You are on page 1of 19

Infection, Genetics and Evolution 112 (2023) 105459

Contents lists available at ScienceDirect

Infection, Genetics and Evolution


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

Review

Efflux pumps and microbial biofilm formation


Mahdyeh Neghabi Hajiagha a, Hossein Samadi Kafil b, *
a
Department of Microbiology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
b
Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran

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

Keywords: Biofilm-related infections are resistant forms of pathogens that are regarded as a medical problem, particularly
Biofilm due to the spread of multiple drug resistance. One of the factors associated with biofilm drug resistance is the
Efflux pumps presence of various types of efflux pumps in bacteria. Efflux pumps also play a role in biofilm formation by
Drug resistance
influencing Physical-chemical interactions, mobility, gene regulation, quorum sensing (QS), extracellular poly­
Infection
meric substances (EPS), and toxic compound extrusion. According to the findings of studies based on efflux pump
Pathogenesis
expression analysis, their role in the anatomical position within the biofilm will differ depending on the biofilm
formation stage, encoding gene expression level, the type and concentration of substrate. In some cases, the
function of the efflux pumps can overlap with each other, so it seems necessary to accurate identify the efflux
pumps of biofilm-forming bacteria along with their function in this process. Such studies will help to choose
treatment strategy, at least in combination with antibiotics. Furthermore, if the goal of treatment is an efflux
pump manipulation, we should not limit it to inhibition.

1. Introduction ((p)ppGpp signaling) (Irving and Corrigan, 2018; Pacios et al., 2020),
toxin-antitoxin (TA) system (Ronneau and Helaine, 2019), drug efflux
Antibiotic resistance has emerged in bacteria, raising the problem of pumps (Kumar et al., 2020), the quorum sensing (QS) system as bacterial
recalcitrant infections (Aslam et al., 2018; Ventola, 2015). Multidrug communication process (Mukherjee and Bassler, 2019).
resistant (MDR) bacteria is a public health crisis that negatively affects Biofilms are constituted of several different microbial species (pol­
the clinical outcomes, particularly in immunocompromised patients or ymicrobial) (Kafil and Mobarez, 2015a; Wolcott et al., 2013) that are
those hospitalized in intensive care units (Bialvaei et al., 2016; Vivas contained in a three-dimensional extracellular matrix made up of
et al., 2019) (Chen et al., 2020; Gasperini et al., 2021; Wang et al., 2017; extracellular polymeric substances (EPS) (Karygianni et al., 2020) and
Wang et al., 2019b). The rapid increase in the development of genetic work synergistically. This matrix, which is known as “safe haven”, serves
basis of antibiotic resistance has been regarded as the primary cause of as a biological barrier against antimicrobial agents and immune com­
this phenomenon. However, bacteria are smart organized microorgan­ ponents, while still having a secure ecological niche for microorganisms
isms that they can also enter to a physiologically dormant state and to survive (Kumar et al., 2019). Indeed, biofilm communities shelter
display phenotypic tolerance to antibiotic treatment. They can manage resistant and persister cells that can tolerate antibiotics or antimicrobial
complicated situations in different environments and correlation with agents and regrow when the antibiotic is withdrawn (Conlon et al.,
other competitions. So, in order to survive and colonise effectively, they 2015; Fisher et al., 2017). Microbial biofilm formation is now recog­
employ a variety of strategies. (Maisonneuve and Gerdes, 2014). Several nized as a significant virulence factor in several localized primary in­
important molecular mechanisms whereby bacteria use them to tolerate fections (Kafil and Mobarez, 2015b; Kafil et al., 2013; Mohamad et al.,
and persistence in stress conditions include: RpoS (Schellhorn, 2020) 2023). Nevertheless, evidence that tolerant and persister bacteria play a
and the general stress response (Hecker and Völker, 2001), the SOS significant role in biofilm-related bacterial relapse and infection recal­
response (Zgur Bertok and Podlesek, 2020), tolerance to reactive oxygen citrance has been increasing in recent years (Yan and Bassler, 2019).
species (ROS) (Johnson and Hug, 2019), energy metabolism/cell Efflux pumps are proteins found in the cytoplasmic membrane of all
respiration(oxygen oxidoreduction by Cytochrome bd) (Borisov et al., cells, including bacteria and eukaryotes (Van Bambeke et al., 2003a;
2020) and Tau metabolism (Javaux et al., 2007), the stringent response Van Bambeke et al., 2003b). These are responsible for secreting toxins or

* Corresponding author at: Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
E-mail address: kafilhs@tbzmed.ac.ir (H.S. Kafil).

https://doi.org/10.1016/j.meegid.2023.105459
Received 13 April 2023; Received in revised form 25 May 2023; Accepted 27 May 2023
Available online 2 June 2023
1567-1348/© 2023 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-
nc-nd/4.0/).
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

antibiotics formed by the cell, as well as the efflux of toxic compounds 2.2. Biofilm formation
found in the bacterial context, such as antibiotics (Marquez, 2005).
Despite the fact that bacterial efflux pumps play a significant role in The development of anti-biofilm strategies is based on a thorough
bacterial resistance, their involvement in bacterial infection dissemi­ comprehension of the biofilm formation process and biofilm-related
nation via biofilm formation cannot be overlooked (Ebbensgaard et al., mechanical parameters. (Boudarel et al., 2018). Biofilm formation can
2020; Webber and Piddock, 2003). Therefore, in current study, we will be induced by a multitude of endogenous and environmental criteria,
investigate the development of biofilm, the effect of bacterial efflux including bacterial cell density, nutritional status, and cellular stress,
pumps on it, and the associated antimicrobial resistance in order to which mostly varies throughout bacteria (Landini et al., 2010; O’Toole
develop a novel treatment strategy, such as multi-targeted or combi­ et al., 2000). Naturally bacteria must approach a surface sufficiently
national therapies. close (10–20 nm) to enhance their chances of adhering. They cling to a
surface either passively or actively, depending on whether they are
2. Biofilm nonmotile bacteria, such as staphylococci or motile bacteria, such as
Pseudomonas aeruginosa (Joo and Otto, 2012). Despite the repellent
William J. Costerton coined the word "biofilm" to describe the forces between the bacteria and the environmental surfaces, attractive
aggregate existence of bacteria adhering to surfaces in 1978 (Costerton van der Waals and electrostatic interactions and the mechanical force of
et al., 1978). Biofilm analysis has developed into a common subject of flagella and fimbriae preparations on the other hand provides the link­
research within microbiology since the beginning of that accomplish­ age (Palmer et al., 2007).
ment (Koo and Yamada, 2016). At that time, the number of articles Biofilm development cycle proceeds in 3 main stages (Fig. 1): 1.
published on biofilm and public health topics has increased from 1459 in Establishment; consist of 2 phases involving 1.1. Reversible attachment,
1992 to 7303 in 2004–2016 (Donlan, 2016). In this regard biofilm also known as early adhesion in which bacteria use extracellular or­
known as architectural colony made by microorganisms such as pro­ ganelles and proteins, such as flagella, pili, fimbriae, curli fibres, and
karyotes (bacteria) and eukaryotes such as fungi and microalgae (mostly outer membrane proteins, to sense and attach to surfaces. It is also
revealed) (Ramage et al., 2012; Yuan et al., 2021) that have adapted influenced by hydrophobic interactions and surface texture. The intra­
their metabolism for the respective environment (Yuan et al., 2021). The cellular levels of (c-diGMP) can affect to regulation of this process
key cause of cell attachment to surfaces and aggregation is the extra­ (Hengge, 2009). 1.2. Irreversible attachment between bacteria and tis­
cellular matrix, which is the consequence of the cells’ natural production sue or inorganic texture and as well as cell-cell adhesion is enabled by
of extracellular polymeric substance (EPS). The multiple gradients that secretion of an extracellular polymeric substance (EPS) or slime that
occur within biofilm matrix create a micro-niche (heterogeneous mi­ consists of DNA (eDNA), proteins, lipids, and lipopolysaccharides,
croenvironments) and provide a framework of cell-to-cell interactions siderophores between cells and surfaces (Flemming and Wingender,
(Flemming et al., 2016). 2010; Passos da Silva et al., 2017). 2. The second phase is growth, which
has two stages: proliferation and maturation. 2.1. Proliferation that in
this process, adherent bacteria multiply and expand to form three-
2.1. Biofilm-related infections: dimensional microcolonies (10–100μ). In through the synthesis of EPS
and the polymerization of a viscoelastic matrix, this community is pre­
Biofilms are now recognized as widely distributed, existing in served from the extracellular environment. Quorum sensing (QS) or
aquatic and agricultural water systems, and also a wide range of eco­ chemical cell-cell communication as the complex regulatory process
systems and medical instruments with public health implications relies on high-density plays a key role in modulating of production and
(Donlan, 2002; Kordmahaleh and Shalke, 2013). Microbial biofilm for­ composition of EPS, and monitoring their extracellular environment.
mation, which was first defined as a plural behavior of microbial com­ 2.2. Mature phase that bacteria replicate and clump together in EPS to
munities, is now considered as a crucial virulence factor extensively in form a three-dimensional structure that protects cells from outside
various localized chronic and even systemic infections including those of pressures. This structure is mushroom or tower like; shape for example
the auditory, cardiovascular, gastrointestinal, integumentary, repro­ in P. aeruginosa (Klausen et al., 2003). Additional biofilm constituents,
ductive, respiratory, and urinary systems (Larsen and Fiehn, 2017; including proteins, DNA, and polysaccharides, will accumulate as the
Miller et al., 2021; Vestby et al., 2020). On the other aspect, beneficial biofilm develops (Koo and Yamada, 2016; Rabin et al., 2015). On the
outcomes of biofilm formation on the body have been mentioned, such other hand Intercellular communication in the biofilm allows bacteria to
as biofilm formation by commensal bacteria such as Staphylococcus acquire specific functions and the bacteria are replaced in different
epidermidis, which stimulates the immune system and thereby inhibits biofilm layers based on their metabolic and respiratory characteristics
pathogenic bacteria colonization (Motta et al., 2021; Nguyen et al., (Rabin et al., 2015). Intercellular degradation factors generate path­
2017). According to the Centers for Disease Control (CDC) and the Na­ ways, which provide essential nutrients to lower cells and properly
tional Institutes of Health (NIH), biofilm formation is associated to 65 dispose of waste (O’Toole et al., 2000). 3. Dispersal as the last stage that
percent and 85 percent of all infections, particularly chronic infections some mature biofilm cells begin to separate and spread into the liquid
(Potera, 1999). phase in duration (McDougald et al., 2012). This stage is critical because
During a period of infection tranquilization, four possible causes for separated cells (planktonic) might possibly start a new cycle of biofilm
the recurrence of biofilm - associated infections by bacteria are inter­ formation. Biofilms spread for a variety of reasons, including nutritional
preted: 1. Biofilm may lead to the adoption of slow growth states such as deficiency, competitive pressures, population outgrowth, and so forth
dormancy and acts as a reservoir of cells in a VBNC state in starvation (Davies, 2011; McDougald et al., 2012; Rabin et al., 2015).
zones of the biofilm. 2. The features of the biofilm matrix may contribute Depending on the type of pathogen, different factors are involved in
to tolerance by trapping or inactivating antimicrobials. 3. Antimicrobial each stage of biofilm development, some of which are mentioned in
resistance may even arise in biofilm communities due to horizontal gene Fig. 1 (Schulze et al., 2021). Kimberly K. Jefferson in 2004 reported a
transfer, which allows resistance genes to spread among cells. 4. In collection of genes involved in biofilm development (the cause genes), as
certain conditions, the ’biogeography of infection’ can change the well as genes that are regulated differently in a developed biofilm (the
steady progress of infection by altering spatial organization and evalu­ effect genes) (Jefferson, 2004). Furthermore, several experiments have
ating population communication in polymicrobial biofilm infections shown that the genes transcribed by biofilm-associated microbes vary
(Flemming et al., 2016; Stacy et al., 2016). In some sorts of circum­ from those transcribed by planktonic counterparts (Donlan, 2002).
stances, bacterial infection is definitely linked to biofilm formation The biofilm is positively and negatively regulated by several physi­
(Table 1). cochemical factors that will be discussed in the following (Kafil et al.,

2
M.N. Hajiagha and H.S. Kafil
Table 1
Biofilm related infections and their most common cause’s bacteria.

Medical
devises Cosmetic
Dental Voice
related Cerebro Tympanostomy Intravenous Biliary Intrauterine Joint
infections

implants prostheses filler


Tubes Dentures Tracheostomy Breast implant
spinal shunts catheter tract stents devices prosthesis
Contact Lenses Speech valve Tubes
Vascular prosthesis and Penile implants Orthopaedic fixation
Nasolacrimal silastic sten prostheses Endotracheal tubes
grafts Urinary tract catheters devices
Ocular abiotic materials
Prosthetic heart valves
Glaucoma tubes
Heart assist devices
Punctual plugs
Cardiac pacemakers
Corned sutures
Scleral buckle
Implanted crystalline
Cochlear implant
Bone-anchored hearing aid
implants
Tissue Chronic otitis media Caries Chronic rhinosinusitis Endocarditis on native Peptic ulcer disease Chronic prostatitis Chronic osteomyelitis Acne
infections Keratitis Chronic Adenoiditis valve Cystitis Necrotising fasciitis
Diffuse lamellar keratitis periodontitis Tonsillitis Pyelonephritis Chronic wounds
3

Endophthalmitis pneumonia Cystic fibrosis Urinary stones Peritoneal dialysis


Sinusitis Adenoid disease pneumonia Bacterial vaginosis catheter
Cholesteatoma Chronic obstructive Burn-related infection
Otitis Media pulmonary disease
(COPD)
Diffuse panbronchiolitis
Bronchicetasia
Common P. aeruginosa MRSA Oral flora P. aeruginosa E. faecalis H. pylori E. coli S. aureus P. acnes
causes CoNS S. pneumonia, R. dentacariosa K. pneumonia Bacteroidetes K. pneumonia S. pneumonie S. pyogenes
bacteria MRSA P. aeruginosa C. albicans P. aeruginosa Enterobacteriaceae P. aeruginosa P. acne S. aureus
Enterococcus spp S. aureus C. tropicalis S. aureus Fusobacterium spp P. mirabilis Propionibacterium spp Klebsiella spp
S. aureus H. influenza R. dentacariosa S. epidermidis Pseudomonadaceae Corynebacterium Clostridium spp
Streptococcus spp P. aeruginosa S. salivarius Streptococcus spp. Bacteroides

Infection, Genetics and Evolution 112 (2023) 105459


S. aureus P. aeruginosa
S. epidermidis
S. pneumonia,
Acinetobacter spp
M. catarrhalis
H. influenza
K. pneumonie Entrobacter
spp
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Fig. 1. Schematic representation of the three major stages of biofilm formation, from adherence to dispersion. In early stages motile or non-motile bacteria adhere to
either biotic or abiotic surfaces through the adhesion factors. The second stage continues with proliferation of attached bacteria and production of multifunctional
biofilm matrix which surrounds the cells. Finally, in third stage cells get released from the mature and 3D structure to re-build a new community.

2016; Liu et al., 2020; Renner and Weibel, 2011; Schulze et al., 2021): 1. 3. Efflux pumps related to multidrug resistance
Surface physical characteristics electrostatic interactions and surface
energy are examples of physical interactions that impact cell attach­ Efflux pumps are proteinaceous transporter machinery system that
ment. Some of these interactions and interfacial forces are hydration localized within the microorganism’s cell membrane. They are usually
forces, hydrophobic interactions, and steric forces.2. Chemical charac­ associated with multidrug resistance due to their ability in extruding of
teristics have an impact on cell adherence to surfaces and the biofilm the several types of antibiotics in either solitude or cooperative manner
formation. Chemical communications between cells and other signalling in bacteria. Moreover, each of efflux pumps can transport a variety of
factors including divalent cationic ions (e.g., Ca2+, Mg2+), nutrients, structurally diverse substances, such as drugs, toxins and metabolites, or
quorum sensing systems (QS), regulatory small RNAs (sRNAs), alterna­ they might be specialized for a single substrate.
tive sigma factors, two-component systems and second messengers, such Up to now, six families of bacterial efflux pumps have been estab­
as c-di-GMP and cAMP-CRP influences the organization of communities. lished to extract internal or entered substances from the bacterial

Fig. 2. Schematic illustration of six families of bacterial transporters along with their energy sources and location in the cell membrane. The major facilitator su­
perfamily (MFS), ATP-binding cassette (ABC), small multidrug resistance (SMR), and major facilitator superfamily (MATE) families are found in both gram-positive
and gram-negative species, whereas the resistance nodulation-cell division (RND) superfamily is present only in gram-negative species. The structure of the Pro­
teobacterial antimicrobial compound efflux (PACE) family has not been determined well yet. OM, outer membrane; IM, inner membrane; S, substrate.

