Vous êtes sur la page 1sur 11

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/268874904

Clinical Implications of Oral Candidiasis: Host Tissue Damage and


Disseminated Bacterial Disease

Article  in  Infection and Immunity · November 2014


DOI: 10.1128/IAI.02843-14 · Source: PubMed

CITATIONS READS

47 663

6 authors, including:

Eric Kong Sona Kucharíková


University of Maryland, Baltimore KU Leuven
12 PUBLICATIONS   401 CITATIONS    51 PUBLICATIONS   1,099 CITATIONS   

SEE PROFILE SEE PROFILE

Patrick Van Dijck Brian Peters


KU Leuven The University of Tennessee Health Science Center
286 PUBLICATIONS   9,253 CITATIONS    98 PUBLICATIONS   3,055 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Chronic orthopaedic biofilm infections View project

Studying the vapour-phase-mediated antimicrobial activity (VMAA) of volatiles View project

All content following this page was uploaded by Mary Ann Jabra-Rizk on 27 January 2015.

The user has requested enhancement of the downloaded file.


Clinical Implications of Oral Candidiasis: Host Tissue Damage and
Disseminated Bacterial Disease
Eric F. Kong,a,b Sona Kucharíková,c,d Patrick Van Dijck,c,d Brian M. Peters,b* Mark E. Shirtliff,e,f Mary Ann Jabra-Rizka,f
Department of Oncology and Diagnostic Sciences, Dental School, University of Maryland, Baltimore, Maryland, USAa; Graduate Program in Life Sciences, Molecular
Microbiology and Immunology Program, University of Maryland, Baltimore, Maryland, USAb; Department of Molecular Microbiology, VIB, KU Leuven, Belgiumc; Laboratory
of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Belgiumd; Department of Microbial Pathogenesis, Dental School, University of Maryland,
Baltimore, Maryland, USAe; Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, Maryland, USAf

The clinical significance of polymicrobial interactions, particularly those between commensal species with high pathogenic po-
tential, remains largely understudied. Although the dimorphic fungal species Candida albicans and the bacterium Staphylococ-
cus aureus are common cocolonizers of humans, they are considered leading opportunistic pathogens. Oral candidiasis specifi-

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


cally, characterized by hyphal invasion of oral mucosal tissue, is the most common opportunistic infection in HIVⴙ and
immunocompromised individuals. In this study, building on our previous findings, a mouse model was developed to investigate
whether the onset of oral candidiasis predisposes the host to secondary staphylococcal infection. The findings demonstrated that
in mice with oral candidiasis, subsequent exposure to S. aureus resulted in systemic bacterial infection with high morbidity and
mortality. Histopathology and scanning electron microscopy of tongue tissue from moribund animals revealed massive C. albi-
cans hyphal invasion coupled with S. aureus deep tissue infiltration. The crucial role of hyphae in the process was demonstrated
using a non-hypha-producing and a noninvasive hypha-producing mutant strains of C. albicans. Further, in contrast to previ-
ous findings, S. aureus dissemination was aided but not contingent upon the presence of the Als3p hypha-specific adhesion. Im-
portantly, impeding development of mucosal C. albicans infection by administering antifungal fluconazole therapy protected
the animals from systemic bacterial disease. The combined findings from this study demonstrate that oral candidiasis may con-
stitute a risk factor for disseminated bacterial disease warranting awareness in terms of therapeutic management of immuno-
compromised individuals.

A considerable number of infectious diseases involve multiple


microbial species coexisting and interacting in a host (1, 2). In
these infections, the presence of one microorganism predisposes
cells to the substrate is followed by proliferation and hypha for-
mation resulting in a network of cells embedded in extracellular
polymeric matrix (21–23). In the oral cavity, C. albicans coexists
the host to colonization by others and in additive polymicrobial and forms tight associations with various commensal bacterial spe-
infections two or more nonpathogenic microorganisms together cies forming complex mucosal biofilms, a phenomenon known to
can cause disease (1, 3). Adherence of organisms to surfaces and to play an important factor in C. albicans colonization (2, 5, 10, 11,
each other is a prerequisite for establishing concurrent infections, 24–26). Thus, when Candida infections arise, they often occur in
which often stem from the formation of polymicrobial biofilms association with bacteria (27). Among the bacterial species, Staph-
that form on biotic or abiotic surfaces (2, 4–6). In the oral cavity, ylococcus aureus specifically is a precarious opportunistic patho-
the oral mucosa serves as an important barrier to the myriad of gen implicated in a variety of diseases, ranging from minor skin
microbial species present in this complex environment. However, infections to more serious invasive diseases (28–30). However, it
the interactions between the various species can be synergistic in
that the presence of one microorganism generates a niche for
other microorganisms (1, 5, 7–11). Received 28 October 2014 Returned for modification 11 November 2014
Candida albicans is an opportunistic fungal species commonly Accepted 13 November 2014
colonizing human mucosal surfaces as a component of the normal Accepted manuscript posted online 24 November 2014
microflora (12–14). However, when host defenses are weakened, Citation Kong EF, Kucharíková S, Van Dijck P, Peters BM, Shirtliff ME, Jabra-Rizk MA.
C. albicans can proliferate causing an array of infections ranging 2015. Clinical implications of oral candidiasis: host tissue damage and
disseminated bacterial disease. Infect Immun 83:604 – 613. doi:10.1128/IAI.02843-
from mucosal such as oral candidiasis (OC) to systemic infections
14.
that are often life-threatening (12, 15–17). Therefore, candidal
Editor: G. S. Deepe, Jr.
infections are unsurprisingly often endogenous occurring when
Address correspondence to Mary Ann Jabra-Rizk, mrizk@umaryland.edu.
there is a disruption in host environment (13). Specifically, OC is
* Present address: Brian M. Peters, Department of Oral and Craniofacial Biology,
considered an AIDS-defining opportunistic infection with up to Dental School, Louisiana State University Health Sciences Center, New Orleans,
80% of HIV⫹ individuals suffering recurrent episodes during the Louisiana, USA.
course of their HIV disease progression (15, 18). E.F.K. and S.K. contributed equally to this article.
The ability of this highly adaptable fungal pathogen to transi- Supplemental material for this article may be found at http://dx.doi.org/10.1128
tion from commensal to pathogen is primarily the result of its /IAI.02843-14.
ability to morphologically switch between yeast and hyphal forms Copyright © 2015, American Society for Microbiology. All Rights Reserved.
(14, 19, 20). In fact, the majority of C. albicans infections are as- doi:10.1128/IAI.02843-14
sociated with its ability to form biofilms where adhesion of yeast

