Você está na página 1de 15

review special focus REVIEW: biofilms

Virulence 2:5, 445-459; September/October 2011; 2011 Landes Bioscience

Staphylococcus aureus biofilms


Properties, regulation and roles in human disease
Nathan K. Archer,1,2 Mark J. Mazaitis,3 J. William Costerton,4 Jeff G. Leid,5 Mary Elizabeth Powers6 and Mark E. Shirtliff1,7,*
1
Department of Microbial Pathogenesis; Dental School; 2Graduate Program in Life Sciences, Microbiology and Immunology Program; 3Institute for Genome Sciences;
University of Maryland; Baltimore, MD USA 4Center for Genomic Sciences; Allegheny-Singer Research Institute; Pittsburgh, PA USA; 5Department of Biological Sciences;
Northern Arizona University; Flagstaff, AZ USA; 6University of Waterloo; Waterloo, ON Canada; 7Department of Microbiology and Immunology; Medical School;
University of Maryland; Baltimore, MD USA

Key words: Staphylococcus aureus, biofilms, MRSA, therapy, vaccine, host response, pathogenesis

Increasing attention has been focused on understanding Although a biofilm can arise from a single cell, differential
bacterial biofilms and this growth modalitys relation environmental conditions throughout the community can poten-
to human disease. In this review we explore the genetic tiate the development of distinct subpopulations. Gradients in
regulation and molecular components involved in biofilm oxygen, nutrients and electron acceptors can cause heterogeneous
formation and maturation in the context of the Gram-positive gene expression throughout a biofilm.4 In a staphylococci in vitro
cocci, Staphylococcus aureus. In addition, we discuss diseases colony biofilm model, four distinct metabolic states were identi-
and host immune responses, along with current therapies fied: cells growing aerobically, fermentatively, dormant (including
associated with S. aureus biofilm infections and prevention very slow growing cells and persisters), or dead. The cells exposed
strategies. to the upper air oxygen-rich interface and lower liquid-nutrient-
rich interface were metabolically active.4 However, the majority
of cells were dormant and located in an anoxic environment.4 In
Staphylococcus aureus biofilm mode of growth is tightly regu- addition, heterogeneity of biofilm protein expression was demon-
lated by complex genetic factors. Host immune responses against strated with multiple cell wall-associated proteins. Expression was
persistent biofilm infections are largely ineffective and lead to shown to vary in cell clusters throughout the biofilm, and in one
chronic disease. However, current research has taken biofilm case, differential expression was visualized on a cell-cell basis.5
formation into account in terms of elucidating host immunity By adopting this sessile mode of life, biofilm-embedded
toward infection, and may lead to the development of efficacious microorganisms benefit from a number of advantages over their
anti-biofilm S. aureus therapies. planktonic counterparts. The extracellular matrix is capable of
sequestering and concentrating environmental nutrients such as
Biofilms carbon, nitrogen and phosphate.6 An additional benefit to the
biofilm growth modality is the ability to evade multiple clearance
A biofilm can be defined as a microbially-derived sessile commu- mechanisms produced by host and synthetic sources. Examples
nity, typified by cells that are attached to a substratum, interface, of ineffective clearance strategies include antimicrobial and anti-
or to each other, are embedded in a matrix of extracellular poly- fouling agents, shear stress, host phagocytic elimination and host
meric substance, and exhibit an altered phenotype with regard to radical and protease defenses. Resistance to antimicrobial factors
growth, gene expression and protein production.1 Biofilm thick- is mediated through a dormant phenotype caused by adaptation
ness can range from a single cell layer to a substantial community to an anoxic environment and nutrient deprivation, and results
encased by a viscous polymeric milieu.2 Structural analyses have in low metabolic levels and radically downregulated rates of cell
shown that in some cases unique pillar or mushroom-shaped division.7 This stressed environment produces many slow grow-
structures can be formed by the micro-colony architecture of ing cells that are tolerant to high levels of antibiotics but also a
these dense biofilms; however, other structures do form depend- proportion of persister cells (no more than ~1% of total popu-
ing on the environmental conditions.2 Intricate channel net- lation).7 These cells are found in biofilm communities but also
works flow through these complex structures and provide some exist in planktonic cultures.8 In addition, persister cells dem-
accessibility to essential nutrients even in the deepest regions of onstrate multidrug tolerance that is inherently different from
the biofilm. Although biofilm formation is not a prerequisite resistance, which is prevention of the antibiotic from hitting its
for persistent infection,3 biofilm eradication is arduous, usually microbial target.7,8 Instead, persister tolerance is accomplished by
requiring surgical intervention and therefore warrants further shutting down the microbial targets or the cellular need for those
investigation. targets by maintaining a metabolically quiescent state, thereby
protecting the cell from even bactericidal antibiotics.7 Although
*Correspondence to: Mark E. Shirtliff; Email: mshirtliff@umaryland.edu the mechanism for achieving this metabolic state in S. aureus
Submitted: 06/04/11; Revised: 08/10/11; Accepted: 08/11/11 is not completely understood, in E. coli it has been implicated
http://dx.doi.org/10.4161/viru.2.5.17724
that it is accomplished through a certain proportion of cells in

www.landesbioscience.com Virulence 445


Figure 1. IgGs against recombinant forms of cell wall-associated biofilm proteins bind to intact S. aureus biofilms. IgG against each selected candi-
date protein was applied followed by the secondary goat anti-rabbit F(ab)2 antibody (upper right in AD). After a washing step, SYTO 9 was applied
to stain all bacterial cells (upper left in AD). Biofilms were probed with (A) anti-SA0486 (autolysin) IgG and secondary antibody, (B) anti-SA0486
(lipoprotein) IgG and secondary antibody, (C) anti-SA0688 (lipoprotein) IgG and secondary antibody and (D) anti-SA0037 (conserved hypothetical) IgG
and secondary antibody. The merged image is shown in the lower part in (AD). The base of the glass is located at the bottom of each image, and each
image is a cross-sectional view of the biofilm from the base into the lumen.

the total microbial population having drastically downregulated nares. In the human population, approximately 2025% have
biosynthetic pathways and toxin/antitoxin production, thereby become persistently colonized and 7580% intermittently or
producing a persister phenotype in this population subset.7 By never colonized.15,16 Previous studies have shown that there is a
protecting these cells within the biofilm, effectors of the immune strong causal connection between S. aureus nasal carriage and
system are prevented from clearing out these populations.9 increased risk of nosocomial infection. Nasal carriage provides a
Therefore, once antibiotic regimens are halted, these persisters staging ground for S. aureus to disseminate to other areas of the
are able to spontaneously shift out of their quiescent state and body where, once transmitted to the circulatory system through
produce a reactivation of infection.9 an epithelial breach, planktonic growth and upregulation of
While low metabolic rates may explain a great deal of the anti- adherence factors occurs.17,18 Invading staphylococci are then
microbial resistance properties of biofilms, other factors may play either removed by the host innate immune response or attach to
a role. One such feature may be the capability of biofilms to act host extracellular matrix proteins and form a biofilm. The cel-
as a diffusion barrier to slow down the infiltration of some anti- lular physiology is then quickly transformed into one reflective of
microbial agents.10 For example, reactive chlorine species (such a biofilm. Owing to the escalating involvement of Staphylococcus
as hypochlorite, chloramines or chlorine dioxide) in a number of aureus in foreign body-related infections, the swift development
antimicrobial/antifouling agents may be deactivated in the sur- and exhibition of multiple-antibiotic resistance, and their predi-
face layers of the biofilm before they are able to disseminate into lection to transform from an acute infection to one that is per-
the lower layers.11 In addition, a recent study showed that several sistent, chronic and recurrent, this pathogen continues to receive
antibiotics (oxacillin, cefotaxime and vancomycin) had reduced considerable attention.
penetration throughout S. aureus and S. epidermidis biofilms.12 Staphylococcus aureus can produce a multilayered biofilm
The final benefit to the biofilm development is the potential embedded within a glycocalyx or slime layer with heteroge-
for seeding dispersal or cellular detachment. Micro-colonies may neous protein expression throughout (see Fig. 1). Early studies
detach under the direction of fluid mechanical shear forces or described the solid component of the glycocalyx as primarily
through a genetically programmed response that mediates the composed of teichoic acids (80%) and staphylococcal and host
seeding dispersal process.13 In a similar fashion to a metastatic proteins (see Fig. 2).19 In later studies, a specific polysaccharide
cancer cell, detached micro-colonies migrate from the original antigen named polysaccharide intercellular antigen (PIA) was
biofilm community to uninfected regions of the host, attach and isolated. PIA is composed of b-1,6-linked N-acetylglucosamine
promote nascent biofilm formation. In addition, while not the residues (8085%) and an anionic fraction with a lower con-
case for non-motile bacteria like S. aureus, seeding dispersal can tent of non-N-acetylated D-glucosaminyl residues that con-
also be mediated by the movement of single, motile cells from tains phosphate and ester-linked succinate (1520%).20
the adherent micro-colony.14 Therefore, this advantage allows an PIA-dependent biofilm formation. PIA is produced in vitro
enduring bacterial source population that is resilient against anti- from UDP-N-acetylglucosamine via products of the intercellular
microbial agents and the host immune response, while simultane- adhesion (ica) locus.21 The genes and products of the ica locus
ously enabling continuous shedding to encourage bacterial spread. [icaR (regulatory) and icaADBC (biosynthetic) genes] have been
demonstrated to be necessary for biofilm formation and viru-
Staphylococcus aureus Biofilms lence and are upregulated in response to anaerobic growth, such
as the conditions seen in the biofilm environment.22 The staphy-
Staphylococcus aureus is a Gram-positive, ubiquitous bacterial spe- lococcal respiratory response regulator, SrrAB, is responsible for
cies. The ecological niche of S. aureus in humans is the anterior PIA induction under anaerobic environments via binding of a

