ABSTRACT
Gardnerella species are associated with bacterial vaginosis (BV) and have been investigated as etiological agents of the condition. Nonetheless, the isolation of this taxon from healthy individuals has raised important questions regarding its etiological role. Recently, using advanced molecular approaches, the Gardnerella genus was expanded to include several different species that exhibit differences in virulence potential. Understanding the significance of these different species with respect to mucosal immunity and the pathogenesis and complications of BV could be crucial to solving the BV enigma. Here, we review key findings regarding the unique genetic and phenotypic diversity within this genus, virulence factors, and effects on mucosal immunity as they stand. We also comment on the relevance of these findings to the proposed role of Gardnerella in BV pathogenesis and in reproductive health and identify key gaps in knowledge that should be explored in the future.
KEYWORDS: Gardnerella, bacterial vaginosis, vaginal microbiome, sialidase, vaginolysin, cytokines, mucosal immunity, biofilms, chemokines, dendritic cells, female reproductive tract
INTRODUCTION
The cervicovaginal microbiome profoundly shapes reproductive health (1). The current consensus asserts that an optimal cervicovaginal microbiome in reproductive-age individuals is dominated by L. crispatus and several other non-iners lactobacilli (2). These lactobacilli confer protection from infections via the production of lactic acid and other antimicrobial products (3). Though several nonoptimal cervicovaginal microbial communities have been associated with clinically defined conditions, such as aerobic vaginitis (4) and vulvovaginal candidiasis (5), bacterial vaginosis (BV) is the most common one in reproductive-age individuals (6). During BV, the vaginal microbiome is characterized by a drastic reduction or complete lack of vaginal lactobacilli and increased abundance and quantities of anaerobic and facultative bacteria, whether examined by phenotypic methods such as Gram stain and light microscopy or by molecular methods using both compositional and quantitative genomics (2, 7–9). In addition, BV has also been linked to several reproductive sequelae, including pregnancy complications and infertility (10, 11), as well as increased susceptibility to sexually transmitted infections (STI), including increased acquisition risk and transmission of human immunodeficiency virus (HIV) (12, 13). The mechanistic link between BV and these associated sequelae remains unclear, though it potentially involves alterations in host mucosal immunity (14). Gardnerella vaginalis has been proposed as an etiological agent of BV as early as the 1950s (15, 16), although the isolation of this taxon from healthy individuals has raised questions regarding its etiological role (15, 17, 18). A critical review of Gardnerella as a cause of BV was recently published (15). More recently, the description of G. vaginalis was modified to include four species and multiple genomospecies (19). This has sparked new research questions regarding the clinical and immunological relevance of these now distinguishable taxa (Fig. 1). Here, we review how the molecular heterogeneity of Gardnerella species reflects differences in virulence potential and host immune responses to better understand their contributions to reproductive health.
FIG 1.
Key gaps in knowledge with respect to Gardnerella heterogeneity and contributions to the vaginal microbiome and to BV pathogenesis.
GARDNERELLA HETEROGENEITY — BEYOND “JUST GENETICS”
The Gardnerella genus of Actinobacteria (which have a high G-C content), includes several thin-walled Gram-positive pleomorphic species (20, 21), that until recently were collectively classified as G. vaginalis. The first Gardnerella isolate was isolated by Leopold in 1953 on modified blood agar from cervical swabs and urine samples (22). Within the following 2 years, two reports by Gardner and Duke described the same bacterium as a proposed agent of BV (nonspecific vaginitis) (16, 23). Initial efforts to differentiate pathogenic and nonpathogenic Gardnerella strains used biotyping based on biochemical tests. Eight Gardnerella biotypes were described based on hippurate hydrolysis, lipase activity, and beta-galactosidase activity (24). Additional Gardnerella biotypes were resolved through added biochemical characterization (25–27). Although certain biotypes (e.g., lipase-positive) appeared to be somewhat associated with BV (28, 29), this was not consistent across studies (30). Additionally, no clear associations were identified between these biotypes and known bacterial virulence factors (31–33). The clinical relevance of these biotypes therefore remains unclear, and the use of this system for the study of Gardnerella isolates has mostly been supplanted by molecular methods.
Earlier molecular attempts to decipher the genetic heterogeneity of Gardnerella, involved the use of restriction fragment length polymorphism. This revealed several genotypes but without a clear clinical relevance (34, 35). The description and taxonomy of G. vaginalis has since been changed following the description of four genetically distinct clades/subgroups identified by whole-genome sequencing and cpn60 barcode sequencing, and further species designation has been added and confirmed with subsequent molecular characterization (19, 36–38). The following Gardnerella species names along with several genomospecies have now been adopted: G. swidsinskii and G. leopoldii (subgroup A/clade 4), G. piotii (subgroup B/clade 2), G. vaginalis (subgroup C/clade 1), and subgroup D/clade 3 members, which include several genomospecies (19, 39) (Table 1). Future designation of additional Gardnerella species is likely with the genomic characterization of additional isolates. The expanded description of subgroup A to include two distinct species, G. leopoldii and G. swidsinskii, is somewhat supported by observations that these species do not tend to cooccur more often than other Gardnerella species (39). These genetically related though distinct genomospecies, are also reliably distinguished using matrix-assisted laser desorption/ionization time of flight mass spectrometry, which may be relevant for clinical tests and future studies of the cervicovaginal microbiome and BV (19). Structural and morphological differences also exist among these newly described Gardnerella species. For example, a G. swidsinskii isolate was found to possess a polysaccharide-like capsule, which was absent in a G. leopoldii isolate (40). Importantly, these structural variations may elicit different immune responses by stimulating different toll-like receptors (TLR) and other pattern recognition receptors (PRR), so further study is important (41, 42). Although 16S rRNA genes microbial profiling is the most commonly used method for the study of the vaginal microbiome, it does not generally discriminate different Gardnerella species (39). This is likely due to a combination of factors, including the presence of multiple copies of this genetic target within a single genome and a reduced barcoding gap compared to protein-encoding gene targets such as cpn60 (43).
TABLE 1.
Gardnerella spp. nomenclature and classification of common isolates, indicating key virulence factorsa
| Species designation | Subgroup/clade | Ecotype | Vaginolysin type (if present) | Sialidase activity (of tested isolates) | Known isolates |
|---|---|---|---|---|---|
| G. vaginalis genome sp. 1 | C/1 | 1A, 1B (JCP7275) | 1A + 1B | −/+ | ATCC 14019, ATCC 14018, ATCC 49145*, JCP7276, UGent 25.49*, HMP9231, UGent 09.01*, 3549624*, 284V, UGent 09.07*, 75712, 315-A, JCP7672, 0288E, JCP7275 |
| Gardnerella sp. genome sp. 2 | C/1 | 1B | 1B | − | JCP8108, 1400E, 55152, 41V |
| Gardnerella sp. genome sp. 3 | B/2 | 2B | 1A + 1B | −/+ | GED7275B*, JCP8017A, JCP8017B, JCP7659, 00703C2mash, JCP7719 |
| G. piotii genome sp. 4 | B/2 | 2A | 1B | + | JCP8151A, JCP8151B, JCP8522, JCP8070, JCP8066, UGent 21.28*, UGent 18.01* |
| G. leopoldii genome sp. 5 | A/4 | 3B | 2 | − | AMD, UGent 06.41*, UGent 09.48*, 6420B |
| G. swidsinskii genome sp. 6 | A/4 | 3B | 2 | − | 5-1, GS 9838-1*, GS 10234*, GV37*, 409-05 |
| Gardnerella sp. genome sp. 7 | A/4 | 3A | 1B + 1C | − | JCP8481A, JCP8481B, PSS_7772B* |
| Gardnerella sp. genome sp. 8 | D/3 | 3A | 1B | − | 101, 00703Dmash |
| Gardnerella sp. genome sp. 9 | D/3 | 3A | 1A | − | 6119V5 |
| Gardnerella sp. genome sp. 10 | D/3 | 3A | 1B | − | 1500E |
| Gardnerella sp. genome sp. 11 | B/2 | − | − | + | GED7760B |
| Gardnerella sp. genome sp. 12 | Unassigned/broadly A/4 | − | 3 | − | CMW7778B |
| Gardnerella sp. genome sp. 13 | Does not fit into the defined subgroups/clades | − | 1B | − | KA00225 |
| Ref. | |||||
| (19) | (36, 37) | (45) | (95) | (19, 104, 111) | (39, 159) |
−, unknown; −/+, sialidase activity confirmed in some isolates (in vitro), but not in others; +, (in vitro) confirmed sialidase activity; *, ecotype designation not available.
Ecotypes are genetically related strains that are otherwise ecologically distinct from others (i.e., possess different ecological roles) (44, 45). Currently proposed hypotheses speculate that different Gardnerella strains possess different ecological roles in BV pathogenesis (46). In line with this, three major Gardnerella ecotypes have been identified so far (using 35 draft and complete genomic sequences), roughly corresponding with the four clade/subgroup descriptions with a few exceptions (Table 1) (45). Ecotype 1 strains possess enriched genetic material related to pentose interconversion, galactose metabolism, and ATP-binding cassette transporters, ecotype 2 strains possess two distinct sialidase encoding genes (among other features), and ecotype 3 encompasses the rest of the species which display underrepresentation of the above genes and overabundance of other unique factors such as metalloendopeptidase related membrane proteins (45). Further characterization of these ecotypes, and expansion of the analysis to include more Gardnerella isolates and description of the ecotypes of other BV-associated bacteria could be useful for deciphering their contributions to BV.
In vitro studies of different Gardnerella isolates (belonging to different subgroups/clades) have provided insight to both phenotypic differences and species interactions. In vitro, subgroups A, B, and C thrive the most when grown as single/monococultured isolates, whereas subgroup D isolates have been observed to increase their growth rate as the number of competitors increases (47). This finding is congruent with cpn60 subgroup or clade-based characterizations of vaginal microbial communities where Gardnerella subgroups A, B, and C tend to dominate communities and reach high bacterial loads, whereas subgroup D is not generally a very abundant taxon and tends to occur more frequently alongside the other clades or other taxa (48–52). This may be due to the ability of subgroup D isolates to utilize a wider range of carbon sources relative to the other Gardnerella subgroups, acting as nutritional generalists, which promotes their growth in the vaginal microbiome through negative frequency dependent selection (53).
Regardless of BV status, subgroup A species are commonly detected at a higher frequency in the vagina compared to the other species (48, 52, 54). Nonetheless, there is no clear consensus regarding Nugent-BV and Gardnerella species associations, possibly due to differences in study populations. For example, a study using a cohort of nonpregnant premenopausal women found clades 1 and 3 were associated with Nugent-BV, whereas clades 4 and 2 did not exhibit these associations; notably clade 2 was found to associate with intermediate Nugent scores (54). In another cohort of women reporting a female sexual partner, clades 1, 2, 3, and microbial communities with multiple clades were associated with BV (52). Using the newer species designation, increased relative abundances of G. piotii, G. vaginalis, and G. swidsinskii were shown to be associated with Nugent-diagnosed BV in vaginal samples from Canadian women (39). In that same study, increased relative abundances of G. vaginalis and G. swidsinskii were associated with the presence of abnormal discharge and vaginal malodor, whereas the other two species were not (39). Whether these associations occur in other populations remains to be determined. With respect to treatment outcomes, G. leopoldii/G. swidsinskii (the primers used did not differentiate between the two) were more abundant among those with recurrent BV, whereas high abundance of genomospecies 7 was associated with a refractory response following oral metronidazole treatment (55). In vitro, clades 3 and 4 are inherently resistant to metronidazole, which could help explain these findings (56). However, in a study of premenopausal individuals with BV, decreases in clade 4 and 1 (measured by qPCR) posttreatment were observed regardless of clinical outcome (57).
