Download as pdf or txt
Download as pdf or txt
You are on page 1of 7

Received: 8 September 2016 Revised: 20 September 2016 Accepted: 22 September 2016

DOI 10.1111/cmi.12674

MICROREVIEW

Polymicrobial infections involving clinically relevant


Gram‐negative bacteria and fungi
Sanjiveeni Dhamgaye1 | Yue Qu1,2 | Anton Y. Peleg1,2

1
Biomedicine Discovery Institute, Department
of Microbiology, Faculty of Medicine, Nursing Abstract
and Health Sciences, Monash University, Interactions between fungi and bacteria and their relevance to human health and disease have
Clayton, Victoria, Australia recently attracted increased attention in biomedical fields. Emerging evidence shows that bacteria
2
Department of Infectious Diseases, The and fungi can have synergistic or antagonistic interactions, each with important implications for
Alfred Hospital and Central Clinical School,
human colonization and disease. It is now appreciated that some of these interactions may be stra-
Monash University, Melbourne, Victoria,
Australia tegic and helps promote the survival of one or both microorganisms within the host. This review
Correspondence will shed light on clinically relevant interactions between fungi and Gram‐negative bacteria. Mech-
Anton Y. Peleg, Department of Infectious anism of interaction, host immune responses, and preventive measures will also be reviewed.
Diseases, Central Clinical School, The Alfred
Hospital and Monash University, Melbourne,
VIC 3004, Australia.
Email: anton.peleg@monash.edu

1 | I N T RO D U CT I O N respiratory tract, the gastrointestinal system, the skin, and the urinary
tract. This is predicted based on the resident microflora of these parts
In their natural habitat, microorganisms often exist in close association of the human body and is now well supported by microbiome studies
with a diversity of other species. These interactions drive evolutionary of healthy humans. The human microbiome project recently analyzed
development and adaptation in each microorganism, which promotes 242 healthy human subjects and revealed that the signature microbes
the survival of the species within a given environmental or of a particular niche differ greatly among individuals (Human
host‐specific niche. To promote survival, the interactions between Microbiome Project, 2012). More recent studies have also focused
microorganisms can either be synergistic or antagonistic and are often on fungal diversity in different parts of the body, also termed the
mediated by metabolic factors, secretion of antimicrobial molecules, or mycobiome (Ghannoum et al., 2010; Cui, Morris, & Ghedin, 2013).
structural properties related to cell–cell interactions (Hogan & Kolter, These studies highlight the dominance of Candida albicans as a resident
2002; Gaddy, Tomaras, & Actis, 2009; Gibson, Sood, & Hogan, 2009; fungus but also highlight the diversity in fungal species that may be
Peleg, Hogan, & Mylonakis, 2010). Within humans or in the natural found in or on the human body including Malassezia, Cladosporium,
environment, microorganisms predominately live as biofilms, which Aureobasidium, Saccharomycetales, Aspergillus, Fusarium, Alternaria,
are complex three‐dimensional structures comprising of cell aggregates Penicillium, and Cryptococcus (Ghannoum et al., 2010; Charlson et al.,
encased within a self‐produced matrix of extracellular polymeric sub- 2012; Findley et al., 2013; Diaz, Strausbaugh, & Dongari‐Bagtzoglou,
stances (Bjarnsholt et al., 2013; Flemming et al., 2016). In reality, these 2014; Goralska, 2014). These fungal species are ubiquitous in the envi-
biofilms are often polymicrobial (Partida‐Martinez, Monajembashi, ronment, and therefore, it is not surprising that they are found in
Greulich, & Hertweck, 2007). The focus of this review is on bacterial– human body sites exposed to the environment (e.g., skin and respira-
fungal interactions and infections involving human pathogens, more tory tract). In a healthy human host, these fungi pose negligible risk
specifically clinically relevant Gram‐negative bacteria. Developing an of acute disease, but when the host immune system is impaired in
understanding of how these microorganisms interact and how the host some way such as post‐chemotherapy or transplant‐related immuno-
immune system responds will help elucidate disease mechanisms and suppression, several of these fungi can become invasive and cause
uncover new insights for novel therapeutic or preventative strategies serious infection. Of all the human fungal pathogens, the most com-
(Kobayashi & Crouch, 2009). mon one that is in constant interaction with a diverse range of bacteria
Clinically, the role of polymicrobial infections on patient outcomes is C. albicans. Hospitalized patients are often heavily colonized with
remains poorly studied. With respect to Gram‐negative bacteria, the C. albicans at many body sites and Candida thrives off forming complex
most common sites for mixed‐species infections with fungi are the biofilms on almost all medical devices, including mechanical ventilator

Cellular Microbiology 2016; 1–7 wileyonlinelibrary.com/journal/cmi © 2016 John Wiley & Sons Ltd 1
2 DHAMGAYE ET AL.

