exhibits a geographical pattern: The EURODIAB Autoimmune Complications of Type 1 Diabetes study (1989-1990) reported incidence rates of ~42.9 per 100,000/year in Finland, compared with 4.6/100,000/year in northern Greece (31). Climate, particularly sunlight exposure, influences microbiota and impacts the immune system. Vitamin D deficiencies and disturbances in the circadian rhythm alter the gut microbiota, contributing to the development of autoimmune disease (2,28,30).

Evidence suggests that vitamin D levels are lower in newly diagnosed patients with T1DM compared with population controls (32,33). Moreover, the supplementation of this vitamin and the risk of T1DM show a dose-response effect (30).

The composition of early-life microbiota undergoes frequent changes influenced by maternal and environmental microbes, as well as exposure to food and animal-borne antigens. The method of birth, whether cesarean or vaginal, has an impact on the neonatal microbiota; findings from the TEDDY study indicate that children delivered vaginally exhibit higher levels of Bacteroides, which are associated with increased diversity and accelerated maturation of gut intestinal flora (14,34).

An increasing number of studies underscore the role of environmental factors in the rising incidence of T1DM, particularly among younger children (35,36); to the best of our knowledge, however, there are few studies from Romania or Eastern Europe (37-39). Given the geographical variation in T1DM, the data available may not entirely align with the actual pathogenesis of T1DM.

Investigating the association between T1DM onset and gut microbiota dysbiosis may foster collaborations between researchers, clinicians and policymakers, leading to the development of novel diagnostic tools, therapeutic interventions and tailored public health policies.

Considering the cultural, dietary and environmental changes that have occurred in Eastern Europe over the past three decades, it has been hypothesized that alterations in the gut microbiome may contribute to a decreased age at onset of T1DM, as well as a more severe clinical presentation. Longitudinal microbiome studies have shown that changes in microbial composition and function, including a reduction in short-chain fatty acid producers and enrichment of pro-inflammatory taxa, occur prior to or around the development of islet autoimmunity and overt T1DM (11,14). In parallel, epidemiological studies from Eastern Europe, including Romania and Poland, have reported an increase in T1DM incidence since the 1990s, with diagnosis occurring at progressively younger ages (37,40). At the time of diagnosis, a notable proportion of patients present with diabetic ketoacidosis, reflecting both delayed recognition and a more severe onset of disease, which may be associated with environmental and microbial risk factors (41). Moreover, it has been well documented that T1DM frequently coexists with other autoimmune diseases, such as thyroid autoimmunity and celiac disease, further supporting a role for shared environmental drivers, including gut microbiome dysregulation, in shaping disease course and comorbidity (42,43).

The aim of the present study was to analyze the gut microbiota composition in Romanian children with newly diagnosed T1DM, and to investigate its potential associations with age of onset, severity at presentation, glycemic control, and co-existing autoimmune conditions.

Materials and methods

Study population

The present retrospective case-control pilot study involved 31 pediatric patients (age, 1-18 years, 16 female and 15 male) diagnosed with T1DM within the past 6 months, based on the criteria set by the American Diabetes Association (44).

The inclusion criteria for the patients were as follows: i) Positive clinical T1DM diagnosis and ii) onset <6 months prior to admission. The exclusion criteria were as follows: i) Antibiotic/probiotic administration in the 2 weeks prior to the admission; ii) T1DM diagnosed >6 months previously and iii) other types of diabetes mellitus.

These inclusion and exclusion criteria for patients in the study were established to identify relevant changes in the gut microbiota in patients with type 1 diabetes, in light of observational evidence showing that individuals with type 1 diabetes exhibit altered fecal and oral microbiota composition, reduced butyrate-producing species, and lower plasma levels of acetate and propionate (45).

Patients were referred either by their primary care physician or transferred from other pediatric hospitals in the southern region of Romania to the Pediatric Endocrinology and Diabetes Department at Elias Emergency and University Hospital, Bucharest (Romania). Being one of three referral centers for evaluating and initiating continuous glucose monitor devices and insulin pumps reimbursed by the National Health System in the southern region, most patients with recently diagnosed T1DM sought treatment here. All participants were of Caucasian ethnicity, as defined by Thomson et al (46). All eligible pediatric patients, admitted between January 2019 and December 2021, were enrolled in chronological order of admission. The study adhered to the Helsinki Declaration of 2013, with parental approval obtained through written informed consent for the use of medical records in scientific research.

Patients with newly diagnosed T1DM were matched with healthy children of similar age (4-18 years), sex, BMI and urban environment. First-degree relatives with known T1DM were also evaluated to assess the influence of genetic and environmental factors. A comprehensive medical history, including demographics, family history of autoimmune diseases and/or diabetes, personal medical history, birth details, breastfeeding practices, infection history and treatment details, was obtained from all participants. Anthropometric measurements were conducted, including age, weight, height, BMI, blood pressure, Tanner stage (47) and waist-to-hip ratio. Data were collected from medical records and laboratory reports, and compiled in an Excel Spreadsheet (version 16.0 (Microsoft Corporation)

Laboratory procedures

A 10 ml peripheral blood sample was obtained 8 h after the last meal to assess hematological and biochemical parameters. The complete blood count and blood chemistry analyses included measurements of glucose, creatinine, total cholesterol, high- and low-density lipoprotein-cholesterol, triglycerides, glycated hemoglobin (HbA1c), hepatic function and reactive C-protein (CRP). Additionally, levels of anti-thyroglobulin and anti-thyroperoxidase (for autoimmune thyroid disorders) and anti-tissue transglutaminase antibodies (for celiac disease) were measured to assess autoimmune conditions. Thyroid function was evaluated based on thyroid-stimulating hormone (TSH) and free thyroxine (fT4) levels. Furthermore, assessments were made for insulin-like growth factor 1 (IGF1), 25-hydroxy vitamin D [25(OH) vitamin D], and phosphorus-calcium metabolism, including measurements of total serum calcium, phosphorus and parathyroid hormone (PTH).

ELISA

The levels of 25(OH) vitamin D, anti-thyroglobulin, anti-thyroperoxidase and anti-tissue transglutaminase antibodies were determined using ELISA on a Chemwell 2010 ELISA system (Awareness Technology, Inc.) using commercially available kits as follows: 25(OH) Vitamin D ELISA kit (Immunodiagnostic Systems, cat. no. AC-57SF1), anti-thyroglobulin (catalog no. ABIN649023, Antibodies-online), anti-thyroperoxidase ELISA kit (cat. no. MBS3800854), and anti-tissue transglutaminase ELISA kit (both MyBioSource, cat. no. MBS7208318). All assays were performed in strict accordance with the manufacturer's instructions.

Chemiluminometric method

The determination of serum PTH levels were determined using the COBAS E 411 analyzer (Roche Diagnostics), a fully automated, random-access, multichannel instrument designed for immunological analysis.