VIRULENCE FACTORS AND PROPOSED ROLES IN BV PATHOGENESIS
Several hypotheses have been proposed regarding the contribution of Gardnerella species to BV. One hypothesis is that every Gardnerella species has the potential to cause BV, while others propose certain Gardnerella strains are genetically driven toward a more pathogenic phenotype (46). In line with these arguments, both BV- and non-BV associated Gardnerella isolates show evidence of horizontal gene transfer, and evidence of adherence and cytotoxicity of epithelial cells, albeit BV-associated Gardnerella isolates appear to adhere and cause cytotoxicity somewhat more efficiently (40, 58, 59). Gardnerella species possess several virulence factors, including a hemolysin, mucus degrading sialidases, and the ability to form biofilms which contribute to their “pathogenic” phenotype. The contributions of each of these different virulence factors have been examined in detail using in vitro and in-silico approaches.
Gardnerella biofilms.
BV is commonly associated with the presence of vaginal squamous epithelial cells studded with bacteria (“clue cells”) that are typically dominated by Gardnerella (16, 60, 61). The importance of Gardnerella to this BV-biofilm structure was not fully recognized until 2005 (62). Interestingly, evidence exists of sexual transmission of Gardnerella biofilms but not dispersed Gardnerella forms (63), which may, in part, explain why some individuals colonized by Gardnerella do not develop BV or transmit it to others. This finding even prompted a proposal to refer to the biofilm form as “gardnerellosis” (63). Gardnerella species have been shown to be superior at biofilm formation compared to other BV-associated bacteria (64), although isolate-specific differences have been noted in vitro (65). Coaggregation or the attachment of bacteria to one another facilitates biofilm attachment and was shown to occur between other BV-associated bacteria and Gardnerella (66). Little is known with respect to how Gardnerella initiates biofilm formation in vivo, though this process likely involves bacterial lectins such as serine-rich-repeat adhesins, which have been predicted in several Gardnerella isolates (67). These carbohydrate-binding proteins can attach to glycosylated components in the mucosa, forming a “two-way” host-Gardnerella interaction that favors bacterial biofilm proliferation. In addition, a collagen-binding Cna protein was identified on the surface of a G. vaginalis isolate (68) and may also play a role in biofilm establishment and potentially in immune evasion via interactions with complement proteins (69). Further characterization of these interactions will be necessary to explore the potential of these carbohydrate-binding and collagen-binding proteins as therapeutic targets or for diagnostics.
Once formed, bacterial biofilms are characterized as aggregated bacterial cells embedded in a sticky extracellular matrix (ECM). The composition of an ECM varies among organisms, but generally consists of macromolecules, and predominantly exopolysaccharides (70). Unsurprisingly, in a G. leopoldii isolate, transcripts associated with adhesion and exopolysaccharide biosynthesis such as glycosyltransferases were upregulated in biofilms compared to planktonic counterparts (71). These Gardnerella biofilms exhibit increased resistance to metronidazole (72) and clindamycin treatment (73), and reduced sensitivity to antibacterial metabolites such as hydrogen peroxide and lactic acid (74). A study of a single Gardnerella (subgroup C) isolate showed its biofilm contains a higher carbohydrate content compared to planktonic cultures, with relatively similar nucleic acid and protein content (74). N-acetylglucosamine appears to be an important contributor to this Gardnerella isolate biofilm ECM, although chitinase treatment did not significantly reduce biofilm thickness (74). In vitro, proteinase treatments appear to reduce Gardnerella biofilms and increase their susceptibility to antimicrobials, possibly due to degradation of bacterial anchor proteins (72, 74).
Extracellular DNA (eDNA) is another important component of bacterial biofilms thought to be released from lysed cells. Degradation of eDNA by DNases appears to inhibit biofilm formation and reduce biofilm density in vitro and decrease (but not eliminate) Gardnerella colonization in a murine model (75). These findings suggest eDNA is vital for Gardnerella biofilm formation and structural integrity, though questions remain regarding where the eDNA is localized within the biofilm, especially in vivo. The contributions (if any) of the host to the biofilm eDNA have not yet been explored either, though given the increases in exfoliated epithelial cells in BV samples (76), this cellular turnover could be a potential contributor, as was previously demonstrated in the gut mucosa (77). ECM composition also affects immune recognition (78). A transcriptomics study of a G. leopoldii isolate showed that at least in vitro, the biofilms exhibit reduced transcription of metabolic and ribosomal genes suggestive of reduced metabolic activity (especially carbon metabolism) (71). This could explain the observed resistance to antibiotics such as metronidazole (56). Metronidazole enters bacterial cells as a prodrug that is reduced intracellularly to an active form (79), and thus reduced metabolic activity reduces activation of metronidazole. Transcripts associated with protein export and amino acid biosynthesis were also highly expressed in these biofilm cultures (71). Amino acids produced by Gardnerella can be utilized mutualistically by Prevotella species and may thus contribute to the growth of other BV-associated bacteria within the biofilm structure (80, 81). BV-associated species can then metabolize the Gardnerella produced amino acids to biogenic amines which are the likely source of malodor during BV (82). Whether or not amino acid metabolism is upregulated in all Gardnerella species biofilms is unclear, although it could certainly shed light on why not all cases of BV result in a positive whiff test.
Quorum sensing (QS) has been identified as a form of communication between bacteria whereby signaling molecules (autoinducing peptides) are released to regulate gene expression in a way that depends on cell density and population needs. Given that regulation of virulence factors is an important component of QS (83), understanding the mechanisms of QS in Gardnerella initiated biofilms could identify potential targets for therapeutics against BV-associated biofilms. Autoinducer 2 (AI-2) transporter transcripts are highly abundant in in vitro G. leopoldii biofilms but not in their planktonic counterparts (71). G. vaginalis also produces the AI-2 signaling molecule required for quorum sensing, which can be inhibited to a degree following benzoyl peroxide (84) as well as by subtilosin (a bacteriocin) treatments (85). Beyond these findings, mechanisms of quorum sensing contributing to Gardnerella biofilm formation and persistence are not fully understood. Models to study Gardnerella-initiated polymicrobial biofilms are being developed (66, 86), and analysis of those systems through transcriptomic, proteomic, and metabolomic approaches will greatly improve our understanding of QS in these biofilms.
In vitro, there is no consensus on which subgroup forms better biofilms (37, 47, 65) and media formulation was shown to affect biofilm formation capabilities of different Gardnerella species isolates (58, 87). G. vaginalis biofilm formation is enhanced at pH ~5 to 6.5, which is the typical pH range during BV, either due to increased adhesion to the epithelia under these settings and/or reduced interference by lactobacilli (88). In contrast, lower pH (<4.5) typically seen in lactobacilli-dominant states and high pH (>7) result in weak or no Gardnerella biofilms, at least in vitro (88). It is crucial to understand what factors promote these apparent differences in biofilm formation and why, as well as which best represent conditions found in vivo to establish more valid in vitro systems to study Gardnerella biofilm formation and their effects on the mucosa.
Vaginolysin.
Vaginolysin, is a cholesterol-dependent hemolysin which lyses susceptible cells upon interaction with the molecule CD59 in a way that forces rapid structural “blebbing” changes in vaginal and cervical epithelial cells (89, 90). Although vaginolysin causes hemolysis of human erythrocytes, bacteremia due to Gardnerella is a relatively rare phenomenon and is not a feature of BV (89, 91). The undecapeptide vaginolysin was shown to be orthologous to other Gram-positive cholesterol-dependent cytolysins and appears to function in a human specific manner in vitro due to its interactions with the complement glycoprotein CD59 (89). Though vaginolysin can be directly secreted, recent evidence suggests it is also packaged within G. vaginalis membrane vesicles, which can be internalized by vaginal epithelial cells (92). Production of vaginolysin as well as other virulence factors in these vesicles is dependent on pH and is not produced under more acidic conditions (such as those found in optimal lactobacilli-dominant vaginal communities) (93). It is unclear how common membrane vesicle formation is among the different Gardnerella species and genomospecies. Like other cytolysins, the epithelial response to vaginolysin induces phosphorylation and activation of p38 mitogen-activated protein kinase, and the effect on the epithelium is concentration dependent (89, 94). Not all Gardnerella isolates possess vaginolysin genes, as several G. piotii isolates and genomospecies 11 seem to lack them, possibly due to purifying selection (59, 65, 95) (Table 1). The gene encoding vaginolysin appears to be exchanged through horizontal gene transfer and is likely not part of the core Gardnerella genome as previously thought (36, 59). More than one Gardnerella species often cooccur in the vaginal microbiome and during BV (18, 48, 95), therefore differential production of vaginolysin could explain the different roles these species play during the establishment of BV. Vaginolysin expression is upregulated in the planktonic state and reduced in biofilms of G. leopoldii (71). Given that vaginolysin is capable of lysing neutrophils (96) and possibly other immune cells such as macrophages, vaginolysin production in the early stages of Gardnerella proliferation could represent an early immune evasion mechanism.
Differences in the genetic makeup of vaginolysin exist within the 13 different Gardnerella genomospecies, and these have recently been typed into 5 vaginolysin groups — types 1A, 1B, 1C (collectively type 1), type 2, and type 3 (95). Type 1 and 2 are more common among the characterized isolates (Table 1), and type 1A appears to induce higher cytotoxicity even upon reduced CD59 expression compared to type 2. Nonetheless, vaginal epithelial cytokine responses to type 1A and type 2 vaginolysins are similar for the most part (with increased cytokine concentration upon treatment) (95). Vaginolysin, has also been shown to activate proinflammatory signaling in cervical HeLa cells (89), though its effects on vaginal epithelial tissue appear to differ based on apical or basolateral activity likely due to differential CD59 expression (97). BV-associated Gardnerella species induced higher cytotoxicity in cervical HeLa cells relative to non-BV isolates, which correlated with increased vaginolysin expression (58, 98). Treatment with L. crispatus reduced Gardnerella cytotoxicity in vitro, and though vaginolysin expression was also generally reduced in BV-Gardnerella isolates, this did not reach statistical significance (98). Specific immune responses to Gardnerella vaginolysin have been detected in individuals with BV (99). Antivaginolysin-specific immunoglobulin A (IgA) response has been demonstrated in both healthy and BV samples (100) and may help prevent adverse health outcomes in pregnant women with BV (101). The presence of these higher IgA responses could also be indicative of a low sialidase environment (102), as will be discussed next.