tubing used in the intensive care unit, vascular and urinary catheters, of the dominant bacterial species found in the gastrointestinal tract
and intra‐abdominal draining tubes (Kojic & Darouiche, 2004). Gram‐ of warm blooded animals (Tenaillon, Skurnik, Picard, & Denamur,
negative bacteria also heavily populate these sites, and the mecha- 2010) and is one of the most common causes of urinary tract and
nisms and proposed impact of their interactions are discussed below intra‐abdominal infections, and community‐acquired bloodstream
and shown in Figure 1. infections. Given that Candida is a commensal of the human gut, stud-
ies have assessed Candida–E. coli interactions in the context of perito-
nitis (Gale & Sandoval, 1957; Burd, Raymond, & Dunn, 1992; Akagawa,
Abe, & Yamaguchi, 1995; Klaerner et al., 1997; Ikeda, Suegara, Abe, &
2 | C L I N I C A L L Y I M P O R T A N T B A C T E R I A L–
Yamaguchi, 1999). Klaerner et al. showed that the interaction between
FUNGAL INTERACTIONS
E. coli and C. albicans was synergistic and increased lethality in mice
when infected intraperitoneally compared to mono‐infection with
2.1 | C. albicans and Escherichia coli either microorganism (Klaerner et al., 1997). Similarly, in the urinary
E. coli is a Gram‐negative bacterium that specializes in transitioning tract, E. coli contributed to the establishment of C. albicans infection
from a commensal to an opportunistic pathogen of humans. It is one in which C. albicans alone failed to cause infection due to its inability

FIGURE 1 Mechanisms and impact of Gram‐negative bacteria and fungal interaction I. Direct cell–cell contact: P. aeruginosa preferentially
attached to C. albicans hyphae forming biofilms on the filaments (Hogan & Kolter, 2002; Gibson et al., 2009) II. Facilitation of tissue attachment:
C. albicans attachment to the bladder mucosa is facilitated by the presence of E. coli (Levison & Pitsakis, 1987). III. Interaction through secretory
metabolites: (a) a feedback mechanism where phenazines from P. aeruginosa stimulate production of ethanol by C. albicans that subsequently
benefits P. aeruginosa (Chen et al., 2014). (b) Candida QS molecule, farnesol inhibits A. baumannii growth and viability (Peleg et al., 2008) and (c)
A. baumannii secretory factor and Pseudomonas QS molecule (C12‐acyl homoserine lactone) inhibits C. albicans yeast to hyphae transition (Cugini
et al., 2007; Gaddy et al., 2009)
DHAMGAYE ET AL. 3