Spectrophotometric methods

Serum biochemical parameters were assessed using standard enzymatic colorimetric methods. Lipids [total cholesterol (cat. no. 72291UD00), HDL-C- kit number: 67136UQ10, triglycerides- kit number:71068UD00), glucose (cat. no. 67921UQ02, creatinine- kit number: 71560UD00, aspartate transaminase (AST; cat. no. 70068UD00, alanine transaminase (ALT) - kit number: 72494UD00, C-reactive protein (CRP)- kit no. 50519Y600, and calcium - kit number: 74372UD00; all Abbott Diagnostics) were measured on the Architect c8000 Clinical Chemistry Analyzer (Abbott Diagnostics, Abbott Park, IL, USA). Assays were performed according to the manufacturer's instructions and quality control procedures.

Serum phosphorus was determined using the Vitros 5,1 FS Chemistry Analyzer and reagent kit (cat. no. 1203-0398-1506, Ortho Clinical Diagnostics).

Flow cytometry

Complete blood count was determined using a Sysmex XN 1000 hematology analyzer (Sysmex Corporation). Flow cytometry was performed on whole blood samples. Cells were washed twice with PBS and incubated with blocking buffer (2% FBS in PBS) for 15 min at 4˚C. Surface markers were detected with directly conjugated monoclonal antibodies, including CD3-FITC (clone UCHT1; cat. no. 555332; BD Biosciences), CD4-PE (clone RPA-T4; cat. no. 555347; BD Biosciences), and CD8-APC (cat. no. 555369; BD Biosciences). For apoptosis analysis, cells were stained with the Annexin V-FITC Apoptosis Detection kit (cat. no. 556547; BD Biosciences) in combination with propidium iodide (PI; cat. no. P4864; Sigma-Aldrich; Merck KGaA). Where secondary detection was required, biotinylated primary antibodies were visualized using streptavidin-PE (cat. no. 554061; BD Biosciences). Data acquisition was performed on a BD FACSCanto II flow cytometer (BD Biosciences), and results were analyzed using FlowJo software, version 10.8 (Tree Star Inc.).

ECLIA IGF-1, TSH and fT4 concentrations were evaluated by ECLIA on a Roche COBAS® e 411 analyzer (Roche Diagnostics). Commercial reagent kits were provided by Immunodiagnostic Systems, including the IDS-iSYS IGF-1 assay (cat. no. IS-3900), IDS-iSYS TSH assay (cat. no. IS-3100), and IDS-iSYS free T4 assay (cat. no. IS-3300). All assays were performed according to the manufacturer's instructions.

High-performance liquid chromatography (HPLC)

HbA1c was measured using the Bio-Rad D-10 Hemoglobin Testing System (Bio-Rad Laboratories, Inc.). The separation was performed on the D-10 cation-exchange cartridge (Bio-Rad Laboratories, Inc.; cat. no. 220-0101) at a controlled temperature of 25˚C. Whole blood samples (5 µl) were injected. The mobile phase consisted of two buffers (Buffer A: phosphate buffer, pH 6.5; Buffer B: phosphate buffer with increased ionic strength). The system was operated at a flow rate of 1.5 ml/min with a programmed gradient increase of Buffer B according to the manufacturer's protocol. An internal calibrator and quality control material (Bio-Rad Laboratories, Inc.; cat. no. 220-0102 and 220-0103) were included, and all procedures were carried out strictly according to the manufacturer's instructions. β-cell function and insulin resistance were evaluated using the homeostasis model assessment indices (HOMA-B and HOMA-IR). HOMA-B was calculated as [20 x fasting insulin [µU/ml])/(fasting glucose [mmol/L] - 3.5), and HOMA-IR as (fasting insulin [µU/ml] x fasting glucose [mmol/L])/22.5, according to Matthews et al (48)

Microbiota analysis

Stool samples were gathered during hospitalization or at home using a standardized procedure that involved antiseptic handling, collection in sterile tubes (without culture media) and immediate freezing at -20˚C. Subsequently, fecal DNA was extracted utilizing the PureLink Microbiome Purification kit (cat. Number A29790, Invitrogen; Thermo Fisher Scientific, Inc.) following the manufacturer's instructions. The concentration of DNA was assessed using a Qubit 4 fluorometer (Thermo Fisher Scientific, Inc.). DNA samples were diluted in DNase-free water to a concentration of 3 ng/µl. Quantitative PCR was performed to determine the relative abundance of intestinal microorganisms in stool DNA isolated from both patients with T1DM and healthy controls, using a ViiA7© Fast Real-Time instrument (Applied Biosystems; Thermo Fisher Scientific, Inc.). Bacterial or fungal group-specific primers (16S and 18S rRNA, respectively; Table I) were used at their designated annealing temperatures. For all other primers, each set was designed to target the 16S rRNA gene, which is used as a molecular marker for bacterial identification and quantification (49). The 16S rRNA gene contains both highly conserved regions, enabling the design of universal primers, and variable regions, allowing species-level discrimination. The control was18S rRN) gene, which serves as a conserved and widely used molecular marker for fungi. These primers were designed to recognize a broad range of clinically and environmentally relevant yeasts, with specificity toward Candida and Saccharomyces spp. The primers were previously described in Trandafir et al 2024, validated for specificity toward the bacterial taxa of interest, ensuring accurate amplification and minimizing off-target effects (49). Each PCR reaction comprised 2.5 nM forward and reverse primers, 9 ng DNA and 2X SYBR Green Master Mix (Applied Biosystems; Thermo Fisher Scientific, Inc.). Negative controls were samples without DNA templates. Thermocycling conditions were as follows: Initial denaturation at 95˚C for 5 min, followed by 40 cycles of 95˚C for 10 sec, 60˚C for 30 sec and 72˚C for 1 sec. Universal 16S rRNA and 18S rRNA primers were used for normalization and relative abundance of bacterial abundance was calculated using the 2-ΔΔCq method (50,51).

Table I

Sequences of the primers.

Table I

Sequences of the primers.

TargetForward primer (5'→3')Reverse primer (5'→3')Target gene
Eubacteria ACTCCTACGGGAGGCAGCAGT ATTACCGCGGCTGCTGGC16S rDNA
Bacteroides spp.CCTACGATGGATAGGGGT T CACGCTACTTGGCTGGTTCAG16S rDNA
Butyricicoccus spp. ACCTGAAGAATAAGCTCC GATAACGCTTGCTCCCTACGT16S rDNA
Gamma proteobacteria GCTAACGCATTAAGTACCCCG GCCATGCAGCACCTGTCT16S rDNA
Akkermansia muciniphila GCGTAGGCTGTTTCGTAAGTC GTGTGTGAAAG GAGTGTTCCCGATATCTACGC ATTTCA16S rDNA
Lactobacillus spp. ACGAGTAGGGAAATCTTCCA CACCGCTACACATGGAG16S rDNA
Clostridium leptum GCACAAGCAGTGGAGT CTTCCTCCGTTTTGTCAA16S rDNA
Clostridium coccoidesGACGCCGCGTGAAGG A AGCCCCAGCCTTTCACATC16S rDNA
Faecalibacterium prausnitzii CCCTTCAGTGCCGCAGT GTCGCAGGATGTCAAGAC16S rDNA
rRNA 18S universal primers ATTGGAGGGCAAGTCTGGTG CCGATCCCTAGTCGGCATAGrRNA 18S
Saccharomyces spp. AGGAGTGCGGTTCTTTG TACTTACCGAGGCAAGCTACArRNA 18S
Candida spp. TTTATCAACTTGTCACACCAGA ATCCCGCCTTACCACTACCGrRNA 18S
Metabolite analysis

Fecal content pellets (0.2 g) were suspended in 1 ml sterile saline solution and incubated at room temperature for 2 min. Subsequently, the sample was vigorously shaken for 4 min to create a slurry. After centrifugation at 1,790 x g for 1 h at 4˚C, the supernatant was collected and subjected to further centrifugation at 28,600 x g for 30 min at 4˚C.

