Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

International Journal of Medical Microbiology


journal homepage: www.elsevier.com/locate/ijmm

Biofilms: Survival and defense strategy for pathogens



Ashutosh Kumara,1, Anwar Alama,1, Mamta Ranib, Nasreen Z. Ehteshamc, Seyed E. Hasnaina,d,e,
a
Molecular Infection and Functional Biology Lab, Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, New Delhi, India
b
School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
c
National Institute of Pathology, Safdarjang Hospital Campus, New Delhi, India
d
JH-Institute of Molecular Medicine, Jamia Hamdard, New Delhi, India
e
Dr Reddy’s Institute of Life Sciences, University of Hyderabad Campus, Hyderabad, India

A R T I C L E I N F O A B S T R A C T

Keywords: Studies on biofilm related infections are gaining prominence owing to their involvement in majority of clinical
Biofilms infections. Biofilm, considered as a generic mechanism for survival used by pathogenic as well as non-pathogenic
Drug tolerance microorganisms, involves surface attachment and growth of heterogeneous cells encapsulated within a matrix.
Microbiota The matrix provides ecological niche where partnership of cells endows a community like behaviour that not
Mycobacteria
only enables the cohort to survive local microenvironment stress but also channelizes them to evolve, dis-
Biofilm regulators
Biofilm disrupters
seminate and cause resurgence of infections. In this mini-review we highlight the mechanisms used by microbes
to develop and sustain biofilms, including the influence of the microbiota. Several strategies to target biofilms
have been validated on certain groups of microorganisms and these basically target different stages in the life
cycle of biofilm, however comprehensive methods to target microbial biofilms are relatively unknown. In the
backdrop of recent reports suggesting that biofilms can harbour multiple species of organisms, we need to relook
and devise newer strategies against biofilms. Effective anti-biofilm strategies cannot be confined to a single
methodology that can disrupt one pathway but should simultaneously target the various routes adopted by the
microorganisms for survival within their ecosystem. An overview of the currently available drugs, their mode of
action, genomic targets and translational therapies against biofilm related infection are discussed.

1. Introduction abscessus (M. abscessus) etc., pose serious health concerns due to re-
calcitrant nature of microbes towards antimicrobial drugs and host
Majority of the microorganisms develop different types of survival immune responses.
mechanisms such as growth regulation, heterogeneity in population, Biofilms are a major health concern contributing to nearly 80% of
proteolytic systems etc. to adapt to stress conditions. Pathogenic mi- the recalcitrant hospital infections (Davies, 2003). Bacteria such as
croorganisms acquire the ability to sustain various host immunological Staphylococcus aureus (S. aureus), P. aeruginosa, Acinetobacter baumannii
responses. Biofilms are heterogeneous congregation of surface asso- and other clinically important microorganisms that thrive on medical
ciated microorganisms encapsulated within a self-produced polymer devices form biofilms which confer them with up to 1000 times more
matrix consisting of polysaccharide, protein and DNA. Several factors resistance and tolerance to antibiocides as compared to their planktonic
such as bacterial defense mechanism, suitable area for colonization, the forms. Nearly 80% of the pathogenic bacteria are associated with
community cooperation related benefits and extraordinary mode of medical device related infections such as ortho-dental prosthetics,
growth in their habitat accelerate biofilm formation (Jefferson, 2004). contact lenses, cardiovascular valves, urinary catheters, pacemakers
The matrix acts as a physical barrier against drugs and provides a and breast implants (Scott, 2009). Bacteria can also modulate the pH of
protective ecological niche for the survival of microorganisms. A single the environment in order to facilitate the formation of biofilm. Klebsiella
species of bacteria or consortia of multispecies microbes can exist and Pseudomonas increase the alkalinity of urine in order to form bio-
within a biofilm. Biofilm associated diseases caused by several species films on urinary catheters. These biofilms cause secondary complica-
of microorganisms such as Mycoplasma pneumoniae, Candida albicans (C. tions when they detach from the catheter causing tissue injury in ur-
albicans), Pseudomonas aeruginosa (P. aeruginosa), Staphylococcus epi- inary bladder (Neethirajan et al., 2014).
dermidis, Mycobacterium tuberculosis (M. tuberculosis), Mycobacterium Biofilm formation by M. tuberculosis for its survival within the host


Corresponding author at: Molecular Infection and Functional Biology Lab, Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, New Delhi, India.
E-mail addresses: seyedhasnain@gmail.com, seh@bioschool.iitd.ac.in (S.E. Hasnain).
1
These authors contributed equally to this work.

http://dx.doi.org/10.1016/j.ijmm.2017.09.016
Received 14 January 2017; Received in revised form 11 September 2017; Accepted 19 September 2017
1438-4221/ © 2017 Elsevier GmbH. All rights reserved.

Please cite this article as: Kumar, A., International Journal of Medical Microbiology (2017), http://dx.doi.org/10.1016/j.ijmm.2017.09.016
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

may be one of the main cause behind long term drug treatment of tu- surface topography and are facilitated by the presence of adhesins on
berculosis patients (Hopewell et al., 2006, 2014). The involvement of bacterial pili. The probability of making initial interaction within the
non-tuberculous mycobacteria (NTM) such as M. abscessus in biofilm substratum is influenced by hydrodynamic forces arising from the
formation in cystic fibrosis (CF) patients has been reported (Qvist et al., viscosity of medium and presence or absence of chemotactic molecules.
2013). CF is caused by mutation in the Cystic Fibrosis Transmembrane Antigen 43 of uropathogenic Escherichia coli (E. coli) mediates attach-
Conductance Regulator (CFTR) gene which is present on the long arm ment to the abiotic surface and also aids in intercommunication be-
of chromosome 7. The mutated/abnormal CFTR protein (a cAMP-acti- tween the bacteria of same species (Ulett et al., 2007). Mutant strains of
vated ion channel) leads to decreased chloride secretion and increased P. aeruginosa lacking CheR1 methyltransferase fail to express chemo-
sodium absorption across the epithelial surface. This results in thick- tactic amino acids that are required for surface sampling, attachment
ened mucus secretions in lung airways where bacteria are trapped and and maturation of biofilm (Schmidt et al., 2011). Flagellate bacteria can
colonize. M. abscessus exists in two phenotypic variants; the smooth encounter hydrodynamic stress and hence have the advantage to form
form expresses glycopeptidolipids whereas the rough form expresses biofilms. In the case of biotic surfaces, attachment of bacteria to the
minimal amounts of glycopeptidolipids (Howard et al., 2006). Different host surface is largely through the interaction of bacterial proteins with
studies have confirmed that smooth variant persists inside biofilm the extracellular proteins or carbohydrate moieties on the surface of the
whereas rough variant can cause persistent and acute infection in the cells/tissues and depends on the hydrophobicity of interacting cell
host (Catherinot et al., 2009; Bernut et al., 2014). M. abscessus can membranes. In P. aeruginosa, surface pili type IV aids in adherence and
convert itself bidirectionally into smooth and rough forms which have movement through the viscous cell surface (Klausen et al., 2003). Pili
been shown in vitro by routine plating (Byrd and Lyons, 1999; Howard type I adhesins, e.g. FimH, bind to mannosylated moieties on host cells
et al., 2006). This ability of M. abscessus perhaps allows it to colonize as and have been implicated in pathogenesis caused by uropathogenic E.
a smooth-morphotype in the lung airways of CF patient and later cause coli (Wright et al., 2007). Several other types of adhesion molecules,
acute respiratory failure upon emergence of an isogenic rough-mor- such as Sag, have since been reported in Enterococcus spp. that bind to
photype (Catherinot et al., 2009). collagen of eukaryotic cells and provide focal points for aggregation
Besides these, the microbiota flourishes along the epithelial walls or and adherence to eukaryotic cells, leading to biofilm formation
exposed tissue surface and locally form biofilms inside the host body (Mohamed et al., 2006). S. epidermidis and S. aureus express microbial
within wounds, urogenital system, respiratory tract or as dental pla- surface components recognizing adhesive matrix molecules that attach
ques. Such biofilms, besides causing local infections also predispose non-covalently to fibronectin, fibrinogen and human matrix proteins
individuals to secondary long term manifestations. For example, (Fey and Olson, 2010). Upon contact with the surface, auto-inducer
Porphyromonas gingivalis causes periodontal biofilms within dental signals initiate the switchover of gene expression that leads to upre-
plaques that also play a major role in the progression of cardiovascular gulation of factors required for sessile colony formation. Master reg-
diseases (Leishman et al., 2010). Several clinically relevant biofilm ulator switches, such as PrfA and SinR, have provided insights into
forming bacteria are listed in Table 1. mechanisms regulating genetic reprogramming in Listeria mono-
cytogenes and Bacillus subtilis biofilms, respectively (Luo et al., 2013a,b;
2. Biofilm stages Newman et al., 2013). The role of small non-coding RNAs (sRNAs) in
regulating the switch between planktonic to biofilm mode and vice
Bacterial biofilms can form on abiotic surfaces as well as on biotic versa is evident from the observation that sequestration of RNA reg-
surfaces and proceed through distinct stages which can be broadly ca- ulatory proteins, such as RsmA with sRNAs, switches development of
tegorized into reversible adhesion stage and irreversible cohesion stage. planktonic form into biofilm stages in P. aeruginosa (Ventre et al.,
In vitro models of biofilm formation does not necessarily represent the 2006).
outcome in in vivo models due to the difference in host environment Attachment of Mycobacterium smegmatis to the substratum induces
where the challenges are different in terms of nutrient availability, synthesis of alginate, glycopeptidolipid and small chain mycolic acids
immune stress etc. Microbial cells within the biofilm evolve multiple aided by the activity of chaperonic proteins such as GroEL-1 (Davies
mechanisms to proceed through the four stages of biofilm development, et al., 1993; Recht and Kolter, 2001). Mycolic acids, being hydrophobic,
namely, (i) attachment to surface, (ii) sessile growth, (iii) colonization, allow non-covalent binding with host cell proteins such as fibronectin,
and (iv) dispersal (Fig. 1). Unique set of genes are expressed in each fibrinogen, collagen IV etc. Alteration in mycolic acid synthesis through
stage of biofilm formation that enable production of extracellular ma- disruption of polyketide synthatase affects both formation of biofilm
trix consisting of polysaccharides, glycopeptidolipids, proteins and fatty and virulence (Ojha et al., 2008; Pang et al., 2012). The attractive or
acids. During the initial stages of reversible adhesion, bacterium can repulsive phenomenon between bacteria in the niche depends on the
aggregate loosely but can dissociate and revert back to planktonic availability of nutrients, pH, presence of ions and temperature. Once
forms. The initial stages of abiotic biofilm formation are driven by the bacterium attaches to the surface they interact with other bacteria
electrostatic interactions between the substratum and the bacteria, of the same species or other species which initiates the process of

