Abstract
Bacterial vaginosis (BV) is a complex polymicrobial vaginal infection that affects a large percentage of women during different stages of life including the reproductive age. In a healthy vaginal environment, the epithelium is colonized by protective Lactobacillus species that make up 90%–95% of the total vaginal microbiota. BV is characterized by a reduction of lactobacilli and a concurrent increase in diverse anaerobic bacteria, including Gardnerella vaginalis, Prevotella bivia, Hoylesella timonensis, and Fannyhessea vaginae. BV is associated with an increased risk of infertility, preterm birth, and a higher susceptibility to sexually transmitted infections (STIs), including Human Immunodeficiency Virus type-1 (HIV-1). This review examines the contribution of individual pathogenic bacteria to the development of BV and the resulting effects on susceptibility to STI. The impact of the different key bacterial virulence factors, such as secreted proteins, biofilm formation, and inflammatory potential on subsequent viral infection are discussed. While antibiotics are commonly prescribed to treat BV, recurrence rates are high, and antimicrobial resistance among BV-associated bacteria is increasingly reported. Understanding the mechanisms underlying BV and the impact of specific bacteria and their virulence factors on viral infections can improve preventive strategies and open up novel therapeutic applications.
Keywords: vaginal microbiota, bacterial vaginosis, Gardnerella vaginalis, Hoylesella timonensis, Prevotella timonensis, Prevotella bivia, Fannyhessea vaginae, Atopobium vaginae, Mobiluncus mulieris, Sneathia amnii, HIV-1
Review of the individual bacterial vaginosis-associated bacteria, their unique virulence factors, and their impact on viral infection, with an emphasis on the molecular mechanisms of microbe–host interactions in the vaginal mucosa.
Introduction
The cervicovaginal epithelium consists of a stratified squamous nonkeratinized epithelium covered in cervicovaginal mucus (CVM) and cervicovaginal fluid (Lacroix et al. 2020). This gel-like mucosa layer serves as a protective barrier against infectious microorganisms, lubricates the vaginal epithelium, and harbors the vaginal microbiome (Allen et al. 1982). The vaginal microbiome is influenced by several factors, including hormonal levels (Farage et al. 2010), race (Sun et al. 2022), number of sexual partners (Nelson et al. 2012, Ma 2022), contraceptive use (Vodstrcil et al. 2017, Achilles et al. 2018), the use of sex toys, and vaginal lubricant (Laniewski et al. 2019).
In healthy women, the vaginal microbiome is dominated by Lactobacillus species including L. crispatus, L. gasseri, L. jensenii, and L. iners. Usually, women are colonized by a single dominant Lactobacillus species that accounts for ~90%–95% of the total microbial community (Auriemma et al. 2021: 2, Pendharkar et al. 2023). Under healthy conditions, the lactobacilli produce lactic acid, which maintains the vaginal pH at ∼4 (O’Hanlon et al. 2013, Ñahui Palomino et al. 2017). In addition to lactic acid (Boris and Barbés 2000, O’Hanlon et al. 2011), lactobacilli also produce antimicrobial compounds, such as hydrogen peroxide (Sgibnev and Kremleva 2015), bacteriocins (Stoyancheva et al. 2014), and arginine deaminase enzymes (Rousseau et al. 2005), that all suppress the growth of anaerobic bacteria. Additionally, lactobacilli strongly adhere to the vaginal epithelium, which serves as a protective mechanism as thereby attachment of anaerobic bacteria is limited (Edelman et al. 2012, Leccese Terraf et al. 2014, He et al. 2020, Parolin et al. 2021, Qian et al. 2021). Several reviews about the protective roles of vaginal Lactobacillus spp. have recently been published (Amabebe and Anumba 2018, Chee et al. 2020, Pendharkar et al. 2023). Overall, the high prevalence of lactobacilli is key for maintaining an acidic vaginal pH and limiting anaerobic bacterial growth.
Bacterial vaginosis (BV) is a polymicrobial infection that occurs when the vaginal microbiome shifts from a Lactobacillus-dominated microbiome toward a higher prevalence of facultative or obligate anaerobic bacteria, such as Gardnerella spp., Hoylesella spp., Prevotella spp., Fannyhessea spp., Mobiluncus spp., Sneathia spp., and BV-associated bacteria (BVAB) 1–3 (Fredricks and Marrazzo 2005, Srinivasan et al. 2012, Gao et al. 2022). The Amsel criteria are commonly used to diagnose BV and are based on an increased vaginal pH (>4.5), thinned vaginal discharge, amine odor after addition of potassium hydroxide, and the presence of “clue cells” (Amsel et al. 1983, Mohammadzadeh et al. 2015). The Nugent score test is also used for BV diagnosis, which involves the quantification of Gram-positive rods, such as lactobacilli (healthy bacteria) and Gram-negative or Gram-variable bacteria (associated with BV) (Nugent et al. 1991, Coleman and Gaydos 2018). BV is associated with infertility (Ravel et al. 2021), adverse pregnancy outcomes such as preterm birth (Dingens et al. 2016), and increased susceptibility to sexually transmitted infections (STIs), including Human Immunodeficiency Virus type-1 (HIV-1) (Sha et al. 2005, Borgdorff et al. 2014, Kyongo et al. 2015, Gosmann et al. 2017), Human Papilloma Virus (HPV) (Gillet et al. 2011, Brotman et al. 2014, Li et al. 2020), and Herpes Simplex Virus type-2 (HSV-2) (Cherpes et al. 2005, Nagot et al. 2007). Virulence factors associated with BV include bacterial adherence to the cervicovaginal epithelium, biofilm formation (Patterson et al. 2010, Machado et al. 2013), and expression of cytotoxins (Gelber et al. 2008, Rampersaud et al. 2011) and enzymes, such as mucinases, sulfatases, galactosidases, and prolidases (Howe et al. 1999, Olmsted et al. 2003, Cauci et al. 2005, Moncla et al. 2016). For instance, sialidases are enzymes found at higher levels in women with BV and have been shown to degrade the protective mucus layer (Briselden et al. 1992, Lewis et al. 2013, Plesniarski et al. 2021) and other glycoproteins, such as the secretory Immunoglobulin A (IgA) (Lewis et al. 2012). Moreover, certain anaerobic bacteria can induce proinflammatory responses, resulting in recruitment of immune cells, some of which can be targets for viruses, such as Langerhans cells and T cells for HIV-1 (Libby et al. 2008, Doerflinger et al. 2014).
Numerous studies have reported associations between an altered vaginal microbiome and increased susceptibility to viral infections that affect millions of women worldwide. For HIV-1 infections, BV is associated with HIV-1 prevalence, increased viral load, and genital HIV-1 RNA shedding (Cherpes et al. 2005, Atashili et al. 2008, Borgdorff et al. 2014, Kyongo et al. 2015, Gosmann et al. 2017). A BV-associated vaginal microbiome is also thought to increase the risk of developing HPV-induced cervical cancer (Gillet et al. 2011, Moscicki et al. 2012, Łaniewski et al. 2018). The HSV-2 virus is the main cause of genital herpes and several studies have suggested an association between BV and an increased risk of infection (Cherpes et al. 2005, Kaul et al. 2007, Nagot et al. 2007). It is evident that the polymicrobial anaerobic colonization in BV greatly enhances susceptibility to viral infections, but the underlying mechanisms and the specific roles of individual bacterial spp. are not fully understood. This review summarizes the current knowledge about virulence traits of specific vaginal BV-associated anaerobic bacteria and their impact on virus infection.
Virulence-associated traits of anaerobic vaginal bacteria
Gardnerella vaginalis
Gardnerella vaginalis is the most extensively studied BV-associated facultative anaerobic bacterium. Within the G. vaginalis clade, there is a high strain diversity and also other non-vaginalis Gardnerella subspecies can play roles in BV (Tortelli et al. 2021). Staining of Gardnerella subspecies can be Gram-variable. Genomic and functional analyses of different G. vaginalis strains demonstrated the presence of a range of virulence traits, including adherence to the epithelium, biofilm formation, hemolytic and cytotoxic abilities, immune evasion, and antimicrobial resistance (Yeoman et al. 2010). Gardnerella vaginalis can adhere efficiently to the vaginal epithelium and produce a biofilm (Yeoman et al. 2010, Alves et al. 2014) and in this process displace adherent lactobacilli (Machado et al. 2013). Therefore, G. vaginalis is considered an important early colonizer that facilitates the subsequent attachment of other anaerobes.
An important virulence factor of G. vaginalis is the cytotoxin vaginolysin that is highly expressed in BV-associated G. vaginalis strains. Vaginolysin targets CD59, also known as MAC-inhibitory protein, on epithelial cells, erythrocytes, and neutrophils resulting in membrane blebbing and pore formation (Patterson et al. 2010, Yeoman et al. 2010, Randis et al. 2013, Anton et al. 2022). Bacterial membrane vesicles can carry vaginolysin and induce cytotoxicity and proinflammatory responses in vaginal epithelial cells (Shishpal et al. 2020). Gardnerella vaginalis strains also encode oxygen-independent coproporphyrinogen III oxygenases (HemN), which are involved in the release and utilization of iron from hemoglobin and myoglobin (Yeoman et al. 2010). Other toxin-related G. vaginalis genes include potential lysozyme-like toxins, toxin–antitoxin genes, and an invasion-associated hydrolase (Yeoman et al. 2010). Together, the G. vaginalis virulence factors promote a proinflammatory response in cervicovaginal epithelial cells (Libby et al. 2008, Doerflinger et al. 2014, Santos et al. 2018, Anton et al. 2022), induce cellular exfoliation (Lewis et al. 2013, Gilbert et al. 2019), and increase epithelial permeability in vitro (Anton et al. 2022, Berard et al. 2023). However, the in vivo importance of the different factors remains unclear.
Similar to the vaginal Lactobacillus spp., G. vaginalis can utilize glycogen, which is an abundant carbon source in the vaginal epithelium (Bhandari and Hill 2023, Jenkins et al. 2023, Navarro et al. 2023, Segui-Perez et al. 2024). In addition, G. vaginalis degrades the protective mucus layer through the production of sialidases that cleave terminal sialic acid from mucins (Gilbert et al. 2019, Agarwal and Lewis 2021). Gardnerella spp. encode up to four sialidases, NanH1, NanH2, NanH3, and NanH4, (Robinson et al. 2019) that can be highly abundant in clinical BV samples (Hardy et al. 2017, Pelayo et al. 2024). Of these four enzymes, only GvNanH2 and GvNanH3 display high sialidase activity in in vitro assays and can effectively remove sialic acids from N-linked and O-linked sialoglycan substrates (Lewis et al. 2013, Robinson et al. 2019, Pelayo et al. 2024). A recent study demonstrated that recombinant sialidases of G. vaginalis efficiently desialylate vaginal epithelial glycans, inducing pathways related to cell death, differentiation, and inflammatory responses (Agarwal et al. 2023). The sialidase activity of G. vaginalis also plays an important role in cellular invasion (Govinden et al. 2018) and the proteolytic degradation of IgA molecules (Lewis et al. 2012). These studies underscore the significance of sialidase activity as a virulence factor in a wide range of pathogenic processes. In summary, G. vaginalis contributes to BV by initial adhesion to the epithelium, degradation of the protective mucus layer, inducing cytotoxicity, promoting inflammation, and compromising epithelial integrity. The main virulence factors of G. vaginalis and the bacteria described below are summarized in Table 1.
Table 1.
