You are on page 1of 12

Microbiological Research 247 (2021) 126722

Contents lists available at ScienceDirect

Microbiological Research
journal homepage: www.elsevier.com/locate/micres

An overview of Acinetobacter baumannii pathogenesis: Motility, adherence


and biofilm formation
Hing Jian Mea a, Phelim Voon Chen Yong a, Eng Hwa Wong b, *
a
School of Biosciences, Faculty of Health and Medical Sciences, Taylor’s University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
b
School of Medicine, Faculty of Health and Medical Sciences, Taylor’s University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia

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

Keywords: The Gram-negative opportunistic pathogen Acinetobacter baumannii has gain notoriety in recent decades, pri­
Acinetobacter baumannii marily due to its propensity to cause nosocomial infections in critically ill patients. Its global spread, multi-drug
Virulence resistance features and plethora of virulence factors make it a serious threat to public health worldwide. Though
Bacterial pathogenesis
much effort has been expended in uncovering its successes, it continues to confound researchers due to its highly
Surface structures
Host-pathogen interaction
adaptive nature, mutating to meet the needs of a given environment. Its persistence in the clinical setting allows
Animal models it to be in close proximity to a potential host, where contact can be made facilitating infection and colonization.
In this article, we aim to provide a current overview of the bacterial virulence factors, specifically focusing on
factors involved in the initial stages of infection, highlighting the role of adaptation facilitated by two-component
systems and biofilm formation. Finally, the study of host-pathogen interactions using available animal models,
their suitability, notable findings and some perspectives moving forward are also discussed.

1. Introduction With such a reputation, it is no surprise that major global health


authorities including the European Centre for Disease Prevention and
The rise of Acinetobacter spp, specifically A. baumannii as a nosoco­ Control (ECDC), Infectious Diseases Society of America (IDSA), WHO
mial pathogen has stunned the world and poses a significant threat to and Center for Disease Control and Prevention of America (CDC) have
the global health care system. Initially regarded as a low-grade path­ declared multi-drug resistant A. baumannii a critical threat to global
ogen, it is now recognized as an opportunistic pathogen, predominantly health, with the latter promoting Carbapenem-Resistant Acinetobacter
infecting hospital patients making it a leading nosocomial pathogen from a ‘serious’ threat level to ‘urgent’ in 2019 (CDC, 2019; Pendleton
with broad clinical manifestations. Most notably in ventilator-associated et al., 2013; Rello et al., 2018; Tacconelli et al., 2017). It has also been
pneumonia, blood stream infections, urinary tract infections and men­ considered the most serious among the ‘ESKAPE’ organisms (Entero­
ingitis (Bergogne-Berezin and Towner 1996; Peleg et al., 2008). Earlier coccus faecium, Staphylococcus aureus, Klebsiella pneumoniae, A. bau­
cases can be traced back to the early 2000s as reports began to surface mannii, Pseudomonas aeruginosa, and Enterobacter species), so named due
from wounded veterans and soldiers who served in Iraq and Afghanistan to their proficient ability to escape the effects of antimicrobial drugs
repatriated to be treated for conflict induced trauma contracting the (Boucher et al., 2009). Furthermore, the WHO has in a recent publica­
infection (Peleg et al., 2008). So common were instances of infection tion considered carbapenem-resistant A. baumannii a priority 1 critical
from Acinetobacter spp., with cases as high as 33 % of all wound and bacteria in their list of bacteria, outscoring even the 2nd priority bac­
blood isolates, that it garnered the nickname ‘Iraqibacter’ (Petersen teria in terms of ranking by 9.3 % within their own predetermined
et al., 2007). Since then, outbreaks have been reported in all major criteria (Tacconelli et al., 2018).
continents (except for Antarctica), with cases cause by multi-drug Collectively, much attention has been directed on studying clinically
resistant isolates reaching as high as 44 %. This has led to poor pa­ relevant A. baumannii, with attention to its virulence and pathogenesis.
tient prognosis which further strains an already struggling global health These studies have illuminated a great deal of knowledge in under­
care infrastructure as our existing antimicrobial solutions are rendered standing the pathogen and what makes it so ‘successful’ with its diverse
ineffective (Dijkshoorn et al., 2007; Giammanco et al., 2017). repertoire of virulence factors, providing significant insights aimed at

* Corresponding author.
E-mail address: EngHwa.Wong@taylors.edu.my (E.H. Wong).

https://doi.org/10.1016/j.micres.2021.126722
Received 24 September 2020; Received in revised form 29 January 2021; Accepted 1 February 2021
Available online 4 February 2021
0944-5013/© 2021 Elsevier GmbH. This article is made available under the Elsevier license (http://www.elsevier.com/open-access/userlicense/1.0/).
H.J. Mea et al. Microbiological Research 247 (2021) 126722

exposing possible weaknesses and targets for the development of novel Table 1
treatment and preventive strategies. Special interest has also been given Identified list of virulence factors along with their roles in pathogenesis for
in understanding the pathogen’s ability to survive, persist and thrive in Acinetobacter baumannii.
diverse environmental conditions especially in clinical settings which is Virulence factor Proposed role in References
believed to be directly linked to its development of multi-drug resistance pathogenesis
and hospital outbreaks. This article seeks to summarize the current Pili Motility, adherence, Piepenbrink et al., 2016;
characterization of virulence factors (Table 1) contributing to patho­ biofilm development Tomaras et al., 2003
genesis focusing on its ability to move, adapt by adhering/attaching and Porin (OmpA, Omp33-36, Porins (OmpA, Omp33- Choi et al., 2005, 2007,
Omp22, CarO, OprD- 36, Omp22, CarO, 2008; Gaddy et al. 2009;
colonize any given environment via forming biofilm along with its reg­
like) OprD-like), host cell Kim et al., 2016; Kwon
ulatory mechanisms and current means of studying the complex host- death et al., 2019; Lee et al.,
pathogen interaction during the initial stages of infection. Special 2001; Smani et al.,
attention is also given to surface related features and proteins that 2012a, 2013a; Rumbo
contribute to its pathogenesis. et al., 2014.
Biofilm associated proteins Adherence; biofilm Brossard and
(BAP) development and Campagnari, 2011;
2. Virulence factors maturation Loehfelm et al., 2008
BAP like Proteins (BLP) Adherence De Gregorio et al., 2015
In the past decade, many have undertaken the task to unravel the Lipopolysaccharide (LPS) Adherence, Serum Bartholomew et al.,
resistance, survival in 2019; McQueary et al.,
unique and complex mechanisms that contribute to the successful
tissue infection, evasion 2012
emergence of A. baumannii to the clinical scene. Interestingly, most of the host immune
nosocomial pathogens like E. coli, P. Aeruginosa and S. Aureus have response
distinct molecular determinant or toxins primarily responsible for Penicillin-binding protein Adherence, serum Lee et al., 2008
causing disease. This has not been the case for A. baumannii, instead 7/8 and b-lactamase resistance, in vivo
PER-1 survival
bacterial pathogenesis has been attributed to a combination of factors Trimeric Autotransporter Adherence, biofilm Bentancor et al., 2012;
and an arsenal of virulence factors working in concert to cause an (Ata) development Thibau et al., 2019;
infection (Fig. 1). It begins with the surface of the bacterial cells which Weidensdorfer et al.,
being made up of a plethora of molecular components interacts with the 2016
FhaBC secretion system Adherence, host cell Pérez et al., 2016
environment. The current known list of virulence factors identified in
death
A. baumannii are summarized in Table 1. Capsular polysaccharide Adherence, growth in Russo et al., 2010;
serum, survival in tissue Lees-Miller et al., 2013
3. Bacterial pathogenesis infection, biofilm
development
Phospholipase (PLC and Serum resistance, Fiester et al., 2016
3.1. Initial stage of infection PLD) invasion, in vivo survival
Outer membrane vesicle Delivery of virulence (Jin et al., 2011).,Jun
For a bacterium to become pathogenic, contact must first be estab­ (OMV) factors, horizontal et al., 2013
lished with a given host surface through adherence before expressing transfer of resistance
gene
virulence factors for colonization (Falagas and Rafailidis, 2007). Here
Iron acquisition system In vivo survival, Fiester et al., 2016;
they transition from a planktonic free-swimming mode of growth to that (acinetobactin and NfuA) persistence, host cell Gaddy et al., 2012
of a surface-orientated one. It has been recognized that one of the major death
contributing factor to the rise of nosocomial A. baumannii stems from its Zinc acquisition system In vivo survival, Lonergan et al., 2019
ability to adhere and colonize biotic and abiotic surfaces, employing a (ZnuABC, ZigA & ZrlA) persistence
Manganese acquisition In vivo survival, Green et al., 2019
change of strategy currently dubbed ‘persist and resist’ rather than the
system (MumC and persistence, immune
traditional toxin expression of other pathogens. This, coupled with an MumT) evasion
equally confounding capacity to survive in normally unfavorable con­ Type II protein secretion In vivo survival Harding et al., 2016
ditions makes A. baumannii a formidable pathogen. system
Type VI protein secretion Killing of competing Repizo et al., 2015;
system bacteria, host Silverman et al., 2012
3.2. In search of a target - motility colonization
Tuf (plasminogen binding Immune evasion, Koenigs et al., 2015
Normally, bacterial cells exist in a planktonic free moving state. Here protein) complement system
they employ a variety of techniques to propel themselves around. It is supression
RecA Motility, In vivo Aranda et al., 2011;
ironic for Acinetobacter spp. to have received such a name as the original survival, biofilm Corral et al., 2020a
Greek word from which it is derived translates to mean nonmotile rod. development
This was initially proposed back in 1954 by Brisou and Prevot, 1954 who GigABCD In vivo survival, killing Gebhardt and Shuman,
attempted to distinguish the organisms based on its motility. This of host cells 2017
AdeRS Antimicrobial Montaña et al., 2015;
however is not the case for the two clinically relevant Acinetobacter
resistance, virulence Richmond et al., 2016;
species, A. baumannii and A. nosocomialis which have been demonstrated regulator Sun et al., 2017
to possess both surface-associated and twitching motility (Eijkelkamp BaeSR Antimicrobial Lin et al., 2014
et al., 2011a; Skiebe et al., 2011). The direct correlation between resistance, virulence
motility and ability to cause disease during the initial phase of infection regulator
BfmRS Csu Pili expression; Kim et al., 2019; Liou
has been established in other pathogens such as E. coli, Salmonella spp. virulence regulator et al., 2014
and P. aeruginosa (Josenhans and Suerbaum, 2002; Kao et al., 2014). CheAY Csu Pili expression; (Chen et al., 2017)
However, unlike other bacteria, A. baumannii along with other species of virulence regulator
Acinetobacter have been shown to lack flagella thus making the link GacAS and PaaE (two Neutrophil recruitment, Cerqueira et al., 2014;
component system & virulence regulator Bhuiyan et al., 2016
between motility and virulence difficult to establish.
phenylacetic acid
Twitching motility is a kind of jerky movement in a wet surface that metabolism)
is flagella-independent and has been characterized and studied in other (continued on next page)
genera like Pseudomonas, Neisseria, Myxococcus and Ralstonia (Corral

