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
International Journal of Molecular Medicine
Join Editorial Board Propose a Special Issue
Print ISSN: 1107-3756 Online ISSN: 1791-244X
Journal Cover
December-2025 Volume 56 Issue 6

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
December-2025 Volume 56 Issue 6

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
Review Open Access

Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review)

  • Authors:
    • Di Cheng
    • Nan Li
    • Qian Sun
    • Kun Wang
    • Fengchun Gao
  • View Affiliations / Copyright

    Affiliations: Department of Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250000, P.R. China, Department of Obstetrics, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250000, P.R. China, Department of Clinical Laboratory, Jinan Maternity and Child Care Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250000, P.R. China
    Copyright: © Cheng et al. This is an open access article distributed under the terms of Creative Commons Attribution License.
  • Article Number: 203
    |
    Published online on: September 23, 2025
       https://doi.org/10.3892/ijmm.2025.5644
  • 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

Preterm birth (PTB) is a global maternal and neonatal health challenge, affecting ~15 million infants each year. Despite advances in obstetric and neonatal care, PTB‑related morbidity and mortality remain high. Emerging evidence implicates dysbiosis of the vaginal microbiota (VMB) as a key contributor to PTB. A healthy VMB is typically dominated by Lactobacillus spp., which maintain an acidic vaginal environment and inhibit pathogen colonization. Conversely, reduced Lactobacilli abundance alongside overgrowth of anaerobic taxa such as Gardnerella, Atopobium and Mycoplasma is strongly associated with spontaneous PTB and preterm premature rupture of membranes. Excessive proliferation of vaginal pathogens may lead to ascending infection and intra‑amniotic inflammation via activation of host Toll‑like receptor signaling and induction of pro‑inflammatory cytokines IL‑1β, IL‑6 and IL‑8. Moreover, VMB‑derived metabolites such as lactate play important roles in immunomodulation and inflammation. Although antibiotics remain the mainstay for treating bacterial vaginosis, their non‑specific effects often disrupt microbial balance and predispose to recurrence. Recently, probiotic therapies and VMB transplantation have emerged as promising alternative or adjunctive strategies for PTB prevention and management. However, variability in probiotic efficacy and lack of standardized intervention protocols remain significant challenges. The present review examined pregnancy‑associated VMB dynamics, the mechanisms linking dysbiosis to PTB risk and future microbiome‑based intervention strategies, with the aim of informing theoretical and practical approaches to reduce the global burden of preterm birth.

Introduction

Preterm birth (PTB) is a global maternal and neonatal health problem, with ~15 million infants born preterm each year, 11% of all live births (1). PTB contributes to 15% of under-five childhood mortality and 35% of neonatal mortality worldwide (2). It is associated with a range of serious complications, including bronchopulmonary dysplasia, respiratory distress syndrome, intraventricular hemorrhage, neurodevelopmental impairment, and necrotizing enterocolitis (2), which rank among the leading direct causes of mortality in children under five. Roughly 40-45% of PTB cases occur spontaneously, 25-30% result from preterm premature rupture of the membranes (PPROM), and the remaining 30-35% are due to medically indicated or elective early deliveries (3). Intra-amniotic infection is estimated to underlie ~35% of spontaneous PTB and 50% of PPROM cases (3,4). Studies have isolated specific bacterial species from the amniotic fluid of PTB patients and confirmed that these organisms are common vaginal colonizers, supporting ascending vaginal infection as the primary route of microbial invasion into the amniotic cavity (4,5).

Vaginal microbiota (VMB) plays a critical role in maintaining maternal and fetal health (6). For example, Tabatabaei et al (7) reported that Lactobacillus spp. may reduce the risk of PTB, whereas community state types (CSTs) associated with bacterial vaginosis (BV) may increase PTB risk. Moreover, a Lactobacillus-depleted CST, accompanied by elevated abundances of Gardnerella or Mycoplasma, is linked to higher PTB incidence (8). Compared with early gestation, the VMB becomes increasingly stable and Lactobacillus-dominated in late pregnancy (9,10), which may represent an evolutionary mechanism to ensure successful pregnancy. By contrast, VMBs dominated by other genera, particularly those associated with BV, are generally considered suboptimal and correlate with greater adverse outcomes (11,12).

While previous reviews, for example Giannella et al (13), have characterized shifts in the maternal microbiome during pregnancy, their focus has been largely limited to oral and gastrointestinal communities with minimal consideration of vaginal populations. Bayar et al (14) examined the association between the pregnancy microbiome and PTB yet did not address how vaginal microbial communities relate to risk factors such as bacterial vaginosis nor did they explore mechanistic pathways linking vaginal microbes, host immunity, inflammation and microbial metabolites to PTB. In response, the present review summarized the composition and dynamic shifts of the VMB, with emphasis on its pregnancy-specific alterations. It examined the potential mechanisms by which vaginal dysbiosis and BV contribute to PTB, including inflammatory responses, immune modulation and the impact of microbial metabolites on pregnancy outcomes. It further explored molecular pathways driving ascending infection following VMB imbalance, such as Toll-like receptor (TLR)/NF-κB signaling activation and subsequent inflammatory cascades. Finally, it critically analyzed current clinical strategies for restoring vaginal ecosystem homeostasis, through probiotics or VMB transplantation to prevent PTB, highlighting uncertainties in probiotic efficacy and outlining questions for future research. Collectively, these insights aimed to inform novel approaches for vaginal microbiome modulation in PTB prevention.

Data collection methods

PubMed and Web of Science were searched from database inception through 31 May 2025. Medical Subject Headings and free text terms 'preterm birth,' 'vaginal microbiota,' 'microbiota,' and 'probiotic' were combined in the search strategy.

Composition and pregnancy-induced dynamics of the vaginal microbiota
Establishment and stability of the vaginal microbial ecosystem

The VMB plays a crucial role in female reproductive tract health and disease (15). A Lactobacillus-dominated VMB is generally considered indicative of a healthy vaginal environment, whereas an unhealthy VMB is characterized by high microbial diversity and heterogeneity, with reduced Lactobacillus abundance and increased loads of anaerobic bacteria, including Gardnerella, Atopobium, Mobiluncus, Prevotella, Streptococcus, Chlamydia and Mycoplasma (16). Following menarche, elevated circulating estrogen promotes vaginal epithelial proliferation and glycogen deposition; the breakdown products of this glycogen serve as preferential carbon sources for Lactobacillus spp., reinforcing their dominance and lowering vaginal pH (17). This acidic milieu, together with bacteriocins and other antimicrobial compounds produced by lactobacilli, establishes a mucosal microenvironment that is inhospitable to most other microorganisms but favorable to Lactobacillus spp. Consequently, Lactobacillus spp. are regarded as keystone members of the VMB during the reproductive years and contribute to protection against BV, pelvic inflammatory disease, candidiasis, sexually transmitted infections and oncogenic human papillomavirus infection (11,18). Ravel et al (18) characterized the VMB of reproductive-age women across four ethnic groups using 16S rRNA gene sequencing and defined five CSTs based on dominant genera and species (as shown in Fig. 1). CSTs I-III and V are LDOM communities (>90% Lactobacillus spp.), dominated respectively by L. crispatus, L. gasseri, L. iners and L. jensenii. By contrast, CST IV is called the Lactobacillus-depleted state and is dominated by strict and facultative anaerobes. It contains almost no Lactobacillus and shows high microbial diversity (19).

Community state types and stability
of the female vaginal microbiota. The vaginal microbiota can be
classified into five CSTs (I-V) based on dominant genera and
species. CST I, II, III and V are each dominated by
Lactobacillus crispatus, L. gasseri, L. iners
and L. jensenii, respectively, whereas CST IV is
characterized by a high diversity of strict and facultative
anaerobes. Vaginal microbial stability correlates positively with
circulating estrogen: during puberty and pregnancy, rising estrogen
levels promote a stable, lactobacilli-dominated community and
acidify vaginal pH. Conversely, estrogen withdrawal leads to
decreased microbial stability and a shift toward more diverse,
non-lactobacilli profiles. CST, community state types; VMB, vaginal
microbiota.

Figure 1

Community state types and stability of the female vaginal microbiota. The vaginal microbiota can be classified into five CSTs (I-V) based on dominant genera and species. CST I, II, III and V are each dominated by Lactobacillus crispatus, L. gasseri, L. iners and L. jensenii, respectively, whereas CST IV is characterized by a high diversity of strict and facultative anaerobes. Vaginal microbial stability correlates positively with circulating estrogen: during puberty and pregnancy, rising estrogen levels promote a stable, lactobacilli-dominated community and acidify vaginal pH. Conversely, estrogen withdrawal leads to decreased microbial stability and a shift toward more diverse, non-lactobacilli profiles. CST, community state types; VMB, vaginal microbiota.

Throughout a woman's life span, both intrinsic and extrinsic factors modulate VMB composition. Prior to menarche, low estrogen levels correlate with a high-diversity microbiota comprising aerobic, anaerobic and enteric bacteria (20). Although some women maintain VMB stability across the menstrual cycle, the endocrine withdrawal of estrogen and progesterone during menses, alongside endometrial shedding and vaginal bleeding, induces transient shifts marked by decreased Lactobacillus abundance that typically rebounds in the follicular phase (21). During pregnancy, hormonal stability, markedly elevated estrogen concentrations and the absence of menses favor a Lactobacillus dominated (LDOM) VMB (22). Estrogen is the principal host factor sustaining the VMB, promoting epithelial thickening and intracellular glycogen synthesis (23). Glycogen is hydrolyzed by host α-amylase into glucose and maltose, substrates fermented by Lactobacillus spp. into lactic acid, thereby reducing local pH (24). Moreover, various vaginal bacteria encode amylase-like enzymes capable of glycogen metabolism (25,26). Glycogen may also originate from epithelial-cell lysis induced by high lactate concentrations and cytolysins secreted by Lactobacilli, a process potentially mediated by hyaluronidase-1 and matrix metalloproteinase (MMP)-8 (27). With the onset of puberty and rising estrogen, vaginal pH acidifies and meta-analyses have demonstrated enhanced VMB stability and Lactobacillus predominance (28,29). Similarly, cyclical and pregnancy-related fluctuations in VMB stability parallel estrogen levels; elevated estrogen is consistently associated with increased VMB stability, which is beneficial for vaginal health. Conversely, decreased estrogen post-pregnancy and in menopause is linked to reduced VMB stability; however, estrogen therapy in postmenopausal women restores a pronounced LDOM state (30,31). Notably, estrogen-containing contraceptives increase Lactobacillus abundance in reproductive-aged women and decrease BV incidence (32,33), further supporting a causal role for estrogen in VMB regulation.

Pregnancy-induced shifts in microbial composition

During pregnancy, the VMB is characterized by decreased richness and diversity, with Lactobacillus spp. emerging as the dominant genus. This maintenance of microbial homeostasis is closely linked to rising estrogen levels (as shown in Fig. 2), which not only stimulate glycogen synthesis but also promote increased lactic acid production by Lactobacillus. As gestation advances into the late trimester, Lactobacillus proliferation is often accompanied by a marked decline in dysbiosis-associated taxa such as G. vaginalis, A. vaginae and P. bivia (34). Studies show that during pregnancy, vaginal microbiota communities dominated by Lactobacillus usually remain stable or slowly change into other community types still dominated by Lactobacillus (8,35,36). Communities showing dysbiosis in mid-gestation often become dominated by Lactobacillus. When Gardnerella vaginalis dominates, its communities are relatively unstable and usually shift to a Lactobacillus iners community (35,36). These dynamic shifts align with the overall trend toward an optimized vaginal microenvironment during pregnancy. As white women generally possess higher baseline Lactobacillus levels prior to conception (36), black women demonstrate a more pronounced transition toward Lactobacillus dominance and enhanced VMB stability throughout pregnancy (35,36), suggesting a stronger modulatory effect of gestation on the VMB in this population. At parturition, the precipitous decline in estrogen precipitates a significant destabilization of the VMB (37,38), leading to community restructuring: Lactobacillus proportions decrease while anaerobic genera such as Peptoniphilus, Prevotella and Anaerococcus proliferate; taxa whose overgrowth is often linked to adverse outcomes (8,37). Moreover, short interpregnancy intervals (<1 year) have been shown to markedly increase the risk of PTB and other obstetric complications (39-41).

Interplay between pregnancy, glycogen
metabolism and vaginal microbial homeostasis. Elevated estrogen
during pregnancy stimulates epithelial glycogen synthesis, which
vaginal bacterial enzymes hydrolyze into glucose and maltose. These
sugars are fermented by lactobacilli to produce lactic acid,
lowering vaginal pH and inhibiting dysbiosis-associated taxa (such
as Gardnerella vaginalis, Atopobium vaginae and
Prevotella bivia). Lactobacilli also secrete cytolysins that
induce epithelial cell lysis, triggering the release of
hyaluronidase-1 and MMP-8, thereby facilitating glycogen liberation
and further supporting lactobacilli growth. After parturition, the
sudden decline in estrogen precipitates microbial community
restructuring: lactobacilli proportions diminish and
dysbiosis-associated organisms expand. MMP, matrix
metalloproteinase.