Fecal content pellets (0.2 g) were suspended in 1 ml sterile saline solution and incubated at room temperature for 2 min. Subsequently, the sample was vigorously shaken for 4 min to create a slurry. The resulting supernatant was transferred to a new tube and filtered using a minisart-GF filter membrane (Sartorius AG) with a 1 ml sterile plastic syringe. Finally, another filtration step was performed using a Whatman-25 mmGD/X0 filter (Merck Millipore) and a 1 ml sterile plastic syringe. Metabolite levels (including butyrate, acetate, propionate, taurine, succinate and lactate) were determined using commercial kits following the manufacturer's instructions (Abbexa, cat. no. abx258338 for butyrate quantification and Sigma-Aldrich for the other metabolites- Cat. Number MAK355, MAK184, MAK065, and MAK086). Optical densities were measured using a spectrophotometer at 455 nm (Mulsiskan FC, Thermo Fisher Scientific, Inc.) and converted to µg/g feces.

Statistical analysis

All data are presented as the mean ± SEM or SD and were analyzed using the GraphPad Prism 9.0 software (Dotmatics). A total of three independent repeats were performed. Power analysis was initially performed with a set power (1) of 0.90 and 0.05 for two groups (Control and T1DM) tested using difference in means and SD as parameters. Standardized statistical test methods were used to analyze the results of demography and laboratory tests. The analysis was performed by a normality test (P<0.05 was considered to be normal and homogeneous) followed by parametric testing (unpaired t-test) and Welch's post hoc correction. Spearman correlation analysis of the association between proinflammatory taxa and early onset T1DM, high-level onset glycaemia and ketoacidosis, insulin dose, inflammation, thyroid autoimmunity, IGF-1 and other clinical parameters regarding growth was performed using SPSS Windows v. 17.0 (SPSS, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

Among the 31 patients, the ratio male: female was 1.11 and the mean BMI was 17.48 kg/m2. The mean age of the onset of the disease was 9.0±0.5 years and the mean glycaemia was 350±98 mg/dl (Table II). The vaginal: caesarian birth ratio was 0.36, (13/31) and 10/31 patients with T1DM had a family history of autoimmune disease.

Table II

Biochemical and immunological parameters.

Table II

Biochemical and immunological parameters.

ParameterMeanSEMStd. DeviationMedianNormal values
Leukocytes, x103/µl6.680.402.226.154.60-15.50
Neutrophils, x103/µl3.370.301.692.951.50-7.00
Lymphocytes, x103/µl2.620.191.092.311.50-10.50
Hemoglobin, g/dl13.420.211.1513.0011.00-15.30
CRP, mg/dl3.582.118.710.55<0.50
ALT, U/l19.291.779.8416.0013.00-26.00
AST, U/l23.451.287.1222.008.00-25.00
Creatinine, mg/dl0.660.030.150.630.57-0.86
Total cholesterol (mg/dl)172.238.1644.70164.50140.00-200.00
Triglycerides (mg/dl)53.233.3118.1551.50>40.00
HDL-cholesterol, mg/dl63.482.4513.6461.00<130.00
LDL-cholesterol, mg/dl102.905.7532.0096.4035.00-150.00
Anti-thyroglobulin antibodies, IU/ml60.8729.48161.4410.001.00-16.00
Anti-thyroid peroxidase antibodies (IU/ml)51.6631.90177.609.00<20.00
Anti-transglutaminase antibodies, IU/ml0.910.401.900.40<10.00
TSH, µIU/ml2.5070.221.182.360.30-3.60
Free T4, ng/dl1.1850.030.161.200.8-1.48
250H vitamin D, ng/ml28.162.1011.4924.28>30.00
Total calcium, mg/dl9.570.070.379.608.40-10.20
Phosphorus, mg/dl4.830.190.654.852.50-4.50
PTH, pg/ml31.362.5213.3127.8915.00-65.00
Insulin, IU/24 h/kg19.234.0821.5811.000.30-1.00

[i] CRP, C-reactive protein; ALT, alanine aminotransferase; AST, aspartate aminotransferase; HDL, high-density lipoprotein; LDL, low-density lipoprotein; TSH, thyroid-stimulating hormone; T4, thyroxine; 25(OH) vitamin D, 25-hydroxyvitamin D; PTH, parathyroid hormone.

In the T1DM group, dysbiosis was evident, characterized by an overabundance of detrimental bacteria such as Clostridium coccoides, Faecalibacterium, Bacteroides and Enterobacteriaceae, as well as fungi including Candida, Saccharomyces and Aspergillus spp., in contrast to the healthy control group (Fig. 1, Fig. 2 and Fig. 3). Furthermore, a notable decrease in Bifidobacterium spp. and elevated presence of A. muciniphila within the beneficial taxa were observed among patients with T1DM compared with the healthy control group (Figs. 1 and 4).

Beneficial bacteria abundance in
T1DM. Relative abundance of (A) Bifidobacterium, (B)
Lactobacillus and (C) A. muciniphila in patients with
T1DM compared with healthy control group. ***P<0.001.
T1DM, type 1 diabetes mellitus.

Figure 1

Beneficial bacteria abundance in T1DM. Relative abundance of (A) Bifidobacterium, (B) Lactobacillus and (C) A. muciniphila in patients with T1DM compared with healthy control group. ***P<0.001. T1DM, type 1 diabetes mellitus.

Mycobiome characterization. Dysbiosis
with increased abundance of (A) Aspergillus (B) Candida
spp. and (C) Saccharomyces in T1DM. The primers were
specific to the ribosomal 18S RNA gene and designed tEo recognize
various Candida and Saccharomyces spp. T1DM, type 1
diabetes mellitus. *P<0.05.

Figure 2

Mycobiome characterization. Dysbiosis with increased abundance of (A) Aspergillus (B) Candida spp. and (C) Saccharomyces in T1DM. The primers were specific to the ribosomal 18S RNA gene and designed tEo recognize various Candida and Saccharomyces spp. T1DM, type 1 diabetes mellitus. *P<0.05.