Table 1
List of clinically relevant biofilm forming bacteria.

Location Organism Reference

Lungs of patients with Cystic Fibrosis Pseudomonas aeruginosa, Staphylococcus aureus, Haemophilus influenza, Burkholderia cepacia complex Lopes et al. (2015)
Pacemakers Staphylococci, Streptococci Greenspon et al. (2008)
Prosthetic heart valves Staphylococcus epidermidis, Staphylococcus aureus, Streptococci, Gram-negative bacilli, Diphtheroids, Enterococci Donlan (2001)
and Candida spp.
Wounds (equine) Staphylococci, Bacillus, Enterococci, Pseudomonas, Clostridium etc. Westgate et al. (2011)
Wounds (dog) Staphylococcus intermedius, Staphylococcus epidermidis, Streptococcus canis Swanson et al. (2014)
Contact Lenses Pseudomonas aeruginosa, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus saprophyticus, El-Ganiny et al. (2017)
Klebsiella spp.
Orthopaedic implants Staphylococci, Pseudomonas genus, Enterococci, Streptococci Arciola et al. (2015)
Breast implants Staphylococcus epidermidis, Pseudomonas aeruginosa Seng et al. (2015)
Dental Streptococcus mutans, Streptococcus sanguinis, Porphyromonas gingivalis, Fusobacterium nucleatum, Lactobacillus Huang et al. (2011)
casei, Actinomyces naeslundii

2
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

Fig. 1. Combination of antibiotic and biofilm dis-


ruptive agents modulate biofilm developmental
stages. Microorganisms undergo four definitive
stages for completion of biofilm cycle involving ad-
herence, sessile growth, colonization and dispersal.
Biofilm matrix impedes entry and interferes with the
interaction of antibiotic with the cell. Strategies di-
rected against biofilm formation involve use of an-
tibiotics in combination with biofilm disrupters,
specific for developmental stages (i–iv). Attachment
of planktonic form to the surface (i) can be in-
capacitated by using silver coatings. Microbial com-
munities coordinate using quorum sensing (QS) and
grow as sessile structure (ii) encapsulated within a
matrix. Use of inhibitors (lactoferrin, DNase,
Clarithromycin) against quorum sensing molecules
and matrix components disrupts sessile growth.
Colonization of persister cells require transcription of
genes – PKA and PNAG, (iii) which may be silenced
using siRNA. Nutritional limitations induce stress
like conditions and favour dispersal of persister cells
(iv). Use of biofilm nano-disrupters (Fe3O4, D-amino
acids) and stress inducers (NOS) can facilitate dis-
ruption of matrix allowing antibiotics to enter the
biofilm shield and act on cells.

cohesion or attachment among microbial cells to form microcolonies. virulence factors. Nutrient availability is an important determinant in
Consequent to these events, bacteria are irreversibly attached to the shape and size of biofilm. A gradient in nutrient and oxygen from the
substratum and can overcome hydrodynamic stresses by virtue of their outer to the inner core causes variability in metabolic activity of cells
colonization and maturation. Motility of bacteria is reduced and the and biodiversification. The outer layer of cells grows faster as compared
cells secrete exopolysaccharides that aid in entrapping nutrients. to the inner core. In such an environment, starved cells achieve static
Transition of single cell into group of cells necessitates the need to growth and hence their numbers get depleted. Such dormant cells
develop intercommunication so as to coordinate growth and maximize achieve antibiotic tolerance and are difficult to detect in chronic biofilm
efficiency. Bacteria coordinate growth through quorum sensing system infections. Sub-inhibitory concentrations of Imipenem or Tobramycin
using several signalling peptides and molecules such as competence may induce alginate production in P. aeruginosa (Bagge et al., 2004).
stimulating peptide, 3,5-cyclic diguanylic acid (c-di-GMP), farnesol, The hallmark of biofilm formation is to achieve greater co-ordination.
rhamnolipids, phenol-soluble modulins etc. Quorum sensing helps to Cells in the outer layer of C. albicans express genes associated with
maintain optimum cell density as high as 107cells/cm2, required for synthesis of proteins required for glycolysis, cholesterol and cell wall
virulence of Vibrio chlorea (Deep et al., 2011). Quorum sensing in gram whereas cells in the inner core express genes associated with enzymes
positive and negative bacteria is predominantly mediated through small required for sulphur metabolism or degradation of the cell wall. This is
peptides and acyl-homoserine lactones, respectively (Taga and Bassler, indicative of the fact that biofilm is an active congregation of cells that
2003). In C. albicans, high levels of farnesol inhibit mycelium conver- have attained the sufficiency to modulate their metabolic engine as
sion and facilitate active division by budding (Hornby et al., 2001). In required in response to the microenvironment challenge.
P. aeruginosa, rhamnolipids are required for intercellular interactions The last phase of biofilm culminates into detachment of cells that
and maintenance of open channels between cellular aggregates that aid causes dissemination of single or clustered cells. The dispersal of the
in distribution of nutrients (Davey et al., 2003). bacterium from the biofilm is not a passive process, rather it is a highly
Studies in uropathogenic E. coli show that cellulose and curli amy- orchestered event that leads to release of specific cells at the final stages
loid fibers are essential for pellicle formation (Cegelski et al., 2009). In of biofilm life cycle. Cells along the inner core of the biofilms lyse prior
patients suffering from CF, P. aeruginosa upregulates proteins such as to their dispersal. These lysed cells provide nutrients to the bacterium
Pel that play crucial role in attachment to the epithelial surface. During that would undergo dispersal at a later stage. The cells undergoing
the colony formation stage new set of proteins such as Psl and CdrA dispersal shut down the genes encoding exopolysaccharides or fimbriae
adhesins are upregulated in response to high levels of signalling mo- and simultaneously upregulate genes encoding chemotactic proteins or
lecules, c-di-GMP, which reinforce matrix components and stabilize flagella required for planktonic lifestyle (Rollet et al., 2009).
pellicle formation (Ma et al., 2006; Borlee et al., 2010). Several proteins
associated with motility and fimbriae formation, lipopolysaccharide
synthesis and cell wall surface are involved in biofilm formation in 3. Biofilm components
bacteria (Niba et al., 2007; Cucarella et al., 2001; Geoghegan et al.,
2010). The composition of the matrix has been studied in bacteria such as
Colonization of bacteria induces secretion of exopolysaccharides, P. aeruginosa, Bacillus spp., Staphylococcus spp. and Streptococcus spp.
proteins, extra-cellular DNA (eDNA) etc. that not only aid in structural (Lopez et al., 2010). The constituents of extracellular matrix depend on
stability but also enhance substrate exchange and distribution of nu- the environment and the bacteria present within the biofilm (Fig. 2).
trients. The components of the matrix vary according to the bacterial During the process of transition from a free living planktonic stage into
species involved in biofilm formation. The genes on plasmids encoding a congregation of cells, bacteria require greater coordination to attain
biofilm proteins are horizontally transferred and aid in transmission of maximal efficiency within the limited space and nutrition. Although
quorum sensing molecules are secreted by free living bacteria as well,