Overview of virulence factors associated with individual anaerobic vaginal bacteria.
| Bacteria | Adherence/biofilm | Secretion of toxins and other compounds | Glycosidases/impact on mucins | Immune system activation or evasion | Antibiotic resistance |
|---|---|---|---|---|---|
| Gardnerella vaginalis | Adheres to the cervicovaginal epithelium, can displace adherent lactobacilli (Machado et al. 2013a), and promotes biofilm formation (Patterson et al. 2010, Yeoman et al. 2010, Alves et al. 2014). Encodes genes for fimbriae/pili (Yeoman et al. 2010). | Produces vaginolysin (Gelber et al. 2008), and encodes other toxins, such as lysozyme-like toxins, oxygen-independent oxidases (HemN), several toxin antitoxins genes, and an invasion-associated hydrolase (Yeoman et al. 2010).It causes cytotoxicity in several epithelial cell lines (Patterson et al. 2010, Castro et al. 2015, Anton et al. 2022, Segui-Perez et al. 2024). | Can utilize glycogen (Bhandari and Hill 2023, Jenkins et al. 2023, Navarro et al. 2023, Segui-Perez et al. 2024) and exhibits sialidase activity, degrading cervicovaginal cell sialoglycans (Gilbert et al. 2019, Robinson et al. 2019), and secretory IgA (Lewis et al. 2012). | Induces IL-8, IL-6, and CCL-20 production by cervicovaginal epithelial cells (Libby et al. 2008, Anton et al. 2022, Segui-Perez et al. 2024), and HeLa cells (Santos et al. 2018). | Some strains are resistant to metronidazole and bleomycin (Yeoman et al. 2010, Petrina et al. 2017). |
| Hoylesella timonensis | Adheres to the cervicovaginal epithelium (Ilhan et al. 2020, Segui-Perez et al. 2024). Forms a sparse biofilm and elongates microvilli in endometrial cells (Ilhan et al. 2020). | Does not have cytotoxic activity on cervicovaginal cells (Segui-Perez et al. 2024). | Can utilize glycogen (Jenkins et al. 2023, Segui-Perez et al. 2024) and degrade the vaginal mucus layer due to high sialidase and fucosidase activities (Jenkins et al. 2023, Pelayo et al. 2024, Segui-Perez et al. 2024). Encodes an extensive array of carbohydrate-active enzymes (CAZymes) (Segui-Perez et al. 2024). | Does not induce a proinflammatory response in vaginal epithelial cells (Ilhan et al. 2020, Segui-Perez et al. 2024), but stimulates dendritic cells toward Th1 polarization (van Teijlingen et al. 2020). Increases HIV-1 uptake in Langerhans cells and DCs (Van Teijlingen et al. 2022) and infection in CD4 + T cells (van Teijlingen et al. 2024). | Some strains are resistant to clindamycin (Petrina et al. 2017). |
| Prevotella bivia | Does not adhere to cervicovaginal epithelial cells (Segui-Perez et al. 2024), but forms a sparse biofilm in endometrial cells (Ilhan et al. 2020). Can join a G. vaginalis-biofilm (Machado et al. 2013, Castro et al. 2021a). | Produces ammonia (Pybus and Onderdonk 1997), polyamines (Łaniewski and Herbst-Kralovetz 2021), and collagenases (Doust and Mobarez 2004). Does not have cytotoxic activity (Segui-Perez et al. 2024). | Can degrade glycogen (Segui-Perez et al. 2024). Expresses sialidase activity, but does not efficiently degrade the glycocalyx in vaginal epithelial cells (Gilbert et al. 2019, Segui-Perez et al. 2024). | Does not induce a proinflammatory response in cervicovaginal epithelial cells in most studies (Doerflinger et al. 2014, Ilhan et al. 2020, Segui-Perez et al. 2024). | Some strains are resistant to clindamycin and metronidazole (Petrina et al. 2017, Veloo et al. 2018). |
| Fannyhessea vaginae | Does not adhere well to the epithelium on its own. Joins G. vaginalis biofilm (Hardy et al. 2016, Castro et al. 2019, 2020). | Does not induce cytotoxicity in ME-180 vaginal epithelial cells(Patterson et al. 2010). | Not described. | Upregulates proinflammatory cytokines TNFα, CCL-20, IL-6, and IL-8 in cervicovaginal epithelial cells (Trama et al. 2007, Libby et al. 2008, Doerflinger et al. 2014). | Some strains are resistant to metronidazole and secnidazole (De Backer et al. 2010, Petrina et al. 2017). |
| Mobiluncus mulieris | Limited adherence to ME-180 cells and can be outcompeted by L. crispatus (Machado et al. 2013b). Does not form a biofilm (Patterson et al. 2010). | Is not cytotoxic (Patterson et al. 2010). Has proteolytic activity through the production of proline aminopeptidase (Schoonmaker et al. 1991). | Not described. | Increases inflammation through flagella-mediated TLR5 activation (McKenzie et al. 2021) | Some strains are resistant to metronidazole (Spiegel 1987). |
| Sneathia amnii | Can adhere to a ME-180 cervical cell line and encodes for a fibronectin-binding protein (Harwich et al. 2012). | Expresses the cytotoxin CptA (Gentile et al. 2020), and encodes for multiple putative invasins (Harwich et al. 2012). Produces polyamines (Łaniewski and Herbst-Kralovetz 2021). | Its genome encodes for a potential O-sialoglycoprotein endopeptidase. Can degrade glycogen (Harwich et al. 2012). | Induces a proinflammatory response in vaginal epithelial cells (Anahtar et al. 2015) and in a human 3D cervical cell model (Łaniewski and Herbst-Kralovetz 2021). | Sensitive to metronidazole (Harwich et al. 2012). |
Hoylesella timonensis (formerly Prevotella timonensis)
Different Prevotella spp. including P. bivia and P. amnii have been implicated in the pathogenesis of BV. Some of these bacteria are closely related with highly similar 16S rRNA genes, which has hampered classification in the past (Ilhan et al. 2020, Hitch et al. 2022). Hoylesella timonensis, a Gram-negative bacterium often present in women with BV (Srinivasan et al. 2012, Gao et al. 2022), was previously classified as Prevotella timonensis, but has recently been reassigned to the new genus Hoylesella based on phylogenetic analysis (Hitch et al. 2022). The pathogenic potential of H. timonensis has been relatively underexplored in comparison to G. vaginalis, but more publications are emerging that emphasize the unique contributions of H. timonensis to BV. Similar to G. vaginalis, H. timonensis can adhere to the vaginal epithelium (Segui-Perez et al. 2024). It can form a sparse biofilm and was shown to induce microvilli elongation in a 3D-endometrial cell model (Ilhan et al. 2020). It remains to be determined if H. timonensis can displace lactobacilli as has been demonstrated for G. vaginalis, but these traits suggest that H. timonensis could have the capacity to be an early colonizer in BV.
The H. timonensis genome does not encode any evident toxin genes. An important aspect of H. timonensis virulence potential seems to be its extensive repertoire of carbohydrate-active enzymes (CAZymes). Hoylesella timonensis encodes a broad array of mucin-degrading enzymes, including fucosidases and sialidases (Segui-Perez et al. 2024). The bacteria express two highly active GH33 sialidases, NanH1 and NanH2 and these H. timonensis enzymes are highly abundant in clinical BV samples as detected by metagenomic and metatranscriptomic analysis (Pelayo et al. 2024). The H. timonensis NanH1 and NanH2 enzymes effectively remove sialic acids from mucin substrates and the vaginal epithelial glycocalyx (Segui-Perez et al. 2024). Interestingly, similarly to G. vaginalis NanH3, the H. timonensis sialidases are also active at low pH, indicating that they potentially remain active in Lactobacillus-dominated communities (Pelayo et al. 2024). The high capacity of these bacteria to degrade mucus perhaps explains the association of H. timonensis with thinned vaginal discharge (Coleman and Gaydos 2018). In addition to the sialidases and fucosidases, H. timonensis encodes a broad spectrum of other CAZymes, including α-glucosidases that allow the bacteria to use glycogen as a sole carbon source (Jenkins et al. 2023, Segui-Perez et al. 2024).
In vitro studies indicate that H. timonensis does not induce production of proinflammatory cytokines by vaginal and endocervical cells (Ilhan et al. 2020, Segui-Perez et al. 2024). In contrast, other vaginal Prevotella spp., such as P. amnii and P. bivia, do induce secretion of IL-1α, IL-1β, and IL-8 from human vaginal epithelial cells (Anahtar et al. 2015, Gosmann et al. 2017). When in contact with dendritic cells, H. timonensis induces a strong proinflammatory response that is more significant than G. vaginalis. When T cells were exposed to H. timonensis, a T helper 1 skewing was observed that might be associated with enhanced susceptibility to STIs and preterm birth in the presence of H. timonensis (van Teijlingen et al. 2020). In conclusion, once H. timonensis establishes itself in an anaerobic vaginal microenvironment, it adheres efficiently to the epithelium, is well-adapted to utilizing glycogen as an energy source and can degrade the mucus layer, which may provide nutrients and adhesion sites for other bacteria. Its ability to control inflammatory responses by the epithelium and immune cells most likely contributes to long-term colonization of the vaginal environment.
Prevotella bivia
Prevotella bivia is another member of the Prevotella clade and has been implicated in the pathogenesis of BV by multiple studies (Smayevsky et al. 2001, Muzny et al. 2018). Like H. timonensis, P. bivia is capable of forming a sparse biofilm in a 3D-endometrial cell model (Ilhan et al. 2020), but it does not strongly adhere to the cervicovaginal epithelium (Segui-Perez et al. 2024). Once G. vaginalis has established a biofilm, P. bivia can join in, suggesting that P. bivia may be a secondary colonizer (Machado et al. 2013, Castro et al. 2021). Prevotella bivia produces ammonia, which promotes the growth of G. vaginalis (Pybus and Onderdonk 1997), and the synergistic relationship between P. bivia and G. vaginalis likely contributes to the establishment of polymicrobial colonization that is characteristic of BV.
Prevotella bivia exhibits several enzymatic activities that may contribute to its pathogenic potential. It produces collagenase, which may promote the detachment of vaginal epithelial cells (Doust and Mobarez 2004, Łaniewski and Herbst-Kralovetz 2021). Prevotella bivia can grow on glycogen, but its mucin-degrading capacity is not as extensive as H. timonensis (Segui-Perez et al. 2024). However, P. bivia does express active fucosidases and sialidases in vitro and in mouse models (Gilbert et al. 2019, Segui-Perez et al. 2024). The P. bivia genome encodes two GH29 fucosidase genes and one GH33 sialidase gene (NanH). Unlike H. timonensis, the P. bivia enzymes are not active at the vaginal epithelial glycocalyx (Segui-Perez et al. 2024). These findings indicate that the differential levels of sialidase activity and substrate specificity among Gardnerella, Hoylesella, and Prevotella spp. are important characteristics that distinguish these bacterial species. Another property of P. bivia is the production of polyamines, such as putrescine, spermidine, and cadaverine, which are compounds that cause the malodor that can be a characteristic of BV and contribute to an increased vaginal pH (Nelson et al. 2015, Łaniewski and Herbst-Kralovetz 2021).
The inflammatory potential of P. bivia in the cervicovaginal environment remains unclear. Prevotella bivia did not induce a proinflammatory response or cytotoxicity in several studies (Doerflinger et al. 2014, Ilhan et al. 2020, Segui-Perez et al. 2024), but was reported to induce IL-6 and IL-8 expression in another study with cervical epithelial cells (Gosmann et al. 2017). It remains to be investigated if these differences are a result of strain variation between different studies. Overall, P. bivia does not appear to be an initial colonizer of the vaginal epithelium. However, once it integrates into the G. vaginalis biofilm, it supports the growth of G. vaginalis and expresses sialidases and collagenases, contributing to the epithelial damage that is characteristic of BV.