2
H.J. Mea et al. Microbiological Research 247 (2021) 126722

Table 1 (continued ) (Piepenbrink et al., 2016; Ronish et al., 2018). The same study also
Virulence factor Proposed role in References attributed TFP to promoting host cell adherence, which will be further
pathogenesis discussed in the next section.
OmpR/EnvZ Killing of host cells, (Tipton and Rather,
An alternative form of motility observed in A. baumannii clinical
virulence regulator 2017) isolates is called surface associated motility seen among bacteria on
PmrAB Antimicrobial Beceiro et al., 2011; Chin semi-solid surfaces. It is sometimes termed as ‘swarming’ motility and
resistance, LPS et al., 2015 was first characterize in Acinetobacter isolates by Barker and Maxted
modification
(1975). Initially thought to be directed by the type IV pili, a study by
Clemmer et al. (2011) showed that the loss of the pilT gene resulted in 54
et al., 2020a, 2020b; Mattick, 2002; Talà et al., 2019). It involves the use % motility reduction, with the residual being attributed to an alternative
of a functional type IV pili (TFP) that cycles between repeated rounds of form of motility. Another study by Wilharm et al. (2013) corroborated
extension and retraction that pulls the bacterial cells forward (Jarrell this finding and added that pilT inactivation can interfere but not abolish
and McBride, 2008; Mattick, 2002). Genes that have been identified to surface motility using a transformed mutant strain. Additionally, LPS
be involved in pili assembly are pilA, pilB, pilC, pilF, pilM, pilN, pilO, pilP, and K1 capsule have also been implicated to have a role in
pilQ, pilZ and pilW, along with genes involved in twitching pilR, pilS, pilT surface-associated motility though more work could be done in vali­
and pilU (Clemmer et al., 2011). Several studies have shown that most of dating this (McQueary et al., 2012). In an attempt to characterize a
the genes mentioned are present across different clonal groups of clin­ larger sample group, Skiebe et al. (2012) found that among 83 different
ically A. baumannii strains and show high levels of conservation, sug­ A. baumannii isolates all but one exhibited motility on a semi-solid
gesting a common feature among the species including the reference surface of 0.5 % agarose. This was challenged by other studies which
strain ATCC® 17978™ & 19606™ (Eijkelkamp et al., 2011a, 2011b; contradicted this claim (Clemmer et al., 2011; Eijkelkamp et al., 2011b),
Eijkelkamp et al., 2014; Piepenbrink et al., 2016). However, high vari­ although it should be noted the experimental conditions differ with one
ations have been recorded primarily in the fimbrial protein PilA which is study by Dahdouh et al. (2018) showing low surfaced associated motility
a major pilin subunit thought to contribute to the mechanism allowing among Spanish isolates but high motility in Lebanese isolates. These
for host immune system evasion. This variation is also observed when studies serve to highlight the need for a comprehensive and represen­
comparing within genera between A. baumannii and A. nosocomialis, tative study to characterize a large sample of A. baumannii clinical iso­
with surprising similarity among other bacterial species compared, lates which should aim at correlating virulence and motility.
implying that functional requirements determine the protein structure Interestingly, several studies have shown that a loss of motility

Fig. 1. A schematic diagram depicting the bacterial cell envelope and surface related virulence factors involved primarily in motility, adherence and biofilm for­
mation. These include Lipooligosaccharides & Lipopolysaccharides (LOS & LOS), Outer membrane protein A, Csu Pili, Type 4 pili (TFP), Biofilm associated protein
(BAP) and trimeric auto transporter (Ata).

3
H.J. Mea et al. Microbiological Research 247 (2021) 126722

resulted in a significant attenuation of the pathogen. An example of this hydrophilic strains. Interestingly, hydrophilic IC II strains adhered bet­
was found when two immotile mutant isolates were found to have had ter to lung epithelial cells and were more virulent in C. elegans and
their motility restored via addition of 1, 3-diaminopropane (DAP) in the murine infection model (Skerniškytė et al., 2019). One study also found
growth media. The mutants were found to have mutation transposon that a mutant A. baumannii strain that lacked a redox protein thioredoxin
insertions in dat and ddc genes preventing the biogenesis of DAP which A was more hydrophobic making it more susceptible to J774 macro­
also resulted in A. baumannii attenuation in a Galleria mellonella cater­ phage uptake which points to better bacterial clearance, although this
pillar infection model (Skiebe et al., 2012). Another example is by was not tested in an in vivo model (May et al., 2019). Both studies do
Pérez-Varela et al. (2017) where an ATCC® 17978™ mutant harboring however suggest that explorations into manipulating cell surface hy­
an amino acid substitution in position 520 or 540 of the RpoB protein drophobicity could lead to uncovering novel therapeutic strategies to
that conferred rifampin resistance was found to have impaired combat nosocomial A. baumannii infections highlighting another aspect
surface-associated motility which also affected the pathogen’s virulence of its pathogenesis.
capabilities in a C. elegans infection model. Other factors that have been
shown to affect motility include two-component system, quorum 3.4. Attachment and the initiation of infection
sensing, light, salinity, iron and lipopolysaccharides demonstrating the
close correlation between motility and extracellular conditions (Eijkel­ 3.4.1. Attachment/adherence
kamp et al., 2014; McQuery et al. 2012; Pérez-Varela et al., 2017; Skiebe In its fight for survival during this critical moment, A. baumannii
et al., 2012; Wood et al., 2018) must decide what to do next whether to remain in a free moving state or
Taken together this presents a promising route of study in the attach to a surface and start colonizing it. Usually, a major determining
development of novel therapeutic strategies specifically targeting factor is nutrient availability particularly low nutrient availability. This
motility against A. baumannii and may have applications for the entire sounds counter intuitive, however bacteria grown in high nutrient en­
genus as this trait seems to be consistent in other species. There are vironments either fail to form biofilm of form loose structures that are
however several challenges with this, one of which have already been easily disrupted by fluid shear (Petrova and Sauer, 2012). This is
highlighted above with regards to the type IV pili where the major pilin consistent for A. baumannii as a study found that under iron limiting
subunit possesses high variations seen in both amino acid sequence and conditions one of the most notable changes other than slow growth rates
glycosylation patterns (Ronish et al., 2018). Another would be was a reduction in motility specifically via the T4P and type I pili (Eij­
phase-variation first reported in 2015 which needs to be accounted for kelkamp et al., 2011b). A comprehensive transcriptome analysis
when studying bacterial virulence which could lead to conflicting results compared mRNA expression profiles of biofilm and planktonic cells
and ineffective therapeutic strategies prompting the need to revisit all found that there was a down-regulation in genes associated with motility
current A. baumannii studies to ascertain certainty (Ahmad et al., 2019; and an upregulation of genes associated with biofilm formation. These
Tipton, Dimitrova and Rather, 2015). same genes contribute to attachment and adherence as the first step in
establishing a mature biofilm (Rumbo-Feal et al., 2013). A combination
3.3. Surface-related features of these 2 approaches applying transcriptome profiles on a wide popu­
lation of A. baumannii strain would validate these correlations especially
Before adherence can take place the repulsive forces that exist be­ in the utilization of model strains representative of the current clinical
tween the surfaces of both bacterial and host target cells due to it being context. Collectively, these studies highlight the sequence of events that
negatively charged must be overcome. This is thought to be accomplish are essential for the pathogen to begin colonization starting with
mainly by establishing initially weak hydrophobic interactions that are attachment. It should be noted that there have emerge some reports that
reversible before more permanent fixtures are established (Rosenberg challenged this claim with clinically isolated A. baumannii lacking the
et al., 2013). Cell Surface hydrophobicity (CSH) is known to be an ability to adhere to A549 cells in an in-vitro study (Lázaro-Díez et al.,
important parameter that enables microorganisms to attach to hydro­ 2016). Hence, existing evidence should be viewed with caution, given
carbon found on surfaces and or cells, moving from water to organic the limitations of these studies which only serve to highlight the need for
phases in a given environment which aides in the organisms survival as a consistent model for virulence and pathogenicity studies on
it seeks for carbon sources (Krasowska and Sigler, 2014). Surfaces which A. baumannii.
are hydrophobic will give rise to adherence from hydrophobic cells and An early study on the subject found that A. baumannii attachment to
vice versa for hydrophilic surfaces. It should also be noted that micro­ abiotic surfaces such as plastic and glass was mediated by a pili assembly
organisms have the capacity to manipulate their cell surface hydro­ that was visualized under electron microscopy. Thorough investigation
phobicity depending on the environmental conditions and growth identified these pili to be the product of a chaperone-usher pili assembly
phases that they are in although this has yet to be confirm for Acineto­ that resembled that of other bacteria later named CsuA/BABCDE-
bacter spp. (khelissa et al., 2017; Vij et al., 2019). This is an important mediated pilus. This pilus has been found to be widely conserved and
area of concern when considering how the plethora of medical devices common in nearly all A. baumannii clinical isolates (Moriel et al., 2013;
and implants such as catheters, mechanical heart valves and respiratory Tomaras et al., 2003). The CsuA/BABCDE-mediated pilus consist of two
tubes are made of hydrophobic materials and have been shown to harbor subassemblies, a short thin tip fibrillum encoded by the csuE gene that is
microbe presence including biofilm-forming A. baumannii (Greene et al., mounted on second subassembly helical rod encoded by the csuA/B gene
2016a). Links between bacterial CSH affecting pathogenicity and sub­ (Pakharukova et al., 2017; Tomaras et al., 2003). They function as a
sequently virulence have been studied among various microorganisms surface recognition adhesion site and pili biogenesis respectively and are
with results varied showing that it may positively or negatively modu­ very much dependent on environmental conditions to regulate expres­
late virulence. Pertaining to A. baumannii, a previous study has shown sion (Hospenthal et al., 2017). Csu functional studies revealed that
that CSH correlated with increased adherence to abiotic surfaces which absence of CsuA/BABCDE-mediated pilus production resulted in the
then leads to biofilm formation explaining bacterial clinical persistence lack of attachment and biofilm-forming capacity of the mutant strain to
(Pour et al., 2011). In addition, a recent study which analyzed 36 clinical plastic. However, attachment was still present on eukaryotic cells,
strains of A. baumannii from both International Clone Lineage I and II (IC mainly surfaces of fungal filaments and A549 cells (Gaddy et al., 2009).
I & IC II) found IC I strains to be more hydrophobic than IC II in general Later studies however found that the CsuA/BABCDE-mediated pilus is
leading to the formation of conventional biofilm and adherence to not involved in the adherence and attachment of A. baumannii to host
abiotic (plastic) surfaces. This observation was also true when cells, in this case bronchial epithelial cells but is essential for biofilm
comparing within IC II clonal lineage cells with more hydrophobic IC II formation on abiotic surfaces (de Breij et al., 2010; Lee et al., 2006).
strains displaying better biofilm and adherence capacity than Therefore, the mechanism of attachment to host cells are independent of