to attach to the bladder mucosal lining (Levison & Pitsakis, 1987). The P. aeruginosa regulated its own production of various quorum‐sensing
mechanism was shown to be agglutination of C. albicans to the man- (QS) molecules that consequentially induced the production of several
nose‐binding E. coli, which facilitated the attachment of Candida to P. aeruginosa virulence factors including pyoverdine, rhamnolipids, and
the bladder mucosa (Figure 1; Levison & Pitsakis, 1987). It has also pyocyanin (Trejo‐Hernandez, Andrade‐Dominguez, Hernandez, &
been reported that E. coli positively modulates biofilm growth of Encarnacion, 2014). There have now been several studies characteriz-
C. albicans in vitro, and this was due to bacterial lipopolysaccharide ing the complex mechanisms of interaction between Candida and
(LPS; Bandara, Yau, Watt, Jin, & Samaranayake, 2009). The mechanism Pseudomonas (Peleg et al., 2010). These studies have shown both
by which E. coli LPS facilitates C. albicans biofilm growth is not known, antagonistic and synergistic interactions, depending on diverse envi-
but a hypothesis is that it is secondary to LPS‐mediated glucocorticoid ronmental factors, timing of interaction, and growth state at the time
production leading to immunosuppression (Besedovsky, del Rey, of interaction. For example, co‐culturing P. aeruginosa and C. albicans
Sorkin, & Dinarello, 1986; Chang, Feddersen, Henson, & Voelkel, in liquid culture showed that P. aeruginosa preferentially adhered to
1987; Burd et al., 1992; Akagawa et al., 1995). Despite this positive and formed biofilms on C. albicans hyphae rather than yeast cells,
association between E. coli and C. albicans in the urinary tract, E. coli and through the secretion of phospholipase C and phenazines, caused
negatively modulated the growth and biofilm formation of other hyphal death (Figure 1; Hogan & Kolter, 2002; Gibson et al., 2009). A
Candida species, including C. tropicalis, C. dubliniensis, C. krusei, and high concentration of phenazines triggered the production of toxic
C. parapsilosis (Bandara et al., 2009). Thus far, the clinical implications reactive oxygen species that killed C. albicans hyphal cells (Hogan &
of this antagonism in a biofilm setting remain unknown, but further Kolter, 2002; Nseir et al., 2007; Morales et al., 2013). A lower concen-
work is welcomed and may uncover relevant mechanisms that could tration of phenazines impaired hyphal growth of C. albicans and more
be exploited as future novel therapeutics. importantly switched fungal respiration to fermentation leading to
the production of ethanol, glycerol, and acetate by C. albicans in glu-
cose containing media (Morales et al., 2013). It was shown that
2.2 | C. albicans and Pseudomonas aeruginosa C. albicans ethanol production not only influenced biofilm maturation
The respiratory tract and the skin are the major human body sites har- but also promoted more phenazine production by P. aeruginosa
boring interactions between C. albicans and P. aeruginosa. The clinical through WspR‐dependent activation of Pel exopolysaccharide (Chen
impact of a polymicrobial infection involving these two microorgan- et al., 2014). The spectrum of P. aeruginosa phenazines produced was
isms has been studied in acute ventilator‐associated pneumonia in favor of those most effective against fungal cells and led to greater
(VAP), as well as in chronic suppurative lung disease such as cystic production of ethanol by C. albicans, forming a feedback loop driving
fibrosis. C. albicans has been frequently isolated from the airways of the polymicrobial interaction towards the protection of P. aeruginosa
patients on mechanical ventilatory support (Hamet et al., 2012) and (Chen et al., 2014).
in general is clinically ignored. It has now been shown that Candida col- Other well‐studied molecular factors governing the interactions
onization of the respiratory tract may promote the development of between P. aeruginosa and C. albicans are their QS systems. QS mole-
pseudomonal VAP and has been associated with the presence of cules play a crucial role in inter‐cellular communication within a given
multidrug‐resistant bacteria (Azoulay et al., 2006; Hamet et al., population of cells, and there has been greater appreciation of their
2012). Furthermore, more severe clinical outcomes have been cross‐kingdom impact. P. aeruginosa produces acyl homoserine lac-
observed in patients with VAP and cystic fibrosis infected with tones, such as 3‐oxo‐C12‐homoserine lactone that inhibits the Ras1–
P. aeruginosa and Candida spp. simultaneously, relative to P. aeruginosa cyclic AMP–protein kinase A pathway required for hyphal growth in
infection alone (Navarro et al., 2001; Hamet et al., 2012). These human C. albicans, thereby inhibiting filamentation of the fungus (Hogan,
data have also been supported by findings from a rat pneumonia model Vik, & Kolter, 2004; Davis‐Hanna, Piispanen, Stateva, & Hogan,
(Roux et al., 2009) and by another clinical study showing that antifun- 2008; McAlester, O'Gara, & Morrissey, 2008). On the other hand,
gal treatment can decrease the risk of developing P. aeruginosa infec- C. albicans modulates P. aeruginosa biological activities through the
tion in patients on mechanical ventilation (Nseir et al., 2007). These production of farnesol, a well‐known eukaryotic QS molecule. Cugini
studies highlight that Candida colonization in the airways may in itself et al. initially showed that farnesol produced by C. albicans reduced
not cause disease, but it may be playing an underappreciated role in the production of Pseudomonas quinolone signal and pyocyanin in
facilitating bacterial infection. P. aeruginosa (Cugini, Calfee, Morales, Pesci, & Hogan, 2007). In a later
Patients with burn wounds are also confronted with Candida– study, they reported that farnesol stimulated the production of Pseudo-
P. aeruginosa co‐colonization and infection (Gupta, Haque, monas quinolone signal, N‐butanoyl‐L‐homoserine lactone and phena-
Mukhopadhyay, Narayan, & Prasad, 2005). Clinical outcome data are zine in P. aeruginosa when the growth environment changed from a
sparse in this area, but a previous study using a murine burn wound liquid culture to a colony biofilm grown on solid medium (Cugini,
infection model has shown that co‐infection led to greater mortality Morales, & Hogan, 2010). The authors speculated that the varied
than monomicrobial infection, and greater fungal burdens were effects of farnesol on P. aeruginosa QS systems were associated with
observed at the burn wound site and internal organs (Neely, Law, & different pathways it activated, either through interaction with specific
Holder, 1986). targets such as QS transcription factors (Cugini et al., 2007), or through
The enhanced virulence observed with Candida–Pseudomonas co‐ promoting the production of reactive oxygen species, which is linked
infection may be partly explained by a recent proteomics study, which to the activation of P. aeruginosa QS systems (Cugini et al., 2010).
found that when co‐cultured with C. albicans in mixed biofilms, Overall, the laboratory studies characterizing the diverse interactions
4 DHAMGAYE ET AL.