Fannyhessea vaginae (formerly Atopobium vaginae)
Fannyhessea vaginae, previously known as Atopobium vaginae, is a facultative anaerobic Gram-positive commonly found in women with BV (Burton et al. 2004, Ferris et al. 2004, De Backer et al. 2007). Fannyhessea vaginae is often found alongside G. vaginalis, and the cooccurrence of these bacteria has been linked to adverse pregnancy outcomes, including miscarriage and preterm birth (Menard et al. 2010, Bretelle et al. 2015). In a competitive setting, L. crispatus can outcompete F. vaginae for attachment to vaginal epithelial cells (Machado et al. 2013) and a preexisting biofilm seems to be necessary for successful F. vaginae colonization (Hardy et al. 2016, Castro et al. 2019, 2020). Fannyhessea vaginae elicits a robust epithelial proinflammatory response in cervicovaginal epithelial cells, characterized by elevated levels of IL-6, IL-8, CCL-20, and TNFα (Trama et al. 2007, Libby et al. 2008, Doerflinger et al. 2014). In conjunction with other anaerobic bacteria, F. vaginae also increases paracellular permeability in ectocervical cells (Hinderfeld et al. 2019). Taken together, F. vaginae is an important secondary colonizer that can exacerbate BV by creating an inflammatory environment that contributes to epithelial disruption and recruitment of immune cells.
Mobiluncus spp.
Mobiluncus bacteria are Gram-positive anaerobes of which several species have been associated with BV (Fredricks et al. 2005, Menard et al. 2008, Oakley et al. 2008). With respect to adherence and competition with healthy lactobacilli, it was shown that M. mulieris was able to displace L. crispatus adhered to glass slides (Machado et al. 2013) and the authors speculate that this Mobiluncus spp. may secrete soluble factors that displace lactobacilli. Contrary to G. vaginalis, M. mulieris did not form a biofilm on wells treated with polystyrene (Patterson et al. 2010), adherence to uterine cells was limited, and could be outcompeted by L. crispatus in one study (Machado et al. 2013). Mobiluncus mulieris is not cytotoxic (Patterson et al. 2010), but it has been shown to have proteolytic activity through proline aminopeptidase (Schoonmaker et al. 1991). Gardnerella vaginalis grows faster in the presence of M. mulieris, which might be mediated by the effect of Mobiluncus peptides on Gardnerella (Machado et al. 2013). These studies suggest that M. mulieris contributes to BV progression by interacting with and promoting an environment that positively affects the growth of BVAB.
Sneathia spp.
Sneathia spp., such as S. amnii, are often found in BV and have been associated with preeclampsia, spontaneous abortion, postpartum bacteremia, as well as with sexually transmitted diseases and cervical cancer (Shukla et al. 2002, Fredricks et al. 2007, Srinivasan and Fredricks 2008, Ling et al. 2010, Nawrot et al. 2010). Sneathia spp. have been isolated from amniotic fluid and its presence can lead to inflammation and cause pregnancy complications (DiGiulio et al. 2008, Han et al. 2009). In a 3D cervical cell model, infection with S. amnii was associated with the presence of clue cells (Łaniewski and Herbst-Kralovetz 2021). It was also reported that S. amnii can damage the fetal membranes, and is possibly related to the expression of the cytopathogenic toxin CptA (Gentile et al. 2020). Like P. bivia, S. amnii produces polyamines that are linked to altering the pH and malodor, which are associated with BV (Łaniewski and Herbst-Kralovetz 2021).
Sneathia amnii can adhere to ME-180 human cervical cancer cells and cause cell cytotoxicity, but does not form a biofilm (Harwich et al. 2012). Adhesion to and damage of the epithelium could be mediated by a putative adhesin, multiple invasins, a fibronectin-binding protein, and the hemolysin CptA that are encoded in the S. amnii genome (Harwich et al. 2012). Sneathia amnii infection leads to secretion of proinflammatory cytokines, increases oxidative stress, and alters cellular metabolism by cervicovaginal epithelial cell models (Anahtar et al. 2015, Łaniewski and Herbst-Kralovetz 2021). With respect to nutrient utilization, S. amnii can degrade glycogen and encodes a potential O-sialoglycoprotein endopeptidase that could be involved in the degradation of sialylated proteins (Harwich et al. 2012). This bacterial species also produces β-glucuronidases that degrade glycosaminoglycans found in the vaginal tissue (Collins et al. 2001), and high levels of these enzymes have been associated with aerobic vaginitis (Wang et al. 2016). Overall, Sneathia spp. harbor multiple virulence traits that allow these bacteria to spread within infected tissues in the female genital tract (FRT) and contribute to severe adverse effects of BV.
Megasphaera spp.
Megasphaera spp., that can be subdivided in phylotype 1 (MP1) and phylotype 2 (MP2), are another type of bacteria associated with BV and pregnancy complications. Megasphaera spp. have been used in combination with other bacteria for molecular diagnosis of BV (Zozaya-Hinchliffe et al. 2008, Fethers et al. 2012, Datcu et al. 2014, Lennard et al. 2017). Both MP1 and MP2 were linked to elevated vaginal pH (Glascock et al. 2021). Women with high prevalence of MP1 and MP2 have an increased risk for HIV acquisition (McClelland et al. 2018, Sabo et al. 2020). MP2 was also associated with the sexually transmitted parasite trichomoniasis, more so than MP1 (Martin et al. 2013, Glascock et al. 2021). A study found MP1 in samples collected from the upper genital tract of women undergoing hysterectomy (Mitchell et al. 2015), suggesting that MP1 may be capable of ascending from the vagina. In contrast to the BV-associated G. vaginalis, both M. elsdenii and H. timonensis induce dendritic cell activation and proinflammatory cytokines (van Teijlingen et al. 2020), suggesting that they might be involved in inducing inflammation. The potential involvement of Megasphaera spp. in epithelial cytotoxicity and mucus degradation has not yet been explored.
BVAB
BVAB 1, 2, and 3 are vaginal bacteria in the Clostridiales order that have recently been identified and linked to BV (Fredricks et al. 2005, Marrazzo et al. 2008, Mitchell et al. 2009). The presence of BVAB 2 was associated with elevated vaginal pH, a hallmark of BV. BVAB bacteria seem to be linked to increased HIV-1 shedding (Mitchell et al. 2009), HPV prevalence, and progression of cervical intraepithelial neoplasia (Mitchell et al. 2009, Naidoo et al. 2022). More studies are needed to investigate the virulence traits of BVAB and unravel how these bacteria contribute to BV.
Effects of healthy status and BV on viral infections
The FRT is exposed to different (sexually transmitted) viral infections, including HIV-1, HPV, and HSV-2. BV patients are more susceptible to these infections. In this section, we describe how the composition of the vaginal microbiota and the BV-status affect the barrier functions of the CVM, the epithelial glycocalyx, epithelial cells and their connecting tight junctions (TJs) and interactions with immune cells that together determine the outcome of viral–host interactions (Fig. 1).
Figure 1.
Bacterial interactions with the vaginal mucosal epithelium in health and disease. Schematic representation of the vaginal epithelium in a healthy state (left) and during BV with HIV-1 coinfection (right). The figure illustrates differences in microbiota composition, epithelial structure, and the presence of immune cells. Figure design by Soledad Cook-Ordonez.
Degradation of protective mucus affects virus infection
The vaginal mucosa is covered with a soluble mucus layer that mediates interactions with bacteria and viruses. A dense mucus “plug” is present in the cervix that limits access of microbes to the uterus. CVM mostly originates from the cervix and, together with antimicrobial compounds, protects the epithelium against chemical and biological agents (Lacroix et al. 2020). The CVM has been demonstrated to reduce the mobility of HIV-1 virions, particularly under acidic conditions (pH ∼4) (Nunn et al. 2015). This effect might be mediated by d-lactic acid produced by lactobacilli, which neutralizes the negative charge on the surface of HIV-1 virions, consequently increasing nonspecific interactions between the viral membrane and mucins (Lai et al. 2009, Nunn et al. 2015, Hoang et al. 2020). In BV, the mucosal defense system is compromised by the presence of elevated concentrations of mucin-degrading enzymes, such as sialidases, galactosidases, and sulfatases (Howe et al. 1999, Olmsted et al. 2003, Cauci et al. 2005, Moncla et al. 2015, 2016). CVM isolated from women with BV is thinner and has reduced viscosity, and the mobility of HIV-1 virions is increased in this diseased mucus (Hoang et al. 2020). The mucin-degrading ability of vaginal anaerobic bacteria is, therefore, an important virulence factor that reduces the ability of the CVM to trap viral particles.
Compromising epithelial barrier function
Once the mucus layer is compromised, bacterial and viral particles come into closer contact with the vaginal epithelium. The vaginal glycocalyx is the carbohydrate-rich layer that coats the surfaces of vaginal epithelial cells and is formed by transmembrane mucins, such as MUC1 and MUC4 (Gipson et al. 1997, Hjelm et al. 2010). This glycocalyx interacts directly with vaginal bacteria and provides attachment sites for commensal lactobacilli and an additional protective layer for pathogens (Agarwal et al. 2022). Bacteria glycosylases secreted by BV-bacteria, such as sialidases and fucosidases, also degrade and modify the glycocalyx. Viral particles can traverse the epithelium via transcytosis, a process involving vesicular trafficking, the paracellular route that occurs through intercellular spaces, or direct infection of the epithelium and the release of novel virions. Studies have demonstrated that HIV-1 virions can undergo transcytosis through cervicovaginal and uterine epithelial cells (Asin et al. 2003, Gupta et al. 2013, Micsenyi et al. 2013), leading to the subsequent transmission of viral particles to CD4+ T cells (Asin et al. 2003, Stoddard et al. 2007, Micsenyi et al. 2013). BVAB such as G. vaginalis and S. amnii express the toxins vaginolysin and CptA, which induce cytotoxicity and release of cells from the epithelium. These so-called “clue cells” are coated with bacteria and used for BV diagnosis in the clinic. Cytotoxicity and clue cell formation contribute to tissue disruption (Cook et al. 1989), which could facilitate viral access deeper into the mucosa.
TJs, composed of transmembrane proteins, such as occludin, claudins, and zonula occludens, seal the intracellular space between neighboring epithelial cells. When intact, TJs form a selective barrier that restricts the passage of viral particles across the cervicovaginal and endocervical epithelial layers (Blaskewicz et al. 2011, Carias et al. 2013). However, under inflammatory conditions, such as bacterial and/or viral infections, TJ complexes become more open, allowing the migration of immune cells and resulting in increased permeability of the tissue (Tugizov 2021, Citi et al. 2024). Interestingly, immune cells such as dendritic cells and Langerhans cells, express TJ proteins that may allow them to open the paracellular space in a zipper-like manner and migrate through the epithelium (Rescigno et al. 2001, Ichiyasu et al. 2004, Zimmerli and Hauser 2007, Kubo et al. 2009). Anaerobic BVAB can modulate TJs and compromise the integrity of the vaginal epithelium through the production of cytotoxins (Patterson et al. 2010, Gilbert et al. 2019, Anton et al. 2022). They also induce the production of proinflammatory cytokines that alter TJ proteins, leading to epithelial cell damage and exfoliation (Hedges et al. 2006, Capaldo et al. 2014, Plesniarski et al. 2021). For intestinal pathogens such as Clostridium perfringens and Campylobacter jejuni, direct targeting of TJs with toxins and proteases has been described (Freedman et al. 2016) (Sharafutdinov et al. 2024). Whether BVAB also target TJs is currently not reported and remains to be investigated. Overall, a compromised epithelial barrier leads to enhanced contact of Langerhans cells and DCs with bacteria and/or viruses, thereby increasing the chance of successful viral infection and transmission.