Figure 2

Interplay between pregnancy, glycogen metabolism and vaginal microbial homeostasis. Elevated estrogen during pregnancy stimulates epithelial glycogen synthesis, which vaginal bacterial enzymes hydrolyze into glucose and maltose. These sugars are fermented by lactobacilli to produce lactic acid, lowering vaginal pH and inhibiting dysbiosis-associated taxa (such as Gardnerella vaginalis, Atopobium vaginae and Prevotella bivia). Lactobacilli also secrete cytolysins that induce epithelial cell lysis, triggering the release of hyaluronidase-1 and MMP-8, thereby facilitating glycogen liberation and further supporting lactobacilli growth. After parturition, the sudden decline in estrogen precipitates microbial community restructuring: lactobacilli proportions diminish and dysbiosis-associated organisms expand. MMP, matrix metalloproteinase.

Vaginal microbiota dysbiosis as a risk factor for PTB
Bacterial vaginosis: A prominent dysbiotic state linked to PTB

Intra-amniotic infection is a major trigger of PTB (42), particularly in early gestation (43). Most intra-amniotic infections originate from ascending colonization of the lower genital tract by bacteria present before or during pregnancy (44) and cultures of preterm fetal membranes have confirmed the involvement of BV-associated species (45). Moreover, asymptomatic BV in pregnant women confers more than a two-fold increased risk of PTB compared with BV-negative women (46); BV diagnosed before 20 weeks' gestation is associated with a five-fold higher risk (47) and diagnosis before 16 weeks with a seven-fold increase (48). Clinical trials have investigated antibiotic treatment of asymptomatic BV to prevent PTB (49,50) and several studies suggest efficacy in high-risk populations (51-53). In addition, BV has been identified as an independent risk factor for PTB (54). The global prevalence of BV ranges from 23-29% (55), characterized by replacement of Lactobacilli with anaerobic Gram-negative bacteria in the VMB (56). A cross-sectional study of 50 BV patients using denaturing gradient gel electrophoresis fingerprinting detected significant enrichment of Firmicutes, Fusobacteria, Bacteroidetes and Acinetobacter (57). In BV-associated VMBs, Gardnerella, Atopobium, Prevotella, Porphyromonas, Sneathia, Mobiluncus, Mycoplasma BVAB1, BVAB2, Mageeibacillus indolicus (BVAB3) and Peptostreptococcus are frequently enriched (56). Moreover, G. vaginalis has been reported in the majority of clinically diagnosed BV cases (58). Numerous G. vaginalis isolates produce vaginolysin, a cholesterol-dependent cytolysin that disrupts mammalian cell membranes and induces cell death. Amino-acid sequence variations among vaginolysin toxins from different G. vaginalis strains underlie differences in pathogenicity (59). G. vaginalis comprises four clades, G. vaginalis, G. leopoldii, G. piotii and G. swidsinskii (60,61), all of which can occur at low abundance in asymptomatic women, often co-existing within the same VMB (62). Overgrowth of these taxa leads to Lactobacillus depletion and disruption of the natural lactic acid defense mechanism, causing vaginal pH to rise above 4.5 (63,64). BV-associated bacteria damage the vaginal mucosal and epithelial barriers by producing biofilms, sialidases and cytolysins, elevating pH and secreting colonization-enhancing enzymes. High expression of cholesterol-dependent cytolysin genes in L. iners and G. vaginalis correlates with reduced Lactobacillus abundance (65). Epithelial cell models indicate that pore forming toxins such as vaginolysin from G. vaginalis (66) and putative inerolysin from L. iners (67) interact with lipid rafts to lyse epithelial cells, suggesting a mechanism for BV pathogenesis. Vaginal biofilms resist clearance of BV associated bacteria by acidic environments, lactobacilli derived antimicrobials, host immunity and antibiotics (56,68). Although their precise role remains unclear, these biofilms are considered critical for BV onset and recurrence (68,69). In vitro studies show that initial attachment of G. vaginalis may be mediated by L. iners or Peptoniphilus spp., while biofilm maturation is enhanced by Atopobium vaginae, Prevotella bivia, Fusobacterium nucleatum and Mobiluncus spp (68,70), indicating that interspecies interactions promote biofilm formation.

Specific microbial profiles associated with increased PTB risk

Abnormal VMB is recognized as a potential risk factor for PTB. Clinical studies show that bacteria isolated from the placenta and fetal membranes of preterm deliveries closely match those in the vagina, supporting the idea that ascending vaginal colonization by pathogens frequently leads to intra-amniotic infection and PTB (71) and can be reproduced in various animal models of ascending infection (72,73). Romero et al (74) compared the VMB of 18 women who delivered preterm with 70 term controls and found no significant differences in bacterial taxa, relative abundances, or CST distributions when stratified by gestational age. A Peruvian cohort likewise reported no association between vaginal CST and PTB (5). These discrepancies may reflect differences in maternal ethnicity, geographic location, and gestational age at sampling. Fettweis et al (35) profiled the VMB and cytokine milieu of 45 African-American women with PTB vs. 90 term-matched controls. Multi-omics analyses showed markedly lower levels of L. crispatus and higher abundances of BVAB1, Sneathia amnii, TM7-H1 and a subset of Prevotella in the PTB group. Term deliveries were more likely to harbor L. crispatus and exhibit reduced prevalence of A. vaginae and G. vaginalis. A small study of nulliparous African-American women noted a nonsignificant trend toward reduced diversity in spontaneous PTB (75), and another black cohort found that although specific Lactobacillus abundances were not linked to PTB risk, lower overall diversity associated with PTB among African-American women (76). In a UK study, L. iners dominance at 16 weeks was markedly associated with cervical shortening and PTB before 34 weeks, whereas L. crispatus dominance strongly predicted term birth; high-diversity communities were not linked to cervical shortening or PTB, but Lactobacillus depletion was relatively uncommon in that cohort (77). The role of L. iners remains ambiguous, as it appears in both healthy and dysbiotic VMBs. Unlike the exclusionary L. crispatus, L. iners has a reduced genome lacking key metabolic functions (78), suggesting that L. iners has evolved to tolerate other vaginal microbes, including pathogens, and is often found in transitional microbiota states. Consequently, L. iners may be unable to prevent pathogen overgrowth during pregnancy, indirectly fostering high-risk VMB profiles. Srinivasan et al (79) reported that high levels of L. iners can be detected in women who are BV positive or BV negative and across a broad range of vaginal pH values. Although this phenomenon has been extensively documented under BV conditions (80), it does not provide direct evidence that the species is pathogenic during dysbiosis. Of women ~50% are thought to harbor L. iners (23), so its persistent detection during BV is unsurprising. Another study showed that the detection rate of L. iners is similar among women with normal, intermediate and BV Nugent scores (81); however, relative abundance may be more decisive and further work is required to quantify both relative and absolute levels under diverse conditions. Shipitsyna et al (82) observed a decline in L. iners during active BV and therefore suggested that it is unlikely to be a primary pathogen; by contrast, other investigators argue that communities dominated by L. iners are more prone to shift toward dysbiosis, whereas those dominated by L. crispatus appear more stable (83). A semi-quantitative qPCR study that sampled Belgian women (predominantly Caucasian) throughout the menstrual cycle found that during active BV, menstruation and after intercourse, the previously dominant L. crispatus sharply decreased, while L. iners became predominant, often accompanied by a concomitant rise in Gardnerella vaginalis (84), indicating greater adaptability of L. iners to fluctuations in the vaginal environment such as BV. Macklaim et al (85) further demonstrated that genes involved in mannose and maltose uptake and glycogen degradation are markedly upregulated in L. iners under BV. Additional studies show that both the abundance and gene expression of L. iners vary with the phase of the menstrual cycle (86); it is also possible that host physiological changes first trigger BV and that L. iners subsequently adapts. Clarifying this temporal sequence will be critical for determining whether, and to what extent, L. iners is harmful to the host.

In a cohort of 111 pregnant women, those harboring only a single Lactobacillus species exhibited higher PTB risk than women with multiple Lactobacillus spp. In addition, this study also indicated that L. iners was the dominant species in PTB cases (87). Tabatabaei et al (7) similarly reported that Lactobacillus spp. may reduce PTB risk, whereas BV-associated CSTs increase it. Another study confirms that the Lactobacillus depleted CST (CST IV), marked by elevated Gardnerella or Mycoplasma, is linked to greater PTB risk (8), underscoring that Lactobacillus species diversity is a key determinant of pregnancy outcome. Four studies of PPROM cases revealed that ~50% of women exhibited moderate or low Lactobacillus dominance and high diversity (88). In individual PPROM patients, ~50% of those with Lactobacillus dominated VMBs prior to membrane rupture developed dysbiosis afterwards and erythromycin treatment exacerbated reductions in Lactobacillus alongside enrichment of potential pathogens (89). In a prospective cohort, 25% of women who later experienced PPROM had low Lactobacillus abundance and high microbial diversity before membrane rupture, compared with just 3% of women who delivered at term with intact membranes (90). Enhanced vaginal microbial diversity, particularly during the mid-gestational period, demonstrated a significant correlation with PPROM, while vaginal microbiota compositions lacking Lactobacillus predominance emerged as a consistent predictive biomarker for PPROM across all gestational age intervals.

Multiple studies have demonstrated significant population specific differences in vaginal microbiome composition and their associations with PTB risk. DiGiulio et al (91) identified ethnicity as a strong determinant of VMB composition and PTB risk. In a predominantly white cohort, CSTs characterized by Lactobacillus depletion, especially those enriched for Gardnerella or Ureaplasma, were associated with shorter gestational duration and higher PTB risk. A subsequent comparison of Californian white and Alabaman black women revealed more frequent Lactobacillus depletion and Gardnerella enrichment among black women, yet these features predicted PTB only in white women; in both groups, L. crispatus dominance conferred protection against early delivery (92). Hyman et al (93) reported greater bacterial diversity among white women who delivered preterm, while overall species diversity was higher in African-American women. A more recent investigation comparing black and non-black American women confirmed that Lactobacillus depletion was more prevalent in black women but increased PTB risk only in white women; it also identified distinct taxa markedly associated with spontaneous PTB, with stronger effects in the black cohort (94). The protective role of L. crispatus has also been validated in European, Middle Eastern and Asian populations, where L. iners dominance associated with higher PTB risk and L. crispatus dominance with term delivery (87). Sun et al (95) showed in stratified analyses that African-American women have increased abundance of Lactobacillus iners and decreased abundance of Lactobacillus crispatus and that lower levels of L. crispatus are associated with increased risk of spontaneous PTB. Another study of Asian and Myanmar/Karen minority women combined microbiome profiling with cytokine analysis and found that signatures composed of specific taxa and inflammatory cytokines predict PTB more accurately than single markers, thus providing a framework for biomarker development in resource limited settings (96). Fettweis et al (35) employed multi omics approaches to identify a marked reduction in L. crispatus and increases in pathogenic taxa such as BVAB1 and Sneathia amnii alongside elevated levels of pro inflammatory cytokines in cases of PTB, highlighting these features as candidate biomarkers across populations. These findings indicate that intervention strategies should prioritize probiotic strains that enhance or maintain protective Lactobacilli colonization in specific populations and that biomarker discovery should integrate microbial and host inflammatory multi omics signatures to achieve precise cross population applicability.