between Candida and Pseudomonas highlight the real complexities of to protect itself from various environmental insults such as UV rays,
their interaction, and questions still remain as to how they apply during temperature shocks, predation by amoebae, and heavy metal toxicity
human infection. (Nosanchuk & Casadevall, 2003; Frases, Chaskes, Dadachova, &
Casadevall, 2006; Kronstad et al., 2012). Melanin production by
C. neoformans is normally dependent on an environmental precursor.
2.3 | C. albicans and Acinetobacter baumannii
A remarkable interaction between C. neoformans and a Gram‐negative
A. baumannii has gained considerable importance as a multidrug‐resis- bacterium, Klebsiella aerogenes, was observed in a laboratory setting
tant, hospital‐associated pathogen (Howard, O'Donoghue, Feeney, & where C. neoformans was able to produce melanin by converting tyro-
Sleator, 2012). It has a particular predilection in causing infections in sine to L‐3,4‐dihydroxyphenylalanine utilizing a bacterial enzyme dur-
patients in intensive care units, with VAP and bloodstream infection ing combined growth (Frases et al., 2006). C. neoformans has also
being the most life‐threatening. A. baumannii is able to survive in the been shown to interact with A. baumannii, whereby enhanced produc-
hospital environment for prolonged periods of time, leading to prob- tion of the polysaccharide capsule was observed for certain serotypes
lematic hospital outbreaks (Garnacho‐Montero et al., 2005). (Abdulkareem, Lee, Ahmadi, & Martinez, 2015). It would appear that
A. baumannii interacts with fungal species, most importantly C. albicans, through evolution, C. neoformans has developed multiple strategies
in critically‐ill patients who are often on mechanical ventilation and for survival, including taking advantage of bacterial neighbors for
have vascular and urinary catheters in situ. The first descriptions of self‐preservation. However, the clinical implications of these findings
A. baumannii–fungal interactions were with the model yeast Saccharo- are not clear, as co‐infections with these Gram‐negative bacteria
myces cerevisiae, where Acinetobacter was able to utilize ethanol pro- would be extremely rare.
duced by the yeast as a carbon source for improved growth (Smith,
Des Etages, & Snyder, 2004). Interestingly, these findings led to subse-
quent studies showing that ethanol mediated increased A. baumannii 2.5 | Aspergillus fumigatus and Gram‐negative
resistance to salt stress, increased growth, virulence gene expression, bacteria
and virulence in multiple non‐mammalian infection model systems Apart from yeast–bacterial interactions, interactions with Aspergillus,
(Smith et al., 2004; Smith et al., 2007; Camarena, Bruno, Euskirchen, the most common mould pathogen of humans, have also been studied
Poggio, & Snyder, 2010). In contrast, laboratory studies assessing inter- (Brandl et al., 2011; Kwon‐Chung & Sugui, 2013). The clinical rele-
actions between A. baumannii and C. albicans have demonstrated an vance of these interactions predominately relates to the respiratory
antagonistic relationship, whereby A. baumannii preferentially targets tract and is especially evident in those with suppurative lung disease
the key virulence characteristic of C. albicans; hyphal development such as patients with cystic fibrosis (Kwon‐Chung & Sugui, 2013).
(Peleg et al., 2008). The A. baumannii outer membrane protein was In the context of cystic fibrosis, Aspergillus interacts with a multitude
implicated as a mechanism of attachment and killing of the filamentous of respiratory Gram‐negative bacteria, including P. aeruginosa,
form of Candida by triggering apoptosis (Gaddy et al., 2009). However, Burkholderia cepacia complex, Stenotrophomonas maltophilia, and
a secretory factor was also thought to be at play (Gaddy et al., 2009). Achromobacter spp. (Brandl et al., 2011; de Vrankrijker et al., 2011;
This polymicrobial interaction was also studied within the nematode Hauser, Jain, Bar‐Meir, & McColley, 2011; Lambiase et al., 2011).
Caenorhabditis elegans, which showed that A. baumannii prevented P. aeruginosa exerts an inhibitory effect on the establishment of
the morphological transition of C. albicans from a yeast to hyphal cell, A. fumigatus biofilms by secreting a small diffusible heat‐stable mole-
and this in turn led to reduced severity of disease (Peleg et al., 2008). cule (Mowat et al., 2010). The effect of this molecule however was
Conversely, when C. albicans was allowed to establish a mature biofilm rendered ineffective once the biofilm was established. Notably, a
before co‐infection with A. baumannii, the growth of the bacteria was P. aeruginosa QS mutant did not show inhibitory effects on A. fumigatus
impaired, and this was shown to be secondary to the secretion of the biofilm formation, indicating involvement of the QS system in the inhi-
Candida QS molecule farnesol (Peleg et al., 2008). Farnesol appears bition, but further investigations are required to identify the exact mol-
to mediate its effect on A. baumannii by interfering with bacterial mem- ecule (Mowat et al., 2010). Whether these interactions impact on the
brane integrity (Kostoulias et al., 2016). Similar to the Candida–Pseudo- co‐existence of these microorganisms and the magnitude of lung
monas interactions, the nature of Candida–A. baumannii interactions destruction in patients with cystic fibrosis remains to be determined.
are diverse and are determined by environmental conditions, initial
inoculum, and growth state of the individual microorganisms.
3 | HOST IMMUNE RESPONSES TO
B A C TE R I A L – FU N G A L I N F E C T I O N S
2.4 | Cryptococcus neoformans and Gram‐negative
bacteria
Very little is known about host immune responses to polymicrobial
Apart from Candida, bacterial–fungal interactions with another yeast infections. It is clear that immune responses against bacteria or fungi
known as C. neoformans have also been reported. C. neoformans is an alone are dissimilar (Allard et al., 2009), but whether dual infection
encapsulated soil fungus that is capable of infecting humans through stimulates both immune pathways or is dominated by one has only
the respiratory tract. It is particularly dangerous in individuals with recently been explored. In a mouse model to investigate lung adaptive
impaired immunity, causing life‐threatening meningitis (Howard et al., immune responses, fungal lysate alone from C. albicans and A. fumigatus
2012). As a survival mechanism, C. neoformans synthesizes melanin led to airway eosinophilia, release of TH2 type cytokines (IL‐4, IL‐5 and
DHAMGAYE ET AL. 5