Roles of different immune cells in HIV-1 infection
The repertoire of immune cells present in the mucosa plays an essential role in the potential success of incoming viruses. Langerhans cells and CD4+ T cells are the initial immune cells that encounter HIV-1 in the lower genital tract (Hladik et al. 2007). Langerhans cells, predominantly present in the stratified squamous nonkeratinized epithelium, efficiently capture and internalize HIV-1 (Valladeau et al. 2000, Turville et al. 2002, Hladik et al. 2007). Once internalized, HIV-1 is targeted into specialized vesicles called Birbeck granules that are unique to Langerhans cells, where the virus is degraded via autophagy, preventing HIV-1 infection and transmission to susceptible CD4 + T cells (de Witte et al. 2007a, Ribeiro et al. 2016). Through this mechanism, Langerhans cells are protective against HIV-1 infection and prevent transmission to CD4 + T cells. Dendritic cells (DCs) are also present in the submucosal lamina propria layer and can detect and respond to HIV-1. DCs can capture HIV-1, but in contrast to Langerhans cells, do not protect against HIV-1 infection (Geijtenbeek et al. 2000a, b, Kwon et al. 2002, Shen et al. 2014). DCs that have captured and internalized HIV-1 can migrate to draining lymph nodes and transmit the virus to CD4 + T cells, thereby facilitating viral spread (Geijtenbeek et al. 2000a, Cameron et al. 1992, Weissman et al. 1995, Granelli-Piperno et al. 1999, Kwon et al. 2002, Shen et al. 2014). Macrophages that are present in submucosal vaginal tissue are also permissive for HIV-1 infection (Shen et al. 2009, 2011). Overall, recent studies suggest that during BV, the repertoire of immune cells present in the vaginal lamina propria is altered, which contributes to enhanced susceptibility to viral infections such as HIV-1.
Proinflammatory immune activation and recruitment of immune cells
In addition to directly compromising epithelial barrier functions, the inflammation induced by BV-associated anaerobic bacteria can promote the production of proinflammatory cytokines and immune cell recruitment (Bleul et al. 1997, Hladik et al. 1999, McKinnon et al. 2011). Several BVAB, such as G. vaginalis, F. vaginae, S. amnii, and M. mulieris induce inflammation and lead to an increased proinflammatory response (Libby et al. 2008, Doerflinger et al. 2014, McKenzie et al. 2021, Łaniewski and Herbst-Kralovetz 2021, Anton et al. 2022). Proinflammatory responses are required to attenuate BV but also affect susceptibility to viral infections by attracting and activating target immune cells, as well as altering the function of immune cells. Under noninflammatory conditions, immature Langerhans cells inhibit HIV-1 infection and dissemination by capturing and degrading virions (de Witte et al. 2007a, Valladeau et al. 2000). Under inflammatory conditions, the antiviral capabilities of activated Langerhans cells are compromised, making them susceptible to HIV-1 infection and transmission to target cells (de Witte et al. 2007a, De Jong and Geijtenbeek 2009). BV is also linked to increased levels of proinflammatory cytokines that can lead to the recruitment and activation of immune cells including CD4 + T cells into vaginal tissue. The influx of T cells into the vaginal lamina propria results in enhanced susceptibility to HIV-1 due to proximity to the virus (Arnold et al. 2016). Furthermore, the number of activated target CD4 + T cells in genital mucosa is enhanced in germ-free mice following intravaginal colonization with BV-associated P. bivia (Gosmann et al. 2017). In addition, when DCs are exposed to BVAB and cocultured with T cells, H. timonensis skews CD4 + T cells toward a T helper 1 (Th1) response (van Teijlingen et al. 2020). A strong Th1 response can lead to uncontrolled inflammation and is typically associated with preterm birth (van Teijlingen et al. 2020). Overall, BVAB promote inflammation, compromise epithelial barrier functions, and alter immune responses, consequently increasing susceptibility to viral infections.
Direct effects of BVAB on viral uptake by immune cells
Recently, we have discovered that BVAB have a direct impact on viral susceptibility by immune cells, specifically for HIV-1. Interestingly, H. timonensis enhances HIV-1 uptake by vaginal Langerhans cells, where the virions are protected from autophagy-mediated degradation and remain infectious for several days (Van Teijlingen et al. 2022). Furthermore, the enhanced viral uptake by Langerhans cells leads to enhanced transmission to susceptible HIV-1 target cells (Van Teijlingen et al. 2022). Moreover, exposure to H. timonensis also enhances HIV-1 uptake by DCs and CD4 + T cells (van Teijlingen et al. 2024, van Smoorenburg et al. 2025). In primary DCs, the increase in uptake results in enhanced transmission to target cells (van Smoorenburg et al. 2025). In vaginal CD4 + T cells, the H. timonensis-induced uptake results in productive infection of CD4 + T cells (van Teijlingen et al. 2024). This demonstrates that in addition to the indirect effects described above, a BV-associated bacterium can also directly affect the viral susceptibility of immune cells. The observation that multiple immune cells are affected by H. timonensis indicates that the underlying mechanism of enhanced uptake may not be restricted to a specific cellular receptor. Which specific virulence factor or property of H. timonensis is responsible for the enhanced viral uptake remains to be identified.
BVAB affect HPV and HSV-2 susceptibility
Besides enhancing susceptibility to HIV-1, BVAB have also been linked to an increased risk of acquiring the STIs, HPV, and HSV-2 (Cherpes et al. 2005, Kaul et al. 2007, Nagot et al. 2007, Gillet et al. 2011, Moscicki et al. 2012, Łaniewski et al. 2018). An increase in vaginal anaerobic bacterial species is observed in HPV-infected women (Shannon et al. 2017). Furthermore, HPV infection has been significantly associated with the presence of F. vaginae (Di Paola et al. 2017) and BVAB bacteria (Naidoo et al. 2022). In addition, sialidases produced by G. vaginalis have been implicated in HPV persistence in cervical infections (Di Paola et al. 2017, Novak et al. 2023), although underlying molecular mechanisms remain to be investigated. Women who acquired an HPV infection show decreased numbers of Langerhans cells in the FRT (Jimenez-Flores et al. 2006), but women who in addition cleared the HPV infection, show increased numbers of endocervical LCs (Shannon et al. 2017). Both Langerhans cells and DCs interact with HPV and can facilitate viral spread (de Witte et al. 2007b, Yan et al. 2004, Bousarghin et al. 2005). In addition, CD4 + T cells can also interact with HPV (Williams et al. 2002), although no recruitment of CD4 + T cells is observed in HPV-infected women (Shannon et al. 2017).
In contrast to HPV, an increased number of activated mucosal CD4 + T cells as well as an enhanced number of cervical DCs are observed in the FRT during an HSV-2 infection (Rebbapragada et al. 2007, Shannon et al. 2014). Furthermore, an HSV-2 infection enhances the risk of acquiring an HIV-1 infection (Wald and Link 2002), which could in part be explained by HSV-2 affecting the antiviral properties of LCs as well as by the enhanced presence of HIV-1 target cells (Rebbapragada et al. 2007, de Jong et al. 2010). Altogether, different immune cells in the vaginal tissue play an essential role in establishing homeostasis and providing an additional line of defense against viral infections. Although more research is required, it is plausible that the same mechanisms by which BVAB enhance the risk of HIV-1 infection may apply to HPV or HSV-2 infections. These mechanisms could include decreased protective effects from Lactobacillus-dominated vaginal microbiota, changes in mucus levels, enhanced epithelial permeability, induction of a proinflammatory response with enhanced immune cell recruitment, and direct immune modulation by bacteria.
Concluding remarks
BV is a polymicrobial infection that significantly impacts women’s quality of life and reproductive health. BV is associated with an increased risk of acquiring STIs, including HIV-1, HPV, and HSV-2. Traditionally, research has focused on G. vaginalis, due to its numerous virulence factors and high prevalence in women with BV. However, BV is a polymicrobial condition, and species within the genera Hoylesella, Prevotella, Fannyhessea, Mobiluncus, and Sneathia also exhibit important virulence traits. Bacterial glycosidases, adhesion, the proinflammatory impact on the epithelium, and recruitment of immune cells are all key factors that increase the risk of viral infection. More insight into the contributions of individual vaginal anaerobic bacteria and their synergistic effects that lead to BV and increased susceptibility to viral infections is necessary to develop new therapeutic interventions.
Contributor Information
Celia Segui-Perez, Section Infection Biology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, the Netherlands.
Marleen Y van Smoorenburg, Amsterdam UMC, location University of Amsterdam, Department of Experimental Immunology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam institute for Immunology and Infectious Diseases, Infectious Diseases, Amsterdam, the Netherlands.
Anna E Maranus, Section Infection Biology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, the Netherlands.
Teunis B H Geijtenbeek, Amsterdam UMC, location University of Amsterdam, Department of Experimental Immunology, Meibergdreef 9, Amsterdam, the Netherlands; Amsterdam institute for Immunology and Infectious Diseases, Infectious Diseases, Amsterdam, the Netherlands.
Karin Strijbis, Section Infection Biology, Division of Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Yalelaan 1, 3584 CL, Utrecht, the Netherlands.
Conflict of interest
The authors declare no conflict of interest.
Funding
This research is supported by a ZonMW TOP grant that was awarded to K. Strijbis and T. B. H. Geijtenbeek (grant number 91218017).
