Sie sind auf Seite 1von 14

FEMS Yeast Research, 15, 2015, fov077

doi: 10.1093/femsyr/fov077
Advance Access Publication Date: 22 August 2015
Minireview

MINIREVIEW

Polymicrobial Candida biofilms: friends and foe

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


in the oral cavity
Lindsay E. O’Donnell, Emma Millhouse, Leighann Sherry, Ryan Kean,
Jennifer Malcolm, Christopher J. Nile and Gordon Ramage∗
Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical,
Veterinary and Life Sciences, University of Glasgow, Glasgow G2 3JZ, UK
∗ Corresponding author: Infection and Immunity Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life
Sciences, University of Glasgow, 378 Sauchiehall Street, Glasgow G2 3JZ, UK. Tel: +44(0)141 211 9752. E-mail: gordon.ramage@glasgow.ac.uk
One sentence summary: An examination of where and how Candida interacts with bacteria in the oral cavity
Editor: Carol Munro

ABSTRACT
The role of polymicrobial biofilm infections in medicine is becoming more apparent. Increasing number of microbiome
studies and deep sequencing has enabled us to develop a greater understanding of how positive and negative microbial
interactions influence disease outcomes. An environment where this is particularly pertinent is within the oral cavity, a
rich and diverse ecosystem inhabited by both bacteria and yeasts, which collectively occupy and coexist within various
niches as biofilm communities. Studies within this environment have however tended to be subject to extensive
independent investigation, in the context of either polymicrobial bacterial communities or yeast biofilms, but rarely both
together. It is clear however that they are not mutually exclusive. Therefore, this review aims to explore the influence of
candidal populations on the composition of these complex aggregates and biofilm communities, to investigate their
mechanistic interactions to understand how these impact clinical outcomes, and determine whether we can translate how
this knowledge can be used to improve patient management.

Keywords: Candida; biofilm; polymicrobial

INTRODUCTION negative) are in fact a highly prevalent and clinically important


entity (Fig. 1).
Candida biofilms, in particular Candida albicans, are an impor-
One location within the body where Candida species are
tant healthcare issue due to ineffective clinical management
readily isolated is within the oral cavity. Traditionally oral mi-
strategies. Over the past 20 years we have learned a great deal
crobiologists have invested significant time and effort unrav-
about their clinical importance, including the mechanisms used
elling the importance of specific bacterial–bacterial interac-
by members of the genus to form biofilms and resist the chal-
tions, while investigations of polymicrobial interactions have
lenge of host and antimicrobial molecules (Nett 2014; Ramage,
not received the same level of attention. This has led to a
Robertson and Williams 2014). However, as our levels of knowl-
disparity of fundamental knowledge on the significance of
edge have increased, in part through the development of more
candidal–bacterial interactions within the oral environment.
sophisticated technologies, there has been a growing awareness
The clinical implications of these polymicrobial biofilm inter-
that Candida rarely exist within a monospecies environment,
actions primarily relate to recalcitrance to antimicrobial treat-
and that heterogeneous biofilm populations consisting of ag-
ment strategies. Moreover, there is growing evidence from the
gregates of other fungi and bacteria (Gram positive and Gram

Received: 10 June 2015; Accepted: 14 August 2015



C FEMS 2015. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

1
2 FEMS Yeast Research, 2015, Vol. 15, No. 7

(A) (C)

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


(B)

Figure 1. Interactions between Candida albicans and bacteria. Candida albicans coaggregating with either (A) Gram-negative and (B) Gram-positive bacteria or (C) a
polymicrobial biofilm aggregate consisting of Gram-positive and Gram-negative oral bacterial species interacting with C. albicans hyphae. White scale bars = 20 μm.
Confocal Image taken by Dr Owain Millington, Biophotonics Unit, Strathclyde University).

literature that polymicrobial interactions may synergize the primarily within biofilm communities. Advances in genome se-
pathogenic potential of one or other microorganism (Stacy et al. quencing are only now beginning to shed light on the impor-
2014). This only serves to highlight the importance of a dual tance of Candida within these complex communities (Nobbs and
approach to microbial analysis, where mycological and bacteri- Jenkinson 2015). Microbiome analysis of the saliva from elderly
ological analysis can have an equal contribution through inter- Dutch patients showed that an increased candidal load was as-
disciplinary collaboration (Holmes, Cannon and Jenkinson 1995). sociated with a dysbiotic bacterial flora that favoured the co-
This review aims to critically evaluate the available evidence existence with oral streptococci to the exclusion of pathogenic
as a means of appraising the clinical importance of Candida anaerobic species (Kraneveld et al. 2012). Candida species have
biofilms in polymicrobial environments, using key oral diseases been isolated from a range of oral environments involving both
and groups of microorganisms to illustrate these points. soft and hard tissue of biological and non-biological origin, illus-
trating the adaptability of candidal yeasts (Fig. 2). The sites from
which C. albicans has been isolated include periodontal pockets,
Polymicrobial candidal interactions in the oral cavity
root canals, orthodontic appliances, enamel, dentures and mu-
Oral candidosis is one of the most well-defined fungal biofilm in- cosal surfaces (Ramage et al. 2004; de Carvalho et al. 2006; Ar-
fections of both soft and hard tissue and is characterized by com- slan et al. 2008; Dongari-Bagtzoglou et al. 2009; Sardi et al. 2010;
plex biofilms which interact with bacteria and the host (Dongari- Freitas et al. 2014). In order for candidal biofilms to flourish in
Bagtzoglou et al. 2009; Rautemaa and Ramage 2011). The oral these environments, moisture, nutrients, hyphal growth and the
cavity provides a key portal of entry within the human host, presence of commensal bacteria are all required which influence
and is home to a rich and diverse microbial flora. Despite being C. albicans architecture and virulence (Bertolini et al. 2015).
bathed in saliva, an important innate defence mechanism con-
taining numerous antimicrobial molecules, the oral cavity is a
Caries
favourable habitat for both prokaryotes and eukaryotes. Within
this, it is suggested that up to 108 microbes per millilitre of saliva Dental caries is one of the most common diseases worldwide,
are present (Guo and Shi 2013). The oral cavity, therefore, acts as impacting 2.43 billion (36% of the global population) (Vos et al.
an important incubator for a complex ‘microbial soup’, in which 2012). Largely influenced by diet caries has a multifactorial ae-
yeasts such as Candida interact with one another and with a tiology involving behavioural, environmental and immunologi-
plethora of cultivatable and non-cultivatable bacterial species, cal factors. Microbial dental plaque biofilms adherent to tooth
O’Donnell et al. 3

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


Figure 2. Oral sites of polymicrobial Candida biofilm diseases. The schematic diagram illustrates site within the oral cavity typically where Candida-bacterial polymi-
crobial biofilms are observed (clockwise from top position): caries, periodontitis, orthodontic, endodontic, angular cheilitis, denture stomatitis.

surfaces play a key role in the development of dental hanced exopolymeric matrix production, facilitated by the in-
caries, through carbohydrate metabolism (predominantly su- creased surface area associated with hyphal networks, supports
crose) leading to production of large quantities of lactic acid, mixed biofilm growth of dense communities cemented to tooth
and ultimately the dissolution of tooth surfaces. Typically, enamel. Based on this and other studies, the interaction be-
caries has been associated primarily with Streptococcus mu- tween candidal yeasts and streptococci is an important area re-
tans and Lactobacillus species (Loesche 1986; Badet and The- quiring further extensive investigation.
baud 2008), although more recently, oral microbiome studies
have highlighted the polymicrobial aetiology of carious lesions
Periodontal disease
(Belda-Ferre et al. 2012; Simon-Soro, Guillen-Navarro and Mira
2014). Historically, candidal yeasts have been isolated in patients Periodontal disease (PD) is a complex disease orchestrated by
with caries (Krasse 1954; Koo and Bowen 2014), though the ev- host–pathogen interactions. It affects almost 50% of the US
idence for their direct role has yet been shown directly. There population under 30 years old, and by the time they reach 65
is now growing evidence that C. albicans actively participates in years of age approximately 70% are affected (Eke et al. 2012).
cariogenic biofilms, through synergistic interaction with St. mu- In its mild and reversible form (gingivitis), the gingival tis-
tans (Metwalli et al. 2013; Koo and Bowen 2014). Evidence of en- sues are characterized by swelling, an inflamed gum line and
4 FEMS Yeast Research, 2015, Vol. 15, No. 7

bleeding, whereas in its severe and irreversible form (periodon- of which species are important. In fact, this is a pertinent point
titis) there is destruction of the supporting periodontal liga- to all oral diseases. Endodontic biofilms tend to reflect their ori-
ments and progressive bone resorption. While dysregulated in- gin, i.e. those from cariogenic lesion on occlusal surfaces may
flammatory responses are pivotal with respect to periodontitis, be more similar to supragingival plaque whereas those in peri-
the initial catalytic stimulus common to both forms of the dis- apical infection may reflect a predominantly anaerobic environ-
ease comes from complex microbial biofilms. These initially es- ment. There is increasing evidence for the involvement of Can-
tablish themselves above the gum line (supragingival plaque), dida species in endodontic infections (Siqueira and Sen 2004).
alter the microenvironment and drive a lower redox and pH, Its role as a dentophilic pathogen is highlighted through in vitro
thus enabling capnophiles and anaerobes to colonize and pro- studies of dentine, where penetration of dentine tubules with
duce subgingival plaque biofilms. The microbiology of supra- C. albicans was demonstrated (Sen, Safavi and Spangberg 1997).
and subgingival plaque is extremely well characterized, with the Subsequent studies have confirmed the presence of C. albicans
influence of defined groupings of commensal and pathogenic from clinical root canal specimens (Baumgartner, Watts and Xia
species accurately mapped to clinical outcomes (Ximenez-Fyvie 2000), with subsequent studies showing an association between