4
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

cytoplasm and transport it toward the external environment (Fig. 2): with cells unable to form biofilms or planktonic cells. In most cases,
ATP-binding cassette (ABC) supplies the required energy to operate by either up-regulation or overexpression and sometimes downregulation
direct hydrolysis of ATP. The multidrug and toxin extrusion (MATE) are observed. A number of articles have compared the expression of
family has been composed of the Na+/H+ drug antiport systems, and genes in biofilm-producing and planktonic cells in the presence or
the major facilitator superfamily (MFS), the small multidrug resistance absence of antibiotics with each other. On the other hand, investigation
(SMR) family, the resistance-nodulation-cell division (RND) superfamily of biofilm producing ability either in sensitive or antibiotic resistant
and the proteobacterial antimicrobial compound efflux (PACE) family strains have been conducted in a few studies.
which work by the proton motive force. Therefore, it can be said that the most important role of efflux pumps
They are usually encoded by chromosomal genes, but they also can in biofilm formation process is to create ideal conditions or at least help
be expressed by transmissible genetic elements in some bacteria. The to establish such conditions. Several functions through which these
gene expression of efflux pumps is regulated by complex and multi­ pumps can perform their role have been proposed based on their fea­
functional systems mediated by local and global regulators. These reg­ tures (Fig. 3): 1. Enhancement of biofilm matrix construction via
ulators are commonly located as repressors upstream of the operon that extruding of the extracellular polymeric substances, QS, and quorum
encodes these genes. Inhibition or mutation of any of the positive or quenching (QQ) molecules, 2. Alteration in the expression of genes
negative regulators, for example, might alter the expression pathway involved in biofilm formation indirectly, 3. Excretion of harmful sub­
and result in overexpression. stances such as secondary metabolites, drugs, toxins and possibly host
Generally, regulation systems classified in three different types: immune molecules, 4. Facilitation of cell adhesion and aggregation
regulation by two-component systems, regulation by transcription and directly, 5. Efflux of biofilm formation stimulating factors which have
post-transcription factors and regulation of AcrB by a small protein. important role in biofilm formation like the iron acquisition agents, 6.
These systems are species-specific and function as an interconnected Influence on osmotic regulation and electrical charge of the bacterial
network that is followed by the expression of other pumps if one pumps surface and around the cell by transit of minerals and ions.
stops expressing. They are also connected with many other cellular Accurate identification of each efflux pump family and discovery of
processes at the transcriptional and posttranscriptional level. Accord­ their special role in biofilm formation can help to specifically inhibit it
ingly, deletion of these transporters not only affects antibiotic resistance and subsequently prevent biofilm development. Since multidrug resis­
but also decreases or disrupts the other functions like adhesion, colo­ tance is recognized as the most important feature of drug efflux pumps,
nization and dissemination, metabolism, invasion, motility, cell–cell thus we will discuss the relationship between this feature and biofilm
communication and biofilm formation. formation in some bacterial strains first. In most cases, upregulation of
There is apparently no detailed and accurate experimental data on pumps encoding genes and increased resistance in biofilm cells have
how efflux pumps contribute to biofilm formation except in a few cases. been reported, but in some cases, it has also been seen to be down­
Blocking (deleting or mutation) the efflux pump expression genes regulated. Physiological heterogeneity within the biofilm probably leads
(Matsumura et al., 2011), as well as inhibiting the efflux pumps them­ to such a pattern of gene expression (Babin et al., 2017; Sankaran et al.,
selves with efflux pump inhibitors in one or two different strains (de 2019; Stewart and Franklin, 2008).
Aguiar Coletti et al., 2017; Kvist et al., 2008), provides information on Indeed, the expression of these genes depends on several factors,
the role of efflux pumps. It usually results in defects or a decline in including the type and concentration of substrate, the stage of biofilm
biofilm formation. However, enhanced biofilm formation has been formation, the anatomical location of the biofilm in which the bacteria
observed in rare cases, which might be caused by harmful substances are located, and the environmental conditions in which the biofilm is
being trapped inside the bacteria. Another group of studies compares the formed (Fig. 4). For example, there is an increased expression of specific
efflux pump’s expressing genes in biofilm-producing cells and persistent antibiotic resistance pumps in surface-attached communities and

Fig. 3. Possible roles of efflux pumps in biofilm development process. This division of functions based on the stage of biofilm formation is not certain and they can
overlap with each other as well.

5
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Fig. 4. Suggested states for function of the efflux pump in biofilm formation based on the type of pump and substrate in different stages. a| First a pump with a wide
range of substrates, the role of which likely varies depending on environmental conditions, anatomical location, and the substance present in each step. b| The second
option is associated with pumps that can transport a multi-functional molecule (regulator, signal molecule, part of the EPS compound or toxic in high concentration).
The pump’s expression level controls the required functional concentration of the compound in each phase, or likewise, variable concentrations of the compound at
each level influence the pump’s expression rate. c| The final model is pumps with entirely specific substrates, which are expressed individually at each stage and
function in a specialized manner.

planktonic bacteria but decreased or no change in the expression of the They actively extrude substrates by using the proton motive force as
same gene in biofilm cells in the middle of the biofilm. In the middle of their energy source.
the biofilm, however, there is an increase in the expression of pumps The RND superfamily comprises mainly seven types that are the
involved in the release of secondary metabolites to remove waste heavy metal efflux (HME), the hydrophobe/amphiphile efflux-1 (Gram-
products. In terms of formation stage, increasing the expression of negative bacteria), the nodulation factor exporter family (NFE), the
pumps involved in adhesion in the early stages of biofilm formation or SecDF protein-secretion accessory protein family, the hydrophobe/
increasing the expression of pumps involved in extracellular polymer amphiphile efflux-2 family, the eukaryotic sterol homeostasis family,
formation can be recorded in the growth and maturation stages of and the hydrophobe/amphiphile efflux-3 family. The idea of the evo­
biofilm. lution process of RND pumps due to selection pressure in response to the
Observing each phenomenon in different stages overlaps with each presence of antibiotics has been limited as a result of functional in­
other not far from expectation, so examining each pump carefully to vestigations and phylogenetic assessments. Researchers claim that these
select it for a hypothetical therapeutic purpose is highly recommended. transporters and associated diverse range of compounds (substrate
According to studies, it seems two RND superfamily and MFS of efflux polyspecificities) evolved from physiological activities.
pumps play a greater role in the biofilm formation process, although So far different roles of these pumps have been reported in cell ho­
more studies in this regard seem necessary. Therefore, we will first meostasis that the most prominent of which include engagement in
describe the RND family and two members of this family and their role bacterial virulence, interactions between bacteria and plants, trafficking
in biofilm specific antibiotic resistance. of quorum sensing molecules (intercellular communication). As well as,
they contribute in detoxification by efflux of either indigenous meta­
3.1. RND transporters bolic intermediates or exogenous toxic compounds such as the antibi­
otics, heavy metals and solvents as the growth inhibitors and extrude the
This family consists of a tripartite combined apparatus (Alvarez- produced antimicrobial substances.
Ortega et al., 2013; Du et al., 2018; Tseng et al., 1999), constitutes of an The broad polyspecificity is due to either structural character of
outer membrane protein channel, a periplasmic membrane-fusion RND’s binding pockets and transition channels like dynamics of the
(adaptor) protein and a trimer cytoplasm-bound membrane pump. loops, a broad range of ligands followed by different binding areas due
Various classes of these types of efflux pumps have particularly defined to the large size of binding pocket, hydration, and activated ligands with
in Gram negative bacteria. Acr pumps of Escherichia coli and Mex pumps aromatic side chains. The significance of two well-known RND efflux
of P. aeruginosa are two well-characterized members of this superfamily. pumps in biofilm-specific antibiotic resistance will be discussed in the

6
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

following sections. Yasufuku et al., 2011). This resistance isn’t entirely due to the efflux of
toxic compounds through these transporters. The increased expression
3.1.1. AcrAB-TolC of MDT pumps can be attributed to the response to stressful conditions
AcrAB-TolC in E.coli and other Enterobacteriaceae is one of the first following the presence of antibiotics (Ma et al., 1995; Zhu et al., 2020).
described RND efflux pumps that are associated with increased levels of In this regard Baye et al., showed in the presence of tetracycline and
multidrug resistance (Bialek-Davenet et al., 2014; Thanassi et al., 1997). tobramycin antibiotics, AcrD and AcrE mutant strains had similar or
The Acr pump may play a role in biofilm resistance to ciprofloxacin at enhanced biofilm growth and viability (Bay et al., 2017). They also
low concentrations (Maira-Litrán et al., 2000). However, at the depth of exhibited higher antibiotic resistance than planktonic to tested antibi­
the biofilm, which has both a most suppressed growth rate and a lower otics (Bay et al., 2017).
concentration of antibiotics due to its lower diffusion (Gilbert and However, E. coli (ΔtolC, ΔacrA and ΔacrB) strains exhibited reduction
McBain, 2001), upregulation of the expression of multidrug resistance in biofilm growth and approximately 2-fold increased antimicrobial
(mar) operons and, as a result, the expression of Acr efflux pumps is susceptibility compared with wild type (Bay et al., 2017). Even in a
evident (Gilbert and McBain, 2001; Weston et al., 2018). This suggests similar study, impaired biofilm growth was observed for acrD and emrE
that Acr’s primary function in these cells cannot be associated with mutants (Matsumura et al., 2011). As well as, antimicrobial RND pump
increased antibiotic resistance in biofilm. There are also other studies inhibitor (verapamil) and the antimicrobial photodynamic therapy
that support this idea by dismissing the release of antimicrobials as the (aPDT) mediated by methylene blue (MB) were assessed in Ecoli biofilm
first function of AcrAB pumps in bacteria (Bialek et al., 2010; Buckner (de Aguiar Coletti et al., 2017). It was found that following the appli­
et al., 2016). cation of 22 J/cm2, 312 μg/mL of VP, and 200 μg/mL of MB, 80% of
Türkel et al. (2018) also examined the expression of AcrA in cipro­ metabolism reduction and 3.4 log10 CFU/mL declined for E. coli. In
floxacin susceptible and resistant strains of ESBL-producing Klebsiella interpreting these conflicting results, we can point to the possible dif­
pneumoniae by using quantitative real-time PCR. They also compared the ferences in the conditions of the two experiments.
ability of the two groups to form biofilms. This group’s findings revealed Likely, a nutritional conditions and growth stages of biofilm play a
that ciprofloxacin susceptible strains had higher levels of AcrA expres­ more important role in its antibacterial resistance than efflux activity of
sion than resistant ones. Sensitive strains, on the other hand, had a pumps, and b. MDTs can indirectly affect biofilm resistance profile
higher propensity for forming biofilms. However, the expression of through their effect on biofilm formation, growth and survival by the
efflux genes had no effect on biofilm growth and no difference in efflux overlapping specificities (Tal and Schuldiner, 2009). Nevertheless,
gene expression was observed among biofilm production. Based on the several research have reported pumps that may be efficient in biofilm
results of their research, the group suggested that the bacterium tries to specific resistance, and recognizing such pumps can be extremely ad­
maintain itself in ciprofloxacin-sensitive conditions with minimal en­ vantageous in biofilm control. For example, PA1874–1877 in
ergy consumption by using biofilm formation or expression of efflux P. aeruginosa has been identified in Pseudomonas biofilm cells, and
pumps (Türkel et al., 2018). According to another investigation, there removing the encoding genes resulted in tobramycin, gentamicin, and
was no difference between MDR and non-MDR K. pneumoniae strains in ciprofloxacin sensitivity in these cells (Zhang and Mah, 2008)
their capacity to form biofilms. It’s even been found that high resistance
profiles were associated with decreased biofilm formation in both Aci­ 3.1.3. MexAB-OprM
netobacter baumannii and Salmonella enterica (Fabrega et al., 2014; Perez, MexAB-OprM, MexCD-OprJ, MexEF-OprN and MexXY-OprM are
2015). In addition, the expression of efflux pumps genes such as acrA, another well-known RND pumps that contribute in antibiotic resistance
emrB, oqxA, and qacEΔ1 in K. pneumoniae is increased in biofilm in P. aeruginosa. The role of this efflux pump in a biofilm-specific anti­
development. biotic resistance has also been reported (Mah and O’Toole, 2001; Olsen,
As a consequence of the results, authors concluded that there is no 2015). It has been observed that expression of MexAB-oprM and/or
association between the quantity and type of efflux pump genes and MexCD-oprJ in the presence of azithromycin pre-inhibitory concentra­
biofilm formation capacity, ruling out the involvement of efflux pump tions is essential for biofilm formation (Gillis et al., 2005). No elevation
expression as the primary determinant of biofilm resistance. They also in resistance to other antibiotics was observed in azithromycin-resistant
observed that using of concentrations above 10 mg of carbonyl biofilm variants (Gillis et al., 2005). The expression of these pumps in
cyanidem-chlorophenyl hydrazine (CCCP) as an inhibitor of efflux some AZM-resistant isolates can be greatly influenced by the mutation of
pumps further inhibits biofilm (Tang et al., 2020). Conversely, when the a negative nfxB regulator or not (Mulet et al., 2009). Although, the
CCCP concentration was less than 10 mg, biofilm formation was sequencing of nfxB from PAO1 biofilm variant (PAO1-BV) in this study
induced. There are possibly some feedback mechanisms that aid to in­ showed that overexpression of MexCD-oprJ can’t be caused by a mu­
crease EPS production or expression of additional efflux pump gene to tation in nfxB (Gillis et al., 2005). It is possible that regulation of RND
compensate for the blocked efflux pumps. efflux pumps are very complex and other factors like brlR, PA0756–0757
and rapA besides nfgx act as regulators which their mutations can lead to
3.1.2. TolC as a component of RND and MFS efflux pumps biofilm-specific resistance (Hall and Mah, 2017). For example the ndvB
In E. coli, some members of RND efflux pumps, along with other gene encodes a glucosyltransferase involved in the synthesis of cyclic-b-
families, form multidrug resistance transporters (MDTs) that are (1, 3)-glucans (Bhagwat et al., 1996). They bind to tobramycin and
responsible for the extracellular efflux of compounds and also connect restrict it from reaching the cellular target as a novel mechanism of
the cytoplasm to periplasm and outer membrane (Saier Jr, 2000). In resistance (Mah et al., 2003).
some of these transporters TolC as an outer membrane protein link to the It has been demonstrated that mutants resistant to AZM exhibit
RND members AcrB, AcrD, AcrF and MFS (EmrB) via periplasmic linkers resistance towards ciprofloxacin and cefepime as well (Macia et al.,
(AcrA, AcrE and EmrA) (Piddock, 2006). MDTs are responsible for the 2006; Mulet et al., 2009). This could be a warning about the effect of
phenomenon of multidrug resistance (MDR) in planktonic cells of E. coli. AZM maintenance therapy on increasing drug resistance in P. aeruginosa
Nevertheless several studies showed they are associated with the sessile isolates from chronic respiratory infections due to overproduction of the
communities and their antibmicrobial resistance pattern as well (Ito MexCD-OprJ and biofilm formation. In a distinct study that focused on
et al., 2009; Masadeh et al., 2013). the ability of bacterial biofilm formation, particularly in those with
In the presence of subinhibitory concentrations of various antibi­ multidrug resistance, upregulation wasn’t observed in the expression of
otics, studies on biofilm formation in E. coli revealed an increase in these two pump’s genes. Interestingly, some instances of down­
biofilm formation (Penesyan et al., 2020) and the induction of expres­ regulation were actually documented (De Kievit et al., 2001). However,
sion of several genes encoding efflux pumps (Ebbensgaard et al., 2020; some other studies have attributed the resistance of biofilm cells,

7
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

especially bacteria in the substratum region, to other types of antibi­ on negatively charged surfaces (Rzhepishevska et al., 2013). Hydro­
otics, including tetracycline, chloramphenicol, quinolones, β-lactams, phobicity has both negative and advantageous effects on bacterial bio­
aztreonam, gentamicin, tetracycline and tobramycin and colistin, to the film formation capacity on biotic and abiotic surfaces (Pandey et al.,
MexAB-oprM pump (Soto, 2013). MexEF-oprN and MexCD-oprJ other 2022).
members of this family contribute in biofilm antibiotic resistance in The CusABCF pump in E. coli and other Gram-negative bacteria is
areas with low oxygen tension (Schaible et al., 2012; Van Acker and responsible for protecting them against lethal concentrations of Cu+ and
Coenye, 2016). The MexEF-oprN coding genes exhibited increased Ag+ by leaving it outside the bacterium (Greene and Koronakis, 2021;
expression in hypoxic cells of the biofilm, whereas no such result was Yun et al., 2010). The presence of low amount of copper or silver ions
observed for the mexCD-oprJ pump genes (Schaible et al., 2012). around the bacteria can be thought to cause the environment to progress
towards a positive charge. Previously investigations has mentioned that
4. Role of efflux pumps in biofilm formation subinhibitory concentrations of some metal can contribute in cell sur­
face adhesion and/or cell-to-cell aggregation process and changes in
4.1. Role in early stage biofilm extracellular polymeric substance (EPS) matrix (de Araújo and
de Oliveira, 2019). Therefore, when selecting these toxic metals as
The early stages of biofilm formation are some complex events that antibacterial compounds, proper application dosage should be
began by sense some especial environmental signals in bacteria. These considered.
environmental signals differ from organism to organism. The initial The ability of bacteria to overcome the osmolarity pressure of the
attachment of bacteria includes of physical and chemical interactions environment promotes the adhesive strength (Liu et al., 2013; Van der
that contribute to cell-surface and cell-cell interactions. Bacterial efflux Waal et al., 2011). There are two general strategies of osmoadaptation
pumps and their components can involve in either physical (direct or using by bacteria to control hyper-osmotic conditions: a) the salt in
indirect effect via influence on attachment structures like pili) or cytoplasm and b) the organic osmolyte such as proline, glycine betaine
chemical interactions (efflux of effective chemical substances). They and choline (Csonka, 1989; Galinski, 1995). Proline transport genes
may have some preventing or promoting role in aggregation. Accord­ (proV, proX, proP, proW) in both gram positive and gram-negative bac­
ingly, depending on their role, they may show up or down-regulation. teria encodes MFS-like efflux pump, which export proline and glycine
Some efflux pumps and their likely substrate and function in the early betaine. The evidence showed representative upregulation of these
stages has been gathered in Table 2. genes during the initial stages of biofilm formation (Mauter et al., 2013;
Resch et al., 2005). The same result has obtained in S. epidermis where
4.1.1. Role in physical and chemical interaction SE0225, a glycine/betaine transporter opuCD encoding gene, revealed
Attachment of bacterial cells to any types of surfaces is influenced by almost 3 fold enhanced expression in biofilm cells in compare with
surface charges and hydrophobicity (Dickson and Koohmaraie, 1989). planktonic cells (Yao et al., 2005). In Enteropathogenic E. coli (EPEC), it
Due to negatively charge of the most bacterial cell surfaces, attachment has been shown that overexpression of the OmpX protein significantly
and biofilm formation in positively charged surfaces is more favorable restored biofilm formation and curli production under sucrose- and
(Song et al., 2015). However, some bacteria, such as Pseudomonas, are NaCl- induced hyper-osmotic conditions in ΔtolC; that previously lacked
able to modify their surface charge to form long-term biofilm structure these properties (Li et al., 2018). Overexpression of OmpX in this