Mechanistic pathways linking vaginal microbial dysbiosis to PTB

Inflammatory pathways mediated by vaginal microbial dysbiosis

Elevated diversity within vaginal microbial communities and the presence of bacterial vaginosis-associated taxa, including Gardnerella vaginalis, Atopobium vaginae and representatives of the Veillonellaceae family, demonstrate a robust association with heightened risk of PTB prior to 34 gestational weeks (7). A study (97) indicates that one pathway by which a diverse VMB elevates PTB risk is through premature activation of parturition-associated inflammatory cascades. In one early clinical investigation, women undergoing cervical cerclage with braided sutures exhibited persistent VMB dysbiosis and local inflammation, accompanied by ultrasound evidence of premature cervical remodeling (97). A UK multicenter retrospective analysis further showed that braided-suture cerclage was associated with a threefold increase in intra-amniotic fetal death and a twofold rise in PTB, whereas monofilament cerclage had no significant impact on the VMB, cytokine concentrations or cervical anatomy. These findings suggest that microbiota-triggered inflammation may act directly on the cervix as pregnancy advances (14). Activation of TLRs is regarded as a critical initiating event for inflammation in both term and preterm labor (PTL). TLRs are evolutionarily conserved pattern recognition receptors that orchestrate innate immune responses by sensing pathogen associated molecular patterns (PAMPs) and damage associated molecular patterns (DAMPs), then triggering downstream signaling to induce cytokine and chemokine release (as illustrated in Fig. 3) (98). Among TLR family members, the functional role of TLR4 is best characterized. TLR4 binds lipopolysaccharide (LPS) and other PAMP/DAMP ligands and its activation state in the uterus correlates with both term and PTL. In mice, TLR4 knockout delays parturition and markedly reduces neonatal survival (99). Pharmacologic blockade of TLR4 signaling with the antagonist (+)-naloxone inhibits Escherichia coli induced inflammatory cascades and prevents PTB (100). More recently, decidual endothelial cell specific TLR4 expression has been shown to be essential: Mice lacking endothelial TLR4 resist LPS-induced PTB, indicating a non-immune-cell locus of the action of TLR4 (101). Clinically, maternal TLR4 single-nucleotide polymorphisms are markedly associated with extreme preterm delivery (<32 weeks) (102). TLR4 and its co-receptor CD14 signal via both MyD88-dependent and TRIF-dependent pathways to regulate distinct inflammatory gene programs. MyD88-deficient mice are completely protected against E. coli induced PTB, underscoring the dominant role of the MyD88 axis in preterm parturition (103). Other TLRs, such as TLR2, also contribute to labor timing (104,105), and fetal TLR4/TLR2 polymorphisms further modulate PTB risk (106,107).

Vaginal microbiota-mediated
inflammatory and immune pathways in preterm birth. Pathogen- and
damage-associated molecular patterns (PAMPs and DAMPs) activate
TLRs, which signal via MyD88- and TRIF-dependent pathways to
assemble inflammasomes and release proinflammatory cytokines. TLR
activation also recruits macrophages, whose NF-κB signaling
upregulates both proinflammatory mediators and
contraction-associated genes. Additionally, NF-κB-driven IL-6/STAT3
activation in perivascular stromal cells induces IL-10 production,
maintaining immune homeostasis under inflammatory stress. Elevated
L. iners abundance has been associated with increased
vaginal IgM, C3b, C5/C5a and IL-6; C5a promotes macrophage MMP-9
secretion, driving cervical remodeling. L. iners and G.
vaginalis have both been linked to higher neutrophil counts in
the vagina, which contribute to cervical shortening during labor
and elevate preterm birth risk. PAMPs, pathogen associated
molecular patterns; DAMPs, damage associated molecular patterns;
TLRs, Toll-like receptors; LPS, lipopolysaccharide.

Figure 3

Vaginal microbiota-mediated inflammatory and immune pathways in preterm birth. Pathogen- and damage-associated molecular patterns (PAMPs and DAMPs) activate TLRs, which signal via MyD88- and TRIF-dependent pathways to assemble inflammasomes and release proinflammatory cytokines. TLR activation also recruits macrophages, whose NF-κB signaling upregulates both proinflammatory mediators and contraction-associated genes. Additionally, NF-κB-driven IL-6/STAT3 activation in perivascular stromal cells induces IL-10 production, maintaining immune homeostasis under inflammatory stress. Elevated L. iners abundance has been associated with increased vaginal IgM, C3b, C5/C5a and IL-6; C5a promotes macrophage MMP-9 secretion, driving cervical remodeling. L. iners and G. vaginalis have both been linked to higher neutrophil counts in the vagina, which contribute to cervical shortening during labor and elevate preterm birth risk. PAMPs, pathogen associated molecular patterns; DAMPs, damage associated molecular patterns; TLRs, Toll-like receptors; LPS, lipopolysaccharide.

TLR activation can subsequently drive inflammasome assembly, sustaining secretion of IL-8, CCL2, IL-1β and IL-6 by placental and decidual tissues (108). These mediators recruit immune cells and induce prostaglandin and MMP production, collectively promoting cervical ripening and uterine contractility. Amniotic cavity macrophage infiltration constitutes a critical pathophysiological mechanism for spontaneous labor initiation, functionally mediated through NF-κB-driven transcriptional activation of uterine activation-associated gene networks (such as PTGFR, GJA1, OXTR and PTGS2) and pro-inflammatory cytokines TNFα, IL-1β, IL-6 and IL-8 (109). Thus, pattern-recognition receptors and downstream effectors orchestrate both physiological and pathological labor initiation.

Immune modulation by the vaginal microbiota in PTB pathogenesis

Various approaches are used to induce PTL and parturition in mice, including immunological, hormonal and environmental triggers. Immunomodulators activate or amplify physiological inflammatory pathways that drive labor onset, recapitulating clinical features of infection and inflammation-associated parturition. The most common murine PTL models involve intrauterine or intraperitoneal administration of LPS or heat-killed whole Escherichia coli cells (110). Although LPS is widely employed to induce PTB via pro-inflammatory responses in mice (72), its role in the human female reproductive tract remains unclear. Clinical studies (35,96,111) report elevated concentrations of pro-inflammatory cytokines and chemokines, such as IL-1β, IL-6, IL-8, IL-10, IL-18, granulocyte-macrophage colony-stimulating factor (GM-CSF), macrophage inflammatory protein-1β (MIP-1β) and eosinophil chemotactic factor, in the amniotic fluid, cervicovaginal fluid and plasma of women who subsequently experienced spontaneous PTB. Moreover, cervical levels of IL-1β, IL-6, IL-8, IL-10, eotaxin, MIP-1β and GM-CSF are associated with VMB composition (112). Animal models demonstrate a central role for macrophages in both term and PTL. Macrophage depletion via anti-F4/80 antibody markedly reduces susceptibility to LPS-induced PTB in mice (113). Mechanistically, macrophages contribute to PTB pathogenesis by secreting TNF-α, IL-1, IL-6, IL-8 and by regulating expression of contraction-associated genes such as MMPs (109). Specifically, IL-1 is critical in inflammatory PTB; exogenous IL-1 induces PTL in mice, whereas blockade of the IL-1 receptor inhibits labor initiation (114), probably through activation of the NF-κB signaling pathway (115). Research indicates that IL-6 deficient mice exhibit delays parturition and resistance to LPS-induced PTB (116). Dominance of L. iners is associated with elevated levels of immunoglobulin M, complement components C3b, C5, C5a, and IL-6, collectively leading to cervical shortening, reduced endometrial receptivity, decreased implantation rates and increased PTB risk (117,118). Complement activation also plays a pivotal role: Clinical data show raised terminal complement fragments C3a, C4a and C5a in women undergoing infection-associated spontaneous PTB (113). In mouse models, C5a receptor (C5aR)-deficient animals are protected from both LPS- and RU486-induced PTB; this protection involves aberrant complement deposition in the cervical epithelium and C5a-dependent macrophage MMP-9 release that drives cervical remodeling (119). IL-10 has a protective effect against PTB. IL-10 knockout or neutralization markedly increases susceptibility to PTB in mice (120). IL-10 deficiency leads to downregulation of inflammatory cytokine gene expression in uterine and placental tissues, suggesting its role in tempering excessive inflammation during labor. Decidual endothelial cell TLR4 signaling at term may activate the IL-6/STAT3 axis via NF-κB, promoting perivascular stromal cell production of IL-10 (101). This mechanism probably maintains immune homeostasis under inflammatory conditions, a balance that is disrupted in PTB. Neutrophils, which contribute to cervical remodeling, may infiltrate the myometrium and facilitate cervical shortening during labor, thereby increasing PTB risk (121,122). Limited data suggest that neutrophil numbers in cervicovaginal fluid decline as gestation advances (123). Notably, women with a low-diversity VMB (CST III, dominated by L. iners) often exhibit elevated vaginal neutrophil counts during pregnancy (123). One study reported a positive association between G. vaginalis (CST IV) abundance and neutrophil levels in samples from women who subsequently delivered preterm (123).

Cezar-de-Mello et al (124) analyzed miRNA cargo in extracellular vesicles secreted by in vitro human VMB models under BV and Lactobacillus crispatus conditions, identifying upregulation of miRNAs targeting the glucocorticoid receptor in the BV environment. The glucocorticoid receptor (GR) pathway mediates canonical anti-inflammatory effects across diverse immune cell types (125). Macrophages expressing pattern recognition receptors, including TLRs, detect danger-associated molecular patterns and initiate inflammasome activation pathways that orchestrate proinflammatory cytokine secretion (126). Glucocorticoids can upregulate NLRP3 inflammasome components to enhance inflammatory responses (126) and GR, in concert with TNF-α, induces TLR2 expression to stimulate innate immunity (127). Conversely, GR can suppress inflammation by promoting macrophage clearance of neutrophils (128). Glucocorticoids also modulate B cell activation, survival, proliferation and differentiation in a dose-dependent manner, reducing B cell counts, progenitor proliferation, and IgG production (129), and they downregulate the anti-apoptotic protein Bcl-2, sensitizing B cells to glucocorticoid-induced apoptosis (125). These findings suggest that vaginal dysbiosis may trigger compensatory local activation of the GR signaling axis to suppress inflammation.

Microbial metabolites as modulators of pregnancy outcomes Lactate: A protective immunoregulatory metabolite

In vaginal epithelial-cell models, both D-lactic and L-lactic acid, alone or in combination with a spectrum of VMB-associated metabolites, induce the anti-inflammatory cytokine interleukin-1 receptor antagonist (IL-1RA) while suppressing pro-inflammatory mediators such as IL-6, IL-8, TNF-α, RANTES and macrophage inflammatory protein-3α (130,131). Organotypic models of the female reproductive-tract epithelium confirm that L-lactic acid upregulates IL-1RA and downregulates IL-8 (130). Although certain in vitro studies have reported pro-inflammatory responses to specific Lactobacillus strains (132,133), these strains are not dominant members of the VMB. In vivo experiments and cervical-epithelial models both demonstrate that lactate production by Lactobacillus spp. and the resulting acidic vaginal pH reinforce epithelial-barrier integrity, thereby inhibiting colonization by anaerobes and pathogens (134,135). Thus, dominance of protective lactobacilli is associated with reduced risk of suboptimal vaginal health. However, excessive lactobacilli proliferation can give rise to cytolytic vaginosis, a condition marked by epithelial-cell lysis, dissolution and shedding attributed to overproduction of lactic acid (136). Although cytolytic vaginosis is less prevalent than BV, its occurrence underscores the clinical importance of quantitatively assessing the VMB.

Short-chain fatty acids (SCFAs): immunological consequences in vaginal health

Beyond the presence or absence of key taxa, the VMB influences immune responses and pregnancy outcomes via production of SCFAs. While SCFAs enhance intestinal-barrier integrity (137), their effects in the vaginal milieu appear deleterious. SCFAs are organic fatty acids typically containing fewer than six carbon atoms; common examples include acetate, propionate and butyrate (138). Elevated levels of SCFAs, such as propionate, succinate, acetate and butyrate, are associated with increased concentrations of IL-1β, IL-2, IL-6, IL-8, IL-10, TNF-α, IFN-γ and RANTES (139). For instance, in early gestation, elevated acetate is associated with PTB under conditions of weakened L. crispatus dominance (139,140). High SCFA concentrations also inhibit expression of neutrophil gelatinase-associated lipocalin, an innate-antimicrobial factor, potentially facilitating overgrowth of BV-associated communities and further contributing to PTB (141). In BV settings, high concentrations of acetate (100 mM) and butyrate (20 mM) selectively activate TLR1/2/3 in cervicovaginal epithelial cells, resulting in increased TNF-α secretion and concomitant suppression of IL-6, RANTES and IP-10 production (131).

Other bioactive metabolites and their effect on parturition

Metabolomic analyses of PTB patients reveal upregulation of glycerophospholipid-metabolism-related metabolites. Glycerophospholipids are fundamental membrane constituents and precursors for bioactive lipids such as arachidonic acid (AA) and lysobisphosphatidic acid (LPA). These bioactive lipids are generated by phospholipase A2 (PLA2) and, in the case of AA, further converted by cyclooxygenase-2 (COX-2) into prostaglandins (PGs) (142,143). LPA3, a G protein-coupled receptor expressed in uterine epithelium, modulates COX-2 activity and PG levels (144). Collectively, these molecules serve as key regulators of inflammation. Increased phosphoinositol, a glycerophospholipid metabolite, depletes glycerophospholipid pools and downstream mediators (LPA and PG), potentially impairing implantation; additionally, elevated phosphoinositol promotes Ca2+ influx and uterine contractility, which may precipitate premature labor (145).