IL‐13), and changes in mucus production, whereas bacterial antigens biofilm‐related infections. It will clearly take a combination of innova-
from P. aeruginosa evoked an inflammatory response dominated by tive strategies to prevent the burgeoning problem of biofilm‐related
neutrophils, secretion of TH1 type cytokines, and minimal mucus pro- infections, in particular those that are polymicrobial (Salwiczek
duction (Allard et al., 2009). Interestingly, co‐administration of bacte- et al., 2014).
rial and fungal antigens activated immune responses characteristic of
bacteria alone, suggesting that at least in this mouse model, bacterial
5 | CO NC LUSIO NS
antigens were able to immunomodulate the response towards fungal
antigens (Allard et al., 2009). How Pseudomonas antigens direct the
Although the seriousness and the impact of bacterial–fungal interac-
immune response towards fungal lysates from TH2 to TH1 is not
tions have been recognized globally, effective measures to deal with
defined, but it appears to be independent of bacterial LPS and TLR4
the problem are lacking. From a mechanistic point of view, opportuni-
(Allard et al., 2009). A subsequent report suggested that Candida can
ties exist for identifying novel therapeutic strategies by exploiting the
also modulate the host immune response, which may promote
ability of one microorganism to inhibit the growth and viability of
pseudomonal infection (Roux et al., 2009). Using a rat pneumonia
another. Also, new combination therapies and surface‐attached antimi-
model, pre‐infection with Candida helped establish P. aeruginosa pneu-
crobials may help to target these interactions. Our understanding of
monia by impairing reactive oxygen species generation by alveolar
polymicrobial infections and their mechanisms of interaction are evolv-
macrophages (Roux et al., 2009). These studies highlight the potential
ing, but it is clear that we still have a great deal to learn about this com-
clinical implications of understanding how the host immune system
plex and diverse problem.
reacts to mixed bacterial–fungal infections and further studies may
provide important insights for the development of future preventative
ACKNOWLEDGMENT
or therapeutic strategies against polymicrobial infections.
A.Y.P was supported by an Australian National Health and Medical
Research Council Career Development Fellowship and Project Grant.

4 | T RE A T M E NT A N D P R EV E N T A T I V E
CONFLICTS OF INTERES T
S T R A T E G I E S A G A I N S T BA C T E R I A L –F U N G A L
INFECTIONS The authors declare that they have no competing interests.

Very little is known about the most effective antimicrobial treatment RE FE RE NC ES

of polymicrobial infections. Standard therapy includes use of conven- Abdulkareem, A. F., Lee, H. H., Ahmadi, M., & Martinez, L. R. (2015). Fungal
serotype‐specific differences in bacterial–yeast interactions. Virulence,
tional antifungal and antibacterial agents based on antimicrobial
6, 652–657.
susceptibility results for individual microorganisms, but reports of
Akagawa, G., Abe, S., & Yamaguchi, H. (1995). Mortality of Candida
greater severity of illness, longer duration of symptoms, and greater albicans‐infected mice is facilitated by superinfection of Escherichia coli
modification of treatment antimicrobials in polymicrobial infections or administration of its lipopolysaccharide. Journal of Infectious Diseases,
171, 1539–1544.
exist (Dyess, Garrison, & Fry, 1985; Tsai et al., 2014). Though
C. albicans was previously considered a colonizer in the respiratory Allard, J. B., Rinaldi, L., Wargo, M. J., Allen, G., Akira, S., Uematsu, S., …
Whittaker, L. A. (2009). Th2 allergic immune response to inhaled fungal
tract, it may actually play an important role in facilitating Gram‐ antigens is modulated by TLR‐4‐independent bacterial products. Euro-
negative bacterial infection through protection within biofilms or alter- pean Journal of Immunology, 39, 776–788.
ation of local host innate immune defenses. It is well established that Azoulay, E., Timsit, J. F., Tafflet, M., de Lassence, A., Darmon, M., Zahar, J.
organisms are more resistant to antimicrobials in biofilms, and this R., … Schlemmer, B. (2006). Candida colonization of the respiratory
tract and subsequent pseudomonas ventilator‐associated pneumonia.
appears to be an even greater problem with polymicrobial biofilms
Chest, 129, 110–117.
(Harriott & Noverr, 2009; Chotirmall et al., 2010; Filkins & O'Toole,
Bandara, H. M., Yau, J. Y., Watt, R. M., Jin, L. J., & Samaranayake, L. P.
2015). The fungal scaffold may provide a shield for the interacting (2009). Escherichia coli and its lipopolysaccharide modulate in vitro Can-
bacteria, and therefore, combining a biofilm‐active antifungal with an dida biofilm formation. Journal of Medical Microbiology, 58, 1623–1631.
effective antibacterial may provide a potential therapeutic strategy for Besedovsky, H., del Rey, A., Sorkin, E., & Dinarello, C. A. (1986). Immuno-
these difficult infections (Qu et al., 2016). regulatory feedback between interleukin‐1 and glucocorticoid
hormones. Science, 233, 652–654.
Polymicrobial biofilms have been implicated in the pathogenesis of
Bjarnsholt, T., Alhede, M., Alhede, M., Eickhardt‐Sorensen, S. R., Moser, C.,
cystic fibrosis and many hospital acquired infections related to
Kuhl, M., … Hoiby, N. (2013). The in vivo biofilm. Trends in Microbiology,
implanted medical devices such as central venous or urinary catheters 21, 466–474.
(Coad, Griesser, Peleg, & Traven, 2016). Developing effective and Brandl, M. T., Carter, M. Q., Parker, C. T., Chapman, M. R., Huynh, S., &
durable medical surface‐attached antimicrobials that have activity Zhou, Y. (2011). Salmonella biofilm formation on Aspergillus niger
against Candida and Gram‐negative or –positive bacteria forms an involves cellulose–chitin interactions. PloS One, 6, e25553.

unmet clinical need. A recent example targeting the Candida biofilm Burd, R. S., Raymond, C. S., & Dunn, D. L. (1992). Endotoxin promotes syn-
ergistic lethality during concurrent Escherichia coli and Candida albicans
structure is the covalent surface attachment of the echinocandin
infection. Journal of Surgical Research, 52, 537–542.
antifungal, caspofungin (Coad et al., 2015; Kucharikova et al., 2016).
Camarena, L., Bruno, V., Euskirchen, G., Poggio, S., & Snyder, M. (2010).
These data hold promise for the prevention of the supporting Candida Molecular mechanisms of ethanol‐induced pathogenesis revealed by
biofilm structure that may be an important driver of polymicrobial RNA‐sequencing. PLoS Pathogens, 6, e1000834.
6 DHAMGAYE ET AL.

