Sie sind auf Seite 1von 18

Critical Reviews in Microbiology

ISSN: 1040-841X (Print) 1549-7828 (Online) Journal homepage: http://www.tandfonline.com/loi/imby20

Etiology of bacterial vaginosis and polymicrobial


biofilm formation

Hyun-Sul Jung, Marthie M. Ehlers, Hennie Lombaard, Mathys J. Redelinghuys


& Marleen M. Kock

To cite this article: Hyun-Sul Jung, Marthie M. Ehlers, Hennie Lombaard, Mathys J. Redelinghuys
& Marleen M. Kock (2017): Etiology of bacterial vaginosis and polymicrobial biofilm formation,
Critical Reviews in Microbiology, DOI: 10.1080/1040841X.2017.1291579

To link to this article: http://dx.doi.org/10.1080/1040841X.2017.1291579

Published online: 30 Mar 2017.

Submit your article to this journal

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=imby20

Download by: [FU Berlin] Date: 31 March 2017, At: 01:13


CRITICAL REVIEWS IN MICROBIOLOGY, 2017
http://dx.doi.org/10.1080/1040841X.2017.1291579

REVIEW ARTICLE

Etiology of bacterial vaginosis and polymicrobial biofilm formation


Hyun-Sul Junga, Marthie M. Ehlersa,b, Hennie Lombaardc, Mathys J. Redelinghuysa and Marleen M. Kocka,b
a
Department of Medical Microbiology, Faculty of Health Sciences, University of Pretoria, Pretoria, South Africa; bDepartment of Medical
Microbiology, Tshwane Academic Division, National Health Laboratory Service (NHLS), Pretoria, South Africa; cGauteng Department of
Health, Rahima Moosa Mother and Child Hospital, Wits Obstetrics and Gynaecology Clinical Research Division, Department of
Obstetrics and Gynaecology, University of Witwatersrand, Johannesburg, South Africa

ABSTRACT ARTICLE HISTORY


Microorganisms in nature rarely exist in a planktonic form, but in the form of biofilms. Biofilms Received 10 June 2016
have been identified as the cause of many chronic and persistent infections and have been impli- Revised 26 January 2017
cated in the etiology of bacterial vaginosis (BV). Bacterial vaginosis is the most common form of Accepted 2 February 2017
vaginal infection in women of reproductive age. Similar to other biofilm infections, BV biofilms Published online 22 March
2017
protect the BV-related bacteria against antibiotics and cause recurrent BV. In this review, an over-
view of BV-related bacteria, conceptual models and the stages involved in the polymicrobial BV KEYWORDS
biofilm formation will be discussed. Biofilm; BVAB; BV;
Gardnerella vaginalis;
pathogenesis

Introduction (Kenyon et al., 2013; Koumans et al., 2007). Bacterial


vaginosis could either be symptomatic or be asymp-
The human vagina is a dynamic, yet balanced ecosys-
tomatic and the rate of symptomatic BV may range
tem that consists of different microbes called the vagi-
from 10% to 66% (Begum et al., 2003; Klebanoff et al.,
nal microbiota (Hyman et al., 2005; Kim et al., 2009).
2004; Schwebke & Desmond, 2007). Women with symp-
The vagina contains a complex population of aerobic
and anaerobic bacteria that can reach up to 109 col- tomatic BV may experience symptoms of vaginal dis-
ony-forming units (CFU) per milliliter of vaginal fluid in comfort (including vaginal or perineal itching) and an
healthy women (Bartlett et al., 1977; Bartlett & Polk, increased amount of thin, foul smelling, homogenous
1984). In most women, the normal vaginal microbiota discharge after unprotected sexual intercourse or at the
are typically dominated by Lactobacillus spp. (Kim time of menstruation (Klebanoff et al., 2004). In contrast,
et al., 2009), which produce lactic acid, hydrogen per- women with asymptomatic BV report no typical BV
oxide (H2O2) (Hawes et al., 1996) and bacteriocins to symptoms and are not generally treated (Klebanoff
inhibit the colonization of other pathogenic microor- et al., 2004; Schwebke & Desmond, 2007).
ganisms (Aroutcheva et al., 2001). Lactobacilli promote Bacterial vaginosis is clinically important, because of
a healthy vaginal environment by acidifying the vagi- the many adverse health sequelae associated with this
nal environment with lactic acid to a pH of 3.5–4.5 condition (Haggerty et al., 2004; Hillier et al., 1995; Ness
(O’Hanlon et al., 2013). It is also known that lactobacilli et al., 2005; Ralph et al., 1999; Wilson et al., 2002). The
enhance the barrier function of the epithelial cells and microbiota associated with BV produce several immuno-
stimulate the host innate immunity (Karczewski et al., modulatory (hemolysins, volatile (VFAs) and nonvolatile
2010). A disturbed vaginal microbiota due to hormonal fatty acids (NVFAs), proteases and sialidases) (Cauci
changes, douching and sexual activity may lead to et al., 2002; Mattsby-Baltzer et al., 1998; Mirmonsef
vaginal colonization with potentially pathogenic bac- et al., 2012) and proinflammatory substances (lipopoly-
teria, typically those causing bacterial vaginosis (BV) saccharides) that induce the production of proinflam-
(Hawes et al., 1996), aerobic vaginitis (AV) (Donders matory cytokines (such as interleukin (IL)-1b, IL-6 and
et al., 2002) and sexually transmitted infections (STIs) tumor necrosis factor (TNF)), which may consequently
(Cherpes et al., 2008). result in preterm birth in pregnant women (Yeganegi
Bacterial vaginosis is the most common reproductive et al., 2009). Sialidase activity may inhibit serum
tract infection in women of reproductive age worldwide immunoglobulin A (sIgA), which allows cytotoxins to

CONTACT Marleen M Kock marleen.kock@up.ac.za Department of Medical Microbiology, University of Pretoria/NHLS, Pretoria, South Africa
ß 2017 University of Pretoria
2 H.-S. JUNG ET AL.

induce premature rupture of membranes (PROM) (Cauci (Fredricks et al., 2005; Hillier et al., 1993). The develop-
et al., 2002). Finally, sialidase and prolidase may also ment of culture-independent molecular techniques like
contribute to the degradation and detachment of vagi- broad-range 16 S ribosomal DNA (rDNA) PCR (Fredricks
nal epithelium and mucins, as well as the increase of et al., 2005), quantitative real-time PCR (qPCR) assays
IL-1b, IL-8 and TNF-a that makes the host more suscep- (Zozaya-Hinchliffe et al., 2010), denaturing gradient gel
tible to human immunodeficiency virus 1 (HIV-1) electrophoresis (DGGE) and 454 pyrosequencing (Ling
(Cauci et al., 2008; Sturm-Ramirez et al., 2000) and et al., 2010; Verstraelen et al., 2004) have improved the
herpes simplex virus 2 (HSV-2) (Cherpes et al., 2003). detection of uncultivable anaerobes, such as BVAB1,
Microbiologically, BV is typically characterized by the BVAB2, BVAB3, Eggerthella spp., Megasphaera type 1,
reduction of H2O2-producing lactobacilli and an over- Leptotrichia spp. and Sneathia spp. from BV-positive
growth of Gardnerella vaginalis and other anaerobic women. BV-related bacteria may have a synergistic rela-
bacteria-like Atopobium vaginae, Bacteroides spp., tionship with each other during BV development and
Mobiluncus spp. and Prevotella spp. (Fredricks et al., may also participate in the formation of a polymicrobial
2005; Ling et al., 2010). These anaerobic bacteria have BV biofilm, which plays an important role in BV etiology
synergistic interactions during BV infection and form a (Alves et al., 2014; Castro & Cerca, 2015).
polymicrobial biofilm, which is considered as one of the
factors contributing to recurrent BV (Swidsinski et al.,
Gardnerella vaginalis
2008).
Biofilm formation is one of the effective strategies of Gardnerella vaginalis is a facultative anaerobic, Gram-
microorganisms to survive in unfavorable environments negative to Gram-variable, nonmotile, small pleo-
(Sapi et al., 2012; Sauer et al., 2002). Biofilms can be morphic rod (Greenwood & Pickett, 1980). Although G.
formed everywhere, either on biotic or on abiotic surfa- vaginalis may not grow at pH 4 (Breshears et al., 2015;
ces, including living or dead tissues or medical devices Greenwood & Pickett, 1979), G. vaginalis can tolerate a
like urinary catheters (Ganderton et al., 1992; Kubota vaginal environment with a high oxidation–reduction
et al., 2008; Lee et al., 2008). Biofilms can play an (redox) potential and can adhere to vaginal epithelial
important pathologic role during the disease process, cells at a vaginal pH between 4 and 5, unlike strict BV
either by preventing full penetration of antibiotics or by anaerobes like Prevotella spp. (Muzny & Schwebke,
allowing microorganisms to escape host defense mech- 2013; Sobel et al., 1981; Udayalaxmi et al., 2012).
anisms (Singh et al., 2010; Thurlow et al., 2011). The Gardnerella vaginalis possesses several virulence factors,
incomplete eradication of biofilms by antibiotics and including biofilm formation, bacteriocins, vaginolysins,
host defenses allows pathogenic bacteria to develop sialidases and proteases (Harwich et al., 2010;
resistance to antibiotic treatment and finally lead to Udayalaxmi et al., 2011; Yeoman et al., 2010) (Table 1).
recurrent infections (Swidsinski et al., 2008, 2011). There is no doubt that G. vaginalis is associated with
This review will provide an overview of the bacteria BV and is involved in the formation of the polymicrobial
associated with BV (BVAB and BV-related bacteria), cur- BV biofilm (Machado et al., 2013; Swidsinski et al., 2005).
rently available conceptual models and theories and However, some G. vaginalis strains can be isolated from
the processes involved in the formation of a polymicro- the vagina of “healthy” women and sexually inexperi-
bial BV biofilm in three main sections. In this review, the enced women and do not necessarily cause BV
term “BV-related bacteria” or “BV-related anaerobes” (Aroutcheva et al., 2001; Fethers et al., 2012). This phe-
will be used to distinguish other bacteria associated nomenon may be explained by a genetic difference
with BV from “BV-associated bacteria 1, 2 and 3 (BVAB1, between commensal and pathogenic strains of G. vagi-
BVAB2 and BVAB3)” that was identified and named by nalis, as reported by several functional and genomic
Fredricks et al. (2005). studies (Castro et al., 2015; Harwich et al., 2010; Yeoman
et al., 2010). The pathogenic strains show greater cyto-
toxicity, greater adherence and biofilm formation ability
Bacteria associated with BV and their roles in
than the commensal strains (Castro et al., 2015; Harwich
BV etiology
et al., 2010; Patterson et al., 2010; Yeoman et al., 2010).
Bacterial vaginosis is a polymicrobial syndrome that
involves multiple facultative anaerobic and strict anaer-
Atopobium vaginae
obic bacteria (Fredricks et al., 2005; Hillier et al., 1993).
Frequently described BV-related anaerobes include Atopobium vaginae is a facultative anaerobic, nonmotile,
A. vaginae, Bacteroides spp., G. vaginalis, Mobiluncus small elongated Gram-positive coccus that occur as
spp., Mycoplasma hominis and Prevotella spp. either single cells, in pairs, or in short chains
Table 1. Virulence factors produced by bacteria associated with BV and their roles in BV etiology.
First association with
Bacterial species BV was reported in Virulence factors Role of virulence factors in BV etiology
Atopobium vaginae 2004 (Ferris et al., 2004;  Elicit proinflammatory responses (e.g. IL-1b, IL-6, IL-8 and  Negatively affect the barrier function of epithelial cells
Verstraelen et al., 2004) TNF-a) in the vaginal epithelium (Doerflinger et al., 2014; (Doerflinger et al., 2014)
Libby et al., 2008)
 Adherence and biofilm formation (coaggregation) (Hardy  Allows bacteria to persist BV treatment (Swidsinski et al.,
et al., 2016; Swidsinski et al., 2005) 2015)
 Production of VFAs and NVFAsa  Induce alterations in the shape and growth of eukaryotic
cells and apoptosis of human splenic B cells (Kurita-Ochiai
et al., 1998)
 Inhibit chemotaxis (Al-Mushrif et al., 2000; Botta et al.,
1985)
 Internalization into the vaginal epithelial cells (Fichorova Allows bacteria to escape antibiotic treatment (Fichorova
et al., 2013) et al., 2013)
BV-associated bacteria 1, 2 and 3 2005 (Fredricks et al., 2005)  Production of polyamines (BVAB1) (Srinivasan et al., 2012)  Decreases the redox potential and increases the vaginal pH
(BVAB1, BVAB2, BVAB3) (Srinivasan et al., 2015)
Gardnerella vaginalis 1954 (Gardner &  Biofilm formation (Swidsinski et al., 2005) Allows to persist BV treatment (Swidsinski et al., 2008, 2011,
2015)
Dukes 1954; Gardner & Dukes,  Production of amino and keto acids (Chen et al., 1979)  Enhances growth of BV-related bacteria like P. bivia and P.
1955) anaerobius (Pybus & Onderdonk, 1997; Pybus & Onderdonk,
1998)
 Increases ammonia and VFA production by P. bivia (Pybus
& Onderdonk, 1997)
 Production of sialidase, phospholipase C and protease  Promotes vaginal sloughing (“clue cells”) (Yeoman et al.,
(Udayalaxmi et al., 2011; Yeoman et al., 2010) 2010)
 Inhibit secretory immunoglobulin (sIgA) (Lewis et al., 2012)
 Production of vaginolysin (cytolysin)  Induces pore formation and lysis of vaginal epithelial cells
(cytotoxic activity) (Randis et al., 2013)
 Internalization into the vaginal epithelial cells (Marrs et al.,  Escape immune responses and the effects of antibiotic
2012) therapy (Marrs et al., 2012)
Mobiluncus spp. 1984 (Spiegel & Roberts, 1984)  Production of VFAs (acetic, formic, butyric, isobutyric, valeric  Same as in A. vaginae
and isovaleric acids) and NVFAs (succinic acid) (Al-Mushrif
et al., 2000)
 Production of malic acid (Spiegel & Roberts, 1984)  May cause vaginal irritation (Spiegel, 1991)
 Production of polyamines (trimethylamine)  Same as in BVAB
 Biofilm formation (coaggregation) (Costello et al., 2007; De  Same as in A. vaginae
Boer & Plantema, 1988)
Gram-positive anaerobic rods 1983 (Spiegel et al., 1983)  Production of VFAs (acetic and formic acids) and NVFAs  Same as in A. vaginae
(Eggerthella spp., Eubacterium (succinic acid) (Kageyama et al., 1999)
spp.)
Gram-negative anaerobic rods 1980 (Spiegel et al., 1980)  Produce a large amount of amino acids and ammonia  Enhances growth of G. vaginalis and P. bivia (Pybus &
(Bacteroides spp., Fusobacterium (Kapatral et al., 2005) Onderdonk, 1997) Increases the vaginal pH to a very high
nucleatum, Prevotella spp. alkaline pH (Pybus & Onderdonk, 1997)
Leptotrichia/Sneathia spp.)
 Production of butyric acid (VFA) (Kapatral et al., 2005) and  Same as in A. vaginae
succinic acid production (NVFA) (Pybus & Onderdonk, 1996)
 Sialidase and glycosidase production (Briselden et al., 1992;  Same as in G. vaginalis
Marconi et al. 2013; Olmsted et al., 2003)
 Production of hemolysin (Sneathia spp.) (Harwich et al.,  Cytotoxic to vaginal epithelial cells (Harwich et al., 2012)
2012)
CRITICAL REVIEWS IN MICROBIOLOGY