3
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

Fig. 2. Extracellular matrix of bacterial biofilms:


Extracellular matrix mainly consists of extracellular
polysaccharides, proteins and extracellular DNA that
are essential for the formation and maintenance of
biofilms. The nature and composition of biofilms
vary between different species and also within
strains of same species and with the environment
(Lopez et al., 2010). The composition of extracellular
matrix of three bacterial species Pseudomonas aeru-
ginosa, Bacillus subtilis and Staphylococcus aureus are
well studied. Extracellular matrix of Pseudomonas
aeruginosa consists of exopolysaccharides, surface
proteins, lectin binding proteins and extracellular
DNA. Extracellular matrix of Bacillus subtilis consists
of exopolysaccharide, Poly-gamma-Glutamate and
extracellular proteins. Extracellular matrix of Sta-
phylococcus aureus consists of polymer of N-acetyl glucosamine, adhesive proteins and extracellular DNA.

these molecules hold greater significance in biofilm biology as they are membrane protein LapG, causing dispersal of cells. P. aeuroginisa uti-
considered viable targets for drugs and could play a crucial role in lizes rhamnolipids to cause dispersal of Klebsiella pneumonia, S. aureus
tackling biofilms. In P. aeruginosa, rhanmnolipids not only act as and C. albicans (Davies and Marques, 2009).
quorum sensor but also block chemotactic activity of polymorpho-
nuclear (PMN) cells and induce necrosis. This is evident from the fact
that newly established biofilms are more susceptible to PMN attack as 5. Biofilm in drug tolerance and persister formation
compared to mature biofilms (O’Loughlin et al., 2013). The main con-
stituents of biofilm produced by P. aeruginosa are polysaccharides such Within biofilm, cells exhibit morphological, replicative and phy-
as Psl, Pel and alginate, that increase extracellular polymeric matrix siological heterogeneity aided by differential gene expression due to the
hydration and decrease flexibility of biofilm (Orgad et al., 2011). Many gradient in diffusible gases, toxic components or nutritional pressure
proteins identified in biofilms are associated with outer membrane of (Stewart, 2003). Depleted nutrient and oxygen within biofilm promote
vesicle and are either secretory or cytoplasmic in nature (Toyofuku asynchronous growth of non-replicative cohorts which exhibit fluc-
et al., 2012). tuations in gene expression and may lead to drug tolerance. Phenotypic
The polymeric components of extracellular matrix entrap alginate, diversity of bacteria within biofilm not only enhances greater co-
extracellular amyloids, iron, pili etc. which contribute to adherence, ordination but also empower them to reprogramme genes required for
hydration, rigidity and matrix formation. Consequently, the extra- efflux of toxins, DNA repair, ion sequestration, lipid biosynthesis, host
cellular matrix provides physical barrier and subdue direct effects of immune modulation etc., which in turn provides persistence and se-
antibiotics on bacterial cells (Orgad et al., 2011; Alsteens et al., 2012; lective propagation of resilient cells enduring stress (Anderson et al.,
Ramsugit et al., 2013). The recalcitrant nature of biofilm is a cumula- 2003; Lewis, 2005; Fux et al., 2005; Lebeaux et al., 2014).
tive effect of inertness of polymeric extracellular components and Within biofilms, fraction of bacteria evolve as persister cells that are
adaptability of bacteria against antibiotics. Physical barrier of matrix genetically similar but are physiologically different compared to parent
leads to a decreasing gradient of drugs along the outer surface towards cells. Persisters are metabolically inert, replicate slowly, modulate
inner core of biofilm (Stewart, 2003). Bacteria depend on host iron to toxin-antitoxin system, upregulate DNA repair and anti-oxidative ma-
upregulate polysaccharide intercellular adhesin (PIA) molecules re- chinery, have enhanced phosphate metabolism, and exhibit unrespon-
quired for fibrin mediated biofilm formation on implanted biomaterials siveness towards minimal inhibitory concentrations of antibiotics
(Lin et al., 2012). Lactoferrin, an iron binding protein, reduces iron (Lewis, 2010). Drug treatment normally kills planktonic cells and the
supply necessary for the survival of bacteria. Bacteria subvert the effect majority of biofilm cells. However, drug tolerant persisters repopulate
of lactoferrin by secretion of proteases against lactoferrin (Schmidtchen the biofilm, disseminate into planktonic forms and start a new cycle of
et al., 2002). biofilm development (Lewis, 2010; Zhang, 2014; Keren et al., 2011)
that increases the duration of treatment of diseases caused by biofilm
forming pathogenic microorganisms (Fig. 3). Low oxygen condition
4. Biofilm regulators
induces biofilm whereas normoxia decreases biofilm formation (Totani
et al., 2017). Enhanced bacterial respiration reduces the persisters in
The master regulatory proteins trigger genes that control expression
bacterial population (Vilchèze et al., 2017).
of factors required for biofilm formation. Several master regulator
Toxin-antitoxin systems that may control bacterial growth and
genes such as transcription activator PrfA in Listeria monocytogenes and
metabolism induce the persister phenotype, pointing to the role of
flagella regulator SinR in Bacillus subtilis have been identified that play
several other growth regulatory proteins of M. tuberculosis in drug tol-
crucial role in biofilm (Luo et al., 2013a,b; Newman et al., 2013). These
erance (Wang and Wood, 2011; Kumar et al., 2017). Mycobacterial
changes in gene expression pattern weaken the argument that biofilm
proteins that inhibit translation by interacting with ribosome (Trauner
formation is part of developmental stage utilized by bacteria to com-
et al., 2012; Kumar et al., 2012; Li et al., 2015) possibly induce persister
plete their life cycle. Rather it is now apparent that biofilm formation is
phenotype and drug tolerance because it has been reported that protein
a process by virtue of which microorganisms alter their phenotype to
synthesis inhibition by tetracycline induces persisters cells and aids in
adapt to environmental stresses or immune responses.
survival (Kwan et al., 2013). The M. tuberculosis PpiB found in the se-
Transcriptomic analysis of genes expressed during various stages of
cretory proteome and membrane fraction (González-Zamorano
biofilm formation fails to provide a consensus pathway due to the im-
et al.,2009; Xiong et al., 2005; Souza et al., 2011) is also involved in
mense physiological heterogeneity and diversity of species harboring
biofilm formation. PpiB can be a viable target for developing new
the biofilm. The consistent factor in the regulation of biofilms is c-di-
strategies for intervention against drug resistant TB (Pandey et al.,
GMP that helps in aggregation of cells and a decrease in its level causes
2016; Hasnain et al., 2017).
dispersal of cells from biofilms. The process of dispersal of bacteria from
the biofilms is also regulated. In Pseudomonas putida, diminishing levels
of c-di-GMP triggers cysteine proteinase activity that degrades

4
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

complement proteins C3b or deposition of IgG on the surface of the


bacterium and provides resistance to phagocytosis by neutrophils. This
has a positive implication because inhibition of biofilm formation could
render the pathogen susceptible to complement mediated lysis (Kristian
et al., 2008). eDNA, generated by autolysis of bacteria, helps in colo-
nization of sessile form, suppresses host innate immune response, in-
creases tolerance to antibiotics and aids in virulence (Nguyen et al.,
2010; Qin et al., 2007; Madsen et al., 2012; Thurlow et al., 2011; Rose
et al., 2015). A combination of DNase with antibiotics such as AM-228,
Moxifloxacin or Clarithromycin, have been shown to be more effective
in mycobacterial killing as compared to antibiotic alone, suggesting
that eDNA can be a suitable target against biofilm formation (Aung
et al., 2016).
Polymicrobial interactions can be either synergistic or antagonistic.
C albicans can coaggregate symbiotically with S. aureus such that the
Candida hyphae penetrate the epithelial barrier and provide route for
entry of S. aureus. The symbiotic association increases virulence and
metabolic cooperation (Peters et al., 2010).

