You are on page 1of 18

Ecotoxicology and Environmental Safety 264 (2023) 115389

Contents lists available at ScienceDirect

Ecotoxicology and Environmental Safety


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

Review

Bacterial biofilm inhibitors: An overview


Vipin Chandra Kalia , Sanjay K.S. Patel , Jung-Kul Lee *
Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea

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

Edited by Professor Bing Yan Bacteria that cause infectious diseases adopt biofilms as one of their most prevalent lifestyles. Biofilms enable
bacteria to tolerate environmental stress and evade antibacterial agents. This bacterial defense mechanism has
Keywords: rendered the use of antibiotics ineffective for the treatment of infectious diseases. However, many highly drug-
Antibiotics resistant microbes have rapidly emerged owing to such treatments. Different signaling mechanisms regulate
Antipathogens
bacterial biofilm formation, including cyclic dinucleotide (c-di-GMP), small non-coding RNAs, and quorum
Biofilm
sensing (QS). A cell density-dependent phenomenon, QS is associated with c-di-GMP (a global messenger), which
Biomolecules
Inhibitors regulates gene expression related to adhesion, extracellular matrix production, the transition from the planktonic
Peptides to biofilm stage, stability, pathogenicity, virulence, and acquisition of nutrients. The article aims to provide
Polysaccharides information on inhibiting biofilm formation and disintegrating mature/preformed biofilms. This treatment en­
Quorum sensing ables antimicrobials to target the free-living/exposed bacterial cells at lower concentrations than those needed to
Healthcare treat bacteria within the biofilm. Therefore, a complementary action of antibiofilm and antimicrobial agents can
be a robust strategic approach to dealing with infectious diseases. Taken together, these molecules have broad
implications for human health.

1. Introduction et al., 2007; Lallukka et al., 2017; Vestby et al., 2020). Despite the di­
versity of infectious diseases, there was a significant reduction in mor­
Among infectious diseases, the contribution of biofilm-forming tality and morbidity between 2000 and 2020. This change is primarily
bacteria has gained attention. Biofilm-forming bacteria cause around due to improved hygiene, higher accessibility to health care, and med­
65–80% of infectious diseases (Lebeaux et al., 2013). Such ical advances, such as vaccines and antimicrobial therapies (Baik et al.,
biofilm-related infectious diseases are typically chronic (Bjarnsholt, 2022). Nevertheless, the social and economic burden remains substan­
2013). Bacteria within the biofilm are challenging to treat with antibi­ tially higher in lower-middle-income countries due to poor sanitation
otics, leading to prolonged illness and contributing significantly to high and limited access to healthcare (Lallukka et al., 2017; Bloom and
morbidity and mortality rates (WHO, 2014). The global economic Cadratte, 2019; Baker et al., 2022). Additionally, the emergence of new
burden due to the emergence of antibiotic-resistant bacteria is estimated infectious diseases and the reemergence of old ones, such as COVID-19,
to reach USD 100 trillion by 2050, and the associated death toll will highlight the ongoing need for vigilance and preparedness in the face of
increase by up to 10 million per year (Gandra et al., 2014; WHO, 2014). infectious diseases (Khabbaz et al., 2015; Morens and Fauci, 2020). It is
A comprehensive global estimate reveals that infectious bacteria cause essential to evaluate the information generated by various studies to
7.7 million deaths, 13.6% of all global deaths. This has drawn the inhibit the different processes leading to biofilm formation and disin­
attention of health authorities to develop strategies to mitigate this tegration the mature biofilms (Kannappan et al., 2017a; Koo et al., 2017;
socio-economic burden. The following five types of biofilm-forming Srinivasan et al., 2018).
bacteria have been reported to be responsible for 54.9% of these Pathogenic bacteria causing various infectious diseases switch be­
deaths: Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa, tween a planktonic state and biofilm form to tolerate environmental
Staphylococcus aureus, and Streptococcus pneumoniae (Donlan, 2001; Tien stress, especially those caused by antimicrobial agents (Roy et al., 2018).

Abbreviations: AHL, acyl homoserine lactone; AI, autoinducer; BP, bacteriophage; c-di-GMP, cyclic dimeric guanosine monophosphate; DNase, deoxyribonuclease;
ECM, extracellular matrix; eDNA, extracellular DNA; EPS, Exopolysaccharide; MRSA, methicillin-resistant Staphylococcus aureus; NP, nanoparticle; QS, quorum
sensing; QSI, QS inhibitor; QSS, QS system; sRNA, small non-coding RNA.
* Corresponding author.
E-mail address: jkrhee@konkuk.ac.kr (J.-K. Lee).

https://doi.org/10.1016/j.ecoenv.2023.115389
Received 18 May 2023; Received in revised form 5 August 2023; Accepted 17 August 2023
Available online 25 August 2023
0147-6513/© 2023 The Authors. Published by Elsevier Inc. This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Biofilm-forming bacteria can often evade the body’s immune system, by aggregating in an aqueous environment (Melaugh et al., 2016). These
making fighting these infections even more challenging. Moreover, aggregated bacteria adhere to a solid surface and produce extracellular
biofilms form on medical devices and implants, increasing the risk of polymeric substrates (EPS), which include polysaccharides, proteins,
infection during surgeries or other medical procedures (Kannappan lipids, and DNA, providing solid support for maintaining the integrity
et al., 2017b, 2019a; b; Kannappan et al., 2020). Preventive measures, and survival of the bacteria termed “matrixome” (Claessen et al., 2014;
such as regular cleaning of medical devices and implants, improved Coughlan et al., 2016; Kannappan et al., 2019a; Karygianni et al.,
hygiene practices in healthcare settings, and the development of new 2020a). Their motility, cell surface proteins, adhesins, protein autolysin,
antimicrobial agents, are crucial in preventing the spread of shear, and electrostatic interactions influence the bacterial adherence
biofilm-forming bacteria and reducing the incidence of infectious dis­ process. In the subsequent stage, bacterial cell proliferation, aggrega­
eases caused by these bacteria. In addition to the pathogens listed above, tion, and differentiation form a mature biofilm (Stoodley et al., 2002).
the few most frequently encountered bacterial biofilms in healthcare Bacterial cells from the mature biofilm can undergo detachment and
centers are Acinetobacter spp., Enterococcus faecalis, and Proteus mirabilis. enter another cycle of biofilm formation, resulting in the spread of in­
The most challenging part of these persistently spreading infections is fections to other parts of the medical devices or organs (Stoodley et al.,
the enhanced tolerance of bacterial pathogens to biocides. This 2002; Fey and Olson, 2010). Exopolysaccharides (EPSs) suppress host
enhanced drug resistance is due to many factors, the most important immune responses and confer antibiotic resistance to pathogens
being the delayed and incomplete penetration of antimicrobials through (Angelin and Kavitha, 2020; Li et al., 2022). Extracellular proteins that
the EPM, the transmission of plasmids conferring drug resistance, and enhance biofilm formation and stability vary considerably among or­
the emergence of persister cells (Borriello et al., 2004; Lewis, 2007; ganisms. eDNA coordinates bacterial twitching motility, metal chela­
Stewart et al., 2009). Biofilm protects microorganisms, making it diffi­ tion, quenching of positively charged antibiotics, and acquisition of
cult to eradicate them compared to their free-living/planktonic coun­ antibiotic resistance genes, contributing to antibiotic resistance, bacte­
terparts. Bacteria within biofilms can tolerate up to 1000-fold higher rial aggregation, and stabilization. The interaction among eDNA, pro­
antibiotic concentrations in extreme cases than their free-living coun­ teins, EPS, and other molecules enables biofilm formation and structural
terparts (Kalia et al., 2019). Antimicrobial peptides, polyamines, phy­ integrity (Panlilio and Rice, 2021). The upregulation of genes encoding
tochemicals, and nanoparticles (NPs) possess specific features that ECM metabolism and cell motility precedes bacterial dispersal. Genes
enable them to inhibit biofilm formation. At the other end of the spec­ responsible for synthesizing EPS and fimbriae are downregulated
trum are enzymes, bacteriophages (BPs), and NPs, which target mature (Armbruster and Parsek, 2018). In the dispersal stage, enzymatic
biofilms (Kumar et al., 2021). This review deals with the various ap­ degradation of EPS disassembles the biofilm matrix, releasing microbes
proaches employed to control biofilms and their potential to treat and expanding infected regions (Fleming and Rumbaugh, 2017).
biofilm-based infectious diseases. Briefly, the antibiofilm agents target Genetic regulation of biofilm formation operates primarily through
the multiple mechanisms and components involved in the biofilm for­ three primary mechanisms, which are: (i) QS, (ii) cyclic dinucleotide,
mation, which include: (i) inhibiting initial adhesion, (ii) removal of and (iii) small non-coding RNAs (sRNAs). A perusal of the various
mature biofilms and degradation of various components of the biofilm, studies which have been targeting these three regulatory systems to
(iii) interference with bacterial communication or quorum sensing (QS), manipulate biofilm formation reveals that only a few of them have re­
and (iv) inhibiting non-QS regulatory processes.. ported the potential of cyclic dinucleotides (López-Baena et al., 2019),
and (ii) small non-coding RNAs (sRNAs) as drug targets (Taylor et al.,
2. Biofilms 2017; Manfiolli et al., 2018). In contrast, most studies have revealed QS
systems (QSS) as the significant drug targets (Subhadra et al., 2018;
Biofilm formation is a multistep process, which involves (i) bacterial Kalia et al., 2019).
attachment, (ii) maturation, and (iii) bacterial release (Flemming et al.,
2016). Free-living or planktonic bacterial cells initiate biofilm formation

Fig. 1. Diagrammatic representation of bacterial biofilm formation and dispersal. Antibiofilm agents act against the signaling systems: cyclic dinucleotide, quorum
sensing, and small non-coding RNAs and the gene expressions responsible for adhesion, extra-cellular matrix production, transition from planktonic to biofilm stage,
stability, pathogenicity, virulence, and acquiring nutrients.

2
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

2.1. Cyclic dinucleotide reduced phosphorylation of MpkA and SakA, have been used to
demonstrate their role in inducing these changes (Manfiolli et al., 2018).
Bacterial regulatory signaling molecules, such as cyclic dimeric Transcriptomic studies have also supported the role of sRNA in
guanosine monophosphate (c-di-GMP), confer abilities to respond and promoting gene expression in the ECM and curli fimbriae in
adapt to changing environments (López-Baena et al., 2019). This ubiq­ P. aeruginosa (Gómez-Lozano et al., 2012). Among over 200 sRNAs re­
uitous bacterial messenger regulates different processes. The intracel­ ported in P. aeruginosa genomes, SrbA was proposed to play a role in
lular fluctuation in its concentration contributes to switching from biofilm formation and pathogenicity. srbA deletion mutants had 66%
motile to sessile and biofilm-forming lifestyles (Moscoso et al., 2011; lower biofilm mass, and their infection resulted in a mortality rate of
Pérez-Mendoza et al., 2014). The major virulent factors include type 39% over 72 h in Caenorhabditus elegans compared to 78% in the control
secretion systems (TSSs) and their associated effector proteins. Some of (Taylor et al., 2017). In P. aeruginosa, the sRNA ErsA contributes to the
these TSS participate in suppressing host immune responses during regulatory system involved in its pathogenicity factors, including
infection. Type 1 secretion systems (T1SS) secrete adhesins, which are motility and biofilm formation. Studies using genetically modified
critical in binding to the host, Type 3 secretion systems secrete effectors P. aeruginosa strains have shown that ErsA influences EPS production
to suppress host immunity, and c-di-GMP also regulates the Type 6 and positively regulates biofilm formation and maturation processes
secretion systems to export ATPase (Tran et al., 2014; Trampari et al., (Ferrara et al., 2015; Falcone et al., 2018). It acts through the
2015; Wang et al., 2017; Kan et al., 2018). T1SS secretes adhesins post-transcriptional modulation of the transcription factor AmrZ by
through the ATP-binding cassette transporter. Protein translocation targeting the algC gene encoding virulence factors (Ferrara et al., 2020).
occurs via a periplasmic intermediate (Smith et al., 2018). The secretion In P. aeruginosa, multiple regulatory systems contribute to their patho­
of Lap adhesins results in the autophosphorylation of the GacS sensor, genicity. A few hundred virulence factors are QS-mediated. The role of
which transmits its phosphate group to transcriptional regulators. This sRNA (Qrrs) in regulating QS-mediated virulence factors has provided
mechanism triggers a regulatory cascade transcribing many genes. significant insights into developing antimicrobial drugs. Although
During the limitation of nutrients, binding occurs between c-di-GMP to transcriptome sequencing has identified potential intergenic sRNAs,
regulatory protein LapD, which induces sequestration of protease LapG. only the roles of a few have been well-defined. The sRNAs PrrH, PhrS,
Under low c-di-GMP concentrations, biofilm dispersal takes place (Yuan PrrF, and AmiL are involved in expressing virulence factors, motility,
et al., 2015). and biofilm formation (Thi Bach Nguyen et al., 2018; Lu et al., 2019; Pu
Norspermidine, a polyamine secreted by Vibrio cholerae, regulates et al., 2022).
biofilm formation. Deleting the nspC gene encoding for the decarbox­
ylase enzyme involved in norspermidine synthesis reduces biofilm for­ 2.3. Quroum sensing
mation (Lee et al., 2009). The NspS/MbaA signaling system binds the
NspS protein to norspermidine, inhibiting MbaA phosphodiesterase ac­ Bacteria express specific characteristics that are only beneficial if
tivity. Furthermore, it also enhances c-di-GMP concentration to promote they are produced in "large" quantities. These genes are expressed in a
biofilm formation. Norspermidine within biofilm also assists in synchronized manner when bacterial population density is high
acquiring iron, which helps strengthen biofilms (Wotanis et al., 2017). A (Papenfort and Bassler, 2016; Kalia et al., 2019). The expression of
few other works have elucidated that c-di-GMP mediates the production QS-based genes leads to biofilm formation, sporulation, and antibiotic
of the bacterial ECM, leading to biofilm formation (De Smet et al., 2021). biosynthesis. The QSS is involved in the biosynthesis of signaling mol­
Bacterial binding to the surface during biofilm formation coincides with ecules released into the milieu and sensed by specific receptors. The
the activation of diguanylate cyclase enzymes (DGCs) involved in signal-receptor complex is translocated into the cell and initiates tran­
C-di-GMP synthesis. It results in loss of motility due to DGC (SadC) and scription upon binding to gene promoters. This QS-mediated gene
an increase in exopolysaccharide production due to another DGC (RoeA) expression begins bacterial biofilm formation by synthesizing auto­
(Merritt et al., 2007, 2010; Christen et al., 2010; Massie et al., 2012). inducer (AI) signal molecules (i) acylhomoserine lactones (AHLs), (ii) AI
cAMP levels increase from low in planktonic cells to high after a few peptides, and (iii) AI-2 (Papenfort and Bassler, 2016; Dong et al., 2021).
rounds of bacterial attachment/detachment. As this process progresses, In addition, biomolecules with signaling characteristics include (i) fatty
irreversibly attached cells transform into strong biofilm structures acids, (ii) quinolones and diketopiperazines (PQS), (iii) ketones, and (iv)
(Armbruster and Parsek, 2018). C-di-GMP is degraded by the phospho­ epinephrine and norepinephrine (Koul et al., 2016; Zhou et al., 2017).
diesterases (PDEs) DipA and RbdA (Basu and Sauer, 2014; Petrova et al., Biofilm-forming QSSs vary among bacterial species; AI-2 regulates ica,
2015). Finally, c-di-GMP is reported to be inducing biofilms’ dispersal to bhp, and icaR in Staphylococcus epidermidis, and las, rhl, PQS, and inte­
initiate subsequent rounds of bacterial infection (De Smet et al., 2021). grated QS in P. aeruginosa (Papenfort and Bassler, 2016; Lee and Yoon,
2017). In contrast to a single QSS, a small number of bacteria possess
2.2. Small non-coding RNAs multiple QSSs, including Bacillus, Clostridium, Pseudomonas, Sino­
rhizobium, Streptococcus, and Vibrio spp. These QSSs operate as hierar­
Multiple mechanisms and pathways regulate biofilm formation. A chical systems or networks (Koul et al., 2016).
signal transduction system mediates microbial adhesion and ECM syn­ Bacterial cyclic dimeric guanosine monophosphate (c-di-GMP) is a
thesis within biofilms through various transcription factors. In addition signaling molecule. It mediates the production of the bacterial ECM,
to transcription factors, small RNA (sRNA) post-transcriptional regula­ leading to biofilm formation. The binding of C-di-GMP to membrane
tors have been shown to play a critical role in biofilm formation proteins LapD and LapA and effector YcgR regulates the transition from
(Fernández et al., 2016; Pusic et al., 2016). Protein kinase C-mediated the planktonic to biofilm mode of lifestyle (López-Baena et al., 2019).
Mitogen-Activated Protein (MAP) kinase pathways and protein phos­ Norspermidine, a polyamine secreted by Vibrio cholerae, regulates bio­
phatases are vital. In Candida albicans, two main transcription factors, film formation. The NspS/MbaA signaling system binds NspS protein to
Bcr1p and Zap1p, regulate the expression of adhesins and the production norspermidine, inhibiting MbaA phosphodiesterase activity. Further­
of β-1,3 glucan during biofilm formation (Nobile et al., 2009; Srikantha more, it also enhances c-di-GMP concentration to promote biofilm for­
et al., 2013). During C. albicans infection transcription factor, Fks1p mation. Norspermidine within biofilm also assists in acquiring iron,
regulates the level of ECM polysaccharide in the biofilm (Taff et al., which helps strengthen biofilms (Wotanis et al., 2017). Levels of cAMP
2012). In Aspergillus fumigatus, a reduction in adhesion and changes in increase from low in planktonic cells to high after a few rounds of
cell surface and ECM production were observed in the absence of MAP bacterial attachment/detachment. As this process progresses, irrevers­
kinase activity, especially those due to SakA, MpkA, and MpkC. Phos­ ibly attached cells transform into strong biofilm structures (Armbruster
phatase mutants (ΔptcB and ΔsitA), having low cellular integrity and and Parsek, 2018).