Table 2
Summary of likely efflux pumps involved in early stage of biofilm.
Efflux pump/ Class of pump Bacterial species Potential substrate/Role Main function
Component

Physico-chemical Hydrophobicity CusABCF CusA (RND),CusB Gram -bacteria Cu/Ag Regulation of the
interactions/ Pili and (MFP), CusC hydrophobicity
surface proteins (OMF) Surface positive charge
TolC RND & MFS Enteroaggregative E. coli 38-KD protein Aggregation
(EAEC) (hydrophobic surface in aqueous environment
protein) secretion Aggregation/Adhesion to
AAF regulation host cells in aqueous
environment
Osmoadaptation Glycine betaine/ ABC transporter Gram – and + bacteria Proline and glycine betaine Control hyper-osmotic
L-proline conditions
transporter
Osmoregulation OmpX – Extraintestinal pathogenic NaCl or Sucrose Curli production under
Escherichia coli (ExPEC) transporter regulation hyper-osmotic conditions
TolC RND & MFS Enteroaggregative E. coli Mg2 transporter regulation Adherence
(EAEC) Curli (csgAB) regulation Adherence to abiotic
S. enterica serovar surfaces under osmotic
Typhimurium condition
Pili production AdeABC & RND A. baumannii Expression of type I pilus Adherence to biotic and
AdeIJK MFS A. baumannii ATCC17978 system abiotic surfaces
A1S_0116 Staphylococcus aureus Ac-505 (a lipopeptide-like Adhesion factor
Tet38 compound) Attachment and
Interacting with CD36 internalization into Eps
Motility MexEF-OprN RND P. aeruginosa PA14 PQS Swarming
MexGHI-opmD ⸗ P. aeruginosa PAO1 ⸗
AcrAB-TolC Salmonella Typhimurium chewy, flgLMK and type III
secretion system
regulation/ fumarate
Gene regulation Lm. G_1771 ABC Listeria monocytogenes Dlt, SrtA & GntR regulation Biofilm formation
SmeGH RND Stenotrophomonas negatively repressor
maltophilia Biofilm formation
triggering compounds

8
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

situation may alter the production of NaCl or sucrose transport related substantial difference in initial attachment. AdeB-deficient mutants also
proteins or signal pathways, increasing or decreasing NaCl or sucrose have higher levels of pil and com (competence) gene expression. In some
efflux or influx through the outer membrane. strains downregulation of AdeABC efflux pump, so increased pili pro­
duction may be associated with the ability of A. baumannii to form a
4.1.2. Pili and surface proteins biofilm on plastic. However, these pumps have an important function in
The bacterial TolC homologues is a multi-functional protein that EPs and mature biofilm formation on biotic surfaces in the other
operate many different functions (Koronakis et al., 2004). Enter­ A. baumannii isolates (Richmond et al., 2016).
oaggregative E. coli (EAEC)’s major properties are its ability to attach to QS-regulated operon A1S_0112–A1S_0119 contains of A1S_0116
Hep-2 cells in vitro and to produce biofilm in the intestinal mucosa gene which encodes an RND efflux pump in A. baumannii ATCC17978
(Wakimoto et al., 2004). Several factors, such as AAF, AggR as the strain. It’s believed that it’s implicated in the secretion of signaling
aggregative factor, and Aap (dispersin) as an anti-aggregative factor molecules or secondary metabolites. Recently, the A1S_0114 gene was
exported by localized outer membrane AatA (homologous to TolC), are reported within the same operon which produces Ac-505 (a lipopeptide-
involved in adhesion and biofilm formation of EAEC (Harrington et al., like compound named acinetin505) (Rumbo-Feal et al., 2017).
2006; Sheikh et al., 2001). In a study conducted by the Naoko et al and A1S_0116 involved in the secretion of Ac-505 as a secondary metabolite.
colleagues on EAEC04 showed less aggregation and adherence for TolC- The exact role of this pump and Ac-505 in early stage of biofilm pro­
deleted mutants (042tolC) than the wild type (Imuta et al., 2008). The duction hasn’t revealed yet, but deficiency of A1S_0116 may result in
loss of the hydrophobicity of the bacterial surface and the decrease in surface motility defect (Blaschke et al., 2021). Tet38 in Staphylococcus
transcription of the aggregative fimbriae (AAF) are two putative aureus has been proposed as an adhesion factor that facilitates epithelial
mechanisms by which the TolC mutant lost the aggregative phenotype. cell attachment and internalization by interacting with the scavenger
Another hypothesis that may be implicated in these phenomena is a receptor CD36 (Truong-Bolduc et al., 2021).
disturbance in the regulation of Mg2 transport, which is most probably
correlated to TolC. According to one study, Mg2 is essential for the 4.1.3. Motility
adhesion of some EAEC strains (Chart et al., 1997). Moreover, there is a Cell surface Motility and its regulation in some motile bacteria like
38-kDa protein (hydrophobic surface protein layer) reported previously Bacillus, Pseudomonas, Vibrio, and Escherichia are required for biofilm
for EAEC (Imuta et al., 2008). It is thought to be a humoral factor formation. Flagella and motility are probably important early in bacte­
secreted by TolC and unlike Aap, functions as an aggregation factor. rial adhesion to locate and interact with a suitable surface. Nevertheless,
A similar result was reported for EPEC TolC-mutant strains as well as in the growing and mature stages of biofilm, functional inhibition of
for wild-type in M9 medium after the administration of efflux pump motility aids in the maintenance of the bacterial aggregation (Gutten­
inhibitors. They also reported reduced curli production and diminished plan and Kearns, 2013). There are 3 types of motilities including
csgB and csgD expression (Hou et al., 2014; Li et al., 2018). The curli swimming, swarming, and twitching which involve in biofilm formation
proteins aid in the adherence of certain bacteria to abiotic surfaces as (Harshey, 2003). Swimming and swarming are related to flagella
well as host cells (Kim et al., 2012; Prigent-Combaret et al., 2000; Tursi biosynthesis, rotation, and chemotaxis while twitching motility is
et al., 2020). The csgBA operon synthesizes the main and minor curli dependent on type IV pili (Aybey et al., 2021). Swarming motility is
subunits. These results indicate the ability of TolC to respond to different often influenced by quarum sensing (Partridge and Harshey, 2013), so
osmotic conditions and regulate biofilm formation based on it. Besides, we can expect to see inhibition of quorum sensing responses in places
low expressed level of curli component coding genes (csgB and csgD) was where decreased motility is helpful for biofilm formation; less motile
seen for S. enterica serovar Typhimurium mutants lacking of Acr-TolC strain of P. aeruginosa is more competent for biofilm formation within
and after using the efflux inhibitor in LB broth without the salt (Baugh the cystic fibrosis (CF) lung (Ramsey and Whiteley, 2004).
et al., 2012). Curli is a crucial component of the Salmonella extracellular Martin et al., showed nfxC-type mutants of P. aeruginosa strain PA14
matrix, and its absence reduces biofilm formation significantly (Baugh (overexpressing MexEF-OprN) had deficiencies in swarming and biofilm
et al., 2012). The exact role of the efflux pump is not recognized, but formation in Tryptic Soy Broth (TSB) due to rapidly pumped of the
some studies have suggested special role by exporting the global regu­ Pseudomonas Quinolone Signal (PQS) Precursor HHQ (4-hydroxy-2-
lator as activator on the transcriptional level or can be due to post- heptylquinoline) (Lamarche and Déziel, 2011). They also observed
transcriptional changes. Other investigators attribute these effect to knocking out the mexE (the membrane fusion) gene in the MGL01 (a
post-transcriptional changes (Baugh et al., 2012). P. aeruginosa mexS- mutant overexpressing MexEFOprN) restores
The AdeABC and AdeIJK pumps in A. baumannii appeared to be swarming motility, pyocyanin production and biofilm formation to wild-
involved in the expression of genes encoding CsuA / B, CsuC, and FimA type in vitro. Surveys showed an increased Pseudomonas quinolone
proteins (type 1 pilus system) by a similar mechanism with Salmonella as signal (PQS) (one of the AIs in QS system) production due to the efflux of
we described above (Yoon et al., 2015). Eun et al., reported overex­ 4-hydroxy-2-heptylquinoline in this strain. This suggests that the
pressed adeABC and adeIJK mutants show lower biofilm formation due MexEF-OprN efflux pump regulates motility and biofilm formation
to lower expression of type I pilus system-related genes (Yoon et al., through secretion of a QS system signaling molecule (Lamarche and
2015). Abiotic surface attachment, colonization, and the formation of Déziel, 2011). It needs to compare mentioned phenotypes in various
microclonies on abiotic surface can be mediated by these proteins. culture media since it’s been reported an overexpression of the mexEF-
Forthermore, DABA-AT that involved in biosynthesis of DAP reduces in oprN operon near to the primary normal human airways epithelial cells
overexpressing adeABC and adeIJK mutants. DAP is a polyamine (Frisk et al., 2004) which represents CF condition. however In another
required for A. baumannii surface-associated motility. So, the reduction study the mexEF mutants exhibited enhanced swarming, rhamnolipid
in DAP production contributes in less motility and biofilm formation. synthesis, and lethality in a mouse infection mode (Vaillancourt et al.,
However, it has not yet been proven that DABA-AT is the substrate of 2021). A gene cluster has been characterized in P. aeruginosa that en­
these pumps. codes MexGHI-opmD pump. Allelic exchange mutants of mexI and opmD
Different A. baumannii adeABC-deleted strains show different ability in P. aeruginosa PAO1 demonstrated poor swarming motility in addition
in biofilm formation on different surfaces (Richmond et al., 2016). In to other QS-related phenotypes (Aendekerk et al., 2002), but biofilm
AYE mutants (adeB-deleted A. baumannii), the ability to form biofilms on formation hasn’t studied yet.
both mucosal and plastic surfaces is impaired, but in S1 mutants (adeAB- Mutants lacking a component of AcrAB-TolC in Salmonella Typhi­
deleted A. baumannii), only biofilm formation on mucosal surfaces is murium represented changed motility and adherence ability. It’s been
reduced. LIVE/DEAD staining and confocal laser scanning microscopy reported less motility in L110 (acrB::aph) than wild-type (SL1344).
on adherent AdeB AYE mutants on mucosal tissue revealed no Lacking acrA, acrB and particularly tolC gene resulted in poorly adhere to

9
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

cells in tissue culture and grow in in anaerobic conditions. By using the interference with one will upregulate or downregulate the other. Efflux
Reverse transcription-PCR and Western blotting, they confirmed pumps could potentially be involved in quorum sensing and its-related
decreased expression of numerous genes encoding cheWY and flgLMK biofilm formation. This involvement performed by influencing the
and 14 Salmonella pathogenicity island (SPI)-1-encoded type III secre­ transmission of signaling molecules, the effect on the expression of genes
tion system genes, including sopE, and associated effector proteins. involved in quorum sensing, and antimicrobial resistance mechanism
However, acrA, acrB, or tolC can be involved in a regulatory network (Lekshmi et al., 2018). In addition, efflux pumps can help in biofilm
that influence in genes that encode proteins involved in this phenomena formation by exporting quorum sensing signals produced as inhibitors of
(Webber et al., 2009). the growth of competing bacteria or noxious QS signals. Besides they
Buckner et al., reported that disruption of acrD in S. enterica serovar may act as a defense system against quorum sensing inhibitors or
Typhimurium led to motility defects caused by changes in intracellular improper signals from other species. The transporters which involved in
concentrations of fumarate (Buckner et al., 2016). According to the QS transportation across the bacterial cell membrane has been listed in
distinct study, S. enterica AcrD mutants were incapable to create a Table 3.
competent biofilm in the crystal violet model (Baugh et al., 2012); This feature was first reported in P. aeruginosa, where the formation
however O-antigen mutants, which are impaired in swarming, were of a controlled biofilm by quorum sensing is well known. It’s been
generally more effective in biofilm production than the wild type claimed that the MexAB-OprM extrude the autoinducer N-(3-oxodode­
(Mireles et al., 2001). They conclude that inhibition of motility is likely canoyl)-L-homoserine lactone (3-oxo-C12-HSL) (Pearson et al., 1999),
to be advantageous to prevent bacterial cell dispersal in vitro, but and MexEF-OprN has role in 4-hydroxy-2-heptylquinoline extrusion
impaired EPs secretion following AcrD inhibition impairs biofilm evo­ (HHQ) and kynurenine, as a precursor of HHQ (Alcalde-Rico et al.,
lution in later stages. 2016). MexB deleted mutants showed an increased QS response due to
As well as reduced swimming and swarming motilities has been the accumulation of acyl-HSLs and its selective binding to LasR (QS
observed in ΔmexB, ΔmexF and mexA mutant in Pseudomonas syringae pv. receptor protein) (Minagawa et al., 2012). Several studies have reported
tabaci 6605. ΔmexA had fewer flagella than wild type (Ichinose et al., decreased expression of virulence factors regulated by QS following
2020). overexpression of MexAB / OprM and MexEF-OprN efflux pumps
(Alcalde-Rico et al., 2020a; Lamarche and Déziel, 2011; Rasamiravaka
4.1.4. Indirect regulation of biofilm-related genes and El Jaziri, 2016). A similar result has been reported for the MexCD-
Although there are only few reports about how efflux pumps OprJ efflux pump (Alcalde-Rico et al., 2018). They believe that the
contribute in biofilm regulation, signal molecule transport by them may release of intracellular autoinducers by the mentioned pumps is the
be considered as a part of this role. Zhu et al. (2008) reported a putative cause of this phenomenon, although this theory has recently been
ABC transporter permease encoded by Lm.G 1771 gene in Listeria mon­ controversial (Alcalde-Rico et al., 2020b).
ocytogenes that its deleted mutant exhibited an enhanced ability for In Acinetobacter Spp, QS also plays an important role in the formation
biofilm formation compared with WT. Proteomic and transcriptomic and maturation of biofilms. Decreased expression and production of
analyses in lm.G_1771 deletion mutant (D1771) showed several cell AHL in A. baumannii leads to a reduction in biofilm formation. The
surface protein encoding genes may influence by lm.G_1771 (Zhu et al., secretion of long-chain AHLs into the extracellular space is mediated
2011). They hypothesized that through a new signal transduction through efflux pumps. In the review of several clinically isolated
pathway the lm. G_1771 negatively regulates the expression of several A. baumannii strains, it has been observed that hyper expression of the
candidate encoding genes. For example, cell surface proteins (Dlt), cell AdeFGH efflux pump, especially the upregulation of abaI (an AHL syn­
surface anchor proteins (SrtA), and transcriptional regulators (GntR) thase) and abeG (a component of the AdeFGH efflux pump) genes induce
that involve in biofilm formation. A similar phenotype was observed in the biofilm formation (He et al., 2015). Nevertheless, In another study,
SmeH (the transporter protein of the SmeGH, a RND efflux pump)- overexpression of the AdeABC, AdeFGH and AdeIJK pumps has reduced
deficient Stenotrophomonas maltophilia mutants. This strain showed a biofilm formation (Yoon et al., 2015). Generally, in-depth function of
significantly increased ability to form biofilms, but no difference in the AcrAB efflux pump in different stages of biofilm formation by QS
swimming motility was observed compared to the wild strain D457. hasn’t characterized yet. It may lead to the uncontrolled release of AHLs
Similar to lm. G_1771, this pump shows a negative effect on the regu­ outward and a decrease in its intracellular concentration. In
lation of genes involved in biofilm development. In addition, the accu­ A. nosocomialis, knocking out of the acrA and/or acrB genes resulted in
mulation of toxic substances following the inactivation of transporter reduced biofilm/pellicle development and antimicrobial resistance.
can trigger a stress respond and subsequently induce the expression of Probably it’s due to a disorder in the formation of the extracellular
biofilm genes. matrix, as no change in secretion of AHLs was observed (Subhadra et al.,
2020). acrAB mutants may use different efflux pumps for extrusion as a
4.2. Effect on growth and maturation compensatory instead of AcrAB efflux pump. EMSAs confirmed limited
AHL secretion in the acrR deleted mutant of A. nosocomialis. According
4.2.1. Quorum sensing to an in-silico analysis, AcrR may bind to motifs in the promoter regions
During the growth and maturation stages, the accumulation of multi- of anoI (encoding AHL synthase) and anoR at a higher level. Even
layered clusters of microbial cells leads to a series of specially controlled removing it may have caused the overexpression of pumps.
activities strongly influenced by the genetic and QS control system In previous studies it’s been showed that the RND efflux pump
(Kjelleberg and Molin, 2002). QS is an intercellular communication BpeAB-OprB in Burkholderia pseudomallei KHW may be involved in
process that allows bacteria to live as multicellular organisms by sensing synthesis or secretion of N-octanoyl-homoserine lactone (C8HSL) (Chan
their environment and regulating their density and behavior (Bassler and Chua, 2005). Likewise, Acinetobacter, it seems AHLs regulate
and Losick, 2006). This system involves cell-to-cell communication BpeAB-OprB efflux pump in B. pseudomallei KHW. The adding of C8HSL
based on the synthesis, release, recognition, and response to extracel­ and C10HSL autoinducers exogenously promotes bpeAB promoter-lacZ
lular signaling molecules known as autoinducers at the group level expression to the early exponential phase (Chan and Chua, 2005).
(Papenfort and Bassler, 2016). There are 3 main QS-related autoinducers However Takehiko et al., obtained the opposite results in B. pseudomallei
consist of Acyl-Homoserine Lactones (AHL) in gram negative bacteria, 1026b BpeAB-OprB mutants (Mima and Schweizer, 2010). They found
autoinducing peptides (AIP) in gram positive bacteria and AI-2 as the no defects in AHLs secretion or biofilm development unless they were
inter-species signaling molecules (Wu and Luo, 2021). accompanied by deficiencies in AmrAB-OprA. Additionally, the Bacter­
There is an interrelationship between efflux pumps and quorum oides fragilis BmeB efflux pump establishes a correlation between QS and
sensing systems. They arise from each other at the same time, and that biofilm formation by regulating the internal concentration of HSLs