Dysbiosis markers in T1DM. Dysbiosis
with increased abundance of (A) Clostridium coccoides and
(B) Bacteroides spp. in T1DM. (C) Dysbiosis with increased
abundance of Enterobacteriaceae in T1DM (P=0.0193). T1DM,
type 1 diabetes mellitus. *P<0.05 and
**P<0.01

Figure 3

Dysbiosis markers in T1DM. Dysbiosis with increased abundance of (A) Clostridium coccoides and (B) Bacteroides spp. in T1DM. (C) Dysbiosis with increased abundance of Enterobacteriaceae in T1DM (P=0.0193). T1DM, type 1 diabetes mellitus. *P<0.05 and **P<0.01

Abundance of butyrate-producing
bacteria (A) Relative abundance of Clostridium leptum. (B)
Dysbiosis with a decrease of Butyricicoccus spp. in T1DM
group. T1DM, type 1 diabetes mellitus. *P<0.05.

Figure 4

Abundance of butyrate-producing bacteria (A) Relative abundance of Clostridium leptum. (B) Dysbiosis with a decrease of Butyricicoccus spp. in T1DM group. T1DM, type 1 diabetes mellitus. *P<0.05.

A decrease in butyrate-producing bacteria was observed in the T1DM group with dysbiosis with increased abundance of Enterobacteriaceae in T1DM (P=0.0193; Fig. 3). This pattern underscores the alteration in the microbial composition associated with T1DM, suggesting a potential role of gut microbiota dysregulation in the pathogenesis of the disease.

Correlation analysis identified significant correlations between specific gut microbiota taxa and clinical parameters in pediatric T1DM (Fig. 5). The relative abundance of Butyricicoccus was positively associated with younger age at onset (r=0.6276, P=0.0018) but negatively linked to onset within 6 months (r=-0.517, P=0.0164), pancreatic β cell reserve (HOMA-B, r=-0.5962, P=0.0034), C-peptide (r=-0.5005, P=0.0344) and phosphorus levels (r=-0.5937, P=0.0036). Bacteroides relative abundance showed negative correlations with early-stage diabetes (r=-0.4431, P=0.0442) and inflammation (indicated by CRP levels; r=-0.4331; P=0.0498). The relative abundance of Clostridium leptum was inversely associated with recent diagnosis (r=-0.6278, P=0.0023) but positively correlated with hemoglobin (r=0.4765, P=0.025) and PTH levels (r=0.6047, P=0.0029). Antibiotic intake was correlated positively with the relative abundance of Candida albicans (r=0.4431, P=0.0442), while the inflammatory index was linked positively to Enterobacteriaceae (r=0.4331, P=0.0441) and negatively with C. coccoides (r=-0.4241, P=0.0492). Hemoglobin was also positively associated with Butyricicoccus (r=0.5857, P =0.0042), highlighting potential microbiota roles in T1DM progression and inflammation.

Spearman correlation coefficients
between clinical parameters (and relative abundance of bacterial
taxa in the microbiome. Red, positive correlation; blue, negative
correlations; intensity of color corresponds to the magnitude of
the correlation. HbA1C, Hemoglobin A1c (glycated hemoglobin);
HOMA-B, Homeostatic Model Assessment of β-cell function; IGF,
Insulin-like Growth Factor; CRP, C-reactive Protein; TTG Ab, Tissue
Transglutaminase Antibodies; fT4, Free Thyroxine; TSH,
Thyroid-Stimulating Hormone; PTH, Parathyroid Hormone.

Figure 5

Spearman correlation coefficients between clinical parameters (and relative abundance of bacterial taxa in the microbiome. Red, positive correlation; blue, negative correlations; intensity of color corresponds to the magnitude of the correlation. HbA1C, Hemoglobin A1c (glycated hemoglobin); HOMA-B, Homeostatic Model Assessment of β-cell function; IGF, Insulin-like Growth Factor; CRP, C-reactive Protein; TTG Ab, Tissue Transglutaminase Antibodies; fT4, Free Thyroxine; TSH, Thyroid-Stimulating Hormone; PTH, Parathyroid Hormone.

The microbiota findings were compared with the metabolite levels in stool samples from both patients with T1DM and healthy controls (Fig. 6). Patients with T1DM exhibited notably decreased levels of butyrate and lactate compared with the healthy controls. Conversely, acetate levels were higher in the T1DM cohort.

Fecal metabolite analysis in patients
with T1DM and healthy controls. In fecal samples, acetate
represented the predominant SCFA, accounting for ~85% in controls
and ~92% in T1DM patients. Lactate and butyrate contributed ~10 and
~5% in controls, compared with ~6 and ~2% in T1DM patients,
respectively. T1DM, type 1 diabetes mellitus.

Figure 6

Fecal metabolite analysis in patients with T1DM and healthy controls. In fecal samples, acetate represented the predominant SCFA, accounting for ~85% in controls and ~92% in T1DM patients. Lactate and butyrate contributed ~10 and ~5% in controls, compared with ~6 and ~2% in T1DM patients, respectively. T1DM, type 1 diabetes mellitus.

Discussion

The present retrospective case-control study was conducted in Romania between January 2019 and December 2021, involving 31 children. The timeline of analysis and manuscript preparation was substantially affected by the coronavirus Disease 2019 (COVID-19) pandemic. Institutional priorities during this period were redirected toward pandemic-related clinical and research activities, and access to laboratories and clinical facilities was intermittently restricted. These factors contributed to an extended delay in data processing, analysis, and publication. Nevertheless, all study data were securely stored, curated, and analyzed in accordance with the approved protocol, ensuring the integrity and validity of the findings.

The role of microbiota in T1DM has garnered increasing attention and investigation: Previous studies have linked specific bacterial taxa such as Bacteroides, Bifidobacterium and Ruminococcus with heightened cytokine levels, pancreatic inflammation, autoimmune reaction and the onset of T1DM (1,2,4). The present study demonstrated dysbiosis characterized by an overabundance of proinflammatory bacteria (including C. coccoides, Faecalibacterium, Bacteroides and Enterobacteriaceae) and fungi in pediatric patients with T1DM from Romania, in comparison with healthy controls.

Contrary to the healthy control group, patients with T1DM exhibited a microbiota predominantly composed of Bacteroides taxa, despite the presence of relatively abundant protective bacteria such as Bifidobacterium and Lactobacillus spp., and A. muciniphila. Notably, earlier onset and more severe presentation of T1DM were positively associated with specific proinflammatory bacterial species, including Bacteroides and Butyricicoccus spp., in alignment with the literature (16,52). C. leptum is another taxon correlated with a younger age at onset and although its role may vary depending on the context, host health, and microbiota composition its altered abundance may contribute to disease susceptibility and progression (53). In gut dysbiosis, its abundance may shift, and interactions with other microbial taxa could influence systemic inflammation indirectly. However, it is primarily reported as a contributor to a balanced, healthy gut environment rather than as a proinflammatory agent (21-23).