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


et al. 2000; Shi et al. 2015). With this historical focus on defined C. albicans and E. faecalis (Peciuliene et al. 2001). In spite of this
bacterial groupings defined by Socransky’s traffic light analogy evidence of polymicrobiality, there are no studies describing the
(Socransky et al. 1998), there has been minimal interest with re- candidal–bacterial interactions in the root canal environment.
spect to the influence of candidal species (Holmstrup 1999). This
is surprising given that Candida species have also been isolated
Denture stomatitis
from subgingival mixed biofilm consortia in patients with severe
chronic periodontitis, where quantitatively high levels of C. albi- Edentulousness is an irreversible clinical condition that can be
cans were shown to correlate with moderate and severe chronic described as an ultimate marker of oral disease burden and is
periodontitis (Canabarro et al. 2012). There is, however, a lack often associated with socioeconomic factors (Jeganathan and
of direct evidence for causality, although in diabetes patients Lin 1992; Cunha-Cruz, Hujoel and Nadanovsky 2007). Denture
the relationship between subgingival candidal colonization and stomatitis (DS) refers to inflammation of the oral mucosa and
periodontitis is more apparent (Sardi et al. 2012; Hammad, pathological changes associated with denture surfaces adjacent
Darwazeh and Idrees 2013). This relationship maybe a conse- to tissue (Jeganathan and Lin 1992). Approximately two thirds
quence of metabolic requirements, with elevated blood sugar of individuals who wear removable complete dentures suffer
levels supporting the growth of Candida species. Further evi- from DS, though most individuals are asymptomatic (Gendreau
dence for Candida’s involvement follows the use of oral contra- and Loewy 2011). With 15 million dentures wearers in the UK,
ceptives (OC), by which several studies have found an increased this is not an inconsequential disease (Coulthwaite and Verran
prevalence of Candida spp. carriage, as well as higher incidences 2007). Many factors influence its onset and severity, including
of oral and vaginal candidiasis amongst OC users (Spinillo et al. salivary flow and denture cleanliness amongst others (Oksala
1995; Kazi, Saleem and Kazi 2012; Zakout, Salih and Ahmed 1990; Soysa, Samaranayake and Ellepola 2004, 2006; Soysa and
2012). Furthermore, the prevalence of severe periodontitis is Ellepola 2005), although microbial factors remain one of the
higher amongst OC users, suggesting that the hormones lead to most important. Dentures support the growth of microbial
the development of a dysbiotic biofilm, enabling Candida yeast biofilms (denture plaque) within tiny cracks and fissures. These
to colonize (Brusca et al. 2010). Irrespective of why Candida spp. polymicrobial biofilm communities dominate the denture sur-
are present in this environment, we do know that the subgin- face, with up to 1011 microbes per milligram of denture plaque
gival environment represents a highly diverse microbial ecosys- (Nikawa, Hamada and Yamamoto 1998), which take advantage
tem comprised of a variety of commensals and pathogens, rang- of the varied topography associated with denture acrylics and
ing from benign streptococcal species to virulent Porphyromonas resins (Ramage et al. 2004). Some of the bacterial species iso-
gingivalis (Socransky and Haffajee 2005; Haffajee and Socransky lated include periodontal pathogens such as Fusobacterium nu-
2006). Here, competition for nutrients, gases and space dictates cleatum, Aggregatibacter actinomycetemcomitans and P. gingivalis
biofilm structure, and it is likely that the larger Candida cells play (Sachdeo, Haffajee and Socransky 2008; Yasui et al. 2012), al-
a significant physical and chemical role. though caries-associated species such as Streptococcus and Lacto-
bacillus species predominate (Teles et al. 2012), possibly through
their ability to coaggregate with C. albicans hyphae (Bilhan et al.
Endodontic infection
2009; Ribeiro et al. 2012). Here they form biofilms analogous to
Endodontitis can result from direct tooth trauma, carious le- that of the enamel surface through pioneer species, followed
sions on the enamel surface, or from periodontal infection pro- by coaggregation and maturation of complex polymicrobial
gressing to the root apex. It is characterized by an infection of biofilms (Fig. 3).
the pulp within the dental root canal system and is the major Unlike the oral diseases described above, DS is generally con-
aetiologic agent of apical periodontitis. The American Associ- sidered to be of yeast aetiology, with the literature dispropor-
ation of Endodontists estimate that over 15 million root canal tionately focused on Candida spp. (Coleman et al. 1997; Bagg et al.
treatments are performed annually in the USA, and these are 2003; Redding et al. 2004; Li, Redding and Dongari-Bagtzoglou
primarily of an infectious origin. Endodontic infections are typi- 2007). Candida albicans is the most frequently isolated yeast from
cally of biofilm aetiology and are associated with key oral bacte- the denture, but C. glabrata, C. dubliniensis, C. tropicalis, C. kru-
rial pathogens from up to 100 different bacterial genera (Siqueira sei and a range of other Candida species have been frequently
and Rocas 2009), by and large from four key phyla (Firmicutes, Bac- isolated (Coco et al. 2008; Williams et al. 2011). Candida albicans
teroidetes, Actinobacteria and Proteobacteria), although Enterococcus accounts for the majority of the inflammatory pathology ob-
faecalis is considered the primary aetiological agent. Nonethe- served clinically (Salerno et al. 2011). It exists as a commensal
less, due consideration should be made to the method employed in the oral cavity of 25–50% of the healthy population, and can
(culture versus non-culture) when assembling the snapshot of become pathogenic under optimal conditions, such as when the
the dominant microbiota, as this heavily biases our perception immune response is compromised (Dagistan et al. 2009). This is
O’Donnell et al. 5

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


Figure 3. Development of polymicrobial Candida biofilm on denture acrylic. This schematic representation of denture biofilm development shows how initial coloniza-
tion by yeast and bacterial species (white), followed by hyphal formation and coaggregation (grey), which then enables the bacterial species to expand and grow into
the spaces unoccupied on the surface of both the acrylic and C. albicans hyphae (black).

Figure 4. Micrographs of polymicrobial Candida denture-related biofilms. (A) Scanning electron micrograph (SEM) and (B) confocal laser scanning micrograph (CLSM) of
complex bacterial communities coaggregating with C. albicans upon denture acrylic. These micrographs show low (SEM) and high (CLSM) magnifications of mixtures
of C. albicans yeast (round) and hyphae (long filaments—white arrows) coaggregated with smaller bacterial species. Confocal Image taken by Dr Owain Millington,
Biophotonics Unit, Strathclyde University).

not surprising given its dimorphic capabilities, i.e. the ability to tiology that includes anatomical issues, dry mouth, immuno-
form hyphae and yeast interchangeably, a requisite of biofilm suppression and the wearing of poor fitting dentures, amongst
formation (Ramage et al. 2002b). The hyphal form has been more many others. Although not particularly common per se, this
commonly isolated in DS sufferers and is assumed to be the disease is of interest as it is often associated with the coisola-
more invasive form of the organism, with an enhanced ability tion of Candida species with Staphylococcus aureus, microorgan-
to adhere to and colonize the prosthesis surface (Gendreau and isms not unaccustomed to one another within the human host
Loewy 2011; Verran et al. 2014). Collectively, these polymicrobial (Tawara, Honma and Naito 1996; Adam, Baillie and Douglas 2002;
biofilms actively release proteolytic and lipolytic enzymes that Baena-Monroy et al. 2005). Both species are leading pathogens
induce inflammation of the palatal surface (Marcos-Arias et al. in blood borne and systemic infections, a major cause of mor-
2011; Ramage et al. 2012), ultimately leading to DS. The scanning bidity and mortality in hospitalized patients. These species are
electron micrograph (SEM) in Fig. 4 illustrates that C. albicans in- of significant interest because of the escalating development
teracts with bacteria on the surface of denture acrylic, with the of antimicrobial resistance and their increasing involvement in
associated confocal micrograph, showing bacteria coaggregating chronic and systemic polymicrobial biofilm infections (Perlroth,
with C. albicans hyphae. Choi and Spellberg 2007), and have been shown to coaggre-
gate together and exist within a dynamic and interactive state
(Shirtliff, Peters and Jabra-Rizk 2009; Peters et al. 2012a,b). The re-
Angular cheilitis
lationship between these two has been described as mutualistic,
Angular cheilitis is an inflammation of one, or more commonly synergistic and antagonistic, yet most of the evidence indicates
both, corners of the mouth. It is a disease of multifactorial ae- synergy, as the majority of their interactions are associated with
6 FEMS Yeast Research, 2015, Vol. 15, No. 7

enhanced pathogenicity and disease severity (Peters and Noverr


2013; Schlecht et al. 2015).

Oropharyngeal and respiratory infection


As described, Candida is one of the main colonizers of the oral
cavity and plays an important role in many oral diseases. How-
ever, there is thought to be a potential link between oral and
pulmonary colonization of Candida, which could contribute to-
ward respiratory infection. Studies have identified respiratory
pathogens colonizing the oral cavity, as well as oral pathogens
colonizing the lungs (El-Solh 2011; Bansal, Khatri and Taneja
2013; Vadiraj et al. 2013; Przybylowska et al. 2015). Amongst