 Production of the 5-nitroimidazole reductase (the nim  Promotes metronidazole resistance (Lubbe et al., 1999;
genes) (Lubbe et al., 1999; Theron et al., 2004) Theron et al., 2004)
3

(continued)
4 H.-S. JUNG ET AL.

(Jovita et al., 1999). Atopobium vaginae was first isolated

 May stimulate the growth of Actinomyces spp. and F. nuclea-

 Hydrolyze phospholipids of vaginal epithelial cells (De Silva


tum (Donelli et al., 2012; Periasamy & Kolenbrander, 2009)
and described in 1999 when it was identified as the
cause of an abscess in the fallopian tube and the ovary

 Hydrolyze urea to ammonia (Masover et al., 1977)


Role of virulence factors in BV etiology
of a woman who had undergone transvaginal oocyte
€rfer et al., 2003).

 Degrades human IgA (Robertson et al., 1984)


recovery (Jovita et al., 1999; Geißdo
An association of A. vaginae with BV was first
reported in 2004 (Ferris et al., 2004; Verstraelen et al.,
2004). A study using DGGE by Ferris et al. (2004) dem-
onstrated that A. vaginae was detected in 55% (12/20)
of BV-positive patients, while only 8.3% (2/24) was
 Same as in G. vaginalis
 Same as in A. vaginae

detected in BV-negative patients. In the same year,


Verstraelen et al. (2004) reported similar findings using
& Quinn, 1986)
 Same as above

16 S rDNA cloning and sequencing, detecting A. vaginae


in 61.3% (8/13) of disturbed vaginal microbiota and in
3.4% (3/87) of normal vaginal microbiota. Atopobium
vaginae is suggested as a marker for abnormal vaginal
microbiota and BV as A. vaginae is present in more than
80% of BV cases (Bradshaw et al., 2006; De Backer et al.,
 Production of propionic and butyric acids (VFA) and succinic

(Ureaplasma urealyticum) (Blanchard et al., 1988; De Silva &


 Production of IgA protease A, phospholipase C and urease

2007; Marconi et al., 2012). Women with A. vaginae are


 Production of the 5-nitroimidazole reductase (the nim

2.6 times (95% confidence intervals (CI): 0.6–11.4] and


19.2 times (95% CI: 3.7–98.7) more likely to be catego-
genes) (Lubbe et al., 1999; Theron et al., 2004)

rized as BV by using the Amsel’s criteria and the Nugent


 Biofilm formation (except Peptoniphilus spp.)

scoring system (Haggerty et al., 2009). In addition, A.


acid (NVFA) (Pybus & Onderdonk, 1998)
Virulence factors

Quinn, 1986; Robertson et al., 1984)

vaginae is rarely detected in BV patients without G. vag-


inalis and this coexistence is also detected in patients
with recurrent BV (Bradshaw et al., 2006; Trama et al.,
2008; Verhelst et al., 2004). However, A. vaginae is also
 Production of sialidase

commonly found in women with normal vaginal micro-


biota – this may also be explained by A. vaginae strain
diversity as recently reported (Mendes-Soares et al.,
2015).
Studies have found that A. vaginae is one of the
dominant bacterial species (along with G. vaginalis) in
the polymicrobial BV biofilm and may represent
1–54.1% of the biofilm mass (Hardy et al., 2016;
1973 (McCormack et al., 1973)
et al. 1982) 2005 (Fredricks
1982 (Veillonella spp.) (Piot

Swidsinski et al., 2005, 2008). This biofilm may allow A.


1980 (Spiegel et al., 1980) &
First association with
BV was reported in

vaginae to be resistant to metronidazole treatment, as


metronidazole does not fully eradicate A. vaginae in the
biofilm and this resistance may explain why A. vaginae
et al., 2005)

NVFAs: nonvolatile fatty acids; VFAs: volatile fatty acids.

often reemerges in recurrent BV (Mayer et al., 2015;


McMillan et al., 2011; Swidsinski et al., 2008).

Bacterial vaginosis-associated bacteria (BVAB)


Anaerobic cocci (Anaerococcus spp.,

In 2005, three bacterial species were newly identified in


Mollicutes (Mycoplasma hominis,
Finegoldia spp., Megasphaera

the vaginal fluid of BV-positive women and were named


Ureaplasma urealyticum)

as BVAB1, BVAB2 and BVAB3 (Fredricks et al., 2005).


Peptostreptococcus spp.,
spp., Peptococcus spp.,
Table 1. Continued

These bacteria were related to bacteria in the phylum


Peptoniphilus spp.,

Clostridium (the order Clostridiales) but were detected


Veillonella spp)
Bacterial species

only in the vaginal fluid of women with BV (Fredricks


et al., 2005). Fluorescence in situ hybridization (FISH)
revealed that BVAB1 appear as thin, curved bacteria;
short, straight rods for BVAB2 and long, wide and
a
CRITICAL REVIEWS IN MICROBIOLOGY 5

straight rods for BVAB3 – these BVAB were attached Prevotella spp.; propionic acids produced by
to vaginal epithelial cells in a similar appearance as Megasphaera spp. and Propionibacterium spp.; butyric,
“clue cells” (Fredricks et al., 2005). A recent study by isobutyric, valeric and isovaleric acids by Mobiluncus
Muzny et al. (2014) suggested that BVAB1 could be the spp. and Prevotella spp. (Al-Mushrif et al., 2000;
dominant curved rods in vaginal smears with BV and Kageyama et al., 1999; Wang et al., 2012). Similarly,
not Mobiluncus spp., which are often identified as NVFAs like succinic acid may be produced by
curved rods in Nugent scoring. In addition, BVAB1 is Actinomyces spp., anaerobic Gram-positive cocci,
associated with a positive whiff test (fishy odor) and are Eggerthella spp., Mobiluncus spp. and Prevotella spp. (Al-
thus thought to produce polyamines (putrescine, cadav- Mushrif et al., 2000; Kageyama et al., 1999; Pybus &
erine and trimethylamine), which are associated with Onderdonk, 1998). Mobiluncus spp. can also produce
the fishy odor (Srinivasan et al., 2012; Wolrath et al., malic acid (NVFA) that causes irritation of vaginal
2001). mucous membranes (Spiegel & Roberts, 1984). Gram-
To date, only BVAB3 have been cultured and bio- negative anaerobes like Bacteroides spp., Leptotrichia
chemically characterized (Austin et al., 2015) and the spp., Megasphaera spp. and Prevotella spp. are sialidase
remaining two BVAB (BVAB1 and BVAB2) have not yet producers (Briselden et al., 1992; Doerflinger et al., 2014;
been isolated by culture. The species name of BVAB3 Marconi et al., 2013). In addition to sialidase, glycosulfa-
was proposed as Mageeibacillus indolicus (Austin et al., tase, collagenase and fibrinolysins produced by
2015). The involvement of BVAB in the formation of a Prevotella spp. and Sneathia spp. (Lewis & Lewis, 2012;
polymicrobial BV biofilm has not yet been confirmed Roberton et al., 2005) and the glycosidase and protein-
and consequently, the role of BVAB in BV etiology has ase produced by anaerobic Gram-negative bacteria are
not been sufficiently investigated. examples of enzymes that degrade mucin in the vaginal
epithelial cells and promotes vaginal sloughing
(Olmsted et al., 2003). Bacteroides spp., Finegoldia spp,
Other BV-related bacteria
Peptostreptococcus spp. and Propionibacterium spp. may
Along with the above-mentioned bacteria, other also produce 5-nitroimidazole reductase from the nim
BV-related bacteria may contribute to BV development. genes, which promotes metronidazole resistance
Other bacteria associated with BV may include (Lubbe et al., 1999; Theron et al., 2004).
Anaerococcus spp., Bacteroides spp. (B. levii, B. Peptoniphilus spp. may help the biofilm formation of
ureolyticus), Dialister spp., Eggerthella spp., Finegoldia G. vaginalis during initial adhesion (Castro et al., 2013;
spp., Fusobacterium nucleatum, Leptotrichia spp., Machado et al., 2013; Patterson et al., 2010). The genus
Megasphaera spp., Mobiluncus spp., Peptococcus spp., Peptoniphilus was proposed in 2001 (Ezaki et al., 2001)
Peptoniphilus spp., Peptostreptococcus spp. (P. anaero- and has been associated with persistent BV as
bius, P. prevotii, P. tetradius), Prevotella spp. (P. amnii, Peptoniphilus spp. were found in 87% (27/31) of persist-
P. bivia, P. buccalis, P. corporis, P. disiens), Mycoplasma ent BV cases in a study by Marrazzo et al. (2008) and
hominis, Sneathia spp., Ureaplasma urealyticum and viri- the risk for persistent BV was significantly higher in
dans streptococci (S. acidominimus, S. constellatus, S. women if Peptoniphilus spp. were detected at baseline
intermedius, S. mitis, S. morbillorum, S. mutans, S. sanguis (before treatment) (risk ratio (RR) 1.7; 95% CI: 0.8–8.3).
II) (Hillier et al. 1993; Srinivasan et al., 2012; Zozaya- In the same study, the risk of persistent BV was higher
Hinchliffe et al., 2010). The list of bacteria associated for women if P. lacrimalis were detected before treat-
with BV may become longer as new culture-independ- ment (RR 3.5; 95% CI: 1.6–15.5) (Marrazzo et al., 2008).
ent techniques identify additional bacterial species
associated with BV as shown in a recent study (Xia
Current conceptual models and new theories
et al., 2016).
in BV etiology
In order to assist with BV development, BV-related
bacteria may produce amino acids, metabolic products Many conceptual models and theories have been pro-
(such as VFAs and NVFAs and polyamines) and enzymes posed for the etiology of BV, yet the exact mechanism
(Al-Mushrif et al., 2000; Briselden et al., 1992; Chen of how BV is triggered remains controversial (Schwebke
et al., 1979) and possibly contribute in polymicrobial BV et al., 2014b). There are currently three major concep-
biofilm formation (Machado et al., 2013; Patterson et al., tual models that are proposed for the etiology of BV,
2010) (Table 1). The VFAs produced by BV-related bac- that is, the “Lactobacillus depletion model”, the “primary
teria as a result of the fermentation of lactic acid pathogen model” and the “polymicrobial pathogen
include formic and acetic acids produced by model” (Table 2) (Muzny & Schwebke, 2013; Srinivasan
Actinomyces spp., Eggerthella lenta, Mobiluncus spp. and & Fredricks, 2008;). These models share characteristic
6 H.-S. JUNG ET AL.