Gardnerella sialidases.
The vaginal mucosa is a protective barrier comprised of sialoglycoproteins called mucins that, among other functions, help prevent adherence of pathogens to the underlying epithelium (103). Gardnerella species possess several hydrolytic enzymes such as sialidases that appear to play a significant role in the pathogenesis of BV. Sialidases cleave sialic acid from mucosal sialoglycans to enable bacterial adherence and biofilm formation and allow its use as a nutritional substrate (104). In line with this, the vaginal epithelial glycocalyx appeared to be reduced in BV-positive participants compared to healthy controls. This was further confirmed to be associated with reduced sialylation of surface N- and O-glycans, and a similar glycan phenotype was produced following treatment with a Gardnerella sialidase. Desialylation of these glycans exposes terminal sugars that can then be used for bacterial adherence (105), perhaps via carbohydrate-binding proteins such as lectins which have been predicted in Gardnerella (67). Sialidases can also cleave immunoglobulins (secreted sialoglycoproteins) likely as an immune evasion strategy, which may also indirectly increase risk to other mucosal pathogens. In support of this, an association between sialidases and IgA cleavage has been demonstrated (102).
Gardnerella isolates possessing sialidase genes have been shown to be associated with BV biofilms (106). This has led to the development of diagnostic tests for BV based on sialidase activity (107–109). In line with this, concentrations of the mucins 5AC and 5B are elevated in cervicovaginal lavage samples with increased bacterial diversity (110). In vitro, some Gardnerella species (especially subgroup B isolates) cause degradation of vaginal mucins, via anchored cell-associated sialidases and secreted sialidases (104, 111). Secretion of cell wall-anchored sialidases can also occur due to cell turnover or proteolytic cleavage (111, 112). Sialidase activity in Gardnerella isolates is primarily attributed to NanH2 and NanH3, whereas NanH1 (sialidase A) genes are also present in isolates that do not show sialidase activity (111). The lack of extracellular sialidase activity associated with NanH1 may be explained by its lack of signal peptide, suggesting it is likely an intracellular enzyme (112). Phenotypic assessments of clinical isolates have shown that sialidase activity varies significantly among the different Gardnerella species, with few clade 1 and many clade 2 isolates possessing sialidase activity, whereas clade 4 members do not (19, 38, 65). Sialidase activity is strongest in G. piotii and genomospecies 3 (subgroup B) isolates (38, 112), and the presence of sialidase genes in other Gardnerella isolates could be due to horizontal gene transfer within the vaginal ecosystem (59). Based on these findings, it can be speculated that colonization by NanH2 and/or NanH3-encoding Gardnerella species (especially subgroup B isolates) may be the first step in the establishment of BV biofilms. However, our understanding is incomplete and sialidase is likely only one of several factors in the formation of biofilms and in the pathogenesis of BV.
Other virulence factors.
Other Gardnerella virulence factors have been recognized but are not as well characterized; these include pili, lipases, and siderophores. Pili have been demonstrated in some Gardnerella isolates (113, 114), and so has the presence of pili-associated genes (40), though the presence of these genes does not always result in pili expression in vitro. Existence of pili in Gardnerella species may contribute to virulence on multiple fronts by interacting with the host immune system, enhancing attachment to tissues, and contributing to horizontal transfer of other virulence genes. Iron acquisition strategies, including via siderophore activity has been demonstrated in several Gardnerella isolates (115) which may explain increases of Gardnerella loads during menses (116). Adequate access to iron is important for the expression of virulence factors (117), and evolution of multiple iron acquisition strategies in some Gardnerella isolates can thus promote their survival in vivo. The presence of lipases has been demonstrated in several Gardnerella isolates (24). Though their role in Gardnerella pathogenicity is not well characterized, bacterial lipases can increase nutrient availability, promoting persistence, and may be involved in immune evasion and/or weakening of the epithelial barrier (118).
IMMUNE RESPONSES TO GARDNERELLA SPECIES — FINDINGS FROM IN VITRO AND ANIMAL STUDIES
The relationship between BV and mucosal immunity is incompletely understood. Clinically, BV is not characterized by leukocyte infiltration, hence the name “vaginosis” and not “vaginitis.” This shouldn’t be surprising given that BV is recognized as a biofilm condition and the nature of bacterial biofilms to promote a more quiescent response (78). Nonetheless, BV is associated with subclinical inflammation characterized by generally high proinflammatory cytokine responses and decreased levels of some chemokines (119–121). Given its predominance in BV biofilms (62), Gardnerella-mediated alternations in mucosal immunity could help explain BV persistence and how BV increases the risk for STI and pregnancy complications. This section focuses on findings from in vitro and murine studies utilizing single Gardnerella isolates to examine how Gardnerella species contribute to cytokine production and cell viability (Table S1).
Immune cell responses to Gardnerella species.
Four main types of antigen-presenting cells are found within the vaginal epithelia and lamina propria: CD14− dendritic cells (DCs), CD14+ DCs, macrophages, and Langerhans cells (122). DCs sample for pathogens and respond to microbial insults by production of cytokines that contribute to T-cell polarization. The production of these cytokines differs among the DC subsets, such that CD14− DCs favor Th2 activation, and CD14+ DCs prime Th1 responses (122). Bertran et al. (123) showed that G. vaginalis did not generally induce cytotoxicity or increases in proinflammatory cytokines (TNF-α, IL-12p70, IFN-γ) in monocyte-derived DCs, suggesting a somewhat quiescent response. A different study utilizing the same isolate (as well as Prevotella and other BV-associated organisms) did show a significant increase in DC activation markers and increased expression of IL-1β, IL-6, IL-8, IL-12A + IL-12B, and TNF cytokine mRNA (124). However, in the case of Gardnerella, the responses across the board were much less pronounced than those in response to Prevotella stimulation. Interestingly, at the protein level only IL-8 secretion was significantly increased in response to Gardnerella-only stimulation, whereas many other cytokines, including TNF-α, IL-1β, and IL-6, were increased in response to Prevotella stimulation (124). The apparent effect on cytokine secretion could be indicative of an altered DC response to G. vaginalis. The findings of the latter study are more in line with available literature showing that DC stimulation with cervicovaginal fluid (as opposed to a single microorganism) from BV patients leads to DC activation (125). T-lymphocyte and peripheral blood mononuclear cells challenged with subgroup C isolates increased levels of some proinflammatory cytokines, including IFN-γ and IL-17 (123, 126) but not IP-10 or IFN-α (126). Lymphocyte proliferation seems to occur only at high, BV-consistent Gardnerella loads (107 CFU/mL) (123). Studies looking at human and mouse macrophage and monocytic cell lines show increases in IL-12 and TNF-α transcription and secretion, as well as IFN-γ, IL-10, and IL-17 secretion (123, 127) and IL-1β transcription and secretion (127–129), which could be suggestive of increases in Th1, Th17 and T-reg responses. However, these models do not capitulate well the diversity of DC effector and tissue macrophage function found in human vaginal mucosal epithelial tissue. Expression of CCR7 and the costimulatory molecule CD80 were also noted on macrophages following treatment with G. vaginalis supernatants, as was the increased production of reactive oxygen species by stimulated macrophages, suggestive of M1 macrophage polarization (127, 128). In other bacterial infections, M1 tissue macrophages are associated with microbial clearance and are activated by the more metabolically active planktonic bacteria (130). It remains unclear whether stimulation of these macrophages with Gardnerella-dominated biofilms will reproduce these findings or rather promote an M2 phenotype as reported in other biofilm conditions (130). In monocytic cells, stimulation with lavage samples from individuals with BV resulted in cell activation via TLR2 mediated response but not TLR4 (131). Within macrophages, the resulting signaling cascade leads to activation of NF-κB which then activates the NLRP3 inflammasome. NLRP3 inflammasome leads to caspase-1 production which then cleaves the pro-IL-1β, and pro-IL-18 proteins leading to their upregulated release and eventually causing cell death via pyroptosis (127, 129). This inflammasome activation has been shown to enhance HIV-1 replication which may help explain the increase in HIV-1 susceptibility in individuals with BV. Other immune cell populations, including myeloid derived suppressor cells and innate T-cells (which are present within the epithelia), have been implicated in other biofilm infections (78) but were not studied extensively in the context of Gardnerella colonization.
Cytokine responses of reproductive epithelia and fetal membranes to Gardnerella challenge.
Pivotal sites of Gardnerella colonization are the stratified squamous epithelium of the vagina and ectocervix, or the simple columnar epithelium of the endocervix and uterus in the case of an ascending infection (132). Therefore, it is not surprising that the majority of in vitro and murine studies to date have focused on the immune responses of these sites to Gardnerella. Cytokine responses appear to differ both depending on the cell model and Gardnerella species tested. For example, results from in vitro studies using the same G. vaginalis isolates have not been consistent (Table S1). HeLa cell stimulation with a large dose of live bacteria induced significant increases in IL-1β, TNF-α, IL-6, and IL-8 (133), whereas treatment of the same cell line with lysates from the same isolate did not (134). The challenge of cervical and uterine carcinoma cell lines with heat-inactivated G. vaginalis isolates also did not cause significant changes in these cytokines (126), though a lower challenge dose was used. Though some studies suggest increased IL-8 production by vaginal and cervical epithelial cells (133, 135–139), BV is not typically associated with an increase in neutrophil counts (140). However, in murine models, inoculation with G. vaginalis does increase vaginal myeloperoxidase activity (141). One possible explanation for this is lysis of neutrophils by vaginolysin in some settings. This IL-8 response also seems to differ depending on the Gardnerella species used, as do other chemokine responses (97). In the lower female genital tract, the vaginal epithelium has a different PRR profile compared to endocervical cells, and to immune cells in the basement membrane which may explain differences in the immune responses to the same pathogen (142). For example, increases in IP-10 production were present following endocervical G. vaginalis challenge but not vaginal or ectocervical challenge (143) though treatment with cell free supernatants from a related isolate did induce IP-10 secretion in ectocervical cells (137). In contrast, in response to G. leopoldii isolate treatment, secreted IP-10 was reduced in both apical and basolateral ectocervical-vaginal compartments (97). No change was observed in response to G. vaginalis isolates (138, 143), and an increase was observed in response to an uncharacterized clinical isolate in vaginal epithelial cells (139). Whether these responses are consistent in vivo or across models, and whether regulation of these occurs at the transcriptional, translational, or secretory level is unclear. Reduced chemokine responses have been described in response to anaerobic oral infections in which Prevotella species are prominent, and with respect to IP-10 and related CXCR3 chemokines in response to intracellular pathogens (144, 145). Reversal of this CXCR3 chemokine response seems to be a marker of successful BV treatment, though a causal link is yet to be elucidated (146). IP-10 and related CXCR3 chemokines have effects on Th1 cells, CD8+ T cells, and other lymphocytes, including natural killer cells and γδ T cells (147), which are understudied in the context of BV. Several hypotheses could help explain how Gardnerella and other BV-associated species exert their effects on mucosal immunity, including via short-chain fatty acid production (148), stimulation of different host signaling pathways, or even production of enzymes and toxins capable of degrading cytokines and mucosal components as reported in other bacterial and biofilm conditions.