References
- Achilles SL, Austin MN, Meyn LA et al. Impact of contraceptive initiation on vaginal microbiota. Am J Obstet Gynecol. 2018;218:622.e1–622.e10. 10.1016/j.ajog.2018.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agarwal K, Choudhury B, Robinson LS et al. Resident microbes shape the vaginal epithelial glycan landscape. Sci Transl Med. 2022;15:eabp9599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agarwal K, Choudhury B, Robinson LS et al. Resident microbes shape the vaginal epithelial glycan landscape. Sci Transl Med. 2023;15:eabp9599. 10.1126/scitranslmed.abp9599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Agarwal K, Lewis AL. Vaginal sialoglycan foraging by Gardnerella vaginalis: mucus barriers as a meal for unwelcome guests?. Glycobiology. 2021;31:667–80. 10.1093/glycob/cwab024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Allen A, Bell A, Mantle M et al. The structure and physiology of gastrointestinal mucus. In: Chantler EN, Elder JB, Elstein M (eds), Mucus in Health and Disease—II. Boston, MA: Springer, 1982, 115–33. [DOI] [PubMed] [Google Scholar]
- Alves P, Castro J, Sousa C et al. Gardnerella vaginalis outcompetes 29 other bacterial species isolated from patients with bacterial vaginosis, using in an in vitro biofilm formation model. J Infect Dis. 2014;210:593–6. 10.1093/infdis/jiu131. [DOI] [PubMed] [Google Scholar]
- Amabebe E, Anumba DOC. The vaginal microenvironment: the physiologic role of lactobacilli. Front Med. 2018;5:181. 10.3389/fmed.2018.00181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Amsel R, Totten PA, Spiegel CA et al. Nonspecific vaginitis. Diagnostic criteria and microbial and epidemiologic associations. Am J Med. 1983;74:14–22. 10.1016/0002-9343(83)91112-9. [DOI] [PubMed] [Google Scholar]
- Anahtar MN, Byrne EH, Doherty KE et al. Cervicovaginal bacteria are a major modulator of host inflammatory responses in the female genital tract. Immunity. 2015;42:965–76. 10.1016/j.immuni.2015.04.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Anton L, Ferguson B, Friedman ES et al. Gardnerella vaginalis alters cervicovaginal epithelial cell function through microbe-specific immune responses. Microbiome. 2022;10:119. 10.1186/s40168-022-01317-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Arnold KB, Burgener A, Birse K et al. Increased levels of inflammatory cytokines in the female reproductive tract are associated with altered expression of proteases, mucosal barrier proteins, and an influx of HIV-susceptible target cells. Mucosal Immunol. 2016;9:194–205. 10.1038/mi.2015.51. [DOI] [PubMed] [Google Scholar]
- Asin SN, Wildt-Perinic D, Mason SI et al. Human Immunodeficiency Virus type 1 infection of human uterine epithelial cells: viral shedding and cell contact-mediated infectivity. J Infect Dis. 2003;187:1522–33. 10.1086/374782. [DOI] [PubMed] [Google Scholar]
- Atashili J, Poole C, Ndumbe PM et al. Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. AIDS. 2008;22:1493–501. 10.1097/QAD.0b013e3283021a37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Auriemma RS, Scairati R, del Vecchio G et al. The vaginal microbiome: a long urogenital colonization throughout woman life. Front Cell Infect Microbiol. 2021;11:686167. 10.3389/fcimb.2021.686167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Berard AR, Brubaker DK, Birse K et al. Vaginal epithelial dysfunction is mediated by the microbiome, metabolome, and mTOR signaling. Cell Rep. 2023;42:112474. 10.1016/j.celrep.2023.112474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bhandari P, Hill JE. Transport and utilization of glycogen breakdown products by Gardnerella spp. from the human vaginal microbiome. Microbiol Spectr. 2023;11:e0443522. 10.1128/spectrum.04435-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Blaskewicz CD, Pudney J, Anderson DJ. Structure and function of intercellular junctions in human cervical and vaginal mucosal epithelia. Biol Reprod. 2011;85:97–104. 10.1095/biolreprod.110.090423. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bleul CC, Wu L, Hoxie JA et al. The HIV coreceptors CXCR4 and CCR5 are differentially expressed and regulated on human T lymphocytes. Proc Natl Acad Sci USA. 1997;94:1925–30. 10.1073/pnas.94.5.1925. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Borgdorff H, Tsivtsivadze E, Verhelst R et al. Lactobacillus-dominated cervicovaginal microbiota associated with reduced HIV/STI prevalence and genital HIV viral load in African women. ISME J. 2014;8:1781–93. 10.1038/ismej.2014.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Boris S, Barbés C. Role played by lactobacilli in controlling the population of vaginal pathogens. Microbes Infect. 2000;2:543–6. 10.1016/S1286-4579(00)00313-0. [DOI] [PubMed] [Google Scholar]
- Bousarghin L, Hubert P, Franzen E et al. Human papillomavirus 16 virus-like particles use heparan sulfates to bind dendritic cells and colocalize with langerin in Langerhans cells. J Gen Virol. 2005;86:1297–305. 10.1099/vir.0.80559-0. [DOI] [PubMed] [Google Scholar]
- Bretelle F, Rozenberg P, Pascal A et al. High Atopobium vaginae and Gardnerella vaginalis vaginal loads are associated with preterm birth. Clin Infect Dis. 2015;60:860–7. 10.1093/cid/ciu966. [DOI] [PubMed] [Google Scholar]
- Briselden AM, Moncla BJ, Stevens CE et al. Sialidases (neuraminidases) in bacterial vaginosis and bacterial vaginosis-associated microflora. J Clin Microbiol. 1992;30:663–6. 10.1128/jcm.30.3.663-666.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brotman RM, Shardell MD, Gajer P et al. Interplay between the temporal dynamics of the vaginal microbiota and human papillomavirus detection. J Infect Dis. 2014;210:1723–33. 10.1093/infdis/jiu330. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Burton JP, Devillard E, Cadieux PA et al. Detection of Atopobium vaginae in postmenopausal women by cultivation-independent methods warrants further investigation. J Clin Microbiol. 2004;42:1829–31. 10.1128/JCM.42.4.1829-1831.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cameron PU, Freudenthal PS, Barker JM et al. Dendritic cells exposed to Human Immunodeficiency Virus type-1 transmit a vigorous cytopathic infection to CD4+ T cells. Science. 1992;257:383–7. 10.1126/science.1352913. [DOI] [PubMed] [Google Scholar]
- Capaldo CT, Farkas AE, Hilgarth RS et al. Proinflammatory cytokine-induced tight junction remodeling through dynamic self-assembly of claudins. MBoC. 2014;25:2710–9. 10.1091/mbc.e14-02-0773. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Carias AM, McCoombe S, McRaven M et al. Defining the interaction of HIV-1 with the mucosal barriers of the female reproductive tract. J Virol. 2013;87:11388–400. 10.1128/JVI.01377-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castro J, Alves P, Sousa C et al. Using an in-vitro biofilm model to assess the virulence potential of bacterial vaginosis or non-bacterial vaginosis Gardnerella vaginalis isolates. Sci Rep. 2015;5:11640. 10.1038/srep11640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castro J, Machado D, Cerca N. Unveiling the role of Gardnerella vaginalis in polymicrobial bacterial vaginosis biofilms: the impact of other vaginal pathogens living as neighbors. ISME J. 2019;13:1306–17. 10.1038/s41396-018-0337-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castro J, Rosca AS, Cools P et al. Gardnerella vaginalis enhances Atopobium vaginae viability in an in vitro model. Front Cell Infect Microbiol. 2020;10:83. 10.3389/fcimb.2020.00083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castro J, Rosca AS, Muzny CA et al. Atopobium vaginae and Prevotella bivia are able to incorporate and influence gene expression in a pre-formed Gardnerella vaginalis biofilm. Pathogens. 2021;10:247. 10.3390/pathogens10020247. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cauci S, McGregor J, Thorsen P et al. Combination of vaginal pH with vaginal sialidase and prolidase activities for prediction of low birth weight and preterm birth. Am J Obstet Gynecol. 2005;192:489–96. 10.1016/j.ajog.2004.07.023. [DOI] [PubMed] [Google Scholar]
- Chee WJY, Chew SY, Than LTL. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb Cell Fact. 2020;19:203. 10.1186/s12934-020-01464-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cherpes TL, Melan MA, Kant JA et al. Genital tract shedding of herpes simplex virus type 2 in women: effects of hormonal contraception, bacterial vaginosis, and vaginal group B Streptococcus colonization. Clin Infect Dis. 2005;40:1422–8. 10.1086/429622. [DOI] [PubMed] [Google Scholar]
- Citi S, Fromm M, Furuse M et al. A short guide to the tight junction. J Cell Sci. 2024;137:jcs261776. 10.1242/jcs.261776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Coleman JS, Gaydos CA. Molecular diagnosis of bacterial vaginosis: an update. J Clin Microbiol. 2018;56:e00342–18. 10.1128/JCM.00342-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Collins MD, Hoyles L, Tornqvist E et al. Characterization of some strains from human clinical sources which resemble “Leptotrichia sanguinegens”: description of Sneathia sanguinegens sp. nov., gen. nov. Syst Appl Microbiol. 2001;24:358–61. 10.1078/0723-2020-00047. [DOI] [PubMed] [Google Scholar]
- Cook RL, Reid G, Pond DG et al. Clue cells in bacterial vaginosis: immunofluorescent identification of the adherent Gram-negative bacteria as Gardnerella vaginalis. J Infect Dis. 1989;160:490–6. 10.1093/infdis/160.3.490. [DOI] [PubMed] [Google Scholar]
- Datcu R, Gesink D, Mulvad G et al. Bacterial vaginosis diagnosed by analysis of first-void-urine specimens. J Clin Microbiol. 2014;52:218–25. 10.1128/JCM.02347-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Backer E, Dubreuil L, Brauman M et al. In vitro activity of secnidazole against Atopobium vaginae, an anaerobic pathogen involved in bacterial vaginosis. Clin Microbiol Infect. 2010;16:470–2. 10.1111/j.1469-0691.2009.02852.x. [DOI] [PubMed] [Google Scholar]
- De Backer E, Verhelst R, Verstraelen H et al. Quantitative determination by real-time PCR of four vaginal Lactobacillus species, Gardnerella vaginalis and Atopobium vaginae indicates an inverse relationship between L. gasseri and L. iners. BMC Microbiol. 2007;7:115. 10.1186/1471-2180-7-115. [DOI] [PMC free article] [PubMed] [Google Scholar]
- de Jong MA, de Witte L, Taylor ME et al. Herpes simplex virus type 2 enhances HIV-1 susceptibility by affecting Langerhans cell function. J Immunol. 2010;185:1633–41. 10.4049/jimmunol.0904137. [DOI] [PubMed] [Google Scholar]
- De Jong M a. WP, Geijtenbeek TBH. Human Immunodeficiency Virus-1 acquisition in genital mucosa: Langerhans cells as key-players. J Intern Med. 2009;265:18–28. 10.1111/j.1365-2796.2008.02046.x. [DOI] [PubMed] [Google Scholar]
- de Witte L, Nabatov A, Pion M et al. Langerin is a natural barrier to HIV-1 transmission by Langerhans cells. Nat Med. 2007a;13:367–71. 10.1038/nm1541. [DOI] [PubMed] [Google Scholar]
- de Witte L, Zoughlami Y, Aengeneyndt B et al. Binding of human papilloma virus L1 virus-like particles to dendritic cells is mediated through heparan sulfates and induces immune activation. Immunobiology. 2007b;212:679–91. 10.1016/j.imbio.2007.09.006. [DOI] [PubMed] [Google Scholar]
- DiGiulio DB, Romero R, Amogan HP et al. Microbial prevalence, diversity and abundance in amniotic fluid during preterm labor: a molecular and culture-based investigation. PLoS One. 2008;3:e3056. 10.1371/journal.pone.0003056. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dingens AS, Fairfortune TS, Reed S et al. Bacterial vaginosis and adverse outcomes among full-term infants: a cohort study. BMC Pregnancy Childbirth. 2016;16:278. 10.1186/s12884-016-1073-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Di Paola M, Sani C, Clemente AM et al. Characterization of cervico-vaginal microbiota in women developing persistent high-risk human papillomavirus infection. Sci Rep. 2017;7:10200. 10.1038/s41598-017-09842-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Doerflinger SY, Throop AL, Herbst-Kralovetz MM. Bacteria in the vaginal microbiome alter the innate immune response and barrier properties of the human vaginal epithelia in a species-specific manner. J Infect Dis. 2014;209:1989–99. 10.1093/infdis/jiu004. [DOI] [PubMed] [Google Scholar]