Table 2. Conceptual models in BV etiology.


Conceptual models Main features Supported by Not supported by
Lactobacillus depletion model  Removal of lactobacilli occurs first  The findings that lactobacilli pro-  The findings that some women
before the rise in the vaginal pH duce H2O2 and other antibacterial maintain a “healthy” vaginal envir-
 Absence of lactic acid-producing substances to maintain normal onment without lactobacilli (Gajer
lactobacilli leads to the rise in the vaginal microbiota (Aroutcheva et al., 2012; Zhou et al., 2004)
vaginal pH et al., 2001; Klebanoff et al., 1991)  Some strains of Atopobium spp.,
 Rise in the vaginal pH promotes  The finding that BV-related bacteria Leptotrichia spp. and Megasphaera
the overgrowth of BV-related bac- are inhibited by low vaginal pH spp. may produce lactic acid (Gajer
teria (Boskey et al., 2001) et al., 2012; Zhou et al., 2004)
 Provides a scientific basis for a res-
toration strategy using probiotics
and acidification agents
Primary pathogen model  Introduction of a single pathogen  The findings that G. vaginalis out-  The findings that some G. vaginalis
(G. vaginalis) by sexual activity to competes other BV-related bacteria strains are present in normal micro-
the vaginal environment by possessing many virulence fac- biota and in sexually inexperienced
 G. vaginalis creates a lower redox tors including biofilm formation women (Fethers et al., 2012;
potential environment to create (Alves et al., 2014; Patterson et al., Schwebke et al., 2014a)
intermediate microbiota 2010)
 Intermediate microbiota promote  The findings that G. vaginalis have
the overgrowth of BV bacteria ability to persist in low pH, while
 Provides a scientific basis for G. others not
vaginalis biofilm disruptors as treat-  The findings that G. vaginalis may
ment strategy produce antagonistic substances to
displace lactobacilli (Nagy et al.,
1991)
Polymicrobial pathogen model  Introduction of a BV consortium of  High bacterial diversity in BV (Ling  Other BV-related bacteria than G.
bacteria by sexual activity to the et al., 2010; Srinivasan et al., 2012) vaginalis are not as virulent as G.
vaginal environment  The involvement of the polymicro- vaginalis (Alves et al., 2014;
 The overgrowth of BV-related bac- bial biofilm formation (Swidsinski Machado et al., 2013; Patterson
teria and removal of lactobacilli et al., 2013) et al., 2010)
take place simultaneously  The finding that other BV-related
 Provides a scientific basis for poly- bacteria than G. vaginalis can
microbial biofilm disruptors as inhibit lactobacilli (Nagy et al.,
treatment strategy 1991)

features like the reduction of lactobacilli, the exposure (“Do€derlein’s bacillus”, identified later as Lactobacillus
and overgrowth of BVAB and BV-related bacteria and acidophilus) from the vaginal fluids of asymptomatic
the increase in the vaginal pH (Muzny & Schwebke, pregnant women (Do €derlein, 1892; Thomas, 1928). In
2013; Srinivasan & Fredricks, 2008). In the “Lactobacillus 1997, Pavlova et al. (1997) suggested that the reduction
depletion model”, the depletion of H2O2-producing lac- of vaginal lactobacilli must be induced by external fac-
tobacilli and a simultaneous rise in the vaginal pH have tors to trigger a rise in the vaginal pH (>4.5) and the
to occur first to trigger the overgrowth of the intro- overgrowth of BV-related bacteria.
duced BV-related anaerobic bacteria, which then results In some women, the absence of H2O2-producing lac-
in BV (Srinivasan & Fredricks, 2008). In the “primary tobacilli strains is associated with the acquisition of BV
pathogen model” and the “polymicrobial pathogen (Cherpes et al., 2008; Hawes et al., 1996). Studies show
model”, the exposure to pathogen/s leads to the dis- that vaginal lactobacilli possess multiple control mecha-
placement of vaginal lactobacilli that in turn leads to BV nisms to maintain the “healthy” vaginal environment,
(Muzny & Schwebke, 2013). The formation of a polymi- such as: (i) vaginal acidification via lactic acid produc-
crobial biofilm is thought to play a key role in BV tion (Boskey et al., 2001); (ii) interference of adhesion
etiology, regardless of which conceptual model is fol- and growth of pathogenic bacteria (Castro et al., 2013;
lowed (Muzny & Schwebke, 2013; Swidsinski et al., 2005, Coudeyras et al., 2008; Velraeds et al., 1996); (iii) produc-
2013). In the following subsections, three conceptual tion of H2O2 and bacteriocins (Aroutcheva et al., 2001;
models and one new theory in BV etiology will be Klebanoff et al., 1991) and (iv) biofilm formation (Terraf
discussed. et al., 2014; Ventolini et al., 2015). Major disturbances of
the vaginal lactobacilli may occur due to excessive
douching (Pavlova & Tao, 2000), use of spermicides
Lactobacillus depletion model
(nonoxynol-9) (Hooton et al., 1991), antibiotics (Vallor
Lactobacilli have been considered to be a part of nor- et al., 2001) and natural causes like phages (Kiliç et al.,
mal vaginal microbiota since 1892, when Albert 2001; Pavlova et al., 1997; Tao et al., 1997). Frequent
€derlein first isolated and described an organism
Do douching is significantly associated with the lack of the
CRITICAL REVIEWS IN MICROBIOLOGY 7

H2O2-producing lactobacilli (1 times per month: OR confirmed this finding (Gajer et al., 2012; Zhou et al.,
1.6; 95% CI: 1.2–2.1 and 2 times per month: OR 2.5; 2004). As lactic acid-producing strains of Atopobium
95% CI: 1.1–6.0) (Beigi et al., 2005; Ness et al., 2002) and spp., Leptotrichia spp. and Megasphaera spp. are found
the acquisition of BV (douching for hygiene: RR 2.1; 95% in some women who still maintained “healthy” vaginal
CI: 1.0–4.3; p ¼ .05 and douching within the past two environments, the presence of non-Lactobacillus vaginal
months: adjusted OR 2.9; 95% CI: 1.5–5.6) (Hawes et al., microbiota and the lack of vaginal lactobacilli may not
1996; Holzman et al., 2001). The usage of spermicides necessarily cause BV (Gajer et al., 2012; Zhou et al.,
(such as nonoxynol-9) may also possibly reduce H2O2- 2004).
producing lactobacilli, as the H2O2-producing strains of
lactobacilli are more susceptible to nonoxynol-9 than
Primary pathogen model
non-producing strains (Hooton et al., 1991).
Alternatively, bacteriophages may reduce vaginal lac- The “primary pathogen model” originated from a study
tobacilli (Kiliç et al., 2001; Pavlova et al., 1997; Tao et al., by Gardner and Dukes in 1955, in which Haemophilus
1997). Pavlova et al. (1997) and Kiliç et al. (2001) found vaginalis (G. vaginalis) was successfully isolated from a
that the proportion of lysogenic phages is higher BV patient and was proposed as the sole etiological
among Lactobacillus strains isolated from BV patients agent for BV (Gardner & Dukes, 1955). Other studies
and that some of the lysogenic phages may become have also confirmed the existence of G. vaginalis (up to
virulent (the lytic stage) in other vaginal lactobacilli 96%) from the vaginal fluid of women with BV
strains due to genetic mutations that may eliminate lac- (Marrazzo et al., 2008; Srinivasan et al., 2012) and the
tobacilli in the vagina (Kiliç et al., 2001; Pavlova et al., causation of BV symptoms like vaginal sloughing by G.
1997). However, Martın et al. (2009) proposed that the vaginalis in an in vivo murine model (Gilbert et al.,
lysogeny of bacteriophages in lactobacilli does not play 2013).
a significant role in the elimination of lactobacilli, as less In contrast to the “Lactobacillus depletion model”,
than half of the 15 prophages that were studied by the “primary pathogen model” suggests that the sexual
them lacked genes for the development of a lytic cycle. introduction of G. vaginalis into the vaginal environ-
No further study has confirmed this finding in the con- ment creates the “intermediate microbiota” with a
text of vaginal lactobacilli. reduced number of lactobacilli, which causes a decrease
In the “Lactobacillus depletion model”, the vaginal in the redox potential of the vagina (increase in the
pH becomes higher (>4.5) once the normal vaginal vaginal pH) and triggers the overgrowth of vaginal
microbiota (vaginal lactobacilli) are disturbed, as the anaerobic bacteria (Muzny & Schwebke, 2013). An
main source for acidity in the vagina is the lactic acid- extensive version of this model was proposed by
producing lactobacilli (Boskey et al., 1999, 2001; Schwebke et al. (2014b) that suggests G. vaginalis as
O’Hanlon et al., 2013). Furthermore, the BVAB and other the key organism in BV etiology. According to
BV-related bacteria like G. vaginalis, Mobiluncus spp., Schwebke et al. (2014b), G. vaginalis is not part of the
Peptostreptococcus anaerobius and Prevotella spp. may normal vaginal microbiota but is introduced through
utilize lactic acid to produce butyric acids (VFA), succinic sexual activity. Schwebke et al. (2014b) also added that
acid (NVFA) and other acids to modulate immune G. vaginalis may survive in the male urethra and semen,
responses and to inhibit the killing activity of the which may later be transmitted back to female partners
remaining lactobacilli (Al-Mushrif et al., 2000; Macklaim through sexual contact.
et al., 2013; Pybus & Onderdonk, 1996), while Prevotella According to the “primary pathogen model”, G. vagi-
amnii, Dialister spp. and Megasphaera spp. may produce nalis first tolerates a high oxidation–reduction (redox)
polyamines (putrescine and spermidine) to relieve the potential vaginal environment (pH 4–5) and adheres to
excess acidity due to lactic acid (Chen et al., 1979; vaginal epithelial cells (Muzny & Schwebke, 2013; Sobel
Macklaim et al., 2013). et al., 1981; Udayalaxmi et al., 2012;). Gardnerella vagina-
The major concern against the “Lactobacillus deple- lis may form biofilms during this stage (Udayalaxmi
tion model” is that this model is based on the notion et al., 2012) and may produce antagonistic substances
that women are colonized by Lactobacillus spp. to main- to displace lactobacilli (Nagy et al., 1991). Gardnerella
tain the “healthy” vaginal environment. According to a vaginalis produces amino and keto acids that enhance
large-scale US cross-sectional study, 27% of “healthy” the growth of BV anaerobes (Chen et al., 1979), in order
women harbored vaginal microbiota that lack a large to create the “intermediate vaginal microbiota (IVM)”.
number of lactobacilli and included facultative or strict The overgrowth of the “IVM” (including P. bivia) raises
anaerobic bacteria (community state types IV (CST IV)) the vaginal pH (>4.5), as amino acids like arginine serve
(Ravel et al., 2011). Smaller-scaled studies also as a precursor for the production of ammonia and
8 H.-S. JUNG ET AL.