BV is associated with increased risk to pregnancy complications, including preterm birth (11), possibly due to ascending infection and/or stimulation of inflammation (149). Treatment of fetal membranes with Gardnerella isolates (mostly G. vaginalis) promoted secretion of the proinflammatory cytokines IL-6, TNF-α, and IL-1β (mostly from the choriodecidual compartment) (136, 150–152), and the immune-modulatory cytokine IL-10 (153–155). Production of beta-defensins such as HBD-1 also increased following Gardnerella stimulation (150). With respect to PRR signaling, TLR-2 and TLR-7 were somewhat upregulated in fetal membranes in response to G. vaginalis although this was not statistically significant (156). Gardnerella has been detected more frequently in the cervix of women who underwent premature rupture of membranes with evidence of intraamniotic microbial invasion but without accompanying intraamniotic inflammation (157). In support of this, the increase in IL-6 in response to Gardnerella challenge of fetal membranes was accompanied by increased production of gp130 and mIL-6R (151). This suggests that IL-6 response to Gardnerella occurs through the gp130 and mIL-6 membrane receptor which induces antiinflammatory action (151). This again supports the hypothesis that as biofilms Gardnerella species either do not induce or somehow suppress some inflammatory responses. To add to this, Gardnerella biofilms within the endometrium and fallopian tubes have been reported in vivo and presence of these biofilms was more likely in pregnant and BV positive individuals (132). Stimulation of in vitro 3-dimensional endometrial epithelial cell models with G. piotii isolates did not elicit any significant cytokine responses, though one of the isolates did slightly increase MIP-3α secretion (158). Gardnerella has also been proposed to enhance ascension of perhaps more pathogenic organisms such as group B streptococci or mycoplasmas into fetal membranes. This was supported in a mouse model, where coinoculation resulted in invasive infection of placental tissue accompanied by placental histopathology (152). In a separate study, G. vaginalis was shown to induce IL-8, IL-6, IL-10, and IL-1β expression in the cervicovaginal space of pregnant mice, but reduced expression of the proinflammatory cytokine TNF-α relative to negative controls. G. vaginalis-induced cervical remodeling/breakdown was also proposed in this model due to increased expression of soluble E-cadherin and Tff-1 (136), possibly due to microRNA regulation. MicroRNAs and gene regulation by Gardnerella metabolites have also been proposed as potential mediators of the link between BV and pregnancy complications. In support of this hypothesis, G. vaginalis supernatant treatment of ectocervical cells resulted in increased expression of miRNAs associated with epithelial barrier disruption and short gestation (137), an effect that was mostly reversed upon treatment with optimal lactobacilli supernatants. Nonetheless, given that pregnancy can alter TLR expression and mucosal antimicrobial properties, validation of these miRNA findings is necessary.
SUMMARY OF KNOWLEDGE GAPS AND CHALLENGES
The role of Gardnerella in reproductive health remains incompletely understood. On the one hand, Gardnerella is a core component of BV biofilms, and on the other, it has also been isolated from healthy individuals. Advances to resolve Gardnerella strains into pathogenic and nonpathogenic categories have been proven challenging. Recently, the Gardnerella genus was emended to include several different species and genomospecies, which do appear to possess different virulence potentials. The regulation of these virulence factors and their effects on the mucosal epithelia remain ambiguous and are currently being investigated through systems biology approaches and in vitro models. Given that the biofilm form of Gardnerella appears to be the most problematic for reproductive health, identification of bacterial components involved in adhesion and biofilm formation will be beneficial for the design of more targeted interventions. Perhaps the most significant gaps in knowledge are with respect to Gardnerella effects on cervicovaginal mucosal immune responses and how those differ among the different genomospecies and the biofilm and planktonic growth states. We reviewed findings from in vitro and murine-controlled Gardnerella challenge studies which mostly used G. vaginalis isolates and show some upregulation in proinflammatory responses in some models but not in others. With respect to some chemokines such as IP-10, in vitro studies seem to show discrepant results in response to Gardnerella. Whether this is due to model and/or Gardnerella species differences is unclear. An area remaining completely uncharacterized with respect to Gardnerella immunobiology is the interactions with myeloid-derived suppressor cells as well as intraepithelial lymphocytes, which have been implicated in other biofilm conditions. Further investigation of these immune responses when combined with consideration of Gardnerella heterogeneity will greatly enhance our understanding of Gardnerella species contributions to reproductive health.
ACKNOWLEDGMENTS
K.S.M. is currently supported by the University of Manitoba Department of Medicine H.E. Sellers Research Chair. K.S.M. received research funding from the Canadian Institutes for Health Research (grant number THA-11960).
E.S.: Conceptualization, Investigation, Writing – Original Draft, Writing – Review and Editing; J.E.H.: Writing – Review and Editing; P.S.: Supervision, Funding Acquisition, Writing – Review and Editing; K.S.M.: Conceptualization, Supervision, Resources, Funding Acquisition, Writing – Review and Editing.
We declare no conflict of interest. The sponsors had no role in the design, execution, interpretation, or writing of the study.
Footnotes
Supplemental material is available online only.
Contributor Information
Kelly S. MacDonald, Email: kelly.macdonald@umanitoba.ca.
Anthony R. Richardson, University of Pittsburgh
REFERENCES
- 1.Martin DH. 2012. The microbiota of the vagina and its influence on women's health and disease. Am J Med Sci 343:2–9. doi: 10.1097/MAJ.0b013e31823ea228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.McKinnon LR, Achilles SL, Bradshaw CS, Burgener A, Crucitti T, Fredricks DN, Jaspan HB, Kaul R, Kaushic C, Klatt N, Kwon DS, Marrazzo JM, Masson L, McClelland RS, Ravel J, van de Wijgert J, Vodstrcil LA, Tachedjian G. 2019. The evolving facets of bacterial vaginosis: implications for HIV transmission. AIDS Res Hum Retroviruses 35:219–228. doi: 10.1089/aid.2018.0304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Petrova MI, Lievens E, Malik S, Imholz N, Lebeer S. 2015. Lactobacillus species as biomarkers and agents that can promote various aspects of vaginal health. Front Physiol 6:81. doi: 10.3389/fphys.2015.00081. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Donders GG, Vereecken A, Bosmans E, Dekeersmaecker A, Salembier G, Spitz B. 2002. Definition of a type of abnormal vaginal flora that is distinct from bacterial vaginosis: aerobic vaginitis. BJOG: An Internal J Obs Gyn 109:34–43. doi: 10.1111/j.1471-0528.2002.00432.x. [DOI] [PubMed] [Google Scholar]
- 5.Goncalves B, Ferreira C, Alves CT, Henriques M, Azeredo J, Silva S. 2016. Vulvovaginal candidiasis: epidemiology, microbiology and risk factors. Crit Rev Microbiol 42:905–927. doi: 10.3109/1040841X.2015.1091805. [DOI] [PubMed] [Google Scholar]
- 6.Javed A, Parvaiz F, Manzoor S. 2019. Bacterial vaginosis: an insight into the prevalence, alternative treatments regimen and it's associated resistance patterns. Microb Pathog 127:21–30. doi: 10.1016/j.micpath.2018.11.046. [DOI] [PubMed] [Google Scholar]
- 7.Nugent RP, Krohn MA, Hillier SL. 1991. Reliability of diagnosing bacterial vaginosis is improved by a standardized method of gram stain interpretation. J Clin Microbiol 29:297–301. doi: 10.1128/jcm.29.2.297-301.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Zozaya-Hinchliffe M, Lillis R, Martin DH, Ferris MJ. 2010. Quantitative PCR assessments of bacterial species in women with and without bacterial vaginosis. J Clin Microbiol 48:1812–1819. doi: 10.1128/JCM.00851-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sha BE, Chen HY, Wang QJ, Zariffard MR, Cohen MH, Spear GT. 2005. Utility of Amsel criteria, Nugent score, and quantitative PCR for Gardnerella vaginalis, Mycoplasma hominis, and Lactobacillus spp. for diagnosis of bacterial vaginosis in human immunodeficiency virus-infected women. J Clin Microbiol 43:4607–4612. doi: 10.1128/JCM.43.9.4607-4612.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Ravel J, Moreno I, Simon C. 2021. Bacterial vaginosis and its association with infertility, endometritis, and pelvic inflammatory disease. Am J Obstet Gynecol 224:251–257. doi: 10.1016/j.ajog.2020.10.019. [DOI] [PubMed] [Google Scholar]
- 11.Hillier SL, Nugent RP, Eschenbach DA, Krohn MA, Gibbs RS, Martin DH, Cotch MF, Edelman R, Pastorek JG, 2nd, Rao AV, et al. 1995. Association between bacterial vaginosis and preterm delivery of a low-birth-weight infant. The Vaginal Infections and Prematurity Study Group N Engl J Med 333:1737–1742. [DOI] [PubMed] [Google Scholar]
- 12.Atashili J, Poole C, Ndumbe PM, Adimora AA, Smith JS. 2008. Bacterial vaginosis and HIV acquisition: a meta-analysis of published studies. AIDS 22:1493–1501. doi: 10.1097/QAD.0b013e3283021a37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cohen CR, Lingappa JR, Baeten JM, Ngayo MO, Spiegel CA, Hong T, Donnell D, Celum C, Kapiga S, Delany S, Bukusi EA. 2012. Bacterial vaginosis associated with increased risk of female-to-male HIV-1 transmission: a prospective cohort analysis among African couples. PLoS Med 9:e1001251. doi: 10.1371/journal.pmed.1001251. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Spear GT, St John E, Zariffard MR. 2007. Bacterial vaginosis and human immunodeficiency virus infection. AIDS Res Ther 4:25. doi: 10.1186/1742-6405-4-25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Morrill S, Gilbert NM, Lewis AL. 2020. Gardnerella vaginalis as a Cause of Bacterial Vaginosis: appraisal of the Evidence From in vivo Models. Front Cell Infect Microbiol 10:168. doi: 10.3389/fcimb.2020.00168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Gardner HL, Dukes CD. 1955. Haemophilus vaginalis vaginitis: a newly defined specific infection previously classified non-specific vaginitis. Am J Obstet Gynecol 69:962–976. doi: 10.1016/0002-9378(55)90095-8. [DOI] [PubMed] [Google Scholar]