7. Anti biofilm approaches

It has been noted that antibiotic tolerant bacteria residing within


biofilms become susceptible to antibiotics upon dispersal from biofilm
suggesting that resilience towards antibiotics is due to phenotypic
Fig. 3. Mechanism of drug tolerance through biofilm: Fractions of bacteria in biofilms are adaptability and not essentially due to genetic adaptability (Fig. 1)
present as persisters that enhance drug tolerance. Drug treatment normally kills plank- (Anwar et al., 1989). Factors such as mechanical stress, enzymatic di-
tonic cells and the majority of biofilm cells and when drug concentration drops, persister gestion, pH, oxygen availability, temperature and limiting nutrition
bacteria re-populate the biofilm due to their drug tolerance nature. Hypoxia enhances
trigger dispersal of cells from the biofilm. Use of silver coating on
pellicle formation whereas normoxia decreases pellicle formation (Totani et al., 2017).
Enhanced bacterial respiration due to presence of cysteine or other small thiols causes
medical devices prevents surface attachment of bacteria. Clari-
decrease in persisters/drug tolerance in bacterial population (Vilchèze et al., 2017). The thromycin blocks biofilm matrix formation in P. aeruginosa (Yasuda
reduction in drug tolerance of bacterial infection shortened disease treatment duration. et al., 1993). Ciprofloxacin reduces the overall thickness of the biofilm
Verified and putative roles are shown with solid and dotted lines, respectively. and exposes the immature biofilm to phagocytosis by polymorpho-
nuclear neutrophils. Streptokinase dissolves the matrix polymer of
6. Evasion of immune responses biofilm in S. aureus (Nemoto et al., 2000). Acyl-homoserine lactone
interferes with cellular signalling mechanisms used for quorum sensing
Adherent bacteria cannot be opsonised easily and block signalling in and affects normal biofilm formation (Parsek and Greenberg, 2000).
polymorphonuclear leukocytes (Gunther et al., 2009; Chorell et al., Epigallocatechin gallate and polyphenol in green tea remodels amyloid
2012; Piatek et al., 2013). In patients with CF, P. aeruginosa produces fibrils in P. aeruginosa biofilm and increases the antibiotic potency
alginate and rhamnolipids to evade macrophages and induction of ne- (Stenvang et al., 2016). As a result, immune cells can easily phagocy-
crosis in polymorphonuclear leukocytes, respectively (Leid et al., 2005; tose bacterial cells or potency of drugs are restored. Vaccine prepared
Van Gennip et al., 2009). S. aureus escapes phagocytosis in blood by from strong biofilm forming bacteria induces antibodies against Poly-N-
producing coagulase that activates prothrombin and polymerizes acetyl-β-glucosamine (PNAG) and improves tolerance against S. aureus
monomeric fibrinogen into insoluble fibrin, a constituent of biofilm infections (Pérez et al., 2009).
matrix (Cheng et al., 2010). Upon contact with macrophages, S. aureus Within the microbiota, different species of microorganism can co-
biofilms release lytic toxins that skew differentiation of activated exist and may either show synergism or compete for space and nutri-
macrophages into M2 lineage. The defection towards M2 lineage re- tion. The facultative anaerobic C. albicans and anaerobic Clostridium
duces inducible nitric oxide synthase (iNOS) and increases arginase 1 difficle (C. difficle) are opportunistic pathogens that reside within the
(enzyme involved in collagen synthesis), consequently leading to fi- human gut. In the presence of C. albicans, C. difficle can grow in
brosis and evasion of recognition by Toll-like receptors (Hanke and anaerobic conditions, which would otherwise be toxic. C. difficle in turn
Kielian, 2012). secretes p-cresol that modulates hyphae formation and ultimately in-
Cells within biofilms monitor the microenvironment within and hibits biofilm formation in C. albicans (van Leeuwen et al., 2016). Non-
outside the physical boundaries to evade host immune responses or pathogenic microbiota can release antimicrobial molecules in the niche
utilize the host machinery to propagate. Biofilms of M. tuberculosis of pathogenic microbes and block adhesion sites normally used by pa-
develop within host tissues to survive the prolonged drug challenge in thogenic microbes. This type of symbiotic association used by non-pa-
TB patients. NTMs, such as M. abscessus and Mycobacterium avium, cause thogenic microbiota increases competition for food or space and also
secondary manifestations in CF patients (Hopewell et al., 2006, 2014; elicits immune response via IgM, that differs in reactivity to pathogens.
Qvist et al., 2013). Within the macrophage, M. tuberculosis requires Within the symbiotic environment of the microbiota, a balance in po-
eukaryotic serine/threonine protein kinase G (PKG) to evade fusion pulation between non-pathogenic and pathogenic microorganisms ex-
with the lysosome. PKG associates with mycobacterial nudix hydro- ists. Disturbing this balance through antibiotics that selectively target
xylase (RenU) and accelerates hydrolysis of NADH or FAD which pathogenic microorganisms may lead to a skewed overpopulation of
maintains an optimum redox potential in macrophage. Disruption of non-pathogenic microorganisms, thereby diminishing the chances of
PKG-RenU pathway impairs mycobacterial sensitivity to oxidative biofilm formation by pathogenic microorganism. The intestinal biofilm
stress, impairs biofilm growth and reduces survival of M. tuberculosis is heavily colonised by wide varieties of bacteria such as Bacteroides
within macrophages (Wolff et al., 2015). fragilis group, enterobacteria etc., that act as cellular factories for the
Staphylococcus epidermidis biofilm interferes with the activation of breakdown of fatty acid and production of metabolic cofactors (vitamin
B12). These bacteria play an important role in host immunity by

5
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

secreting antimicrobial peptides or competing with pathogenic micro- quorum inhibitors, nanoparticles, biofilm disrupters and antibiotics to
organisms for space and nutrition (Macfarlane et al., 2005). Treatment simultaneously target various aspects of microbial physiology can
with antibiotics may sometimes disturb the gut microflora and cause possibly attenuate their survival.
susceptibility to infection caused by Clostridium spp. (Buffie et al.,
2012). The gut symbiota (probiotics) play an important role in main- 8. Conclusion
taining microbial composition, metabolism and immunity of gut by
immunomodulating systemic immunity and pH (Singh et al., 2013). Although a wide array of antibiotics have been developed since the
They compete with pathogens for binding sites and neutralize toxins discovery of Penicillin nearly a century ago, the knockout punch re-
released by pathogens. Microbiota as probiotics have potentials for use quired for eradicating infections is still eluding researchers. The use of
against biofilms associated with dental plaque, chronic wounds and antibiotics can successfully suppress infection but in majority of cases,
urogenital infections (Singh and Hasnain, 2014; Vuotto et al., 2014). there has been a resurgence of infections. Although survival mechan-
The extracellular matrix proteins, when injected intradermally in isms employed by microorganisms have been substantively deciphered
mice, induce humoral immune response and increase Th-2 response. and their genomic or proteomic targets identified, rapid evolution of
These mechanisms facilitate phagocytosis of S. aureus and limits bac- microorganisms outpace the current profile of chemotherapeutics.
teria, undergoing dispersive stage of biofilm cycle, within the tissue (Gil Initial drug resistance endowed by mutations in genes were supposed to
et al., 2014). Addition of antibiotics during this treatment regime may contribute to resilience of infections, but research over the last decade
help in an enhanced clearance of bacteria. Although use of whole has shown that biofilms are formed by microorganism across the
protein lysate from biofilm may induce a non-specific immune re- spectrum and have been identified in majority of clinical infections.
sponse, recombinant form of specific antigens such as Biofilm asso- Cells within biofilms exhibit differential gene expression which pro-
ciated protein (Bap), accumulation associated protein (Aap) and surface vides heterogeneity to the cohort in space and time. Thus, a certain
exposed protein induce production of specific antibodies and suppress genetic pathway active in one cell may be shut off in other cells of the
bacterial growth (Fattahian et al., 2011; Shahrooei et al., 2012; Yan cohort and subvert the effectiveness of a singular therapeutic strategy.
et al., 2014). Besides, biofilm matrix impedes interaction of drugs with microorgan-
Antibiotics such as Pyrazinamide shorten treatment duration by isms and provides a protective barrier against immune surveillance.
depleting membrane energy and inhibit trans-translation in persister Within biofilm, a small subpopulation of cells evolve into phenotypi-
cells (Zhang et al., 2003; Shi et al., 2011). The ability to target persisters cally dormant persister cells that later disseminate from the biofilm,
can significantly improve efforts in eradicating biofilm related infec- show enhanced tolerance to drugs and lead to recalcitrance of infec-
tions. It has been reported that cis-2-decanoic acid reverts persister cells tions. The mechanistics of biofilm formation have clearly outlined de-
to drug-susceptible state by improving metabolic activity. A combina- finitive pathways involving a cycle of attachment, sessile growth, co-
tion of cis-2-decanoic acid and antimicrobials could thus inhibit bac- lonization and dissipation. A silver lining to this observation is that
terial viability (Marques et al., 2014). Bacteria develop resistance to several genes involved in biofilm cycle are common in both pathogenic
current antibiotics by modifying regulatory pathways that are the tar- as well as non-pathogenic genre and offer an exciting platform for
gets of antibiotics, synthesize enzymes for degrading antibiotics, de- targeting using inhibitors. It is hypothesised that targeting biofilms
velop efflux mechanisms (Siddiqi et al., 2004), modify cell wall com- could prove to be the masterstroke required for tackling a wide spec-
ponents etc. (Wright, 2005). Nanoparticles can be easily modified for trum of microorganisms, particularly those involved in pathogenesis in
surface charge and ligand binding properties. They exhibit anti- humans. A multi-targeted strategy against biofilm formation would
microbial activity by affecting the cell permeability, generation of re- therefore limit resilience of infections commonly observed in hospital
active oxygen species or by competitive inhibition of key enzymes. infections.
Since their mode of action is completely different as compared to an-
tibiotics, bacteria have lesser chance of developing resistance against Funding
nanoparticles. Catalytic nanoparticles containing Fe3O4 have shown
promising peroxidase like activity and trigger formation of free radicals SEH and NZE thank the DBT, Ministry of S & T, Government of India
that disrupt biofilm matrix and kill bacteria encapsulated within the for a Centre of Excellence Grant.
biofilm matrix (Gao et al., 2016). The different infection stages of
mycobacteria confer differential susceptibility towards antibiotics, the Competing interest
inactive stationary phase being more resilient towards drugs. While
antibiotics are effective in inhibiting the growth of bacteria by targeting The authors declare that we have no conflict of interests.
key transcriptional, translational or metabolic pathways, their efficacy
is redundant on non-living polymeric extracellular matrix. Anionic al- Acknowledgements
ginates bind to positively charged antibiotics such as fluoroquinolones
or aminoglycosides, impede the penetration of antibiotics and thus re- SEH is a JC Bose National Fellow, Department of Science and
quire higher inhibitory concentration of drugs. P. aeruginosa possesses Technology, Government of India and Robert Koch Fellow, Robert Koch
high affinity siderophores that aid in iron uptake from the host. In CF Institute, Berlin, Germany. AK is a recipient of Research Associateship
patients, free iron is often recorded in sputum and clinical isolates of from the Centre of Excellence Grant (BT/PR12817/COE/34/23/2015)
lungs (Smith et al., 2013). Use of metal chelators or nanomolar con- to SEH and NZE from the Department of Biotechnology (DBT), Ministry
centrations of D-amino acids for dispersal of biofilm prior to treatment of Science and Technology, Government of India. AA is a recipient of
with drugs are emerging as novel strategies for combating biofilm Research Associateship from Department of Biotechnology, GoI, India.
problem (Banin et al., 2006; Kolodkin-Gal et al., 2010).
Since aggregation of cells constitutes the first step in biofilm for- References
mation, strategies to disrupt aggregation by use of quorum sensing in-
hibitors would suppress formation of multicellular structure. There has Alsteens, D., Ramsook, C.B., Lipke, P.N., Dufrêne, Y.F., 2012. Unzipping a functional
been an alarming increase in the number of resistant pathogens due to microbial amyloid. ACS Nano 6 (9), 7703–7711.
Anderson, G.G., Palermo, J.J., Schilling, J.D., Roth, R., Heuser, J., Hultgren, S.J., 2003.
indiscriminate use of antibiotics and the inherent ability of micro- Intracellular bacterial biofilm-like pods in urinary tract infections. Science 301,
organisms to evolve in order to subvert drug targets, induce immune 105–107.
skewness or form biofilm resistance. Whereas antibiotics primarily slow Anwar, H., van Biesen, T., Dasgupta, M., Lam, K., Costerton, J.W., 1989. Interaction of
biofilm bacteria with antibiotics in a novel in vitro chemostat system. Antimicrob.
down the growth of microbes, newer strategies combining the use of