polyamines, such as putrescine, cadaverine and tyram- G. vaginalis as the sole etiological agent for BV, reported
ine (Srinivasan et al., 2015; Wolrath et al., 2001), which that one or more other bacteria than G. vaginalis were
further stimulates the growth of G. vaginalis and other isolated from 56 BV-positive women (56/141; 39.7%)
BV-related anaerobes and confirms the establishment of and that eleven women developed BV without the pres-
the BV microbiota (Pybus & Onderdonk, 1997, 1998; ence of G. vaginalis. Furthermore, Gardner & Dukes
Muzny & Schwebke, 2013). (1955) reported that the inoculation of women with
The “primary pathogen model” is not free of argu- vaginal fluid was more successful in causing BV symp-
ment against it, as G. vaginalis can still be commonly toms than inoculation with a G. vaginalis pure culture
found in the vaginal microbiota of women without BV (11/15; 73% versus 1/13; 8%) (Gardner & Dukes, 1955).
(Hyman et al., 2005; Schwebke et al., 2014a) and even in The involvement of a polymicrobial biofilm in BV is
sexually inexperienced women (Fethers et al., 2012). evident, as the BV biofilms found on the vaginal epithe-
These findings may indicate that G. vaginalis can be a lium do not only contain G. vaginalis, but also a variety
common inhabitant of the vaginal microbiota in some of BV-related bacteria, such as A. vaginae, Bacteroides
women without causing BV (Fethers et al., 2012; spp., Streptococcus spp. and Veillonella spp. (Swidsinski
Schwebke et al., 2014a). The possible explanation for et al., 2005). In fact, a recent study highlighted the syn-
this can be the existence of different genotypic strains ergy between A. vaginae and G. vaginalis in the forma-
(Jayaprakash et al., 2012; Lopes Dos Santos Santiago tion of a BV biofilm, indicating the polymicrobial nature
et al., 2011) and biotypes (Piot et al., 1984) – especially of this biofilm (Hardy et al., 2016). Furthermore,
biotype 5 (Piot’s biotype) is significantly associated with Mobiluncus spp. may also possibly participate in the for-
the normal vaginal microbiota (Aroutcheva et al., 2001). mation of a polymicrobial BV biofilm, as Mobiluncus
spp. (M. curtisii and M. mulieris) are able to form a bio-
film (Costello et al., 2007; De Boer & Plantema, 1988).
Polymicrobial pathogen model
Similar to the above-mentioned two conceptual
The “polymicrobial pathogen model” is similar to the models, there also exists a controversy in this model. As
“primary pathogen model” but is different in that the previously demonstrated by several studies (Alves et al.,
sexual exposure of BV-related bacteria (including G. vag- 2014; Machado, 2013; Patterson et al., 2010), many BV-
inalis; both from men and women) to the vaginal micro- related bacteria may not be as virulent as pathogenic
biota directly leads to the simultaneous displacement of strains of G. vaginalis, in terms of cytotoxicity, adher-
lactobacilli and a BV condition without having ence and biofilm formation. Therefore, more evidence is
“intermediate microbiota” (Muzny & Schwebke, 2013). needed to prove that the primary trigger that initiates
Instead of a single primary pathogen like G. vaginalis in BV is indeed a polymicrobial consortium of BV-related
the “primary pathogen model”, a consortium of BV- bacteria.
related anaerobes may show antibiosis against lactoba-
cilli or produce amino acids and other virulence factors
Internalization of BV-related bacteria in the
to enhance the growth of BV-related anaerobes (Nagy
vaginal epithelial cells: a new escaping
et al., 1991; Pybus & Onderdonk, 1996). According to a
mechanism?
study by Nagy et al. (1991), some strains of M. curtisii,
Peptostreptococcus spp., Prevotella bivius and P. disiens The internalization theory of the BV-related bacteria
can inhibit the growth of lactobacilli, possibly indicating was introduced only in 2012 and so far, there are only
the ability of BV-related anaerobes to displace lactoba- two studies that support this theory (Fichorova et al.,
cilli. Mobiluncus spp. may also produce cytotoxins that 2013; Marrs et al., 2012). Marrs et al. (2012) suggested
could damage vaginal epithelial cells similar to G. vagi- that G. vaginalis may use the internalization mechanism
nalis (Taylor-Robinson et al., 1993). Furthermore, P. bivia into vaginal epithelial cells as a strategy to escape the
and Bifidobacterium spp. can produce amino acids to host defenses and antibiotics like clindamycin and
enhance the growth of other anaerobic bacteria like metronidazole. According to their study, the uptake of
Eggerthella lenta and G. vaginalis (Africa et al., 2014; live G. vaginalis cells by the VK2 vaginal epithelial cells
Pybus & Onderdonk, 1996). was 21.8-fold higher than the uptake of G. vaginalis cells
The “polymicrobial pathogen model” is supported by killed by heat. Furthermore, the internalization process
the fact that BV involves multiple anaerobic bacteria of G. vaginalis was inhibited when the VK2 vaginal epi-
and that the BV microbiota are taxonomically more thelial cells were pretreated with cytochalasin-D, an
diverse than normal vaginal microbiota (Ling et al., inhibitor of actin polymerization in the cell cytoskeleton
2010; Srinivasan et al., 2012). Even the original study by (Marrs et al., 2012). However, a recent study found that
Gardner & Dukes (1955), where the authors suggested G. vaginalis tends to exist in a dispersed form or forms
CRITICAL REVIEWS IN MICROBIOLOGY 9

an extracellular biofilm to escape immune responses polymorphisms and BV and indicated that the role of
and was less efficiently internalized by dendritic cells intrinsic host factors in the etiology of BV is still unclear.
and T lymphocytes than lactobacilli (Bertran et al.,
2016). The internalization mechanism of G. vaginalis
Biofilm formation in bacterial vaginosis
into the vaginal epithelial cells thus requires further
investigation. It is well known that the polymicrobial biofilm in BV
Additionally, A. vaginae and P. bivia can maintain via- plays a key role in its pathogenesis (Muzny & Schwebke,
bility by internalizing into the vaginal epithelial cells 2015). The importance of biofilms in BV was first high-
(Fichorova et al., 2013). According to Fichorova et al. lighted in 2005 when Swidsinski et al. (2005) demon-
(2013), A. vaginae and P. bivia can persist in high num- strated that only G. vaginalis and A. vaginae produce a
bers on Trichomonas vaginalis-infected vaginal epithelial characteristic “adherent” biofilm in women with BV. This
cells and that these bacteria could use the internaliza- dense “adherent” biofilm found in BV patients con-
tion mechanism as protection against antibiotics and tained short rods (G. vaginalis and A. vaginae) that were
attacks by T. vaginalis. This mechanism warrants further tightly attached to each other on the vaginal epithelial
investigation as it may possibly be one of the factors surface, “resembling brickwork” (Swidsinski et al., 2005).
leading to BV and also recurrent BV. A polymicrobial BV biofilm may ensure the survival of G.
vaginalis and other BV-related bacteria (Schwebke et al.,
2014a). Patterson et al. (2007) observed a greater toler-
Additional theories for the development of BV ance to 5-fold higher concentrations of H2O2 and 4- to
Sexual activity is not the sole determinant for the occur- 8-fold higher concentrations of lactic acid in G. vaginalis
rence of BV and as such BV can be classified as a sexu- biofilms as compared to planktonic cells, which may
ally associated infection rather than a sexually suggest that biofilm formation contributes to the sur-
transmitted infection (Lepp€aluoto, 2011). The alkaline vival of G. vaginalis.
buffering action of semen decreases the acidity of the Biofilm formation is a dynamic and complex process
that involves multiple interactions between single or
vagina (increase in pH) for several hours after inter-
multiple bacterial species (Machado et al., 2013;
course (Boskey et al., 1999) which, combined with the
Whittaker et al., 1996). To date, the exact process of the
effects of vaginal coital transudate and vaginal neuro-
polymicrobial biofilm formation in BV remains unknown
genic transudate, could directly result in a physiological
(Machado et al., 2013). However, it can be hypothesized
post-coital condition (i.e. BV) for protection of ejacu-
that the BV biofilm follows the same route as the forma-
lated spermatozoa (Lepp€aluoto, 2011). This post-coital
tion of oral biofilms, which is a well-described example
condition is characterized by “pure” Gardnerella flora
of a polymicrobial biofilm (Machado et al., 2013). In the
replacing Lactobacillus; a polymicrobial form of BV is
following subsections, the current review will attempt
suggested to be autoinfection of the postcoital physio-
to discuss each stage of polymicrobial biofilm formation
logical G. vaginalis flora, mixed with M. hominis and
in the context of BV, namely (i) adhesion, (ii) coaggrega-
anaerobic bacteria at lower vaginal acidity (Lepp€aluoto, tion (microcolony formation); (iii) maturation and (iv)
2011). dispersion.
In addition, genetic polymorphisms within a popula-
tion may be contributing factors in the development of
BV and its associated complications (Witkin, 2007; Adhesion
Turovskiy et al., 2011). An example may include the As the vaginal lactobacilli may interfere with the adhe-
increased risk of women of African ethnicity to develop sion of BV-related bacteria by colonizing the vaginal
BV; however, the exact reasons for the predisposition of epithelium (Castro et al., 2013; Coudeyras et al., 2008;
this population to BV have yet to be elucidated Velraeds et al., 1996), it is possible that the BV-related
(Danielsson et al., 2011). Research on genetic polymor- bacteria first need to inhibit and to displace the colon-
phisms and BV has up to now been confined to the ized lactobacilli in order to initiate the biofilm formation
innate immune system (Turovskiy et al., 2011); polymor- process. In fact, through a yet unknown mechanism,
phisms in genes coding 19 for components of the BV-related anaerobes, such as P. bivius, P. disiens, G. vag-
innate immune system (e.g. IL-1b) have been shown to inalis, M. curtisii, M. mulieris and Peptostreptococcus spp.
influence the quantitative bacterial composition of the may inhibit the growth of lactobacilli and may displace
vagina (Witkin, 2007). However, Turovskiy et al. (2011) lactobacilli (except L. iners) (Castro et al., 2013; Nagy
have pointed out the inconsistent findings of the few et al., 1991). It is, however, also possible that G. vaginalis
studies that investigated the link between genetic may first need to initiate the biofilm formation process,
10 H.-S. JUNG ET AL.

as only G. vaginalis is able to strongly adhere to vaginal species indicating the vaginal microbiota shifting to the
epithelial cells in the presence of L. crispatus and to BV microbiota (Ferris et al., 2007; Yoshimura et al.,
form a biofilm regardless of the pH (Machado et al., 2011). Indeed, a study by Castro et al. (2013) revealed
2013; Udayalaxmi et al., 2012). As several reports indi- that L. iners enhanced the adherence of G. vaginalis to
cate that G. vaginalis may act as an early colonizer spe- epithelial cells rather than inhibiting the bacteria.
cies in the polymicrobial BV biofilm (Alves et al., 2014; Peptoniphilus spp. show similar adherence ability as G.
Patterson et al., 2010; Machado et al., 2013), the current vaginalis on ME-180 vaginal epithelial cells (bacteria
review will focus on the “primary pathogen model” in attached on 75% to 100% of ME-180 cells in all viewed
an attempt to explain the adhesion process of a polymi- microscopic fields) (Patterson et al., 2010). However,
crobial BV biofilm. Peptoniphilus spp. have received almost no attention in
Bacterial adhesion to vaginal epithelial cells may be research, despite the bacteria’s good adherence ability
mediated by interactions between cell appendages (pili, and virulence potential in BV (Africa et al., 2014).
fimbriae or flagella), carbohydrates and cell surface Therefore, more studies confirming the virulence poten-
adhesins (McMillan et al., 2012; Peeters & Piot, 1985; tial and the role of Peptoniphilus spp. during the initial
Terraf et al., 2014). adhesion process of BV biofilm formation would be
Gardnerella vaginalis harbors genes encoding type I, beneficial in delineating the etiology of BV.
II and IV fimbriae/pili, as well as a biofilm-associated
protein (BAP) family gene (bapL) (Harwich et al., 2010;
Microcolony formation and coaggregation
Yeoman et al., 2010). The presence of pili was confirmed
by an earlier electron microscopy study (Johnson & After adhesion, G. vaginalis may first develop microcolo-
Davies, 1984). The BapL protein is a BAP found in nies before biofilm maturation, as demonstrated by
Listeria monocytogenes that is involved in surface Muli & Struthers (1998) and Hymes et al. (2013). Thin
attachment during biofilm formation (Jordan et al., sections of the G. vaginalis biofilms grown in a Sorbarod
2008). biofilm system contained numerous microcolonies that
According to Patterson (2010), the bapL gene, which were 30 lm to 50 lm in diameter (Muli & Struthers,
encodes the BapL protein of G. vaginalis, is highly 1998). In addition, a study by Hymes et al. (2013)
expressed only during biofilm formation, indicating the showed that G. vaginalis forms smaller microcolonies
involvement of this protein in biofilm formation. In add- with no extracellular polymeric substance (EPS) matrix
ition, partial genome and amino acid sequence analyses when G. vaginalis strains were grown on a glass surface
of the BapL have showed that the BapL contain nine in the presence of DNase, while G. vaginalis formed a
Rib domains and a cell-wall anchoring motif (LPxTG) fully developed three-dimensional biofilm containing
(Patterson, 2010). The name “Rib domains”, originated the EPS matrix when grown without DNase. This finding
from the Rib cell surface protein (resistance to pro- may thus suggest that the microcolony formation of G.
teases, immunity and group B) of group B streptococci vaginalis may occur before the production of the EPS
that contains repetitive elements (Stålhammar- matrix.
Carlemalm et al., 1993). A whole genome sequencing In oral biofilms, the attachment of one species is fol-
study by Harwich et al. (2010) confirmed that the BapL lowed by the attachment of a second colonizing species
protein of G. vaginalis contains Rib domains, which is (coaggregation) (Nagaoka et al., 2008; Okuda et al.,
highly repetitive in the central repeat region. Harwich 2012). The coaggregation may occur through two differ-
et al. (2010) showed that the BV isolates of G. vaginalis ent mechanisms: (i) the second colonizing species starts
(strains AMD, 101 and 551) tended to produce thicker the coaggregation cascade first in the suspension and
biofilms than non-BV isolates (strains 5–1 and 465). It the formed microcolonies subsequently bind on the
was proposed that the number and distribution of Rib surface of developing biofilms and/or (ii) individual cells
domain repeats within the central repeat region might of a second colonizing species in the suspension recog-
have played a role in biofilm formation capacity, as the nize and adhere to the bacterial cells of the early colo-
distribution of Rib domains differed between a BV iso- nizing species, forming an aggregate in developing
late (strain AMD) and a non-BV isolate (strain 5–1) biofilms (Rickard et al., 2003). It is not known whether
(Harwich et al., 2010). second/third colonizing species in BV use the first or
Lactobacillus iners and Peptoniphilus spp. may assist second mechanism of coaggregation; however, it is pos-
G. vaginalis during the initial adhesion process (Castro sible that A. vaginae, F. nucleatum and Mobiluncus spp.
et al., 2013; Machado et al., 2013). Even though lactoba- may coaggregate with G. vaginalis (De Boer & Plantema,
cilli are known as indicator organisms for normal vagi- 1988; Patterson et al., 2010). Mobiluncus spp. may likely
nal microbiota, L. iners is proposed as a transitional follow the first route, as Mobiluncus spp. can form
CRITICAL REVIEWS IN MICROBIOLOGY 11