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


these, Candida has been found to be one of the most predomi-
nant pathogens in the lungs, particularly in those suffering from
lung cancer and chronic pulmonary disease (Biswas et al. 2010;
Laroumagne et al. 2011, 2013). Aspiration of oral material into the
lungs is thought to be the primary entry route of oral pathogens.
Therefore, given that the oral carriage rate of Candida is approx-
imately 50% (Darwazeh, Hammad and Al-Jamaei 2010), and that
roughly 45% of healthy individuals aspirate oropharyngeal con-
tents into their lungs whilst sleeping, this puts a high num- Figure 5. Candida albicans and S. aureus polymicrobial biofilm. This confocal scan-
ber at risk of pulmonary colonization by Candida (Gleeson, Eggli ning laser micrograph shows the close interactions between clusters of S. aureus
and Maxwell 1997). Yet, despite the potential to cause infection, (yellow) and C. albicans hyphae (white/green). The appearance of these interac-
tions demonstrates close attachment between the two and three-dimensional
Candida colonization of the lungs is not necessarily detrimental,
space, suggesting structural stability and an element of cooperation with one an-
particularly when Pseudomonas aeruginosa is also isolated (Ader
other. Confocal Image taken by Dr Owain Millington, Biophotonics Unit, Strath-
et al. 2011). Pseudomonas aeruginosa is frequently related with clyde University).
ventilator-associated pneumonia and cystic fibrosis, with C. albi-
cans often coisolated. Many studies have investigated their inter-
actions, yet have produced conflicting results with some identi- (Millsap et al. 2001). The more intricate details involved in these
fying a synergistic relationship (Roux et al. 2009); however, the interactions require further investigation; however, what is clear
vast majority provide stronger evidence for an antagonistic re- is that disease resulting from microbial colonization of a VP is
lationship (Morales et al. 2010; Bandara et al. 2013a). Pseudomonas very much polymicrobial in nature.
aeruginosa gains the upper hand the majority of the time by pre-
venting biofilm formation via killing of C. albicans hyphal fil-
Mechanisms of polymicrobial biofilm interaction
aments (Hogan and Kolter 2002; Hogan, Vik and Kolter 2004).
Nonetheless, recently it has been shown in a murine model that Staphylococcal interactions
lung injury caused by Ps. aeruginosa infection is alleviated if pre- The interaction between C. albicans and S. aureus has been as-
ceded by a short-term C. albicans colonization (Ader et al. 2011). sociated with enhanced pathogenic behaviour, disease severity
This was due to C. albicans activation of innate lymphoid cells, and morbidity (Nair et al. 2014). They form mixed polymicrobial
which produced IL-22, providing protection against P. aeruginosa biofilms in which S. aureus cells are found attached to C. albicans
induced injury (Mear et al. 2014). hyphal filaments (Peters et al. 2010; Lin et al. 2013) (Fig. 5). Their
Candida polymicrobial biofilm formation is the predominant colocalization within biofilms is still unclear, as some describe
problem associated with voice box prosthesis (VP) (Talpaert et al. them interspersed throughout the biofilm three-dimensional
2015). Silicone is the most commonly used material used for structure (Peters et al. 2010), whereas others describe them as
VP; however, silicone is a favourable material for microbial at- only found attached within the upper layers of the biofilm
tachment and can very quickly become colonized (Busscher, (Harriott and Noverr 2009). This disparity could be explained by
Geertsema-Doornbusch and van der Mei 1997). Biofilm forma- different experimental conditions (e.g. growth medium). The ini-
tion can lead to valve malfunctioning, causing seepage of oe- tial colonizing species plays a key role in dictating their interac-
sophageal contents into the trachea, which could potentially tion, as it has been shown that C. albicans biofilm formation was
cause aspiration pneumonia (van Weissenbruch et al. 1997a,b). delayed when S. aureus colonized first, yet when added simul-
Candida albicans is the most common yeast associated with VP taneously biofilms formed rapidly (Lin et al. 2013). The reason
colonization, though C. glabrata and C. tropicalis are also fre- for this inhibition is unknown; perhaps S. aureus secretes an in-
quently isolated (Bauters et al. 2002). Streptococcus spp. and Lacto- hibitory molecule preventing Candida adhesion.
bacillus spp. are the predominant bacterial species isolated (Neu Studies in S. epidermidis have shown that extracellular DNA
et al. 1994); however, the majority of mature biofilms had Can- (eDNA) release through autolysis is an important entity in sup-
dida and lactobacilli as their primary components (Buijssen et al. porting mixed biofilm growth (Pammi et al. 2013), and is also a
2007). The success of polymicrobial biofilms forming on VP is critical feature for C. albicans biofilm extracellular matrix (ECM)
likely due to the location, which is difficult for host immune de- integrity (Rajendran et al. 2014; Sapaar et al. 2014). Therefore, it
fences to access. For the most part, it is very unusual to find a is not surprising that eDNA and the ECM from both C. albicans
biofilm from a VP that is not comprised of both fungal and bac- and S. aureus biofilms are both involved in affecting the action of
terial components. Before Candida can colonize the VP, there is antibacterial agents. In fact, it has been shown that S. aureus is
strong evidence that bacteria must be adhered first; thus, such protected against vancomycin treatment using concentrations
fungal–bacteria interactions are critical for biofilm formation as high as 1600 mg/mL within the mixed biofilm environment,
O’Donnell et al. 7

through C. albicans ECM preventing diffusion and access to S. au- duce phagocytosis by granulocytes (Fehrmann et al. 2013). In
reus (Harriott and Noverr 2009). There are, however, other adap- order to gain a better understanding of the molecular interac-
tive resistance mechanisms that play a role in this resistance tion between C. albicans and S. aureus, Peters et al. (2010) un-
phenotype (Harriott and Noverr 2010). dertook a proteomics approach to identify proteins upregulated
It has also been shown that S. aureus adhere to hyphal fila- during their interaction. The majority of the 27 proteins that
ments by relying on the adhesion to the C. albicans agglutanin- were upregulated were involved in processes, including stress
like sequence 3 protein (Als3p) (Peters et al. 2010, 2012), though and growth responses, and metabolism. Staphylococcus aureus
it is likely that other proteins are involved. Staphylococcus epi- upregulated stress-related genes in response to both yeast and
dermidis has also been shown to preferentially adhere to hy- hyphae, yet interestingly most of these genes were upregulated
phae, with forces between single bacterial and fungal germ in response to yeast rather than hyphal biofilms. As for C. albi-
tubes showing large adhesion forces (∼5 nN) (Beaussart et al. cans, yeast cells increased a number of stress-related proteins
2013). Studies have shown that S. aureus binding to C. albicans such as Tsa1p and aconitate hydratase, yet C. albicans in hyphal
hyphae was significantly stronger than all other bacteria tested, formation showed minimal changes in gene expression in re-

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


including P. aeruginosa (Peters et al. 2010). Interestingly, it was re- sponse to S. aureus. These results suggest that both organisms
ported that none of the members of the ALS family of adhesins induce a stress response on their initial encounter with one an-
(ALS1–7 and ALS9), including ALS3, are involved in interspecies other, particularly whilst Candida exists in yeast form. However,
adhesion (Harriott and Noverr 2010). Thus, further insight as they mature and develop into a hyphal biofilm, they may
is required before we can fully understand the mechanisms re- downregulate these genes as a survival strategy, facilitating sur-
sponsible for adherence, yet it is likely that this is a complex vival within the host.
process in which a multitude of proteins are involved. Never- Clearly, these two pathogens have the ability to influence one
theless, it is thought that adhesion to hyphae may assist S. au- another’s behaviour, so care must be taken in their clinical man-
reus in penetrating into the host (Schlecht et al. 2015), a man- agement. Broad-spectrum antimicrobial activity is crucial, ac-
ner analogous to injection from a needle-stick injury. This has counting for both prokaryote and eukaryote. The use of ethanol
been demonstrated in mice studies, in which mixed infections has been shown to be effective at preventing both mono-and
with C. albicans als3 strains together with S. aureus were unable polymicrobial biofilms (Peters et al. 2013). However, the success-
to invade the tongue, whereas the wild-type infections demon- ful use of miconazole in angular cheilitis is interesting given no
strated coinfection (Peters et al. 2012). The ramifications of this precise mechanism of action for this azole to S. aureus (Sud and
enhanced invasive capacity have been shown historically to im- Feingold 1982). It could therefore be hypothesized that given the
pact mortality, where synergism between the coinfected species polymicrobiality of the disease miconazole acts by exhibiting
administered intraperitoneally in a mouse model lead to 100% C. albicans activity, thereby destabilizing S. aureus colonization,
mortality, whereas monospecies infections caused no mortality which is physically supported by the hyphal biofilm meshwork.
whatsoever (Carlson 1983). Whether or not the relationship be- What is clear though is that these organisms are no strangers to
tween the two organisms is physical or chemical remains to be one another.
determined, although there is evidence that growth related syn-
ergy is an important factor in their cohabitation of microniches
Streptococcal interactions
(Carlson and Johnson 1985). Indeed, the physical relationship be-
tween the organisms is important, but not fundamental. Recent Streptococci are amongst the primary colonizers of the oral cav-
studies indicated that morphogenesis, i.e. the presence of hy- ity and compromise a large proportion of the overall flora (Syed
phae, is not critical for their pathogenic potential, as demon- and Loesche 1978; Moore et al. 1982). Oral streptococcal species
strated in some intricate murine studies using C. albicans genet- are often termed as the mitis group streptococci (MGS), which
ically locked into the yeast state (Nash et al. 2014). This suggests include St. mitis, St. oralis, St. gordonii, St. sanguinis and St. parasan-
that physical cellular interactions are not solely responsible. guinis species (Kawamura et al. 1995). MGS streptococci are tra-
Metabolic signalling between C. albicans and S. aureus may ditionally known to be early colonizers of dental surfaces, com-
play an important role in orchestrating this relationship. Chem- prising approximately 60–80% of the flora (Diaz et al. 2012), al-
ically mediated signalling in the form of quorum sensing (QS) though use of high-throughput gene sequencing technology has
could potentiate both positive and negative interactions be- revealed them to also be predominant colonizers of oral mucosal
tween these two microorganisms, which may inadvertently im- surfaces (Diaz et al. 2012).
pact clinical outcomes. Candida albicans secretion of farnesol, a The relationship between Candida and streptococci is gen-
QS molecule, decreases S. aureus biofilm formation, as well as erally considered to be synergistic, with advanced microscopy
increasing its susceptibility to antibiotics (Akiyama et al. 2002; showing streptococcal interactions with the hyphal filaments of
Jabra-Rizk et al. 2006; Unnanuntana et al. 2009). Moreover, it was Candida (Dutton et al. 2014). Streptococci provide Candida with
shown to competitively inhibit S. aureus lipase activity (Kuroda nutrients from the salivary pellicle, such as lactate and glu-
et al. 2007). However, Lin et al. (2013) found that S. aureus condi- cose, which Candida utilizes as a source of carbon (Holmes et al.
tioned media had a striking impact on C. albicans biofilm growth 2006). Furthermore, streptococci are aciduric and thus create an
rate, indicating that S. aureus secretes a reciprocal QS molecule acid environment through the fermentation of carbohydrates
that stimulates C. albicans growth (Lin et al. 2013).Nonetheless, (Takahashi and Nyvad 2011). At low pH, Candida grows in its
whether C. albicans secretes sufficient farnesol in vivo to have an yeast form. However, when cocolonized with streptococci, Can-
effect on S. aureus remains unknown. Yet despite these conflict- dida can grow and survive at a lower pH (<4.5), and the H2 O2 pro-
ing results, the majority of studies support the idea of a syner- duced by streptococci can induce hyphal growth by inducing ox-
gistic relationship between the two. idative stress (Jenkinson, Lala and Shepherd 1990; Nasution et al.
Indeed, affinity panning of a S. aureus phage display library 2008). This interaction is bidirectional, as C. albicans can promote
against C. albicans biofilms demonstrated that S. aureus released the survival of streptococci by lowering oxygen tension levels to
extracellular fibrinogen-binding protein (Efb) during the inter- that more acceptable for streptococcal growth, as well as provid-
action. This was shown to coat C. albicans yeast cells and re- ing nutrients to stimulate bacterial growth (Douglas 2002). This
8 FEMS Yeast Research, 2015, Vol. 15, No. 7