3
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Other molecules that regulate the transition of planktonic bacteria to killing bacterial pathogens to regulating pathogenicity and virulence
the biofilm stage are sRNAs. Environmental stress induces sRNA pro­ factors (Subhadra et al., 2018; Kalia et al., 2019). Antibiofilm strategies
duction. This system promotes gene expression in the ECM and curli have been targeting either the formation stages or the pre­
fimbriae and represses flagellar genes (Taylor et al., 2017). Microbial formed/mature biofilm through the disintegration of the preformed
adhesion and ECM synthesis within biofilms are mediated by a signal biofilms, inhibiting QS, and non-QS regulatory mechanisms. Bacterial
transduction system, in which kinases (SakA, MpkA, and MpkC) are surface, membrane, and secretory molecules contribute to their ability
highly active. Phosphatase mutants (ΔptcB and ΔsitA), which have low to evade the host immune system and adhere to host cells. A preventive
cellular integrity and reduced phosphorylation of MpkA and SakA, have measure to hind infections is to target bacterial adhesion, the initial step
been used to demonstrate their role in inducing these changes (Manfiolli in biofilm formation (Sharma et al., 2017; Dufrêne and Viljoen, 2020).
et al., 2018). Anti-adhesion strategy has the latent advantage of not hindering bac­
terial growth resulting in minimal selective pressure (Parrino et al.,
2.4. eDNA 2018). In subsequent stages of infection, mature biofilms can be
removed by degrading their matrix components. A few enzymes, such as
Bacterial cell lysis releases genomic DNA (gDNA) into biofilms. The (i) deoxyribonucleases (DNases), (ii) polysaccharide-degraders, and (iii)
gDNA may or may not undergo structural changes (Turnbull et al., 2016; proteases, can effectively metabolize the various components of the
Deng et al., 2020). A comparison between strong and weak biofilm, leading to its disintegration. Mature biofilms are more difficult
biofilm-forming strains of S. epidermidis showed that the former pro­ to eradicate as these are more tolerant to antimicrobial agents due to an
duced more eDNA (Montanaro et al., 2011). This observation was alteration in the growth rate of bacterial cells embedded in the biofilm
further supported by a comparison S. epidermidis and S. aureus, which matrix, horizontal transmission of genes conferring resistance, and the
showed that higher biofilm formation in the former was due to the emergence of resistance mutants (Ito et al., 2009; Savage et al., 2013). In
mechanism of eDNA release via autolysin protein (AtlE) and cell lysis addition to biological techniques, some studies have used electro­
(Dusane, 2017; Zatorska et al., 2017). In gram-negative bacteria, such as chemical and photodynamic approaches to eradicate biofilms (Sultana
Burkholderia pseudomallei, the quantity of eDNA in the biofilm is posi­ et al., 2016; Darmani et al., 2018; Gholibegloo et al., 2018; Srinivasan
tively correlated with biomass thickness, independent of the ratio of et al., 2021).
living and dead bacteria (Pakkulnan et al., 2019). The production and
release of eDNA follow different mechanisms; however, they generally 3.1. Anti-Adhesion
occur through cell lysis (Ibáñez de Aldecoa et al., 2017) or via mem­
brane vesicles (Toyofuku et al., 2019). QS-mediated release of eDNA The adhesion of bacterial cells to the surfaces involves various fac­
also occurs through 4–hydroxy-2–alkylquinolines (HAQs). In tors, such as surface proteins, pili, and exopolysaccharides (Asadi et al.,
P. aeruginosa, HAQs produce reactive oxygen species (ROS) that damage 2019). The most prevalent antibiofilm approaches involve biocides,
the cell membrane and release eDNA (Tahrioui et al., 2019). Abundant antibiotics, and ion coatings. The non-specific adhesion of E. coli can be
biofilms then form around the fragmented eDNA. The addition of DNase inhibited by the herbicide 2,4-dichlorophenoxyacetic acid (Bhat et al.,
I-digested gDNA to P. aeruginosa enhances its biofilm-forming ability 2018). Coating medical devices, such as catheters, with antibiotics, such
(Deng et al., 2020). as rifampin, minocycline, gentamicin, and norfloxacin, could signifi­
eDNA contributes to biofilm fitness through different mechanisms. It cantly reduce S. aureus and UTIs (Ramos et al., 2011). Telithromycin, a
regulates phenazine retention, redox recycling, and extracellular elec­ semi-synthetic antibiotic, is effective against biofilms formed by
tron transfer to the bacterial populations within biofilms (Saunders S. aureus, E. faecalis, and E. faecium strains. A 35–40% reduction has
et al., 2020). It also promotes antibiotic resistance in bacteria within been reported against biofilm formation and for treating established
biofilms by inactivating cationic antibiotics through HGT (Keeling and biofilms. The effect of telithromycin could be enhanced from 40% to
Palmer, 2008; Saxena et al., 2019). eDNA interacts with various biofilm 70% in combination with ampicillin (Xiong et al., 2021). Antimicrobial
components, specifically with a high affinity or non-specifically with a peptides, such as peptide 1018 and lantibiotics, have been found to
low affinity. eDNA binds to various virulence factors and ribosomal interfere with the biofilm formation process in several pathogens,
proteins to promote biofilm formation (Graf et al., 2019). eDNA-protein including P. aeruginosa, E. coli, Acinetobacter baumannii, K. pneumoniae,
interactions stabilize anionic eDNA and facilitate bacterial aggregation S. aureus, Salmonella Typhimurium, and Burkholderia cenocepacia (De la
(Arenas and Tommassen, 2017; Kavanaugh et al., 2019). Complexes of Fuente-Núñez et al., 2014), as well as S. aureus, and S. epidermidis
host factors and histone-like proteins of the DNABII protein family bind (Saising et al., 2012). A peptide derived from β-neurexin could block
to DNA with high affinity in the extracellular matrix of biofilms (Devaraj surface attachment and adhesion by bacteria, leading to the complete
et al., 2018). Interactions among eDNA, EPS, and PsI in P. aeruginosa inhibition of biofilm accumulation (Feuillie et al., 2017). Immobilizing
promote bacterial adhesion and growth (Wang et al., 2015). In addition, antimicrobial agents, such as N-alkylpyridinium bromide, to poly
eDNA interacts with phenazine, which participates in cell signalling, (4-vinyl-N-hexylpyridine) resulted in a 99% inhibition of infections on
biofilm formation, iron acquisition, and antibiotic resistance (Schiessl medical devices caused by S. epidermidis, E. coli, and P. aeruginosa.
et al., 2019; Zhu et al., 2019). In P. aeruginosa, phenazine enhances Exposure to subinhibitory concentrations of sodium hypochloride in­
electron transfer, leading to stronger binding of eDNA to the cell surface duces structural changes in Listeria monocytogenes biofilms and strongly
and higher viscosity of the DNA and sputum (Das et al., 2013, 2015). The reduces the ability of cells to adhere to polystyrene surfaces. Inciden­
more virulent behavior of pathogenic bacteria and their higher stress tally, it also represses the expression of genes responsible for motility
resistance are attributed to divalent cation sequestration by eDNA and QS signaling activity (Bansal et al., 2021).
(Mulcahy et al., 2008; Johnson et al., 2013; Lin et al., 2018). Although Thymol (a phenol derivative) treatment (100 μg/mL) was effective
marked biotechnological advances have been made to overcome the in reducing MRSA adhesion on various surfaces coated with human
biological factors that lead to infectious diseases, the challenges of eDNA plasma. It also inhibited biofilm formation by 88%. This treatment is
and its interaction with various biofilm components are still difficult to mediated by the repression of sarA, which reduces the production of
circumvent (Panlilio and Rice, 2021). adhesins and hemolysins (Valliammai et al., 2020). A unique feature of
polyphenols extracted from fruits is their ability to inhibit biofilm for­
3. Antibiofilm strategies mation and destroy preformed biofilms inhabited by Rothia dentocariosa
and other Streptococcus spp., such as S. mutans, S. oralis, and S. mitis. A
In view of the rapid emergence of antibiotic resistance and its reduction in the adhesion of bacteria to surfaces results in the inhibition
persistence, there has been a shift in the focus of drug development from of biofilm formation by up to 97.3%. In addition, disruption of

4
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

preformed biofilms by up to 80% has been reported after treatment with et al., 2007). DNase 1 (5.0 μg/mL) caused a 40% decrease in biofilm
various combinations of phytocompounds (Sateriale et al., 2020). biomass. DNase 1 in combination with β-lactams resulted in an addi­
Poloxamer 338, with surfactant properties, has been proposed as an tional 10- to 15-fold decrease in viable bacterial counts. DNase 1 plus
anti-biofilm agent. It targets catheter-associated urinary tract infections azithromycin decreased cell viability in biofilms by 2- to 4- fold in
by repressing the adhesion of pathogenic bacteria. A P388-coating of Gram-positive bacteria (S. aureus, and Streptococcus pyogenes) and by
35 nm on catheters could reduce 0.51–0.83 log10 of E. coli strains 20-fold in Haemophilus influenzae (Tetz et al., 2009). DNase (NucB) ob­
Ec5FSL and Ec9FSL (Stirpe et al., 2020). Biosurfactants have been shown tained from Bacillus licheniformis disperses eDNA present in the EPS
to exert anti-adhesive activity against bacterial pathogens. The bio­ matrix of biofilms formed by Gram-negative and Gram-positive bacteria,
surfactant produced by the probiotics Lactobacillus acidophilus and which demonstrates that bacteria can exploit these enzymes to disperse
L. rhamnosus inhibits S. mutans biofilm formation by downregulating the biofilms formed by their competitors (Nijland et al., 2010). The inhibi­
expression of gtfB/C and ftfL (Satpute et al., 2019; Tahmourespour et al., tory effects of the extracellular protease and DNase from Bdellovibrio
2019). bacteriovorus HD100 on S. aureus biofilms have been demonstrated. This
Screening studies identified monoclonal antibodies as anti-adhesive has been proposed as an approach for preventing the invasion of
agents that prevent interactions involving S. aureus I adhesin, Cn, and S. aureus into human epithelial cells (Monnappa et al., 2014).
laminin (Herman-Bausier et al., 2016). The adhesion of bacterial cells to Treating the biofilms during the early development stage with DNase
surfaces, such as the surface protein SdrC of S. aureus, is a critical I (0.1 U/100 μl) significantly inhibited growth and effectively disrupted
component of biofilm formation (Wang et al., 2021). Another technique (>85%) 72-h-old mature C. jejuni biofilms (Kim et al., 2017)(Table 2). A
for inhibiting bacterial adhesion involves blocking combination of DNaseI-benzalkonium chloride (BAC) caused a 3–5 log
carbohydrate-binding adhesins. Chemically-modified Galα1–4 Gal de­ CFU/mL disruption in a 48-h-old dual (E. coli + L. monocytogenes)
rivatives inhibit streptococcal adhesin SadP (Haataja et al., 2018). pathogenic species biofilm on a stainless-steel surface.
Coating titanium with DNase I inhibits bacterial adhesion and Cellulase/pronase-BAC combination was less effective than DnaseI. The
biofilm-forming abilities (Ye et al., 2017). Chelators reduce the avail­ efficiency of DnaseI-BAC was also effective in Pseudomonas fluorescens –
ability of metal ions in the biofilm matrix. Silver ions (AgNPs), along L. monocytogenes mixed culture biofilms. For effective biofilm removal
with antibiotics, show enhanced antibacterial activity by interfering with cellulase, a concentration of 1000 µg/mL was required (Rodrí­
with the DNA replication, protein synthesis, and respiratory process of guez-López et al., 2017). Taking advantage of the competitive behavior
E. coli, S. aureus, Klebsiella species, P. aeruginosa, S. typhimurium, Candida of two types of foodborne pathogens, the supernatants of the
albicans (Secinti et al., 2011; Chernousova and Epple, 2013; Besinis non-biofilm formers degraded C. jejuni NCTC11168 biofilm owing to a
et al., 2017). Despite the demonstration of various anti-adhesive agents, 16% reduction in eDNA. This approach could eradicate the pre-existing
their application in treating infections caused by pathogens at the clin­ biofilm entirely (Jung et al., 2017). DNase I has been successfully used to
ical level is still lacking. These can be recommended as supplementary target eDNA to assist the action of antibiotics in eliminating CF-related
approaches (Vuotto and Donelli, 2019). P. aeruginosa biofilms. A combination therapy (GSH + DNase I + cip­
rofloxacin 5 µg/mL) could significantly reduce biofilm viability by
3.2. Anti-extracellular matrix components 13–35%. An additional benefit of this combined treatment is the GSH
based assistance in the re-growth of the damaged tissue because of the
An approach to degrade biofilm targets its major components: exo­ recovery of the host cells (Das et al., 2017).
polysaccharides (EPS), extracellular DNAs (eDNAs), and proteins. Pre-treatment of P. aeruginosa PAO1 in the presence of 5 µg/mL
(Flemming and Wingender, 2010; Mann and Wozniak, 2012). EPS and DNase caused a 68.6% reduction in biofilm formation over a period of
eDNA are the major factors that regulate and promote antibiotic resis­ 24 h. In contrast, biofilms treated with 10 µg/mL DNase exhibited a 70%
tance by delaying the diffusion of antimicrobials or promoting the reduction. Here, the addition of Mg2+ ions (10 mM) enhanced the effi­
transmission of antibiotic-resistance genes (Mulcahy et al., 2008). The ciency of post-treatment DNase activity (5 µg/mL). It resulted in a 90%
disintegration of the biofilm exposes the sessile cells, making them reduction in the preformed biofilm within 5 min of contact time. How­
susceptible to host immune defense, and much lower concentrations of ever, DNase treatment was less effective against mixed-species biofilms
antibiotics than those required against their free-living counterparts formed by P. aeruginosa PAO1, Klebsiella spp., E. faecalis, and
(Kaplan, 2009). S. Typhimurium, and S. aureus (Sharma and Pagedar Singh, 2018). Under
eDNA, an integral element of the extracellular polymeric matrix in vitro conditions, biofilm treatment with DNase1 (0.001 or
(EPM). Deoxyribonuclease I digests eDNA present in the biofilms of a 0.002 mg/mL) significantly affected (reduced) the growth (log10 CFU)
wide range of bacterial pathogens: Staphylococcus strains, A. baumannii, Streptococcus mutans UA 159 (OMZ 918), and S. oralis SK 248 (OMZ 607)
E. coli, Haemophilus influenzae, K. pneumoniae, Psuedomonas aeruginosa, induced biofilm. However, proteinase K treatment alone or in combi­
etc. (Kaplan et al., 2012). Campylobacter jejuni strain RM1221 is a poor nation with DNase I showed an enhanced growth of S. mutans UA 159
biofilm former due to the presence of genes encoding eDNase. eDNase (OMZ 918), and S. oralis SK 248 (OMZ 607) induced biofilms. Confocal
from strain RM1221 could efficiently disrupt C. jejuni NCTC 11168 images of biofilms treated with a combination of DNaseI and proteinase
biofilms formed on steel surfaces. A surface treatment with eDNase can K resulted in less dense biofilms. Complete degradation of eDNA within
disrupt even the preformed biofilms (Brown et al., 2015). biofilms treated with DNase I was also recorded with Actinomyces oris
DNases have generally been observed to prevent biofilm formation OMZ 745, Candida albicans OMZ 110, and Fusobacterium nucleatum
rather than degrade pre-formed biofilms (Okshevsky et al., 2015). KP-F2 (OMZ 596) (Karygianni et al., 2020b). Various studies have been
DNAse treatment has been reported to be effective during the early conducted using commercially available DNases, such as DNase I
stages of biofilm formation (< 6 h old). Dornase alfa (engineered human derived from bovine pancreas, DNase 1L2 from human keratinocytes,
DNAse I) is used to treat patients with cystic fibrosis (CF). The degra­ Dornase alfa (recombinant human DNase), λ exonuclease (viral DNase),
dation of eDNA reduces spitum viscosity (Wagener and Kupfer, 2012). NucB, and streptodornase produced by Bacillus licheniformis and Strep­
DNase breaks down the phosphodiester linkages of eDNA, leading to the tococcus spp., to disperse pathogenic bacterial biofilms (Fleming and
inhibition of biofilm formation in Gram-positive and Gram-negative Rumbaugh, 2017). It may be concluded that a combination therapy of
bacteria. Treatment leads to changes in various characteristics of the DNase I and antimicrobials can be more effective than either alone.
bacterial community, including biomass, morphology, and architecture. Although eDNA is a critical target for inhibiting biofilm formation,
Treatment with DNase I-like 2 (DNase IL2) (present in human skin) its interactions with other molecules can also be exploited as targets for
efficiently suppresses biofilm formation by P. aeruginosa and S, aureus, anti-biofilm drugs. DNase treatment increases biofilm permeability.
which demonstrates the role of eDNA in biofilm development (Eckhart Monoclonal antibodies (MAbs) specifically targeting DNABII disrupt

5
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Table 1
Bioactive molecules inhibiting quorum sensing mediated bacterial biofilms.
Bioactive molecule Pathogen QS inhibition Surface Biofilm inhibition Effective Applications References
(Source) (I); concentration (EC);
Removal of Applied
preformed (R) (%) concentration (AC)
of inhibitor

Phytochemicals Pseudomonas AHL PS MTP I: 11.82–84.37; R: MEKB: methanolic For treating Sarkar et al. (2015)
(Kalanchoe aeruginosa biosynthesis 46.8–91.7 (viable extract of K. pathogenic infections
blossfeldiana) cells) blossfeldiana in humans
EC: 30, and 62.5 μg/
mL;
AC: 15, 30, 62.5,
125 μg/mL; (MIC:
250 μg/mL)
Curcumin (280 µg/ Aeromonas sobria AHL PS MTP I: 93.95%; Curcumin liposomes Treating opportunistic Ding et al. (2017)
mL) biosynthesis R: NR EC: 280 µg/mL; food pathogens
AC: 280–420 µg/mL
AHL lactonase Acinetobacter AHL Glass I: 80% with Aii20J; EC: Aii20J at 20 μg/ Prevention and Mayer et al. (2020)
(Aii20JB) and baumannii ATCC® biosynthesis coverslips 20% with DNase; mL; treatment of bacterial
DNase 17978™ R: NR AC: Aii20J at 20 μg/ infections such as
mL, DNase at 2 U/ pneumonia,
mL; α-amylase septicemia, skin,
(10 U/mL) (alone or wound, and urinary
in combinations) tract infections
Spathulenol P. aeruginosa AHL PS: MTP I: 80% by plant EC: 10 μl of 0.1 mg/ Potential use as an Luciardi et al. (2016)
(Merremia biosynthesis extract); mL extracts effective
dissecta) R:NR AC: 0.1 mg/mL antipathogenic drug
extracts in
DMSO–H2O (50:50)
Lactonase (SsoPox-I P. aeruginosa AHL MTP Variable; - SsoPox-I: Therapeutic usage Hraiech et al. (2014)
protein) degradation EC: 1 mg/mL against pneumonia
(250 μl) (Rat
model);
AC:0.05–50 µg
(100 μl)
Lactonase (Bacillus P. aeruginosa PAO1 AHL MTP 60; - EC: Cell extracts Suppression of Rajesh and Ravishankar
and Enterobacter) and PAO1-JP2. degradation 100 μg/mL virulence in humans Rai (2014)
AC: Cell extracts and plants
100 μg/mL
Lactonase Acinetobacter AHL Medium 68.75%; 2 µm3/ EC: AHL-lactonases, Effective Chow et al. (2014)
(Geobacillus baumannii S1 degradation plate μm2 10 μl of purified antipathogens against
kaustophilus) enzyme (40 mg/ human bacterial
mL); pathogens
AC: AHL-lactonases,
10–30 μl of purified
enzyme (40 mg/mL)
Fatty acids P. aeruginosa PA01 AHL PS 48.29–53.99; - EC: Not defined Promising medicinal Pejin et al. (2014)
(Ochridaspongia degradation AC: 0.5 MIC; MIC properties
rotunda) was 30 mg/
mL O. rotunda
samples
Hexasaccharide Staphylococcus AHL PS 100; 71.5 EC: 5 mg/mL CHS Prevent bacterial Srivastava et al. (2015)
(Equus caballus) aureus degradation AC: 2–10 mg/mL for pathogenicity
MIC
AHL lactonase P. aeruginosa AHL Medium 60;- EC: 20 μl of purified Potential use as El Aichar et al. (2022)
(Bacillus degradation plate cell extracts (PCE) probiotics and
thuringiensis) AC: 20 μl PCE of the antibiofilm agent
Bacillus strains
dilutions from 10− 1
to 10− 5
AHL lactonase Streptococcus AHL Medium 63;- EC: 20 μl of purified Potential use as
(Bacillus subtilis) mutans degradation plate cell extracts (PCE) probiotics and
AC: 20 μl PCE of the antibiofilm agent
Bacillus strains
dilutions from 10− 1
to 10− 5
Diketopiperazines Lelliottia amnigena AHL Medium 70–80;- EC: 100 μl of crude Reduce pathogenicity. Kachhadia et al. (2022)
(by Bacillus degradation plate extract Reduction in the
cereus) AC: 100 μl of crude emergence of
extract antibiotic-resistant
bacteria
Passiflora edulis Chromobacterium AHL Medium 51–90;- EC: 2 mg/mL Reduced biofilm Venkatramanan et al.
violaceum degradation plate (P. edulis extract) formation (2020)
AC: 0.5, 1.0, and
2.0 mg/mL
(continued on next page)

6
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Table 1 (continued )
Bioactive molecule Pathogen QS inhibition Surface Biofilm inhibition Effective Applications References
(Source) (I); concentration (EC);
Removal of Applied
preformed (R) (%) concentration (AC)
of inhibitor

Coumarin Porphyromonas Autoinducer-2 MTP -;40.4 EC: 200 μM Treatment of He et al. (2022)
gingivalis inhibition AC: 25–200 μM periodontitis
(periodontal
multispecies biofilm)
Isothiocyanates P. aeruginosa Block signal MTP NR;- EC: 200 μM Treating opportunistic Ganin et al. (2013)
(Broccoli) receptor (Sulforaphane) bacterial pathogens.
AC:50–200 μM
(Sulforaphane)
Phenolic compound P. aeruginosa Block signal MTP Variable;- EC: 1.36 mM Suppressing virulence Gutiérrez-Barranquero
(Coumarin) receptor Coumarin and broad spectrum et al. (2015)
AC: 10.0, 5.0, 2.0, drug
1.71, 1.36 and
1.0 mM Coumarin
Polyphenols from P. aeruginosa Block signal MTP >90;- EC: 4.5 μg/mL Affect Quorum- Prateeksha et al. (2017)
honey (HP receptor SeNPs@HP sensing mediated
AC: SeNPs: 7.5 and pathogenicity; Anti-
25 μg/mL; virulence activities
HP: 0.6% and 5%; (in-vitro and in-vivo)
SeNPs@HP: 4.5 and
25 μg/mL
Hamamelitannin S. aureus Inhibits gene Artificial 90.9% (in CFUs/ EC: HAM (120 μg/ Wound healing Brackman et al. (2016)
+ antibiotic transcription dermis biofilm, mL), VAN (250 μg/
(Vancomycin) P. aeruginosa); 70% mL);
(in CFUs/ biofilm in AC: HAM
S. aureus Mu50); NS (0.5–500 μg/mL)
and vancomycin
(1–500 μg/mL)
Dioon spinulosum Porphyromonas Down- MTP 77.1; - EC: Not defined Application in treating Elekhnawy et al. (2022)
extract gingivalis regulation of AC: 125–500 μg/mL wounds, respiratory
genes for QS Dyer Ex Eichler and urinary tract
and biofilm extract infections

‘-‘ Not available; AHL: Acylhomoserine lactone; PS: Polystyrene; MTP: Microtiterplate.