Studies conducted in a Romanian center on patients with type 2 diabetes mellitus (T2DM) or metabolic syndrome have revealed similar microbial compositions, particularly the presence of Enterobacteriaceae, which is correlated with inflammation and other chronic diseases (53,54).

The Spearman correlation analysis demonstrated associations between specific microbial taxa and clinical markers, including those of inflammation, metabolic status and endocrine function, suggesting a potential role of the gut microbiome in modulating health outcomes in the context of metabolic disorder.

The presence of proinflammatory bacteria such as Enterobacteriaceae showed a positive association with the systemic inflammatory index; relative abundance of anti-inflammatory bacteria as Bacteroides and C. coccoides showed a negative association with CRP levels and with the systemic inflammatory index (23-25).

Antibiotic use during acute infections in the 2 months preceding diagnosis in children with T1DM may have perturbed the microbiota, resulting in an overabundance of Candida spp (45,53,54). Additionally, T1DM is associated with an increased susceptibility to recurrent infections and alterations in the immune system within the first year of the disease, as well as lower complement component 4) levels and a higher helper/suppressor T lymphocyte ratio (55,56). These findings are consistent with those reported by other authors (55-57).

Assessing growth and bone density is key in children with T1DM and T2DM, given the link between impaired bone health and dysbiosis characterized by an elevated presence of Lactobacilli (58). In the present study, dysbiosis impacted calcium-phosphorous metabolism; phosphorus levels negatively correlated with Butyricicoccus and PTH levels were positively correlated with C. leptum (56,59,60).

The role of Bacteroides in intestinal homeostasis, particularly in T1DM, is complex and context-dependent. Bacteroides spp. exhibit both pro-inflammatory and anti-inflammatory properties, depending on species, strain and host factors. On one hand, certain Bacteroides strains (such as B. fragilis, producing polysaccharide A) have been shown to exert anti-inflammatory effects by promoting regulatory T cell response and maintaining mucosal barrier integrity (61,62) On the other hand, other species or imbalances in Bacteroides abundance are associated with pro-inflammatory states, including increased intestinal permeability and heightened immune activation, which are relevant to the pathogenesis of T1DM (11).

Here, the enrichment of Bacteroides in patients with T1DM may reflect a shift toward a dysbiotic profile that contributes to inflammation and autoimmune activation. However, not all Bacteroides are pathogenic, and their impact may vary with microbial context, age and diet. Future strain-level or functional metagenomic analyses may clarify the specific contributions of Bacteroides in the T1DM gut microbiome.

Patients with T1DM exhibited notably decreased levels of butyrate and lactate compared with healthy controls, with higher acetate levels in the T1DM cohort, diverging from other studies that found decreased plasma levels of acetate and propionate and similar levels of plasma butyrate in controls compared with patients with T1DM (11,24). Measurements of SCFA levels (butyrate, lactate and acetate) were obtained from fecal samples, which primarily reflect microbial fermentation activity in the colon and the unabsorbed SCFA fraction. This contrasts with other studies reporting SCFA concentrations in plasma, which are influenced not only by gut microbial production but also by host absorption efficiency, hepatic metabolism and systemic circulation dynamics (62,63). Therefore, direct comparisons between fecal and plasma SCFA levels must be interpreted with caution. The increase in fecal acetate alongside decreased butyrate and lactate in the present T1DM cohort may indicate altered microbial metabolism or impaired SCFA absorption in these patients, rather than a contradiction of plasma findings reported in previous studies (63,64).

The present study demonstrated the role of gut microbiota in pediatric patients with T1DM. It highlighted dysbiosis characterized by an overabundance of proinflammatory bacteria such as Bacteroides, Enterobacteriaceae and C. coccoides, alongside fungi such as Candida spp, in patients with T1DM compared with healthy controls. This aligns with existing research, underscoring the association of these taxa with inflammation, autoimmune reaction and metabolic imbalances (11,14). The study also demonstrated correlations between microbial taxa and clinical markers, such as the systemic inflammatory index, CRP levels and calcium-phosphorous metabolism, emphasizing the microbiota influence on systemic inflammation, bone health and disease progression.

Moreover, the perturbation of microbiota following antibiotic use and the increased prevalence of recurrent infections in patients with T1DM highlight the dynamic interactions between external factors, immune function and the gut microbiome. By revealing specific microbiota compositions associated with earlier disease onset, severity and metabolic markers, the present study demonstrated how gut dysbiosis impacts T1DM pathophysiology. These findings may facilitate research into microbiota-targeted interventions, such as dietary modification or probiotic therapy, to improve disease management and patient outcomes in T1DM.

However, the present study had limitations, including the heterogeneous composition of the study group, its retrospective nature with a small patient cohort and the commencement of enrollment a few months before the onset of the COVID-19 pandemic and lockdown, primarily involving an urban population. Additionally, the microbiota study excluded patients who received antibiotics following infections, particularly those with diabetic ketoacidosis.

Future research directions include proteomics and metabolomics profiling to evaluate functional changes in the microbiome of patients with T1DM. Longitudinal studies observing disease onset and progression in patients with dysbiosis are also warranted. Furthermore, the associations between dysbiosis and other autoimmune diseases (such as Hashimoto's thyroiditis and celiac disease) need to be investigated with regard to the correlation between antibody positivity and clinical disease onset.

The present data may be used to estimate the risk of developing T1DM and other autoimmune diseases in the siblings of these patients. Risk calculators that consider specific genetic traits (human leukocyte antigen haplotypes), antibody presence and family history of the disease may predict the likelihood of developing diabetes before symptoms manifest. Early intervention, when a reasonable number of insulin-producing β cells are present in the pancreas, may positively influence immune responses by modifying gut microbiota, potentially slowing disease progression and improving outcomes.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by Executive Agency for Higher Education, Research, Development and Innovation Funding (project ID no. PN-III-P1-1.1-PD-2019-0499, grant no. 224/2021).

Availability of data and materials

The data generated in the present study are not publicly available due to participant privacy but may be requested from the corresponding author.