6
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

Agents Chemother. 33, 1824–1826. endocarditis: the important role of methicillin-resistant Staphylococcus aureus. PACE
Arciola, C.R., Campoccia, D., Ehrlich, G.D., Montanaro, L., 2015. Biofilm-based implant – Pacing Clin. Electrophysiol. 31, 548–553. http://dx.doi.org/10.1111/j.1540-8159.
infections in orthopaedics. Adv. Exp. Med. Biol. 830, 29–46. http://dx.doi.org/10. 2008.01039.x.
1007/978-3-319-11038-7_2. Gunther, F., Wabnitz, G.H., Stroh, P., Prior, B., Obst, U., Samstag, Y., Wagner, C., Hansch,
Aung, T.T., Yam, J.K.H., Lin, S., Salleh, S.M., Givskov, M., et al., 2016. Biofilms of pa- G.M., 2009. Host defense against Staphylococcus aureus biofilms infection: phago-
thogenic nontuberculous mycobacteria targeted by new therapeutic approaches. cytosis of biofilms by polymorphonuclear neutrophils (PMN). Mol. Immunol. 46,
Antimicrob. Agents Chemother. 60, 24–35. 1805–1813.
Bagge, N., Schuster, M., Hentzer, M., Ciofu, O., Givskov, M., Greenberg, E.P., Høiby, N., Hanke, M.L., Kielian, T., 2012. Deciphering mechanisms of Staphylococcal biofilm eva-
2004. Pseudomonas aeruginosa biofilms exposed to imipenem exhibit changes in sion of host immunity. Front. Cell. Infect. Microbiol. 2, 62.
global gene expression and β-Lactamase and alginate production. Antimicrob. Agents Hasnain, S.E., Ehtesham, N.Z., Tripathi, D., Grover, S., Kumar, A., Alam, A., Pandey, S.,
Chemother. 48, 1175–1187. http://dx.doi.org/10.1128/AAC.48.4.1175-1187.2004. 2017. A medicament for the treatment of diseases by biofilm forming microorgan-
Banin, E., Brady, K.M., Greenberg, E.P., 2006. Chelator-induced dispersal and killing of isms. Indian Patent 201711014310. April 21.
Pseudomonas aeruginosa cells in a biofilm. Appl. Environ. Microbiol. 72 (3), Hopewell, P.C., Pai, M., Maher, D., Uplekar, M., Raviglione, M.C., 2006. International
2064–2069. standards for tuberculosis care. Lancet Infect. Dis. 6 (11), 710–725.
Bernut, A., Herrmann, J.-L., Kissa, K., Dubremetz, J.-F., Gaillard, J.-L., Lutfalla, G., Hopewell, P.C., Fair, E.L., Uplekar, M., 2014. Updating the international standards for
Kremer, L., 2014. Mycobacterium abscessus cording prevents phagocytosis and pro- tuberculosis care. Entering the era of molecular diagnostics. Ann. Am. Thorac. Soc.
motes abscess formation. Proc. Natl. Acad. Sci. U. S. A. 111, E943–E952. http://dx. 11, 277–285. http://dx.doi.org/10.1513/AnnalsATS.201401-004AR.
doi.org/10.1073/pnas.1321390111. Hornby, J.M., Jensen, E.C., Lisec, A.D., Tasto, J.J., Jahnke, B., Shoemaker, R., Dussault,
Borlee, B.R., Goldman, A.D., Murakami, K., Samudrala, R., Wozniak, D.J., Parsek, M.R., P., Nickerson, K.W., 2001. Quorum sensing in the dimorphic fungus candida albicans
2010. Pseudomonas aeruginosa uses a cyclic-di-GMP-regulated adhesin to reinforce is mediated by farnesol. Appl. Environ. Microbiol. 67, 2982–2992. http://dx.doi.org/
the biofilm extracellular matrix. Mol. Microbiol. 75, 827–842. http://dx.doi.org/10. 10.1128/AEM.67.7.2982-2992.2001.
1111/j.1365-2958.2009.06991.x. Howard, S.T., Rhoades, E., Recht, J., Pang, X., Alsup, A., Kolter, R., Lyons, C.R., Byrd,
Buffie, C.G., Jarchum, I., Equinda, M., Lipuma, L., Gobourne, A., Viale, A., Ubeda, C., T.F., 2006. Spontaneous reversion of Mycobacterium abscessus from a smooth to a
Xavier, J., Pamer, E.G., 2012. Profound alterations of intestinal microbiota following rough morphotype is associated with reduced expression of glycopeptidolipid and
a single dose of clindamycin results in sustained susceptibility to Clostridium difficile- reacquisition of an invasive phenotype. Microbiology 152, 1581–1590. http://dx.doi.
induced colitis. Infect. Immun. 80, 62–73. http://dx.doi.org/10.1128/IAI.05496-11. org/10.1099/mic.0.28625-0.
Byrd, T.F., Lyons, C.R., 1999. Preliminary characterization of a Mycobacterium abscessus Huang, R., Li, M., Gregory, R.L., 2011. Bacterial interactions in dental biofilm. Virulence
mutant in human and murine models of infection. Infect. Immun. 67, 4700–4707. 2, 435–444. http://dx.doi.org/10.4161/viru.2.5.16140.
Catherinot, E., Roux, A.L., Macheras, E., Hubert, D., Matmar, M., Dannhoffer, L., Chinet, Jefferson, K.K., 2004. What drives bacteria to produce a biofilm? FEMS Microbiol. Lett.
T., Morand, P., Poyart, C., Heym, B., Rottman, M., Gaillard, J.L., Herrmann, J.L., 236 (2), 163–173. http://dx.doi.org/10.1016/j.femsle.2004.06.005.
2009. Acute respiratory failure involving an R variant of mycobacterium abscessus. J. Keren, I., Minami, S., Rubin, E., Lewis, K., 2011. Characterization and transcriptome
Clin. Microbiol. 47, 271–274. http://dx.doi.org/10.1128/JCM.01478-08. analysis of mycobacterium tuberculosis persisters. MBio 2, e00100–e00111.
Cegelski, L., Pinkner, J.S., Hammer, N.D., Cusumano, C.K., Hung, C.S., Chorell, E., Åberg, Klausen, M., Heydorn, A., Ragas, P., Lambertsen, L., Aaes-Jørgensen, A., Molin, S., Tolker-
V., Walker, J.N., Seed, P.C., Almqvist, F., Chapman, M.R., Hultgren, S.J., 2009. Small- Nielsen, T., 2003. Biofilm formation by Pseudomonas aeruginosa wild type, flagella
molecule inhibitors target Escherichia coli amyloid biogenesis and biofilm formation. and type IV pili mutants. Mol. Microbiol. 48, 1511–1524. http://dx.doi.org/10.1046/
Nat. Chem. Biol. 5, 913–919. http://dx.doi.org/10.1038/nchembio.242. j.1365-2958.2003.03525.x.
Cheng, A.G., McAdow, M., Kim, H.K., Bae, T., Missiakas, D.M., Schneewind, O., 2010. Kolodkin-Gal, I., Romero, D., Cao, S., Clardy, J., Kolter, R., Losick, R., 2010. D-amino
Contribution of coagulases towards Staphylococcus aureus disease and protective acids trigger biofilm disassembly. Science 328 (5978), 627–629.
immunity. PLoS Pathog. 6 (8), e1001036. Kristian, S.A., Birkenstock, T.A., Sauder, U., Mack, D., Gotz, F., Landmann, R., 2008.
Chorell, E., Pinkner, J.S., Bengtsson, C., Edvinsson, S., Cusumano, C.K., et al., 2012. Biofilm formation induces C3a release and protects staphylococcus epidemidis from
Design and synthesis of fluorescent pilicides and curlicides: bioactive tools to study IgG and complement deposition and from neutrophil dependent killing. J. Inf. Dis.
bacterial virulence mechanisms. Chem. Eur. J. 18, 4522–4532. 197, 1028–1035.