Enhanced primary bile-acid and steroid-hormone metabolism pathways have also been observed in PTB cohorts, consistent with findings by Menon et al (146) and Lizewska et al (147). Bile acids, synthesized from cholesterol in the liver, are modified by the gut microbiota and facilitate dietary-fat absorption (148). Pregnancy-related hormonal changes (such as cholestasis) can disrupt bile acid clearance, leading to elevated circulating levels that induce fetal stress (148). Steroid hormones, also derived from cholesterol, govern development, osmoregulation, metabolism and stress responses. Maturation of the fetal hypothalamic-pituitary-adrenal axis and increased fetal adrenal production of C19 steroids (dehydroepiandrosterone sulfate) and cortisol are pivotal for labor initiation, whereas placental progesterone maintains uterine quiescence during gestation (149). Perturbations in steroid synthesis, metabolism or transport may likewise trigger labor onset.

Hydrogen peroxide (H2O2) produced by H2O2-generating Lactobacillus spp., notably L. crispatus, L. jensenii, L. gasseri and L. vaginalis (150), inhibits pathogens such as G. vaginalis and Candida albicans via oxidation of proteins, essential thiol groups in enzymes, DNA and other cellular components (151). This activity diminishes pathogen adhesion to the vaginal epithelium and ascension into the uterus (150). H2O2 also modulates immune responses by suppressing pro-inflammatory cytokines, including IL-1β (152).

Probiotic interventions targeting vaginal microbial homeostasis for PTB prevention

The VMB can be modulated by various interventions, including probiotics, prebiotics, antibiotics and pH modulators (153). When treating infectious diseases related to BV, the most commonly used and fastest-acting method is antibiotic therapy. Although antibiotics are effective against infection, they also disrupt the overall microbial balance, including beneficial lactobacilli, leading to high recurrence rates (154) and increased risk of spontaneous PTB (12). As an alternative or adjunct to antibiotics, several approaches have been proposed to restore a Lactobacillus-dominated vaginal community. Oral probiotics, leveraging cross-talk between the gut and vaginal microbiomes, are considered a promising method. Probiotics are live, beneficial strains capable of colonizing the vagina and improving its microbial composition, such as Lactobacillus crispatus, L. jensenii and L. gasseri (12,155). These probiotics can be administered orally or vaginally, in formulations including capsules, suppositories or creams (156,157). Most intervention studies have focused on using Lactobacilli to alleviate BV and aerobic vaginitis, both of which are dysbiotic states associated with increased risk of spontaneous PTB. Results have been mixed: some studies report efficacy, while others find no significant effect. Probiotic studies are summarized in Table I. For example, one study (158) showed that oral capsules containing Lactobacillus rhamnosus GR-1 and L. fermentum RC-14 reduced vaginal yeast and E. coli counts, increased lactobacilli levels and restored an asymptomatic BV microbiota to a healthy Lactobacilli-dominated state. Ho et al's (159) study of group B Streptococcus (GBS)-positive pregnant women at 35-37 weeks demonstrated that daily oral GR-1 + RC-14 until delivery markedly lowered GBS positivity compared with placebo. In 40 BV-afflicted black women, GR-1 + RC-14 achieved higher cure rates than metronidazole (160). Maternal oral supplementation with Lactobacillus rhamnosus GR-1 and Lactobacillus fermentum RC-14 probiotic capsules markedly reduced vaginal microbial α diversity and species richness, while concurrently suppressing Gardnerella vaginalis colonization. The probiotic cohort exhibited enrichment of Lactobacillus crispatus, Lactobacillus iners and the gut-associated species Prevotella copri, taxa positively associated with VMB stability (157). Other studies have shown that probiotic intervention reduces expression of inflammatory factors, a known risk factor for PTB. In a double-blind trial of 159 volunteers, vaginal tablets containing Lactobacillus brevis CD2, L. salivarius subsp. salicinius and L. plantarum achieved clinical cure in nearly 80% of BV patients, with 32% restoring a normal VMB (161). Post-treatment vaginal lavage fluids showed significant decreases in IL-1β and IL-6, whereas the control group showed no change in local pro-inflammatory cytokines (161). Bayar et al (155) reported that administering LACTIN-V to high-risk pregnant women markedly reduced early PTB incidence. The protective effect of LACTIN-V may be attributed to its inhibition of pro-inflammatory responses to uropathogens. However, in a randomized, double-blind trial, Husain et al (162) demonstrated that daily oral administration of a probiotic preparation comprising Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14 from early pregnancy did not modify the vaginal microbiota of pregnant women nor prevent bacterial vaginosis. In a similar study, Gille et al observed that eight weeks of daily oral administration of L. rhamnosus GR-1 or L. reuteri RC-14, compared with placebo, yielded no significant difference in the incidence of BV as assessed by Nugent score before and after intervention (163). These divergent findings may reflect limitations in study design. In particular, the proportion of women with abnormal vaginal microbiota in Gille et al's (163) intervention arm was substantially lower than that reported in general populations, warranting validation in larger and more representative cohorts. Moreover, an unexpectedly high loss to follow up may have reduced the statistical power to detect intergroup differences. The two trials employed oral delivery, which may not ensure sufficient vaginal colonization or confer protection against spontaneous PTB. Consequently, the investigators recommend that future research prioritize the identification of probiotic strains and delivery modalities capable of beneficially modulating the vaginal microbiota during pregnancy before assessing their efficacy in preventing PTB (162). VMB transplantation (VMT) is another strategy under consideration. A recent case series reported successful engraftment of healthy vaginal communities into patients with refractory BV, with four out of five cases achieving sustained remission for up to 21 months (164). Although limited by sample size, these findings underscore the need to assess the long-term efficacy of both probiotic and VMT approaches in pregnant populations.

Table I

Effects of probiotic interventions on the female reproductive tract microbiota, bacterial vaginosis, and pregnancy outcomes.

Table I

Effects of probiotic interventions on the female reproductive tract microbiota, bacterial vaginosis, and pregnancy outcomes.

Authors, yearStudy populationIntervention protocolOutcome(Refs.)
Smith et al, 202064 healthy womenOral administration of freeze-dried capsules (containing Lactobacillus rhamnosus GR-1 and Lactobacillus fermentum RC-14), once daily for 60 days.Compared with placebo, oral probiotic therapy markedly increased vaginal Lactobacilli counts (logarithmic increase, P=0.01) within 4 weeks, while markedly decreasing yeast and E. coli counts. Vaginal health improvement was perceived by 30% in the probiotic group vs. 12% in the placebo group (P=0.17).(148)
Tuckey et al, 2005110 pregnant women (35-37 weeks), vaginal GBS-positiveTwo probiotic capsules daily at bedtime, each capsule containing 1×109 CFU live Lactobacillus rhamnosus GR-1 and Lactobacillus fermentum RC-14.Oral probiotics containing L. rhamnosus GR-1 and L. fermentum RC-14 markedly reduced vaginal and rectal GBS colonization. GBS negativity rate was 42.9% in probiotic group vs. 18.0% in placebo group.(149)
Wilks et al, 200440 women with BVTwo probiotic capsules daily at bedtime (Lactobacillus rhamnosus GR-1 and Lactobacillus fermentum RC-14).Probiotic treatment achieved markedly higher BV cure rates compared with metronidazole treatment.(150)
Lizewska et al, 2018140 BV-positive pregnant womenTopical clindamycin treatment (2% gel, 5 g, intravaginally at bedtime for 7 days), followed by randomization to oral probiotics or placebo, one capsule daily for 30 days.The probiotic group showed significant reduction in vaginal α-diversity and species richness, decreased Gardnerella vaginalis abundance, and increased abundance of L. crispatus, L. iners, and the gut-associated species Prevotella copri, indicative of a healthy vaginal microbiota.(147)
Srinivasan et al, 201561 pregnant women at risk for preterm birthLACTIN-V administration (2×109 CFU) starting at 14 weeks gestation, daily for 5 consecutive days, then once weekly for 6 weeks.The probiotic group exhibited a PTB rate of 3.3% before 34 weeks, markedly lower than the 7% rate observed in a background population of 2190 women with similar PTB risk.(145)
O'Hanlon et al, 201167 BV patientsIntravaginal probiotic tablets containing a mixture of Lactobacillus brevis CD2, L. salivarius subsp. salicinius, and L. plantarum (≥1×109 CFU/tablet), administered nightly for 8 consecutive days.Clinical cure was achieved in nearly 80% of the BV patients receiving probiotics, with 32% restoring a normal vaginal microbiota. Levels of IL-1β and IL-6 in vaginal lavage fluid markedly decreased.(151)

[i] GBS, group B Streptococcus.

Conclusions and future perspectives

Conclusions

PTB remains a major global health challenge, with 35-50% of spontaneous PTB and PPROM cases attributed to ascending vaginal infections. A growing body of evidence indicates that dynamic shifts in the VMB critically influence pregnancy outcomes. Vaginal microbiota dominated by Lactobacillus species, particularly L. crispatus, confer protective effects through three synergistic mechanisms: Lactic acid-driven pH homeostasis, bacteriocin-mediated pathogen suppression, and immunoregulatory pathways that attenuate proinflammatory cytokine signaling, including IL-6, IL-8, and TNF-α. By contrast, high-diversity, Lactobacillus-depleted (CST IV) vaginal microbiota enriched with opportunistic pathogens such as Gardnerella vaginalis, Atopobium vaginae, and Prevotella spp. demonstrate a strong association with increased PTB risk. These pathogens compromise epithelial integrity through biofilm formation, protease secretion and production of SCFAs, exacerbating local inflammation and activating TLR/NF-κB signaling cascades.

Future perspectives

The field continues to confront several persistent challenges despite recent advancements. While observational studies have reproducibly linked vaginal microbial dysbiosis to PTB risk, establishing definitive causal relationships requires longitudinal investigations capable of tracing microbial community trajectories across pregnancy trimesters. Such studies would clarify whether specific pathogen colonization patterns precede cervical remodeling events and identify predictive microbial signatures. A parallel knowledge gap exists in understanding functional differences between Lactobacillus species, particularly the clinically relevant dichotomy between L. crispatus' protective mucosal interactions and L. iners' ambiguous ecological role, which demands comparative multi-omic analyses spanning genomic, proteomic and metabolomic dimensions. Current microbiome profiling techniques have limitations; for example, 16S rRNA sequencing lacks resolution at the species level and does not capture functional information. Future studies should integrate metagenomic and multi-omics approaches to achieve a more comprehensive characterization of both microbial communities and their metabolite profiles. Current therapeutic development efforts face dual limitations: heterogeneous probiotic formulations lacking strain-specific rationale and clinical trials inadequately powered to detect ethnic-geographic variations in microbial-host interactions. Progress necessitates standardized intervention protocols paired with mechanistic studies decoding how microbial metabolites interface with cervical tissue immune networks at single-cell resolution. Addressing these interconnected biological and methodological complexities could enable microbiota-informed risk stratification systems and next-generation therapeutic strategies tailored to individual microbial ecologies.

Limitations

Although the present review was based on a comprehensive search and synthesis of PubMed and Web of Science from inception to May 2025, it still has several limitations. First, by restricting its analysis to English-language publications, it may have overlooked important studies in other languages. Second, the included studies vary in their definitions of bacterial vaginosis, sampling time points and sequencing platforms, making direct comparisons difficult. Third, as a narrative review, it did not conduct a formal risk of bias assessment or quantitative meta-analysis, so it cannot rule out the influence of publication bias. Future research would benefit from standardized protocols, broader inclusion criteria, and multicenter collaboration to validate and build upon these findings.

Availability of data and materials

Not applicable.

Authors' contributions

DC, FG and KW reviewed literature and wrote the manuscript. NL and QS collected data and prepared figures. Data authentication is not applicable. All authors reviewed and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests

Acknowledgements

Not applicable.

Funding

The present review was supported by the Science and Technology Development Program of Jinan Municipal Health Commission (grant no. 2024203001).