- 17.Hickey RJ, Forney LJ. 2014. Gardnerella vaginalis does not always cause bacterial vaginosis. J Infect Dis 210:1682–1683. doi: 10.1093/infdis/jiu303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Janulaitiene M, Paliulyte V, Grinceviciene S, Zakareviciene J, Vladisauskiene A, Marcinkute A, Pleckaityte M. 2017. Prevalence and distribution of Gardnerella vaginalis subgroups in women with and without bacterial vaginosis. BMC Infect Dis 17:394. doi: 10.1186/s12879-017-2501-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Vaneechoutte M, Guschin A, Van Simaey L, Gansemans Y, Van Nieuwerburgh F, Cools P. 2019. Emended description of Gardnerella vaginalis and description of Gardnerella leopoldii sp. nov., Gardnerella piotii sp. nov. and Gardnerella swidsinskii sp. nov., with delineation of 13 genomic species within the genus Gardnerella. Int J Syst Evol Microbiol 69:679–687. doi: 10.1099/ijsem.0.003200. [DOI] [PubMed] [Google Scholar]
- 20.Sadhu K, Domingue PA, Chow AW, Nelligan J, Cheng N, Costerton JW. 1989. Gardnerella vaginalis has a gram-positive cell-wall ultrastructure and lacks classical cell-wall lipopolysaccharide. J Med Microbiol 29:229–235. doi: 10.1099/00222615-29-3-229. [DOI] [PubMed] [Google Scholar]
- 21.Muli FW, Struthers JK, Tarpey PA. 1999. Electron microscopy studies on Gardnerella vaginalis grown in conventional and biofilm systems. J Med Microbiol 48:211–213. doi: 10.1099/00222615-48-2-211. [DOI] [PubMed] [Google Scholar]
- 22.Leopold S. 1953. Heretofore undescribed organism isolated from the genitourinary system. U S Armed Forces Med J 4:263–266. [PubMed] [Google Scholar]
- 23.Gardner HL, Dukes CD. 1954. New etiologic agent in nonspecific bacterial vaginitis. Science 120:853. doi: 10.1126/science.120.3125.853. [DOI] [PubMed] [Google Scholar]
- 24.Piot P, Van Dyck E, Peeters M, Hale J, Totten PA, Holmes KK. 1984. Biotypes of Gardnerella vaginalis. J Clin Microbiol 20:677–679. doi: 10.1128/jcm.20.4.677-679.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Benito R, Vazquez JA, Berron S, Fenoll A, Saez-Neito JA. 1986. A modified scheme for biotyping Gardnerella vaginalis. J Med Microbiol 21:357–359. doi: 10.1099/00222615-21-4-357. [DOI] [PubMed] [Google Scholar]
- 26.Pedraza-Aviles AG, Inzunza-Montiel AE, Ortiz-Zaragoza MC, Morales-Espinosa MR, Ponce-Rosas ER. 1995. Gardnerella vaginalis biotypes: modification of a proposed system. Rev Latinoam Microbiol 37:19–26. [PubMed] [Google Scholar]
- 27.Gonzalez-Pedraza Aviles A, Ortiz-Zaragoza MC, Inzunza-Montiel AE, Ponce-Rosas ER. 1996. Biotypes of Gardnerella vaginalis isolated from urinary tract. Rev Latinoam Microbiol 38:75–80. [PubMed] [Google Scholar]
- 28.Briselden AM, Hillier SL. 1990. Longitudinal study of the biotypes of Gardnerella vaginalis. J Clin Microbiol 28:2761–2764. doi: 10.1128/jcm.28.12.2761-2764.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nisha K, Antony B, Udayalaxmi J. 2019. Comparative analysis of virulence factors & /biotypes of Gardnerella vaginalis isolated from the genital tract of women with & without bacterial vaginosis. Indian J Med Res 149:57–61. doi: 10.4103/ijmr.IJMR_1674_16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Aroutcheva AA, Simoes JA, Behbakht K, Faro S. 2001. Gardnerella vaginalis isolated from patients with bacterial vaginosis and from patients with healthy vaginal ecosystems. Clin Infect Dis 33:1022–1027. doi: 10.1086/323030. [DOI] [PubMed] [Google Scholar]
- 31.Scott TG, Smyth CJ, Keane CT. 1987. In vitro adhesiveness and biotype of Gardnerella vaginalis strains in relation to the occurrence of clue cells in vaginal discharges. Genitourin Med 63:47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Pleckaityte M, Janulaitiene M, Lasickiene R, Zvirbliene A. 2012. Genetic and biochemical diversity of Gardnerella vaginalis strains isolated from women with bacterial vaginosis. FEMS Immunol Med Microbiol 65:69–77. doi: 10.1111/j.1574-695X.2012.00940.x. [DOI] [PubMed] [Google Scholar]
- 33.Udayalaxmi J, Bhat GK, Kotigadde S. 2011. Biotypes and virulence factors of Gardnerella vaginalis isolated from cases of bacterial vaginosis. Indian J Med Microbiol 29:165–168. doi: 10.4103/0255-0857.81798. [DOI] [PubMed] [Google Scholar]
- 34.Simoes JA, Hashemi FB, Aroutcheva AA, Heimler I, Spear GT, Shott S, Faro S. 2001. Human immunodeficiency virus type 1 stimulatory activity by Gardnerella vaginalis: relationship to biotypes and other pathogenic characteristics. J Infect Dis 184:22–27. doi: 10.1086/321002. [DOI] [PubMed] [Google Scholar]
- 35.Nath K, Devlin D, Beddoe AM. 1992. Heterogeneity in restriction patterns of Gardnerella vaginalis isolates from individuals with bacterial vaginosis. Res Microbiol 143:199–209. doi: 10.1016/0923-2508(92)90009-D. [DOI] [PubMed] [Google Scholar]
- 36.Ahmed A, Earl J, Retchless A, Hillier SL, Rabe LK, Cherpes TL, Powell E, Janto B, Eutsey R, Hiller NL, Boissy R, Dahlgren ME, Hall BG, Costerton JW, Post JC, Hu FZ, Ehrlich GD. 2012. Comparative genomic analyses of 17 clinical isolates of Gardnerella vaginalis provide evidence of multiple genetically isolated clades consistent with subspeciation into genovars. J Bacteriol 194:3922–3937. doi: 10.1128/JB.00056-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Paramel Jayaprakash T, Schellenberg JJ, Hill JE. 2012. Resolution and characterization of distinct cpn60-based subgroups of Gardnerella vaginalis in the vaginal microbiota. PLoS One 7:e43009. doi: 10.1371/journal.pone.0043009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schellenberg JJ, Paramel Jayaprakash T, Withana Gamage N, Patterson MH, Vaneechoutte M, Hill JE. 2016. Gardnerella vaginalis Subgroups Defined by cpn60 Sequencing and Sialidase Activity in Isolates from Canada, Belgium and Kenya. PLoS One 11:e0146510. doi: 10.1371/journal.pone.0146510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Hill JE, Albert AYK, Group VR, the VOGUE Research Group . 2019. Resolution and cooccurrence patterns of Gardnerella leopoldii, G. swidsinskii, G. piotii, and G. vaginalis within the vaginal microbiome. Infect Immun 87:e00532-19. doi: 10.1128/IAI.00532-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Harwich MD, Jr, Alves JM, Buck GA, Strauss JF, 3rd, Patterson JL, Oki AT, Girerd PH, Jefferson KK. 2010. Drawing the line between commensal and pathogenic Gardnerella vaginalis through genome analysis and virulence studies. BMC Genomics 11:375. doi: 10.1186/1471-2164-11-375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Kawasaki T, Kawai T. 2014. Toll-like receptor signaling pathways. Front Immunol 5:461. doi: 10.3389/fimmu.2014.00461. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Nicholson LB. 2016. The immune system. Essays Biochem 60:275–301. doi: 10.1042/EBC20160017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Links MG, Dumonceaux TJ, Hemmingsen SM, Hill JE. 2012. The chaperonin-60 universal target is a barcode for bacteria that enables de novo assembly of metagenomic sequence data. PLoS One 7:e49755. doi: 10.1371/journal.pone.0049755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Koeppel A, Perry EB, Sikorski J, Krizanc D, Warner A, Ward DM, Rooney AP, Brambilla E, Connor N, Ratcliff RM, Nevo E, Cohan FM. 2008. Identifying the fundamental units of bacterial diversity: a paradigm shift to incorporate ecology into bacterial systematics. Proc Natl Acad Sci U S A 105:2504–2509. doi: 10.1073/pnas.0712205105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Cornejo OE, Hickey RJ, Suzuki H, Forney LJ. 2018. Focusing the diversity of Gardnerella vaginalis through the lens of ecotypes. Evol Appl 11:312–324. doi: 10.1111/eva.12555. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Muzny CA, Taylor CM, Swords WE, Tamhane A, Chattopadhyay D, Cerca N, Schwebke JR. 2019. An updated conceptual model on the pathogenesis of bacterial vaginosis. J Infect Dis 220:1399–1405. doi: 10.1093/infdis/jiz342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Khan S, Voordouw MJ, Hill JE. 2019. Competition among Gardnerella subgroups from the human vaginal microbiome. Front Cell Infect Microbiol 9:374. doi: 10.3389/fcimb.2019.00374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Shipitsyna E, Krysanova A, Khayrullina G, Shalepo K, Savicheva A, Guschin A, Unemo M. 2019. Quantitation of all four gardnerella vaginalis clades detects abnormal vaginal microbiota characteristic of bacterial vaginosis more accurately than putative g. vaginalis sialidase a gene count. Mol Diagn Ther 23:139–147. doi: 10.1007/s40291-019-00382-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Freitas AC, Bocking A, Hill JE, Money DM, Group VR, the VOGUE Research Group . 2018. Increased richness and diversity of the vaginal microbiota and spontaneous preterm birth. Microbiome 6:117. doi: 10.1186/s40168-018-0502-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Albert AY, Chaban B, Wagner EC, Schellenberg JJ, Links MG, van Schalkwyk J, Reid G, Hemmingsen SM, Hill JE, Money D, Group VR, VOGUE Research Group . 2015. A study of the vaginal microbiome in healthy canadian women utilizing cpn60-based molecular profiling reveals distinct gardnerella subgroup community state types. PLoS One 10:e0135620. doi: 10.1371/journal.pone.0135620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Schellenberg JJ, Oh AY, Hill JE. 2017. Microbial profiling of cpn60 universal target sequences in artificial mixtures of vaginal bacteria sampled by nylon swabs or self-sampling devices under different storage conditions. J Microbiol Methods 136:57–64. doi: 10.1016/j.mimet.2017.03.003. [DOI] [PubMed] [Google Scholar]
- 52.Plummer EL, Vodstrcil LA, Murray GL, Fairley CK, Danielewski JA, Garland SM, Chow EPF, Bulach DM, Fethers KA, Hocking JS, Bradshaw CS. 2020. Gardnerella vaginalis clade distribution is associated with behavioral practices and nugent score in women who have sex with women. J Infect Dis 221:454–463. [DOI] [PubMed] [Google Scholar]