synergistic relationship can prove disparaging for the host. Stud- gordonii is able to suppress farnesol induced inhibition of biofilm
ies have shown that streptococci augment the persistence of formation, via autoinducer 2 (AI-2), as luxS mutants were less
Candida spp. Xu et al. (2014 ) demonstrated that coinfection with effective at permitting hyphal formation; however, the mode of
C. albicans and St. oralis resulted in a more pathogenic inflamma- action has yet to be elucidated (Bamford et al. 2009). Farnesol
tory response compared with infection with either microorgan- has also been shown to inhibit St. mutans biofilm accumulation
ism alone, as demonstrated through an exaggerated upregula- and polysaccharide production (Koo et al. 2003). Based on this
tion of TLR2 dependent inflammatory genes (Dutton et al. 2014). and further work, it has been suggested that it may be used
Adherence to mucosal surfaces occurs through binding inter- to control its competitiveness in mixed species biofilms and
actions with components of the salivary pellicle; however, there could be used as a means of a chemotherapeutic strategy (Jeon
is a limited number of niches for C. albicans to inhabit. Thus, et al. 2011). AI-2 is the primary QS molecule secreted by bac-
C. albicans has to compete with other microbes (Kolenbrander teria that allows interspecies communication (Vendeville et al.
et al. 2002). To overcome this problem, C. albicans has evolved 2005). The luxS gene is associated with AI-2 production and luxS
a mechanism allowing it to bind directly to MGS species, in- streptococcal mutants can form monospecies biofilms. How-

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


cluding St. oralis, St. mitis and St. gordonii (Jenkinson, Lala and ever, when cocolonized with C. albicans, biofilm formation be-
Shepherd 1990). This interaction is mutually beneficial as C. albi- comes abrogated, suggesting that this molecule is involved in
cans can support the outgrowth of streptococci by enabling them cellular communication (McNab et al. 2003; Bamford et al. 2009).
to form more robust oral biofilms (Xu et al. 2014). Adherence be- Another important signalling mechanism in streptococci, in-
tween these two species occurs via interactions of the C. albicans cluding St. gordonii, is through the comCDE operon, which en-
hyphal cell wall protein Als3, and the streptococcal cell surface codes a sensor-regulator system (ComDE) activated by the comC
adhesins SspA and SspB (Holmes, McNab and Jenkinson 1996), gene product competence stimulating peptide (CSP). Strepto-
proteins that belong to the antigen I/II polypeptide family (Bam- coccus gordonii–C. albicans biofilms formed with comCDE or
ford et al. 2009). Als3p is one of eight Als protein family members comC mutants showed increased biomass compared to wild-
expressed in C. albicans (Als1p-7p, Als9p). Direct binding of SspB type biofilms. Interestingly, more eDNA was observed in the
and Als3 is required for bacterial–fungal attachment. Interaction mixed comCDE mutant biofilms. Although purified CSP did not
between these molecules is associated with the N-terminal do- affect C. albicans hyphal formation. Contrary to earlier findings
main of Als3 (Bamford et al. 2015), as deletions at the N-terminus (Jarosz et al. 2009), it did inhibit monospecies biofilm formation,
abrogated binding to St. gordonii. Hoyer et al. (2014) have demon- suggesting that the St. gordonii comCDE QS system modulates
strated that this interaction may be more complex than origi- the production of eDNA (Jack et al. 2015), and important compo-
nally thought by showing that the peptide-binding domain (PBD) nent of candidal ECM (Rajendran et al. 2014).
of C. albicans is essential for C. albicans–St. gordonii adherence.
The PBD functions by binding to the free C-terminus; however,
Candidal interactions
in St. gordonii the SspB C-terminus is covalently linked to pep-
tidoglycan, and is thus unavailable to bind. Further investiga- Hyphae provide C. albicans with an advantage over many of its
tion is required before we can fully understand the mechanism competitors in terms of size and surface area, enabling them to
behind this interaction, though recent studies suggest that the take advantage of more sites for adhesion and occupation of a
early stage of cell wall O-mannosylation may be important in the variety of niches. This is why it is a more successful pathogen
development of these polymicrobial communities (Dutton et al. than other members of the genus. Nonetheless, there is hypoth-
2014). esis that Candida spp., in particular C. glabrata benefit from C. al-
An important component of a biofilm is the ECM, which con- bicans. There have been suggestions that DS pathology may be
fers protection to antimicrobials (Xu et al. 2014). The ECM of promoted by the synergistic interaction between these species
streptococcal biofilms is composed of α-glucans (Gregoire et al. within denture biofilms. Coco et al. (2008) first reported that C.
2011), whereas Candida biofilm ECM is primarily composed of β- glabrata and C. albicans were often coisolated from patients, par-
glucans (Al-Fattani and Douglas 2006; Taff et al. 2012). Streptococ- ticularly those with severe inflammation. The authors hypoth-
cus mutans utilizes its ECM components to enhance adhesion to esized that pathogenic synergy existed between the two Can-
fungal cells by depositing α-glucans on the surface of hyphae dida species. Candida glabrata, devoid of hyphae, forms relatively
(Gregoire et al. 2011). Moreover, interaction between St. mutans structurally poor and unstable biofilms, yet is associated with
and C. albicans is promoted by glucosyltransferase-derived ECM disease. Therefore, it was hypothesized to use C. albicans as a
and expression of the St. mutans virulence gene gtfB (Falsetta structural scaffold to gain entry into the host. Further studies
et al. 2014). It was also shown in this study that Candida-derived have confirmed this, where C. albicans appeared to assist the
β1,3-glucans contribute to ECM matrix structure, whilst fungal invasive capacity of C. glabrata within an in vitro reconstituted
β-glucan and mannan provide sites for GtfB binding and activ- epithelial biofilm model (Silva et al. 2011). The mechanistic of
ity. Furthermore, β-glucans are found on the surface of hyphae this interaction is at present unknown; however, we can specu-
as well as in the matrix (Dongari-Bagtzoglou et al. 2009), thus late that tissue destruction through proteolytic and lipolytic en-
suggesting that streptococci utilize these proteins to adhere to zymes augments the invasive capacity of the hyphae and allows
candidal hyphae. Collectively, this suggests that the biofilm ECM coaggregative C. glabrata to enter and contribute to pathogen-
contributes to this mutualistic behaviour, favouring their coex- esis. Further work by this group has shown similar data with
istence in the oral environment to the detriment of the host. work in a reconstituted human vaginal epithelial model, where
As with Candida–S. aureus interactions, QS is an important C. glabrata individually caused minimal tissue damage, though
factor in the relationship between Candida and streptococci. there was a significant increase in C. glabrata colonization and
Farnesol, a tetraprenoid alcohol and a key intermediate in invasiveness in combination with C. albicans (Alves et al. 2014).
the sterol biosynthetic pathway in eukaryotic cells, represents Damage was primarily dependent on the process of invasion,
the primary QS molecule associated with C. albicans, its main with key virulence genes upregulated (HWP1, PLD1 and ALS3).
role being repression of hyphal growth and biofilm formation Further studies using in vivo models to investigate the patho-
(Ramage et al. 2002a). However, one study has suggested that St. genesis of DS would be useful in this context (Nett et al. 2010),
O’Donnell et al. 9

although as described above there is mounting evidence that broad-spectrum antibiotic therapy or pre-existing medical con-
hitchhiking through adhesion to hyphae is not a limited phe- ditions, including diabetes (Rams, Babalola and Slots 1990; Sardi
nomenon and may also be important with respect to C. glabrata et al. 2010; Al Mubarak et al. 2013).
using C. albicans to gain entry to the host (Schlecht et al. 2015).
Facultative Gram-positive interactions
Anaerobic Gram-negative interactions
Candida species and E. faecalis have become increasingly noted
Life in subgingival plaque is highly anaerobic, favouring many for their coisolation within endodontic infections, both of which
obligate PD pathogens such as P. gingivalis, F. nucleatum and play an important role in nosocomial infection. Interestingly,
Prevotella intermedia. However, given the undefined relationship data from a longitudinal study carried out over two years at a
between Candida spp. and PD, then this remains a relatively German teaching hospital found that Candida-positive patients
neglected area of research. Studies regarding C. albicans and P. (blood, CSF, skin, feaces or sputum) were twice as likely to be co-
gingivalis have produced conflicting results It was shown that colonized by E. faecalis (Hermann et al. 1999). Enterococcus faecalis