604 iai.asm.org Infection and Immunity February 2015 Volume 83 Number 2


Oral Candidiasis and Systemic Bacterial Infection

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


FIG 1 Infection timeline. A timeline for infection and antifungal therapy model beginning with immunosuppression the day prior to infection with C. albicans
until animals are euthanized 5 days after C. albicans infection is shown.

primarily exists as a commensal organism with approximately transcriptional repressor of filamentous growth and its parent strain
30% of healthy humans colonized in their nasopharynx or on their (SN250) (45, 46), the als3⌬ strain lacking the hyphal specific adhesin
skin. Although colonization itself is a harmless state, colonized Als3p (47, 48) and an MRSA strain (MRSA-M2) recently sequenced by
individuals are at risk of endogenous infection when S. aureus us (GenBank accession number AMTC00000000) (49) were used in
enters otherwise sterile sites via wounds or indwelling medical the present study. C. albicans strains were grown in yeast peptone dex-
devices. With the emergence of methicillin-resistant S. aureus trose broth (Difco Laboratories) overnight at 30°C, and cells were equili-
brated in fresh medium to an optical density at 600 nm of 1.0. S. aureus
(MRSA), this ubiquitous pathogen is becoming an even greater
cultures were grown overnight in Trypticase soy broth (TSB; Difco) at
therapeutic challenge (31–33). 37°C and then grown in fresh TSB to mid-log phase. The cells were har-
Our in vitro studies have demonstrated that S. aureus exhibits vested, washed with phosphate-buffered saline (PBS), and then sus-
high affinity to the C. albicans hyphae as these species coexist in pended in Hanks’ balanced salt solution (Fisher Scientific), and suspen-
biofilm and identified the C. albicans hyphal specific adhesin sions were maintained at 30°C. C. albicans was used at a final cell density of
Als3p to be involved in the coadherence process (34, 35). Further, 2 ⫻ 107 cells/ml, and S. aureus was used at a final cell density of 1 ⫻ 106
our previous in vivo studies have indicated that the interaction of cells/ml.
these species may carry important clinical implications (36, 37). In Animals. Three-month-old pathogen-free female C57BL/6 mice
fact, several studies have reported the coisolation of these diverse (Charles River Laboratories) were used in these studies. Mice were housed at
species from a multitude of diseases such as periodontitis, denture a maximum of five animals per cage. To suppress bacterial flora and allow C.
stomatitis, cystic fibrosis, keratitis, ventilator-associated pneumo- albicans colonization, mice received ampicillin at a dose of 0.5 mg/ml in
nia, urinary tract catheters and burn wound infections (38–42). their drinking water 2 days prior to infection and throughout the study.
Infection model. The coinfection mouse model was performed in
Given the prevalence of OC as an opportunistic infection in
accordance with an established model of OC (50) with modification.
immunocompromised populations and the established clinical Time-line of infection and treatment is illustrated in Fig. 1. Mice were
similarity of OC in mice to that in the human host, we developed rendered susceptible to candidiasis by subcutaneous administration
a mouse model to assess whether OC predisposes to secondary (0.2 ml) of cortisone acetate (225 mg/kg of body weight) in the dorsum
bacterial infection. Further, hypha-deficient mutant strains of C. of the neck every other day starting 1 day before infection (total of
albicans were used to demonstrate the crucial role of hyphae in the three injections). Animals were divided into groups, with five mice in
disease process and antifungal therapy was implemented as a each group: mono- or coinfected groups with WT or mutant strains
strategy to impede the development of bacterial dissemination. and groups with or without therapy. On the day of infection, mice were
anesthetized by intraperitoneal injections (0.5 ml) of Tribromoetha-
MATERIALS AND METHODS nol (Sigma-Aldrich; 250 mg/kg of body weight). While under anesthe-
Ethics statement. All animal experiments were conducted at the AAA- sia, the animals were placed on heating pads (Braintree Scientific)
LAAC accredited Animal Facility of the University of Maryland in accor- maintained at 37°C. Anesthetized animals were weighed and then
dance with the USA Animal Welfare Act as regulated by the U.S. Depart- orally inoculated by placing calcium alginate swabs (Fisher Scientific)
ment of Agriculture. Animal studies were approved by the University of saturated for 5 min with C. albicans yeast cell suspension sublingually for
Maryland Animal Care and Use Committee (IACUC protocol 0814012). 50 min (Fig. 2A). Animals were placed in a supine position on isothermal
This institution has an Animal Welfare Assurance on file with the Office of pads and monitored until they recovered from anesthesia. Two days after
Laboratory Animal Welfare, National Institutes of Health. The assurance C. albicans infection, the animals were similarly infected with S. aureus cell
number is A-3199-01. suspension; however, in addition, S. aureus was also added to drinking
Strains and growth conditions. The C. albicans wild-type strain water at a final cell density of 106 cells/ml for the duration of experiments.
SC5314 (43), the non-hypha-producing double mutant cph1⌬/efg1⌬ Animals were monitored daily for any developing clinical signs of distress,
strain (HLC54) (44), the tup1⌬ mutant strain (BCa2-10) lacking the and those showing signs of severe disease or loss of significant body weight

February 2015 Volume 83 Number 2 Infection and Immunity iai.asm.org 605


Kong et al.

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


FIG 2 Mouse infection model. (A) Anesthetized mice were infected sublingually using swabs saturated with C. albicans cell suspensions. (B) At 3 days
postinfection with the wild-type strains, white lesions can be seen on the surface of the tongue, and at the day animals are euthanized, advanced oral candidiasis
characterized by white plaques is seen covering the tongue and other oral surfaces (red arrows). (C) A similar clinical picture is seen in animals infected with the
als3 strain (red arrows). (D) No lesions, however, were visible in mice infected with C. albicans receiving fluconazole therapy.