- Doust RH, Mobarez AM. Collagenase activity in Prevotella bivius isolated from patients with premature rupture of membranes. Med J Islamic Repub Iran. 2004;18:61–6. [Google Scholar]
- Edelman SM, Lehti TA, Kainulainen V et al. Identification of a high-molecular-mass Lactobacillus epithelium adhesin (LEA) of Lactobacillus crispatus ST1 that binds to stratified squamous epithelium. Microbiology. 2012;158:1713–22. 10.1099/mic.0.057216-0. [DOI] [PubMed] [Google Scholar]
- Farage MA, Miller KW, Sobel JD. Dynamics of the vaginal ecosystem—hormonal influences. Infect Dis. 2010;3:IDRT.S3903. 10.4137/IDRT.S3903. [DOI] [Google Scholar]
- Ferris MJ, Masztal A, Aldridge KE et al. Association of Atopobium vaginae, a recently described metronidazole resistant anaerobe, with bacterial vaginosis. BMC Infect Dis. 2004;4:5. 10.1186/1471-2334-4-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fethers K, Twin J, Fairley CK et al. Bacterial vaginosis (BV) candidate bacteria: associations with BV and behavioural practices in sexually-experienced and inexperienced women. PLoS One. 2012;7:e30633. 10.1371/journal.pone.0030633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fredricks DN, Fiedler TL, Marrazzo JM. Molecular identification of bacteria associated with bacterial vaginosis. N Engl J Med. 2005;353:1899–911. 10.1056/NEJMoa043802. [DOI] [PubMed] [Google Scholar]
- Fredricks DN, Fiedler TL, Thomas KK et al. Targeted PCR for detection of vaginal bacteria associated with bacterial vaginosis. J Clin Microbiol. 2007;45:3270–6. 10.1128/JCM.01272-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fredricks DN, Marrazzo JM. Molecular methodology in determining vaginal flora in health and disease: its time has come. Curr Infect Dis Rep. 2005;7:463–70. 10.1007/s11908-005-0049-2. [DOI] [PubMed] [Google Scholar]
- Freedman JC, Shrestha A, McClane BA. Clostridium perfringens enterotoxin: action, genetics, and translational applications. Toxins. 2016;8:73. 10.3390/toxins8030073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gao J, Peng Y, Jiang N et al. High-throughput sequencing-based analysis of changes in the vaginal microbiome during the disease course of patients with bacterial vaginosis: a case-control study. Biology. 2022;11:1797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Geijtenbeek TB, Kwon DS, Torensma R et al. DC-SIGN, a dendritic cell-specific HIV-1-binding protein that enhances trans-infection of T cells. Cell. 2000a;100:587–97. 10.1016/S0092-8674(00)80694-7. [DOI] [PubMed] [Google Scholar]
- Geijtenbeek TB, Torensma R, van Vliet SJ et al. Identification of DC-SIGN, a novel dendritic cell-specific ICAM-3 receptor that supports primary immune responses. Cell. 2000b;100:575–85. 10.1016/S0092-8674(00)80693-5. [DOI] [PubMed] [Google Scholar]
- Gelber SE, Aguilar JL, Lewis KLT et al. Functional and phylogenetic characterization of vaginolysin, the human-specific cytolysin from Gardnerella vaginalis. J Bacteriol. 2008;190:3896–903. 10.1128/JB.01965-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gentile GL, Rupert AS, Carrasco LI et al. Identification of a cytopathogenic toxin from Sneathia amnii. J Bacteriol. 2020;202:e00162–20. 10.1128/JB.00162-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gilbert NM, Lewis WG, Li G et al. Gardnerella vaginalis and Prevotella bivia trigger distinct and overlapping phenotypes in a mouse model of bacterial vaginosis. J Infect Dis. 2019;220:1099–108. 10.1093/infdis/jiy704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gillet E, Meys JF, Verstraelen H et al. Bacterial vaginosis is associated with uterine cervical human papillomavirus infection: a meta-analysis. BMC Infect Dis. 2011;11:10. 10.1186/1471-2334-11-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gipson IK, Ho SB, Spurr-Michaud SJ et al. Mucin genes expressed by human female reproductive tract epithelia. Biol Reprod. 1997;56:999–1011., 10.1095/biolreprod56.4.999. [DOI] [PubMed] [Google Scholar]
- Glascock AL, Jimenez NR, Boundy S et al. Unique roles of vaginal megasphaera phylotypes in reproductive health. Microb Genom. 2021;7:000526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gosmann C, Anahtar MN, Handley SA et al. Lactobacillus-deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African women. Immunity. 2017;46:29–37. 10.1016/j.immuni.2016.12.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Govinden G, Parker JL, Naylor KL et al. Inhibition of sialidase activity and cellular invasion by the bacterial vaginosis pathogen Gardnerella vaginalis. Arch Microbiol. 2018;200:1129–33. 10.1007/s00203-018-1520-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Granelli-Piperno A, Finkel V, Delgado E et al. Virus replication begins in dendritic cells during the transmission of HIV-1 from mature dendritic cells to T cells. Curr Biol. 1999;9:21–9. 10.1016/S0960-9822(99)80043-8. [DOI] [PubMed] [Google Scholar]
- Gupta S, Gach JS, Becerra JC et al. The neonatal Fc receptor (FcRn) enhances Human Immunodeficiency Virus type 1 (HIV-1) transcytosis across epithelial cells. PLoS Pathog. 2013;9:e1003776. 10.1371/journal.ppat.1003776. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Han YW, Shen T, Chung P et al. Uncultivated bacteria as etiologic agents of intra-amniotic inflammation leading to preterm birth. J Clin Microbiol. 2009;47:38–47. 10.1128/JCM.01206-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hardy L, Jespers V, Abdellati S et al. A fruitful alliance: the synergy between Atopobium vaginae and Gardnerella vaginalis in bacterial vaginosis-associated biofilm. Sex Transm Infect. 2016;92:487–91. 10.1136/sextrans-2015-052475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hardy L, Jespers V, Van Den Bulck M et al. The presence of the putative Gardnerella vaginalis sialidase A gene in vaginal specimens is associated with bacterial vaginosis biofilm. PLoS One. 2017;12:e0172522. 10.1371/journal.pone.0172522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Harwich MD, Serrano MG, Fettweis JM et al. Genomic sequence analysis and characterization of Sneathia amnii sp. nov. BMC Genomics. 2012;13:S4. 10.1186/1471-2164-13-S8-S4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- He Y, Niu X, Wang B et al. Evaluation of the inhibitory effects of Lactobacillus gasseri and Lactobacillus crispatus on the adhesion of seven common lower genital tract infection-causing pathogens to vaginal epithelial cells. Front Med. 2020;7:284. 10.3389/fmed.2020.00284. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hedges SR, Barrientes F, Desmond RA et al. Local and systemic cytokine levels in relation to changes in vaginal flora. J Infect Dis. 2006;193:556–62. 10.1086/499824. [DOI] [PubMed] [Google Scholar]
- Hinderfeld AS, Phukan N, Bär A-K et al. Cooperative interactions between Trichomonas vaginalis and associated bacteria enhance paracellular permeability of the cervicovaginal epithelium by dysregulating tight junctions. Infect Immun. 2019;87:e00141–19. 10.1128/IAI.00141-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hitch TCA, Bisdorf K, Afrizal A et al. A taxonomic note on the genus Prevotella: description of four novel genera and emended description of the genera Hallella and Xylanibacter. Syst Appl Microbiol. 2022;45:126354. 10.1016/j.syapm.2022.126354. [DOI] [PubMed] [Google Scholar]
- Hjelm BE, Berta AN, Nickerson CA et al. Development and characterization of a three-dimensional organotypic human vaginal epithelial cell Model1. Biol Reprod. 2010;82:617–27. 10.1095/biolreprod.109.080408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hladik F, Lentz G, Delpit E et al. Coexpression of CCR5 and IL-2 in human genital but not blood T cells: implications for the ontogeny of the CCR5+ Th1 phenotype. J Immunol. 1999;163:2306–13. 10.4049/jimmunol.163.4.2306. [DOI] [PubMed] [Google Scholar]
- Hladik F, Sakchalathorn P, Ballweber L et al. Initial events in establishing vaginal entry and infection by Human Immunodeficiency Virus type-1. Immunity. 2007;26:257–70. 10.1016/j.immuni.2007.01.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hoang T, Toler E, DeLong K et al. The cervicovaginal mucus barrier to HIV-1 is diminished in bacterial vaginosis. PLoS Pathog. 2020;16:e1008236. 10.1371/journal.ppat.1008236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Howe L, Wiggins R, Soothill PW et al. Mucinase and sialidase activity of the vaginal microflora: implications for the pathogenesis of preterm labour. Int J STD AIDS. 1999;10:442–7. 10.1258/0956462991914438. [DOI] [PubMed] [Google Scholar]
- Ichiyasu H, McCormack JM, McCarthy KM et al. Matrix metalloproteinase-9-deficient dendritic cells have impaired migration through tracheal epithelial tight junctions. Am J Respir Cell Mol Biol. 2004;30:761–70. 10.1165/rcmb.2003-0370OC. [DOI] [PubMed] [Google Scholar]
- Ilhan ZE, Łaniewski P, Tonachio A et al. Members of Prevotella genus distinctively modulate innate immune and barrier functions in a human three-dimensional endometrial epithelial cell model. J Infect Dis. 2020;222:2082–92. 10.1093/infdis/jiaa324. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jenkins DJ, Woolston BM, Hood-Pishchany MI et al. Bacterial amylases enable glycogen degradation by the vaginal microbiome. Nat Microbiol. 2023;8:1641–52. 10.1038/s41564-023-01447-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jimenez-Flores R, Mendez-Cruz R, Ojeda-Ortiz J et al. High-risk human papilloma virus infection decreases the frequency of dendritic Langerhans’ cells in the human female genital tract. Immunology. 2006;117:220–8. 10.1111/j.1365-2567.2005.02282.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kaul R, Nagelkerke NJ, Kimani J et al. Prevalent herpes simplex virus type 2 infection is associated with altered vaginal flora and an increased susceptibility to multiple sexually transmitted infections. J Infect Dis. 2007;196:1692–7. 10.1086/522006. [DOI] [PubMed] [Google Scholar]
- Kubo A, Nagao K, Yokouchi M et al. External antigen uptake by Langerhans cells with reorganization of epidermal tight junction barriers. J Exp Med. 2009;206:2937–46. 10.1084/jem.20091527. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kwon DS, Gregorio G, Bitton N et al. DC-SIGN-mediated internalization of HIV is required for trans-enhancement of T cell infection. Immunity. 2002;16:135–44. 10.1016/S1074-7613(02)00259-5. [DOI] [PubMed] [Google Scholar]
- Kyongo JK, Crucitti T, Menten J et al. Cross-sectional analysis of selected genital tract immunological markers and molecular vaginal microbiota in sub-Saharan African women, with relevance to HIV risk and prevention. Clin Vaccine Immunol. 2015;22:526–38. 10.1128/CVI.00762-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lacroix G, Gouyer V, Gottrand F et al. The cervicovaginal mucus barrier. Int J Mol Sci. 2020;21:8266. 10.3390/ijms21218266. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lai SK, Wang Y-Y, Hanes J. Mucus-penetrating nanoparticles for drug and gene delivery to mucosal tissues. Adv Drug Deliv Rev. 2009;61:158–71. 10.1016/j.addr.2008.11.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Łaniewski P, Barnes D, Goulder A et al. Linking cervicovaginal immune signatures, HPV and microbiota composition in cervical carcinogenesis in non-Hispanic and Hispanic women. Sci Rep. 2018;8:7593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Łaniewski P, Herbst-Kralovetz MM. Bacterial vaginosis and health-associated bacteria modulate the immunometabolic landscape in 3D model of human cervix. npj Biofilms Microbiomes. 2021;7:1–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Laniewski P, Owen KA, Brotman RM et al. Clinical and personal lubricants impact growth of vaginal Lactobacillus species and colonization of vaginal epithelial cells. Am J Obstetr Gynecol. 2019;221:694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Leccese Terraf MC, Mendoza LM, Juárez Tomás MS et al. Phenotypic surface properties (aggregation, adhesion and biofilm formation) and presence of related genes in beneficial vaginal lactobacilli. J Appl Microbiol. 2014;117:1761–72. 10.1111/jam.12642. [DOI] [PubMed] [Google Scholar]