446 Virulence Volume 2 Issue 5


100 bp DNA sequence upstream of the icaADBC operon.23
Other environmental factors can also play a role in regulation of
ica, including glucose, ethanol, osmolarity, temperature and anti-
biotics such as tetracycline.18 In the homologous Staphylococcus
epidermidis locus, regulation of ica can occur via reversible inacti-
vation by insertion sequence (IS256) phase variation in 2533%
of variants,24 and this has been observed in some S. aureus strains
as well.25 In addition, PIA expression is repressed by TcaR, a tran-
scriptional regulator of the teicoplanin-associated locus; however,
deletion of the tcaR gene had no effect on PIA synthesis.26 Strong
negative regulation is conferred by IcaR, through binding of
the ica cluster promoter and deletion of the icaR gene results in
enhanced ica cluster gene expression.26,27 The protein regulator of
biofilm formation, Rbf, however, represses transcription of IcaR,
albeit indirectly, leading to augmented ica gene expression, PIA
production and biofilm formation.28 In addition, Spx, a global
regulator of stress response genes, was shown to have a negative Figure 2. SEM micrograph from an implant recovered at five days
showing massive numbers of cocci partially occluded by dehydrated
regulatory impact on biofilm formation, seemingly by modulat-
material. Bar = 5 m.
ing IcaR.29
PIA-independent biofilm formation. Despite the impor-
tance of the ica gene locus in biofilm development, biofilms adhesion can substitute PIA-mediated biofilm development in
can occur in an ica-independent fashion. The arlRS two-com- ica-independent biofilms.
ponent system was shown to repress biofilm development, and eDNA and biofilm formation. Another important component
when deleted led to enhanced attachment and PIA production.30 of the staphylococcal biofilm is extracellular DNA (eDNA). The
However, biofilm synthesis was unaffected by additional dele- discovery that this substance is an important component of bio-
tion of the icaADBC operon, suggesting that in this double dele- films was previously made in P. aeruginosa.39-41 This is supported
tion mutant, PIA was not essential for biofilm development.30 by the in vitro finding that early, immature biofilm clearance
The S. aureus clinical isolate, UAMS-1 (University of Arkansas could be attained by DNase treatment.39 In addition, support of
Medical System-1), had unabated biofilm formation in vitro and the importance of eDNA in biofilms in vivo includes the success-
in vivo in a catheter infection model even with mutation of the ful clinical usage of DNase concurrently with antibiotic therapy
ica cluster.17 In a guinea pig model of biofilm infection, dele- in the treatment of cystic fibrosis patients,42 and the finding that
tion of ica and thus, lack of PIA production caused no decrease DNase found on skin cells can lessen biofilm formation.43 Rice et
in virulence.31 In addition, Fitzpatrick et al.32 showed that bio- al.44 discovered that mutation of cidA, a gene encoding an effec-
film formation in MRSA strain BH1CC was unaffected by ica tor of murein hydrolase activity and regulating cell death, led to
locus deletion. However, other mutant strains lost the ability to a less adherent biofilm that contained lower levels of genomic
form biofilm. Interestingly, when S. aureus icaADBC operon- DNA. In addition, treatment with DNase I minimally affected
deletion mutants are categorized by methicillin susceptibility, biofilm formation in the cidA mutant, whereas the parental strain
MRSA strains are capable of biofilm development, whereas had a dramatic inhibition. The cidA gene has been shown to be a
MSSA strains are impaired in biofilm formation.33 These data holin homolog involved in cell lysis, and these data suggest that
propose that biofilm formation in an ica-independent manner this cell lysis activity is necessary for DNA release and biofilm
is strain specific. In an ica-deletion mutant S. aureus strain, pro- development.44 Cell lysis and subsequent genomic DNA release
tein A (SpA) production was found essential for biofilm forma- must also occur early in cell attachment for proper biofilm for-
tion.34 Furthermore, biofilm development could be recovered in mation, as shown by Mann et al.45 This cell lysis and the biofilm
spa mutants by addition of exogenous SpA, indicating that it matrix interspersed between whole intact bacterial cells can be
is not necessary for SpA to be covalently anchored to the cell readily seen in Figures 3 and 4. The lrg gene, another regula-
wall.34 The fibronectin-binding proteins (FnBPs) can also arbi- tor of murein hydrolase and cell death, counteracts the effects of
trate biofilm formation through an essential role by the major cidA DNA release as revealed by enhanced biofilm attachment
autolysin (Atl) and sigB regulation,35 and in Staphylococcus epi- and matrix-associated eDNA in lrgAB mutants.45 Other globally
dermidis, PIA-independent biofilms were mediated through the upregulated genes in S. aureus biofilms included autolysin, an
accumulation-associated protein (Aap).36,37 In addition, biofilm- enzyme that hydrolyzes staphylococcal cells, and phage genes due
associated protein (Bap) and Bap-related proteins of S. aureus to the change from a lysogenic to lytic form in the stressed envi-
can confer biofilm development independent of PIA production ronment of the biofilm.46,47 Therefore, S. aureus has a number of
through cell-to-cell aggregation, and are characterized by their modalities to add eDNA into the early biofilm matrix, thereby
high-molecular weight, presence on the bacterial surface, role providing the structural support for biofilm formation. Control
as a virulence factor and occasional containment in mobile ele- of cell lysis and eDNA release is another branch in the complex
ments.38 These reports suggest that proteinaceous cell-to-cell regulation of biofilm formation by S. aureus.

www.landesbioscience.com Virulence 447


system has been shown to downregulate genes of cell wall-associ-
ated adherence factors.54 This would lead to lesser adherence and
thus, indirectly, decreased initial biofilm formation. Repression
of agr has been shown to be necessary for biofilm formation and
induction of agr through auto-inducing peptides (AIP), an essen-
tial component of the quorum sensing system, results in seeding
dispersal in mature biofilms.55 This mechanism occurs in a pro-
tease-dependent manner and requires an active agr system since
both protease inhibitors and inhibitory AIP block seeding disper-
sal.55,56 As well, agr has been shown to upregulate the expression
of detergent-like peptides and nucleases that seem to increase bio-
film detachment.53,57 Therefore, the staphylococcal global regula-
tors function in a complementary and opposing fashion where
sarA induces attachment and early biofilm formation conceivably
by repressing nucleolytic and proteolytic extracellular enzymes,
and once biofilms have attained a significant quorum population,
agr expression leads to upregulation of a number of virulence
factors to thwart the host immune response and supports seed-
ing dispersal in a nuclease, protease and surfactant-dependent
manner.
Biofilm regulation by sigB. Additional levels of con-
trol are accomplished through the sigB operon product B in
Staphylococcus aureus (regulated by operon genes rsbU, rsbW and
rsbV ).58 Factors necessary for the early stages of biofilm forma-
tion, including clumping factor, fibronectin binding protein A
(FnbpA) and coagulase, are all upregulated by B.59,60 In addi-
tion, factors that correlate with seeding dispersal and a plank-
tonic phenotype, including -hemolysin, enterotoxin B, serine
Figure 3. TEM micrograph of a cross-section through a biofilm recov- protease (SplA), cysteine protease (SplB), the metalloprotease
ered from an indwelling medical device implanted in the peritoneum Aur, staphopain and leukotoxin D are all negatively controlled.61
of a rat for five days. Bacteria are apparent on the surface of the biofilm
and interspersed with host material and lysed bacteria within the
Rachid et al. revealed that a sigB-deficient S. aureus could not
biofilm. Bar = 5 m. form biofilm and resulted in upregulation of RNAIII, a vital
component of the anti-biofilm promoting agr system.62 This is
exemplified in the case of the laboratory strain RN6390, which
Biofilm regulation by agr/sarA. The major global regulators, lacks B and is deficient in biofilm formation.63 Additional dele-
staphylococcal accessory regulator (sarA) and accessory gene reg- tion of agr or use of protease inhibitors reverted S. aureus to a
ulator (agr), have also been implicated in biofilm formation. A biofilm-positive phenotype.63 Jefferson et al.27 discovered that
two component regulatory gene locus encoded by arlRS, a mem- IcaR binds to a 42-bp region just upstream of icaA and hypoth-
ber of the OmpR-PhoB family of response regulators, is regu- esize that its role is to sterically hinder the binding of B, thus
lated by the agr and sarA loci.48 When upregulated, the product preventing activation of the ica locus. In contrast, Valle et al.49
of arlS prevents biofilm formation and may mediate attachment found that a sigB deletion mutant still effectively developed bio-
to polymer surfaces by affecting peptidoglycan hydrolase activity. film. These conflicting reports suggest that the sigB operon may
Transcripts of sarA are upregulated in biofilms when compared act in a strain-specific manner in controlling biofilm formation.
with planktonic cultures.49 In addition, capacity to form biofilm Transcriptomic and proteomic expression results in bio-
is reduced in sarA mutants.50 Biofilm formation can be recov- films. Based on studies from other researchers and our own
ered in sarA mutants by concurrently mutating the gene encoding laboratory, we have found a staged progression of virulence gene
the S. aureus thermostable nuclease, nuc or addition of protease expression in the biofilm under control of the sigB, agr, sarA and
inhibitors, as nuc and extracellular proteases were found to be sae regulons.17,46,64 In the planktonic mode of growth, the micro-
transcriptionally upregulated in sarA mutants.51 By inhibiting bial surface components recognizing adhesive matrix molecules
nuc expression or protease activity, sarA expression may prevent (MSCRAMMS) are upregulated while the staphylococci are in
degradation of extracellular DNA and protein, important biofilm a low concentration such as that seen in a case of early sepsis.64
structural components. Although SarA affects and is affected by Once an early biofilm develops, transient upregulation of sarA
agr, the effect of agr in vitro mutants on biofilm development and agr systems downregulate the adhesins and upregulates a
is minimal and has been shown to be an agr-independent path- number of immunoavoidance factors and toxins that cause dam-
way.49,50,52,53 Despite this, there is evidence that the agr locus is age to the host.65 In a fully mature biofilm, staphylococci are slow
associated with biofilm development. The agr quorum sensing growing and nutrient deprived resulting in the downregulation

448 Virulence Volume 2 Issue 5


Figure 4. TEM micrograph depicts bacteria within a medical implant biofilm. The bacteria are associated with large amounts of host extracellular
matrix materials and lysed bacteria. Fibrils are apparent radiating from the bacterial cell surface in a clear halo surrounding the bacteria (arrow).
Bar = 1 m.

in virulence factor production and the upregulation of stress Osteomyelitis. Osteomyelitis is defined as an infection of
response genes, phages genes and systems for maintaining pH the bone, and can be caused by a variety of bacteria or fungi.
homeostasis.64 The implications of these various gene expression However, S. aureus provides the majority of cases.69 Bacteria can
changes can only be understood in the context of the human be introduced through a hematogenous route or direct inocula-
host and will be explored more thoroughly below in the dis- tion during surgery, trauma or an overlying infection.70 While in
cussion of one of the well studied diseases caused by S. aureus: the blood in the planktonic mode of growth, S. aureus upregu-
osteomyelitis. lates the production of adhesins for many host adhesive matrix
For a summary diagram of the regulatory factors involved in molecules (e.g., fibrinogen, fibrin, osteopontin, fibronectin, colla-
biofilm formation, maintenance and detachment, please refer to gen, elastin, etc.).64 Once in the bone, S. aureus attaches to local-
Figure 5. ized areas of trauma and divides by binary fission to form an early
biofilm (see Fig. 6) while constitutively expressing a low level of
S. aureus Biofilm-Related Diseases the auto-inducing peptide (AIP) of the quorum sensing system.70
As the population expands and the AIP levels increase, a quorum
Staphylococcus aureus has re-emerged as a clinically relevant is attained and the agr quorum sensing system is activated, result-
pathogen due to its resistance to antibiotics and the increased ing in the production of toxins, immunoavoidance factors and
use of indwelling medical devices. Infections associated with other agr-controlled virulence factors with a concomitant down-
S. aureus in the US have a crude mortality rate of 25% along regulation in microbial adhesins.65 Simultaneously, a number
with hospitalizations resulting in approximately twice the length of microbial inflammatory mediators such as formylmethionyl
of stay, deaths and medical costs of typical hospitalizations.66,67 peptides, low level hemolysins, lipotechoic acids, peptidogly-
S. aureus biofilms, once established, are recalcitrant to antimi- can, phenol soluble modulins and staphylococcal DNA (due to
crobial treatment and the host response, and therefore are the unmethylated cytosine-phosphate-guanosine [CpG] motifs) call
etiological agent of many recurrent infections.68 Following is an the innate inflammatory response effectors (e.g., PMNs) to bear
incomplete list of S. aureus diseases that have a demonstrated bio- on the invading pathogen.71 However, PMNs attempt to attack
film component. Osteomyelitis will be first discussed in detail the virulent early biofilm in a non-specific way and are met with
in the context of S. aureus stages of colonization, infection and an armamentarium of staphylococcal toxins and immunoavoid-
maturation of the biofilm as this progression of disease is mim- ance factors resulting in host cell lysis and frustrated phagocyto-
icked in many other diseases and host sites. sis.71 The end result is a maturing S. aureus biofilm population

www.landesbioscience.com Virulence 449


Figure 5. For figure legend, see page 451.

with detaching flocs and tissue damage due to PMN-induced col- that the biofilm and devitalized tissue must be surgically debrided
lateral host tissue damage, thereby providing the essential devi- in order resolve the infection.69 However, in cases of juvenile
talized surface for further biofilm development (see Fig. 7 and osteomyelitis, children are often able to resorb the dead bone and
Movie S1).70,72 In adults, the resulting tissue destruction means tissue during antimicrobial therapy, resulting in the elimination