were euthanized. Animals were euthanized 5 days postinfection with C. until they were euthanized, and then the tissue was harvested and pro-
albicans by CO2 inhalation, followed by cervical dislocation, and then cessed as described. Control animals were treated with sterile PBS. Exper-
weighed. The tongues and kidneys were harvested, weighed, homoge- iments were performed to demonstrate lack of any effect for fluconazole
nized, and cultured in triplicate on bacterial and yeast chromogenic media on S. aureus colonization.
CHROMagar (DRG International, Inc.). The plates were incubated for 48 Histopathology of infected tongue tissue. One half of harvested
h at 37°C, and viable counts were enumerated and expressed as the log tongues were fixed and processed for histopathology. In addition, kidneys
CFU/g of tissue. from mice with severe disease were also subjected to histopathology. The
Antifungal therapy. In order to demonstrate that S. aureus does not tissue was embedded in paraffin and sectioned, the sections were depar-
disseminate in the absence of OC, some animals were given daily intra- affinized with xylene and stained with periodic acid-Schiff (PAS) or Gram
peritoneal injections (0.2 ml) of fluconazole (West-Ward Pharmaceutical stain for better visualization of bacteria, and the slides were examined
Corp.; 16 mg/kg of body weight) beginning the day of C. albicans infection using light microscopy. In order to rule out the gastrointestinal tract as a

606 iai.asm.org Infection and Immunity February 2015 Volume 83 Number 2


Oral Candidiasis and Systemic Bacterial Infection

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


FIG 3 Tissue microbial recovery from mono- and coinfected mice with or without fluconazole therapy. At 5 days postinfection with C. albicans, mice were
euthanized, and tissue samples were harvested and assessed for microbial burden. (A) Tongue fungal burden. A high level of C. albicans colonization was seen in
mice infected with the wild-type alone (WT) or in combination with S. aureus (WT⫹SA). In contrast, no C. albicans was recovered from mice infected with the
efg1/cph1 strain with some recovery from mice coinfected with S. aureus. On the other hand, mice infected with the asl3 strain with or without S. aureus recovery
of als3 was comparable to that for the wild type. (B) Recovery of S. aureus from tongues. S. aureus was recovered from the majority of mice infected with S. aureus
alone (SA) or in combination with the efg1/cph1 strain. However, significantly higher level of colonization was noted in mice coinfected with wild-type C. albicans
(WT⫹SA) compared to infection with the efg1/cph1 strain. (C) C. albicans and S. aureus recovery from kidneys of coinfected animals. The microbial presence in
kidneys was assessed as an indication of systemic disease. S. aureus was consistently recovered from the kidneys of all mice coinfected with wild-type strain
(WT⫹SA) concomitant with clinical signs of disease. Similarly, although not consistent, S. aureus was recovered from kidneys of mice coinfected with the als3
strain (als3⫹SA). No organisms were recovered from the kidneys of all other groups, i.e., animals monoinfected or coinfected with C. albicans mutants. (D) C.
albicans and S. aureus recovery from the tongues of fluconazole-treated and untreated animals. No C. albicans was recovered from any of the mice receiving
fluconazole therapy with (CA/SA/F) or without (CA/F) S. aureus. However, S. aureus was recovered from coinfected fluconazole treated (SA/CA/F) mice
although at significantly lower levels than the coinfected group not treated with fluconazole (SA/CA). Five animals were included in each experimental and
control group, and all experiments were performed on at least three separate occasions. Since some groups served as controls for subsequent experiments, higher
number of animals were tested, and therefore more data points were available for some experimental groups. Error bars represent standard error of the means;
P values of ⬍0.05 (*) were considered significant. NS, not significant.

portal of entry for S. aureus dissemination, sections from the small and present the data. Since the experiments were dynamic (systemic infection,
large intestines were also harvested and processed. testing mutant strains, fluconazole therapy, etc.), they were conducted
SEM of infected tongue tissue. For scanning electron microscopy sequentially in phases, and several experimental sessions were performed
(SEM) analysis, tongue tissue was fixed in 2% paraformaldehyde–2.5% to complete the database. Since some groups served as controls for subse-
glutaraldehyde and, after washing steps with PBS, postfixed with 1% quent experimental sets (such as coinfected), the number of animals in
osmium tetroxide, rinsed with PBS, and dehydrated using a series of these groups was substantially higher than a total of 15 mice. Therefore,
washes with ethyl alcohol (30 to 100%). Samples were dried by critical more data points for these groups were included in the analysis.
point drying using an Autosamdri-810 (Tousimi), mounted on alumi- All statistical analysis was performed using GraphPad Prism 5.0 soft-
num stubs, sputter coated with 10 to 20 nm of platinum/palladium, ware. A Kruskal-Wallis one-way analysis of variance test was used to com-
and imaged with a Quanta 200 scanning electron microscope (FEI Co., pare differences between multiple groups, and Dunn’s multiple-compar-
Hillsboro, OR). ison test was used to determine whether the difference between two
Data analysis. The database for statistical analysis was built up by samples was statistically significant. A Student unpaired t test was used to
incremental addition of experimental data. In order to determine the compare differences between two samples (Fig. 3D). P values of ⬍0.05
optimal variables and conditions (infectious doses, endpoint of infection, were considered significant.
etc.) and standardize the model, initial experiments were performed using
small groups of animals. For the main experiments, a total of 11 experi-
mental groups were tested, each including five randomly selected mice for RESULTS
each strain and experimental condition analyzed. All experiments were Tissue microbial burden. To determine whether OC constitutes a
performed on at least three separate occasions, and averages were used to risk factor for the development of secondary bacterial infection, 5

February 2015 Volume 83 Number 2 Infection and Immunity iai.asm.org 607


Kong et al.

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


FIG 4 Histopathology images of tissue sections of tongues from coinfected animals. Tongues were also subjected for histopathology and examined by light
microscopy. (A) Representative image from PAS-stained section of tongues from mice coinfected with S. aureus and wild-type C. albicans demonstrating
extensive presence and penetration of epithelial tissue by the invasive C. albicans hyphae. (B) Upon magnification, massive amounts of S. aureus could be seen
on the outer epithelial layer intertwined with the invading hyphae with marked presence of host inflammatory cells. (C) Tissue sections were also stained with
Gram stain for better visualization of the bacteria. An image of tissue from within the epithelium shows bacterial cells adhering to penetrated hyphae. (D) Image
of Gram-stained kidney tissue sections from coinfected mice with systemic disease showing the presence of S. aureus cells. Arrows: white, hyphae; black, S. aureus.
Bar, 20 ␮m.