- Lennard K, Dabee S, Barnabas SL et al. Microbial composition predicts genital tract inflammation and persistent bacterial vaginosis in South African adolescent females. Infect Immun. 2017;86:e00410–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis WG, Robinson LS, Gilbert NM et al. Degradation, foraging, and depletion of mucus sialoglycans by the vagina-adapted actinobacterium Gardnerella vaginalis. J Biol Chem. 2013;288:12067–79. 10.1074/jbc.M113.453654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lewis WG, Robinson LS, Perry J et al. Hydrolysis of secreted sialoglycoprotein immunoglobulin A (IgA) in ex vivo and biochemical models of bacterial vaginosis. J Biol Chem. 2012;287:2079–89. 10.1074/jbc.M111.278135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li Y, Yu T, Yan H et al. Vaginal microbiota and HPV infection: novel mechanistic insights and therapeutic strategies. Infect Drug Resist. 2020;13:1213–20. 10.2147/IDR.S210615. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Libby EK, Pascal KE, Mordechai E et al. Atopobium vaginae triggers an innate immune response in an in vitro model of bacterial vaginosis. Microbes Infect. 2008;10:439–46. 10.1016/j.micinf.2008.01.004. [DOI] [PubMed] [Google Scholar]
- Ling Z, Kong J, Liu F et al. Molecular analysis of the diversity of vaginal microbiota associated with bacterial vaginosis. BMC Genomics. 2010;11:488. 10.1186/1471-2164-11-488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ma Z (Sam). Microbiome transmission during sexual intercourse appears stochastic and supports the red Queen hypothesis. Front Microbiol. 2022;12:789983. 10.3389/fmicb.2021.789983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Machado A, Jefferson KK, Cerca N. Interactions between Lactobacillus crispatus and bacterial vaginosis (BV)-associated bacterial species in initial attachment and biofilm formation. Int J Mol Sci. 2013;14:12004–12. 10.3390/ijms140612004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Machado A, Salgueiro D, Harwich M et al. Quantitative analysis of initial adhesion of bacterial vaginosis-associated anaerobes to ME-180 cells. Anaerobe. 2013;23:1–4. 10.1016/j.anaerobe.2013.07.007. [DOI] [PubMed] [Google Scholar]
- Marrazzo JM, Thomas KK, Fiedler TL et al. Relationship of specific vaginal bacteria and bacterial vaginosis treatment failure in women who have sex with women: a cohort study. Ann Intern Med. 2008;149:20–8. 10.7326/0003-4819-149-1-200807010-00006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Martin DH, Zozaya M, Lillis RA et al. Unique vaginal microbiota that includes an unknown mycoplasma-like organism is associated with Trichomonas vaginalis infection. J Infect Dis. 2013;207:1922–31. 10.1093/infdis/jit100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McClelland RS, Lingappa JR, Srinivasan S et al. Key vaginal bacteria associated with increased risk of HIV acquisition in African women: a nested case-control study. Lancet Infect Dis. 2018;18:554–64. 10.1016/S1473-3099(18)30058-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKenzie R, Maarsingh JD, Łaniewski P et al. Immunometabolic analysis of Mobiluncus mulieris and Eggerthella sp. reveals novel insights into their pathogenic contributions to the hallmarks of bacterial vaginosis. Front Cell Infect Microbiol. 2021;11. 10.3389/fcimb.2021.759697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKinnon LR, Nyanga B, Chege D et al. Characterization of a human cervical CD4+ T cell subset coexpressing multiple markers of HIV susceptibility. J Immunol. 2011;187:6032–42. 10.4049/jimmunol.1101836. [DOI] [PubMed] [Google Scholar]
- Menard J-P, Fenollar F, Henry M et al. Molecular quantification of Gardnerella vaginalis and Atopobium vaginae loads to predict bacterial vaginosis. Clin Infect Dis. 2008;47:33–43. 10.1086/588661. [DOI] [PubMed] [Google Scholar]
- Menard JP, Mazouni C, Salem-Cherif I et al. High vaginal concentrations of Atopobium vaginae and Gardnerella vaginalis in women undergoing preterm labor. Obstet Gynecol. 2010;115:134–40. 10.1097/AOG.0b013e3181c391d7. [DOI] [PubMed] [Google Scholar]
- Micsenyi AM, Zony C, Alvarez RA et al. Postintegration HIV-1 infection of cervical epithelial cells mediates contact-dependent productive infection of T cells. J Infect Dis. 2013;208:1756–67. 10.1093/infdis/jit362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitchell C, Moreira C, Fredricks D et al. Detection of fastidious vaginal bacteria in women with HIV infection and bacterial vaginosis. Infect Dis Obstet Gynecol. 2009;2009:236919. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mitchell CM, Haick A, Nkwopara E et al. Colonization of the upper genital tract by vaginal bacterial species in non-pregnant women. Am J Obstet Gynecol. 2015;212:611.e1–9. 10.1016/j.ajog.2014.11.043. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mohammadzadeh F, Dolatian M, Jorjani M et al. Diagnostic value of Amsel’s clinical criteria for diagnosis of bacterial vaginosis. Glob J Health Sci. 2015;7:8–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moncla BJ, Chappell CA, Debo BM et al. The effects of hormones and vaginal microflora on the glycome of the female genital tract: cervical-vaginal fluid. PLoS One. 2016;11:e0158687. 10.1371/journal.pone.0158687. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moncla BJ, Chappell CA, Mahal LK et al. Impact of bacterial vaginosis, as assessed by Nugent criteria and hormonal status on glycosidases and lectin binding in cervicovaginal lavage samples. PLoS One. 2015;10:e0127091. 10.1371/journal.pone.0127091. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Moscicki A-B, Schiffman M, Burchell A et al. Updating the natural history of human papillomavirus and anogenital cancers. Vaccine. 2012;30:F24–33. 10.1016/j.vaccine.2012.05.089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Muzny CA, Blanchard E, Taylor CM et al. Identification of key bacteria involved in the induction of incident bacterial vaginosis: a prospective study. J Infect Dis. 2018;218:966–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nagot N, Ouedraogo A, Defer M-C et al. Association between bacterial vaginosis and Herpes simplex virus type-2 infection: implications for HIV acquisition studies. Sex Transm Infect. 2007;83:365–8. 10.1136/sti.2007.024794. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ñahui Palomino RA, Zicari S, Vanpouille C et al. Vaginal Lactobacillus inhibits HIV-1 replication in human tissues ex vivo. Front Microbiol. 2017;8:906. 10.3389/fmicb.2017.00906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Naidoo K, Abbai N, Tinarwo P et al. BV associated bacteria specifically BVAB 1 and BVAB 3 as biomarkers for HPV risk and progression of cervical neoplasia. Infect Dis Obstet Gynecol. 2022;2022:9562937. 10.1155/2022/9562937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Navarro S, Abla H, Delgado B et al. Glycogen availability and pH variation in a medium simulating vaginal fluid influence the growth of vaginal Lactobacillus species and Gardnerella vaginalis. BMC Microbiol. 2023;23:186. 10.1186/s12866-023-02916-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nawrot R, Kamieniarz K, Malinowska M et al. The prevalence of Leptotrichia amnionii in cervical swabs of HPV positive and negative women. Eur J Gynaecol Oncol. 2010;31:425–8. [PubMed] [Google Scholar]
- Nelson DE, Dong Q, Van der Pol B et al. Bacterial communities of the coronal sulcus and distal urethra of adolescent males. PLoS One. 2012;7:e36298. 10.1371/journal.pone.0036298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nelson TM, Borgogna J-LC, Brotman RM et al. Vaginal biogenic amines: biomarkers of bacterial vaginosis or precursors to vaginal dysbiosis?. Front Physiol. 2015;6:253. 10.3389/fphys.2015.00253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Novak J, Belleti R, da Silva Pinto GV et al. Cervicovaginal Gardnerella sialidase-encoding gene in persistent human papillomavirus infection. Sci Rep. 2023;13:14266. 10.1038/s41598-023-41469-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nugent RP, Krohn MA, Hillier SL. Reliability of diagnosing bacterial vaginosis is improved by a standardized method of Gram stain interpretation. J Clin Microbiol. 1991;29:297–301. 10.1128/jcm.29.2.297-301.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nunn KL, Wang Y-Y, Harit D et al. Enhanced trapping of HIV-1 by human cervicovaginal mucus is associated with Lactobacillus crispatus-dominant microbiota. mBio. 2015;6:e01084–01015. 10.1128/mBio.01084-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Oakley BB, Fiedler TL, Marrazzo JM et al. Diversity of human vaginal bacterial communities and associations with clinically defined bacterial vaginosis. Appl Environ Microbiol. 2008;74:4898–909. 10.1128/AEM.02884-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Hanlon DE, Moench TR, Cone RA. In vaginal fluid, bacteria associated with bacterial vaginosis can be suppressed with lactic acid but not hydrogen peroxide. BMC Infect Dis. 2011;11:200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- O’Hanlon DE, Moench TR, Cone RA. Vaginal pH and microbicidal lactic acid when lactobacilli dominate the microbiota. PLoS One. 2013;8:e80074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Olmsted SS, Meyn LA, Rohan LC et al. Glycosidase and proteinase activity of anaerobic Gram-negative bacteria isolated from women with bacterial vaginosis. Sex Transm Dis. 2003;30:257–61. 10.1097/00007435-200303000-00016. [DOI] [PubMed] [Google Scholar]
- Parolin C, Croatti V, Laghi L et al. Lactobacillus biofilms influence anti-Candida activity. Front Microbiol. 2021;12:750368. 10.3389/fmicb.2021.750368. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Patterson JL, Stull-Lane A, Girerd PH et al. Analysis of adherence, biofilm formation and cytotoxicity suggests a greater virulence potential of Gardnerella vaginalis relative to other bacterial-vaginosis-associated anaerobes. Microbiology. 2010;156:392–9. 10.1099/mic.0.034280-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pelayo P, Hussain FA, Werlang CA et al. Prevotella are major contributors of sialidases in the human vaginal microbiome. Proc Natl Acad Sci. 2024;121:e2400341121. 10.1073/pnas.2400341121. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pendharkar S, Skafte-Holm A, Simsek G et al. Lactobacilli and their probiotic effects in the vagina of reproductive age women. Microorganisms. 2023;11:636. 10.3390/microorganisms11030636. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petrina MAB, Cosentino LA, Rabe LK et al. Susceptibility of bacterial vaginosis (BV)-associated bacteria to secnidazole compared to metronidazole, tinidazole and clindamycin. Anaerobe. 2017;47:115–9. 10.1016/j.anaerobe.2017.05.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plesniarski A, Siddik AB, Su R-C. The microbiome as a key regulator of female genital tract barrier function. Front Cell Infect Microbiol. 2021;11:790627. 10.3389/fcimb.2021.790627. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pybus V, Onderdonk AB. Evidence for a commensal, symbiotic relationship between Gardnerella vaginalis and Prevotella bivia involving ammonia: potential significance for bacterial vaginosis. J Infect Dis. 1997;175:406–13. 10.1093/infdis/175.2.406. [DOI] [PubMed] [Google Scholar]