450 Virulence Volume 2 Issue 5


Figure 5 (See opposite page). Flowchart of regulatory factors involved in S. aureus biofilm formation, maintenance and detachment. (A) PIA-de-
pendent biofilm formationexpression of the icaADBC gene cluster results in PIA expression and biofilm formation.21 Expression of icaADBC can be
suppressed by production of tcaR and icaR, resulting in downregulation of PIA and thus biofilm formation. 26,27 In the case of the icaR gene, expression
can be up or downregulated by the proteins Spx and Rbf, respectively. 28,29 Consequently, Spx induction of icaR expression results in downregulation
of icaADBC expression, PIA production and biofilm formation.29 Conversely, Rbf inhibits icaR expression leading to upregulation of icaADBC expres-
sion, PIA production and biofilm formation.28 Additionally, anaerobic conditions induce production of SrrAB, causing expression of the icaADBC gene
cluster, PIA production and biofilm formation.23 PIA-independent biofilm formation*in ica-deletion mutants, PIA-independent biofilm formation can
be mediated through cell wall-associated protein cell-to-cell adhesion (MRSA-specific). 33 Examples of proteins arbitrating biofilm formation include
SpA, FnBPs and Bap. 34,35,38 icaADBC, intercellular adhesion biosynthetic genes; PIA, polysaccharide intercellular antigen; tcaR, transcriptional regulator
of the teicoplanin-associated locus; icaR, intercellular adhesion regulatory gene; Spx, global regulator of stress response genes; Rbf, protein regulator
of biofilm formation; SrrAB, staphylococcal respiratory response regulator; SpA, S. aureus protein A; FnBPs, fibronectin-binding proteins; Bap, biofilm-
associated protein. (B) eDNA and biofilm formationeDNA leads to enhanced biofilm formation.39-41 DNase treatment degrades eDNA and inhibits
eDNA-mediated biofilm formation. 39,42,43 DNA release is arbitrated through cell lysis and controlled by lrg and cidA gene expression.44,45 Upregulation
of the lrg gene results in inhibition of cellular lysis, DNA release and biofilm formation.45 Conversely, cidA gene expression enhances cellular lysis, DNA
release and biofilm formation.44 eDNA, extracellular DNA; lrg, regulator of murein hydrolase and cell death; cidA, regulator of murein hydrolase and
cell death. (C) agr/sarA/sigB regulationexpression of the sarA gene results in downregulation of proteases and the thermostable nuclease, allowing
for development of an immature biofilm. 51 Expression of sigB similarly downregulates protease production, but additionally promotes expression of
adherence factors that aid in initial biofilm formation.59-61 The immature biofilm increases in cell density until a mature biofilm develops. At this stage,
the density of AIPs throughout the bacterial community reaches a quorum sensing threshold and induces expression of the agr gene. 55 Induction of
agr results in upregulation of detergent-like peptide, protease and thermostable nuclease expression; leading to release of bacterial cells from the
mature biofilm, termed seeding dispersal. 53,55-57 sarA, staphylococcal accessory regulator; sigB, sigmaB; AIP, auto-inducing peptides; agr, accessory gene
regulator.

of the bacterial attachment surface. Therefore, this type of bio-


film infection in juveniles is one of the few types of biofilm infec-
tions that can be successfully resolved with antibiotic therapy
alone.69 This mode of infection is much like other modes of
S. aureus chronic infection such as those discussed below.
Indwelling medical device infection. Millions of indwelling
medical devices are implanted into hosts every year and S. aureus
is the major culprit for their infection and failure.73 S. aureus
has been known to infect and form a chronic biofilm infection
most often on orthopedic implants including prosthetic joints,
wires, pins, external fixators, plates, screws, nails and mini-large
fragment implants.74 However, other indwelling medical devices
that are prone to staphylococcal biofilm infection include stents,
ventilators, intravenous catheters, invasive blood pressure units,
infusion pumps, cardiac defibrillators, mechanical heart valves,
aspirators, pacemakers, stitch materials, ear and central nervous
system shunts, cosmetic surgical implants, penile implants and
orthopedic devices.74 During cases of implant infection, the
implant becomes coated with host derived extracellular matrix
proteins, providing a rich surface for bacterial attachment.75
Whether infected during implantation, during subsequent
trauma or hematogenously, the result is the same. In order to
resolve the infection, the device must be removed to completely
resolve the infection since antibiotics cannot eliminate this bio-
film population.
Periodontitis and peri-implantitis. Periodontitis is defined
as inflammation and infection of the ligaments and bones
that support the teeth and peri-implantitis as destructive
inflammation of soft and hard tissues surrounding a den-
tal implant.76 S. aureus biofilm formation in the oral cavity
can act as a reservoir for oral infections, and the prevalence of
S. aureus in the periodontal pocket is 13.4% and 15.8% in Figure 6. SEM micrograph demonstrating cocci adherent to a bone
the oral cavity.77 S. aureus has a high affinity to titanium sur- sequestra. Collapsed glycocalyx material is apparent associated with
faces and is recovered at high levels from infected peri-implant the bacterial biofilm (arrow). Bar = 5 m.
pockets. Although this pathogen is not strongly associated with

www.landesbioscience.com Virulence 451


Figure 7. (A) Nasal colonization confers Staphylococcus aureus a reservoir for dissemination onto skin and various other host epithelial surfaces. Dam-
age to the epithelia can occur from extraneous sources. (B) Damage allows S. aureus to breach the epithelial layer and bind to host matrix via surface
expressed colonization factors. Map, MHC class II analog protein; Cna, collagen-binding adhesin; FnbB, fibronectin binding protein B; FnbA, fibronec-
tin binding protein A; Fib, fibrinogen binding protein; FbpA, fibronectin binding protein; CflA, clumping factor A; EbpS, elastin binding protein. (C) Ini-
tial attachment and cell division produces an early S. aureus biofilm. The quorum sensing compound of S. aureus, termed auto-inducing peptide (AIP),
is secreted and taken up by the microcolony. SarA expression is upregulated, leading to production of virulence and immunoavoidance factors (CHIPS,
chemotaxis inhibitory protein of staphylococci; Eap, extracellular adherence protein; SCIN, staphylococcal complement inhibitor). Additionally, AIP has
reached quorum threshold levels, resulting in agr activation and a downregulation of adhesins and an upregulation of virulence factor expression that
cause damage to the host and evade the immune response. The nascent S. aureus biofilm avoids immunological destruction by initiating leukocyte
apoptosis and/or developing a protective environment via inflammation and tissue damage. (D) Mature S. aureus biofilm is encapsulated by polysac-
charide intercellular antigen (PIA), protein and extracellular DNA (eDNA). AIP has reached quorum threshold levels, resulting in agr gene expression.
Subsequently, protease and detergent-like peptides are secreted into the biofilm and promote seeding dispersal. Large biofilm aggregates detach
forming flocs and planktonic bacterial cells migrate from the mature biofilm into the circulatory system, where adherence to distal tissue and forma-
tion of a nascent biofilm can occur to repeat the cycle.

periodontitis, it is associated with the more difficult-to-treat techniques. Diabetic foot wound patients with S. aureus wound
refractory cases.76 colonization have a 2-fold increase in healing time.81 Similarly, a
Chronic wound infection. Staphylococcus aureus biofilms have murine cutaneous wound model demonstrated that S. aureus bio-
been implicated in cases of chronic wound infections such as dia- film delays re-epithelialization and healing.82 In addition, delayed
betic foot ulcers, venous stasis ulcers and pressure sores. In fact, re-epithelialization was shown to be specifically dependent on
S. aureus is the most commonly isolated bacteria from such wound S. aureus biofilm development.82 However, with modern molec-
infections and studies involving patients with chronic venous leg ular and culture independent techniques, this prevalence may
ulcers found S. aureus positive cultures in 8893.5% of infec- change in the future.
tions.78,79 Davis et al.80 observed S. aureus biofilm structures in Chronic rhinosinusitis. Chronic rhinosinusitis (CRS) is an
a porcine wound colonization model using multiple microscopic inflammatory disorder with an unknown etiopathogenesis.83

452 Virulence Volume 2 Issue 5


Bacterial biofilms have been repeatedly demonstrated on the This early inflammatory response may be ineffective at
mucosal surface of CRS patients and correlate with more severe microbial clearance since the host produces IgG subclasses of
disease.84,85 Staphylococcus aureus is recovered from 50% of CRS IgG2a and IgG2b that are very effective at fixing complement to
cases, and more interestingly, is associated with more severe, sur- which Gram positive bacteria like staphylococci are inherently
gically recalcitrant infection and unfavorable post-operative evo- resistant.93 Also, S. aureus possesses a staphylococcal comple-
lution.83,86 Despite the connections between biofilm and CRS, ment inhibitor (SCIN) that binds to and stabilizes C3 conver-
little is known of the pathogenesis conferred by S. aureus biofilms. tases, interfering with additional C3b deposition through the
Endocarditis. Infection of the heart valve with S. aureus car- classical, lectin and alternative complement pathways.94 This
ries a high rate of morbidity and mortality. The left heart valve is leads to a substantial decrease in phagocytosis and killing of
most often infected and while the mortality rate is low in persons S. aureus by human neutrophils [polymorphonuclear leukocytes
under the age of 50, it easily surpass 50% in older patients clini- (PMN)].
cally diagnosed with S. aureus endocarditis.87 The treatment is While neutrophils comprise the immune systems primary
most often monitoring and antimicrobial therapy since biofilms defense against extracellular pathogens, S. aureus has evolved
are often sloughed and limited in size due to the high shear forces. a number of different strategies to disarm this early inflamma-
Ocular infection. Ocular infections associated with S. aureus tory response.95 Some of these mechanisms include the release of
biofilms include conjunctivitis, keratitis and endophthalmi- chemotaxis inhibitory protein of staphylococci (CHIPS), SCIN,
tis.88,89 Staphylococci, specifically S. aureus, had the highest isola- clumping factor A (ClfA) and extracellular adherence protein
tion rate from patients with conjunctivitis, and it is predicted that (Eap),71 not to mention the vast array of staphylococcal toxins.
MRSA cultures are more common in serious ocular infections The ability to develop biofilms may also be an effective means of
than methicillin-susceptible strains.88 In addition, S. aureus is the evading PMN phagocytosis. Neutrophils were able to penetrate,
second most common cause of post-operative endophthalmitis but not phagocytose biofilm when cultured with S. aureus bio-
and biofilm formation may play a large role in such infection.89 films.96 Neutrophils co-cultured with planktonic S. aureus cul-
Polymicrobial biofilm infections. Microorganisms rarely tures, however, were able to internalize S. aureus bacterial cells.96
live in isolated environments, but rather exist in polymicrobial A study by Guenther et al. contradicts this by showing that
communities. Recent interest has been formed in understanding S. aureus biofilms are not protected by the effects of PMN phago-
the relationships between multi-species biofilms and infection. cytosis, and in fact, PMNs release lactoferrin and elastase, along
A mixed microorganism interaction with medical importance with DNA.98 This difference may be due to the strain of S. aureus
exists between S. aureus and Candida albicans, a pathogenic used, duration of biofilm development, growth conditions or acti-
fungus associated with nosocomial infections in immuno-com- vation state of co-cultured neutrophils. The opsonization activity
promised patients. Candida albicans and S. aureus have been of IgG can also influence PMN action. For example, addition
co-cultured from oral and vaginal mucosal surfaces, both exhib- of serum IgG to S. aureus/neutrophil co-cultures resulted in
iting biofilm modes of growth.90 A recent study by Peters et al.90 enhanced biofilm clearance, not by increased de-granulation or
demonstrates the ability of S. aureus to favorably bind the hyphal adherence, but rather by upregulated oxygen radical production
form of C. albicans and develop biofilm. Interestingly, C. albicans in PMNs.99
hyphae can penetrate host epithelial cells, potentially providing Early activation of the inflammatory response may not only
an entrance for S. aureus local and systemic infections.91 be ineffective but may also even be detrimental to the host due
to the PMN activation and inflammatory mediator influx result-
Host Response to S. aureus Biofilm Infection ing in concomitant host tissue damage. Interestingly, S. aureus
biofilms, and not planktonic cultures, were conferred a growth
Biofilm-associated infections result in chronic disease that advantage in the presence of IL-1.100 This insinuates that induc-
are resistant to the host immune response. Although a myriad tion of inflammatory mediators, such as IL-1, may bolster
of information exists on biofilm development, little is known biofilm formation and therefore mitigate the immune systems
how the immune system reacts to infection. Recent research ability to clear S. aureus infection. In addition, recent studies in
has focused on elucidating the host responses to S. aureus bio- our laboratory have shown that the development of chronic infec-
film infections and delineating which confer clearance or foster tion was prevented by the early inhibition of Th1/Th17 through
persistence. the administration of monoclonal antibodies against the IL12p40
In a mouse tibial implant model of S. aureus biofilm osteo- antigen common to Th1/Th17 activation through the preven-
myelitis, a pro-inflammatory Th1/Th17 and anti-inflammatory tion of the early and non-specific host damage elicited by the
Th2/Treg paradigm was discovered. C57BL/6J mice were shown inflammatory response. Th2/Treg anti-inflammatory responses
to hold a chronic infection and resulted in upregulation of IL-2, and effective IgG1 antibodies, which may inhibit chronic infec-
IL-12p70, TNF and IL-1 (Th1-associated cytokines) and IL-6 tion, are not activated until persistent infection has already devel-
and IL-17 (Th17-associated cytokines).92 In addition, IgG2a and oped.101 However, even though the host produces these IgG1
IgG2b (Th1-associated antibodies) levels were increased early in antibodies, they may also be less effective due to the heteroge-
infection, and IgG1 (Th2-associated Ab) and suppressive Tregs neous antigenic character of the biofilm where an antigen may
were not upregulated until late in infection.92 This data suggests be produced in certain areas of the biofilm but completely absent
that pro-inflammatory Th1/Th17 responses occur early. from other areas (see Fig. 1).