preformed biofilms (Novotny et al., 2016). In contrast, proteinase K antibiotic and 50% and 10% respectively with disinfectant (Dieltjens
treatment did not cause any additional changes in permeability (Ran­ et al., 2020) (Table 2). Preformed biofilms can be destabilized and dis­
drianjatovo-Gbalou et al., 2017). It has been suggested that targeting rupted by EPS metabolism. The action of hydrolytic enzymes on EPS
DNABII is more effective because it is an integral part of the vertices of components can significantly alter biofilm integrity (Kalia et al., 2018).
DNA strands (Novotny et al., 2016). DNase I and alginate lyase disin­ The role of proteases in biofilm dispersal has been demonstrated for
tegrate the biofilms formed by Enterococcus faecalis and E. faecium in the many pathogens: (i) Aureolysin and V8 serine protease (SspA) act against
urinary tract. These enzymatic treatments enable vancomycin to S. aureus biofilms by degrading Bap and clumping factor b (Gustafsson and
significantly reduce biofilm hydrophobicity and structure, and cell Oscarsson, 2008), (ii) Trypsin disperses P. aeruginosa biofilms (Banar et al.,
viability (Torelli et al., 2017). 2016). Thymol in thyme oil (0.5 mg/mL), and Proteinase K (50 and
The major ECM degrading enzymes are hydrolases, proteases, and 100 µg/mL) combinations effectively inhibited adhesion and proliferation
nucleases (Li and Lee, 2017). Dispersin B (glycoside hydrolase) hydro­ of E. coli O157:H7. On the other hand, (50–100 µg/mL PK +
lyzes poly-N-acetylglucosamine (PNAG), leading to a marked reduction 0.25/0.5 mg/mL TO) had synergistic biofilm-dispersal effects at 24 h (Cui
in the infections caused by Staphylococcus spp. and E. coli. A combination et al., 2016). A few other examples of the role of proteases are as follows:
of Dispersin B and triclosan reduced biofilm forming ability of S. aureus, (i) LapG Protease A triggers dispersal by modifying the EPS-binding pro­
S. epidermidis, and E. coli (Darouiche et al., 2009). Proteinase K cleaves tein LapA; (ii) proteinase K targets peptide bonds and disperses biofilms
the protein component of the biofilm leading to its dispersal. Dispersin B produced by many bacteria; (iii) Spl protease A disperses S. aureus by
treatment followed by Proteinase K or trypsin was more effective in cleaving EbpS proteins associated with the cell wall; (iv) streptococcal
eradicating Staphylococcus biofilms. The major limitation of this cysteine protease (SpeB) A hydrolyzes surface proteins M and F1; and (v)
approach is the allergic reactions elicited by the host (Chen et al., 2013). protease surface-protein-releasing enzyme (SPRE) acts by releasing anti­
The quantity of EPS present in the ECM regulates the penetration and gen P1 (Fleming and Rumbaugh, 2017).
diffusion of antimicrobial compounds into Salmonella enterica Typhi­ A crude extract of Adiantum philippense L. decreased the EPS content
murium ATCC14028 biofilms. Salmonella biofilms produced by an of biofilms. It attacks adhesins and disrupts preformed biofilms. At MIC the
isogenic ΔcgsD mutant had significantly reduced EPS production, which inhibition (%) of adhesion and of preformed biofilms by A. philippense was
reduced cell adhesion and 72% lower biofilm biomass. EPS inhibitor quite varied, ranging from as low as 37.34 and 44.10 for Shigella flexneri,
(2-cyclopentenyl-5-(4-chlorophenyl)− 2-aminoimidazole dissolved in 50.26% and 56.54% for P. aeruginosa, 54.73 and 62.72 for E. coli, and
DMSO) reduced biofilm cells in the wild-type strain from 2.3 C to 60.92% and 70.58% for S. aureus, respectively (Adnan et al., 2020). The
1.8 CFU x 109, whereas in the sensitive strain, the reduction was from human systemic amyloid precursor transthyretin eradicates bacterial
1.8 to 1.2 CFU x 109. The impact on biofilm biomass (OD 570) WT was biofilms by inhibiting CsgA amyloid-β aggregation. Curli amyloid fibers
0.28 and 0.1; in contrast, in the sensitive strain, it got reduced from 0.24 along with cellulose are an integral component of EPM, in E. coli and
to 0.02. The sensitivity to ciprofloxacin and hydrogen peroxide (H2O2) Bacillus subtilis biofilms. Engineered nontetramer-forming monomer
on monoculture biofilms of wild-type strain ATCC14028 and isogenic (M-TTR) was more efficient than wildtype-TTR tetramer (Jain et al.,
ΔcsgD mutant (as survival %) were 17% and 2%, respectively, with 2017).

7
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Table 2
Bioactive molecules inhibiting biofilm matrix components.
Bioactive molecule Bacterial pathogen Effective [Applied] Biofilm inhibition/ References
concentration of inhibitor removal (%)
Name Activity

DNase I Oxidative Campylobacter jejuni EC: 4 U/mL Complete Brown et al. (2015)
AC: 4 U/mL
DNase I Oxidative C. jejuni EC: 0.1 U/100 μl -a;> 85 Kim et al. (2017)
AC: 0.1, 0.01, and 0.001 U/100 μl
DNase I + benzalkonium Oxidative Escherichia coli EC: 400 µg/mL and 600 µg/mL 3–5 log CFU/mL reduction Rodríguez-López
chloride (BAC) and + Proteolytic + Listeria monocytogenes AC: et al. (2017)
Pronase-BAC Enzymes: 200, 400, 700, and
1000 µg/mL;
BAC (Benzalkonium chloride): 25, 50,
100, 250, and 500 µg/mL
BAC-PRN/DnaseI: 400 µg/mL
(enzyme)
Dnase I Oxidative Campylobacter jejuni and EC: 50 μl of ~108 CFU of the cell 16% reduction in eDNA; Significant Jung et al. (2017)
C. coli suspensions
AC: 50 μl of ~108 CFU of the cell
suspensions
DNase I + GSH Oxidative Pseudomonas aeruginosa EC: DNase I (40 U) or GSH (5 mM); Combination Therapy (CT) (GSH + Das et al. (2017)
+ Pyocyanin ciprofloxacin (5 µg/mL), DNase I + ciprofloxacin 5 µg/mL)]:
AC: DNase I (40 U) or GSH (5 mM); biofilm viability significantly reduced
ciprofloxacin (5, 10, and 15 µg/mL) to 13–35%.
DNase I + GSH Oxidative P. aeruginosa EC: DNase I (40 U) or GSH (5 mM); -;> 85
+ Ciprofloxacin ciprofloxacin (5 µg/mL),
AC: DNase I (40 U) or GSH (5 mM);
ciprofloxacin (5, 10, and 15 µg/mL)
DNase I Oxidative P. aeruginosa PAO1 EC: DNase I (5–10 µg/mL);, 68.6;- Sharma and
AC: DNase I (0–50 µg/mL) Pagedar Singh
DNase I + Mg2+ ions Oxidative P. aeruginosa PAO1 EC: DNase I (5 µg/mL);, -;90 (2018)
AC: DNase I (0–50 µg/mL)
DNase I Oxidative Streptococcus mutans UA EC: DNase I (0.001 mg/mL); 100 reduction in eDNA;- Karygianni et al.
159 (OMZ 918), and AC: DNase I (0.001–0.002 mg/mL) (2020b)
S. oralis SK 248 (OMZ
607)
2-cyclopentenyl-5-(4- EPS degrading Salmonella enterica EC: EPS inhibitor# - 50 μM Reduced EPS and cell adhesion. Dieltjens et al.
chlorophenyl)− 2- Typhimurium AC: EPS inhibitor - 50 μM Biofilm cell reduction inn resistant (2020)
aminoimidazole ATCC14028 strain 2.3–1.8 CFU x 109 where as in
the sensitive strain the reduction was
from 1.8 to 1.2 CFU x 109.
Aureolysin and V8 serine EPS degrading S. aureus EC: ND Degrading Bap and clumping factor Gustafsson and
protease (SspA) AC: ND Oscarsson (2008)
α-mannosidase, EPS degrading P. aeruginosa ATCC EC: mannosidases: 0.02 unit/mL and Degrading biofilms: 5–60% Banar et al. (2016)
β-mannosidase 27853 trypsin: 0.75 μg/mL Best: Trypsin
(Hydrolases) and AC: mannosidases: 0.005, 0.01, 0.015,
trypsin (Protease) 0.02 and 0.03 unit/mL and trypsin:
0.08, 0.175, 0.35, 0.75 and 1.5 μg/mL
Proteinase K + thyme oil Antibacterial E. coli O157:H7 EC: PK - 50 and 100 µg/mL; TO - 3.84–3.89 logs lower cells: Inhibit Cui et al. (2016)
(TO) 0.5 mg/mL) (Biofilm inhibition) adhesion and proliferation. Biofilm
EC: PK - 50 and 100 µg/mL; TO - 0.25/ dispersal (Not quantified)
0.5 mg/mL) (Biofilm dispersal).
AC: PK - 25, 50 and 100 µg/mL; TO -
4.0, 2.0, 1.0, 0.5 and 0.25 mg/mL)
Adiantum philippense L.+ EPS degrading Shigella flexneri, P. EC: 200 μl (MIC) + 100 μl (1000 μg/ 37–60.92; 44.10–70.58 Adnan et al. (2020)
chloramphenicol aeruginosa, E. coli, and S. mL)
aureus AC: 100–200 μl (plant extract -
1000 μg/mL) + chloramphenicol:
100 μl (1000 μg/mL)
Transthyretin (TTR) Anti-ECM E. coli and Bacillus EC: 1:1 CsgA: M-TTR 80;- Jain et al. (2017)
subtilis AC: CsgA (15 µM); 1:1 CsgA: M-TTR
and 50 µM M-TTR
Human monoclonal Anti-ECM Salmonella EC: 0.5 mg/mL 70 (Reduced staining); 40 Tursi et al. (2020)
antibody (mAb) 3H3 Typhimurium AC: 0.5 mg/mL
Vancomycin and Anti-ECM MSSA and MRSA EC: 10 mg/mL (Erdosteine) Reduced IC50 values Pani et al. (2022)
linezolid + erdosteine AC: 2, 5 and 10 mg/mL (Erdosteine)
alone or in combination with
Antibiotics (0, 0.5, 1, 5, 10, 20, 50,
100 and 200 times the MIC)
a
Not available; EPS: Exopolysaccharides; GSH: Reduced glutathione; IC50, half-maximal inhibitory concentration; MRSA: methicillin-resistant S. aureus; MSSA:
methicillin-sensitive S. aureus. #: 2-cyclopentenyl-5-(4-chlorophenyl)− 2-aminoimidazole dissolved in DMSO

The biofilm matrix formed by Salmonella spp. and E. coli contains vancomycin and linezolid against MSSA- and MRSA-induced biofilms
approximately 85% curli amyloid fibers. Human monoclonal antibody was further enhanced in the presence of erdosteine, a thiol-based drug,
(mAb) 3H3, which binds pan-amyloid epitopes, plays a dual role in over a period of 24 h. Erdosteine facilitated the penetration of the
preventing and eradicating biofilms (Tursi et al., 2020). The activity of antibiotic into the biofilm and disrupted EPS (Pani et al., 2022). Various

8
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

studies have revealed co-applicating these enzymes results in synergistic subsequent pathogenicity. The enzymatic degradation of AHLs in­
degradation of ECM components. activates QSSs. Lactonases and acylases produced by bacteria inactivate
A few other antibiofilm agents observed to act against pathogenic AHLs by opening the lactone ring and splitting the acyl chain, respec­
bacterial infections are the free fatty acids. P. aeruginosa produces cis-2- tively (Kalia, 2013). Bacillus extracts, purified lactonases, and the het­
decenoic acid to inhibit preformed biofilms induced by E. coli, erologous expression of the aiiA gene can inhibit AHL-mediated QS
K. pneumoniae, P. mirabilis, S. pyogenes, B. subtilis, S. aureus, and (Kumar et al., 2013; Noor et al., 2022). Biofilm formation by
C. albicans (Abdel-Mawgoud et al., 2010). Xanthomonas campestris pro­ P. aeruginosa in a rat model with highly lethal acute pneumonia was
duce cis-11-methyl-2-decanoic acid, which can induce biofilm dispersal observed to be reduced by lactonase (SsoPox-I) as a QSI. The reduction
by regulating the production of EPS-degrading enzyme (Deng et al., was dose-dependent with a [C1/2]~170 µg/mL. The treatment led to a
2014). However, fatty acids promote biofilm formation in B. subtilis 55% reduction (from 75% to 20%) in the mortality rate (Table 1)
during the initial stages (Pedrido et al., 2013). Although it has been (Hraiech et al., 2014). Similarly, the lactonases of the endophytic bac­
observed that L-amino acids promote biofilm formation in P. aeruginosa, teria Bacillus firmus PT18 and Enterobacter asburiae PT39 (extracts
however, D- and L- forms of tryptophan have the dual abilities of 100 μg/mL) inhibited biofilm formation by P. aeruginosa PAO1 and
inhibiting biofilm formation and it dispersal (Brandenburg et al., 2013). PAO1-JP2. The inactivation of C4-HSL by the cell-free lysate of B. firmus
Induction of biofilm dispersal in P. aeruginosa has been reported using PT18 and E. asburiae PT39 inhibited biofilm formation of > 80% (Rajesh
nitric oxide (NO), which can be generated from various compounds such and Ravishankar Rai, 2014). The wild-type GKL did not show any lac­
as sodium nitroprusside (SNP), S-nitroso-L-glutathione (GSNO) and tonase activity against 3-OH-C12-HSL but could effectively hydrolyze
S-nitroso-N-acetylpenicillamine (SNAP). A synergistic effect of NO was 3-OH-C10-HSL. However, mutant GKL enzyme (E101G/R230C) could
reported along with antibiotics leading to dispersal of P. aeruginosa inactivate 3-OH-C10-HSL and 3-OH-C12-HSL, which resulted in a
biofilms formed in CF sputums (Howlin et al., 2017). 68.75% reduction in biofilm biomass (μm3/μm2) with respect to the
inactive mutant (Chow et al., 2014). The QSI present in crude extracts
3.3. Anti-QS-mediated biofilm formation from the freshwater sponge Ochridaspongia rotunda causes a 48.29%
(methanol extract) and 53.99% (acetone extract) inhibition of biofilm
Bioactive molecules produced by prokaryotes and eukaryotes can formation by P. aeruginosa. The treatment also causes a 42–49%
potentially prevent QS-mediated biofilm formation (Kalia, 2013; reduction in pyocyanin production. The expression of QSI also affects
Gómez-Gómez et al., 2019). These QS inhibitors (QSIs) may target QSSs flagellar motility and twitching activity (Pejin et al., 2014). The hex­
at various stages, including signal biosynthesis, degradation of signal asaccharide present in mare colostrum causes AHL degradation and
molecules, competition for signals binding to cognate receptors, and suppresses QS-mediated activities, such as biofilm formation and path­
repression of QS-mediated gene transcription. As QSIs act below the ogenicity, by S. aureus. At 5 mg/mL, it curtails the antibiotic resistance
minimum inhibitory concentration (MIC) and do not restrict microbial of methicillin-resistant S. aureus (MRSA) (Srivastava et al., 2015). The
growth, we anticipate significantly reduced drug-resistant pathogens’ irreversible hydrolysis of AHL by acylase PvdQ activity in a mouse
emergence rates (Koul et al., 2016; Koul and Kalia, 2017). model demonstrated its potential as a therapeutic agent (Utari et al.,
2018). The AHL inhibitors 4-methoxycinnamic acid and 4-dimethylami­
3.3.1. Affecting signal biosynthesis nocinnamic acid have dual benefits, namely the inhibition of biofilm
Several QSIs have been shown to inhibit AHL biosynthesis, and formation and the enhancement of susceptibility to tobramycin (Cheng
consequently, QS-mediated biofilm formation (Table 1). Kalanchoe et al., 2020).
blossfeldiana leaf extract suppresses P. aeruginosa pathogenicity by Screening halotolerant strains of Bacillus revealed the presence of the
interfering with AHL biosynthesis. This results in 11.8–84.37% inhibi­ lactonase-encoding aii gene. The AHL degrading ability of Bacillus
tion of biofilm formation (Sarkar et al., 2015). The use of Merremia thuringiensis DZ16 caused a 60% inhibition of biofilm formation by
dissecta creeper extract, a medicinal plant, interferes with AHL biosyn­ P. aeruginosa. Lactonase of B. subtilis DZ17 reduced Streptococcus mutans
thesis by 63–75%, and the biofilm-forming ability of P. aeruginosa ATCC ATCC 25175 biofilm formation and biomass up to 63% in the xCELLi­
27853 by 55%. Spathulenol present in M. dissecta inhibits the gence model (El Aichar et al., 2022). Prominent biomolecules produced
biofilm-forming potential by up to 80% in P. aeruginosa and by 71% in by Bacillus cereus RC1 were diketopiperazines, viz. cyclo(D-phenyl­
S. aureus (Luciardi et al., 2016). Curcumin (2.5 μl of 280 µg/mL), alanyl-L-prolyl), cyclo Phe-Val, cyclo(Pro-Ala), cyclo(L-prolyl-L-valine),
entrapped in liposomes, interacts with the LuxI-type protein and inhibits cyclo (Leu-Leu), and cyclo(-Leu-Pro). These biomolecules caused an
the production of AHL. The production of C6-HSL, C10-HSL, and attenuation of 88.78%, 91.22%, and 97.59% of soft rot caused by Lel­
C12-HSL was completely inhibited with curcumin liposomes, whereas liottia amnigena RCE in cucumber, carrot, and potato, respectively. This
C4-HSL and C14-HSL were reduced up to 64.6% and 73.0% with free treatment reduced pathogenicity, especially biofilm formation, by
curcumin, compared to 71.35% and 93.55% with curcumin liposomes, 70–80% from 24 to 96 h after incubation without developing antibiotic
respectively. As a result of the reduced AHL concentration, biofilm resistance (Kachhadia et al., 2022). Passiflora edulis extract containing
formation was reduced by 93.35% in contrast to its control at 420 µg/mL hexadecanoic acid and 2-hydroxy-1-(hydroxymethyl) Et ester in­
and free curcumin, which resulted in a reduction of only 51.9%. In activates AHLs. Inhibition in EPS production by 35.9%, 52.2%, and
addition, curcumin reduces virulence factors’ expression in Aeromonas 72.5% and reduction in biofilm formation by 51.8%, 69.4%, and 90.7%
sobria (Ding et al., 2017). In A. baumannii, QSS-abaI/R mediates motility were observed at the sub-MIC levels of 0.5, 1, and 2 mg/mL, respectively
and biofilm formation. The purified QSI enzyme Aii20J reduced biofilm (Venkatramanan et al., 2020). Periodontitis caused by biofilm-forming
formation in A. baumannii by 80% after 24 h. However, DNase signifi­ Porphyromonas gingivalis is inhibited by coumarin. The downregulation
cantly reduced biofilm formation by 20%. A combined treatment of biofilm-related gene expression was observed, which led to the in­
involving Aii20J and DNase did not lead to any improvement in the hibition of AI-2 activity. The antimicrobial effect of coumarin (200 μM)
results than those obtained with the QSI enzyme. In contrast, the against P. gingivalis was observed regarding reduced planktonic growth
application of α-amylase did not affect biofilm formation. A strategy and membrane permeability. The most effective reducing activities were
involving induced mutations in AHL synthase AbaI and treatment with observed at 200 μM, affecting late-stage biofilm formation and dispersed
the QSI enzyme Aii20J and DNase can significantly contribute to pre­ preformed biofilms (He et al., 2022).
venting and treating A. baumannii infections (Mayer et al., 2020).
3.3.3. Blocking receptors
3.3.2. Inactivating signals Inhibition of QS-mediated gene expression can be achieved by
Degrading QS signals may block QS-mediated biofilm formation and blocking receptors with analogs of signaling molecules.