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


P. gingivalis suppressed Candida biofilm formation through a re- has been found to incorporate itself into Candida biofilms, and is
duction in the number of viable yeast cells coincidental with an the third most predominant bacterial species found in mucosal
increasing P. gingivalis concentration (Thein, Samaranayake and fungal biofilms (Dongari-Bagtzoglou et al. 2009; Fox et al. 2014). It
Samaranayake 2006). Conversely, it was also shown that P. gin- was shown to adhere to Candida in both hyphal and yeast forms,
givalis induces germ-tube formation in C. albicans, producing a yet caused a reduction in the overall biofilm biomass (Fox et al.
more invasive phenotype, and thus increasing the risk of infec- 2014). However, Cruz et al. (2013) demonstrated that E. faecalis
tion (Nair, Anil and Samaranayake 2001). Furthermore, both mi- inhibited hyphal morphogenesis, which was partially depen-
crobes appear to have an antagonistic effect on one another in dent on the Fsr QS system, a major regulator of E. faecalis viru-
relation to host cell adhesion, as P. gingivalis inhibited adhesion lence. Collectively, these effects both impacted virulence during
of C. albicans to buccal epithelial cells (Nair and Samaranayake coinfection when compared to monospecies infection, suggest-
1996), whilst the presence of C. albicans did not enhance adhe- ing that they both negatively influence one another’s virulence
sion to gingival epithelial cells or gingival fibroblasts by P. gingi- and help maintain a commensal relationship (Garsin and Lorenz
valis (Tamai, Sugamata and Kiyoura 2011). Yet, in the same study 2013). Further work has revealed that C. albicans releases a sur-
pre-exposure of gingival epithelial cells and fibroblasts to C. albi- face protein Msb2, which binds to host antimicrobial peptides
cans enhanced cell invasion by P. gingivalis. Clearly, further stud- as well as antibiotics, thus conferring protection to both organ-
ies are required to decipher how these microorganisms interact isms (Swidergall, Ernst and Ernst 2013). Furthermore, evaluating
with one another. the influence of C. albicans on the dynamics of the bacterial mi-
As for F. nucleatum, coaggregation studies have revealed its crobiome following antibiotic treatment found that bacterial re-
ability to adhere to C. albicans species (Grimaudo and Nesbitt colonization was enhanced in the presence of C. albicans (Mason
1997), as well as C. dubliniensis (Jabra-Rizk et al. 1999). However, et al. 2012). Moreover, C. albicans reduced Lactobacillus spp. whilst
the interaction with C. albicans may be temperature dependent enhancing E. faecalis numbers, which led to the persistence of E.
as C. albicans grown at 37◦ C did not coaggregate with F. nuclea- faecalis long term. This effect was not apparent in subjects when
tum, yet the two species did coaggregate when grown at 25 and C. albicans was absent. Whether this effect was due to a syner-
45◦ C (Jabra-Rizk et al. 1999). The exact mechanistic behind these gistic relationship with E. faecalis or an antagonistic interaction
interactions remains unknown; however, these observations in- with lactobacilli remains to be elucidated.
dicate that C. albicans–F. nucleatum interactions may be an impor- There is a conceived dogma that lactobacilli antagonize can-
tant factor in oral colonization by yeasts. didal colonization (Young, Krasner and Yudkofsky 1956). This
Studies using lipopolysaccharide from a variety of Gram- forms the basis of why they play a key role in probiotics. It is
negative strains have shown that hyphal formation is inhib- well documented that probiotics reduces candidal levels at sev-
ited as is biofilm formation in a number of Candida spp. eral sites, including oral cavity, bloodstream and urinary tract
(Bandara et al. 2010), indicating that physical interaction may (Mendonca et al. 2012; Kumar et al. 2013). Early observations in-
be an important factor in defining their subgingival niches. dicate that C. albicans decreased in the presence of lactobacilli
Subsequent work in Escherichia coli demonstrated that secreted through provision of nutrients for lactobacilli that leads to lac-
elements also play an important role in affecting hyphal tic acid production, thus hindering candidal growth through
formation (Bandara et al. 2013b). This is also true of the relation- pH-dependent inhibition. This dynamic relationship suggests
ship between the capnophilic bacterium A. actinomycetemcomi- that there is a close association between the two, but to date
tans where it has been shown that its release of AI-2 was actively this has mainly been observed in vaginal infection. Our own
involved in inhibiting C. albicans hyphal growth and biofilm for- microbiome studies of the denture plaque have shown that
mation (Bachtiar et al. 2014). Given the complexity of various C. albicans and lactobacilli are positively associated in disease
metabolites and QS molecules in subgingival plaque, such as (unpublished work). The role of lactobacilli in maintaining
volatile sulphur compounds, fatty acids and AI-2 (Kurita-Ochiai, homeostasis at the vaginal mucosa initially came to light due
Fukushima and Ochiai 1995, Huang et al. 2011; Jang et al. 2013; to the occurrence of vaginal candidiasis during treatment with
Basic and Dahlen 2014), it is likely that these also impact hyphal systemic antibiotics. The mechanisms by which Lactobacillus
formation and Candida’s ability to contribute to PD (Noverr and species inhibits growth and virulence of Candida spp. are not
Huffnagle 2004). This anoxic environment has been shown to re- yet fully understood, but perhaps the production of hydrogen
sult in significant transcriptional changes in C. albicans, includ- peroxide as it has been shown to cause anti-candidal activity,
ing the upregulation of WOR1, which is a transcriptional regula- albeit in some strains of lactobacilli (Strus et al. 2005). This sug-
tor central to phenotypic switching (Fox et al. 2014). Based on the gests that other interactive mechanisms are involved in disease,
available literature it could be surmised that subgingival plaque including the modulation of the host response whereby lacto-
is most likely to antagonize yeast proliferation, except in cases bacilli cells have been shown to upregulate inflammatory cy-
where there is dysbiosis of the biofilm ecology, such as following tokines when cocultured with C. albicans (Martinez et al. 2009),
10 FEMS Yeast Research, 2015, Vol. 15, No. 7

potentially assisting in the clearance of candidal infection. De- Arslan SG, Akpolat N, Kama JD, et al. One-year follow-up of the
spite the overwhelming evidence of an antagonistic interaction, effect of fixed orthodontic treatment on colonization by oral
certain species of oral Lactobacillus, namely Lactobacillus casei, Candida. J Oral Pathol Med 2008;37:26–9.
have demonstrated a stimulatory effect on C. albicans hyphal Bachtiar EW, Bachtiar BM, Jarosz LM, et al. AI-2 of Aggregatibacter
growth (Orsi et al. 2014), and in fact it has been demonstrated actinomycetemcomitans inhibits Candida albicans biofilm for-
that candidal hyphae have the capacity to coaggregate and sup- mation. Front Cell Infect Microbiol 2014;4:94.
port lactobacilli levels in patients with higher levels of oral dis- Badet C, Thebaud NB. Ecology of lactobacilli in the oral cavity: a
ease (Bilhan et al. 2009). Nevertheless, further studies are re- review of literature. Open Microbiol J 2008;2:38–48.
quired to investigate these interactions in detail to determine Baena-Monroy T, Moreno-Maldonado V, Franco-Martinez F, et al.
the true extent of the dynamic relationship; particularly as the Candida albicans, Staphylococcus aureus and Streptococcus mu-
conceived antagonism may only exist for C. albicans. For exam- tans colonization in patients wearing dental prosthesis. Med
ple, recent studies have shown that only one of six probiotic Lac- Oral Patol Oral 2005;10(Suppl 1):E27–39.
tobacillus species had an inhibitory effect on C. glabrata growth Bagg J, Sweeney MP, Lewis MA, et al. High prevalence of non-

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


(Jiang et al. 2015). This suggests that the interaction between Can- albicans yeasts and detection of anti-fungal resistance in
dida and lactobacilli may be dependent on the particular envi- the oral flora of patients with advanced cancer. Palliative Med
ronment they cohabit. 2003;17:477–81.
Bamford CV, d’Mello A, Nobbs AH, et al. Streptococcus gordonii
CONCLUSIONS modulates Candida albicans biofilm formation through inter-
generic communication. Infect Immun 2009;77:3696–704.
Collectively, these data demonstrate that the interaction be- Bamford CV, Nobbs AH, Barbour ME, et al. Functional regions of
tween candidal species and other microoorganisms may be de- Candida albicans hyphal cell wall protein Als3 that determine
pendent on the nature of the interaction (chemical, physical or interaction with the oral bacterium Streptococcus gordonii. Mi-
both) and the particular environment they cohabit. It is clear crobiology 2015;161:18–29.
from many of these studies that the interaction between C. albi- Bandara HM, Cheung BPK, Watt RM, et al. Pseudomonas aeruginosa
cans hyphae and different bacterial species is important in defin- lipopolysaccharide inhibits Candida albicans hyphae forma-
ing their interaction, whether mutualistic or antagonistic in na- tion and alters gene expression during biofilm development.
ture. The secretion of signalling molecules from the myriad of Mol Oral Microbiol 2013a;28:54–69.
microorganisms in the oral cavity such as AI-2, farnesol and Bandara HM, Cheung BP, Watt RM, et al. Secretory products
other small molecules is clearly important, with recent stud- of Escherichia coli biofilm modulate Candida biofilm forma-
ies supporting the notion that the metabolome plays an inte- tion and hyphal development. J Investig Clin Dent 2013b;4:
gral part in defining the interaction between the host, Candida 186–99.
and microbiota such as lactobacilli (Romani et al. 2015). Under- Bandara HM, Lam OL, Watt RM, et al. Bacterial lipopolysaccha-
standing how each of these specific interactions influences one rides variably modulate in vitro biofilm formation of Candida
another and Candida’s pathogenicity will enable us to target this species. J Med Microbiol 2010;59:1225–34.
medically important yeast rationally. Though, we must be cog- Bansal M, Khatri M, Taneja V. Potential role of periodontal in-
nizant of the negative influences of changing its role within fection in respiratory diseases - a review. J Med Life 2013;6:
complex oral biofilm communities and the consequences of dys- 244–8.
biosis (McLean, 2014), as this may support the unnecessary pro- Basic A, Dahlen G. Hydrogen sulfide production from subgingival
liferation and overgrowth of candidal yeasts that leads to oral plaque samples. Anaerobe 2014;35:21–7.
disease. Baumgartner JC, Watts CM, Xia T. Occurrence of Candida albicans
Conflict of interest. None declared. in infections of endodontic origin. J Endod 2000;26:695–8.
Bauters TG, Moerman M, Vermeersch H, et al. Colonization
of voice prostheses by albicans and non-albicans Candida
REFERENCES
species. Laryngoscope 2002;112:708–12.
Adam B, Baillie GS, Douglas LJ. Mixed species biofilms of Can- Beaussart A, Herman P, El-Kirat-Chatel S, et al. Single-cell
dida albicans and Staphylococcus epidermidis. J Med Microbiol force spectroscopy of the medically important Staphy-
2002;51:344–9. lococcus epidermidis-Candida albicans interaction. Nanoscale
Ader F, Jawhara S, Nseir S, et al. Short term Candida albicans 2013;5:10894–900.
colonization reduces Pseudomonas aeruginosa-related lung Belda-Ferre P, Alcaraz LD, Cabrera-Rubio R, et al. The oral
injury and bacterial burden in a murine model. Crit Care metagenome in health and disease. ISME J 2012;6:46–56.
2011;15:R150. Bertolini MM, Xu H, Sobue T, et al. Candida-streptococcal mu-
Akiyama H, Oono T, Huh WK, et al. Actions of farnesol and cosal biofilms display distinct structural and virulence char-
xylitol against Staphylococcus aureus. Chemotherapy 2002;48: acteristics depending on growth conditions and hyphal mor-
122–8. photypes. Mol Oral Microbiol. 2015;30:307–22.
Al-Fattani MA, Douglas LJ. Biofilm matrix of Candida albicans and Bilhan H, Sulun T, Erkose G, et al. The role of Candida albicans
Candida tropicalis: chemical composition and role in drug re- hyphae and Lactobacillus in denture-related stomatitis. Clin
sistance. J Med Microbiol 2006;55:999–1008. Oral Investig 2009;13:363–8.
AlMubarak S, Robert AA, Baskaradoss JK, et al. The preva- Biswas D, Agarwal S, Sindhwani G, et al. Fungal colonization in
lence of oral Candida infections in periodontitis patients patients with chronic respiratory diseases from Himalayan
with type 2 diabetes mellitus. J Infect Public Health 2013;6: region of India. Ann Clin Microbiol Antimicrob 2010;9:28.
296–301. Brusca MI, Rosa A, Albaina O, et al. The impact of oral contra-
Alves CT, Wei XQ, Silva S, et al. Candida albicans promotes inva- ceptives on women’s periodontal health and the subgingival
sion and colonisation of Candida glabrata in a reconstituted occurrence of aggressive periodontopathogens and Candida
human vaginal epithelium. J Infect 2014;69:396–407. species. J Periodontol 2010;81:1010–8.
O’Donnell et al. 11