Chang, S. W., Feddersen, C. O., Henson, P. M., & Voelkel, N. F. (1987). Gale, D., & Sandoval, B. (1957). Response of mice to the inoculations of
Platelet‐activating factor mediates hemodynamic changes and lung both Candida albicans and Escherichia coli. I. The enhancement phenom-
injury in endotoxin‐treated rats. Journal of Clinical Investigation, 79, enon. Journal of Bacteriology, 73, 616–624.
1498–1509. Garnacho‐Montero, J., Ortiz‐Leyba, C., Fernandez‐Hinojosa, E., Aldabo‐
Charlson, E. S., Diamond, J. M., Bittinger, K., Fitzgerald, A. S., Yadav, A., Pallas, T., Cayuela, A., Marquez‐Vacaro, J. A., … Jimenez‐Jimenez, F. J.
Haas, A. R., … Collman, R. G. (2012). Lung‐enriched organisms and aber- (2005). Acinetobacter baumannii ventilator‐associated pneumonia: Epi-
rant bacterial and fungal respiratory microbiota after lung transplant. demiological and clinical findings. Intensive Care Medicine, 31, 649–655.
American Journal of Respiratory and Critical Care Medicine, 186, 536– Ghannoum, M. A., Jurevic, R. J., Mukherjee, P. K., Cui, F., Sikaroodi, M.,
545. Naqvi, A., & Gillevet, P. M. (2010). Characterization of the oral fungal
Chen, A. I., Dolben, E. F., Okegbe, C., Harty, C. E., Golub, Y., Thao, S., … microbiome (mycobiome) in healthy individuals. PLoS Pathogens, 6,
Hogan, D. A. (2014). Candida albicans ethanol stimulates Pseudomonas e1000713.
aeruginosa WspR‐controlled biofilm formation as part of a cyclic rela- Gibson, J., Sood, A., & Hogan, D. A. (2009). Pseudomonas aeruginosa‐
tionship involving phenazines. PLoS Pathogens, 10, e1004480. Candida albicans interactions: Localization and fungal toxicity of a
Chotirmall, S. H., O'Donoghue, E., Bennett, K., Gunaratnam, C., O'Neill, S. J., phenazine derivative. Applied and Environmental Microbiology, 75,
& McElvaney, N. G. (2010). Sputum Candida albicans presages FEV(1) 504–513.
decline and hospital‐treated exacerbations in cystic fibrosis. Chest, Goralska, K. (2014). Interactions between potentially pathogenic fungi and
138, 1186–1195. natural human microbiota. Annals of Parasitology, 60, 159–168.
Coad, B. R., Griesser, H. J., Peleg, A. Y., & Traven, A. (2016). Anti‐infective Gupta, N., Haque, A., Mukhopadhyay, G., Narayan, R. P., & Prasad, R.
surface coatings: Design and therapeutic promise against device‐ (2005). Interactions between bacteria and Candida in the burn wound.
associated infections. PLoS Pathogens, 12, e1005598. Burns, 31, 375–378.
Coad, B. R., Lamont‐Friedrich, S. J., Gwynne, L., Jasieniak, M., Griesser, S. S., Hamet, M., Pavon, A., Dalle, F., Pechinot, A., Prin, S., Quenot, J. P., &
Traven, A., … Griesser, H. J. (2015). Surface coatings with covalently Charles, P. E. (2012). Candida spp. airway colonization could promote
attached caspofungin are effective in eliminating fungal pathogens. antibiotic‐resistant bacteria selection in patients with suspected venti-
Journal of Materials Chemistry B, 3, 8469–8476. lator‐associated pneumonia. Intensive Care Medicine, 38, 1272–1279.
Cugini, C., Calfee, M. W., Farrow, J. M. III, Morales, D. K., Pesci, E. C., & Harriott, M. M., & Noverr, M. C. (2009). Candida albicans and Staphylococ-
Hogan, D. A. (2007). Farnesol, a common sesquiterpene, inhibits PQS cus aureus form polymicrobial biofilms: Effects on antimicrobial
production in Pseudomonas aeruginosa. Molecular Microbiology, 65, resistance. Antimicrobial Agents and Chemotherapy, 53, 3914–3922.
896–906.
Hauser, A. R., Jain, M., Bar‐Meir, M., & McColley, S. A. (2011). Clinical sig-
Cugini, C., Morales, D. K., & Hogan, D. A. (2010). Candida albicans‐produced nificance of microbial infection and adaptation in cystic fibrosis. Clinical
farnesol stimulates Pseudomonas quinolone signal production in Microbiology Reviews, 24, 29–70.
LasR‐defective Pseudomonas aeruginosa strains. Microbiology, 156,
Hogan, D. A., & Kolter, R. (2002). Pseudomonas–Candida interactions: An
3096–3107.
ecological role for virulence factors. Science, 296, 2229–2232.