- 53.Khan S, Vancuren SJ, Hill JE. 2021. A generalist lifestyle allows rare Gardnerella spp. to persist at low levels in the vaginal microbiome. Microb Ecol 82:1048–1060. doi: 10.1007/s00248-020-01643-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Balashov SV, Mordechai E, Adelson ME, Gygax SE. 2014. Identification, quantification and subtyping of Gardnerella vaginalis in noncultured clinical vaginal samples by quantitative PCR. J Med Microbiol 63:162–175. doi: 10.1099/jmm.0.066407-0. [DOI] [PubMed] [Google Scholar]
- 55.Turner E, Sobel JD, Akins RA. 2021. Prognosis of recurrent bacterial vaginosis based on longitudinal changes in abundance of Lactobacillus and specific species of Gardnerella. PLoS One 16:e0256445. doi: 10.1371/journal.pone.0256445. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Schuyler JA, Mordechai E, Adelson ME, Sobel JD, Gygax SE, Hilbert DW. 2016. Identification of intrinsically metronidazole-resistant clades of Gardnerella vaginalis. Diagn Microbiol Infect Dis 84:1–3. doi: 10.1016/j.diagmicrobio.2015.10.006. [DOI] [PubMed] [Google Scholar]
- 57.Hilbert DW, Schuyler JA, Adelson ME, Mordechai E, Sobel JD, Gygax SE. 2017. Gardnerella vaginalis population dynamics in bacterial vaginosis. Eur J Clin Microbiol Infect Dis 36:1269–1278. doi: 10.1007/s10096-017-2933-8. [DOI] [PubMed] [Google Scholar]
- 58.Castro J, Alves P, Sousa C, Cereija T, França Â, Jefferson KK, Cerca N. 2015. Using an in-vitro biofilm model to assess the virulence potential of bacterial vaginosis or non-bacterial vaginosis Gardnerella vaginalis isolates. Sci Rep 5:11640. doi: 10.1038/srep11640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Bohr LL, Mortimer TD, Pepperell CS. 2020. Lateral gene transfer shapes diversity of Gardnerella spp. Front Cell Infect Microbiol 10:293. doi: 10.3389/fcimb.2020.00293. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Cook RL, Reid G, Pond DG, Schmitt CA, Sobel JD. 1989. Clue cells in bacterial vaginosis: immunofluorescent identification of the adherent gram-negative bacteria as Gardnerella vaginalis. J Infect Dis 160:490–496. doi: 10.1093/infdis/160.3.490. [DOI] [PubMed] [Google Scholar]
- 61.Amsel R, Totten PA, Spiegel CA, Chen KC, Eschenbach D, Holmes KK. 1983. Nonspecific vaginitis. Diagnostic criteria and microbial and epidemiologic associations. Am J Med 74:14–22. doi: 10.1016/0002-9343(83)91112-9. [DOI] [PubMed] [Google Scholar]
- 62.Swidsinski A, Mendling W, Loening-Baucke V, Ladhoff A, Swidsinski S, Hale LP, Lochs H. 2005. Adherent biofilms in bacterial vaginosis. Obstet Gynecol 106:1013–1023. doi: 10.1097/01.AOG.0000183594.45524.d2. [DOI] [PubMed] [Google Scholar]
- 63.Swidsinski A, Doerffel Y, Loening-Baucke V, Swidsinski S, Verstraelen H, Vaneechoutte M, Lemm V, Schilling J, Mendling W. 2010. Gardnerella biofilm involves females and males and is transmitted sexually. Gynecol Obstet Invest 70:256–263. doi: 10.1159/000314015. [DOI] [PubMed] [Google Scholar]
- 64.Patterson JL, Stull-Lane A, Girerd PH, Jefferson KK. 2010. Analysis of adherence, biofilm formation and cytotoxicity suggests a greater virulence potential of Gardnerella vaginalis relative to other bacterial-vaginosis-associated anaerobes. Microbiology (Reading) 156:392–399. doi: 10.1099/mic.0.034280-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Janulaitiene M, Gegzna V, Baranauskiene L, Bulavaitė A, Simanavicius M, Pleckaityte M. 2018. Phenotypic characterization of Gardnerella vaginalis subgroups suggests differences in their virulence potential. PLoS One 13:e0200625. doi: 10.1371/journal.pone.0200625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Castro J, Machado D, Cerca N. 2019. Unveiling the role of Gardnerella vaginalis in polymicrobial Bacterial Vaginosis biofilms: the impact of other vaginal pathogens living as neighbors. ISME J 13:1306–1317. doi: 10.1038/s41396-018-0337-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Bonnardel F, Haslam SM, Dell A, Feizi T, Liu Y, Tajadura-Ortega V, Akune Y, Sykes L, Bennett PR, MacIntyre DA, Lisacek F, Imberty A. 2021. Proteome-wide prediction of bacterial carbohydrate-binding proteins as a tool for understanding commensal and pathogen colonisation of the vaginal microbiome. NPJ Biofilms Microbiomes 7:49. doi: 10.1038/s41522-021-00220-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Marín E, Haesaert A, Padilla L, Adán J, Hernáez ML, Monteoliva L, Gil C. 2018. Unraveling Gardnerella vaginalis surface proteins using cell shaving proteomics. Front Microbiol 9:975. doi: 10.3389/fmicb.2018.00975. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Álvarez S, Leiva-Sabadini C, Schuh CMAP, Aguayo S. 2022. Bacterial adhesion to collagens: implications for biofilm formation and disease progression in the oral cavity. Crit Rev Microbiol 48:83–95. doi: 10.1080/1040841X.2021.1944054. [DOI] [PubMed] [Google Scholar]
- 70.Jefferson KK. 2004. What drives bacteria to produce a biofilm? FEMS Microbiol Lett 236:163–173. doi: 10.1016/j.femsle.2004.06.005. [DOI] [PubMed] [Google Scholar]
- 71.Castro J, Franca A, Bradwell KR, Serrano MG, Jefferson KK, Cerca N. 2017. Comparative transcriptomic analysis of Gardnerella vaginalis biofilms vs. planktonic cultures using RNA-seq. NPJ Biofilms Microbiomes 3:3. doi: 10.1038/s41522-017-0012-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Gottschick C, Szafranski SP, Kunze B, Sztajer H, Masur C, Abels C, Wagner-Döbler I. 2016. Screening of compounds against Gardnerella vaginalis biofilms. PLoS One 11:e0154086. doi: 10.1371/journal.pone.0154086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li T, Zhang Z, Wang F, He Y, Zong X, Bai H, Liu Z. 2020. Antimicrobial Susceptibility Testing of Metronidazole and Clindamycin against Gardnerella vaginalis in Planktonic and Biofilm formation. Can J Infect Dis Med Microbiol 2020:1361825. doi: 10.1155/2020/1361825. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Patterson JL, Girerd PH, Karjane NW, Jefferson KK. 2007. Effect of biofilm phenotype on resistance of Gardnerella vaginalis to hydrogen peroxide and lactic acid. Am J Obstet Gynecol 197:170.e1-7–170.e7. doi: 10.1016/j.ajog.2007.02.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Hymes SR, Randis TM, Sun TY, Ratner AJ. 2013. DNase inhibits Gardnerella vaginalis biofilms in vitro and in vivo. J Infect Dis 207:1491–1497. doi: 10.1093/infdis/jit047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Amegashie CP, Gilbert NM, Peipert JF, Allsworth JE, Lewis WG, Lewis AL. 2017. Relationship between nugent score and vaginal epithelial exfoliation. PLoS One 12:e0177797. doi: 10.1371/journal.pone.0177797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Macierzanka A, Mackie AR, Bajka BH, Rigby NM, Nau F, Dupont D. 2014. Transport of particles in intestinal mucus under simulated infant and adult physiological conditions: impact of mucus structure and extracellular DNA. PLoS One 9:e95274. doi: 10.1371/journal.pone.0095274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Yamada KJ, Kielian T. 2019. Biofilm-leukocyte cross-talk: impact on immune polarization and immunometabolism. J Innate Immun 11:280–288. doi: 10.1159/000492680. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Löfmark S, Edlund C, Nord CE. 2010. Metronidazole is still the drug of choice for treatment of anaerobic infections. Clin Infect Dis 50 Suppl 1:S16–23. doi: 10.1086/647939. [DOI] [PubMed] [Google Scholar]
- 80.Chen KC, Forsyth PS, Buchanan TM, Holmes KK. 1979. Amine content of vaginal fluid from untreated and treated patients with nonspecific vaginitis. J Clin Invest 63:828–835. doi: 10.1172/JCI109382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Pybus V, Onderdonk AB. 1997. Evidence for a commensal, symbiotic relationship between Gardnerella vaginalis and Prevotella bivia involving ammonia: potential significance for bacterial vaginosis. J Infect Dis 175:406–413. doi: 10.1093/infdis/175.2.406. [DOI] [PubMed] [Google Scholar]
- 82.Nelson TM, Borgogna JL, Brotman RM, Ravel J, Walk ST, Yeoman CJ. 2015. Vaginal biogenic amines: biomarkers of bacterial vaginosis or precursors to vaginal dysbiosis? Front Physiol 6:253. doi: 10.3389/fphys.2015.00253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Rutherford ST, Bassler BL. 2012. Bacterial quorum sensing: its role in virulence and possibilities for its control. Cold Spring Harb Perspect Med 2:a012427. doi: 10.1101/cshperspect.a012427. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Algburi A, Zehm S, Netrebov V, Weeks R, Zubovskiy K, Chikindas ML. 2018. Benzoyl peroxide inhibits quorum sensing and biofilm formation by gardnerella vaginalis 14018. Infect Dis Obstet Gynecol 2018:1426109. doi: 10.1155/2018/1426109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Algburi A, Zehm S, Netrebov V, Bren AB, Chistyakov V, Chikindas ML. 2017. Subtilosin prevents biofilm formation by inhibiting bacterial quorum sensing. Probiotics & Antimicro Prot 9:81–90. doi: 10.1007/s12602-016-9242-x. [DOI] [PubMed] [Google Scholar]
- 86.Rosca AS, Castro J, França Â, Vaneechoutte M, Cerca N. 2021. Gardnerella vaginalis dominates multi-species biofilms in both pre-conditioned and competitive in vitro biofilm formation models. Microb Ecol 84: 1278–1287. doi: 10.1007/s00248-021-01917-2. [DOI] [PubMed] [Google Scholar]
- 87.Rosca AS, Castro J, Cerca N. 2020. Evaluation of different culture media to support in vitro growth and biofilm formation of bacterial vaginosis-associated anaerobes. PeerJ 8:e9917. doi: 10.7717/peerj.9917. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.He Y, Na R, Niu X, Xiao B, Yang H. 2021. Lactobacillus rhamnosus and Lactobacillus casei affect various stages of gardnerella species biofilm Formation. Front Cell Infect Microbiol 11:568178. doi: 10.3389/fcimb.2021.568178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Gelber SE, Aguilar JL, Lewis KL, Ratner AJ. 2008. Functional and phylogenetic characterization of Vaginolysin, the human-specific cytolysin from Gardnerella vaginalis. J Bacteriol 190:3896–3903. doi: 10.1128/JB.01965-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Randis TM, Zaklama J, LaRocca TJ, Los FC, Lewis EL, Desai P, Rampersaud R, Amaral FE, Ratner AJ. 2013. Vaginolysin drives epithelial ultrastructural responses to Gardnerella vaginalis. Infect Immun 81:4544–4550. doi: 10.1128/IAI.00627-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Reimer LG, Reller LB. 1984. Gardnerella vaginalis bacteremia: a review of thirty cases. Obstet Gynecol 64:170–172. [PubMed] [Google Scholar]