days following infection with C. albicans, mice were euthanized, to 25% of loss in body weight was seen concomitant with high
and tissue samples were harvested for assessment of oral coloni- morbidity and S. aureus presence in the kidneys.
zation and systemic infection. Findings demonstrated that all In contrast to the wild-type C. albicans strains, mice infected
mice infected with the wild-type C. albicans strains (SC5314 and with the non-hypha-producing mutant strain (efg1/cph1) did not
SN250) developed clinical OC where, 3 days postinfection, white develop any clinical disease, and no C. albicans was recovered
lesions could be seen which rapidly progressed into overt candi- from the tongues (Fig. 3A). Importantly, no organisms were re-
diasis (white plaques) covering the tongue surface (Fig. 2B) with covered from the kidneys from any of the mice in this group.
high level of C. albicans recovered. In the group coinfected with S. To provide more insight into the mechanism leading to S. au-
aureus and wild-type strains, in addition to OC, animals exhibited reus dissemination, two additional C. albicans mutant strains were
symptoms indicative of severe morbidity, including weight loss, tested in the animal model in order to assess the importance of
hunch back, ruffled fur, and lethargy with a mortality rate of up to hyphal tissue invasiveness for disseminated disease (tup1 mutant
20% and extensive recovery of both species from homogenized strain) and to determine whether the staphylococcal dissemina-
tongues (see Fig. 3A and B). More significantly, S. aureus was tion is contingent upon the presence of the C. albicans hypha-
recovered from the kidneys of all moribund animals, and in some specific Als3 adhesin (als3 mutant strain). As expected, the find-
(30%) C. albicans was also recovered from kidneys. In contrast, ings demonstrated that the tup1 did not colonize the oral cavity,
mice infected with S. aureus or C. albicans individually, although affirming the importance of hyphae not only in causing disease
organisms were recovered from the tongues (Fig. 3B), animals did but also in mediating colonization (data not shown). On the other
not show any signs of systemic disease with no microbial presence hand, animals infected with the als3 mutant developed OC (Fig.
in kidneys. Interestingly, significantly higher level of S. aureus col- 2C), and the recovery of both species from the tongues was com-
onization was seen when C. albicans was present (Fig. 3B). Al- parable to that observed with the parent strain (Fig. 3A and B).
though otherwise healthy, mice infected with C. albicans alone However, in contrast to the parent strain, S. aureus was recovered
exhibited some weight loss, likely due to interference of lesions on from the kidneys of 50% of mice coinfected with S. aureus and the
ability to eat; however, in animals coinfected with both species, up als3 mutant.

608 iai.asm.org Infection and Immunity February 2015 Volume 83 Number 2


Oral Candidiasis and Systemic Bacterial Infection

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


FIG 5 Scanning electron micrographs of outer tongue epithelium from mice with oral candidiasis and systemic infection. (A) Low-magnification (⫻100)
overview image of an excised tongue with advanced OC showing the thick biofilm formed on the surface. (B) Magnified image (⫻2,000) of the surface of tongue
showing the massive matrix consisting of C. albicans hyphae invading the tissue with S. aureus intertwined with the hyphae. (C) Higher-magnification (⫻5,000)
image of the outer surface of the tongue showing the spiny layer of the tongue surface with hyphae penetrating the surface from the sublingual area where C.
albicans was inoculated. (D) Magnified image (⫻10,000) showing a significant gap in the tissue caused by hyphae invading from the sublingual area as it emerges
through the tongue surface.

Fluconazole treatment. Of the mice receiving antifungal ther- analysis of tongue tissue, the als3 strain exhibited invasive ability
apy, those infected with the wild-type strain and administered comparable to that of the wild-type strain (see Fig. S2A in the
fluconazole did not develop OC (Fig. 2D), and no C. albicans was supplemental material). Although S. aureus could be seen within
recovered from tongues with or without S. aureus coinfection (Fig. the damaged epithelium, its association with the invasive hyphae
3D). Similarly, significantly less S. aureus was recovered from the was to a lesser extent than that with the wild-type strain (see Fig.
tongues of the coinfected group treated with fluconazole com- S2B, C, and D in the supplemental material). Histopathology
pared to those not treated (Fig. 3D). Notably, no organisms were analysis of gastrointestinal tissue sections indicated no signs of
recovered from the kidneys of any of the animals receiving flu- hyphal invasion or pathology to the mucosal tissue (data not
conazole. shown).
Histopathology of infected tissue. In addition to assessing mi- SEM of infected tongue tissue. In addition, infected tongues
crobial burden, tissue harvested from animals was also processed were also subjected to SEM analysis. Consistent with histopathol-
for histopathology in order to visualize the tissue invasion process. ogy, SEM analysis revealed a thick matrix covering the whole sur-
Microscopic analysis of PAS-stained (Fig. 4A and B) and Gram- face of the tongue consisting of C. albicans hyphae, S. aureus, and
stained (Fig. 4C) tissue sections revealed extensive fungal adher- host cells (Fig. 5A and B). Upon higher magnification, C. albicans
ence and hyphal penetration of the epithelial tissue with a massive hyphae could be seen penetrating the outer epithelial layer of the
influx of inflammatory cells. Significantly, extensive and deep tongue from the sublingual area where it was inoculated (Fig. 5C),
tongue tissue infiltration of the typically noninvasive S. aureus was leaving gaps in the tissue at sites of emergence (Fig. 5D). In addi-
seen (see Fig. S1 in the supplemental material) and, more impor- tion to the outer surface, tongues were also sectioned prior to
tantly, in mice with severe disease S. aureus was also seen in stained processing in order to visualize the subepithelial tissue (Fig. 6A,
kidney sections (Fig. 4D). Similarly, based on histopathology red box). Analysis of the deep tissue area revealed extensive pres-

February 2015 Volume 83 Number 2 Infection and Immunity iai.asm.org 609


Kong et al.