Cucarella, C., Solano, C., Valle, J., Amorena, B., Lasa, I., Penadé, J.R., 2001. Bap, a Kumar, A., Majid, M., Kunisch, R., Rani, P.S., Qureshi, I.A., Lewin, A., Hasnain, S.E.,
Staphylococcus aureus surface protein involved in biofilm formation. J. Bacteriol. Ahmed, N., 2012. Mycobacterium tuberculosis DosR regulon gene Rv0079 encodes a
183, 2888–2896. http://dx.doi.org/10.1128/JB.183.9.2888-2896.2001. putative, dormancy associated translation inhibitor (DATIN). PLoS One 7. http://dx.
Davey, M.E., Caiazza, N.C., O’Toole, G.A., 2003. Rhamnolipid surfactant production af- doi.org/10.1371/journal.pone.0038709.
fects biofilm architecture in Pseudomonas aeruginosa PAO1. J. Bacteriol. 185, Kumar, A., Rani, M., Ehtesham, N.Z., Hasnain, S.E., 2017. Commentary: modification of
1027–1036. http://dx.doi.org/10.1128/JB.185.3.1027-1036.2003. host responses by mycobacteria. Front. Immunol. 8, 466. http://dx.doi.org/10.3389/
Davies, D.G., Marques, C.N.H., 2009. A fatty acid messenger is responsible for inducing fimmu.2017.00466.
dispersion in microbial biofilms. J. Bacteriol. 191, 1393–1403. http://dx.doi.org/10. Kwan, B.W., Valenta, J.A., Benedik, M.J., Wood, T.K., 2013. Arrested protein synthesis
1128/JB.01214-08. increases persister-like cell formation. Antimicrob. Agents Chemother. 57,
Davies, D.G., Chakrabarty, A.M., Geesey, G.G., 1993. Exopolysaccharide production in 1468–1473. http://dx.doi.org/10.1128/AAC.02135-12.
biofilms: substratum activation of alginate gene expression by Pseudomonas aeru- Lebeaux, D., Ghigo, J.M., Beloin, C., 2014. Biofilm-related infections: bridging the gap
ginosa. Appl. Environ. Microbiol. 59, 1181–1186. between clinical management and fundamental aspects of recalcitrance towards an-
Davies, D., 2003. Understanding biofilm resistance to antibacterial agents. Nat. Rev. Drug tibiotics. Microbiol. Mol. Biol. Rev. 78, 510–543.
Discov. 2, 114–122. http://dx.doi.org/10.1038/nrd1008. Leid, J.G., Willson, C.J., Shirtliff, M.E., Hassett, D.J., Parsek, M.R., Jeffers, A.K., 2005. The
Deep, A., Chaudhary, U., Gupta, V., 2011. Quorum sensing and bacterial pathogenicity: exopolysaccharide alginate protects Pseudomonas aeruginosa biofilm bacteria
from molecules to disease. J. Lab Physicians. 3 (1), 4–11. fromIFN-gamma-mediatedmacrophage killing. J. Immunol. 175, 7512–7518.
Donlan, R.M., 2001. Biofilms and device-associated infections. Emerging Infectious Leishman, S.J., Do, H.L., Ford, P.J., 2010. Cardiovascular disease and the role of oral
Diseases. pp. 277–281. http://dx.doi.org/10.3201/eid0702.700277. bacteria. J. Oral Microbiol. 21, 2.
El-Ganiny, A.M., Shaker, G.H., Aboelazm, A.A., El-Dash, H.A., 2017. Prevention of bac- Lewis, K., 2005. Persister cells and the riddle of biofilm survival. Biochemistry 70,
terial biofilm formation on soft contact lenses using natural compounds. J. 267–274.
Ophthalmic. Inflamm. Infect. 7 (December (1)), 11. http://dx.doi.org/10.1186/ Lewis, K., 2010. Persister cells. Annu. Rev. Microbiol. 64, 357–372.
s12348-017-0129-0. Epub 2017 Apr 18. Li, X., Sun, Q., Jiang, C., Yang, K., Hung, L.W., Zhang, J., Sacchettini, J.C., 2015.
Fattahian, Y., Rasooli, I., Mousavi Gargari, S.L., Rahbar, M.R., Darvish Alipour Astaneh, Structure of ribosomal silencing factor bound to mycobacterium tuberculosis ribo-
S., Amanih, J., 2011. Protection against Acinetobacter baumannii infection via its some. Structure 23, 1858–1865. http://dx.doi.org/10.1016/j.str.2015.07.014.
functional deprivation of biofilm associated protein (Bap). Microb. Pathog. 51, Lin, M.H., Shu, J.C., Huang, H.Y., Cheng, Y.C., 2012. Involvement of iron in biofilm
402–406. formation by Staphylococcus aureus. PLoS One 7. http://dx.doi.org/10.1371/
Fey, P.D., Olson, M.E., 2010. Current concepts in biofilm formation of Staphylococcus journal.pone.0034388.
epidermidis. Future Microbiol. 5, 917–933. http://dx.doi.org/10.2217/fmb.10.56. Lopes, S.P., Azevedo, N.F., Pereira, M.O., 2015. Microbiome in cystic fibrosis: shaping
Fux, C.A., Costerton, J.W., Stewart, P.S., Stoodley, P., 2005. Survival strategies of in- polymicrobial interactions for advances in antibiotic therapy. Crit. Rev. Microbiol.
fectious biofilms. Trends Microbiol. 13, 34–40. 41, 353–365. http://dx.doi.org/10.3109/1040841X.2013.847898.
Gao, L., Liu, Y., Kim, D., Li, Y., Hwang, G., Naha, P.C., Cormode, D.P., Koo, H., 2016. Lopez, D., Vlamakis, H., Kolter, R., 2010. Biofilms. Cold Spring Harb Perspect Biol. 2,
Nanocatalysts promote streptococcus mutans matrix degradation and enhance bac- a000398. http://dx.doi.org/10.1101/cshperspect.a000398.
terial killing to suppress dental caries in vivo. Biomaterials 101, 272–284. Luo, Q., Shang, J., Feng, X., Guo, X., Zhang, L., Zhou, Q., 2013a. PrfA led to reduced
Geoghegan, J.A., Corrigan, R.M., Gruszka, D.T., Speziale, P., O’Gara, J.P., Potts, J.R., biofilm formation and contributed to altered gene expression patterns in biofilm-
Foster, T.J., 2010. Role of surface protein SasG in biofilm formation by forming Listeria monocytogenes. Curr. Microbiol. 67, 372.
Staphylococcus aureus. J. Bacteriol. 192, 5663–5673. http://dx.doi.org/10.1128/JB. Luo, Q., Shang, J., Feng, X., Guo, X., Zhang, L., Zhou, Q., 2013b. PrfA led to reduced
00628-10. biofilm formation and contributed to altered gene expression patterns in biofilm-
Gil, C., Solano, C., Burgui, S., Latasa, C., García, B., Toledo-Arana, A., et al., 2014. Biofilm forming Listeria monocytogenes. Curr. Microbiol. 67 (September (3)), 372–378.
matrix exoproteins induce a protective immune response against Staphylococcus Ma, L., Jackson, K.D., Landry, R.M., Parsek, M.R., Wozniak, D.J., 2006. Analysis of
aureus biofilm infection. Infect. Immun. 82, 1017–1029. Pseudomonas aeruginosa conditional psl variants reveals roles for the psl poly-
González-Zamorano, M., Hernández, G.M., Xolalpa, W., et al., 2009. Mycobacterium tu- saccharide in adhesion and maintaining biofilm structure postattachment. J.
berculosis glycoproteomics based on ConA-lectin affinity capture of mannosylated Bacteriol. 188, 8213–8221. http://dx.doi.org/10.1128/JB.01202-06.
proteins. J. Proteome Res. 8, 721–733. Macfarlane, S., Woodmansey, E.J., Macfarlane, G.T., 2005. Colonization of mucin by
Greenspon, A.J., Rhim, E.S., Mark, G., Desimone, J., Ho, R.T., 2008. Lead-associated human intestinal bacteria and establishment of biofilm communities in a two-stage