Buijssen KJ, Harmsen HJ, van der Mei HC, et al. Lactobacilli: im- Fehrmann C, Jurk K, Bertling A, et al. Role for the fibrinogen-
portant in biofilm formation on voice prostheses. Otolaryng binding proteins coagulase and Efb in the Staphylococcus
Head Neck 2007;137:505–7. aureus-Candida interaction. Int J Med Microbiol 2013;303:230–
Busscher HJ, Geertsema-Doornbusch GI, van der Mei HC. Adhe- 8.
sion to silicone rubber of yeasts and bacteria isolated from Fox EP, Cowley ES, Nobile CJ, et al. Anaerobic bacteria grow within
voice prostheses: influence of salivary conditioning films. J Candida albicans biofilms and induce biofilm formation in
Biomed Mater Res 1997;34:201–9. suspension cultures. Curr Biol 2014;24:2411–6.
Canabarro A, Valle C, Farias MR, et al. Association of subgingival Freitas AO, Marquezan M, Nojima Mda C, et al. The influence of
colonization of Candida albicans and other yeasts with sever- orthodontic fixed appliances on the oral microbiota: a sys-
ity of chronic periodontitis. J Periodontal Res 2012;48:428–32. tematic review. Dental Press J Orthod 2014;19:46–55.
Carlson E. Effect of strain of Staphylococcus aureus on synergism Garsin DA, Lorenz MC. Candida albicans and Enterococcus fae-
with Candida albicans resulting in mouse mortality and mor- calis in the gut: synergy in commensalism? Gut Microbes
bidity. Infect Immun 1983;42:285–92. 2013;4:409–15.

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


Carlson E, Johnson G. Protection by Candida albicans of Staphylo- Gendreau L, Loewy ZG. Epidemiology and etiology of denture
coccus aureus in the establishment of dual infection in mice. stomatitis. J Prosthodont 2011;20:251–60.
Infect Immun 1985;50:655–9. Gleeson K, Eggli DF, Maxwell SL. Quantitative aspira-
Coco BJ, Bagg J, Cross LJ, et al. Mixed Candida albicans and Can- tion during sleep in normal subjects. Chest 1997;111:
dida glabrata populations associated with the pathogenesis 1266–72.
of denture stomatitis. Oral Microbiol Immunol 2008;23:377–83. Gregoire S, Xiao J, Silva BB, et al. Role of glucosyltransferase B in
Coleman D, Sullivan D, Harrington B, et al. Molecular and phe- interactions of Candida albicans with Streptococcus mutans and
notypic analysis of Candida dubliniensis: a recently identified with an experimental pellicle on hydroxyapatite surfaces.
species linked with oral candidosis in HIV-infected and AIDS Appl Environ Microb 2011;77:6357–67.
patients. Oral Dis 1997;3(Suppl 1):S96–101. Grimaudo NJ, Nesbitt WE. Coaggregation of Candida albicans
Coulthwaite L, Verran J. Potential pathogenic aspects of denture with oral Fusobacterium species. Oral Microbiol Immunol
plaque. Brit J Biomed Sci 2007;64:180–9. 1997;12:168–73.
Cruz MR, Graham CE, Gagliano BC, et al. Enterococcus faecalis in- Guo L, Shi W. Salivary biomarkers for caries risk assessment. J
hibits hyphal morphogenesis and virulence of Candida albi- Calif Dent Assoc 2013;41:107–9, 112–8.
cans. Infect Immun 2013;81:189–200. Haffajee AD, Socransky SS. Introduction to microbial aspects
Cunha-Cruz J, Hujoel PP, Nadanovsky P. Secular trends in socio- of periodontal biofilm communities, development and treat-
economic disparities in edentulism: USA, 1972–2001. J Dent ment. Periodontology 2000 2006;42:7–12.
Res 2007;86:131–6. Hammad MM, Darwazeh AM, Idrees MM. The effect of glycemic
Dagistan S, Aktas AE, Caglayan F, et al. Differential diagnosis of control on Candida colonization of the tongue and the sub-
denture-induced stomatitis, Candida, and their variations in gingival plaque in patients with type II diabetes and peri-
patients using complete denture: a clinical and mycological odontitis. Oral Surg Oral Med O 2013;116:321–6.
study. Mycoses 2009;52:266–71. Harriott MM, Noverr MC. Candida albicans and Staphylococcus au-
Darwazeh AM, Hammad MM, Al-Jamaei AA. The relation- reus form polymicrobial biofilms: effects on antimicrobial re-
ship between oral hygiene and oral colonization with Can- sistance. Antimicrob Agents Ch 2009;53:3914–22.
dida species in healthy adult subjects. Int J Dent Hygiene Harriott MM, Noverr MC. Ability of Candida albicans mutants
2010;8:128–33. to induce Staphylococcus aureus vancomycin resistance dur-
de Carvalho FG, Silva DS, Hebling J, et al. Presence of mu- ing polymicrobial biofilm formation. Antimicrob Agents Ch
tans streptococci and Candida spp. in dental plaque/dentine 2010;54:3746–55.
of carious teeth and early childhood caries. Arch Oral Biol Hermann C, Hermann J, Munzel U, et al. Bacterial flora ac-
2006;51:1024–8. companying Candida yeasts in clinical specimens. Mycoses
Diaz PI, Dupuy AK, Abusleme L, et al. Using high throughput se- 1999;42:619–27.
quencing to explore the biodiversity in oral bacterial com- Hogan DA, Kolter R. Pseudomonas-Candida interactions: an
munities. Mol Oral Microbiol 2012;27:182–201. ecological role for virulence factors. Science 2002;296:
Dongari-Bagtzoglou A, Kashleva H, Dwivedi P, et al. Charac- 2229–32.
terization of mucosal Candida albicans biofilms. PLoS One Hogan DA, Vik A, Kolter R. A Pseudomonas aeruginosa quorum-
2009;4:e7967. sensing molecule influences Candida albicans morphology.
Douglas HFJaLJ. Chapter 18: Interactions between Candida Mol Microbiol 2004;54:1212–23.
species and bacteria in mixed infections. In: Brogden K, Holmes AR, Cannon RD, Jenkinson HF. Interactions of Candida
Guthmiller J (eds). Polymicrobial Diseases. Washington, DC: albicans with bacteria and salivary molecules in oral biofilms.
ASM Press, 2002, 357–74. J Ind Microbiol 1995;15:208–13.
Dutton LC, Nobbs AH, Jepson K, et al. O-mannosylation in Can- Holmes AR, McNab R, Jenkinson HF. Candida albicans binding
dida albicans enables development of interkingdom biofilm to the oral bacterium Streptococcus gordonii involves multiple
communities. mBio 2014;5:e00911. adhesin-receptor interactions. Infect Immun 1996;64:4680–5.
Eke PI, Dye BA, Wei L, et al. Prevalence of periodontitis in adults Holmes AR, van der Wielen P, Cannon RD, et al. Candida albicans
in the United States: 2009 and 2010. J Dent Res 2012;91:914–20. binds to saliva proteins selectively adsorbed to silicone. Oral
El-Solh AA. Association between pneumonia and oral care in Surg Oral Med O 2006;102:488–94.
nursing home residents. Lung 2011;189:173–80. Holmstrup P. Non-plaque-induced gingival lesions. Ann Peri-
Falsetta ML, Klein MI, Colonne PM, et al. Symbiotic relation- odontol 1999;4:20–31.
ship between Streptococcus mutans and Candida albicans syn- Hoyer LL, Oh SH, Jones R, et al. A proposed mechanism for the
ergizes virulence of plaque biofilms in vivo. Infect Immun interaction between the Candida albicans Als3 adhesin and
2014;82:1968–81. streptococcal cell wall proteins. Front Microbiol 2014;5:564.
12 FEMS Yeast Research, 2015, Vol. 15, No. 7

Huang CB, Alimova Y, Myers TM, et al. Short- and medium-chain Laroumagne S, Lepage B, Hermant C, et al. Bronchial colonisation
fatty acids exhibit antimicrobial activity for oral microorgan- in patients with lung cancer: a prospective study. Eur Respir
isms. Arch Oral Biol 2011;56:650–4. J 2013;42:220–9.
Jabra-Rizk MA, Falkler WA, Jr, Merz WG, et al. Coaggregation of Laroumagne S, Salinas-Pineda A, Hermant C, et al. Incidence
Candida dubliniensis with Fusobacterium nucleatum. J Clin Micro- and characteristics of bronchial colonisation in patient with
biol 1999;37:1464–8. lung cancer: a retrospective study of 388 cases. Rev Mal Respir
Jabra-Rizk MA, Meiller TF, James CE, et al. Effect of farnesol 2011;28:328–35.
on Staphylococcus aureus biofilm formation and antimicrobial Li L, Redding S, Dongari-Bagtzoglou A. Candida glabrata:
susceptibility. Antimicrob Agents Ch 2006;50:1463–9. an emerging oral opportunistic pathogen. J Dent Res
Jack AA, Daniels DE, Jepson MA, et al. Streptococcus gordonii com- 2007;86:204–15.
CDE (competence) operon modulates biofilm formation with Lin YJ, Alsad L, Vogel F, et al. Interactions between Candida albi-
Candida albicans. Microbiology 2015;161:411–21. cans and Staphylococcus aureus within mixed species biofilms.
Jang YJ, Choi YJ, Lee SH, et al. Autoinducer 2 of Fusobacterium nu- BIOS 2013;84:30–9.