Cui, L., Morris, A., & Ghedin, E. (2013). The human mycobiome in health and Hogan, D. A., Vik, A., & Kolter, R. (2004). A Pseudomonas aeruginosa
disease. Genome Medicine, 5, 63. quorum‐sensing molecule influences Candida albicans morphology.
Davis‐Hanna, A., Piispanen, A. E., Stateva, L. I., & Hogan, D. A. (2008). Molecular Microbiology, 54, 1212–1223.
Farnesol and dodecanol effects on the Candida albicans Ras1‐cAMP Howard, A., O'Donoghue, M., Feeney, A., & Sleator, R. D. (2012).
signalling pathway and the regulation of morphogenesis. Molecular Acinetobacter baumannii: An emerging opportunistic pathogen. Viru-
Microbiology, 67, 47–62. lence, 3, 243–250.
de Vrankrijker, A. M., van der Ent, C. K., van Berkhout, F. T., Stellato, R. K., Human Microbiome Project, C (2012). Structure, function and diversity of
Willems, R. J., Bonten, M. J., & Wolfs, T. F. (2011). Aspergillus fumigatus the healthy human microbiome. Nature, 486, 207–214.
colonization in cystic fibrosis: Implications for lung function? Clinical
Ikeda, T., Suegara, N., Abe, S., & Yamaguchi, H. (1999). Efficacy of
Microbiology and Infection, 17, 1381–1386.
antibacterial drugs in mice with complex infection by Candida albicans
Diaz, P. I., Strausbaugh, L. D., & Dongari‐Bagtzoglou, A. (2014). Fungal– and Escherichia coli. Journal of Antibiotics, 52, 552–558.
bacterial interactions and their relevance to oral health: Linking the
Klaerner, H. G., Uknis, M. E., Acton, R. D., Dahlberg, P. S., Carlone‐Jambor,
clinic and the bench. Frontiers in Cellular and Infection Microbiology, 4,
C., & Dunn, D. L. (1997). Candida albicans and Escherichia coli are syner-
101.
gistic pathogens during experimental microbial peritonitis. Journal of
Dyess, D. L., Garrison, R. N., & Fry, D. E. (1985). Candida sepsis. Implications Surgical Research, 70, 161–165.
of polymicrobial blood‐borne infection. Archives of Surgery, 120,
Kobayashi, D. Y., & Crouch, J. A. (2009). Bacterial/Fungal interactions:
345–348.
From pathogens to mutualistic endosymbionts. Annual Review of Phyto-
Filkins, L. M., & O'Toole, G. A. (2015). Cystic fibrosis lung infections: pathology, 47, 63–82.
Polymicrobial, complex, and hard to treat. PLoS Pathogens, 11,
Kojic, E. M., & Darouiche, R. O. (2004). Candida infections of medical
e1005258.
devices. Clinical Microbiology Reviews, 17, 255–267.
Findley, K., Oh, J., Yang, J., Conlan, S., Deming, C., Meyer, J. A., … Segre, J.
Kostoulias, X., Murray, G. L., Cerqueira, G. M., Kong, J. B., Bantun, F.,
A. (2013). Topographic diversity of fungal and bacterial communities in
Mylonakis, E., … Peleg, A. Y. (2016). Impact of a cross‐kingdom signaling
human skin. Nature, 498, 367–370.
molecule of Candida albicans on Acinetobacter baumannii physiology.
Flemming, H. C., Wingender, J., Szewzyk, U., Steinberg, P., Rice, S. A., & Antimicrobial Agents and Chemotherapy, 60, 161–167.
Kjelleberg, S. (2016). Biofilms: An emergent form of bacterial life.
Kronstad, J., Saikia, S., Nielson, E. D., Kretschmer, M., Jung, W., Hu, G., …
Nature Reviews: Microbiology, 14, 563–575.
Attarian, R. (2012). Adaptation of Cryptococcus neoformans to mamma-
Frases, S., Chaskes, S., Dadachova, E., & Casadevall, A. (2006). Induction by lian hosts: Integrated regulation of metabolism and virulence. Eukaryotic
Klebsiella aerogenes of a melanin‐like pigment in Cryptococcus Cell, 11, 109–118.
neoformans. Applied and Environmental Microbiology, 72, 1542–1550. Kucharikova, S., Gerits, E., De Brucker, K., Braem, A., Ceh, K., Majdic, G., …
Gaddy, J. A., Tomaras, A. P., & Actis, L. A. (2009). The Acinetobacter Thevissen, K. (2016). Covalent immobilization of antimicrobial agents
baumannii 19606 OmpA protein plays a role in biofilm formation on abi- on titanium prevents Staphylococcus aureus and Candida albicans coloni-
otic surfaces and in the interaction of this pathogen with eukaryotic zation and biofilm formation. Journal of Antimicrobial Chemotherapy, 71,
cells. Infection and Immunity, 77, 3150–3160. 936–945.
DHAMGAYE ET AL. 7