4
H.J. Mea et al. Microbiological Research 247 (2021) 126722

CsuA/BABCDE-mediated pilus. For a more comprehensive review of the portion (Bartholomew et al., 2019). Functionally, the outer membrane
pilus biogenesis in Gram-negative bacteria the reader is referred to the allows for more transmembrane transport across compared to the
following article (Hospenthal et al., 2017). cytoplasmic membrane. This is mainly facilitated by porins which can
A. baumannii has also been found to produce biofilm-associated make up about 50 % of the outer membrane. Porins in A. baumannii also
protein (Bapab) which is part of the biofilm forming and maturation serve similar functions mainly controlling the movement of solutes
process first characterized in S. aureus. It is also discovered to play a across the OM but have also been shown to contribute to antimicrobial
critical role in adherence (Brossard and Campagnari, 2011; Loehfelm resistance and as a virulence factor which is common among most
et al., 2008). Initial sequence analysis revealed a repetitive structure gram-negative bacteria.
consistent with bacterial cell surface adhesins which are large surface The first porin to be identified and characterize is OmpA (first
proteins possessing core domain tandem repeats. Most identified Aci­ identified as Omp 38). It is a β-barrel porin with a 2-nm pore diameter
netobacter genus homologues have a 80–100 aa long Ig-like repeats and a C-terminal periplasmic globular extension which accommodates
which share structural and functional similarities but not similar pri­ molecules up to 500 Da in size (Sugawara and Nikaido, 2012). It is
mary sequence, hence displaying clear heterogeneity (De Gregorio et al., highly conserved among bacterial species and has also been found in
2015). This protein was found to be prevalent in most A. baumannii A. nosocomialis, which is another clinically relevant species of the family
strains, with one study reporting presence in all clinically relevant with a 92 % amino acid sequence similarity (Kim et al., 2016). This
strains along with an avirulent strain with the only exception from a protein has been found to have low permeability when compared with
community acquired strain (Eijkelkamp et al., 2014; Yang et al., 2019). similarly sized porins of other bacteria (e.g. OprF of E. coli) thus impli­
Similarly, all but one isolate from an Australian Hospital spanning a cating it in antibiotic resistance (Kwon et al., 2019; Sugawara and
decade were found to possess the bap gene (Goh et al., 2013). Bap has Nikaido, 2012). OmpA has also been discovered to contribute to bac­
been found to modulate the cell surface hydrophobicity by increasing it, terial virulence by adhering to host lung epithelial cells through direct
which results in improve observed adherence to epithelial cells (Bros­ interactions with fibronectin on cell surfaces (Gaddy et al., 2009; Smani
sard and Campagnari, 2011). Recently, two additional proteins were et al., 2012a, 2012b). This induces an inflammatory response which
discovered to share a large sequence motif at the NH2 terminus with BAP could also lead to cellular invasion and subsequent host cell death
subsequently called Bap-like proteins, designated BLP1 & BLP2. In one through the activation of caspase-3 as OmpA has been detected to
study, BLP1 has been found to be unevenly distributed in A. baumannii localize in both mitochondria and nuclei (Choi et al., 2005, 2007; 2008;
but present in other Acinetobacter spp. with a large sequence from 3044 Lee et al., 2001). In addition, OmpA has been found to constitute a large
up to 3356 aa. Whereas BLP2 is more conserve in A. baumannii and abundance in these outer membrane vesicles (OMV) secreted by
found in all ACB complexes tested with a smaller size of 728aa protein. A. baumannii, conferring upon it cytotoxic effects (Jin et al., 2011). This
Both proteins are co-expressed with BAP suggesting complimentary is further supported by clinical data that demonstrates high bacterial
functions with significant drop in adherence to A549 cells observed in OmpA expression directly correlates with increase mortality rates.
Δblp1 and Δblp2 mutant (De Gregorio et al., 2015). Conversely, attenuated phenotype was observed in murine and
In addition, other notable factors involved in attachment and G. mellonella models infected with ΔAbOmpA mutant clinical strains
adherence of A. baumannii is the expression of blaPER-1 a broad range (Sánchez-Encinales et al., 2017).
β-lactamase gene (Lee et al., 2008). The extent of involvement of this Other porins more recently discovered include Omp34 (previously
gene with adherence is still not fully established as studies regarding its known as omp33-36), which similar to OmpA also contributes to bac­
distribution among clinical isolates found that not all strains harbor the terial pathogenesis, adhering and inducing lung epithelial cell death via
gene. Although initially thought to contribute to biofilm formation, its activation of caspases (Rumbo et al., 2014; Smani et al., 2013). Adher­
positive correlations through prevalence studies have been under­ ence using Omp34 has also been attributed to be fibronectin-dependent
whelming (Bardbari et al., 2017; Yang et al., 2019). The Acinetobacter (Smani et al., 2012b). Another porin CarO, is responsible for carbape­
trimeric autotransporter adhesin (Ata) first identified in A. baumannii nem resistance in A. baumannii functions similarly to the other porins
ATCC® 17878™ was found to mediate bacterial adhesion to extracel­ also promoting cell adhesion. It is also capable of manipulating host cell
lular matrix proteins and forms an important virulence factor in most immune response by reducing NF-κB pathway expressing genes, ach­
Gram-negative bacterium (Bentancor et al., 2012; Weidensdorfer et al., ieved through suppressing IκBβ phosphorylation. The exact mechanism
2016). Ata deleted mutants were found to be significantly attenuated in is still uncertain but current observations suggest its role in bacterial
Galleria mellonella larvae models as adhesion was completely abolished. persistence within a given host (Sato et al., 2017; Zhang et al., 2020). To
In addition, Ata was found to induce strong immunogenic response the best of our knowledge, since most of the studied porins have high
demonstrating its critical role in the infection process and potential use immunogenic, cytotoxic effects and contribute to host cell adherence, it
as a target for vaccine and therapeutic development (Thibau et al., 2019; is no surprise that they have been the subject of intense vaccine target
Weidensdorfer et al., 2016). Some notable mentions include fibronectin, studies. Suffice to say, porins are a major virulent factor and are thus a
poly-β-1,6-N-acetylglucosamine (PNAG) and porins which will be dis­ prime target for controlling A. baumannii pathogenesis before and dur­
cussed below. It is also worth mentioning that many of the components ing infections. Particular attention could be paid to what governs porin
brought up thus far play multifaceted roles especially in initiating and expression as disruption have been seen to affect outer membrane sta­
supporting biofilm growth and maturation which is integral to bility and reduce antimicrobial resistance (Kwon et al., 2019)
A. baumannii’s pathogenesis to successfully colonize.
3.5. Two component regulatory systems
3.4.2. Porins/OMP
Another notable feature of A. baumannii are the outer membrane Much of the bacterial virulence factors stated in this review and used
proteins (OMP) that litter the surface of the outer membrane (OM). by A baumannii are regulated by two-component systems that directly
Surrounded by two concentric lipid bilayer membranes, gram negative affect the genetic expression of the pathogen in response to environ­
bacteria have a lipid component of the inner cytoplasmic membrane mental signals. This by itself is a large topic and deserves a separate
composed of phospholipids distributed across the inner and outer leaf­ review on its own, as such we will only briefly look into some common
lets (Fig. 1). The inner leaflet has a composition that is similar to the ones that directly affect the pathogenesis in the initial phases of infec­
cytoplasmic membrane. In contrast, the outer leaflet is asymmetric tion. Two component systems (TCS) are ubiquitous signal transduction
being made up of mainly lipopolysaccharides (LPS) (or lip­ systems that consist of sensor kinases embedded in the cytoplasmic
ooligosaccharides (LOS) in the case of A. baumannii due to the absence of membrane that sense surrounding extracellular and intracellular stimuli
o-polysaccharides) on the outer portion and lipid bilayer in the inner (Groisman, 2016). These sensors usually respond to environmental

5
H.J. Mea et al. Microbiological Research 247 (2021) 126722

physiological stimuli like pH, osmotic pressure and envelope stresses affect he pathogenesis of A. baumannii and effectively control infection
leading to the relaying of the sensor kinase via phosphorylation. This is presents an interesting area for further investigation.
followed by a conformational change of the transcription factor which
leads to DNA binding and transcription (Kröger et al., 2017). A feedback 3.6. Biofilm
mechanism is vital for the process to function optimally ensuring that
output is adjusted to reflect the organism’s needs at a given situation. Upon successful attachment to a host/surface, A. baumannii will
This is also evident in all two component systems which play a role in begin to express several virulence factors to colonize this new space by
not only stress response but, pathogenesis, symbiotic relations with host the formation of a biofilm. Some of those factors have already been
cells, antimicrobial synthesis with competing microbes and antibiotic discussed in the previous sections and will be further expounded here as
resistance (Groisman, 2016). they also play a significant role in biofilm formation and maturation.
Current studies into A. baumannii TCS have revealed seven systems This highly regulated event starts with the bacteria forming tertiary
that contribute to virulence. These include AdeRS, BaeSR, BfmRS, structures where close contact is made between each bacterial cell to be
CheAY, GacSA, ompR-envZ and PmrAB. AdeRS TCS controls the encased in an extracellular matrix that comprises of carbohydrates,
expression of three RND efflux pumps AdeABC, AdeFGH and AdeIJK; proteins, nuclei acids and various other macromolecules (Dufour et al.,
which have been characterized to be closely associated with conferring a 2010). Biofilms provide a means for the bacteria to survive in harsh
multidrug resistant phenotype against aminoglycosides, tetracyclines, environments and have also been associated with the pathogen’s ability
tigecycline, erythromycin, fluroquinones and chloramphenicol (Mon­ to resist various antimicrobial assaults occurring in three stages; early
taña et al., 2015). Studies have shown that AdeRS deletion resulted in development, matrix formation and maturation (Hoyle and Costerton,
more than 579 genes differentially expressed. These included genes 1991). Tests have shown that clinically isolated A. baumannii biofilm
related to virulence, most notably a drop in biofilm formation, antimi­ forming cells are able to survive desiccation twice as long when
crobial resistance, and host killing capacity during infection (Richmond compared to their non-biofilm forming counterparts. However, these
et al., 2016; Sun et al., 2017). Similarly, BaeSR also regulates expression same studies have found that the strong biofilm forming isolates seem to
of major efflux pumps, though this has been proposed to be of an indirect be less resistant to antimicrobial agents suggesting a certain trade-off
nature (Lin et al., 2014). BfmRS was discovered to affect the expression between antibiotic resistance and desiccation tolerance (Espinal, Marti
of CsuA/BABCDE, thus also affecting morphology, biofilm expression, & Vila 2011; Greene et al., 2016a). This seeming contradiction may be
attachment to both abiotic and biotic surfaces and microbial resistance. due to the use of different strains in these studies which highlights the
Disruption of BfmRS prevents host cell colonization and subsequent need to standardize and create a more consistent experimental designed
infection. Interest into this TCS as a potential target for antimicrobial starting with genetic profiling of the strains used.
treatment has yielded mixed findings. One example is the identification Indeed, biofilm production has been observed in A. baumannii and is
of the TCS’s role in bacterial capsule production and cell wall homeo­ considered a major virulence factor that greatly enhances its survival
stasis which when disrupted reduces bacterial survival in human serum and persistence. Consequently, A. baumannii biofilm have been found to
but subsequently increases host cell cytotoxicity due to an increase in be present on a whole host of medical devices such as catheters, endo­
OMV production (Kim et al., 2019; Liou et al., 2014). These studies also tracheal tubes of respirators, dialysis tube bottles, stainless steel, and
recorded increase bacterial resistance to several antibiotics, hence the polycarbonate surfaces (Gaddy and Actis 2009; Greene et al., 2016b).
role of BfmRS in modulating multiple virulence factors. CheAY, a more Greene et al. (2016b) showed that among 6 surfaces tested for
recently discovered TCS, has been implicated in affecting virulence A. baumannii biofilm growth, polycarbonate which is a durable, low-cost
through the decrease in homoserine lactone synthase expression. This plastic favored for its ability to undergo autoclaving was the most ac­
results in impaired cell motility and biofilm formation regulated by commodating surface for biofilm growth, making it an ideal reservoir.
CsuA/BABCDE chaperone/usher pili in an AbaI-dependent manner Additionally, one study found that despite long term desiccation and
(Chen et al., 2017). starvation up to 60 days, five clinical isolated strains of A. baumannii
PmrAB contributes to polymyxin B and colistin-resistance strains not were able to maintain virulence on surfaces such as plastic, glass and
just for A. baumannii but also in P. aeruginosa and K. pneumoniae (Beceiro even lab coats though at decreases culturability (Chapartegui-González
et al., 2011). This is believed to be possible through the regulation of et al., 2018). With such persistence within a hospital environment, it is
genes involved in lipopolysaccharide modification, with phosphoetha­ natural for the pathogen to be eventually the cause of numerous hospital
nolamine conferring colistin resistance that’s results from an increase in outbreaks documented over the decades (Dijkshoorn et al., 2007; Greene
bacterial surface charge that repels cationic polymyxins (Beceiro et al., et al., 2016b).
2011; Chin et al., 2015). Another more recently discovered TCS of Although, many studies have suggested that the formation of biofilm
A. baumannii is the ompR-envZ which regulates porin expression trig­ varies from strain to strain, some commonalities have been observed
gered by environmental osmolarity differences. Interestingly, a muta­ with most A. baumannii initiating biofilm formation through the
tion to the this TCS resulted in decreased G. mellonella virulence and expression of type I chaperone-usher pilus system. Studies have shown
drastically increased colony morphology switching frequency from that a large majority of clinical A. baumannii isolates can form biofilms,
opaque to translucent, which is an indication of phase variation (Tipton which is significantly higher when compared to environmental isolates
and Rather, 2017). First identified through a mutagenesis in a trans­ (Bardbari et al., 2017; Eijkelkamp et al., 2011a, 2011b). They also
poson screen ATCC® strain 19606™, the TCS GacAS sensor kinase GacS observed that strains from International Clone II usually form larger
was identified when the mutant was found to be unable to metabolize biofilm than their counterparts, begging the question of their correlation
citrate as a sole carbon source (Dorsey et al., 2002). Later revealed to be to pathogenicity. The Csu pili is interesting as it has dual roles in bac­
directly involved in bacterial virulence, GacAS deletion affected more terial adherence and biofilm formation. Its deactivation resulted in the
than 674 genes expression with a wide range of functions, including loss of adherence to abiotic surface and biofilm forming capabilities (de
motility (specifically type I pili) and the CsuA/BABCDE chaperone-usher Breij et al., 2009; Pakharukova et al., 2018). This however is not the case
pili assembly system, resulting in attenuated virulence in a mice septi­ with regards to interactions with biotic surfaces as adherence of Csu
cemia model (Cerqueira et al., 2014). Collectively, TCS are vital for mutants were still observed, suggesting an alternative means of adher­
bacterial responses to environmental changes, regulating gene expres­ ence (Gaddy & Actis 2009). This poses a challenge to therapeutic
sion in order to adapt and survive. Although substantial, studies into development aimed against the Csu pili as its usefulness would be
identifying similar trends in clinical strains are still needed in order to limited to controlling environmental and clinical devices colonization
validate TCS role in virulence as most of the studies quoted employed and not necessarily suited for infection control.
reference ATCC® strains. Targeting and disrupting TCS to consequently Part of the biofilm initiation strategy once attachment has been