10
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Table 3
Summary of potential efflux pump involved in growth and maturation stages of biofilm.
Efflux pump/ Class of pump Bacterial species Potential substrate/Role Main function
Component

Quorum sensing MexAB-OprM RND P. aeruginosa 3-oxo-C12-HSL AHL


MexCD-OprJ P. aeruginosa HHQ & kynurenine
MexEF-OprN A. baumannii (OH-dDHL)
AdeFGH B. pseudomallei KHW C8HSL
AdeIJK
AdeABC
BpeAB-OprB
MsrA ABC Staphylococcus spp - AIP
transporter
Secondary AcrEF RND E. coli Indole Signaling molecules in low
metabolites Mtr transporter RND E. coli Nitrosyl indole derivatives concentrations
MdtEF RND Salmonella spp Diamide, H2O2, Paraquat, ADBAC, detergents, toxic in high concentrations
MdsABC RND P. aeruginosa gold, dyes, SDS, novobiocin Toxic in anaerobic respiration and
MexEF-OprN RND E. coli, Salmonella and Phenazines stress conditions
MexGHI-OpmD Shigella Bile salts Toxic metabolites in stress
AcrAB conditions
Iron acquisition, signal molecule &
electron mediator
Toxic in high concentrations
Biofilm inducer in low
concentrations
MexCD-OprJ,
MexCD-OprN RND P. aeruginosa Kynurenine Precursor of anthranilate & AQs
MexEF-OprN SMR E. coli K-12 Spermidine Swarming & antibiotic resistance
MdtIJ RND Gram negative bacteria Cu/Ag Biofilm formation stimulator at low
CusABCF MFS S. aureus Siderophore concentrations
NorA RND P. aeruginosa Siderophore Toxic in high concentrations
MexXY-OprM RND B. pseudomallei KHW Siderophore production Toxic heavy metal
MexAB-OprM RND E. coli & S. enterica PPIX Iron acquisition
BpeAB-OprB A unique E. coli Nickel Iron acquisition
MacAB family S. aureus QAC Iron acquisition
RcnAB MFS E. coli pHBA Haem homeostasis
MdeA ND E. coli Non-metabolic carbohydrates Curli production in low
NorB MFS concentrations
NorC Toxic in high concentrations
AaeAB Biofilm formation stimulator at low
SetAB concentrations
Toxic in high concentrations
Toxic in high concentrations
Toxic in high concentrations

(Pumbwe et al., 2008). Streptococcus. suis is involved in efflux pump SatAB regulation (Wang
In most Gram-positive QS bacteria AIP is processed and released by et al., 2019c). It’s been suggested that determined fluoroquinolones
ABC transporters (Bassler, 1999). The MsrA efflux pump is involved in susceptibility in ΔluxS strains is due to overexpression of efflux pump
the expression of the Agr locus (a Gram-positive two-component QS SatAB (Wang et al., 2019c).
system). On the other hand, the agr locus regulates biofilm formation in As a result, incorrect antibiotic dose can not only fail to eradicate the
Staphylococcus spp. Using of baicalin as a MsrA efflux pump inhibitor pathogen, but also aid in the development of biofilms by triggering the
reduced transcription levels of agrA, agrC, RNAIII, and sarA (quorum- expression of QS- and efflux pump-related genes. In a study by Dawan
sensing system regulators) and the formation of biofilm (Wang et al., et al. On S. Typhimurium strains, this result was clearly observed (Dawan
2019a). et al., 2022). S. Typhimurium ATCC 19585 (STWT) and S. Typhimurium
AI-2 is transferred by LsrC, LsrD, and the lsrA (ATPbinding protein), CCARM 8009 (STCI) strains that were only exposed to sub-MICs of CEF,
but the exact mechanism isn’t characterized yet. The lsr operon is CHL, CIP, ERY, NOR and TET antibiotics showed a high ability to form
regulated by LsrR as a repressor protein. As well as it’s been determined biofilms, while the using of these antibiotics in combination with PAβN
that MdtH, an MSF multidrug efflux pump in E. coli, regulated by LsrR. It (an efflux pump inhibitor) exhibited significantly decreased the biofilm-
needs to assay relationship between AI2/LsrR and MdtH, and their formation.
impact on biofilm formation (Yu et al., 2020).
Rahmati introduces a model in which the actual role of drug efflux 4.2.2. Secondary metabolites
pumps is extruding QS molecules that results in their over production in Multidrug efflux pumps can help in maintenance of homeostatic
response to cell density. According to this, SdiA (suppressor of division levels of biofilm metabolites by extruding the exogenous components
inhibition) seems to control AcrAB efflux pump production positively to including produced toxic compounds by competitors, heavy metals, and
mediate cell–cell communication (Rahmati et al., 2002). Recently, a organic pollutants and endogenous metabolites (Table 3). The precise
novel transcriptional regulator called AnoR has been identified in in function of metabolites, whether they act as hazardous or beneficial
A. nosocomialis which has crucial role in AHL production (Subhadra external agents in the formation of biofilms, depends on their intrinsic
et al., 2020). In addition, reduction of surface motility and biofilm for­ properties or their intra- and extracellular concentration. Besides the
mation were also reported in its deleted mutants. The study of these location of biofilm within the body, the developmental stage of it, and
strains showed a decrease in the expression of acrA and acrB genes, the anatomical location of bacterial cells within the biofilm structure can
which indicates the effects of AHLs in the expression of AcrAB multidrug be as determinants of metabolites function. In the microbial population
efflux system. Moreover, LuxS/AI-2 quorum-sensing system in enclosed in the biofilm matrix, the oxygen concentration is gradually

11
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

reduced from top to bottom. With anaerobic conditions in the biofilm toxic phenazines. Especially the steady-state in the production and
center, either anaerobic bacteria predominate or optional anaerobic release of phenazines is controlled by MexG/HI-OpmD (Huang et al.,
bacteria activate the anaerobic respiration pathway for energy. 2018; Jin et al., 2015; Sakhtah et al., 2016; Wolloscheck et al., 2018). In
a study conducted by the author and his colleagues, this issue was
4.2.2.1. Indole. A tryptophan metabolite synthesized by gut bacteria, examined in detail and the results obtained confirmed the above
functions as metabolites-mediated QS signal and is involved in biofilm mechanism (Wolloscheck et al., 2018). They observed a significant
formation in a different ways (Lee and Lee, 2010). Decreased extracel­ decrease in extracellular pyocyanin so that the concentration of it in
lular secretion of indole or increased intracellular concentration of its culture medium supernatant was measured almost 2-fold lower
harmful metabolic byproduct following the elimination of their exporter compared to the wild strain. Also, the high amount of pigment in the
may disrupt biofilm formation (Kim and Park, 2015). culture of PAO1 (overexpressing MexGHI-OpmD or MexHI-OpmD
The AcrEF-TolC and Mtr transporter proteins are reported to be mutant) indicated the role of this pump in pyocyanin production.
responsible in the export and import of indole in E. coli (Heatwole and Conversely, these phenazines also induce upregulation the MexGHI-
Somerville, 1991). The mtr gene-deleted mutants showed growth defi­ OpmD expression by a SoxR (transcription factor)- dependent mecha­
cient on tryptophan-free medium, which is non-inducible by adding the nism (Dietrich et al., 2006; Du et al., 2015; Sakhtah et al., 2016).
external indole (Yanofsky et al., 1991). In the acrEF mutant, there was an
increase in intracellular indole concentration and a decrease in indole 4.2.2.3. Bile salts. A group of bactericidal exogenous metabolite which
excretion (Kawamura-Sato et al., 1999). Furthermore, in another study, are synthesized in the liver and presence in the duodenum, jejunum, and
the use of exopolysaccharide secreted by Lactobacillus plantarum and proximal ileum. However, Bile is lethal at high concentrations induces
Bacillus spp. against E. coli resulted in inhibition of biofilm formation expression of virulence genes at low concentrations, including efflux
and efflux pumps (Mahdhi et al., 2018). This phenomenon may be pumps. Intestinal bacteria are resistant to these compounds by using
related to the ability of EPS to alter the physiology of the bacterial multiple strategies. Since bile salts are considered as one of the sub­
membrane or to reduce indole production and secretion (Mahdhi et al., strates of efflux pumps, it seems that exporters protect the bacteria
2018). Other studies, however, refuse the protein-dependent transfer of against the toxic amounts of bile salts. The AcrAB efflux pump is one of
indole and tryptophan. They claimed that indole transfer across the lipid the most crucial RND families in E. coli, Salmonella and Shigella is used to
layer directly (Piñero-Fernandez et al., 2011; Zarkan et al., 2020). There resist bile salts (Buckley et al., 2006; Nickerson et al., 2017; Nikaido and
are some evidences that indicated some efflux pumps is regulated by Zgurskaya, 2001). ΔacrB mutant in Shigella was unable to growth in
indole; for example the expression of AcrAB multidrug efflux pump in 0.4% and 2% bile salts (Nickerson et al., 2017). As well as in Salmonella
S. enterica serovar Typhimurium and the expression of mdtABC and acrD the acrA, acrB, and tolC mutants could not adapt to bile (Urdaneta and
in E. coli are mediated by indole (Hirakawa et al., 2005; Nikaido et al., Casadesús, 2018). However, in both bacteria, long-term incubation with
2011). sub-inhibitory concentrations has been shown to induce the expression
Under respiratory stress RND efflux pump, MdtEF, extrudes indole of these pumps. Quantitative reverse-transcription (qRT)-PCR analysis
nitrosative derivatives, which are produced during anaerobic respira­ revealed that acrB and some others genes including S2558, ospE1-ospE2
tion of nitrate (Zhang et al., 2011). It has been shown that expression of and yhcN of Shigella were induced almost 2-fold in prolonged exposure
MdtEF in E. coli upregulated more than 20-fold anaerobically (Deng to 0.4% bile salts (Nickerson et al., 2017). In a similar investigation the
et al., 2013). It is unlikely that the expression of this pump in the center expression level of the acrA and acrB genes in the presence of pre-
of the biofilm, where anaerobic conditions prevail, will act as an inhibitory amount of DOC was obtained approximately 10 fold during
exporter of these compounds due to anaerobic respiration. It is necessary exponential growth (Urdaneta and Casadesús, 2018). The AcrAB-TolC
to study the expression of this pump in biofilm members in the presence efflux pump in Salmonella is regulated by ramA (an efflux transcrip­
or absence of antibiotics in order to discover its physiological signifi­ tional regulator). Sylvie et al identified the bile-mediated activation of
cance. MdsABC, a RND efflux pump, in Salmonella also protects biofilm acrB and tolC proceed in a in a ramA-dependent manner (Baucheron
cells from stressful conditions due to its wide range of substrates such as et al., 2014). Bile specifically binds to RamR (a repressor of ramA) and
diamide, hydrogen peroxide and Paraquat, alkyldimethylbenzylammo­ transcriptionally actives ramA (Ricci et al., 2012).Not only in these two
nium chloride (ADBAC), detergents, gold, dyes, SDS and novobiocin bacteria but also in several intestinal pathogens such a bile salt-
(Baugh et al., 2012; Sun et al., 2014). Decreased ability to form biofilms mediated transcription of efflux pump has been reported (Nishino
has already been observed in mutants without this pump (Baugh et al., et al., 2009).
2012). It has also been shown that long-term exposure to bile increases
biofilm formation in some bacterial species like Clostridium difficile,
4.2.2.2. Phenazines. The effect of phenazines in P. aeruginosa on biofilm Vibrio cholera and Shigella flexneri that is known to enteric bile salt-
formation varies from beneficial (as signal molecules) to harmful induced biofilm (Dubois et al., 2019; Hung et al., 2006; Nickerson and
(generating toxic superoxide species). Two samples of phenazine com­ Faherty, 2018). This is a transient phenomenon that prepares a rapid
pounds, phenazine-1-carboxylic acid (PCA) and pyocyanin (PYO), have dispersion opportunity to bacteria to colonize in the epithelial cell sur­
been studied for their effect on biofilm formation. PCA enhances biofilm faces in the small intestine and then reside in the colon where includes
formation in P. aeruginosa mutant lacking the ability to produce the low concentrations of the bile (Gholizadeh et al., 2019; Gupta et al.,
siderophores pyoverdine and pyochelin, by iron acquisition through the 2016). The VexAB pump of V. cholerae, the CmeABC Pump in
reduction of Fe (III) to Fe (II) (Wang et al., 2011). In contrast to PCA, Campylobacter jejuni, the HefC Efflux Pump in Helicobacter pylori,
pyocyanin (PYO) may promote biofilm development through a different EmrAB of the MFS in E. coli are the other examples of efflux pump which
mechanism. It is a virulence factor that acts as a signal molecule in are contributed in protection of toxic bile metabolites. However, a
addition to being an electron mediator. 5-methylphenazine-1-carbox­ definitive association between overexpression of the pump and bile-
ylate (5-Me-PCA) another intermediate compound with high redox po­ induced biofilm has not yet been investigated. Nevertheless,
tential has been introduced recently, which is involved in conversion of increasing multidrug resistance or enhances the pump of substrates
PCA to pyocyanin. It has been revealed to be essential for biofilm for­ involved in the extracellular component can be suggested as its func­
mation because it promotes extracellular electron transfer in the biofilm tional role in biofilm development. Furthermore, by removing the
(Sakhtah et al., 2016). In P. aeruginosa, MexEF-OprN and MexGHI- inducing amounts of bile salts, it will lead to more dispersion in the
OpmD pumps, in addition to the effect on fluoroquinolone resistance, lower parts of the gastrointestinal tract. Biofilm formation in efflux
are involved in controlling the concentrations of both toxic and non- pump mutants in the presence of inducing level of bile can assist to

12
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

elucidate this topic substantially. contributed in siderophore secretion. It has been suggested that the
transcription of this pump is inversely related to the environmental
4.2.2.4. Kynurenine. An intermediate metabolite in the tryptophan concentration of the iron and is in fact iron responsive. They showed that
degradation pathway, is used as a precursor of anthranilate and alkyl- the addition of FeCl3 in chemically defined medium (CDM) inhibited the
quinolone signal (AQs) molecules. This metabolite is also associated expression of the pump while the ferric uptake regulator protein (Fur)
with some phenotypes like swarming, antibiotic resistance (Butt et al., regulated its expression positively (Deng et al., 2012). Decreased biofilm
2016). Results revealed that kynurenine extrusion is mediated by formation and biofilm maturation in S. aureus according to combina­
MexCD-OprJ and MexCD-OprN in the P. aeruginosa which their over­ tional treatment of nilotinib (as an efflux pump inhibitor) and the flu­
expression lead to an impaired QS response (Alcalde-Rico et al., 2018) oroquinolone ciprofloxacin indicates importance of this pump in biofilm
and biofilm formation (Favre-Bonté et al., 2003). Despite the existence production process (Zimmermann et al., 2019).
of a wide range of different substrates for these pumps and other reasons Another study reported that MexAB-OprM and MexXY is overex­
for justifying this result, the author believed that this impairment is pressed in P. aeruginosa in iron depletion conditions, implying that these
mainly due to excessive removal of kynurenine as a precursor of HHQ in pumps is critical for P. aeruginosa survival under these conditions (Poole
the MexEF-OprN overexpressing mutant (Alcalde-Rico et al., 2018; et al., 1993). BpeAB-OprB was also shown to facilitate the production of
Olivares et al., 2012). siderophore in B. pseudomallei KHW. bpeAB null mutant and bpeR
(regulator of bpeAB) over expressing strains were impaired in side­
4.2.2.5. Spermidine. Spermidine is a polyamine compound that found rophore production and biofilm formation (Mima and Schweizer, 2010).
in many bacterial cells. It has role in various cellular processes like Therefore, the simultaneous use of iron chelators and inhibitors of these
regulation of biofilm formation (Thongbhubate et al., 2021), but the pumps could synergistically prevent the growth and formation of bio­
overaccumulation of this polyamine secondary metabolite can cause the films (Liu et al., 2010).
cell toxicity (Fukuchi et al., 1995). MdtIJ belongs to SMR superfamily is Excess haem (iron-containing molecule) acquisition into the bacte­
responsible for transporting excess spermidine out of the cell (Higashi rial cell will also disrupt haem natural homeostasis, resulting in the
et al., 2008). Considering that intracellular spermidine stimulates bio­ accumulation of an immediate heme precursor called protoporphyrin IX
film formation, it can be expected that deleting this pump will lead to (PPIX). Bacteria use a pump belonging to the ABC family (MacAB) to
increased biofilm formation. An increase in biofilm mass has previously regulate the intracellular concentration of these metabolite, which has
been reported in mdtJ-deficient mutants in E. coli K-12 (Bay et al., 2017). been reported in E. coli and Salmonella (Turlin et al., 2014). Deletion of
However, Thongbhubate and colleagues reported no difference in the this pump in S. enterica has led to a reduction in biofilm formation,
capacity of the wild type and deleted-mdtJ strains to achieve this phe­ which may also indicate its role in natural haem homeostasis in biofilm
nomena (Thongbhubate et al., 2021). In another study, even an increase cells (Baugh et al., 2012).
in the expression of this pump was observed during the growth of the 4.2.2.6.3. Nickel. It was confirmed that nickel triggers biofilm for­
biofilm (Kvist et al., 2008). mation by inducing the curli expression (csgBA genes) in sub-inhibitory
concentrations in E. coli (Perrin et al., 2009). In the presence of higher
4.2.2.6. Heavy metals concentrations, RcnAB efflux pump is used to transport the excess
4.2.2.6.1. Copper and silver. Copper and silver are two very toxic amount of nickel. The mutant ARY023 strain lacking rcnA (yohM) dis­
compounds for bacterial cells (Ameh et al., 2022) that have previously played significantly more susceptibility to Ni and Co and increased
been mentioned for their possible positive effects in the early stages of intracellular accumulation of nickel (Rodrigue et al., 2005).
biofilm formation. Therefore, reducing their intracellular concentration 4.2.2.6.4. Quaternary ammonium compound:. Different types of
by CusABCF pump (Betts, 2020), a heavy metal RND efflux pump (HME- multidrug-resistance efflux pumps for the extrusion of quaternary
RND), seems to be vital for bacterial survival and biofilm structure ammonium compounds have been investigated in both Gram-positive
maintenance. and Gram-negative bacteria that are also involved in the formation of
4.2.2.6.2. Iron. Iron mediates biofilm formation through oxidative biofilms (Buffet-Bataillon et al., 2016). For example, the members of
stress, regulating surface motility, stabilizing the polysaccharide matrix, MFS superfamily MdeA, NorB and NorC encoding genes, which function
serving as the signal agent and etc. On the other hand, intracellular as QAC transporter in S. aureus were upregulated in biofilm growth stage
overaccumulation poses iron-mediated oxidative stress that potentially (He and Ahn, 2011). Some studies have demonstrated that the presence
damage to bacterial cell. Acquisition of iron is mediated by siderophore, of these compounds in the environment trigger the expression of efflux
a small, high-affinity iron-chelating compound with high affinity for pumps and biofilm formation (Buffet-Bataillon et al., 2016). This in­
extracellular Fe3+. siderophore is secreted by efflux pumps, which in duction is probably due to the triggering of stress responses following
gram-negative bacteria due to the presence of a dual membranes it needs exposure to QACs, which is also effective in increasing the level of
to be exported in a multistep process. First both the ABC transporter and surviving persisters and dormant cells (Buffet-Bataillon et al., 2016).
major facilitator superfamily (MFS) transport siderophore through the Some efflux pumps are particular transporters of a specific metabo­
inner membrane. Second TolC-like efflux pumps excrete it across the lite that is upregulated in high amounts to protect the bacterium from
outer membrane. EntS (MFS-class efflux pump) in E. coli is responsible the compound’s harmful metabolites. The administration of p-hydrox­
for the export of enterobactin (known as strongest siderophore) from the ybenzoic acid (pHBA) increased the expression of yhcRQP, in E. coli.
cytoplasm to the periplasm. Then, enterobactin is transported across the pHBA is an aromatic organic acid metabolite produced during the ubi­
outer membrane by the coordination of the inner-membrane RND-class quinone production process, which is generally found in small amounts.
efflux pump (AcrB, AcrD, or MdtABC) with the outer membrane protein Hence they renamed this family to aromatic carboxylic acid efflux
channel TolC (Horiyama and Nishino, 2014). There are various RND (AaeAB efflux system) as metabolic relief valve to keep bacteria
pumps in other bacterial species as well, which are involved in side­ balanced metabolically (Van Dyk et al., 2004). Another example is SetA
rophore secretion, like the ApeX in Bacillus anthracis for petrobactin, and SetB in E. coli that belongs to MFS family is involved in glucose
VexGH in V. cholerae for vibriobactin, and both MmpL4 and MmpL5 in extrusion and upregulated during the biofilm maturation. One of the
Mycobacterium tuberculosis for mycobactin and carboxymycobactin. functions of this pump is most likely to protect bacterial cells from the
PvdRT-OpmQ and MdtABC-OpmB in P. aeruginosa for pyoverdine, accumulation of non-metabolic carbohydrates like IPTG and methyl-β-D-
MacAB in S. enterica and E. coli for salmochelin and enterotoxin are thiogalactoside (TMG) that are harmful to them (Liu et al., 1999a; Liu
examples of ABC siderophore transporters (Liu et al., 2022). et al., 1999b).
NorA (MFS efflux pump) as a main efflux pump of S. aureus is