microcolonies on its own (De Boer & Plantema, 1988). eDNA) with a concentration of 100 lg/mL on G. vagina-
Another possibility of coaggregation can be between lis biofilms resulted in up to 80% reduction in biofilm
the uropathogenic bacteria like Escherichia coli (E. coli) mass. In addition, the authors also showed that the
and Enterococcus faecalis (E. faecalis) and the G. vagina- release of eDNA was maximal during the early exponen-
lis biofilms as the presence of E. coli and E. faecalis in tial growth phase, which may indicate that eDNA is
the G. vaginalis biofilms could enhance the growth of G. actively involved in the biofilm formation of G. vaginalis
vaginalis (Castro et al., 2016). (Hymes et al., 2013).
The possible candidate bridge species between early In addition to eDNA, the pathogenic strains of G.
and late colonizers may be F. nucleatum (Patterson vaginalis (317 and 594) may encode glycosyltransferases
et al., 2010). Fusobacterium nucleatum was originally (GT) (family I, II and IV) that seem to be involved in EPS
described as the causative organism of periodontal production (Yeoman et al., 2010). Glycosyltransferases
infections (e.g. gingivitis and chronic periodontitis), but are essential for the biosynthesis of EPS in bacteria,
this organism can also reside in the vagina (in both BV such as E. faecalis (Theilacker et al., 2011) and
and non-BV cases) (Cassini et al., 2013; Nikolaitchouk Xanthomonas campestris (Tao et al., 2010) and are
et al., 2008; Oakley et al., 2008). Fusobacterium nuclea- thought to be essential in G. vaginalis as well (Yeoman
tum is well known as the bridge species between early et al., 2010).
and late colonizers in oral biofilms (Kaplan et al., 2009;
Okuda et al., 2012). Although F. nucleatum does not
Dispersion
adhere well on vaginal epithelial cells (Patterson et al.,
2010), F. nucleatum is known to express receptors that Dispersion is an essential step in biofilm formation that
facilitate coaggregation of Prevotella spp. and contributes to bacterial survival and disease transmis-
Bifidobacterium spp. (Nagaoka et al., 2008; Okuda et al., sion (Stoodley et al., 2001). The exact dispersal mechan-
2012). Alternatively, the following candidate bacteria ism of polymicrobial BV biofilm is not known as it has
may coaggregate as late colonizers as these bacteria not yet been studied. However, it can be hypothesized
have shown a synergistic interaction in a dual-species that both active and passive dispersion might occur
biofilm model: Actinomyces neuii, Bacillus firmus, during BV. Active dispersion might occur when the
Brevibacterium ravenspurgense, Corynebacterium spp., vaginal environment becomes favorable during men-
Enterococcus faecalis, Escherichia coli, Nosocomiicoccus struation or through the overgrowth of BV-related bac-
ampullae, P. bivia, Propionibacterium acnes, teria, as the vaginal pH is increased by menstrual blood
Staphylococcus spp. and Streptococcus spp. (Castro & (pH 7.32) (Hovhannisyan & Grigoryan, 2014) and poly-
Cerca, 2015). amines (Wolrath et al., 2001). In contrast, passive disper-
sion (erosion and sloughing) might possibly occur when
the biofilm is exposed to the shear forces of vaginal
Biofilm maturation
sloughing induced by sialidase, glycosulfatase, glycosi-
After successful microcolony formation/coaggregation, dase, proteinase, collagenase and fibrinolysins (Lewis &
G. vaginalis may release extracellular DNA (eDNA) that Lewis 2012; Olmsted et al., 2003) and when the biofilm
stimulates the EPS matrix production (Hymes et al., is exposed to the shear forces of menstrual flow.
2013). It is possible that the EPS production of BV-
related bacteria may be controlled by quorum-sensing
Conclusions
molecules (molecules used in a special cell-to-cell sig-
naling/communication mechanism, such as cyclic-di- The formation of a polymicrobial biofilm is essential in
GMP observed in Pseudomonas aeruginosa) (Borlee the etiology of BV, as it increases the chances of survival
et al., 2010), though the quorum sensing in a polymicro- of BV-related bacteria during BV infection (Swidsinski
bial BV biofilm has not yet been studied. The fully et al., 2014). The current review suggests that BV-related
matured in vivo BV biofilm may appear like a bacteria can cooperate and form synergistic relation-
“brickwork”, a highly organized structure without spaces ships with each other during BV and during the forma-
between bacterial cells (Swidsinski et al., 2005). tion of a polymicrobial BV biofilm (Machado et al., 2013;
Extracellular DNA is thought to originate from lysed Pybus & Onderdonk, 1997). There is evidence suggest-
cells and provides structural integrity and stability to ing G. vaginalis as an early colonizer species and other
biofilms in Bacillus cereus, L. monocytogenes and S. BV-related bacteria as second/third colonizer species
aureus (Whitchurch et al., 2002). A study by Hymes et al. during the development of a polymicrobial BV biofilm
(2013) showed that the G. vaginalis biofilms contain the (Alves et al., 2014; Machado et al., 2013; Patterson et al.,
eDNA and that DNase treatment (which degrades 2010); however, there is still a possibility that another
12 H.-S. JUNG ET AL.

BV-related anaerobe like Peptoniphilus spp. may initiate Aroutcheva AA, Simoes JA, Behbakht K, Faro S. (2001).
the biofilm process (Patterson et al., 2010). Gardnerella vaginalis isolated from patients with bacterial
vaginosis and from patients with healthy vaginal ecosys-
The limitation of BV biofilm research is that suitable
tems. Clin Infect Dis 33:1022–7.
in vivo models are lacking (Machado & Cerca, 2015). The Aroutcheva AA, Simoes JA, Faro S. (2001). Antimicrobial pro-
in vitro studies can provide some evidence on how BV tein produced by vaginal Lactobacillus acidophilus that
could be caused; however, the information obtained inhibits Gardnerella vaginalis. Infect Dis Obstet Gynecol
from in vitro studies might not guarantee the under- 9:33–9.
Austin MN, Rabe LK, Srinivasan S, et al. (2015). Anaerobe
standing or improve results in vivo (i.e. in the vagina).
Mageeibacillus indolicus gen. nov., sp. nov.: a novel bacter-
Conversely, in vivo models like the murine vaginal epi- ium isolated from the female genital tract. Anaerobe
thelium may also not be successful in studying BV bio- 32:37–42.
films, as the murine vaginal epithelium does not Bartlett JG, Onderdonk AB, Drude E, et al. (1977).
resemble the in vivo environment of the human vagina Quantitative bacteriology of the vaginal flora. J Infect Dis
136:271–7.
(such as keratinized epithelial cells and the higher pH Bartlett JG, Polk BF. (1984). Bacterial flora of the vagina:
than that of the human vagina) (Breshears et al., 2015; quantitative study. Rev Infect Dis 6:S67–S72.
Herbst-Kralovetz et al., 2016). Therefore, we might need Begum A, Nilufar S, Akther K, et al. (2003). Prevalence of
to use every available in vitro and in vivo system, or use selected reproductive tract infections among pregnant
women attending an urban childcare unit in Dhaka,
a combination of both systems to decipher the complex
Bangladesh. J Heal Popul Nutr 21:112–16.
interactions involved in BV biofilms (Herbst-Kralovetz Beigi RH, Wiesenfeld HC, Hillier SL, et al. (2005). Factors asso-
et al., 2016). The choice of which model to use for such ciated with absence of H2O2-producing Lactobacillus
studies will depend on the research question being among women with bacterial vaginosis. J Infect Dis
asked (Herbst-Kralovetz et al., 2016). In future studies, 191:924–9.
Bertran T, Brachet P, Vareille-Delarbre M, et al. (2016). Slight
the following research gaps could be essential to better
pro-inflammatory immunomodulation properties of den-
understand and to define the etiology of BV, such as: (i) dritic cells by Gardnerella vaginalis: the “invisible man” of
the dispersal mechanism in a polymicrobial BV biofilm; bacterial vaginosis? J Immunol Res 2016:1–13.
(ii) transcriptomic studies to study gene expression dur- Blanchard A, Razin S, Kenny GE, Barile MF. (1988).
ing the formation of a polymicrobial BV biofilm and (iii) Characteristics of Ureaplasma urealyticum urease. J
Bacteriol 170:2692–7.
quorum sensing in a polymicrobial BV biofilm. Borlee BR, Goldman AD, Murakami K, et al. (2010).
Pseudomonas aeruginosa uses a cyclic-di-GMP-regulated
adhesin to reinforce the biofilm extracellular matrix. Mol
Disclosure statement Microbiol 75:827–42.
The authors declare no potential conflict of financial interest. Boskey ER, Cone RA, Whaley KJ, Moench TR. (2001). Origins
The opinions expressed in this review are those of the of vaginal acidity: high D/L lactate ratio is consistent with
bacteria being the primary source. Hum Reprod
authors and do not necessarily reflect the opinions of the
16:1809–13.
MRC of South Africa.
Boskey ER, Telsch KM, Whaley KJ, et al. (1999). Acid produc-
tion by vaginal flora in vitro is consistent with the rate and
extent of vaginal acidification. Infect Immun 67:5170–5.
Funding Botta GA, Eftimiadi C, Costa A, et al. (1985). Influence of vola-
This work was supported by the South African Medical tile fatty acids on human granulocyte chemotaxis. FEMS
Research Council (MRC). Microbiol Lett 27:69–72.
Bradshaw CS, Tabrizi SN, Fairley CK, et al. (2006). The associ-
ation of Atopobium vaginae and Gardnerella vaginalis with
References bacterial vaginosis and recurrence after oral metronidazole
therapy. J Infect Dis 194:828–36.
Africa CWJ, Nel J, Stemmet M. (2014). Anaerobes and bacter- Breshears LM, Edwards VL, Ravel J, Peterson ML. (2015).
ial vaginosis in pregnancy: virulence factors contributing Lactobacillus crispatus inhibits growth of Gardnerella vagi-
to vaginal colonisation. Int J Environ Res Public Health nalis and Neisseria gonorrhoeae on a porcine vaginal
11:6979–7000. mucosa model. BMC Microbiol 15:276.
Al-Mushrif S, Eley A, Jones BM. (2000). Inhibition of chemo- Briselden AM, Moncla BJ, Stevens CE, Hillier SL. (1992).
taxis by organic acids from anaerobes may prevent a puru- Sialidases (neuraminidases) in bacterial vaginosis and bac-
lent response in bacterial vaginosis. J Med Microbiol terial vaginosis-associated microflora. J Clin Microbiol
49:1023–30. 30:663–6.
Alves P, Castro J, Sousa C, et al. (2014). Gardnerella vaginalis Cassini MA, Pilloni A, Condo  SG, et al. (2013). Periodontal
outcompetes 29 other bacterial species isolated from bacteria in the genital tract: are they related to adverse
patients with bacterial vaginosis, using in an in vitro bio- pregnancy outcome?. Int J Immunopathol Pharmacol
film formation model. J Infect Dis 210:593–6. 26:931–9.
CRITICAL REVIEWS IN MICROBIOLOGY 13