Kwon‐Chung, K. J., & Sugui, J. A. (2013). Aspergillus fumigatus – what makes Peleg, A. Y., Tampakakis, E., Fuchs, B. B., Eliopoulos, G. M., Moellering, R. C.
the species a ubiquitous human fungal pathogen? PLoS Pathogens, 9, Jr., & Mylonakis, E. (2008). Prokaryote–eukaryote interactions identi-
e1003743. fied by using Caenorhabditis elegans. Proceedings of the National
Lambiase, A., Catania, M. R., Del Pezzo, M., Rossano, F., Terlizzi, V., Sepe, Academy of Sciences of the United States of America, 105, 14585–14590.
A., & Raia, V. (2011). Achromobacter xylosoxidans respiratory tract infec- Qu, Y., Locock, K., Verma‐Gaur, J., Hay, I. D., Meagher, L., & Traven, A.
tion in cystic fibrosis patients. European Journal of Clinical Microbiology (2016). Searching for new strategies against polymicrobial biofilm infec-
and Infectious Diseases, 30, 973–980. tions: Guanylated polymethacrylates kill mixed fungal/bacterial
Levison, M. E., & Pitsakis, P. G. (1987). Susceptibility to experimental Can- biofilms. Journal of Antimicrobial Chemotherapy, 71, 413–421.
dida albicans urinary tract infection in the rat. Journal of Infectious Roux, D., Gaudry, S., Dreyfuss, D., El‐Benna, J., de Prost, N., Denamur, E., …
Diseases, 155, 841–846. Ricard, J. D. (2009). Candida albicans impairs macrophage function and
McAlester, G., O'Gara, F., & Morrissey, J. P. (2008). Signal‐mediated inter- facilitates Pseudomonas aeruginosa pneumonia in rat. Critical Care Med-
actions between Pseudomonas aeruginosa and Candida albicans. icine, 37, 1062–1067.
Journal of Medical Microbiology, 57, 563–569. Salwiczek, M., Qu, Y., Gardiner, J., Strugnell, R. A., Lithgow, T., McLean,
Morales, D. K., Grahl, N., Okegbe, C., Dietrich, L. E., Jacobs, N. J., & Hogan, K. M., & Thissen, H. (2014). Emerging rules for effective antimicrobial
D. A. (2013). Control of Candida albicans metabolism and biofilm forma- coatings. Trends in Biotechnology, 32, 82–90.
tion by Pseudomonas aeruginosa phenazines. MBio, 4, e00526–e00512. Smith, M. G., Des Etages, S. G., & Snyder, M. (2004). Microbial synergy via
Mowat, E., Rajendran, R., Williams, C., McCulloch, E., Jones, B., Lang, S., & an ethanol‐triggered pathway. Molecular and Cellular Biology, 24,
Ramage, G. (2010). Pseudomonas aeruginosa and their small diffusible 3874–3884.
extracellular molecules inhibit Aspergillus fumigatus biofilm formation. Smith, M. G., Gianoulis, T. A., Pukatzki, S., Mekalanos, J. J., Ornston, L. N.,
FEMS Microbiology Letters, 313, 96–102. Gerstein, M., & Snyder, M. (2007). New insights into Acinetobacter
Navarro, J., Rainisio, M., Harms, H. K., Hodson, M. E., Koch, C., Mastella, G., baumannii pathogenesis revealed by high‐density pyrosequencing and
… McKenzie, S. G. (2001). Factors associated with poor pulmonary transposon mutagenesis. Genes and Development, 21, 601–614.
function: Cross‐sectional analysis of data from the ERCF. European Epi- Tenaillon, O., Skurnik, D., Picard, B., & Denamur, E. (2010). The population
demiologic Registry of Cystic Fibrosis. European Respiratory Journal, 18, genetics of commensal Escherichia coli. Nature Reviews: Microbiology, 8,
298–305. 207–217.
Neely, A. N., Law, E. J., & Holder, I. A. (1986). Increased susceptibility to Trejo‐Hernandez, A., Andrade‐Dominguez, A., Hernandez, M., &
lethal Candida infections in burned mice preinfected with Pseudomonas Encarnacion, S. (2014). Interspecies competition triggers virulence and
aeruginosa or pretreated with proteolytic enzymes. Infection and Immu- mutability in Candida albicans–Pseudomonas aeruginosa mixed biofilms.
nity, 52, 200–204. The ISME Journal, 8, 1974–1988.
Nosanchuk, J. D., & Casadevall, A. (2003). The contribution of melanin to Tsai, M. H., Chu, S. M., Hsu, J. F., Lien, R., Huang, H. R., Chiang, M. C., …
microbial pathogenesis. Cellular Microbiology, 5, 203–223. Huang, Y. C. (2014). Polymicrobial bloodstream infection in neonates:
Nseir, S., Jozefowicz, E., Cavestri, B., Sendid, B., Di Pompeo, C., Dewavrin, Microbiology, clinical characteristics, and risk factors. PloS One, 9,
F., … Durocher, A. (2007). Impact of antifungal treatment on Candida– e83082.
Pseudomonas interaction: A preliminary retrospective case–control
study. Intensive Care Medicine, 33, 137–142.
How to cite this article: Dhamgaye, S., Qu, Y., and Peleg, A. Y.
Partida‐Martinez, L. P., Monajembashi, S., Greulich, K. O., & Hertweck, C.
(2016), Polymicrobial infections involving clinically relevant
(2007). Endosymbiont‐dependent host reproduction maintains bacte-
rial‐fungal mutualism. Current Biology, 17, 773–777. Gram‐negative bacteria and fungi, Cellular Microbiology, doi:
Peleg, A. Y., Hogan, D. A., & Mylonakis, E. (2010). Medically important bac- 10.1111/cmi.12674
terial–fungal interactions. Nature Reviews: Microbiology, 8, 340–349.

You might also like