- 92.Shishpal P, Kasarpalkar N, Singh D, Bhor VM. 2020. Characterization of Gardnerella vaginalis membrane vesicles reveals a role in inducing cytotoxicity in vaginal epithelial cells. Anaerobe 61:102090. doi: 10.1016/j.anaerobe.2019.102090. [DOI] [PubMed] [Google Scholar]
- 93.Shishpal P, Patel V, Singh D, Bhor VM. 2021. pH Stress mediated alteration in protein composition and reduction in cytotoxic potential of Gardnerella vaginalis membrane vesicles. Front Microbiol 12:723909. doi: 10.3389/fmicb.2021.723909. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Pleckaityte M. 2020. Cholesterol-dependent cytolysins produced by vaginal bacteria: certainties and controversies. Front Cell Infect Microbiol 9:452. doi: 10.3389/fcimb.2019.00452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Garcia EM, Serrano MG, Edupuganti L, Edwards DJ, Buck GA, Jefferson KK. 2021. Sequence comparison of Vaginolysin from different Gardnerella species. Pathogens 10:86. doi: 10.3390/pathogens10020086. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Rottini G, Dobrina A, Forgiarini O, Nardon E, Amirante GA, Patriarca P. 1990. Identification and partial characterization of a cytolytic toxin produced by Gardnerella vaginalis. Infect Immun 58:3751–3758. doi: 10.1128/iai.58.11.3751-3758.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Garcia EM, Kraskauskiene V, Koblinski JE, Jefferson KK. 2019. Interaction of Gardnerella vaginalis and Vaginolysin with the apical versus basolateral face of a three-dimensional model of vaginal epithelium. Infect Immun 87:e00646-18–e00618. doi: 10.1128/IAI.00646-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Castro J, Martins AP, Rodrigues ME, Cerca N. 2018. Lactobacillus crispatus represses vaginolysin expression by BV associated Gardnerella vaginalis and reduces cell cytotoxicity. Anaerobe 50:60–63. doi: 10.1016/j.anaerobe.2018.01.014. [DOI] [PubMed] [Google Scholar]
- 99.Cauci S, Guaschino S, Driussi S, De Santo D, Lanzafame P, Quadrifoglio F. 2002. Correlation of local interleukin-8 with immunoglobulin A against Gardnerella vaginalis hemolysin and with prolidase and sialidase levels in women with bacterial vaginosis. J Infect Dis 185:1614–1620. doi: 10.1086/340417. [DOI] [PubMed] [Google Scholar]
- 100.Cauci S, Monte R, Ropele M, Missero C, Not T, Quadrifoglio F, Menestrina G. 1993. Pore-forming and haemolytic properties of the Gardnerella vaginalis cytolysin. Mol Microbiol 9:1143–1155. doi: 10.1111/j.1365-2958.1993.tb01244.x. [DOI] [PubMed] [Google Scholar]
- 101.Cauci S, Thorsen P, Schendel DE, Bremmelgaard A, Quadrifoglio F, Guaschino S. 2003. Determination of immunoglobulin A against Gardnerella vaginalis hemolysin, sialidase, and prolidase activities in vaginal fluid: implications for adverse pregnancy outcomes. J Clin Microbiol 41:435–438. doi: 10.1128/JCM.41.1.435-438.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Cauci S, Monte R, Driussi S, Lanzafame P, Quadrifoglio F. 1998. Impairment of the mucosal immune system: IgA and IgM cleavage detected in vaginal washings of a subgroup of patients with bacterial vaginosis. J Infect Dis 178:1698–1706. doi: 10.1086/314505. [DOI] [PubMed] [Google Scholar]
- 103.Amabebe E, Anumba DOC. 2018. The vaginal microenvironment: the physiologic role of Lactobacilli. Front Med 5:181. doi: 10.3389/fmed.2018.00181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lewis WG, Robinson LS, Gilbert NM, Perry JC, Lewis AL. 2013. Degradation, foraging, and depletion of mucus sialoglycans by the vagina-adapted Actinobacterium Gardnerella vaginalis. J Biol Chem 288:12067–12079. doi: 10.1074/jbc.M113.453654. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Agarwal K, Choudhury B, Robinson LS, Allsworth JE, Lewis AL, Lewis WG. 2022. Resident microbes shape the vaginal epithelial glycan landscape. medRxiv. doi: 10.1101/2022.02.23.22271417. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Hardy L, Jespers V, Van den Bulck M, Buyze J, Mwambarangwe L, Musengamana V, Vaneechoutte M, Crucitti T. 2017. The presence of the putative Gardnerella vaginalis sialidase A gene in vaginal specimens is associated with bacterial vaginosis biofilm. PLoS One 12:e0172522. doi: 10.1371/journal.pone.0172522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Rodriguez-Nava C, Cortes-Sarabia K, Avila-Huerta MD, Ortiz-Riano EJ, Estrada-Moreno AK, Alarcon-Romero LDC, Mata-Ruiz O, Medina-Flores Y, Vences-Velazquez A, Morales-Narvaez E. 2021. Nanophotonic sialidase immunoassay for bacterial vaginosis diagnosis. ACS Pharmacol Transl Sci 4:365–371. doi: 10.1021/acsptsci.0c00211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Wu S, Lin X, Hui KM, Yang S, Wu X, Tan Y, Li M, Qin AQ, Wang Q, Zhao Q, Ding P, Shi K, Li XJ. 2019. A biochemiluminescent sialidase assay for diagnosis of bacterial vaginosis. Sci Rep 9:20024. doi: 10.1038/s41598-019-56371-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Coleman JS, Gaydos CA. 2018. Molecular diagnosis of bacterial vaginosis: an update. J Clin Microbiol 56:e00342-18–e00318. doi: 10.1128/JCM.00342-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Borgdorff H, Gautam R, Armstrong SD, Xia D, Ndayisaba GF, van Teijlingen NH, Geijtenbeek TB, Wastling JM, van de Wijgert JH. 2016. Cervicovaginal microbiome dysbiosis is associated with proteome changes related to alterations of the cervicovaginal mucosal barrier. Mucosal Immunol 9:621–633. doi: 10.1038/mi.2015.86. [DOI] [PubMed] [Google Scholar]
- 111.Robinson LS, Schwebke J, Lewis WG, Lewis AL. 2019. Identification and characterization of NanH2 and NanH3, enzymes responsible for sialidase activity in the vaginal bacterium Gardnerella vaginalis. J Biol Chem 294:5230–5245. doi: 10.1074/jbc.RA118.006221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Kurukulasuriya SP, Patterson MH, Hill JE. 2021. Slipped-strand mispairing in the gene encoding sialidase NanH3 in Gardnerella spp. Infect Immun 89:e00583-20–e00520. doi: 10.1128/IAI.00583-20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Boustouller YL, Johnson AP, Taylor-Robinson D. 1987. Pili on Gardnerella vaginalis studied by electronmicroscopy. J Med Microbiol 23:327–329. doi: 10.1099/00222615-23-4-327. [DOI] [PubMed] [Google Scholar]
- 114.Johnson AP, Davies HA. 1984. Demonstration by electron microscopy of pili on Gardnerella vaginalis. Br J Vener Dis 60:396–397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Jarosik GP, Land CB, Duhon P, Chandler R, Mercer T. 1998. Acquisition of iron by Gardnerella vaginalis. Infect Immun 66:5041–5047. doi: 10.1128/IAI.66.10.5041-5047.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Srinivasan S, Liu C, Mitchell CM, Fiedler TL, Thomas KK, Agnew KJ, Marrazzo JM, Fredricks DN. 2010. Temporal variability of human vaginal bacteria and relationship with bacterial vaginosis. PLoS One 5:e10197. doi: 10.1371/journal.pone.0010197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Litwin CM, Calderwood SB. 1993. Role of iron in regulation of virulence genes. Clin Microbiol Rev 6:137–149. doi: 10.1128/CMR.6.2.137. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Chen X, Alonzo F. 2019. Bacterial lipolysis of immune-activating ligands promotes evasion of innate defenses. Proc Natl Acad Sci U S A 116:3764–3773. doi: 10.1073/pnas.1817248116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Thurman AR, Kimble T, Herold B, Mesquita PM, Fichorova RN, Dawood HY, Fashemi T, Chandra N, Rabe L, Cunningham TD, Anderson S, Schwartz J, Doncel G. 2015. Bacterial vaginosis and subclinical markers of genital tract inflammation and mucosal immunity. AIDS Res Hum Retroviruses 31:1139–1152. doi: 10.1089/aid.2015.0006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Masson L, Barnabas S, Deese J, Lennard K, Dabee S, Gamieldien H, Jaumdally SZ, Williamson AL, Little F, Van Damme L, Ahmed K, Crucitti T, Abdellati S, Bekker LG, Gray G, Dietrich J, Jaspan H, Passmore JS. 2019. Inflammatory cytokine biomarkers of asymptomatic sexually transmitted infections and vaginal dysbiosis: a multicentre validation study. Sex Transm Infect 95:5–12. doi: 10.1136/sextrans-2017-053506. [DOI] [PubMed] [Google Scholar]
- 121.Mitchell C, Marrazzo J. 2014. Bacterial vaginosis and the cervicovaginal immune response. Am J Reprod Immunol 71:555–563. doi: 10.1111/aji.12264. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Monin L, Whettlock EM, Male V. 2020. Immune responses in the human female reproductive tract. Immunology 160:106–115. doi: 10.1111/imm.13136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Bertran T, Brachet P, Vareille-Delarbre M, Falenta J, Dosgilbert A, Vasson MP, Forestier C, Tridon A, Evrard B. 2016. Slight pro-inflammatory immunomodulation properties of dendritic cells by Gardnerella vaginalis: the “invisible man” of bacterial vaginosis? J Immunol Res 2016:9747480. doi: 10.1155/2016/9747480. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.van Teijlingen NH, Helgers LC, Zijlstra-Willems EM, van Hamme JL, Ribeiro CMS, Strijbis K, Geijtenbeek TBH. 2020. Vaginal dysbiosis associated-bacteria Megasphaera elsdenii and Prevotella timonensis induce immune activation via dendritic cells. J Reprod Immunol 138:103085. doi: 10.1016/j.jri.2020.103085. [DOI] [PubMed] [Google Scholar]