13
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Table 4
Summary of potential efflux pumps involved in production of biofilm exopolymeric substances (EPS).
Efflux pump/ Class of Bacterial species Potential substrate/Role Main function
Component pump

Exopolymeric Exopolysaccharides SetB MFS E. coli Glucose Biofilm matrix generation


substance (EPS) EmrE SMR E. coli Arabinose Exopolysaccharide matrix
AraE - Salmonella spp Arabinose generation
A1S_1117 - A. baumannii Sugars Repressor role in regulation and
TetA(C) MFS Salmonella & Tetracycline production of curli and c-diGMP
Alg44 - E. coli Associated with the ABC transporter- Biofilm matrix generation
LT42 ABC Pseudomonas dependent EPS assembly system/ Promotion of colanic acid
S. mutants interaction with AlgE transporter (cspE) expression
Glucosyltransferase Synthesis and secretion of alginate
Sucrose-associated colonization in
vivo
Amino acids and YhdWXYZ ABC E. coli L-leucine Stability of the aggregation
peptides YfiK & YdeD E. coli L-cysteine Stability of the aggregation
eDNA PMT MFS A. baumannii eDNA Adherence to abiotic (polystyrene)
AIIMS 7 & E. coli and biotic (S. cerevisiae/HeLa)
surfaces

4.2.3. Biofilm matrix membrane protein) are two component involved in synthesis and
secretion of alginate, respectively (Hay et al., 2010). The periplasmic C
4.2.3.1. Exopolysaccharides. Sugars, often in the form of poly­ terminus of Alg44 is hypothesized to be homologous to membrane
saccharides in combination with other substances, make up a large fusion proteins implicated in periplasmic transforming in efflux pumps
portion of the polymer structure. Some of well-known biofilm matrix (Remminghorst and Rehm, 2006). Previous studies have reported a
sugars are including polyanionic (alginate and colanic acid) and Poly­ decrease in algE expression in mutants without Alg44 (Oglesby et al.,
cationic (N-acetylglucosamine) exopolysaccharides (Allison, 2003; 2008). Alg44, which is associated with the ABC transporter-dependent
Flemming and Wingender, 2010). EPS assembly system model and drug efflux pumps, might also
The uptake and export of many hexoses like glucose, galactose, interact with AlgE (Oglesby et al., 2008). In agreement with role of
mannose, and fructose in bacteria generally are mediated by sugar efflux Alg44 in synthesis and secretion of the exopolysaccharide alginate, the
transporter, a group of transporters similar to MFS proteins (Liu et al., alginate co-polymerase Alg44 deleted strains produced a thin and low
1999b). It seems the function of these transporters affect the composi­ cohesive biofilm in compared with wild strain (Noirot-Gros et al., 2019;
tion of the biofilm matrix (Pasqua et al., 2019). The previously described Whitney et al., 2015).
upregulation of SetB pump in E. coli can be expanded to participation in An ABC transporter (LT42) and a putative multidrug efflux pump
the biofilm matrix generation by glucose excretion as well (Pasqua et al., (LT43) have been identified in S. mutants which seem important in
2019). Arabinose is a 5-carbon monosaccharide that uses AraJ (Reeder sucrose-associated colonization in vivo. The deleted multidrug efflux
and Schleif, 1991), Cmr (mdfA) (Kredich and Tomkins, 1966), EmrD pump mutant was impaired to colonize the rats otherwise had normal
(Koita and Rao, 2012) and YeA (Bost et al., 1999) pumps to transmit glucosyltransferase activities (Tao and Tanzer, 2002).
through bacterial membranes. The emrE lacking strains showed a
decrease in biofilm formation that some investigations proposed the 4.2.3.2. Amino acids and peptides. Unlike D-amino acids, positively
contribution of arabinose in exopolysaccharide matrix (Alav et al., 2018; charged L-amino acids help in the stability of the aggregation in biofilm
Matsumura et al., 2011). Nevertheless Erin et al., observed a hyper­ by interacting with other biofilm matrix components (Flemming and
biofilm structure in araE-deleted mutant of Salmonella in the presence of Wingender, 2010). YhdWXYZ system, an ABC-type transporter, in E. coli
L-arabinose (Vasicek et al., 2021). Subsequently the addition of arabi­ has role in L-leucine transportation (Zheng et al., 2018). An almost two-
nose to the culture medium increased the thickness of the biofilm fold increase in yhdX gene expression in biofilm cells has been reported
without causing dispersion. It is probable that arabinose suppresses the compared to planktonic cells (Riley et al., 2006). YfiK incorporation
expression and production of curli fimbriae and GMP as two controlling with YdeD protein exported L-cysteine cross the membrane in E. coli
factors of the extracellular matix. (Vasicek et al., 2021). The biofilm (Franke et al., 2003).
cells, not the planktonic cells in Acinetobacter, encode the efflux gene
A1S_1117, which belongs to the msf family (Rumbo-Feal et al., 2013). 4.2.3.3. Extracellular DNA (eDNA). In some bacteria, such as S. aureus
Several studies have suggested that this pump may be involved in sugar and A. baumannii, eDNA is one of the primary biofilm matrix compo­
transport and biofilm matrix formation, but this has not yet been nents, which could lead to a more compressed and stable biofilm. A MFS
confirmed (Alav et al., 2018; AlMatar et al., 2021). transporter-like ORF (PMT) of 453 in a pathogenic strain A. baumannii
Colanic acid in bacteria such as Salmonella and E. coli contribute in AIIMS 7 has been described that may has a role in adherence to abiotic
biofilm matrix formation and its encoding genes are upregulated in (polystyrene) and biotic (S. cerevisiae/HeLa) surfaces (Sahu et al.,
during biofilm formation (Prigent-Combaret et al., 1999; Prigent-Com­ 2012). The results confirmed a 2.58-fold increase in the production of
baret et al., 2000). It’s been confirmed an overexpression of the tetra­ eDNA in PMT gene-cloned E. coli in comparison with E. coli harboring a
cycline resistance efflux pump TetA(C) pump in response to osmotic control plasmid.
stress condition. Results showed enhanced expression levels of colanic There is a list of efflux pumps in Table 4 which have probable role in
acid transporter (cspE) in presence of 50 mM NaCl and antibiotic secretion or production of exopolymeric substances according to the
(Ampicillin and Tetracycline) in that indicate contribution of high-level studies.
TetA(C) pumps in promotion of colanic acid biosynthesis and biofilm
maturation (May et al., 2009). 5. Conclusion
Alginates are the most abundant exopolysaccharides in the Pseudo­
monas biofilm matrix in the lungs of cystic fibrosis patients (Hentzer Bacterial biofilms have always been one of the dilemmas in the field
et al., 2001). Alg44 (membrane-anchored protein) and AlgE (outer of the health-care system, especially in relation to catheters, artificial

14
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

organs and other medical equipment. The biofilm structure possesses its keeping the robustness and the plasticity of this intercellular signaling network.
bioRxiv. https://doi.org/10.1101/2020.03.10.986737.
resistance to drugs and conditions to several mechanisms that make it
Alcalde-Rico, M., Olivares-Pacheco, J., Halliday, N., Cámara, M., Martínez, J.L., 2020b.
difficult and sometimes impossible to overcome with it. Efflux pumps, The impaired quorum sensing response of MexAB-OprM efflux pump overexpressing
which are usually known for their role in multidrug resistance, seem to mutants is not due to non-physiological efflux of 3-oxo-C12-HSL. Environ. Microbiol.
be effective in the formation of biofilm structure in addition to drug 22.
Allison, D.G., 2003. The biofilm matrix. Biofouling 19, 139–150.
extrusion. Based on the studies, it seems that the expression of efflux AlMatar, M., Albarri, O., Makky, E.A., Köksal, F., 2021. Efflux pump inhibitors: new
pumps is different in various places and stages of biofilm formation updates. Pharmacol. Rep. 73, 1–16.
under the influence of their role in interactions, mobility, gene regula­ Alvarez-Ortega, C., Olivares, J., Martínez, J.L., 2013. RND multidrug efflux pumps: what
are they good for? Front. Microbiol. 4, 7.
tion, QS, secretion of EPS, and the secondary metabolites. Therefore, it is Ameh, T., Gibb, M., Stevens, D., Pradhan, S.H., Braswell, E., Sayes, C.M., 2022. Silver and
possible that the incorrect use of antibiotics or efflux pump inhibitors, copper nanoparticles induce oxidative stress in bacteria and mammalian cells.
instead of proper treatment, can induce the expression of a pump Nanomaterials 12, 2402.
Aslam, B., Wang, W., Arshad, M.I., Khurshid, M., Muzammil, S., Rasool, M.H., Nisar, M.
effective in biofilm formation or inhibit a pump with a negative effect on A., Alvi, R.F., Aslam, M.A., Qamar, M.U., 2018. Antibiotic resistance: a rundown of a
it, respectively. Therefore, in order to choose the correct combat strat­ global crisis. Infect. Drug Resist. 11, 1645.
egy, it is recommended to carefully examine the role of pumps in biofilm Aybey, A., Usta, A., Demirkan, E., 2021. Effects of psychotropic drugs as bacterial efflux
pump inhibitors on quorum sensing regulated behaviors. J. Microbiol. Biotechnol.
formation in each bacterium, range of substrates, and their expression Food Sci. 2021, 128–131.
level separately. Babin, B.M., Atangcho, L., van Eldijk, M.B., Sweredoski, M.J., Moradian, A., Hess, S.,
Tolker-Nielsen, T., Newman, D.K., Tirrell, D.A., 2017. Selective proteomic analysis of
antibiotic-tolerant cellular subpopulations in Pseudomonas aeruginosa biofilms. MBio
Funding
8 e01593-01517.
Bassler, B.L., 1999. How bacteria talk to each other: regulation of gene expression by
This study was supported by Drug Applied Reserahc Center, Tabriz quorum sensing. Curr. Opin. Microbiol. 2, 582–587.
Bassler, B.L., Losick, R., 2006. Bacterially speaking. Cell 125, 237–246.
University of Medical Sciences as a grant to Dr Hossein Samadi Kafil.
Baucheron, S., Nishino, K., Monchaux, I., Canepa, S., Maurel, M.-C., Coste, F.,
Roussel, A., Cloeckaert, A., Giraud, E., 2014. Bile-mediated activation of the acrAB
Code availability and tolC multidrug efflux genes occurs mainly through transcriptional derepression
of ramA in Salmonella enterica serovar Typhimurium. J. Antimicrob. Chemother. 69,
2400–2406.
Not applicable. Baugh, S., Ekanayaka, A.S., Piddock, L.J., Webber, M.A., 2012. Loss of or inhibition of all
multidrug resistance efflux pumps of Salmonella enterica serovar Typhimurium
Ethics approval results in impaired ability to form a biofilm. J. Antimicrob. Chemother. 67,
2409–2417.
Bay, D.C., Stremick, C.A., Slipski, C.J., Turner, R.J., 2017. Secondary multidrug efflux
This study was a literature review and Ethic was not applicable. pump mutants alter Escherichia coli biofilm growth in the presence of cationic
antimicrobial compounds. Res. Microbiol. 168, 208–221.
Betts, H.D., 2020. An investigation of the chemistry of silver in biological systems, and
Authors’ contributions the development of silver-containing materials for use as antibacterial agents.
Bhagwat, A.A., Gross, K.C., Tully, R.E., Keister, D.L., 1996. Beta-glucan synthesis in
H.S.K participated in data collection, annotation, drafting manu­ Bradyrhizobium japonicum: characterization of a new locus (ndvC) influencing beta-
(1–> 6) linkages. J. Bacteriol. 178, 4635–4642.
script, writing and final approve. Bialek, S., Lavigne, J.-P., Chevalier, J., Marcon, E., Leflon-Guibout, V., Davin, A.,
M.N.H participated in data collection, annotation, drafting manu­ Moreau, R., Pagès, J.-M., Nicolas-Chanoine, M.-H., 2010. Membrane efflux and
script, writing and final approve. influx modulate both multidrug resistance and virulence of Klebsiella pneumoniae in a
Caenorhabditis elegans model. Antimicrob. Agents Chemother. 54, 4373–4378.
Bialek-Davenet, S., Criscuolo, A., Ailloud, F., Passet, V., Jones, L., Delannoy-Vieillard, A.-
Consent to participate S., Garin, B., Le Hello, S., Arlet, G., Nicolas-Chanoine, M.-H., 2014. Genomic
definition of hypervirulent and multidrug-resistant Klebsiella pneumoniae clonal
groups. Emerg. Infect. Dis. 20, 1812.
Not applicable. Bialvaei, A.Z., Kafil, H.S., Asgharzadeh, M., Yousef Memar, M., Yousefi, M., 2016.
Current methods for the identification of carbapenemases. J. Chemother. 28, 1–19.
Consent for publication Blaschke, U., Skiebe, E., Wilharm, G., 2021. Novel genes required for surface-associated
motility in Acinetobacter baumannii. Curr. Microbiol. 78, 1509–1528.
Borisov, V.B., Siletsky, S.A., Paiardini, A., Hoogewijs, D., Forte, E., Giuffrè, A., Poole, R.
Not applicable. K., 2020. Bacterial oxidases of the cytochrome bd family: redox enzymes of unique
structure, function, and utility as drug targets. In: Antioxidants & Redox Signaling.
Bost, S., Silva, F., Belin, D., 1999. Transcriptional activation of ydeA, which encodes a
Declaration of Competing Interest member of the major facilitator superfamily, interferes with arabinose accumulation
and induction of the Escherichia coli arabinose PBAD promoter. J. Bacteriol. 181,
None to declare. 2185–2191.
Boudarel, H., Mathias, J.-D., Blaysat, B., Grédiac, M., 2018. Towards standardized
mechanical characterization of microbial biofilms: analysis and critical review. NPJ
Data availability Biofilms Microbiom. 4, 1–15.
Buckley, A.M., Webber, M.A., Cooles, S., Randall, L.P., La Ragione, R.M., Woodward, M.
J., Piddock, L.J., 2006. The AcrAB–TolC efflux system of Salmonella enterica serovar
No data was used for the research described in the article. Typhimurium plays a role in pathogenesis. Cell. Microbiol. 8, 847–856.
Buckner, M.M., Blair, J.M., La Ragione, R.M., Newcombe, J., Dwyer, D.J., Ivens, A.,
References Piddock, L.J., 2016. Beyond antimicrobial resistance: evidence for a distinct role of
the AcrD efflux pump in Salmonella biology. MBio 7, e01916.
Buffet-Bataillon, S., Tattevin, P., Maillard, J.-Y., Bonnaure-Mallet, M., Jolivet-
Aendekerk, S., Ghysels, B., Cornelis, P., Baysse, C., 2002. Characterization of a new efflux
Gougeon, A., 2016. Efflux pump induction by quaternary ammonium compounds
pump, MexGHI-OpmD, from Pseudomonas aeruginosa that confers resistance to
and fluoroquinolone resistance in bacteria. Future Microbiol. 11, 81–92.
vanadium. Microbiology 148, 2371–2381.
Butt, A., Halliday, N., Williams, P., Atkins, H.S., Bancroft, G.J., Titball, R.W., 2016.
Alav, I., Sutton, J.M., Rahman, K.M., 2018. Role of bacterial efflux pumps in biofilm
Burkholderia pseudomallei kynB plays a role in AQ production, biofilm formation,
formation. J. Antimicrob. Chemother. 73, 2003–2020.
bacterial swarming and persistence. Res. Microbiol. 167, 159–167.
Alcalde-Rico, M., Hernando-Amado, S., Blanco, P., Martínez, J.L., 2016. Multidrug efflux
Chan, Y.Y., Chua, K.L., 2005. The Burkholderia pseudomallei BpeAB-OprB efflux pump:
pumps at the crossroad between antibiotic resistance and bacterial virulence. Front.
expression and impact on quorum sensing and virulence. J. Bacteriol. 187,
Microbiol. 7, 1483.
4707–4719.
Alcalde-Rico, M., Olivares-Pacheco, J., Alvarez-Ortega, C., Cámara, M., Martínez, J.L.,
Chart, H., Spencer, J., Smith, H.R., Rowe, B., 1997. Magnesium ions are required for
2018. Role of the multidrug resistance efflux pump MexCD-OprJ in the Pseudomonas
HEp-2 cell adhesion by enteroaggregative strains of Escherichia coli O126: H27 and
aeruginosa quorum sensing response. Front. Microbiol. 2752.
O44: H18. FEMS Microbiol. Lett. 148, 49–52.
Alcalde-Rico, M., Olivares-Pacheco, J., Halliday, N., Cámara, M., Martínez, J.L., 2020a.
Chen, F., Pang, X.-Y., Shen, C., Han, L.-Z., Deng, Y.-X., Chen, X.-S., Zhang, J.-J., Xia, Q.,
The analysis of the role of MexAB-OprM on quorum sensing homeostasis shows that
Qian, Y.-B., 2020. High mortality associated with gram-negative bacterial
the apparent redundancy of Pseudomonas aeruginosa multidrug efflux pumps allows