www.landesbioscience.com Virulence 453


Therapy and Prophylaxis of S. aureus Biofilm only when equally efficacious and less toxic alternative agents are
Infections unavailable. Aminoglycosides can safely achieve high levels at the
site of infection when implanted close to the infectious nidus.112
In terms of the annual burden of morbidity and mortality it Glycopeptides are usually thought of as having a time-depen-
imposes on society, S. aureus is arguably the most significant bac- dent mode of action. Vancomycin is the principal agent used
terial pathogen in the western world. However, no concomitant for treatment of methicillin resistant biofilm infection; the rec-
advances in therapy or prevention have been forthcoming. This is ommended regimen in osteomyelitis being 15 mg/kg IV every
particularly so in the case of chronic, S. aureus biofilm infections. 24 h.110 The MICs to vancomycin of S. aureus strains have risen
The biofilm mode of growth is now recognized as a major over the most recent ten years, but aiming for slightly higher
mediator of infection, with an estimated 80% of all infections minimum serum concentrations most commonly overcomes
caused by biofilms.102 Resolution of S. aureus biofilm infections this. Resistance to vancomycin among staphylococci remains
continues to require surgical removal of the nidus of infection rare; less than 15 cases have been reported worldwide.113 One
due to innate resistance to antimicrobial and host response fac- study reported that a use of continuous infusion vancomycin
tors.103 However, attempts to develop an effective anti-S. aureus at a high dose was linked to an increased risk of occurrence of
vaccine have often ignored this mode of growth in favor of using antibiotic-associated diarrhea caused by Clostridium difficile.
planktonic cells and infection models.104-106 This is further com- Should this become a significant problem, linezolid or dapto-
plicated by the fact that S. aureusduring biofilm formation mycin may be effective alternatives for treatment of S. aureus
and maturationreleases a number of extracellular factors that biofilm infection.114
induce an inflammatory, Th1-type immune response rather than Rifampin has the capacity to kill metabolically dormant ses-
the more effective Th2-type as mentioned before.92 sile bacteria, making it highly useful for musculoskeletal infec-
Antimicrobial therapy. It is important to note that the vast tions of which a biofilm is the focus.115 Furthermore, it exhibits
majority of S. aureus biofilm infections must be either prevented high bioavailability and has very few known side-effects. In
from forming or be surgically removed once formed in order to order to prevent emergence of resistance to rifampin, it should
resolve the infection. In general, antimicrobial therapy alone is be only used in combination with another antibiotic agent that
not effective. In conjunction with surgical intervention such as is active against S. aureus, such as vancomycin or one of the
debridement, incision and drainage, indwelling medical device fluroquinolones.
removal, antimicrobial therapy is often prolonged and often takes Inclusion of antimicrobial agents at the site of infection.
place in the outpatient setting. Some individuals require extended A more preventative alternative to post-infection surgical man-
parenteral therapy due to either of the agent itself (e.g., vanco- agement and antibiotic treatment is inclusion of the agent in an
mycin) or difficulties with absorption from the gastrointestinal implanted medical device. This was initially performed with
tract.107 However, the evidence suggesting that parenteral admin- permanent, antibiotic-containing bone cement. Use of cement or
istration of vancomycin results in better outcomes than oral is calcium sulfate116 beads has shown some efficacy in preventing
limited. Parenteral antimicrobial therapy as an outpatient is both infection.117 These provide a more rapid release of high concen-
convenient and cost-effective, but is appropriate only for those trations of antibiotics and may be useful for situations such as
patients who meet the Infectious Disease Society of America cri- infection prophylaxis following open fractures.116 Calcium sul-
teria.107 The majority of patients with surgically managed biofilm fate beads also dissolve and so do not require surgical removal.
infections can be treated orally with one or more of several antibi- Additionally, they do not generate heat during formation, facili-
otics, so long as the causative organism is not resistant to them.108 tating use of a greater variety of antibiotics.
The requirement for prolonged treatment regimens means it is The antibiotic selected for inclusion in beads must be (1) active
vital to take into consideration toxicities and drug interactions against the causative microbes, (2) available as a powder that
when selecting a regimen. will harden properly and (3) able to maintain activity despite the
-lactam agents are often used in the treatment of biofilm heat generated during the polymerization process.110 The most
infections such as osteomyelitis. They demonstrate time-depen- commonly used agents are gentamicin in mainland Europe, and
dent killing, maximal when the serum concentration is ca. four in the United States and United Kingdom, tobramycin. Release
times the MIC.109 -lactams do not show a post-antibiotic effect; of antibiotic agents from cement or beads of any type shows a
i.e., antimicrobial activity after antibiotic concentrations have biphasic profile. The majority of the agent is released in the first
decreased below therapeutic levels. Penicillin G (1220 million few hours to days after implantation, with the remainder leach-
U every 24 h IV) and oxacillin (12 g IV every 46 h) are the ing out slowly over a matter of weeks, months or in some cases,
current recommended first-line therapy for osteomyelitis caused years.118
by penicillin-sensitive and -resistant S. aureus, respectively.110 While antibiotics and novel derivatives have been the mainstay
Unlike -lactams, fluoroquinolones and aminoglycosides of therapeutic alternatives, modern advances using other antimi-
demonstrate both concentration-dependent activity and a post- crobial agents such as nanosilver,119 cytokines such as IL-12,120
antibiotic effect.111 Dosing of such agents is limited by their nitric oxide,121 or anti-staphylococcal phages122 are being tested.
toxicity (aminoglycoside nephrotoxicity). Therefore, systemic However, the widespread clinical utility of these other agents
aminoglycosides are usually recommended for prolonged therapy have yet to be demonstrated.

454 Virulence Volume 2 Issue 5


Vaccines vivo expression technology (RIVET) and signature-tagged muta-
genesis.141,142 RIVET allows for identification of in vivo expressed
The greatest obstacle to development of a vaccine effective against antigens during infection whereas signature-tagged mutagenesis
S. aureus biofilm infection remains selection of appropriate anti- identifies antigens required for pathogenesis and survival. These
gens. The sequencing of the whole genome of multiple S. aureus techniques have the advantage of discovering vaccine candidates
strains,123 followed by those of methicillin-resistant124,125 and that in vitro methods may have missed. Additionally, these tech-
vancomycin-intermediate126 and epidemic127 strains undoubtedly niques do not require selective pressure on the bacteria and there-
marked a significant step forward. The subsequent development fore allow for a natural progression of infection.141
of bioinformatics to manipulate and analyze these data has facili- Proteomics and its derivatives. Proteomic profiling identi-
tated high-throughput genomic, transcriptomic and proteomic fies the spectrum of proteins expressed by bacteria under varying
analyses of microbial growth and pathogenesis.128,129 growth conditions using a combination of two-dimensional gel
The first question that must be answered is which compo- electrophoresis (2DGE) and mass spectrometry (MS). Detection
nent of the biofilm should be targeted. Broadly speaking, two of membrane and cell wall proteins is a limitation of proteomic
alternatives exist: bacterial cells within the biofilm and the bio- profiling due to (1) their relatively low abundance and (2) solu-
film matrix itself. The biofilm matrix may be composed of poly- bility constraints due to hydrophobicity (the presence of varying
saccharides, protein or extracellular DNA, in proportions that numbers of transmembrane domains), and an isoelectric point
vary between bacterial genera, species and strains. Most anti- at pH 8 +.143 Since vaccine development focuses on surface-asso-
biofilm vaccine efforts to-date have been directed toward the ciated proteins, use of extraction protocols that solubilize mem-
extracellular matrix.130 Perhaps the best example of this is the brane proteins or isoelectric focusing performed in the alkaline
staphylococcal polysaccharide inter-cellular adhesin (PIA), or pH range are essential. Reference maps of the surface proteomes
poly-N-acetyl--1,6-glucosamine (PNAG), production of which of S. aureus strains Phillips and VISA generated by lysostaphin
is encoded by the icaADBC locus.131 PIA is produced by both extraction are available.144,145 Another novel antigen discovery
S. epidermidis132 and S. aureus21 and is involved in adherence of strategy involves identification of surface proteins shaved from
S. epidermidis to host tissues133 and biomaterials.134 However, group A Streptococcus cells by trypsin digestion.146,147 Use of this
only 57% of icaADBC-positive strains135 produced a biofilm in technique has led to discovery of 42 S. aureus surface proteins
vitro,136 suggesting distinct strain differences in any correlation that may have potential as vaccine antigens.148
of PIA and biofilm formation. The proportion of ica-positive Serological probing of proteomic samples, known as immu-
S. aureus clinical isolates varies according to the clinical origin noproteomics, followed by peptide identification using matrix-
and even between infection sites that are both biofilm-mediated. assisted laser desorption ionization time-of-flight MS is a direct
For example, the proportion of icaADBC-positive S. aureus strains method for defining antigenic proteins. An initial 2DGE
was higher in orthopedic prosthesis-associated infections (92%) immunoproteomic study of S. aureus identified 15 known and
than in those that were catheter-associated (63%).137 There is also novel proteins that were immunoreactive with patient sera.149
some evidence that PIA expression undergoes phase variation.138 Additionally, subtractive proteome analysis was used to iden-
Although it has been tested against planktonic-type infection in tify immunogenic anchorless S. aureus surface proteins. Proteins
animal models,139 the efficacy of vaccination with PIA in prevent- reacting with an intravenous immunoglobulin (IVIG) prepara-
ing biofilm-type infections remains to be determined. tion (pooled IgG extracted from sera of multiple donors) but not
Genomics and its derivatives. Genome-based technologies with IVIG depleted of S. aureus-specific opsonizing antibodies
facilitated rapid identification of vaccine candidates compared were considered vaccine candidates.150 Of a total of ~40 anchor-
with the more conventional approach of identifying and ana- less cell wall proteins identified in this manner, three were cloned.
lyzing individual virulence factors from pathogens cultured in Although not tested in a biofilm model of infection, recombinant
vitro.140 Identification of putative antigensprincipally surface versions thereof provided partial protection in a mouse sepsis
proteinsby systematic searching of the pathogens genome model.150 Such anchorless wall proteins lack either a conserved
using bioinformatics is termed reverse vaccinology.140 This has a signal peptide or LPXTG motif, and so may have been omitted
number of advantages compared with traditional methodologies: from classical reverse vaccinology screens.151
(1) there is no need for in vitro culture and (2) antigen selec- An additional consideration when developing vaccines effec-
tion can proceed independent of in vivo expression levels and/or tive against chronic, biofilm-mediated S. aureus infection is the
immunogenicity. As a result, many antigens that would have been physiological heterogeneity of these communities (see Fig. 1).
passed over in conventional studies may be detected. However, Biofilm communities are inherently complex systems, usually
such analyses yield only limited information regarding levels of existing in close proximity to a surface. This complexity arises
expression of vaccine candidate antigens during pathogenesis. from a number of factors. First, distinct physicochemical gradi-
Transcriptomic analysis of bacterial pathogens in vivo using ents are found within microbial biofilm communities.4 In most
microarrays has the potential to generate such data; however, the cases, organic compounds, oxygen, or water enter the biofilm
large quantity of host RNA in any sample makes this problem- from the surrounding bulk fluid and diffuse through the matrix
atic. This need for information concerning in vivo expression of to the depths closer to the surface.152 Bacteria resident within a
potential vaccine antigens led to development of positive- and biofilm consume these compounds at varying rates, resulting in
negative-selection technologies such as recombination-based in differential availability of nutrients, dependent on the location