7
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

continuous culture system. Appl. Environ. Microbiol. 71 (11), 7483–7492. et al., 2012. Vaccination with SesC decreases Staphylococcus epidermidis biofilm
Marques, C.N., Morozov, A., Planzos, P., Zelaya, H.M., 2014. The fatty acid signalling formation. Infect. Immun. 80, 3660–3668.
molecule cis-2-decenoic acid increases metabolic activity and reverts persister cells to Shi, W., Zhang, X., Jiang, X., Yuan, H., Lee, J.S., Barry, C.E., Wang, H., Zhang, W., Zhang,
an antimicrobial-susceptible state. Appl. Environ. Microbiol. 80 (22), 6976–6991. Y., 2011. Pyrazinamide inhibits trans-translation in Mycobacterium tuberculosis.
Mohamed, J.A., Teng, F., Nallapareddy, S.R., Murray, B.E., 2006. Pleiotrophic effects of 2 Science 333, 1630–1632.
Enterococcus faecalissagA-like genes, salA and salB, which encode proteins that are Siddiqi, N., Das, R., Pathak, N., Banerjee, S., Ahmed, N., Katoch, V.M., Hasnain, S.E.,
antigenic during human infection, on biofilm formation and binding to collagen type 2004. Mycobacterium tuberculosis isolate with a distinct genomic identity over-
I and fibronectin. J. Infect. Dis. 193, 231–240. expresses a TAP like efflux pump. Infection 32, 989–1012.
Neethirajan, S., Clond, M.A., Vogt, A., 2014. Medical biofilms-nanotechnology ap- Singh, Y., Hasnain, S.E., 2014. Holobionts: emerging strategy for interventions against
proaches. J. Biomed. Nanotechnol. 10, 2806–2827. http://dx.doi.org/10.1166/jbn. infectious diseases, metabolic disorders and cancers. Indian J. Med. Res. 140 (1),
2014.1892. 11–14.
Nemoto, K., Hirota, K., Ono, T., et al., 2000. Effect of varidase (streptokinase) on biofilm Singh, Y., Ahmad, J., Musarrat, J., Ehtesham, N.Z., Hasnain, S.E., 2013. Emerging im-
formed by Staphylococcus aureus. Chemotherapy 46, 111–115. portance of holobionts in evolution and in probiotics. Gut Pathog. 5 (1), 12.
Newman, J.A., Rodrigues, C., Lewis, R.L., 2013. Molecular basis of the activity of SinR Smith, D.J., Lamont, I.L., Anderson, G.J., Reid, D.W., 2013. Targeting iron uptake to
protein, the master regulator of biofilm formation in bacillus subtilis. J. Biol. Chem. control Pseudomonas aeruginosa infections in cystic fibrosis. Eur. Respir. J. http://
288, 10766–10778. dx.doi.org/10.1183/09031936.00124012.
Nguyen, K.T., Piastro, K., Gray, T.A., Derbyshire, K.M., 2010. Mycobacterial biofilms Souza, G.A.de., Leversen, N.A., Målen, H., Wiker, H.G., 2011. Bacterial proteins with
facilitate horizontal DNA transfer between strains of Mycobacterium smegmatis. J. cleaved or uncleaved signal peptides of the general secretory pathway. J. Proteomics
Bacteriol. 192, 5134–5142. 75 (2), 502–510.
Niba, E.T.E., Naka, Y., Nagase, M., Mori, H., Kitakawa, M., 2007. A genome-wide ap- Stenvang, M., Dueholm, M.S., Vad, B.S., Seviour, T.W., Zeng, G., et al., 2016.
proach to identify the genes involved in biofilm formation in E. coli. DNA Res. 14, Epigallocatechin gallate remodels overexpressed functional amyloids in
237–246. http://dx.doi.org/10.1093/dnares/dsm024. Pseudomonas aeruginosa and increases biofilm susceptibility to antibiotic treatment.
O’Loughlin, C.T., Miller, L.C., Siryaporn, A., Drescher, K., Semmelhack, M.F., Bassler, J. Biol. Chem. 291, 26540–26553.
B.L., 2013. A quorum-sensing inhibitor blocks Pseudomonas aeruginosa virulence Stewart, P.S., 2003. Diffusion in biofilms. J. Bacteriol. http://dx.doi.org/10.1128/jb.185.
and biofilm formation. Proc. Natl. Acad. Sci. U. S. A. 110, 17981–17986. http://dx. 5.1485-1491.2003.
doi.org/10.1073/pnas.1316981110. Swanson, E.A., Freeman, L.J., Seleem, M.N., Snyder, P.W., 2014. Biofilm-infected wounds
Ojha, A.K., Hatfull, G.F., Baughn, A.D., Sambandan, D., Hsu, T., et al., 2008. Growth of in a dog. J. Am. Vet. Med. Assoc. 244, 699–707. http://dx.doi.org/10.2460/javma.
Mycobacterium tuberculosis biofilms containing free mycolic acids and harbouring 244.6.699.
drug-tolerant bacteria. Mol. Microbiol. 69 (1), 164–174. Taga, M.E., Bassler, B.L., 2003. Chemical communication among bacteria. Proc. Natl.
Orgad, O., Oren, Y., Walker, S.L., Herzberg, M., 2011. The role of alginate in Acad. Sci. U. S. A. 100, 14549–14554. http://dx.doi.org/10.1073/pnas.1934514100.
Pseudomonas aeruginosa EPS adherence, viscoelastic properties and cell attachment. Thurlow, L.R., Hanke, M.L., Fritz, T., Angle, A., Aldrich, A., et al., 2011. Staphylococcus
Biofouling 27, 787–798. http://dx.doi.org/10.1080/08927014.2011.603145. aureus biofilms prevent macrophage phagocytosis and attenuate inflammation in
Pérez, M.M., Prenafeta, A., Valle, J., Penadés, J., Rota, C., Solano, C., et al., 2009. vivo. J. Immunol. 186, 6585–6596.
Protection from Staphylococcus aureus mastitis associated with poly- N-acetyl b-1,6 Totani, T., Nishiuchi, Y., Tateishi, Y., Yoshida, Y., Kitanaka, H., Niki, M., Kaneko, Y.,
glucosamine specific antibody production using biofilmembedded bacteria. Vaccine Matsumoto, S., 2017. Effects of nutritional and ambient oxygen condition on biofilm
27, 2379–2386. formation in Mycobacterium avium subsp. hominissuis via altered glycolipid ex-
Pandey, S., Sharma, A., Tripathi, D., Kumar, A., Khubaib, M., et al., 2016. Mycobacterium pression. Sci. Rep. 7, 41775. http://dx.doi.org/10.1038/srep41775.
tuberculosis peptidyl-Prolyl isomerases also exhibit chaperone like activity in-Vitro Toyofuku, M., Roschitzki, B., Riedel, K., Eberl, L., 2012. Identification of proteins asso-
and In-Vivo. PLoS One 11 (3), e0150288. ciated with the pseudomonas aeruginosa biofilm extracellular matrix. J. Proteome
Pang, J.M., Layre, E., Sweet, L., Sherrid, A., Moody, D.B., Ojha, A., Sherman, D.R., 2012. Res. 11, 4906–4915. http://dx.doi.org/10.1021/pr300395j.
The polyketide Pks1 contributes to biofilm formation in Mycobacterium tuberculosis. Trauner, A., Lougheed, K.E.A., Bennett, M.H., Hingley-Wilson, S.M., Williams, H.D.,
J. Bacteriol. 194, 715–721. 2012. The dormancy regulator DosR controls ribosome stability in hypoxic myco-
Parsek, M.R., Greenberg, E.P., 2000. Acyl-homoserine lactone quorum sensing in gram- bacteria. J. Biol. Chem. 287, 24053–24063. http://dx.doi.org/10.1074/jbc.M112.
negative bacteria: a signaling mechanism involved in associations with higher or- 364851.
ganisms. Proc. Natl. Acad. Sci. U. S. A. 97, 8789–8793. Ulett, G.C., Valle, J., Beloin, C., Sherlock, O., Ghigo, J.M., Schembri, M.A., 2007.
Peters, B.M., Jabra-Rizk, M.A., Scheper, M.A., Leid, J.G., Costerton, J.W., Shirtliff, M.E., Functional analysis of antigen 43 in uropathogenic Escherichia coli reveals a role in