15
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

bloodstream infection in liver transplant recipients undergoing immunosuppression Guttenplan, S.B., Kearns, D.B., 2013. Regulation of flagellar motility during biofilm
reduction. World J. Gastroenterol. 26, 7191. formation. FEMS Microbiol. Rev. 37, 849–871.
Conlon, B.P., Rowe, S.E., Lewis, K., 2015. Persister cells in biofilm associated infections. Hall, C.W., Mah, T.-F., 2017. Molecular mechanisms of biofilm-based antibiotic
In: Biofilm-Based Healthcare-Associated Infections, pp. 1–9. resistance and tolerance in pathogenic bacteria. FEMS Microbiol. Rev. 41, 276–301.
Costerton, J.W., Geesey, G.G., Cheng, K.-J., 1978. How bacteria stick. Sci. Am. 238, Harrington, S.M., Dudley, E.G., Nataro, J.P., 2006. Pathogenesis of enteroaggregative
86–95. Escherichia coli infection. FEMS Microbiol. Lett. 254, 12–18.
Csonka, L.N., 1989. Physiological and genetic responses of bacteria to osmotic stress. Harshey, R.M., 2003. Bacterial motility on a surface: many ways to a common goal.
Microbiol. Rev. 53, 121–147. Annu. Rev. Microbiol. 57, 249–273.
Davies, D.G., 2011. Biofilm dispersion. In: Biofilm Highlights. Springer, pp. 1–28. Hay, I.D., Rehman, Z.U., Rehm, B.H., 2010. Membrane topology of outer membrane
Dawan, J., Li, Y., Lu, F., He, X., Ahn, J., 2022. Role of efflux pump-mediated antibiotic protein AlgE, which is required for alginate production in Pseudomonas aeruginosa.
resistance in quorum sensing-regulated biofilm formation by Salmonella typhimurium. Appl. Environ. Microbiol. 76, 1806–1812.
Pathogens 11, 147. He, X., Ahn, J., 2011. Differential gene expression in planktonic and biofilm cells of
de Aguiar Coletti, T.M.S.F., De Freitas, L.M., Almeida, A.M.F., Fontana, C.R., 2017. multiple antibiotic-resistant Salmonella typhimurium and Staphylococcus aureus. FEMS
Optimization of antimicrobial photodynamic therapy in biofilms by inhibiting efflux Microbiol. Lett. 325, 180–188.
pump. Photomed. Laser Surg. 35, 378–385. He, X., Lu, F., Yuan, F., Jiang, D., Zhao, P., Zhu, J., Cheng, H., Cao, J., Lu, G., 2015.
de Araújo, L.C.A., de Oliveira, M.B.M., 2019. Effect of heavy metals on the biofilm Biofilm formation caused by clinical Acinetobacter baumannii isolates is associated
formed by microorganisms from impacted aquatic environments. In: Bacterial with overexpression of the AdeFGH efflux pump. Antimicrob. Agents Chemother. 59,
Biofilms. IntechOpen. 4817–4825.
De Kievit, T.R., Parkins, M.D., Gillis, R.J., Srikumar, R., Ceri, H., Poole, K., Iglewski, B.H., Heatwole, V.M., Somerville, R.L., 1991. Cloning, nucleotide sequence, and
Storey, D.G., 2001. Multidrug efflux pumps: expression patterns and contribution to characterization of mtr, the structural gene for a tryptophan-specific permease of
antibiotic resistance in Pseudomonas aeruginosa biofilms. Antimicrob. Agents Escherichia coli K-12. J. Bacteriol. 173, 108–115.
Chemother. 45, 1761–1770. Hecker, M., Völker, U., 2001. General Stress Response of Bacillus subtilis and Other
Deng, X., Sun, F., Ji, Q., Liang, H., Missiakas, D., Lan, L., He, C., 2012. Expression of Bacteria.
multidrug resistance efflux pump gene norA is iron responsive in Staphylococcus Hengge, R., 2009. Principles of c-di-GMP signalling in bacteria. Nat. Rev. Microbiol. 7,
aureus. J. Bacteriol. 194, 1753–1762. 263–273.
Deng, Z., Shan, Y., Pan, Q., Gao, X., Yan, A., 2013. Anaerobic expression of the gadE- Hentzer, M., Teitzel, G.M., Balzer, G.J., Heydorn, A., Molin, S., Givskov, M., Parsek, M.R.,
mdtEF multidrug efflux operon is primarily regulated by the two-component system 2001. Alginate overproduction affects Pseudomonas aeruginosa biofilm structure and
ArcBA through antagonizing the H-NS mediated repression. Front. Microbiol. 4, 194. function. J. Bacteriol. 183, 5395–5401.
Dickson, J.S., Koohmaraie, M., 1989. Cell surface charge characteristics and their Higashi, K., Ishigure, H., Demizu, R., Uemura, T., Nishino, K., Yamaguchi, A.,
relationship to bacterial attachment to meat surfaces. Appl. Environ. Microbiol. 55, Kashiwagi, K., Igarashi, K., 2008. Identification of a spermidine excretion protein
832–836. complex (MdtJI) in Escherichia coli. J. Bacteriol. 190, 872–878.
Dietrich, L.E., Price-Whelan, A., Petersen, A., Whiteley, M., Newman, D.K., 2006. The Hirakawa, H., Inazumi, Y., Masaki, T., Hirata, T., Yamaguchi, A., 2005. Indole induces
phenazine pyocyanin is a terminal signalling factor in the quorum sensing network the expression of multidrug exporter genes in Escherichia coli. Mol. Microbiol. 55,
of Pseudomonas aeruginosa. Mol. Microbiol. 61, 1308–1321. 1113–1126.
Donlan, R.M., 2002. Biofilms: microbial life on surfaces. Emerg. Infect. Dis. 8, 881. Horiyama, T., Nishino, K., 2014. AcrB, AcrD, and MdtABC multidrug efflux systems are
Donlan, R.M., 2016. Microbial Biofilms. involved in enterobactin export in Escherichia coli. PLoS One 9, e108642.
Du, X., Li, Y., Zhou, Q., Xu, Y., 2015. Regulation of gene expression in Pseudomonas Hou, B., Meng, X.-R., Zhang, L.-Y., Tan, C., Jin, H., Zhou, R., Gao, J.-F., Wu, B., Li, Z.-L.,
aeruginosa M18 by phenazine-1-carboxylic acid. Appl. Microbiol. Biotechnol. 99, Liu, M., 2014. TolC promotes ExPEC biofilm formation and curli production in
813–825. response to medium osmolarity. BioMed Res. Int. 2014.
Du, D., Wang-Kan, X., Neuberger, A., Van Veen, H.W., Pos, K.M., Piddock, L.J., Luisi, B. Huang, Y., Zhou, E., Jiang, C., Jia, R., Liu, S., Xu, D., Gu, T., Wang, F., 2018. Endogenous
F., 2018. Multidrug efflux pumps: structure, function and regulation. Nat. Rev. phenazine-1-carboxamide encoding gene PhzH regulated the extracellular electron
Microbiol. 16, 523–539. transfer in biocorrosion of stainless steel by marine Pseudomonas aeruginosa.
Dubois, T., Tremblay, Y.D., Hamiot, A., Martin-Verstraete, I., Deschamps, J., Monot, M., Electrochem. Commun. 94, 9–13.
Briandet, R., Dupuy, B., 2019. A microbiota-generated bile salt induces biofilm Hung, D.T., Zhu, J., Sturtevant, D., Mekalanos, J.J., 2006. Bile acids stimulate biofilm
formation in Clostridium difficile. npj Biofilms Microbiom. 5, 1–12. formation in Vibrio cholerae. Mol. Microbiol. 59, 193–201.
Ebbensgaard, A.E., Løbner-Olesen, A., Frimodt-Møller, J., 2020. The role of efflux pumps Ichinose, Y., Nishimura, T., Harada, M., Kashiwagi, R., Yamamoto, M., Noutoshi, Y.,
in the transition from low-level to clinical antibiotic resistance. Antibiotics 9, 855. Toyoda, K., Taguchi, F., Takemoto, D., Matsui, H., 2020. Role of two sets of RND-
Fabrega, A., Soto, S.M., Ballesté-Delpierre, C., Fernández-Orth, D., Jiménez de Anta, M. type multidrug efflux pump transporter genes, mexAB-oprM and mexEF-oprN, in
T., Vila, J., 2014. Impact of quinolone-resistance acquisition on biofilm production virulence of Pseudomonas syringae pv. tabaci 6605. Plant Pathol. J. 36, 148.
and fitness in Salmonella enterica. J. Antimicrob. Chemother. 69, 1815–1824. Imuta, N., Nishi, J., Tokuda, K., Fujiyama, R., Manago, K., Iwashita, M., Sarantuya, J.,
Favre-Bonté, S., Köhler, T., Van Delden, C., 2003. Biofilm formation by Pseudomonas Kawano, Y., 2008. The Escherichia coli efflux pump TolC promotes aggregation of
aeruginosa: role of the C4-HSL cell-to-cell signal and inhibition by azithromycin. enteroaggregative E. coli 042. Infect. Immun. 76, 1247–1256.
J. Antimicrob. Chemother. 52, 598–604. Irving, S.E., Corrigan, R.M., 2018. Triggering the stringent response: signals responsible
Fisher, R.A., Gollan, B., Helaine, S., 2017. Persistent bacterial infections and persister for activating (p) ppGpp synthesis in bacteria. Microbiology 164, 268–276.
cells. Nat. Rev. Microbiol. 15, 453. Ito, A., Taniuchi, A., May, T., Kawata, K., Okabe, S., 2009. Increased antibiotic resistance
Flemming, H.-C., Wingender, J., 2010. The biofilm matrix. Nat. Rev. Microbiol. 8, of Escherichia coli in mature biofilms. Appl. Environ. Microbiol. 75, 4093–4100.
623–633. Javaux, C., Joris, B., De Witte, P., 2007. Functional characteristics of TauA binding
Flemming, H.-C., Wingender, J., Szewzyk, U., Steinberg, P., Rice, S.A., Kjelleberg, S., protein from TauABC Escherichia coli system. Protein J. 26, 231–238.
2016. Biofilms: an emergent form of bacterial life. Nat. Rev. Microbiol. 14, 563. Jefferson, K.K., 2004. What drives bacteria to produce a biofilm? FEMS Microbiol. Lett.
Franke, I., Resch, A., Daßler, T., Maier, T., Böck, A., 2003. YfiK from Escherichia coli 236, 163–173.
promotes export of O-acetylserine and cysteine. J. Bacteriol. 185, 1161–1166. Jin, K., Zhou, L., Jiang, H., Sun, S., Fang, Y., Liu, J., Zhang, X., He, Y.-W., 2015.
Frisk, A., Schurr, J.R., Wang, G., Bertucci, D.C., Marrero, L., Hwang, S.H., Hassett, D.J., Engineering the central biosynthetic and secondary metabolic pathways of
Schurr, M.J., 2004. Transcriptome analysis of Pseudomonas aeruginosa after Pseudomonas aeruginosa strain PA1201 to improve phenazine-1-carboxylic acid
interaction with human airway epithelial cells. Infect. Immun. 72, 5433–5438. production. Metab. Eng. 32, 30–38.
Fukuchi, J.-i., Kashiwagi, K., Yamagishi, M., Ishihama, A., Igarashi, K., 1995. Decrease in Johnson, L.A., Hug, L.A., 2019. Distribution of reactive oxygen species defense
cell viability due to the accumulation of spermidine in spermidine acetyltransferase- mechanisms across domain bacteria. Free Radic. Biol. Med. 140, 93–102.
deficient mutant of Escherichia coli (*). J. Biol. Chem. 270, 18831–18835. Joo, H.-S., Otto, M., 2012. Molecular basis of in vivo biofilm formation by bacterial
Galinski, E.A., 1995. Osmoadaptation in bacteria. Adv. Microb. Physiol. 37, 273–328. pathogens. Chem. Biol. 19, 1503–1513.
Gasperini, B., Cherubini, A., Lucarelli, M., Espinosa, E., Prospero, E., 2021. Multidrug- Kafil, H.S., Mobarez, A.M., 2015a. Assessment of biofilm formation by enterococci
resistant bacterial infections in geriatric hospitalized patients before and after the isolates from urinary tract infections with different virulence profiles. J. King Saud
COVID-19 outbreak: results from a retrospective observational study in two geriatric Univ.-Sci. 27, 312–317.
wards. Antibiotics 10, 95. Kafil, H.S., Mobarez, A.M., 2015b. Spread of enterococcal surface protein in antibiotic
Gholizadeh, P., Mahallei, M., Pormohammad, A., Varshochi, M., Ganbarov, K., resistant entero-coccus faecium and Enterococcus faecalis isolates from urinary tract
Zeinalzadeh, E., Yousefi, B., Bastami, M., Tanomand, A., Mahmood, S.S., Yousefi, M., infections. Open Microbiol. J. 9, 14–17.
Asgharzadeh, M., Kafil, H.S., 2019. Microbial balance in the intestinal microbiota Kafil, H.S., Mobarez, A.M., Moghadam, M.F., 2013. Adhesion and virulence factor
and its association with diabetes, obesity and allergic disease. Microb. Pathog. 127, properties of Enterococci isolated from clinical samples in Iran. Indian J. Pathol.
48–55. Microbiol. 56, 238.
Gilbert, P., McBain, A.J., 2001. Biofilms: their impact on health and their recalcitrance Kafil, H.S., Mobarez, A.M., Moghadam, M.F., Hashemi, Z.S., Yousefi, M., 2016.
toward biocides. Am. J. Infect. Control 29, 252–255. Gentamicin induces efaA expression and biofilm formation in Enterococcus faecalis.
Gillis, R.J., White, K.G., Choi, K.-H., Wagner, V.E., Schweizer, H.P., Iglewski, B.H., 2005. Microb. Pathog. 92, 30–35.
Molecular basis of azithromycin-resistant Pseudomonas aeruginosa biofilms. Karygianni, L., Ren, Z., Koo, H., Thurnheer, T., 2020. Biofilm matrixome: extracellular
Antimicrob. Agents Chemother. 49, 3858–3867. components in structured microbial communities. Trends Microbiol 28, 668–681.
Greene, N.P., Koronakis, V., 2021. Bacterial metal resistance: coping with copper without Kawamura-Sato, K., Shibayama, K., Horii, T., Iimuma, Y., Arakawa, Y., Ohta, M., 1999.
cooperativity? mBio 12, e0065321. Role of multiple efflux pumps in Escherichia coli in indole expulsion. FEMS
Gupta, P., Sarkar, S., Das, B., Bhattacharjee, S., Tribedi, P., 2016. Biofilm, pathogenesis Microbiol. Lett. 179, 345–352.
and prevention—a journey to break the wall: a review. Arch. Microbiol. 198, 1–15.