www.landesbioscience.com Virulence 455


of a particular cell within the community. This effect has been In conjunction with the multitude and redundancy of its viru-
observed experimentally in the case of oxygen tension.152 The lence factors in avoiding host responses and influencing disease,
situation is further complicated by very low metabolic levels S. aureus is able to form intricate micro-colonies termed bio-
and radically downregulated rates of cell division of the deeply films. Although neutrophils are capable of invading the biofilm,
entrenched microorganisms.153 the bacterial community is able to thwart this attack and may
Brady and coworkers used immunoproteomics to elucidate also skew the immune response to survive attack. Staphylococcus
S. aureus surface proteins that were immunogenic in the rab- aureus is the etiological agent to a myriad of human acute infec-
bit model154 of osteomyelitis.46 In subsequent work, the same tions, however, its ability to form biofilm in host emanates into
group demonstrated that expression of these antigens during chronic and recalcitrant disease. Current therapies for treating
S. aureus biofilm growth in vitro was spatially heterogeneous.5 and preventing chronic biofilm-mediated infections are limited
Based on these data, four antigens were included in a quadri- to surgical intervention and prolonged antibiotic regiments or
valent vaccine administered to rabbits that were then infected addition of antimicrobial compounds to indwelling-medical
with S. aureus M2 in a biofilm model of infection of osteomy- devices. Vaccination studies have begun to take biofilm develop-
elitis. Vaccination, in combination with post-infection vancomy- ment into consideration, and with the combination of genomic
cin therapy, was effective at preventing development of chronic and proteomic-based techniques have identified numerous poten-
S. aureus osteomyelitis.155 tial vaccination candidates. However, the physiological heteroge-
Despite the considerable human and fiscal resources that have neity and subsequent multifarious protein expression throughout
been expended, the search for a vaccine effective against S. aureus the biofilm must be carefully examined for development of an
biofilm infections continues. A caveat to consider in developing efficacious S. aureus vaccine. Alternatively, modulating the host
anti-S. aureus biofilm therapies are the potential losses of treat- immune response may prove advantageous in resolving chronic
ment efficacy when given to immunocompromised patients at S. aureus infections and warrants further investigation.
risk of infection, specifically in the case of vaccination. It seems
likely that the answer lies in an increased understanding of Acknowledgments
S. aureus biofilm development in vivo, and particularly its inter- Completion of this article was funded by the National Institute
actions with the immune system. of Allergy and Infectious Diseases, National Institutes of Health
(grant R01 AI69568-01A2).
Conclusion
Note
Staphylococcus aureus is a clinically relevant pathogen due to its Supplemental material can be found at:
antimicrobial resistance and evasion of the host immune system. www.landesbioscience.com/journals/virulence/article/17724
7. Lewis K. Persister cells. Annu Rev Microbiol 2010; 16. DallAntonia M, Coen PG, Wilks M, Whiley A,
References
64:357-72; PMID:20528688; DOI:10.1146/annurev. Millar M. Competition between methicillin-sensitive
1. Donlan RM, Costerton JW. Biofilms: survival mech- micro.112408.134306. and -resistant Staphylococcus aureus in the anterior
anisms of clinically relevant microorganisms. Clin 8. Spoering AL, Lewis K. Biofilms and planktonic cells nares. J Hosp Infect 2005; 61:62-7; PMID:15893854;
Microbiol Rev 2002; 15:167-93; PMID:11932229; of Pseudomonas aeruginosa have similar resistance to DOI:10.1016/j.jhin.2005.01.008.
DOI:10.1128/CMR.15.2.167-93.2002. killing by antimicrobials. J Bacteriol 2001; 183:6746- 17. Beenken KE, Dunman PM, McAleese F, Macapagal
2. Costerton JW, Lewandowski Z, Caldwell DE, Korber 51; PMID:11698361; DOI:10.1128/JB.183.23.6746- D, Murphy E, Projan SJ, et al. Global gene expres-
DR, Lappin-Scott HM. Microbial biofilms. Annu 51.2001. sion in Staphylococcus aureus biofilms. J Bacteriol
Rev Microbiol 1995; 49:711-45; PMID:8561477; 9. Lewis K. Persister cells and the riddle of biofilm 2004; 186:4665-84; PMID:15231800; DOI:10.1128/
DOI:10.1146/annurev.mi.49.100195.003431. survival. Biochemistry (Mosc) 2005; 70:267-74; JB.186.14.4665-84.2004.
3. Kristian SA, Golda T, Ferracin F, Cramton SE, PMID:15807669; DOI:10.1007/s10541-005-0111-6. 18. Fitzpatrick F, Humphreys H, OGara JP. The genetics
Neumeister B, Peschel A, et al. The ability of bio- 10. Xu KD, McFeters GA, Stewart PS. Biofilm resistance to of staphylococcal biofilm formationwill a greater
film formation does not influence virulence of antimicrobial agents. Microbiology 2000; 146:547-9; understanding of pathogenesis lead to better manage-
Staphylococcus aureus and host response in a mouse PMID:10746758. ment of device-related infection? Clin Microbiol Infect
tissue cage infection model. Microb Pathog 2004; 2005; 11:967-73; PMID:16307550; DOI:10.1111/
11. De Beer D, Srinivasan R, Stewart PS. Direct measure-
36:237-45; PMID:15043859; DOI:10.1016/j.mic- j.1469-0691.2005.01274.x.
ment of chlorine penetration into biofilms during dis-
path.2003.12.004. 19. Hussain M, Wilcox MH, White PJ. The slime of coag-
infection. Appl Environ Microbiol 1994; 60:4339-44;
4. Rani SA, Pitts B, Beyenal H, Veluchamy RA, PMID:7811074. ulase-negative staphylococci: biochemistry and relation
Lewandowski Z, Davison WM, et al. Spatial patterns to adherence. FEMS Microbiol Rev 1993; 10:191-207;
12. Singh R, Ray P, Das A, Sharma M. Penetration of anti-
of DNA replication, protein synthesis and oxygen PMID:8318256.
biotics through Staphylococcus aureus and Staphylococcus
concentration within bacterial biofilms reveal diverse 20. Mack D, Fischer W, Krokotsch A, Leopold K,
epidermidis biofilms. J Antimicrob Chemother 2010;
physiological states. J Bacteriol 2007; 189:4223-33; Hartmann R, Egge H, et al. The intercellular adhesin
65:1955-8; PMID:20615927; DOI:10.1093/jac/
PMID:17337582; DOI:10.1128/JB.00107-07. involved in biofilm accumulation of Staphylococcus epi-
dkq257.
5. Brady RA, Leid JG, Kofonow J, Costerton JW, dermidis is a linear beta-1,6-linked glucosaminoglycan:
13. Boyd A, Chakrabarty AM. Role of alginate lyase in cell
Shirtliff ME. Immunoglobulins to surface-associated purification and structural analysis. J Bacteriol 1996;
detachment of Pseudomonas aeruginosa. Appl Environ
biofilm immunogens provide a novel means of visu- 178:175-83; PMID:8550413.
Microbiol 1994; 60:2355-9; PMID:8074516.
alization of methicillin-resistant Staphylococcus aureus 21. Cramton SE, Gerke C, Schnell NF, Nichols WW,
14. Sauer K, Camper AK, Ehrlich GD, Costerton JW,
biofilms. Appl Environ Microbiol 2007; 73:6612-9; Gotz F. The intercellular adhesion (ica) locus is pres-
Davies DG. Pseudomonas aeruginosa displays mul-
PMID:17720840; DOI:10.1128/AEM.00855-07. ent in Staphylococcus aureus and is required for bio-
tiple phenotypes during development as a biofilm.
6. Beveridge TJ, Makin SA, Kadurugamuwa JL, Li film formation. Infect Immun 1999; 67:5427-33;
J Bacteriol 2002; 184:1140-54; PMID:11807075;
Z. Interactions between biofilms and the environ- PMID:10496925.
DOI:10.1128/jb.184.4.1140-54.2002.
ment. FEMS Microbiol Rev 1997; 20:291-303;
15. Kluytmans J, van Belkum A, Verbrugh H. Nasal car-
PMID:9299708; DOI:10.1111/j.1574-6976.1997.
riage of Staphylococcus aureus: epidemiology, underlying
tb00315.x.
mechanisms and associated risks. Clin Microbiol Rev
1997; 10:505-20; PMID:9227864.