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


cleatum as a target molecule to inhibit biofilm formation of Loesche WJ. Role of Streptococcus mutans in human dental decay.
periodontopathogens. Arch Oral Biol 2013;58:17–27. Microbiol Rev 1986;50:353–80.
Jarosz LM, Deng DM, van der Mei HC, et al. Streptococcus mu- McLean RJ. Normal bacterial flora may inhibit Candida albicans
tans competence-stimulating peptide inhibits Candida albi- biofilm formation by Autoinducer-2. Front Cell Infect Microbiol
cans hypha formation. Eukaryot Cell 2009;8:1658–64. 2014;4:117.
Jeganathan S, Lin CC. Denture stomatitis-a review of the aetiol- McNab R, Ford SK, El-Sabaeny A, et al. LuxS-based signaling in
ogy, diagnosis and management. Aust Dent J 1992;37:107–14. Streptococcus gordonii: autoinducer 2 controls carbohydrate
Jenkinson HF, Lala HC, Shepherd MG. Coaggregation of Strepto- metabolism and biofilm formation with Porphyromonas gin-
coccus sanguis and other streptococci with Candida albicans. givalis. J Bacteriol 2003;185:274–84.
Infect Immun 1990;58:1429–36. Marcos-Arias C, Eraso E, Madariaga L, et al. Phospholipase and
Jeon JG, Pandit S, Xiao J, et al. Influences of trans-trans far- proteinase activities of Candida isolates from denture wear-
nesol, a membrane-targeting sesquiterpenoid, on Streptococ- ers. Mycoses 2011;54:e10–6.
cus mutans physiology and survival within mixed-species Martinez RC, Seney SL, Summers KL, et al. Effect of Lactobacillus
oral biofilms. Int J Oral Sci 2011;3:98–106. rhamnosus GR-1 and Lactobacillus reuteri RC-14 on the ability
Jiang Q, Stamatova I, Kari K, et al. Inhibitory activity in vitro of of Candida albicans to infect cells and induce inflammation.
probiotic lactobacilli against oral Candida under different fer- Microbiol Immunol 2009;53:487–95.
mentation conditions. Benef Microbes 2015;6:361–8. Mason KL, Erb Downward JR, Mason KD, et al. Candida albicans
Kawamura Y, Hou XG, Sultana F, et al. Determination of 16S and bacterial microbiota interactions in the cecum during
rRNA sequences of Streptococcus mitis and Streptococcus gor- recolonization following broad-spectrum antibiotic therapy.
donii and phylogenetic relationships among members of the Infect Immun 2012;80:3371–80.
genus Streptococcus. Int J Syst Bacteriol 1995;45:406–8. Mear JB, Gosset P, Kipnis E, et al. Candida albicans airway ex-
Kazi YF, Saleem S, Kazi N. Investigation of vaginal microbiota posure primes the lung innate immune response against
in sexually active women using hormonal contraceptives in Pseudomonas aeruginosa infection through innate lymphoid
Pakistan. BMC Urol 2012;12:22. cell recruitment and interleukin-22-associated mucosal re-
Kolenbrander PE, Andersen RN, Blehert DS, et al. Communi- sponse. Infect Immun 2014;82:306–15.
cation among oral bacteria. Microbiol Mol Biol R 2002;66: Mendonca FH, Santos SS, Faria Ida S, et al. Effects of probiotic
486–505. bacteria on Candida presence and IgA anti-Candida in the oral
Koo H, Bowen WH. Candida albicans and Streptococcus mutans: a cavity of elderly. Braz Dent J 2012;23:534–8.
potential synergistic alliance to cause virulent tooth decay Metwalli KH, Khan SA, Krom BP, et al. Streptococcus mutans, Can-
in children. Future Microbiol 2014;9:1295–7. dida albicans, and the human mouth: a sticky situation. PLoS
Koo H, Hayacibara MF, Schobel BD, et al. Inhibition of Strepto- Pathog 2013;9:e1003616.
coccus mutans biofilm accumulation and polysaccharide pro- Millsap KW, Bos R, van der Mei HC, et al. Adhesive interac-
duction by apigenin and tt-farnesol. J Antimicrob Chemoth tions between voice prosthetic yeast and bacteria on silicone
2003;52:782–9. rubber in the absence and presence of saliva. Anton Leeuw
Kraneveld EA, Buijs MJ, Bonder MJ, et al. The relation between 2001;79:337–43.
oral Candida load and bacterial microbiome profiles in Dutch Moore WEC, Holdeman LV, Smibert RM, et al. Bacteriology of ex-
older adults. PLoS One 2012;7:e42770. perimental gingivitis in young-adult humans. Infect Immun
Krasse B. The relationship between lactobacilli, Candida and 1982;38:651–67.
streptococci and dental caries; examination of saliva and Morales DK, Jacobs NJ, Rajamani S, et al. Antifungal mechanisms
plaque material collected on the same occasion. Odontol Revy by which a novel Pseudomonas aeruginosa phenazine toxin
1954;5:241–61. kills Candida albicans in biofilms. Mol Microbiol 2010;78:1379–
Kumar S, Singhi S, Chakrabarti A, et al. Probiotic use and preva- 92.
lence of candidemia and candiduria in a PICU. Pediatr Crit Nair N, Biswas R, Gotz F, et al. Impact of Staphylococcus aureus
Care Me 2013;14:e409–15. on pathogenesis in polymicrobial infections. Infect Immun
Kurita-Ochiai T, Fukushima K, Ochiai K. Volatile fatty acids, 2014;82:2162–9.
metabolic by-products of periodontopathic bacteria, inhibit Nair RG, Anil S, Samaranayake LP. The effect of oral bacteria on
lymphocyte proliferation and cytokine production. J Dent Res Candida albicans germ-tube formation. APMIS 2001;109:147–
1995;74:1367–73. 54.
Kuroda M, Nagasaki S, Ito R, et al. Sesquiterpene farnesol as a Nair RG, Samaranayake LP. The effect of oral commensal bacte-
competitive inhibitor of lipase activity of Staphylococcus au- ria on candidal adhesion to human buccal epithelial cells in
reus. FEMS Microbiol Lett 2007;273:28–34. vitro. J Med Microbiol 1996;45:179–85.
O’Donnell et al. 13

Nash EE, Peters BM, Palmer GE, et al. Morphogenesis is Ramage G, Saville SP, Wickes BL et al. Inhibition of Candida
not required for Candida albicans-Staphylococcus aureus albicans biofilm formation by farnesol, a quorum-sensing
intra-abdominal infection-mediated dissemination and molecule. Appl Environ Microb 2002a;68:5459–63.
lethal sepsis. Infect Immun 2014;82:3426–35. Ramage G, Tomsett K, Wickes BL, et al. Denture stomatitis: a role
Nasution O, Srinivasa K, Kim M, et al. Hydrogen peroxide in- for Candida biofilms. Oral Surg Oral Med O 2004;98:53–9.
duces hyphal differentiation in Candida albicans. Eukaryot Cell Ramage G, VandeWalle K, Lopez-Ribot JL, et al. The filamentation
2008;7:2008–11. pathway controlled by the Efg1 regulator protein is required
Nett JE. Future directions for anti-biofilm therapeutics targeting for normal biofilm formation and development in Candida al-
Candida. Expert Rev Anti-Infe 2014;12:375–82. bicans. FEMS Microbiol Lett 2002b;214:95–100.
Nett JE, Marchillo K, Spiegel CA, et al. Development and valida- Ramage G, Zalewska A, Cameron DA, et al. A comparative in
tion of an in vivo Candida albicans biofilm denture model. In- vitro study of two denture cleaning techniques as an ef-
fect Immun 2010;78:3650–9. fective strategy for inhibiting Candida albicans biofilms on
Neu TR, Verkerke GJ, Herrmann IF, et al. Microflora on explanted denture surfaces and reducing inflammation. J Prosthodont