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


FIG 6 Representative SEM images from within deep tissue of mice with oral candidiasis and systemic infection. (A) Lower-magnification (⫻200) image
demonstrating the massive biofilm matrix formed on the surface of the tongue with sloughing of tissue. (B) Image (⫻5,000) from within the tongue tissue (red
box) showing deep hyphal invasion and the associated presence of S. aureus. (C and D) High-magnification images (⫻8,000 and ⫻10,000, respectively) showing
hyphae penetrating crevices within the tissue, with an overwhelming amount of S. aureus adhering to tissue within the crevices.

ence of both hyphae and S. aureus (Fig. 6B). Importantly, upon to deep tissues and organs. C. albicans and S. aureus are frequent
higher magnification, significant amount of S. aureus was seen colonizers of human mucosal surfaces; using confocal fluorescent
within the tissue in some cases associated with the hyphae (Fig. 6C and atomic force microscopy, our previous in vitro studies re-
and D). Figure 7 is a false-colored image for better visualization of vealed a unique mixed biofilm architecture wherein S. aureus as-
the deep copenetration of C. albicans hyphae and S. aureus in sociated closely with the hyphae (34, 52). However, in order to
coinfected mice. investigate the clinical implications of this interaction during co-
colonization of host mucosal tissue, a suitable animal model is
DISCUSSION needed.
The human mouth with its diverse niches and ample supply of In the present study, at 3 days after oral infection with C. albi-
nutrients is undoubtedly conducive for the unrestricted forma- cans, localized lesions consistent with OC were readily seen on the
tion of natural microbial biofilms, such as those found on the oral tongues rapidly progressing into overt candidiasis. Given the mas-
mucosal tissue, where a multitude of microbial species coexist (10, sive hyphal invasion and deep infiltration of S. aureus observed
51). Mucosal biofilms have been implicated in a wide variety of upon microscopic analysis of tongues, it was not surprising that
infections, and it is therefore imperative that we understand the the mice with OC became moribund within 2 days of exposure to
implications of the interactions between colonizing microbial spe- S. aureus, indicating that S. aureus rapidly gained entry into the
cies as they coexist on host tissue surfaces (7). circulatory system.
Oral candidiasis is perhaps the most common oral mucosal The ability of C. albicans to switch morphology between a yeast
infection particularly in immunocompromised individuals. Al- and hyphal form is central to its pathogenesis (19). Where the
though C. albicans is commonly encountered as a commensal in yeast form is associated with systemic disease the hyphae are adept
healthy individuals, this opportunistic pathogen is capable of in- at causing mucosal infections such as OC and Candida vaginitis,
vading virtually any site of the human host, from superficial sites and therefore mutant strains of C. albicans that do not produce

610 iai.asm.org Infection and Immunity February 2015 Volume 83 Number 2


Oral Candidiasis and Systemic Bacterial Infection

plays a role in S. aureus adherence to the hyphae, the als3 mutant


strain was also used to coinfect a group of mice. Although our in
vitro adherence studies demonstrated diminished S. aureus adher-
ence to the hyphae, in the animal model the level of S. aureus
recovery from tongues was comparable to that with the wild type.
However, S. aureus was only recovered from the kidneys of 50% of
the mice coinfected with the als3 mutant strain. Interestingly, in
our previous study using a different model, animals were only
directly inoculated once with S. aureus simultaneously with C.
albicans to investigate whether Als3p is crucial during the early
stages of colonization prior to development of OC (37). In con-
trast to the findings from the present study, Als3p was found to be
necessary for S. aureus to disseminate (37). Combined, these find-
ings are in line with the hypothesis that the process of C. albicans-
mediated staphylococcal dissemination is multifactorial and OC

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


development prior to bacterial exposure is sufficient to mediate
systemic disease, and although not contingent upon, the process is
likely augmented by the ability of S. aureus to adhere to the inva-
sive hyphae via Als3p.
The azole fluconazole is the most commonly used antifungal
agent for the prevention and treatment of OC (22, 54). Therefore,
FIG 7 False-colored SEM image. In order to better visualize the coinvasion of to further affirm the significant clinical implications of OC, a
C. albicans hyphae and S. aureus into the subepithelial tissue, a captured image group of animals were administered daily fluconazole treatments.
was false colored using Adobe Photoshop. The micrograph shows the invasive In contrast to the untreated group, those receiving therapy did not
hyphae (purple) within the tissue, with infiltrated S. aureus adhering to the
hyphae and the surrounding tissue.
develop OC or systemic disease. Notably, significantly less S. au-
reus was recovered from the treated group, highlighting the im-
portance of enhanced colonization and persistence of biofilm-
associated microbial communities. Our findings are in line with
hyphae are not capable of causing mucosal infections (44, 46, 48). those from a recent study by Roux et al. (55), who, using a rat
The importance of the hypha-mediated tissue damage manifested model, showed that C. albicans airway colonization favors the de-
as OC in the development of secondary bacterial infection was velopment of bacterial pneumonia. Importantly, and similar to
clearly established when a mutant strain of C. albicans unable to our findings, antifungal treatment decreased airway fungal colo-
form hyphae (efg1/cph1) and a strain producing noninvasive hy- nization and in turn the risk of bacterial pneumonia. These find-
phae (tup1) failed to induce staphylococcal infection. Interest- ings are significant since they not only solidify the importance of
ingly, however, although the mutant strain was not efficient at implementing antifungal therapy upon the indication of OC but
colonizing the oral cavity by itself, some recovery was noted in also suggest that antibiotic therapy may be similarly warranted in
coinfected mice, indicating that the presence of S. aureus aided the immunocompromised individuals, including HIV⫹ patients, who
mutant in colonization. Along these lines, it was also interesting are prone to recurrent episodes of OC.
that although the C. albicans was not recovered from the kidneys It is important to note, however, that although individuals with
of any of the monoinfected mice, in some cases, C. albicans dis- immune suppression are the most vulnerable, other mundane fac-
seminated in some coinfected mice. Although it is not clear tors are also considered risk factors for candidiasis; these factors
whether S. aureus contributed to C. albicans dissemination, it is include steroid and antibiotic usage, hormonal imbalances, nutri-
noteworthy that a study by Xu et al. (53) demonstrated that mu- tional deficiency, age, diabetes, metabolic disorders, and denture
cosal commensal bacteria, namely, Streptococcus oralis, modified usage, among others (13, 56, 57). In fact, denture stomatitis stem-
C. albicans virulence, suggesting pathogenic synergy. Whether a ming from the adherence of C. albicans to denture material, fol-
similar process occurs between C. albicans and S. aureus, however, lowed by hyphal infiltration of the denture-associated palatal tis-
remains to be investigated. sue, is prevalent in approximately 70% of denture wearers (58).
The dissemination of S. aureus was clearly via the extensive Therefore, although our model is an immunocompromised
tissue damage caused by the invading hyphae. In fact, in some of model, it is conceivable to speculate that under certain conditions,
our SEM images red blood cells could be seen around the hyphae potentially other individuals with OC could also be predisposed to
and S. aureus as they coinvaded the tissue (data not shown), indi- secondary bacterial infections.
cating that the process of invasion caused injury to the vascular In the case of S. aureus, nasal carriage is a well-defined risk
system, thereby allowing S. aureus to gain entry. Adherence of S. factor of infection with this bacterium, supported by the fact that
aureus to the C. albicans biofilm matrix also likely played a key in most of the cases strains isolated from colonization and infec-
role in the process by enhancing its colonization and persis- tion sites are indistinguishable (59). Therefore, given the demon-
tence on the tissue within the biofilm hyphal matrix as indi- strated propensity of staphylococci to disseminate in our animal
cated by the significantly higher level of S. aureus recovery in the model, it is feasible to speculate that a similar phenomenon may
presence of C. albicans. In fact, in some images of infected tissue, take place in a susceptible human host as these species cocolonize
infiltrating bacterial cells could be seen adhered to the hyphae. the oral and nasal mucosa. It is important to note, however, that
Since we have previously shown that the Als3p hyphal adhesin although we focused here on C. albicans and S. aureus, C. albicans