References

1 

Ohuma EO, Moller AB, Bradley E, Chakwera S, Hussain-Alkhateeb L, Lewin A, Okwaraji YB, Mahanani WR, Johansson EW, Lavin T, et al: National, regional, and global estimates of preterm birth in 2020, with trends from 2010: A systematic analysis. Lancet. 402:1261–1271. 2023. View Article : Google Scholar : PubMed/NCBI

2 

Saigal S and Doyle LW: An overview of mortality and sequelae of preterm birth from infancy to adulthood. Lancet. 371:261–269. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Goldenberg RL, Culhane JF, Iams JD and Romero R: Epidemiology and causes of preterm birth. Lancet. 371:75–84. 2008. View Article : Google Scholar : PubMed/NCBI

4 

Romero R, Gomez-Lopez N, Winters AD, Jung E, Shaman M, Bieda J, Panaitescu B, Pacora P, Erez O, Greenberg JM, et al: Evidence that intra-amniotic infections are often the result of an ascending invasion-a molecular microbiological study. J Perinat Med. 47:915–931. 2019. View Article : Google Scholar : PubMed/NCBI

5 

Witkin SS: The vaginal microbiome, vaginal anti-microbial defence mechanisms and the clinical challenge of reducing infection-related preterm birth. BJOG. 122:213–218. 2015. View Article : Google Scholar

6 

Fox C and Eichelberger K: Maternal microbiome and pregnancy outcomes. Fertil Steril. 104:1358–1363. 2015. View Article : Google Scholar : PubMed/NCBI

7 

Tabatabaei N, Eren AM, Barreiro LB, Yotova V, Dumaine A, Allard C and Fraser WD: Vaginal microbiome in early pregnancy and subsequent risk of spontaneous preterm birth: A case-control study. BJOG. 126:349–358. 2019. View Article : Google Scholar

8 

Liu L, Yin T, Zhang X, Sun L and Yin Y: Temporal and spatial variation of the human placental microbiota during pregnancy. Am J Reprod Immunol. 92:e700232024. View Article : Google Scholar : PubMed/NCBI

9 

Conde-Agudelo A, Papageorghiou AT, Kennedy SH and Villar J: Novel biomarkers for the prediction of the spontaneous preterm birth phenotype: A systematic review and meta-analysis. BJOG. 118:1042–1054. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Meertens LJE, van Montfort P, Scheepers HCJ, van Kuijk SMJ, Aardenburg R, Langenveld J, van Dooren IMA, Zwaan IM, Spaanderman MEA and Smits LJM: Prediction models for the risk of spontaneous preterm birth based on maternal characteristics: A systematic review and independent external validation. Acta Obstet Gynecol Scand. 97:907–920. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Hickey RJ, Zhou X, Pierson JD, Ravel J and Forney LJ: Understanding vaginal microbiome complexity from an ecological perspective. Transl Res. 160:267–282. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Happel AU, Kullin B, Gamieldien H, Wentzel N, Zauchenberger CZ, Jaspan HB, Dabee S, Barnabas SL, Jaumdally SZ, Dietrich J, et al: Exploring potential of vaginal Lactobacillus isolates from South African women for enhancing treatment for bacterial vaginosis. PLoS Pathog. 16:e10085592020. View Article : Google Scholar : PubMed/NCBI

13 

Giannella L, Grelloni C, Quintili D, Fiorelli A, Montironi R, Alia S, Delli Carpini G, Di Giuseppe J, Vignini A and Ciavattini A: Microbiome changes in pregnancy disorders. Antioxidants (Basel). 12:4632023. View Article : Google Scholar : PubMed/NCBI

14 

Bayar E, Bennett PR, Chan D, Sykes L and MacIntyre DA: The pregnancy microbiome and preterm birth. Semin Immunopathol. 42:487–499. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Bachmann NL, Rockett RJ, Timms VJ and Sintchenko V: Advances in clinical sample preparation for identification and characterization of bacterial pathogens using metagenomics. Front Public Health. 6:3632018. View Article : Google Scholar

16 

Sroka-Oleksiak A, Gosiewski T, Pabian W, Gurgul A, Kapusta P, Ludwig-Słomczyńska AH, Wołkow PP and Brzychczy-Włoch M: Next-generation sequencing as a tool to detect vaginal microbiota disturbances during pregnancy. Microorganisms. 8:18132020. View Article : Google Scholar : PubMed/NCBI

17 

Linhares IM, Summers PR, Larsen B, Giraldo PC and Witkin SS: Contemporary perspectives on vaginal pH and lactobacilli. Am J Obstet Gynecol. 204:120.e1–e5. 2011. View Article : Google Scholar

18 

Ravel J, Gajer P, Abdo Z, Schneider GM, Koenig SS, McCulle SL, Karlebach S, Gorle R, Russell J, Tacket CO, et al: Vaginal microbiome of reproductive-age women. Proc Natl Acad Sci USA. 108(Suppl 1): S4680–S4687. 2011. View Article : Google Scholar

19 

Blostein F, Gelaye B, Sanchez SE, Williams MA and Foxman B: Vaginal microbiome diversity and preterm birth: Results of a nested case-control study in Peru. Ann Epidemiol. 41:28–34. 2020. View Article : Google Scholar

20 

Stennett CA, Dyer TV, He X, Robinson CK, Ravel J, Ghanem KG and Brotman RM: A cross-sectional pilot study of birth mode and vaginal microbiota in reproductive-age women. PLoS One. 15:e02285742020. View Article : Google Scholar : PubMed/NCBI

21 

Chaban B, Links MG, Jayaprakash TP, Wagner EC, Bourque DK, Lohn Z, Albert AY, van Schalkwyk J, Reid G, Hemmingsen SM, et al: Characterization of the vaginal microbiota of healthy Canadian women through the menstrual cycle. Microbiome. 2:232014. View Article : Google Scholar : PubMed/NCBI

22 

Roy EJ and Mackay R: The concentration of oestrogens in blood during pregnancy. J Obstet Gynaecol Br Emp. 69:13–17. 1962. View Article : Google Scholar : PubMed/NCBI

23 

Ma B, Forney LJ and Ravel J: Vaginal microbiome: Rethinking health and disease. Annu Rev Microbiol. 66:371–389. 2012. View Article : Google Scholar : PubMed/NCBI

24 

van de Wijgert J and Verwijs MC: Lactobacilli-containing vaginal probiotics to cure or prevent bacterial or fungal vaginal dysbiosis: A systematic review and recommendations for future trial designs. BJOG. 127:287–299. 2020. View Article : Google Scholar

25 

Bhandari P, Tingley JP, Palmer DRJ, Abbott DW and Hill JE: Characterization of an α-Glucosidase enzyme conserved in gardnerella spp. isolated from the human vaginal microbiome. J Bacteriol. 203:e00213212021. View Article : Google Scholar

26 

Rosca AS, Castro J, Sousa LGV and Cerca N: Gardnerella and vaginal health: The truth is out there. FEMS Microbiol Rev. 44:73–105. 2020. View Article : Google Scholar

27 

Tachedjian G, Aldunate M, Bradshaw CS and Cone RA: The role of lactic acid production by probiotic Lactobacillus species in vaginal health. Res Microbiol. 168:782–792. 2017. View Article : Google Scholar : PubMed/NCBI

28 

Kaur H, Merchant M, Haque MM and Mande SS: Crosstalk between female gonadal hormones and vaginal microbiota across various phases of women's gynecological lifecycle. Front Microbiol. 11:5512020. View Article : Google Scholar : PubMed/NCBI

29 

Anderson DJ, Marathe J and Pudney J: The structure of the human vaginal stratum corneum and its role in immune defense. Am J Reprod Immunol. 71:618–623. 2014. View Article : Google Scholar : PubMed/NCBI

30 

Shen J, Song N, Williams CJ, Brown CJ, Yan Z, Xu C and Forney LJ: Effects of low dose estrogen therapy on the vaginal microbiomes of women with atrophic vaginitis. Sci Rep. 6:243802016. View Article : Google Scholar : PubMed/NCBI

31 

Cauci S, Driussi S, De Santo D, Penacchioni P, Iannicelli T, Lanzafame P, De Seta F, Quadrifoglio F, de Aloysio D and Guaschino S: Prevalence of bacterial vaginosis and vaginal flora changes in peri- and postmenopausal women. J Clin Microbiol. 40:2147–2152. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Brooks JP, Edwards DJ, Blithe DL, Fettweis JM, Serrano MG, Sheth NU, Strauss JF III, Buck GA and Jefferson KK: Effects of combined oral contraceptives, depot medroxyprogesterone acetate and the levonorgestrel-releasing intrauterine system on the vaginal microbiome. Contraception. 95:405–413. 2017. View Article : Google Scholar

33 

Bradshaw CS, Vodstrcil LA, Hocking JS, Law M, Pirotta M, Garland SM, De Guingand D, Morton AN and Fairley CK: Recurrence of bacterial vaginosis is significantly associated with posttreatment sexual activities and hormonal contraceptive use. Clin Infect Dis. 56:777–786. 2013. View Article : Google Scholar

34 

Garcia-Garcia RM, Arias-Álvarez M, Jordán-Rodríguez D, Rebollar PG, Lorenzo PL, Herranz C and Rodríguez JM: Female reproduction and the microbiota in mammals: Where are we? Theriogenology. 194:144–153. 2022. View Article : Google Scholar : PubMed/NCBI

35 

Fettweis JM, Serrano MG, Brooks JP, Edwards DJ, Girerd PH, Parikh HI, Huang B, Arodz TJ, Edupuganti L, Glascock AL, et al: The vaginal microbiome and preterm birth. Nat Med. 25:1012–1021. 2019. View Article : Google Scholar : PubMed/NCBI

36 

Serrano MG, Parikh HI, Brooks JP, Edwards DJ, Arodz TJ, Edupuganti L, Huang B, Girerd PH, Bokhari YA, Bradley SP, et al: Racioethnic diversity in the dynamics of the vaginal microbiome during pregnancy. Nat Med. 25:1001–1011. 2019. View Article : Google Scholar : PubMed/NCBI

37 

MacIntyre DA, Chandiramani M, Lee YS, Kindinger L, Smith A, Angelopoulos N, Lehne B, Arulkumaran S, Brown R, Teoh TG, et al: The vaginal microbiome during pregnancy and the postpartum period in a European population. Sci Rep. 5:89882015. View Article : Google Scholar : PubMed/NCBI

38 

Zhang X, Zhai Q, Wang J, Ma X, Xing B, Fan H, Gao Z, Zhao F and Liu W: Variation of the vaginal microbiome during and after pregnancy in Chinese women. Genomics Proteomics Bioinformatics. 20:322–333. 2022. View Article : Google Scholar : PubMed/NCBI

39 

Shachar BZ, Mayo JA, Lyell DJ, Baer RJ, Jeliffe-Pawlowski LL, Stevenson DK and Shaw GM: Interpregnancy interval after live birth or pregnancy termination and estimated risk of preterm birth: A retrospective cohort study. BJOG. 123:2009–2017. 2016. View Article : Google Scholar : PubMed/NCBI

40 

McKinney D, House M, Chen A, Muglia L and DeFranco E: The influence of interpregnancy interval on infant mortality. Am J Obstet Gynecol. 216:316.e1–316.e9. 2017. View Article : Google Scholar

41 

Kangatharan C, Labram S and Bhattacharya S: Interpregnancy interval following miscarriage and adverse pregnancy outcomes: Systematic review and meta-analysis. Hum Reprod Update. 23:221–231. 2017.

42 

Bobitt JR and Ledger WJ: Unrecognized amnionitis and prematurity: A preliminary report. J Reprod Med. 19:8–12. 1977.PubMed/NCBI

43 

Andrews WW, Hauth JC, Goldenberg RL, Gomez R, Romero R and Cassell GH: Amniotic fluid interleukin-6: Correlation with upper genital tract microbial colonization and gestational age in women delivered after spontaneous labor versus indicated delivery. Am J Obstet Gynecol. 173:606–612. 1995. View Article : Google Scholar : PubMed/NCBI

44 

Andrews WW, Goldenberg RL, Hauth JC, Cliver SP, Conner M and Goepfert AR: Endometrial microbial colonization and plasma cell endometritis after spontaneous or indicated preterm versus term delivery. Am J Obstet Gynecol. 193:739–745. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Hillier SL, Krohn MA, Cassen E, Easterling TR, Rabe LK and Eschenbach DA: The role of bacterial vaginosis and vaginal bacteria in amniotic fluid infection in women in preterm labor with intact fetal membranes. Clin Infect Dis. 20(Suppl 2): S276–S278. 1995. View Article : Google Scholar : PubMed/NCBI

46 

Leitich H and Kiss H: Asymptomatic bacterial vaginosis and intermediate flora as risk factors for adverse pregnancy outcome. Best Pract Res Clin Obstet Gynaecol. 21:375–390. 2007. View Article : Google Scholar : PubMed/NCBI

47 

Hay PE, Lamont RF, Taylor-Robinson D, Morgan DJ, Ison C and Pearson J: Abnormal bacterial colonisation of the genital tract and subsequent preterm delivery and late miscarriage. BMJ. 308:295–298. 1994. View Article : Google Scholar : PubMed/NCBI

48 

Leitich H, Bodner-Adler B, Brunbauer M, Kaider A, Egarter C and Husslein P: Bacterial vaginosis as a risk factor for preterm delivery: A meta-analysis. Am J Obstet Gynecol. 189:139–147. 2003. View Article : Google Scholar : PubMed/NCBI