456 Virulence Volume 2 Issue 5


22. Cramton SE, Ulrich M, Gotz F, Doring G. Anaerobic 34. Merino N, Toledo-Arana A, Vergara-Irigaray M, Valle 49. Valle J, Toledo-Arana A, Berasain C, Ghigo JM, Amorena
conditions induce expression of polysaccharide intercel- J, Solano C, Calvo E, et al. Protein A-mediated mul- B, Penades JR, et al. SarA and not sigmaB is essential
lular adhesin in Staphylococcus aureus and Staphylococcus ticellular behavior in Staphylococcus aureus. J Bacteriol for biofilm development by Staphylococcus aureus.
epidermidis. Infect Immun 2001; 69:4079-85; 2009; 191:832-43; PMID:19047354; DOI:10.1128/ Mol Microbiol 2003; 48:1075-87; PMID:12753197;
PMID:11349079; DOI:10.1128/IAI.69.6.4079- JB.01222-08. DOI:10.1046/j.1365-2958.2003.03493.x.
85.2001. 35. Houston P, Rowe SE, Pozzi C, Waters EM, OGara 50. Beenken KE, Blevins JS, Smeltzer MS. Mutation of
23. Ulrich M, Bastian M, Cramton SE, Ziegler K, Pragman JP. Essential role for the major autolysin in the fibro- sarA in Staphylococcus aureus limits biofilm formation.
AA, Bragonzi A, et al. The staphylococcal respiratory nectin-binding protein-mediated Staphylococcus aureus Infect Immun 2003; 71:4206-11; PMID:12819120;
response regulator SrrAB induces ica gene transcription biofilm phenotype. Infect Immun 2011; 79:1153-65; DOI:10.1128/IAI.71.7.4206-11.2003.
and polysaccharide intercellular adhesin expression, PMID:21189325; DOI:10.1128/IAI.00364-10. 51. Tsang LH, Cassat JE, Shaw LN, Beenken KE, Smeltzer
protecting Staphylococcus aureus from neutrophil killing 36. Hennig S, Nyunt Wai S, Ziebuhr W. Spontaneous MS. Factors contributing to the biofilm-deficient
under anaerobic growth conditions. Mol Microbiol switch to PIA-independent biofilm formation in an phenotype of Staphylococcus aureus sarA mutants. PLoS
2007; 65:1276-87; PMID:17697253; DOI:10.1111/ ica-positive Staphylococcus epidermidis isolate. Int J ONE 2008; 3:3361; PMID:18846215; DOI:10.1371/
j.1365-2958.2007.05863.x. Med Microbiol 2007; 297:117-22; PMID:17292669; journal.pone.0003361.
24. Conlon KM, Humphreys H, OGara JP. icaR encodes DOI:10.1016/j.ijmm.2006.12.001. 52. Cheung AL, Projan SJ. Cloning and sequencing of
a transcriptional repressor involved in environmen- 37. Rohde H, Burandt EC, Siemssen N, Frommelt L, sarA of Staphylococcus aureus, a gene required for the
tal regulation of ica operon expression and biofilm Burdelski C, Wurster S, et al. Polysaccharide intercel- expression of agr. J Bacteriol 1994; 176:4168-72;
formation in Staphylococcus epidermidis. J Bacteriol lular adhesin or protein factors in biofilm accumulation PMID:8021198.
2002; 184:4400-8; PMID:12142410; DOI:10.1128/ of Staphylococcus epidermidis and Staphylococcus aureus 53. Beenken KE, Mrak LN, Griffin LM, Zielinska AK,
JB.184.16.4400-8.2002. isolated from prosthetic hip and knee joint infections. Shaw LN, Rice KC, et al. Epistatic relationships between
25. Kiem S, Oh WS, Peck KR, Lee NY, Lee JY, Song Biomaterials 2007; 28:1711-20; PMID:17187854; sarA and agr in Staphylococcus aureus biofilm forma-
JH, et al. Phase variation of biofilm formation in DOI:10.1016/j.biomaterials.2006.11.046. tion. PLoS ONE 2010; 5:10790; PMID:20520723;
Staphylococcus aureus by IS 256 insertion and its impact 38. Lasa I, Penades JR. Bap: a family of surface proteins DOI:10.1371/journal.pone.0010790.
on the capacity adhering to polyurethane surface. J involved in biofilm formation. Res Microbiol 2006; 54. Chan WC, Coyle BJ, Williams P. Virulence regulation
Korean Med Sci 2004; 19:779-82; PMID:15608385; 157:99-107; PMID:16427771; DOI:10.1016/j.res- and quorum sensing in staphylococcal infections: com-
DOI:10.3346/jkms.2004.19.6.779. mic.2005.11.003. petitive AgrC antagonists as quorum sensing inhibitors.
26. Jefferson KK, Pier DB, Goldmann DA, Pier GB. 39. Whitchurch CB, Tolker-Nielsen T, Ragas PC, Mattick J Med Chem 2004; 47:4633-41; PMID:15341477;
The teicoplanin-associated locus regulator (TcaR) JS. Extracellular DNA required for bacterial biofilm DOI:10.1021/jm0400754.
and the intercellular adhesin locus regulator (IcaR) formation. Science 2002; 295:1487; PMID:11859186; 55. Boles BR, Horswill AR. Agr-mediated dispersal of
are transcriptional inhibitors of the ica locus in DOI:10.1126/science.295.5559.1487. Staphylococcus aureus biofilms. PLoS Pathog 2008;
Staphylococcus aureus. J Bacteriol 2004; 186:2449- 40. Steinberger RE, Holden PA. Extracellular DNA 4:1000052; PMID:18437240; DOI:10.1371/journal.
56; PMID:15060048; DOI:10.1128/JB.186.8.2449- in single- and multiple-species unsaturated bio- ppat.1000052.
56.2004. films. Appl Environ Microbiol 2005; 71:5404-10; 56. Lauderdale KJ, Boles BR, Cheung AL, Horswill AR.
27. Jefferson KK, Cramton SE, Gotz F, Pier GB. PMID:16151131; DOI:10.1128/AEM.71.9.5404- Interconnections between SigmaB, agr and proteolytic
Identification of a 5-nucleotide sequence that controls 10.2005. activity in Staphylococcus aureus biofilm maturation.
expression of the ica locus in Staphylococcus aureus and 41. Allesen-Holm M, Barken KB, Yang L, Klausen M, Infect Immun 2009; 77:1623-35; PMID:19188357;
characterization of the DNA-binding properties of IcaR. Webb JS, Kjelleberg S, et al. A characterization of DNA DOI:10.1128/IAI.01036-08.
Mol Microbiol 2003; 48:889-99; PMID:12753184; release in Pseudomonas aeruginosa cultures and biofilms. 57. Kong KF, Vuong C, Otto M. Staphylococcus quorum
DOI:10.1046/j.1365-2958.2003.03482.x. Mol Microbiol 2006; 59:1114-28; PMID:16430688; sensing in biofilm formation and infection. Int J
28. Cue D, Lei MG, Luong TT, Kuechenmeister L, DOI:10.1111/j.1365-2958.2005.05008.x. Med Microbiol 2006; 296:133-9; PMID:16487744;
Dunman PM, ODonnell S, et al. Rbf promotes bio- 42. Gibson RL, Burns JL, Ramsey BW. Pathophysiology DOI:10.1016/j.ijmm.2006.01.042.
film formation by Staphylococcus aureus via repression and management of pulmonary infections in cystic 58. Pan-Farr J, Jonas B, Forstner K, Engelmann S, Hecker
of icaR, a negative regulator of icaADBC. J Bacteriol fibrosis. Am J Respir Crit Care Med 2003; 168:918- M. The sigmaB regulon in Staphylococcus aureus and its
2009; 191:6363-73; PMID:19684134; DOI:10.1128/ 51; PMID:14555458; DOI:10.1164/rccm.200304- regulation. Int J Med Microbiol 2006; 296:237-58;
JB.00913-09. 505SO. PMID:16644280; DOI:10.1016/j.ijmm.2005.11.011.
29. Pamp SJ, Frees D, Engelmann S, Hecker M, Ingmer H. 43. Eckhart L, Fischer H, Barken KB, Tolker-Nielsen T, 59. Nicholas RO, Li T, McDevitt D, Marra A, Sucoloski
Spx is a global effector impacting stress tolerance and Tschachler E. DNase1L2 suppresses biofilm formation S, Demarsh PL, et al. Isolation and characterization of
biofilm formation in Staphylococcus aureus. J Bacteriol by Pseudomonas aeruginosa and Staphylococcus aureus. a sigB deletion mutant of Staphylococcus aureus. Infect
2006; 188:4861-70; PMID:16788195; DOI:10.1128/ Br J Dermatol 2007; 156:1342-5; PMID:17459041; Immun 1999; 67:3667-9; PMID:10377157.
JB.00194-06. DOI:10.1111/j.1365-2133.2007.07886.x. 60. Nair SP, Bischoff M, Senn MM, Berger-Bachi B.
30. Toledo-Arana A, Merino N, Vergara-Irigaray M, 44. Rice KC, Mann EE, Endres JL, Weiss EC, Cassat JE, The sigmaB regulon influences internalization of
Debarbouille M, Penades JR, Lasa I. Staphylococcus Smeltzer MS, et al. The cidA murein hydrolase regula- Staphylococcus aureus by osteoblasts. Infect Immun
aureus develops an alternative, ica-independent biofilm tor contributes to DNA release and biofilm develop- 2003; 71:4167-70; PMID:12819110; DOI:10.1128/
in the absence of the arlRS two-component system. ment in Staphylococcus aureus. Proc Natl Acad Sci USA IAI.71.7.4167-70.2003.
J Bacteriol 2005; 187:5318-29; PMID:16030226; 2007; 104:8113-8; PMID:17452642; DOI:10.1073/ 61. Kullik I, Giachino P. The alternative sigma factor
DOI:10.1128/JB.187.15.5318-29.2005. pnas.0610226104. sigmaB in Staphylococcus aureus: regulation of the sigB
31. Francois P, Tu Quoc PH, Bisognano C, Kelley WL, 45. Mann EE, Rice KC, Boles BR, Endres JL, Ranjit operon in response to growth phase and heat shock.
Lew DP, Schrenzel J, et al. Lack of biofilm contri- D, Chandramohan L, et al. Modulation of eDNA Arch Microbiol 1997; 167:151-9; PMID:9042755;
bution to bacterial colonisation in an experimental release and degradation affects Staphylococcus DOI:10.1007/s002030050428.
model of foreign body infection by Staphylococcus aureus biofilm maturation. PLoS ONE 2009; 62. Rachid S, Ohlsen K, Wallner U, Hacker J, Hecker M,
aureus and Staphylococcus epidermidis. FEMS Immunol 4:5822; PMID:19513119; DOI:10.1371/journal. Ziebuhr W. Alternative transcription factor sigma(B)
Med Microbiol 2003; 35:135-40; PMID:12628549; pone.0005822. is involved in regulation of biofilm expression in
DOI:10.1016/S0928-8244(02)00463-7. 46. Brady RA, Leid JG, Camper AK, Costerton JW, a Staphylococcus aureus mucosal isolate. J Bacteriol
32. Fitzpatrick F, Humphreys H, OGara JP. Evidence for Shirtliff ME. Identification of Staphylococcus aureus 2000; 182:6824-6; PMID:11073930; DOI:10.1128/
icaADBC-independent biofilm development mech- proteins recognized by the antibody-mediated immune JB.182.23.6824-6.2000.
anism in methicillin-resistant Staphylococcus aureus response to a biofilm infection. Infect Immun 2006; 63. Cassat J, Dunman PM, Murphy E, Projan SJ, Beenken
clinical isolates. J Clin Microbiol 2005; 43:1973-6; 74:3415-26; PMID:16714572; DOI:10.1128/ KE, Palm KJ, et al. Transcriptional profiling of a
PMID:15815035; DOI:10.1128/JCM.43.4.1973- IAI.00392-06. Staphylococcus aureus clinical isolate and its isogenic
6.2005. 47. Resch A, Fehrenbacher B, Eisele K, Schaller M, agr and sarA mutants reveals global differences in com-
33. ONeill E, Pozzi C, Houston P, Smyth D, Humphreys Gotz F. Phage release from biofilm and plankton- parison to the laboratory strain RN6390. Microbiology
H, Robinson DA, et al. Association between methicillin ic Staphylococcus aureus cells. FEMS Microbiol Lett 2006; 152:3075-90; PMID:17005987; DOI:10.1099/
susceptibility and biofilm regulation in Staphylococcus 2005; 252:89-96; PMID:16213676; DOI:10.1016/j. mic.0.29033-0.
aureus isolates from device-related infections. J Clin femsle.2005.08.048. 64. Resch A, Leicht S, Saric M, Pasztor L, Jakob A, Gotz F,
Microbiol 2007; 45:1379-88; PMID:17329452; 48. Fournier B, Klier A, Rapoport G. The two-component et al. Comparative proteome analysis of Staphylococcus
DOI:10.1128/JCM.02280-06. system ArlS-ArlR is a regulator of virulence gene aureus biofilm and planktonic cells and correlation with
expression in Staphylococcus aureus. Mol Microbiol transcriptome profiling. Proteomics 2006; 6:1867-77;
2001; 41:247-61; PMID:11454217; DOI:10.1046/ PMID:16470655; DOI:10.1002/pmic.200500531.
j.1365-2958.2001.02515.x.