Authors' contributions

AAI, SF and GGP conceived the study. AAI and GGP confirm the authenticity of all the raw data. SI designed the methodology. TP analyzed data. AAI and GGP performed experiments. AAI wrote and edited the manuscript, constructed figures and supervised the study. GGP edited the manuscript. SF and GGP acquired funding. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

The present study was conducted according to the guidelines of The Declaration of Helsinki and was approved by the Ethics Committee of University of Bucharest (protocol code CEC reg. no. 235/9.10.2019) and the Institutional Review Board of the Elias Hospital (approval no. 1695, 12.03.2019; both Bucharest, Romania). Informed consent was obtained from the parents of patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Ogle GD, Wang F, Haynes A, Gregory GA, King TW, Deng K, Dabelea D, James S, Jenkins AJ, Li X, et al: Global type 1 diabetes prevalence, incidence, and mortality estimates 2025: Results from the International Diabetes Federation Atlas, 11th Edition, and the T1D Index Version 3.0. Diabetes Res Clin Pract. 225(112277)2025.PubMed/NCBI View Article : Google Scholar

2 

Dedrick S, Sundaresh B, Huang Q, Brady C, Yoo T, Cronin C, Rudnicki C, Flood M, Momeni B, Ludvigsson J and Altindis E: The role of gut microbiota and environmental factors in type 1 diabetes pathogenesis. Front Endocrinol (Lausanne). 11(78)2020.PubMed/NCBI View Article : Google Scholar

3 

Penders J, Thijs C, Vink C, Stelma FF, Snijders B, Kummeling I, Van den Brandt PA and Stobberingh EE: Factors influencing the composition of the intestinal microbiota in early infancy. Pediatrics. 118:511–521. 2006.PubMed/NCBI View Article : Google Scholar

4 

Gradisteanu Pircalabioru G, Corcionivoschi N, Gundogdu O, Chifiriuc MC, Marutescu LG, Ispas B and Savu O: Dysbiosis in the development of type I diabetes and associated complications: From mechanisms to targeted gut microbes manipulation therapies. Int J Mol Sci. 22(2763)2021.PubMed/NCBI View Article : Google Scholar

5 

Rampanelli E and Nieuwdorp M: Gut microbiome in type 1 diabetes: The immunological perspective. Expert Rev Clin Immunol. 19:93–109. 2023.PubMed/NCBI View Article : Google Scholar

6 

Abuqwider J, Corrado A, Scidà G, Lupoli R, Costabile G, Mauriello G and Bozzetto L: Gut microbiome and blood glucose control in type 1 diabetes: A systematic review. Front Endocrinol (Lausanne). 14(1265696)2023.PubMed/NCBI View Article : Google Scholar

7 

Wen L, Ley RE, Volchkov PY, Stranges PB, Avanesyan L, Stonebraker AC, Hu C, Wong FS, Szot GL, Bluestone JA, et al: Innate immunity and intestinal microbiota in the development of type 1 diabetes. Nature. 455:1109–1113. 2008.PubMed/NCBI View Article : Google Scholar

8 

Markle JG, Frank DN, Mortin-Toth S, Robertson CE, Feazel LM, Rolle-Kampczyk U, von Bergen M, McCoy KD, Macpherson AJ and Danska JS: Sex differences in the gut microbiome drive hormone-dependent regulation of autoimmunity. Science. 339:1084–1088. 2013.PubMed/NCBI View Article : Google Scholar

9 

Hansen CH, Krych L, Nielsen DS, Vogensen FK, Hansen LH, Sørensen SJ, Buschard K and Hansen AK: Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse. Diabetologia. 55:2285–2294. 2012.PubMed/NCBI View Article : Google Scholar

10 

Maffeis C, Martina A, Corradi M, Quarella S, Nori N, Torriani S, Plebani M, Contreas G and Felis GE: Association between intestinal permeability and faecal microbiota composition in Italian children with beta cell autoimmunity at risk for type 1 diabetes. Diabetes Metab. Res Rev. 32:700–709. 2016.PubMed/NCBI View Article : Google Scholar

11 

Kostic AD, Gevers D, Siljander H, Vatanen T, Hyötyläinen T, Hämäläinen AM, Peet A, Tillmann V, Pöhö P, Mattila I, et al: The dynamics of the human infant gut microbiome in development and in progression toward type 1 diabetes. Cell Host Microbe. 17:260–273. 2015.PubMed/NCBI View Article : Google Scholar

12 

Ziegler AG, Hoffmann GF, Hasford J, Larsson HE, Danne T, Berner R, Penno M, Koralova A, Dunne J and Bonifacio E: Screening for asymptomatic β-cell autoimmunity in young children. Lancet Child Adolesc Health. 3:288–290. 2019.PubMed/NCBI View Article : Google Scholar

13 

Vaarala O: Gut microbiota and type 1 diabetes. Rev Diabet Stud. 9:251–259. 2012.PubMed/NCBI View Article : Google Scholar

14 

Vatanen T, Franzosa EA, Schwager R, Tripathi S, Arthur TD, Vehik K, Lernmark Å, Hagopian WA, Rewers MJ, She JX, et al: The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature. 562:589–594. 2018.PubMed/NCBI View Article : Google Scholar

15 

Bäckhed F, Ley RE, Sonnenburg JL, Peterson DA and Gordon JI: Host-bacterial mutualism in the human intestine. Science. 307:1915–1920. 2025.PubMed/NCBI View Article : Google Scholar

16 

Murri M, Leiva I, Gomez-Zumaquero JM, Tinahones FJ, Cardona F, Soriguer F and Queipo-Ortuño MI: Gut microbiota in children with type 1 diabetes differs from that in healthy children: A case-control study. BMC Med. 11(46)2013.PubMed/NCBI View Article : Google Scholar

17 

Arts RJW, Joosten LAB and Netea MG: The potential role of trained immunity in autoimmune and autoinflammatory disorders. Front Immunol. 9(298)2018.PubMed/NCBI View Article : Google Scholar

18 

Marchesi JR and Ravel J: The vocabulary of microbiome research: A proposal. Microbiome. 3(31)2015.PubMed/NCBI View Article : Google Scholar

19 

Jung TH, Han KS, Park JH and Hwang HJ: Butyrate modulates mucin secretion and bacterial adherence in LoVo cells via MAPK signaling. PLoS One. 17(e0269872)2022.PubMed/NCBI View Article : Google Scholar

20 

Vatanen T, de Beaufort C, Marcovecchio ML, Overbergh L, Brunak S, Peakman M, Mathieu C and Knip M: INNODIA consortium. Gut microbiome shifts in people with type 1 diabetes are associated with glycaemic control: An INNODIA study. Diabetologia. 67:1930–1942. 2024.PubMed/NCBI View Article : Google Scholar

21 

Flint HJ, Scott KP, Duncan SH, Louis P and Forano E: Microbial degradation of complex carbohydrates in the gut. Gut Microbes. 3:289–306. 2012.PubMed/NCBI View Article : Google Scholar

22 

Hague A, Butt AJ and Paraskeva C: The role of butyrate in human colonic epithelial cells: An energy source or inducer of differentiation and apoptosis? Proc Nutr Soc. 55:937–943. 1996.PubMed/NCBI View Article : Google Scholar

23 

Lopetuso LR, Scaldaferri F, Petito V and Gasbarrini A: Commensal Clostridia: Leading players in the maintenance of gut homeostasis. Gut Pathog. 5(23)2013.PubMed/NCBI View Article : Google Scholar

24 

Leiva-Gea I, Sánchez-Alcoholado L, Martín-Tejedor B, Castellano-Castillo D, Moreno-Indias I, Urda-Cardona A, Tinahones FJ, Fernández-García JC and Queipo-Ortuño MI: Gut microbiota differs in composition and functionality between children with type 1 Diabetes and MODY2 and healthy control subjects: A case-control study. Diabetes Care. 41:2385–2395. 2018.PubMed/NCBI View Article : Google Scholar