Castro J, Alves P, Sousa C, et al. (2015). Using an in-vitro bio- and its meaning for the puerperal fever]. Leipzig: Verlag
film model to assess the virulence potential of bacterial von Eduard Besold.
vaginosis or non-bacterial vaginosis Gardnerella vaginalis Doerflinger SY, Throop AL, Herbst-Kralovetz MM. (2014).
isolates. Sci Rep 5:1–10. Bacteria in the vaginal microbiome alter the innate
Castro J, Cerca N. (2015). BV and non-BV associated immune response and barrier properties of the human
Gardnerella vaginalis establish similar synergistic interac- vaginal epithelia in a species-specific manner. J Infect Dis
tions with other BV-associated microorganisms in dualspe- 209:1989–99.
cies biofilms. Anaerobe 36:56–9. Donders GGG, Vereecken A, Bosmans E, et al. (2002).
Castro J, Henriques A, Machado A, et al. (2013). Reciprocal Definition of a type of abnormal vaginal flora that is dis-
interference between Lactobacillus spp. and Gardnerella tinct from bacterial vaginosis: aerobic vaginitis. BJOG
vaginalis on initial adherence to epithelial cells. Int J Med 109:34–43.
Sci 10:1193–8. Donelli G, Vuotto C, Cardines R, Mastrantonio P. (2012).
Castro J, Machado D, Cerca N. (2016). Escherichia coli and Biofilm-growing intestinal anaerobic bacteria. FEMS
Enterococcus faecalis are able to incorporate and enhance Immunol Med Microbiol 65:318–25.
a pre-formed Gardnerella vaginalis biofilm. Pathog Dis Ezaki T, Kawamura Y, Li N, et al. (2001). Proposal of the gen-
74:ftw007. era Anaerococcus gen. nov., Peptoniphilus gen. nov. and
Cauci S, Culhane JF, Di Santolo M, McCollum K. (2008). Gallicola gen. nov for members of the genus
Among pregnant women with bacterial vaginosis, the Peptostreptococcus. Int J Syst Evol Microbiol 51:1521–8.
hydrolytic enzymes sialidase and prolidase are positively Ferris MJ, Masztal A, Aldridge KE, et al. (2004). Association of
associated with interleukin-1. Am J Obstet Gynecol Atopobium vaginae, a recently described metronidazole
198:132.e1–e7. resistant anaerobe, with bacterial vaginosis. BMC Infect Dis
Cauci S, Guaschino S, Driussi S, et al. (2002). Correlation of 4:5.
local interleukin-8 with immunoglobulin A against Ferris MJ, Norori J, Zozaya-Hinchliffe M, Martin DH. (2007).
Gardnerella vaginalis hemolysin and with prolidase and Cultivation-independent analysis of changes in bacterial
sialidase levels in women with bacterial vaginosis. J Infect vaginosis flora following metronidazole treatment. J Clin
Dis 185:1614–20. Microbiol 45:1016–18.
Chen KCS, Forsyth PS, Buchanan TM, Holmes KK. (1979). Fethers K, Twin J, Fairley CK, et al. (2012). Bacterial vaginosis
Amine content of vaginal fluid from untreated and treated (BV) candidate bacteria: associations with BV and behav-
patients with nonspecific vaginitis. J Clin Invest 63:828–35. ioural practices in sexually-experienced and inexperienced
Cherpes TL, Hillier SL, Meyn LA, et al. (2008). A delicate bal- women. PLoS One 7:e30633.
ance: risk factors for acquisition of bacterial vaginosis Fichorova RN, Buck OR, Yamamoto HS, et al. (2013). The vil-
include sexual activity, absence of hydrogen peroxide-pro- lain team-up or how Trichomonas vaginalis and bacterial
ducing lactobacilli, black race, and positive herpes simplex vaginosis alter innate immunity in concert. Sex Transm
virus type 2 serology. Sex Transm Dis 35:78–83. Infect 89:460–6.
Cherpes TL, Meyn LA, Krohn MA, et al. (2003). Association Fredricks DN, Fiedler TL, Marrazzo JM. (2005). Molecular iden-
between acquisition of herpes simplex virus type 2 in tification of bacteria associated with bacterial vaginosis. N
women and bacterial vaginosis. Clin Infect Dis 37:319–25. Engl J Med 353:1899–911.
Costello J, Blaize J, L’Amoreaux WJ, McCoy EC. (2007). Gajer P, Brotman RM, Bai G, et al. (2012). Temporal dynamics
Ultrastructure of the pathogenic bacteria Mobiluncus mulie- of the human vaginal microbiota. Sci Transl Med
ris. MAM 13:336–7. 4:132ra52.
Coudeyras S, Jugie G, Vermerie M, Forestier C. (2008). Ganderton L, Chawla J, Winters C, et al. (1992). Scanning
Adhesion of human probiotic Lactobacillus rhamnosus to electron microscopy of bacterial biofilms on indwelling
cervical and vaginal cells and interaction with vaginosisas- bladder catheters. Eur J Clin Microbiol Infect Dis
sociated pathogens. Infect Dis Obstet Gynecol 11:789–96.
2008:549640. doi:10.1155/2008/549640 Gardner H, Dukes C. (1955). Haemophilus vaginalis vaginitis: a
Danielsson D, Teigen PK, Moi H. (2011). The genital econi- newly defined specific infection previously classified
che:focus on microbiota and bacterial vaginosis. Ann N Y “nonspecific” vaginitis. Am J Obstet Gynecol 69:962–76.
Acad Sci 1230:48–58. Gardner HL, Dukes CD. (1954). New etiologic agent in non-
De Backer E, Verhelst R, Verstraelen H, et al. (2007). specific bacterial vaginitis. Science 120:853.
Quantitative determination by real-time PCR of four vagi- Geißdo €rfer W, Bo
€hmer C, Pelz K, et al. (2003). Tuboovarian
nal Lactobacillus species, Gardnerella vaginalis and abscess caused by Atopobium vaginae following transvagi-
Atopobium vaginae indicates an inverse relationship nal oocyte recovery. J Clin Microbiol 41:2788–90.
between L. gasseri and L. iners. BMC Microbiol 7:115. Gilbert NM, Lewis WG, Lewis AL. (2013). Clinical features of
De Boer JM, Plantema FHF. (1988). Ultrastructure of the in bacterial vaginosis in a murine model of vaginal infection
situ adherence of Mobiluncus to vaginal epithelial cells. with Gardnerella vaginalis. PLoS One 8:e59539.
Can J Microbiol 34:757–66. Greenwood JR, Pickett MJ. (1979). Salient features of
De Silva NS, Quinn PA. (1986). Endogenous activity of phos- Haemophilus vaginalis. J Clin Microbiol 9:200–4.
pholipases A and C in Ureaplasma urealyticum. J Clin Greenwood JR, Pickett MJ. (1980). Transfer of Haemophilus
Microbiol 23:354–9. vaginalis Gardner and Dukes to a new genus, Gardnerella:
€derlein A. (1892). Das Scheidensekret und seine
Do G. vaginalis (Gardner and Dukes) comb. nov. Int J Syst
Bedeutung fu €r das Puerperalfieber [The vaginal secretion Bacteriol 30:170–8.
14 H.-S. JUNG ET AL.

Haggerty CL, Hillier SL, Bass DC, Ness RB. (2004). Bacterial Kageyama A, Benno Y, Nakase T. (1999). Phylogenetic evi-
vaginosis and anaerobic bacteria are associated with endo- dence for the transfer of Eubacterium lentum to the genus
metritis. Clin Infect Dis 39:990–5. Eggerthella as Eggerthella lenta gen. nov., comb. nov. Int J
Haggerty CL, Totten PA, Ferris M, et al. (2009). Clinical charac- Syst Bacteriol 49:1725–32.
teristics of bacterial vaginosis among women testing posi- Kapatral V, Anderson I, Ivanova N, et al. (2005). Genome
tive for fastidious bacteria. Sex Transm Infect 85:242–8. sequence and analysis of the oral bacterium Fusobacterium
Hardy L, Jespers V, Abdellati S, et al. (2016). A fruitful alliance: nucleatum strain ATCC 25586. J Bacteriol 184:2005–18.
the synergy between Atopobium vaginae and Gardnerella Kaplan CW, Lux R, Haake SK, Shi W. (2009). The
vaginalis in bacterial vaginosis-associated biofilm. Sex Fusobacterium nucleatum outer membrane protein RadD is
Transm Infect 92:487–91. an arginine-inhibitable adhesin required for inter-species
Harwich MD, Alves JM, Buck GA, et al. (2010). Drawing the adherence and the structured architecture of multispecies
line between commensal and pathogenic Gardnerella vagi- biofilm. Mol Microbiol 71:35–47.
nalis through genome analysis and virulence studies. BMC Karczewski J, Troost FJ, Konings I, et al. (2010). Regulation of
Genomics 11:375. human epithelial tight junction proteins by Lactobacillus
Harwich MD, Serrano MG, Fettweis JM, et al. (2012). Genomic plantarum in vivo and protective effects on the epithelial
sequence analysis and characterization of Sneathia amnii barrier. Am J Physiol - Gastrointest Liver Physiol 298:851–9.
sp. nov. BMC Genomics 13:S4. Kenyon C, Colebunders R, Crucitti T. (2013). The global epi-
Hawes SE, Hillier SL, Benedetti J, et al. (1996). Hydrogen per- demiology of bacterial vaginosis: a systematic review. Am
oxide-producing lactobacilli and acquisition of vaginal J Obstet Gynecol 209:505–23.
infections. J Infect Dis 174:1058–63. Kiliç AO, Pavlova SI, Alpay S, et al. (2001). Comparative study
Herbst-Kralovetz MM, Pyles RB, Ratner AJ, et al. (2016). New of vaginal Lactobacillus phages isolated from women in
systems for studying intercellular interactions in bacterial the United States and Turkey: prevalence, morphology,
vaginosis. J Infect Dis 214:S6–S13. host range, and DNA homology. Clin Diagn Lab Immunol
Hillier SL, Krohn MA, Rabe LK, et al. (1993). The normal vagi- 8:31–9.
nal flora, H2O2-producing lactobacilli, and bacterial vagin- Kim TK, Thomas SM, Ho M, et al. (2009). Heterogeneity of
osis in pregnant women. Clin Infect Dis 16:S273–S81. vaginal microbial communities within individuals. J Clin
Hillier SL, Nugent RP, Eschenbach DA, et al. (1995). Microbiol 47:1181–9.
Association between bacterial vaginosis and preterm deliv- Klebanoff MA, Schwebke JR, Zhang J, et al. (2004).
ery of low-birth-weight infant. N Engl J Med 333:1737–42. Vulvovaginal symptoms in women with bacterial vaginosis.
Holzman C, Leventhal JM, Qiu H, et al. (2001). Factors linked Obstet Gynecol 104:267–72.
to bacterial vaginosis in nonpregnant women. Am J Public Klebanoff SJ, Hillier SL, Eschenbaeh DA, Waltersdorph AM.
Health 91:1664–70. (1991). Control of the microbial flora of the vagina by
Hooton TM, Fennell CL, Clark AM, Stamm WE. (1991). H2O2-generating lactobacilli. J Infect Dis 164:94–100.
Nonoxynol-9: differential antibacterial activity and Koumans EH, Sternberg M, Bruce C, et al. (2007). The preva-
enhancement of bacterial adherence to vaginal epithelial lence of bacterial vaginosis in the United States, 2001-
cells. J Infect Dis 164:1216–19. 2004; associations with symptoms, sexual behaviors, and
Hovhannisyan HG, Grigoryan GG. (2014). A new sustainable reproductive health. Sex Transm Dis 34:864–9.
symbiotic association of lactic acid cocci and bacilli for col- Kubota H, Senda S, Nomura N, et al. (2008). Biofilm formation
onization/recolonization of vagina and prevention of bac- by lactic acid bacteria and resistance to environmental
terial vaginosis. AJBIO 2:84–8. stress. J Biosci Bioeng 106:381–6.
Hyman RW, Fukushima M, Diamond L, et al. (2005). Microbes Kurita-Ochiai T, Ochiai K, Fukushima K. (1998). Volatile fatty
on the human vaginal epithelium. Proc Natl Acad Sci U S acid, metabolic byproduct of periodontopathic bacteria,
A 102:7952–7. induces apoptosis in WEHI 231 and RAJI B lymphoma cells
Hymes SR, Randis TM, Sun TY, Ratner AJ. (2013). DNase inhib- and splenic B cells. Infect Immun 66:2587–94.
its Gardnerella vaginalis biofilms in vitro and in vivo. Lee H-W, Koh YM, Kim J, et al. (2008). Capacity of multidrug-
J Infect Dis 207:1491–7. resistant clinical isolates of Acinetobacter baumannii to
Jayaprakash TP, Schellenberg JJ, Hill JE. (2012). Resolution form biofilm and adhere to epithelial cell surfaces. Clin
and characterization of distinct cpn60-based subgroups of Microbiol Infect 14:49–54.
Gardnerella vaginalis in the vaginal microbiota. PLoS One Lepp€aluoto PA. (2011). Bacterial vaginosis: what is physio-
7:e43009. logical in vaginal bacteriology? An update and opinion.
Johnson AP, Davies HA. (1984). Demonstration by electron Acta Obstet Gynecol Scand 90:1302–6.
microscopy of pili on Gardnerella vaginalis. Br J Vener Dis Lewis AL, Lewis WG. (2012). Host sialoglycans and bacterial
60:396–7. sialidases: a mucosal perspective. Cell Microbiol
Jordan SJ, Perni S, Glenn S, et al. (2008). Listeria monocyto- 14:1174–82.
genes biofilm-associated protein (BapL) may contribute to Lewis WG, Robinson LS, Perry J, et al. (2012). Hydrolysis of
surface attachment of L. monocytogenes but is absent from secreted sialoglycoprotein immunoglobulin A (IgA) in ex
many field isolates. Appl Environ Microbiol 74:5451–6. vivo and biochemical models of bacterial vaginosis. J Biol
Jovita MR, Collins MD, Sjo €den B, Falsen E. (1999). Chem 287:2079–89.
Characterization of a novel Atopobium isolate from the Libby EK, Pascal KE, Mordechai E, et al. (2008). Atopobium
human vagina: description of Atopobium vaginae sp. nov. vaginae triggers an innate immune response in an in vitro
Int J Syst Bacteriol 49:1573–6. model of bacterial vaginosis. Microbes Infect 10:439–46.
CRITICAL REVIEWS IN MICROBIOLOGY 15