- 125.St John EP, Martinson J, Simoes JA, Landay AL, Spear GT. 2007. Dendritic cell activation and maturation induced by mucosal fluid from women with bacterial vaginosis. Clin Immunol 125:95–102. doi: 10.1016/j.clim.2007.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Nicolo S, Tanturli M, Mattiuz G, Antonelli A, Baccani I, Bonaiuto C, Baldi S, Nannini G, Menicatti M, Bartolucci G, Rossolini GM, Amedei A, Torcia MG. 2021. Vaginal Lactobacilli and vaginal dysbiosis-associated bacteria differently affect cervical epithelial and immune homeostasis and anti-viral defenses. Int J Mol Sci 22:6487. doi: 10.3390/ijms22126487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Xiang N, Yin T, Chen T. 2021. Gardnerella vaginalis induces NLRP3 inflammasome-mediated pyroptosis in macrophages and THP-1 monocytes. Exp Ther Med 22:1174. doi: 10.3892/etm.2021.10609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Liu CW, Su BC, Chen JY. 2021. Tilapia Piscidin 4 (TP4) reprograms M1 macrophages to M2 phenotypes in cell models of Gardnerella vaginalis-induced vaginosis. Front Immunol 12:773013. doi: 10.3389/fimmu.2021.773013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Vick EJ, Park HS, Huff KA, Brooks KM, Farone AL, Farone MB. 2014. Gardnerella vaginalis triggers NLRP3 inflammasome recruitment in THP-1 monocytes. J Reprod Immunol 106:67–75. doi: 10.1016/j.jri.2014.08.005. [DOI] [PubMed] [Google Scholar]
- 130.Paharik AE, Horswill AR. 2016. The staphylococcal biofilm: adhesins, regulation, and host response. Microbiol Spectr 4. doi: 10.1128/microbiolspec.VMBF-0022-2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Mares D, Simoes JA, Novak RM, Spear GT. 2008. TLR2-mediated cell stimulation in bacterial vaginosis. J Reprod Immunol 77:91–99. doi: 10.1016/j.jri.2007.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Swidsinski A, Verstraelen H, Loening-Baucke V, Swidsinski S, Mendling W, Halwani Z. 2013. Presence of a polymicrobial endometrial biofilm in patients with bacterial vaginosis. PLoS One 8:e53997. doi: 10.1371/journal.pone.0053997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Santos CMA, Pires MCV, Leão TL, Silva AKS, Miranda LS, Martins FS, Silva AM, Nicoli JR. 2018. Anti-inflammatory effect of two Lactobacillus strains during infection with Gardnerella vaginalis and Candida albicans in a HeLa cell culture model. Microbiology (Reading) 164:349–358. doi: 10.1099/mic.0.000608. [DOI] [PubMed] [Google Scholar]
- 134.Hashemi FB, Ghassemi M, Roebuck KA, Spear GT. 1999. Activation of human immunodeficiency virus type 1 expression by Gardnerella vaginalis. J Infect Dis 179:924–930. doi: 10.1086/314674. [DOI] [PubMed] [Google Scholar]
- 135.Brosnahan AJ, Merriman JA, Salgado-Pabón W, Ford B, Schlievert PM. 2013. Enterococcus faecalis inhibits superantigen toxic shock syndrome toxin-1-induced interleukin-8 from human vaginal epithelial cells through tetramic acids. PLoS One 8:e61255. doi: 10.1371/journal.pone.0061255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Sierra LJ, Brown AG, Barila GO, Anton L, Barnum CE, Shetye SS, Soslowsky LJ, Elovitz MA. 2018. Colonization of the cervicovaginal space with Gardnerella vaginalis leads to local inflammation and cervical remodeling in pregnant mice. PLoS One 13:e0191524. doi: 10.1371/journal.pone.0191524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Anton L, Sierra LJ, DeVine A, Barila G, Heiser L, Brown AG, Elovitz MA. 2018. Common cervicovaginal microbial supernatants alter cervical epithelial function: mechanisms by which Lactobacillus crispatus contributes to cervical health. Front Microbiol 9:2181. doi: 10.3389/fmicb.2018.02181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Manhanzva MT, Abrahams AG, Gamieldien H, Froissart R, Jaspan H, Jaumdally SZ, Barnabas SL, Dabee S, Bekker LG, Gray G, Passmore JS, Masson L. 2020. Inflammatory and antimicrobial properties differ between vaginal Lactobacillus isolates from South African women with non-optimal versus optimal microbiota. Sci Rep 10:6196. doi: 10.1038/s41598-020-62184-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Fichorova RN, DeLong AK, Cu-Uvin S, King CC, Jamieson DJ, Klein RS, Sobel JD, Vlahov D, Yamamoto HS, Mayer KH. 2021. Protozoan-viral-bacterial co-infections alter galectin levels and associated immunity mediators in the female genital tract. Front Cell Infect Microbiol 11:649940. doi: 10.3389/fcimb.2021.649940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Cauci S, Guaschino S, De Aloysio D, Driussi S, Santo DD, Penacchioni P, Quadrifoglio F. 2003. Interrelationships of interleukin-8 with interleukin-1beta and neutrophils in vaginal fluid of healthy and bacterial vaginosis positive women. Mol Hum Reprod 9:53–58. doi: 10.1093/molehr/gag003. [DOI] [PubMed] [Google Scholar]
- 141.Choi SI, Won G, Kim Y, Kang CH, Kim GH. 2022. Lactobacilli strain mixture alleviates bacterial vaginosis through antibacterial and antagonistic activity in gardnerella vaginalis-infected C57BL/6 Mice. Microorganisms 10:471. doi: 10.3390/microorganisms10020471. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Nasu K, Narahara H. 2010. Pattern recognition via the toll-like receptor system in the human female genital tract. Mediators Inflamm 2010:976024. doi: 10.1155/2010/976024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Eade CR, Diaz C, Wood MP, Anastos K, Patterson BK, Gupta P, Cole AL, Cole AM. 2012. Identification and characterization of bacterial vaginosis-associated pathogens using a comprehensive cervical-vaginal epithelial coculture assay. PLoS One 7:e50106. doi: 10.1371/journal.pone.0050106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Antonia AL, Gibbs KD, Trahair ED, Pittman KJ, Martin AT, Schott BH, Smith JS, Rajagopal S, Thompson JW, Reinhardt RL, Ko DC. 2019. Pathogen evasion of chemokine response through suppression of CXCL10. Front Cell Infect Microbiol 9:280. doi: 10.3389/fcimb.2019.00280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Jauregui CE, Wang Q, Wright CJ, Takeuchi H, Uriarte SM, Lamont RJ. 2013. Suppression of T-cell chemokines by Porphyromonas gingivalis. Infect Immun 81:2288–2295. doi: 10.1128/IAI.00264-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Joag V, Obila O, Gajer P, Scott MC, Dizzell S, Humphrys M, Shahabi K, Huibner S, Shannon B, Tharao W, Mureithi M, Oyugi J, Kimani J, Kaushic C, Ravel J, Anzala O, Kaul R. 2019. Impact of standard bacterial vaginosis treatment on the genital microbiota, immune milieu, and ex vivo human immunodeficiency virus susceptibility. Clin Infect Dis 68:1675–1683. doi: 10.1093/cid/ciy762. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Groom JR, Luster AD. 2011. CXCR3 ligands: redundant, collaborative and antagonistic functions. Immunol Cell Biol 89:207–215. doi: 10.1038/icb.2010.158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Delgado-Diaz DJ, Tyssen D, Hayward JA, Gugasyan R, Hearps AC, Tachedjian G. 2020. Distinct immune responses elicited from cervicovaginal epithelial cells by lactic acid and short chain fatty acids associated with optimal and non-optimal vaginal microbiota. Front Cell Infect Microbiol 9:446. doi: 10.3389/fcimb.2019.00446. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Pararas MV, Skevaki CL, Kafetzis DA. 2006. Preterm birth due to maternal infection: causative pathogens and modes of prevention. Eur J Clin Microbiol Infect Dis 25:562–569. doi: 10.1007/s10096-006-0190-3. [DOI] [PubMed] [Google Scholar]
- 150.Zaga-Clavellina V, Martha RV, Flores-Espinosa P. 2012. In vitro secretion profile of pro-inflammatory cytokines IL-1β, TNF-α, IL-6, and of human beta-defensins (HBD)-1, HBD-2, and HBD-3 from human chorioamniotic membranes after selective stimulation with Gardnerella vaginalis. Am J Reprod Immunol 67:34–43. doi: 10.1111/j.1600-0897.2011.01054.x. [DOI] [PubMed] [Google Scholar]
- 151.Noda-Nicolau NM, Polettini J, da Silva MG, Peltier MR, Menon R. 2018. Polybacterial stimulation suggests discrete IL-6/IL-6R signaling in human fetal membranes: potential implications on IL-6 bioactivity. J Reprod Immunol 126:60–68. doi: 10.1016/j.jri.2018.02.007. [DOI] [PubMed] [Google Scholar]
- 152.Gilbert NM, Foster LR, Cao B, Yin Y, Mysorekar IU, Lewis AL. 2021. Gardnerella vaginalis promotes group B Streptococcus vaginal colonization, enabling ascending uteroplacental infection in pregnant mice. Am J Obstet Gynecol 224:530.e1–530.e17. doi: 10.1016/j.ajog.2020.11.032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Noda-Nicolau NM, Polettini J, Peltier MR, da Silva MG, Menon R. 2016. Combinations and loads of bacteria affect the cytokine production by fetal membranes: an in vitro study. Am J Reprod Immunol 76:504–511. doi: 10.1111/aji.12596. [DOI] [PubMed] [Google Scholar]
- 154.Zaga-Clavellina V, Flores-Espinosa P, Pineda-Torres M, Sosa-González I, Vega-Sánchez R, Estrada-Gutierrez G, Espejel-Núñez A, Flores-Pliego A, Maida-Claros R, Estrada-Juárez H, Chávez-Mendoza A. 2014. Tissue-specific IL-10 secretion profile from term human fetal membranes stimulated with pathogenic microorganisms associated with preterm labor in a two-compartment tissue culture system. J Matern Fetal Neonatal Med 27:1320–1327. doi: 10.3109/14767058.2013.857397. [DOI] [PubMed] [Google Scholar]
- 155.Peltier MR, Drobek CO, Bhat G, Saade G, Fortunato SJ, Menon R. 2012. Amniotic fluid and maternal race influence responsiveness of fetal membranes to bacteria. J Reprod Immunol 96:68–78. doi: 10.1016/j.jri.2012.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Abrahams VM, Potter JA, Bhat G, Peltier MR, Saade G, Menon R. 2013. Bacterial modulation of human fetal membrane Toll-like receptor expression. Am J Reprod Immunol 69:33–40. doi: 10.1111/aji.12016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Kacerovsky M, Pliskova L, Bolehovska R, Lesko D, Gerychova R, Janku P, Matlak P, Simetka O, Stranik J, Faist T, Mls J, Vescicik P, Jacobsson B, Musilova I. 2021. Cervical Gardnerella vaginalis in women with preterm prelabor rupture of membranes. PLoS One 16:e0245937. doi: 10.1371/journal.pone.0245937. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Łaniewski P, Gomez A, Hire G, So M, Herbst-Kralovetz MM. 2017. Human three-dimensional endometrial epithelial cell model to study host interactions with vaginal bacteria and Neisseria gonorrhoeae. Infect Immun 85:e01049-16–e01016. doi: 10.1128/IAI.01049-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.Schellenberg JJ, Patterson MH, Hill JE. 2017. Gardnerella vaginalis diversity and ecology in relation to vaginal symptoms. Res Microbiol 168:837–844. doi: 10.1016/j.resmic.2017.02.011. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Table S1. Download iai.00390-22-s0001.pdf, PDF file, 0.4 MB (367.6KB, pdf)