25 

Brown CT, Davis-Richardson A, Giongo A, Gano KA, Crabb DB, Mukherjee N, Casella G, Drew JC, Ilonen J, Knip M, et al: Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes. PLoS One. 6(e25792)2011.PubMed/NCBI View Article : Google Scholar

26 

Valladares R, Sankar D, Li N, Williams E, Lai KK, Abdelgeliel AS, Gonzalez CF, Wasserfall CH, Larkin J, Schatz D, et al: Lactobacillus johnsonii N6.2 mitigates the development of type 1 diabetes in BB-DP rats. PLoS One. 5(e10507)2010.PubMed/NCBI View Article : Google Scholar

27 

Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature. 486:207–214. 2012.PubMed/NCBI View Article : Google Scholar

28 

Hypponen E: Vitamin D and increasing incidence of type 1 diabetes-evidence for an association? Diabetes Obes Metab. 12:737–743. 2010.PubMed/NCBI View Article : Google Scholar

29 

Chia LW, Mank M, Blijenberg B, Aalvink S, Bongers RS, Stahl B, Knol J and Belzer C: Bacteroides thetaiotaomicron fosters the growth of butyrate-producing Anaerostipes caccae in the presence of lactose and total human milk carbohydrates. Microorganisms. 8(1513)2020.PubMed/NCBI View Article : Google Scholar

30 

Zipitis CS and Akobeng AK: Vitamin D supplementation in early childhood and risk of type 1 diabetes: A systematic review and meta-analysis. Arch Dis Child. 93:512–517. 2008.PubMed/NCBI View Article : Google Scholar

31 

Karvonen M, Viik-Kajander M, Moltchanova E, Libman I, LaPorte R and Tuomilehto J: Incidence of childhood type 1 diabetes worldwide. Diabetes Mondiale (DiaMond) Project Group. Diabetes Care. 23:1516–1526. 2000.PubMed/NCBI View Article : Google Scholar

32 

Pozzilli P, Manfrini S, Crinò A, Picardi A, Leomanni C, Cherubini V, Valente L, Khazrai M and Visalli N: IMDIAB group. Low levels of 25-hydroxyvitamin D3 and 1,25-dihydroxyvitamin D3 in patients with newly diagnosed type 1 diabetes. Horm Metab Res. 37:680–683. 2005.PubMed/NCBI View Article : Google Scholar

33 

Borkar VV, Devidayal Verma S and Bhalla AK: Low levels of vitamin D in North Indian children with newly diagnosed type 1 diabetes. Pediatr Diabetes. 11:345–50. 2010.PubMed/NCBI View Article : Google Scholar

34 

Hagopian WA, Erlich H, Lernmark Å, Rewers M, Ziegler AG, Simell O, Akolkar B, Vogt R Jr, Blair A, Ilonen J, et al: The environmental determinants of diabetes in the young (TEDDY): Genetic criteria and international diabetes risk screening of 421,000 infants. Pediatr Diabetes. 12:733–743. 2011.PubMed/NCBI View Article : Google Scholar

35 

Lönnrot M, Lynch KF, Elding Larsson H, Lernmark Å, Rewers MJ, Törn C, Burkhardt BR, Briese T, Hagopian WA, She JX, et al: TEDDY study group: Respiratory infections are temporally associated with initiation of type 1 diabetes autoimmunity: The TEDDY study. Diabetologia. 60:1931–1940. 2017.PubMed/NCBI View Article : Google Scholar

36 

Rewers M and Ludvigsson J: Environmental risk factors for type 1 diabetes. Lancet. 387:2340–2348. 2016.PubMed/NCBI View Article : Google Scholar

37 

Vlad A, Serban V, Green A, Möller S, Vlad M, Timar B and Sima A: ONROCAD Study Group. Time trends, regional variability and seasonality regarding the incidence of type 1 diabetes mellitus in Romanian children aged 0-14 years, between 1996 and 2015. J Clin Res Pediatr Endocrinol. 10:92–99. 2018.PubMed/NCBI View Article : Google Scholar

38 

Ionescu-Tîrgoviște C, Guja C, Călin A and Moţa M: An increasing trend in the incidence of type 1 diabetes mellitus in children aged 0-14 years in Romania-ten years (1988-1997) EURODIAB study experience. J Pediatr Endocrinol Metab. 17:983–991. 2004.PubMed/NCBI View Article : Google Scholar

39 

Gyürüs EK, Patterson C and Soltész G: Hungarian Childhood Diabetes Epidemiology Group. Twenty-one years of prospective incidence of childhood type 1 diabetes in Hungary-the rising trend continues (or peaks and highlands?). Pediatr Diabetes. 13:21–25. 2012.PubMed/NCBI View Article : Google Scholar

40 

Jarosz-Chobot P, Polańska J, Szadkowska A, Kretowski A, Bandurska-Stankiewicz E, Ciechanowska M, Deja G, Mysliwiec M, Peczynska J, Rutkowska J, et al: Rapid increase in the incidence of type 1 diabetes in Polish children from 1989 to. 2004, and predictions for 2010 to 2025. Diabetologia. 54:508–515. 2011.PubMed/NCBI View Article : Google Scholar

41 

Usher-Smith JA, Thompson M, Ercole A and Walter FM: Variation between countries in the frequency of diabetic ketoacidosis at first presentation of type 1 diabetes in children: A systematic review. Diabetologia. 55:2878–2894. 2012.PubMed/NCBI View Article : Google Scholar

42 

Triolo TM, Armstrong TK, McFann K, Yu L, Rewers MJ, Klingensmith GJ, Eisenbarth GS and Barker JM: Additional autoimmune disease found in 33% of patients at type 1 diabetes onset. Diabetes Care. 34:1211–1213. 2011.PubMed/NCBI View Article : Google Scholar

43 

Akhtadze E, Cilio CM, Hansen I, Gerdes AM, Mortensen HB, Pociot F and Nerup J: Distribution of autoimmune diseases in Danish children and adolescents with type 1 diabetes mellitus. Horm Res. 65:193–198. 2006.

44 

ElSayed NA, Aleppo G, Aroda VR, Bannuru RR, Brown FM, Bruemmer D, Collins BS, Hilliard ME, Isaacs D, Johnson EL, et al: 2. Classification and diagnosis of diabetes: Standards of Medical Care in Diabetes-2023. Diabetes Care. 46 (Suppl 1):S19–S40. 2023.PubMed/NCBI View Article : Google Scholar

45 

de Groot PF, Belzer C, Levels JH, Aalvink S, Boot F, Holleman F, Van Raalte DH, Scheithauer TP, Simsek S, Schaap FG, et al: Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS One. 12(e0188475)2017.PubMed/NCBI View Article : Google Scholar

46 

Thomson G, Valdes AM, Noble JA, Kockum I, Grote MN, Najman J, Erlich HA, Cucca F, Pugliese A, Steenkiste A, et al: Relative predispositional effects of HLA class II DRB1-DQB1 haplotypes genotypes on type 1 diabetes: a meta-analysis. Tissue Antigens. 70:110–127. 2007.PubMed/NCBI View Article : Google Scholar

47 

Emmanuel M and Bokor BR: Tanner Stages. 2022 Dec 11. In StatPearls. StatPearls Publishing, Treasure Island, FL, 2025. https://www.ncbi.nlm.nih.gov/books/NBK470280/.