16
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Kim, J., Park, W., 2015. Indole: a signaling molecule or a mere metabolic byproduct that Matsumura, K., Furukawa, S., Ogihara, H., Morinaga, Y., 2011. Roles of multidrug efflux
alters bacterial physiology at a high concentration? J. Microbiol. 53, 421–428. pumps on the biofilm formation of Escherichia coli K-12. Biocontrol Sci. 16, 69–72.
Kim, S.-M., Lee, H.-W., Choi, Y.-W., Kim, S.-H., Lee, J.-C., Lee, Y.-C., Seol, S.-Y., Cho, D.- Mauter, M., Fait, A., Elimelech, M., Herzberg, M., 2013. Surface cell density effects on
T., Kim, J., 2012. Involvement of curli fimbriae in the biofilm formation of Escherichia coli gene expression during cell attachment. Environ. Sci. Technol. 47,
Enterobacter cloacae. J. Microbiol. 50, 175–178. 6223–6230.
Kjelleberg, S., Molin, S., 2002. Is there a role for quorum sensing signals in bacterial May, T., Ito, A., Okabe, S., 2009. Induction of multidrug resistance mechanism in
biofilms? Curr. Opin. Microbiol. 5, 254–258. Escherichia coli biofilms by interplay between tetracycline and ampicillin resistance
Klausen, M., Heydorn, A., Ragas, P., Lambertsen, L., Aaes-Jørgensen, A., Molin, S., genes. Antimicrob. Agents Chemother. 53, 4628–4639.
Tolker-Nielsen, T., 2003. Biofilm formation by Pseudomonas aeruginosa wild type, McDougald, D., Rice, S.A., Barraud, N., Steinberg, P.D., Kjelleberg, S., 2012. Should we
flagella and type IV pili mutants. Mol. Microbiol. 48, 1511–1524. stay or should we go: mechanisms and ecological consequences for biofilm dispersal.
Koita, K., Rao, C.V., 2012. Identification and analysis of the putative pentose sugar efflux Nat. Rev. Microbiol. 10, 39–50.
transporters in Escherichia coli. Miller, A.L., Bessho, S., Grando, K., Tükel, Ç., 2021. Microbiome or infections: amyloid-
Koo, H., Yamada, K.M., 2016. Dynamic cell–matrix interactions modulate microbial containing biofilms as a trigger for complex human diseases. Front. Immunol. 12,
biofilm and tissue 3D microenvironments. Curr. Opin. Cell Biol. 42, 102–112. 514.
Kordmahaleh, F.A., Shalke, S.E., 2013. Bacterial biofilms: microbial life on surfaces. Mima, T., Schweizer, H.P., 2010. The BpeAB-OprB efflux pump of Burkholderia
J. Biol. 2, 242–248. pseudomallei 1026b does not play a role in quorum sensing, virulence factor
Koronakis, V., Eswaran, J., Hughes, C., 2004. Structure and function of TolC: the production, or extrusion of aminoglycosides but is a broad-spectrum drug efflux
bacterial exit duct for proteins and drugs. Annu. Rev. Biochem. 73, 467–489. system. Antimicrob. Agents Chemother. 54, 3113–3120.
Kredich, N.M., Tomkins, G.M., 1966. The enzymic synthesis of L-cysteine in Escherichia Minagawa, S., Inami, H., Kato, T., Sawada, S., Yasuki, T., Miyairi, S., Horikawa, M.,
coli and Salmonella typhimurium. J. Biol. Chem. 241, 4955–4965. Okuda, J., Gotoh, N., 2012. RND type efflux pump system MexAB-OprM of
Kumar, S., Chandra, N., Singh, L., Hashmi, M.Z., Varma, A., 2019. Biofilms in Human Pseudomonas aeruginosa selects bacterial languages, 3-oxo-acyl-homoserine lactones,
Diseases: Treatment and Control. Springer. for cell-to-cell communication. BMC Microbiol. 12, 1–10.
Kumar, S., Lekshmi, M., Parvathi, A., Ojha, M., Wenzel, N., Varela, M.F., 2020. Mireles, J.R., Toguchi, A., Harshey, R.M., 2001. Salmonella enterica serovar Typhimurium
Functional and structural roles of the major facilitator superfamily bacterial swarming mutants with altered biofilm-forming abilities: surfactin inhibits biofilm
multidrug efflux pumps. Microorganisms 8, 266. formation. J. Bacteriol. 183, 5848–5854.
Kvist, M., Hancock, V., Klemm, P., 2008. Inactivation of efflux pumps abolishes bacterial Mohamad, F., Alzahrani, R.R., Alsaadi, A., Alrfaei, B.M., Yassin, A.E.B., Alkhulaifi, M.M.,
biofilm formation. Appl. Environ. Microbiol. 74, 7376–7382. Halwani, M., 2023. An explorative review on advanced approaches to overcome
Lamarche, M.G., Déziel, E., 2011. MexEF-OprN efflux pump exports the Pseudomonas bacterial resistance by curbing bacterial biofilm formation. Infect. Drug Resist.
quinolone signal (PQS) precursor HHQ (4-hydroxy-2-heptylquinoline). PLoS One 6, 19–49.
e24310. Motta, J.-P., Wallace, J.L., Buret, A.G., Deraison, C., Vergnolle, N., 2021. Gastrointestinal
Landini, P., Antoniani, D., Burgess, J.G., Nijland, R., 2010. Molecular mechanisms of biofilms in health and disease. Nat. Rev. Gastroenterol. Hepatol. 1–21.
compounds affecting bacterial biofilm formation and dispersal. Appl. Microbiol. Mukherjee, S., Bassler, B.L., 2019. Bacterial quorum sensing in complex and dynamically
Biotechnol. 86, 813–823. changing environments. Nat. Rev. Microbiol. 17, 371–382.
Larsen, T., Fiehn, N.E., 2017. Dental biofilm infections–an update. Apmis 125, 376–384. Mulet, X., Maciá, M.D., Mena, A., Juan, C., Pérez, J.L., Oliver, A., 2009. Azithromycin in
Lee, J.-H., Lee, J., 2010. Indole as an intercellular signal in microbial communities. FEMS Pseudomonas aeruginosa biofilms: bactericidal activity and selection of nfxB mutants.
Microbiol. Rev. 34, 426–444. Antimicrob. Agents Chemother. 53, 1552–1560.
Lekshmi, M., Parvathi, A., Kumar, S., Varela, M.F., 2018. Efflux pump-mediated quorum Nguyen, T.H., Park, M.D., Otto, M., 2017. Host response to Staphylococcus epidermidis
sensing: new avenues for modulation of antimicrobial resistance and bacterial colonization and infections. Front. Cell. Infect. Microbiol. 7, 90.
virulence. In: Biotechnological Applications of Quorum Sensing Inhibitors. Springer, Nickerson, K.P., Faherty, C.S., 2018. Bile salt-induced biofilm formation in enteric
pp. 127–142. pathogens: techniques for identification and quantification. JoVE (J. Visual. Exp.)
Li, B., Huang, Q., Cui, A., Liu, X., Hou, B., Zhang, L., Liu, M., Meng, X., Li, S., 2018. e57322.
Overexpression of outer membrane protein X (OmpX) compensates for the effect of Nickerson, K.P., Chanin, R.B., Sistrunk, J.R., Rasko, D.A., Fink, P.J., Barry, E.M.,
TolC inactivation on biofilm formation and curli production in extraintestinal Nataro, J.P., Faherty, C.S., 2017. Analysis of Shigella flexneri resistance, biofilm
pathogenic Escherichia coli (ExPEC). Front. Cell. Infect. Microbiol. 8, 208. formation, and transcriptional profile in response to bile salts. Infect. Immun. 85
Liu, J.Y., Miller, P.F., Gosink, M., Olson, E.R., 1999a. The identification of a new family e01067-01016.
of sugar efflux pumps in Escherichia coli. Mol. Microbiol. 31, 1845–1851. Nikaido, H., Zgurskaya, H.I., 2001. AcrAB and related multidrug efflux pumps of
Liu, J.Y., Miller, P.F., Willard, J., Olson, E.R., 1999b. Functional and biochemical Escherichia coli. J. Mol. Microbiol. Biotechnol. 3, 215–218.
characterization of Escherichia coli sugar efflux transporters. J. Biol. Chem. 274, Nikaido, E., Shirosaka, I., Yamaguchi, A., Nishino, K., 2011. Regulation of the AcrAB
22977–22984. multidrug efflux pump in Salmonella enterica serovar Typhimurium in response to
Liu, Y., Yang, L., Molin, S., 2010. Synergistic activities of an efflux pump inhibitor and indole and paraquat. Microbiology 157, 648–655.
iron chelators against Pseudomonas aeruginosa growth and biofilm formation. Nishino, K., Nikaido, E., Yamaguchi, A., 2009. Regulation and physiological function of
Antimicrob. Agents Chemother. 54, 3960–3963. multidrug efflux pumps in Escherichia coli and Salmonella. Biochim. Biophys. Acta
Liu, C., Niu, Y., Zhou, X., Zhang, K., Cheng, L., Li, M., Li, Y., Wang, R., Yang, Y., Xu, X., (BBA)-Proteins Proteom. 1794, 834–843.
2013. Hyperosmotic response of Streptococcus mutans: from microscopic physiology Noirot-Gros, M.-F., Forrester, S., Malato, G., Larsen, P.E., Noirot, P., 2019. CRISPR
to transcriptomic profile. BMC Microbiol. 13, 1–9. interference to interrogate genes that control biofilm formation in Pseudomonas
Liu, C., Sun, D., Zhu, J., Liu, J., Liu, W., 2020. The regulation of bacterial biofilm fluorescens. Sci. Rep. 9, 1–14.
formation by cAMP-CRP: a mini-review. Front. Microbiol. 11. Oglesby, L.L., Jain, S., Ohman, D.E., 2008. Membrane topology and roles of Pseudomonas
Liu, L., Wang, W., Wu, S., Gao, H., 2022. Recent advances in the siderophore biology of aeruginosa Alg8 and Alg44 in alginate polymerization. Microbiology 154, 1605.
Shewanella. Front. Microbiol. 13, 823758. Olivares, J., Alvarez-Ortega, C., Linares, J.F., Rojo, F., Köhler, T., Martínez, J.L., 2012.
Ma, D., Cook, D.N., Alberti, M., Pon, N.G., Nikaido, H., Hearst, J.E., 1995. Genes acrA Overproduction of the multidrug efflux pump MexEF-OprN does not impair
and acrB encode a stress-induced efflux system of Escherichia coli. Mol. Microbiol. 16, Pseudomonas aeruginosa fitness in competition tests, but produces specific changes in
45–55. bacterial regulatory networks. Environ. Microbiol. 14, 1968–1981.
Macia, M., Borrell, N., Segura, M., Gomez, C., Perez, J., Oliver, A., 2006. Efficacy and Olsen, I., 2015. Biofilm-specific antibiotic tolerance and resistance. Eur. J. Clin.
potential for resistance selection of antipseudomonal treatments in a mouse model of Microbiol. Infect. Dis. 34, 877–886.
lung infection by hypermutable Pseudomonas aeruginosa. Antimicrob. Agents O’Toole, G., Kaplan, H.B., Kolter, R., 2000. Biofilm formation as microbial development.
Chemother. 50, 975–983. Annu. Rev. Microbiol. 54, 49–79.
Mah, T.-F.C., O’Toole, G.A., 2001. Mechanisms of biofilm resistance to antimicrobial Pacios, O., Blasco, L., Bleriot, I., Fernandez-Garcia, L., Ambroa, A., López, M., Bou, G.,
agents. Trends Microbiol. 9, 34–39. Cantón, R., Garcia-Contreras, R., Wood, T.K., 2020. ppGpp and its role in bacterial
Mah, T.-F., Pitts, B., Pellock, B., Walker, G.C., Stewart, P.S., O’Toole, G.A., 2003. persistence: new challenges. Antimicrob. Agents Chemother. 64.
A genetic basis for Pseudomonas aeruginosa biofilm antibiotic resistance. Nature 426, Palmer, J., Flint, S., Brooks, J., 2007. Bacterial cell attachment, the beginning of a
306–310. biofilm. J. Ind. Microbiol. Biotechnol. 34, 577–588.
Mahdhi, A., Leban, N., Chakroun, I., Bayar, S., Mahdouani, K., Majdoub, H., Kouidhi, B., Pandey, R.P., Mukherjee, R., Chang, C.-M., 2022. Emerging concern with imminent
2018. Use of extracellular polysaccharides, secreted by Lactobacillus plantarum and therapeutic strategies for treating resistance in biofilm. Antibiotics 11, 476.
Bacillus spp., as reducing indole production agents to control biofilm formation and Papenfort, K., Bassler, B.L., 2016. Quorum sensing signal-response systems in Gram-
efflux pumps inhibitor in Escherichia coli. Microb. Pathog. 125, 448–453. negative bacteria. Nat. Rev. Microbiol. 14, 576–588.
Maira-Litrán, T., Allison, D., Gilbert, P., 2000. Expression of the multiple antibiotic Partridge, J.D., Harshey, R.M., 2013. Swarming: flexible roaming plans. J. Bacteriol. 195,
resistance operon (mar) during growth of Escherichia coli as a biofilm. J. Appl. 909–918.
Microbiol. 88, 243–247. Pasqua, M., Grossi, M., Zennaro, A., Fanelli, G., Micheli, G., Barras, F., Colonna, B.,
Maisonneuve, E., Gerdes, K., 2014. Molecular mechanisms underlying bacterial Prosseda, G., 2019. The varied role of efflux pumps of the MFS family in the
persisters. Cell 157, 539–548. interplay of bacteria with animal and plant cells. Microorganisms 7, 285.
Marquez, B., 2005. Bacterial efflux systems and efflux pumps inhibitors. Biochimie 87, Passos da Silva, D., Schofield, M.C., Parsek, M.R., Tseng, B.S., 2017. An update on the
1137–1147. sociomicrobiology of quorum sensing in gram-negative biofilm development.
Masadeh, M.M., Gharaibeh, S.F., Alzoubi, K.H., Al-Azzam, S.I., Obeidat, W.M., 2013. Pathogens 6, 51.
Antimicrobial activity of common mouthwash solutions on multidrug-resistance Pearson, J.P., Van Delden, C., Iglewski, B.H., 1999. Active efflux and diffusion are
bacterial biofilms. J. Clin. Med. Res. 5, 389. involved in transport of Pseudomonas aeruginosa cell-to-cell signals. J. Bacteriol. 181,
1203–1210.