9
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Cinnamaldehyde and its derivatives exhibit vigorous inhibitory activity addition, α-CD (10 mM at 120 min) caused around 64% inhibition of
against QS-mediated biofilm formation in P. aeruginosa and E. coli (Kalia QS-mediated bioluminescence in Aliivibrio fischeri (Molnár et al., 2021).
et al., 2019). Plant biomolecules (from broccoli), such as isothiocyanates In contrast to the studies mentioned above, others have shown the
and glucosinolates, act as QSIs against E. coli and P. aeruginosa. Sulfo­ potential of CDs in releasing antibiotics and QSIs in a sustained manner.
raphane inhibited the AHL receptor LasR binding at 50 µM, whereas The QSI hamamelitannin (HAM), a nonpeptide analog, inhibits the RAP/
12 µM was sufficient to hinder biofilm formation by P. aeruginosa. In the TRAP QS system in S. aureus, thereby increasing the susceptibility of
presence of either of the isothiocyanates (100 µM), namely sulforaphane biofilms toward conventional antibiotics. This dual functionality of
or erucin, a 70% reduction in QS-mediated pyocyanin synthesis in HAM has several advantages. Wound care gauzes made of cellulose
P. aeruginosa was observed. The difference in the activities between the functionalized with non-toxic hydroxypropyl-β-cyclodextrin were found
two compounds seems to be due to differences in their hydrophobicity or to release antibiotics (vancomycin) and QSI (HAM) in a sustained
the slower oxidation of erucin (Ganin et al., 2013). Coumarin, a sec­ manner, causing: (i) a variation from 20% to 55% in biofilm formation,
ondary metabolite present in cassia, bison grass, melilot, tonka bean (ii) in the case of P. aeruginosa-induced biofilm, a 90.9% reduction in
woodruff, and cinnamon, inhibits QS-mediated biofilm formation by CFU/biofilm, and (iii) in the case of S. aureus Mu50, a non-significant
E. coli, Vibrio anguillarum, Edwardsiella tarda, and S. aureus. The phenolic reduction in single-species biofilms but a significant reduction (around
compounds of coumarin act as QSI by inhibiting AHLs, AI-2, and agr 70%) in mixed-species biofilms. This enabled overcoming the difficulties
QSS, which negatively affects rhlI- and pqsA-mediated gene expression in treating chronic wound infections caused by multispecies biofilms
responsible for biofilm formation with other virulence factors in (Brackman et al., 2016). CD-mediated QSI has been observed to exploit
P. aeruginosa (Gutiérrez-Barranquero et al., 2015). The Serratia marces­ the high solubility of CD in water as a mechanism to effectively deliver
cens biofilm-associated exopolysaccharides production, hydrophobicity, QS signal (AHL) analogs, which thus compete with AHL for receptor
and swarming motility were found to be inhibited by Piper betle extract. sites. Here, inclusion into β-CDs inhibited P. aeruginosa virulence (host
At 500 µg/mL, the PBE inhibited biofilm formation and EPS production evasion and biofilm development) in Caenorhabditis elegans (Ziegler
by 65% and 43%, respectively, in S. marcescens. Bioactive compounds et al., 2021). These approaches will likely provide solutions that may be
act primarily by downregulating the expression of the QS genes fimA, superior to antibiofilm agents compared to conventional techniques,
and fimC, which encode for fimbrial proteins responsible for adherence such as antibiotics through wound care gauzes.
and colonization (Srinivasan et al., 2016, 2017). Similar QS gene Extracts from Allium sativum L., Armoracia ruticana, Scutellaria bai­
downregulation due to QS receptor antagonists (tocopherols and phytol) calensis Georgi, and Phyllanthus amarus diterpene phytols have been
has also been reported in Diplocyclos palmatus leaf extract (Alexpandi reported to inhibit biofilm formation by P. aeruginosa, Pectobacterium
et al., 2019). The S. epidermidis biofilm inhibition efficacy of geraniol in carotovorum, and Vibrio campbellii (Kalia et al., 2019). The
rats was shown through oral supplementation at a rate of 200 mg/kg indole-mediated inhibition of QS-regulated biofilm formation, EPS
body weight for 3 days (Arunachalam et al., 2018). Selenium NPs, production, and motility is owing to its effects on the three-channel QSS.
intracellular drug delivery agents, and polyphenols present in honey Microalgae are a source of auxins such as indole-3-acetic acid and
(HP) have shown superior QSI activity, targeting the LasR receptor. The -acetamide. These biomolecules significantly reduce the pathogenicity
biofilm-forming ability of P. aeruginosa PAO1 was significantly reduced of V. campbellii. This leads to an enhancement in the survival rate of
by > 90%. Polyphenols of honey alone were ineffective at HP MIC 5% Macrobrachium rosenbergii, a giant river prawn (Yang et al., 2017). Dioon
and HP Sub-MIC 0.6% (Prateeksha et al., 2017). Phytochemicals, such as spinulosum extract against P. aeruginosa downregulated the expression of
garlic extract, act as QSIs by competitively blocking the binding of QS the QS genes lasI, lasR, rhlI, rhlR, and ndvB, which resulted in the inhi­
signal molecules to transcriptional regulators such as LuxR and LasR in bition of EPS production up to 51.4% and biofilm formation up to
P. aeruginosa (Mishra et al., 2020). Indole and its derivatives as QS signal 77.1%. This process has applications in treating wounds and respiratory
analogs exhibit a dose-dependent QSI effect, suppressing biofilm for­ and UTIs (Elekhnawy et al., 2022).
mation, prodigiosin production, and motility at a MIC over a range of
2.5–5.0 mM. Indole derivatives cause 40–75% inhibition of biofilm 3.4. Anti-c-di-GMP-mediated biofilm formation
formation by S. marcescens (Sethupathy et al., 2020).
Targeting the c-di-GMP signaling system is a promising strategy for
3.3.4. Inhibiting gene transcription developing antimicrobial drugs, primarily due to its effect on critical
The targeting of gene expression related to biofilm formation and bacterial lifestyle features without affecting their growth. Recent efforts
pathogenicity by QSIs can help in treating infectious pathogens. Among have focused on targeting bacterial pathogenicity and biofilm forma­
a wide range of biofilm inhibitors, cyclodextrins (CDs) (e.g., α- and tion. Another feature that favors c-di-GMP as a drug target is its absence
β-CDs) and their derivatives (methylated, quaternary amino, and poly­ in eukaryotes, reducing any potential threat to the infected host (Val­
mer) act as QSIs. Several studies have shown that QSI is induced by CD- entini and Filloux, 2019). Non-antimicrobial agents, including small
encapsulated QS signals, preventing binding to their cognate receptors. molecules, could inhibit the c-di-AMP-mediated transition of planktonic
It thus promotes the inhibition of QS-mediated pathogenicity, including to sessile lifestyles (Jenal and Dorman, 2009; Römling and Balsalobre,
biofilm formation. Synthetic CD derivatives (6-alkylamino-α- or γ-CDs, 2012). Recent works have focused on screening different compounds to
2-alkylamino-CDs, and 6,6’-dioctylamino-β-CD) act as inhibitors of QS- inhibit the functioning of c-di-GMP signaling. Thus, the inhibition of
mediated elastase production, a key regulator of antibiotic-resistant biofilm formation and virulence factors were observed by (i) inhibiting
biofilm produced by P. aeruginosa PAO1, violacein pigment production c-di-GMP synthesis through (a) DI-3 (N-4-anilinophenyl-benzamide) in
of Chromobacterium violaceum, and the prodigiosin production of P. aeruginosa and Vibrio cholerae and (b) DI-8 (N-2-henylethyl-amino­
S. marcescens (Morohoshi et al., 2013). Hydroxypropyl-β-CDs have been carbonothioyl-benzamide) in V. cholerae (Sambanthamoorthy et al.,
shown to reduce the ability of P. aeruginosa outer-membrane vesicles, 2012); (ii) reducing global levels of c-diGMP using (a) azathiopine in
which fuse with the lipid rafts of epithelial cells, to reduce CFTR Cl- E. coli and P. aeruginosa (Antoniani et al., 2013), (b) 6-mercaptopurine in
secretion. Hydroxypropyl-β-cyclodextrin (HPβCD) or methyl-­ E. coli (Migliore et al., 2018), and (c) terrein in P. aeruginosa (Kim et al.,
β-cyclodextrin (MβCD) are equally effective in suppressing planktonic 2018); (iii) competing with c-di-GMP for its binding sites or its seques­
growth and the biofilm-forming ability of P. aeruginosa (Barnaby et al., tration of (a) thiol-benzo-triazolo-quinazolinones in P. aeruginosa (Zhou
2019). α-CD and its methylated form can inhibit 90–98% of the et al., 2013), (b) ebselen in P. aeruginosa (Lieberman et al., 2014), and
QS-mediated biofilm formation by P. aeruginosa. Changes in tempera­ (c) proline-rich tetrapeptide Gup-Gup-Nap-Arg in P. aeruginoisa (Foletti
ture were found to reduce the ability of α-CD at 0.1 mM from 78% at et al., 2018). Certain peptides structurally resemble effector proteins,
30 ◦ C to non-significant levels at 22 ◦ C after 6 h (Berkl et al., 2022). In sequestering c-di-GMP, which inhibits biofilm formation by

10
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

P. aeruginosa (Hee et al., 2020). Diguanylate cyclases and degradative preformed biofilms by 52.69%, 59.79%, and 65% in E. coli, S. aureus,
phosphodiesterases also regulate the levels of c-di-GMP. Raffinose, a and P. aeruginosa, respectively (Husain et al., 2022). Selenium NPs as
galactotrisaccharide, inhibits biofilm matrix formation by enhancing the intracellular drug delivery agents have shown superior QSI activity,
activity of c-di-GMP-specific phosphodiesterase, thereby reducing the targeting the LasR receptor. Biofilm formation was inhibited > 90% at
concentration of c-di-GMP (Kim et al., 2016). Agents that reduce 4.5 μg/mL SeNPs@HP (Prateeksha et al., 2017). AgNPs coated with
c-di-GMP levels by activating esterases or inhibiting diguanylate cy­ thioether-bridged mesoporous organo-silica bind to ceftazidime, and
clases can be envisaged as potential mechanisms to counter biofilm alginate lyase causes degradation of P. aeruginosa biofilms. In mouse
formation. Diguanylate cyclase inhibitors include D-di-GMP and GTP. A lungs the eradication of P. aeruginosa PAO1 was observed to be effective
major limitation of this approach is the high variability between the two with nanocomposites, via two routes (i) 68% and 85% inhibition and (ii)
enzymes (Cho et al., 2020). Anti-c-di-GMP peptides (YfiN interacting 47% and 72a% with Ag@MON-CE (NPs decorated with PEI (poly­
peptides) have been identified in lytic bacteriophage PB1. These bind to etherimide) loaded with ceftazidime) and Ag@MON-AE (NPs decorated
the diguanylate cyclase YfiN of P. aeruginosa and negatively impact the with alginate lyase and ceftazidime), respectively (Wan et al., 2020). Ag
functioning of c-di-GMP signaling, interfering with bacterial motility and ZnO NPs altered biofilm formation and iron homeostasis in P. aer­
and biofilm formation (De Smet et al., 2021). uginosa. AgNPs resulted in increased modifications in cell viability and
Reactive Oxygen Species production, leading to significantly high
3.5. Other approaches disruption of the biofilm formation process. Cell viability decreased by
90% at 50 mg/L and 99.7% at 100 mg/L of ZnO NPs, whereas Ag NPs
3.5.1. Bacteriophages did not show any significant changes. The impact on eDNA and carbo­
BPs prevent biofilm formation by gram-negative and gram-positive hydrate content of the biofilm matrix decreased by 32.82% and 59,92%
pathogenic bacteria. Screening of BP lysin CF-301 revealed that com­ in the presence of 1 mg/L Ag NPs, respectively. These changes disturbed
plete eradication of S. aureus biofilms formed on the surfaces of catheters the biofilm architecture (de Celis et al., 2022). Phloroglucinol encap­
and surgical meshes could be observed at ≤ 0.25 μg/mL within 1 h sulated in chitosan NPs effectively inhibited biofilms formed by mono-
(Schuch et al., 2017). Antibiotic-resistant Vibrio alginolyticus was and dual-species of S. aureus, S. mutans, and K. pneumoniae. MIC values
inhibited in the planktonic state by BP pVa-21 (Myoviridae). However, of PG-CSNPs (10 mg/mL) against planktonic cells of S. aureus, S.
up to 5 h is required to eliminate the bacteria present within the biofilm mutans, and K. pneumoniae were found to be 2048 µg/mL, whereas the
(Kim et al., 2019). Biofilm formation by E. faecalis EF54 on natural tooth C. albicans was found to be higher than 2048 µg/mL. Sub-minimal
roots was cleared by BP application (SHEF2). Furthermore, the survival inhibitory concentrations (sub- MIC) of PG-CSNPs against preformed
rate of zebrafish infected with E. faecalis strain OS16 was improved by mono/dual species biofilms was 256–2048 µg/mL Mature biofilm
up to 84% (Al-Zubidi et al., 2019). Endolysin from BP vB_EfaS_PHB08 formed by mono/dual species had an ½ BMIC80 AntiMicrobial
eradicated biofilms by reducing E. faecalis by 1 × 105 CFU. Thus, BPs + PGCSNPs of 1024 µg/mL (Khan et al., 2022). AgNPs synthesized using
can treat pathogenic infections in wounds and the urinary tract (Yang an extract of Bothriochloa laguroides proved inhibitory to the
et al., 2020). The efficacy of BPs can be enhanced in combination with biofilm-forming abilities of S. aureus strains MRSA and MSSA (94%) and
antibiotics; biofilms caused by MSSA and P. aeruginosa in patients with Yersinia enterocolitica strains 8081 (91%) and ME110 (96%). An addi­
multi-organ failure on ventricular assistance are highly susceptible to tional advantage is their potential to eradicate mature biofilms by 99%
lytic BPs in conjunction with antibiotics. This has enabled successful for strain 8081, 94% for ME110 and 87% for MRSA and 82% for MSSA
heart transplantations (Aslam et al., 2019). The lytic phage (Toranzo et al., 2022). Niosome NP-encapsulated imipenem affects the
ɸWL-3/fosfomycin combination was effective against sensitivity and biofilms formed by S. epidermidis by reducing the
ciprofloxacin-resistant E. coli biofilms (Wang et al., 2020). The intra­ expression of icaD, FnbA, and EbpS (Piri-Gharaghie et al., 2022). The
operative administration of BP cocktails along with antibiotics stopped essential oils at 200 mg/mL had strong antimicrobial activity against
the recurrence of infections in patients with musculoskeletal infections P. mirabilis. Here, 92% deactivated survival rate with the Solanum nigrum
(Onsea et al., 2019). Recurrent urinary tract infections caused by the essential oils was recorded (Khaled et al., 2021). Inhibition of
XDR K. pneumoniae strain CX10301 were successfully treated with a A. baumannii biofilms was achieved with chitosan NPs loaded with
cocktail of lytic BPs and antibiotics (sulfamethoxazole-trimethoprim) essential oil (150 µg/mL) (Govindan et al., 2022). Encapsulation of
(Bao et al., 2020). Colonization of the gut by E. coli strain ST131-H30R alizarin liposomes in chitosan-gum arabic nanocarriers resulted in an
was successfully reduced by a cocktail of BPs and E. coli Nissle-1917, improved kinetic release mechanism. It efficiently inhibits 95% of the
which produces microcin C7 (Porter et al., 2022). A cocktail of lytic biofilms formation by S. aureus (MRSA) MW2 and Candida albicans
BPs was used in phase I/II clinical trials to treat P. aeruginosa infections DAY185 and ATCC10231 at CGL-Alz NCs (Alizarin: 50 μg/mL). The
in patients with burns. The trial was terminated prematurely because of effect was evident in mono- and dual species biofilms and cell aggre­
the poor efficacy of PP1131 (Jault et al., 2019). gation by C. albicans (Raj et al., 2022). The critical characteristics that
regulate the efficacy of NPs are their size, shape, surface charge, and
3.5.2. Nanoparticles hydrophobicity. In contrast, the biofilm features that influence the ef­
A significant limitation of the use of antibiotics to treat pathogenic ficiency of NPs are EPS matrix viscosity, compactness, liquid flow, and
bacterial biofilms is their burst release. NPs have proven effective in other interactions (Robino and Scavone, 2020).
achieving a sustained release of antibiotics (Al-Wrafy et al., 2022). The
primary advantage of NPs as carriers is targeted drug delivery. Coating 4. Stumbling blocks
NPs with antibiotics enhances their stability and antimicrobial potency.
Enzymatic biofilm degradation can be achieved by targeting matrix Despite enormous research efforts to treat infectious diseases, there
components of the matrixome (Karygianni et al., 2020a; b). Coating of are no assured treatment options. Antibiofilm agents that target their
biopolymer NPs with amino-cellulose and hyaluronic acid inhibited formation and stability have been evaluated, and very few products have
biofilm formation by up to 40% in E. coli and 94% in the case of S. aureus been tested in clinical trials (Lu et al., 2022). There is a paucity of in vitro
(Ivanova et al., 2018) (Table 3). Multimetallic (ZnO:MgO) NPs are an studies on multi-species biofilms. Few studies have evaluated cytotox­
advancement for disrupting P. mirabilis cell membranes. These NPs icity toward the host (Fleming and Rumbaugh, 2017). Another bacterial
cause protein dysfunction and degrade biofilm DNA (Basavegowda and characteristic that needs to be considered is their ability to switch back
Baek, 2021). Multi-functional ZnO-NPs at ½ × MIC of Zn-NPs produced to free-living planktonic entities owing to active self-dispersal. As
using phytocompounds (from Plumbago zeylanica) inhibited biofilm dispersed cells are more lethal than planktonic cells, it is important to
formation by 60%, 70%, and 77.69%, and could also eradicate the manage them post-biofilm disintegration. Thus, antibiofilm agents may

11
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Table 3
Nanoparticles as therapeutic agents against biofilm-forming microbial pathogens.
Pathogen Inhibitor Support Mechanism Inhibitor concentration Applications Reference
(Source) (EC: Effective; AC: Applied)

Staphylococcus Amino-cellulose Nanoparticles Interacts with EC: 4 × 1010 NPs/mL Inhibit biofilm Ivanova et al.
aureus, and hyaluronic (NPs) phospholipids of the AC: 1 × 1010, 2 × 1010, and formation in E. coli (2018)
Escherichia coli acid (Biocompatible bacterial cell membrane 4 × 1010 NPs/mL (40%) and S. aureus
polymer) (94%). It did not
influence the human cell
viability
S. aureus, E. coli, Plumbago Zinc oxide (ZnO)- Effects EC: ½ × MIC of Zn-NPs Inhibit biofilm Husain et al.
Pseudomonas zeylanica NPs Exopolysaccharide AC:1⁄₁₆, 1/8 and ¼ × MIC of ZnO- formation (up to 77%) (2022)
aeruginosa (extract) production NPs, and eliminate
MIC: E. coli, and S. aureus 200 µg/ preformed biofilms (up
mL, to 65%
P. aeruginosa 400 µg/mL
S. aureus, S. mutans, Phloroglucinol Chitosan NPs Interact with the EC: PG-CSNPs at 1024 μg/mL for Promotes antibacterial Khan et al.
Klebsiella negatively charged sugar dual sp. biofilm (BF); 256 μg/mL activity 86–92% BF (2022)
pneumoniae, polymers and eDNA of against C. albicans; 512 μg/mL inhibition. Prevent the
Candida albicans the ‘matrixome’ against S. mutans and S. aureus; spread of the epidemic
1024 μg/mL against Klebsiella and treat mixed-species
pneumoniae biofilms
AC: Preformed BF: PG-CSNPs
(10 mg/mL)
Sub-MIC 256–2048 µg/mL
Mature BF: ½ BMIC80
Antimicrobial + PGCSNPs
1024 µg/mL
S. aureus, Yersinia Bothriochloa Gold NPs (AgNPs) Inhibited planktonic and Biofilm inhibition: Potential for developing Toranzo et al.
enterocolitica laguroides sessile growth EC: 125 pmol/L (8 × MIC) for a new class of (2022)
(extract) Y. enterocolitica strains; 250 pmol/ antibacterial agents
L for S. aureus strains
Mature biofilm eradication:
EC: 500 pmol/L (30 × MIC) for
Y. enterocolitica strains; 250 pmol/
L for S. aureus strains
AC: 16 pmol/L for Y. enterica and
64–125 pmol/L for S. aureus
S. aureus, C. Alizarin Gum arabic Negatively charged gum Biofilm inhibition (Mono/Dual): Inhibit biofilm Raj et al. (2022)
albicans chitosan Arabic with improved EC: CGL-Alz NCs (Alizarin: 50 μg/ formation of
kinetic release mL). multispecies pathogens
AC: MIC: C. albicans 400 μg/mL,
and S. aureus 600 μg/mL; CGL-Alz
NCs (Alizarin: 0–50 μg/mL).
P. aeruginosa Polyphenols from Selenium NPs Affect QS-mediated Biofilm formation Anti-virulence activities Prateeksha
honey pathogenicity EC: 4.5 μg/mL, SeNPs@HP, (in-vitro and in-vivo) et al. (2017)
AC: Chromobacterium violaceum
CV12472 − 10 μg/mL; P.
aeruginosa PAO1–15 μg/mL
P. aeruginosa Thioether- Silver NPs (AgNPs) Binding to ceftazidime Biofilm formation and Inhibition of biofilm Wan et al.
bridged and alginate lyase Degradation: formation and rapid (2020)
mesoporous EC: ceftazidime: 0.656 mg/mL degradation of biofilms
organo-silica Ag@MON-CE/AE - 1.72 mg/mL in mouse lungs.
AC: ceftazidime: 0.27–0.656 mg/ Reduced lung injuries
mL and no deaths or serious
Ag@MON-CE/AE - 0.07, 0.14, side effects
0.21, 0.43, 0.86, and 1.72 mg/mL
P. aeruginosa. NPs Ag- and ZnO-NPs Effect growth, biofilm EC: Cell viability at ZnO 100 mg/ Higher disruption in de Celis et al.
formation, ROS L; biofilm formation (2022)
production, iron eDNA and carbohydrates by 1 mg/
homeostasis, eDNA L Ag NPs.
AC: Ag (1 and 5 mg/L) and ZnO
(50 and 100 mg/L) NPs
Proteus mirabilis NPs ZnO:MgO-NPs Disrupt cell membranes; EC: No specific information Biofilm degradation Basavegowda
protein dysfunction and AC: No specific information and Baek
degrade biofilm DNA (2021)
Acinetobacter Plant essential Chitosan NPs Effective removal of EC: 150 µg/mL Effective against Govindan et al.
baumannii oils biofilm formation AC: 150 µg/mL nosocomial and urinary (2022)
tract infections
P. mirabilis Essential oils None Effect survival and EC: 200 µg/mL Active against urinary Khaled et al.
(Solanum nigrum) virulence factors AC: 25–200 µg/mL tract infectious bacteria (2021)

PG-CSNPs: phloroglucinol-chitosan nanoparticles; sub-MIC: Sub-minimal inhibitory concentrations.