6
H.J. Mea et al. Microbiological Research 247 (2021) 126722

established is the formation of extracellular polymeric substance (EPS) attenuated virulence profile. Therefore, although there still exists gap in
matrix that is composed of polysaccharides, extracellular DNA (eDNA), our knowledge of the biogenesis of A. baumannii biofilm formation, the
lipids and proteins. The EPS forms an irreversible surface attachment bulk of the work on biofilms has shown that it is an integral part of the
resulting in bacterial persistence (Petrova and Sauer, 2012). This has yet virulence and persistence strategy employed by the pathogen. As such, it
to be clearly defined and establish in A. baumannii studies. One of the follows reasonably that the biofilm has been targeted for the develop­
well documented polysaccharides that is produced by A. baumannii ment of novel therapeutic strategies.
during this process is poly-β-(1-6)-N-acetylglucosamine (PNAG) which is
common among other bacteria such as E.coli (Choi et al., 2009; Nait 4. Host-pathogen interaction studies
Chabane et al., 2014). Encoded by the pgaABCD gene cluster which
shares close homology with the pga locus of E.coli, PNAG produced by Due to the urgent need to understand the pathogenesis and virulence
the bacteria helps form the landscape of the biofilm which allows the of the bacteria especially in understanding its interaction with host
bacteria to attach facilitating surface-to-cell and cell-to-cell adhesion as during infection, studies employing host-pathogen interaction models
well as pellicle formation. Deletion of this gene resulted in bacteria that have been employed. It ranges from in vitro to in vivo models providing
could aggregate but not form multilayer three-dimensional structures the necessary means to evaluate novel treatments, characterize host
(Choi et al., 2009; Nait Chabane et al., 2014). In addition, upregulation immune response and identify novel bacterial virulence factors. Animal
of PNAG expression and its transporter was observed in a colistin models in particular have been the mainstay in studying A. baumannii
resistant strain which points to it being regulated by the PmrAB two pathogenesis especially due to the wide array of clinical manifestations
component system (Park et al., 2015). Another EPS confirmed to be perpetrated by the pathogen. Two major challenges exist when
release by A. baumannii is extracellular DNA (eDNA). Essentially, eDNA employing the use of animal models, primarily the ability to properly
is made up of the genomic DNA of the bacteria and found to be release in and accurately mimic human disease and the bacteria strains used when
three ways: (i) active release in a free form, (ii) encapsulation by conducting these interaction studies.
membrane vesicles and released (early stages of growth), and (iii) pas­
sive release during cell death and lysis (late stages of growth). eDNA 4.1. In-vivo models
results in biofilm augmentation as its introduction in cell media pro­
duces a larger biomass. Conversely, DNAase I presence leads to 4.1.1. Mammalian models
observable reduction in biofilm mass (Sahu et al., 2012). Though an The primary choice for mammalian in vivo studies has been to use
important component of biofilm formation, it is unclear precisely how murine models with some studies employing other mammals such as
does eDNA contribute or regulate this process. With reference to rabbits and guinea pigs although this is rather rare (Pachón-Ibáñez et al.,
P. aeruginosa, eDNA was observed to act as a scaffold, intracellular 2010). These studies were initially carried out to evaluate the antibiotic
connector and molecular linker due to it being polyionic (Whitchurch efficacy during Acinetobacter infection but has since shifted to investi­
et al., 2002; Yang et al., 2007). Similar connections could be made that gate bacterial pathogenesis, host response & immunity along with new
require further investigations into the role of the various biofilm matrix alternative therapeutic strategies (Bentancor et al., 2012; Cerqueira
components, many of which have yet to be studied in A. baumannii. et al., 2014; Jolly-Guillou et al., 1997; Sánchez-Encinales et al., 2017;
As the biofilm continues to develop and mature, the process is Skerniškytė et al., 2019). Although, these studies could never realisti­
believed to be linked to a ubiquitous intracellular messenger signaling cally reproduce human disease infection process, they have nonetheless
molecule cyclic-di-GMP, a cyclic-di-nucleotide recently identified in provided a wealth of data to glean in our understanding of bacterial
A. baumannii regulating biofilm formation and surface associated pathogenesis, providing a clearer picture from whence to approach
motility (Ahmad et al., 2020). Cyclic-di-GMP and cyclic-di-AMP have effective infection control. In addition, the wide range and availability of
been identified in other bacteria to be an allosteric activator of exopo­ murine models coupled with cost and diverse use case solidifies it as a
lysaccharide biosynthesis which directly contributes to virulence and staple infection model of choice. As A. baumannii can cause quite a broad
biofilm expression of Vibrio cholerae and Pseudomonas aeruginosa, mak­ range of infections, it is imperative that a suitable selection of murine
ing its disruption a viable option in therapeutic development (Pestrak strains be conducted to best acquire the closest resemblance to infections
and Wozniak, 2020; Young et al., 2020). Furthermore, a key phenom­ in humans. Some more common inbred and outbred strains which have
enon that helps sustain the biofilm is quorum sensing (QS). Quorum been successfully used include A/J, BALB/c, C57BL/6 and Swiss
sensing is the mode of communication shared among the bacteria to Webster mice. Whichever strain chosen can affect the outcome of the
maintain the biofilm and population density via signal molecules called infection studies, where similar ATCC® bacteria strains when used re­
auto inducers. Changes in expression and concentration in these auto sults in differences to mice survival, tissue load and clearance and im­
inducers as the biofilm matures through the increase in bacterial pop­ mune response (Álvarez-Fraga et al., 2018; Palmer et al., 2019).
ulation results in signal transduction cascade that informs the bacteria
on how to best adapt (Saipriya et al., 2019). One well documented 4.1.2. Pneumonia model
means by which this is achieved is through the expression of N-acyl-­ As pneumonia represents the highest clinical manifestation of
homoserine lactones (AHL), which is an auto inducer signal molecule. A. baumannii infections, lung infection studies using murine models have
The QS occurs when AbaI, a sensor protein, functions as a synthase to also steadily increased. The first study to have used this model was
produce AHL signal molecule that bind with AbaR receptor to produce a performed in 1997 by Joly-Guillou and colleagues testing the efficacy of
signal cascade. CsuA/BABCDE chaperone-usher system expression was imipenem against acute A. baumannii pneumonia. The goal of this model
also upregulated when bacterial cells were cultured with non-native is to introduce bacteria into the mouse in order to induce inflammatory
AHLs, increasing biofilm density (Luo et al., 2015). responses consistent with acute pneumonia. From there researchers are
Some previously mentioned contributors in the development and also able to evaluate the virulence expression of the pathogen and have
maturation of biofilm includes: blaPER-1 gene where positive expression made some interesting discoveries. So far, we identified two approaches
resulted in an increase in biofilm formation (Lee et al., 2008); Biofilm to achieve this immune response: intra-tracheal and intranasal in­
associated protein (BAP) found to be widely present in many troductions of bacteria. The former involves using a blunt ended needle
A. baumannii strains (Goh et al., 2013) and biofilm like proteins (BLP) to cannulate the trachea of an anesthetized mice inoculated with bac­
whose gene disruption lead to impaired biofilm formation (De Gregario terial suspension, while the latter involves bacterial suspension is
et al., 2015). Overall, many virulence factors play a multi-concerted role directly applied to the nostrils as the mouse inhales breathing the sus­
of both adhesion and biofilm formation, with any disruption of the pension in (Joly-Guillou et al., 1997; Palmer et al., 2019). Usually, these
former leading to the absence of biofilm formation and thus an models struggle to induce a proper infection in immunocompetent mice

7
H.J. Mea et al. Microbiological Research 247 (2021) 126722

which leads to a difficulty in mimicking the actual disease as seen in (amoeba), Galleria mellonella (greater wax moth) and Danio rerio (zebra
clinical settings. This is usually overcome by inducing neutropenia or fish).
using neutropenic mice (Harris et al., 2013). However, some studies Caenorhabditis elegans is a soil inhabitant nematode that is micro­
have proposed that this step could be omitted if a sufficiently virulent scopic in size with a length of approximately 1 mm when fully matured.
strain of bacteria is used, giving us greater insight into the pathogenesis It has a short generation time of approximately 3 days and is able to
of the bacteria (Harris et al., 2013). reproduce up to 400 progenies from a single adult as it is a hermaph­
Studies have also shown the usefulness of pneumonia models in rodite. As a result of its well characterized genome, anatomical
highlighting the host-response to A. baumannii infections. This was simplicity and transparent body, C. elegans is an appealing host for
achieved by using transgenic mice with well-defined genetic changes infection studies as seen in studying bacterial pathogenesis of Salmonella
which allows for more specific characterization of host response on a enterica, Listeria monocytogenes, Burkholderia pseudomallei and Legionella
molecular level. One such example was the knockout of CD14 and Toll- pneumophila (Anju et al., 2020; Brassinga and Sifri, 2019). Though
like receptors 2 and 4 resulting in a difficulty in bacterial clearance, simpler in nature, the innate immune system of the nematode has sur­
which highlights the role played by these receptors effective immune prising similarities with that of humans. Seen primarily in its conserve
response (Knapp et al., 2006). Another study using gp91phox− /− and immune regulators that facilitate host’ identification of pathogens at the
NOS− /− mice shows the essential role of NADPH phagocyte oxidase and onset of infection. This allows for anti-infective agents screening which,
NOS2 has in limiting bacterial growth and replication (Qiu et al., 2008). if found to be protective to the nematode, is useful as a potential com­
While using transgenic mice have shown great potential, as seen in these pound for further animal trials and subsequent human trials
examples, it has yet to be widely adopted in studying A. baumannii (Skerniškytė et al., 2019). The first describe use of C. elegans to study A
pneumonia model. In addition, pneumonia model has also been used to baumannii was by Smith et al. (2004) which investigated the effects of
study the effectiveness of vaccines which saw immunization of immu­ ethanol as a signaling molecule for various Acinetobacter spp. That study
nocompetent mice prior to pneumonia infection leading to a drop in found that ethanol positively triggered A. baumannii virulence which
bacterial load post infection when compared to the non-immunized negatively affected worm proliferation and brood size. Since then,
group (Bentancor et al., 2012). C. elegans have also been adopted to study eukaryote and prokaryote
relationships and anti-virulence characteristics of various compounds
4.1.3. Sepsis models (Khadke et al., 2019; Raorane et al., 2019). Interestingly, one study
Bacteremia represents the second most common clinical manifesta­ modified Smith’s work and demonstrated the successful use of C. elegans
tion of nosocomial A. baumannii infections mainly triggered by host toll in A. baumannii pathogenesis study. Concluding that fertility assay was
-like receptor 4 (TLR4) activation by bacterial lipopolysaccharide (LPS). more accurate than killing assay and validated by intraperitoneal in­
In severe cases, this could be a result of bacterial dissemination into the jection murine sepsis model (Vallejo et al., 2015) The only disadvantage
bloodstream from an initial pneumonic infection. This process has been is the incubation temperature of C. elegans is relatively low at 25 ◦ C
successfully demonstrated using murine pneumonia models where a compared to the usual growth temperature of A baumannii at 37 ◦ C. This
depletion of alveolar macrophages and neutrophils resulted in increased could affect its virulence and survivability translating into a possible
morbidity and mortality with extrapulmonary bacterial dissemination discrepancy.
consistent with sepsis (de Breij et al., 2012 & Lee et al., 2020) To date, Used alongside C. elegans, Dictyostelium discoideum is a unicellular
sepsis models also employed the use of similar murine strains as used in amoeba that has also been proposed mainly for its ability to feed on
pneumonia models which generally involved the use of two main ap­ bacteria by phagocytosis, an ability which lends itself to being used as a
proaches. The first and most widely adopted model is the murine peri­ model for human macrophage-bacterial interactions (Anju et al., 2020).
toneal sepsis model which saw extensive used in other pathogens such as One of the major advantages is its ability to also grow at 37 ◦ C, allowing
K. pneumoniae and later adopted in A. baumannii antibiotic and subse­ for greater relevance to actual bacterial pathogenesis in human host.
quently virulence studies (Ko et al., 2004; Smani et al., 2013; Tsai et al., One study aimed at identifying mutant A. baumannii strains, employed
2011). This model has been used extensively to establish minimum le­ the coculturing of amoeba with bacteria. Since bacteria grows faster
thal doses (MLDs) part of characterizing the many clinical strains iso­ than the amoeba, it was able to form colonies first before consumption
lated for pathogenesis and virulence studies which when compared to by amoeba which forms visible plaques on the bacterial lawn. Bacteria
ATCC® reference strains help to highlight the evolution undertaken by with plaques were successfully identified as mutant A. baumannii which
the pathogen through the decades. A rarer approach involves the use of lack certain virulence factors allowing for rapid screening of transposon
inducing systemic infection via introducing the pathogen retro-orbitally mutant libraries for future pathogenesis studies (Smith et al., 2007). This
which has the added advantage of reducing complications and distress provides an interesting opportunity for rapid screening of large pools of
from the animal though a highly skilled operator is required (Yardeni clinical isolates collected across countries around world for virulence
et al., 2011). This approached has been successfully used to study the characterization.
role of TLR9 signaling in response to A. baumannii infection in both Galleria mellonella is the caterpillar of the greater wax moth and has
pneumonic and systemic infection (Noto et al., 2015) been utilized in host-entomopathogenic microbe interaction studies
prior to its utilization as a model for human pathogens. Its low cost,
4.1.4. Non-mammalian models convenience and ethically acceptable ranking has made it rather
Interestingly, although mammalian models still provide the best favorable for use as a model, in addition to its ability to grow well at 37
means to study host pathogen interaction, interest in non-mammalian ◦
C. It has been used extensively in several other pathogenic bacteria,
models has recently grown as they offer several advantages over their both gram positive and negative species with substantial utility in
mammalian counterparts. Firstly, non-mammalian models are not A. baumannii studies as well (Anju et al., 2020; Peleg et al., 2009). Its
bound by ethical restrictions. Next, compared to mammalian models complex and unique immune system shares some similarities with the
which require costly holding facilities, non-mammalian models are mammalian immune system, specifically the presence of phagocytic
relatively cheaper. These factors allow researchers to employ large cells, antimicrobial peptides, encapsulation and clotting factors and
quantities of organisms in any given study, facilitating high-throughput complement like proteins (Tsai et al., 2016). Its introduction as a host for
screening for bacterial virulence factors, toxins and general bacterial A. baumannii was demonstrated by Peleg (2009) and colleagues where
virulence. Such studies can utilize not only large number of host but a they demonstrated the direct correlation of host death to bacterial load
diverse range of bacterial strains which would expedite the character­ and prolonged survival when antibiotics was administered. Since then,
ization of clinical strains as they emerge. Some of the models used thus many more studies have incorporated this model in studies involving
far include Caenorhabditis elegans (nematode), Dictyostelium discoideum antimicrobial efficacy, bacterial virulence expression and pathogenicity