49 

Carey JC, Klebanoff MA, Hauth JC, Hillier SL, Thom EA, Ernest JM, Heine RP, Nugent RP, Fischer ML, Leveno KJ, et al: Metronidazole to prevent preterm delivery in pregnant women with asymptomatic bacterial vaginosis. National institute of child health and human development network of maternal-fetal medicine units. N Engl J Med. 342:534–540. 2000. View Article : Google Scholar : PubMed/NCBI

50 

Klebanoff MA, Carey JC, Hauth JC, Hillier SL, Nugent RP, Thom EA, Ernest JM, Heine RP, Wapner RJ, Trout W, et al: Failure of metronidazole to prevent preterm delivery among pregnant women with asymptomatic Trichomonas vaginalis infection. N Engl J Med. 345:487–493. 2001. View Article : Google Scholar : PubMed/NCBI

51 

Brocklehurst P, Hannah M and McDonald H: Interventions for treating bacterial vaginosis in pregnancy. Cochrane Database Syst Rev. Jan 20–2000.Epub ahead of print. PubMed/NCBI

52 

Guise JM, Mahon SM, Aickin M, Helfand M, Peipert JF and Westhoff C: Screening for bacterial vaginosis in pregnancy. Am J Prev Med. 20(Suppl 3): S62–S72. 2001. View Article : Google Scholar

53 

Koumans EH, Markowitz LE and Hogan V; CDC BV Working Group: Indications for therapy and treatment recommendations for bacterial vaginosis in nonpregnant and pregnant women: A synthesis of data. Clin Infect Dis. 35(Suppl 2): S152–S172. 2002. View Article : Google Scholar : PubMed/NCBI

54 

Petrova MI, van den Broek M, Balzarini J, Vanderleyden J and Lebeer S: Vaginal microbiota and its role in HIV transmission and infection. FEMS Microbiol Rev. 37:762–792. 2013. View Article : Google Scholar : PubMed/NCBI

55 

Peebles K, Velloza J, Balkus JE, McClelland RS and Barnabas RV: High global burden and costs of bacterial vaginosis: A systematic review and meta-analysis. Sex Transm Dis. 46:304–311. 2019. View Article : Google Scholar : PubMed/NCBI

56 

Onderdonk AB, Delaney ML and Fichorova RN: The human microbiome during bacterial vaginosis. Clin Microbiol Rev. 29:223–238. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Ling Z, Kong J, Liu F, Zhu H, Chen X, Wang Y, Li L, Nelson KE, Xia Y and Xiang C: Molecular analysis of the diversity of vaginal microbiota associated with bacterial vaginosis. BMC Genomics. 11:4882010. View Article : Google Scholar : PubMed/NCBI

58 

Muzny CA and Schwebke JR: Gardnerella vaginalis: Still a prime suspect in the pathogenesis of bacterial vaginosis. Curr Infect Dis Rep. 15:130–135. 2013. View Article : Google Scholar : PubMed/NCBI

59 

Garcia EM, Serrano MG, Edupuganti L, Edwards DJ, Buck GA and Jefferson KK: Sequence comparison of vaginolysin from different gardnerella species. Pathogens. 10:862021. View Article : Google Scholar : PubMed/NCBI

60 

Vaneechoutte M, Guschin A, Van Simaey L, Gansemans Y, Van Nieuwerburgh F and Cools P: Emended description of Gardnerella vaginalis and description of Gardnerella leopoldii sp. nov., Gardnerella piotii sp. nov. and Gardnerella swidsinskii sp. nov., with delineation of 13 genomic species within the genus Gardnerella. Int J Syst Evol Microbiol. 69:679–687. 2019. View Article : Google Scholar : PubMed/NCBI

61 

Ahmed A, Earl J, Retchless A, Hillier SL, Rabe LK, Cherpes TL, Powell E, Janto B, Eutsey R, Hiller NL, et al: Comparative genomic analyses of 17 clinical isolates of Gardnerella vaginalis provide evidence of multiple genetically isolated clades consistent with subspeciation into genovars. J Bacteriol. 194:3922–3937. 2012. View Article : Google Scholar : PubMed/NCBI

62 

Balashov SV, Mordechai E, Adelson ME and Gygax SE: Identification, quantification and subtyping of Gardnerella vaginalis in noncultured clinical vaginal samples by quantitative PCR. J Med Microbiol. 63:162–175. 2014. View Article : Google Scholar

63 

Georgijević A, Cjukić-Ivancević S and Bujko M: Bacterial vaginosis. Epidemiology and risk factors. Srp Arh Celok Lek. 128:29–33. 2000.In Serbian.

64 

van de Wijgert JH, Borgdorff H, Verhelst R, Crucitti T, Francis S, Verstraelen H and Jespers V: The vaginal microbiota: what have we learned after a decade of molecular characterization? PLoS One. 9:e1059982014. View Article : Google Scholar : PubMed/NCBI

65 

France MT, Fu L, Rutt L, Yang H, Humphrys MS, Narina S, Gajer PM, Ma B, Forney LJ and Ravel J: Insight into the ecology of vaginal bacteria through integrative analyses of metagenomic and metatranscriptomic data. Genome Biol. 23:662022. View Article : Google Scholar : PubMed/NCBI

66 

Garcia EM, Kraskauskiene V, Koblinski JE and Jefferson KK: Interaction of gardnerella vaginalis and vaginolysin with the apical versus basolateral face of a three-dimensional model of vaginal epithelium. Infect Immun. 87:e00646–18. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Ragaliauskas T, Plečkaitytė M, Jankunec M, Labanauskas L, Baranauskiene L and Valincius G: Inerolysin and vaginolysin, the cytolysins implicated in vaginal dysbiosis, differently impair molecular integrity of phospholipid membranes. Sci Rep. 9:106062019. View Article : Google Scholar : PubMed/NCBI

68 

Jung HS, Ehlers MM, Lombaard H, Redelinghuys MJ and Kock MM: Etiology of bacterial vaginosis and polymicrobial biofilm formation. Crit Rev Microbiol. 43:651–667. 2017. View Article : Google Scholar : PubMed/NCBI

69 

Swidsinski A, Dörffel Y, Loening-Baucke V, Schilling J and Mendling W: Response of Gardnerella vaginalis biofilm to 5 days of moxifloxacin treatment. FEMS Immunol Med Microbiol. 61:41–46. 2011. View Article : Google Scholar

70 

Castro J, Machado D and Cerca N: Unveiling the role of Gardnerella vaginalis in polymicrobial Bacterial Vaginosis biofilms: The impact of other vaginal pathogens living as neighbors. ISME J. 13:1306–1317. 2019. View Article : Google Scholar : PubMed/NCBI

71 

Jones HE, Harris KA, Azizia M, Bank L, Carpenter B, Hartley JC, Klein N and Peebles D: Differing prevalence and diversity of bacterial species in fetal membranes from very preterm and term labor. PLoS One. 4:e82052009. View Article : Google Scholar : PubMed/NCBI

72 

Suff N, Karda R, Diaz JA, Ng J, Baruteau J, Perocheau D, Tangney M, Taylor PW, Peebles D, Buckley SMK and Waddington SN: Ascending vaginal infection using bioluminescent bacteria evokes intrauterine inflammation, preterm birth, and neonatal brain injury in pregnant mice. Am J Pathol. 188:2164–2176. 2018. View Article : Google Scholar : PubMed/NCBI

73 

Randis TM, Gelber SE, Hooven TA, Abellar RG, Akabas LH, Lewis EL, Walker LB, Byland LM, Nizet V and Ratner AJ: Group B Streptococcus β-hemolysin/cytolysin breaches maternal-fetal barriers to cause preterm birth and intrauterine fetal demise in vivo. J Infect Dis. 210:265–273. 2014. View Article : Google Scholar : PubMed/NCBI

74 

Romero R, Hassan SS, Gajer P, Tarca AL, Fadrosh DW, Bieda J, Chaemsaithong P, Miranda J, Chaiworapongsa T and Ravel J: The vaginal microbiota of pregnant women who subsequently have spontaneous preterm labor and delivery and those with a normal delivery at term. Microbiome. 2:182014. View Article : Google Scholar : PubMed/NCBI

75 

Nelson DB, Shin H, Wu J and Dominguez-Bello MG: The gestational vaginal microbiome and spontaneous preterm birth among Nulliparous African American Women. Am J Perinatol. 33:887–893. 2016. View Article : Google Scholar : PubMed/NCBI

76 

Stout MJ, Zhou Y, Wylie KM, Tarr PI, Macones GA and Tuuli MG: Early pregnancy vaginal microbiome trends and preterm birth. Am J Obstet Gynecol. 217:356.e1–356.e18. 2017. View Article : Google Scholar

77 

Kindinger LM, Bennett PR, Lee YS, Marchesi JR, Smith A, Cacciatore S, Holmes E, Nicholson JK, Teoh TG and MacIntyre DA: The interaction between vaginal microbiota, cervical length, and vaginal progesterone treatment for preterm birth risk. Microbiome. 5:62017. View Article : Google Scholar : PubMed/NCBI

78 

Petrova MI, Reid G, Vaneechoutte M and Lebeer S: Lactobacillus iners: Friend or Foe? Trends Microbiol. 25:182–191. 2017. View Article : Google Scholar

79 

Srinivasan S, Hoffman NG, Morgan MT, Matsen FA, Fiedler TL, Hall RW, Ross FJ, McCoy CO, Bumgarner R, Marrazzo JM and Fredricks DN: Bacterial communities in women with bacterial vaginosis: High resolution phylogenetic analyses reveal relationships of microbiota to clinical criteria. PLoS One. 7:e378182012. View Article : Google Scholar : PubMed/NCBI

80 

Santiago GL, Cools P, Verstraelen H, Trog M, Missine G, El Aila N, Verhelst R, Tency I, Claeys G, Temmerman M and Vaneechoutte M: Longitudinal study of the dynamics of vaginal microflora during two consecutive menstrual cycles. PLoS One. 6:e281802011. View Article : Google Scholar : PubMed/NCBI

81 

Tamrakar R, Yamada T, Furuta I, Cho K, Morikawa M, Yamada H, Sakuragi N and Minakami H: Association between Lactobacillus species and bacterial vaginosis-related bacteria, and bacterial vaginosis scores in pregnant Japanese women. BMC Infect Dis. 7:1282007. View Article : Google Scholar : PubMed/NCBI

82 

Shipitsyna E, Roos A, Datcu R, Hallén A, Fredlund H, Jensen JS, Engstrand L and Unemo M: Composition of the vaginal microbiota in women of reproductive age-sensitive and specific molecular diagnosis of bacterial vaginosis is possible? PLoS One. 8:e606702013. View Article : Google Scholar

83 

Verstraelen H, Verhelst R, Claeys G, De Backer E, Temmerman M and Vaneechoutte M: Longitudinal analysis of the vaginal microflora in pregnancy suggests that L. crispatus promotes the stability of the normal vaginal microflora and that L. gasseri and/or L. iners are more conducive to the occurrence of abnormal vaginal microflora. BMC Microbiol. 9:1162009. View Article : Google Scholar : PubMed/NCBI

84 

Santiago GL, Tency I, Verstraelen H, Verhelst R, Trog M, Temmerman M, Vancoillie L, Decat E, Cools P and Vaneechoutte M: Longitudinal qPCR study of the dynamics of L. crispatus, L. iners, A. vaginae, (sialidase positive) G. vaginalis, and P. bivia in the vagina. PLoS One. 7:e452812012. View Article : Google Scholar : PubMed/NCBI

85 

Macklaim JM, Fernandes AD, Di Bella JM, Hammond JA, Reid G and Gloor GB: Comparative meta-RNA-seq of the vaginal microbiota and differential expression by Lactobacillus iners in health and dysbiosis. Microbiome. 1:122013. View Article : Google Scholar

86 

Gajer P, Brotman RM, Bai G, Sakamoto J, Schütte UM, Zhong X, Koenig SS, Fu L, Ma ZS, Zhou X, et al: Temporal dynamics of the human vaginal microbiota. Sci Transl Med. 4:132ra522012. View Article : Google Scholar : PubMed/NCBI

87 

Petricevic L, Domig KJ, Nierscher FJ, Sandhofer MJ, Fidesser M, Krondorfer I, Husslein P, Kneifel W and Kiss H: Characterisation of the vaginal Lactobacillus microbiota associated with preterm delivery. Sci Rep. 4:51362014. View Article : Google Scholar : PubMed/NCBI

88 

Baldwin EA, Walther-Antonio M, MacLean AM, Gohl DM, Beckman KB, Chen J, White B, Creedon DJ and Chia N: Persistent microbial dysbiosis in preterm premature rupture of membranes from onset until delivery. PeerJ. 3:e13982015. View Article : Google Scholar : PubMed/NCBI