Ag@MON-AE: NPs decorated with alginate lyase and ceftazidime, while Ag@MON-CE: Ag@MON-PEI (polyetherimide) loaded with ceftazidime

12
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

be supplemented with other bioactive molecules such as nitrous oxide Al-Zubidi, M., Widziolek, M., Court, E.K., Gains, A.F., Smith, R.E., Ansbro, K.,
Alrafaie, A., Evans, C., Murdoch, C., Mesnage, S., Douglas, C.W., Rawlinson, A.,
(from sodium nitroprusside) (Uppuluri and Lopez-Ribot, 2016). The use
Stafford, G.P., 2019. Identification of novel bacteriophages with therapeutic
of QSIs as antipathogens interferes with biofilm formation and patho­ potential that target Enterococcus faecalis. Infect. Immun. 87, e00512–e00519.
genicity without killing the pathogens. Therefore, free-living cells can be https://doi.org/10.1128/IAI.00512-19.
exposed to QSIs along with low doses of antibiotics, reducing the risk of An, A.Y., Choi, K.Y.G., Baghela, A.S., Hancock, R.E.W., 2021. An overview of biological
and computational methods for designing mechanism-informed anti-biofilm agents.
the emergence of drug-resistant microbes (Kalia et al., 2019). The Front. Microbiol. 12, 640787 https://doi.org/10.3389/fmicb.2021.640787.
integration of biological and in silico techniques may facilitate the future Angelin, J., Kavitha, M., 2020. Exopolysaccharides from probiotic bacteria and their
design of antibiofilm agents (An et al., 2021). health potential. Int. J. Biol. Macromol. 162, 853–865. https://doi.org/10.1016/j.
ijbiomac.2020.06.190.
Antoniani, D., Rossi, E., Rinaldo, S., Bocci, P., Lolicato, M., Paiardini, A., Raffaelli, N.,
5. Conclusions Cutruzzola, F., Landini, P., 2013. The immunosuppressive drug azathioprine inhibits
biosynthesis of the bacterial signal molecule cyclic-di-GMP by interfering with
intracellular nucleotide pool availability. Appl. Microbiol. Biotechnol. 97,
Targeting the biofilm formation process is more desirable than tar­ 7325–7336. https://doi.org/10.1007/s00253-013-4875-0.
geting fully formed biofilms for the treatment of infectious diseases. This Arenas, J., Tommassen, J., 2017. Meningococcal biofilm formation: Let’s stick together.
scenario is more complex than is envisaged. Coating medical equipment Trends Microbiol 25, 113–124. https://doi.org/10.1016/j.tim.2016.09.005.
Armbruster, C.R., Parsek, M.R., 2018. New insight into the early stages of biofilm
with biomolecules, including QSIs, that restrict bacterial adhesion, formation. Proc. Natl. Acad. Sci. USA 115, 4317–4319. https://doi.org/10.1073/
biofilm formation, and disruption of mature biofilms has been proposed. pnas.1804084115.
Antibiofilm-forming agents have gained importance because of their Arunachalam, K., Ramar, M., Ramanathan, S., Govindaraju, A., Karutha Pandian, S.,
Kandasamy, R., Ravi, A.V., 2018. In vivo protective effect of geraniol on colonization
wide range of applications in aquaculture, water purification, waste­
of Staphylococcus epidermidis in rat jugular vein catheter model. Pathog. Dis. 76,
water treatment, shipping, agriculture, and the maintenance of fty055. https://doi.org/10.1093/femspd/fty055.
archaeological monuments. Despite the promising discovery of anti­ Asadi, A., Razavi, S., Talebi, M., Gholami, M., 2019. A review on anti-adhesion therapies
of bacterial diseases. Infection 47, 13–23. https://doi.org/10.1007/s15010-018-
biofilm agents, tremendous efforts are required to translate their po­
1222-5.
tential for the treatment of infectious diseases. Aslam, S., Pretorius, V., Lehman, S.M., Morales, S., Schooley, R.T., 2019. Novel
bacteriophage therapy for treatment of left ventricular assist device infection.
CRediT authorship contribution statement J. Heart Lung Transplant. 38, 475–476. https://doi.org/10.1016/j.
healun.2019.01.001.
Baik, D., Kim, B.-W., Ki, M., 2022. Increasing trends in mortality and costs of infectious
Vipin Chandra Kalia: Conceptualization, Writing – original draft diseases in Korea: trends in mortality and costs of infectious diseases. Epidemiol.
preparation, Visualization, Writing – review & editing. Sanjay Kumar Health 44, e2022010. https://doi.org/10.4178/epih.e2022010.
Baker, R.E., Mahmud, A.S., Miller, I.F., Rajeev, M., Rasambainarivo, F., Rice, B.L.,
Singh Patel: Writing – original draft preparation. Jung-Kul Lee: Writing – Takahashi, S., Tatem, A.J., Wagner, C.E., Wang, L.-F., Wesolowski, A., Metcalf, C.J.
review & editing, Resources, Funding acquisition, Supervision, Project E., 2022. Infectious disease in an era of global change. Nat. Rev. Microbiol. 20,
administration. 193–205. https://doi.org/10.1038/s41579-021-00639-z.
Banar, M., Emaneini, M., Satarzadeh, M., Abdellahi, N., Beigverdi, R., van Leeuwen, W.
B., Jabalameli, F., 2016. Evaluation of mannosidase and trypsin enzymes effects on
Declaration of Competing Interest biofilm production of Pseudomonas aeruginosa isolated from burn wound infections.
PLoS One 11, e0164622 https://doi.org/10.1371/journal.pone.0164622.
Bansal, M., Dhowlaghar, N., Nannapaneni, R., Kode, D., Chang, S., Sharma, C.S.,
The authors declare that they have no known competing financial McDaniel, C., Kiess, A., 2021. Decreased biofilm formation by planktonic cells of
interests or personal relationships that could have appeared to influence Listeria monocytogenes in the presence of sodium hypochlorite. Food Microbiol 96,
the work reported in this paper. 103714. https://doi.org/10.1016/j.fm.2020.103714.
Bao, J., Wu, N., Zeng, Y., Chen, L., Li, L., Yang, L., et al., 2020. Non-active antibiotic and
bacteriophage synergism to successfully treat recurrent urinary tract infection
Data Availability caused by extensively drug-resistant Klebsiella pneumoniae. Emerg. Microbes Infect.
9, 771–774. https://doi.org/10.1080/22221751.2020.1747950.
Barnaby, R., Koeppen, K., Stanton, B.A., 2019. Cyclodextrins reduce the ability of
I have mentioned all the references.
Pseudomonas aeruginosa outer-membrane vesicles to reduce CFTR Cl_ secretion. Am.
J. Physiol. Lung Cell. Mol. Physiol. 316, L206–L215. https://doi.org/10.1152/
Acknowledgements ajplung.00316.2018.
Basavegowda, N., Baek, K.-H., 2021. Multimetallic nanoparticles as alternative
antimicrobial agents: Challenges and perspectives. Molecules 26, 912. https://doi.
This research was supported by the Basic Science Research Program org/10.3390/molecules26040912.
through the National Research Foundation of Korea (NRF), funded by Basu, R.A., Sauer, K., 2014. Diguanylate cyclase NicD-based signalling mechanism of
the Ministry of Science, ICT & Future Planning (NRF-2022M3A9I30823 nutrient-induced dispersion by Pseudomonas aeruginosa. Mol. Microbiol. 94,
771–793. https://doi.org/10.1111/mmi.12802.
66, NRF-2021R1A2B5B03002728, and 2022M3A9I5015091). The Berkl, Z., Fekete-Kertész, I., Buda, K., Vaszita, E., Fenyvesi, É., Szente, L., Molnár, M.,
sponsor(s) had no role in the study design; in the collection, analysis, 2022. Effect of cyclodextrins on the biofilm formation capacity of Pseudomonas
and interpretation of data; in the writing of the report; and in the de­ aeruginosa PAO1. Molecules 27, 3603. https://doi.org/10.3390/
molecules27113603.
cision to submit the article for publication. Besinis, A., Hadi, S.D., Le, H.R., Tredwin, C., Handy, R.D., 2017. Antibacterial activity
and biofilm inhibition by surface modified titanium alloy medical implants following
References application of silver, titanium dioxide and hydroxyapatite nanocoatings.
Nanotoxicology 11, 327–338. https://doi.org/10.1080/17435390.2017.1299890.
Bhat, S.V., Kamencic, B., Körnig, A., Shahina, Z., Dahms, T.E.S., 2018. Exposure to sub-
Abdel-Mawgoud, A.M., Lépine, F., Déziel, E., 2010. Rhamnolipids: diversity of structures,
lethal 2.,4-Dichlorophenoxyacetic acid arrests cell division and alters cell surface
microbial origins and roles. Appl. Microbiol. Biotechnol. 86, 1323–1336. https://doi.
properties in Escherichia coli. Front. Microbiol. 9, 44. https://doi.org/10.3389/
org/10.1007/s00253-010-2498-2.
fmicb.2018.00044.
Adnan, M., Patel, M., Deshpande, S., Alreshidi, M., Siddiqui, A.J., Reddy, M.N.,
Bjarnsholt, T., 2013. The role of bacterial biofilms in chronic infections. APMIS 121,
Emira, N., Feo, V.N., 2020. Effect of Adiantum philippense extract on biofilm
1–58. https://doi.org/10.1111/apm.12099.
formation., adhesion with its antibacterial activities against foodborne pathogens.,
Bloom, D.E., Cadratte, D., 2019. Infectious diseases threats in the twenty-first century:
and characterization of bioactive metabolites: an in vitro-in silico approach. Front.
strengthening the global response. Front Immunol. 20, 549. https://doi.org/
Microbiol. 11, 823. https://doi.org/10.3389/fmicb.2020.00823.
10.3389/fimmu.2019.00549.
Alexpandi, R., Prasanth, M.I., Ravi, A.V., Balamurugan, K., Durgadevi, R., Srinivasan, R.,
Borriello, G., Werner, E., Roe, F., Kim, A.M., Ehrlich, G.D., Stewart, P.S., 2004. Oxygen
de Mesquita, J.F., Karutha Pandian, S., 2019. Protective effect of neglected plant
limitation contributes to antibiotic tolerance of Pseudomonas aeruginosa in biofilms.
Diplocyclos palmatus on quorum sensing mediated infection of Serratia marcescens and
Antimicrob. Agents Chemother. 48, 2659–2664. https://doi.org/10.1128/
UV-A induced photoaging in model Caenorhabditis elegans. J. Photochem. Photobiol.
AAC.48.7.2659-2664.2004.
B Biol. 201, 111637 https://doi.org/10.1016/j.jphotobiol.2019.111637.
Brackman, G., Garcia-Fernandez, M.J., Lenoir, J., De Meyer, L., Remon, J.P., De Beer, T.,
Al-Wrafy, F.A., Al-Gheethi, A.A., Ponnusamy, S.K., Noman, E.A., Fattah, S.A., 2022.
Concheiro, A., Alvarez-Lorenzo, C., Coenye, T., 2016. Dressings loaded with
Nanoparticles approach to eradicate bacterial biofilm-related infections: a critical
cyclodextrin-hamamelitannin complexes increase Staphylococcus aureus
review. Chemosphere 288, 132603. https://doi.org/10.1016/j.
susceptibility toward antibiotics both in single as well as in mixed biofilm
chemosphere.2021.132603.

13
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

communities. Macromol. Biosci. 16, 859–869. https://doi.org/10.1002/ evolutionarily robust anti-biofilm strategy. Nat. Commun. 11, 107. https://doi.org/
mabi.201500437. 10.1038/s41467-019-13660-x.
Brandenburg, K.S., Rodriguez, K.J., McAnulty, J.F., Murphy, C.J., Abbott, N.L., Ding, T., Li, T., Wang, Z., Li, J., 2017. Curcumin liposomes interfere with quorum sensing
Schurr, M.J., Czuprynski, C.J., 2013. Tryptophan inhibits biofilm formation by system of Aeromonas sobria and in silico analysis. Sci. Rep. 7, 8612. https://doi.org/
Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 57, 1921–1925. https:// 10.1038/s41598-017-08986-9.
doi.org/10.1128/AAC.00007-13. Dong, X., Grenier, D., Wang, K., Wang, Y., 2021. Research progress of bacterial quorum
Brown, H.L., Reuter, M., Hanman, K., Betts, R.P., van Vliet, A.H., 2015. Prevention of sensing receptors: Classification, structure, function and characteristics. Sci. Total
biofilm formation and removal of existing biofilms by extracellular DNases of Environ. 763, 143031 https://doi.org/10.1016/j.scitotenv.2020.143031.
Campylobacter jejuni. PLoS One 10, e0121680. https://doi.org/10.1371/journal. Donlan, R.M., 2001. Biofilms and device-associated infections. Emerg. Infect. Dis. 7,
pone.0121680. 277–281. https://doi.org/10.3201/eid0702.010226.
de Celis, M., Belda, I., Marquina, D., Santos, A., 2022. Phenotypic and transcriptional Dufrêne, Y.F., Viljoen, A., 2020. Binding strength of gram-positive bacterial adhesins.
study of the antimicrobial activity of silver and zinc oxide nanoparticles on a Front. Microbiol. 11, 1457. https://doi.org/10.3389/fmicb.2020.01457.
wastewater biofilm-forming Pseudomonas aeruginosa strain. Sci. Total Environ. 826, Dusane, D.H., 2017. CORR insights®: does extracellular DNA production vary in
153915 https://doi.org/10.1016/j.scitotenv.2022.153915. staphylococcal biofilms isolated from infected implants versus controls? Clin.
Chen, M., Yu, Q., Sun, H., 2013. Novel strategies for the prevention and treatment of Orthop. Relat. Res. 475, 2114–2116. https://doi.org/10.1007/s11999-017-5360-3.
biofilm related infections. Int. J. Mol. Sci. 14, 18488–18501. https://doi.org/ Eckhart, L., Fischer, H., Barken, K.B., Tolker-Nielsen, T., Tschachler, E., 2007. DNase1L2
10.3390/ijms140918488. suppresses biofilm formation by Pseudomonas aeruginosa and Staphylococcus aureus.
Cheng, W.-J., Zhou, J.-W., Zhang, P.-P., Luo, H.-Z., Tang, S., Li, J.-J., Deng, S.-M., Jia, A.- Br. J. Dermatol. 156, 1342–1345. https://doi.org/10.1111/j.1365-
Q., 2020. Quorum sensing inhibition and tobramycin acceleration in 2133.2007.07886.x.
Chromobacterium violaceum by two natural cinnamic acid derivatives. Appl. El Aichar, F., Muras, A., Parga, A., Otero, A., Nateche, F., 2022. Quorum quenching and
Microbiol. Biotechnol. 104, 5025–5037. https://doi.org/10.1007/s00253-020- anti-biofilm activities of halotolerant Bacillus strains isolated in different
10593-10590. environments in Algeria. J. Appl. Microbiol. 132, 1825–1839. https://doi.org/
Chernousova, S., Epple, M., 2013. Silver as antibacterial agent: ion, nanoparticle, and 10.1111/jam.15355.
metal. Angew. Chem. Int. Ed. 52, 1636–1653. https://doi.org/10.1002/ Elekhnawy, E., Negm, W.A., El-Aasr, M., Kamer, A.A., Alqarni, M., El-Saber Batiha, G.,
anie.201205923. Obaidullah, A.J., Fawzy, H.M., 2022. Histological assessment, anti-quorum sensing,
Cho, K.H., Tryon, R.G., Kim, J.-H., 2020. Screening for diguanylate cyclase (DGC) and anti-biofilm activities of Dioon spinulosum extract: in vitro and in vivo approach.
inhibitors mitigating bacterial biofilm formation. Front. Chem. 8, 264. https://doi. Sci. Rep. 12, 180. https://doi.org/10.1038/s41598-021-03953-x.
org/10.3389/fchem.2020.00264. Falcone, M., Ferrara, S., Rossi, E., Johansen, H.K., Molin, S., Bertoni, G., 2018. The small
Chow, J.Y., Yang, Y., Tay, S.B., Chua, K.L., Yew, W.S., 2014. Disruption of biofilm RNA ErsA of Pseudomonas aeruginosa contributes to biofilm development and
formation by the human pathogen Acinetobacter baumannii using engineered motility through post-transcriptional modulation of AmrZ. Front. Microbiol. 9, 238.
quorum-quenching lactonases. Antimicrob. Agents Chemother. 58, 1802–1805. https://doi.org/10.3389/fmicb.2018.00238.
https://doi.org/10.1128/AAC.02410-13. Fernández, L., Breidenstein, E.B.M., Taylor, P.K., Bains, M., 2016. Interconnection of
Christen, M., Kulasekara, H.D., Christen, B., Kulasekara, B.R., Hoffman, L.R., Miller, S.I., post- transcriptional regulation: the RNA-binding protein Hfq is a novel target of the
2010. Asymmetrical distribution of the second messenger c-di-GMP upon bacterial Lon protease in Pseudomonas aeruginosa. Sci. Rep. 6, 26811. https://doi.org/
cell division. Science 328, 1295–1297. https://doi.org/10.1126/science.1188658. 10.1038/srep26811.
Claessen, D., Rozen, D.E., Kuipers, O.P., Søgaard-Andersen, L., van Wezel, G.P., 2014. Ferrara, S., Carloni, S., Fulco, R., Falcone, M., Macchi, R., Bertoni, G., 2015. Post-
Bacterial solutions to multicellularity: a tale of biofilms, filaments and fruiting transcriptional regulation of the virulence-associated enzyme AlgC by the σ22-
bodies. Nat. Rev. Microbiol.. 12, 115–124. https://doi.org/10.1038/nrmicro3178. dependent small RNA ErsA of Pseudomonas aeruginosa. Environ. Microbiol. 17,
Coughlan, L.M., Cotter, P.D., Hill, C., Alvarez-Ordóñez, A., 2016. New weapons to fight 199–214. https://doi.org/10.1111/1462-2920.12590.
old enemies: Novel strategies for the (bio)control of bacterial biofilms in the food Ferrara, S., Rossi, A., Ranucci, S., De Fino, I., Bragonzi, A., Cigana, C., Bertoni, G., 2020.
industry. Front. Microbiol. 7, 1641. https://doi.org/10.1038/fmicb.2016.01641. The small RNA ErsA plays a role in the regulatory network of Pseudomonas aeruginosa
Cui, H., Ma, C., Lin, L., 2016. Co-loaded proteinase K/thyme oil liposomes for pathogenicity in airway infections. mSphere 5, e00909–e00920. https://doi.org/
inactivation of Escherichia coli O157:H7 biofilms on cucumber. Food Funct. 7, 10.1128/mSphere.00909-20.
4030–4040. https://doi.org/10.1039/c6fo01201a. Feuillie, C., Formosa-Dague, C., Hays, L.M., Vervaeck, O., Derclaye, S., Brennan, M.P.,
Darmani, H., Tawalbeh, K.H., Al-Hiyasat, A.S., Al-Akhras, M.-A., 2018. Comparison of Foster, T.J., Geoghegan, J.A., Dufrene, Y.F., 2017. Molecular interactions and
the photosensitivity of biofilms of different genera of cariogenic bacteria in tooth inhibition of the staphylococcal biofilm-forming protein SdrC. Proc. Natl. Acad. Sci.
slices. Pol. J. Microbiol. 67, 455. https://doi.org/10.21307/pjm-2018-053. USA 114, 3738–3743. https://doi.org/10.1073/pnas.1616805114.
Darouiche, R.O., Mansouri, M.D., Gawande, P.V., Madhyastha, S., 2009. Antimicrobial Fey, P.D., Olson, M.E., 2010. Current concepts in biofilm formation of Staphylococcus
and antibiofilm efficacy of triclosan and dispersin B combination. J. Antimicrob. epidermidis. Future Microbiol 5, 917–933. https://doi.org/10.2217/fmb.10.56.
Chemother. 64, 88–93. https://doi.org/10.1093/jac/dkp158. Fleming, D., Rumbaugh, K., 2017. Approaches to dispersing medical biofilms.
Das, T., Kutty, S.K., Kumar, N., Manefield, M., 2013. Pyocyanin facilitates extracellular Microorganisms 5, 15. https://doi.org/10.3390/microorganisms5020015.
DNA binding to Pseudomonas aeruginosa influencing cell surface properties and Flemming, H.C., Wingender, J., 2010. The biofilm matrix. Nat. Rev. Microbiol. 8,
aggregation. PLoS One 8, e58299. https://doi.org/10.1371/journal.pone.0058299. 623–633. https://doi.org/10.1038/nrmicro2415.
Das, T., Kutty, S.K., Tavallaie, R., Ibugo, A.I., Panchompoo, J., Sehar, S., Aldous, L., Flemming, H.-C., Wingender, J., Szewzyk, U., Steinberg, P., Rice, S.A., Kjelleberg, S.,
Yeung, A.W.S., Thomas, S.R., Kumar, N., Gooding, J.J., Manefield, M., 2015. 2016. Biofilms: an emergent form of bacterial life. Nat. Rev. Microbiol. 14, 563–575
Phenazine virulence factor binding to extracellular DNA is important for https://doi.org/10.1038.nrmicro.2016.94.
Pseudomonas aeruginosa biofilm formation. Sci. Rep. 5, 8398. https://doi.org/ Foletti, C., Kramer, R.A., Mauser, H., Jenal, U., Bleicher, K.H., Wennemers, H., 2018.
10.1038/srep08398. Functionalized proline-rich peptides bind the bacterial second messenger c-di-GMP.
Das, T., Simone, M., Ibugo, A.I., Witting, P.K., Manefield, M., Manos, J., 2017. Angew. Chem. Int. Ed. Engl. 57, 7729–7733. https://doi.org/10.1002/
Glutathione enhances antibiotic efficiency and effectiveness of DNase I in disrupting anie.201801845.
Pseudomonas aeruginosa biofilms while also inhibiting pyocyanin activity, thus Gandra, S., Barter, D.M., Laxminarayan, R., 2014. Economic burden of antibiotic
facilitating restoration of cell enzymatic activity, confluence and viability. Front. resistance: how much do we really know? Clin. Microbiol. Infect. 20, 973–979.
Microbiol. 8, 2429. https://doi.org/10.3389/fmicb.2017.02429. https://doi.org/10.1111/1469-0691.12798.
De la Fuente-Núñez, C., Reffuveille, F., Haney, E.F., Straus, S.K., Hancock, R.E., 2014. Ganin, H., Rayo, J., Amara, N., Levy, N., Krief, P., Meijler, M.M., 2013. Sulforaphane and
Broad-spectrum anti-biofilm peptide that targets a cellular stress response. PLoS erucin, natural isothiocyanates from broccoli, inhibit bacterial quorum sensing. Med.
Pathog. 10, e1004152 https://doi.org/10.1371/journal.ppat.1004152. Chem. Commun. 4, 175–179. https://doi.org/10.1039/C2MD20196H.
De Smet, J., Wagemans, J., Hendrix, H., Staes, I., Visnapuu, A., Horemans, B., Gholibegloo, E., Karbasi, A., Pourhajibagher, M., Chiniforush, N., Ramazani, A.,
Aertsen, A., Lavigne, R., 2021. Bacteriophage-mediated interference of the c-di-GMP Akbari, T., Bahdor, A., Khoobi, M., 2018. Carnosine-graphene oxide conjugates
signalling pathway in Pseudomonas aeruginosa. Microb. Biotechnol. 14, 967–978. decorated with hydroxyapatite as promising nanocarrier for ICG loading with
https://doi.org/10.1111/1751-7915.13728. enhanced antibacterial effects in photodynamic therapy against Streptococcus
Deng, B., Ghatak, S., Sarkar, S., Singh, K., Das Ghatak, P., Mathew-Steiner, S.S., Roy, S., mutans. J. Photochem. Photobiol. B Biol. 181, 14–22. https://doi.org/10.1016/j.
Khanna, S., Wozniak, D.J., McComb, D.W., Sen, C.K., 2020. Novel bacterial diversity jphotobiol.2018.02.004.
and fragmented eDNA identified in hyperbiofilm-forming Pseudomonas aeruginosa Gómez-Gómez, B., Arregui, L., Serrano, S., Santos, A., Perez-Corona, T., Madrid, Y.,
rugose small colony variant. iScience 23, 100827. https://doi.org/10.1016/j. 2019. Unravelling mechanisms of bacterial quorum sensing disruption by metal-
isci.2020.100827. based nanoparticles. Sci. Total Environ. 696, 133869 https://doi.org/10.1016/j.
Deng, Y., Lim, A., Lee, J., Chen, S., An, S., Dong, Y.H., Zhang, L.H., 2014. Diffusible scitotenv.2019.133869.
signal factor (DSF) quorum sensing signal and structurally related molecules Gómez-Lozano, M., Marvig, R.L., Molin, S., Long, K.S., 2012. Genome-wide identification
enhance the antimicrobial efficacy of antibiotics against some bacterial pathogens. of novel small RNAs in Pseudomonas aeruginosa. Environ. Microbiol 14, 2006–2016.
BMC Microbiol 14, 51. https://doi.org/10.1186/1471-2180-14-51. https://doi.org/10.1111/j.1462-2920.2012.02759.x.
Devaraj, A., Buzzo, J., Rocco, C.J., Bakaletz, L.O., Goodman, S.D., 2018. The DNABII Govindan, R., Chackaravarthi, G., Ramachandran, G., Chelliah, C.K., Muthuchamy, M.,
family of proteins is comprised of the only nucleoid associated proteins required for Quero, F., Mothana, R.A., Noman, O.M., Siddiqui, N.A., Li, W.-J., 2022. Effective
nontypeable Haemophilus influenzae biofilm structure. MicrobiologyOpen 7, e00563. removal of biofilm formation in Acinetobacter baumannii using chitosan nanoparticles
https://doi.org/10.1002/mbo3.563. loaded plant essential oils. J. King Saud. Univ. Sci. 34, 101845 https://doi.org/
Dieltjens, L., Appermans, K., Lissens, M., Lories, B., Kim, W., Van der Eycken, E.V., 10.1016/j.jksus.2022.101845.
Foster, K.R., Steenackers, H.P., 2020. Inhibiting bacterial cooperation is an Graf, A.C., Leonard, A., Schäuble, M., Rieckmann, L.M., Hoyer, J., Maass, S., Lalk, M.,
Becher, D., Pané-Farré, J., Riedel, K., 2019. Virulence factors produced by