8
H.J. Mea et al. Microbiological Research 247 (2021) 126722

(Gaddy et al., 2012; Richmond et al., 2016; Sánchez-Encinales et al., Recently, infection studies involving in-vitro models have seen the
2017; Skiebe et al., 2012). use of a host cell line co-cultured with immune cells. It has been
Danio rerio, more commonly known as the zebrafish is a more recent established using murine models that immune cells are critical during
host-pathogen model with limited uses in A. baumannii infection studies. the early stages of infection, producing pro-inflammatory cytokines such
Favored for its quick breeding and development with innate immune as TNF-α, IL-6, MCP-1 and MIP-2 in the lungs which supports the
cells appearing 25 h post fertilization. Studies into zebrafish have pri­ clearance of A. baumannii infection (van Faassen et al., 2007; de Breij
marily been focused on high throughput screening of drugs meant for et al., 2012). In-vitro studies utilizing just immune cells such as those
cancer, inflammatory diseases and infectious diseases due to some carried out by Qiu et al. (2012), help shed light on the important role
orthologous genes shared with mammals (van der Sar et al., 2004). played by these immune cells which are part of the innate immune
Many studies have successfully used transgenic zebrafish with fluores­ system’s first response to infection with phagocytosis observed within
cent tags attached to various immune cells allowing for greater in-depth 10 min of co-incubation. Interestingly, a later study incorporated both
visualization of the immune response during infection (Tobin et al., macrophage and neutrophil co-culture with either A. baumannii or
2012; Zhang et al., 2019)). Thus far, one study which utilized Zebrafish A. pittii clinical and reference strains show that neutrophils are more
showed that A. baumannii mutant lacking ΔgacS led to neutrophils crucial in in controlling infection with it being more effective at killing
migrating to the site of infection but failed to migrate away, suggesting bacteria and essential for the recruitment and activation of macrophages
that there exists a bacterial driven metabolic by-product that was (Lázaro-Díez et al., 2017).The same research group further expanded on
responsible as a neutrophil chemotaxis. Bhuiyan et al. (2016) was able this by elucidating the neutrophil’s response to A. baumannii infection
to study the migration pattern using fluorophore-marked bacteria and through gene expression and cytokine production assays uncovering the
selective fluorescent immune cells to track their movements and in­ upregulation and production of key pro-inflammatory mediators crucial
teractions within the host at real-time. This provides a whole new for effective infection control (Lázaro-Díez et al., 2020). These advances
avenue of study that can shed greater light on both host immune demonstrate the growing knowledge and understanding of the not only
response and bacterial immune evasion simultaneously. It should be the pathogen but also host response which is a step in the right direction
noted that studies employing these models cannot stand alone and and is critical for future translation to novel therapeutic strategies. Ul­
mammalian models will remain the mainstay approach to studying timately, cell lines provide important information not immediately
A. baumannii virulence and pathogenesis. It can, however, serve to accessible from in vivo or clinical observations by limiting the scope to
provide rapid bacterial characterization of various clinical strains and molecular mechanisms expressed by the host and pathogen to establish a
evaluation of novel antimicrobial efficacy. successful infection. Providing useful data for understanding the infec­
tion process starting from the lungs extending to bacteremia, which to
4.2. In-vitro model the best of our knowledge remains an untapped avenue for research.

In addition to in vivo models, many studies aimed at elucidating 5. Conclusion future perspective
bacterial virulence and pathogenesis have also utilized in vitro models to
get a preliminary glimpse into the complex host-pathogen interactions. This review has shown the considerable advancement made in un­
Being simpler and more cost-effective, these studies have provided the derstanding the pathogenesis and virulence characteristics harbored by
basis on which many in vivo studies may take place with complementing A. baumannii in the initial stages of infection, but also comments on
data to substantiate any findings. Since pathogens first contact with a some gaps that still exist. Much of the advances stem from the adoption
given host upon successful entry is with the epithelial mucous mem­ of many notable in vivo and in vitro models that allowed for an in-depth
brane cells, it has become the first choice in most in vitro studies. This is study and characterization of various clinical strains isolated. This
true for a broad range of diseases and surfaces on the human body and coupled with a large catalogue of over 2000 genome sequence publicly
evaluating this interaction provides vital information in terms of path­ available, positions researchers to be more readily able to do large
ogen virulence properties, immunomodulation and antimicrobial ac­ comparative genomic studies in mapping the bacteria’s core genome
tivities in the given context as the infection progresses (Pedersen et al., and compare the variations that exist around the world in terms of
2018). virulence and pathogenicity. Additionally, categorization studies to
Most of the in vitro studies involved use epithelial cells of a given part identify which clonal lineages the many strains belong to could serve to
of the body co-infected with a bacterial strain specific to the site of expand our understanding of the spread of the pathogen along with its
infection. This is first to first assess the adherence and cytotoxicity evolution in acquiring multi-drug resistance. With evidence of genome
properties of the pathogen. Comparison is usually made between plasticity arising in this pathogen, the clock is ticking as we encounter an
reference ATCC® and clinically isolated contemporary strains which increasing number of pan-drug resistant A. baumannii strains where
provides important phenotypic information that can be further explored current existing antimicrobial agents are no longer capable at treating its
through comparative genomic sequence analysis. A. baumannii adher­ infections (Traglia et al., 2018). Thankfully, the current employment of
ence and biofilm forming characteristics were identified in similar ATCC® 17978™ and ®ATCC 19606™ as reference standards serve well
fashion which led to future studies employing similar in vitro models in as controls in many comparative studies. However, they do highlight the
order to assess the virulence of clinically isolated strains (Tomaras et al., need for consistency in identifying and characterizing clinically relevant
2003). In vitro models also provide useful initial host immunological strains using well-established combined in vivo and in vitro models and
response to the presence of pathogens as co-infection and bacterial procedures highlighted in this review. With such a coordinated effort,
adherence to the host triggers an inflammatory response aimed the direct comparison between studies would no doubt pave a promising
recruiting immune cells necessary for bacterial clearance. Though most future as new innovative techniques continue to develop and optimize
of the immune response to a foreign pathogen follows a general pattern, allowing for a greater understanding of the pathogen, spearheading
some unique bacterial surface proteins are capable of elucidating a development of more effective novel therapeutic strategies.
varied response that give clues to the molecular mechanism behind
bacterial pathogenesis which can vary depending on the site of infection Author contributions
(March et al., 2010; Vijayakumar et al., 2016). This variation can be
explained by the need of the bacteria to adapt to a given environment, Hing Jian Mea carried out the literature search, planning and writing
expressing virulence specifically catered to its needs for survival as can of the manuscript draft. Eng Hwa Wong and Phelim Voon Chen Yong
be seen in biofilm and motility variations observed in A. baumannii edited and contributed to the structure and designed. All authors have
isolated from sputum or the blood (Vijayakumar et al., 2016). reviewed and approved the final manuscript draft before submission.