89 

Brown RG, Marchesi JR, Lee YS, Smith A, Lehne B, Kindinger LM, Terzidou V, Holmes E, Nicholson JK, Bennett PR and MacIntyre DA: Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med. 16:92018. View Article : Google Scholar : PubMed/NCBI

90 

Brown RG, Al-Memar M, Marchesi JR, Lee YS, Smith A, Chan D, Lewis H, Kindinger L, Terzidou V, Bourne T, et al: Establishment of vaginal microbiota composition in early pregnancy and its association with subsequent preterm prelabor rupture of the fetal membranes. Transl Res. 207:30–43. 2019. View Article : Google Scholar : PubMed/NCBI

91 

DiGiulio DB, Callahan BJ, McMurdie PJ, Costello EK, Lyell DJ, Robaczewska A, Sun CL, Goltsman DS, Wong RJ, Shaw G, et al: Temporal and spatial variation of the human microbiota during pregnancy. Proc Natl Acad Sci USA. 112:11060–11065. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Callahan BJ, DiGiulio DB, Goltsman DSA, Sun CL, Costello EK, Jeganathan P, Biggio JR, Wong RJ, Druzin ML, Shaw GM, et al: Replication and refinement of a vaginal microbial signature of preterm birth in two racially distinct cohorts of US women. Proc Natl Acad Sci USA. 114:9966–9971. 2017. View Article : Google Scholar : PubMed/NCBI

93 

Hyman RW, Fukushima M, Jiang H, Fung E, Rand L, Johnson B, Vo KC, Caughey AB, Hilton JF, Davis RW and Giudice LC: Diversity of the vaginal microbiome correlates with preterm birth. Reprod Sci. 21:32–40. 2014. View Article : Google Scholar :

94 

Elovitz MA, Gajer P, Riis V, Brown AG, Humphrys MS, Holm JB and Ravel J: Cervicovaginal microbiota and local immune response modulate the risk of spontaneous preterm delivery. Nat Commun. 10:13052019. View Article : Google Scholar : PubMed/NCBI

95 

Sun S, Serrano MG, Fettweis JM, Basta P, Rosen E, Ludwig K, Sorgen AA, Blakley IC, Wu MC, Dole N, et al: Race, the vaginal microbiome, and spontaneous preterm birth. mSystems. 7:e00017222022. View Article : Google Scholar : PubMed/NCBI

96 

Kumar M, Murugesan S, Singh P, Saadaoui M, Elhag DA, Terranegra A, Kabeer BSA, Marr AK, Kino T, Brummaier T, et al: Vaginal microbiota and cytokine levels predict preterm delivery in Asian women. Front Cell Infect Microbiol. 11:6396652021. View Article : Google Scholar : PubMed/NCBI

97 

Kindinger LM, MacIntyre DA, Lee YS, Marchesi JR, Smith A, McDonald JA, Terzidou V, Cook JR, Lees C, Israfil-Bayli F, et al: Relationship between vaginal microbial dysbiosis, inflammation, and pregnancy outcomes in cervical cerclage. Sci Transl Med. 8:350ra1022016. View Article : Google Scholar : PubMed/NCBI

98 

Green ES and Arck PC: Pathogenesis of preterm birth: Bidirectional inflammation in mother and fetus. Semin Immunopathol. 42:413–429. 2020. View Article : Google Scholar : PubMed/NCBI

99 

Wahid HH, Dorian CL, Chin PY, Hutchinson MR, Rice KC, Olson DM, Moldenhauer LM and Robertson SA: Toll-Like receptor 4 is an essential upstream regulator of on-time parturition and perinatal viability in mice. Endocrinology. 156:3828–3841. 2015. View Article : Google Scholar : PubMed/NCBI

100 

Chin PY, Dorian CL, Hutchinson MR, Olson DM, Rice KC, Moldenhauer LM and Robertson SA: Novel Toll-like receptor-4 antagonist (+)-naloxone protects mice from inflammation-induced preterm birth. Sci Rep. 6:361122016. View Article : Google Scholar : PubMed/NCBI

101 

Deng W, Yuan J, Cha J, Sun X, Bartos A, Yagita H, Hirota Y and Dey SK: Endothelial cells in the decidual bed are potential therapeutic targets for preterm birth prevention. Cell Rep. 27:1755–1768.e4. 2019. View Article : Google Scholar : PubMed/NCBI

102 

Liassides C, Papadopoulos A, Siristatidis C, Damoraki G, Liassidou A, Chrelias C, Kassanos D and Giamarellos-Bourboulis EJ: Single nucleotide polymorphisms of Toll-like receptor-4 and of autophagy-related gene 16 like-1 gene for predisposition of premature delivery: A prospective study. Medicine (Baltimore). 98:e173132019. View Article : Google Scholar : PubMed/NCBI

103 

Filipovich Y, Lu SJ, Akira S and Hirsch E: The adaptor protein MyD88 is essential for E coli-induced preterm delivery in mice. Am J Obstet Gynecol. 200:93.e1–8. 2009. View Article : Google Scholar : PubMed/NCBI

104 

Montalbano AP, Hawgood S and Mendelson CR: Mice deficient in surfactant protein A (SP-A) and SP-D or in TLR2 manifest delayed parturition and decreased expression of inflammatory and contractile genes. Endocrinology. 154:483–498. 2013. View Article : Google Scholar :

105 

Patni S, Wynen LP, Seager AL, Morgan G, White JO and Thornton CA: Expression and activity of Toll-like receptors 1-9 in the human term placenta and changes associated with labor at term. Biol Reprod. 80:243–248. 2009. View Article : Google Scholar

106 

Krediet TG, Wiertsema SP, Vossers MJ, Hoeks SB, Fleer A, Ruven HJ and Rijkers GT: Toll-like receptor 2 polymorphism is associated with preterm birth. Pediatr Res. 62:474–476. 2007. View Article : Google Scholar : PubMed/NCBI

107 

Lorenz E, Hallman M, Marttila R, Haataja R and Schwartz DA: Association between the Asp299Gly polymorphisms in the Toll-like receptor 4 and premature births in the Finnish population. Pediatr Res. 52:373–376. 2002. View Article : Google Scholar : PubMed/NCBI

108 

Romero R, Xu Y, Plazyo O, Chaemsaithong P, Chaiworapongsa T, Unkel R, Than NG, Chiang PJ, Dong Z, Xu Z, et al: A role for the inflammasome in spontaneous labor at term. Am J Reprod Immunol. 79:e124402018. View Article : Google Scholar

109 

Lindström TM and Bennett PR: The role of nuclear factor kappa B in human labour. Reproduction. 130:569–581. 2005. View Article : Google Scholar : PubMed/NCBI

110 

McCarthy R, Martin-Fairey C, Sojka DK, Herzog ED, Jungheim ES, Stout MJ, Fay JC, Mahendroo M, Reese J, Herington JL, et al: Mouse models of preterm birth: Suggested assessment and reporting guidelines. Biol Reprod. 99:922–937. 2018.PubMed/NCBI

111 

Zhu B, Tao Z, Edupuganti L, Serrano MG and Buck GA: Roles of the microbiota of the female reproductive tract in gynecological and reproductive health. Microbiol Mol Biol Rev. 86:e00181212022. View Article : Google Scholar : PubMed/NCBI

112 

Yudin MH, Landers DV, Meyn L and Hillier SL: Clinical and cervical cytokine response to treatment with oral or vaginal metronidazole for bacterial vaginosis during pregnancy: A randomized trial. Obstet Gynecol. 102:527–534. 2003.PubMed/NCBI

113 

Soto E, Romero R, Richani K, Yoon BH, Chaiworapongsa T, Vaisbuch E, Mittal P, Erez O, Gotsch F, Mazor M and Kusanovic JP: Evidence for complement activation in the amniotic fluid of women with spontaneous preterm labor and intra-amniotic infection. J Matern Fetal Neonatal Med. 22:983–992. 2009. View Article : Google Scholar : PubMed/NCBI

114 

Romero R and Tartakovsky B: The natural interleukin-1 receptor antagonist prevents interleukin-1-induced preterm delivery in mice. Am J Obstet Gynecol. 167:1041–1045. 1992. View Article : Google Scholar : PubMed/NCBI

115 

Nadeau-Vallée M, Quiniou C, Palacios J, Hou X, Erfani A, Madaan A, Sanchez M, Leimert K, Boudreault A, Duhamel F, et al: Novel noncompetitive IL-1 receptor-biased ligand prevents infection- and inflammation-induced preterm birth. J Immunol. 195:3402–3415. 2015. View Article : Google Scholar : PubMed/NCBI

116 

Robertson SA, Christiaens I, Dorian CL, Zaragoza DB, Care AS, Banks AM and Olson DM: Interleukin-6 is an essential determinant of on-time parturition in the mouse. Endocrinology. 151:3996–4006. 2010. View Article : Google Scholar : PubMed/NCBI

117 

Di Simone N, Santamaria Ortiz A, Specchia M, Tersigni C, Villa P, Gasbarrini A, Scambia G and D'Ippolito S: Recent insights on the maternal microbiota: Impact on pregnancy outcomes. Front Immunol. 11:5282022020. View Article : Google Scholar : PubMed/NCBI

118 

Chan D, Bennett PR, Lee YS, Kundu S, Teoh TG, Adan M, Ahmed S, Brown RG, David AL, Lewis HV, et al: Microbial-driven preterm labour involves crosstalk between the innate and adaptive immune response. Nat Commun. 13:9752022. View Article : Google Scholar : PubMed/NCBI

119 

Gonzalez JM, Franzke CW, Yang F, Romero R and Girardi G: Complement activation triggers metalloproteinases release inducing cervical remodeling and preterm birth in mice. Am J Pathol. 179:838–849. 2011. View Article : Google Scholar : PubMed/NCBI

120 

Robertson SA, Skinner RJ and Care AS: Essential role for IL-10 in resistance to lipopolysaccharide-induced preterm labor in mice. J Immunol. 177:4888–4896. 2006. View Article : Google Scholar : PubMed/NCBI

121 

Osman I, Young A, Ledingham MA, Thomson AJ, Jordan F, Greer IA and Norman JE: Leukocyte density and pro-inflammatory cytokine expression in human fetal membranes, decidua, cervix and myometrium before and during labour at term. Mol Hum Reprod. 9:41–45. 2003. View Article : Google Scholar : PubMed/NCBI

122 

Rinaldi SF, Catalano RD, Wade J, Rossi AG and Norman JE: Decidual neutrophil infiltration is not required for preterm birth in a mouse model of infection-induced preterm labor. J Immunol. 192:2315–2325. 2014. View Article : Google Scholar : PubMed/NCBI

123 

Mohd Zaki A, Hadingham A, Flaviani F, Haque Y, Mi JD, Finucane D, Dalla Valle G, Mason AJ, Saqi M, Gibbons DL and Tribe RM: Neutrophils dominate the cervical immune cell population in pregnancy and their transcriptome correlates with the microbial vaginal environment. Front Microbiol. 13:9044512022. View Article : Google Scholar : PubMed/NCBI

124 

Cezar-de-Mello PFT, Ryan S and Fichorova RN: The microRNA cargo of human vaginal extracellular vesicles differentiates parasitic and pathobiont infections from colonization by homeostatic bacteria. Microorganisms. 11:5512023. View Article : Google Scholar : PubMed/NCBI

125 

Zen M, Canova M, Campana C, Bettio S, Nalotto L, Rampudda M, Ramonda R, Iaccarino L and Doria A: The kaleidoscope of glucorticoid effects on immune system. Autoimmun Rev. 10:305–310. 2011. View Article : Google Scholar : PubMed/NCBI

126 

Busillo JM, Azzam KM and Cidlowski JA: Glucocorticoids sensitize the innate immune system through regulation of the NLRP3 inflammasome. J Biol Chem. 286:38703–38713. 2011. View Article : Google Scholar : PubMed/NCBI

127 

Hermoso MA, Matsuguchi T, Smoak K and Cidlowski JA: Glucocorticoids and tumor necrosis factor alpha cooperatively regulate toll-like receptor 2 gene expression. Mol Cell Biol. 24:4743–4756. 2004. View Article : Google Scholar : PubMed/NCBI

128 

Baschant U and Tuckermann J: The role of the glucocorticoid receptor in inflammation and immunity. J Steroid Biochem Mol Biol. 120:69–75. 2010. View Article : Google Scholar : PubMed/NCBI

129 

Franchimont D: Overview of the actions of glucocorticoids on the immune response: A good model to characterize new pathways of immunosuppression for new treatment strategies. Ann NY Acad Sci. 1024:124–137. 2004. View Article : Google Scholar : PubMed/NCBI