14
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Staphylococcus aureus biofilms have a moonlighting function contributing to biofilm Kannappan, A., Gowrishankar, S., Srinivasan, R., Karutha Pandian, S., Ravi, A.V., 2017a.
integrity. Mol. Cell. Proteom. 18, 1036–1053. https://doi.org/10.1074/mcp. Antibiofilm activity of Vetiveria zizanioides root extract against methicillin-resistant
RA118.001120. Staphylococcus aureus. Microb. Pathog. 110, 313–324. https://doi.org/10.1016/j.
Gustafsson, E., Oscarsson, J., 2008. Maximal transcription of aur (aureolysin) and sspA micpath.2017.07.016.
(serine protease) in Staphylococcus aureus requires staphylococcal accessory Kannappan, A., Sivaranjani, M., Srinivasan, R., Rathna, J., Karutha Pandian, S., Ravi, A.
regulator R (sarR) activity. FEMS Microbiol. Lett. 284, 158–164. https://doi.org/ V., 2017b. Inhibitory efficacy of geraniol on biofilm formation and development of
10.1111/j.1574-6968.2008.01198.x. adaptive resistance in Staphylococcus epidermidis RP62A. J. Med. Microbiol 66,
Gutiérrez-Barranquero, J.A., Reen, F.J., McCarthy, R.R., O’Gara, F., 2015. Deciphering 1506–1515. https://doi.org/10.1099/jmm.0.000570.
the role of coumarin as a novel quorum sensing inhibitor suppressing virulence Kannappan, A., Balasubramaniam, B., Ranjitha, R., Srinivasan, R., Packiavathy, I.A.S.V.,
phenotypes in bacterial pathogens. Appl. Microbiol. Biotechnol. 99, 3303–3316. Balamurugan, K., Karutha Pandian, S., Ravi, A.V., 2019a. In vitro and in vivo biofilm
https://doi.org/10.1007/s00253-015-6436-1. inhibitory efficacy of geraniol-cefotaxime combination against Staphylococcus spp.
Haataja, S., Verma, P., Fu, O., Papageorgiou, A.C., Pöysti, S., Pieters, R.J., Nilsson, U.J., Food Chem. Toxicol. 125, 322–332. https://doi.org/10.1016/j.fct.2019.01.008.
Finne, J., 2018. Rationally designed chemically modified glycodendrimer inhibits Kannappan, A., Srinivasan, R., Nivetha, A., Annapoorani, A., Karutha Pandian, S.,
Streptococcus suis adhesin sadp at picomolar concentrations. Chemistry 24, Ravi, A.V., 2019b. Anti-virulence potential of 2-hydroxy-4- methoxybenzaldehyde
1905–1912. https://doi.org/10.1002/chem.201704493. against methicillin-resistant Staphylococcus aureus and its clinical isolates. Appl.
He, Z., Jiang, W., Jiang, Y., Dong, J., Song, Z., Xu, J., Zhou, W., 2022. Anti-biofilm Microbiol. Biotechnol. 103, 6747–6758. https://doi.org/10.1007/s00253-019-
activities of coumarin as quorum sensing inhibitor for Porphyromonas gingivalis. 09941-6.
J. Oral. Microbiol 14, 2055523. https://doi.org/10.1080/20002297.2022.2055523. Kannappan, A., Durgadevi, R., Srinivasan, R., Lagoa, R.J.L., Packiavathy, I.A.S.V.,
Hee, C.-S., Habazettl, J., Schmutz, C., Schirmer, T., Jenal, U., Grzesiek, S., 2020. Karutha Pandian, S., 2020. 2-Hydroxy-4-methoxybenzaldehyde from Hemidesmus
Intercepting second-messenger signaling by rationally designed peptides indicus is antagonistic to Staphylococcus epidermidis biofilm formation. Biofouling 36,
sequestering c-di-GMP. Proc. Natl. Acad. Sci. USA 117, 17211–17220. https://doi. 549–563. https://doi.org/10.1080/08927014.2020.1777989.
org/10.1073/pnas.2001232117. Kaplan, J.B., 2009. Therapeutic potential of biofilm-dispersing enzymes. Int. J. Artif.
Herman-Bausier, P., Valotteau, C., Pietrocola, G., Rindi, S., Alsteens, D., Foster, T.J., Organs 32, 545–554. https://doi.org/10.1177/039139880903200903.
Speziale, P., Dufrene, Y.F., 2016. Mechanical strength and inhibition of the Kaplan, J.B., LoVetri, K., Cardona, S.T., Madhyastha, S., Sadovskaya, I., Jabbouri, S.,
Staphylococcus aureus collagen-binding protein Cna. e01529–16 mBio 7. https://doi. Izano, E.A., 2012. Recombinant human DNase I decreases biofilm and increases
org/10.1128/mBio.01529-16. antimicrobial susceptibility in Staphylococci. J. Antibiot. 65, 73–77. https://doi.org/
Howlin, R.P., Cathie, K., Hall-Stoodley, L., Cornelius, V., Duignan, C., Allan, R.N., 10.1038/ja.2011.113.
Fernandez, B.O., Barraud, N., Bruce, K.D., Jefferies, J., et al., 2017. Low-dose nitric Karygianni, L., Ren, Z., Koo, H., Thumheer, T., 2020a. Biofilm matrixome: Extracellular
oxide as targeted anti-biofilm adjunctive therapy to treat chronic Pseudomonas components in structured microbial communities. Trends Microbiol 28, 668–681.
aeruginosa infection in cystic fibrosis. Mol. Ther. 25, 2104–2116. https://doi.org/ https://doi.org/10.1016/j.tim.2020.03.016.
10.1016/j.ymthe.2017.06.021. Karygianni, L., Attin, T., Thurnheer, T., 2020b. Combined DNase and proteinase
Hraiech, S., Hiblot, J., Lafleur, J., Lepidi, H., Papazian, L., Rolain, J.M., Raoult, D., treatment interferes with composition and structural integrity of multispecies oral
Elias, M., Silby, M.W., Bzdrenga, J., Bregeon, F., Chabriere, E., 2014. Inhaled biofilms. J. Clin. Med. 9, 983. https://doi.org/10.3390/jcm9040983.
lactonase reduces Pseudomonas aeruginosa quorum sensing and mortality in rat Kavanaugh, J.S., Flack, C.E., Lister, J., Ricker, E.B., Ibberson, C.B., Jenul, C.,
pneumonia. PLoS One 9, e107125. https://doi.org/10.1371/journal.pone.0107125. Moormeier, D.E., Delmain, E.A., Bayles, K.W., Horswill, A.R., 2019. Identification of
Husain, F.M., Qais, F.A., Ahmad, I., Hakeem, M.J., Baig, M.H., 2022. Biosynthesized zinc extracellular DNA-binding proteins in the biofilm matrix. e01137–19 mBio 10.
oxide nanoparticles disrupt established biofilms of pathogenic bacteria. Appl. Sci. https://doi.org/10.1128/mBio.01137-19.
12, 710. https://doi.org/10.3390/app12020710. Keeling, P.J., Palmer, J.D., 2008. Horizontal gene transfer in eukaryotic evolution. Nat.
Ibáñez de Aldecoa, A.L., Zafra, O., González-Pastor, J.E., 2017. Mechanisms and Rev. Genet. 9, 605–618. https://doi.org/10.1038/nrg2386.
regulation of extracellular DNA release and its biological roles in microbial Khabbaz, R., Bell, B.P., Schuchat, A., Ostroff, S.M., Moseley, R., Levitt, A., Hughes, J.M.,
communities. Front. Microbiol. 8, 1390. https://doi.org/10.3389/ (2015). Emerging and reemerging infectious disease threats. Mandell, Douglas, and
fmicb.2017.01390. Bennett’s Principles and Practice of Infectious Diseases, 158–177.e6. https://doi.
Ito, A., Taniuchi, A., May, T., Kawata, K., Okabe, S., 2009. Increased antibiotic resistance org/10.1016/B978–1-4557–4801-3.00014-X.
of Escherichia coli in mature biofilms. Appl. Environ. Microbiol. 75, 4093–4100. Khaled, J.M., Alyahya, S.A., Kanisha, C.C., Alharbi, N.S., Kadaikunnan, S.,
https://doi.org/10.1128/AEM.02949-08. Ramachandran, G., Alanzi, K.F., Rajivgandhi, G., Vimala, R.T.V., Manoharan, N.,
Ivanova, A., Ivanova, K., Hoyo, J., Heinze, T., Sanchez-Gomez, S., Tzanov, T., 2018. 2021. Anti-biofilm activity of Lc-MS based Solanum nigrum essential oils against
Layer-by-layer decorated nanoparticles with tunable antibacterial and antibiofilm multi drug resistant biofilm forming P. mirabilis. Saudi J. Biol. Sci. 28, 302–309.
properties against both Gram-positive and Gram-negative bacteria. ACS Appl. Mater. https://doi.org/10.1018/j.sjbs.2020.10.003.
Interfaces 10, 3314–3323. https://doi.org/10.1021/acsami.7b16508. Khan, F., Oh, D., Chandika, P., Jo, D.M., Bamunarachchi, N.I., Jung, W.K., Kim, Y.-M.,
Jain, N., Ådén, J., Nagamatsu, K., Evans, M.L., Li, X., McMichael, B., Ivanova, M.I., 2022. Inhibitory activities of phloroglucinol-chitosan nanoparticles on mono- and
Almqvist, F., Buxbaum, J.N., Chapman, M.R., 2017. Inhibition of curli assembly and dual-species biofilms of Candida albicans and bacteria. Colloids Surf. B Biointerfaces
E. coli biofilm formation by the human systemic amyloid precursor transthyretin. 211, 112307. https://doi.org/10.1016/j.colsurfb.2021.112307.
Proc. Natl. Acad. Sci. USA 1141, 12184–12189. https://doi.org/10.1073/ Kim, B., Park, J.S., Choi, H.Y., Yoon, S.S., Kim, W.G., 2018. Terrein is an inhibitor of
pnas.1708805114. quorum sensing and c-di-GMP in Pseudomonas aeruginosa: a connection between
Jault, P., Leclerc, T., Jennes, S., Pirnay, J.P., Que, Y.-A., Resch, G., Rousseau, A.F., quorum sensing and c-di-GMP. Sci. Rep. 8, 8617. https://doi.org/10.1038/s41598-
Ravat, F., Carsin, H., Floch, R.L., Schaal, J.V., Soler, C., Fevre, C., Arnaud, I., 018-26974-5.
Bretaudeau, L., Gabard, J., 2019. Efficacy and tolerability of a cocktail of Kim, H.-S., Cha, E., Kim, Y., Jeon, Y.H., Olson, B.H., Byun, Y., Park, H.-D., 2016.
bacteriophages to treat burn wounds infected by Pseudomonas aeruginosa Raffinose, a plant galactoside, inhibits Pseudomonas aeruginosa biofilm formation via
(PhagoBurn): a randomised, controlled, double-blind phase 1/2 trial. Lancet Infect. binding to LecA and decreasing cellular cyclic diguanylate levels. Sci. Rep. 6, 25318.
Dis. 19, 35–45. https://doi.org/10.1016/S1473-3099(18)30482-1. https://doi.org/10.1038/srep25318.
Jenal, U., Dorman, C.J., 2009. Small molecule signaling. Curr. Opin. Microbiol. 12, Kim, S.G., Jun, J.W., Giri, S.S., Yun, S., Kim, H.J., Kim, S.W., Kang, J.W., Han, S.J.,
125–128. https://doi.org/10.1016/jmib.2009.02.002. Jeong, D., Park, S.C., 2019. Isolation and characterization of pVa-21, a giant
Johnson, L., Horsman, S.R., Charron-Mazenod, L., Turnbull, A.L., Mulcahy, H., bacteriophage with anti-biofilm potential against Vibrio alginolyticus. Sci. Rep. 9,
Surette, M.G., Lewenza, S., 2013. Extracellular DNA-induced antimicrobial peptide 6284. https://doi.org/10.1038/s41598-019-42681-1.
resistance in Salmonella enterica serovar Typhimurium. BMC Microbiol. 13, 115. Kim, S.-H., Park, C., Lee, E.-J., Bang, W.-S., Kim, Y.-J., Kim, J.-S., 2017. Biofilm
https://doi.org/10.1186/1471-2180-13-115. formation of Campylobacter strains isolated from raw chickens and its reduction with
Jung, G.H., Lim, E.S., Woo, M.-A., Lee, J.Y., Kim, J.-S., Paik, H.-D., 2017. Inverse DNase I treatment. Food Control 71, 94–100. https://doi.org/10.1016/j.
correlation between extracellular DNase activity and biofilm formation among foodcont.2016.06.038.
chicken-derived Campylobacter strains. J. Mol. Biol. 27, 1942–1951. https://doi.org/ Koo, H., Allan, R.N., Howlin, R.P., Stoodley, P., Hall-Stoodley, L., 2017. Targeting
10.4014/jmb.1703.03052. microbial biofilms: current and prospective therapeutic strategies. Nat. Revs.
Kachhadia, R., Kapadia, C., Singh, S., Gandhi, K., Jajda, H., Alfarraj, S., et al., 2022. Microbiol. 15, 740–755. https://doi.org/10.1038/nrmicro.2017.99.
Quorum sensing inhibitory and quenching activity of Bacillus cereus RC1 extracts on Koul, S., Kalia, V.C., 2017. Multiplicity of quorum quenching enzymes: A potential
soft rot-causing bacteria Lelliottia amnigena. ACS Omega 7, 25291–25308. https:// mechanism to limit quorum sensing bacterial population. Indian J. Microbiol. 57,
doi.org/10.1021/acsomega.2c02202. 100–108. https://doi.org/10.1007/s12088-016-0633-1.
Kalia, V.C., 2013. Quorum sensing inhibitors: an overview. Biotechnol. Adv. 31, Koul, S., Prakash, J., Mishra, A., Kalia, V.C., 2016. Potential emergence of multi-quorum
224–245. https://doi.org/10.1016/j.biotechadv.2012.10.004. sensing inhibitor resistant (MQSIR) bacteria. Indian J. Microbiol. 56, 1–18. https://
Kalia, V.C., Koul, S., Ray, S., Prakash, J., 2018. Targeting quorum sensing mediated doi.org/10.1007/s12088-015-0558-0.
Staphylococcus aureus biofilms: A proteolytic approach. In: Kalia, V.C. (Ed.), Kumar, M., Sarma, D.K., Shubham, S., Kumawat, M., Verma, V., Nina, P.B., Jp, D.,
Biotechnological Applications of Quorum Sensing Inhibitors. Springer, Singapore, Kumar, S., Singh, B., Tiwari, R.R., 2021. Futuristic non-antibiotic therapies to
pp. 23–32. https://doi.org/10.1007/978-981-10-9026-4_2. combat antibiotic resistance: a review. Front. Microbiol. 12, 609459 https://doi.org/
Kalia, V.C., Patel, S.K.S., Kang, Y.C., Lee, J.-K., 2019. Quorum sensing inhibitors as 10.3389/fmicb.2021.609459.
antipathogens: biotechnological applications. Biotechnol. Adv. 37, 68–90. https:// Kumar, P., Patel, S.K.S., Lee, J.K., Kalia, V.C., 2013. Extending the limits of Bacillus for
doi.org/10.1016/j.biotechadv.2018.11.006. novel biotechnological applications. Biotechnol. Adv. 31, 1543–1561. https://doi.
Kan, J., An, L., Wu, Y., Long, J., Song, L., Fang, R., Jia, Y., 2018. A dual role for proline org/10.1016/j.biotechadv.2013.08.007.
iminopeptidase in the regulation of bacterial motility and host immunity. Mol. Plant Lallukka, T., Milllear, A., Pain, A., Cortinovis, M., Giussani, G., 2017. Global, regional,
Pathol. 9, 2011–2024. https://doi.org/10.1111/mpp.12677. and national life expectancy, all-cause mortality, and cause-specific mortality for