www.landesbioscience.com Virulence 457


65. Dunman PM, Murphy E, Haney S, Palacios D, Tucker- 81. Bowling FL, Jude EB, Boulton AJ. MRSA and diabetic 97. Guenther F, Stroh P, Wagner C, Obst U, Hansch GM.
Kellogg G, Wu S, et al. Transcription profiling-based foot wounds: contaminating or infecting organisms? Phagocytosis of staphylococci biofilms by polymor-
identification of Staphylococcus aureus genes regulated Curr Diab Rep 2009; 9:440-4; PMID:19954689; phonuclear neutrophils: S. aureus and S. epidermidis
by the agr and/or sarA loci. J Bacteriol 2001; 183:7341- DOI:10.1007/s11892-009-0072-z. differ with regard to their susceptibility towards the
53; PMID:11717293; DOI:10.1128/JB.183.24.7341- 82. Schierle CF, De la Garza M, Mustoe TA, Galiano host defense. Int J Artif Organs 2009; 32:565-73;
53.2001. RD. Staphylococcal biofilms impair wound heal- PMID:19856266.
66. Wenzel RP, Edmond MB. The impact of hospital- ing by delaying reepithelialization in a murine cuta- 98. Meyle E, Stroh P, Gunther F, Hoppy-Tichy T, Wagner
acquired bloodstream infections. Emerg Infect Dis neous wound model. Wound Repair Regen 2009; C, Hansch GM. Destruction of bacterial biofilms by
2001; 7:174-7; PMID:11294700; DOI:10.3201/ 17:354-9; PMID:19660043; DOI:10.1111/j.1524- polymorphonuclear neutrophils: relative contribution
eid0702.010203. 475X.2009.00489.x. of phagocytosis, DNA release and degranulation. Int J
67. Rubin RJ, Harrington CA, Poon A, Dietrich K, 83. Foreman A, Wormald PJ. Different biofilms, differ- Artif Organs 2010; 33:608-20; PMID:20890882.
Greene JA, Moiduddin A. The economic impact of ent disease? A clinical outcomes study. Laryngoscope 99. Stroh P, Gunther F, Meyle E, Prior B, Wagner C,
Staphylococcus aureus infection in New York City hospi- 2010; 120:1701-6; PMID:20641074; DOI:10.1002/ Hansch GM. Host defence against Staphylococcus
tals. Emerg Infect Dis 1999; 5:9-17; PMID:10081667; lary.21024. aureus biofilms by polymorphonuclear neutrophils:
DOI:10.3201/eid0501.990102. 84. Ferguson BJ, Stolz DB. Demonstration of biofilm in oxygen radical production but not phagocytosis
68. Jones SM, Morgan M, Humphrey TJ, Lappin-Scott human bacterial chronic rhinosinusitis. Am J Rhinol depends on opsonisation with immunoglobulin G.
H. Effect of vancomycin and rifampicin on meticillin- 2005; 19:452-7; PMID:16270598. Immunobiology 2011; 216:351-7; PMID:20850891;
resistant Staphylococcus aureus biofilms. Lancet 2001; 85. Psaltis AJ, Weitzel EK, Ha KR, Wormald PJ. The DOI:10.1016/j.imbio.2010.07.009.
357:40-1; PMID:11197363; DOI:10.1016/S0140- effect of bacterial biofilms on post-sinus surgical out- 100. McLaughlin RA, Hoogewerf AJ. Interleukin-1beta-
6736(00)03572-8. comes. Am J Rhinol 2008; 22:1-6; PMID:18284851; induced growth enhancement of Staphylococcus aure-
69. Lew DP, Waldvogel FA. Osteomyelitis. Lancet 2004; DOI:10.2500/ajr.2008.22.3119. us occurs in biofilm but not planktonic cultures.
364:369-79; PMID:15276398; DOI:10.1016/S0140- 86. Stephenson MF, Mfuna L, Dowd SE, Wolcott RD, Microb Pathog 2006; 41:67-79; PMID:16769197;
6736(04)16727-5. Barbeau J, Poisson M, et al. Molecular characteriza- DOI:10.1016/j.micpath.2006.04.005.
70. Ziran BH. Osteomyelitis. J Trauma 2007; tion of the polymicrobial flora in chronic rhinosinus- 101. Prabhakara RLJ, Harro JM, Costerton JW, Shirtliff
62:59-60; PMID:17556977; DOI:10.1097/ itis. J Otolaryngol Head Neck Surg 2010; 39:182-7; ME. Immune response to Staphylococcus aureus biofilm
TA.0b013e318065abbd. PMID:20211106. infections. 5th ASM Conference on Biofilms. Cancun,
71. Anwar S, Prince LR, Foster SJ, Whyte MK, Sabroe 87. Rder BL, Wandall DA, Frimodt-Moller N, Espersen Mexico 2009.
I. The rise and rise of Staphylococcus aureus: laughing F, Skinhoj P, Rosdahl VT. Clinical features of 102. NIH NH, Lung and Blood Institiute Research on
in the face of granulocytes. Clin Exp Immunol 2009; Staphylococcus aureus endocarditis: a 10-year experi- Microbial Biofilms 2002.
157:216-24; PMID:19604261; DOI:10.1111/j.1365- ence in Denmark. Arch Intern Med 1999; 159:462-9; 103. OMay GA, Brady RA, Prabhakara R, Leid JG,
2249.2009.03950.x. PMID:10074954; DOI:10.1001/archinte.159.5.462. Calhoun JH, Shirtliff ME, Eds. Osteomyelitis. In
72. Akiyama H, Torigoe R, Arata J. Interaction of 88. Murugan K, Usha M, Malathi P, Al-Sohaibani AS, Biofilm Infections: New York: Springer 2010.
Staphylococcus aureus cells and silk threads in vitro Chandrasekaran M. Biofilm forming multi drug 104. Kim HK, Cheng AG, Kim HY, Missiakas DM,
and in mouse skin. J Dermatol Sci 1993; 6:247- resistant Staphylococcus spp. among patients with Schneewind O. Nontoxigenic protein A vaccine
57; PMID:8136322; DOI:10.1016/0923- conjunctivitis. Pol J Microbiol 2010; 59:233-9; for methicillin-resistant Staphylococcus aureus infec-
1811(93)90046-R. PMID:21466040. tions in mice. J Exp Med 2010; 207:1863-70;
73. Baldoni D, Haschke M, Rajacic Z, Zimmerli W, 89. Leid JG, Costerton JW, Shirtliff ME, Gilmore PMID:20713595; DOI:10.1084/jem.20092514.
Trampuz A. Linezolid alone or combined with MS, Engelbert M. Immunology of Staphylococcal 105. Kuklin NA, Clark DJ, Secore S, Cook J, Cope
rifampin against methicillin-resistant Staphylococcus biofilm infections in the eye: new tools to LD, McNeely T, et al. A novel Staphylococcus aureus
aureus in experimental foreign-body infection. study biofilm endophthalmitis. DNA Cell vaccine: iron surface determinant B induces rapid
Antimicrob Agents Chemother 2009; 53:1142-8; Biol 2002; 21:405-13; PMID:12167243; antibody responses in rhesus macaques and specific
PMID:19075065; DOI:10.1128/AAC.00775-08. DOI:10.1089/10445490260099692. increased survival in a murine S. aureus sepsis model.
74. Costerton JW, Montanaro L, Arciola CR. Biofilm in 90. Peters BM, Jabra-Rizk MA, Scheper MA, Leid JG, Infect Immun 2006; 74:2215-23; PMID:16552052;
implant infections: its production and regulation. Int J Costerton JW, Shirtliff ME. Microbial interactions DOI:10.1128/IAI.74.4.2215-23.2006.
Artif Organs 2005; 28:1062-8; PMID:16353112. and differential protein expression in Staphylococcus 106. Stranger-Jones YK, Bae T, Schneewind O. Vaccine
75. Herrmann M, Vaudaux PE, Pittet D, Auckenthaler R, aureus-Candida albicans dual-species biofilms. assembly from surface proteins of Staphylococcus aure-
Lew PD, Schumacher-Perdreau F, et al. Fibronectin, FEMS Immunol Med Microbiol 2010; 59:493-503; us. Proc Natl Acad Sci USA 2006; 103:16942-7;
fibrinogen and laminin act as mediators of adherence PMID:20608978. PMID:17075065; DOI:10.1073/pnas.0606863103.
of clinical staphylococcal isolates to foreign mate- 91. Sudbery P, Gow N, Berman J. The distinct morpho- 107. Tice AD, Rehm SJ, Dalovisio JR, Bradley JS, Martinelli
rial. J Infect Dis 1988; 158:693-701; PMID:3171224; genic states of Candida albicans. Trends Microbiol LP, Graham DR, et al. Practice guidelines for outpa-
DOI:10.1093/infdis/158.4.693. 2004; 12:317-24; PMID:15223059; DOI:10.1016/j. tient parenteral antimicrobial therapy. IDSA guidelines.
76. Heitz-Mayfield LJ, Lang NP. Comparative biology of tim.2004.05.008. Clin Infect Dis 2004; 38:1651-71; PMID:15227610;
chronic and aggressive periodontitis vs. peri-implantitis. 92. Prabhakara R, Harro JM, Leid JG, Harris M, DOI:10.1086/420939.
Periodontol 2000 2010; 53:167-81; PMID:20403112; Shirtliff ME. Murine Immune Response to a Chronic 108. Osmon DR, Berbari EF. Outpatient intravenous antimi-
DOI:10.1111/j.1600-0757.2010.00348.x. Staphylococcus aureus Biofilm Infection. Infect Immun crobial therapy for the practicing orthopaedic surgeon.
77. Cuesta AI, Jewtuchowicz V, Brusca MI, Nastri ML, 2011; 79:1789-96; PMID:21282411; DOI:10.1128/ Clin Orthop Relat Res 2002; 80-6; PMID:12360011;
Rosa AC. Prevalence of Staphylococcus spp. and IAI.01386-10. DOI:10.1097/00003086-200210000-00013.
Candida spp. in the oral cavity and periodontal pockets 93. Muschel LH, Jackson JE. The reactivity of serum 109. McKinnon PS, Davis SL. Pharmacokinetic and phar-
of periodontal disease patients. Acta Odontol Latinoam against protoplasts and spheroplasts. J Immunol 1966; macodynamic issues in the treatment of bacterial
2010; 23:20-6; PMID:20645638. 97:46-51; PMID:4958551. infectious diseases. Eur J Clin Microbiol Infect Dis
78. Gjdsbl K, Christensen JJ, Karlsmark T, Jorgensen 94. Rooijakkers SH, Ruyken M, Roos A, Daha MR, 2004; 23:271-88; PMID:15015030; DOI:10.1007/
B, Klein BM, Krogfelt KA. Multiple bacterial spe- Presanis JS, Sim RB, et al. Immune evasion by a staphy- s10096-004-1107-7.
cies reside in chronic wounds: a longitudinal study. lococcal complement inhibitor that acts on C3 conver- 110. Rao N, Ziran BH, Lipsky BA. Treating osteomy-
Int Wound J 2006; 3:225-31; PMID:16984578; tases. Nat Immunol 2005; 6:920-7; PMID:16086019; elitis: antibiotics and surgery. Plast Reconstr Surg
DOI:10.1111/j.1742-481X.2006.00159.x. DOI:10.1038/ni1235. 2011; 127:177-87; PMID:21200289; DOI:10.1097/
79. Hansson C, Hoborn J, Moller A, Swanbeck G. The 95. Kobayashi SD, DeLeo FR. Role of neutrophils in PRS.0b013e3182001f0f.
microbial flora in venous leg ulcers without clinical innate immunity: a systems biology-level approach. 111. Jacobs MR. Optimisation of antimicrobial ther-
signs of infection. Repeated culture using a validated Wiley Interdiscip Rev Syst Biol Med 2009; 1:309-33; apy using pharmacokinetic and pharmacodynamic
standardised microbiological technique. Acta Derm PMID:20836000; DOI:10.1002/wsbm.32. parameters. Clin Microbiol Infect 2001; 7:589-
Venereol 1995; 75:24-30; PMID:7747531. 96. Leid JG, Shirtliff ME, Costerton JW, Stoodley P. 96; PMID:11737083; DOI:10.1046/j.1198-
80. Davis SC, Ricotti C, Cazzaniga A, Welsh E, Eaglstein Human leukocytes adhere to, penetrate and respond 743x.2001.00295.x.
WH, Mertz PM. Microscopic and physiologic evidence to Staphylococcus aureus biofilms. Infect Immun 112. Andes D, Craig WA. Pharmacokinetics and phar-
for biofilm-associated wound colonization in vivo. 2002; 70:6339-45; PMID:12379713; DOI:10.1128/ macodynamics of outpatient intravenous antimicro-
Wound Repair Regen 2008; 16:23-9; PMID:18211576; IAI.70.11.6339-45.2002. bial therapy. [vi.]. Infect Dis Clin North Am 1998;
DOI:10.1111/j.1524-475X.2007.00303.x. 12:849-60; PMID:9888026; DOI:10.1016/S0891-
5520(05)70024-6.