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


silicone rubber voice prostheses: taxonomy, hydrophobicity 2012;21:516–22.
and electrophoretic mobility. J Appl Bacteriol 1994;76:521–8. Rams TE, Babalola OO, Slots J. Subgingival occurrence of en-
Nikawa H, Hamada T, Yamamoto T. Denture plaque—past and teric rods, yeasts and staphylococci after systemic doxycy-
recent concerns. J Dent 1998;26:299–304. cline therapy. Oral Microbiol Immun 1990;5:166–8.
Nobbs AH, Jenkinson HF. Interkingdom networking within the Rautemaa R, Ramage G. Oral candidosis–clinical challenges of a
oral microbiome. Microbes Infect 2015;17:484–92. biofilm disease. Crit Rev Microbiol 2011;37:328–36.
Noverr MC, Huffnagle GB. Regulation of Candida albicans Redding SW, Marr KA, Kirkpatrick WR, et al. Candida glabrata sep-
morphogenesis by fatty acid metabolites. Infect Immun sis secondary to oral colonization in bone marrow transplan-
2004;72:6206–10. tation. Med Mycol 2004;42:479–81.
Oksala E. Factors predisposing to oral yeast infections. Acta Odon- Ribeiro DG, Pavarina AC, Dovigo LN, et al. Prevalence of Candida
tol Scand 1990;48:71–4. spp. associated with bacteria species on complete dentures.
Orsi CF, Sabia C, Ardizzoni A, et al. Inhibitory effects of different Gerodontology 2012;29:203–8.
lactobacilli on Candida albicans hyphal formation and biofilm Romani L, Zelante T, Palmieri M, et al. The cross-talk between op-
development. J Biol Reg Homeos Ag 2014;28:743–52. portunistic fungi and the mammalian host via microbiota’s
Pammi M, Liang R, Hicks J, et al. Biofilm extracellular DNA en- metabolism. Semin Immunopathol 2015;37:163–71.
hances mixed species biofilms of Staphylococcus epidermidis Roux D, Gaudry S, Dreyfuss D, et al. Candida albicans impairs
and Candida albicans. BMC Microbiol 2013;13:257. macrophage function and facilitates Pseudomonas aeruginosa
Peciuliene V, Reynaud AH, Balciuniene I, et al. Isolation of yeasts pneumonia in rat. Crit Care Med 2009;37:1062–7.
and enteric bacteria in root-filled teeth with chronic apical Sachdeo A, Haffajee AD, Socransky SS. Biofilms in the edentu-
periodontitis. Int Endod J 2001;34:429–34. lous oral cavity. J Prosthodont 2008;17:348–56.
Perlroth J, Choi B, Spellberg B. Nosocomial fungal infections: epi- Salerno C, Pascale M, Contaldo M, et al. Candida-associated den-
demiology, diagnosis, and treatment. Med Mycol 2007;45:321– ture stomatitis. Med Oral Patol Oral 2011;16:e139–43.
46. Sapaar B, Nur A, Hirota K, et al. Effects of extracellular DNA from
Peters BM, Jabra-Rizk MA, O’May GA, et al. Polymicrobial interac- Candida albicans and pneumonia-related pathogens on Can-
tions: impact on pathogenesis and human disease. Clin Mi- dida biofilm formation and hyphal transformation. J Appl Mi-
crobiol Rev 2012a;25:193–213. crobiol 2014;116:1531–42.
Peters BM, Jabra-Rizk MA, Scheper MA, et al. Microbial inter- Sardi JC, Duque C, Mariano FS, et al. Candida spp. in periodontal
actions and differential protein expression in Staphylococcus disease: a brief review. J Oral Sci 2010;52:177–85.
aureus-Candida albicans dual-species biofilms. FEMS Immunol Sardi JC, Duque C, Mariano FS, et al. Adhesion and invasion of
Med Mic 2010;59:493–503. Candida albicans from periodontal pockets of patients with
Peters BM, Noverr MC. Candida albicans-Staphylococcus aureus chronic periodontitis and diabetes to gingival human fibrob-
polymicrobial peritonitis modulates host innate immunity. lasts. Med Mycol 2012;50:43–9.
Infect Immun 2013;81:2178–89. Schlecht LM, Peters BM, Krom BP, et al. Systemic Staphylococcus
Peters BM, Ovchinnikova ES, Krom BP, et al. Staphylococcus au- aureus infection mediated by Candida albicans hyphal inva-
reus adherence to Candida albicans hyphae is mediated by the sion of mucosal tissue. Microbiology 2015;161:168–81.
hyphal adhesin Als3p. Microbiol-Sgm 2012;158:2975–86. Sen BH, Safavi KE, Spangberg LS. Growth patterns of Candida al-
Peters BM, Ovchinnikova ES, Krom BP, et al. Staphylococcus aureus bicans in relation to radicular dentin. Oral Surg Oral Med O
adherence to Candida albicans hyphae is mediated by the hy- 1997;84:68–73.
phal adhesin Als3p. Microbiology 2012b;158:2975–86. Shi B, Chang M, Martin J, et al. Dynamic changes in the subgin-
Peters BM, Ward RM, Rane HS, et al. Efficacy of ethanol against gival microbiome and their potential for diagnosis and prog-
Candida albicans and Staphylococcus aureus polymicrobial nosis of periodontitis. mBio 2015;6:e01926–14.
biofilms. Antimicrob Agents Ch 2013;57:74–82. Shirtliff ME, Peters BM, Jabra-Rizk MA. Cross-kingdom inter-
Przybylowska D, Rubinsztajn R, Chazan R, et al. The prevalence actions: Candida albicans and bacteria. FEMS Microbiol Lett
of oral inflammation among denture wearing patients with 2009;299:1–8.
chronic obstructive pulmonary disease. Adv Exp Med Biol Silva S, Henriques M, Hayes A, et al. Candida glabrata and Can-
2015;858:87–91. dida albicans co-infection of an in vitro oral epithelium. J Oral
Rajendran R, Sherry L, Lappin DF, et al. Extracellular DNA release Pathol Med 2011;40:421–7.
confers heterogeneity in Candida albicans biofilm formation. Simon-Soro A, Guillen-Navarro M, Mira A. Metatranscriptomics
BMC Microbiol 2014;14:303. reveals overall active bacterial composition in caries lesions.
Ramage G, Robertson SN, Williams C. Strength in numbers: an- J Oral Microbiol 2014;6:25443.
tifungal strategies against fungal biofilms. Int J Antimicrob Ag Siqueira JF, Jr, Rocas IN. Diversity of endodontic microbiota re-
2014;43:114–20. visited. J Dent Res 2009;88:969–81.
14 FEMS Yeast Research, 2015, Vol. 15, No. 7

Siqueira JF, Jr, Sen BH. Fungi in endodontic infections. Oral Surg Thein ZM, Samaranayake YH, Samaranayake LP. Effect of oral
Oral Med O 2004;97:632–41. bacteria on growth and survival of Candida albicans biofilms.
Socransky SS, Haffajee AD. Periodontal microbial ecology. Peri- Arch Oral Biol 2006;51:672–80.
odontology 2000 2005;38:135–87. Unnanuntana A, Bonsignore L, Shirtliff ME, et al. The effects of
Socransky SS, Haffajee AD, Cugini MA, et al. Microbial complexes farnesol on Staphylococcus aureus biofilms and osteoblasts. An
in subgingival plaque. J Clin Periodontol 1998;25:134–44. in vitro study. J Bone Jt Surg 2009;91:2683–92.
Soysa NS, Ellepola AN. The impact of cigarette/tobacco smoking Vadiraj S, Nayak R, Choudhary GK, et al. Periodontal pathogens
on oral candidosis: an overview. Oral Dis 2005;11:268–73. and respiratory diseases-evaluating their potential associa-
Soysa NS, Samaranayake LP, Ellepola AN. Cytotoxic drugs, radio- tion: a clinical and microbiological study. J Contemp Dent Pract
therapy and oral candidiasis. Oral Oncol 2004;40:971–8. 2013;14:610–5.
Soysa NS, Samaranayake LP, Ellepola AN. Diabetes mellitus van Weissenbruch R, Albers FW, Bouckaert S, et al. Deterioration
as a contributory factor in oral candidosis. Diabet Med of the Provox silicone tracheoesophageal voice prosthesis:
2006;23:455–9. microbial aspects and structural changes. Acta Oto-laryngol

Downloaded from https://academic.oup.com/femsyr/article-abstract/15/7/fov077/641473 by guest on 03 July 2020


Spinillo A, Capuzzo E, Nicola S, et al. The impact of oral 1997a;117:452–458.
contraception on vulvovaginal candidiasis. Contraception van Weissenbruch R, Bouckaert S, Remon JP, et al. Chemoprophy-
1995;51:293–7. laxis of fungal deterioration of the Provox silicone tracheoe-
Stacy A, Everett J, Jorth P, et al. Bacterial fight-and-flight re- sophageal prosthesis in postlaryngectomy patients. Ann Otol
sponses enhance virulence in a polymicrobial infection. P Rhinol Laryngol 1997b;106:329–37.
Natl Acad Sci USA 2014;111:7819–24. Vendeville A, Winzer K, Heurlier K, et al. Making ‘sense’ of
Strus M, Kucharska A, Kukla G, et al. The in vitro activity of vagi- metabolism: autoinducer-2, LuxS and pathogenic bacteria.
nal Lactobacillus with probiotic properties against Candida. In- Nat Rev Microbiol 2005;3:383–96.
fect Dis Obstet Gynecol 2005;13:69–75. Verran J, Jackson S, Coulthwaite L, et al. The effect of dentifrice
Sud IJ, Feingold DS. Action of antifungal imidazoles on Staphylo- abrasion on denture topography and the subsequent reten-
coccus aureus. Antimicrob Agents Ch 1982;22:470–4. tion of microorganisms on abraded surfaces. J Prosthet Dent
Swidergall M, Ernst AM, Ernst JF. Candida albicans mucin Msb2 2014;112:1513–22.
is a broad-range protectant against antimicrobial peptides. Vos T, Flaxman AD, Naghavi M, et al. Years lived with disability
Antimicrob Agents Ch 2013;57:3917–22. (YLDs) for 1160 sequelae of 289 diseases and injuries 1990–
Syed SA, Loesche WJ. Bacteriology of human experimental gin- 2010: a systematic analysis for the global burden of disease
givitis: effect of plaque age. Infect Immun 1978;21:821–9. study 2010. Lancet 2012;380:2163–96.
Taff HT, Nett JE, Zarnowski R, et al. A Candida biofilm-induced Williams DW, Kuriyama T, Silva S, et al. Candida biofilms and oral
pathway for matrix glucan delivery: implications for drug re- candidosis: treatment and prevention. Periodontology 2000
sistance. PLoS Pathog 2012;8:e1002848. 2011;55:250–65.
Takahashi N, Nyvad B. The role of bacteria in the caries process: Ximenez-Fyvie LA, Haffajee AD, Socransky SS. Comparison of
ecological perspectives. J Dent Res 2011;90:294–303. the microbiota of supra- and subgingival plaque in health
Talpaert MJ, Balfour A, Stevens S, et al. Candida biofilm formation and periodontitis. J Clin Periodontol 2000;27:648–57.
on voice prostheses. J Med Microbiol 2015;64:199–208. Xu H, Sobue T, Thompson A, et al. Streptococcal co-infection
Tamai R, Sugamata M, Kiyoura Y. Candida albicans enhances augments Candida pathogenicity by amplifying the mucosal
invasion of human gingival epithelial cells and gingival fi- inflammatory response. Cell Microbiol 2014;16:214–31.
broblasts by Porphyromonas gingivalis. Microb Pathog 2011;51: Yasui M, Ryu M, Sakurai K, et al. Colonisation of the oral cavity
250–4. by periodontopathic bacteria in complete denture wearers.
Tawara Y, Honma K, Naito Y. Methicillin-resistant Staphylococcus Gerodontology 2012;29:e494–502.
aureus and Candida albicans on denture surfaces. Bull Tokyo Young G, Krasner RI, Yudkofsky PL. Interactions of oral strains of
Dent Coll 1996;37:119–28. Candida albicans and lactobacilli. J Bacteriol 1956;72:525–9.
Teles FR, Teles RP, Sachdeo A, et al. Comparison of microbial Zakout YM, Salih MM, Ahmed HG. Frequency of Candida species
changes in early redeveloping biofilms on natural teeth and in papanicolaou smears taken from Sudanese oral hormonal
dentures. J Periodontol 2012;83:1139–48. contraceptives users. Biotech Histochem 2012;87:95–7.

Das könnte Ihnen auch gefallen