February 2015 Volume 83 Number 2 Infection and Immunity iai.asm.org 611


Kong et al.

coexists in the oral cavity with more than 700 different bacterial cally important Candida species, p 11–25. In Calderone RA (ed), Candida
species, so it is also conceivable that similar interactions with clin- and candidiasis. ASM Press, Washington, DC.
15. Fidel PLJ. 2011. Candida-host interactions in HIV disease implications
ical relevance may occur between C. albicans and other species.
for oropharyngeal candidiasis. Adv Dent Res 23:45– 49. http://dx.doi.org
As our knowledge of interspecies interactions on mucosal /10.1177/0022034511399284.
niches expands, the role of C. albicans in pathogenesis might prove 16. Pfaller MA, Diekema DJ. 2007. Epidemiology of invasive candidiasis: a
to be more intricate than currently recognized. The identification persistent public health problem. Clin Microbiol Rev 20:133–163. http:
of oral candidiasis as a risk factor for disseminated bacterial dis- //dx.doi.org/10.1128/CMR.00029-06.
17. Perlroth J, Choi B, Spellberg B. 2007. Nosocomial fungal infections:
ease carries serious clinical implications warranting awareness in
epidemiology, diagnosis, and treatment. Med Mycol 45:321–346. http:
terms of therapeutic management, particularly in immunocom- //dx.doi.org/10.1080/13693780701218689.
promised individuals who experience recurrent episodes of oral 18. de Repentigny L, Lewandowski D, Jolicoeur P. 2004. Immunopatho-
candidiasis. genesis of oropharyngeal candidiasis in human immunodeficiency virus
infection. Clin Microbiol Rev 17:729 –759. http://dx.doi.org/10.1128
ACKNOWLEDGMENTS /CMR.17.4.729-759.2004.
19. Saville SP, Lazzell AL, Monteagudo C, Lopez-Ribot JL. 2003. Engineered
This study was supported by the National Institutes of Health grant control of cell morphology in vivo reveals distinct roles for yeast and fila-
DE14424 to M.A.J.-R., by the Interuniversity Attraction Poles Programme mentous forms of Candida albicans during infection. Eukaryot Cell
initiated by the Belgian Science Policy Office (P.V.D. and M.A.J.-R.), by