17
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Penesyan, A., Paulsen, I.T., Gillings, M.R., Kjelleberg, S., Manefield, M.J., 2020. Sankaran, J., Tan, N.J., But, K.P., Cohen, Y., Rice, S.A., Wohland, T., 2019. Single
Secondary effects of antibiotics on microbial biofilms. Front. Microbiol. 11, 2109. microcolony diffusion analysis in Pseudomonas aeruginosa biofilms. npj Biofilms
Perez, L.R.R., 2015. Acinetobacter baumannii displays inverse relationship between Microbiom. 5, 1–10.
meropenem resistance and biofilm production. J. Chemother. 27, 13–16. Schaible, B., Taylor, C.T., Schaffer, K., 2012. Hypoxia increases antibiotic resistance in
Perrin, C., Briandet, R., Jubelin, G., Lejeune, P., Mandrand-Berthelot, M.-A., Pseudomonas aeruginosa through altering the composition of multidrug efflux pumps.
Rodrigue, A., Dorel, C., 2009. Nickel promotes biofilm formation by Escherichia coli Antimicrob. Agents Chemother. 56, 2114–2118.
K-12 strains that produce curli. Appl. Environ. Microbiol. 75, 1723–1733. Schellhorn, H.E., 2020. Function, evolution, and composition of the RpoS regulon in
Piddock, L.J., 2006. Multidrug-resistance efflux pumps? Not just for resistance. Nat. Rev. Escherichia coli. Front. Microbiol. 11, 2282.
Microbiol. 4, 629–636. Schulze, A., Mitterer, F., Pombo, J.P., Schild, S., 2021. Biofilms by bacterial human
Piñero-Fernandez, S., Chimerel, C., Keyser, U., Summers, D., 2011. Indole transport pathogens: clinical relevance-development, composition and regulation-
across Escherichia coli membranes. J. Bacteriol. 193, 1793–1798. therapeutical strategies. Microb. Cell 8, 28.
Poole, K., Krebes, K., McNally, C., Neshat, S., 1993. Multiple antibiotic resistance in Sheikh, J., Hicks, S., Dall’Agnol, M., Phillips, A.D., Nataro, J.P., 2001. Roles for Fis and
Pseudomonas aeruginosa: evidence for involvement of an efflux operon. J. Bacteriol. YafK in biofilm formation by enteroaggregative Escherichia coli. Mol. Microbiol. 41,
175, 7363–7372. 983–997.
Potera, C., 1999. Forging a Link Between Biofilms and Disease. American Association for Song, F., Koo, H., Ren, D., 2015. Effects of material properties on bacterial adhesion and
the Advancement of Science. biofilm formation. J. Dent. Res. 94, 1027–1034.
Prigent-Combaret, C., Vidal, O., Dorel, C., Lejeune, P., 1999. Abiotic surface sensing and Soto, S.M., 2013. Role of efflux pumps in the antibiotic resistance of bacteria embedded
biofilm-dependent regulation of gene expression in Escherichia coli. J. Bacteriol. 181, in a biofilm. Virulence 4, 223–229.
5993–6002. Stacy, A., McNally, L., Darch, S.E., Brown, S.P., Whiteley, M., 2016. The biogeography of
Prigent-Combaret, C., Prensier, G., Le Thi, T.T., Vidal, O., Lejeune, P., Dorel, C., 2000. polymicrobial infection. Nat. Rev. Microbiol. 14, 93–105.
Developmental pathway for biofilm formation in curli-producing Escherichia coli Stewart, P.S., Franklin, M.J., 2008. Physiological heterogeneity in biofilms. Nat. Rev.
strains: role of flagella, curli and colanic acid. Environ. Microbiol. 2, 450–464. Microbiol. 6, 199–210.
Pumbwe, L., Skilbeck, C.A., Wexler, H.M., 2008. Presence of quorum-sensing systems Subhadra, B., Surendran, S., Lim, B.R., Yim, J.S., Kim, D.H., Woo, K., Kim, H.-J., Oh, M.
associated with multidrug resistance and biofilm formation in Bacteroides fragilis. H., Choi, C.H., 2020. Regulation of the AcrAB efflux system by the quorum-sensing
Microb. Ecol. 56, 412–419. regulator AnoR in Acinetobacter nosocomialis. J. Microbiol. 58, 507–518.
Rabin, N., Zheng, Y., Opoku-Temeng, C., Du, Y., Bonsu, E., Sintim, H.O., 2015. Biofilm Sun, J., Deng, Z., Yan, A., 2014. Bacterial multidrug efflux pumps: mechanisms,
formation mechanisms and targets for developing antibiofilm agents. Future Med. physiology and pharmacological exploitations. Biochem. Biophys. Res. Commun.
Chem. 7, 493–512. 453, 254–267.
Rahmati, S., Yang, S., Davidson, A.L., Zechiedrich, E.L., 2002. Control of the AcrAB Tal, N., Schuldiner, S., 2009. A coordinated network of transporters with overlapping
multidrug efflux pump by quorum-sensing regulator SdiA. Mol. Microbiol. 43, specificities provides a robust survival strategy. Proc. Natl. Acad. Sci. 106,
677–685. 9051–9056.
Ramage, G., Rajendran, R., Sherry, L., Williams, C., 2012. Fungal biofilm resistance. Int. Tang, M., Wei, X., Wan, X., Ding, Z., Ding, Y., Liu, J., 2020. The role and relationship
J. Microbiol. 2012. with efflux pump of biofilm formation in Klebsiella pneumoniae. Microb. Pathog. 147,
Ramsey, M.M., Whiteley, M., 2004. Pseudomonas aeruginosa attachment and biofilm 104244.
development in dynamic environments. Mol. Microbiol. 53, 1075–1087. Tao, L., Tanzer, J., 2002. Novel sucrose-dependent adhesion co-factors in Streptococcus
Rasamiravaka, T., El Jaziri, M., 2016. Quorum-sensing mechanisms and bacterial mutans. J. Dent. Res. 81, 505–510.
response to antibiotics in P. aeruginosa. Curr. Microbiol. 73, 747–753. Thanassi, D.G., Cheng, L.W., Nikaido, H., 1997. Active efflux of bile salts by Escherichia
Reeder, T., Schleif, R., 1991. Mapping, sequence, and apparent lack of function of araJ, a coli. J. Bacteriol. 179, 2512–2518.
gene of the Escherichia coli arabinose regulon. J. Bacteriol. 173, 7765–7771. Thongbhubate, K., Nakafuji, Y., Matsuoka, R., Kakegawa, S., Suzuki, H., 2021. Effect of
Remminghorst, U., Rehm, B.H., 2006. Alg44, a unique protein required for alginate spermidine on biofilm formation in Escherichia coli K-12. J. Bacteriol. 203 e00652-
biosynthesis in Pseudomonas aeruginosa. FEBS Lett. 580, 3883–3888. 00620.
Renner, L.D., Weibel, D.B., 2011. Physicochemical regulation of biofilm formation. MRS Truong-Bolduc, Q., Wang, Y., Hooper, D., 2021. Staphylococcus aureus Tet38 efflux pump
Bull. 36, 347–355. structural modeling and roles of essential residues in drug efflux and host cell
Resch, A., Rosenstein, R., Nerz, C., Götz, F., 2005. Differential gene expression profiling internalization. Infect. Immun. 89, e00811–e00820.
of Staphylococcus aureus cultivated under biofilm and planktonic conditions. Appl. Tseng, T.-T., Gratwick, K.S., Kollman, J., Park, D., Nies, D.H., Goffeau, A., Saier Jr., M.H.,
Environ. Microbiol. 71, 2663–2676. 1999. The RND permease superfamily: an ancient, ubiquitous and diverse family that
Ricci, V., Busby, S.J., Piddock, L.J., 2012. Regulation of RamA by RamR in Salmonella includes human disease and development proteins. J. Mol. Microbiol. Biotechnol. 1,
enterica serovar Typhimurium: isolation of a RamR superrepressor. Antimicrob. 107–125.
Agents Chemother. 56, 6037–6040. Türkel, İ., Yıldırım, T., Yazgan, B., Bilgin, M., Başbulut, E., 2018. Relationship between
Richmond, G.E., Evans, L.P., Anderson, M.J., Wand, M.E., Bonney, L.C., Ivens, A., antibiotic resistance, efflux pumps, and biofilm formation in extended-spectrum
Chua, K.L., Webber, M.A., Sutton, J.M., Peterson, M.L., 2016. The Acinetobacter β-lactamase producing Klebsiella pneumoniae. J. Chemother. 30, 354–363.
baumannii two-component system AdeRS regulates genes required for multidrug Turlin, E., Heuck, G., Simões Brandão, M.I., Szili, N., Mellin, J., Lange, N.,
efflux, biofilm formation, and virulence in a strain-specific manner. MBio 7 e00430- Wandersman, C., 2014. Protoporphyrin (PPIX) efflux by the MacAB-TolC pump in
00416. Escherichia coli. Microbiol. Open 3, 849–859.
Riley, M., Abe, T., Arnaud, M.B., Berlyn, M.K., Blattner, F.R., Chaudhuri, R.R., Glasner, J. Tursi, S.A., Puligedda, R.D., Szabo, P., Nicastro, L.K., Miller, A.L., Qiu, C., Gallucci, S.,
D., Horiuchi, T., Keseler, I.M., Kosuge, T., 2006. Escherichia coli K-12: a cooperatively Relkin, N.R., Buttaro, B.A., Dessain, S.K., 2020. Salmonella typhimurium biofilm
developed annotation snapshot—2005. Nucleic Acids Res. 34, 1–9. disruption by a human antibody that binds a pan-amyloid epitope on curli. Nat.
Rodrigue, A., Effantin, G., Mandrand-Berthelot, M.-A., 2005. Identification of rcnA Commun. 11, 1–13.
(yohM), a nickel and cobalt resistance gene in Escherichia coli. J. Bacteriol. 187, Urdaneta, V., Casadesús, J., 2018. Adaptation of Salmonella enterica to bile: essential role
2912–2916. of AcrAB-mediated efflux. Environ. Microbiol. 20, 1405–1418.
Ronneau, S., Helaine, S., 2019. Clarifying the link between toxin–antitoxin modules and Vaillancourt, M., Limsuwannarot, S.P., Bresee, C., Poopalarajah, R., Jorth, P., 2021.
bacterial persistence. J. Mol. Biol. 431, 3462–3471. Pseudomonas aeruginosa mexR and mexEF antibiotic efflux pump variants exhibit
Rumbo-Feal, S., Gomez, M.J., Gayoso, C., Álvarez-Fraga, L., Cabral, M.P., Aransay, A.M., increased virulence. Antibiotics 10, 1164.
Rodríguez-Ezpeleta, N., Fullaondo, A., Valle, J., Tomás, M., 2013. Whole Van Acker, H., Coenye, T., 2016. The role of efflux and physiological adaptation in
transcriptome analysis of Acinetobacter baumannii assessed by RNA-sequencing biofilm tolerance and resistance. J. Biol. Chem. 291, 12565–12572.
reveals different mRNA expression profiles in biofilm compared to planktonic cells. Van Bambeke, F., Glupczynski, Y., Plesiat, P., Pechere, J., Tulkens, P.M., 2003a.
PLoS One 8, e72968. Antibiotic efflux pumps in prokaryotic cells: occurrence, impact on resistance and
Rumbo-Feal, S., Pérez, A., Ramelot, T.A., Álvarez-Fraga, L., Vallejo, J.A., Beceiro, A., strategies for the future of antimicrobial therapy. J. Antimicrob. Chemother. 51,
Ohneck, E.J., Arivett, B.A., Merino, M., Fiester, S.E., 2017. Contribution of the 1055–1065.
A. baumannii A1S_0114 gene to the interaction with eukaryotic cells and virulence. Van Bambeke, F., Michot, J.-M., Tulkens, P.M., 2003b. Antibiotic efflux pumps in
Front. Cell. Infect. Microbiol. 7, 108. eukaryotic cells: occurrence and impact on antibiotic cellular pharmacokinetics,
Rzhepishevska, O., Hakobyan, S., Ruhal, R., Gautrot, J., Barbero, D., Ramstedt, M., 2013. pharmacodynamics and toxicodynamics. J. Antimicrob. Chemother. 51, 1067–1077.
The surface charge of anti-bacterial coatings alters motility and biofilm architecture. Van der Waal, S., Van der Sluis, L., Özok, A., Exterkate, R., Van Marle, J., Wesselink, P.,
Biomater. Sci. 1, 589–602. de Soet, J., 2011. The effects of hyperosmosis or high pH on a dual-species biofilm of
Sahu, P.K., Iyer, P.S., Gaikwad, M.B., Talreja, S.C., Pardesi, K.R., Chopade, B.A., 2012. An Enterococcus faecalis and Pseudomonas aeruginosa: an in vitro study. Int. Endod. J. 44,
MFS transporter-like ORF from MDR Acinetobacter baumannii AIIMS 7 is associated 1110–1117.
with adherence and biofilm formation on biotic/abiotic surface. Int. J. Microbiol. Van Dyk, T.K., Templeton, L.J., Cantera, K.A., Sharpe, P.L., Sariaslani, F.S., 2004.
2012. Characterization of the Escherichia coli AaeAB efflux pump: a metabolic relief valve?
Saier Jr., M.H., 2000. A functional-phylogenetic classification system for transmembrane J. Bacteriol. 186, 7196–7204.
solute transporters. Microbiol. Mol. Biol. Rev. 64, 354–411. Vasicek, E.M., O’Neal, L., Parsek, M.R., Fitch, J., White, P., Gunn, J.S., 2021. L-arabinose
Sakhtah, H., Koyama, L., Zhang, Y., Morales, D.K., Fields, B.L., Price-Whelan, A., transport and metabolism in Salmonella influences biofilm formation. Front. Cell.
Hogan, D.A., Shepard, K., Dietrich, L.E., 2016. The Pseudomonas aeruginosa efflux Infect. Microbiol. 609.
pump MexGHI-OpmD transports a natural phenazine that controls gene expression Ventola, C.L., 2015. The antibiotic resistance crisis: part 1: causes and threats.
and biofilm development. Proc. Natl. Acad. Sci. 113, E3538–E3547. PharmacyTherap. 40, 277.
Vestby, L.K., Grønseth, T., Simm, R., Nesse, L.L., 2020. Bacterial biofilm and its role in
the pathogenesis of disease. Antibiotics 9, 59.

18
M.N. Hajiagha and H.S. Kafil Infection, Genetics and Evolution 112 (2023) 105459

Vivas, R., Barbosa, A.A.T., Dolabela, S.S., Jain, S., 2019. Multidrug-resistant bacteria and Yasufuku, T., Shigemura, K., Shirakawa, T., Matsumoto, M., Nakano, Y., Tanaka, K.,
alternative methods to control them: an overview. Microb. Drug Resist. 25, 890–908. Arakawa, S., Kinoshita, S., Kawabata, M., Fujisawa, M., 2011. Correlation of
Wakimoto, N., Nishi, J., Sheikh, J., Nataro, J.P., Sarantuya, J., Iwashita, M., Manago, K., overexpression of efflux pump genes with antibiotic resistance in Escherichia coli
Tokuda, K., Yoshinaga, M., Kawano, Y., 2004. Quantitative biofilm assay using a strains clinically isolated from urinary tract infection patients. J. Clin. Microbiol. 49,
microtiter plate to screen for enteroaggregative Escherichia coli. Am. J. Trop. Med. 189–194.
Hyg. 71, 687–690. Yoon, E.-J., Nait Chabane, Y., Goussard, S., Snesrud, E., Courvalin, P., Dé, E., Grillot-
Wang, Y., Wilks, J.C., Danhorn, T., Ramos, I., Croal, L., Newman, D.K., 2011. Phenazine- Courvalin, C., 2015. Contribution of resistance-nodulation-cell division efflux
1-carboxylic acid promotes bacterial biofilm development via ferrous iron systems to antibiotic resistance and biofilm formation in Acinetobacter baumannii.
acquisition. J. Bacteriol. 193, 3606–3617. MBio 6, e00309–e00315.
Wang, J., Seebacher, N., Shi, H., Kan, Q., Duan, Z., 2017. Novel strategies to prevent the Yu, L., Li, W., Li, Q., Chen, X., Ni, J., Shang, F., Xue, T., 2020. Role of LsrR in the
development of multidrug resistance (MDR) in cancer. Oncotarget 8, 84559. regulation of antibiotic sensitivity in avian pathogenic Escherichia coli. Poult. Sci. 99,
Wang, J., Jiao, H., Meng, J., Qiao, M., Du, H., He, M., Ming, K., Liu, J., Wang, D., Wu, Y., 3675–3687.
2019a. Baicalin inhibits biofilm formation and the quorum-sensing system by Yuan, H., Wang, Y., Lai, Z., Zhang, X., Jiang, Z., Zhang, X., 2021. Analyzing microalgal
regulating the MsrA drug efflux pump in Staphylococcus saprophyticus. Front. biofilm structures formed under different light conditions by evaluating cell–cell
Microbiol. 10, 2800. interactions. J. Colloid Interface Sci. 583, 563–570.
Wang, M., Wei, H., Zhao, Y., Shang, L., Di, L., Lyu, C., Liu, J., 2019b. Analysis of Yun, B.-Y., Xu, Y., Piao, S., Kim, N., Yoon, J.-H., Cho, H.-S., Lee, K., Ha, N.-C., 2010.
multidrug-resistant bacteria in 3223 patients with hospital-acquired infections (HAI) Periplasmic domain of CusA in an Escherichia coli Cu+/Ag+ transporter has metal
from a tertiary general hospital in China. Bosn. J. Basic Med. Sci. 19, 86. binding sites. J. Microbiol. 48, 829–835.
Wang, Y., Liu, B., Li, J., Gong, S., Dong, X., Mao, C., Yi, L., 2019c. LuxS/AI-2 system is Zarkan, A., Liu, J., Matuszewska, M., Gaimster, H., Summers, D.K., 2020. Local and
involved in fluoroquinolones susceptibility in Streptococcus suis through universal action: the paradoxes of indole signalling in bacteria. Trends Microbiol. 28,
overexpression of efflux pump SatAB. Vet. Microbiol. 233, 154–158. 566–577.
Webber, M., Piddock, L., 2003. The importance of efflux pumps in bacterial antibiotic Zgur Bertok, D., Podlesek, Z., 2020. The DNA damage inducible SOS response is aa key
resistance. J. Antimicrob. Chemother. 51, 9–11. player in generation of bacterial persister cells and population wide tolerance. Front.
Webber, M.A., Bailey, A.M., Blair, J.M., Morgan, E., Stevens, M.P., Hinton, J.C., Ivens, A., Microbiol. 11, 1785.
Wain, J., Piddock, L.J., 2009. The global consequence of disruption of the AcrAB- Zhang, L., Mah, T.-F., 2008. Involvement of a novel efflux system in biofilm-specific
TolC efflux pump in Salmonella enterica includes reduced expression of SPI-1 and resistance to antibiotics. J. Bacteriol. 190, 4447–4452.
other attributes required to infect the host. J. Bacteriol. 191, 4276–4285. Zhang, Y., Xiao, M., Horiyama, T., Zhang, Y., Li, X., Nishino, K., Yan, A., 2011. The
Weston, N., Sharma, P., Ricci, V., Piddock, L.J.V., 2018. Regulation of the AcrAB-TolC multidrug efflux pump MdtEF protects against nitrosative damage during the
efflux pump in Enterobacteriaceae. Res. Microbiol. 169, 425–431. anaerobic respiration in Escherichia coli. J. Biol. Chem. 286, 26576–26584.
Whitney, J.C., Whitfield, G.B., Marmont, L.S., Yip, P., Neculai, A.M., Lobsanov, Y.D., Zheng, B., Ma, X., Wang, N., Ding, T., Guo, L., Zhang, X., Yang, Y., Li, C., Huo, Y.-X.,
Robinson, H., Ohman, D.E., Howell, P.L., 2015. Dimeric c-di-GMP is required for 2018. Utilization of rare codon-rich markers for screening amino acid overproducers.
post-translational regulation of alginate production in Pseudomonas aeruginosa. Nat. Commun. 9, 1–11.
J. Biol. Chem. 290, 12451–12462. Zhu, X., Long, F., Chen, Y., Knøchel, S., She, Q., Shi, X., 2008. A putative ABC transporter
Wolcott, R., Costerton, J., Raoult, D., Cutler, S., 2013. The polymicrobial nature of is involved in negative regulation of biofilm formation by Listeria monocytogenes.
biofilm infection. Clin. Microbiol. Infect. 19, 107–112. Appl. Environ. Microbiol. 74, 7675–7683.
Wolloscheck, D., Krishnamoorthy, G., Nguyen, J., Zgurskaya, H.I., 2018. Kinetic control Zhu, X., Liu, W., Lametsch, R., Aarestrup, F., Shi, C., She, Q., Shi, X., Knøchel, S., 2011.
of quorum sensing in Pseudomonas aeruginosa by multidrug efflux pumps. ACS Infect. Phenotypic, proteomic, and genomic characterization of a putative ABC-transporter
Dis. 4, 185–195. permease involved in Listeria monocytogenes biofilm formation. Foodborne Pathog.
Wu, L., Luo, Y., 2021. Bacterial quorum-sensing systems and their role in intestinal Dis. 8, 495–501.
bacteria-host crosstalk. Front. Microbiol. 101. Zhu, Z., Surujon, D., Ortiz-Marquez, J.C., Huo, W., Isberg, R.R., Bento, J., van
Yan, J., Bassler, B.L., 2019. Surviving as a community: antibiotic tolerance and Opijnen, T., 2020. Entropy of a bacterial stress response is a generalizable predictor
persistence in bacterial biofilms. Cell Host Microbe 26, 15–21. for fitness and antibiotic sensitivity. Nat. Commun. 11, 1–15.
Yanofsky, C., Horn, V., Gollnick, P., 1991. Physiological studies of tryptophan transport Zimmermann, S., Klinger-Strobel, M., Bohnert, J.A., Wendler, S., Rödel, J., Pletz, M.W.,
and tryptophanase operon induction in Escherichia coli. J. Bacteriol. 173, 6009–6017. Löffler, B., Tuchscherr, L., 2019. Clinically approved drugs inhibit the Staphylococcus
Yao, Y., Sturdevant, D.E., Otto, M., 2005. Genomewide analysis of gene expression in aureus multidrug NorA efflux pump and reduce biofilm formation. Front. Microbiol.
Staphylococcus epidermidis biofilms: insights into the pathophysiology of 2762.
S. epidermidis biofilms and the role of phenol-soluble modulins in formation of
biofilms. J Infect Dis 191, 289–298.

19

You might also like