9
H.J. Mea et al. Microbiological Research 247 (2021) 126722

Funding Choi, C., Lee, E., Lee, Y., Park, T., Kim, H., Hyun, S., et al., 2005. Outer membrane
protein 38 of Acinetobacter baumannii localizes to the mitochondria and induces
apoptosis of epithelial cells. Cell. Microbiol. 7 (8), 1127–1138.
This study was financially supported by the Taylor’s University Choi, C., Hyun, S., Lee, J., Lee, J., Lee, Y., Kim, S., et al., 2007. Acinetobacter baumannii
Research Grant – Scheme Project No: TRGS/MFS/2017/SBS/003; outer membrane protein A targets the nucleus and induces cytotoxicity. Cell.
TRGS/MFS/2/2014/SOM/005. The funders had no role in the design, Microbiol. 10 (2), 309–319. https://doi.org/10.1111/j.1462-5822.2007.01041.x.
Choi, C., Lee, J., Lee, Y., Park, T., Lee, J., 2008. Acinetobacter baumannii invades epithelial
collection, and analysis of the data, manuscript draft preparation, or cells and outer membrane protein A mediates interactions with epithelial cells. BMC
decision to publish. Microbiol. 8 (1), 216.
Choi, A., Slamti, L., Avci, F., Pier, G., Maira-Litrán, T., 2009. The pgaABCD Locus of
Acinetobacter baumannii Encodes the Production of Poly-β-1-6-N-
Declaration of Competing Interest Acetylglucosamine, Which Is Critical for Biofilm Formation. J. Bacteriol. 191 (19),
5953–5963.
Clemmer, K., Bonomo, R., Rather, P., 2011. Genetic analysis of surface motility in
The authors report no declarations of interest. Acinetobacter baumannii. Microbiology 157 (9), 2534–2544.
Corral, J., Pérez-Varela, M., Barbé, J., Aranda, J., 2020a. Direct interaction between
Acknowledgements RecA and a CheW-like protein is required for surface-associated motility, chemotaxis
and the full virulence of Acinetobacter baumannii strain ATCC 17978. Virulence 11
(1), 315–326.
This review was supported by Taylor’s University Research Grant – Corral, J., Sebastià, P., Coll, N., Barbé, J., Aranda, J., Valls, M., 2020b. Twitching and
Scheme Project No: TRGS/MFS/2017/SBS/003; TRGS/MFS/2/2014/ swimming motility play a role in Ralstonia solanacearum pathogenicity. Msphere 5
(2).
SOM/005. Dahdouh, E., Suarez Rodriguez, M., Daoud, Z., 2018. Comparison of virulence
determinants among Acinetobacter baumannii Clinical isolates obtained from Spain
References and Lebanon. J. Infect. Dev. 12 (02.1), 15S.
de Breij, A., Gaddy, J., van der Meer, J., Koning, R., Koster, A., van den Broek, P., et al.,
2009. CsuA/BABCDE-dependent pili are not involved in the adherence of
Ahmad, I., Karah, N., Nadeem, A., Wai, S., Uhlin, B., 2019. Analysis of colony phase
Acinetobacter baumannii ATCC19606T to human airway epithelial cells and their
variation switch in Acinetobacter baumannii clinical isolates. PLoS One 14 (1) p.
inflammatory response. Res. Microbiol. 160 (3), 213–218.
e0210082.
de Breij, A., Dijkshoorn, L., Lagendijk, E., van der Meer, J., Koster, A., Bloemberg, G.,
Ahmad, I., Nygren, E., Khalid, F., Myint, S., Uhlin, B., 2020. A Cyclic-di-GMP signalling
et al., 2010. Do biofilm formation and interactions with human cells explain the
network regulates biofilm formation and surface associated motility of Acinetobacter
clinical success of Acinetobacter baumannii? PLoS One 5 (5) p. e10732.
baumannii 17978. Sci. Rep. 10 (1).
de Breij, A., Eveillard, M., Dijkshoorn, L., van den Broek, P., Nibbering, P., Joly-
Álvarez-Fraga, L., Vázquez-Ucha, J., Martínez-Guitián, M., Vallejo, J., Bou, G.,
Guillou, M., 2012. Differences in Acinetobacter baumannii strains and host innate
Beceiro, A., Poza, M., 2018. Pneumonia infection in mice reveals the involvement of
immune response determine morbidity and mortality in experimental pneumonia.
the feoA gene in the pathogenesis of Acinetobacter baumannii. Virulence 9 (1),
PLoS One 7 (2) p. e30673.
496–509.
De Gregorio, E., Del Franco, M., Martinucci, M., Roscetto, E., Zarrilli, R., Di Nocera, P.,
Anju, V., Siddhardha, B., Dyavaiah, M., 2020. Animal models to understand
2015. Biofilm-associated proteins: news from Acinetobacter. BMC Genomics 16 (1).
Host–Pathogen interactions. Model Organisms For Microbial Pathogenesis. Biofilm
Dijkshoorn, L., Nemec, A., Seifert, H., 2007. An increasing threat in hospitals: multidrug-
Formation And Antimicrobial Drug Discovery, pp. 393–411.
resistant Acinetobacter baumannii. Nat. Rev. Microbiol. 5 (12), 939–951.
Aranda, J., Bardina, C., Beceiro, A., Rumbo, S., Cabral, M., Barbe, J., Bou, G., 2011.
Dorsey, C., Tomaras, A., Actis, L., 2002. Genetic and phenotypic analysis of
Acinetobacter baumannii RecA protein in repair of dNA damage, antimicrobial
Acinetobacter baumannii insertion derivatives generated with a transposome
resistance, general stress response, and virulence. J. Bacteriol. 193 (15), 3740–3747.
system. Appl. Environ. Microbiol. 68 (12), 6353–6360.
Bardbari, A., Arabestani, M., Karami, M., Keramat, F., Alikhani, M., Bagheri, K., 2017.
Dufour, D., Leung, V., Lévesque, C., 2010. Bacterial biofilm: structure, function, and
Correlation between ability of biofilm formation with their responsible genes and
antimicrobial resistance. Endod. Topics 22 (1), 2–16.
MDR patterns in clinical and environmental Acinetobacter baumannii isolates.
Eijkelkamp, B., Hassan, K., Paulsen, I., Brown, M., 2011a. Investigation of the human
Microb. Pathog. 108, 122–128.
pathogen Acinetobacter baumannii under iron limiting conditions. BMC Genomics
Barker, J., Maxted, H., 1975. Observations on the growth and movement of acinetobacter
12 (1).
on semi-solid media. J. Med. Microbiol. 8 (3), 443–446.
Eijkelkamp, B., Stroeher, U., Hassan, K., Papadimitrious, M., Paulsen, I., Brown, M.,
Bartholomew, T., Kidd, T., Sá Pessoa, J., Conde Álvarez, R., Bengoechea, J., 2019. 2-
2011b. Adherence and motility characteristics of clinical Acinetobacter baumannii
Hydroxylation of Acinetobacter baumannii lipid a contributes to virulence. Infect.
isolates. FEMS Microbiol. Lett. 323 (1), 44–51.
Immun. 87 (4).
Eijkelkamp, B., Stroeher, U., Hassan, K., Paulsen, I., Brown, M., 2014. Comparative
Beceiro, A., Llobet, E., Aranda, J., Bengoechea, J., Doumith, M., Hornsey, M., et al.,
analysis of surface-exposed virulence factors of Acinetobacter baumannii. BMC
2011. Phosphoethanolamine modification of lipid A in colistin-resistant variants of
Genomics 15 (1), 1020.
Acinetobacter baumannii mediated by the pmrAB two-component regulatory system.
Falagas, M., Rafailidis, P., 2007. Attributable mortality of Acinetobacter baumannii: no
Antimicrob. Agents Chemother. 55 (7), 3370–3379.
longer a controversial issue. Crit. Care 11 (3), 134.
Bentancor, L., Camacho-Peiro, A., Bozkurt-Guzel, C., Pier, G., Maira-Litran, T., 2012.
Fiester, S., Arivett, B., Schmidt, R., Beckett, A., Ticak, T., Carrier, M., et al., 2016. Iron-
Identification of Ata, a multifunctional trimeric autotransporter of Acinetobacter
regulated phospholipase C activity contributes to the cytolytic activity and virulence
baumannii. J. Bacteriol. 194 (15), 3950–3960.
of Acinetobacter baumannii. PLoS One 11 (11) p. e0167068.
Bhuiyan, M., Ellett, F., Murray, G., Kostoulias, X., Cerqueira, G., Schulze, K., et al., 2016.
Gaddy, J., Arivett, B., McConnell, M., López-Rojas, R., Pachón, J., Actis, L., 2012. Role of
Acinetobacter baumannii phenylacetic acid metabolism influences infection
acinetobactin-mediated Iron acquisition functions in the interaction of Acinetobacter
outcome through a direct effect on neutrophil chemotaxis. Proc. Natl. Acad. Sci. 113
baumannii strain ATCC 19606Twith human lung epithelial cells, galleria mellonella
(34), 9599–9604.
caterpillars, and mice. Infect. Immun. 80 (3), 1015–1024.
Boucher, H., Talbot, G., Bradley, J., Edwards, J., Gilbert, D., Rice, L., et al., 2009. Bad
Gebhardt, M., Shuman, H., 2017. GigA and GigB are master regulators of antibiotic
bugs, No drugs: No ESKAPE! An update from the infectious diseases society of
resistance, stress responses, and virulence in Acinetobacter baumannii. J. Bacteriol.
america. Clin. Infect. Dis. 48 (1), 1–12.
199 (10).
Brassinga, A., Sifri, C., 2019. The Caenorhabditis elegans model of Legionella infection.
Giammanco, A., Calà, C., Fasciana, T., Dowzicky, M., 2017. Global assessment of the
Methods Mol. Biol. 371–397.
activity of tigecycline against multidrug-resistant gram-negative pathogens between
Brisou, J., Prevot, A., 1954. Studies on bacterial taxonomy. X. The revision of species
2004 and 2014 as part of the tigecycline evaluation and surveillance trial. Msphere 2
under Acromobacter group. Ann. Inst. Pasteur (Paris) 86 (6), 722–728.
(1).
Brossard, K., Campagnari, A., 2011. The Acinetobacter baumannii biofilm-associated
Goh, H., Beatson, S., Totsika, M., Moriel, D., Phan, M., Szubert, J., et al., 2013. Molecular
protein plays a role in adherence to human epithelial cells. Infect. Immun. 80 (1),
analysis of the Acinetobacter baumannii biofilm-associated protein. Appl. Environ.
228–233.
Microbiol. 79 (21), 6535–6543.
Centre of Disease Control and Prevention, 2019. Antibiotic Threats in the United States.
Green, E., Juttukonda, L., Skaar, E., 2019. The manganese-responsive transcriptional
Cerqueira, G., Kostoulias, X., Khoo, C., Aibinu, I., Qu, Y., Traven, A., Peleg, A., 2014.
regulator MumR protects Acinetobacter baumannii from oxidative stress. Infect.
A global virulence regulator in Acinetobacter baumannii and its control of the
Immun. 88 (3).
phenylacetic acid catabolic pathway. J. Infect. Dis. 210 (1), 46–55.
Greene, C., Vadlamudi, G., Newton, D., Foxman, B., Xi, C., 2016a. The influence of
Chapartegui-González, I., Lázaro-Díez, M., Bravo, Z., Navas, J., Icardo, J., Ramos-
biofilm formation and multidrug resistance on environmental survival of clinical and
Vivas, J., 2018. Acinetobacter baumannii maintains its virulence after long-time
environmental isolates of Acinetobacter baumannii. Am. J. Infect. Control 44 (5),
starvation. PLoS One 13 (8) p. e0201961.
e65–e71.
Chen, R., Lv, R., Wang, M., Du, Z., Tan, Y., Cui, Y., 2017. A1S_2811, a CheA/Y-like
Greene, C., Wu, J., Rickard, A., Xi, C., 2016b. Evaluation of the ability of Acinetobacter
hybrid two-component regulator from Acinetobacter baumannii ATCC17978, is
baumannii to form biofilms on six different biomedical relevant surfaces. Lett. Appl.
involved in surface motility and biofilm formation in this bacterium.
Microbiol. 63 (4), 233–239.
MicrobiologyOpen 6 (5), e00510. https://doi.org/10.1002/mbo3.510. In press.
Groisman, E., 2016. Feedback control of two-component regulatory systems. Annu. Rev.
Chin, C., Gregg, K., Napier, B., Ernst, R., Weiss, D., 2015. A PmrB-Regulated deacetylase
Microbiol. 70 (1), 103–124.
required for lipid a modification and polymyxin resistance in Acinetobacter
baumannii. Antimicrob. Agents Chemother. 59 (12), 7911–7914.