458 Virulence Volume 2 Issue 5


113. Gould IM. VRSA-doomsday superbug or damp squib? 128. Kaushik DK, Sehgal D. Developing antibacterial 143. Fountoulakis M, Takacs B. Effect of strong deter-
Lancet Infect Dis 2010; 10:816-8; PMID:21109164; vaccines in genomics and proteomics era. Scand J gents and chaotropes on the detection of pro-
DOI:10.1016/S1473-3099(10)70259-0. Immunol 2008; 67:544-52; PMID:18397199; teins in two-dimensional gels. Electrophoresis
114. Fraimow HS. Systemic antimicrobial therapy DOI:10.1111/j.1365-3083.2008.02107.x. 2001; 22:1593-602; PMID:11425215;
in osteomyelitis. Semin Plast Surg 2009; 23:90-9; 129. Zagursky RJ, Anderson AS. Application of genomics DOI:10.1002/1522-2683(200105)22:9<1593::AID-
PMID:20567731; DOI:10.1055/s-0029-1214161. in bacterial vaccine discovery: a decade in review. Curr ELPS1593>3.0.CO;2-6.
115. Perlroth J, Kuo M, Tan J, Bayer AS, Miller LG. Opin Pharmacol 2008; 8:632-8; PMID:18625342; 144. Gatlin CL, Pieper R, Huang ST, Mongodin E,
Adjunctive use of rifampin for the treatment of DOI:10.1016/j.coph.2008.06.009. Gebregeorgis E, Parmar PP, et al. Proteomic profiling
Staphylococcus aureus infections: a systematic review 130. Schaffer AC, Lee JC. Vaccination and passive of cell envelope-associated proteins from Staphylococcus
of the literature. Arch Intern Med 2008; 168:805-19; immunisation against Staphylococcus aureus. Int J aureus. Proteomics 2006; 6:1530-49; PMID:16470658;
PMID:18443255; DOI:10.1001/archinte.168.8.805. Antimicrob Agents 2008; 32:71-8; PMID:18757184; DOI:10.1002/pmic.200500253.
116. Wahlig H, Dingeldein E. Antibiotics and bone cements. DOI:10.1016/j.ijantimicag.2008.06.009. 145. Nandakumar R, Nandakumar MP, Marten MR, Ross
Experimental and clinical long-term observations. Acta 131. Joyce JG, Abeygunawardana C, Xu Q, Cook JC, JM. Proteome analysis of membrane and cell wall asso-
Orthop Scand 1980; 51:49-56; PMID:7376844; Hepler R, Przysiecki CT, et al. Isolation, struc- ciated proteins from Staphylococcus aureus. J Proteome
DOI:10.3109/17453678008990768. tural characterization and immunological evalua- Res 2005; 4:250-7; PMID:15822900; DOI:10.1021/
117. Klemm K. [Gentamicin-PMMA-beads in treating bone tion of a high-molecular-weight exopolysaccharide pr049866k.
and soft tissue infections (authors transl)]. Zentralbl from Staphylococcus aureus. Carbohydr Res 2003; 146. Musser JM. The next chapter in reverse vaccinology.
Chir 1979; 104:934-42; PMID:494865. 338:903-22; PMID:12681914; DOI:10.1016/S0008- Nat Biotechnol 2006; 24:157-8; PMID:16465156;
118. Wininger DA, Fass RJ. Antibiotic-impregnated cement 6215(03)00045-4. DOI:10.1038/nbt0206-157.
and beads for orthopedic infections. Antimicrob Agents 132. McKenney D, Hubner J, Muller E, Wang Y, Goldmann 147. Rodrguez-Ortega MJ, Norais N, Bensi G, Liberatori
Chemother 1996; 40:2675-9; PMID:9124821. DA, Pier GB. The ica locus of Staphylococcus epider- S, Capo S, Mora M, et al. Characterization and
119. Alt V, Bechert T, Steinrucke P, Wagener M, Seidel midis encodes production of the capsular polysac- identification of vaccine candidate proteins through
P, Dingeldein E, et al. An in vitro assessment of the charide/adhesin. Infect Immun 1998; 66:4711-20; analysis of the group A Streptococcus surface proteome.
antibacterial properties and cytotoxicity of nanopartic- PMID:9746568. Nat Biotechnol 2006; 24:191-7; PMID:16415855;
ulate silver bone cement. Biomaterials 2004; 25:4383- 133. Costa AR, Henriques M, Oliveira R, Azeredo J. The DOI:10.1038/nbt1179.
91; PMID:15046929; DOI:10.1016/j.biomateri- role of polysaccharide intercellular adhesin (PIA) in 148. Solis N, Larsen MR, Cordwell SJ. Improved accuracy
als.2003.10.078. Staphylococcus epidermidis adhesion to host tissues and of cell surface shaving proteomics in Staphylococcus
120. Boyce BM, Lindsey BA, Clovis NB, Smith ES, Hobbs subsequent antibiotic tolerance. Eur J Clin Microbiol aureus using a false-positive control. Proteomics
GR, Hubbard DF, et al. Additive effects of exogenous Infect Dis 2009; 28:623-9; PMID:19130107; 2010; 10:2037-49; PMID:20217865; DOI:10.1002/
IL-12 supplementation and antibiotic treatment in DOI:10.1007/s10096-008-0684-2. pmic.200900564.
infection prophylaxis. J Orthop Res 2011; In press; 134. Olson ME, Garvin KL, Fey PD, Rupp ME. 149. Vytvytska O, Nagy E, Bluggel M, Meyer HE,
PMID:21815205; DOI:10.1002/jor.21520. Adherence of Staphylococcus epidermidis to bioma- Kurzbauer R, Huber LA, et al. Identification of
121. Friedman A, Blecher K, Sanchez D, Tuckman-Vernon terials is augmented by PIA. Clin Orthop Relat Res vaccine candidate antigens of Staphylococcus aureus
C, Gialanella P, Friedman JM, et al. Susceptibility of 2006; 451:21-4; PMID:16906069; DOI:10.1097/01. by serological proteome analysis. Proteomics 2002;
Gram-positive and -negative bacteria to novel nitric blo.0000229320.45416.0c. 2:580-90; PMID:11987132; DOI:10.1002/1615-
oxide-releasing nanoparticle technology. Virulence 135. Arciola CR, Baldassarri L, Montanaro L. Presence 9861(200205)2:5<580::AID-PROT580>3.0.CO;2-G.
2011; 2:217-21; PMID:21577055; DOI:10.4161/ of icaA and icaD genes and slime production in a 150. Glowalla E, Tosetti B, Kronke M, Krut O. Proteomics-
viru.2.3.16161. collection of staphylococcal strains from catheter- based identification of anchorless cell wall proteins
122. Matsuzaki S, Rashel M, Uchiyama J, Sakurai S, Ujihara associated infections. J Clin Microbiol 2001; 39:2151- as vaccine candidates against Staphylococcus aureus.
T, Kuroda M, et al. Bacteriophage therapy: a revital- 6; PMID:11376050; DOI:10.1128/JCM.39.6.2151- Infect Immun 2009; 77:2719-29; PMID:19364833;
ized therapy against bacterial infectious diseases. J 6.2001. DOI:10.1128/IAI.00617-08.
Infect Chemother 2005; 11:211-9; PMID:16258815; 136. Knobloch JK, Horstkotte MA, Rohde H, Mack D. 151. Chhatwal GS. Anchorless adhesins and invasins of
DOI:10.1007/s10156-005-0408-9. Evaluation of different detection methods of biofilm Gram-positive bacteria: a new class of virulence factors.
123. Holden MT, Feil EJ, Lindsay JA, Peacock SJ, Day NP, formation in Staphylococcus aureus. Med Microbiol Trends Microbiol 2002; 10:205-8; PMID:11973142;
Enright MC, et al. Complete genomes of two clinical Immunol (Berl) 2002; 191:101-6; PMID:12410349; DOI:10.1016/S0966-842X(02)02351-X.
Staphylococcus aureus strains: evidence for the rapid DOI:10.1007/s00430-002-0124-3. 152. de Beer D, Stoodley P, Roe F, Lewandowski Z.
evolution of virulence and drug resistance. Proc Natl 137. Rohde H, Knobloch JK, Horstkotte MA, Mack D. Effects of biofilm structures on oxygen distribution and
Acad Sci USA 2004; 101:9786-91; PMID:15213324; Correlation of Staphylococcus aureus icaADBC genotype mass transport. Biotechnol Bioeng 1994; 43:1131-8;
DOI:10.1073/pnas.0402521101. and biofilm expression phenotype. J Clin Microbiol PMID:18615526; DOI:10.1002/bit.260431118.
124. Chua K, Seemann T, Harrison PF, Davies JK, Coutts 2001; 39:4595-6; PMID:11797608; DOI:10.1128/ 153. Brown MR, Allison DG, Gilbert P. Resistance of
SJ, Chen H, et al. Complete genome sequence of JCM.39.12.4595-6.2001. bacterial biofilms to antibiotics: a growth-rate related
Staphylococcus aureus strain JKD6159, a unique 138. Ziebuhr W, Heilmann C, Gotz F, Meyer P, Wilms K, effect? J Antimicrob Chemother 1988; 22:777-80;
Australian clone of ST93-IV community methicil- Straube E, et al. Detection of the intercellular adhesion PMID:3072331; DOI:10.1093/jac/22.6.777.
lin-resistant Staphylococcus aureus. J Bacteriol 2010; gene cluster (ica) and phase variation in Staphylococcus 154. Mader JTSM. The rabbit model of bacterial osteomy-
192:5556-7; PMID:20729356; DOI:10.1128/ epidermidis blood culture strains and mucosal isolates. elitis of the tibia. In: Zak OSMA, Ed. In Handbook
JB.00878-10. Infect Immun 1997; 65:890-6; PMID:9038293. of Animal Models of Infection London UK: Academic
125. Gill SR, Fouts DE, Archer GL, Mongodin EF, Deboy 139. Harro JM, Peters BM, OMay GA, Archer N, Kerns Press Ltd. 1999; 581-91.
RT, Ravel J, et al. Insights on evolution of virulence P, Prabhakara R, et al. Vaccine development in 155. Brady RA, OMay GA, Leid JG, Prior ML, Costerton
and resistance from the complete genome analysis Staphylococcus aureus: taking the biofilm phenotype JW, Shirtliff ME. Resolution of Staphylococcus aure-
of an early methicillin-resistant Staphylococcus aureus into consideration. FEMS Immunol Med Microbiol us biofilm infection using vaccination and antibi-
strain and a biofilm-producing methicillin-resistant 2010; 59:306-23; PMID:20602638. otic treatment. Infect Immun 2011; 79:1797-803;
Staphylococcus epidermidis strain. J Bacteriol 2005; 140. Rappuoli R. Reverse vaccinology. Curr Opin Microbiol PMID:21220484; DOI:10.1128/IAI.00451-10.
187:2426-38; PMID:15774886; DOI:10.1128/ 2000; 3:445-50; PMID:11050440; DOI:10.1016/
JB.187.7.2426-38.2005. S1369-5274(00)00119-3.
126. Howden BP, Seemann T, Harrison PF, McEvoy CR, 141. Lowe AM, Beattie DT, Deresiewicz RL. Identification
Stanton JA, Rand CJ, et al. Complete genome sequence of novel staphylococcal virulence genes by in vivo
of Staphylococcus aureus strain JKD6008, an ST239 expression technology. Mol Microbiol 1998;
clone of methicillin-resistant Staphylococcus aureus with 27:967-76; PMID:9535086; DOI:10.1046/j.1365-
intermediate-level vancomycin resistance. J Bacteriol 2958.1998.00741.x.
2010; 192:5848-9; PMID:20802046; DOI:10.1128/ 142. Coulter SN, Schwan WR, Ng EY, Langhorne MH,
JB.00951-10. Ritchie HD, Westbrock-Wadman S, et al. Staphylococcus
127. Diep BA, Gill SR, Chang RF, Phan TH, Chen JH, aureus genetic loci impacting growth and survival in
Davidson MG, et al. Complete genome sequence of multiple infection environments. Mol Microbiol 1998;
USA300, an epidemic clone of community-acquired 30:393-404; PMID:9791183; DOI:10.1046/j.1365-
meticillin-resistant Staphylococcus aureus. Lancet 2006; 2958.1998.01075.x.
367:731-9; PMID:16517273; DOI:10.1016/S0140-
6736(06)68231-7.

www.landesbioscience.com Virulence 459

Você também pode gostar