2010. Microbial interactions and differential protein expression in Staphylococcus long-term persistence in the urinary tract. Infect. Immun. 75, 3233–3244. http://dx.
aureus—Candida albicans dual-species biofilms. FEMS Immunol. Med. Microbiol. 59, doi.org/10.1128/IAI.01952-06. IAI.01952-06[pii]\n.
493–503. http://dx.doi.org/10.1111/j.1574-695X.2010.00710.x. Van Gennip, M., Christensen, L.D., Alhede, M., Phipps, R., Jensen, P.O., et al., 2009.
Piatek, R., Zalewska-Piatek, B., Dzierzbicka, K., Makowiec, S., Pilipczuk, J., et al., 2013. Inactivation of the rhlA gene in Pseudomonas aeruginosa prevents rhamnolipid
Pilicides inhibit the FGL chaperone/usher assisted biogenesis of the Dr fimbrial production, disabling the protection against polymorphonuclear leukocytes. APMIS
polyadhesin from uropathogenic Escherichia coli. BMC Microbiol. 13, 131–142. 117, 537–546.
Qin, Z., Ou, Y., Yang, L., Zhu, Y., Tolker Nielsen, T., Molin, S., Qu, D., 2007. Role of Ventre, I., Goodman, A.L., Vallet-Gely, I., et al., 2006. Multiple sensors control reciprocal
autolysinmediated DNA release in biofilm formation of Staphylococcus epidermidis. expression of Pseudomonas aeruginosa regulatory RNA and virulence genes. Proc.
Microbiology 153, 2083–2092. Natl. Acad. Sci. U. S. A. 103 (1), 171–176.
Qvist, T., Eickhardt-Sørensen, S., Pressler, T., Katzenstein, T.L., Andersen, C.B., Iversen, Vilchèze, C., Hartman, T., Weinrick, B., Jain, P., Weisbrod, T.R., Leung, L.W., Freundlich,
M., Bjarnsholt, T., Hoiby, N., 2013. First evidence of Mycobacterium abscessus bio- J.S., Jacobs, W.R., 2017. Enhanced respiration prevents drug tolerance and drug
film in the lungs of chronically infected CF patients. J. Cyst. Fibros. 12 (Suppl. 1), S2. resistance in Mycobacterium tuberculosis. Proc. Natl. Acad. Sci. U. S.A. 201704376.
Ramsugit, S., Guma, S., Pillay, B., Jain, P., Larsen, M.H., Danaviah, S., Pillay, M., 2013. http://dx.doi.org/10.1073/pnas.1704376114.
Pili contribute to biofilm formation in vitro in Mycobacterium tuberculosis. Antonie Vuotto, C., Longo, F., Donelli, G., 2014. Probiotics to counteract biofilm-associated in-
van Leeuwenhoek. Int. J. Gen. Mol. Microbiol. 104, 725–735. http://dx.doi.org/10. fections: promising and conflicting data. Int. J. Oral Sci. 6 (4), 189–194.
1007/s10482-013-9981-6. Wang, X., Wood, T.K., 2011. Toxin-antitoxin systems influence biofilm and persister cell
Recht, J., Kolter, R., 2001. Glycopeptidolipid acetylation affects sliding motility and formation and the general stress response. Appl. Environ. Microbiol. 77, 5577–5583.
biofilm formation in Mycobacterium smegmatis. J. Bacteriol. 183, 5718–5724. http://dx.doi.org/10.1128/AEM.05068-11.
Rollet, C., Gal, L., Guzzo, J., 2009. Biofilm-detached cells, a transition from a sessile to a Westgate, S.J., Percival, S.L., Knottenbelt, D.C., Clegg, P.D., Cochrane, C.A., 2011.
planktonic phenotype: a comparative study of adhesion and physiological char- Microbiology of equine wounds and evidence of bacterial biofilms. Vet. Microbiol.
acteristics in Pseudomonas aeruginosa. FEMS Microbiol. Lett. 290, 135–142. http:// 150, 152–159. http://dx.doi.org/10.1016/j.vetmic.2011.01.003.
dx.doi.org/10.1111/j.1574-6968.2008.01415.x. Wolff, K.A., Peria, A.H., Nguyen, H.T., Pham, T.H., Amzel, A.M., Gabelli, S.B., Nguyen, L.,
Rose, S.J., Babrak, L.M., Bermudez, L.E., 2015. Mycobacterium avium possess extra- 2015. A redox regulatory system critical for mycobacterial survival in macrophages
cellular DNA that contributes to biofilm formation structural integrity and tolerance and biofilm development. PLoS Pathog. 11 (4), e1004839.
to antibiotics. PLoS One 10 (5), e0128772. Wright, G.D., 2005. Bacterial resistance to antibiotics: enzymatic degradation and mod-
Schmidt, J., Müsken, M., Becker, T., Magnowska, Z., Bertinetti, D., Möller, S., ification. Adv. Drug Deliv. Rev. 57 (10), 1451–1470.
Zimmermann, B., Herberg, F.W., Jänsch, L., Häussler, S., 2011. The Pseudomonas Wright, K.J., Seed, P.C., Hultgren, S.J., 2007. Development of intracellular bacterial
aeruginosa chemotaxis methyltransferase CheR1 impacts on bacterial surface sam- communities of uropathogenic Escherichia coli depends on type 1 pili. Cell Microbiol.
pling. PLoS One 6. http://dx.doi.org/10.1371/journal.pone.0018184. 9, 2230–2241.
Schmidtchen, A., Frick, I.M., Andersson, E., Tapper, H., Bjorck, L., 2002. Proteinases of Xiong, Y., Chalmers, M.J., Gao, F.P., Cross, T.A., Marshall, A.G., 2005. Identification of
common pathogenic bacteria degrade and inactivate the antibacterial peptide LL-37. Mycobacterium tuberculosis H37Rv integral membrane proteins by one-dimensional
Mol. Microbiol. 46, 157–168. gel electrophoresis and liquid chromatography electrospray ionization tandem mass
Scott, R.D.I., 2009. The Direct Medical Costs of Healthcare-Associated Infections in U.S. spectrometry. J. Proteome Res. 4 (3), 855–861.
Hospitals and the Beneftis of Prevention. Division of Healthcare Quality Promotion Yan, L., Zhang, L., Ma, H., Chiu, D., Bryers, J.D., 2014. A single B-repeat of
National Center for Preparedness, Detection and Control of Infectious Diseases. Staphylococcus epidermidis accumulation-associated protein induces protective im-
Seng, P., Bayle, S., Alliez, A., Romain, F., Casanova, D., Stein, A., 2015. The microbial mune responses in an experimental biomaterial-associated infection mouse model.
epidemiology of breast implant infections in a regional referral centre for plastic and Clin. Vaccine Immunol. 21, 1206–1214.
reconstructive surgery in the south of France. Int. J. Infect. Dis. Jun 35, 62–66. Yasuda, H., Ajiki, Y., Koga, T., Kawada, H., Yokota, T., 1993. Interaction between biofilms
http://dx.doi.org/10.1016/j.ijid.2015.04.010. formed by Pseudomonas aeruginosa and clarithromycin. Antimicrob. Agents
Shahrooei, M., Hira, V., Khodaparast, L., Stijlemans, B., Kucharíková, S., Burghout, P., Chemother. 37, 1749–1755.

8
A. Kumar et al. International Journal of Medical Microbiology xxx (xxxx) xxx–xxx

Zhang, Y., Wade, M.M., Scorpio, A., Zhang, H., Sun, Z., 2003. Mode of action of pyr- van Leeuwen, P.T., van der Peet, J.M., Bikker, F.J., Hoogenkamp, M.A., Oliveira Paiva,
azinamide: disruption of Mycobacterium tuberculosis membrane transport and en- A.M., Kostidis, S., Mayboroda, O.A., Smits, W.K., Krom, B.P., 2016. Interspecies in-
ergetics by pyrazinoic acid. J. Antimicrob. Chemother. 52, 790–795. teractions between clostridium difficile and candida albicans. mSphere 1 (6) pii:
Zhang, Y., 2014. Persisters, persistent infections and the Yin-Yang model. Emerg. Microb. e00187-16.
Infect. 3, e3.

You might also like