10
H.J. Mea et al. Microbiological Research 247 (2021) 126722

Harding, C., Kinsella, R., Palmer, L., Skaar, E., Feldman, M., 2016. Medically relevant Loehfelm, T., Luke, N., Campagnari, A., 2008. Identification and characterization of an
Acinetobacter species require a type II secretion system and specific membrane- Acinetobacter baumannii biofilm-associated protein. J. Bacteriol. 190 (3),
associated chaperones for the export of multiple substrates and full virulence. PLoS 1036–1044.
Pathog. 12 (1) p. e1005391. Lonergan, Z., Nairn, B., Wang, J., Hsu, Y., Hesse, L., Beavers, W., et al., 2019. An
Harris, G., Kuo Lee, R., Lam, C., Kanzaki, G., Patel, G., Xu, H., Chen, W., 2013. A mouse Acinetobacter baumannii, zinc-regulated peptidase maintains cell wall integrity
model of Acinetobacter baumannii-associated pneumonia using a clinically isolated during immune-mediated nutrient sequestration. Cell Rep. 26 (8), 2009–2018 e6.
hypervirulent strain. Antimicrob. Agents Chemother. 57 (8), 3601–3613. Luo, L., Wu, L., Xiao, Y., Zhao, D., Chen, Z., Kang, M., et al., 2015. Enhancing pili
Hospenthal, M., Costa, T., Waksman, G., 2017. A comprehensive guide to pilus assembly and biofilm formation in Acinetobacter baumannii ATCC19606 using non-
biogenesis in Gram-negative bacteria. Nat. Rev. Microbiol. 15 (6), 365–379. native acyl-homoserine lactones. BMC Microbiol. 15 (1).
Hoyle, B., Costerton, J., 1991. Bacterial Resistance to Antibiotics: the Role of Biofilms. Mattick, J., 2002. Type IV pili and twitching motility. Annu. Rev. Microbiol. 56 (1),
Progress In Drug Research / Fortschritte Der Arzneimittelforschung / Progrès Des 289–314.
Recherches Pharmaceutiques, pp. 91–105. May, H., Yu, J., Shrihari, S., Seshu, J., Klose, K., Cap, A., et al., 2019. Thioredoxin
Jarrell, K., McBride, M., 2008. The surprisingly diverse ways that prokaryotes move. Nat. modulates cell surface hydrophobicity in Acinetobacter baumannii. Front. Microbiol.
Rev. Microbiol. 6 (6), 466–476. 10.
Jin, J., Kwon, S., Moon, D., Gurung, G., Lee, J., Kim, S., et al., 2011. Acinetobacter McQueary, C., Kirkup, B., Si, Y., Barlow, M., Actis, L., Craft, D., Zurawski, D., 2012.
baumannii secretes cytotoxic outer membrane protein A via outer membrane Extracellular stress and lipopolysaccharide modulate Acinetobacter baumannii
vesicles. PLoS One 6 (2), e17027. https://doi.org/10.1371/journal.pone.0017027. surface-associated motility. J. Microbiol. 50 (3), 434–443.
In press. Montaña, S., Vilacoba, E., Traglia, G., Almuzara, M., Pennini, M., Fernández, A., et al.,
Joly-Guillou, M., Wolff, M., Pocidalo, J., Walker, F., Carbon, C., 1997. Use of a new 2015. Genetic variability of AdeRS two-component system associated with
mouse model of Acinetobacter baumannii pneumonia to evaluate the postantibiotic tigecycline resistance in XDR-Acinetobacter baumannii isolates. Curr. Microbiol. 71
effect of imipenem. Antimicrob. Agents Chemother. 41 (2), 345–351. (1), 76–82.
Josenhans, C., Suerbaum, S., 2002. The role of motility as a virulence factor in bacteria. Moriel, D., Beatson, S., Wurpel, D., Lipman, J., Nimmo, G., Paterson, D., Schembri, M.,
Int. J. Med. Microbiol. 291 (8), 605–614. 2013. Identification of novel vaccine candidates against multidrug-resistant
Jun, S., Lee, J., Kim, B., Kim, S., Park, T., Lee, J., Lee, Y., 2013. Acinetobacter baumannii Acinetobacter baumannii. PLoS One 8 (10) p. e77631.
outer membrane vesicles elicit a potent innate immune response via membrane Nait Chabane, Y., Marti, S., Rihouey, C., Alexandre, S., Hardouin, J., Lesouhaitier, O.,
proteins. PLoS One 8 (8) p. e71751. et al., 2014. Characterisation of pellicles formed by Acinetobacter baumannii at the
Khadke, S., Lee, J., Woo, J., Lee, J., 2019. Inhibitory effects of Honokiol and magnolol on air-liquid interface. PLoS One 9 (10) p. e111660.
biofilm formation by Acinetobacter baumannii. Biotechnol. Bioprocess Eng. 24 (2), Noto, M., Boyd, K., Burns, W., Varga, M., Peek, R., Skaar, E., 2015. Toll-like receptor 9
359–365. contributes to defense against Acinetobacter baumannii infection. Infect. Immun. 83
khelissa, S., Jama, C., Abdallah, M., Boukherroub, R., Faille, C., Chihib, N., 2017. Effect (10), 4134–4141.
of incubation duration, growth temperature, and abiotic surface type on cell surface Pachón-Ibáñez, M., Docobo-Pérez, F., López-Rojas, R., Dominguez-Herrera,
́ J., Jiménez-
properties, adhesion and pathogenicity of biofilm-detached Staphylococcus aureus Mejias, M., Garcia-Curiel,
́ A., et al., 2010. Efficacy of rifampin and its combinations
cells. AMB Express 7 (1). with imipenem, sulbactam, and colistin in experimental models of infection caused
Kim, S., Oh, M., Jun, S., Jeon, H., Kim, S., Kim, K., et al., 2016. Outer membrane Protein by Imipenem-Resistant Acinetobacter baumannii. Antimicrob. Agents Chemother. 54
A plays a role in pathogenesis of Acinetobacter nosocomialis. Virulence 7 (4), (3), 1165–1172.
413–426. Pakharukova, N., Tuittila, M., Paavilainen, S., Zavialov, A., 2017. Methylation,
Kim, S., Kim, M., Kim, S., Son, J., Kim, S., Lee, Y., et al., 2019. The sensor kinase BfmS crystallization and SAD phasing of the Csu pilus CsuC–CsuE chaperone–adhesin
controls production of outer membrane vesicles in Acinetobacter baumannii. BMC subunit pre-assembly complex from Acinetobacter baumannii. Acta Crystallogr. F
Microbiol. 19 (1). Struct. Biol. Commun. 73 (8), 450–454.
Knapp, S., Wieland, C., Florquin, S., Pantophlet, R., Dijkshoorn, L., Tshimbalanga, N., Pakharukova, N., Tuittila, M., Paavilainen, S., Malmi, H., Parilova, O., Teneberg, S.,
et al., 2006. Differential roles of CD14 and toll-like receptors 4and 2 in et al., 2018. Structural basis for Acinetobacter baumannii biofilm formation. Proc.
MurineAcinetobacterPneumonia. Am. J. Respir. Crit. Care Med. 173 (1), 122–129. Natl. Acad. Sci. 115 (21), 5558–5563.
Ko, W., Lee, H., Chiang, S., Yan, J., Wu, J., Lu, C., Chuang, Y., 2004. In vitro and in vivo Palmer, L., Green, E., Sheldon, J., Skaar, E., 2019. Assessing Acinetobacter baumannii
activity of meropenem and sulbactam against a multidrug-resistant Acinetobacter virulence and persistence in a murine model of lung infection. Methods Mol. Biol.
baumannii strain. J. Antimicrob. Chemother. 53 (2), 393–395. 289–305.
Koenigs, A., Zipfel, P., Kraiczy, P., 2015. Translation elongation factor tuf of Park, Y., Lee, J., Ko, K., 2015. Transcriptomic analysis of colistin-susceptible and colistin-
Acinetobacter baumannii is a plasminogen-binding protein. PLoS One 10 (7) p. resistant isolates identifies genes associated with colistin resistance in Acinetobacter
e0134418. baumannii. Clin. Microbiol. Infect. 21 (8), 765.e1–765.e7.
Krasowska, A., Sigler, K., 2014. How microorganisms use hydrophobicity and what does Pedersen, R., Grønnemose, R., Stærk, K., Asferg, C., Andersen, T., Kolmos, H., et al.,
this mean for human needs? Front. Cell. Infect. Microbiol. 4. 2018. A method for quantification of epithelium colonization capacity by pathogenic
Kröger, C., Kary, S., Schauer, K., Cameron, A., 2017. Genetic regulation of virulence and Bacteria. Front. Cell. Infect. Microbiol. 8.
antibiotic resistance in Acinetobacter baumannii. Genes 8 (1), 12. Peleg, A., Seifert, H., Paterson, D., 2008. Acinetobacter baumannii: emergence of a
Kwon, H., Kim, S., Oh, M., Shin, M., Lee, J., 2019. Distinct role of outer membrane successful pathogen. Clin. Microbiol. Rev. 21 (3), 538–582.
protein A in the intrinsic resistance of Acinetobacter baumannii and Acinetobacter Peleg, A., Jara, S., Monga, D., Eliopoulos, G., Moellering, R., Mylonakis, E., 2009.
nosocomialis. Infect. Genet. Evol. 67, 33–37. Galleria mellonella as a model system to study Acinetobacter baumannii
Lázaro-Díez, M., Navascués-Lejarza, T., Remuzgo-Martínez, S., Navas, J., Icardo, J., pathogenesis and therapeutics. Antimicrob. Agents Chemother. 53 (6), 2605–2609.
Acosta, F., et al., 2016. Acinetobacter baumannii and A. Pittii clinical isolates lack Pendleton, J., Gorman, S., Gilmore, B., 2013. Clinical relevance of the ESKAPE
adherence and cytotoxicity to lung epithelial cells in vitro. Microbes Infect. 18 (9), pathogens. Expert Rev. Anti. Ther. 11 (3), 297–308.
559–564. Pérez, A., Merino, M., Rumbo-Feal, S., Álvarez-Fraga, L., Vallejo, J., Beceiro, A., et al.,
Lázaro-Díez, M., Chapartegui-González, I., Redondo-Salvo, S., Leigh, C., Merino, D., 2016. The FhaB/FhaC two-partner secretion system is involved in adhesion
Segundo, D., et al., 2017. Human neutrophils phagocytose and kill Acinetobacter ofAcinetobacter baumanniiAbH12O-A2 strain. Virulence 8 (6), 959–974.
baumannii and A. Pittii. Sci. Rep. 7 (1). Pérez-Varela, M., Corral, J., Vallejo, J., Rumbo-Feal, S., Bou, G., Aranda, J., Barbé, J.,
Lázaro-Díez, M., Chapartegui-González, I., Suberbiola, B., Ocejo-Vinyals, J., López- 2017. Mutations in the β-Subunit of the RNA polymerase impair the surface-
Hoyos, M., Ramos-Vivas, J., 2020. Gene expression profiling in human neutrophils associated motility and virulence of Acinetobacter baumannii. Infect. Immun. 85 (8).
after infection with Acinetobacter baumannii in vitro. PLoS One 15 (11) p. Pestrak, M., Wozniak, D., 2020. Regulation of cyclic di-GMP signaling in Pseudomonas
e0242674. aeruginosa. Microbial Cyclic Di-Nucleotide Signal. 471–486.
Lee, J., Oh, J., Kim, K., Jeong, Y., Park, J., CHO, J., 2001. Apoptotic cell death induced by Petersen, K., Riddle, M., Danko, J., Blazes, D., Hayden, R., Tasker, S., Dunne, J., 2007.
Acinetobacter baumannii in epithelial cells through caspase-3 activation note. APMIS Trauma-related infections in battlefield casualties from Iraq. Ann. Surg. 245 (5),
109 (10), 679–684. 803–811.
Lee, J., Koerten, H., van den Broek, P., Beekhuizen, H., Wolterbeek, R., van den Petrova, O., Sauer, K., 2012. Sticky situations: key components that control bacterial
Barselaar, M., et al., 2006. Adherence of Acinetobacter baumannii strains to human surface attachment. J. Bacteriol. 194 (10), 2413–2425.
bronchial epithelial cells. Res. Microbiol. 157 (4), 360–366. Piepenbrink, K., Lillehoj, E., Harding, C., Labonte, J., Zuo, X., Rapp, C., et al., 2016.
Lee, H., Koh, Y., Kim, J., Lee, J., Lee, Y., Seol, S., et al., 2008. Capacity of multidrug- Structural diversity in the type IV pili of multidrug-resistant Acinetobacter. J. Biol.
resistant clinical isolates of Acinetobacter baumannii to form biofilm and adhere to Chem. 291 (44), 22924–22935.
epithelial cell surfaces. Clin. Microbiol. Infect. 14 (1), 49–54. Pour, N., Dusane, D., Dhakephalkar, P., Zamin, F., Zinjarde, S., Chopade, B., 2011.
Lees-Miller, R., Iwashkiw, J., Scott, N., Seper, A., Vinogradov, E., Schild, S., Feldman, M., Biofilm formation byAcinetobacter baumanniistrains isolated from urinary tract
2013. A common pathway forO-linked protein-glycosylation and synthesis of capsule infection and urinary catheters. FEMS Immunol. Med. Microbiol. 62 (3), 328–338.
inAcinetobacter baumannii. Mol. Microbiol. 89 (5), 816–830. Qiu, H., KuoLee, R., Harris, G., Chen, W., 2008. Role of NADPH phagocyte oxidase in
Lin, M., Lin, Y., Yeh, H., Lan, C., 2014. Role of the BaeSR two-component system in the host defense against acute respiratory Acinetobacter baumannii infection in mice.
regulation of Acinetobacter baumannii adeAB genes and its correlation with Infect. Immun. 77 (3), 1015–1021.
tigecycline susceptibility. BMC Microbiol. 14 (1), 119. Qiu, H., KuoLee, R., Harris, G., Van Rooijen, N., Patel, G., Chen, W., 2012. Role of
Liou, M., Soo, P., Ling, S., Kuo, H., Tang, C., Chang, K., 2014. The sensor kinase BfmS macrophages in early host resistance to respiratory Acinetobacter baumannii
mediates virulence in Acinetobacter baumannii. J. Microbiol. Immunol. Infect. 47 infection. PLoS One 7 (6) p. e40019.
(4), 275–281. Raorane, C., Lee, J., Kim, Y., Rajasekharan, S., García-Contreras, R., Lee, J., 2019.
Antibiofilm and Antivirulence Efficacies of flavonoids and curcumin against
Acinetobacter baumannii. Front. Microbiol. 10.