130 

Hearps AC, Tyssen D, Srbinovski D, Bayigga L, Diaz DJD, Aldunate M, Cone RA, Gugasyan R, Anderson DJ and Tachedjian G: Vaginal lactic acid elicits an anti-inflammatory response from human cervicovaginal epithelial cells and inhibits production of pro-inflammatory mediators associated with HIV acquisition. Mucosal Immunol. 10:1480–1490. 2017. View Article : Google Scholar : PubMed/NCBI

131 

Delgado-Diaz DJ, Tyssen D, Hayward JA, Gugasyan R, Hearps AC and Tachedjian G: Distinct immune responses elicited from cervicovaginal epithelial cells by lactic acid and short chain fatty acids associated with optimal and non-optimal vaginal microbiota. Front Cell Infect Microbiol. 9:4462020. View Article : Google Scholar : PubMed/NCBI

132 

Galdeano CM and Perdigón G: The probiotic bacterium Lactobacillus casei induces activation of the gut mucosal immune system through innate immunity. Clin Vaccine Immunol. 13:219–226. 2006. View Article : Google Scholar : PubMed/NCBI

133 

Rocha-Ramírez LM, Pérez-Solano RA, Castañón-Alonso SL, Moreno Guerrero SS, Ramírez Pacheco A, García Garibay M and Eslava C: Probiotic lactobacillus strains stimulate the inflammatory response and activate human macrophages. J Immunol Res. 2017:46074912017. View Article : Google Scholar : PubMed/NCBI

134 

Anton L, Sierra LJ, DeVine A, Barila G, Heiser L, Brown AG and Elovitz MA: Common cervicovaginal microbial supernatants alter cervical epithelial function: Mechanisms by which lactobacillus crispatus contributes to cervical health. Front Microbiol. 9:21812018. View Article : Google Scholar : PubMed/NCBI

135 

Dizzell S, Nazli A, Reid G and Kaushic C: Protective effect of probiotic bacteria and estrogen in preventing HIV-1-mediated impairment of epithelial barrier integrity in female genital tract. Cells. 8:11202019. View Article : Google Scholar : PubMed/NCBI

136 

Qi W, Li H, Wang C, Li H, Zhang B, Dong M, Fan A, Han C and Xue F: Recent advances in presentation, diagnosis and treatment for mixed vaginitis. Front Cell Infect Microbiol. 11:7597952021. View Article : Google Scholar : PubMed/NCBI

137 

Dong M, Dong Y, Bai J, Li H, Ma X, Li B, Wang C, Li H, Qi W, Wang Y, et al: Interactions between microbiota and cervical epithelial, immune, and mucus barrier. Front Cell Infect Microbiol. 13:11245912023. View Article : Google Scholar : PubMed/NCBI

138 

Schönfeld P and Wojtczak L: Short- and medium-chain fatty acids in energy metabolism: The cellular perspective. J Lipid Res. 57:943–954. 2016. View Article : Google Scholar : PubMed/NCBI

139 

Aldunate M, Srbinovski D, Hearps AC, Latham CF, Ramsland PA, Gugasyan R, Cone RA and Tachedjian G: Antimicrobial and immune modulatory effects of lactic acid and short chain fatty acids produced by vaginal microbiota associated with eubiosis and bacterial vaginosis. Front Physiol. 6:1642015. View Article : Google Scholar : PubMed/NCBI

140 

Amabebe E and Anumba DOC: The vaginal microenvironment: The physiologic role of lactobacilli. Front Med (Lausanne). 5:1812018. View Article : Google Scholar : PubMed/NCBI

141 

Beghini J, Giraldo PC, Linhares IM, Ledger WJ and Witkin SS: Neutrophil gelatinase-associated lipocalin concentration in vaginal fluid: relation to bacterial vaginosis and vulvovaginal candidiasis. Reprod Sci. 22:964–968. 2015. View Article : Google Scholar : PubMed/NCBI

142 

Stafford GP, Parker JL, Amabebe E, Kistler J, Reynolds S, Stern V, Paley M and Anumba DOC: spontaneous preterm birth is associated with differential expression of vaginal metabolites by lactobacilli-dominated microflora. Front Physiol. 8:6152017. View Article : Google Scholar : PubMed/NCBI

143 

Amabebe E and Anumba DOC: A combination of cervicovaginal fluid glutamate, acetate and D-Lactate identified asymptomatic low-risk women destined to deliver preterm: A prospective cohort study. Reprod Sci. 29:915–922. 2022. View Article : Google Scholar

144 

Ghartey J, Bastek JA, Brown AG, Anglim L and Elovitz MA: Women with preterm birth have a distinct cervicovaginal metabolome. Am J Obstet Gynecol. 212:776.e1–776.e12. 2015. View Article : Google Scholar : PubMed/NCBI

145 

Srinivasan S, Morgan MT, Fiedler TL, Djukovic D, Hoffman NG, Raftery D, Marrazzo JM and Fredricks DN: Metabolic signatures of bacterial vaginosis. mBio. 6:e002042015. View Article : Google Scholar : PubMed/NCBI

146 

Menon R, Jones J, Gunst PR, Kacerovsky M, Fortunato SJ, Saade GR and Basraon S: Amniotic fluid metabolomic analysis in spontaneous preterm birth. Reprod Sci. 21:791–803. 2014. View Article : Google Scholar : PubMed/NCBI

147 

Lizewska B, Teul J, Kuc P, Lemancewicz A, Charkiewicz K, Goscik J, Kacerovsky M, Menon R, Miltyk W and Laudanski P: Maternal plasma metabolomic profiles in spontaneous preterm birth: Preliminary results. Mediators Inflamm. 2018:93628202018. View Article : Google Scholar : PubMed/NCBI

148 

Smith DD and Rood KM: Intrahepatic cholestasis of pregnancy. Clin Obstet Gynecol. 63:134–151. 2020. View Article : Google Scholar

149 

Tuckey RC: Progesterone synthesis by the human placenta. Placenta. 26:273–281. 2005. View Article : Google Scholar : PubMed/NCBI

150 

Wilks M, Wiggins R, Whiley A, Hennessy E, Warwick S, Porter H, Corfield A and Millar M: Identification and H(2) O(2) production of vaginal lactobacilli from pregnant women at high risk of preterm birth and relation with outcome. J Clin Microbiol. 42:713–717. 2004. View Article : Google Scholar : PubMed/NCBI

151 

O'Hanlon DE, Moench TR and Cone RA: In vaginal fluid, bacteria associated with bacterial vaginosis can be suppressed with lactic acid but not hydrogen peroxide. BMC Infect Dis. 11:2002011. View Article : Google Scholar : PubMed/NCBI

152 

Mitchell C, Fredricks D, Agnew K and Hitti J: Hydrogen peroxide-producing lactobacilli are associated with lower levels of vaginal interleukin-1β, independent of bacterial vaginosis. Sex Transm Dis. 42:358–363. 2015. View Article : Google Scholar : PubMed/NCBI

153 

Hemmerling A, Harrison W, Schroeder A, Park J, Korn A, Shiboski S, Foster-Rosales A and Cohen CR: Phase 2a study assessing colonization efficiency, safety, and acceptability of Lactobacillus crispatus CTV-05 in women with bacterial vaginosis. Sex Transm Dis. 37:745–750. 2010. View Article : Google Scholar : PubMed/NCBI

154 

Mayer BT, Srinivasan S, Fiedler TL, Marrazzo JM, Fredricks DN and Schiffer JT: Rapid and profound shifts in the vaginal microbiota following antibiotic treatment for bacterial vaginosis. J Infect Dis. 212:793–802. 2015. View Article : Google Scholar : PubMed/NCBI

155 

Bayar E, MacIntyre DA, Sykes L, Mountain K, Parks TP, Lee PP and Bennett PR: Safety, tolerability, and acceptability of Lactobacillus crispatus CTV-05 (LACTIN-V) in pregnant women at high-risk of preterm birth. Benef Microbes. 14:45–56. 2023. View Article : Google Scholar : PubMed/NCBI

156 

Tomusiak A, Strus M, Heczko PB, Adamski P, Stefański G, Mikołajczyk-Cichońska A and Suda-Szczurek M: Efficacy and safety of a vaginal medicinal product containing three strains of probiotic bacteria: A multicenter, randomized, double-blind, and placebo-controlled trial. Drug Des Devel Ther. 9:5345–5354. 2015. View Article : Google Scholar : PubMed/NCBI

157 

Vasundhara D, Raju VN, Hemalatha R, Nagpal R and Kumar M: Vaginal & gut microbiota diversity in pregnant women with bacterial vaginosis & effect of oral probiotics: An exploratory study. Indian J Med Res. 153:492–502. 2021. View Article : Google Scholar : PubMed/NCBI

158 

Reid G, Charbonneau D, Erb J, Kochanowski B, Beuerman D, Poehner R and Bruce AW: Oral use of Lactobacillus rhamnosus GR-1 and L. fermentum RC-14 significantly alters vaginal flora: Randomized, placebo-controlled trial in 64 healthy women. FEMS Immunol Med Microbiol. 35:131–134. 2003. View Article : Google Scholar : PubMed/NCBI

159 

Ho M, Chang YY, Chang WC, Lin HC, Wang MH, Lin WC and Chiu TH: Oral Lactobacillus rhamnosus GR-1 and Lactobacillus reuteri RC-14 to reduce Group B Streptococcus colonization in pregnant women: A randomized controlled trial. Taiwan J Obstet Gynecol. 55:515–518. 2016. View Article : Google Scholar : PubMed/NCBI

160 

Anukam KC, Osazuwa E, Osemene GI, Ehigiagbe F, Bruce AW and Reid G: Clinical study comparing probiotic Lactobacillus GR-1 and RC-14 with metronidazole vaginal gel to treat symptomatic bacterial vaginosis. Microbes Infect. 8:2772–2776. 2006. View Article : Google Scholar : PubMed/NCBI

161 

Hemalatha R, Mastromarino P, Ramalaxmi BA, Balakrishna NV and Sesikeran B: Effectiveness of vaginal tablets containing lactobacilli versus pH tablets on vaginal health and inflammatory cytokines: A randomized, double-blind study. Eur J Clin Microbiol Infect Dis. 31:3097–3105. 2012. View Article : Google Scholar : PubMed/NCBI

162 

Husain S, Allotey J, Drymoussi Z, Wilks M, Fernandez-Felix BM, Whiley A, Dodds J, Thangaratinam S, McCourt C, Prosdocimi EM, et al: Effects of oral probiotic supplements on vaginal microbiota during pregnancy: A randomised, double-blind, placebo-controlled trial with microbiome analysis. BJOG. 127:275–284. 2020. View Article : Google Scholar

163 

Gille C, Böer B, Marschal M, Urschitz MS, Heinecke V, Hund V, Speidel S, Tarnow I, Mylonas I, Franz A, et al: Effect of probiotics on vaginal health in pregnancy. EFFPRO, a randomized controlled trial. Am J Obstet Gynecol. 215:608.e1–608.e7. 2016. View Article : Google Scholar

164 

Govinden G, Parker JL, Naylor KL, Frey AM, Anumba DOC and Stafford GP: Inhibition of sialidase activity and cellular invasion by the bacterial vaginosis pathogen Gardnerella vaginalis. Arch Microbiol. 200:1129–1133. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

  • Abstract
  • View
  • Download
  • Twitter
Copy and paste a formatted citation
Spandidos Publications style
Cheng D, Li N, Sun Q, Wang K and Gao F: Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review). Int J Mol Med 56: 203, 2025.
APA
Cheng, D., Li, N., Sun, Q., Wang, K., & Gao, F. (2025). Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review). International Journal of Molecular Medicine, 56, 203. https://doi.org/10.3892/ijmm.2025.5644
MLA
Cheng, D., Li, N., Sun, Q., Wang, K., Gao, F."Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review)". International Journal of Molecular Medicine 56.6 (2025): 203.
Chicago
Cheng, D., Li, N., Sun, Q., Wang, K., Gao, F."Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 203. https://doi.org/10.3892/ijmm.2025.5644
Copy and paste a formatted citation
x
Spandidos Publications style
Cheng D, Li N, Sun Q, Wang K and Gao F: Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review). Int J Mol Med 56: 203, 2025.
APA
Cheng, D., Li, N., Sun, Q., Wang, K., & Gao, F. (2025). Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review). International Journal of Molecular Medicine, 56, 203. https://doi.org/10.3892/ijmm.2025.5644
MLA
Cheng, D., Li, N., Sun, Q., Wang, K., Gao, F."Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review)". International Journal of Molecular Medicine 56.6 (2025): 203.
Chicago
Cheng, D., Li, N., Sun, Q., Wang, K., Gao, F."Vaginal microbiome and preterm birth: Composition, mechanisms and microbiota‑directed therapies (Review)". International Journal of Molecular Medicine 56, no. 6 (2025): 203. https://doi.org/10.3892/ijmm.2025.5644
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