48 

Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF and Turner RC: Homeostasis model assessment: Insulin resistance and β-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 28:412–419. 1985.PubMed/NCBI View Article : Google Scholar

49 

Trandafir M, Pircalabioru GG and Savu O: Microbiota analysis in individuals with type two diabetes mellitus and end-stage renal disease: A pilot study. Exp Ther Med. 27(211)2024.PubMed/NCBI View Article : Google Scholar

50 

Srinivasan R, Karaoz U, Volegova M, MacKichan J, Kato-Maeda M, Miller S, Nadarajan R, Brodie EL and Lynch SV: Use of 16S rRNA gene for identification of a broad range of clinically relevant bacterial pathogens. PLoS One. 10(e0117617)2015.PubMed/NCBI View Article : Google Scholar

51 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

52 

Giongo A, Gano KA, Crabb DB, Mukherjee N, Novelo LL, Casella G, Drew JC, Ilonen J, Knip M, Hyöty H, et al: Toward defining the autoimmune microbiome for type 1 diabetes. ISME J. 5:82–91. 2011.PubMed/NCBI View Article : Google Scholar

53 

Gradisteanu Pircalabioru G, Chifiriuc MC, Picu A, Petcu LM, Trandafir M and Savu O: Snapshot into the type-2-diabetes-associated microbiome of a romanian cohort. Int J Mol Sci. 23(15023)2022.PubMed/NCBI View Article : Google Scholar

54 

Gradisteanu Pircalabioru G, Ilie I, Oprea L, Picu A, Petcu LM, Burlibasa L, Chifiriuc MC and Musat M: Microbiome, mycobiome and related metabolites alterations in patients with metabolic syndrome-a pilot study. Metabolites. 12(218)2022.PubMed/NCBI View Article : Google Scholar

55 

Hull CM, Peakman M and Tree TIM: Regulatory T cell dysfunction in type 1 diabetes: what's broken and how can we fix it? Diabetologia. 60:1839–1850. 2017.PubMed/NCBI View Article : Google Scholar

56 

Alnek K, Tagoma A, Metsküla K, Talja I, Janson H, Mandel M, Vorobjova T, Oras A, Sepp H, Pruul K, et al: Comparison of immunological and immunogenetic markers in recent-onset type 1 diabetes among children and adults. Sci Rep. 15(15491)2025.PubMed/NCBI View Article : Google Scholar

57 

Mazzoni MB, Roversi M, Maltoni G, Biasucci G, Berardi A, Tonielli E, Cicognani A and Salardi S: Gualandi. Recurrent infections in children and adolescents with type 1 diabetes mellitus: A case-control study. BMC Infect Dis. 18(599)2018.

58 

Hygum K, Starup-Linde J, Harsløf T, Vestergaard P and Langdahl BL: Mechanisms in endocrinology: Diabetes mellitus, a state of low bone turnover-a systematic review and meta-analysis. Eur J Endocrinol. 176:R137–R157. 2017.PubMed/NCBI View Article : Google Scholar

59 

Knudsen JK, Leutscher P and Sørensen S: Gut microbiota in bone health and diabetes. Curr Osteoporos Rep. 19:462–479. 2021.PubMed/NCBI View Article : Google Scholar

60 

Janner M and Saner C: Impact of type 1 diabetes mellitus on bone health in children. Horm Res Paediatr. 95:205–214. 2022.PubMed/NCBI View Article : Google Scholar

61 

Round JL and Mazmanian SK: Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota. Proc Natl Acad Sci USA. 107:12204–12209. 2010.PubMed/NCBI View Article : Google Scholar

62 

Mazmanian SK, Round JL and Kasper DL: A microbial symbiosis factor prevents intestnal inflammatory disease. Nature. 453:620–625. 2008.PubMed/NCBI View Article : Google Scholar

63 

den Besten G, van Eunen K, Groen AK, Venema K, Reijngoud DJ and Bakker BM: The role of short-chain fatty acids in the interplay between diet, gut microbiota and host energy metabolism. J Lipid Res. 54:2325–2340. 2013.PubMed/NCBI View Article : Google Scholar

64 

de Groot PF, Belzer C, Aydin Ö, Levin E, Levels JH, Aalvink S, Boot F, Holman R, Reitsma PH, Gerdes VE and Nieuwdorp M: Distinct fecal and oral microbiota composition in human type 1 diabetes, an observational study. PLoS ONE. 12(e0188475)2017.PubMed/NCBI View Article : Google Scholar

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Arhire AI, Papuc T, Ioacără S, Gradisteanu Pircalabioru G and Barbu CG: Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients. Biomed Rep 24: 1, 2026.
APA
Arhire, A.I., Papuc, T., Ioacără, S., Gradisteanu Pircalabioru, G., & Barbu, C.G. (2026). Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients. Biomedical Reports, 24, 1. https://doi.org/10.3892/br.2025.2074
MLA
Arhire, A. I., Papuc, T., Ioacără, S., Gradisteanu Pircalabioru, G., Barbu, C. G."Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients". Biomedical Reports 24.1 (2026): 1.
Chicago
Arhire, A. I., Papuc, T., Ioacără, S., Gradisteanu Pircalabioru, G., Barbu, C. G."Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients". Biomedical Reports 24, no. 1 (2026): 1. https://doi.org/10.3892/br.2025.2074
Copy and paste a formatted citation
x
Spandidos Publications style
Arhire AI, Papuc T, Ioacără S, Gradisteanu Pircalabioru G and Barbu CG: Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients. Biomed Rep 24: 1, 2026.
APA
Arhire, A.I., Papuc, T., Ioacără, S., Gradisteanu Pircalabioru, G., & Barbu, C.G. (2026). Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients. Biomedical Reports, 24, 1. https://doi.org/10.3892/br.2025.2074
MLA
Arhire, A. I., Papuc, T., Ioacără, S., Gradisteanu Pircalabioru, G., Barbu, C. G."Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients". Biomedical Reports 24.1 (2026): 1.
Chicago
Arhire, A. I., Papuc, T., Ioacără, S., Gradisteanu Pircalabioru, G., Barbu, C. G."Unveiling the gut connection: Exploring the link between microbiota and type 1 diabetes onset in pediatric patients". Biomedical Reports 24, no. 1 (2026): 1. https://doi.org/10.3892/br.2025.2074
Follow us
  • Twitter
  • LinkedIn
  • Facebook
About
  • Spandidos Publications
  • Careers
  • Cookie Policy
  • Privacy Policy
How can we help?
  • Help
  • Live Chat
  • Contact
  • Email to our Support Team