Ling Z, Kong J, Liu F, et al. (2010). Molecular analysis of the mediator for persistence in the vagina?. Reprod Sci
diversity of vaginal microbiota associated with bacterial 20:791–6.
vaginosis. BMC Genomics 11:488. Mendes-Soares H, Krishnan V, Settles ML, et al. (2015).
Lopes Dos Santos Santiago G, Deschaght P, El Aila N, et al. Finescale analysis of 16S rRNA sequences reveals a high
(2011). Gardnerella vaginalis comprises three distinct geno- level of taxonomic diversity among vaginal Atopobium
types of which only two produce sialidase. Am J Obstet spp. Pathog Dis 73:ftv020.
Gynecol 204:450.e1–e7. Mirmonsef P, Zariffard MR, Gilbert D, et al. (2012). Short-chain
Lubbe MM, Stanley K, Chalkley LJ. (1999). Prevalence of nim fatty acids induce pro-inflammatory cytokine production
genes in anaerobic/facultative anaerobic bacteria isolated alone and in combination with Toll-like receptor ligands.
in South Africa. FEMS Microbiol Lett 172:79–83. Am J Reprod Immunol 67:391–400.
Machado A, Cerca N. (2015). Influence of biofilm formation Muli FW, Struthers JK. (1998). The growth of Gardnerella vagi-
by Gardnerella vaginalis and other anaerobes on bacterial nalis and Lactobacillus acidophilus in Sorbarod biofilms. J
vaginosis. J Infect Dis 212:1856–61. Med Microbiol 47:401–5.
Machado A, Jefferson KK, Cerca N. (2013). Interactions Muzny CA, Schwebke JR. (2013). Gardnerella vaginalis: still a
between Lactobacillus crispatus and bacterial vaginosis prime suspect in the pathogenesis of bacterial vaginosis.
(BV)-associated bacterial species in initial attachment and Curr Infect Dis Rep 15:130–5.
biofilm formation. Int J Mol Sci 14:12004–12. Muzny CA, Schwebke JR. (2015). Biofilms: an underappreci-
Machado A, Salgueiro D, Harwich M, et al. (2013). ated mechanism of treatment failure and recurrence in
Quantitative analysis of initial adhesion of bacterial vagin- vaginal infections. Clin Infect Dis 61:601–6.
osis-associated anaerobes to ME-180 cells. Anaerobe Muzny CA, Sunesara IR, Griswold ME, et al. (2014).
23:1–4. Association between BVAB1 and high Nugent scores
Macklaim JM, Fernandes AD, Di Bella JM, et al. (2013). among women with bacterial vaginosis. Diagn Microbiol
Comparative meta-RNA-seq of the vaginal microbiota and Infect Dis 80:321–3.
differential expression by Lactobacillus iners in health and Nagaoka S, Hojo K, Murata S, et al. (2008). Interactions
dysbiosis. Microbiome 1:12. between salivary Bifidobacterium adolescentis and other
Marconi C, Cruciani F, Vitali B, Donders GGG. (2012). oral bacteria: in vitro coaggregation and coadhesion
Correlation of Atopobium vaginae amount with bacterial assays. FEMS Microbiol Lett 281:183–9.
vaginosis markers. J Low Genit Tract Dis 16:127–32. Nagy E, Petterson M, Mårdh PA. (1991). Antibiosis between
Marconi C, Donders GG, Parada CMGL, et al. (2013). Do bacteria isolated from the vagina of women with and
Atopobium vaginae, Megasphaera sp. and Leptotrichia sp. without signs of bacterial vaginosis. APMIS 99:739–44.
change the local innate immune response and sialidase Ness RB, Hillier SL, Richter HE, et al. (2002). Douching in rela-
activity in bacterial vaginosis?. Sex Transm Infect 89:167–73. tion to bacterial vaginosis, lactobacilli, and facultative bac-
Marrazzo JM, Thomas KK, Fiedler TL, et al. (2008). teria in the vagina. Obstet Gynecol 100:765–72.
Relationship of specific vaginal bacteria and bacterial Ness RB, Kip KE, Hillier SL, et al. (2005). A cluster analysis of
vaginosis treatment failure in women who have sex with bacterial vaginosis-associated microflora and pelvic inflam-
women. Ann Intern Med 149:20–8. matory disease. Am J Epidemiol 162:585–90.
Marrs CN, Knobel SM, Zhu WQ, et al. (2012). Evidence for Nikolaitchouk N, Andersch B, Falsen E, et al. (2008). The lower
Gardnerella vaginalis uptake and internalization by squa- genital tract microbiota in relation to cytokine-, SLPI- and
mous vaginal epithelial cells: implications for the patho- endotoxin levels: application of checkerboard DNA-DNA
genesis of bacterial vaginosis. Microbes Infect 14:500–8. hybridization (CDH). APMIS 116:263–77.
Martın R, Sobero n N, Escobedo S, Suarez JE. (2009). O’Hanlon DE, Moench TR, Cone RA. (2013). Vaginal pH and
Bacteriophage induction versus vaginal homeostasis: role microbicidal lactic acid when lactobacilli dominate the
of H2O2 in the selection of Lactobacillus defective pro- microbiota. PLoS One 8:1–8.
phages. Int Microbiol 12:131–6. Oakley BB, Fiedler TL, Marrazzo JM, Fredricks DN. (2008).
Masover GK, Razin S, Hayflick L. (1977). Effects of carbon Diversity of human vaginal bacterial communities and
dioxide, urea, and ammonia on growth of Ureaplasma ure- associations with clinically defined bacterial vaginosis.
alyticum (T-strain mycoplasma). J Bacteriol 130:292–6. Appl Environ Microbiol 74:4898–909.
Mattsby-Baltzer I, Platz-Christensen JJ, Hosseini N, Rosen P. Okuda T, Kokubu E, Kawana T, et al. (2012). Synergy in bio-
(1998). IL-1_, IL-6, TNFa, fetal fibronectin, and endotoxin in film formation between Fusobacterium nucleatum and
the lower genital tract of pregnant women with bacterial Prevotella species. Anaerobe 18:110–16.
vaginosis. Acta Obstet Gynecol Scand 77:701–6. Olmsted SS, Meyn LA, Rohan LC, Hillier SL. (2003).
Mayer BT, Srinivasan S, Fiedler TL, et al. (2015). Rapid and Glycosidase and proteinase activity of anaerobic Gram-
profound shifts in the vaginal microbiota following anti- negative bacteria isolated from women with bacterial
biotic treatment for bacterial vaginosis. J Infect Dis vaginosis. Sex Transm Dis 30:257–61.
212:793–802. Patterson JL. (2010). Characterization of adherence, cytotox-
McCormack WM, Braun P, Lee Y-H, et al. (1973). The genital icity and biofilm formation by Gardnerella vaginalis [disser-
mycoplasmas. N Engl J Med 288:78–89. tation]. Richmond (VA), USA: Virginia Commonwealth
McMillan A, Dell M, Zellar MP, et al. (2011). Disruption of uro- University.
genital biofilms by lactobacilli. Colloids Surf B Biointerfaces Patterson JL, Girerd PH, Karjane NW, Jefferson KK. (2007).
86:58–64. Effect of biofilm phenotype on resistance of Gardnerella
McMillan A, Macklaim JM, Burton JP, Reid G. (2012). Adhesion vaginalis to hydrogen peroxide and lactic acid. Am J
of Lactobacillus iners AB-1 to human fibronectin: a key Obstet Gynecol 197:170.e1–e7.
16 H.-S. JUNG ET AL.