11
H.J. Mea et al. Microbiological Research 247 (2021) 126722

Rello, J., Kalwaje Eshwara, V., Lagunes, L., Alves, J., Wunderink, R., Conway-Morris, A., Tacconelli, E., Carrara, E., Savoldi, A., Harbarth, S., Mendelson, M., Monnet, D., et al.,
et al., 2018. A global priority list of the TOp TEn resistant Microorganisms (TOTEM) 2018. Discovery, research, and development of new antibiotics: the WHO priority
study at intensive care: a prioritization exercise based on multi-criteria decision list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect. Dis. 18 (3),
analysis. Eur. J. Clin. Microbiol. Infect. Dis. 38 (2), 319–323. 318–327.
Repizo, G., Gagné, S., Foucault-Grunenwald, M., Borges, V., Charpentier, X., Talà, L., Fineberg, A., Kukura, P., Persat, A., 2019. Pseudomonas aeruginosa orchestrates
Limansky, A., et al., 2015. Differential role of the T6SS in Acinetobacter baumannii twitching motility by sequential control of type IV pili movements. Nat. Microbiol. 4
virulence. PLoS One 10 (9) p. e0138265. (5), 774–780.
Richmond, G., Evans, L., Anderson, M., Wand, M., Bonney, L., Ivens, A., et al., 2016. The Thibau, A., Dichter, A., Vaca, D., Linke, D., Goldman, A., Kempf, V., 2019.
Acinetobacter baumannii two-component system AdeRS regulates genes required for Immunogenicity of trimeric autotransporter adhesins and their potential as vaccine
multidrug efflux, biofilm formation, and virulence in a strain-specific manner. Mbio targets. Med. Microbiol. Immunol. 209 (3), 243–263.
7 (2). Tipton, K., Rather, P., 2017. An ompR-envZ two-component system ortholog regulates
Ronish, L., Lillehoj, E., Fields, J., Sundberg, E., Piepenbrink, K., 2018. The structure of phase variation, osmotic tolerance, motility, and virulence in Acinetobacter
PilA from Acinetobacter baumannii AB5075 suggests a mechanism for functional baumannii strain AB5075. J. Bacteriol. 199 (3).
specialization in Acinetobacter type IV pili. J. Biol. Chem. 294 (1), 218–230. Tobin, D., May, R., Wheeler, R., 2012. Zebrafish: a see-through host and a fluorescent
Rosenberg, E., DeLong, E., Lory, S., Stackebrandt, E., Thompson, F., 2013. The toolbox to probe host–pathogen interaction. PLoS Pathog. 8 (1) p. e1002349.
Prokaryotes: Human Microbioogy, 4th edn. Springer, Berlin, Heidelberg, Tomaras, A., Dorsey, C., Edelmann, R., Actis, L., 2003. Attachment to and biofilm
pp. 107–123. formation on abiotic surfaces by Acinetobacter baumannii: involvement of a novel
Rumbo, C., Tomás, M., Fernández Moreira, E., Soares, N., Carvajal, M., Santillana, E., chaperone-usher pili assembly system. Microbiology 149 (12), 3473–3484.
et al., 2014. The Acinetobacter baumannii Omp33-36 porin is a virulence factor that Traglia, G., Chiem, K., Quinn, B., Fernandez, J., Montaña, S., Almuzara, M., et al., 2018.
induces apoptosis and modulates autophagy in human cells. Infect. Immun. 82 (11), Genome sequence analysis of an extensively drug-resistant Acinetobacter baumannii
4666–4680. indigo-pigmented strain depicts evidence of increase genome plasticity. Sci. Rep. 8
Rumbo-Feal, S., Gómez, M., Gayoso, C., Álvarez-Fraga, L., Cabral, M., Aransay, A., et al., (1).
2013. Whole transcriptome analysis of Acinetobacter baumannii assessed by RNA- Tsai, Y., Fung, C., Lin, J., Chen, J., Chang, F., Chen, T., Siu, L., 2011. Klebsiella
Sequencing reveals different mRNA expression profiles in biofilm compared to pneumoniaeOuter membrane porins OmpK35 and OmpK36 play roles in both
planktonic cells. PLoS One 8 (8) p. e72968. antimicrobial resistance and virulence. Antimicrob. Agents Chemother. 55 (4),
Russo, T., Luke, N., Beanan, J., Olson, R., Sauberan, S., MacDonald, U., et al., 2010. The 1485–1493.
K1 capsular polysaccharide of Acinetobacter baumannii strain 307-0294 is a major Tsai, C., Loh, J., Proft, T., 2016. Galleria mellonella infection models for the study of
virulence factor. Infect. Immun. 78 (9), 3993–4000. bacterial diseases and for antimicrobial drug testing. Virulence 7 (3), 214–229.
Sahu, P., Iyer, P., Oak, A., Pardesi, K., Chopade, B., 2012. Characterization of eDNA from Vallejo, J., Beceiro, A., Rumbo-Feal, S., Rodríguez-Palero, M., Russo, T., Bou, G., 2015.
the Clinical StrainAcinetobacter baumanniiAIIMS 7 and its Role in Biofilm Optimisation of the Caenorhabditis elegans model for studying the pathogenesis of
Formation. Sci. World J. 2012, 1–10. opportunistic Acinetobacter baumannii. Int. J. Antimicrob. Agents.
Sánchez-Encinales, V., Álvarez-Marín, R., Pachón-Ibáñez, M., Fernández-Cuenca, F., van der Sar, A., Appelmelk, B., Vandenbroucke-Grauls, C., Bitter, W., 2004. A star with
Pascual, A., Garnacho-Montero, J., et al., 2017. Overproduction of outer membrane stripes: zebrafish as an infection model. Trends Microbiol. 12 (10), 451–457.
protein A (OmpA) by Acinetobacter baumannii is a risk factor for nosocomial van Faassen, H., KuoLee, R., Harris, G., Zhao, X., Conlan, J., Chen, W., 2007. Neutrophils
pneumonia, bacteremia and mortality increase. J. Infect. Dis. p. jix010. play an important role in host resistance to respiratory infection with Acinetobacter
Sato, Y., Unno, Y., Kawakami, S., Ubagai, T., Ono, Y., 2017. Virulence characteristics of baumannii in mice. Infect. Immun. 75 (12), 5597–5608.
Acinetobacter baumannii clinical isolates vary with the expression levels of omps", Vij, R., Crawford, C., Casadevall, A., 2019. Variation in cell surface hydrophobicity
in. J. Med. Microbiol. 66 (2), 203–212. among Cryptococcus neoformans strains influences interactions with amoeba.
Silverman, J., Brunet, Y., Cascales, E., Mougous, J., 2012. Structure and regulation of the Msphere.
type VI secretion system. Annu. Rev. Microbiol. 66 (1), 453–472. Vijayakumar, S., Rajenderan, S., Laishram, S., Anandan, S., Balaji, V., Biswas, I., 2016.
Skerniškytė, J., Krasauskas, R., Péchoux, C., Kulakauskas, S., Armalytė, J., Biofilm formation and motility depend on the nature of the Acinetobacter baumannii
Sužiedėlienė, E., 2019. Surface-related features and virulence among Acinetobacter clinical isolates. Front. Public Health 4.
baumannii clinical isolates belonging to international clones I and II. Front. Weidensdorfer, M., Chae, J., Makobe, C., Stahl, J., Averhoff, B., Müller, V., et al., 2016.
Microbiol. 9. Analysis of endothelial adherence of Bartonella henselae and Acinetobacter
Skiebe, E., de Berardinis, V., Morczinek, P., Kerrinnes, T., Faber, F., Lepka, D., et al., baumannii using a dynamic HumanEx vivo infection model. Infect. Immun. 84 (3),
2012. Surface-associated motility, a common trait of clinical isolates of 711–722.
Acinetobacter baumannii, depends on 1,3-diaminopropane. Int. J. Med. Microbiol. Whitchurch, C., Tolker-Nielsen, T., Rages, P., Mattick, J., 2002. Extracellular DNA
302 (3), 117–128. required for bacterial biofilm formation. Science 295 (5559), pp. 1487-1487.
Smani, Y., Docobo-Pérez, F., López-Rojas, R., Domínguez-Herrera, J., Ibáñez- Wilharm, G., Piesker, J., Laue, M., Skiebe, E., 2013. DNA uptake by the nosocomial
Martínez, J., Pachón, J., 2012a. Platelet-activating Factor Receptor Initiates Contact pathogen Acinetobacter baumannii occurs during movement along wet surfaces.
of Acinetobacter baumanniiExpressing Phosphorylcholine with Host Cells. J. Biol. J. Bacteriol. 195 (18), 4146–4153.
Chem. 287 (32), 26901–26910. Wood, C., Ohneck, E., Edelmann, R., Actis, L., 2018. A light-regulated type I pilus
Smani, Y., McConnell, M., Pachón, J., 2012b. Role of fibronectin in the adhesion of contributes toAcinetobacter baumanniiBiofilm, motility, and virulence functions.
Acinetobacter baumannii to host cells. PLoS One 7 (4) p. e33073. Infect. Immun. 86 (9).
Smani, Y., Dominguez-Herrera, J., Pachon, J., 2013. Association of the outer membrane Yang, L., Barken, K., Skindersoe, M., Christensen, A., Givskov, M., Tolker-Nielsen, T.,
protein Omp33 with fitness and virulence of Acinetobacter baumannii. J. Infect. Dis. 2007. Effects of iron on DNA release and biofilm development by Pseudomonas
208 (10), 1561–1570. aeruginosa. Microbiology 153 (5), 1318–1328.
Smith, M., Des Etages, S., Snyder, M., 2004. Microbial synergy via an ethanol-triggered Yang, C., Su, P., Moi, S., Chuang, L., 2019. Biofilm formation in Acinetobacter
pathway. Mol. Cell. Biol. 24 (9), 3874–3884. baumannii: genotype-phenotype correlation. Molecules 24 (10), 1849.
Smith, M., Gianoulis, T., Pukatzki, S., Mekalanos, J., Ornston, L., Gerstein, M., Young, E., Bonds, G., Karatan, E., 2020. Signals modulating cyclic di-GMP pathways in
Snyder, M., 2007. New insights into Acinetobacter baumannii pathogenesis revealed Vibrio cholerae. Microb. Cyclic Di-Nucl. Signal. 357–378.
by high-density pyrosequencing and transposon mutagenesis. Genes Dev. 21 (5), Zhang, X., de Boer, L., Stockhammer, O., Grijpma, D., Spaink, H., Zaat, S., 2019.
601–614. A zebrafish embryo model for in vivo visualization and intravital analysis of
Sun, J., Chiang, Y., Shang, H., Perng, C., Yang, Y., Chiueh, T., 2017. Phenotype biomaterial-associated Staphylococcus aureus infection. J. Vis. Exp. (143) p. e58523.
microarray analysis of the AdeRS two-component system in Acinetobacter Zhang, L., Liang, W., Xu, S., Mei, J., Di, Y., Lan, H., et al., 2020. CarO promotes adhesion
baumannii. Eur. J. Clin. Microbiol. Infect. Dis. 36 (12), 2343–2353. and colonization of Acinetobacter baumannii through inhibiting NF-кB pathways.
Int. J. Clin. Exp. Med. 12 (3), 2518–2524.

12

You might also like