15
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

249 causes of death, 1980-2015: a systematic analysis for the Global Burden of human epithelial cells. Sci. Rep. 4, 1–8. https://doi.org/10.1007/978-3-319-23534-
Disease study 2015. Lancet 388, 1459–1544. https://doi.org/10.1016/S0140-6736 9_1.
(16)31012-1. Montanaro, L., Poggi, A., Visai, L., Ravaioli, S., Campoccia, D., Speziale, P., Arciola, C.R.,
Lebeaux, D., Chauhan, A., Rendueles, O., Beloin, C., 2013. From in vitro to in vivo 2011. Extracellular DNA in biofilms. Int. J. Art. Organs 34, 824–831. https://doi.
models of bacterial biofilm-related infections. Pathogens 2, 288–356. https://doi. org/10.5301/ijao.5000051.
org/10.3390/pathogens2020288. Morens, D.M., Fauci, A.S., 2020. Emerging pandemic diseases: how we got to COVID-19.
Lee, J., Sperandio, V., Frantz, D.E., Longgood, J., Camilli, A., Phillips, M.A., Michael, A. Cell 182, 1077–1092. https://doi.org/10.1016/j.cell.2020.08.021.
J., 2009. An alternative polyamine biosynthetic pathway is widespread in bacteria Morohoshi, T., Tokita, K., Ito, S., Saito, Y., Maeda, S., Kato, N., Ikeda, T., 2013. Inhibition
and essential for biofilm formation in Vibrio cholerae. J. Biol. Chem. 284, 9899–9907. of quorum sensing in Gram-negative bacteria by alkylamine-modified cyclodextrins.
https://doi.org/10.1074/jbc.M900110200. J. Biosci. Bioeng. 116, 175–179. https://doi.org/10.1016/j.jbiosc.2013.01.022.
Lee, K., Yoon, S.S., 2017. Pseudomonas aeruginosa biofilm., a programmed bacterial life Moscoso, J.A., Mikkelsen, H., Heeb, S., Williams, P., Filloux, A., 2011. The Pseudomonas
for fitness. J. Microbiol. Biotechnol. 27, 1053–1064. https://doi.org/10.4014/ aeruginosa sensor RetS switches type III and type VI secretion via cdi-GMP signaling.
jmb.1611.11056. Environ. Microbiol 13, 3128–3138. https://doi.org/10.1111/j.1462-
Lewis, K., 2007. Persister cells, dormancy and infectious disease. Nat. Rev. Microbiol. 5, 2920.2011.02595.x.
48–56. https://doi.org/10.1038/nmicro1557. Mulcahy, H., Charron-Mazenod, L., Lewenza, S., 2008. Extracellular DNA chelates
Li, X.H., Lee, J.H., 2017. Antibiofilm agents: a new perspective for antimicrobial strategy. cations and induces antibiotic resistance in Pseudomonas aeruginosa biofilms. PLoS
J. Microbiol. 55, 753–766. https://doi.org/10.1007/s12275-017-7274-x. Pathog. 4, e1000213 https://doi.org/10.1371/journal.ppat.1000213.
8 Li, Y., Wang, H., Xu, C., Sun, S.-H., Xao, K., Huang, X., 2022. Two strategies of Nijland, R., Hall, M.J., Burgess, J.G., 2010. Dispersal of biofilms by secreted, matrix
stubborn biofouling strains surviving from NaClO membrane cleaning: EPS degrading, bacterial DNase. PLoS One 5, e15668. https://doi.org/10.1371/journal.
shielding and/or quorum sensing. Sci. Total Environ. 838, 156421 https://doi.org/ pone.0015668.
10.1016/j.scitotenv.2022.156421. Nobile, C.J., Nett, J.E., Hernday, A.D., Homann, O.R., Deneault, J.S., Nantel, A.,
Lieberman, O.J., Orr, M.W., Wang, Y., Lee, V.T., 2014. High-throughput screening using Mitchell, A.P., 2009. Biofilm ECM regulation by Candida albicans Zap1. PLoS Biol. 7,
the differential radial capillary action of ligand assay identifies ebselen as an e1000133 https://doi.org/10.1371/journal.pbio.1000133.
inhibitor of diguanylate cyclases. ACS Chem. Biol. 9, 183–192. https://doi.org/ Noor, A.O., Almasri, D.M., Basyony, A.F., Albohy, A., Almutairi, L.S., Alhammadi, S.S.,
10.1021/cb400485k. Alkhamisi, M.A., Alsharif, S.A., Elfaky, M.A., 2022. Biodiversity of N-acyl
Lin, H., Wang, C., Zhao, H., Chen, G., Chen, X., 2018. Interaction between copper and homoserine lactonase (aiiA) gene from Bacillus subtilis. Microb. Pathog. 166, 105543
extracellular nucleic acids in the EPS of unsaturated Pseudomonas putida CZ1 biofilm. https://doi.org/10.1016/j.micpath.2022.105543.
Environ. Sci. Pollut. Res. 25, 24172–24180. https://doi.org/10.1007/s11999-017- Novotny, L.A., Jurcisek, J.A., Goodman, S.D., Bakaletz, L.O., 2016. Monoclonal
5266-0. antibodies against DNA-binding tips of DNABII proteins disrupt biofilms in vitro and
López-Baena, F.J., Vinardell, J.M., Medina, C., 2019. Regulation of protein secretion induce bacterial clearance in vivo. EBioMed 10, 33–44. https://doi.org/10.1016/j.
systems mediated by cyclic diguanylate in plant interacting bacteria. Front. ebiom.2016.06.022.
Microbiol. 10, 1289. https://doi.org/10.3389/fmicb.2019.01289. Okshevsky, M., Regina, V.R., Meyer, R.L., 2015. Extracellular DNA as a target for biofilm
Lu, L., Li, M., Yi, G., Liao, L., Cheng, Q., Zhu, J., Zhang, B., Wang, Y., Chen, Y., Zeng, M., control. Curr. Opin. Biotechnol. 33, 73–80. https://doi.org/10.1016/j.
2022. Screening strategies for quorum sesning inhibitors in combating bacterial copbio.2014.12.002.
infections. J. Pharm. Anal. 12, 1–14. https://doi.org/10.1016/j.jpha.2021.03.009. Onsea, J., Soentjens, P., Djebara, S., Merabishvili, M., Depypere, M., Spriet, I., Munter, P.
Lu, Y., Li, H., Pu, J., Xiao, Q., Zhao, C., Cai, Y., Liu, Y., Wang, L., Li, Y., Huang, B., D., Debaveye, Y., Nijs, S., Vanderschot, P., Wagemans, J., Pirnay, J.-P., Lavigne, R.,
Zeng, J., Chen, C., 2019. Identification of a novel RhlI/R-PrrH-LasI/Phzc/PhzD Metsemakers, W.-J., 2019. Bacteriophage application for difficult-to-treat
signalling cascade and its implication in P aeruginosa virulence. Emerg. Microbes musculoskeletal infections: development of a standardized multidisciplinary
Infect. 8, 1658–1667. https://doi.org/10.1080/22221751.2019.1687262. treatment protocol. Viruses 11, 891. https://doi.org/10.3390/v11100891.
Luciardi, M.C., Pérez Hernández, M.V., Muruaga, N., Bardón, A., Arena, M.E., Pakkulnan, R., Anutrakunchai, C., Kanthawong, S., Taweechaisupapong, S.,
Cartagena, E., 2016. Volatiles from subtropical convolvulaceae that interfere with Chareonsudjai, P., Chareonsudjai, S., 2019. Extracellular DNA facilitates bacterial
bacterial cell-to-cell communication as potential antipathogenic drugs. Evid. Based adhesion during Burkholderia pseudomallei biofilm formation. PLOS One 14,
Complement. Altern. Med. 2016, 7890260. https://doi.org/10.1155/2016/ e0213288. https://doi.org/10.1371/journal.pone.0213288.
7890260. Pani, A., Lucini, V., Dugnani, S., Scaglione, F., 2022. Erdosteine enhances antibiotic
Manfiolli, A.O., Dos Reis, T.F., de Assis, L.J., de Castro, P.A., Silva, L.P., Hori, J.I., activity against bacteria within biofilm. Int. J. Antimicrob. Agents 59, 106529.
Walker, L.A., Munro, C.A., Rajendran, R., Ramage, G., Goldman, G.H., 2018. https://doi.org/10.1016/j.ijantimicag.2022.106529.
Mitogen activated protein kinases (MAPK) and protein phosphatases are involved in Panlilio, H., Rice, C.V., 2021. The role of extracellular DNA in the formation,
Aspergillus fumigatus adhesion and biofilm formation. Cell Surf. 1, 43–56. https://doi. architecture, stability, and treatment of bacterial biofilms. Biotechnol. Bioeng. 118,
org/10.1016/j.tcsw.2018.03.002. 2129–2141. https://doi.org/10.1002/bit.27760.
Mann, E.E., Wozniak, D.J., 2012. Pseudomonas biofilm matrix composition and niche Papenfort, K., Bassler, B., 2016. Quorum-sensing signal-response systems in gram-
biology. FEMS Microbiol. Rev. 36, 893–916. https://doi.org/10.1111/j.1574- negative bacteria. Nat. Rev. Microbiol. 14, 576–588. https://doi.org/10.1038/
6976.2011.00322.x. nrmicro.2016.89.
Massie, J.P., Reynolds, E.L., Koestler, B.J., Cong, J.P., Agostoni, M., Waters, C.M., 2012. Parrino, B., Diana, P., Cirrincione, G., Cascioferro, S., 2018. Bacterial biofilm inhibition
Quantification of high-specificity cyclic diguanylate signaling. Proc. Natl. Acad. Sci. in the development of effective anti-virulence strategy. Open Med. Chem. J. 12,
USA 109, 12746–12751. https://doi.org/10.1073/pnas.1115663109. 84–87. https://doi.org/10.2174/1874104501812010084.
Mayer, C., Muras, A., Parga, A., Romero, M., Rumbo-Feal, S., Poza, M., Ramos-Vivas, J., Pedrido, M.E., de Oña, P., Ramirez, W., Leñini, C., Goñi, A., Grau, R., 2013. Spo0a links
Otero, A., 2020. Quorum sensing as a target for controlling surface associated de novo fatty acid synthesis to sporulation and biofilm development in Bacillus
motility and biofilm formation in Acinetobacter baumannii ATCC® 17978TM. Front. subtilis. Mol. Microbiol. 87, 348–367. https://doi.org/10.1111/mmi.12102.
Microbiol. 11, 565548 https://doi.org/10.3389/fmicb.2020.565548. Pejin, B., Talevksa, A., Ciric, A., Glamoclija, J., Nikolic, M., Talevski, T., Sokovic, M.,
Melaugh, G., Hutchison, J., Kragh, K.N., Irie, Y., Roberts, A., Bjarnsholt, T., Diggle, S.P., 2014. Anti-quorum sensing activity of selected sponge extracts: A case study of
Gordon, V.D., Allen, R.J., 2016. Shaping the growth behaviour of biofilms initiated Pseudomonas aeruginosa. Nat. Prod. Res. 28, 2330–2333. https://doi.org/10.1080/
from bacterial aggregates. PLOS One 11, e0149683. https://doi.org/10.1371/ 14786419.2014.934239.
journal.pone.0149683. Pérez-Mendoza, D., Aragón, I.M., Prada-Ramírez, H.A., Romero-Jiménez, L., Ramos, C.,
Merritt, J.H., Brothers, K.M., Kuchma, S.L., O’Toole, G.A., 2007. SadC reciprocally Gallegos, M.T., Sanjuan, J., 2014. Responses to elevated c-di-GMP levels in
influences biofilm formation and swarming motility via modulation of mutualistic and pathogenic plant-interacting bacteria. PLoS One 9, e91645. https://
exopolysaccharide production and flagellar function. J. Bacteriol. 189, 8154–8164. doi.org/10.1371/journal.pone.0091645.
https://doi.org/10.1128/JB.00585-07. Petrova, O.E., Cherny, K.E., Sauer, K., 2015. The diguanylate cyclase GcbA facilitates
Merritt, J.H., Ha, D.G., Cowles, K.N., Lu, W., Morales, D.K., Rabinowitz, J., Gitai, Z., Pseudomonas aeruginosa biofilm dispersion by activating BdlA. J. Bacteriol. 197,
O’Toole, G.A., 2010. Specific control of Pseudomonas aeruginosa surface-associated 174–187. https://doi.org/10.1128/JB.02244-14.
behaviors by two c-di-GMP diguanylate cyclases. e00183–10 mBio 1. https://doi. Piri-Gharaghie, T., Jegargoshe-Shirin, N., Saremi-Nouri, S., Khademhosseini, S.H.,
org/10.1128/mBio.00183-10. Hoseinnezhad-Lazarjani, E., Mousavi, A., Kabiri, H., Rajaei, N., Riahi, A., Farhadi-
Migliore, F., Macchi, R., Landini, P., Paroni, M., 2018. Phagocytosis and epithelial cell Biregani, A., Fatehi-Ghahfarokhi, S., 2022. Effects of Imipenem-containing Niosome
invasion by Crohn’s disease-associated adherent-invasive Escherichia coli are nanoparticles against high prevalence methicillin-resistant Staphylococcus epidermidis
inhibited by the anti-inflammatory drug 6-mercaptopurine. Front. Microbiol. 9, 964. biofilm formed. Sci. Rep. 12, 5140. https://doi.org/10.1038/s41598-022-09195-9.
https://doi.org/10.3389/fmicb.2018.00964. Porter, S.B., Johnston, B.D., Kisiela, D., Clabots, C., 2022. Bacteriophage cocktail and
Mishra, R., Panda, A.K., De Mandal, S., Shakeel, M., Bisht, S.S., Khan, J., 2020. Natural microcin-producing probiotic Escherichia coli protect mice against gut colonization
anti-biofilm agents: strategies to control biofilm-forming pathogens. Front. with multidrug-resistant Escherichia coli. Front. Microbiol. 13, 887799 https://doi.
Microbiol. 11, 566325 https://doi.org/10.3389/fmicb.2020.566325. org/10.3389/fmicb.2022.887799.
Molnár, M., Fenyvesi, É., Berkl, Z., Németh, I., Fekete-Kertész, I., Márton, R., Vaszita, E., Prateeksha, Singh, B.R., Shoeb, M., Sharma, S., Naqvi, A.H., Gupta, V.K., Singh, B.N.,
Varga, E., Ujj, D., Szente, L., 2021. Cyclodextrin-mediated quorum quenching in the 2017. Scaffold of selenium nanovectors and honey phytochemicals for inhibition of
Aliivibrio fischeri bioluminescence model system—Modulation of bacterial Pseudomonas aeruginosa quorum sensing and biofilm formation. Front. Cell Infect.
communication. Int. J. Pharm. 594, 120–150. https://doi.org/10.1016/j. Microbiol. 7, 93. https://doi.org/10.3389/fcimb.2017.00093.
ijpharm.2020.120150. Pu, J., Zhang, S., He, X., Zeng, J., Shen, C., Luo, Y., Li, H., Long, Y., Liu, J., Xiao, Q.,
Monnappa, A.K., Dwidar, M., Seo, J.K., Hur, J.-H., Mitchell, R.J., 2014. Bdellovibrio Lu, Y., Huang, B., Chen, C., 2022. The small RNA AmiL regulates quorum sensing-
bacteriovorus inhibits Staphylococcus aureus biofilm formation and invasion into mediated virulence in Pseudomonas aeruginosa PAO1. Microbiol. Spectr. 10,
e0221121 https://doi.org/10.1128/spectrum.02211-21.