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


2:1053–1060. http://dx.doi.org/10.1128/EC.2.5.1053-1060.2003.
Flemish Science Foundation (FWO) grant WO.026.11N (P.V.D. and 20. Chauvel M, Nesseir A, Cabral V, Znaidi S, Goyard S, Bachellier-Bassi S,
M.A.J.-R.), and by FWO postdoctoral fellowships awarded to S.K. Firon A, Legrand M, Diogo D, Rossignol T, Naulleau C, d’Enfert C.
We thank Scott Filler for his input and assistance. We also thank Ru- 2012. A Versatile overexpression strategy in the pathogenic yeast Candida
Ching Hsia and the University of Maryland Core Imaging Facility for albicans: identification of regulators of morphogenesis and fitness. PLoS
assistance with electron microscopy, Shariq Khan and Christina Tsui for One 7:e45912. http://dx.doi.org/10.1371/journal.pone.0045912.
their contributions, and Vincent Bruno for providing C. albicans strains. 21. Nett JE, Sanchez H, Andes DR. 2011. Interface of Candida albicans biofilm
matrix-associated drug resistance and cell wall integrity regulation. Eu-
REFERENCES karyot Cell 10:1660 –1669. http://dx.doi.org/10.1128/EC.05126-11.
22. Tournu H, Van Dijck P. 2012. Candida biofilms and the host: models and
1. Brogden KA, Guthmiller JM, Taylor CE. 2005. Human polymicrobial in- new concepts for eradication. Int J Microbiol 2012:845352. http://dx.doi
fections. Lancet 365:253–255. http://dx.doi.org/10.1016/S0140-6736(05) .org/10.1155/2012/845352.
17745-9. 23. Al-Fattani MA, Douglas LJ. 2006. Biofilm matrix of Candida albicans and
2. Jabra-Rizk MA. 2011. Pathogenesis of polymicrobial biofilms. Open My- Candida tropicalis: chemical composition and role in drug resistance. J
col J 5:39 – 43. http://dx.doi.org/10.2174/1874437001105010039. Med Microbiol 55:999 –1008. http://dx.doi.org/10.1099/jmm.0.46569-0.
3. O’Connell HA, Kottkamp GS, Eppelbaum JL, Stubblefield BA, Gilbert 24. Holmes AR, Cannon RD, Jenkinson HF. 1995. Interactions of Candida
SE, Gilbert ES. 2006. Influences of biofilm structure and antibiotic resis- albicans with bacteria and salivary molecules in oral biofilms. J Ind Micro-
tance mechanisms on indirect pathogenicity in a model polymicrobial biol 15:208 –213. http://dx.doi.org/10.1007/BF01569827.
biofilm. Appl Environ Microbiol 72:5013–5019. http://dx.doi.org/10 25. Holmes AR, McNab R, Jenkinson HF. 1996. Candida albicans binding to
.1128/AEM.02474-05. the oral bacterium Streptococcus gordonii involves multiple adhesin-
4. El-Azizi MA, Starks SE, Khardori N. 2004. Interactions of Candida
receptor interactions. Infect Immun 64:4680 – 4685.
albicans with other Candida spp. and bacteria in the biofilms. J Appl Mi-
26. Dongari-Bagtzoglou A, Kashleva H, Dwivedi P, Diaz P, Vasilakos J.
crobiol 96:1067–1073.
2009. Characterization of mucosal Candida albicans biofilms. PLoS One
5. Demuyser L, Jabra-Rizk MA, Van Dijck P. 2014. Microbial cell surface
4:e7967. http://dx.doi.org/10.1371/journal.pone.0007967.
proteins and secreted metabolites involved in multispecies biofilms. Path
27. Klotz SA, Gaur NK, De Armond R, Sheppard D, Khardori N, Edwards
Dis 70:219 –230. http://dx.doi.org/10.1111/2049-632X.12123.
JE, Lipke PN, El-Azizi M. 2007. Candida albicans Als proteins mediate
6. Peters B, Jabra-Rizk MA, O’May GA, Costerton JW, Shirtliff ME. 2012.
Polymicrobial interactions in biofilms: impact on pathogenesis and hu- aggregation with bacteria and yeasts. Med Mycol 45:363–370. http://dx
man disease. Clin Microbiol Rev 25:193–213. http://dx.doi.org/10.1128 .doi.org/10.1080/13693780701299333.
/CMR.00013-11. 28. Rehm SJ. 2008. Staphylococcus aureus: the new adventures of a legendary
7. Brogden KA, Guthmiller JM. 2002. Chapter 21, Polymicrobial diseases: pathogen. Cleveland Clin J Med 75:177–192. http://dx.doi.org/10.3949
current and future research. In Brogden KA, Guthmiller JM (ed), Polymi- /ccjm.75.3.177.
crobial diseases. ASM Press, Washington, DC. 29. Otto M. 2013. Staphylococcal infections: mechanisms of biofilm maturation
8. Jenkinson HF, Douglas JL. 2002. Chapter 18, Interactions between Can- and detachment as critical determinants of pathogenicity. Annu Rev Med
dida species and bacteria in mixed infections. In Brogden KA, Guthmiller 64:175–188. http://dx.doi.org/10.1146/annurev-med-042711-140023.
JM (ed), Polymicrobial diseases. ASM Press, Washington, DC. 30. Thammavongsa V, Missiakas DM, Schneewind O. 2013. Staphylococcus
9. Jenkinson HF, Lala HC, Shepherd MG. 1990. Coaggregation of Strepto- aureus degrades neutrophil extracellular traps to promote immune cell
coccus sanguis and other streptococci with Candida albicans. Infect Immun death. Science 342:863– 866. http://dx.doi.org/10.1126/science.1242255.
58:1429 –1436. 31. Sakoulas G, Moellering RC. 2008. Increasing antibiotic resistance among
10. Jenkinson HF, Lamont RJ. 2005. Oral microbial communities in sickness methicillin-resistant Staphylococcus aureus strains. Clin Infect Dis 46:
and in health. Trends Microbiol 13:589 –595. http://dx.doi.org/10.1016/j S360 –S367. http://dx.doi.org/10.1086/533592.
.tim.2005.09.006. 32. Kraus D, Peschel A. 2008. Staphylococcus aureus evasion of innate anti-
11. Falsetta ML, Klein MI, Colone PM, Scott-Anne K, Gregoire S, Pai C-H, microbial defense. Future Microbiol 3:437– 451. http://dx.doi.org/10
Gonzalez M, Watson G, Krysan DJ, Bowen WH, Koo H. 2014. Symbi- .2217/17460913.3.4.437.
otic relationship between Streptococcus mutans and Candida albicans syn- 33. Gordon RJ, Lowy FD. 2008. Pathogenesis of Methicillin-Resistant Staph-
ergizes the virulence of plaque biofilms in vivo. Infect Immun 82:1968 – ylococcus aureus infection. Clin Infect Dis 46(Suppl 5):S350 –S359. http:
1981. http://dx.doi.org/10.1128/IAI.00087-14. //dx.doi.org/10.1086/533591.
12. Ganguly S, Mitchell AP. 2011. Mucosal biofilms of Candida albicans. 34. Peters B, Ovchinnikova E, Schlecht LM, Hoyer LL, Busscher HJ, van
Curr Opin Microbiol 14:380 –385. http://dx.doi.org/10.1016/j.mib.2011 der Mei HC, Krom BP, Jabra-Rizk MA, Shirtliff ME. 2012. Staphylococ-
.06.001. cus aureus adherence to Candida albicans hyphae is mediated by the hy-
13. Williams DW, Jordan RPC, Wei XQ, Alves CT, Wise MP, Wilson MJ, phal adhesin Als3p. Microbiol 158(Pt 12):2975–2986. http://dx.doi.org
Lewis MAO. 2013. Interactions of Candida albicans with host epithelial /10.1099/mic.0.062109-0.
surfaces. J Oral Microbiol 2013:5. http://dx.doi.org/10.3402/jom.v5i0 35. Peters BM, Jabra-Rizk MA, Scheper MA, Leid JG, Costerton JW,
.22434. Shirtliff ME. 2010. Microbial interactions and differential protein expres-
14. Moran G, Coleman D, Sullivan D. 2012. An introduction to the medi- sion in Staphylococcus aureus and Candida albicans dual-species biofilms.