- Qian Z, Zhu H, Zhao D et al. Probiotic Lactobacillus sp. strains inhibit growth, adhesion, biofilm formation, and gene expression of bacterial vaginosis-inducing Gardnerella vaginalis. Microorganisms. 2021;9:728. 10.3390/microorganisms9040728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rampersaud R, Planet PJ, Randis TM et al. Inerolysin, a cholesterol-dependent cytolysin produced by Lactobacillus iners. J Bacteriol. 2011;193:1034–41. 10.1128/JB.00694-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Randis TM, Zaklama J, LaRocca TJ et al. Vaginolysin drives epithelial ultrastructural responses to Gardnerella vaginalis. Infect Immun. 2013;81:4544–50. 10.1128/IAI.00627-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ravel J, Moreno I, Simón C. Bacterial vaginosis and its association with infertility, endometritis, and pelvic inflammatory disease. Am J Obstet Gynecol. 2021;224:251–7. 10.1016/j.ajog.2020.10.019. [DOI] [PubMed] [Google Scholar]
- Rebbapragada A, Wachihi C, Pettengell C et al. Negative mucosal synergy between Herpes simplex type 2 and HIV in the female genital tract. AIDS. 2007;21:589–98. 10.1097/QAD.0b013e328012b896. [DOI] [PubMed] [Google Scholar]
- Rescigno M, Rotta G, Valzasina B et al. Dendritic cells shuttle microbes across gut epithelial monolayers. Immunobiology. 2001;204:572–81. 10.1078/0171-2985-00094. [DOI] [PubMed] [Google Scholar]
- Ribeiro CM, Sarrami-Forooshani R, Setiawan LC et al. Receptor usage dictates HIV-1 restriction by human TRIM5alpha in dendritic cell subsets. Nature. 2016;540:448–52. 10.1038/nature20567. [DOI] [PubMed] [Google Scholar]
- Robinson LS, Schwebke J, Lewis WG et al. Identification and characterization of NanH2 and NanH3, enzymes responsible for sialidase activity in the vaginal bacterium Gardnerella vaginalis. J Biol Chem. 2019;294:5230–45. 10.1074/jbc.RA118.006221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rousseau V, Lepargneur JP, Roques C et al. Prebiotic effects of oligosaccharides on selected vaginal lactobacilli and pathogenic microorganisms. Anaerobe. 2005;11:145–53. 10.1016/j.anaerobe.2004.12.002. [DOI] [PubMed] [Google Scholar]
- Sabo MC, Lehman DA, Wang B et al. Associations between vaginal bacteria implicated in HIV acquisition risk and pro-inflammatory cytokines and chemokines. Sex Transm Infect. 2020;96:3–9. 10.1136/sextrans-2018-053949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Santos CMA, Pires MCV, Leão TL et al. Anti-inflammatory effect of two Lactobacillus strains during infection with Gardnerella vaginalis and Candida albicans in a HeLa cell culture model. Microbiology. 2018;164:349–58. 10.1099/mic.0.000608. [DOI] [PubMed] [Google Scholar]
- Schoonmaker JN, Lunt BD, Lawellin DW et al. A new proline aminopeptidase assay for diagnosis of bacterial vaginosis. Am J Obstetr Gynecol. 1991;165:737–42. [DOI] [PubMed] [Google Scholar]
- Segui-Perez C, de Jongh R, Jonkergouw RLW et al. Prevotella timonensis degrades the vaginal epithelial glycocalyx through high fucosidase and sialidase activities. mBio. 2024;15:e0069124. 10.1128/mbio.00691-24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sgibnev A, Kremleva E. Vaginal protection by H2O2-producing lactobacilli. Jundishapur J Microbiol. 2015;8:e22913. 10.5812/jjm.22913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sha BE, Zariffard MR, Wang QJ et al. Female genital-tract HIV load correlates inversely with Lactobacillus species but positively with bacterial vaginosis and Mycoplasma hominis. J Infect Dis. 2005;191:25–32. 10.1086/426394. [DOI] [PubMed] [Google Scholar]
- Shannon B, Yi TJ, Perusini S et al. Association of HPV infection and clearance with cervicovaginal immunology and the vaginal microbiota. Mucosal Immunol. 2017;10:1310–9. 10.1038/mi.2016.129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shannon B, Yi TJ, Thomas-Pavanel J et al. Impact of asymptomatic Herpes Simplex Virus Type 2 infection on mucosal homing and immune cell subsets in the blood and female genital tract. J Immunol. 2014;192:5074–82. 10.4049/jimmunol.1302916. [DOI] [PubMed] [Google Scholar]
- Sharafutdinov I, Tegtmeyer N, Rohde M et al. Campylobacter jejuni surface-bound protease HtrA, but not the secreted protease nor protease in shed membrane vesicles, disrupts epithelial cell-to-cell junctions. Cells. 2024;13:224. 10.3390/cells13030224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen R, Kappes JC, Smythies LE et al. Vaginal myeloid dendritic cells transmit founder HIV-1. J Virol. 2014;88:7683–8. 10.1128/JVI.00766-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen R, Richter HE, Clements RH et al. Macrophages in vaginal but not intestinal mucosa are monocyte-like and permissive to Human Immunodeficiency Virus type 1 infection. J Virol. 2009;83:3258–67. 10.1128/JVI.01796-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shen R, Richter HE, Smith PD. Early HIV-1 target cells in human vaginal and ectocervical mucosa. Am J Reprod Immunol. 2011;65:261–7. 10.1111/j.1600-0897.2010.00939.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shishpal P, Kasarpalkar N, Singh D et al. Characterization of Gardnerella vaginalis membrane vesicles reveals a role in inducing cytotoxicity in vaginal epithelial cells. Anaerobe. 2020;61:102090. 10.1016/j.anaerobe.2019.102090. [DOI] [PubMed] [Google Scholar]
- Shukla SK, Meier PR, Mitchell PD et al. Leptotrichia amnionii sp. nov., a novel bacterium isolated from the amniotic fluid of a woman after intrauterine fetal demise. J Clin Microbiol. 2002;40:3346–9. 10.1128/JCM.40.9.3346-3349.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Smayevsky J, Canigia LF, Lanza A et al. Vaginal microflora associated with bacterial vaginosis in nonpregnant women: reliability of sialidase detection. Infect Dis Obstet Gynecol. 2001;9:17–22. 10.1155/S1064744901000047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Spiegel CA. Susceptibility of Mobiluncus species to 23 antimicrobial agents and 15 other compounds. Antimicrob Agents Chemother. 1987;31:249–52. 10.1128/AAC.31.2.249. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srinivasan S, Fredricks DN. The human vaginal bacterial biota and bacterial vaginosis. Interdiscip Perspect Infect Dis. 2008;2008:750479. 10.1155/2008/750479. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Srinivasan S, Hoffman NG, Morgan MT et al. Bacterial communities in women with bacterial vaginosis: high resolution phylogenetic analyses reveal relationships of microbiota to clinical criteria. PLoS One. 2012;7:e37818. 10.1371/journal.pone.0037818. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stoddard E, Cannon G, Ni H et al. gp340 expressed on human genital epithelia binds HIV-1 envelope protein and facilitates viral Transmission1. J Immunol. 2007;179:3126–32. 10.4049/jimmunol.179.5.3126. [DOI] [PubMed] [Google Scholar]
- Stoyancheva G, Marzotto M, Dellaglio F et al. Bacteriocin production and gene sequencing analysis from vaginal Lactobacillus strains. Arch Microbiol. 2014;196:645–53. 10.1007/s00203-014-1003-1. [DOI] [PubMed] [Google Scholar]
- Sun S, Serrano MG, Fettweis JM et al. Race, the vaginal microbiome, and spontaneous preterm birth. mSystems. 2022;7:e00017–22. 10.1128/msystems.00017-22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tortelli BA, Lewis AL, Fay JC. The structure and diversity of strain-level variation in vaginal bacteria. Microb Genom. 2021;7:mgen000543. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trama J, Libby E, Mordechai E et al. Cervicovaginal epithelial cell cytokine production in response to microorganisms associated with bacterial vaginosis (43.40). J Immunol. 2007;178:S44. 10.4049/jimmunol.178.Supp.43.40. [DOI] [Google Scholar]
- Tugizov S. Virus-associated disruption of mucosal epithelial tight junctions and its role in viral transmission and spread. Tissue Barriers. 2021;9:1943274. 10.1080/21688370.2021.1943274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Turville SG, Cameron PU, Handley A et al. Diversity of receptors binding HIV on dendritic cell subsets. Nat Immunol. 2002;3:975–83. 10.1038/ni841. [DOI] [PubMed] [Google Scholar]
- Valladeau J, Ravel O, Dezutter-Dambuyant C et al. Langerin, a novel C-type lectin specific to Langerhans cells, is an endocytic receptor that induces the formation of Birbeck granules. Immunity. 2000;12:71–81. 10.1016/S1074-7613(00)80160-0. [DOI] [PubMed] [Google Scholar]
- van Smoorenburg MY, Remmerswaal EBM, Segui-Perez C et al. Vaginal Prevotella timonensis bacteria enhance HIV-1 uptake and differentially affect transmission by distinct primary dendritic cell subsets. Eur J Immunol. 2025;55:e202451192. 10.1002/eji.202451192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van Teijlingen NH, Helgers LC, Sarrami-Forooshani R et al. Vaginal bacterium Prevotella timonensis turns protective Langerhans cells into hiv-1 reservoirs for virus dissemination. EMBO J. 2022;41:e110629. 10.15252/embj.2022110629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- van Teijlingen NH, Helgers LC, Zijlstra-Willems EM et al. Vaginal dysbiosis associated-bacteria Megasphaera elsdenii and Prevotella timonensis induce immune activation via dendritic cells. J Reprod Immunol. 2020;138:103085. 10.1016/j.jri.2020.103085. [DOI] [PubMed] [Google Scholar]
- van Teijlingen NH, van Smoorenburg MY, Sarrami-Forooshani R et al. Prevotella timonensis bacteria associated with vaginal dysbiosis enhance Human Immunodeficiency Virus type 1 susceptibility of vaginal CD4+ T cells. J Infect Dis. 2024;230:e43–7. 10.1093/infdis/jiae166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veloo ACM, Chlebowicz M, Winter HLJ et al. Three metronidazole-resistant Prevotella bivia strains harbour a mobile element, encoding a novel nim gene, nimK, and an efflux small MDR transporter. J Antimicrob Chemother. 2018;73:2687–90. 10.1093/jac/dky236. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vodstrcil LA, Twin J, Garland SM et al. The influence of sexual activity on the vaginal microbiota and Gardnerella vaginalis clade diversity in young women. PLoS One. 2017;12:e0171856. 10.1371/journal.pone.0171856. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wald A, Link K. Risk of Human Immunodeficiency Virus infection in herpes simplex virus type 2-seropositive persons: a meta-analysis. J Infect Dis. 2002;185:45–52. 10.1086/338231. [DOI] [PubMed] [Google Scholar]
- Wang Z, Fu L, Xiong Z et al. Diagnosis and microecological characteristics of aerobic vaginitis in outpatients based on preformed enzymes. Taiwanese J Obstetr Gynecol. 2016;55:40–4. 10.1016/j.tjog.2015.06.012. [DOI] [PubMed] [Google Scholar]
- Weissman D, Li Y, Ananworanich J et al. Three populations of cells with dendritic morphology exist in peripheral blood, only one of which is infectable with Human Immunodeficiency Virus type 1. Proc Natl Acad Sci USA. 1995;92:826–30. 10.1073/pnas.92.3.826. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Williams OM, Hart KW, Wang EC et al. Analysis of CD4(+) T-cell responses to human papillomavirus (HPV) type 11 L1 in healthy adults reveals a high degree of responsiveness and cross-reactivity with other HPV types. J Virol. 2002;76:7418–29. 10.1128/JVI.76.15.7418-7429.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yan M, Peng J, Jabbar IA et al. Despite differences between dendritic cells and Langerhans cells in the mechanism of papillomavirus-like particle antigen uptake, both cells cross-prime T cells. Virology. 2004;324:297–310. 10.1016/j.virol.2004.03.045. [DOI] [PubMed] [Google Scholar]
- Yeoman CJ, Yildirim S, Thomas SM et al. Comparative genomics of Gardnerella vaginalis strains reveals substantial differences in metabolic and virulence potential. PLoS One. 2010;5:e12411. 10.1371/journal.pone.0012411. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zimmerli SC, Hauser C. Langerhans cells and lymph node dendritic cells express the tight junction component claudin-1. J Invest Dermatol. 2007;127:2381–90. 10.1038/sj.jid.5700882. [DOI] [PubMed] [Google Scholar]
- Zozaya-Hinchliffe M, Martin DH, Ferris MJ. Prevalence and abundance of uncultivated megasphaera-like bacteria in the human vaginal environment. Appl Environ Microbiol. 2008;74:1656–9. 10.1128/AEM.02127-07. [DOI] [PMC free article] [PubMed] [Google Scholar]