16
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Pusic, P., Tata, M., Wolfinger, M.T., Sonnleitner, E., Häussler, S., Bläsi, U., 2016. Cross- Smith, T.J., Sondermann, H., O’Toole, G.A., 2018. Type 1 does the two-step: type 1
regulation by CrcZ RNA controls anoxic biofilm formation in Pseudomonas secretion substrates with a functional periplasmic intermediate. e00168–e00118
aeruginosa. Sci. Rep. 6, 39621. https://doi.org/10.1038/srep39621. J. Bacteriol. 200. https://doi.org/10.1128/JB.00168-18.
Raj, V., Kim, Y., Kim, Y.G., Lee, J.H., Lee, J., 2022. Chitosan-gum arabic embedded Srikantha, T., Daniels, K.J., Pujol, C., Kim, E., Soll, D.R., 2013. Identification of genes
alizarin nanocarriers inhibit biofilm formation of multispecies microorganisms. upregulated by the transcription factor Bcr1 that are involved in impermeability,
Carbohydr. Polym. 284, 118959 https://doi.org/10.1016/j.carbpol.2021.118959. impenetrability and drug-resistance of Candida albicans a/alpha biofilms. Eukaryot.
Rajesh, P.S., Ravishankar Rai, V., 2014. Quorum quenching activity in cell-free lysate of Cell. 12, 875–888.
endophytic bacteria isolated from Pterocarpus santalinus Linn., and its effect on Srinivasan, R., Devi, K.R., Kannappan, A., Karutha Pandian, S., Ravi, A.V., 2016. Piper
quorum sensing regulated biofilm in Pseudomonas aeruginosa PAO1. Microb. Res 169, betle and its bioactive metabolite phytol mitigates quorum sensing mediated
561–569. https://doi.org/10.1016/j.micres.2013.10.005. virulence factors and biofilm of nosocomial pathogen Serratia marcescens in vitro.
Ramos, E.R., Reitzel, R., Jiang, Y., Hachem, R.Y., Chaftari, A.M., Chemaly, R.F., J. Ethnopharmacol. 193, 592–603. https://doi.org/10.1016/j.jeo.2016.10.017.
Hackett, B., Pravinkumar, S.E., Nates, J., Tarrand, J.J., et al., 2011. Clinical Srinivasan, R., Mohankumar, R., Kannappan, A., Karthick Raja, V., Archunan, G.,
effectiveness and risk of emerging resistance associated with prolonged use of Karutha Pandian, S., Ruckmani, K., Ravi, A.V., 2017. Exploring the anti-quorum
antibiotic-impregnated catheters: More than 0.5 million catheter days and 7 years of sensing and antibiofilm efficacy of phytol against Serratia marcescens associated
clinical experience. Crit. Care Med. 39, 245–251. https://doi.org/10.1097/ acute pyelonephritis infection in Wistar rats. Front. Cell. Infect. Microbiol. 7, 498.
CCM.0b013e3181feb83e. https://doi.org/10.1016/j.jphotobiol.2019.111637.
Randrianjatovo-Gbalou, I., Rouquette, P., Lefebvre, D., Girbal-Neuhauser, E., Marcato- Srinivasan, R., Vigneshwari, L., Rajavel, T., Durgadevi, R., Kannappan, A.,
Romain, C.-E., 2017. In situ analysis of Bacillus licheniformis biofilms: Amyloid-like Balamurugan, K., Devi, K.S., Ravu, A.V., 2018. Biogenic synthesis of silver
polymers and eDNA are involved in the adherence and aggregation of the nanoparticles using Piper betle aqueous extract and evaluation of its anti-quorum
extracellular matrix. J. Appl. Microbiol. 122, 1262–1274. https://doi.org/10.1111/ sensing and antibiofilm potential against uropathogens with cytotoxic effects: an in
jam.13423. vitro and in vivo approach. Environ. Sci. Pollut. Res. Int. 25, 10538–10554. https://
Robino, L., Scavone, P., 2020. Nanotechnology in biofilm prevention. Future Microbiol doi.org/10.1007/s11356-017-1049-0.
15, 377–379. https://doi.org/10.2217/fmb-2019-0327. Srinivasan, R., Santhakumari, S., Poonguzhali, P., Geetha, M., Dyavaiah, M.,
Rodríguez-López, P., Carballo-Justo, A., Draper, L.A., Cabo, M.L., 2017. Removal of Xiangmin, L., 2021. Bacterial biofilm inhibition: a focused review on recent
Listeria monocytogenes dual-species biofilms using combined enzyme-benzalkonium therapeutic strategies for combating the biofilm mediated infections. Front.
chloride treatments. Biofouling 33, 45–58. https://doi.org/10.1080/ Microbiol. 12, 676458 https://doi.org/10.3389/fmicb.2021.676458.
08927014.2016.1261847. Srivastava, A., Singh, B.N., Deepak, D., Rawat, A.K., Singh, B.R., 2015. Colostrum
Römling, U., Balsalobre, C., 2012. Biofilm infections, their resilience to therapy and hexasaccharide., a novel Staphylococcus aureus quorum sensing inhibitor.
innovative treatment strategies. J. Intern. Med. 272, 541–561. https://doi.org/ Antimicrob. Agents Chemother. 59, 2169–2178. https://doi.org/10.1128/
10.1111/joim.12004. AAC.03722-14.
Roy, R., Tiwari, M., Donelli, G., Tiwari, V., 2018. Strategies for combating bacterial Stewart, P.S., Davison, W.M., Steenbergen, J.N., 2009. Daptomycin rapidly penetrates a
biofilms: A focus on anti-biofilm agents and their mechanisms of action. Virulence 9, Staphylococcus epidermidis biofilm. Antimicrob. Agents Chemother. 53, 3505–3507.
522–554. https://doi.org/10.1080/21505594.2017.1313372. https://doi.org/10.1128/AAC.01728-08.
Saising, J., Dube, L., Ziebandt, A.K., Voravuthikunchai, S.P., Nega, M., Götz, F., 2012. Stirpe, M., Brugnoli, B., Donelli, G., Francolini, I., Vuotto, C., 2020. Poloxamer 338
Activity of gallidermin on Staphylococcus aureus and Staphylococcus epidermidis affects cell adhesion and biofilm formation in Escherichia coli: potential applications
biofilms. Antimicrob. Agents Chemother. 56, 5804–5810. https://doi.org/10.1128/ in the management of catheter-associated urinary tract infections. Pathogens 9, 885.
AAC.01296-12. https://doi.org/10.3390/pathogens9110z85.
Sambanthamoorthy, K., Sloup, R.E., Parashar, V., Smith, J.M., Kim, E.E., Stoodley, P., Sauer, K., Davies, D.G., Costerton, J.W., 2002. Biofilms as complex
Semmeelhack, M.F., Neiditch, M.B., Waters, C.M., 2012. Identification of small differentiated communities. Annu. Rev. Microbiol. 56, 187–209. https://doi.org/
molecules that antagonize diguanylate cyclase enzymes to inhibit biofilm formation. 10.1146/annurev.micro.56.012302.160705.
Antimicrob. Agents Chemother. 56, 5202–5211. https://doi.org/10.1128/ Subhadra, B., Kim, D.H., Woo, K., Surendran, S., Choi, C.H., 2018. Control of biofilm
AAC.01396-12. formation in healthcare: recent advances exploiting quorum-sensing interference
Sarkar, R., Mondal, C., Bera, R., Chakraborty, S., Barik, R., Roy, P., Kumar, A., Yadav, K. strategies and multidrug efflux pump inhibitors. Materials 11, e1676. https://doi.
K., Choudhury, J., Chaudhary, S.K., Samanta, S.K., Karmakar, S., Das, S., org/10.3390/ma11091676.
Mukherjee, P.K., Mukherjee, J., Sen, T., 2015. Antimicrobial properties of Kalanchoe Sultana, S.T., Call, D.R., Beyenal, H., 2016. Eradication of Pseudomonas aeruginosa
blossfeldiana: a focus on drug resistance with particular reference to quorum sensing- biofilms and persister cells using an electrochemical scaffold and enhanced
mediated bacterial biofilm formation. J. Pharm. Pharm. 67, 951–962. https://doi. antibiotic susceptibility. NPJ Biofilms Microb. 2, 2. https://doi.org/10.1038/
org/10.1111/jphp.12397. s41522-016-0003-0.`.
Sateriale, D., Imperatore, R., Colicchio, R., Pagliuca, C., Varricchio, E., Volpe, M.G., Taff, H.T., Nett, J.E., Zarnowski, R., Ross, K.M., Sanchez, H., Cain, M.T., Andes, D.R.,
Salvatore, P., Paolucci, M., Pagliarulo, C., 2020. Phytocompounds vs. dental plaque 2012. A Candida biofilm-induced pathway for ECM glucan delivery: implications for
bacteria: in vitro effects of myrtle and pomegranate polyphenolic extracts against drug resistance. PLoS Pathog. 8, e1002848 https://doi.org/10.1371/journal.
single-species and multispecies oral biofilms. Front. Microbiol. 11, 592265 https:// ppat.1002848.
doi.org/10.3389/fmicb.2020.592265. Tahmourespour, A., Kasra-Kermanshahi, R., Salehi, R., 2019. Lactobacillus rhamnosus
Satpute, S.K., Mone, N.S., Das, P., Banat, I.M., Banpurkar, A.G., 2019. Inhibition of biosurfactant inhibits biofilm formation and gene expression of caries-inducing
pathogenic bacterial biofilms on PDMS based implants by L. acidophilus derived Streptococcus mutans. Dent. Res. J. 16, 87–94.
biosurfactant. BMC Microbiol 19, 39. https://doi.org/10.1186/s12866-019-1412-z. Tahrioui, A., Duchesne, R., Bouffartigues, E., Rodrigues, S., Maillot, O., Tortuel, D.,
Saunders, S.H., Tse, E.C.M., Yates, M.D., Otero, F.J., Trammell, S.A., Stemp, E.D.A., Hardouin, J., Taupin, L., Groleau, M.C., Dufour, A., Déziel, E., Brenner-Weiss, G.,
Barton, J.K., Tender, L.M., Newman, D.K., 2020. Extracellular DNA promotes Feuilloley, M., Orange, N., Lesouhaitier, O., Cornelis, P., Chevalier, S., 2019.
efficient extracellular electron transfer by pyocyanin in Pseudomonas aeruginosa Extracellular DNA release, quorum sensing, and PrrF1/F2 small RNAs are key
biofilms. Cell 182, 919–932. https://doi.org/10.1016/j.cell.2020.07.006. players in Pseudomonas aeruginosa tobramycin-enhanced biofilm formation. NPJ
Savage, V.J., Chopra, I., O’Neill, A.J., 2013. Staphylococcus aureus biofilms promote Biofilms Microb. 5, 15. https://doi.org/10.1038/s41522-019-0088-3.
horizontal transfer of antibiotic resistance. Antimicrob. Agents Chemother. 57, Taylor, P.K., Van Kessel, A.T.M., Colavita, A., Hancock, R.E.W., Mah, T.-F., 2017. A novel
1968–1970. https://doi.org/10.1128/AAC.02008-12. small RNA is important for biofilm formation and pathogenicity in Pseudomonas
Saxena, P., Joshi, Y., Rawat, K., Bisht, R., 2019. Biofilms: Architecture, resistance, aeruginosa. PLoS One 12, e0182582. https://doi.org/10.1371/journal.
quorum sensing and control mechanisms. Indian J. Microbiol. 59, 3–12. https://doi. pone.0182582.
org/10.1007/s12088-018-0757-6. Tetz, G.V., Artemenko, N.K., Tetz, V.V., 2009. Effect of DNase and antibiotics on biofilm
Schiessl, K.T., Hu, F., Jo, J., Nazia, S.Z., Wang, B., Price-Whelan, A., Min, W., Dietrich, L. characteristics. Antimicrob. Agents Chemother. 53, 1204–1209. https://doi.org/
E.P., 2019. Phenazine production promotes antibiotic tolerance and metabolic 10.1128/aac.00471-08.
heterogeneity in Pseudomonas aeruginosa biofilms. Nat. Commun. 10, 762. https:// Thi Bach Nguyen, H., Romero, A.D., Amman, F., Sorger-Domenigg, T., Tata, M.,
doi.org/10.1038/s41467-019-08733-w. Sonnleitner, E., Bläsi, U., 2018. Negative control of RpoS synthesis by the sRNA ReaL
Schuch, R., Khan, B.K., Raz, A., Rotolo, J.A., Wittekind, M., 2017. Bacteriophage lysin in Pseudomonas aeruginosa. Front. Microbiol. 9, 2488. https://doi.org/10.3389/
CF-301., a potent antistaphylococcal biofilm agent. e02666–16 Antimicrob. Agents fmicb.2018.02488.
Chemother. 61. https://doi.org/10.1128/AAC.02666-16. Tien, H.C., Battad, A., Bryce, E.A., Fuller, J., Mulvey, M., Bernard, K., Brisebois, R.,
Secinti, K.D., Özalp, H., Attar, A., Sargon, M.F., 2011. Nanoparticle silver ion coatings Doucet, J.J., Rizoli, S.B., Fowler, R., 2007. Multidrug resistant Acinetobacter
inhibit biofilm formation on titanium implants. J. Clin. Neurosci. 18, 391–395. infections in critically injured Canadian forces soldiers. BMC Infect. Dis. 7, 95.
https://doi.org/10.1016/j.jocn.2010.06.022. https://doi.org/10.1186/1471-2334-7-95.
Sethupathy, S., Sathiyamoorthi, E., Kim, Y.-G., Lee, J.-H., Lee, J., 2020. Antibiofilm and Toranzo, A., Bustos, P.S., Ortega, M.G., Páez, P.L., Lucero-Estrada, C., 2022. Biologically
antivirulence properties of indoles against Serratia marcescens. Front. Microbiol. 11, synthesized silver nanoparticles, mediated by Bothriochloa laguroides, inhibit biofilm
584812 https://doi.org/10.3389/fmicb.2020.584812. formation and eradicate mature biofilm of Yersinia enterocolitica and Staphylococcus
Sharma, A.K., Dhasmana, N., Dubey, N., Kumar, N., Gangwal, A., Gupta, M., Singh, Y., aureus. J. Appl. Microbiol. 132, 209–220. https://doi.org/10.1111/jam.15195.
2017. Bacterial virulence factors: secreted for survival. Indian J. Microbiol. 57, 1–10. Torelli, R., Cacaci, M., Papi, M., Sterbini, F.P., Martini, C., Posteraro, B., Palmieri, V., De
https://doi.org/10.1007/s12088-016-0625-1. Spirito, M., Sanguinetti, M., Bugli, F., 2017. Different effects of matrix degrading
Sharma, K., Pagedar Singh, A., 2018. Antibiofilm effect of DNase against single and enzymes towards biofilms formed by E. faecalis and E. faecium clinical isolates.
mixed species biofilm. Foods 7, 42. https://doi.org/10.3390/foods7030042. Colloids Surf. B Biointerfaces 158, 349–355. https://doi.org/10.1016/j.
colsurfb.2017.07.010.

17
V.C. Kalia et al. Ecotoxicology and Environmental Safety 264 (2023) 115389

Toyofuku, M., Nomura, N., Eberl, L., 2019. Types and origins of bacterial membrane Antimicrob. Agents 56, 106200. https://doi.org/10.1016/j.
vesicles. Nat. Rev. Microbiol. 17, 13–24. https://doi.org/10.1038/s41579-018- ijantimicag.2020.106200.
0112-2. Wang, S., Liu, X., Liu, H., Zhang, L., Guo, Y., Yu, S., Wozniak, D.J., Ma, L.Z., 2015. The
Trampari, E., Stevenson, C.E., Little, R.H., Wilhelm, T., Lawson, D.M., Malone, J.G., exopolysaccharide Psl-eDNA interaction enables the formation of a biofilm skeleton
2015. Bacterial rotary export ATPases are allosterically regulated by the nucleotide in Pseudomonas aeruginosa. Environ. Microbiol. Rep. 7, 330–340. https://doi.org/
second messenger cyclic-di-GMP. J. Biol. Chem. 290, 24470–24483. https://doi.org/ 10.1111/1758-2229.12252.
10.1074/jbc.M115.661439. Wang, Y., Li, Y., Wang, J., Wang, X., 2017. FleQ regulates both the type VI secretion
Tran, C.S., Rangel, S.M., Almblad, H., Kierbel, A., Givskov, M., Tolker-Nielsen, T., system and flagella in Pseudomonas putida. Biotechnol. Appl. Biochem. 65, 419–427.
Hauser, A.R., Engel, J.N., 2014. The Pseudomonas aeruginosa type III translocon is https://doi.org/10.1002/bab.1611.
required for biofilm formation at the epithelial barrier. PLoS Pathog. 10, e1004479 World Health Organization, 2014. Antimicrobial Resistance: Global Report on
https://doi.org/10.1371/journal.ppat.1004479. Surveillance 2014. World Health Organization, Geneva, Switzerland. 〈http://apps.
Turnbull, L., Toyofuku, M., Hynen, A.L., Kurosawa, M., Pessi, G., Petty, N.K., Osvath, S. who.int/iris/bitstream/10665/112642/1/9789241564748_eng.pdf?ua=1〉.
R., Cárcamo-Oyarce, G., Gloag, E.S., Shimoni, R., Omasits, U., Ito, S., Yap, X., Accessed 2023.03.23.
Monahan, L.G., Cavaliere, R., Ahrens, C.H., Charles, I.G., Nomura, N., Eberl, L., Wotanis, C.K., Brennan III, W.P., Angotti, A.D., Villa, E.A., Zayner, J.P., Mozina, A.,
Whitchurch, C.B., 2016. Explosive cell lysis as a mechanism for the biogenesis of Rutkovsky, A.C., Sobe, R.C., Bond, W.G., Karatan, E., 2017. Relative contributions of
bacterial membrane vesicles and biofilms. Nat. Commun. 7, 11220. https://doi.org/ norspermidine synthesis and signaling pathways to the regulation of Vibrio cholerae
10.1038/ncomms11220. biofilm formation. PLoS One 12, e0186291. https://doi.org/10.1371/journal.
Tursi, S.A., Puligedda, R.D., Szabo, P., Nicastro, L.K., Miller, A.L., Qiu, C., Gallucci, S., pone.0186291.
Relkin, N.R., Buttaro, B.A., Dessain, S.K., Tukel, C., 2020. Salmonella Typhimurium Xiong, Y., Chen, J., Sun, X., Xu, G., Li, P., Deng, Q., Yu, Z., Chen, Z., Zheng, J., 2021. The
biofilm disruption by a human antibody that binds a pan-amyloid epitope on curli. antibacterial and antibiofilm activity of telithromycin against Enterococcus spp.
Nat. Commun. 11, 1007. https://doi.org/10.1038/s41467-020-14685-3. isolated from patients in China. Front. Microbiol. 11, 616797 https://doi.org/
Uppuluri, P., Lopez-Ribot, J.L., 2016. Go forth and colonize: dispersal from clinically 10.3389/fmicb.2020.616797.
important microbial biofilms. PLoS Pathog. 12, e1005397 https://doi.org/10.1371/ Yang, D., Chen, Y., Sun, E., Hua, L., Peng, Z., Wu, B., 2020. Characterization of a lytic
journal.ppat.1005397. bacteriophage vB_EfaS_PHB08 harboring endolysin lys08 against Enterococcus
Utari, P.D., Setroikromo, R., Melgert, B.N., Quax, W.J., 2018. PvdQ quorum quenching faecalis biofilms. Microrganisms 8, 1332. https://doi.org/10.3390/
acylase attenuates Pseudomonas aeruginosa virulence in a mouse model of pulmonary microorganisms8091332.
infection. Front. Cell Infect. Microbiol. 8, 119. https://doi.org/10.3389/ Yang, Q., Pande, G.S.J., Wang, Z., Lin, B., Rubin, R.A., Defoirdt, T., 2017. Indole
fcimb.2018.00119. signalling and microalgal auxins decrease the virulence of Vibrio campbellii, a major
Valentini, M., Filloux, A., 2019. Multiple roles of c-di-GMP signaling in bacterial pathogen of aquatic organisms. Environ. Microbiol. 19, 1987–2004. https://doi.org/
pathogenesis. Annu. Rev. Microbiol 73, 387–406. https://doi.org/10.1146/annurev- 10.1111/1462-2920.13714.
micro-020518-115555. Ye, J., Shao, C., Zhang, X., Guo, X., Gao, P., Cen, Y., Ma, S., Liu, Y., 2017. Effects of
Valliammai, A., Selvaraj, A., Yuvashree, U., Aravindraja, C., Pandian, S.K., 2020. sarA- DNase I coating of titanium on bacteria adhesion and biofilm formation. Mater. Sci.
Dependent antibiofilm activity of thymol enhances the antibacterial efficacy of Eng. C: Mater. Biol. Appl. 78, 738–747. https://doi.org/10.1016/j.
rifampicin against Staphylococcus aureus. Front. Microbiol. 11, 1744. https://doi. msec.2017.04.078.
org/10.3389/fmicb.2020.01744. Yuan, X., Khokhani, D., Wu, X., Yang, F., Biener, G., Koestler, B.J., Raicu, V., He, C.,
Venkatramanan, M., Sankar Ganesh, P., Senthil, R., Akshay, J., Veera Ravi, A., Waters, C.M., Sundin, G.W., Tian, F., Yang, C.H., 2015. Cross-talk between a
Langeswaran, K., Vadivelu, J., Nagarajan, S., Rajendran, K., Shankar, E.M., 2020. regulatory small RNA, cyclic-di-GMP signalling and flagellar regulator FlhDC for
Inhibition of quorum sensing and biofilm formation in Chromobacterium violaceum by virulence and bacterial behaviours. Environ. Microbiol. 17, 4745–4763. https://doi.
fruit extracts of Passiflora edulis. ACS Omega 5, 25605–25616. https://doi.org/ org/10.1111/1462-2920.13029.
10.1021/acsomega.0c02483. Zatorska, B., Groger, M., Moser, D., Diab-Elschahawi, M., Lusignani, L.S., Presterl, E.,
Vestby, L.K., Grønseth, T., Simm, R., Nesse, L.L., 2020. Bacterial biofilm and its role in 2017. Does extracellular DNA production vary in staphylococcal biofilms isolated
the pathogenesis of disease. Antibiotics 9, 59. https://doi.org/10.3390/ from infected implants versus controls? Clin. Orthop. Rel. Res. 475, 2105–2113.
antibiotics9020059. https://doi.org/10.1007/s11999-017-5266-0.
Vuotto, C., Donelli, G., 2019. Novel treatment strategies for biofilm-based infections. Zhou, J., Watt, S., Wang, J., Nakayama, S., Sayre, D.A., Lam, Y.-F., Lee, V.Y., Sintim, H.
Drugs 79, 1635–1655. https://doi.org/10.1007/s40265-019-01184-z. O., 2013. Potent suppression of c-di-GMP synthesis via I-site allosteric inhibition of
Wagener, J.S., Kupfer, O., 2012. Dornase alfa (pulmozyme). Curr. Opin. Pulm. Med. 18, diguanylate cyclases with 2′-F-c-di-GMP. Bioorg. Med. Chem. 21, 4396–4404.
609–614. https://doi.org/10.1097/MCP.0b013e328358d51f. https://doi.org/10.1016/j.bmc.2013.04.050.
Wan, B., Zhu, Y., Tao, J., Zhu, F., Chen, J., Li, L., Zhao, J., Wang, L., Sun, S., Yang, Y., Zhou, L., Zhang, L.H., Cámara, M., He, Y.W., 2017. The DSF family of quorum sensing
Zhang, X., Zhang, Y., 2020. Alginate lyase guided silver nanocomposites for signals: diversity, biosynthesis, and turnover. Trends Microbiol. 25, 293–303.
eradicating Pseudomonas aeruginosa from lungs. ACS Appl. Mater. Interfaces 12, https://doi.org/10.1016/j.tim.2016.11.013.
9050–9061. https://doi.org/10.1021/acsami.9b21815. Zhu, K., Chen, S., Sysoeva, T.A., You, L., 2019. Universal antibiotic tolerance arising from
Wang, J., Zhang, M., Wang, M., Zang, J., Zhang, X., Hang, T., 2021. Structural insights antibiotic-triggered accumulation of pyocyanin in Pseudomonas aeruginosa. PLOS
into the intermolecular interaction of the adhesion SdrC in the pathogenicity of Biol. 17, e3000573 https://doi.org/10.1371/journal.pbio.3000573.
Staphylococcus aureus. Acta Crystallogr. F. Struct. Biol. Commun. F77, 47–53. Ziegler, E.W., Brown, A.B., Nesnas, N., Chouinard, C.D., Mehta, A.K., Palmer, A.G., 2021.
https://doi.org/10.1107/S2053230X21000741. β-Cyclodextrin encapsulation of synthetic AHLs: Drug delivery implications and
Wang, L., Tkhilaishvili, T., Bernal Andres, B., Trampuz, A., Gonzalez Moreno, M., 2020. quorum-quenching exploits. Chem. Biochem. 22, 1292–1301. https://doi.org/
Bacteriophage-antibiotic combinations against ciprofloxacin/ceftriaxone-resistant 10.1002/cbic.202000773.
Escherichia coli in vitro and in an experimental Galleria mellonella model. Int. J.

18

You might also like