612 iai.asm.org Infection and Immunity February 2015 Volume 83 Number 2


Oral Candidiasis and Systemic Bacterial Infection

FEMS Immunol Med Microbiol 59:493–503. http://dx.doi.org/10.1111/j 48. Phan QT, Belanger PH, Filler SG. 2000. Role of hyphal formation in
.1574-695X.2010.00710.x. interactions of Candida albicans with endothelial cells. Infect Immun 68:
36. Peters BM, Noverr MC. 2013. Candida albicans-Staphylococcus aureus 3485–3490. http://dx.doi.org/10.1128/IAI.68.6.3485-3490.2000.
polymicrobial peritonitis modulates host innate immunity. Infect Immun 49. Harro JM, Daugherty S, Bruno VM, Jabra-Rizk MA, Rasko DA, Shirtliff
81:2178 –2189. http://dx.doi.org/10.1128/IAI.00265-13. ME. 2013. Draft genome sequence of the methicillin-resistant Staphylo-
37. Schlecht LS, Peters BM, Krom B, Hänsch GM, Filler SG, Jabra-Rizk coccus aureus isolate MRSA-M2. Genome Announc 1:e00037–12. http:
MA, Shirtliff ME. 2014. Systemic Staphylococcus aureus infection medi- //dx.doi.org/10.1128/genomeA.00037-12.
ated by Candida albicans hyphal invasion of mucosal tissue. Microbiology 50. Solis NV, Filler SG. 2012. Mouse model of oropharyngeal candidiasis.
http://dx.doi.org/10.1099/mic.0.083485-0. Nat Protoc 7:637– 642. http://dx.doi.org/10.1038/nprot.2012.011.
38. Timsit JF, Cheval C, Gachot B, Bruneel F, Wolff M, Carlet J, Regnier B. 51. Nasidze I, Quinque D, Li J, Li M, Tang K, Stoneking M. 2009. Com-
2001. Usefulness of a strategy based on bronchoscopy with direct exami- parative analysis of human saliva microbiome diversity by barcoded py-
nation of bronchoalveolar lavage fluid in the initial antibiotic therapy of rosequencing and cloning approaches. Anal Biochem 391:64 – 68. http:
suspected ventilator-associated pneumonia. Intensive Care Med 27:640 – //dx.doi.org/10.1016/j.ab.2009.04.034.
647. http://dx.doi.org/10.1007/s001340000840. 52. Beaussart A, Herman P, El-Kirat-Chatel S, Lipke PN, Kucharıková S,
39. Pate JC, Jones DB, Wilhelmus KR. 2006. Prevalence and spectrum of Van Dijck P, Dufrêne YF. 2013. Single-cell force spectroscopy of the
bacterial coinfection during fungal keratitis. Br J Ophthalmol 90:289 –292. medically important Staphylococcus epidermidis-Candida albicans interac-
http://dx.doi.org/10.1136/bjo.2005.081869. tion. Nanoscale 5:10894 –10900. http://dx.doi.org/10.1039/c3nr03272h.
40. Tawara Y, Honma K, Naito Y. 1996. Methicillin-resistant Staphylococcus 53. Xu H, Sobue T, Thompson A, Xie Z, Poon K, Ricker A, Cervantes J, Diaz
aureus and Candida albicans on denture surfaces. Bull Tokyo Dent Coll

Downloaded from http://iai.asm.org/ on January 15, 2015 by guest


PI, Dongari-Bagtzoglou A. 2014. Streptococcal coinfection augments Can-
37:119 –128.
dida pathogenicity by amplifying the mucosal inflammatory response.
41. Gupta N, Haque A, Mukhopadhyay G, Narayan RP, Prasad R. 2005.
Cell Microbiol 16:214 –231. http://dx.doi.org/10.1111/cmi.12216.
Interactions between bacteria and Candida in the burn wound. Burns
54. Martin MV. 1999. The use of fluconazole and itraconazole in the treat-
31:375–378. http://dx.doi.org/10.1016/j.burns.2004.11.012.
ment of Candida albicans infections: a review. J Antimicrob Chemother
42. Baena-Monroy T, Moreno-Maldonado V, Franco-Martínez F, Aldape-
44:429 – 437. http://dx.doi.org/10.1093/jac/44.4.429.
Barrios B, Quindós G, Sánchez-Vargas LO. 2005. Candida albicans,
Staphylococcus aureus, and Streptococcus mutans colonization in patients 55. Roux D, Gaudry S, Khoy-Ear L, Aloulou M, Phillips-Houlbracq M, Bex
wearing dental prosthesis. Med Oral Patol Oral Circ Bucal 10:E27–E39. J, Skurnik D, Denamur E, Monteiro RC, Dreyfuss D, Ricard JD. 2013.
43. Gillum AM, Tsay EY, Kirsch DR. 1984. Isolation of the Candida albicans Airway fungal colonization compromises the immune system allowing
gene for orotidine =5=-phosphate decarboxylase by complementation of bacterial pneumonia to prevail. Crit Care Med 41:191–199. http://dx.doi
Saccharomyces cerevisiae ura3 and Escherichia coli pyrF mutations. Mol .org/10.1097/CCM.0b013e31828a25d6.
Gen Genet 198:179 –182. http://dx.doi.org/10.1007/BF00328721. 56. Harriott MM, Lilly EA, Rodriguez TE, Fidel PL, Jr, Noverr MC. 2010.
44. Lo H-L, Kohler JR, DiDomenico D, Loebenberg D, Cacciapuoti A, Fink Candida albicans forms biofilms on the vaginal mucosa. Microbiol 156:
GR. 1997. Nonfilamentous Candida albicans mutants are avirulent. Cell 3635–3644. http://dx.doi.org/10.1099/mic.0.039354-0.
90:939 –949. http://dx.doi.org/10.1016/S0092-8674(00)80358-X. 57. Nett JE, Marchillo K, Spiegel CA, Andes DR. 2010. Development and
45. Braun BR, Johnson AD. 1997. Control of filament formation in Candida validation of an in vivo Candida albicans biofilm denture model. Infect
albicans by the transcriptional repressor TUP1. Science 277:105–109. http: Immun 78:3650 –3659. http://dx.doi.org/10.1128/IAI.00480-10.
//dx.doi.org/10.1126/science.277.5322.105. 58. Salerno C, Pascale M, Contaldo M, Esposito V, Busciolano M, Guida A,
46. Braun BR, Head WS, Wang MX, Johnson AD. 2000. Identification and Milillo L, Petruzzi M, Serpico R. 2011. Candida-associated denture
characterization of TUP1-regulated genes in Candida albicans. Genetics stomatitis. Med Oral Patol Oral Cir Bucal 16:139 –143.
156:31– 44. 59. Edwards JS, Betts L, Frazier ML, Pollet RM, Kwong SM, Walton WG,
47. Phan QT, Myers CL, Fu F, Sheppard DC, Yeaman MR, Welch WH, Ballentine WK, Huang JJ, Habibi S, Del Campo M, Meier JL, Dervan
Ibrahim AS, Edwards JE, Filler SG. 2007. Als3 is a Candida albicans PB, Firth N, Redinbo MR. 2013. Molecular basis of antibiotic multire-
invasin that binds to cadherins and induces endocytosis by host cells. PLoS sistance transfer in Staphylococcus aureus. Proc Natl Acad Sci U S A 110:
Biol 5:e64. http://dx.doi.org/10.1371/journal.pbio.0050064. 2804 –2809. http://dx.doi.org/10.1073/pnas.1219701110.

February 2015 Volume 83 Number 2 Infection and Immunity iai.asm.org 613

View publication stats

Vous aimerez peut-être aussi