Patterson JL, Stull-Lane A, Girerd PH, Jefferson KK. (2010). Schwebke JR, Flynn MS, Rivers CA. (2014a). Prevalence of
Analysis of adherence, biofilm formation and cytotoxicity Gardnerella vaginalis among women with Lactobacillus-pre-
suggests a greater virulence potential of Gardnerella vagi- dominant vaginal flora. Sex Transm Infect 90:61–3.
nalis relative to other bacterial-vaginosis-associated anae- Schwebke JR, Muzny CA, Josey WE. (2014b). Role of
robes. Microbiology (Reading, Engl) 156:392–9. Gardnerella vaginalis in the pathogenesis of bacterial
Pavlova SI, Kiliç AO, Mou SM, Tao L. (1997). Phage infection vaginosis: a conceptual model. J Infect Dis 210:338–43.
in vaginal lactobacilli: an in vitro study. Infect Dis Obstet Singh R, Ray P, Das A, Sharma M. (2010). Penetration of
Gynecol 5:36–44. antibiotics through Staphylococcus aureus and
Pavlova SI, Tao L. (2000). In vitro inhibition of commercial Staphylococcus epidermidis biofilms. J Antimicrob
douche products against vaginal microflora. Infect Dis Chemother 65:1955–8.
Obstet Gynecol 8:99–104. Sobel JD, Schneider J, Kaye D, Levison ME. (1981). Adherence
Peeters M, Piot P. (1985). Adhesion of Gardnerella vaginalis to of bacteria to vaginal epithelial cells at various times in
vaginal epithelial cells: variables affecting adhesion and the menstrual cycle. Infect Immun 32:194–7.
inhibition by metronidazole. Genitourin Med 61:391–5. Spiegel CA. (1991). Bacterial vaginosis. Clin Microbiol Rev
Periasamy S, Kolenbrander PE. (2009). Aggregatibacter actino- 4:458–502.
mycetemcomitans builds mutualistic biofilm communities Spiegel CA, Amsel R, Eschenbach D, et al. (1980). Anaerobic
with Fusobacterium nucleatum and Veillonella species in bacteria in nonspecific vaginitis. N Engl J Med 303:601–7.
saliva. Infect Immun 77:3542–51. Spiegel CA, Davick P, Totten PA, et al. (1983). Gardnerella
Piot P, Van Dyck E, Godts P, Vanderheyden J. (1982). The vaginalis and anaerobic bacteria in the etiology of bacter-
vaginal microbial flora in non-specific vaginitis. Eur J Clin ial (nonspecific) vaginosis. Scand J Infect Dis 40:41–6.
Microbiol 1:301–6. Spiegel CA, Roberts M. (1984). Mobiluncus gen. nov.,
Piot P, van Dyck E, Peeters M, et al. (1984). Biotypes of Mobiluncus curtisii subsp. curtisii sp. nov., Mobiluncus curti-
Gardnerella vaginalis. J Clin Microbiol 20:677–9. sii subsp. holmesii subsp. nov., and Mobiluncus mulieris sp.
Pybus V, Onderdonk AB. (1996). The effect of pH on growth nov., curved rods from the human vagina. Int J Syst
and succinate production by Prevotella bivia. Microb Ecol Bacteriol 34:177–84.
Health Dis 9:19–25. Srinivasan S, Fredricks DN. (2008). The human vaginal bacter-
Pybus V, Onderdonk AB. (1997). Evidence for a commensal,
ial biota and bacterial vaginosis. Interdiscip Perspect Infect
symbiotic relationship between Gardnerella vaginalis and
Dis 2008:750479.
Prevotella bivia involving ammonia: potential significance
Srinivasan S, Hoffman NG, Morgan MT, et al. (2012). Bacterial
for bacterial vaginosis. J Infect Dis 175:406–13.
communities in women with bacterial vaginosis: high reso-
Pybus V, Onderdonk AB. (1998). A commensal symbiosis
lution phylogenetic analyses reveal relationships of micro-
between Prevotella bivia and Peptostreptococcus anaerobius
biota to clinical criteria. PLoS One 7:e37818.
involves amino acids: potential significance to the patho-
Srinivasan S, Morgan MT, Fiedler TL, et al. (2015). Metabolic
genesis of bacterial vaginosis. FEMS Immunol Med
signatures of bacterial vaginosis. MBio 6:1–16.
Microbiol 22:317–27.
Stålhammar-Carlemalm M, Stenberg L, Lindahl G. (1993).
Ralph SG, Rutherford AJ, Wilson JD. (1999). Influence of bac-
Protein Rib: a novel group B streptococcal cell surface pro-
terial vaginosis on conception and miscarriage in the first
tein that confers protective immunity and is expressed by
trimester: cohort study. BMJ 319:220–3.
Randis TM, Zaklama J, LaRocca TJ, et al. (2013). Vaginolysin most strains causing invasive infections. J Exp Med
drives epithelial ultrastructural responses to Gardnerella 177:1593–603.
vaginalis. Infect Immun 81:4544–50. Stoodley P, Wilson S, Hall-Stoodley L, et al. (2001). Growth
Ravel J, Gajer P, Abdo Z, et al. (2011). Vaginal microbiome of and detachment of cell clusters from mature mixed-spe-
reproductive-age women. Proc Natl Acad Sci USA cies biofilms. Appl Environ Microbiol 67:5608–13.
108:4680–7. Sturm-Ramirez K, Gaye-Diallo A, Eisen G, et al. (2000). High
Rickard AH, Gilbert P, High NJ, et al. (2003). Bacterial coag- levels of tumor necrosis factor-a and interleukin-1b in bac-
gregation: an integral process in the development of terial vaginosis may increase susceptibility to human
multi-species biofilms. Trends Microbiol 11:94–100. immunodeficiency virus. J Infect Dis 182:467–73.
Roberton AM, Wiggins R, Horner PJ, et al. (2005). A novel Swidsinski A, Do €rffel Y, Loening-Baucke V, et al. (2011).
bacterial mucinase, glycosulfatase, is associated with bac- Response of Gardnerella vaginalis biofilm to 5 days of
terial vaginosis. J Clin Microbiol 43:5504–8. moxifloxacin treatment. FEMS Immunol Med Microbiol
Robertson JA, Stemler ME, Stemke GW. (1984). 61:41–6.
Immunoglobulin A protease activity of Ureaplasma urealyti- Swidsinski A, Loening-baucke V, Mendling W, et al. (2014).
cum. J Clin Microbiol 19:255–8. Infection through structured polymicrobial Gardnerella bio-
Sapi E, Bastian SL, Mpoy CM, et al. (2012). Characterization of films (StPM-GB). Histol Histopathol 29:567–87.
biofilm formation by Borrelia burgdorferi in vitro. PLoS One Swidsinski A, Loening-Baucke V, Swidsinski S, Verstraelen H.
7:1–11. (2015). Polymicrobial Gardnerella biofilm resists repeated
Sauer K, Camper AK, Ehrlich GD, et al. (2002). Pseudomonas intravaginal antiseptic treatment in a subset of women
aeruginosa displays multiple phenotypes during develop- with bacterial vaginosis: a preliminary report. Arch Gynecol
ment as a biofilm. J Bacteriol 184:1140–54. Obstet 291:605–9.
Schwebke JR, Desmond R. (2007). Natural history of asymp- Swidsinski A, Mendling W, Loening-Baucke V, et al. (2005).
tomatic bacterial vaginosis in a high-risk group of women. Adherent biofilms in bacterial vaginosis. Obstet Gynecol
Sex Transm Dis 34:876–7. 106:1013–23.
CRITICAL REVIEWS IN MICROBIOLOGY 17

Swidsinski A, Mendling W, Loening-baucke V, et al. (2008). An Ventolini G, Mitchell E, Salazar M. (2015). Biofilm formation
adherent Gardnerella vaginalis biofilm persists on the vagi- by vaginal Lactobacillus in vivo. Med Hypotheses
nal epithelium after standard therapy with oral metronida- 84:417–420.
zole. Am J Obstet Gynecol 198:97.e1–.e6. Verhelst R, Verstraelen H, Claeys G, et al. (2004). Cloning of
Swidsinski A, Verstraelen H, Loening-Baucke V, et al. (2013). 16S rRNA genes amplified from normal and disturbed
Presence of a polymicrobial endometrial biofilm in patients vaginal microflora suggests a strong association between
with bacterial vaginosis. PLoS One 8:e53997. Atopobium vaginae, Gardnerella vaginalis and bacterial
Tao F, Swarup S, Zhang LH. (2010). Quorum sensing modula- vaginosis. BMC Microbiol 4:16.
tion of a putative glycosyltransferase gene cluster essential Verstraelen H, Verhelst R, Claeys G, et al. (2004).
for Xanthomonas campestris biofilm formation. Environ Cultureindependent analysis of vaginal microflora: the
Microbiol 12:3159–70. unrecognized association of Atopobium vaginae with bac-
Tao L, Pavlova SI, Mou SM, et al. (1997). Analysis of terial vaginosis. Am J Obstet Gynecol 191:1130–2.
Lactobacillus products for phages and bacteriocins that Wang X, Li X, Zhao C, et al. (2012). Correlation between com-
inhibit vaginal lactobacilli. Infect Dis Obstet Gynecol position of the bacterial community and concentration of
5:244–51. volatile fatty acids in the rumen during the transition
Taylor-Robinson AW, Borriello SP, Taylor-Robinson D. (1993). period and ketosis in dairy cows. Appl Environ Microbiol
Identification and preliminary characterization of a cytotoxin 78:2386–92.
isolated from Mobiluncus spp. Int J Exp Pathol 74:357–66. Whitchurch CB, Tolker-Nielsen T, Ragas PC, Mattick JS. (2002).
Terraf MCL, Mendoza LM, Tomas MSJ, et al. (2014). Extracellular DNA required for bacterial biofilm formation.
Phenotypic surface properties (aggregation, adhesion and Science 295:1487.
biofilm formation) and presence of related genes in bene- Whittaker CJ, Klier CM, Kolenbrander PE. (1996). Mechanisms
ficial vaginal lactobacilli. J Appl Microbiol 117:1761–72. of adhesion by oral bacteria. Annu Rev Microbiol
Theilacker C, Sava I, Sanchez-Carballo P, et al. (2011). 50:513–52.
Deletion of the glycosyltransferase bgsB of Enterococcus Wilson JD, Ralph SG, Rutherford AJ. (2002). Rates of bacterial
vaginosis in women undergoing in vitro fertilisation for dif-
faecalis leads to a complete loss of glycolipids from the
ferent types of infertility. BJOG 109:714–17.
cell membrane and to impaired biofilm formation. BMC
Witkin SS, Linhares IM, Giraldo P. (2007). Bacterial flora of the
Microbiol 11:67.
female genital tract: function and immune regulation. Best
Theron MM, Janse van Rensburg MN, Chalkley LJ. (2004).
Pract Res Clin Obstet Gynaecol 21:347–54.
Nitroimidazole resistance genes (nimB) in anaerobic Gram-
Wolrath H, Forsum U, Larsson PG, Bor en H. (2001). Analysis of
positive cocci (previously Peptostreptococcus spp.). J
bacterial vaginosisrelated amines in vaginal fluid by gas
Antimicrob Chemother 54:240–2.
chromatography and mass spectrometry. J Clin Microbiol
Thomas S. (1928). Do €derlein’s bacillus: Lactobacillus acidoph-
39:4026–31.
ilus. J Infect Dis 43:218–27.
Xia Q, Cheng L, Zhang H, et al. (2016). Identification of vagi-
Thurlow LR, Hanke ML, Fritz T, et al. (2011). Staphylococcus
nal bacteria diversity and it’s association with clinically
aureus biofilms prevent macrophage phagocytosis and diagnosed bacterial vaginosis by denaturing gradient gel
attenuate inflammation in vivo. J Immunol 186:6585–96. electrophoresis and correspondence analysis. Infect Genet
Turovskiy Y, Noll KS, Chikindas ML. (2011). The aetiology of Evol 44:479–86.
bacterial vaginosis. J Appl Microbiol 110:1105–28. Yeganegi M, Watson CS, Martins A, et al. (2009). Effect of
Trama JP, Pascal KE, Zimmerman J, et al. (2008). Rapid detec- Lactobacillus rhamnosus GR-1 supernatant and fetal sex on
tion of Atopobium vaginae and association with organisms lipopolysaccharide-induced cytokine and prostaglandin-
implicated in bacterial vaginosis. Mol Cell Probes regulating enzymes in human placental trophoblast cells:
22:96–102. implications for treatment of bacterial vaginosis and pre-
Udayalaxmi J, Bhat G, Kotigadde S, Kotian S. (2012). Effect of vention of p. Am J Obstet Gynecol 200:532.e1–8.
pH on the adherence, surface hydrophobicity and the bio- Yeoman CJ, Yildirim S, Thomas SM, et al. (2010). Comparative
film formation of Gardnerella vaginalis. J Clin Diagnostic genomics of Gardnerella vaginalis strains reveals substan-
Res 6:967–9. tial differences in metabolic and virulence potential. PLoS
Udayalaxmi J, Bhat GK, Kotigadde S. (2011). Biotypes and One 5:e12411.
virulence factors of Gardnerella vaginalis isolated from Yoshimura K, Morotomi N, Fukuda K, et al. (2011).
cases of bacterial vaginosis. Indian J Med Microbiol Intravaginal microbial flora by the 16S rRNA gene
29:165–8. sequencing. Am J Obstet Gynecol 205:235.e1–e9.
Vallor AC, Antonio MA, Hawes SE, Hillier SL. (2001). Factors Zhou X, Bent SJ, Schneider MG, et al. (2004). Characterization
associated with acquisition of, or persistent colonization of vaginal microbial communities in adult healthy women
by, vaginal lactobacilli: role of hydrogen peroxide produc- using cultivation-independent methods. Microbiology
tion. J Infect Dis 184:1431–6. (Reading, Engl) 150:2565–73.
Velraeds MMC, Van der Mei HC, Reid G, Busscher HJ. (1996). Zozaya-Hinchliffe M, Lillis R, Martin DH, Ferris MJ. (2010).
Inhibition of initial adhesion of uropathogenic Quantitative PCR assessments of bacterial species in
Enterococcus faecalis by biosurfactants from Lactobacillus women with and without bacterial vaginosis. J Clin
isolates. Appl Environ Microbiol 62:1958–63. Microbiol 48:1812–29.

Das könnte Ihnen auch gefallen